WO2017152874A1 - 一种脲类化合物、其制备方法及其医药用途 - Google Patents

一种脲类化合物、其制备方法及其医药用途 Download PDF

Info

Publication number
WO2017152874A1
WO2017152874A1 PCT/CN2017/076265 CN2017076265W WO2017152874A1 WO 2017152874 A1 WO2017152874 A1 WO 2017152874A1 CN 2017076265 W CN2017076265 W CN 2017076265W WO 2017152874 A1 WO2017152874 A1 WO 2017152874A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
tert
butyl
benzimidazol
urea
Prior art date
Application number
PCT/CN2017/076265
Other languages
English (en)
French (fr)
Inventor
司聚同
姜美锋
Original Assignee
恩瑞生物医药科技(上海)有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 恩瑞生物医药科技(上海)有限公司 filed Critical 恩瑞生物医药科技(上海)有限公司
Priority to JP2018547295A priority Critical patent/JP6927994B2/ja
Priority to EP17762560.5A priority patent/EP3424924B1/en
Priority to RU2018131539A priority patent/RU2741596C2/ru
Priority to BR112018016554-9A priority patent/BR112018016554B1/pt
Priority to US16/078,054 priority patent/US10647680B2/en
Priority to DK17762560.5T priority patent/DK3424924T3/da
Priority to CN201780026742.3A priority patent/CN109071523B/zh
Priority to AU2017230437A priority patent/AU2017230437B2/en
Priority to KR1020187028521A priority patent/KR102430942B1/ko
Publication of WO2017152874A1 publication Critical patent/WO2017152874A1/zh
Priority to HK19100900.8A priority patent/HK1258545A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • 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
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/28Oxygen atom
    • C07D473/30Oxygen atom attached in position 6, e.g. hypoxanthine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • 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

Definitions

  • the present invention relates to a novel urea compound, a process for the preparation thereof, and a pharmaceutical composition containing the same, and its use as a FLT3 tyrosine protein kinase inhibitor, particularly in the prevention and/or treatment of cancer.
  • Cancer is one of the major diseases of human clinical death, especially malignant tumors such as lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, intestinal cancer, esophageal cancer and leukemia, and the mortality rate is extremely high. To date, there are no effective methods and drugs to prevent, cure and eradicate cancer. There is an urgent need for high-quality anticancer drugs and treatment methods with good specificity, high activity, low toxicity, and no drug resistance.
  • Leukemia a blood cancer
  • Leukemia is a clonal malignant disease of hematopoietic stem cells. Due to the ability of leukemia cells to lose their ability to differentiate into mature functional blood cells, they are arrested at different stages of cell development, and leukemia cells accumulate in bone marrow and other hematopoietic tissues. Infiltrating other organs and tissues, normal hematopoiesis is inhibited, and clinical manifestations include anemia, hemorrhage, infection, and infiltration of various organs. The incidence of leukemia accounts for 6/7 of all tumors, especially in children and the elderly. Leukemia is a heterogeneous malignant tumor of the cell. It has a wide variety of causes and complex etiology. It has different clinical characteristics.
  • AML acute myeloid leukemia
  • the clinical treatment of blood cancer can be performed by various methods such as chemotherapy, radiotherapy, immunotherapy, targeted therapy, differentiation therapy, and bone marrow/stem cell transplantation
  • the clinical treatment of acute myeloid leukemia has hardly changed in the past 40 years.
  • the standard treatment method still relies on induction remission as the "7+3" basic therapy (daunorubicin 25-45mg/m 2 , IV 1-3 days, cytarabine 100mg/m 2 , IV 1-7 Day, except for acute promyelocytic leukemia APL).
  • Targeted therapy and tumor immunotherapy are the development direction of clinical cancer treatment today, in which targeted drug therapy has good specificity, small side effects and obvious curative effect.
  • Many targeted drugs have been successfully applied to different types of cancer including certain types of leukemia such as Gleevec (CML).
  • CML Gleevec
  • the mutation rate of genes involved in cell signaling pathway accounts for about 50-60%, DNA methylation-related genes. Abnormality accounted for 44%, chromosome modification gene variation was about 30%, pith The transcription factor gene abnormality accounted for 20-25%, the transcription factor fusion gene incidence rate was about 18%, and the tumor suppressor gene mutation accounted for 14%.
  • the most common genetic abnormalities are FLT3-ITD (19-28%), FLT3-TKD (5-10%), NPM1 (mutation is 27-35%), DNMTA (mutation is 26%), NRAS.
  • Protein kinase is a protein amino acid phosphorylase essential for important physiological functions such as cell growth, development, differentiation, metabolism, aging and apoptosis, and is mainly composed of transmembrane and cytoplasmic protein kinases. Abnormalities in protein kinases can directly lead to clinically different types of diseases such as cancer, inflammation, the immune system, the nervous system, and cardiovascular and cerebrovascular diseases.
  • protein kinases such as EGFR, HER2/3/4, VEGFRs, PDGFR, c-MET, IGF-1R, FGFRs, CSF-1R, TRK receptors, Ephrin receptors, TAM receptors , TIE-2, FLT-3, RET, ALK, BCR-ABL, JAKs, SRC, FAK, BTK, SYK, BLK, CDK, PI3K, MEK/RAS/RAF, etc. have been identified as target protein molecules for different clinical diseases. .
  • FMS-Like Tyrosine Kinase 3 FMS-Like Tyrosine Kinase 3 (FLT3) or fetal liver kinase-2 (FLK-2) or human stem cell kinase-1 (STK) -1
  • FLK-3 fetal liver kinase-2
  • STK human stem cell kinase-1
  • the kinase family includes the colony stimulating factor 1 receptor CSF1R, the platelet-derived growth factor PDGFR ⁇ / ⁇ , and the stem cell factor receptor KIT.
  • FLT3 gene In the normal growth and development physiological environment, the expression of FLT3 gene is mainly concentrated in the early development of brain, liver, placenta, gonads and hematopoietic cells.
  • FLT3 gene and its ligand gene FLT3-L are highly expressed.
  • FLT3-L binds to FLT3 to induce autophosphorylation of FLT3 protein, activates FLT3 enzyme activity and mediates downstream Signaling pathways such as PI3K, JAK/STAT and RAS are involved in the biological functions of growth, development, proliferation and differentiation of blood cells (Drexler HG et al., FLT3: receptor and ligand. Growth Factors.
  • the FLT3 tyrosine kinase activating mutation is one of the most important mutations in AML and one of the major causes of AML. Due to the selective growth advantage of FLT3-ITD clones, it is difficult to cure such leukemias by using common chemotherapeutic drugs alone. At the same time, patients with such leukemias have strong tolerance to chemotherapy drugs, and the clinical outcome is poor.
  • FLT3 tyrosine kinase activating mutation has become an important target for AML targeted therapy (Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002; 100 (5): 1532-1542. Kiyoi H. et al., Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product. Leukemia. 1998; 12(9): 1333-1337).
  • FLT3-ITD and FLT3-TKD inhibitors have been one of the most closely developed drug developments in the field of AML research. To date, preclinical studies have found that nearly 100 different types of small molecule compounds are capable of selective/non-selective inhibition or partial inhibition of FLT3 protein kinase activity in vitro and in vitro proliferation of FLT3 mutant-positive leukemia cell lines or leukemia patient sample cells.
  • Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid Leukemia.Blood.2013;122(22):3607-15;Safaian NN et al., Sorafenib (Nexavar) induces molecular remission and regression of extramedullary disease in a patient with FLT3-ITD+acute myeloid leukemia.Leuk Res.2009;33 (2): 348-50; Zhang W et al, Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia.
  • the three human leukemia cell lines, MV4-11, MOLM-13 and MOLM-14, are the most widely used FLT3-ITD expressing cells.
  • MV4-11 cells contain FLT3 ITD homozygous mutation (+/+), which belongs to human acute lymphatic myelomonocytic leukemia.
  • MOLM-13 and MOLM-14 are sister cell lines from the same patient, containing FLT3 ITD/WT heterozygous mutation (+/-), belonging to human acute myeloid leukemia (Quentmeier H et al, FLT3 mutations in acute myeloid leukemia cell lines . Leukemia. 2003; 17(1): 120-124).
  • the present invention utilizes (1) FLT3-ITD expression positive cell lines MV4-11 and MOLM-13; (2) FLT3 gene wild type high expression positive cell line RS4; 11; (3) other clinically common oncogene expression positive leukemia
  • FLT3-ITD expression positive cell lines MV4-11 and MOLM-13 FLT3 gene wild type high expression positive cell line RS4; 11; (3) other clinically common oncogene expression positive leukemia
  • cell lines and different types of solid tumor cell lines are dedicated to the development of a novel compound with high activity, high selectivity, good pharmaceutical properties and low side effects of metabolic parameters in vivo, which can be used as an effective FLT3 cheese.
  • a selective inhibitor of a protein kinase (activating mutation) for the prevention and/or treatment of cancer, particularly leukemia.
  • the present invention relates to a novel urea compound which is capable of effectively inhibiting the growth of the FLT3-ITD mutant-expressing leukemia cell lines MV4-11 and MOLM-13, and inducing apoptosis, the GI50 of which is the subnanomolar range, and There was no significant inhibitory effect on the growth of wild-type FLT3 wild-type or normal or non-expressing cancer cell lines.
  • the compounds of the present invention are capable of inhibiting tumor growth in FLT3-ITD leukemia cells MV4-11 xenograft animals efficiently, rapidly, and in a concentration-dependent manner. Further pharmacokinetic and pharmacological studies have found that the compounds of the invention exhibit good pharmaceutical properties in rats.
  • the present invention provides a compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof,
  • X, Y are each independently selected from N or C-BR 1 ;
  • A, A 1 are each independently selected from N or C-BR 1 ;
  • W and Z are each independently selected from N or C-BR 1 ;
  • B is the same or different and is each independently selected from: hydrogen, halogen, alkyl, alkenyl, alkynyl, cyano, cycloalkyl, heterocyclyl, aryl, or heteroaryl.
  • the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl group is optionally further substituted with one or more Q groups;
  • B is the same or different and is each independently selected from: -O- or -NR 4 -; and R 1 are the same or different and are each independently selected from: hydrogen, alkyl, alkenyl, alkynyl , cycloalkyl, heterocyclic, aryl, heteroaryl, -R u OR x , -R u C(O)OR x , -R u N(R y )(R z ), -C(O) N(R y )(R z ), -R u S(O) n N(R y )(R z ), -R u S(O) n R x ; the alkyl group, alkenyl group, alkynyl group, The cycloalkyl, heterocyclic, aryl, heteroaryl group is optionally further selected from the group consisting of halogen, cyano, hydroxy, alkyl, alkoxy, hydroxy
  • G is selected from an aryl group, a heteroaryl group or a heterocyclic group, which is optionally further selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, Substituted with one or more groups of alkoxy, hydroxy, amino, acyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, wherein said alkyl, alkenyl, alkynyl, alkoxy Or an acyl group, a cycloalkyl group, a heterocyclic group, an aryl group or a heteroaryl group, optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkenyl, alkynyl, aryl, hydroxy, alkoxy, haloalkoxy, Substituted by one or more groups of a cycloalkyl group, an ester group, and a cyano group; or,
  • G and R 2 together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl group, which is optionally further selected from the group consisting of halogen, alkyl, and alkene.
  • Substituted by one or more groups of a group, an alkynyl group, an alkoxy group, a hydroxyl group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group wherein the alkyl group, the alkenyl group, and the alkynyl group are substituted Alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally further selected from the group consisting of halogen, alkyl, haloalkyl, alkenyl, alkynyl, aryl, hydroxy, alkoxy, haloalkoxy Substituted by one or more groups of a group, a cycloalkyl, alkeny
  • R 3 is selected from the group consisting of Q
  • R u is selected from a bond, an alkylene group, an alkenylene group, or an alkynylene group;
  • R x is selected from hydrogen, alkyl, hydroxyalkyl, haloalkyl, alkenyl, or alkynyl; or
  • R y and R z are each independently selected from hydrogen, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, or haloalkyl; or
  • R y and R z together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl group, which is optionally further selected from the group consisting of halogen, haloalkyl, alkyl, alkenyl and alkynyl. Substituted by one or more groups;
  • Q is selected from the group consisting of hydrogen, halogen, hydroxy, amino, alkyl, alkoxy, cycloalkyl, alkenyl, alkynyl, cyano, aryl, heterocyclyl or heteroaryl, said amino, alkyl, alkane
  • An oxy, cycloalkyl, alkenyl, alkynyl, aryl, heterocyclyl or heteroaryl group is optionally further substituted with one or more groups selected from the group consisting of hydroxyl, halogen, alkyl;
  • n is an integer from 0-2.
  • the compound of the formula (I) according to the invention or a pharmaceutically acceptable salt, solvate or prodrug thereof,
  • X, Y, A, and A 1 are selected from the following structures:
  • R 5 1 , R 5 2 , R 5 3 , R 5 4 , R 5 5 , R 5 6 , R 5 7 , R 5 8 and R 5 9 are each independently selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, alkane Oxy, alkenyl, alkynyl, -N(R y )(R z ), cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkoxy, alkenyl, alkynyl —N(R y )(R z ), cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally further selected from the group consisting of halogen, hydroxy, alkyl, alkoxy, haloalkyl, haloalkoxy, Substituted by one or more groups of a cycloalkyl group or an ester group.
  • R 1 , B, R y , R z are as defined in the formula (I).
  • the compound of the formula (I) according to the present invention or a pharmaceutically acceptable salt, solvate or prodrug thereof,
  • X, Y, A, and A 1 are selected from the following structures:
  • R 5 1 , R 5 2 , R 5 3 , R 5 4 and R 5 5 are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, -N(R y )(R z ), haloalkyl, haloalkane Oxygen.
  • R 1 , B, R y , R z are as defined in the formula (I).
  • the compound of the formula (I) according to the present invention or a pharmaceutically acceptable salt, solvate or prodrug thereof,
  • X, Y, A, and A 1 are selected from the following structures:
  • R 5 1 , R 5 2 and R 5 3 are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, -N(R y )(R z ), haloalkyl, haloalkoxy.
  • R 1 , B, R y , R z are as defined in the formula (I).
  • R 5 1 , R 5 2 , and R 5 3 are each independently selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, -N(R y )(R z ), C 1 a -C 6 haloalkyl group, a C 1 -C 6 haloalkoxy group, wherein R y and R z are each independently selected from hydrogen, C 1 -C 6 alkyl.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof according to the present invention wherein W and Z are selected from the following four modes :
  • W is CQ and Z is N;
  • Q is selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 3 -C 7 cycloalkyl, C 5 -C 7 aryl, 5- to 7-membered heterocyclic or 5- to 7-membered heteroaryl.
  • R 1 is absent
  • B is the same or different and is independently selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 Haloalkoxy.
  • B is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl;
  • B is the same or different and is each independently selected from: -O- or -NR 4 -, preferably -O-; and, R 1 are the same or different and are each independently selected from: hydrogen, C 1 a -C 10 alkyl group, which is optionally further selected from the group consisting of halogen, cyano, hydroxy, C 1 -C 6 alkoxy, 4-6-membered heterocyclic, C 5 -C 7 aryl, C 5 Substituting one or more groups of a -C 7 haloaryl group, a 5 to 7 membered heteroaryl group, a C 3 -C 6 cycloalkyl group, a 5 to 7 membered heteroaryl group, wherein the 4 to 6 membered hetero group
  • the cyclo group is preferably a 4- to 6-membered heterocyclic group containing oxygen or nitrogen; the C 5 -C 7 aryl or C 5 -C 7 haloaryl group is preferably a
  • R 4 is as defined in the formula (I).
  • B is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl;
  • R 1 When R 1 is present, B is the same or different and is each independently selected from: -O- or -NR 4 -, preferably -O-; and, R 1 are the same or different and are each independently selected from: -R u OR x , wherein R u is selected from C 1 -C 6 alkylene, R x is selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 haloalkyl;
  • R 4 is as defined in the formula (I).
  • B is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl;
  • B is the same or different and is each independently selected from: -O- or -NR 4 -, preferably -O-; and, R 1 are the same or different and are each independently selected from: -C(O N(R y )(R z ), wherein R y and R z are each independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1- C 6 haloalkoxy, C 3 -C 7 cycloalkyl; or,
  • R y and R z together with the nitrogen atom to which they are attached form a 5- to 7-membered heterocyclic group or a 5- to 7-membered heteroaryl group, preferably a 6-membered heterocyclic group or a 6-membered heteroaryl group, more preferably a morpholinyl group or a piperazine.
  • Pyridyl, piperazinyl, pyridyl, pyrimidinyl, the 5- to 7-membered heterocyclic or 5- to 7-membered heteroaryl optionally further selected from halogen, C 1 -C 6 alkyl, C 1 -C Substituted by one or more groups of 6 haloalkyl;
  • R 4 is as defined in the formula (I).
  • B is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl;
  • R 1 is present, B is the same or different and is each independently selected from: -O- or -NR 4 -, preferably -O-; and, R 1 are the same or different and are each independently selected from: -R u N (R y )(R z ), wherein R u is selected from C 1 -C 6 alkylene; R y and R z are each independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy a C 1 -C 6 haloalkyl group, a C 1 -C 6 haloalkoxy group, a C 3 -C 7 cycloalkyl group; or
  • R y and R z together with the nitrogen atom to which they are attached form a 5- to 7-membered heterocyclic group or a 5- to 7-membered heteroaryl group, preferably morpholinyl, piperidinyl, piperazinyl, azepanyl, a pyridyl group, a pyrimidinyl group, the 5- to 7-membered heterocyclic group or a 5- to 7-membered heteroaryl group, optionally further selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or Substituted by a variety of groups;
  • R 4 is as defined in the formula (I).
  • B is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl;
  • R 1 is present, B is the same or different and is each independently selected from: -O- or -NR 4 -, preferably -O-; and, R 1 are the same or different and are each independently selected from: -R u C (O)OR x , wherein: R u is selected from C 1 -C 6 alkylene; R x is selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 haloalkane base;
  • R 4 is as defined in the formula (I).
  • B is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl;
  • B is the same or different and is each independently selected from: -O- or -NR 4 -, preferably -O-; and, R 1 are the same or different and are each independently selected from: 5 to 7 Aryl or 5- to 7-membered heteroaryl, preferably thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, triazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrrole , N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, phenyl, pyridyl, pyrimidinyl, the 5 to 7-membered aryl or 5 to 7-membered heteroaryl optionally Further substituted with one or more groups selected from the group consisting of a C3-C6 cycloalkyl group, a 5- to
  • R 4 is as defined in the formula (I).
  • R 4 is selected from hydrogen or C 1 -C 6 alkyl, or R 4 and R 1 together with the nitrogen atom to which they are bonded form a 5- to 7-membered heterocyclic group or a 5- to 7-membered heteroaryl group, preferably piperidinyl or piperidin a pyridyl group, a morpholinyl group, a pyridyl group, a pyrimidinyl group, the 5- to 7-membered heterocyclic group or a 5- to 7-membered heteroaryl group, optionally further selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C Substitution of one or more groups of 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy.
  • G is selected from a C 5 -C 7 aryl group, a 5 to 7 membered heteroaryl group or a 5 to 7 membered heterocyclic group, preferably
  • the C 5 -C 7 aryl, 5- to 7-membered heteroaryl or 5- to 7-membered heterocyclic group is optionally further selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1- C 6 alkoxy, C 1 -C 6 haloalkoxy, hydroxy, amino, acyl, C 3 -C 7 cycloalkyl, 5- to 7-membered heterocyclic, C 5 -C7 aryl, 5-7 Substituted by one or more groups of a heteroaryl group; the C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group, a C 3 -C 7 cycloalkyl group, a 5-7 membered heterocyclic group a C 5 -C7 aryl group, a 5- to 7-membered heteroaryl group, optionally further selected from the group consisting of halogen, hydroxy, C 1
  • R 2 is not hydrogen
  • G and R 2 together with the nitrogen atom to which they are attached form a 5-7 membered heterocyclic group or a 5-7 membered heteroaryl group, preferably pyrrolyl, pyrazolyl, imidazolyl
  • the 5-7 membered heterocyclyl or 5-7 membered heteroaryl is optionally further selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 One or more groups of -C 6 haloalkoxy, hydroxy, amino, C 3 -C 7 cycloalkyl, 5- to 7-membered heterocyclic, C 5 -C7 aryl, 5- to 7-membered heteroaryl Replaced.
  • R 3 is selected from the group consisting of hydrogen, halogen, hydroxy, amino, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 3 -C a 7- cycloalkyl group, a cyano group, a C 5 -C 7 -membered aryl group, a 5- to 7-membered heterocyclic group or a 5- to 7-membered heteroaryl group.
  • Typical compounds of the formula (I) of the invention include, but are not limited to:
  • Another aspect of the present invention provides a process for the preparation of a compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, which comprises the steps of:
  • the compound of formula (II) is reacted with a compound of formula (III) under the action of a base at a suitable temperature, pH and a suitable solvent to provide a compound of formula (I);
  • the solvent is preferably THF, acetonitrile, dichloromethane, toluene, and the base is preferably triethylamine, N,N-diisopropylethylamine, DMAP, pyridine;
  • X, Y, A, A1, Z, W, R1, B, R2, G, R3 are as defined in the general formula (I).
  • Another aspect of the present invention provides a process for the preparation of a compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, which comprises the steps of:
  • the compound of formula (IV) is reacted with a compound of formula (V) under the action of a base at a suitable temperature, pH and a suitable solvent to provide a compound of formula (I);
  • the solvent is preferably THF, acetonitrile, dichloromethane, toluene, and the base is preferably triethylamine, N,N-diisopropylethylamine, DMAP, pyridine;
  • X, Y, A, A1, Z, W, R1, B, R2, G, R3 are as defined in the general formula (I).
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the formula (I) according to the present invention or a pharmaceutically acceptable salt, solvate or prodrug thereof as an active ingredient, and a Or a plurality of pharmaceutically acceptable carriers.
  • the invention further relates to the use of a compound of the formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising the same, for the preparation of a FLT3 tyrosine protein kinase inhibitor.
  • the present invention further relates to a compound represented by the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition containing the same, which is prepared for use in the prevention and/or treatment of mammals including humans
  • drugs for cancer include, but are not limited to, non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, gastric cancer, breast cancer, pancreatic cancer, liver cancer, and colon cancer.
  • the present invention further relates to a compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising the same, which is useful as a FLT3 tyrosine protein kinase inhibitor use.
  • the present invention further relates to a compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition containing the same, which is useful as a prophylactic and/or therapeutic mammal, including a human
  • drugs for cancer include, but are not limited to, non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, gastric cancer, breast cancer, pancreatic cancer, liver cancer, and colon cancer.
  • the invention further relates to a method of inhibiting FLT3 tyrosine protein kinase comprising administering to a patient in need thereof an inhibitory effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or pharmaceutically acceptable salt thereof A drug, or a pharmaceutical composition containing the same.
  • the invention further relates to a method of preventing and/or treating cancer in a mammal, including a human, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof , a solvate or a prodrug, or a pharmaceutical composition containing the same.
  • cancers include, but are not limited to, non-solid tumors such as leukemia, solid tumors such as skin cancer, melanoma, lung cancer, gastric cancer, breast cancer, pancreatic cancer, liver cancer, and colon cancer.
  • alkyl refers to a saturated aliphatic hydrocarbon group, including straight chain and branched chain groups of 1 to 20 carbon atoms. It includes a linear or branched alkyl group having from 1 to 18 carbon atoms, preferably from 1 to 10 carbon atoms, more preferably from 1 to 6 carbon atoms, even more preferably from 1 to 4 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl, Isopyl, n-heptyl, isoheptyl, n-octyl, isooctyl, n-decyl, n-decyl and the like.
  • the "alkyl group” further includes a cyclic alkyl group having 3 to 10 carbon atoms, preferably 3 to 8 carbon atoms, more preferably 4 to 6 carbon atoms, such as a cyclopropyl group, a cyclobutyl group, a cyclopentyl group or a ring. Hexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclodecyl, decahydronaphthyl, norbornane, adamantyl.
  • the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups, independently selected from alkyl, alkenyl, Alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycle Alkoxy, cycloalkylthio, heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate.
  • alkenyl refers to a straight or branched hydrocarbon chain radical containing at least one double bond consisting of carbon and hydrogen atoms and attached to the remainder of the molecule by a single or double bond. It preferably has 2 to 10 carbon atoms, more preferably 2 to 6 carbon atoms, and even more preferably 2 to 4 carbon atoms. Non-limiting examples include ethenyl, propenyl, butenyl, pentenyl, pentadienyl, hexenyl.
  • Alkenyl can be substituted or not Substituted, when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halo, thiol , hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo An amino group, a halogenated alkyl group, a hydroxyalkyl group, a carboxyl group or a carboxylate group.
  • alkynyl refers to a straight or branched hydrocarbon chain radical containing at least one triple bond consisting of a carbon atom and a hydrogen atom and attached to the remainder of the molecule by a single or triple bond. It preferably has 2 to 10 carbon atoms, more preferably 2 to 6 carbon atoms, and even more preferably 2 to 4 carbon atoms. Non-limiting examples include ethynyl, propynyl, butynyl, pentynyl, hexynyl.
  • the alkynyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, hetero A cycloalkylthio group, an oxo group, an amino group, a halogenated alkyl group, a hydroxyalkyl group, a carboxyl group or a carboxylate group.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably the cycloalkyl ring comprises from 3 to The 10 carbon atoms, most preferably the cycloalkyl ring contains 3 to 7 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptene
  • the alkenyl group, the cyclooctyl group and the like are preferably a cyclopropyl group or a cyclohexenyl group.
  • Polycyclic cycloalkyl groups include spiro, fused, and bridged cycloalkyl groups.
  • the cycloalkyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, Heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate groups.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O) m
  • a hetero atom (where m is an integer of 0 to 2), but does not include a ring moiety of -OO-, -OS- or -SS-, and the remaining ring atoms are carbon. It preferably comprises from 3 to 12 ring atoms, wherein from 1 to 4 are heteroatoms, more preferably the heterocyclyl ring contains from 3 to 10 ring atoms, more preferably the heterocyclyl ring contains from 5 to 7 ring atoms.
  • Non-limiting examples of monocyclic heterocyclic groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, tetrahydrofuranyl and the like.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • the heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, Heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate groups.
  • the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cyclo
  • aryl refers to an all-carbon monocyclic or fused polycyclic ring (i.e., a ring that shares a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably from 5 to 10 members, more preferably from 5 to 7 members. Even more preferred is phenyl and The naphthyl group is most preferably a phenyl group.
  • the aryl group may be a completely aromatic group such as a phenyl group, a naphthyl group, an anthracenyl group, a phenanthryl group or the like.
  • the aryl group may also contain a combination of an aromatic ring and a non-aromatic ring, for example, ruthenium, osmium, and iridium.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples comprising:
  • the aryl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups, independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkane.
  • Base amino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkane A thio group, an amino group, a halogenated alkyl group, a hydroxyalkyl group, a carboxyl group or a carboxylate group.
  • heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 members, more preferably 5 to 7 members, even more preferably 5 or 6 members, such as thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, or the like.
  • heteroaryl ring may be fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples of which include:
  • the heteroaryl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkanethio Base, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, Heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate groups.
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl, cycloalkyl are as defined above.
  • Non-limiting examples include methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy and the like.
  • the alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, and an alkane group.
  • haloalkyl refers to an alkyl group wherein one or more hydrogen atoms are replaced by a halogen, wherein alkyl is as defined above.
  • Non-limiting examples include chloromethyl, trifluoromethyl, 1-chloro-2-fluoroethyl, 2,2-difluoro Ethyl, 2-fluoropropyl, 2-fluoropropan-2-yl, 2,2,2-trifluoroethyl, 1,1-difluoroethyl, 1,3-difluoro-2-methylpropane , 2,2-difluorocyclopropyl, (trifluoromethyl)cyclopropyl, 4,4-difluorocyclohexyl and 2,2,2-trifluoro-1,1-dimethyl-ethyl .
  • halogen includes fluoro, chloro, bromo and iodo.
  • cyano refers to -CN.
  • hydroxy refers to an -OH group.
  • hydroxyalkyl refers to an alkyl group substituted by a hydroxy group, wherein the alkyl group is as defined above.
  • hydroxyalkoxy refers to an alkoxy group substituted by a hydroxy group, wherein the alkoxy group is as defined above.
  • acyl refers to -C(O)R, wherein R refers to alkyl, cycloalkyl, alkenyl, alkynyl, wherein alkyl, cycloalkyl, alkenyl, alkynyl are as defined above.
  • R refers to alkyl, cycloalkyl, alkenyl, alkynyl, wherein alkyl, cycloalkyl, alkenyl, alkynyl are as defined above.
  • Non-limiting examples include acetyl, propionyl, butyryl, valeryl, hexanoyl, vinyl, acryloyl groups.
  • amido refers to -NHC(O)R, wherein R refers to alkyl, alkenyl, alkynyl, wherein alkyl, alkenyl, alkynyl are as defined above.
  • R refers to alkyl, alkenyl, alkynyl, wherein alkyl, alkenyl, alkynyl are as defined above.
  • Non-limiting examples include formamide, acetamido, propionamide, butanamide, pentanoamide, hexanoamido, vinyl amide, acrylamide.
  • ester group refers to -C(O)OR, wherein R refers to alkyl or cycloalkyl, wherein alkyl, cycloalkyl are as defined above.
  • R refers to alkyl or cycloalkyl, wherein alkyl, cycloalkyl are as defined above.
  • Non-limiting examples include ethyl ester groups, propyl ester groups, butyl ester groups, amyl ester groups, cyclopropyl ester groups, cyclobutyl ester groups, cyclopentyl ester groups, cyclohexyl ester groups.
  • substituents are selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, a halogenated alkyl group, an alkoxy group, an aryl group, a halogenated aryl group, an aryloxy group, an aralkyl group, an aralkyloxy group, and a hetero group.
  • substituents can also be further substituted.
  • the alkyl group as a substituent is also optionally selected from one or more groups selected from a halogen atom, a hydroxyl group, an alkoxy group, an alkylamino group, a pyrrolidinyl group, a phenyl group, a pyridyl group, or a halogenated phenyl group.
  • the heterocyclic group as a substituent is also optionally substituted with one or more groups selected from a halogen atom, an alkyl group, and an alkoxy group.
  • the present invention mainly employs the following synthetic routes and technical solutions.
  • the first synthetic method of the compound of the present invention is such that the structure of the compound of the formula (I) of the present invention is divided into a part A and a part B as above, wherein the part A is an amine intermediate compound of the formula (II), and the part B is an active ester intermediate. a compound of formula (III).
  • an intermediate product is obtained by a substitution reaction in a suitable solvent under a suitable temperature and pH conditions;
  • the base may be, for example, potassium carbonate or cesium carbonate.
  • Etc. the solvent may be, for example, DMF, acetonitrile or the like.
  • the nitro group of the intermediate product is then reduced to the amino group to give intermediates 1, 2, 3; the reduction of the nitro group can be achieved, for example, in an iron powder ammonium chloride system or a H 2 /palladium carbon system.
  • the synthesis method is the same as that of the above-mentioned benzimidazole intermediate, except that the corresponding starting material is used instead of the benzimidazole intermediate M to obtain a corresponding other type of intermediate.
  • Step 1 Intermediate M1 and p-bromonitrobenzene are subjected to Buchwald reaction in a suitable solvent under the action of a base, a catalyst and a ligand to obtain an intermediate M2;
  • the solvent is preferably dioxane, toluene, and the base is preferably tert-butyl.
  • the catalyst is preferably (pd) 2 (dba) 3 , palladium acetate, pd (dba) 2 ;
  • the ligand is preferably Xphos, BINAP;
  • Step 2 Reducing the amino group of the two nitro groups of the intermediate M2 under reducing conditions to obtain the intermediate M3, and the reducing conditions may be, for example, an iron powder ammonium chloride system or a H 2 /palladium carbon system;
  • Step 3 Under acidic conditions, intermediate M3 and formic acid undergo a ring closure reaction at elevated temperature to obtain intermediate 4, the acidic condition such as hydrochloric acid;
  • Step 4 When R 0 is selected from a methoxy group: the intermediate 4 is heated under reflux under acidic conditions to obtain an intermediate 5, for example under HBr;
  • Step 5 The intermediate 5 is reacted with the corresponding active halogenated product under the catalysis of a base in a suitable solvent at a temperature to obtain an intermediate 6, such as sodium hydroxide, potassium carbonate or the like, for example, the solvent DMF, etc.;
  • Step 1 Substituting 2-nitro-5-fluoroaniline with an intermediate M4 in the presence of a base to obtain an intermediate M5, which may be, for example, potassium carbonate or the like, in a suitable solvent.
  • an intermediate M5 which may be, for example, potassium carbonate or the like, in a suitable solvent.
  • Step 2 Intermediate M5 and p-bromonitrobenzene are subjected to Buchwald reaction in a suitable solvent under the action of a base, a catalyst and a ligand to obtain an intermediate M6;
  • the solvent is preferably dioxane, toluene, and the base is preferably tert-butyl.
  • the catalyst is preferably (pd) 2 (dba) 3 , palladium acetate, pd (dba) 2 ;
  • the ligand is preferably Xphos, BINAP;
  • Step 3 reducing the amino group of the intermediate M6 to an amino group under reducing conditions to obtain an intermediate M7, which may be, for example, an iron powder ammonium chloride system or a H 2 /palladium carbon system;
  • Step 4 Under acidic conditions, intermediate M7 and formic acid undergo a ring closure reaction at elevated temperature to give intermediate 7, the acidic conditions such as hydrochloric acid.
  • Step 1 The intermediate M8 is substituted with the intermediate M9 in the presence of a base in a suitable solvent to obtain the intermediate M10,
  • the base may be, for example, triethylamine, and the solvent may be, for example, DMSO;
  • Step 2 reducing the amino group of the intermediate M10 to an amino group under reducing conditions to obtain an intermediate M11, which may be, for example, an iron powder ammonium chloride system or a H 2 /palladium carbon system;
  • Step 3 Under acidic conditions and high temperature conditions, intermediate M11 and formic acid undergo a ring closure reaction to obtain intermediate 8, the acidic condition such as hydrochloric acid;
  • the pyrazole intermediate is obtained by reaction under acid catalysis in a suitable solvent at a suitable temperature and pH.
  • the solvent may be, for example, ethanol
  • the acid may be, for example, hydrochloric acid.
  • Part B intermediate has only one urea forming site, it is made into an active ester, as shown in Scheme 7 below.
  • the phenyl chloroformate is reacted with the corresponding amine (intermediate M12) under suitable conditions of a suitable temperature and pH in a suitable solvent to give the corresponding active ester
  • the solvent may be, for example, Ethyl acetate, dichloromethane, tetrahydrofuran, acetone, acetonitrile, water, etc.
  • the base may be, for example, pyridine, sodium hydrogencarbonate, potassium carbonate, triethylamine, sodium hydroxide or the like. Since the groups attached to the amino group are different, resulting in different activities, the base selected in the reaction is slightly different, and those skilled in the art can make conventional selection according to the general technical knowledge in the art.
  • the B moiety intermediate has two urea sites (-NH- and -NH 2 -), for example:
  • the A part of the amine intermediate is then made into an active ester. That is, as a second synthesis method of the compound of the present invention, the structure of the compound of the general formula (I) of the present invention is divided into a part A and a part B, wherein the part A is an active ester intermediate compound of the formula (IV), and the part B is an intermediate of the amine.
  • Compound of formula (V) Compound of formula (V).
  • the preparation method of the A-part active ester intermediate compound of the formula (IV) is similar to the preparation method of the active ester in the above section B. That is, at a suitable temperature and pH, in a suitable solvent, in the base The A-part amine intermediate compound of formula (II) is reacted with phenyl chloroformate under catalysis to obtain a Part A active ester intermediate.
  • the active ester is deprived of one molecule of phenol to obtain the corresponding isocyanate intermediate, and then reacted with the corresponding amine under the action of a base to form a final urea compound, that is, a compound of the formula (I).
  • the solvent is preferably THF, acetonitrile, dichloromethane, toluene
  • the base is preferably triethylamine, N,N-diisopropylethylamine, DMAP, pyridine;
  • X, Y, A, A 1 , Z, W, R 1 , B, R 2 , G, R 3 are as defined in Formula 1, and R 0 , R 0 1 , R 0 2 are not specifically stated , the same as the definition of -BR 1 .
  • the compound represented by the formula (I) of the present invention, the prodrug thereof should follow the design principle of the prodrug, and can release the original active formula by enzymatic hydrolysis, hydrolysis, acid hydrolysis or metabolic degradation under normal physiological conditions in the living body.
  • Compound shown by I). This includes, but is not limited to, the lipidation of hydroxyl groups on the compound (such as the formation of phosphates and carbonates), the protection of amino groups and carboxyl groups.
  • the pharmaceutically acceptable salt of the compound of the formula (I) of the present invention may be an acid addition salt or a base addition salt.
  • the acid may be a mineral acid including, but not limited to, hydrochloric acid, sulfuric acid, phosphoric acid, hydrobromic acid; or may be an organic acid including, but not limited to, citric acid, maleic acid, oxalic acid, formic acid, acetic acid, propionic acid, valeric acid.
  • glycolic acid glycolic acid, benzoic acid, fumaric acid, trifluoroacetic acid, succinic acid, tartaric acid, lactic acid, glutamic acid, aspartic acid, salicylic acid, pyruvic acid, methanesulfonic acid, benzenesulfonic acid, p-benzenesulfonic acid And dextran camphorsulfonic acid and the like.
  • the base may be an inorganic base including, but not limited to, sodium hydroxide, potassium hydroxide, magnesium hydroxide, calcium hydroxide; or may be an organic base including, but not limited to, ammonium hydroxide, triethylamine, N, N- Dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-benzyl phenyl An amine, arginine or lysine; or may be an alkali metal salt, including but not limited to: lithium, potassium and sodium salts; or may be an alkaline earth metal salt, including but not limited to: barium, calcium and magnesium salts;
  • the transition metal salt includes, but is not limited to, a zinc salt; or other metal salts including, but not limited to, sodium hydrogen phosphate and disodium hydrogen phosphate.
  • the compound of the formula (I) or a pharmaceutically acceptable salt or prodrug is prepared into a clinically usable pharmaceutical composition.
  • the pharmaceutical preparations thereof include, but are not limited to, oral preparations such as tablets, gels, soft/hard capsules, emulsions, dispersible powders, granules, water/oil suspoemulsions; injections Including intravenous injection, intramuscular injection, intraperitoneal injection, rectal suppository, intracranial injection, these dosage forms may be aqueous solutions or oily solutions; topical preparations include creams, ointments, gels, water/oil solutions and packs Formulations; inhalation dosage forms include fine powders, liquid aerosols, and various dosage forms suitable for in vivo implantation.
  • the pharmaceutical composition of the present invention may be added with a pharmaceutically acceptable carrier, diluent or excipient as needed.
  • a pharmaceutically acceptable carrier diluent or excipient as needed.
  • Carriers for solid oral formulations include, but are not limited to, mannitol, lactose, starch, magnesium stearate, cellulose, glucose, sucrose, cyclodextrin, and the intestinal absorption molecular carrier vitamin E-PEG 1000.
  • Oral formulations may incorporate suitable colorants, sweeteners, flavoring agents, and preservatives.
  • the compound of the formula (I) or a pharmaceutically acceptable salt or prodrug of the present invention is administered to a warm-blooded animal at a unit dose of 0.01 to 100 mg/kg.
  • the dosage of the drug to be administered depends on a variety of factors including, but not limited to, the following factors: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the patient's The route, the patient's diet, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, and the like.
  • the optimal mode of treatment such as the mode of treatment, the daily amount of the compound of formula (I) or the type of pharmaceutically acceptable salt, can be verified according to conventional treatment regimens.
  • the compound of the formula (I) or a pharmaceutically acceptable salt or prodrug of the present invention can be used alone or in combination with radiotherapy, chemotherapy, immunotherapy, and tumors conventionally used in clinical treatment of cancer.
  • Combination therapy with one or more methods of viral, RNAi, cancer adjuvant therapy, and bone marrow transplantation and stem cell transplantation including but not limited to the following anti-tumor drugs and treatments:
  • alkylating agents such as cisplatin, cisplatin, oxaliplatin, chlorambucil, carbophosphoramide, nitrogen mustard, melphalan, temozolomide, busulfan, nitrosourea.
  • anti-tumor antibiotics such as doxorubicin, bleomycin, doxorubicin, daunorubicin, epirubicin, idarubicin, mitomycin C, actinomycin, genus Anti-mitotic drugs such as vincristine, vinblastine, vindesine, vinorelbine, paclitaxel, taxotere, Polo kinase inhibitors.
  • Antimetabolites and antifolates such as fluoropyrimidine, rametamine, cytarabine, azacitidine, decitabine, trebuta, hydroxyurea, IDH1/IDH2 mutant inhibitors.
  • Topoisomerase inhibitors such as epipodophyllotoxin, camptothecin, and irinotecan.
  • Cell growth inhibitors such as antiestrogens/antiandrogens.
  • antiestrogens/antiandrogens such as tamoxifen, fulvestine, toremifene, raloxifene, ranoxifene, oxycidifene, bicalutamide, flutamide, nilutamide, cyproterone acetate;
  • LHRH antagonists or LHRH agonists such as goserelin, leuprolide, and buserelin, progestogens such as megestrol acetate;
  • Aromatase inhibitors such as anastrozole, letrozole, vorozole, exemestane, 5a-reductase inhibitors such as finasteride.
  • Anti-invasive agents such as c-Src kinase family inhibitors, metalloproteinase inhibitors, inhibitors of urokinase plasminogen activator receptor function or heparanase-like antibodies.
  • Inhibitors of cell growth function include tyrosine kinase inhibitors and inhibitors of serine/threonine kinases such as Ras/Raf signaling inhibitors, cell signaling inhibitors of MEK and/or AKT kinase, c-Kit inhibition Agent, c-Met inhibitor, PDGFR inhibitor, ABL kinase inhibitor, PI3 kinase inhibitor, CSF-1R kinase inhibitors, EGFR family kinase inhibitors, FGFR family kinase inhibitors, IGF receptor kinase inhibitors, Aurora kinase inhibitors, cyclin-dependent kinase inhibitors such as CDK2 and/or CDK4, CDK6 inhibitors, Nuclear transporter CRM1 inhibitor, a Wnt/beta-catenin inhibitor.
  • tyrosine kinase inhibitors and inhibitors of serine/threonine kinases such as Ras/Raf signaling inhibitors, cell signal
  • Inhibitors of anti-apoptotic proteins such as BCL2 inhibitors (Venetoclax) and MCL1 inhibitors.
  • PARP inhibitors such as Olaparib and Rucaparib.
  • anti-angiogenic inhibitors such as VEGFR inhibitors.
  • Epigenetic inhibitors such as histone deacetylase (HDAC) inhibitors and DNA methyltransferase (DNMT) inhibitors.
  • HDAC histone deacetylase
  • DNMT DNA methyltransferase
  • Tumor immunotherapy includes any in vitro and in vivo method of increasing the immunogenicity of a patient's tumor cells.
  • cytokine IL-2, IL-4 or GM-CSF for transfection; methods for reducing T cell ineffectiveness such as anti-PD-1/PD-L mAb; transfected immune cells such as cytokine transfected trees Method of squamous cell; method of cytokine transfected tumor cell line; reduction of immunosuppressive cells such as regulatory T cells, myeloid suppressor cells, or dendrites expressing guanamine 2,3-deoxygenase A functional method of cells; and a method of cancer vaccine consisting of tumor-associated antigenic proteins or peptides.
  • Tumor gene therapy such as CRISPR-Cas 9, RNAi and gene transduction.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • is given in units of 10 -6 (ppm).
  • the NMR was measured by a (Bruker AVANCE-400) nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), internal standard. It is tetramethylsilane (TMS).
  • the MS was measured by a liquid chromatography mass spectrometer (Thermo, Ultimate 3000/MSQ).
  • the HPLC was measured using a high pressure liquid chromatograph (Agilent 1260 Infinity, Gemini C18 250 x 4.6 mm, 5u column).
  • the silica gel plate HSGF245 used for thin layer chromatography has a specification of 0.15 mm to 0.2 mm, and the specification for separation and purification of thin layer chromatography is 0.9 mm to 1.0 mm (Yantai Yellow Sea).
  • the known starting materials of the present invention can be synthesized by or according to methods known in the art, or purchased from Shanghai Darui Fine Chemicals Co., Ltd., Shanghai Titan Technology Co., Ltd., Shanghai Runjie Chemical Reagent Co., Ltd., TCI, Aldrich Chemical Company.
  • the experimental methods in the examples which do not specify the specific conditions are usually carried out according to conventional conditions or according to the conditions recommended by the raw material or commodity manufacturer. Reagents without specific source are routine reagents purchased from the market.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature and is 20 ° C to 30 ° C.
  • Step 1 1 - Preparation of (4-nitro-phenyl)-1H-benzimidazole
  • Benzimidazole (23.6 g, 0.2 mol) and p-fluoronitrobenzene (31.0 g, 0.22 mol) were dissolved in 300 ml of DMF at room temperature, and anhydrous potassium carbonate (69.0 g, 0.5 mol) was added. The mixture was heated to 90 ° C and allowed to react for 6 hours. After cooling the reaction solution to room temperature, it was poured slowly into water, stirred at room temperature for 0.5 hour, and filtered. The resulting solid was washed with water and dried in vacuo overnight. The obtained solid was slurried with methyl tert-butyl ether to give 46 g of 1-(4-nitro-phenyl)-1H-benzimidazole as a yellow solid.
  • Step 3 Preparation of oxazol-3-yl-carbamic acid phenyl ester (active ester)
  • Step 4 Preparation of 1-(4-benzimidazol-1-yl-phenyl)-3-isoxazole-3-yl-urea
  • Step 5 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-hydroxy-benzimidazol-1-yl)-phenyl]-urea
  • Step 2 Same as Step 5 of Example 4, except that 4-(5-ethoxy-benzimidazol-1-yl)-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzimidazole -5-Alcohol, 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-ethoxy-benzimidazol-1-yl)-phenyl]- Urea.
  • Step 2 Same as Step 5 of Example 4 except that 4-(5-hexyloxy-benzimidazol-1-yl)-phenylamine was used in place of 1-(4-amino-phenyl)-1H-benzimidazole -5-Alcohol, 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-hexyloxy-benzimidazol-1-yl)-phenyl]- Urea.
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3- ⁇ 4-[5-(2-methoxy-ethoxy)-benzimidazol-1-yl]- Preparation of phenyl ⁇ -urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-[5-(2-methoxy-ethoxy)-benzimidazol-1-yl]-phenylamine was substituted for 1-(4-amino-phenyl)- 1H-benzimidazol-5-ol gives 1-(5-tert-butyl-isoxazol-3-yl)-3- ⁇ 4-[5-(2-methoxy-ethoxy)-benzene And imidazol-1-yl]-phenyl ⁇ -urea.
  • Example 7 The same procedure as in Example 7 was carried out except that 2-chloroethyl ethyl ether (Dari) was used instead of 1-chlorohexane in Step 1, to give 1-(5-tert-butyl-isoxazole-3- Benzyl-3- ⁇ 4-[5-(2-ethoxy-ethoxy)-benzimidazol-1-yl]-phenyl ⁇ -urea.
  • Example 7 The same procedure as in Example 7 was carried out except that 2-chloroethoxyethanol (Titan) was used instead of 1-chlorohexane in Step 1, to give 1-(5-tert-butyl-isoxazol-3-yl). -3-(4- ⁇ 5-[2-(2-Hydroxy-ethoxy)-ethoxy]-benzimidazol-1-yl ⁇ -phenyl)-urea.
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3- ⁇ 4-[5-(2-morpholin-4-yl-ethoxy)-benzimidazole-1- Preparation of phenyl]-phenyl ⁇ -urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-[5-(2-morpholin-4-yl-ethoxy)-benzimidazol-1-yl]-phenylamine was substituted for 1-(4-amino-benzene -1H-benzimidazol-5-ol to give 1-(5-tert-butyl-isoxazol-3-yl)-3- ⁇ 4-[5-(2-morpholin-4-yl- Ethoxy)-benzimidazol-1-yl]-phenyl ⁇ -urea.
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(5-trifluoromethoxy-benzimidazol-1-yl)-phenyl]-urea preparation
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(5-trifluoromethoxy-benzimidazol-1-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole- 5-Alcohol, 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-trifluoromethoxy-benzimidazol-1-yl)-phenyl]- Urea.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-fluoro-benzimidazol-1-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(5-fluoro-benzimidazol-1-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(5-fluoro-benzimidazol-1-yl)-phenyl]-urea is obtained.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-trifluoromethyl-benzimidazol-1-yl)-phenyl]-urea
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-methyl-benzimidazol-1-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(5-methyl-benzimidazol-1-yl)-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzimidazole-5-ol 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(5-trifluoromethyl-benzimidazol-1-yl)-phenyl]-urea was obtained.
  • Step 4 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(6-methoxy-benzimidazol-1-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(6-methoxy-benzimidazol-1-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5- Alcohol gives 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(6-methoxy-benzimidazol-1-yl)-phenyl]-urea.
  • Step 6 1-(5-tert-Butyl-isoxazol-3-yl)-3- ⁇ 4-[6-(2-methoxy-ethoxy)-benzimidazol-1-yl]- Preparation of phenyl ⁇ -urea
  • Step 6 1-(5-tert-Butyl-isoxazol-3-yl)-3- ⁇ 4-[6-(2-morpholin-4-yl-ethoxy)-benzimidazole-1- Preparation of phenyl]-phenyl ⁇ -urea
  • the product obtained in the step 5 is 4-[6-(2-morpholin-4-yl-ethoxy)-benzimidazol-1-yl]-phenylamine (100 mg, 0.295 mmol), (5-tert-butyl-iso)
  • the oxazol-3-yl)-carbamic acid phenyl ester (115.3 mg, 0.443 mmol) and triethylamine (90 mg, 0.888 mmol) were dissolved in 10 ml of THF and refluxed overnight. On the next day, the reaction mixture was concentrated under reduced pressure.
  • Step 1 Preparation of N 1 -(2-dimethylamino-ethyl)-N 1 -methyl-4-nitrobenzene-1,3-diamine
  • Step 2 N 1 -(2-Dimethylamino-ethyl)-N 1 -methyl-4-nitro-N 3 -(4-nitro-phenyl)-benzene-1,3-diamine Preparation
  • Step 3 Preparation of N 2 -(4-amino-phenyl)-N 4 -(2-dimethylamino-ethyl)-N 4 -methyl-phenyl-1,2,4-triamine
  • Step 4 Preparation of N-[3-(4-amino-phenyl)-3H-benzimidazol-5-yl]-N,N,N'-trimethylethane-1,2-diamine
  • the product obtained in the step 3 (1.1 g, 4.37 mmol) was dissolved in 40 ml of hydrochloric acid (4 mol/L), 1.1 ml of formic acid was added at room temperature, and the mixture was heated to 120 ° C for 1 h.
  • the reaction solution was treated with ammonia in an ice bath
  • the residue was purified by column chromatography (eluent: dichloromethane / methanol) to yield
  • Step 5 1-(5-tert-Butyl-isoxazol-3-yl)-3-(4- ⁇ 6-[(2-dimethylamino-ethyl)-methyl-amino]-benzo Preparation of imidazol-1-yl ⁇ -phenyl)-urea.
  • N-[3-(4-Amino-phenyl)-3H-benzimidazol-5-yl]-N,N,N'-trimethylethane-1,2-diamine obtained in Step 4 100 mg, 0.323 mmol
  • oxazol-3-yl-carbamic acid phenyl ester 168 mg, 0.647 mmol
  • triethylamine 100 mg, 0.99 mmol
  • Step 3 Preparation of N 2 -(4-amino-phenyl)-4-(4-methyl-piperazin-1-yl)-benzene-1,2-diamine
  • the product obtained in the step 3 was obtained by dissolving N2-(4-amino-phenyl)-4-(4-methyl-piperazin-1-yl)-benzene-1,2-diamine (1.3 g, 4.37 mmol). In 50 ml of hydrochloric acid (4 mol/L), 1.5 ml of formic acid was added at room temperature, and the mixture was heated to 120 ° C for 1 h. The reaction mixture was diluted with EtOAc EtOAc (EtOAc)EtOAc. The residue was purified by column chromatography (eluent: dichloromethane / methanol) toield aniline.
  • Step 5 1-(5-tert-Butyl-isoxazol-3-yl)-3- ⁇ 4-[6-(4-methyl-piperazin-1-yl)-benzimidazol-1-yl Preparation of ]-phenyl ⁇ -urea
  • step 1 of Example 7 Same as step 1 of Example 7, except that 3-(4-amino-phenyl)-3H-benzimidazole-4-ol was substituted for 1-(4-amino-phenyl)-1H-benzimidazole-5 -Alcohol, and replacing 1-chlorohexane with N-(2-chloroethyl)morpholine hydrochloride to give 4-[7-(2-morpholin-4-yl-ethoxy)-benzimidazole -1-yl]-aniline.
  • Step 3 1-(5-tert-Butyl-isoxazol-3-yl)-3- ⁇ 4-[7-(2-morpholin-4-yl-ethoxy)-benzimidazole-1- Preparation of phenyl]-phenyl ⁇ -urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-[7-(2-morpholin-4-yl-ethoxy)-benzimidazol-1-yl]-phenylamine was substituted for 1-(4-amino-benzene -1H-benzimidazol-5-ol to give 1-(5-tert-butyl-isoxazol-3-yl)-3- ⁇ 4-[7-(2-morpholin-4-yl- Ethoxy)-benzimidazol-1-yl]-phenyl ⁇ -urea.
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(5,6-dimethyl-benzimidazol-1-yl)-phenyl]-urea preparation
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(5,6-dimethyl-benzimidazol-1-yl)-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzimidazole- 5-Alcohol to give 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5,6-dimethyl-benzimidazol-1-yl)-phenyl] -urea.
  • step 5 of Example 4 Same as step 5 of Example 4 except that (5-fluoro-7-methyl-benzimidazol-1-yl)-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzimidazole-5 -Alcohol to give 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-fluoro-7-methyl-benzimidazol-1-yl)-phenyl] -urea.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(4-fluoro-benzimidazol-1-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(4-fluoro-benzimidazol-1-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(4-fluoro-benzimidazol-1-yl)-phenyl]-urea was obtained.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(2-methyl-benzimidazol-1-yl)-phenyl]-urea
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(2-chlorobenzimidazol-1-yl)-phenyl]-urea
  • Step 1 Preparation of 4-benzimidazol-1-yl-3-methyl-phenylamine.
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-3-methyl-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • step 5 of Example 4 Same as step 5 of Example 4, except that 4-benzimidazol-1-yl-3-methyl-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • 1-(4-Benzimidazol-1-yl-3-methyl-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea is obtained.
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-3-chloro-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • step 5 of Example 4 Same as step 5 of Example 4, except that 4-benzimidazol-1-yl-3-chloro-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-3-fluoro-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • step 5 of Example 4 Same as step 5 of Example 4, except that 4-benzimidazol-1-yl-3-fluoro-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-3,5-difluoro-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-2-chloro-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • step 5 of Example 4 Same as step 5 of Example 4, except that 4-benzimidazol-1-yl-2-chloro-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • Step 2 Preparation of 1-(6-benzimidazol-1-yl-pyridin-3-yl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • Step 1 1 - Preparation of (5-nitro-pyrimidin-2-yl)-1H-benzimidazole
  • step 2 of Example 1 Same as step 2 of Example 1, except that 1-(5-nitro-pyrimidin-2-yl)-1H-benzimidazole is substituted for 1-(4-nitro-phenyl)-1H-benzene in Step 2. And imidazole gives 2-benzimidazol-1-yl-pyrimidin-5-ylamine.
  • Step 3 Preparation of 1-(2-benzimidazol-1-yl-pyrimidin-5-yl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-(4-indol-1-yl-phenyl)-urea
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-(4-oxazol-1-yl-phenyl)-urea
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(6-fluoro-oxazol-1-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(6-fluoro-oxazol-1-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(6-fluoro-oxazol-1-yl)-phenyl]-urea is obtained.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(7-fluoro-oxazol-1-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(7-fluoro-oxazol-1-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(7-fluoro-oxazol-1-yl)-phenyl]-urea is obtained.
  • Step 2 Preparation of 1-(4-benzotriazol-1-yl-phenyl)-3-(5-tert-butyl-isoxazol-3-yl)-urea
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-(4-pyrrolo[2,3-b]pyridin-1-yl-phenyl)-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-pyrrolo[2,3-b]pyridin-1-yl-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzimidazole-5-ol 1-(5-tert-Butyl-isoxazol-3-yl)-3-(4-pyrrolo[2,3-b]pyridin-1-yl-phenyl)-urea was obtained.
  • Step 1 Preparation of [4-(3-nitro-pyridin-2-ylamino)-phenyl]-carbamic acid tert-butyl ester
  • Step 2 Preparation of [4-(3-amino-pyridin-2-ylamino)-phenyl]-carbamic acid tert-butyl ester
  • [4-(3-Amino-pyridin-2-ylamino)-phenyl]-carbamic acid tert-butyl ester (1 g, 3.33 mmol) was dissolved in 30 ml of hydrochloric acid (4 mol/L) at room temperature, and 1 ml of formic acid was added. The mixture was heated to 120 ° C for 1 h. The reaction mixture was diluted with EtOAc EtOAc (EtOAc)EtOAc. A crude product of 340 mg was obtained. It was used directly in the next reaction without purification.
  • Step 4 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-(4-imidazo[4,5-b]pyridin-3-yl-phenyl)-urea
  • Step 1 Preparation of [4-(5-bromo-3-nitro-pyridin-2-ylamino)-phenyl]-carbamic acid tert-butyl ester
  • Step 2 Preparation of [4-(3-amino-5-bromo-pyridin-2-ylamino)-phenyl]-carbamic acid tert-butyl ester
  • Step 4 1-[4-(6-Bromo-imidazo[4,5-b]pyridin-3-yl)-phenyl]-3-(5-tert-butyl-isoxazol-3-yl) -Preparation of urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(6-bromo-imidazo[4,5-b]pyridin-3-yl)-phenylamine was used in place of 1-(4-amino-phenyl)-1H-benzene. And imidazole-5-ol, 1-[4-(6-bromo-imidazo[4,5-b]pyridin-3-yl)-phenyl]-3-(5-tert-butyl-iso- Zyrid-3-yl)-urea.
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(5-chloroimidazo[4,5-b]pyridin-3-yl)-phenyl]- Preparation of urea
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(5-methylimidazo[4,5-b]pyridin-3-yl)-phenyl] -Preparation of urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(5-methylimidazo[4,5-b]pyridin-3-yl)-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzene. And imidazole-5-ol, 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(5-methylimidazo[4,5-b]pyridine-3- Base)-phenyl]-urea.
  • Step 2 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(7-methylimidazo[4,5-b]pyridin-3-yl)-phenyl]- Preparation of urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-(7-methylimidazo[4,5-b]pyridin-3-yl)-phenylamine was used in place of 1-(4-amino-phenyl)-1H-benzene. And imidazole-5-ol, 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(7-methylimidazo[4,5-b]pyridin-3-yl) )-Phenyl]-urea.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-(4-imidazo[4,5-b]pyridin-1-yl-phenyl)-urea
  • step 5 of Example 4 Same as step 5 of Example 4 except that 4-imidazo[4,5-b]pyridin-1-yl-phenylamine was substituted for 1-(4-amino-phenyl)-1H-benzimidazole-5-ol 1-(5-tert-Butyl-isoxazol-3-yl)-3-(4-imidazo[4,5-b]pyridin-1-yl-phenyl)-urea was obtained.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(6-methoxy-indol-7-yl)-phenyl]-urea
  • step 5 of Example 4 Same as step 5 of Example 4, except that 4-(6-methoxy-indol-7-yl)-phenylamine was used instead of 1-(4-amino-phenyl)-1H-benzimidazole-5-ol.
  • 1-(5-tert-Butyl-isoxazol-3-yl)-3-[4-(6-methoxy-indol-7-yl)-phenyl]-urea was obtained.
  • Step 1 Same as the preparation method in Step 1 to Step 2 of Example 57, which is the same reaction as the other product in Example 57 Step 1 - Step 2.
  • Step 2 Preparation of 1-(5-tert-butyl-isoxazol-3-yl)-3-[4-(6-dimethylamino-indol-7-yl)-phenyl]-urea
  • Step 1 Preparation of thiazol-2-yl-carbamic acid phenyl ester (active ester)
  • step 3 of Example 1 The same as step 3 of Example 1, except that 2-aminoisoxazole (TCI) was used instead of 3-aminoisoxazole to give thiazol-2-yl-carbamic acid phenyl ester.
  • TCI 2-aminoisoxazole
  • step 4 of Example 1 Same as step 4 of Example 1, except that phenyloxazol-2-yl-carbamic acid phenyl ester (active ester) was used in place of oxazol-3-yl-carbamic acid phenyl ester (active ester) to give 1-(4) -Benzimidazol-1-yl-phenyl)-3-thiazol-2-yl-urea.
  • Step 1 Preparation of (4-methyl-thiazol-2-yl)-carbamic acid phenyl ester (active ester)
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-phenyl)-3-(4-methyl-thiazol-2-yl)-urea
  • step 4 of Example 1 Same as step 4 of Example 1, except that (4-methyl-thiazol-2-yl)-carbamic acid phenyl ester (active ester) was substituted for the oxazol-3-yl-carbamic acid phenyl ester (active ester) ), 1-(4-benzimidazol-1-yl-phenyl)-3-(4-methyl-thiazol-2-yl)-urea was obtained.
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-phenyl)-3-[1,3,4]thiadiazol-2-yl-urea
  • Step 1 Preparation of (5-tert-butyl-[1,3,4]thiadiazol-2-yl)-carbamic acid phenyl ester (active ester)
  • step 3 of Example 1 Same as step 3 of Example 1, except that 2-amino-5-tert-butylthiadiazole (Dari) was used instead of 3-aminoisoxazole to give 1-(4-benzimidazol-1-yl-benzene 3-(5-tert-butyl-[1,3,4]thiadiazol-2-yl)-urea.
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-phenyl)-3-(5-tert-butyl-[1,3,4]thiadiazol-2-yl)-urea
  • step 4 of Example 1 Same as step 4 of Example 1, except that (5-tert-butyl-[1,3,4]thiadiazol-2-yl)-carbamic acid phenyl ester (active ester) was substituted for isoxazol-3-yl - phenyl carbamate (active ester) to give 1-(4-benzimidazol-1-yl-phenyl)-3-(5-tert-butyl-[1,3,4]thiadiazole-2 -Base)-urea.
  • Step 1 Preparation of (4-benzimidazol-1-yl-phenyl)-carbamic acid phenyl ester
  • Step 1 Preparation of (4-benzimidazol-1-yl-phenyl)-carbamic acid phenyl ester
  • Step 2 Preparation of 1-(4-benzimidazol-1-yl-phenyl)-3-(5-tert-butyl-2H-pyrazol-3-yl)-urea
  • Example 126 Same as the preparation method of Example 68 except that 4-(5-trifluoromethyl-benzimidazol-1-yl)-phenylamine (synthesized in Step 1 of Example 23) was used instead of 4-benzimidazole in Step 1.
  • 1-yl-aniline to give 1-(5-tert-butyl-2H-pyrazol-3-yl)-3-[4-(5-trifluoromethyl-benzimidazol-1-yl)-benzene Base]-urea.
  • Compound 126 was also obtained (see Example 126).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

提供一种通式(I)所示的脲类化合物、其药学上可接受的盐,及其制备方法,以及其作为FLT3酪氨酸蛋白激酶抑制剂,特别是在预防和/或治疗癌症中的用途。

Description

一种脲类化合物、其制备方法及其医药用途 技术领域
本发明涉及一种新型的脲类化合物、其制备方法及含有其的药物组合物,以及其作为FLT3酪氨酸蛋白激酶抑制剂的用途,特别是在预防和/或治疗癌症中的用途。
背景技术
癌症是人类临床死亡的主要疾病之一,特别是恶性肿瘤例如肺癌、胃癌、乳腺癌、胰腺癌、肝癌、肠癌、食管癌以及白血病,其死亡率极高。至今为止仍没有有效的方法与药物可以预防、治愈和根除癌症。临床上急需特异性好、活性高、毒性小、无耐药性产生的优质抗癌药物和治疗方法。
白血病即血癌,是造血干细胞异常的克隆性恶性疾病,因白血病细胞失去分化为成熟功能性血细胞的能力而停滞在细胞发育的不同阶段恶性增殖,在骨髓和其他造血组织中白血病细胞大量增生积聚并浸润其他器官和组织,正常造血受到抑制,临床表现为贫血、出血、感染及各器官浸润等症状。白血病的发病率在所有肿瘤中占第6/7位,特别是在儿童和老人中有较高的发病率。白血病属于细胞异质性恶性肿瘤,种类繁多,病因复杂,临床上表现出不同的特征,其中有的白血病发病快,死亡率高,存活期短,易复发,愈后不良,极难治愈。例如急性髓细胞白血病(AML)在60岁以上病人中其5年存活率仅为10%-20%,60岁以下存活率为40%-50%(
Figure PCTCN2017076265-appb-000001
 H等人,Acute Myeloid Leukemia.N Engl J Med.2015;373(12):1136-52)。
虽然血癌的临床治疗可采用多种方法如化疗、放疗、免疫治疗、靶向治疗、诱导分化治疗以及骨髓/干细胞移植,但是近40年来急性髓细胞白血病的临床治疗方法几乎无大的改变,其标准治疗方法仍然以诱导缓解为主即“7+3”基本疗法(柔红霉素25-45mg/m2,IV第1-3天,阿糖胞苷100mg/m2,IV第1-7天,急性早幼粒细胞白血病APL除外)。经典的化疗方法虽然能够在短期内有效的诱导缓解AML病情,抑制或者杀死癌细胞,但是此类药物副作用大,选择性差,容易复发和容易产生药物抗性,无法得到根治。靶向治疗与肿瘤免疫治疗是当今癌症临床治疗的发展方向,其中靶向药物治疗特异性好,副作用小,疗效明显。许多靶向药物已成功的应用于不同类型的癌症包括某些类型的白血病如格列卫(治疗CML),然而至今为止全球尚未有被批准用于AML治疗的有效靶向药物上市,临床上急需大量针对AML靶向治疗的药品。
在AML病人临床样本的细胞遗传基因组大数据分析中发现,基因突变频繁发生是AML的一个主要特征,例如细胞信号传导途径相关基因的突变发生率约占50-60%,DNA甲基化相关基因异常占44%,染色体修饰基因变异约为30%,髓 系转录因子基因异常占20-25%,转录因子融合基因发生率为18%左右,肿瘤抑制基因突变占14%。例如在AML病人中,最为常见基因异常有FLT3-ITD(19-28%)、FLT3-TKD(5-10%)、NPM1(突变为27-35%)、DNMTA(突变为26%)、NRAS(突变为8-9%)、ASXL1(突变为17-19%)、CEBPA(突变占4-6%)、TET2(突变在8-27%范围)、WT1(基因异常为8%)、IDH2(点突变为8-9%)、IDH1(突变占9%)、KIT(突变率为2-4%)、RUNX1(基因突变范围为5-10%)、MLL-PTD(5%)、PHF6(占3%)、KRAS(突变为2-4%)、TP53(突变率在2-8%范围)、EZH2(突变约为2%)、JAK2(突变在1-3%范围)(Coombs CC等人,Molecular therapy for acute myeloid leukaemia.Nat Rev Clin Oncol.2016;13,305-318。Welch JS等人,The origin and evolution of mutations in acute myeloid leukemia.Cell.2012;150:264-278。Kandoth C等人,Mutational landscape and significance across 12 major cancer types.Nature 2013;502(7471):333-339。Ding L等人,Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing.Nature.2012;481:506-510。Hanahan D等人,The hallmarks of cancer.Cell.2000;100:57-70。The Cancer Genome Atlas Research Network Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia.N.Engl.J.Med.2013;368:2059-2074)。
蛋白激酶是细胞生长、发育、分化、代谢、老化与凋亡等重要生理功能所必须的蛋白质氨基酸磷酸化酶,主要有跨膜和胞质蛋白激酶两大类型。蛋白激酶的异常可直接导致临床上不同类型的疾病例如癌症、炎症、免疫系统、神经系统以及心脑血管类疾病。人们经过几十年的不断努力,许多蛋白激酶例如EGFR、HER2/3/4、VEGFRs、PDGFR、c-MET、IGF-1R、FGFRs、CSF-1R、TRK受体、Ephrin受体、TAM受体、TIE-2、FLT-3、RET、ALK、BCR-ABL、JAKs、SRC、FAK、BTK、SYK,BLK、CDK、PI3K、MEK/RAS/RAF等已被鉴定为临床不同疾病的靶蛋白分子。其中一些蛋白激酶抑制剂作为靶向治疗药物已成功的应用于临床,并且呈现出良好的治疗效果,例如BCR-ABL、EGFR/HER2、ALK、BTK、VEGFR、JAK等蛋白激酶抑制剂。FMS样酪氨酸激酶3(FMS-Like Tyrosine Kinase 3,FLT3)或称为胎肝激酶2(fetal liver kinase-2,FLK-2)或称为人干细胞激酶1(human stem cell kinase-1,STK-1),属于III型受体酪氨酸激酶。该激酶家族包括集落刺激因子1受体CSF1R、血小板衍生生长因子PDGFRα/β、干细胞因子受体KIT。在正常生长发育生理环境下,FLT3基因的表达主要集中在脑、肝、胎盘、生殖腺体和造血细胞的早期发育中。在髓系和淋巴系干细胞生长发育过程中,FLT3基因及其配体基因FLT3-L都呈现高表达,FLT3-L与FLT3结合诱导FLT3蛋白自身磷酸化,激活FLT3酶活性以及其介导的下游PI3K、JAK/STAT和RAS等信号传导途径,参与血液细胞的生长、发育、增殖与分化等生物学功能(Drexler HG等人,FLT3:receptor and ligand.Growth Factors.2004;22(2):71-3. Review)。例如,在FLT3基因缺失实验鼠中,髓系和淋巴系祖细胞数量减少。然而,当FLT3基因发生异常时如异常表达或突变,正常血细胞发生癌变,形成白血病。如上所述大约30%的急性髓系白血病(AML)患者发生FLT3内部串联重复(ITD,19-28%)和酪氨酸激酶域(TKD,5-10%)突变;中等或高危险骨髓增生异常综合征(MDS)病人中,FLT3突变率为2%;APL病人中,FLT3突变率小于5%;在ALL中的发生率小于1%,且主要见于双表型的ALL病例。
FLT3的上述两种突变,包括FLT3-ITD/FLT3-TKD双突变,均能引起FLT3蛋白自身磷酸化,导致FLT3配体非依赖性激活及其下游信号传导的异常,从而起到促进白血病细胞恶性增殖以及抑制细胞的正常凋亡。FLT3酪氨酸激酶激活突变是AML中最主要的突变之一,也是AML的主要病因之一。由于FLT3-ITD克隆具有选择性生长优势,单独使用普通化疗药物难以治愈此类白血病,同时此类白血病患者耐受化疗药物的作用力强,临床愈后差,患者易对化疗药物产生抗性并且容易复发,所以FLT3酪氨酸激酶激活突变已成为AML靶向治疗的重要靶标(Gilliland DG,Griffin JD.The roles of FLT3 in hematopoiesis and leukemia.Blood.2002;100(5):1532-1542。Kiyoi H.等人,Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product.Leukemia.1998;12(9):1333-1337)。
FLT3-ITD和FLT3-TKD抑制剂一直是AML研究领域备受关注的药物研发之一。至今为止,临床前研究发现近上百个不同类型的小分子化合物能够体外选择性/非选择性抑制或部分抑制FLT3蛋白激酶活性以及FLT3突变型表达阳性白血病细胞系或白血病病人样本细胞的体外增值和异种移植体内肿瘤生长,其中有的已进入临床试验的不同阶段,例如CEP701、CHIR-258、PKC412、MLN-518、舒尼替尼、AC220、XL-999、索拉非尼、帕纳替尼、Crenolanib、ASP2215、AKN-028、TAK-659、E6201、卡博替尼、PLX3387和FLX925(Smith BD等人,Single-agent CEP-701,a novel FLT3 inhibitor,shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia.Blood.2004;103(10):3669-76;Lopes de Menezes DE等人,CHIR-258:a potent inhibitor of FLT3 kinase in experimental tumor xenograft models of human acute myelogenous leukemia.Clin Cancer Res.2005;11(14):5281-91;Weisberg E等人,Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412.Cancer Cell.2002;1(5):433-43;Zarrinkar PP等人,AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia(AML).Blood.2009;114(14):2984-92;Kelly LM等人,CT53518,a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia(AML).Cancer Cell.2002;1(5):421-32;Griswold IJ等人,Effects of MLN518,a dual FLT3 and KIT inhibitor,on normal and malignant hematopoiesis.Blood.2004;104(9):2912-8;Smith CC等人,Crenolanib  is a selective type I pan-FLT3 inhibitor.Proc Natl Acad Sci U S A.2014;111(14):5319-24;Zimmerman EI等人,Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid leukemia.Blood.2013;122(22):3607-15;Safaian NN等人,Sorafenib(Nexavar) induces molecular remission and regression of extramedullary disease in a patient with FLT3-ITD+acute myeloid leukemia.Leuk Res.2009;33(2):348-50;Zhang W等人,Mutant FLT3:a direct target of sorafenib in acute myelogenous leukemia.J Natl Cancer Inst.2008;100(3):184-98)。尽管有些FLT3抑制剂在临床试验早期呈现鼓舞人心的结果,AML病人病情得到改善,但大多数化合物在临床试验的后期无论单独应用或联合治疗AML病人时,并没有呈现出预期的临床效果,至今为止全球尚没有一个FLT3选择性抑制剂被批准用于AML临床治疗。
由于AML疾病的特殊性,综合与分析现有FLT3抑制剂临床前和临床试验数据,我们发现上述多数FLT3抑制剂存在不同的问题,限制其临床效果例如(1)严重副作用;(2)影响正常血细胞的生长与发育,降低病人自身免疫力;(3)获得性药物抗性;(4)融瘤副作用;(5)病人应答率低以及(6)易复发。其主要原因与化合物的选择性、活性、体内代谢、毒性及有效性相关(Kadia TM等人,New Drugs in Acute Myeloid Leukemia(AML).Ann Oncol.2016;27(5):770-8。Stein EM等人,Emerging therapeutic drugs for AML.Blood.2016;127(1):71-8。Stein EM,Molecularly targeted therapies for acute myeloid leukemia.Hematology Am Soc Hematol Educ Program.2015;(1):579-83)。
MV4-11、MOLM-13和MOLM-14三种人类白血病细胞系是当今应用最广的FLT3-ITD表达细胞。其中MV4-11细胞含有FLT3 ITD纯合子突变(+/+),属于人急性淋巴髓单核细胞白血病。MOLM-13和MOLM-14是来自同一病人的姊妹细胞株,含有FLT3 ITD/WT杂合体突变(+/-),属于人急性髓系白血病(Quentmeier H等人,FLT3 mutations in acute myeloid leukemia cell lines.Leukemia.2003;17(1):120-124)。许多研究已证实靶向抑制FLT3-ITD就能有效抑制这三种白血病细胞的体内和体外生长,这些细胞系特别是MV4-11已成为FLT3-ITD选择性抑制剂筛选与鉴定的常用细胞模型。
本发明利用(1)FLT3-ITD表达阳性细胞系MV4-11和MOLM-13;(2)FLT3基因野生型高表达阳性细胞系RS4;11;(3)其它临床上常见的癌基因表达阳性白血病细胞系以及不同类型的实体瘤细胞系作为细胞模型,致力于开发一种高活性、高选择性、良好药学性质和生物体内代谢参数的低副作用的新型化合物,其可以作为一种有效的FLT3酪氨酸蛋白激酶(激活型突变)选择性抑制剂,用于预防和/或治疗癌症特别是白血病。
发明内容
本发明涉及一种新型的脲类化合物,其能够有效的抑制FLT3-ITD突变型表达白血病细胞系MV4-11和MOLM-13的体外生长,诱导细胞凋亡,其GI50为亚纳摩尔范围,而对FLT3野生型高表达或正常表达或无表达癌细胞系生长相对无明显抑制作用。同时本发明化合物能够有效地、快速地、浓度依赖性地抑制FLT3-ITD白血病细胞MV4-11异种移植动物体内的肿瘤生长。进一步的药物代谢动力学和药学研究发现本发明化合物在大鼠体内呈现良好的药学性质。
因此,本发明提供一种通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
Figure PCTCN2017076265-appb-000002
其中:
X、Y各自独立地选自N或者C-BR1
A、A1各自独立地选自N或者C-BR1
W、Z各自独立地选自N或者C-BR1
当R1不存在时,B相同或不同且各自独立地选自:氢、卤素、烷基、烯基、炔基、氰基、环烷基、杂环基、芳基、或杂芳基,所述烷基、烯基、炔基、环烷基、杂环基、芳基、或杂芳基任选进一步被一个或多个Q基团取代;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-;并且,R1相同或不同且各自独立地选自:氢、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基、-RuORx、-RuC(O)ORx、-RuN(Ry)(Rz)、-C(O)N(Ry)(Rz)、-RuS(O)nN(Ry)(Rz)、-RuS(O)nRx;所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氰基、羟基、烷基、烷氧基、羟烷基、羟烷氧基、酰胺基、环烷基、杂环基、芳基、卤代芳基、杂芳基、环烷基-杂芳基的一种或多种基团取代;R4选自氢、烷基、烯基和炔基,或者R4、R1与相连接的氮原子一起形成杂环基或杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基的一种或多种基团取代;
当R2为氢时,G选自芳基、杂芳基或杂环基,所述芳基、杂芳基或杂环基任选进一步被选自卤素、烷基、烯基、炔基、烷氧基、羟基、氨基、酰基、环烷基、杂环基、芳基、杂芳基的一种或多种基团所取代,其中所述烷基、烯基、炔基、烷氧基、酰基、环烷基、杂环基、芳基或杂芳基任选进一步被选自卤素、烷基、 卤代烷基、烯基、炔基、芳基、羟基、烷氧基、卤代烷氧基、环烷基、酯基和氰基的一种或多种基团所取代;或者,
当R2不为氢时,G与R2与它们所连接的氮原子一起形成杂环基或杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、烷基、烯基、炔基、烷氧基、羟基、氨基、环烷基、杂环基、芳基、和杂芳基的一种或多种基团所取代,其中所述烷基、烯基、炔基、烷氧基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、烷基、卤代烷基、烯基、炔基、芳基、羟基、烷氧基、卤代烷氧基、环烷基、酯基和氰基的一种或多种基团所取代;
R3选自Q基团;
Ru选自一个键、亚烷基、亚烯基、或亚炔基;
Rx选自氢、烷基、羟烷基、卤代烷基、烯基、或者炔基;或者,
-RuORx-中氧与相连接的Ru和Rx一起形成含氧的3-7元杂环,所述杂环任选被一种或多种Q基团取代;
Ry和Rz各自独立地选自氢、烷基、烷氧基、烯基、炔基、环烷基、或者卤代烷基;或者,
Ry和Rz与它们所连接的氮原子一起形成杂环基或者杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、卤代烷基、烷基、烯基和炔基的一种或多种基团所取代;
Q选自氢、卤素、羟基、氨基、烷基、烷氧基、环烷基、烯基、炔基、氰基、芳基、杂环基或杂芳基,所述氨基、烷基、烷氧基、环烷基、烯基、炔基、芳基、杂环基或杂芳基任选进一步被选自羟基、卤素、烷基的一种或多种基团取代;
n为0-2的整数。
在本发明一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
其中:
X、Y、A、A1选自如下结构:
Figure PCTCN2017076265-appb-000003
R5 1、R5 2、R5 3、R5 4、R5 5、R5 6、R5 7、R5 8和R5 9各自独立地选自氢、卤素、羟基、烷基、烷氧基、烯基、炔基、-N(Ry)(Rz)、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烷氧基、烯基、炔基、-N(Ry)(Rz)、环烷基、杂环基、芳基或杂芳 基任选进一步被选自卤素、羟基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、酯基的一种或多种基团所取代。
R1、B、Ry、Rz如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
其中:
X、Y、A、A1选自如下结构:
Figure PCTCN2017076265-appb-000004
R5 1、R5 2、R5 3、R5 4和R5 5各自独立地选自氢、卤素、烷基、烷氧基、-N(Ry)(Rz)、卤代烷基、卤代烷氧基。
R1、B、Ry、Rz如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
其中:
X、Y、A、A1选自如下结构:
Figure PCTCN2017076265-appb-000005
R5 1、R5 2、R5 3各自独立地选自氢、卤素、烷基、烷氧基、-N(Ry)(Rz)、卤代烷基、卤代烷氧基。
R1、B、Ry、Rz如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
R5 1、R5 2、R5 3各自独立地选自氢、卤素、C1-C6烷基、C1-C6烷氧基、-N(Ry)(Rz)、C1-C6卤代烷基、C1-C6卤代烷氧基,其中Ry、Rz各自独立地选自氢、C1-C6烷基。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,W、Z选自如下4种方式:
a)W、Z为CQ;
b)W、Z为N;
c)W为CQ且Z为N;
d)Z为CQ且W为N;
其中,Q选自氢、卤素、羟基、氨基、氰基、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、C3-C7环烷基、C5-C7芳基、5至7元杂环基或5至7元杂芳基。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
R1不存在,B相同或不同且各自独立地选自:氢、卤素、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:氢、C1-C10烷基,所述烷基任选进一步被选自卤素、氰基、羟基、C1-C6烷氧基、4~6元杂环基、C5~C7芳基、C5~C7卤代芳基、5~7元杂芳基、C3~C6环烷基、5~7元杂芳基的一种或多种基团取代,其中所述4~6元杂环基优选含氧或氮的4~6元杂环基;所述C5~C7芳基或C5~C7卤代芳基优选苯基或卤代苯基;
R4如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-RuORx,其中Ru选自C1-C6亚烷基,Rx选自氢、C1-C6烷基、C1-C6羟烷基、C1-C6卤代烷基;
R4如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-C(O)N(Ry)(Rz),其中Ry和Rz各自独立地选自氢、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C3-C7环烷基;或者,
Ry和Rz与它们所连接的氮原子一起形成5~7元杂环基或5~7元杂芳基,优选6元杂环基或6元杂芳基,更优选吗啉基、哌啶基、哌嗪基、吡啶基、嘧啶基, 所述5~7元杂环基或5~7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基的一种或多种基团所取代;
R4如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-RuN(Ry)(Rz),其中Ru选自C1-C6亚烷基;Ry和Rz各自独立地选自氢、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C3-C7环烷基;或者,
Ry和Rz与它们所连接的氮原子一起形成5~7元杂环基或者5~7元杂芳基,优选吗啉基、哌啶基、哌嗪基、氮杂环庚烷基、吡啶基、嘧啶基,所述5~7元杂环基或5~7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基的一种或多种基团所取代;
R4如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-RuC(O)ORx,其中:Ru选自C1-C6亚烷基;Rx选自氢、C1-C6烷基、C1-C6羟烷基、C1-C6卤代烷基;
R4如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:5~7元芳基或5~7元杂芳基,优选噻二唑基、吡唑基、噁唑基、噁二唑基、咪唑基、三唑基、噻唑基、呋喃基、噻吩基、吡啶基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、咪唑基、四唑基、苯基、吡啶基、嘧啶基,所述5~7元芳基或5~7元杂芳基任选进一步被选自C3~C6环烷基、5~7元杂环基、酰胺基的一种或多种基团取代;
R4如通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
R4选自氢或C1-C6烷基,或者R4、R1与相连接的氮原子一起形成5~7元杂环基或5~7元杂芳基,优选哌啶基、哌嗪基、吗啉基、吡啶基、嘧啶基,所述5~7元杂环基或5~7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基的一种或多种基团取代。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
当R2为氢时,G选自C5~C7芳基、5至7元杂芳基或5至7元杂环基,优选
Figure PCTCN2017076265-appb-000006
所述C5~C7芳基、5至7元杂芳基或5至7元杂环基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、羟基、氨基、酰基、C3~C7环烷基、5~7元杂环基、C5~C7芳基、5~7元杂芳基的一种或多种基团所取代;所述C1-C6烷基、C1-C6烷氧基、C3~C7环烷基、5~7元杂环基、C5~C7芳基、5~7元杂芳基任选进一步被选自卤素、羟基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、酯基和氰基的一种或多种基团所取代;或者,
当R2不为氢时,G与R2与它们所连接的氮原子一起形成5-7元杂环基或5-7元杂芳基,优选吡咯基、吡唑基、咪唑基,所述5-7元杂环基或5-7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、羟基、氨基、C3~C7环烷基、5~7元杂环基、C5~C7芳基、5~7元杂芳基的一种或多种基团所取代。
在本发明另一个优选的实施方案中,根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
R3选自氢、卤素、羟基、氨基、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、C3~C7环烷基、氰基、C5~C7元芳基、5~7元杂环基或5~7元杂芳基。
本发明典型的通式(I)所示的化合物包括但不限于:
1-(4-苯并咪唑-1-基-苯基)-3-异噁唑-3-基-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-异噁唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-羟基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[2-(2-羟基-乙氧基)-乙氧基]-苯并咪唑-1-基}-苯基)-脲;
吗啉-4-羧酸-1-{4-[3-(5-叔丁基-异噁唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基酯;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-二甲基氨基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-{6-[(2-二甲基氨基-乙基)-甲基-氨基]-苯并咪唑-1-基}-苯基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-{4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(4-氟-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-氯苯并咪唑-1-基)-苯基]-脲;
1-(4-苯并咪唑-1-基-3-甲基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(4-苯并咪唑-1-基-3-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(4-苯并咪唑-1-基-3-氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(4-苯并咪唑-1-基-3,5-二氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(4-苯并咪唑-1-基-2-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(6-苯并咪唑-1-基-吡啶-3-基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(2-苯并咪唑-1-基-嘧啶-5-基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-吲哚-1-基-苯基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-吲唑-1-基-苯基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-氟-吲唑-1-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-氟-吲唑-1-基)-苯基]-脲;
1-(4-苯并三唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-吡咯并[2,3-b]吡啶-1-基-苯基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-3-基-苯基)-脲;
1-[4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯基]-3-(5-叔丁基-异噁唑-3-基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-甲基咪唑[4,5-b]吡啶-3-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-1-基-苯基)-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-嘌呤-7-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基嘌呤-9-基)-苯基]-脲;
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-二甲基氨基-嘌呤-7-基)-苯基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-噻唑-2-基-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(4-甲基-噻唑-2-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-[1,3,4]噻二唑-2-基-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-[1,3,4]噻二唑-2-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-1H-吡唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-苯基-1H-吡唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-环丙基-2H-吡唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-三氟甲基-2H-吡唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2H-吡唑-3-基)-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲;
1-[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-3-(5-叔丁基-2H-吡唑-3-基)-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丁氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-3-甲氧基-丙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二甲氨基-丙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二丁基氨基丙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氰基甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-2H-吡唑-3-基)-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-氟-苄氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[4-(1-环己基-1H-四唑-5-基)-丁氧基]-苯并咪唑-1-基}-苯基)-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(4-吗啉-4-基-[1,2,5]噻二唑-3-基氧基)-苯并咪唑-1-基]-苯基}-脲;
4-(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H苯并咪唑-5-基氧基)-吡啶-2-羧酸甲基胺;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-甲基-2H-吡唑-3-基)-脲;
1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-甲氧基)-苯并咪唑-1-基]-苯基}-脲;
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-羟基-乙基)-2H-吡唑-3-基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-苯基-2H-吡唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-对甲苯基-2H-吡唑-3-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-甲氧基-苯基)-2H-吡唑-3-基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-三氟甲氧基-苯基)-2H-吡唑-3-基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-氟-苯基)-2H-吡唑-3-基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-氟-苯基)-2H-吡唑-3-基]-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-吡啶-2-基-2H-吡唑-3-基)-脲;
4-{5-[3-(4-苯并咪唑-1-基-苯基)-脲基]-3-叔丁基-吡唑-1-基}-苯甲酸乙酯;
1-(2-丙烯酰基-5-叔丁基-2H-吡唑-3-基)-3-(4-咪唑-1-基-苯基)-脲;
3-氨基-5-甲基吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
5-氨基-3-环丙基吡唑-1-甲酸(4-苯并咪唑-1-基-苯基)-酰胺;
5-氨基-3-三氟甲基吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-己氧基-苯并咪唑-1-基)-苯基]-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-酰胺;
(1-{4-[(5-氨基-3-叔丁基-吡唑-1-羰基)-氨基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯;
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-氟-苯并咪唑-1-基)-苯基]-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-酰胺;
4-(1-{4-[(5-氨基-3-叔丁基-吡唑-1-羰基)-氨基]-苯基}-1H-苯并咪唑-5-基氧基)-吡啶-2-羧酸甲基酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-酰胺;
3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
1-(4-苯并咪唑-1-基-苯基)-3-(3,4-二甲基-异噁唑-5-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(3-异丙基-异噁唑-5-基)-脲;
1-(4-苯并咪唑-1-基-苯基)-3-(3-叔丁基-异噁唑-5-基)-脲。
本发明另一方面提供通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的制备方法,其包括以下步骤:
Figure PCTCN2017076265-appb-000007
在适宜的温度、pH和适当的溶剂中,在碱的作用下,使式(II)化合物与式(III)化合物反应,得到通式(I)的化合物;
所述溶剂优选THF、乙腈、二氯甲烷、甲苯,所述碱优选三乙胺、N,N-二异丙基乙胺、DMAP、吡啶;
X、Y、A、A1、Z、W、R1、B、R2、G、R3如通式(I)中所定义。
本发明另一方面提供通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的制备方法,其包括以下步骤:
Figure PCTCN2017076265-appb-000008
在适宜的温度、pH和适当的溶剂中,在碱的作用下,使式(IV)化合物与式(V)化合物反应,得到通式(I)的化合物;
所述溶剂优选THF、乙腈、二氯甲烷、甲苯,所述碱优选三乙胺、N,N-二异丙基乙胺、DMAP、吡啶;
X、Y、A、A1、Z、W、R1、B、R2、G、R3如通式(I)中所定义。
本发明进一步涉及一种药物组合物,其含有治疗有效量的根据本发明的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药作为活性成分,以及一种或多种药学上可接受的载体。
本发明进一步涉及通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者含有其的药物组合物,在制备FLT3酪氨酸蛋白激酶抑制剂中的用途。
本发明进一步涉及通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者含有其的药物组合物,在制备用于预防和/或治疗哺乳动物包括人中癌症的药物的用途。所述癌症包括但不限于,非实体瘤如白血病,实体瘤如皮肤癌、黑色素瘤、肺癌、胃癌、乳腺癌、胰腺癌、肝癌和结肠癌等。
本发明进一步涉及一种通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者含有其的药物组合物,其用作FLT3酪氨酸蛋白激酶抑制剂的用途。
本发明进一步涉及一种通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者含有其的药物组合物,其用作预防和/或治疗哺乳动物包括人中癌症的药物的用途。所述癌症包括但不限于,非实体瘤如白血病,实体瘤如皮肤癌、黑色素瘤、肺癌、胃癌、乳腺癌、胰腺癌、肝癌和结肠癌等。
本发明进一步涉及一种抑制FLT3酪氨酸蛋白激酶的方法,其包括向需要其的患者施用抑制有效量的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者含有其的药物组合物。
本发明进一步涉及一种预防和/或治疗治疗哺乳动物包括人中癌症的方法,其包括向需要其的患者施用治疗有效量的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者含有其的药物组合物。所述癌症包括但不限于,非实体瘤如白血病,实体瘤如皮肤癌、黑色素瘤、肺癌、胃癌、乳腺癌、胰腺癌、肝癌和结肠癌等。
具体实施方式
除非另有规定,本文使用的所有技术和科学术语具有与本领域技术人员的通常理解相同含义。所有专利、申请、公开的申请和其他出版物均以全部内容并入作为参考。倘若对于本文使用的术语有多个定义,除非另有说明,以本节中的为准。如果任何给定取代基的数量没有规定,则可以存在一个或多个取代基。例如“卤代烷基”可以含有一个或多个相同或不同的卤素。在本文的描述中,如果化学结构和化学名称彼此矛盾时,则是以其化学结构为准。当在本文使用时,对于任何保护基团、氨基酸和其他化合物的缩写,除非另有说明,以其常用的公认缩写表示,或根据IUPAC-IUB Commission on Biochemical Nomenclature表示(参见,Biochem.1972,77:942-944)。
除非有相反陈述,否则下列用在说明书和权利要求书中的术语具有下述含义。
术语“烷基”指饱和的脂族烃基团,包括1至20个碳原子的直链和支链基团。其包括碳数1~18、优选碳数1~10、更优选碳数1~6、甚至更优选碳数1~4的直链或支链烷基。非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、异戊基、新戊基、己基、异己基、正庚基、异庚基、正辛基、异辛基、正壬基、正癸基等。本说明书中,“烷基”还包括碳数3~10、优选碳数3~8、更优选碳数4~6的环状烷基,例如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基、环癸基、十氢萘基、降冰片烷、金刚烷基。烷基可以是取代的或未取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,优选为一个或多个以下基团,独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“烯基”指由碳和氢原子组成的含有至少一个双键的直链或支链的烃链基团,并通过单键或双键与分子的其余部分连接。优选具有2~10个碳原子,更优选具有2~6个碳原子,甚至更优选具有2~4个碳原子。非限制性实施例包括乙烯基、丙烯基、丁烯基、戊烯基、戊二烯基、己烯基。烯基可以是取代的或非 取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“炔基”指由碳原子和氢原子组成的含有至少一个三键的直链或支链的烃链基团,并通过单键或三键与分子的其余部分连接。优选具有2~10个碳原子,更优选具有2~6个碳原子,甚至更优选具有2~4个碳原子。非限制性实施例包括乙炔基、丙炔基、丁炔基、戊炔基、己炔基。炔基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,其包括3至20个碳原子,优选包括3至12个碳原子,更优选环烷基环包含3至10个碳原子,最优选环烷基环包含3至7个碳原子。单环环烷基的非限制性实施例包含环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等,优选环丙基、环己烯基。多环环烷基包括螺环、稠环和桥环的环烷基。环烷基可以是任选取代的或未取代的,当被取代时,取代基优选为一个或多个以下基团,独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包括3至20个环原子,其中一个或多个环原子选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包括3至12个环原子,其中1~4个是杂原子,更优选杂环基环包含3至10个环原子,更优选杂环基环包含5至7个环原子。单环杂环基的非限制性实施例包含吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吡喃基、四氢呋喃基等。多环杂环基包括螺环、稠环和桥环的杂环基。杂环基可以是任选取代的或未取代的,当被取代时,取代基优选为一个或多个以下基团,独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为5至10元,更优选5至7元,甚至更优选苯基和 萘基,最优选苯基。芳基可以是完全芳香族的基团,例如苯基、萘基、蒽基、菲基等。芳基也可以含有芳香环与非芳香环的组合,例如,茚、芴和苊等。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,非限制性实施例包含:
Figure PCTCN2017076265-appb-000009
芳基可以是取代的或未取代的,当被取代时,取代基优选为一个或多个以下基团,独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,更优选5至7元,甚至更优选为5元或6元,例如噻二唑基、吡唑基、噁唑基、噁二唑基、咪唑基、三唑基、噻唑基、呋喃基、噻吩基、吡啶基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、咪唑基、四唑基等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2017076265-appb-000010
杂芳基可以是任选取代的或未取代的,当被取代时,取代基优选为一个或多个以下基团,独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“烷氧基”指-O-(烷基)和-O-(未取代的环烷基),其中烷基、环烷基的定义如上所述。非限定性实例包括甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基等。烷氧基可以是任选取代的或未取代的,当被取代时,取代基优选为一个或多个以下基团,独立地选自为烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氨基、卤代烷基、羟烷基、羧基或羧酸酯基。
术语“卤代烷基”指其中一个或多个氢原子被卤素取代的烷基,其中烷基的定义如上所述。非限定性实例包括氯甲基、三氟甲基、1-氯-2-氟乙基、2,2-二氟 乙基、2-氟丙基、2-氟丙-2-基、2,2,2-三氟乙基、1,1-二氟乙基、1,3-二氟-2-甲基丙基、2,2-二氟环丙基、(三氟甲基)环丙基、4,4-二氟环己基和2,2,2-三氟-1,1-二甲基-乙基。
术语“卤素”包括氟、氯、溴和碘。
术语“氰基”指-CN。
术语“羟基”指-OH基团。
术语“羟烷基”指被羟基取代的烷基,其中烷基的定义如上所述。
术语“羟烷氧基”指被羟基取代的烷氧基,其中烷氧基的定义如上所述。
术语“酰基”指-C(O)R,其中R指烷基、环烷基、烯基、炔基,其中烷基、环烷基、烯基、炔基的定义如上所述。非限定性实例包括乙酰基、丙酰基、丁酰基、戊酰基、己酰基、乙烯酰基、丙烯酰基。
术语“酰胺基”指-NHC(O)R,其中R指烷基、烯基、炔基,其中烷基、烯基、炔基的定义如上所述。非限定性实例包括甲酰胺基、乙酰氨基、丙酰胺基、丁酰胺基、戊酰胺基、己酰胺基、乙烯酰胺基、丙烯酰胺基。
术语“酯基”指-C(O)OR,其中R指烷基或环烷基,其中烷基、环烷基的定义如上所述。非限定性实例包括乙酯基、丙酯基、丁酯基、戊酯基、环丙酯基、环丁酯基、环戊酯基、环己酯基。
本说明书中的“任选取代”是指未取代或被一个或多个(例如2、3、4个)取代基取代。其中取代基选自下组:卤素原子、烷基、烯基、炔基、卤代烷基、烷氧基、芳基、卤代芳基、芳氧基、芳烷基、芳烷基氧基、杂环基烷氧基、卤代芳基烷基氧基、烷基氨基、烷基酰基、氰基、或杂环基等。这些取代基还可以进一步被取代。例如,作为取代基的烷基还任选被选自卤素原子、羟基、烷氧基、烷基氨基、吡咯烷基、苯基、吡啶基、或卤代苯基中的一个或多个基团取代。作为取代基的杂环基还任选被选自卤素原子、烷基、烷氧基中的一个或多个基团取代。
本发明通式(I)所示的化合物的制备方法。
为了完成本发明化合物的目的,本发明主要采用如下合成路线与技术方案。
Figure PCTCN2017076265-appb-000011
本发明化合物的第一种合成方法为,将本发明通式(I)化合物的结构如上划分为A部分和B部分,其中A部分为胺类中间体式(II)化合物,B部分为活泼酯中间体式(III)化合物。
Figure PCTCN2017076265-appb-000012
1、A部分胺类中间体式(II)化合物的合成方法如下所示。
方法1:通过取代反应合成A部分胺类中间体
a)苯并咪唑类中间体1或中间体2或中间体3的合成如下方案1所示,
Figure PCTCN2017076265-appb-000013
首先,以苯并咪唑类中间体M起始,在适当的温度和pH条件下,在碱催化下,在适当的溶剂中通过取代反应得到中间产物;所述碱可以为例如碳酸钾、碳酸铯等,所述溶剂可以为例如DMF、乙腈等。然后,将中间产物的硝基还原为氨基,得到中间体1、2、3;硝基的还原可以在例如铁粉氯化铵体系或H2/钯碳体系下实现。
b)其它类型中间体的合成如下方案2所示,
Figure PCTCN2017076265-appb-000014
与上述苯并咪唑类中间体的合成方法相同,只是用相应的起始原料代替苯并咪唑类中间体M得到相应的其它类型的中间体。
方法2:通过关环反应合成A部分胺类中间体
a)苯并咪唑类中间体4、中间体5、中间体6和中间体7的合成如下方案3(方法1)和方案4(方法2)所示,
Figure PCTCN2017076265-appb-000015
步骤1:中间体M1与对溴硝基苯于适当溶剂中在碱、催化剂和配体作用下进行Buchwald反应得到中间体M2;所述溶剂优选二氧六环、甲苯,所述碱优选叔丁醇钠、叔丁醇钾、碳酸铯,所述催化剂优选(pd)2(dba)3、醋酸钯、pd(dba)2;所述配体优选Xphos、BINAP;
步骤2:在还原条件下,将中间体M2的两个硝基还原氨基,得到中间体M3,所述还原条件可以为例如铁粉氯化铵体系或H2/钯碳体系;
步骤3:在酸性条件下,高温下中间体M3和甲酸经历关环反应,得到中间体4,所述酸性条件例如盐酸;
步骤4:R0选自甲氧基时:在酸性条件下,加热回流中间体4得到中间体5,所述酸性条件例如在HBr下;
步骤5:在碱的催化下,在适当的溶剂中和温度下,使中间体5与相应的活泼卤代物反应得到中间体6,所述碱例如氢氧化钠、碳酸钾等,所述溶剂例如DMF等;
Figure PCTCN2017076265-appb-000016
步骤1:在碱存在下,在适当的溶剂中,使2-硝基-5-氟苯胺与中间体M4发生取代反应,得到中间体M5,所述碱可以为例如碳酸钾等,所述溶剂可以为如DMF等;
步骤2:中间体M5与对溴硝基苯于适当溶剂中在碱、催化剂和配体作用下进行Buchwald反应得到中间体M6;所述溶剂优选二氧六环、甲苯,所述碱优选叔丁醇钠、叔丁醇钾、碳酸铯,所述催化剂优选(pd)2(dba)3、醋酸钯、pd(dba)2;所述配体优选Xphos、BINAP;
步骤3:在还原条件下,将中间体M6的两个硝基还原氨基,得到中间体M7,所述还原条件可以为例如铁粉氯化铵体系或H2/钯碳体系;
步骤4:在酸性条件下,高温下中间体M7和甲酸经历关环反应,得到中间体7,所述酸性条件例如盐酸。
b)其它类型中间体的合成如下方案5所示
Figure PCTCN2017076265-appb-000017
步骤1:在碱存在下,在适当的溶剂中,使中间体M8与中间体M9发生取代反应,得到中间体M10,所述碱可以为例如三乙胺,所述溶剂可以为例如DMSO;
步骤2:在还原条件下,将中间体M10的两个硝基还原氨基,得到中间体M11,所述还原条件可以为例如铁粉氯化铵体系或H2/钯碳体系;
步骤3:在酸性条件与高温条件下,中间体M11和甲酸经历关环反应,得到中间体8,所述酸性条件例如盐酸;
2、B部分活泼酯中间体式(III)化合物的合成
1)吡唑类中间体的合成如下方案6所示,
Figure PCTCN2017076265-appb-000018
在适宜的温度与pH条件下,在适当的溶剂中,在酸催化下反应得到吡唑类中间体,所述溶剂可以为例如乙醇,所述酸可以为例如盐酸。
2)其它异噁唑类、噁唑类、噻唑类、噻二唑类中间体均可以商业获得。
3)活泼酯的合成方法
如果B部分中间体只有一个成脲位点,则将其制成活泼酯,如下方案7所示,
Figure PCTCN2017076265-appb-000019
在适宜的温度与pH条件下,在适当的溶剂中,在碱的催化下,使氯甲酸苯酯与相应的胺(中间体M12)反应,得到相应的活泼酯,其中所述溶剂可以为例如乙酸乙酯、二氯甲烷、四氢呋喃、丙酮、乙腈、水等,所述碱可以为例如吡啶、碳酸氢钠、碳酸钾、三乙胺、氢氧化钠等。由于与氨基相连的基团各不相同,导致其活性不尽相同,因此反应时所选择的碱略有不同,本领域技术人员可以根据本领域普通技术知识进行常规选择。
如果B部分中间体有两个可成脲位点(-NH-和-NH2-),例如:
Figure PCTCN2017076265-appb-000020
则将A部分胺类中间体制成活泼酯。即成为本发明化合物的第二种合成方法,将本发明通式(I)化合物的结构划分为A部分和B部分,其中A部分为活泼酯中间体式(IV)化合物,B部分为胺类中间体式(V)化合物。
Figure PCTCN2017076265-appb-000021
其中,A部分活泼酯中间体式(IV)化合物的制备方法与如上B部分为活泼酯的制备方法相似。即为,在适宜的温度与pH条件下,在适当的溶剂中,在碱的 催化下,将A部分的胺类中间体式(II)化合物与氯甲酸苯酯反应,得到A部分活泼酯中间体。
最后,如前所述,将中间体式(II)化合物与式(III)化合物,或者中间体式(IV)化合物与式(V)化合物,在适宜的温度、pH和适当的溶剂中,在碱的作用下,使活泼酯脱掉一分子苯酚得到相应的异氰酸酯中间体,再在碱的作用下和相应的胺反应,生成最终的脲类化合物即通式(I)的化合物。所述溶剂优选THF、乙腈、二氯甲烷、甲苯,所述碱优选三乙胺、N,N-二异丙基乙胺、DMAP、吡啶;
其中,X、Y、A、A1、Z、W、R1、B、R2、G、R3如通式1中所定义,R0、R0 1、R0 2在无特殊说明下,与-BR1的定义相同。
本发明通式(I)所示的化合物,其前药应遵照前药设计原则,在生物体内正常生理状况下,能够通过酶解、水解、酸解或代谢降解,释放出原活性通式(I)所示的化合物。这里包括但不限于化合物上羟基基团的脂化(如形成磷酸脂和碳酸脂)、氨基基团和羧基基团的保护。前药设计参照(1)Karaman R,Prodrugs design based on inter-and intramolecular chemical processes.Chem Biol Drug Des.82(6):643-68,2013;(2)Rautio J等,Prodrugs:design and clinical applications.Nat Rev Drug Discov.7(3):255-702008;(3)Jampilek J,Prodrugs:pharmaceutical design and current perspectives.Curr Pharm Des.17(32):3480-1,2011;(4)Bundgaard H.Design of Progrugs.Elservier,1985。
本发明通式(I)所示的化合物在药学上可接受的盐,可以为酸加成盐或碱加成盐。酸可以为无机酸,包括但不限于:盐酸、硫酸、磷酸、氢溴酸;或可以为有机酸,包括但不限于:柠檬酸、马来酸、草酸,甲酸、乙酸、丙酸、戊酸、乙醇酸、苯甲酸、富马酸、三氟乙酸、琥珀酸、酒石酸、乳酸、谷氨酸、天门冬氨酸、水杨酸、丙酮酸、甲磺酸、苯磺酸、对苯磺酸和右旋樟脑磺酸等。碱可以为无机碱,包括但不限于:氢氧化钠、氢氧化钾、氢氧化镁、氢氧化钙;或可以为有机碱,包括但不限于:氢氧化铵、三乙胺、N,N-二苄基乙二胺、氯普鲁卡因、胆碱、氨、二乙醇胺和其他羟基烷基胺、乙二胺、N-甲基葡糖胺、普鲁卡因、N-苄基苯乙胺、精氨酸或赖氨酸;或可以为碱金属盐,包括但不限于:锂、钾和钠盐;或可以为碱土金属盐,包括但不限于:钡、钙和镁盐;或可以为过渡金属盐,包括但不限于锌盐;或其他金属盐,包括但不限于:磷酸氢钠和磷酸氢二钠。
本发明另一方面将通式(I)所示的化合物或药学上可接受的盐或前药制备成临床上可使用的药用组合物。根据临床适应症,给药途径与方式,其药用制剂包括但不限于口服制剂如片剂、凝胶剂、软/硬胶囊、乳剂、分散性粉剂、颗粒剂、水/油悬乳剂;注射剂包括静脉注射剂、肌肉注射剂、腹腔注射剂、直肠给药栓剂、颅内注射剂,这些剂型可为水溶液也可为油类溶液;局部制剂包括霜剂、软膏剂、凝胶剂、水/油溶液以及包合物制剂;吸入剂型包括细粉、液体气溶胶以及适合于体内植入的各种剂型。
本发明的药物组合物可以根据需要加入药学上可接受的载体、稀释剂或赋形剂。这些载体、稀释剂或赋形剂应符合药物制剂制备工艺规则,与活性成分相兼容。固体口服制剂的载体包括但不限于甘露醇、乳糖、淀粉、硬脂酸镁、纤维素、葡萄糖、蔗糖、环糊精以及促进肠吸收分子载体维生素E-PEG1000。口服制剂可加入适当的着色剂、甜味剂、矫味剂及防腐剂。
本发明通式(I)所示的化合物或药学上可接受的盐或前药,按0.01-100mg/kg单位剂量给予温血动物。但如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行被、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合等。因此,最佳的治疗方式如治疗的模式、通式(I)化合物的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
本发明通式(I)所示的化合物或药学上可接受的盐或前药,在上述癌症治疗中,可单独使用,或与临床上常规使用的放射疗法、化学疗法、免疫疗法、融瘤病毒、RNAi、癌症辅助治疗以及骨髓移植和干细胞移植的一种或多种方法联合治疗,其中包括但不限于以下抗肿瘤类药物和治疗方法:
1)烷化剂如顺铂、顺铂、奥沙利铂、苯丁酸氮芥、卡环磷酰胺,氮芥、美法仑、替莫唑胺、白消安、亚硝基脲类。
2)抗肿瘤抗生素类如阿霉素、博来霉素、多柔比星、道诺霉素、表柔比星、伊达比星、丝裂霉素C、放线菌素、光神霉素;抗有丝分裂药如长春新碱、长春碱、长春地辛、长春瑞滨、紫杉醇、泰索帝、Polo激酶抑制剂。
3)抗代谢和抗叶酸剂如氟嘧啶、雷甲氨蝶呤、阿糖胞苷、阿扎胞苷、地西他滨、替曲塞、羟基脲、IDH1/IDH2突变株抑制剂。
4)拓扑异构酶抑制剂如表鬼臼毒素、喜树碱、伊立替康。
5)细胞生长抑制剂如抗雌激素/抗雄激素类药物。如他莫昔芬、氟维司群、托瑞米芬、雷诺昔芬、屈诺昔芬、碘昔芬、比卡鲁胺、氟他胺、尼鲁米特、醋酸环丙孕酮;
LHRH拮抗剂或LHRH激动剂如戈舍瑞林、亮丙瑞林、和布舍瑞林、孕激素类如醋酸甲地孕酮;
芳香酶抑制剂如阿那曲唑、来曲唑、伏罗唑、伊西美坦、5a-还原酶抑制剂如非那雄胺。
6)抗侵袭剂如c-Src激酶家族抑制剂、金属蛋白酶抑制剂、尿激酶纤溶酶原激活物受体功能的抑制剂或者类肝素酶的抗体。
7)细胞生长功能抑制剂包括酪氨酸激酶抑制剂以及丝氨酸/苏氨酸激酶的抑制剂如Ras/Raf信号传导抑制剂,MEK和/或AKT激酶的细胞信号传导抑制剂、c-Kit抑制剂、c-Met抑制剂、PDGFR抑制剂、ABL激酶抑制剂、PI3激酶抑制剂、 CSF-1R激酶抑制剂、EGFR家族激酶抑制剂、FGFR家族激酶抑制剂、IGF受体激酶抑制剂,极光激酶抑制剂,细胞周期蛋白依赖性激酶抑制剂如CDK2和/或CDK4,CDK6抑制剂,核转运蛋白CRM1抑制剂,Wnt/beta-catenin抑制剂。
8)抗凋亡蛋白的抑制剂如BCL2抑制剂(Venetoclax)和MCL1抑制剂。
9)PARP抑制剂如Olaparib和Rucaparib等。
10抗血管形成抑制剂如VEGFR抑制剂。
11)表观遗传抑制剂如组蛋白去乙酰化酶(HDAC)抑制剂和DNA甲基转移酶(DNMT)抑制剂。
12)肿瘤免疫治疗法包括任何提高患者肿瘤细胞的免疫原性的体外和体内方法。如细胞因子IL-2、IL-4或者GM-CSF进行转染;降低T细胞无效能的方法如抗PD-1/PD-L单抗;使用转染的免疫细胞如细胞因子转染的树突状细胞的方法;使用细胞因子转染的肿瘤细胞系的方法;降低免疫抑制性细胞如调节性T细胞、髓源性抑制细胞、或表达吲哚胺2,3-脱氧酶的树突状细胞的功能方法;以及肿瘤相关抗原蛋白类或肽类组成的癌症疫苗的方法。
13)嵌合抗原受体T细胞免疫疗(CAR T)。
14)肿瘤基因治疗如CRISPR-Cas 9,RNAi和基因转导。
实施例
以下结合实施例进一步描述本发明,但这些实施例并非限制本发明的范围。
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker AVANCE-400)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDCl3)、氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。
MS的测定用液相色谱质谱联用仪(Thermo,Ultimate3000/MSQ)。
HPLC的测定使用高压液相色谱仪(安捷伦1260Infinity,Gemini C18 250×4.6mm,5u色谱柱)。
薄层色谱法(TLC)使用的硅胶板HSGF245采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.9mm~1.0mm(烟台黄海)。
柱层析色谱法一般使用200~300目硅胶为载体(烟台黄海硅胶)。
本发明的已知起始原料可以采用或按照本领域已知的方法来合成,或购买自上海达瑞精细化学品有限公司、上海泰坦科技股份有限公司、上海润捷化学试剂有限公司、TCI、Aldrich Chemical Company。实施例中未注明具体条件的实验方法,通常按照常规条件,或按照原料或商品制造厂商所建议的条件。未注明具体来源的试剂,为市场购买的常规试剂。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例1
1-(4-苯并咪唑-1-基-苯基)-3-异噁唑-3-基-脲(化合物1)的制备
Figure PCTCN2017076265-appb-000022
步骤1:1-(4-硝基-苯基)-1H-苯并咪唑的制备
于室温,将苯并咪唑(23.6g,0.2mol)和对氟硝基苯(31.0g,0.22mol)溶解于300mlDMF中,加入无水碳酸钾(69.0g,0.5mol)。将混合物加热至90℃,反应6小时。将反应液冷却至室温后,缓慢倒入水中,室温搅拌0.5小时,过滤。所得固体水洗,真空干燥过夜。将得到的固体用甲基叔丁基醚打浆,得46g黄色固体状的1-(4-硝基-苯基)-1H-苯并咪唑。
步骤2:4-苯并咪唑-1-基-苯胺的制备
将步骤1中得到的1-(4-硝基-苯基)-1H-苯并咪唑(46g,0.19mol)、还原铁粉(53.9g,0.96mol)、氯化铵(81.3g,1.52mol)加入乙醇(500ml)/水(125ml)中,并将得到的混合物加热至80℃,反应4h。将反应液冷却至室温后,缓慢倒入饱和碳酸氢钠水溶液中(1L),用乙酸乙酯萃取(500ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品37g黄色固体状的4-苯并咪唑-1-基-苯胺。产品无需纯化,直接用于下一步反应。
步骤3:异噁唑-3-基-氨基甲酸苯基酯(活泼酯)的制备
将3-氨基异噁唑(TCI)(840mg,10mmol)溶解于20mlTHF中,于室温加入吡啶(2.37g,30mmol)。将混合物在冰浴中降温至0~5℃,缓慢滴加氯甲酸苯酯(2.34g,15mmol),滴毕撤走冰浴,缓慢升至室温。TLC检测反应完毕后, 将反应液倒入水中,用乙酸乙酯(100×2)萃取,有机相用饱和NaCl溶液洗涤(200mL),无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得到1.5g白色固体状的异噁唑-3-基-氨基甲酸苯基酯。
步骤4:1-(4-苯并咪唑-1-基-苯基)-3-异噁唑-3-基-脲的制备
将步骤2得到的4-苯并咪唑-1-基-苯胺(100mg,0.478mmol)、步骤3得到的异噁唑-3-基-氨基甲酸苯基酯(146.4mg,0.717mmol)和三乙胺(145mg,1.43mmol)溶解于10mlTHF中,并回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到50mg白色固体状的1-(4-苯并咪唑-1-基-苯基)-3-异噁唑-3-基-脲。
1HNMR(DMSO-d6,400MHz)δ:9.81(s,1H),9.36(s,1H),8.76-8.77(d,1H),8.52(s,1H),7.77-7.79(m,1H),7.70-7.72(d,2H),7.60-7.63(d,2H),7.57-7.60(m,1H),7.29-7.36(m,2H),6.88-6.89(d,1H)。
LC-MS(ESI):320.2(M+H)+
实施例2
1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-异噁唑-3-基)-脲(化合物2)的制备
Figure PCTCN2017076265-appb-000023
与实施例1中的制备方法相同,除了用5-甲基-3-氨基异噁唑(TCI)代替步骤3中的3-氨基异噁唑,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.55(s,1H),9.09(s,1H),8.51(s,1H),7.76-7.79(m,1H),7.68-7.71(d,2H),7.60-7.62(d,2H),7.57-7.60(m,1H),7.30-7.34(m,2H),6.57(d,1H),2.38(s,3H)。
LC-MS(ESI):334.2(M+H)+
实施例3
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物3)的制备
Figure PCTCN2017076265-appb-000024
与实施例1中的制备方法相同,除了用5-叔丁基-3-氨基异噁唑(TCI)代替步骤3中的3-氨基异噁唑,得到白色固体状的1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.73(s,1H),9.34(s,1H),8.51(s,1H),7.76-7.78(m,1H),7.69-7.71(d,2H),7.57-7.62(m,3H),7.30-7.35(m,2H),6.54(d,1H),1.31(s,9H)。
LC-MS(ESI):376.1(M+H)+
实施例4
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-羟基-苯并咪唑-1-基)-苯基]-脲(化合物4)的制备
Figure PCTCN2017076265-appb-000025
步骤1:(4-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺的制备
氮气氛下,将2-硝基-4-甲氧基苯胺(1.5g,8.92mmol,达瑞)、对溴硝基苯(3.6g,17mmol)、Xphos(425mg)、Pd2(dba)3(408mg)、叔丁醇钠(1.71g,17.8mmol)溶解于30ml甲苯中,并于90℃反应3小时。将反应液冷却至室温,加入100ml二氯甲烷,室温搅拌5分钟,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得2.5g红黑色固体状的(4-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺。
步骤2:(4-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺的制备
将步骤1中所得产物(4-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺(2.5g,8.65mmol)、还原铁粉(2.91g,52.0mmol)、氯化铵(4.62g,86.5mmol)加入乙醇(50ml)/水(12.5ml)中,并将得到的混合物加热至90℃,反应1h。将反应液冷却至室温后,缓慢倒入饱和碳酸氢钠水溶液中(150ml),用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品1.8g黄色固体状的(4-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺。产品无需纯化,直接用于下一步反应。
步骤3:4-(5-甲氧基-苯并咪唑-1-基)-苯胺的制备
将步骤2中所得产物(4-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺(1.8g,7.86mmol)溶解于60ml盐酸中(4mol/L),于室温加入2ml甲酸,将混合物加热至120℃,反应1h。将反应液冷却,并在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到1.2g黄色固体状的4-(5-甲氧基-苯并咪唑-1-基)-苯胺。
步骤4:1-(4-氨基-苯基)-1H-苯并咪唑-5-醇的制备
于15ml40%氢溴酸中,将步骤3中所得产物4-(5-甲氧基-苯并咪唑-1-基)-苯胺(875mg,3.66mmol)加热至120℃,反应6h。将反应液冷却至室温,有灰白固体析出。将固体过滤,并溶解于70ml水中。过滤,取滤液用氨水调节至约PH=7,有白色固体析出,过滤,水洗固体,真空干燥得600mg灰白色固体状1-(4-氨基-苯基)-1H-苯并咪唑-5-醇。
1HNMR(DMSO-d6,400MHz)δ:9.13(s,1H),8.19(s,1H),7.23-7.25(d,1H),7.19-7.22(d,2H),7.02(d,1H),6.76-6.79(dd,1H),6.70-6.73(d,2H),5.41(s,2H)。
LC-MS(ESI):226.1(M+H)+
步骤5:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-羟基-苯并咪唑-1-基)-苯基]-脲的制备
将步骤4得到的1-(4-氨基-苯基)-1H-苯并咪唑-5-醇(100mg,0.444mmol)、(5-叔丁基-异噁唑-3-基)-氨基甲酸苯基酯(173.3mg,0.666mmol)和三乙胺(137.5mg,1.332mmol)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到80mg白色固体状的1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-羟基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.27(s,1H),9.11(s,1H),8.38(s,1H),7.66-7.68(d,2H),7.56-7.58(d,2H),7.37-7.39(d,1H),7.06-7.07(d,1H),6.81-6.84(dd,1H),6.52(s,1H),1.30(s,9H)。
LC-MS(ESI):392.1(M+H)+
实施例5
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物5)的制备
Figure PCTCN2017076265-appb-000026
与实施例4中的制备方法相同,除了用4-(5-甲氧基-苯并咪唑-1-基)-苯胺代替步骤5中的1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.59(s,1H),9.06(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.58-7.60(d,2H),7.46-7.48(d,1H),7.29-7.30(d,1H),6.94-6.97(dd,1H),6.53(s,1H),3.82(s,3H),1.30(s,9H)。
LC-MS(ESI):406.1(M+H)+
实施例6
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲(化合物6)的制备
Figure PCTCN2017076265-appb-000027
步骤1:4-(5-乙氧基-苯并咪唑-1-基)-苯胺的制备
将1-(4-氨基-苯基)-1H-苯并咪唑-5-醇(实施例4步骤4中合成)(300mg,1.33mmol)、碘乙烷(311mg,2.0mmo)、氢氧化锂(96mg,3.98mmol)于20ml乙醇中60℃反应过夜。次日,将反应液冷却至室温,并倒入水中(80ml),用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得400mg黑黄固体状的4-(5-乙氧基-苯并咪唑-1-基)-苯胺。
步骤2:与实施例4的步骤5相同,除了用4-(5-乙氧基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.08(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.57-7.60(d,2H),7.45-7.47(d,1H),7.27-7.28(d,1H),6.93-6.96(dd,1H),6.53(s,1H),4.05-4.11(q,2H),1.34-1.38(t,3H),1.31(s,9H)。
LC-MS(ESI):420.2(M+H)+
实施例7
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲(化合物7)的制备
Figure PCTCN2017076265-appb-000028
步骤1:4-(5-己氧基-苯并咪唑-1-基)-苯胺的制备
将1-(4-氨基-苯基)-1H-苯并咪唑-5-醇(实施例4步骤4中合成)(300mg,1.33mmol)、1-氯己烷(240mg,2.0mmol,TCI)、氢氧化钠(160mg,4mmol)加入20mlDMF中,将混合物加热至90℃,反应2h。将反应液冷却至室温,并倒入水中(80ml),用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品400mg4-(5-己氧基-苯并咪唑-1-基)-苯胺。产品无需纯化,直接用于下一步反应。
步骤2:与实施例4的步骤5相同,除了用4-(5-己氧基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.58-7.60(d,2H),7.45-7.47(d,1H),7.28-7.29(d,1H),6.94-6.96(dd,1H), 6.53(s,1H),4.00-4.03(t,2H),1.72-1.76(m,2H),1.41-1.46(m,2H),1.32-1.36(m,4H),1.31(s,9H),0.87-0.91(t,3H)。
LC-MS(ESI):ESI 476.1(M+H)+
实施例8
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲(化合物8)的制备
Figure PCTCN2017076265-appb-000029
与实施例6中的制备方法相同,除了用碘代异丙烷(TCI)代替步骤1中的碘乙烷,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.46(s,1H),7.68-7.70(d,2H),7.58-7.61(d,2H),7.45-7.47(d,1H),7.31-7.32(d,1H),6.93-6.95(dd,1H),6.53(s,1H),4.61-4.67(m,1H),1.31(s,9H),1.29-1.30(d,6H)。
LC-MS(ESI):434.1(M+H)+
实施例9
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲(化合物9)的制备
Figure PCTCN2017076265-appb-000030
与实施例7中的制备方法相同,除了用3-氯甲基-3-甲基氧杂环丁烷(达瑞)代替步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.59(s,1H),9.07(s,1H),8.45(s,1H),7.68-7.70(d,2H),7.58-7.60(d,2H),7.47-7.49(d,1H),7.35-7.36(d,1H),6.99-7.01(dd,1H),6.53(s,1H),4.52-4.54(d,2H),4.32-4.34(d,2H),4.11(s,2H),1.40(s,3H),1.31(s,9H)。
LC-MS(ESI):ESI 476.1(M+H)+
实施例10
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(四氢呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲(化合物10)的制备
Figure PCTCN2017076265-appb-000031
与实施例7中的制备方法相同,除了用2-氯甲基四氢呋喃(达瑞)代替步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(四氢呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.59(s,1H),9.06(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.58-7.60(d,2H),7.45-7.48(d,1H),7.29-7.30(d,1H),6.94-6.97(dd,1H),6.53(s,1H),4.16-4.22(m,1H),3.95-4.04(m,2H),3.78-3.83(m,1H),3.67-3.72(m,1H),1.98-2.05(m,1H),1.81-1.93(m,2H),1.68-1.73(m,1H),1.31(s,9H)。
LC-MS(ESI):ESI 476.1(M+H)+
实施例11
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物11)的制备
Figure PCTCN2017076265-appb-000032
与实施例7中的制备方法相同,除了用溴乙醇(TCI)代替步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.58-7.60(d,2H),7.46-7.48(d,1H),7.29-7.30(d,1H),6.95-6.98(dd,1H),6.53(s,1H),4.87-4.90(t,1H),4.02-4.06(m,2H),3.73-3.77(m,2H),1.31(s,9H)。
LC-MS(ESI):ESI 436.1(M+H)+
实施例12
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物12)的制备
Figure PCTCN2017076265-appb-000033
步骤1:4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
Figure PCTCN2017076265-appb-000034
于室温,将1-(4-氨基-苯基)-1H-苯并咪唑-5-醇(实施例4的步骤4制备)(18.0g,0.08mol)溶解于DMF(200ml)中,加入氢氧化钠(9.6g,0.24mol),室温搅拌30分钟,过程中有固体析出。于室温,加入氯乙基甲基醚(11.34g,0.12mol,TCI),并将混合物加热至90℃反应1.5小时。将反应液冷却至室温,并缓慢倒入600ml水中,有固体析出,室温搅拌30分钟。过滤,所得固体水洗,真空干燥得20.5g粉红固体状的4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺。
1HNMR(DMSO-d6,400MHz)δ:8.27(s,1H),7.32-7.35(d,1H),7.26(d,1H),7.22-7.24(d,2H),6.90-6.93(dd,1H),6.72-6.74(d,2H),5.41(s,2H),4.12-4.15(m,2H),3.67-3.70(m,2H),3.33(s,3H)。
LC-MS(ESI):284.1(M+H)+。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲的制备
与实施例4的步骤5相同,除了用4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.08(s,1H),8.52(s,1H),7.68-7.70(d,2H),7.59-7.62(d,2H),7.47-7.50(d,1H),7.38(d,1H),6.98-7.01(dd,1H),6.53(s,1H),4.13-4.15(t,2H),3.68-3.71(t,2H),3.33(s,3H),1.30(s,9H)。
LC-MS(ESI):450.1(M+H)+
实施例13
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物13)的制备
Figure PCTCN2017076265-appb-000035
与实施例7的制备方法相同,除了用2-氯乙基乙基醚(达瑞)代替步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.46(s,1H),7.67-7.70(d,2H),7.58-7.60(d,2H),7.46-7.48(d,1H),7.31(d,1H),6.96-6.99(dd,1H),6.53(s,1H),4.13-4.16(t,2H),3.72-3.74(t,2H),3.50-3.55(q,2H),1.31(s,9H),1.13-1.17(t,3H)。
LC-MS(ESI):ESI 464.1(M+H)+
实施例14
1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[2-(2-羟基-乙氧基)-乙氧基]-苯并咪唑-1-基}-苯基)-脲(化合物14)的制备
Figure PCTCN2017076265-appb-000036
与实施例7的制备方法相同,除了用2-氯乙氧基乙醇(泰坦)代替步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[2-(2-羟基-乙氧基)-乙氧基]-苯并咪唑-1-基}-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.47(s,1H),7.68-7.70(d,2H),7.59-7.61(d,2H),7.47-7.49(d,1H),7.32-7.33(d,1H),6.96-6.99(dd,1H),6.53(s,1H),4.66(br,1H),4.15-4.16(t,2H),3.77-3.79(t,2H),3.49-3.52(m,4H),1.30(s,9H)。
LC-MS(ESI):ESI 480.1(M+H)+
实施例15
吗啉-4-羧酸-1-{4-[3-(5-叔丁基-异噁唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基酯(化合物15)的制备
Figure PCTCN2017076265-appb-000037
将化合物7(41mg,0.105mmol)、三乙胺(32mg,0.315mmol)和DMAP(3mg)溶解于10mlTHF中,于室温加入4-吗啉碳酰氯(19mg,0.126mmol,达瑞),室温反应过夜。次日,将反应液倒入水中(30ml),用乙酸乙酯萃取(30ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得20mg白色固体状的吗啉-4-羧酸-1-{4-[3-(5-叔丁基-异噁唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基酯。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.13(s,1H),8.55(s,1H),7.69-7.71(d,2H),7.60-7.63(d,2H),7.54-7.56(d,1H),7.52-7.53(d,1H),7.10-7.13(dd,1H),6.53(s,1H),3.58-3.62(m,8H),1.30(s,9H)。
LC-MS(ESI):505.2(M+H)+
实施例16
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物16)的制备
Figure PCTCN2017076265-appb-000038
步骤1:4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
Figure PCTCN2017076265-appb-000039
将1-(4-氨基苯基)-1H-苯并咪唑-5-醇(实施例4的步骤4制备)(3.0g,0.013mol)溶解于DMF(200ml)中,于室温加入氢氧化钠(1.60g,0.04mol)、N-(2-氯乙基)吗啉盐酸盐(3.76g,0.020mol,TCI),室温搅拌30分钟。将混合物加热至90℃,反应过夜。将反应液冷却至室温,并缓慢倒入100ml水中,有固体析出。室温搅拌30分钟,过滤,所得固体水洗,真空干燥得3.4g黄色固体状的4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺。
1HNMR(DMSO-d6,400MHz)δ:8.27(s,1H),7.32-7.34(d,1H),7.27(d,1H),7.22-7.24(d,2H),6.89-6.92(dd,1H),6.72-6.74(d,2H),5.41(s,2H),4.11-4.14(t,2H),3.58-3.60(t,4H),2.70-2.73(t,2H),2.48-2.50(m,4H)。
LC-MS(ESI):339.1(M+H)+。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲的制备
与实施例4的步骤5相同,除了用4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(CDCl3-d,400MHz)δ:9.61(s,1H),8.20(s,1H),8.06(s,1H,),7.75-7.77(d,2H),7.46-7.48(d,2H),7.38-7.41(d,1H),7.35-7.36(d,1H),6.99-7.02(dd,1H),5.94(s,1H),4.20-4.23(t,2H),3.76-3.79(t,4H),2.86-2.89(t,2H),2.62-2.64(t,4H),1.39(s,9H)。
LC-MS(ESI):505.3(M+H)+
实施例17
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物17)的制备
Figure PCTCN2017076265-appb-000040
与实施例7的制备方法相同,除了用1-(2-氯-乙基)-哌啶盐酸盐(润捷)替代步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.59(s,1H),9.07(s,1H),8.43(s,1H),7.67-7.69(d,2H),7.57-7.59(d,2H),7.45-7.47(d,1H),7.29-7.30(d,1H),6.94-6.96(dd,1H),6.52(s,1H),4.11-4.14(t,2H),2.67-2.70(t,2H),2.45(m,4H),1.48-1.54(m,4H),1.38-1.41(m,2H),1.30(s,9H)。
LC-MS(ESI):503.3(M+H)+
实施例18
1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-异噁唑-3-基)-脲(化合物18)的制备
Figure PCTCN2017076265-appb-000041
与实施例7的制备方法相同,除了用2-(环己亚氨基)乙基氯盐酸盐(润捷)替代步骤1中的1-氯己烷,得到1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.62(s,1H),9.13(s,1H),8.43(s,1H),7.67-7.69(d,2H),7.57-7.59(d,2H),7.45-7.48(d,1H),7.30(d,1H),6.94-6.97(dd,1H),6.52(s,1H),4.11-4.14(t,2H),2.95(m,2H),2.77(m,4H),1.62(m,4H),1.55(m,4H),1.30(s,9H)。
LC-MS(ESI):517.3(M+H)+
实施例19
1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲(化合物19)的制备
Figure PCTCN2017076265-appb-000042
与实施例7的制备方法相同,除了用1-(3-氯丙基)-4-甲基哌嗪二盐酸盐(TCI)替代步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.08(s,1H),8.43(s,1H),7.67-7.69(d,2H),7.57-7.59(d,2H),7.45-7.47(d,1H),7.27-7.28(d,1H),6.93-6.96(dd,1H),6.53(s,1H),4.04-4.07(t,2H),2.43-2.46(t,2H),2.35(m,8H),2.16(s,3H),1.87-1.90(m,2H),1.30(s,9H)。
LC-MS(ESI):532.3(M+H)+
实施例20
1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-二甲基氨基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物20)的制备
Figure PCTCN2017076265-appb-000043
与实施例7的制备方法相同,除了用二甲氨基氯乙烷盐酸盐(润捷)代替步骤1中的1-氯己烷,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-二甲基氨基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(CDCl3-d,400MHz)δ:9.56(s,1H),8.89(s,1H),8.04(s,1H),7.71-7.73(d,2H),7.41-7.43(d,2H),7.34-7.36(m,2H),6.99-7.02(dd,1H),6.11(s,1H),4.20-4.22(t,2H),2.89-2.92(t,2H),2.47(s,6H),1.37(s,9H)。
LC-MS(ESI):463.2(M+H)+
实施例21
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物21)的制备
Figure PCTCN2017076265-appb-000044
Figure PCTCN2017076265-appb-000045
步骤1:4-(5-三氟甲氧基-苯并咪唑-1-基)-苯胺的制备
与实施例4的步骤1~3相同,除了用2-硝基-4-三氟甲氧基苯胺代替步骤1中的2-硝基-4-甲氧基苯胺,得到4-(5-三氟甲氧基-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5-三氟甲氧基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.62(s,1H),9.10(s,1H),8.66(s,1H,),7.79-7.80(d,1H),7.70-7.72(d,2H),7.61-7.64(m,3H),7.32-7.35(dd,1H),6.53(s,1H),1.31(s,9H)。
LC-MS(ESI):ESI 460.0(M+H)+
实施例22
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲(化合物22)的制备
Figure PCTCN2017076265-appb-000046
Figure PCTCN2017076265-appb-000047
步骤1:4-(5-氟-苯并咪唑-1-基)-苯胺的制备
与实施例4的步骤1~3相同,除了用2-硝基-4-氟苯胺代替步骤1中的2-硝基-4-甲氧基苯胺,得到4-(5-氟-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5-氟-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.10(s,1H),8.57(s,1H),7.68-7.71(d,2H),7.56-7.62(m,4H),7.20-7.23(m,1H),6.52(s,1H),1.28(s,9H)。
LC-MS(ESI):394.1(M+H)+
实施例23
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲(化合物23)的制备
Figure PCTCN2017076265-appb-000048
Figure PCTCN2017076265-appb-000049
步骤1:4-(5-三氟甲基-苯并咪唑-1-基)-苯胺的制备
与实施例4的步骤1~3相同,除了用2-硝基-4-三氟甲基苯胺代替步骤1中的2-硝基-4-甲氧基苯胺,得到4-(5-三氟甲基-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5-三氟甲基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.62(s,1H),9.12(s,1H),8.73(s,1H),8.16(d,1H),7.75-7.77(d,1H),7.71-7.73(d,2H),7.63-7.66(m,3H),6.53(s,1H),1.30(s,9H)。
LC-MS(ESI):444.1(M+H)+
实施例24
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基-苯并咪唑-1-基)-苯基]-脲(化合物24)的制备
Figure PCTCN2017076265-appb-000050
Figure PCTCN2017076265-appb-000051
步骤1:4-(5-甲基-苯并咪唑-1-基)-苯胺的制备
与实施例4的步骤1~3相同,除了用2-硝基-4-甲基苯胺代替步骤1中的2-硝基-4-甲氧基苯胺,得到4-(5-甲基-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5-甲基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.45(s,1H),7.68-7.70(d,2H),7.58-7.60(d,2H),7.56(m,1H),7.46-7.48(d,1H),7.14-7.17(dd,1H),6.53(s,1H),2.45(s,3H),1.31(s,9H)。
LC-MS(ESI):390.1(M+H)+
实施例25
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物25)的制备
Figure PCTCN2017076265-appb-000052
Figure PCTCN2017076265-appb-000053
步骤1:(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺的制备
在氮气氛下,将2-硝基-5-甲氧基苯胺(1.5g,8.92mmol)、对溴硝基苯(3.6g,17mmol)、Xphos(425mg)、Pd2(dba)3(408mg)、叔丁醇钠(1.71g,17.8mmol)溶解于30ml甲苯中,将混合物加热至90℃,反应3小时。将反应液冷却到室温后,加入100ml二氯甲烷,室温搅拌5分钟,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得1.7g红黑色固体状的(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺。
步骤2:(5-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺的制备
将步骤1中得到的(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺(1.7g,5.88mmol)、还原铁粉(2.63g,47.0mmol)、氯化铵(3.15g,58.8mmol)于乙醇(50ml)/水(12.5ml)中加热至90℃,反应1h。将反应液冷却至室温,并缓 慢倒入饱和碳酸氢钠水溶液中(150ml),用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品1.3g。无需纯化,直接用于下一步反应。
步骤3:4-(6-甲氧基-苯并咪唑-1-基)-苯胺的制备
将步骤2中得到的(5-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺(1.3g,5.67mmol)溶于50ml盐酸中(4mol/L),于室温加入1.5ml甲酸,将混合物加热至120℃,反应1h。然后,将反应液在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到1.0g黑黄色固体状的4-(6-甲氧基-苯并咪唑-1-基)-苯胺。
步骤4:1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(6-甲氧基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.07(s,1H),8.35(s,1H),7.69-7.71(d,2H),7.63-7.66(d,1H),7.59-7.62(d,2H),7.01-7.02(d,1H),6.90-6.93(dd,1H),6.53(s,1H),3.79(s,3H),1.31(s,9H)。
LC-MS(ESI):406.1(M+H)+
实施例26
1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物26)的制备
Figure PCTCN2017076265-appb-000054
Figure PCTCN2017076265-appb-000055
步骤1:(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺的制备
在氮气氛下,将2-硝基-5-甲氧基苯胺(1.5g,8.92mmol)、对溴硝基苯(3.6g,17mmol)、Xphos(425mg)、Pd2(dba)3(408mg)、叔丁醇钠(1.71g,17.8mmol)溶解于30ml甲苯中,将混合物加热至90℃,反应3小时。将反应液冷却到室温后,加入100ml二氯甲烷,室温搅拌5分钟,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得1.7g红黑色固体状的(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺。
步骤2:(5-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺的制备
将步骤1中所得产物(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺(1.7g,5.88mmol)、还原铁粉(2.63g,47.0mmol)、氯化铵(3.15g,58.8mmol)于乙醇(50ml)/水(12.5ml)中加热至90℃,反应1h。将反应液冷却至室温,并缓慢倒入饱和碳酸氢钠水溶液中(150ml),用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品1.3g。无需纯化,直接用于下一步反应。
步骤3:4-(6-甲氧基-苯并咪唑-1-基)-苯胺的制备
将步骤2中所得产物(5-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺(1.3g,5.67mmol)溶于50ml盐酸中(4mol/L),于室温加入1.5ml甲酸,将混合物加热至120℃,反应1h。然后,将反应液在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压 浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到1.0g黑黄色固体状的4-(6-甲氧基-苯并咪唑-1-基)-苯胺。
步骤4:3-(4-氨基-苯基)-3H-苯并咪唑-5-醇的制备
于15ml40%氢溴酸中,将步骤3中所得产物4-(6-甲氧基-苯并咪唑-1-基)-苯胺(600mg,3.66mmol)加热至120℃,反应6h。然后,将反应液冷却至室温,有灰白固体析出,过滤,将所得固体溶解于70ml水中。过滤,取滤液氨水调节至约PH=7,过程中有白色固体析出。过滤,固体水洗,真空干燥,得485mg灰白色固体状的3-(4-氨基-苯基)-3H-苯并咪唑-5-醇。
步骤5:4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
将步骤4中所得产物3-(4-氨基-苯基)-1H-苯并咪唑-5-醇(300mg,1.33mmol)、2-氯乙基甲基醚(372.0mg,2.0mmol)、氢氧化钠(160mg,4mmol)于20mlDMF中加热至90℃,反应2h。将反应液冷却至室温后,倒入水中(80ml),用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品360mg。无需纯化,直接用于下一步反应。
步骤6:1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲的制备
将步骤5得到的4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺(100mg,0.295mmol)、(5-叔丁基-异噁唑-3-基)-氨基甲酸苯基酯(115.3mg,0.443mmol)和三乙胺(90mg,0.888mmol)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得60mg白色固体状的1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.36(s,1H),7.68-7.70(d,2H,),7.63-7.65(d,1H),7.59-7.61(d,2H),7.03-7.04(d,1H),6.91-6.94(dd,1H),6.53(s,1H),4.11-4.13(m,2H),3.65-3.68(m,2H),3.31(s,3H),1.31(s,9H)。
LC-MS(ESI):ESI 450.2(M+H)+
实施例27
1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物27)的制备
Figure PCTCN2017076265-appb-000056
Figure PCTCN2017076265-appb-000057
步骤1:(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺的制备
在氮气氛下,将2-硝基-5-甲氧基苯胺(1.5g,8.92mmol,达瑞)、对溴硝基苯(3.6g,17mmol)、Xphos(425mg)、Pd2(dba)3(408mg)、叔丁醇钠(1.71g,17.8mmol)溶解于30ml甲苯中,将混合物加热至90℃,反应3小时。将反应液冷却到室温后,加入100ml二氯甲烷,室温搅拌5分钟,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得1.7g红黑色固体状的(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺。
步骤2:(5-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺的制备
将步骤1中所得产物(5-甲氧基-2-硝基-苯基)-(4-硝基-苯基)-胺(1.7g,5.88mmol)、还原铁粉(2.63g,47.0mmol)、氯化铵(3.15g,58.8mmol)于乙醇(50ml)/水(12.5ml)中加热至90℃,反应1h。将反应液冷却至室温,并缓慢倒入饱和碳酸氢钠水溶液中(150ml),用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品1.3g。无需纯化,直接用于下一步反应。
步骤3:4-(6-甲氧基-苯并咪唑-1-基)-苯胺的制备
将步骤2中所得产物(5-甲氧基-2-氨基-苯基)-(4-氨基-苯基)-胺(1.3g,5.67mmol)溶于50ml盐酸中(4mol/L),于室温加入1.5ml甲酸,将混合物加热至120℃,反应1h。然后,将反应液在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到1.0g黑黄色固体状的4-(6-甲氧基-苯并咪唑-1-基)-苯胺。
步骤4:3-(4-氨基-苯基)-3H-苯并咪唑-5-醇的制备
于15ml40%氢溴酸中,将步骤3中所得产物4-(6-甲氧基-苯并咪唑-1-基)-苯胺(600mg,3.66mmol)加热至120℃,反应6h。然后,将反应液冷却至室温,有灰白固体析出,过滤,将所得固体溶解于70ml水中。过滤,取滤液用氨水调节至约PH=7,过程中有白色固体析出。过滤,固体水洗,真空干燥,得485mg灰白色固体状的3-(4-氨基-苯基)-3H-苯并咪唑-5-醇。
步骤5:4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
将步骤4中所得产物3-(4-氨基-苯基)-1H-苯并咪唑-5-醇(300mg,1.33mmol)、4-(2-氯-乙基)-吗啉盐酸盐(372.0mg,2.0mmol)、氢氧化钠(160mg,4mmol)于20mlDMF中加热至90℃,反应2h。将反应液冷却至室温后,倒入水中(80ml),用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品360mg。无需纯化,直接用于下一步反应。
步骤6:1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲的制备
将步骤5所得产物4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺(100mg,0.295mmol)、(5-叔丁基-异噁唑-3-基)-氨基甲酸苯基酯(115.3mg,0.443mmol)和三乙胺(90mg,0.888mmol)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得60mg白色固体状的1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(CDCl3-d,400MHz)δ:9.64(s,1H),8.04(s,1H),8.00(s,1H,),7.74-7.79(m,3H),7.45-7.49(d,2H),6.97-7.01(m,2H),5.93(s,1H),4.12-4.15(t,2H),3.74-3.77(t,4H),2.83-2.86(t,2H),2.60-2.62(t,4H),1.39(s,9H)。
LC-MS(ESI):505.3(M+H)+
实施例28
1-(5-叔丁基-异噁唑-3-基)-3-(4-{6-[(2-二甲基氨基-乙基)-甲基-氨基]-苯并咪唑-1-基}-苯基)-脲(化合物28)的制备
Figure PCTCN2017076265-appb-000058
Figure PCTCN2017076265-appb-000059
步骤1:N1-(2-二甲基氨基-乙基)-N1-甲基-4-硝基苯-1,3-二胺的制备
将5-氟-2-硝基苯胺(2.5g,0.016mol,TCI)、N,N,N′-三甲基乙二胺(2.45g,0.024mmol)、无水碳酸钾(6.8g,0.048mol)于50mlDMF中加热至90℃,反应3小时。将反应液冷却至室温,并倒入水中,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得黄色固体。将所得固体用甲基叔丁基醚打浆,得2.66g黄色固体。
步骤2:N1-(2-二甲基氨基-乙基)-N1-甲基-4-硝基-N3-(4-硝基-苯基)-苯-1,3-二胺的制备
氮气氛下,将步骤1中所得产物(2.66g,0.011mmol)、对溴硝基苯(4.70g,0.023mmol)、Xphos(530mg)、Pd2(dba)3(510mg)、叔丁醇钠(2.2g,0.023mmol)溶解于80ml甲苯中,于90℃搅拌反应过夜。将反应液冷却至室温,并加入100ml二氯甲烷,于室温搅拌5分钟,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得1.3g红黑色固体。
步骤3:N2-(4-氨基-苯基)-N4-(2-二甲基氨基-乙基)-N4-甲基-苯基-1,2,4-三胺的制备
将步骤2中所得产物(1.3g,3.62mmol)、还原铁粉(1.01g,18.1mmol)、氯化铵(1.55g,29.0mmol)于乙醇(40ml)/水(10ml)中加热至90℃,反应1h。将反应液冷却至室温,并缓慢倒入饱和碳酸氢钠水溶液中(150ml),用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩得粗品1.1g。无需纯化,直接用于下一步反应。
步骤4:N-[3-(4-氨基-苯基)-3H-苯并咪唑-5-基]-N,N,N′-三甲基乙烷-1,2-二胺的制备
将步骤3中所得产物(1.1g,4.37mmol)溶解于40ml盐酸中(4mol/L),于室温加入1.1ml甲酸,将混合物加热至120℃,反应1h。将反应液在冰浴下用氨 水调节至PH>9,用乙酸乙酯萃取(60ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得500mg产品。
步骤5:1-(5-叔丁基-异噁唑-3-基)-3-(4-{6-[(2-二甲基氨基-乙基)-甲基-氨基]-苯并咪唑-1-基}-苯基)-脲的制备。
将步骤4得到的N-[3-(4-氨基-苯基)-3H-苯并咪唑-5-基]-N,N,N′-三甲基乙烷-1,2-二胺(100mg,0.323mmol)、异噁唑-3-基-氨基甲酸苯基酯(168mg,0.647mmol)和三乙胺(100mg,0.99mmol)溶解于THF中,并回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到白色固体32mg。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.07(s,1H),8.18(s,1H),7.66-7.68(d,2H),7.56-7.58(d,2H),7.53-7.55(d,1H),6.80-6.82(dd,1H),6.67-6.68(d,1H),6.52(s,1H),3.42(m,2H),2.90(s,3H),2.37-2.40(t,2H),2.16(s,6H),1.30(s,9H)。
LC-MS(ESI):476.2(M+H)+
实施例29
1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯基}-脲(化合物29)的制备
Figure PCTCN2017076265-appb-000060
Figure PCTCN2017076265-appb-000061
步骤1:5-(4-甲基-哌嗪-1-基)-2-硝基-苯胺的制备
将5-氟-2-硝基苯胺(2.5g,0.016mol)、N-甲基哌嗪(2.4g,0.024mmol,达瑞)、无水碳酸钾(6.8g,0.048mol)于50mlDMF中加热至90℃,反应3小时。 将反应液冷却至室温,并倒入水中,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得黄色固体。将所得固体用甲基叔丁基醚打浆,得3g黄色固体状的5-(4-甲基-哌嗪-1-基)-2-硝基-苯胺。
步骤2:[5-(4-甲基-哌嗪-1-基)-2-硝基-苯基]-(4-硝基-苯基)-胺的制备
氮气氛下,将步骤1中得到的5-(4-甲基-哌嗪-1-基)-2-硝基-苯胺(2.5g,0.011mmol)、对溴硝基苯(4.28g,0.021mmol)、Xphos(480mg)、Pd2(dba)3(460mg)、叔丁醇钠(2g,0.021mmol)溶解于80ml甲苯中,于90℃搅拌反应过夜。将反应液冷却至室温,并加入100ml二氯甲烷,于室温搅拌5分钟,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得1.8g红黑色固体状的[5-(4-甲基-哌嗪-1-基)-2-硝基-苯基]-(4-硝基-苯基)-胺。
步骤3:N2-(4-氨基-苯基)-4-(4-甲基-哌嗪-1-基)-苯-1,2-二胺的制备
将步骤2中所得产物[5-(4-甲基-哌嗪-1-基)-2-硝基-苯基]-(4-硝基-苯基)-胺(1.8g,5.04mmol)、还原铁粉(2.26g,40.3mmol)、氯化铵(2.70g,50.4mmol)于乙醇(40ml)/水(10ml)中加热至90℃,反应1h。将反应液冷却至室温,并缓慢倒入饱和碳酸氢钠水溶液中(150ml),用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩得粗品1.3g。无需纯化,直接用于下一步反应。
步骤4:4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯胺的制备
将步骤3中所得产物N2-(4-氨基-苯基)-4-(4-甲基-哌嗪-1-基)-苯-1,2-二胺(1.3g,4.37mmol)溶解于50ml盐酸中(4mol/L),于室温加入1.5ml甲酸,将混合物加热至120℃,反应1h。将反应液在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(60ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到600mg4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯胺。
步骤5:1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯基}-脲的制备
将步骤4得到的4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯胺(100mg,0.326mmol)、异噁唑-3-基-氨基甲酸苯基酯(127mg,0.490mmol)和三乙胺(100mg,0.99mmol)溶解于THF中,并回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到白色固体状的1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.08(s,1H),8.27(s,1H),7.67-7.69(d,2H),7.56-7.60(m,3H),7.03-7.05(dd,1H),6.91-6.92(d,1H),6.52(s,1H),3.13(m,4H),2.51(m,4H),2.25(s,3H),1.30(s,9H)。
LC-MS(ESI):474.2(M+H)+
实施例30
1-(5-叔丁基-异噁唑-3-基)-3-{4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物30)的制备
Figure PCTCN2017076265-appb-000062
步骤1:3-(4-氨基-苯基)-3H-苯并咪唑-4-醇的制备
与实施例4中的步骤1-4相同,除了用2-氨基-3-硝基苯酚代替步骤1中的2-硝基-4-甲氧基苯胺,得到3-(4-氨基-苯基)-3H-苯并咪唑-4-醇。
步骤2:4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
与实施例7的步骤1相同,除了用3-(4-氨基-苯基)-3H-苯并咪唑-4-醇代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,并且用N-(2-氯乙基)吗啉盐酸盐代替1-氯己烷,得到4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺。
步骤3:1-(5-叔丁基-异噁唑-3-基)-3-{4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲的制备
与实施例4的步骤5相同,除了用4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,得到1-(5-叔丁基-异噁唑-3-基)-3-{4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.57(s,1H),9.04(s,1H),8.22(s,1H),7.58-7.60(d,2H),7.44-7.46(d,2H),7.32-7.34(d,1H),7.15-7.19(t,1H),6.84-6.86(d,1H),6.53(s,1H),4.06-4.09(t,2H),3.43-3.45(t,4H),2.45-2.47(t,2H),2.17(m,4H),1.31(s,9H)。
LC-MS(ESI):505.2(M+H)+
实施例31
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物31)的制备
Figure PCTCN2017076265-appb-000063
步骤1:4-(5,6-二甲氧基-苯并咪唑-1-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用5,6-二甲氧基苯并咪唑(达瑞)代替步骤1中的苯并咪唑,得到4-(5,6-二甲氧基-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5,6-二甲氧基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.06(s,1H),8.28(s,1H,),7.68-7.70(d,2H),7.59-7.61(d,2H),7.31(s,1H),7.04(s,1H),6.52(s,1H),3.82(s,3H),3.79(s,3H),1.31(s,9H)。
LC-MS(ESI):436.1(M+H)+
实施例32
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲基-苯并咪唑-1-基)-苯基]-脲(化合物32)的制备
Figure PCTCN2017076265-appb-000064
步骤1:4-(5,6-二甲基-苯并咪唑-1-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用5,6-二甲基苯并咪唑代替步骤1中的苯并咪唑,得到4-(5,6-二甲基-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5,6-二甲基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.08(s,1H),8.36(s,1H),7.67-7.70(d,2H),7.57-7.59(d,2H),753(s,1H),7.37(s,1H),6.53(s,1H),2.34(s,6H),1.31(s,9H)。
LC-MS(ESI):403.9(M+H)+
实施例33
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲(化合物33)的制备
Figure PCTCN2017076265-appb-000065
步骤1:(5-氟-7-甲基-苯并咪唑-1-基)-苯胺的制备
与实施例4的步骤1-步骤3中的制备方法相同,除了用2-硝基-4-氟-6-甲基苯胺(TCI)代替步骤1中的2-硝基-4-甲氧基苯胺,得到(5-氟-7-甲基-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用(5-氟-7-甲基-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.64(s,1H),9.12(s,1H),8.30(s,1H),7.62-7.65(d,2H),7.49-7.51(d,2H),7.37-7.40(dd,1H),6.94-6.97(dd,1H),6.53(s,1H),2.03(s,3H),1.30(s,9H)。
LC-MS(ESI):408.1(M+H)+
实施例34
1-(5-叔丁基-异噁唑-3-基)-3-[4-(4-氟-苯并咪唑-1-基)-苯基]-脲(化合物34)的制备
Figure PCTCN2017076265-appb-000066
Figure PCTCN2017076265-appb-000067
步骤1:4-(4-氟-苯并咪唑-1-基)-苯胺的制备
与实施例4的步骤1-步骤3中的制备方法相同,除了用2-硝基-3-氟苯胺代替步骤1中的2-硝基-4-甲氧基苯胺,得到4-(4-氟-苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(4-氟-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(4-氟-苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(4-氟-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.62(s,1H),9.08(s,1H),8.46(s,1H),7.55-7.65(m,5H),7.25-7.30(m,1H),7.12-7.17(m,1H),6.53(s,1H),1.30(s,9H)。
LC-MS(ESI):394.1(M+H)+
实施例35
1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-甲基-苯并咪唑-1-基)-苯基]-脲(化合物35)的制备
Figure PCTCN2017076265-appb-000068
步骤1:4-(2-甲基苯并咪唑-1-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用2-甲基苯并咪唑代替步骤1中的苯并咪唑,得到4-(2-甲基苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-甲基-苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(2-甲基苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-甲基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.63(s,1H),9.12(s,1H),7.69-7.71(d,2H),7.61-7.63(m,1H),7.46-7.48(d,2H),7.16-7.23(m,2H),7.10-7.12(m,1H),6.53(s,1H),2.43(s,3H),1.31(s,9H)。
LC-MS(ESI):389.9(M+H)+
实施例36
1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-氯苯并咪唑-1-基)-苯基]-脲(化合物36)的制备
Figure PCTCN2017076265-appb-000069
步骤1:4-(2-氯苯并咪唑-1-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用2-氯苯并咪唑代替步骤1中的苯并咪唑,得到4-(2-氯苯并咪唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-氯苯并咪唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(2-氯苯并咪唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-氯苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.64(s,1H),9.16(s,1H),7.70-7.73(m,3H),7.50-7.53(d,2H),7.29-7.34(m,2H),7.17-7.19(m,1H),6.54(s,1H),1.31(s,9H)。
LC-MS(ESI):410.1(M+H)+
实施例37
1-(4-苯并咪唑-1-基-3-甲基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物37)的制备
Figure PCTCN2017076265-appb-000070
步骤1:4-苯并咪唑-1-基-3-甲基-苯胺的制备。
与实施例1的步骤1-步骤2中的制备方法相同,除了用3-甲基-4-氟硝基苯(润捷)代替步骤1中对氟硝基苯,得到4-苯并咪唑-1-基-3-甲基-苯胺。
步骤2:1-(4-苯并咪唑-1-基-3-甲基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-苯并咪唑-1-基-3-甲基-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(4-苯并咪唑-1-基-3-甲基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.63(s,1H),9.05(s,1H),8.41(s,1H),7.80-7.83(m,1H),7.61-7.62(d,1H),7.48-7.51(dd,1H),7.35-7.38(d,1H),7.29-7.31(m,2H),7.15-7.17(m,1H),6.54(s,1H),2.00(s,3H),1.31(s,9H)。
LC-MS(ESI):390.2(M+H)+
实施例38
1-(4-苯并咪唑-1-基-3-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物38)的制备
Figure PCTCN2017076265-appb-000071
步骤1:4-苯并咪唑-1-基-3-氯-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用3-氯-4-氟硝基苯代替步骤1中的对氟硝基苯,得到4-苯并咪唑-1-基-3-氯-苯胺。
步骤2:1-(4-苯并咪唑-1-基-3-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-苯并咪唑-1-基-3-氯-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(4-苯并咪唑-1-基-3-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.77(s,1H),9.29(s,1H),8.40(s,1H),8.03-8.04(d,1H),7.76-7.79(m,1H),7.60-7.63(d,1H),7.51-7.54(dd,1H),7.29-7.31(m,2H),7.20-7.21(m,1H),6.55(s,1H),1.31(s,9H)。
LC-MS(ESI):410.1(M+H)+
实施例39
1-(4-苯并咪唑-1-基-3-氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物39)的制备
Figure PCTCN2017076265-appb-000072
步骤1:4-苯并咪唑-1-基-3-氟-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用3,4-二氟硝基苯代替步骤1中的对氟硝基苯,制得4-苯并咪唑-1-基-3-氟-苯胺。
步骤2:1-(4-苯并咪唑-1-基-3-氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-苯并咪唑-1-基-3-氟-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(4-苯并咪唑-1-基-3-氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.74(s,1H),9.30(s,1H),8.45(s,1H),7.80-7.84(dd,1H),7.77-7.79(m,1H),7.63-7.67(t,1H),7.30-7.39(m,4H),6.54(s,1H),1.31(s,9H)。
LC-MS(ESI):ESI 393.9(M+H)+
实施例40
1-(4-苯并咪唑-1-基-3,5-二氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物40)的制备
Figure PCTCN2017076265-appb-000073
步骤1:4-苯并咪唑-1-基-3,5-二氟-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用3,4,5-三氟硝基苯代替步骤1中的对氟硝基苯,得到4-苯并咪唑-1-基-3,5-二氟-苯胺。
步骤2:1-(4-苯并咪唑-1-基-3,5-二氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-苯并咪唑-1-基-3,5-二氟-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(4-苯并咪唑-1-基-3,5-二氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.91(s,1H),9.46(s,1H),8.48(s,1H),7.78-7.81(m,1H),7.55-7.58(d,2H),7.31-7.35(m,3H),6.54(s,1H),1.31(s,9H)。
LC-MS(ESI):412.1(M+H)+
实施例41
1-(4-苯并咪唑-1-基-2-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物41)的制备
Figure PCTCN2017076265-appb-000074
步骤1:4-苯并咪唑-1-基-2-氯-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用2-氯-4-氟硝基苯代替步骤1中的对氟硝基苯,制得4-苯并咪唑-1-基-2-氯-苯胺。
步骤2:1-(4-苯并咪唑-1-基-2-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-苯并咪唑-1-基-2-氯-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(4-苯并咪唑-1-基-2-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:10.32(s,1H),8.89(s,1H),8.56(s,1H),8.39-8.41(d,1H),7.90-7.91(d,1H),7.77-7.79(m,1H),7.67-7.69(dd,1H),7.61-7.63(m,1H),7.30-7.37(m,2H),6.49(s,1H),1.31(s,9H)。
LC-MS(ESI):410.1(M+H)+
实施例42
1-(6-苯并咪唑-1-基-吡啶-3-基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物42)的制备
Figure PCTCN2017076265-appb-000075
步骤1:6-苯并咪唑-1-基-吡啶-3-基胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用2-氯-5-硝基吡啶代替步骤1中的对氟硝基苯,制得6-苯并咪唑-1-基-吡啶-3-基胺。
步骤2:1-(6-苯并咪唑-1-基-吡啶-3-基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用6-苯并咪唑-1-基-吡啶-3-基胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(6-苯并咪唑-1-基-吡啶-3-基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.80(s,1H),9.20(s,1H),8.88(s,1H),8.68-8.69(d,1H),8.19-8.22(m,2H),7.89-7.91(d,1H),7.76-7.78(m,1H),7.33-7.38(m,2H),6.53(s,1H),1.31(s,9H)。
LC-MS(ESI):377.1(M+H)+
实施例43
1-(2-苯并咪唑-1-基-嘧啶-5-基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物43)的制备
Figure PCTCN2017076265-appb-000076
步骤1:1-(5-硝基-嘧啶-2-基)-1H-苯并咪唑的制备
于室温,将2-氯-5-硝基嘧啶(118mg,1mmol)和苯并咪唑(175.5mg,1.1mmol)溶解于15ml乙腈中,加入无水碳酸钾(414mg,3mmol),室温反应过夜。次日,将反应液倒入水中,用乙酸乙酯萃取(50×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩得黄黑色固体。无需纯化,直接用于下一步。
步骤2:2-苯并咪唑-1-基-嘧啶-5-基胺的制备
与实施例1的步骤2相同,除了1-(5-硝基-嘧啶-2-基)-1H-苯并咪唑代替步骤2中的1-(4-硝基-苯基)-1H-苯并咪唑,得到2-苯并咪唑-1-基-嘧啶-5-基胺。
步骤3:1-(2-苯并咪唑-1-基-嘧啶-5-基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用2-苯并咪唑-1-基-嘧啶-5-基胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(2-苯并咪唑-1-基-嘧啶-5-基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:10.01(s,1H),9.25(s,1H),9.06(s,1H),9.05(s,2H),8.51-8.53(d,1H),7.78-7.80(d,1H),7.42-7.46(m,1H),7.35-7.39(m,1H),6.52(s,1H),1.30(s,9H)。
LC-MS(ESI):378.2(M+H)+
实施例44
1-(5-叔丁基-异噁唑-3-基)-3-(4-吲哚-1-基-苯基)-脲(化合物44)的制备
Figure PCTCN2017076265-appb-000077
步骤1:4-吲哚-1-基-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用吲哚代替步骤1中的苯并咪唑,得到4-吲哚-1-基-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-(4-吲哚-1-基-苯基)-脲的制备
与实施例4的步骤5相同,除了用4-吲哚-1-基-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-(4-吲哚-1-基-苯基)-脲。
1HNMR(CDCl3-d,400MHz)δ:9.46(s,1H),8.66(s,1H),7.69-7.71(m,3H),7.53-7.55(d,1H),7.48-7.51(d,2H),7.32-7.33(d,1H),7.16-7.26(m,2H),6.69-6.70(d,1H),5.97(s,1H),1.39(s,9H)。
LC-MS(ESI):374.9(M+H)+
实施例45
1-(5-叔丁基-异噁唑-3-基)-3-(4-吲唑-1-基-苯基)-脲(化合物45)的制备
Figure PCTCN2017076265-appb-000078
步骤1:4-吲唑-1-基-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用吲唑代替步骤1中的苯并咪唑,制得4-吲唑-1-基-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-(4-吲唑-1-基-苯基)-脲的制备
与实施例4的步骤5相同,除了用4-吲唑-1-基-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-(4-吲唑-1-基-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.58(s,1H),9.03(s,1H),8.35(d,1H),7.88-7.90(d,1H),7.79-7.81(d,1H),766-7.71(m,4H),7.47-7.51(m,1H),7.25-7.28(m,1H),6.54(s,1H),1.31(s,9H)。
LC-MS(ESI):376.0(M+H)+
实施例46
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-氟-吲唑-1-基)-苯基]-脲(化合物46)的制备
Figure PCTCN2017076265-appb-000079
步骤1:4-(6-氟-吲唑-1-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用6-氟吲唑代替步骤1中的苯并咪唑,制得4-(6-氟-吲唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-氟-吲唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(6-氟-吲唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-氟-吲唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.58(s,1H),9.04(s,1H),8.37(s,1H),7.92-7.95(m,1H),7.65-7.70(m,4H),7.57-7.59(m,1H),7.13-7.18(m,1H),6.53(s,1H),1.30(s,9H)。
LC-MS(ESI):394.1(M+H)+
实施例47
1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-氟-吲唑-1-基)-苯基]-脲(化合物47)的制备
Figure PCTCN2017076265-appb-000080
Figure PCTCN2017076265-appb-000081
步骤1:4-(7-氟-吲唑-1-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用7-氟吲唑代替步骤1中的苯并咪唑,制得4-(7-氟-吲唑-1-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-氟-吲唑-1-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(7-氟-吲唑-1-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-氟-吲唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.63(s,1H),9.16-9.17(d,1H),9.08(s,1H),8.03-8.05(d,2H),7.67-7.69(d,2H),7.59-7.61(d,1H),7.05-7.13(m,2H),6.54(s,1H),1.31(s,9H)。
LC-MS(ESI):394.1(M+H)+
实施例48
1-(4-苯并三唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲(化合物48)的制备
Figure PCTCN2017076265-appb-000082
步骤1:4-苯并三唑-1-基-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用苯并三氮唑(TCI)代替步骤1中的苯并咪唑,制得4-苯并三唑-1-基-苯胺。
步骤2:1-(4-苯并三唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-苯并三唑-1-基-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(4-苯并三唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(CDCl3-d,400MHz)δ:9.65(s,1H),8.16-8.18(d,1H),8.12(s,1H),7.82-7.85(d,2H),7.73-7.79(m,3H),7.55-7.59(m,1H),7.44-7.48(m,1H),5.94(s,1H),1.40(s,9H)。
LC-MS(ESI):376.9(M+H)+
实施例49
1-(5-叔丁基-异噁唑-3-基)-3-(4-吡咯并[2,3-b]吡啶-1-基-苯基)-脲(化合物49)的制备
Figure PCTCN2017076265-appb-000083
步骤1:4-吡咯并[2,3-b]吡啶-1-基-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用1H-吡咯并[2,3-b]吡啶(润捷)代替步骤1中的苯并咪唑,得到4-吡咯并[2,3-b]吡啶-1-基-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-(4-吡咯并[2,3-b]吡啶-1-基-苯基)-脲的制备
与实施例4的步骤5相同,除了用4-吡咯并[2,3-b]吡啶-1-基-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-(4-吡咯并[2,3-b]吡啶-1-基-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.55(s,1H),8.98(s,1H),8.30-8.32(m,1H),8.07-8.09(m,1H),7.90-7.91(d,1H),7.80-7.82(d,2H),7.61-7.63(d,2H),7.18-7.22(m,1H),6.70-6.71(d,1H),6.53(s,1H),1.30(s,9H)。
LC-MS(ESI):376.2(M+H)+
实施例50
1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-3-基-苯基)-脲(化合物50)的制备
Figure PCTCN2017076265-appb-000084
步骤1:[4-(3-硝基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯的制备
2-氯-3-硝基吡啶(1g,6.3mmol,TCI)、(4-氨基-苯基)-氨基甲酸叔丁基酯(达瑞)(1.6g,7.56mmol)、三乙胺(3.2g,31.5mmol)于DMSO(30ml)中85℃反应过夜。次日,将反应液倒入水中(100mg),用乙酸乙酯萃取(60ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压 浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得1.25g[4-(3-硝基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯。
步骤2:[4-(3-氨基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯的制备
将[4-(3-硝基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯(1.25g,3.81mol)、还原铁粉(1.1g,19.05mmol)、氯化铵(1.63g,30.48mmol)于乙醇(40ml)/水(10ml)中回流反应2h。将反应液冷却至室温后,倒入饱和碳酸氢钠水溶液中,用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品1g。无需纯化,直接用于下一步反应。
步骤3:4-(咪唑并[4,5-b]吡啶-3-基)-苯胺的制备
于室温,将[4-(3-氨基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯(1g,3.33mmol)溶解于30ml盐酸中(4mol/L),加入1ml甲酸,将混合物加热至120℃反应1h。将反应液在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。得粗品340mg。无需纯化,直接用于下一步反应。
步骤4:1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-3-基-苯基)-脲的制备
与实施例4的步骤5相同,除了用4-(咪唑并[4,5-b]吡啶-3-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-3-基-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.58(s,1H),9.05(s,1H),8.85(s,1H),8.43-8.44(dd,1H),8.20-8.22(dd,1H),7.85-7.87(d,2H),7.67-7.69(d,2H),7.37-7.41(q,1H),6.53(s,1H),1.31(s,9H)。
LC-MS(ESI):377.2(M+H)+
实施例51
1-[4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯基]-3-(5-叔丁基-异噁唑-3-基)-脲(化合物51)的制备
Figure PCTCN2017076265-appb-000085
Figure PCTCN2017076265-appb-000086
步骤1:[4-(5-溴-3-硝基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯的制备
将2-氯-3-硝基-5-溴吡啶(1g,4.23mmol,TCI)、(4-氨基-苯基)-氨基甲酸叔丁基酯(1.06g,5.08mmol,达瑞)、三乙胺(2.14g,0.021mol)于DMSO(30ml)中80℃反应4h。将反应液冷却至室温,并倒入水中(100mg),用乙酸乙酯萃取(60ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚/乙酸乙酯)纯化,得700mg[4-(5-溴-3-硝基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯。
步骤2:[4-(3-氨基-5-溴-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯的制备
将步骤1得到的[4-(5-溴-3-硝基-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯(700mg,1.72mol)、还原铁粉(480mg,8.58mmol)、氯化铵(734mg,1.37mmol)于乙醇(30ml)/水(8ml)中回流反应1h。将反应液冷却至室温,并倒入饱和碳酸氢钠水溶液中,用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品550mg。无需纯化,直接用于下一步反应。
步骤3:4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯胺的制备
于室温,将步骤2得到的[4-(3-氨基-5-溴-吡啶-2-基氨基)-苯基]-氨基甲酸叔丁基酯(550mg,1.45mmol)溶解于20ml盐酸中(4mol/L),加入0.5ml甲酸,将混合物加热至120℃反应1h。将反应液在冰浴下用氨水调节至PH>9,用乙酸乙酯萃取(50ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得到230mg黄色固体状的4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯胺。
步骤4:1-[4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯基]-3-(5-叔丁基-异噁唑-3-基)-脲的制备
与实施例4的步骤5相同,除了用4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-[4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯基]-3-(5-叔丁基-异噁唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.59(s,1H),9.06(s,1H),8.90(s,1H),8.51(s,1H),8.52(s,1H),7.80-7.82(d,2H),7.67-7.69(d,2H),6.54(s,1H),1.30(s,9H)。
LC-MS(ESI):455.0\457.0(M+H)+
实施例52
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯基]-脲(化合物52)的制备
Figure PCTCN2017076265-appb-000087
步骤1:4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯胺的制备
与实施例51的步骤1-步骤3的制备方法相同,除了用5-三氟甲基-3-硝基吡啶(达瑞)代替步骤1中的2-氯-3-硝基-5-溴吡啶,得到4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.09(s,1H),9.05(s,1H),8.82(d,1H),8.67(d,1H),7.82-7.84(d,2H),7.69-7.71(d,2H),6.53(s,1H),1.30(s,9H)。
LC-MS(ESI):445.2(M+H)+
实施例53
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯基]-脲(化合物53)的制备
Figure PCTCN2017076265-appb-000088
步骤1:4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯胺的制备
与实施例51的步骤1-步骤3的制备方法相同,除了用2,6-二氯-3-硝基吡啶(TCI)代替步骤1中的2-氯-3-硝基-5-溴吡啶,得到4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.08(s,1H),8.85(s,1H),8.25-8.27(d,1H),7.75-7.77(d,2H),7.68-7.70(d,2H),7.44-7.46(d,1H),6.53(s,1H),1.30(s,9H)。
LC-MS(ESI):411.1(M+H)+
实施例54
1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯基]-脲(化合物54)的制备
Figure PCTCN2017076265-appb-000089
步骤1:4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯胺的制备
与实施例51的步骤1-步骤3的制备方法相同,除了用6-甲基-3-硝基吡啶(达瑞)代替步骤1中的2-氯-3-硝基-5-溴吡啶,得到4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.59(s,1H),9.04(s,1H),8.71(s,1H),8.05-8.07(d,1H),7.81-7.83(d,2H),7.65-7.68(d,2H),7.23-7.25(d,1H),6.53(s,1H),2.58(s,3H),1.30(s,9H)。
LC-MS(ESI):391.2(M+H)+
实施例55
1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-甲基咪唑[4,5-b]吡啶-3-基)-苯基]-脲(化合物55)的制备
Figure PCTCN2017076265-appb-000090
步骤1:4-(7-甲基咪唑并[4,5-b]吡啶-3-基)-苯胺的制备
与实施例51的步骤1-步骤3的制备方法相同,除了用2-氯-3-硝基-4甲基吡啶(润捷)代替步骤1中的2-氯-3-硝基-5-溴吡啶,得到4-(7-甲基咪唑并[4,5-b]吡啶-3-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-甲基咪唑[4,5-b]吡啶-3-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(7-甲基咪唑并[4,5-b]吡啶-3-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-甲基咪唑[4,5-b]吡啶-3-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.58(s,1H),9.05(s,1H),8.78(s,1H),8.28-8.29(d,1H),7.84-7.86(d,2H),7.66-7.68(d,2H),7.21-7.22(m,1H),6.53(s,1H),2.63(s,3H),1.31(s,9H)。
LC-MS(ESI):391.1(M+H)+
实施例56
1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-1-基-苯基)-脲(化合物56)的制备
Figure PCTCN2017076265-appb-000091
步骤1:4-咪唑并[4,5-b]吡啶-1-基-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用4-氮杂苯并咪唑(达瑞)代替步骤1中的苯并咪唑,得到4-咪唑并[4,5-b]吡啶-1-基-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-1-基-苯基)-脲的制备
与实施例4的步骤5相同,除了用4-咪唑并[4,5-b]吡啶-1-基-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-1-基-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.61(s,1H),9.09(s,1H),8.80(s,1H),8.51-8.52(dd,1H),8.03-8.05(dd,1H),7.70-7.72(d,2H),7.63-7.66(d,2H),7.35-7.38(q,1H),6.53(s,1H),1.31(s,9H)。
LC-MS(ESI):377.2(M+H)+
实施例57
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-嘌呤-7-基)-苯基]-脲(化合物57)的制备
Figure PCTCN2017076265-appb-000092
步骤1:4-(6-甲氧基-嘌呤-7-基)-苯胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用6-甲氧基嘌呤(泰坦)代替步骤1中的苯并咪唑,得到4-(6-甲氧基-嘌呤-7-基)-苯胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-嘌呤-7-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用4-(6-甲氧基-嘌呤-7-基)-苯胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-嘌呤-7-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.60(s,1H),9.08(s,1H),8.76(s,1H),8.60(s,1H),7.79-7.81(d,2H),7.67-7.69(d,2H),6.53(s,1H),4.14(s,3H),1.31(s,9H)。
LC-MS(ESI):408.1(M+H)+
实施例58
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基嘌呤-9-基)-苯基]-脲(化合物58)的制备
Figure PCTCN2017076265-appb-000093
步骤1:与实施例57的步骤1-步骤2中的制备方法相同,为实施例57步骤1-步骤2中同一反应另一产物。
1HNMR(DMSO-d6,400MHz)δ:9.63(s,1H),9.07(s,1H),8.70(s,1H),8.63(s,1H),7.62-7.64(d,2H),7.56-7.58(d,2H),6.53(s,1H),3.98(s,3H),1.31(s,9H)。
LC-MS(ESI):407.9(M+H)+
实施例59
1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-二甲基氨基-嘌呤-7-基)-苯基]-脲(化合物59)的制备
Figure PCTCN2017076265-appb-000094
步骤1:[7-(4-氨基-苯基)-7H-嘌呤-6-基]-二甲基-胺的制备
与实施例1的步骤1-步骤2中的制备方法相同,除了用6-二甲基氨基嘌呤(达瑞)代替步骤1中的苯并咪唑,得到[7-(4-氨基-苯基)-7H-嘌呤-6-基]-二甲基-胺。
步骤2:1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-二甲基氨基-嘌呤-7-基)-苯基]-脲的制备
与实施例4的步骤5相同,除了用[7-(4-氨基-苯基)-7H-嘌呤-6-基]-二甲基-胺代替1-(4-氨基-苯基)-1H-苯并咪唑-5-醇,制得1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-二甲基氨基-嘌呤-7-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.57(s,1H),9.05(s,1H),8.52(s,1H),8.27(s,1H),7.76-7.78(d,2H),7.64-7.66(d,2H),6.53(s,1H),3.49(s,6H),1.30(s,9H)。
LC-MS(ESI):421.2(M+H)+
实施例60
1-(4-苯并咪唑-1-基-苯基)-3-噻唑-2-基-脲(化合物60)的制备
Figure PCTCN2017076265-appb-000095
步骤1:噻唑-2-基-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3相同,除了用2-氨基噻唑(TCI)代替3-氨基异噁唑,得到噻唑-2-基-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-噻唑-2-基-脲的制备
与实施例1的步骤4相同,除了用噻唑-2-基-氨基甲酸苯基酯(活泼酯)代替异噁唑-3-基-氨基甲酸苯基酯(活泼酯),得到1-(4-苯并咪唑-1-基-苯基)-3-噻唑-2-基-脲。
1HNMR(DMSO-d6,400MHz)δ:10.65(s,1H),9.24(s,1H),8.52(s,1H),7.77-7.79(m,1H),7.73-7.75(d,2H),7.62-7.64(d,2H),7.57-7.60(m,1H),7.39-7.40(d,1H),7.29-7.34(m,2H),7.13-7.14(d,1H)。
LC-MS(ESI):336.1(M+H)+
实施例61
1-(4-苯并咪唑-1-基-苯基)-3-(4-甲基-噻唑-2-基)-脲(化合物61)的制备
Figure PCTCN2017076265-appb-000096
步骤1:(4-甲基-噻唑-2-基)-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3相同,除了用2-氨基-4-甲基噻唑(TCI)代替3-氨基异噁唑,得到(4-甲基-噻唑-2-基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(4-甲基-噻唑-2-基)-脲的制备
与实施例1的步骤4相同,除了用(4-甲基-噻唑-2-基)-氨基甲酸苯基酯(活泼酯)代替异噁唑-3-基-氨基甲酸苯基酯(活泼酯),得到1-(4-苯并咪唑-1-基-苯基)-3-(4-甲基-噻唑-2-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:10.50(s,1H),9.27(s,1H),8.51(s,1H),7.76-7.79(m,1H),7.73-7.75(d,2H),7.61-7.63(d,2H),7.57-7.59(m,1H),7.29-7.35(m,2H),6.66(s,1H),2.24(s,3H)。
LC-MS(ESI):350.1(M+H)+
实施例62
1-(4-苯并咪唑-1-基-苯基)-3-[1,3,4]噻二唑-2-基-脲(化合物62)的制备
Figure PCTCN2017076265-appb-000097
步骤1:[1,3,4]噻二唑-2-基-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3相同,除了用2-氨基噻二唑(泰坦)代替3-氨基异噁唑,制得[1,3,4]噻二唑-2-基-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-[1,3,4]噻二唑-2-基-脲的制备
与实施例1的步骤4相同,除了[1,3,4]噻二唑-2-基-氨基甲酸苯基酯(活泼酯)代替异噁唑-3-基-氨基甲酸苯基酯(活泼酯),得到1-(4-苯并咪唑-1-基-苯基)-3-[1,3,4]噻二唑-2-基-脲。
1HNMR(DMSO-d6,400MHz)δ:11.10(s,1H),9.36(s,1H),9.08(s,1H),8.53(s,1H),7.73-7.79(m,3H),7.58-7.65(m,3H),7.29-7.36(m,2H)。
LC-MS(ESI):337.1(M+H)+
实施例63
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-[1,3,4]噻二唑-2-基)-脲(化合物63)的制备
Figure PCTCN2017076265-appb-000098
步骤1:(5-叔丁基-[1,3,4]噻二唑-2-基)-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3相同,除了用2-氨基-5-叔丁基噻二唑(达瑞)代替3-氨基异噁唑,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-[1,3,4]噻二唑-2-基)-脲。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-[1,3,4]噻二唑-2-基)-脲的制备
与实施例1的步骤4相同,除了(5-叔丁基-[1,3,4]噻二唑-2-基)-氨基甲酸苯基酯(活泼酯)代替异噁唑-3-基-氨基甲酸苯基酯(活泼酯),得到1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-[1,3,4]噻二唑-2-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:11.09(s,1H),9.35(s,1H),8.56(s,1H),7.80-7.82(m,1H),7.75-7.77(d,2H),7.61-7.64(d,2H),7.57-7.60(m,1H),7.30-7.36(m,2H),1.37(s,9H)。
LC-MS(ESI):393.1(M+H)+
实施例64
1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-1H-吡唑-3-基)-脲(化合物64)的制备
Figure PCTCN2017076265-appb-000099
步骤1:(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯的制备
将4-苯并咪唑-1-基-苯胺(实施例1步骤2中合成)(10.0g,0.048mol)、吡啶(11.33g,0.143mol)溶解于100mlTHF中;冰浴下缓慢滴加氯甲酸苯酯(11.23g,0.072mol);滴毕,移走冰浴缓慢升至室温反应3小时。将反应液倒入水中,用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得棕黄色固体。将所得固体用甲基叔丁基醚/石油醚(1∶1)打浆得14g黄色固体状的(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-1H-吡唑-3-基)-脲的制备
将步骤1中得到的(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯(100mg,0.304mmol)、三乙胺(92mg,0.912mmol)、3-氨基-5-甲基吡唑(50.7mg,0.365mmol,TCI)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得40mg白色固体状的1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-1H-吡唑-3-基)-脲;同时得到10mg化合物113(见实施例113)。
1HNMR(DMSO-d6,400MHz)δ:11.97(s,1H),9.42(s,1H),8.96(s,1H),8.49(s,1H),7.76-7.78(m,1H),7.67-7.70(d,2H),7.56-7.59(m,3H),7.28-7.35(m,2H),6.02(s,1H),2.20(s,3H)。
LC-MS(ESI):333.1(M+H)+
实施例65
1-(4-苯并咪唑-1-基-苯基)-3-(5-苯基-1H-吡唑-3-基)-脲(化合物65)的制备
Figure PCTCN2017076265-appb-000100
与实施例64的制备方法相同,除了用3-氨基-5-苯基吡唑(达瑞)代替步骤2中的3-氨基-5-甲基吡唑,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-苯基-1H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:12.80(s,1H),9.27(s,1H),9.13(s,1H),8.51(s,1H),7.71-7.79(m,5H),7.58-7.61(m,3H),7.45-7.48(m,2H),7.29-7.38(m,3H),6.72(s,1H)。
LC-MS(ESI):395.1(M+H)+
实施例66
1-(4-苯并咪唑-1-基-苯基)-3-(5-环丙基-2H-吡唑-3-基)-脲(化合物66)的制备
Figure PCTCN2017076265-appb-000101
与实施例64的制备方法相同,除了用3-环丙基-1H-吡唑-5-氨基(达瑞)代替步骤2中的3-氨基-5-甲基吡唑,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-环丙基-2H-吡唑-3-基)-脲。同时得到化合物114(见实施例114)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.37(s,1H),8.96(s,1H),8.50(s,1H),7.76-7.79(m,1H),7.67-7.70(d,2H),7.56-7.59(m,3H),7.30-7.34(m,2H),5.92(s,1H),1.85-1.89(m,1H),0.91-0.95(m,2H),0.66-0.70(m,2H)。
LC-MS(ESI):359.2(M+H)+
实施例67
1-(4-苯并咪唑-1-基-苯基)-3-(5-三氟甲基-2H-吡唑-3-基)-脲(化合物67)的制备
Figure PCTCN2017076265-appb-000102
Figure PCTCN2017076265-appb-000103
与实施例64的制备方法相同,除了用5-氨基-3-三氟甲基吡唑(达瑞)代替步骤2中的3-氨基-5-甲基吡唑,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-三氟甲基-2H-吡唑-3-基)-脲。同时得到化合物115(见实施例115)。
1HNMR(DMSO-d6,400MHz)δ:13.30(s,1H),9.33(s,1H),9.33(s,1H),8.50(s,1H),7.76-7.78(m,1H),7.70-7.72(d,2H),7.57-7.62(m,3H),7.28-7.36(m,2H),6.42(s,1H)。
LC-MS(ESI):387.1(M+H)+
实施例68
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2H-吡唑-3-基)-脲(化合物68)的制备
Figure PCTCN2017076265-appb-000104
步骤1:(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯的制备
将4-苯并咪唑-1-基-苯胺(实施例1步骤2中合成)(10.0g,0.048mol)、吡啶(11.33g,0.143mol)溶解于100mlTHF中;冰浴下缓慢滴加氯甲酸苯酯(11.23g,0.072mol);滴毕,移走冰浴缓升温室温反应3小时。将反应液倒入水中,用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩得棕黄色固体。将所得固体用甲基叔丁基醚/石油醚(1∶1)打浆,得14g黄色固体状的(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2H-吡唑-3-基)-脲的制备
将步骤1得到的(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯(100mg,0.304mmol)、三乙胺(92mg,0.912mmol)、3-叔丁基-吡唑-5-胺(50.7mg,0.365mmol)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得40mg白色固体状的1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2H-吡唑-3-基)-脲。同时得到12mg化合物116(见实施例116)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.41(s,1H),9.00(s,1H),8.50(s,1H),7.76-7.78(m,1H),7.67-7.70(d,2H),7.57-7.59(m,3H),7.28-7.35(m,2H),6.03(s,1H),1.27(s,9H)。
LC-MS(ESI):375.2(M+H)+
实施例69
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲(化合物69)的制备
Figure PCTCN2017076265-appb-000105
与实施例68的制备方法相同,除了用4-(5-氟-苯并咪唑-1-基)-苯胺(实施例22步骤1中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲。同时得到化合物125(见实施例125)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.43(s,1H),8.99(s,1H),8.56(s,1H),7.67-7.70(d,2H),7.55-7.60(m,4H),7.17-7.22(m,1H),6.03(s,1H),1.27(s,9H)。
LC-MS(ESI):393.1(M+H)+
实施例70
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲(化合物70)的制备
Figure PCTCN2017076265-appb-000106
与实施例68的制备方法相同,除了用4-(5-三氟甲基-苯并咪唑-1-基)-苯胺(实施例23步骤1中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲。同时得到化合物126(见实施例126)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.44(s,1H),9.00(s,1H),8.72(s,1H),8.15(d,1H),7.75-7.77(d,1H),7.70-7.72(d,2H),7.64-7.67(dd,1H),7.60-7.63(d,2H),6.03(s,1H),1.27(s,9H)。
LC-MS(ESI):443.1(M+H)+
实施例71
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物71)的制备
Figure PCTCN2017076265-appb-000107
与实施例68的制备方法相同,除了用4-(5-甲氧基-苯并咪唑-1-基)-苯胺(实施例4步骤3中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.43(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.46-7.48(d,1H),7.29-7.30(d,1H),6.94-6.96(dd,1H),6.02(s,1H),3.82(s,3H),1.27(s,9H)。
LC-MS(ESI):405.1(M+H)+
实施例72
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲(化合物72)的制备
Figure PCTCN2017076265-appb-000108
与实施例68的制备方法相同,除了用4-(5-乙氧基-苯并咪唑-1-基)-苯胺(实施例6步骤1中合成)代替步骤1中的中间体4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.40(s,1H),8.98(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.44-7.47(d,1H),7.27(d,1H),6.92-6.95(dd,1H),6.02(s,1H),4.06-4.11(q,2H),1.34-1.38(t,3H),1.27(s,9H)。
LC-MS(ESI):419.2(M+H)+
实施例73
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲(化合物73)的制备
Figure PCTCN2017076265-appb-000109
与实施例68的制备方法相同,除了用4-(5-己氧基-苯并咪唑-1-基)-苯胺(实施例7步骤1中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基- 2H-吡唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲。同时得到化合物117(见实施例117)。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.44-7.46(d,1H),7.27-7.28(d,1H),6.92-6.95(dd,1H),6.02(s,1H),4.00-4.03(t,2H),1.70-1.76(m,2H),1.41-1.47(m,2H),1.30-1.35(m,4H),1.27(s,9H),0.87-0.91(t,3H)。
LC-MS(ESI):475.2(M+H)+
实施例74
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲(化合物74)的制备
Figure PCTCN2017076265-appb-000110
与实施例68的制备方法相同,除了用4-(5-异丙氧基-苯并咪唑-1-基)-苯胺(实施例8中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.41(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.44-7.46(d,1H),7.27-7.28(d,1H),6.91-6.94(dd,1H),6.02(s,1H),4.61-4.67(m,1H),1.28-1.30(d,6H),1.27(s,9H)。
LC-MS(ESI):433.1(M+H)+
实施例75
1-[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-3-(5-叔丁基-2H-吡唑-3-基)-脲(化合物75)的制备
Figure PCTCN2017076265-appb-000111
Figure PCTCN2017076265-appb-000112
步骤1:4-(5-仲丁氧基-苯并咪唑-1-基)-苯胺的制备
于室温,将1-(4-氨基-苯基)-1H-苯并咪唑-5-醇(实施例4步骤4制备)(300mg,1.22mol)溶解于DMF(20ml)中,加入氢氧化钠(98mg,2.44mmol),室温搅拌30分钟,过程中有固体析出。于室温,加入氯代仲丁烷(TCI)(170g,1.83mmol),将混合物加热至90℃,反应1.5小时。将反应液冷却至室温后,缓慢倒入100ml水中,用乙酸乙酯萃取(60ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。得350mg黄黑色油状物,直接用于下一步反应,无需纯化。
步骤2:[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-氨基甲酸苯基酯的制备
将步骤1得到的4-(5-仲丁氧基-苯并咪唑-1-基)-苯胺(350g,1.24mmol)、吡啶(294mg,3.72mmol)溶解于THF(100ml)中,冰浴下滴加氯甲酸苯酯(291g,1.86mol),过程中有固体析出。滴毕,升至室温反应1.5h后,加水100ml淬灭,用乙酸乙酯萃取(60ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得350mg[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-氨基甲酸苯基酯。
步骤3:1-[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-3-(5-叔丁基-2H-吡唑-3-基)-脲的制备
与实施例68的步骤2相同,除了用[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-氨基甲酸苯基酯代替中的(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯,制得1-[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-3-(5-叔丁基-2H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.44-7.46(d,1H),7.27-7.28(d,1H),6.92-6.95(dd,1H),6.02(s,1H),4.38-4.45(m,1H),1.54-1.74(m,2H),1.27(s,9H),1.25-1.26(d,3H),0.94-0.97(t,3H)。
LC-MS(ESI):447.2(M+H)+
实施例76
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丁氧基-苯并咪唑-1-基)-苯基]-脲(化合物76)的制备
Figure PCTCN2017076265-appb-000113
与实施例75的制备方法相同,除了用1-氯-2-甲基丙烷(TCI)代替步骤1中的氯代仲丁烷,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丁氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.41(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.26-7.27(d,1H),6.94-6.97(dd,1H),6.02(s,1H),3.80-3.81(d,2H),2.02-2.08(m,1H),1.27(s,9H),1.00-1.02(d,6H)。
LC-MS(ESI):447.3(M+H)+
实施例77
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物77)的制备
Figure PCTCN2017076265-appb-000114
步骤1:4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
于室温,将1-(4-氨基苯基)-1H-苯并咪唑-5-醇(实施例4步骤4制备)(18.0g,0.08mol)溶解于DMF(200ml)中,加入氢氧化钠(9.6g,0.24mol),室温搅拌30分钟,过程中有固体析出。于室温,加入氯乙基甲基醚(11.34g, 0.12mol),将混合物加热至90℃,反应1.5小时。将反应液冷却至室温,缓慢倒入600ml水中,有固体析出,室温搅拌30分钟。过滤,所得固体水洗,真空干燥,得20.5g粉红固体状的4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺。
步骤2:{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-氨基甲酸苯基酯的制备
将步骤1得到的4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺(10.0g,0.035mol)、吡啶(8.37g,0.106mol)溶解于THF(100ml)中,冰浴下滴加氯甲酸苯酯(6.63g,0.042mol),过程中有固体析出。滴毕,升至室温反应1.5h后,加水200ml淬灭,用乙酸乙酯萃取(100ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得15g{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-氨基甲酸苯基酯。LC-MS(ESI):404.1(M+H)+
步骤3:1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲
与实施例68的步骤2相同,除了用{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-氨基甲酸苯基酯代替步骤2中的(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。同时得到化合物120(见实施例120)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.41(s,1H),8.99(s,1H),8.43(s,1H),7.65-7.68(d,2H),7.55-7.57(d,2H),7.45-7.48(d,1H),7.30-7.31(d,1H),6.95-6.98(dd,1H),6.02(s,1H),4.14-4.16(m,2H),3.68-3.71(m,2H),3.33(s,3H),1.27(s,9H)。
LC-MS(ESI):449.2(M+H)+
实施例78
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物78)的制备
Figure PCTCN2017076265-appb-000115
与实施例77的步骤1-步骤3的制备方法相同,除了用2-乙氧基氯乙烷代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.40(s,1H),8.98(s,1H),8.43(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.30(d,1H),6.95-6.98(dd, 1H),6.02(s,1H),4.13-4.16(m,2H),3.72-3.74(m,2H),3.50-3.55(q,2H),1.27(s,9H),1.13-1.17(t,3H)。
LC-MS(ESI):463.2(M+H)+
实施例79
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物79)的制备
Figure PCTCN2017076265-appb-000116
与实施例77的步骤1-步骤3的制备方法相同,除了用溴乙醇代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。同时得到化合物121(见实施例121)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.42(s,1H),8.99(s,1H),8.43(s,1H),7.66-7.68(d,2H),7.55-7.58(d,2H),7.46-7.48(d,1H),7.28-7.29(d,1H),6.95-6.98(dd,1H),6.02(s,1H),4.90(m,1H),4.03-4.06(t,2H),3.75-3.76(m,2H),1.27(s,9H)。
LC-MS(ESI):435.2(M+H)+
实施例80
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-3-甲氧基-丙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物80)的制备
Figure PCTCN2017076265-appb-000117
Figure PCTCN2017076265-appb-000118
与实施例77的步骤1-步骤3的制备方法相同,除了用1-氯-3-甲氧基-2-丙醇(达瑞)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-3-甲氧基-丙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.44(s,1H),9.00(s,1H),8.46(s,1H),7.67-7.69(d,2H),7.55-7.58(d,2H),7.46-7.48(d,1H),7.28(d,1H),6.96-6.99(dd,1H),6.03(s,1H),5.14(s,1H),3.93-4.02(m,3H),3.42-3.48(m,2H),3.30(s,3H),1.27(s,9H)。
LC-MS(ESI):479.1(M+H)+
实施例81
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二甲氨基-丙氧基)-苯并咪唑-1-基]-苯基}-脲化合物81)的制备
Figure PCTCN2017076265-appb-000119
与实施例77的步骤1-步骤3的制备方法相同,除了N,N-二甲氨基氯丙烷盐酸盐(泰坦)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二甲氨基-丙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.04(s,1H),9.59(s,1H),9.09(s,1H),8.42(s,1H),7.66-7.69(d,2H),7.54-7.57(d,2H),7.45-7.47(d,1H),7.27(d,1H),6.94-6.96(dd,1H),6.04(s,1H),4.04-4.07(t,2H),2.41-2.45(t,2H),2.19(s,6H),1.87-1.90(m,2H),1.27(s,9H)。
LC-MS(ESI):476.2(M+H)+
实施例82
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二丁基氨基丙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物82)的制备
Figure PCTCN2017076265-appb-000120
与实施例77的步骤1-步骤3的制备方法相同,除了用N-(3-氯丙基)二丁基胺(泰坦)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二丁基氨基丙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.52(s,1H),9.02(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.54-7.56(d,2H),7.47-7.50(d,1H),7.32(d,1H),6.95-6.97(dd,1H),6.03(s,1H),4.11-4.14(t,2H),3.12(m,2H),2.95(m,4H),2.10(m,2H),1.58(m,4H),1.29-1.35(m,4H),1.27(s,9H),0.88-1.92(t,6H)。
LC-MS(ESI):560.2(M+H)+
实施例83
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氰基甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物83)的制备
Figure PCTCN2017076265-appb-000121
与实施例77的步骤1-步骤3的制备方法相同,除了用氯乙腈代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氰基甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.51(s,1H),7.67-7.69(d,2H),7.57-7.59(d,2H),7.53-7.55(d,1H),7.51-7.52(d,1H),7.06-7.08(dd,1H),6.02(s,1H),5.24(s,2H),1.27(s,9H)。
LC-MS(ESI):430.2(M+H)+
实施例84
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物84)的制备
Figure PCTCN2017076265-appb-000122
与实施例68的制备方法相同,除了用4-(5-三氟甲氧基-苯并咪唑-1-基)-苯胺(实施例21中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5三氟甲氧基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.42(s,1H),9.01(s,1H),8.65(s,1H),7.79(d,1H),7.69-7.71(d,2H),7.64-7.67(d,1H),7.59-7.61(d,2H),7.32-7.35(dd,1H),6.03(s,1H),1.27(s,9H)。
LC-MS(ESI):459.1(M+H)+
实施例85
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲(化合物85)的制备
Figure PCTCN2017076265-appb-000123
与实施例77的步骤1-步骤3的制备方法相同,除了用3-氯甲基-3-甲基氧杂环丁烷(达瑞)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.44(s,1H),7.67-7.69(d,2H),7.55-7.58(d,2H),7.47-7.49(d,1H),7.35-7.36(d,1H),6.98-7.01(dd,1H),6.02(s,1H),4.52-4.54(d,2H),4.32-4.34(d,2H),4.11(s,2H),1.40(s,3H),1.27(s,9H)。
LC-MS(ESI):475.2(M+H)+
实施例86
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲(化合物86)的制备
Figure PCTCN2017076265-appb-000124
与实施例77的步骤1-步骤3的制备方法相同,除了用2-氯甲基四氢呋喃(达瑞)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲。同时合成化合物123(见实施例123)。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.43(s,1H),7.66-7.69(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.29-7.30(d,1H),6.94-6.97(dd,1H),6.02(s,1H),4.16-4.22(m,1H),3.95-4.04(m,2H),3.78-3.84(m,1H),3.67-3.72(m,1H),1.98-2.07(m,1H),1.81-1.95(m,2H),1.66-1.75(m,1H),1.27(s,9H)。
LC-MS(ESI):475.2(M+H)+
实施例87
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲(化合物87)的制备
Figure PCTCN2017076265-appb-000125
与实施例77的步骤1-步骤3的制备方法相同,除了用2-(氯甲基)四氢吡喃(达瑞)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲。同时得到化合物122(见实施例122)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.42(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.27-7.28(d,1H),6.94-6.97(dd,1H),6.02(s,1H),3.89-3.97(m,3H),3.63-3.69(m,1H),3.47-3.41(m,1H),1.82-1.84(m,1H),1.67-1.70(m,1H),1.46-1.54(m,4H),1.27(s,9H)。
LC-MS(ESI):489.2(M+H)+
实施例88
(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯(化合物88)的制备
Figure PCTCN2017076265-appb-000126
与实施例77的步骤1-步骤3的制备方法相同,除了用氯乙酸乙酯(达瑞)代替步骤1中的2-氯乙基甲基醚,得到(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯。同时得到化合物124(见实施例124)。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.44(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.47-7.49(d,1H),7.26-7.27(d,1H),6.98-7.01(dd,1H),6.02(s,1H),4.84(s,2H),4.16-4.21(q,2H),1.27(s,9H),1.21-1.25(t,3H)。
LC-MS(ESI):477.2(M+H)+
实施例89
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物89)的制备
Figure PCTCN2017076265-appb-000127
Figure PCTCN2017076265-appb-000128
步骤1:{4-[5-(3-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-氨基甲酸苯基酯的制备
将4-[5-(3-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯胺(实施例16步骤1中合成)(300mg,0.887mmol)、吡啶(210mg,2.66mmol)溶解于THF(20ml)中,冰浴下滴加氯甲酸苯酯(277.6mg,1.74mmol),过程中有固体析出。滴毕,升至室温反应2h后,加水50ml淬灭,用乙酸乙酯萃取(30ml×2),有机相用饱和NaCl溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得280mg{4-[5-(3-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-氨基甲酸苯基酯。LC-MS(ESI):459.2(M+H)+
步骤2:1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲
与实施例68的步骤2中制备方法相同,除了用4-[5-(3-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-氨基甲酸苯基酯代替(4-苯并咪唑-1-基-苯基)-氨基甲酸苯基酯,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。同时得到化合物119(见实施例119)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.41(s,1H),8.99(s,1H),8.43(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.31-7.32(d,1H),6.94-6.97(dd,1H),6.02(s,1H),4.14-4.17(m,2H),3.60(m,4H),2.73(m,2H),2.51(m,4H),1.27(s,9H)。
LC-MS(ESI):504.1(M+H)+
实施例90
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物90)的制备
Figure PCTCN2017076265-appb-000129
Figure PCTCN2017076265-appb-000130
与实施例77的步骤1-步骤3的制备方法相同,除了用1-(2-氯乙基)哌啶盐酸盐代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.41(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.53-7.57(d,2H),7.45-7.47(d,1H),7.30(d,1H),6.94-6.96(dd,1H),6.02(s,1H),4.12-4.14(t,2H),2.70(m,2H),2.47(m,4H),1.49-1.54(m,4H),1.39-1.42(m,2H),1.27(s,9H)。
LC-MS(ESI):502.2(M+H)+
实施例91
1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-2H-吡唑-3-基)-脲化合物91)的制备
Figure PCTCN2017076265-appb-000131
与实施例77的步骤1-步骤3的制备方法相同,除了用2-(环己亚胺基)乙基氯(润捷)代替步骤1中的2-氯乙基甲基醚,得到1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-2H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.44(s,1H),9.00(s,1H),8.43(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.30-7.31(d,1H),6.94-6.97(dd,1H),6.02(s,1H),4.12-4.15(t,2H),2.97(m,2H),2.79(m,4H),1.63(m,4H),1.55(m,4H),1.27(s,9H)。
LC-MS(ESI):516.3(M+H)+
实施例92
1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲(化合物92)的制备
Figure PCTCN2017076265-appb-000132
Figure PCTCN2017076265-appb-000133
与实施例77的步骤1-步骤3的制备方法相同,除了用1-(3-氯丙基)-4-甲基哌嗪(润捷)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲。同时合成化合118(见实施例118)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.50(s,1H),9.03(s,1H),8.42(s,1H),7.67-7.69(d,2H),7.54-7.56(d,2H),7.44-7.46(d,1H),7.27(d,1H),6.92-6.95(dd,1H),6.03(s,1H),4.03-4.06(t,2H),2.25-2.46(m,10H),2.17(s,3H),1.85-1.92(m,2H),1.27(s,9H)。
LC-MS(ESI):531.3(M+H)+
实施例93
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-氟-苄氧基)-苯并咪唑-1-基]-苯基}-脲(化合物93)的制备
Figure PCTCN2017076265-appb-000134
与实施例77的步骤1-步骤3的制备方法相同,除了用3-氟溴苄(达瑞)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-氟-苄氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.50(s,1H),9.02(s,1H),8.47(s,1H),7.67-7.69(d,2H),7.55-7.57(d,2H),7.42-7.50(m,2H),7.31-7.38(m,3H),7.14-7.18(m,1H),7.05-7.07(m,1H),6.04(s,1H),5.22(s,2H),1.27(s,9H)。
LC-MS(ESI):499.2(M+H)+
实施例94
1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[4-(1-环己基-1H-四唑-5-基)-丁氧基]-苯并咪唑-1-基}-苯基)-脲化合物94)的制备
Figure PCTCN2017076265-appb-000135
与实施例77的步骤1-步骤3的制备方法相同,除了用1-环己基-5-(4-氯丁基)-四氮唑(润捷)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[4-(1-环己基-1H-四唑-5-基)-丁氧基]-苯并咪唑-1-基}-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.41(s,1H),8.99(s,1H),8.42(s,1H),7.66-7.68(d,2H),7.55-7.57(d,2H),7.45-7.47(d,1H),7.30-7.31(d,1H),6.93-6.96(dd,1H),6.02(s,1H),4.38-4.45(m,1H),4.08-4.11(t,2H),2.99-3.02(t,2H),1.66-1.98(m,12H),1.39-1.48(m,2H),1.27(s,9H)。
LC-MS(ESI):597.3(M+H)+
实施例95
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(4-吗啉-4-基-[1,2,5]噻二唑-3-基氧基)-苯并咪唑-1-基]-苯基}-脲(化合物95)的制备
Figure PCTCN2017076265-appb-000136
与实施例77的步骤1-步骤3的制备方法相同,除了用3-氯-4-吗啉基-1,2,5-噻二唑(润捷)代替步骤1中的2-氯乙基甲基醚,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(4-吗啉-4-基-[1,2,5]噻二唑-3-基氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.43(s,1H),9.00(s,1H),8.58(s,1H),7.75-7.76(d,1H),7.69-7.71(d,2H),7.60-7.63(m,3H),7.29-7.32(dd,1H),6.03(s,1H),3.76-3.78(t,4H),3.55-3.57(t,4H),1.27(s,9H)。
LC-MS(ESI):560.1(M+H)+
实施例96
4-(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H苯并咪唑-5-基氧基)-吡啶-2-羧酸甲基胺(化合物96)的制备
Figure PCTCN2017076265-appb-000137
与实施例77的步骤1-步骤3的制备方法相同,除了用N-甲基-4-氯-2-吡啶甲酰胺(泰坦)代替步骤1中的2-氯乙基甲基醚,得到4-(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H苯并咪唑-5-基氧基)-吡啶-2-羧酸甲酰胺。同时得到化合物127(见实施例127)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.44(s,1H),9.01(s,1H),8.77-8.80(m,1H),8.62(s,1H),8.51-8.52(d,1H),763-7.72(m,6H),7.39-7.40(d,1H),7.18-7.21(d,2H),6.03(s,1H),2.77-2.79(d,3H),1.27(s,9H)。
LC-MS(ESI):525.2(M+H)+
实施例97
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲(化合物97)的制备
Figure PCTCN2017076265-appb-000138
与实施例68的制备方法相同,除了用4-(5-氟-7-甲基-苯并咪唑-1-基)-苯胺(实施例33步骤1中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.42(s,1H),9.02(s,1H),8.27(s,1H),7.61-7.64(d,2H),7.45-7.48(d,2H),7.36-7.39(dd,1H),6.93-6.96(dd,1H),6.03(s,1H),2.04(s,3H),1.27(s,9H)。
LC-MS(ESI):407.1(M+H)+
实施例98
1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物98)的制备
Figure PCTCN2017076265-appb-000139
步骤1:4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺的制备
与实施例27的步骤1至步骤5的制备方法相同,除了用2-氯乙基甲基醚代替步骤5中的4-(2-氯-乙基)-吗啉盐酸盐,得到4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺。
步骤2:1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲
与实施例68的制备方法相同,除了用4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。同时得到化合物128(见实施例128)。
1HNMR(DMSO-d6,400MHz)δ:12.03(s,1H),9.40(s,1H),9.00(s,1H),8.35(s,1H),7.67-7.69(d,2H,),7.63-7.65(d,1H),7.56-7.59(d,2H),7.03-7.04(d,1H),6.91- 6.94(dd,1H),6.02(s,1H),4.11-4.13(m,2H),3.65-3.68(m,2H),3.31(s,3H),1.27(s,9H)。
LC-MS(ESI):ESI 449.1(M+H)+
实施例99
1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲(化合物99)的制备
Figure PCTCN2017076265-appb-000140
与实施例68的制备方法相同,除了用4-(5,6-二甲氧基-苯并咪唑-1-基)-苯胺(实施例31步骤1中合成)代替步骤1中的4-苯并咪唑-1-基-苯胺,得到1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲。同时得到化合129(见实施例129)。
1HNMR(DMSO-d6,400MHz)δ:12.02(s,1H),9.40(s,1H),9.00(s,1H,),8.27(s,1H),7.67-7.69(d,2H),7.56-7.58(d,2H),7.31(s,1H),7.04(s,1H),6.02(s,1H),3.82(s,3H),3.79(s,3H),1.27(s,9H)。
LC-MS(ESI):ESI 435.1(M+H)+
实施例100
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-甲基-2H-吡唑-3-基)-脲(化合物100)的制备
Figure PCTCN2017076265-appb-000141
步骤1:(5-叔丁基-2-甲基-2H-吡唑-3-基)-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3相同,除了用5-氨基-3-叔丁基-1-甲基吡唑(达瑞)代替步骤3中的3-氨基异噁唑,得到(5-叔丁基-2-甲基-2H-吡唑-3-基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-甲基-2H-吡唑-3-基)-脲的制备
与实施例1的步骤4相同,除了用(5-叔丁基-2-甲基-2H-吡唑-3-基)-氨基甲酸苯基酯(活泼酯)代替异噁唑-3-基-氨基甲酸苯基酯(活泼酯),得到1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-甲基-2H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.17(s,1H),8.58(s,1H),8.51(s,1H),7.77-7.79(m,1H),7.69-7.72(d,2H),7.57-7.60(m,3H),7.29-7.36(m,2H),6.08(s,1H),3.63(s,3H),1.23(s,9H)。
LC-MS(ESI):389.2(M+H)+
实施例101
1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲(化合物101)的制备
Figure PCTCN2017076265-appb-000142
步骤1:1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲的制备
将4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯胺(实施例77步骤1中制得)(107mg,0.378mmol)、(5-叔丁基-2-甲基-2H-吡唑-3-基)-氨基甲酸苯基酯(实施例100步骤1中制得)(156.0mg,0.571mmol)和三乙胺(115mg,1.14mmol)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得56mg白色固体的1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.16(s,1H),8.57(s,1H),8.43(s,1H),7.67-7.69(d,2H,),7.56-7.58(d,2H),7.45-7.47(d,1H),7.29-7.30(d,1H),6.94-6.97(dd,1H),6.08(s,1H),4.14-4.16(t,2H),3.68-3.70(t,2H),3.62(s,3H),3.33(s,3H),1.22(s,9H)。
LC-MS:ESI 463.3(M+H)+
实施例102
1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-甲氧基)-苯并咪唑-1-基]-苯基}-脲(化合物102)的制备
Figure PCTCN2017076265-appb-000143
步骤1:1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-甲氧基)-苯并咪唑-1-基]-苯基}-脲的制备
将4-[5-(2-吗啉-4-基-甲氧基)-苯并咪唑-1-基]-苯胺(实施例16步骤1中制得)(125mg,0.368mmol)、(5-叔丁基-2-甲基-2H-吡唑-3-基)-氨基甲酸苯基酯(实施例100步骤1中制得)(152.0mg,0.555mmol)和三乙胺(112mg,1.11mmol)溶解于10mlTHF中,回流反应过夜。次日,将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得60mg白色固体状的1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-甲氧基)-苯并咪唑-1-基]-苯基}-脲。
1HNMR(DMSO-d6,400MHz)δ:9.17(s,1H),8.57(s,1H),8.43(s,1H),7.67-7.70(d,2H,),7.55-7.57(d,2H),7.45-7.47(d,1H),7.30-7.31(d,1H),6.94-6.96(dd,1H),6.08(s,1H),4.13-4.16(t,2H),3.62(s,3H),3.58-3.60(t,4H),2.71-2.74(t,2H),2.49-2.51(m,4H),1.22(s,9H)。
LC-MS:ESI 518.2(M+H)+
实施例103
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-羟基-乙基)-2H-吡唑-3-基]-脲(化合物103)的制备
Figure PCTCN2017076265-appb-000144
步骤1:2-(5-氨基-3-叔丁基-吡唑-1-基)-乙醇的制备
将2-肼基乙醇(6.69g,0.088mol,泰坦)、氰基频呐酮(10g,0.08mol,达瑞)、浓盐酸(0.2ml)于80ml乙醇中回流反应5小时。反应液减压浓缩,将所得油状物放置过夜,次日有固体析出。将所得固体用甲基叔丁基醚打浆,得12g黄白固体状的2-(5-氨基-3-叔丁基-吡唑-1-基)-乙醇。
步骤2:5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基胺的制备
氮气氛下,将步骤1得到的2-(5-氨基-3-叔丁基-吡唑-1-基)-乙醇(6.6g,0.0361mol)、TBDMSCl(6.49g,0.0433mmol)、咪唑(6.13g,0.09mol)溶解于100mlDMF中,室温反应过夜。次日,将反应液倒入水中,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品6g。无需纯化,直接用于下一步反应。
步骤3:{5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基}-氨基甲酸苯基酯的制备
与实施例1的步骤3的制备方法相同,除了用5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基胺代替步骤3中的3-氨基异噁唑,制得{5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基}-氨基甲酸苯基酯。
步骤4:1-(4-苯并咪唑-1-基-苯基)-3-{5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基}-脲的制备
与实施例1的步骤4的制备方法相同,除了用{5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基}-氨基甲酸苯基酯代替步骤4中的异噁唑-3-基-氨基甲酸苯基酯,得到1-(4-苯并咪唑-1-基-苯基)-3-{5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基}-脲。
步骤5:1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-羟基-乙基)-2H-吡唑-3-基]-脲的制备
于室温,将步骤4得到的1-(4-苯并咪唑-1-基-苯基)-3-{5-叔丁基-2-[2-(叔丁基-二甲基-硅烷氧基)-乙基]-2H-吡唑-3-基}-脲(1.05g,1.97mmol)溶解于30mlTHF中,滴加TBAF(1M的THF溶液)3ml,滴毕室温反应15分钟。将反应液倒入水中,用乙酸乙酯萃取(80ml×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得930mg黄色固体状的1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-羟基-乙基)-2H-吡唑-3-基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.38(s,1H),8.51(s,1H),8.50(s,1H),7.76-7.78(m,1H),7.69-7.71(d,2H),7.57-7.60(m,3H),7.31-7.33(m,2H),6.12(s,1H),5.11-5.13(t,1H),4.00-4.02(m,2H),3.68-3.73(m,2H),1.23(s,9H)。
LC-MS(ESI):418.9(M+H)+
实施例104
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-苯基-2H-吡唑-3-基)-脲(化合物104)的制备
Figure PCTCN2017076265-appb-000145
步骤1:5-叔丁基-2-苯基-2H-吡唑-3-基胺的制备
与实施例103的步骤1相同,除了用苯肼代替2-肼基乙醇,得到5-叔丁基-2-苯基-2H-吡唑-3-基胺。
步骤2:(5-叔丁基-2-苯基-2H-吡唑-3-基)-氨基甲酸苯基酯的制备
与实施例1的步骤3相同,除了用5-叔丁基-2-苯基-2H-吡唑-3-基胺代替3-氨基异噁唑,得到(5-叔丁基-2-苯基-2H-吡唑-3-基)-氨基甲酸苯基酯。
步骤3:1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-苯基-2H-吡唑-3-基)-脲的制备
与实施例1的步骤4相同,除了用(5-叔丁基-2-苯基-2H-吡唑-3-基)-氨基甲酸苯基酯代替异噁唑-3-基-氨基甲酸苯基酯,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-苯基-2H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.30(s,1H),8.50(s,1H),8.50(s,1H),7.76-7.78(m,1H),7.64-7.66(d,2H),7.54-7.58(m,7H),7.41-7.44(m,1H),7.30-7.35(m,2H),6.41(s,1H),1.29(s,9H)。
LC-MS(ESI):451.2(M+H)+
实施例105
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-对甲苯基-2H-吡唑-3-基)-脲(化合物105)的制备
Figure PCTCN2017076265-appb-000146
与实施例104的步骤1-步骤3中的制备方法相同,除了用4-甲基苯肼代替步骤1中的苯肼,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-对甲苯基-2H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.29(s,1H),8.49(s,1H),8.43(s,1H),7.76-7.78(m,1H),7.63-7.66(d,2H),7.55-7.59(m,3H),7.41-7.43(d,2H),7.29-7.36(m,4H),6.39(s,1H),2.39(s,3H),1.29(s,9H)。
LC-MS(ESI):465.2(M+H)+
实施例106
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-甲氧基-苯基)-2H-吡唑-3-基]-脲(化合物106)的制备
Figure PCTCN2017076265-appb-000147
与实施例104的步骤1-步骤3中的制备方法相同,除了用4-甲氧基苯肼代替步骤1中的苯肼,得到1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-甲氧基-苯基)-2H-吡唑-3-基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.28(s,1H),8.50(s,1H),8.39(s,1H),7.76-7.78(m,1H),7.63-7.66(d,2H),7.55-7.59(m,3H),7.42-7.46(d,2H),7.28-7.34(m,1H),7.08-7.12(d,2H),6.38(s,1H),3.83(s,3H),1.29(s,9H)。
LC-MS(ESI):481.2(M+H)+
实施例107
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-三氟甲氧基-苯基)-2H-吡唑-3-基]-脲(化合物107)的制备
Figure PCTCN2017076265-appb-000148
与实施例104的步骤1-步骤3中的制备方法相同,除了用4-三氟甲氧基苯肼代替步骤1中的苯肼,得到1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-三氟甲氧基-苯基)-2H-吡唑-3-基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.29(s,1H),8.58(s,1H),8.51(s,1H),7.78-7.80(m,1H),7.65-7.72(m,4H),7.54-7.59(m,5H),7.29-7.36(m,2H),6.43(s,1H),1.30(s,9H)。
LC-MS(ESI):535.2(M+H)+
实施例108
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-氟-苯基)-2H-吡唑-3-基]-脲(化合物108)的制备
Figure PCTCN2017076265-appb-000149
与实施例104的步骤1-步骤3中的制备方法相同,除了用4-氟苯肼代替步骤1中的苯肼,得到1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-氟-苯基)-2H-吡唑-3-基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.27(s,1H),8.49(s,1H),8.47(s,1H),7.76-7.78(m,1H),7.64-7.66(d,2H),7.55-7.61(m,5H),7.37-7.41(m,2H),7.28-7.34(m,2H),6.40(s,1H),1.29(s,9H)。
LC-MS(ESI):469.2(M+H)+
实施例109
1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-氟-苯基)-2H-吡唑-3-基]-脲(化合物109)的制备
Figure PCTCN2017076265-appb-000150
与实施例104的步骤1-步骤3中的制备方法相同,除了用2-氟苯肼代替步骤1中的苯肼,得到1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-氟-苯基)-2H-吡唑-3-基]-脲。
1HNMR(DMSO-d6,400MHz)δ:9.17(s,1H),8.49(s,1H),8.47(s,1H),7.76-7.78(m,1H),7.63-7.65(d,2H),7.55-7.60(m,5H),7.53-7.48(m,1H),7.39-7.43(m,1H),7.28-7.35(m,2H),6.42(s,1H),1.28(s,9H)。
LC-MS(ESI):469.2(M+H)+
实施例110
1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-吡啶-2-基-2H-吡唑-3-基)-脲(化合物110)的制备
Figure PCTCN2017076265-appb-000151
与实施例104的步骤1-步骤3中的制备方法相同,除了用2-肼吡啶(达瑞)代替步骤1中的苯肼,得到1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-吡啶-2-基-2H-吡唑-3-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:11.32(s,1H),10.22(s,1H),8.53(s,1H),8.50-8.52(m,1H),8.01-8.05(m,1H),7.92-7.94(d,1H),7.77-7.79(m,3H),7.59-7.64(m,3H),7.29-7.37(m,3H),6.65(s,1H),1.31(s,9H)。
LC-MS(ESI):452.2(M+H)+
实施例111
4-{5-[3-(4-苯并咪唑-1-基-苯基)-脲基]-3-叔丁基-吡唑-1-基}-苯甲酸乙酯(化合物111)的制备
Figure PCTCN2017076265-appb-000152
与实施例104的步骤1-步骤3中的制备方法相同,除了用4-肼基苯甲酸乙酯(达瑞)代替步骤1中的苯肼,得到4-{5-[3-(4-苯并咪唑-1-基-苯基)-脲基]-3-叔丁基-吡唑-1-基}-苯甲酸乙酯。
1HNMR(DMSO-d6,400MHz)δ:9.32(s,1H),8.64(s,1H),8.49(s,1H),8.09-8.11(d,2H),7.75-7.78(m,3H),7.64-7.67(d,2H),7.55-7.58(m,3H),7.28-7.34(m,2H),6.45(s,1H),4.32-4.37(q,2H),1.32-1.36(t,3H),1.31(s,9H)。
LC-MS(ESI):523.2(M+H)+
实施例112
1-(2-丙烯酰基-5-叔丁基-2H-吡唑-3-基)-3-(4-咪唑-1-基-苯基)-脲(化合物112)的制备
Figure PCTCN2017076265-appb-000153
步骤1:1-(2-丙烯酰基-5-叔丁基-2H-吡唑-3-基)-3-(4-咪唑-1-基-苯基)-脲的制备
将1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2H-吡唑-3-基)-脲(化合物68)(150mg,0.40mmol)、N,N-二异丙基乙胺(77.4mg,0.60mmol)溶解于10mlTHF中,冰浴下(0-5℃)滴加丙烯酰氯(43.4mg,0.48mmol)。滴毕,在冰浴下反应30分钟后,加水(30ml)淬灭,用乙酸乙酯萃取(50×2),有机相用饱和NaCl溶液洗涤两次,无水硫酸钠干燥,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷/甲醇)纯化,得80mg白色固体状的1-(2-丙烯酰基-5-叔丁基-2H-吡唑-3-基)-3-(4-咪唑-1-基-苯基)-脲。
1HNMR(DMSO-d6,400MHz)δ:10.36(s,1H),10.13(s,1H),8.52(s,1H),7.77-7.79(m,1H),7.72-7.75(d,2H),7.61-7.63(d,2H),7.58-7.60(m,1H),7.48-7.55(q,1H),7.31-7.34(m,2H),6.72(s,1H),6.61-6.66(m,1H),6.20-6.23(m,1H),1.29(s,9H)。
LC-MS(ESI):429.1(M+H)+
实施例113
3-氨基-5-甲基吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺(化合物113)的制备
Figure PCTCN2017076265-appb-000154
在实施例64中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.91(s,1H),8.53(s,1H),7.92-7.94(d,2H),7.77-7.79(m,1H),7.62-7.65(d,2H),7.59-7.61(m,1H),7.31-7.34(m,2H),5.71(d,1H),5.34(s,2H),2.48(d,3H)。
LC-MS(ESI):333.1(M+H)+
实施例114
5-氨基-3-环丙基吡唑-1-甲酸(4-苯并咪唑-1-基-苯基)-酰胺(化合物114)的制备
Figure PCTCN2017076265-appb-000155
在实施例66中同步合成。
1HNMR(DMSO-d6,400MHz)δ:10.07(s,1H),8.54(s,1H),7.94-7.96(d,2H),7.77-7.79(m,1H),7.65-7.67(d,2H),7.61-7.63(m,1H),7.31-7.34(m,2H),6.46(s,2H),5.06(s,1H),1.83-1.88(m,1H),0.88-0.93(m,2H),0.70-0.74(m,2H)。
LC-MS(ESI):359.1(M+H)+
实施例115
5-氨基-3-三氟甲基吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺(化合物115)的制备
Figure PCTCN2017076265-appb-000156
在实施例67中同步合成。
1HNMR(DMSO-d6,400MHz)δ:10.40(s,1H),8.55(s,1H),7.92-7.95(d,2H),7.77-7.79(m,1H),7.68-7.71(d,2H),7.62-7.64(m,1H),7.30-7.37(m,2H),6.90(s,2H),5.76(s,1H)。
LC-MS(ESI):387.0(M+H)+
实施例116
5-氨基-3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺(化合物116)的制备
Figure PCTCN2017076265-appb-000157
在实施例68中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.80(s,1H),8.56(s,1H),7.91-7.94(d,2H),7.77-7.79(m,1H),7.68-7.70(d,2H),7.62-7.64(m,1H),7.31-7.35(m,2H),6.42(s,2H),5.32(s,1H),1.27(s,9H)。
LC-MS(ESI):375.0(M+H)+
实施例117
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-己氧基-苯并咪唑-1-基)-苯基]-酰胺(化合物117)的制备
Figure PCTCN2017076265-appb-000158
在实施例73中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.48(s,1H),7.90-7.92(d,2H),7.65-7.67(d,2H),7.49-7.51(d,1H),7.28-7.29(d,1H),6.94-6.96(dd,1H),6.41(s,2H),5.32(s,1H),4.01-4.04(t,2H),1.71-1.76(m,2H),1.43-1.47(m,2H),1.31-1.35(m,4H),1.27(s,9H),0.87-0.91(t,3H)。
LC-MS(ESI):475.2(M+H)+
实施例118
5-氨基-3-叔丁基-吡唑-1-羧酸(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-酰胺(化合物118)的制备
Figure PCTCN2017076265-appb-000159
在实施例92中同步合成。
1HNMR(CDCl3-d,400MHz)δ:9.28(s,1H),8.06(s,1H),7.78-7.80(d,2H),7.49-7.52(d,2H),7.39-7.41(d,1H),7.34-7.35(d,1H),6.97-7.00(dd,1H),5.44(s,2H),4.10-4.13(t,2H),2.50-2.70(m,10H),2.37(s,3H),2.01-2.08(m,2H),1.31(s,9H)。
LC-MS(ESI):531.2(M+H)+
实施例119
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺(化合物119)的制备
Figure PCTCN2017076265-appb-000160
在实施例89中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.49(s,1H),7.90-7.92(d,2H),7.65-7.67(d,2H),7.50-7.52(d,1H),7.32-7.33(d,1H),6.96-6.98(dd,1H),6.42(s,2H),5.32(s,1H),4.15-4.17(t,2H),3.59-3.61(t,4H),2.73-2.76(t,2H),2.49-2.51(m,4H),1.27(s,9H)。
LC-MS(ESI):504.2(M+H)+
实施例120
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺(化合物120)的制备
Figure PCTCN2017076265-appb-000161
在实施例77中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.49(s,1H),7.90-7.92(d,2H),7.65-7.68(d,2H),7.51-7.53(d,1H),7.31-7.32(d,1H),6.96-6.99(dd,1H),6.42(s,2H),5.32(s,1H),4.15-4.17(m,2H),3.69-3.71(m,2H),3.33(s,3H),1.27(s,9H)。
LC-MS(ESI):449.2(M+H)+
实施例121
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺(化合物121)的制备
Figure PCTCN2017076265-appb-000162
在实施例79中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.49(s,1H),7.90-7.92(d,2H),7.65-7.68(d,2H),7.51-7.53(d,1H),7.30(d,1H),6.97-6.99(dd,1H),6.41(s,2H),5.32(s,1H),4.89-4.92(t,1H),4.04-4.06(t,2H),3.74-3.78(m,2H),1.27(s,9H)。
LC-MS(ESI):435.2(M+H)+
实施例122
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-酰胺(化合物122)的制备
Figure PCTCN2017076265-appb-000163
在实施例87中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.48(s,1H),7.89-7.91(d,2H),7.65-7.67(d,2H),7.50-7.52(d,1H),7.28-7.29(d,1H),6.96-6.98(dd,1H),6.41(s,2H),5.32(s,1H),3.89-3.98(m,3H),3.63-3.69(m,1H),3.34-3.42(m,1H),1.82-1.84(m,1H),1.67-1.70(m,1H),1.46-1.54(m,4H),1.26(s,9H)。
LC-MS(ESI):489.2(M+H)+
实施例123
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-酰胺(化合物123)的制备
Figure PCTCN2017076265-appb-000164
在实施例86中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.49(s,1H),7.90-7.92(d,2H),7.65-7.68(d,2H),7.50-7.53(d,1H),7.31-7.32(d,1H),6.96-6.99(dd,1H),6.41(s,2H),5.32(s,1H),4.18-4.20(m,1H),3.98-4.02(m,2H),3.78-3.82(m,1H),3.69-3.71(m,1H),1.67-2.07(m,4H),1.27(s,9H)。
LC-MS(ESI):475.1(M+H)+
实施例124
(1-{4-[(5-氨基-3-叔丁基-吡唑-1-羰基)-氨基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯(化合物124)的制备
Figure PCTCN2017076265-appb-000165
在实施例88中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.79(s,1H),8.50(s,1H),7.90-7.92(d,2H),7.65-7.67(d,2H),7.52-7.54(d,1H),7.28(d,1H),6.99-7.02(dd,1H),6.41(s,2H),5.32(s,1H),4.84(s,2H),4.16-4.21(q,2H),1.27(s,9H),1.21-1.25(t,3H)。
LC-MS(ESI):477.1(M+H)+
实施例125
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-氟-苯并咪唑-1-基)-苯基]-酰胺(化合物125)的制备
Figure PCTCN2017076265-appb-000166
在实施例69中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.81(s,1H),8.61(s,1H),7.91-7.93(d,2H),7.67-7.69(d,2H),7.58-7.64(m,2H),7.19-7.25(m,1H),6.41(s,2H),5.33(s,1H),1.26(s,9H)。
LC-MS(ESI):393.2(M+H)+
实施例126
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-酰胺(化合物126)的制备
Figure PCTCN2017076265-appb-000167
在实施例70中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.83(s,1H),8.77(s,1H),8.17(d,1H),7.94-7.96(d,2H),7.80-7.82(d,1H),7.71-7.73(d,2H),7.66-7.68(dd,1H),6.41(s,2H),5.33(s,1H),1.27(s,9H)。
LC-MS(ESI):443.1(M+H)+
实施例127
4-(1-{4-[(5-氨基-3-叔丁基-吡唑-1-羰基)-氨基]-苯基}-1H-苯并咪唑-5-基氧基)-吡啶-2-羧酸甲基酰胺(化合物127)的制备
Figure PCTCN2017076265-appb-000168
在实施例96中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.82(s,1H),8.78-8.79(m,1H),8.68(s,1H),8.50-8.52(d,1H),7.93-7.95(d,2H),7.72-7.75(m,3H),7.67(d,1H),7.39-7.40(d,1H),7.18-7.23(m,2H),6.42(s,2H),5.33(s,1H),2.77-2.79(d,3H),1.27(s,9H)。
LC-MS(ESI):525.1(M+H)+
实施例128
5-氨基-3-叔丁基-吡唑-1-羧酸{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺(化合物128)的制备
Figure PCTCN2017076265-appb-000169
在实施例98中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.80(s,1H),8.45(s,1H),7.91-7.93(d,2H,),7.68-7.70(m,3H),7.08(d,1H),6.95-6.98(dd,1H),6.42(s,2H),5.33(s,1H),4.13-4.15(m,2H),3.66-3.68(m,2H),3.31(s,3H),1.27(s,9H)。
LC-MS(ESI):ESI 449.1(M+H)+
实施例129
5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-酰胺(化合物129)的制备
Figure PCTCN2017076265-appb-000170
在实施例99中同步合成。
1HNMR(DMSO-d6,400MHz)δ:9.78(s,1H),8.33(s,1H),7.90-7.92(d,2H,),7.67-7.69(d,2H),7.32(s,1H),7.08(s,1H),6.41(s,2H),5.32(s,1H),3.83(s,3H),3.81(s,3H),1.27(s,9H)。
LC-MS(ESI):ESI 435.1(M+H)+
实施例130
3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺(化合物130)的制备
Figure PCTCN2017076265-appb-000171
与实施例68的制备方法相同,除了用3-(叔丁基)-1H-吡唑代替步骤2中的3-叔丁基-吡唑-5-胺,得到3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺。
1HNMR(CDCl3-d,400MHz)δ:9.26(s,1H),8.23-8.24(d,1H),8.14(s,1H),7.89-7.92(m,1H),7.86-7.88(d,2H),7.54-7.57(m,3H),7.36-7.38(m,2H),6.40-6.41(d,1H),1.39(s,9H)。
LC-MS(ESI):360.1(M+H)+
实施例131
1-(4-苯并咪唑-1-基-苯基)-3-(3,4-二甲基-异噁唑-5-基)-脲(化合物131)的制备
Figure PCTCN2017076265-appb-000172
步骤1:(3,4-二甲基-异噁唑-5-基)-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3中的制备方法相同,除了用3,4-二甲基-5-氨基异噁唑代替3-氨基异噁唑,得到(3,4-二甲基-异噁唑-5-基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(3,4-二甲基-异噁唑-5-基)-脲
与实施例1的步骤4中的制备方法相同,除了用(3,4-二甲基-异噁唑-5-基)-氨基甲酸苯基酯代替异噁唑-3-基-氨基甲酸苯基酯,得到1-(4-苯并咪唑-1-基-苯基)-3-(3,4-二甲基-异噁唑-5-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:9.22(s,1H),9.18(s,1H),8.50(s,1H),7.76-7.78(m,1H),7.69-7.71(d,2H),7.57-7.61(m,3H),7.30-7.33(m,2H),2.16(s,3H),1.85(s,3H)。
LC-MS(ESI):348.1(M+H)+
实施例132
1-(4-苯并咪唑-1-基-苯基)-3-(3-异丙基-异噁唑-5-基)-脲(化合物132)的制备
Figure PCTCN2017076265-appb-000173
步骤1:(3-异丙基-异噁唑-5-基)-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3中的制备方法相同,除了用3-异丙基-5-氨基异噁唑代替3-氨基异噁唑,得到(3-异丙基-异噁唑-5-基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(3-异丙基-异噁唑-5-基)-脲
与实施例1的步骤4中的制备方法相同,除了用(3-异丙基-异噁唑-5-基)-氨基甲酸苯基酯代替异噁唑-3-基-氨基甲酸苯基酯,得到1-(4-苯并咪唑-1-基-苯基)-3-(3-异丙基-异噁唑-5-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:10.21(s,1H),9.13(s,1H),8.51(s,1H),7.76-7.78(m,1H),7.70-7.72(d,2H),7.61-7.63(d,2H),7.57-7.59(m,1H),7.28-7.36(m,2H),6.05(s,1H),2.90-2.96(m,1H),1.21-1.23(d,6H)。
LC-MS(ESI):362.1(M+H)+
实施例133
1-(4-苯并咪唑-1-基-苯基)-3-(3-叔丁基-异噁唑-5-基)-脲(化合物133)的制备
Figure PCTCN2017076265-appb-000174
步骤1:(3-叔丁基-异噁唑-5-基)-氨基甲酸苯基酯(活泼酯)的制备
与实施例1的步骤3中的制备方法相同,除了用3-叔丁基-5-氨基异噁唑代替3-氨基异噁唑,得到(3-叔丁基-异噁唑-5-基)-氨基甲酸苯基酯。
步骤2:1-(4-苯并咪唑-1-基-苯基)-3-(3-叔丁基-异噁唑-5-基)-脲的制备
与实施例1的步骤4中的制备方法相同,除了用(3-叔丁基-异噁唑-5-基)-氨基甲酸苯基酯代替异噁唑-3-基-氨基甲酸苯基酯,得到1-(4-苯并咪唑-1-基-苯基)-3-(3-叔丁基-异噁唑-5-基)-脲。
1HNMR(DMSO-d6,400MHz)δ:10.21(s,1H),9.12(s,1H),8.51(s,1H),7.76-7.79(m,1H),7.70-7.73(d,2H),7.61-7.63(d,2H),7.57-7.60(m,1H),7.29-7.35(m,2H),6.10(s,1H),1.27(s,9H)。
LC-MS(ESI):376.1(M+H)+
实施例134化合物成盐实验
(1)称取本发明化合物100mg-1000mg于50ml单口瓶中;
(2)加入2-12ml甲醇室温搅拌,为浑浊分散体系;
(3)称取3个当量的对应的酸溶解于少量甲醇中滴加其中,滴加过程中体系溶清;
(4)继续室温搅拌2h;
(5)后处理:
1)如有固体析出,加10倍甲醇体积量的乙酸乙酯搅拌30min,抽滤,所得固体用乙酸乙酯洗涤,干燥得产品。
2)如没有固体析出,反应体系浓缩得粗品,粗品溶于少量甲醇中(溶清为限),滴加10倍甲醇体积量的乙酸乙酯或甲基叔丁基醚析出固体,搅拌1h-2h,抽滤,所得固体用乙酸乙酯洗涤,干燥得产品。
3)如没有固体析出,反应体系浓缩得粗品,粗品溶于少量甲醇中(溶清为限),滴加10倍甲醇体积量的乙酸乙酯或甲基叔丁基醚,没有固体析出或者析出固体性状不好(粘稠),则继续浓缩,浓缩完全后油泵抽气30min-1h,所得粗品加乙酸乙酯或甲基叔丁基醚后慢慢碾压成颗粒状,打浆成均匀分散体系,加1/20倍乙酸乙酯体积量的甲醇,继续搅拌30min,抽滤,所得固体用乙酸乙酯洗涤,干燥得产品。
实施例135化合物水溶性和稳定性实验
(1)水溶性实验
称取研成细粉的供试品,于25℃±2℃一定容量的水中,每隔五分钟强力振摇30秒,观察30分钟内的溶解情况。如果无目视可见的溶质颗粒,即视为完全溶解。易溶系指供试品1g能在水1ml~10ml中溶解;溶解系指供试品1g能在水10ml~30ml中溶解;略溶系指供试品1g能在水30ml~100ml中溶解;微溶系指供试品1g能在水100ml~1000ml中溶解;几乎不溶或不溶系指供试品1g在溶剂10000ml中不能完全溶解。
(2)稳定性实验
1)高温试验
供试品开口置于综合药物稳定试验箱中(永生仪器),于60℃放置10天,于第5天和第10天取样,按药物稳定性考察指标进行检测。
2)高湿度试验
供试品开口置于综合药物稳定试验箱中(永生仪器),在25℃相对湿度90%±5%条件下放置10天,于第5天和第10天取样,按药物稳定性考察指标进行检测。
3)强光照试验
供试品开口置于综合药物稳定试验箱中(永生仪器),在4500LX±500LX光照条件下放置10天,于第5天和第10天取样,按药物稳定性考察指标进行检测。
(3)HPLC检测
取供试品适量,用乙腈-水(1∶1)溶解,并定量稀释制成约0.3mg/ml的溶液,取20ul注入液相色谱仪(安捷伦1260Infinity),记录色谱图,按百分面积法计算。流动相为磷酸盐缓冲液和乙腈水溶液,梯度洗脱。
表1为本发明化合物成盐后水溶性及稳定性实验结果。
表1本发明化合物盐的水溶性和稳定性实验结果
Figure PCTCN2017076265-appb-000175
*nd:无测定
试验例1
本发明化合物对表达FLT3野生型和内部串联重复(ITD)突变型白血病细胞系 体外50%生长抑制活性(GI50)浓度范围的测定
实验材料与方法
1、细胞系及细胞培养
肿瘤细胞系是研究肿瘤体外生长抑制的有效细胞模型。本发明选择具有代表性的肿瘤细胞系应用于本发明化合物细胞生长抑制活性的测定。所有使用的细胞系分别来源于ATCC、DSMZ和中科院细胞库。细胞培养条件与方法按每种细胞系要求进行。每次体外培养不超过3次传代,根据需要,可对细胞系进行单克隆纯化与鉴定。
细胞培养基分别选用RPMI1640(Gibco)、MEM(Gibco)、McCOY′S5A(Gibco)、IMDM(Gibco),加入5-20%胎牛血清(Gibco)、1%双抗、2mM谷氨酰胺或者1mM丙酮酸钠。
(1)表达FLT3野生型(WT)和内部串联重复(ITD)突变型白血病细胞系
人急性淋巴髓单核细胞白血病细胞系MV4-11(FLT3ITD+/+突变型,ATCC)用1×IMDM加10%FBS的完全培养基培养。人急性髓系白血病细胞系MOLM-13(FLT3ITD-/+突变型,DSMZ)、人急性淋巴白血病细胞系RS4;11(FLT3野生型,ATCC)、人慢性粒细胞白血病细胞系K562(FLT3表达阴性,BCR-ABL融合蛋白表达阳性,ATCC)、人早幼粒白血病细胞细胞系HL-60(FLT3野生型,ATCC)、人B淋巴细胞白血病细胞系RAMOS(Brutons酪蛋白激酶表达阳性,ATCC)、人急性髓细胞白血病细胞系Kasumi-1(c-Kit N822K突变型,ATCC)、人急性单核细胞白血病细胞株U937(FLT3野生型,ATCC)、人急性髓细胞性白血病细胞系OCI-AML3(NPMc+突变型,ATCC)、和人急性髓细胞白血病细胞系KG-1(表达FGFR1OP2-FGFR1融合蛋白,ATCC),均用1×RPMI1640加10%FBS的完全培养基培养。
(2)EGFR表达细胞系
人表皮癌细胞系A431(EGFR野生型/高表达,中科院上海细胞库);人肺癌细胞系NCI-H292(EGFR野生型,中科院上海细胞库);非小细胞肺癌细胞PC-9(ATCC)和HCC827细胞(中科院上海细胞库)为EGFR Exon19(E746-A750)缺失型突变细胞系;人非小细胞肺腺癌细胞NCI-H1975,表达EGFR L858R/T790M双突变(ATCC),第一代EGFR抑制剂抗性。以上细胞用1×RPMI1640加10%FBS的完全培养基培养。
(3)HER2/ErbB2基因扩增/高表达或突变的肿瘤细胞系
人胃癌细胞系NCI-N87(HER2/ErbB2amp,ATCC)、人乳腺癌细胞株HCC1954(HER2/ErbB2amp,ATCC)和ZR-75-30(HER2/ErbB2amp,ATCC)、 人腺癌细胞系AU565(HER2/ErbB2amp,ATCC)、人肺鳞状细胞癌细胞系NCI-H2170(HER2/ErbB2amp,ATCC)、人支气管肺泡腺癌细胞系NCI-H1781(HER2/ErbB2Ins G776V,C突变型,ATCC)分别用1×RPMI1640完全培养基(10%FBS)培养;人肺腺癌细胞系Calu-3(HER2/ErbB2amp,ATCC)培养基为1×MEM、10%FBS、1%双抗、1%NEAA(Gibco)、2mM谷氨酰胺和1mM丙酮酸钠。人乳腺癌细胞系SK-BR-3(HER2/ErbB2amp,ATCC)用McCOY′S5A完全培养基(10%FBS)培养。
(4)c-MET酪氨酸蛋白激酶基因扩增/过表达的肿瘤细胞系
人胃癌细胞系MKN-45(c-MET amp,ATCC)和非小细胞肺癌细胞系NCI-H1993(c-MET amp,ATCC)分别用1×RPMI1640加10%FBS的完全培养基培养。
(5)表达ALK融合蛋白和ALK基因突变的肿瘤细胞系
人非小细胞肺癌细胞系NCI-H2228(表达EML4-ALK融合基因,ATCC)和人间变性大细胞淋巴瘤细胞系Karpas-299(表达NPM-ALK融合基因,ATCC)分别用1xRPMI1640(10%FBS)完全培养基培养。神经母细胞瘤细胞SH-SY5Y(表达ALK F1174L突变蛋白,中科院上海细胞库)用MEM完全培养基(1xMEM,10%FBS,1%NEAA,1mM丙酮酸钠)培养。
(6)FGFR1基因扩增/高表达的肿瘤细胞系
人非小细胞肺癌细胞NCI-H1581(FGFR1amp,ATCC)是FGFR1基因扩增/过表达肿瘤细胞系,用1xRPMI1640(10%FBS)完全培养基培养。
(7)表达RAS和RAF基因突变的肿瘤细胞系
人大细胞肺癌细胞NCI-H460(KRAS G61H突变型,ATCC)、人非小细胞肺癌细胞株H1299(NRAS Q61K突变型,ATCC)、人黑色素瘤细胞系A375(BRAF V600E突变型,中科院上海细胞库)、人结肠癌细胞株HCT116(KRAS G13D突变型,中科院上海细胞库)用1xRPMI1640(10%FBS)完全培养基培养。人非小细胞肺癌细胞A549(KRAS G12S突变型,中科院上海细胞库)用1×Ham’S F12K,加10%FBS、1%双抗、2mM谷氨酰胺的完全培养基培养。
2、药物处理
贴壁细胞用0.25%胰酶-EDTA(Gibco)消化。悬浮培养细胞直接离心收集(1700rpm,3分钟),弃上清,计数细胞(
Figure PCTCN2017076265-appb-000176
自动细胞计数仪,Invitrogen)。根据每种细胞生长周期,配制不同的细胞浓度(每毫升5-10×104细胞),接种到96孔板(Corning),每孔100微升,37℃、5%CO2培养过夜。第二天,加入待测化合物到培养细胞中(平行2孔),溶剂对照孔加入相同体积溶剂,其中溶剂终浓度小于千分之一。细胞继续培养3-5天,MTT测定。待测本发明化合物与对照化合物分别用DMSO(Sigma)溶解,化合物纯度达98%以上。化合物贮存浓度为10mM,-20℃保存,使用前对倍或者10倍系列稀释。
本发明选用FLT3-ITD高特异性化合物AC220做为对照化合物,按照原研公司制备方法由本公司合成(Chao Q等人,Identification of N-(5-tert-butyl-isoxazol-3-yl)-N′-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}ureadihydrochloride(AC220),a uniquely potent,selective,and efficacious FMS-like tyrosine kinase-3(FLT3)inhibitor.J Med Chem.2009;52(23):7808-16)。
3、MTT检测及GI50计算
MTT检测试剂为Dojindo CCK8试剂盒,酶标测定仪为THERMO MULTISKAN FC仪。
将贴壁细胞培养基吸出,立即加入新配制的含10%CCK8的完全培养基(5%FBS),每孔100ul。悬浮细胞可直接加入CCK8试剂,终浓度为10%,继续培养1-4小时,当溶剂对照孔呈现暗黄色时,测OD450nm光吸收值,按以下公式计算细胞生长率,
细胞生长率(%)=100*(T-T0)/(C-T0)
T=药物处理细胞孔光密度值-空白对照孔光密度值;T0=药物处理前细胞孔光密度值-空白对照孔光密度值;C=溶剂对照细胞孔光密度-空白对照孔光密度值。通过药物浓度与细胞生长率曲线,计算细胞生长50%抑制的药物浓度即GI50。试验重复进行1~3次,并对数据进行生物学统计分析。
4、实验结果
表2总结本发明化合物对表达FLT3-ITD突变型和野生型白血病细胞系生长抑制活性GI50浓度范围的测定结果。GI50值越小,细胞生长抑制活性越强。如果化合物对FLT3-ITD表达细胞(MV4-11和MOLM-13)生长抑制活性强(即GI50低),而对FLT3野生型高表达(如RS4;11)或低表达或无表达(如K562)细胞的生长抑制弱或相对无作用时(即GI50浓度高),说明该类化合物对FLT3-ITD激活型突变的选择性高,具有治疗FLT3-ITD相关疾病的潜在开发价值。
Figure PCTCN2017076265-appb-000177
Figure PCTCN2017076265-appb-000178
Figure PCTCN2017076265-appb-000179
Figure PCTCN2017076265-appb-000180
Figure PCTCN2017076265-appb-000181
Figure PCTCN2017076265-appb-000182
由上表2可知,本发明化合物能够有效的抑制FLT3-ITD表达阳性白血病细胞MV4-11和MOLM-13的生长(或诱导细胞凋亡),其GI50值可达亚纳摩尔。与对照化合物AC220相比,本发明化合物(如化合物11、71、77、79、81和89)对MV4-11和MOLM-13细胞的生长呈现更强抑制活性。进一步地,本发明化合物对FLT3野生型高表达细胞(RSV4;11)或正常表达细胞(HL-60、Ramos、U937、Kasumi-1、KG-1和OCI-AML3)或无表达细胞(K562)的生长呈现相对较弱或无抑制作用。这些结果说明本发明化合物对表达FLT3-ITD激活型突变的白血病细胞具有高选择性和高活性,是一种潜在的新型高活性FLT3-ITD选择性抑制剂。
试验例2
本发明化合物对不同肿瘤细胞体外50%生长抑制活性值(GI50)的测定
选择本发明化合物4、11、12、16、27、68、71、77、79、89和116作为代表例进行不同类型的肿瘤细胞生长抑制试验。化合物以5,000nM为起始浓度,对倍或10倍系列稀释至0.01nM,按照GI50测定与计算方法获得每种测试化合物的GI50值。试验重复进行1~3次,并对数据进行生物学统计分析,结果见表3。
Figure PCTCN2017076265-appb-000183
上述表3结果显示本发明化合物12、16、27、68、71、77、89和116对FLT3-ITD表达阳性细胞系MV4-11具有高的生长抑制活性,GI50值为亚纳摩尔范围(0.039~0.8nM),但对FLT3野生型高表达细胞RS4;11需要相对高的浓度才呈现明显的抑制作用,GI50在微摩尔范围。测试化合物4、11和79对表达FLT3-ITD阳性细胞系MV4-11具有强的生长抑制作用,但对FLT3野生型高表达细胞RS4;11同样呈现一定的生长抑制活性,但需要相对较高的抑制浓度,GI50值在50~800nM范围。表3结果进一步显示本发明化合物(12、16、27、68、71、77、89和116)对EGFR、HER2/ERBB2、FGFR1、RAS、BRAF、cMET和ALK基因异常表达的肿瘤细胞生长无明显抑制作用,GI50值在微摩尔范围,是FLT3-ITD基因表达阳性细胞MV4-11生长抑制浓度GI50的1000倍以上。
试验例3
肿瘤细胞体内生长抑制实验
免疫缺陷小鼠的异种移植是测试化合物动物体内抗肿瘤活性的有效模型。Bab/c免疫缺陷小鼠是最常用的肿瘤细胞异种移植动物之一。为了测试本发明化合物是否能够有效抑制FLT3-ITD表达阳性白血病细胞的体内生长,本发明选用MV4-11细胞Bab/c裸鼠肿瘤模型进行测试。将生长对数期的MV4-11细胞收集于50ml离心管(Corning),1700rm离心3分钟,弃上清。用50ml 1xRPMI1640无血清培养基悬浮细胞,离心,弃上清,再悬浮,细胞计数(
Figure PCTCN2017076265-appb-000184
自动细胞计数仪,Invitrogen),配制成10x107细胞/毫升,置冰上,皮下接种10x106(0.2ml)细胞于6~8周龄(重20克左右)雌性Bab/c裸鼠右侧背部(购于上海西普尔-必凯实验动物有限公司,上海中医药大学动物中心饲养,上海中医药大学伦理委员会批准)。当肿瘤块生长到大小在100-200mm3体积时,随机分组,耳扎标记,称重。治疗组,每组3~6只裸鼠,药物分别用溶剂PTP液(30%聚乙二醇400,0.5%吐温-80,2.5%丙二醇)配制成不同药物浓度乳状混悬液,以0.1ml/每10克体重,每日灌胃一次,连续给药,药量范围为0.1~200mg/kg(本发明测试化合物纯度在99%以上,单杂不高于0.1%)。对照组,每组3~6只裸鼠,给相同体积(0.1ml/每10克体重)的溶剂PTP液,每周测量肿瘤3~4次,当对照组肿瘤平均体积达到1500mm3时,或给药到28天时,实验结束,称重肿瘤块和进行分子病理学分析。
抑瘤率=(1-治疗组相对肿瘤体积TRTV/对照组相对肿瘤体积CRTV)×100%。肿瘤体积按V=1/2a×b2计算,a为长,b为宽。根据测量的结果计算出相对肿瘤体积(relative tumor volume,RTV),计算公式为:RTV=Vt/V0。其中V0为分笼给药时(即d0)测量所得肿瘤体积,Vt为每次测量时的肿瘤体积。具体结果见表4。
表4本发明化合物对肿瘤细胞体内生长的抑瘤率
Figure PCTCN2017076265-appb-000185
*nd:无测定
上述表4结果显示本发明化合物对MV4-11细胞体内肿瘤呈现浓度和时间依赖性的生长抑制活性。化合物68在剂量≥25mg/kg时,给药后第6天,肿瘤生长抑制率已达80%以上,第28天时,肿瘤完全消失。化合物71的肿瘤生长抑制活性强于化合物68,剂量在1~2.5mg/kg时仍呈现明显的肿瘤生长抑制活性。同时与溶剂对照组相比,化合物68和71给药组在剂量≤25mg/kg时,对裸鼠体重无明显影响,剂量≥50mg/kg时,裸鼠体重相对减少10~20%。测试化合物77和16呈现更强的MV4-11细胞体内肿瘤生长抑制活性,其药物剂量分别为2.5mg/kg时,给药第15天,肿瘤完全消失,与溶剂对照组相比,对裸鼠体重无明显影响。
试验例4
hERG试验
以本发明化合物77为代表例进行全细胞模式hERG试验,30μM奎尼丁(Sigma)为阳性对照。
具体方法为,过表达hERG钾离子通道HEK-293细胞(科瑞斯生物),在37℃ 5%CO2培养箱中培养。培养基为DMEM加15%胎牛血清和1%青霉素-链霉素。将稳转的细胞接种于玻片上,细胞密度低于50%,培养过夜。将实验用细胞转移到一个嵌于倒置显微镜平台(Diaphot,Nikon)的约1mL的浴槽中,灌流细胞外液,灌流速度为2.7mL/min。稳定5分钟细胞沉淀后即可开始实验。采用HEKA EPC-10膜片钳放大器和PATCHMASTER采集系统记录膜电流(HEKA Instruments Inc.,D-67466Lambrecht/Pfalz Germany)。所有实验均在室温(22-24℃)完成。实验中使用P-97微电极拉制仪(Sutter Instrument Company,One Digital Drive,Novato,CA 94949)拉直电极(BF150-110-10)。电极内径为1-1.5mm,充满内液后的入水电阻为2-4MΩ。hERG钾通道的电生理刺激方案:首先将膜电压钳制在-80mV,给予细胞持续2s、+20mV电压刺激,激活hERG钾通道,再复极化至-50mV、持续5s,产生外向尾电流,刺激频率每15s一次。电流值为尾电流的峰值。参数分析通过测量对照组与药物处理组的电流最大值,计算处理组最大电流值所占对照组最大电流值的比率(Mean±SE),评估待测化合物在测试浓度下对hERG钾离子通道的作用效果。实验数据采用Origin 8.5(OriginLab Corporation,Northampton,MA)软件进行分析和统计。不同浓度的化合物77阻断hERG钾通道后的电流比例结果见表5。
表5
Figure PCTCN2017076265-appb-000186
结果显示本发明化合物77在测试浓度范围内对hERG钾通道有一定的阻断作用,IC50为18.2μM。
试验例5
本发明化合物在大鼠肝细胞中的稳定性研究
以本发明化合物77为代表例进行在大鼠肝细胞的稳定性实验。
具体方法为,将大鼠肝细胞William’s Medium E混合反应液(2*106cells/mL,BD Gentest)置于二氧化碳培养箱中,将一定体积的受试物或对照品加入反应体系启动反应,最终反应体系中受试物或阳性对照睾酮的浓度均为1μM。分别在反应后0、5、15、30、45、60和120min取样至新的离心管(每个时间点平行准备3份),立即加入3倍体积含内标的冰甲醇溶液终止反应。然后15000rpm离心5分钟沉淀蛋白后,取100μL上清液至自动进样小瓶中进行LC-MS/MS分析 (Waters公司ACQUITY UPLC,美国应用生物系统公司质谱仪API 400)。计算各时间点化合物77平均峰面积与内标平均峰面积的比值,并计算该比值相对于0时间点的百分率,并以此百分率的变化表示化合物77代谢稳定性。数据处理系统为美国应用生物系统公司Analyst 1.5软件。
结果显示化合物77在大鼠肝细胞中的半衰期大于120min。
试验例6
本发明化合物在大鼠肝微粒体中的代谢稳定性研究
以本发明化合物77为代表例进行在大鼠肝微粒体中的稳定性实验。
具体方法为,按照一定比例,配制含有还原性辅酶II(1mg/ml,Roche 10621706001)、大鼠肝微粒体(0.5mg/ml,BD Gentest公司)、磷酸盐缓冲液(0.1M)和去离子水的混合反应液,然后将反应液置于37℃的水浴锅中预热2分钟。将一定体积的化合物77或对照品加入已配制好的混合反应液中启动反应。最终反应体系中化合物77或阳性对照咪达唑仑(中国药品生物制品检定所)的浓度均为2μM,以水代替还原性辅酶II作为阴性对照。分别在反应后0、10、15、30、45和60min从反应溶液中取样(50μl)至新的离心管,每个时间点平行准备2份,立即加入3倍体积含内标的冰甲醇溶液终止反应。然后15000rpm离心5分钟沉淀蛋白后,取100μL上清液至自动进样小瓶中进行LC-MS/MS分析(Waters公司ACQUITY UPLC,美国应用生物系统公司质谱仪API4000)。计算各时间点化合物77平均峰面积与内标平均峰面积的比值,并计算该比值相对于0时间点的百分率,并以此百分率的变化表示化合物77代谢稳定性。数据处理系统为美国应用生物系统公司Analyst1.5软件。
结果显示化合物77在大鼠肝微粒体稳定性的拟合曲线为y=-0.0031x-2.3407(R2=0.9454),消除速率常数为0.0031,半衰期为224min。
试验例7
本发明化合物与大鼠血浆蛋白的结合率试验
应用透析法评估本发明化合物77在大鼠血浆中浓度2μM时与血浆蛋白的结合率。
具体方法为,一定体积的化合物77供试品DMSO储备液(10mM)或阳性对照普萘洛尔(USP)加入到500μL空白血浆中,血浆中供试品终浓度为2μM,阳性对照终浓度为1μM,平行制备三份。300μL制备好的血浆样品加入到血浆室,缓冲液室加入500μL的透析缓冲液(磷酸盐缓冲液PBS中含有100mM磷酸钠和150mM氯化钠,PH 7.2)。密封,在37℃条件下100rpm振荡孵育4h。孵育结束后,从血浆室及缓冲液室分别取50μL样品,50μLPBS加入到血浆室样品中,50μL空白血浆加入到缓冲液室样品中。立即加入3倍体积含内标的甲醇溶液以沉 淀血浆蛋白。取离心后的上清液进行LC/MS/MS分析。根据以下公式计算血浆蛋白结合率:%游离态=(缓冲液室峰面积比/血浆室峰面积比)×100%,%结合态=100%-%游离态。化合物77及普萘洛尔在大鼠血浆蛋白结合率见表6。
表6化合物77及普萘洛尔大鼠血浆蛋白结合率
Figure PCTCN2017076265-appb-000187
结果显示,化合物77与大鼠血浆蛋白结合率为99.54%,阳性对照普萘洛尔在大鼠血浆中的蛋白结合率分别为90.11%。
试验例8
本发明化合物对P450酶抑制活性的测定
以本发明化合物77为代表例进行人肝微粒体8种细胞色素p450酶抑制反应实验。
主要反应体系和过程为,在37℃水浴条件下,受试化合物分别以7个浓度(0、0.25、0.5、5、10、25、50μM;体系中甲醇终浓度均为1%)与人肝微粒体(BD Gentest)(0.5mg/mL)和还原性辅酶II(Roche 10621706001)(1mg/mL)以及相应细胞色素p450酶的探针底物共孵育5~20分钟。其中,(1)CYP1A2的探针底物为非那西丁(Phenacetin,20μM)(Sigma),选择性抑制剂α-萘黄酮(alpha-naphthoflavone,2μM)(Sigma)平行检测作为阳性对照,共孵育20分钟。(2)CYP2b6的探针底物为安非他酮(Bupropion,40μM)(Sigma),选择性抑制剂噻氯匹定(Ticlopidine,10μM)(Sigma)平行检测作为阳性对照,共孵育20分钟。(3)CYP2C8的探针底物为阿莫地喹(Amodiaquine,1μM)(Sigma),选择性抑制剂孟鲁司特(Montelukast,10μM)(Sigma)平行检测作为阳性对照,共孵育20分钟。(4)CYP2C9的探针底物为双氯酚酸(Diclofenac, 2.5μM)(Sigma),选择性抑制剂磺胺苯吡唑(Sulphaphenazole,10μM)(Sigma)平行检测作为阳性对照,共孵育20分钟。(5)CYP2C19的探针底物为美芬妥因(Mephenytoin,40μM)(Sigma),选择性抑制剂噻氯匹定(Ticlopidine,10μM)(Sigma)平行检测作为阳性对照,共孵育20分钟。(6)CYP2D6的探针底物为右美沙芬(Dextromethorphan,5μM)(Sigma),选择性抑制剂奎尼丁(Quinidine,10μM)(Sigma)平行检测作为阳性对照,共孵育20分钟。(7)CYP3A4-TCYP3A4-M的探针底物分别为睾酮(Testosterone,5μM)(Sigma)和咪达唑仑(Midazolam,5μM)(Sigma),选择性抑制剂为酮康唑(Ketoconazole,1μM)(Sigma),平行检测作为阳性对照,共孵育5分钟。通过在上述反应体系中加入含有相应内标的甲醇终止反应,内标化合物为甲苯磺丁脲(Tolbutamide)(Sigma)。用LC/MS/MS测定相应代谢产物的浓度。数据分析采用加入化合物77后的标本代谢产物峰面积相对于溶剂对照标本的代谢产物峰面积的减少,用以计算IC50(50%抑制时的受试物浓度)。加入不同浓度化合物77及选择性抑制剂后CYP1A2、CYP2B6、CYP2C8、CYP2C9、CYP2C19、CYP2D6、CYP3A4-T和CYP3A4-M探针底物的代谢产物峰面积相对于溶剂对照峰面积百分比见表7。
表7不同浓度化合物77及阳性对照与溶剂对照峰面积平均百分比
Figure PCTCN2017076265-appb-000188
如表7所示,上述8种酶的阳性对照中代谢产物峰面积明显低于溶剂对照,提示酶活性被抑制。当化合物77浓度50μM时,未见对CYP3A4-T和CYP3A4-M具有明显抑制作用;CYP1A2,CYP2C19和CYP2D6半数抑制浓(IC50)≥50 μM,CYP2C8的半数抑制浓(IC50)大于或接近于50μM,CYP2B6的半数抑制浓度(IC50)为38.98μM,CYP2C9的半数抑制浓度(IC50)为41.56μM。
试验例9
化合物在Caco-2单层细胞模型中的双向跨膜转运及P-gp外排特性的研究
以本发明化合物77为代表例进行Caco-2细胞转运实验。
主要过程为:Caco-2细胞(ATCC,HTB-37)培养于高糖DMEM培养液(Hyclone)中,培养液中含有10%胎牛血清(GIBCO)和青链霉素(各100单位/mL)(Sigma),细胞在37℃,含5%CO2的培养箱中进行培养。转运实验中,将Caco-2细胞以2x105个/孔的密度接种于12孔Transwell培养板(Corning Costar,货号#3401,1.12cm2,0.4μm孔径)的滤膜上,接种后的Caco-2细胞将于21天后形成完整的细胞单层,包括P-gp外排转运体表达和跨上皮细胞电阻(TEER)形成,期间每隔一天更换一次培养液,Transwell滤膜的顶侧(Apical Side,A)和基底侧(Basolateral Side,B)的培养液容量分别为0.5mL和1.5mL。转运实验开始前(第21天),首先用预热的HBSS缓冲液(137mM NaCl,4.17mM NaHCO3,0.34mM Na2HPO4,5.37mM KCl,0.44mM KH2PO4,1.26mM CaCl2,0.49mM MgCl2,0.41mM MgSO4,5.55mM D-Glucose,10mM HEPES,pH 7.4)将Caco-2细胞单层洗3次,并置于37℃条件下孵育30分钟,然后用细胞电阻仪(Millicell-ERS2)测定TEER值来确认细胞单层的完整性和紧密型。当将Transwell滤膜A侧或者B侧的HBSS缓冲液替换为测试化合物后,转运实验即已开始,最后经过在37℃下孵育2小时,转运实验即终止,在Transwell滤膜的两侧各取出50μL样品溶液加入100μL含有内标化合物的乙腈并混匀,然后以13000rpm,4℃离心10min,最后吸取10μL(或特定体积)上清液用于LC-MS/MS样品分析。对于每一个化合物,A→B和B→A方向的转运实验均为三复管,即n=3。
LC-MS/MS样品分析:本研究中使用的高效液相色谱(HPLC)系统是由两个泵(岛津LC-20AD)及一个自动进样器(岛津SIL-20AC)组成。高效液相色谱系统通过串联与API4000三重四级质谱(Applied Biosystems)相连接,其中API4000三重四级质谱配备了一个电喷雾离子化(ESI)源。超纯度氮气用作气帘气、GAS1、辅助气(GAS2)及碰撞气,其流速分别为20L/h、55L/h、65L/h及6L/hr。离子源的雾化温度设定为500℃。数据通过Analyst 1.5软件进行采集。
数据分析:表观渗透系数(Papp)可以通过化合物透过Caco-2细胞单层的速度来计算,其数值大小与该化合物在体内的吸收相关,通过LC-MS/MS方法测定A侧和B侧的化合物浓度后,按照下列公式计算A→B或B→A方向的Papp值:
Papp(A→B)or Papp(B→A)=(dQ/dt)/(A*C0)=(C2h*V)/(t*πr2*C0)
上述公式中,dQ/dt为渗透速率,即在dt时间内透过的化合物量,C0为给药侧的药物初始浓度,A为细胞单层的表面积即膜面积。
在获得化合物的Papp(A→B)和Papp(B→A)值后,化合物的外排比(Efflux Ratio,ER)即可通过下列公式计算得到:
Efflux Ratio(ER)=Papp(B→A)/Papp(A→B)。
当外排比≥2时,可认为该化合物为外排转运体的底物。
表8 测试化合物77的Papp及溢出率ER值
Figure PCTCN2017076265-appb-000189
实验结论:(I)对照化合物Digoxin在Caco-2单层细胞模型中表现出较低的跨膜通透性(Papp(A→B)值小于1x10-6cm/s),作为P-gp外排底物其ER值为321.9,此结果证明了本研究的准确性。(2)化合物77在Caco-2单层细胞模型中表现出中等的跨膜通透性(Papp(A→B)介于1~10x10-6cm/s之间),以及P-gp外排转运体的底物(ER值在不加入P-gp抑制剂GF120918条件下为5.8,而在加入P-gp抑制剂GF120918条件下为1.1)。
试验例10
本发明化合物在大鼠体内药物代谢动力学的研究
以本发明化合物77为代表例进行大鼠体内药物代谢动力学实验。
具体方法为,挑选6只SD雄性大鼠(~200g,购于上海西普尔-必凯实验动物有限公司),按照实验设计表分为两组(表9),分别通过静脉和口服给药,给药当天给药1次,给药剂量分别为5mg/kg和10mg/kg。
药物配制方法为,取所需量供试品,加入到一定体积40%HP-β-CD水溶液中,加热搅拌溶解(温度25~45℃),之后再加入相同体积的去离子水,NaOH调节pH到5,配成所需药物浓度的20%HP-β-CD澄清水溶液。
表9 化合物77大鼠体内药物代谢动力学实验设计
Figure PCTCN2017076265-appb-000190
Figure PCTCN2017076265-appb-000191
*在口服给药前,所有动物禁食过夜(10-14小时),给药后4小时给食。
静脉给药组在给药前和给药后2min、5min、15min、30min、1h、2h、4h、6h、8h和24h从颈静脉采集血样;口服给药组在给药前和给药后5min、15min、30min、1h、2h、4h、6h、8h、10h和24h从颈静脉采集血样。血液样本采集后于30分钟之内离心分离血浆(离心条件:8000转/分钟,6分钟,2-8℃)。血浆样本在分析前存放于-80℃冰箱内。血浆样本进行LC-MS/MS方法开发和样品检测。
采用WinNonlin Professional v 5.2(Pharsight,USA)计算以下药代动力学参数:AUC(0-t)、AUC(0-∞)、T1/2、MRT(0-∞)、Cmax、Tmax、F。此外,生物利用度(F)将通过下面的公式进行计算。
Figure PCTCN2017076265-appb-000192
SD大鼠给予化合物77甲磺酸盐后的药代动力学参数见表10和表11。
表10 SD大鼠单次静脉给予化合物77甲磺酸盐的药代动力学参数
Figure PCTCN2017076265-appb-000193
表11 SD大鼠单次口服给予化合物77甲磺酸盐的药代动力学参数
Figure PCTCN2017076265-appb-000194
结果显示在SD大鼠静脉给予5mg/kg化合物77甲磺酸盐后的Cmax为18688ng/mL,AUC(0-t)为14409h*ng/mL;SD大鼠口服给予10mg/kg化合物77甲磺酸盐后的Cmax为1051ng/mL,AUC(0-t)为6771h*ng/mL,生物利用度为23.50%。
试验例11
本发明化合物对小鼠血液细胞的影响
以本发明化合物77为代表例进行小鼠长期口服毒理实验。
具体方法为,挑选健康6周龄ICR小鼠(购于上海西普尔-必凯实验动物有限公司),雌雄各半,随机分组,每组6只。试验组与对照组每日分别通过口服灌胃一次,连续给药到40天,每日观测与记录小鼠体重和生理病理特征。试验组药物剂量分别为500mg/kg、350mg/kg和250mg/kg的化合物77甲磺酸盐/20%HP-β-CD澄清水溶液(配制方法与试验例10相同),对照组为20%HP-β-CD水溶液。
结果显示化合物77甲磺酸盐连续给药到40天,500mg/kg、350mg/kg和250mg/kg 3个药物剂量的死亡率分别为66.7%、16.7%和0%。与对照组相比,其体重分别下降34.7%、23.1%和8.2%。血细胞参数分析发现化合物77甲磺酸盐除对网织红细胞呈现一定的影响外(下降10~30%),对其它血细胞数无显著影响。

Claims (23)

  1. 一种通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
    Figure PCTCN2017076265-appb-100001
    其中:
    X、Y各自独立地选自N或者C-BR1
    A、A1各自独立地选自N或者C-BR1
    W、Z各自独立地选自N或者C-BR1
    当R1不存在时,B相同或不同且各自独立地选自:氢、卤素、烷基、烯基、炔基、氰基、环烷基、杂环基、芳基、或杂芳基,所述烷基、烯基、炔基、环烷基、杂环基、芳基、或杂芳基任选进一步被一个或多个Q基团取代;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-;并且,R1相同或不同且各自独立地选自:氢、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基、-RuORx、-RuC(O)ORx、-RuN(Ry)(Rz)、-C(O)N(Ry)(Rz)、-RuS(O)nN(Ry)(Rz)、-RuS(O)nRx;所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氰基、羟基、烷基、烷氧基、羟烷基、羟烷氧基、酰胺基、环烷基、杂环基、芳基、卤代芳基、杂芳基、环烷基-杂芳基的一种或多种基团取代;R4选自氢、烷基、烯基和炔基,或者R4、R1与相连接的氮原子一起形成杂环基或杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基的一种或多种基团取代;
    当R2为氢时,G选自芳基、杂芳基或杂环基,所述芳基、杂芳基或杂环基任选进一步被选自卤素、烷基、烯基、炔基、烷氧基、羟基、氨基、酰基、环烷基、杂环基、芳基、杂芳基的一种或多种基团所取代,其中所述烷基、烯基、炔基、烷氧基、酰基、环烷基、杂环基、芳基或杂芳基任选进一步被选自卤素、烷基、卤代烷基、烯基、炔基、芳基、羟基、烷氧基、卤代烷氧基、环烷基、酯基和氰基的一种或多种基团所取代;或者,
    当R2不为氢时,G与R2与它们所连接的氮原子一起形成杂环基或杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、烷基、烯基、炔基、烷氧基、羟基、 氨基、环烷基、杂环基、芳基、和杂芳基的一种或多种基团所取代,其中所述烷基、烯基、炔基、烷氧基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、烷基、卤代烷基、烯基、炔基、芳基、羟基、烷氧基、卤代烷氧基、环烷基、酯基和氰基的一种或多种基团所取代;
    R3选自Q基团;
    Ru选自一个键、亚烷基、亚烯基、或亚炔基;
    Rx选自氢、烷基、羟烷基、卤代烷基、烯基、或者炔基;或者,
    -RuORx-中氧与相连接的Ru和Rx一起形成含氧的3-7元杂环,所述杂环任选被一种或多种Q基团取代;
    Ry和Rz各自独立地选自氢、烷基、烷氧基、烯基、炔基、环烷基、或者卤代烷基;或者,
    Ry和Rz与它们所连接的氮原子一起形成杂环基或者杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、卤代烷基、烷基、烯基和炔基的一种或多种基团所取代;
    Q选自氢、卤素、羟基、氨基、烷基、烷氧基、环烷基、烯基、炔基、氰基、芳基、杂环基或杂芳基,所述氨基、烷基、烷氧基、环烷基、烯基、炔基、芳基、杂环基或杂芳基任选进一步被选自羟基、卤素、烷基的一种或多种基团取代;
    n为0-2的整数。
  2. 根据权利要求1所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
    其中:
    X、Y、A、A1选自如下结构:
    Figure PCTCN2017076265-appb-100002
    R5 1、R5 2、R5 3、R5 4、R5 5、R5 6、R5 7、R5 8和R5 9各自独立地选自氢、卤素、羟基、烷基、烷氧基、烯基、炔基、-N(Ry)(Rz)、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烷氧基、烯基、炔基、-N(Ry)(Rz)、环烷基、杂环基、芳基或杂芳基任选进一步被选自卤素、羟基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、酯基的一种或多种基团所取代;
    R1、B、Ry、Rz如权利要求1中所定义。
  3. 根据权利要求1或2所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
    其中:
    X、Y、A、A1选自如下结构:
    Figure PCTCN2017076265-appb-100003
    R5 1、R5 2、R5 3、R5 4和R5 5各自独立地选自氢、卤素、烷基、烷氧基、-N(Ry)(Rz)、卤代烷基、卤代烷氧基;
    R1、B、Ry、Rz如权利要求1中所定义。
  4. 根据权利要求1至3中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,
    其中:
    X、Y、A、A1选自如下结构:
    Figure PCTCN2017076265-appb-100004
    R5 1、R5 2、R5 3各自独立地选自氢、卤素、烷基、烷氧基、-N(Ry)(Rz)、卤代烷基、卤代烷氧基;
    R1、B、Ry、Rz如权利要求1中所定义。
  5. 根据权利要求1至4中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    R5 1、R5 2、R5 3各自独立地选自氢、卤素、C1-C6烷基、C1-C6烷氧基、-N(Ry)(Rz)、C1-C6卤代烷基、C1-C6卤代烷氧基,其中Ry、Rz各自独立地选自氢、C1-C6烷基。
  6. 根据权利要求1至5中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,W、Z选自如下4种方式:
    a)W、Z为CQ;
    b)W、Z为N;
    c)W为CQ且Z为N;
    d)Z为CQ且W为N;
    其中,Q选自氢、卤素、羟基、氨基、氰基、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、C3-C7环烷基、C5-C7芳基、5至7元杂环基或5至7元杂芳基。
  7. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    R1不存在,B相同或不同且各自独立地选自:氢、卤素、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基。
  8. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:氢、C1-C10烷基,所述烷基任选进一步被选自卤素、氰基、羟基、C1-C6烷氧基、4~6元杂环基、C5~C7芳基、C5~C7卤代芳基、5~7元杂芳基、C3~C6环烷基-5~7元杂芳基的一种或多种基团取代,其中所述4~6元杂环基优选含氧或氮的4~6元杂环基;所述C5~C7芳基或C5~C7卤代芳基优选苯基或卤代苯基;
    R4如权利要求1中所定义。
  9. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-RuORx,其中Ru选自C1-C6亚烷基,Rx选自氢、C1-C6烷基、C1-C6羟烷基、C1-C6卤代烷基;
    R4如权利要求1中所定义。
  10. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-C(O)N(Ry)(Rz),其中Ry和Rz各自独立地 选自氢、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C3-C7环烷基;或者,
    Ry和Rz与它们所连接的氮原子一起形成5~7元杂环基或5~7元杂芳基,优选6元杂环基或6元杂芳基,更优选吗啉基、哌啶基、哌嗪基、吡啶基、嘧啶基,所述5~7元杂环基或5~7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基的一种或多种基团所取代;
    R4如权利要求1中所定义。
  11. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-RuN(Ry)(Rz),其中Ru选自C1-C6亚烷基;Ry和Rz各自独立地选自氢、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C3-C7环烷基;或者,
    Ry和Rz与它们所连接的氮原子一起形成5~7元杂环基或者5~7元杂芳基,优选吗啉基、哌啶基、哌嗪基、氮杂环庚烷基、吡啶基、嘧啶基,所述5~7元杂环基或5~7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基的一种或多种基团所取代;
    R4如权利要求1中所定义。
  12. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:-RuC(O)ORx,其中:Ru选自C1-C6亚烷基;Rx选自氢、C1-C6烷基、C1-C6羟烷基、C1-C6卤代烷基;
    R4如权利要求1中所定义。
  13. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R1不存在时,B选自氢、卤素、C1-C6烷基、C1-C6卤代烷基;
    当R1存在时,B相同或不同且各自独立地选自:-O-或-NR4-,优选为-O-;并且,R1相同或不同且各自独立地选自:5~7元芳基或5~7元杂芳基,优选噻二唑基、吡唑基、噁唑基、噁二唑基、咪唑基、三唑基、噻唑基、呋喃基、噻吩基、吡啶基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、咪唑基、四唑基、苯基、吡 啶基、嘧啶基,所述5~7元芳基或5~7元杂芳基任选进一步被选自C3~C6环烷基、5~7元杂环基、酰胺基的一种或多种基团取代;
    R4如权利要求1中所定义。
  14. 根据权利要求8至13中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    R4选自氢或C1-C6烷基,或者
    R4、R1与相连接的氮原子一起形成5~7元杂环基或5~7元杂芳基,优选哌啶基、哌嗪基、吗啉基、吡啶基、嘧啶基,所述5~7元杂环基或5~7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基的一种或多种基团取代。
  15. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    当R2为氢时,G选自C5~C7芳基、5至7元杂芳基或5至7元杂环基,优选
    Figure PCTCN2017076265-appb-100005
    所述C5~C7芳基、5至7元杂芳基或5至7元杂环基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、羟基、氨基、酰基、C3~C7环烷基、5~7元杂环基、C5~C7芳基、5~7元杂芳基的一种或多种基团所取代;所述C1-C6烷基、C1-C6烷氧基、C3~C7环烷基、5~7元杂环基、C5~C7芳基、5~7元杂芳基任选进一步被选自卤素、羟基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、酯基和氰基的一种或多种基团所取代;或者,
    当R2不为氢时,G与R2与它们所连接的氮原子一起形成5-7元杂环基或5-7元杂芳基,优选,吡咯基、吡唑基、咪唑基,所述5-7元杂环基或5-7元杂芳基任选进一步被选自卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、羟基、氨基、C3~C7环烷基、5~7元杂环基、C5~C7芳基、5~7元杂芳基的一种或多种基团所取代。
  16. 根据权利要求1至15中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中,
    R3选自氢、卤素、羟基、氨基、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C1-C6卤代烷氧基、C3~C7环烷基、氰基、C5~C7芳基、5~7元杂环基或5~7元杂芳基。
  17. 根据权利要求1至16中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,其中所述化合物选自:
    1-(4-苯并咪唑-1-基-苯基)-3-异噁唑-3-基-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-异噁唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-羟基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[2-(2-羟基-乙氧基)-乙氧基]-苯并咪唑-1-基}-苯基)-脲;
    吗啉-4-羧酸-1-{4-[3-(5-叔丁基-异噁唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基酯;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[5-(2-二甲基氨基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-{6-[(2-二甲基氨基-乙基)-甲基-氨基]-苯并咪唑-1-基}-苯基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[6-(4-甲基-哌嗪-1-基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-{4-[7-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5,6-二甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(4-氟-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(2-氯苯并咪唑-1-基)-苯基]-脲;
    1-(4-苯并咪唑-1-基-3-甲基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-3-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-3-氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-3,5-二氟-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-2-氯-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(6-苯并咪唑-1-基-吡啶-3-基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(2-苯并咪唑-1-基-嘧啶-5-基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-吲哚-1-基-苯基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-吲唑-1-基-苯基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-氟-吲唑-1-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-氟-吲唑-1-基)-苯基]-脲;
    1-(4-苯并三唑-1-基-苯基)-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-吡咯并[2,3-b]吡啶-1-基-苯基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-3-基-苯基)-脲;
    1-[4-(6-溴-咪唑并[4,5-b]吡啶-3-基)-苯基]-3-(5-叔丁基-异噁唑-3-基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-三氟甲基-咪唑[4,5-b]吡啶-3-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-氯咪唑并[4,5-b]吡啶-3-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(5-甲基咪唑并[4,5-b]吡啶-3-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(7-甲基咪唑[4,5-b]吡啶-3-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-(4-咪唑并[4,5-b]吡啶-1-基-苯基)-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基-嘌呤-7-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-甲氧基嘌呤-9-基)-苯基]-脲;
    1-(5-叔丁基-异噁唑-3-基)-3-[4-(6-二甲基氨基-嘌呤-7-基)-苯基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-噻唑-2-基-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(4-甲基-噻唑-2-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-[1,3,4]噻二唑-2-基-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-[1,3,4]噻二唑-2-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-甲基-1H-吡唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-苯基-1H-吡唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-环丙基-2H-吡唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-三氟甲基-2H-吡唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2H-吡唑-3-基)-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-乙氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-己氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丙氧基-苯并咪唑-1-基)-苯基]-脲;
    1-[4-(5-仲丁氧基-苯并咪唑-1-基)-苯基]-3-(5-叔丁基-2H-吡唑-3-基)-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-异丁氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-乙氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-羟基-3-甲氧基-丙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二甲氨基-丙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-二丁基氨基丙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氰基甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-三氟甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-甲基-氧杂环丁烷-3-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-脲;
    (1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(2-哌啶-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-{4-[5-(2-氮杂环庚烷-1-基-乙氧基)-苯并咪唑-1-基]-苯基}-3-(5-叔丁基-2H-吡唑-3-基)-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(3-氟-苄氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-(4-{5-[4-(1-环己基-1H-四唑-5-基)-丁氧基]-苯并咪唑-1-基}-苯基)-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[5-(4-吗啉-4-基-[1,2,5]噻二唑-3-基氧基)-苯并咪唑-1-基]-苯基}-脲;
    4-(1-{4-[3-(5-叔丁基-2H-吡唑-3-基)-脲基]-苯基}-1H苯并咪唑-5-基氧基)-吡啶-2-羧酸甲基胺;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5-氟-7-甲基-苯并咪唑-1-基)-苯基]-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2H-吡唑-3-基)-3-[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-甲基-2H-吡唑-3-基)-脲;
    1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(5-叔丁基-2-甲基-2H-吡唑-3-基)-3-{4-[5-(2-吗啉-4-基-甲氧基)-苯并咪唑-1-基]-苯基}-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-羟基-乙基)-2H-吡唑-3-基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-苯基-2H-吡唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-对甲苯基-2H-吡唑-3-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-甲氧基-苯基)-2H-吡唑-3-基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-三氟甲氧基-苯基)-2H-吡唑-3-基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(4-氟-苯基)-2H-吡唑-3-基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-[5-叔丁基-2-(2-氟-苯基)-2H-吡唑-3-基]-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(5-叔丁基-2-吡啶-2-基-2H-吡唑-3-基)-脲;
    4-{5-[3-(4-苯并咪唑-1-基-苯基)-脲基]-3-叔丁基-吡唑-1-基}-苯甲酸乙酯;
    1-(2-丙烯酰基-5-叔丁基-2H-吡唑-3-基)-3-(4-咪唑-1-基-苯基)-脲;
    3-氨基-5-甲基吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
    5-氨基-3-环丙基吡唑-1-甲酸(4-苯并咪唑-1-基-苯基)-酰胺;
    5-氨基-3-三氟甲基吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-己氧基-苯并咪唑-1-基)-苯基]-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸(4-{5-[3-(4-甲基-哌嗪-1-基)-丙氧基]-苯并咪唑-1-基}-苯基)-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-吗啉-4-基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(2-羟基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(四氢-吡喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸{4-[5-(四氢-呋喃-2-基甲氧基)-苯并咪唑-1-基]-苯基}-酰胺;
    (1-{4-[(5-氨基-3-叔丁基-吡唑-1-羰基)-氨基]-苯基}-1H-苯并咪唑-5-基氧基)-乙酸乙酯;
    5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-氟-苯并咪唑-1-基)-苯基]-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5-三氟甲基-苯并咪唑-1-基)-苯基]-酰胺;
    4-(1-{4-[(5-氨基-3-叔丁基-吡唑-1-羰基)-氨基]-苯基}-1H-苯并咪唑-5-基氧基)-吡啶-2-羧酸甲基酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸{4-[6-(2-甲氧基-乙氧基)-苯并咪唑-1-基]-苯基}-酰胺;
    5-氨基-3-叔丁基-吡唑-1-羧酸[4-(5,6-二甲氧基-苯并咪唑-1-基)-苯基]-酰胺;
    3-叔丁基-吡唑-1-羧酸(4-苯并咪唑-1-基-苯基)-酰胺;
    1-(4-苯并咪唑-1-基-苯基)-3-(3,4-二甲基-异噁唑-5-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(3-异丙基-异噁唑-5-基)-脲;
    1-(4-苯并咪唑-1-基-苯基)-3-(3-叔丁基-异噁唑-5-基)-脲。
  18. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的制备方法,其包括以下步骤:
    Figure PCTCN2017076265-appb-100006
    在适宜的温度、pH和适当的溶剂中,在碱的作用下,使式(II)化合物与式(III)化合物反应,得到通式(I)的化合物;
    所述溶剂优选THF、乙腈、二氯甲烷、甲苯,所述碱优选三乙胺、N,N-二异丙基乙胺、DMAP、吡啶;
    X、Y、A、A1、Z、W、R1、B、R2、G、R3如权利要求1中所定义。
  19. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的制备方法,其包括以下步骤:
    Figure PCTCN2017076265-appb-100007
    在适宜的温度、pH和适当的溶剂中,在碱的作用下,使式(IV)化合物与式(V)化合物反应,得到通式(I)的化合物;
    所述溶剂优选THF、乙腈、二氯甲烷、甲苯,所述碱优选三乙胺、N,N-二异丙基乙胺、DMAP、吡啶;
    X、Y、A、A1、Z、W、R1、B、R2、G、R3如权利要求1中所定义。
  20. 一种药物组合物,其含有治疗有效量的根据权利要求1至17中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,以及一种或多种药学上可接受的载体。
  21. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者根据权利要求20所述的药物组合物,在制备FLT3酪氨酸蛋白激酶抑制剂中的用途。
  22. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或者根据权利要求20所述的药物组合物,在制备用于预防和/或治疗哺乳动物包括人中癌症的药物的用途。
  23. 根据权利要求22所述的用途,其中所述癌症包括非实体瘤如白血病,实体瘤如皮肤癌、黑色素瘤、肺癌、胃癌、乳腺癌、胰腺癌、肝癌、结肠癌。
PCT/CN2017/076265 2016-03-11 2017-03-10 一种脲类化合物、其制备方法及其医药用途 WO2017152874A1 (zh)

Priority Applications (10)

Application Number Priority Date Filing Date Title
JP2018547295A JP6927994B2 (ja) 2016-03-11 2017-03-10 尿素系化合物、その製造方法及びその医薬用途
EP17762560.5A EP3424924B1 (en) 2016-03-11 2017-03-10 Urea compound, preparation method therefor and medical use thereof
RU2018131539A RU2741596C2 (ru) 2016-03-11 2017-03-10 Соединение мочевины, способ его получения и его медицинское применение
BR112018016554-9A BR112018016554B1 (pt) 2016-03-11 2017-03-10 Composto de uréia, método de preparação do mesmo, composição farmacêutica, uso do composto e uso da composição farmacêutica
US16/078,054 US10647680B2 (en) 2016-03-11 2017-03-10 Urea compound, preparation method therefor and medical use thereof
DK17762560.5T DK3424924T3 (da) 2016-03-11 2017-03-10 Ureaforbindelse, fremgangsmåde til fremstilling deraf og medicinsk anvendelse deraf
CN201780026742.3A CN109071523B (zh) 2016-03-11 2017-03-10 一种脲类化合物、其制备方法及其医药用途
AU2017230437A AU2017230437B2 (en) 2016-03-11 2017-03-10 Urea compound, preparation method therefor and medical use thereof
KR1020187028521A KR102430942B1 (ko) 2016-03-11 2017-03-10 우레아 화합물, 이의 제조 방법 및 이의 의학적 용도
HK19100900.8A HK1258545A1 (zh) 2016-03-11 2019-01-18 一種脲類化合物、其製備方法及其醫藥用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610142049.XA CN107176951A (zh) 2016-03-11 2016-03-11 一种脲类化合物、其制备方法及其医药用途
CN201610142049.X 2016-03-11

Publications (1)

Publication Number Publication Date
WO2017152874A1 true WO2017152874A1 (zh) 2017-09-14

Family

ID=59790078

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/076265 WO2017152874A1 (zh) 2016-03-11 2017-03-10 一种脲类化合物、其制备方法及其医药用途

Country Status (10)

Country Link
US (1) US10647680B2 (zh)
EP (1) EP3424924B1 (zh)
JP (1) JP6927994B2 (zh)
KR (1) KR102430942B1 (zh)
CN (2) CN107176951A (zh)
AU (1) AU2017230437B2 (zh)
DK (1) DK3424924T3 (zh)
HK (1) HK1258545A1 (zh)
RU (1) RU2741596C2 (zh)
WO (1) WO2017152874A1 (zh)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018161033A1 (en) * 2017-03-02 2018-09-07 Wright, Adrian Small molecule ire1-alpha inhibitors
US10125123B2 (en) 2016-03-03 2018-11-13 Cornell University Small molecule IRE1-α inhibitors
US10647680B2 (en) 2016-03-11 2020-05-12 Ancureall Pharmaceutical (Shanghai) Co., Ltd. Urea compound, preparation method therefor and medical use thereof
US11084825B2 (en) 2018-12-31 2021-08-10 Biomea Fusion, Llc Substituted pyridines as irreversible inhibitors of menin-MLL interaction
US11174263B2 (en) 2018-12-31 2021-11-16 Biomea Fusion, Inc. Inhibitors of menin-MLL interaction
US12018032B2 (en) 2021-08-20 2024-06-25 Biomea Fusion, Inc. Crystalline forms of N-[4-[4-(4-morpholinyl)-7H-pyrrolo[2,3-d]pyrimidin-6-yl]phenyl]-4-[[3(r)-[(1-oxo-2-propen-1-yl)amino]-1-piperidinyl]methyl]-2-pyridinecarboxamide as an irreversible inhibitor of menin-MLL interaction

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110016014B (zh) * 2018-01-08 2023-02-17 中国科学院上海药物研究所 Ezh2抑制剂及其制备和抗肿瘤治疗中的应用
CN112552302B (zh) * 2019-09-26 2022-05-13 深圳市塔吉瑞生物医药有限公司 取代的芳香稠合环衍生物及其组合物及用途
WO2021104461A1 (zh) * 2019-11-29 2021-06-03 南京明德新药研发有限公司 二氮杂吲哚类衍生物及其作为Chk1抑制剂的应用
CN114341128B (zh) * 2020-07-24 2024-06-25 恩瑞生物医药科技(上海)有限公司 双功能蛋白降解靶向嵌合体类化合物及其制备方法和医药用途
CN113861179A (zh) * 2021-10-22 2021-12-31 上海应用技术大学 一种新型flt3激酶抑制剂及其合成与应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080119466A1 (en) * 2004-02-23 2008-05-22 Chugai Seiyaku Kabushiki Kaisha Heteroarylphenylurea Derivative
CN102153551A (zh) * 2010-04-02 2011-08-17 济南海乐医药技术开发有限公司 基于吲唑或氮杂吲唑的双芳基脲或硫脲类结构抗肿瘤药物
CN102319242A (zh) * 2011-10-13 2012-01-18 济南海乐医药技术开发有限公司 吲唑双芳基脲化合物作为蛋白激酶抑制剂的应用
CN103864792A (zh) * 2012-12-12 2014-06-18 山东亨利医药科技有限责任公司 作为酪氨酸激酶抑制剂的含氮并环类化合物

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1567393A (en) * 1925-03-16 1925-12-29 Schlaich Herman Radiator cap
EP0242322B1 (en) * 1986-03-18 1992-12-16 Sandoz Ag Phenylureas
GB9823873D0 (en) * 1998-10-30 1998-12-30 Pharmacia & Upjohn Spa 2-ureido-thiazole derivatives,process for their preparation,and their use as antitumour agents
WO2005042495A1 (en) * 2003-10-21 2005-05-12 Imclone Systems Incorporated (benzimidazol-2-yl)-phenyl-phenyl-urea compounds and methods for inhibiting heparanase activity
TW200810754A (en) * 2006-03-23 2008-03-01 Synta Pharmaceuticals Corp Benzimidazolyl-pyridine compounds for inflammation and immune-related uses
CN101239978A (zh) * 2008-03-05 2008-08-13 南方医科大学 一种咪唑并吡啶类化合物
JP6261340B2 (ja) * 2010-11-18 2018-01-17 リガンド ファーマシューティカルズ インコーポレイテッド 造血成長因子模倣体の使用
CN103450152B (zh) * 2012-06-04 2015-11-18 济南海乐医药技术开发有限公司 基于吲唑、吲哚或氮杂吲唑、氮杂吲哚的双芳基脲类结构抗肿瘤药物
CN104513252B (zh) * 2013-09-26 2017-11-10 广东东阳光药业有限公司 取代脲衍生物及其在药物中的应用
CN107176951A (zh) 2016-03-11 2017-09-19 恩瑞生物医药科技(上海)有限公司 一种脲类化合物、其制备方法及其医药用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080119466A1 (en) * 2004-02-23 2008-05-22 Chugai Seiyaku Kabushiki Kaisha Heteroarylphenylurea Derivative
CN102153551A (zh) * 2010-04-02 2011-08-17 济南海乐医药技术开发有限公司 基于吲唑或氮杂吲唑的双芳基脲或硫脲类结构抗肿瘤药物
CN102319242A (zh) * 2011-10-13 2012-01-18 济南海乐医药技术开发有限公司 吲唑双芳基脲化合物作为蛋白激酶抑制剂的应用
CN103864792A (zh) * 2012-12-12 2014-06-18 山东亨利医药科技有限责任公司 作为酪氨酸激酶抑制剂的含氮并环类化合物

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE REGISTRY 12 April 2006 (2006-04-12), ANONYMOUS, XP055576441, retrieved from STN Database accession no. 880176-45-4 *
DATABASE REGISTRY April 2006 (2006-04-01), Database accession no. 851676-04- 5 *
DATABASE REGISTRY Database accession no. 863707-78-2 *
DATABASE REGISTRY Database accession no. 863707-79-3 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10125123B2 (en) 2016-03-03 2018-11-13 Cornell University Small molecule IRE1-α inhibitors
US10988461B2 (en) 2016-03-03 2021-04-27 Cornell University Small molecule IRE1-α inhibitors
US10647680B2 (en) 2016-03-11 2020-05-12 Ancureall Pharmaceutical (Shanghai) Co., Ltd. Urea compound, preparation method therefor and medical use thereof
WO2018161033A1 (en) * 2017-03-02 2018-09-07 Wright, Adrian Small molecule ire1-alpha inhibitors
US11084825B2 (en) 2018-12-31 2021-08-10 Biomea Fusion, Llc Substituted pyridines as irreversible inhibitors of menin-MLL interaction
US11174263B2 (en) 2018-12-31 2021-11-16 Biomea Fusion, Inc. Inhibitors of menin-MLL interaction
US11702421B2 (en) 2018-12-31 2023-07-18 Biomea Fusion, Llc Substituted pyridines as irreversible inhibitors of menin-MLL interaction
US11845753B2 (en) 2018-12-31 2023-12-19 Biomea Fusion, Inc. Inhibitors of menin-mll interaction
US12018032B2 (en) 2021-08-20 2024-06-25 Biomea Fusion, Inc. Crystalline forms of N-[4-[4-(4-morpholinyl)-7H-pyrrolo[2,3-d]pyrimidin-6-yl]phenyl]-4-[[3(r)-[(1-oxo-2-propen-1-yl)amino]-1-piperidinyl]methyl]-2-pyridinecarboxamide as an irreversible inhibitor of menin-MLL interaction

Also Published As

Publication number Publication date
EP3424924A1 (en) 2019-01-09
RU2018131539A (ru) 2020-04-13
US10647680B2 (en) 2020-05-12
KR102430942B1 (ko) 2022-08-08
US20190047964A1 (en) 2019-02-14
AU2017230437A1 (en) 2018-09-06
RU2018131539A3 (zh) 2020-04-13
HK1258545A1 (zh) 2019-11-15
CN109071523A (zh) 2018-12-21
AU2017230437B2 (en) 2020-11-26
EP3424924B1 (en) 2023-05-24
KR20180132664A (ko) 2018-12-12
JP6927994B2 (ja) 2021-09-01
JP2019507779A (ja) 2019-03-22
EP3424924A4 (en) 2019-08-07
BR112018016554A2 (pt) 2018-12-26
CN109071523B (zh) 2022-05-31
DK3424924T3 (da) 2023-08-28
CN107176951A (zh) 2017-09-19
RU2741596C2 (ru) 2021-01-27

Similar Documents

Publication Publication Date Title
KR102430942B1 (ko) 우레아 화합물, 이의 제조 방법 및 이의 의학적 용도
US10478423B2 (en) Substituted indazole derivatives active as kinase inhibitiors
KR101624296B1 (ko) Axl 억제제로서의 약학적 활성 화합물
CN101679401B (zh) 用于治疗肿瘤的作为met激酶抑制剂的2-氧代-3-苄基-苯并*唑-2-酮衍生物及相关化合物
WO2018045956A1 (zh) 苯并咪唑类化合物激酶抑制剂及其制备方法和应用
BR112018004175B1 (pt) Composto pirazolo[3,4-d]pirimidina, composição farmacêutica, inibidor de her2 e agente antitumor contendo o dito composto e usos terapêuticos do dito composto
BR112018010291B1 (pt) Composto, composição farmacêutica, e, uso de um composto
IL234313A (en) Aromatic heterocyclic compounds containing nitrogen
WO2018018986A1 (zh) 二苯氨基嘧啶及三嗪化合物、其药用组合物及用途
WO2014082578A1 (zh) 杂芳基炔烃化合物及其应用
EA019534B1 (ru) ПРОИЗВОДНЫЕ 3-(3-ПИРИМИДИН-2-ИЛБЕНЗИЛ)-1,2,4-ТРИАЗОЛО[4,3-b]ПИРИДАЗИНА В КАЧЕСТВЕ ИНГИБИТОРОВ Met КИНАЗЫ
TW201934546A (zh) 嘧啶類化合物、其製備方法及其醫藥用途
EP3176160A1 (en) Pyridine-substituted 2-aminopyridine protein kinase inhibitors
WO2018137639A1 (zh) 一种组蛋白甲基转移酶ezh2抑制剂、其制备方法及其医药用途
JP2017511309A (ja) Wntシグナル伝達経路の阻害剤
CN110467616B (zh) 含杂芳基取代哒嗪酮结构的三唑并吡嗪类化合物的制备及应用
WO2023109751A1 (zh) 嘧啶或吡啶类衍生物及其医药用途
US20180044306A1 (en) 1,3,4-thiadiazol-2-yl-benzamide derivatives as inhibitors of the wnt signalling pathway
WO2022017442A1 (zh) 双功能蛋白降解靶向嵌合体类化合物及其制备方法和医药用途
JP2017521465A (ja) グルコース輸送阻害剤
WO2023046114A1 (zh) 蝶啶酮衍生物及其应用
WO2021020586A1 (ja) 複素環化合物
BR112018016554B1 (pt) Composto de uréia, método de preparação do mesmo, composição farmacêutica, uso do composto e uso da composição farmacêutica
CN116444599A (zh) 甾体类化合物及制备方法和用途
WO2021004482A1 (zh) 取代的吡唑并喹唑啉酮化合物及其应用

Legal Events

Date Code Title Description
REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018016554

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2017230437

Country of ref document: AU

Date of ref document: 20170310

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018547295

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20187028521

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2017762560

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017762560

Country of ref document: EP

Effective date: 20181002

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17762560

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 112018016554

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180814