WO2017124657A1 - 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法 - Google Patents

一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法 Download PDF

Info

Publication number
WO2017124657A1
WO2017124657A1 PCT/CN2016/079491 CN2016079491W WO2017124657A1 WO 2017124657 A1 WO2017124657 A1 WO 2017124657A1 CN 2016079491 W CN2016079491 W CN 2016079491W WO 2017124657 A1 WO2017124657 A1 WO 2017124657A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenoxyphenyl
pyrazole
reaction
amino
pyrimidin
Prior art date
Application number
PCT/CN2016/079491
Other languages
English (en)
French (fr)
Inventor
李英富
黄浩喜
刘冠锋
陈垌珲
杜振军
Original Assignee
成都倍特药业有限公司
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 成都倍特药业有限公司 filed Critical 成都倍特药业有限公司
Publication of WO2017124657A1 publication Critical patent/WO2017124657A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention belongs to the field of medicinal chemistry, and in particular to a Bruton tyrosine kinase inhibitor having a spiro or bridged ring structure, and a method for preparing such a compound and a method and use for inhibiting BTK activity using the novel compounds.
  • B cell malignancies include chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), small lymphocytic lymphoma (SLL), mantle cell lymphoma (MCL), follicular lymphoma (FL), diffuse large B cell lymphoma (DLBCL), multiple myeloma (MM) and Macroglobulinemia (WM).
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphoblastic leukemia
  • SLL small lymphocytic lymphoma
  • MCL mantle cell lymphoma
  • FL follicular lymphoma
  • FL diffuse large B cell lymphoma
  • MM multiple myeloma
  • WM Macroglobulinemia
  • Bruton's tyrosine kinase is a member of a family of non-receptor protein tyrosine kinases that are key signaling enzymes expressed in all hematopoietic cell types except T lymphocytes and natural killer cells. It plays a crucial role in the B cell signaling pathway and is closely related to B lymphocyte development, differentiation, signaling and survival. The important role of BTK in the B cell receptor (BCR) signaling pathway makes it a hot target for the treatment of B cell malignancies.
  • BCR B cell receptor
  • the first oral BTK inhibitor ibrutinib, which has been approved by the FDA, has been approved for four indications. Its inhibitor has strong selectivity and low side effects. It is called a “breakthrough” new drug and its main mechanism of action. It acts on the thiol group of the BTK enzyme cysteine acid (Cys481) residue to form a covalent bond, which inactivates the BTK enzyme.
  • cysteine acid Cys481
  • the present invention provides a Bruton tyrosine kinase inhibitor having a spiro or bridged ring structure, in particular a compound of the formula (I), (II) and an optical isomer thereof, or
  • a pharmaceutically acceptable salt, hydrate or solvate, alone or in combination with other drugs, as a BTK inhibitor in particular for the prevention or treatment of cell proliferative diseases such as cancer, especially in prevention or Use in the treatment of B cell malignancies such as mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL).
  • MCL mantle cell lymphoma
  • CLL chronic lymphocytic leukemia
  • a Bruton tyrosine kinase inhibitor of a spiro or bridged ring structure comprising formula (I):
  • Y is selected from the group consisting of:
  • R1, R2 may be independently selected from a hydrogen atom, a fluorine atom, a bromine atom, an iodine atom, an amino group, a cyano group, a fluorenyl group, a C 1 -C 5 linear or branched alkyl group; a plurality of hydrogen atoms may be substituted by one or more halogen, amino, cyano, hydroxy, thio groups;
  • R 3 and R 4 may be independently selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, an iodine atom, an amino group, a cyano group, a fluorenyl group, a C 1 -C 5 linear or branched alkyl group;
  • One or more hydrogen atoms on the group may be substituted by one or more halogen, amino, cyano, hydroxy, thiol groups.
  • R 3 and R 4 may be independently selected from a halogen atom, a C 1 -C 3 linear or branched alkyl group; one or more hydrogen atoms on the alkyl group may be one or more halogen, amino, Cyano, hydroxy, thiol substituted.
  • Bruton tyrosine kinase inhibitor of the spiro or bridged ring structure is selected from the following structures:
  • a Bruton's tyrosine kinase inhibitor having a spiro or bridged ring structure is selected from the following structures:
  • the invention provides a Bruton tyrosine kinase inhibitor having a spiro or bridged ring structure and a preparation method thereof, which have the following beneficial effects:
  • the Bruton tyrosine kinase inhibitor provided by the present invention has strong inhibitory activity against BTK and cell proliferation, and is nearly 8-fold more potent than ibrutinib, so that prevention can be achieved by using a smaller dose. And the treatment effect, high utilization.
  • the spiro or bridged ring compound designed by the present invention can be used as a BTK inhibitor with broad anti-malignant tumor application prospects, such as treatment of chronic lymphocytic lymphoma, chronic lymphocytic leukemia (CLL), B-cell young Lymphocytic leukemia, lymphoplasmacytic lymphoma, spleen marginal lymphoma, acute lymphoblastic leukemia (ALL), small lymphocytic lymphoma (SLL) plasma cell myeloma, plasmacytoma, extranodal marginal B-cell Lymphoma, multiple myeloma (MM), chromosomal deletion of chronic lymphocytic leukemia, intraductal marginal B-cell lymphoma, mantle cell lymphoma (MCL), intravascular large B-cell lymphoma, primary exudative Lymphoma, diffuse B-cell lymphoma (DLBCL), follicular lymphoma, Waldenstrom's macroglobulinemia,
  • DMSO dimethyl sulfoxide
  • DMF for N,N-dimethylformamide
  • LDA lithium diisopropylamide
  • LHDMS lithium bis(trimethylsilyl)amide
  • LAH means lithium aluminum hydride
  • DCM means dichloromethane
  • TEA means triethylamine
  • TFA trifluoroacetic acid
  • THF means tetrahydrofuran
  • PPh 3 means triphenylphosphine
  • EA means ethyl acetate
  • MeOH means methanol
  • DIEA means N , N diisopropylethylamine
  • (BOC) 2 O represents di-tert-butyl dicarbonate
  • OTs means p-toluenesulfonate
  • OMs means methylsulfonate
  • 9-BBN means 9-boron bicyclol [3.3.1] decane
  • rt represents room temperature
  • DIAD represents diisoprop
  • the derivative of the formula (I) of the present invention can be produced by the following reaction formula:
  • the starting material is a commercialized Ibrutinib intermediate A and a protected group such as a spiro ring or a bridged ring.
  • the intermediate B is obtained by a mitsunobu reaction or a direct substitution reaction, and the intermediate B is deprotected to form an intermediate C.
  • the resulting C is further reacted with acryloyl chloride to give the target compound.
  • the structure of the compound was determined by H NMR and LC-MS.
  • the HNMR measurement was carried out in a ppm unit using a Bruker 400 (400 MHz) spectrometer, 1 H chemical shift, and the solvent was deuterated chloroform or deuterated DMSO, the internal standard was tetramethylsilane, and the chemical shift was given in ppm.
  • the LC-MS was measured using the Agilent 1200 Series, 6110 Series, and 6120 Series using electrospray ionization mode with the following conditions: Waters X Bridge C18 column (50 mm x 4.6 mm x 3.5 um), flow rate: 2.0 mL/min, column temperature: 40 °C.
  • a thin layer of silica gel was used with a GF254 silica gel plate.
  • Step 1 Preparation of N,N-bis(2-chloroethyl)carbamic acid tert-butyl ester (1b)
  • Step 2 Preparation of spiro[ ⁇ -1,4-piperidine]-1-carboxylic acid tert-butyl ester (1d)
  • Step 3 Preparation of 3-hydroxy-2,3-dihydrospiro[ ⁇ -1,4'-piperidine]-1'-carboxylic acid tert-butyl ester (1e)
  • Step 4 3-(4-Amino-3-(phenoxyphenyl)-1H pyrazole[3,4-d]pyrimidin-1-yl)-2,3-dihydrospiro[ ⁇ -1,4 Preparation of tert-butyl ester of '-piperidine]-1'-yl)prop-2-en-1-carboxylate (1f)
  • Step 5 1-(2,3-Dihydrospiro[ ⁇ -1,4'-piperidinyl]-3-yl)-3-(4-phenoxyphenyl)-1H-pyrazole [3,4 -d]Preparation of pyrimidine-4-amine (1g);
  • Step 6 1-(3-(Amino-3-(4-phenoxyphenyl)-3a,7a-1H-pyrazole[3,4-d]pyrimidin-1-yl)-2,3-di Preparation of hydrogen snail [ ⁇ -1,4'-piperidin]-1'-yl)prop-2-en-1-one (1)
  • Step 2 3-Exo-hydroxy-8-aza bridge [3.2.1] octane-8-carboxylic acid tert-butyl ester, 3-endo-hydroxy-8-aza bridge [3.2.1] octane- Preparation of 8-carboxylic acid tert-butyl ester
  • Step 2 Preparation of 3-carbonyl-9-azabicyclo[3.3.1]nonane-9-carboxylic acid tert-butyl ester
  • Step 3 Preparation of 3-hydroxy-9-azabicyclo[3.3.1]nonane-9-carboxylic acid tert-butyl ester
  • Step 4 3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-9-azabicyclo[3.3.1 Preparation of tert-butyl 9-carboxylate
  • Step 6 1-(3-(4-Amine-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-9-azabicyclo[3.3 .1] Preparation of decane-9-yl)propan-2-en-1-one
  • Step 1 Preparation of 4-allyl-4hydroxypiperidine-1-carboxylic acid tert-butyl ester
  • allyl bromide was slowly added dropwise to a solution of magnesium powder (3.2 g, 13.33 mmol) in 20 mL of dry diethyl ether at room temperature under nitrogen atmosphere. The reaction mixture was stirred with heating and then slowly added dropwise after the reaction was initiated. Allyl bromide, the dropping rate is controlled to maintain the reflux state of the reaction, allyl bromide (13.3g, 0.11mol) is added, after the reaction is completed, it is cooled to -15 ° C, and N-tert-butoxycarbonyl-4-piperidyl is slowly added.
  • Step 2 Preparation of 3-hydroxy-1-oxa-8-azaspiro[4.5]decane-8-carboxylic acid tert-butyl ester
  • Step 3 3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-1-oxa-8-aza Preparation of spirocyclo[4.5]decane-8-carboxylic acid tert-butyl ester
  • Step 5 1-(3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-1-oxa-8 -Preparation of azaspiro[4.5]decane-8-yl)propane-2-en-1-one
  • Step 1 Preparation of quinoline-1(2H)-carboxylic acid tert-butyl ester
  • Step 2 Preparation of 4-bromo-3-hydroxy-3,4-dihydroquinolin-1(2H)-carboxylic acid tert-butyl ester
  • Step 3 3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-4-hydroxy-3,4-di Preparation of Hydroquinolin-1(2H)-carboxylic acid tert-butyl ester
  • Step 4 3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-1,2,3,4-tetra Preparation of hydrogen quinolin-4-ol
  • Step 5 1-(3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-4-hydroxy-3, Preparation of 4-dihydroquinoline-1(2H)-yl)propan-2-en-1-one
  • Step 1 Preparation of 4-carbonylspiro[chroman-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester
  • Step 2 Preparation of 4-hydroxyspiro[chroman-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester
  • Step 3 4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl) snail [Color-2,4'- Preparation of piperidine]-1'-carboxylic acid tert-butyl ester
  • Step 5 1-(4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl) snail [color full-2,4 Preparation of '-piperidine-1'-upper) prop-2-en-1-one
  • Step 1 Preparation of 7-fluoro-4-carbonylspiro[chroman-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester
  • N-tert-Butoxycarbonyl-4-piperidone (4.4 g, 20.76 mmol) was added to a solution of 4-fluoro-2-hydroxyacetophenone (3.4 g, 20.76 mmol) in 75 mL of methanol at room temperature.
  • Step 2 Preparation of 7-fluoro-4-hydroxyspiro[chroman-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester
  • Step 3 4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-7-fluorospiro [Color-2 Of 4'-piperidine]-1'-carboxylic acid tert-butyl ester
  • Step 4 1-(7-Fluorospiro[chroman-2,4'-piperidinyl]-4-yl)-3-(4-phenoxyphenyl)-1H-pyrazole [3,4-d Preparation of pyrimidine-4-amine
  • Step 5 1-(4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-7-fluorospiro Preparation of full-2,4'-piperidinyl-1'-yl)propan-2-en-1-one
  • Step 1 Preparation of 3-allyl-3-hydroxypyrrole-1-carboxylic acid tert-butyl ester
  • Step 2 Preparation of tert-butyl 3-(2,3-dibromopropyl)-3-hydroxypyrrole-1-carboxylate
  • Step 3 Preparation of 3-bromo-1-carbonyl-7-azaspiro[4.4]decane-7-carboxylic acid tert-butyl ester
  • Step 4 3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-1-oxo-7-aza Preparation of spiro[4.4]decane-7-carboxylic acid tert-butyl ester
  • Step 6 1-(3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-1-oxo-7 -Preparation of azaspiro[4.4]decane-7-yl)propan-2-en-1-one
  • Step 1 Preparation of 3-hydroxy-3,4-dihydropyridine-1(2H)-carboxylic acid benzyl ester
  • Step 2 Preparation of 4-hydroxy-2-azabicyclo[4.1.0]heptane-2-carboxylic acid tert-butyl ester
  • Step 3 4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-2-azabicyclo[4.1.0 Preparation of benzyl heptane-2-carboxylate
  • Step 5 1-(4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)-2-azabicyclo[ 4.1.0] Preparation of decane-2-yl)propan-2-en-1-one
  • Test Example 1 In vitro BTK inhibitory kinase activity assay
  • the BTK in vitro inhibitory activity test (Test Example 1 and Test Example 2) was determined by Shanghai Ruizhi Chemical Research Co., Ltd.
  • Substrate polypeptide FAM-P2 (GL Biochem product, Cat. No. 112394, batch number P100804-XZ112394);
  • Adenosine triphosphate (Sigma product, batch number A7699-1G, batch number 987-65-5);
  • Diaminoethane tetraacetate (Sigma product, item number E5134, batch number 60-00-4);
  • staurosporine (Sigma product, article number S4400-1MG, lot number 046K4080);
  • Mobility-Shift Assay a microfluidic chip technology that applies the basic idea of capillary electrophoresis to a microfluidic environment.
  • the substrate used for the experiment is a fluorescently labeled peptide, which is enzymed in the reaction system. Under the action of the substrate, the substrate is transformed into a product, and the charge it carries also changes accordingly.
  • the Mobility-Shift Assay separates the two by using the difference in charge between the substrate and the product, and separately detects it. The test results are expressed by the conversion rate.
  • the 10 concentrations of the compound were diluted 10-fold with 1 ⁇ kinase buffer; wherein the kinase buffer contained hydroxyethylpiperazine ethanesulfonic acid at a concentration of 50 mmol/L and a pH of 7.5, 0.01%.
  • adding a substrate solution to the 384-well plate adding 10 ⁇ l of a 2.5-fold substrate solution to the 384-well reaction plate;
  • Test Example 2 Determination of in vitro cell proliferation inhibitory activity of compounds using different cell lines
  • RPMI1640 medium (INVITROGEN product, Cat. No. 61870-127);
  • Luminescent cell viability assay kit Promega product, item number G7572
  • the drug to be tested was prepared into a 10 mmol/L solution using DMSO, and was dissolved and stored for use.
  • the selected two cells were cultured in RPMI1640 medium containing 10% fetal bovine serum, and incubated at 37 ° C under 5% CO 2 humidification conditions.
  • the cells were digested from the cell culture plate using trypsin, and the cell density was measured after resuspending using complete medium (RPMI1640 medium containing 10% fetal bovine serum);
  • the compound test initial concentration is 50 umol/L, and use a DMSO gradient dilution at a concentration of 200 times concentration of 10 mmol/L, the dilution factor is 3 Times, 9 concentration points;
  • the reference compound test initial concentration is 1umol / L, 200 times the concentration of 0.2mmol / L as the starting concentration using DMSO gradient dilution, dilution factor of 3 times, 9 concentration points);
  • the cell culture plate to which the compound was added was returned to the incubator, and incubated at 37 ° C under a humidified condition of 5% CO 2 for 72 hours.
  • the time to read the board is set to 0.5 seconds per hole.
  • % inhibition rate (maximum signal value - compound signal value) / (maximum signal value - minimum signal value) ⁇ 100.
  • the maximum signal value was obtained from cells treated with dimethyl sulfoxide for 72 h; the minimum signal value was obtained from the medium alone (zero cell number).
  • Examples 2, 3, 4, 5, 8, and 9 all have significant inhibitory effects on BTK activity, which is roughly equivalent to the positive control ibrutinib, indicating that the piperidine ring is replaced by The inhibition of compounds such as spiro or bridged loops has little effect, but still exhibits potent BTK kinase inhibitory activity.
  • both of the two compounds tested, Example 4 and Example 8 exhibited more potent cell proliferation inhibitory activity than the positive control, especially the inhibitory activity of Example 4 on DOHH2 cells was increased. Almost 8 times. Therefore, the spiro or bridged ring compound designed by the present invention can be used as a BTK inhibitor, and has a broad application prospect against malignant tumors.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明公开了一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法,该抑制剂包括式(I)化合物,具有抑制BTK活性及抗恶性肿瘤作用。

Description

一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法 技术领域
本发明属于药物化学领域,具体涉及一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,以及此类化合物的制备方法和使用这些新型化合物抑制BTK活性的方法和用途。
背景技术
近年来,血液系统恶性肿瘤的治疗方法正逐步从常规的化学免疫疗法向靶向治疗转变,与传统化疗药物不同,靶向治疗可以精确定位快速生长的癌细胞,同时又能避免对正常细胞的伤害,降低传统化疗药物的脱发、胃肠道反应和骨髓抑制等毒副作用;另一方面随着阿法替尼、卡那替尼、来那替尼等共价靶向抗肿瘤药物特别是大量针对激酶的替尼类药物的产生,使人们又更多地关注小分子共价抑制剂药物。小分子共价抑制剂(covalent inhibitors)也称不可逆抑制剂(irreversible inhibitours),是通过共价键与靶蛋白残基发生不可逆结合,从而发挥其生物功能的一类抑制剂。
B细胞恶性肿瘤包括慢性淋巴细胞白血病(CLL)、急性淋巴细胞白血病(ALL)、小淋巴细胞性淋巴瘤(SLL)、套细胞淋巴瘤(MCL)、滤泡性淋巴瘤(FL)、弥漫大B细胞淋巴瘤(DLBCL)、多发性骨髓瘤(MM)和
Figure PCTCN2016079491-appb-000001
巨球蛋白血症(WM)。对于上述病症传统药物虽然可以缓解症状,但均不能有效延长患者生存时间。
布鲁顿酪氨酸激酶(BTK)是一种非受体蛋白酪氨酸激酶家族的成员,是除了T淋巴细胞和自然杀伤细胞之外的所有造血细胞类型中表达的关键信号酶。 在B细胞信号传导途径中扮演至关重要的角色,与B淋巴细胞发育、分化、信号传递和存活密切相关。BTK在B细胞受体(BCR)信号通路的重要作用,使其成为B细胞恶性肿瘤治疗的热门靶点。
目前第一个获得FDA批准上市的口服BTK抑制剂依鲁替尼,已经获批四种适应症,其抑制剂选择性强,毒副作用低,被称为“突破性”新药,其主要作用机制是与BTK酶半胱按酸(Cys481)残基的巯基发生作用,形成共价键,使BTK酶失去活性。但在给药过程中,依然存在易被代谢,生物利用度低(仅仅7~23%),临床给药量大(560mg/天)等问题。
发明内容
本发明针对上述不足之处而提供的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,特别是通式(Ⅰ)、(Ⅱ)化合物及其光学异构体或其药学上接受的盐、水合物或溶剂化物在单独或与其他药物联合作为BTK抑制剂在药物中的用途,具体而言是在预防或治疗细胞增生疾病如癌症中的用途,尤其是在预防或治疗套细胞淋巴瘤(MCL)和慢性淋巴细胞白血病(CLL)等B细胞恶性肿瘤药物中的用途。
一种螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,包括式(Ⅰ):
Figure PCTCN2016079491-appb-000002
其中Y选自下列基团:
Figure PCTCN2016079491-appb-000003
其中,R1、R2可以独立地选自氢原子、氟原子、溴原子、碘原子、氨基、氰基、巯基、C1~C5直链或支链烷基;所述烷基上的一个或多个氢原子可以被一个或者多个卤素、氨基、氰基、羟基、硫基取代;
m1、m2均为C1~C5烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、-S=O、-S(=O)2取代;
n1、n2均为C0~C5烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、-S=O、-S(=O)2取代;
G为C、O、S、-S=O、-S(=O)2或NR4;所述R4可以是氢、C1~C6直链或支链烷基、C1~C6直链或支链杂烷基;其中C1~C6直链或支链烷基、C1~C6直链或支链杂烷基上一个或多个氢原子可以进一步被一个或多个氨基、氰基、羟基、硫基、卤素取代。
进一步优选为含式(Ⅱ)的化合物:
Figure PCTCN2016079491-appb-000004
其中,R3和R4可以独立地选自氢原子、氟原子、氯原子、溴原子、碘原子、氨基、氰基、巯基、C1~C5直链或支链烷基;所述烷基上的一个或多个氢原子可以被一个或者多个卤素、氨基、氰基、羟基、巯基取代。
进一步优选为含式(III)的化合物:
Figure PCTCN2016079491-appb-000005
其中,m1、m2均为C1~C2烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、S=O、S(=O)2取代;
n1、n2均为C1~C3烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、S=O、S(=O)2取代;
G为C、O、S、S=O、S(=O)2或NR4;所述R4可以是氢、C1~C4直链或支链烷基、C1~C4直链或支链杂烷基。
进一步优选为含式(IV)的化合物:
Figure PCTCN2016079491-appb-000006
其中,R3和R4可以独立地选自卤素原子、C1~C3直链或支链烷基;所述烷基上的一个或多个氢原子可以被一个或者多个卤素、氨基、氰基、羟基、巯基取代。
进一步地,螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,选自以下结构:
Figure PCTCN2016079491-appb-000007
Figure PCTCN2016079491-appb-000008
Figure PCTCN2016079491-appb-000009
Figure PCTCN2016079491-appb-000010
进一步地,一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,选自以下结构:
Figure PCTCN2016079491-appb-000011
本发明提供的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法,具有以下几种有益效果:
(1)本发明提供的布鲁顿酪氨酸激酶抑制剂对BTK和细胞增殖具有较强的抑制活性,比依鲁替尼药效高近8倍,因此使用较小药量即可达到预防和治疗效果,利用率高。
(2)本发明所设计的螺环或桥环化合物可用作BTK抑制剂具有广阔的抗恶性肿瘤的应用前景,如可治疗慢性淋巴细胞淋巴瘤、慢性淋巴细胞白血病(CLL)、B细胞幼淋巴细胞白血病、淋巴质浆细胞淋巴瘤、脾脏边缘带淋巴瘤、急性淋巴细胞白血病(ALL)、小淋巴细胞性淋巴瘤(SLL)浆细胞性骨髓瘤、浆细胞瘤、结外边缘带B细胞淋巴瘤、多发性骨髓瘤(MM)、染色体缺失的慢性淋巴细胞白血病、结内边缘带B细胞淋巴瘤、套细胞淋巴瘤(MCL)、血管内大B细胞淋巴瘤、原发性渗出性淋巴瘤、弥漫性B细胞淋巴瘤(DLBCL)、滤泡性淋巴瘤、Waldenstrom氏巨球蛋白血症等;还可用于系统性红斑狼疮、类风湿性关节炎、系统性血管炎、硬皮病、天疱疮、皮肌炎、混合性结缔组织病、自身免疫性溶血性贫血、甲状腺自身免疫病、溃疡性结肠炎、阴道炎、过敏性鼻炎、支气管炎、乳腺炎、纤维组织炎、宫颈炎、卵巢炎、前列腺炎、鼻炎、鼻窦炎、扁桃体炎、心包炎、肺炎、肾炎、肠炎、阑尾炎、胆囊炎、脑炎、结膜炎、子宫内膜炎、脑膜炎、咽炎等的治疗。
具体实施方式
下面的缩写具有如下所示的意义:DMSO表示二甲亚砜;DMF表示N,N-二甲基甲酰胺;LDA表示二异丙基氨基锂;LHDMS表示二(三甲基硅基)氨基锂;LAH表示氢化铝锂;DCM表示二氯甲烷;TEA表示三乙胺;TFA表示三氟乙酸;THF表示四氢呋喃;PPh3表示三苯基膦;EA表示乙酸乙酯;MeOH表示甲醇;DIEA表示N,N二异丙基乙胺,(BOC)2O表示二碳酸二叔丁酯;OTs是表示对甲基 苯磺酸酯;OMs是表示甲基磺酸酯;9-BBN表示9-硼双环[3.3.1]壬烷;rt表示室温;DIAD表示偶氮二甲酸二异丙酯。
本发明的通式(Ⅰ)所示的衍生物可以通过以下反应式制备:
Figure PCTCN2016079491-appb-000012
起始原料为商品化的依鲁替尼中间体A与带有保护基的螺环或者桥环等片断通过mitsunobu反应或者直接取代反应得到中间体B,中间体B通过脱保护反应生成中间体C,生成的C进一步和丙烯酰氯作用得到目标化合物。通过HNMR和LC-MS来确定化合物的结构。
HNMR测定是用Bruker 400(400MHz)spectrometer,1H化学位移以ppm单位给出,测定溶剂为氘代氯仿或者氘代DMSO,内标为四甲基硅烷,化学位移以ppm作为单位给出。
LC-MS测定是用Agilent 1200系列、6110系列、6120系列,采用电喷射离子化模式,条件是:Waters X Bridge C18柱(50mmx4.6mmx3.5um),流速:2.0mL/min,柱温:40℃。
薄层硅胶使用GF254硅胶板。
柱层析使用烟台黄海硅胶200-300目硅胶为载体。
实施例1:1-(3-(氨基-3-(4-苯氧基苯基)-3a,7a-1H-吡唑[3,4-d]嘧啶-1-基)-2,3- 二氢螺[茚-1,4'-哌啶]-1'-基)丙-2-烯-1-酮的制备,
合成步骤如下所示:
Figure PCTCN2016079491-appb-000013
步骤1:N,N-双(2-氯乙基)氨基甲酸叔丁酯(1b)的制备
在冰浴条件下,向装有二(2-氯乙基)胺盐酸盐(19.99g,0.112mmol)和(Boc)2O(26.84g,0.123mmol)的300mL DCM溶液中慢慢加入TEA(34.00g,0.336mmol),待反应混合物自然升高到室温并继续搅拌5h,TLC显示原料反应完成以后,反应液分别用水和饱和食盐水洗涤,有机相用无水Na2SO4充分干燥,真空蒸发后通过快速色谱法(PE:EA=100:1)纯化得到24.4g目标化合物,为无色油状物;
步骤2:螺[茚-1,4-哌啶]-1-羧酸叔丁酯(1d)的制备
在氮气保护下,向装有茚(2.50g,21.52mmol)的50mL THF溶液中在-10℃下慢慢滴加LiTMDS(43.04mL,43.04mmol,1mol/L THF),该反应混合物在-10℃ 下搅拌30min以后,在-10℃下向上述反应混合物中滴加N,N-双(2-氯乙基)氨基甲酸叔丁酯(5.21g,21.52mmol)的5mL THF溶液,该反应混合物在-10℃下搅拌2h,然后在室温搅拌30min,TLC显示反应完成后,水加入反应混合物中然后用EA萃取(50mLX3),有机相用无水Na2SO4充分干燥,真空蒸发后通过快速色谱法(PE:EA=50:1~30:1)纯化得到3.6g目标化合物,为黄色固体;
步骤3:3-羟基-2,3-二氢螺[茚-1,4'-哌啶]-1'-羧酸叔丁酯(1e)的制备
在氮气保护下,向装有螺[茚-1,4-哌啶]-1-羧酸叔丁酯(2.85g,0.01mmol)的30mL THF溶液中(0℃)慢慢滴加9-BBN(40.00mL,0.02mol,0.5mol/L THF);该反应混合物在70℃下搅拌过夜,然后向上述反应混合物中慢慢滴加1mol/L NaOH(20.00mL)和H2O2(4.00mL)淬灭反应,然后冷却至室温后用EA萃取(50mLX3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=5:1)纯化得到2.85g目标化合物,为黄色油状物;
1H NMR(400MHz,CDCl3)δ1.39(1H,dd,J=13.2Hz,2.0Hz),1.49(9H,s),1.63(1H,dd,J=13.2Hz,2.4Hz),1.71-1.78(1H,m),1.90-1.98(2H,m),2.52(1H,dd,J=13.6Hz,7.2Hz),2.88-2.96(2H,m),5.26-5.29(1H,m),7.18-7.20(1H,m),7.28-7.35(2H,m),7.41-7.43(1H,m).
步骤4:3-(4-氨基-3-(苯氧基苯基)-1H吡唑[3,4-d]嘧啶-1-基)-2,3-二氢螺[茚-1,4'-哌啶]-1'-基)丙-2-烯-1-甲酸叔丁酯(1f)的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(304mg,1.00mmol)、3-羟基-2,3-二氢螺[茚-1,4'-哌啶]-1'-羧酸叔丁酯(456mg,1.50mmol)和PPh3(515mg,1.50mmol)的20mL THF溶液中(0℃)慢慢滴加DIAD(303mg,1.50mmol),该反应混合物在室温下搅拌3h后慢慢滴加水淬灭反应,EA萃 取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1~10:1)纯化得到280mg目标化合物,为白色固体,产率为47.6%。
步骤5:1-(2,3-二氢螺[茚-1,4'-哌啶]-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(1g)的制备;
在室温下,向装有3-(4-氨基-3-(苯氧基苯基)-1H吡唑[3,4-d]嘧啶-1-基)-2,3-二氢螺[茚-1,4'-哌啶]-1'-基)丙-2-烯-1-甲酸叔丁酯(500mg,0.85mmol)的20mLDCM溶液中慢慢滴加TFA(2mL),该反应混合物在室温下搅拌2h,然后慢慢滴加NaHCO3(1mol/L)至pH大于7,分出有机相,水相用DCM萃取(20mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=10:1~1:1)纯化得到320mg目标化合物,为白色固体,产率为77.1%;
步骤6:1-(3-(氨基-3-(4-苯氧基苯基)-3a,7a-1H-吡唑[3,4-d]嘧啶-1-基)-2,3-二氢螺[茚-1,4'-哌啶]-1'-基)丙-2-烯-1-酮(1)的制备
在氮气保护下,向装有1-(2,3-二氢螺[茚-1,4'-哌啶]-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(320mg,0.65mmol)、TEA(132mg,1.31mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(56mg,0.62mmol),该反应混合物在室温下搅拌3h,然后慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=20:1)纯化得到220mg目标化合物,为白色固体,产率为62.3%;
1H NMR(400MHz,d6-DMSO)δ1.64-1.70(2H,m),1.89-2.02(2H,m),2.55-2.58(2H,m),2.88-2.93(2H,m),3.20-3.26(0.5H,m),3.45-3.51(0.5H,m),4.10-4.15(1H,m),4.50-4.54(1H,m),5.69(1H,dd,J=10.4Hz,2.4Hz),6.14(1H,dd,J=16.4Hz,2.4Hz),6.55(1H,t,J=7.6Hz),6.85-6.92(2H,m),7.08-7.19(6H,m),7.29(1H,t,J=7.6Hz),7.36-7.43(3H,m),7.59(2H,d,J=8.8Hz),8.32(1H,s).
EM(计算值):542.2;MS(ESI)m/e(M+1H)+:543.3
实施例2
内型-1-((1R,3s,5S)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-8-氮杂桥[3.2.1]辛烷-8-基)丙-2-烯-1-酮和外型-1-((1R,3s,5S)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-8-氮杂桥[3.2.1]辛烷-8-基)丙-2-烯-1-酮的制备
Figure PCTCN2016079491-appb-000014
步骤1:N-叔丁氧羰基-去甲托品酮的制备
Figure PCTCN2016079491-appb-000015
在氮气保护下,向装有去甲托品酮盐酸盐(4.8g,29.70mmol)和TEA(4.50g,44.55mmol)的DCM(150mL)溶液中慢慢加入(Boc)2O(56mg,44.55mmol),该反应混合物在室温下搅拌过夜,反应完成后再添加水(50mL)淬灭反应,,分出有机相,水相用DCM萃取(30mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=5:1~1:1)纯化得到5.0g目标化合物,为白色固体,产率为74.6%;
步骤2:3-外型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯、3-内型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯制备
Figure PCTCN2016079491-appb-000016
在室温下,向装有N-叔丁氧羰基-去甲托品酮(2.25g,10.00mmol)的25mLTHF和10mL MeOH的混合溶液中分批加入NaBH4(756mg,20.00mmol),该反应混合物在室温下搅拌1h,反应完成后再慢慢滴加饱和NH4Cl淬灭反应,用EA萃取(40mLx3),无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=4:1~3:1)纯化得到722mg目标化合物3-外型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯,产率31.8%;得到3-内型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯1.48g,产率65.2%;
3-外型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯:1H NMR(400MHz,CDCl3)δ1.47(9H,s),1.60-1.62(4H,m),1.94-1.99(4H,m),4.06-4.14(1H,m),4.19-4.28(2H,m).
3-内型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯:1H NMR(400MHz,CDCl3)δ1.46(9H,s),1.69-1.73(2H,m),1.92-1.95(2H,m),2.02-2.17(4H,m),4.14-4.21(3H,m).
步骤3:外型-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-8-氮杂桥[3.2.1]辛烷-8甲酸叔丁酯制备
Figure PCTCN2016079491-appb-000017
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(500mg,1.65mmol)、3-内型-羟基-8-氮杂桥[3.2.1]辛烷-8-甲酸叔丁酯(568mg,2.50mmol)和PPh3(865mg,3.30mmol)的20mLTHF溶液中(0℃)慢慢滴加DIAD(667mg,3.30mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加水淬灭反应,EA萃取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1~10:1)纯化得到600mg目标化合物,为白色固体,产率为70.9%;
内型-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-8-氮杂桥[3.2.1]辛烷-8甲酸叔丁酯的制备。
Figure PCTCN2016079491-appb-000018
方法同上,为白色固体,产率为62.7%;
步骤4:外型-8-氮杂桥[3.2.1]辛烷-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺制备
Figure PCTCN2016079491-appb-000019
在室温下,向装有外型-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-8-氮杂桥[3.2.1]辛烷-8甲酸叔丁酯(512mg,1.00mmol)的20mL DCM溶液中慢慢滴加TFA(2mL),该反应混合物在室温下搅拌反应2h,然后再慢慢滴加NaHCO3(1mol/L)至pH大于7,分出有机相,水相用DCM萃取(20mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=10:1~1:1)纯化得到250mg目标化合物,为白色固体,产率为60.7%;
内型-8-氮杂桥[3.2.1]辛烷-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺制备
Figure PCTCN2016079491-appb-000020
方法同上,为白色固体,产率为30.6%;
步骤5:外型-3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-8-氮杂桥[3.2.1]辛烷-8-基)丙烷-2-烯-1-酮的制备
Figure PCTCN2016079491-appb-000021
在氮气保护下,向装有外型-8-氮杂桥[3.2.1]辛烷-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(103mg,0.25mmol)、TEA(51mg,0.50mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(22mg,0.24mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=50:1~20:1)纯化得到60mg目标化合物,为白色固体,产率为51.3%;
1H NMR(400MHz,d6-DMSO)δ1.91-2.02(5H,m),2.07-2.27(3H,m),4.68-4.69(2H,m),4.84-4.87(1H,m),5.26-5.35(1H,m),5.73(1H,dd,J=10.4Hz,2.4Hz),6.23(1H,dd,J=16.4Hz,2.4Hz),6.79(1H,dd,J=16.4Hz,10.4Hz),7.11-7.20(6H,m),7.41-7.45(2H,m),7.62(2H,d,J=8.4Hz),8.26(1H,s).
EM(计算值):466.2;MS(ESI)m/e(M+1H)+:467.2
内型-3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-8-氮杂桥[3.2.1]辛烷-8-基)丙烷-2-烯-1-酮(2)的制备
Figure PCTCN2016079491-appb-000022
方法同上,为白色固体,产率为48.5%;
1H NMR(400MHz,d6-DMSO)δ1.81-1.87(1H,m),1.92-2.09(3H,m),2.33-2.46(4H,m),4.60-4.63(2H,m),4.84-4.87(1H,m),5.73(1H,dd,J=10.4Hz,2.4Hz),6.23(1H,dd,J=16.4Hz,2.4Hz),6.78(1H,dd,J=16.4Hz,10.4Hz),6.85-6.92(2H,m),7.12-7.21(6H,m),7.42-7.46(2H,m),7.68(2H,d,J=8.4Hz),8.23(1H,s).
EM(计算值):466.2;MS(ESI)m/e(M+1H)+:467.2
实施例3
1-((1R,5S)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-9-氮杂桥[3.3.1]壬烷-9-基)丙-2-烯-1-酮的制备
合成步骤如下所示:
Figure PCTCN2016079491-appb-000023
步骤1:9-苄基-9-氮杂双环[3.3.1]壬-3-酮的制备
在冰浴下,向装有苄胺盐酸盐(3.8g,26.46mmol)、戊二醛(25%戊二醛水溶液,8.8mL,21.90mmol)的20mL水中加入1,3-丙酮二羧酸(3.2g,21.90mmol)和10%的乙酸钠(7.5mL),之后撤去冰浴,该反应混合物在室温下搅拌2h,然后在50℃下搅拌12h,然后慢慢加固体NaHCO3至pH大于7,分出有机相,水相用DCM萃取(20mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=10:1)纯化得到2.3g目标化合物,为白色固体,产率为71.9%;
步骤2:3-羰基-9-氮杂双环[3.3.1]壬烷-9-甲酸叔丁酯的制备
在室温下,向装有9-苄基-9-氮杂双环[3.3.1]壬-3-酮(2.3g,10.03mmol)的80mL EA溶液中加入(Boc)2O(2.3g,10.53mmol),10%钯碳(250mg),该反应混合物在氢气氛围下(2atm)室温下搅拌过夜,反应完成后抽滤除去钯碳,真空 蒸发后通过色谱法(PE:EA=10:1)纯化得到1.7g目标化合物,为白色固体,产率为70.8%;
步骤3:3-羟基-9-氮杂双环[3.3.1]壬烷-9-甲酸叔丁酯的制备
在室温下,向装有3-羰基-9-氮杂双环[3.3.1]壬烷-9-甲酸叔丁酯(1.7g,7.10mmol)的25mL THF溶液中分批加入NaBH4(537mg,14.20mmol),该反应混合物在室温下搅拌1h,然后慢慢滴加饱和NH4Cl淬灭反应,用EA萃取(40mLx3),无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=10:1~3:1)纯化得到1.45g目标化合物,为无色油状物,产率84.8%;
1H NMR(400MHz,CDCl3)δ1.29-1.38(2H,m),1.45(9H,s),1.50-1.68(5H,m),2.03-2.14(1H,m),2.27-2.38(2H,m),3.66-3.70(1H,m),4.35-4.38(1H,m),4.48-4.50(1H,m).
步骤4:3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-9-氮杂双环[3.3.1]壬烷-9-甲酸叔丁酯的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(1.2g,3.96mmol)、3-羟基-9-氮杂双环[3.3.1]壬烷-9-甲酸叔丁酯(1.5g,6.22mmol)和PPh3(2.1g,7.92mmol)的20mL THF溶液中(0℃)慢慢滴加DIAD(1.6g,7.92mmol),该反应混合物在室温下搅拌3h,然后慢慢滴加水淬灭反应,EA萃取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1~10:1)纯化得到800mg目标化合物,为白色固体,产率为38.1%;
步骤5:1-(9-氮杂双环[3.3.1]壬烷-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺的制备
在室温下,向装有3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-9- 氮杂双环[3.3.1]壬烷-9-甲酸叔丁酯(700mg,1.33mmol)的20mL DCM溶液中慢慢滴加TFA(2mL),该反应混合物在室温下搅拌2h,然后慢慢滴加NaHCO3(1mol/L)至pH大于7,分出有机相,水相用DCM萃取(20mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=10:1~1:1)纯化得到450mg目标化合物,为白色固体,产率为79.4%;
步骤6:1-(3-(4-胺-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-9-氮杂双环[3.3.1]壬烷-9-基)丙烷-2-烯-1-酮的制备
在氮气保护下,向装有1-(9-氮杂双环[3.3.1]壬烷-3-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(400mg,0.94mmol)、TEA(190mg,1.88mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(81mg,0.89mmol),该反应混合物在室温下搅拌3h,然后向上述反应混合物中慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=50:1~20:1)纯化得到90mg目标化合物,为白色固体,产率为19.9%。
1H NMR(400MHz,d6-DMSO)δ1.77-1.92(5H,m),2.03-2.13(3H,m),2.30-2.38(2H,m),4.51(1H,s),4.86(1H,s),5.70(1H,dd,J=10.4Hz,2.4Hz),5.77-5.85(1H,m),6.17(1H,dd,J=16.4Hz,2.4Hz),6.87(1H,dd,J=16.4Hz,10.4Hz),7.10-7.21(6H,m),7.41-7.45(2H,m),7.64(2H,d,J=8.8Hz),8.27(1H,s).
EM(计算值):480.2;MS(ESI)m/e(M+1H)+:481.3
实施例4
1-(3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺[4.5]葵烷-8-基)丙-2-烯-1-酮的制备
合成步骤如下所示:
Figure PCTCN2016079491-appb-000024
步骤1:4-烯丙基-4羟基哌啶-1-甲酸叔丁酯的制备
在室温下氮气氛围中,向装有镁粉(3.2g,13.33mmol)的20mL干乙醚溶液中慢慢滴加少量烯丙基溴,该反应混合物加热搅拌,等反应引发后继续慢慢滴加烯丙基溴,滴加速度控制在维持反应回流状态,共加入烯丙基溴(13.3g,0.11mol),反应完成以后冷却至-15℃,慢慢加入N-叔丁氧羰基-4-哌啶酮(10.0g,50.19mmol)的THF溶液(100mL),反应体系在室温下搅拌5h,反应完成之后冷却至0℃,然后慢慢加入饱和NH4Cl萃灭反应,分出有机相,水相用EA萃取(30mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后得到12g目标化合物,为淡黄色油状物,产率为98.9%,所得目标化合物直接用于下一步反应,无须进一步纯化;
步骤2:3-羟基-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯的制备
在室温下,向装有4-烯丙基-4羟基哌啶-1-甲酸叔丁酯(11.3g,46.82mmol) 的100mL叔丁醇和40mL水的混合溶液中慢慢加入NaIO4(11.1g,51.51mmol),该反应混合物在50℃下搅拌0.5h,然后慢慢滴加Na2S2O5(9.8g,51.55mmol)的水溶液30mL,反应混合物在50℃下再反应7h,室温搅拌48h,然后再慢慢加入水萃灭反应,EA萃取(50mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=10:1~1:1)纯化得到2.5g目标化合物,为黄色油状物,产率为20.8%;
1H NMR(400MHz,CDCl3)δ1.46(9H,s),1.48-1.53(2H,m),1.4-1.56(2H,m),1.64-1.71(2H,m),1.96(1H,dd,J=13.6Hz,6.4Hz),3.3-3.38(2H,m),3.59-3.60(1H,m),3.80-3.83(1H,m),3.92(1H,dd,J=10.0Hz,4.0Hz),4.50-4.52(1H,m).
步骤3:3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(1.2g,3.96mmol)、3-羟基-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯(1.6g,6.22mmol)和PPh3(2.1g,7.92mmol)的20mL THF溶液中(0℃)慢慢滴加DIAD(1.6g,7.92mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加水淬灭反应,EA萃取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1~10:1)纯化得到1.3g目标化合物,为白色固体,产率为60.5%;
1H NMR(400MHz,d6-DMSO)δ1.47(9H,s),1.58-1.83(4H,m),2.36-2.38(2H,m),3.28-3.29(2H,m),3.42-3.51(2H,m),4.04-4.08(1H,m),4.20-4.24(1H,m),5.53-5.57(1H,m),7.12-7.21(6H,m),7.42-7.46(3H,m),7.66(3H,d,J=8.4Hz),8.25(1H,s).
步骤4:3-(4-苯氧基苯基)-1-(1-氧杂-8-氮杂螺[4.5]癸烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺的制备
在室温下,向装有3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯(1.5g,2.76mmol)的40mL DCM溶液中慢慢滴加TFA(5mL),该反应混合物在室温下搅拌2h,然后慢慢滴加NaHCO3(1mol/L)至pH大于7,分出有机相,水相用DCM萃取(20mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=10:1~1:1)纯化得到1.0g目标化合物,为白色固体,产率为82.0%;EM(计算值):442.2;MS(ESI)m/e(M+1H)+:443.2
步骤5:1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-1-氧杂-8-氮杂螺[4.5]癸烷-8-yl)丙烷-2-烯-1-酮的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1-(1-氧杂-8-氮杂螺[4.5]癸烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺(751mg,1.70mmol)、TEA(344mg,3.40mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(139mg,1.53mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=30:1)纯化得到350mg目标化合物,为白色固体,产率为41.5%;
1H NMR(400MHz,d6-DMSO)δ1.66-1.94(4H,m),2.44(2H,d,J=7.2Hz),3.29-3.26(1H,m),3.57-3.59(1H,m),3.73-3.77(1H,m),3.89-3.91(1H,m),4.14(1H,dd,J=8.8Hz,6.0Hz),4.28-4.32(1H,m),5.60-5.64(1H,m),5.71(1H,d,J=10.0Hz),6.14(1H,dd,J=16.8Hz,2.0Hz),6.85-6.91(1H,m),7.18-7.27(5H,m),7.49(2H,t,J=8.0Hz),7.72(2H,d,J=8.8Hz),8.31(1H,s).
EM(计算值):496.2;MS(ESI)m/e(M+1H)+:497.3
实施例5
1-(3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-4-羟基-3,4-二氢喹啉-1(2H)-基)丙-2-烯-1-酮的制备
合成步骤如下所示:
Figure PCTCN2016079491-appb-000025
步骤1:喹啉-1(2H)-羧酸叔丁酯的制备
在氮气保护下,向装有喹啉(9.0g,69.68mmol)70mL THF溶液中在0℃下慢慢滴加DIBAl(76.36mL,76.6mmol,1mol/L的THF溶液),滴加时保持反应温度低于45℃,之后室温反应1h,TLC检测完成以后向上述反应混合物中加入TEA(12mL,86.50mmol),再滴加入(Boc)2O(19.0g,87.06mmol)的甲苯溶液40mL,加入时也保持反应温度低于45℃,之后继续向反应体系中加入DMAP(8.5g,69.58mmol)并于室温下搅拌2h,TCL检测反应完成以后用稀HCl调pH值至3~5,EA萃取(50mLx3),真空蒸发后通过色谱法(PE:EA=200:1)纯化得到2.0g目标化合物,为淡黄色油状物,产率为12.4%;
1H NMR(400MHz,CDCl3)δ1.52(9H,s),4.35-4.37(2H,m),5.97-6.01(1H,m),6.47(1H,d,J=8.4Hz),7.03-7.05(2H,m),7.15-7.19(1H,m),7.55(1H,d,J=8.0Hz).
步骤2:4-溴-3-羟-3,4-二氢喹啉-1(2H)-羧酸叔丁酯的制备
在室温下,向装有喹啉-1(2H)-羧酸叔丁酯(3.3g,14.27mmol)的53mL DMSO和0.53mL水的混合溶液中分批加入NBS(3.8g,21.35mmol),该反应混合物在室温下搅拌0.5h,然后慢慢滴加NaHCO3(1mol/L)至pH大于7,EA萃取(40mLx3),合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=30:1~10:1)纯化得到1.0g目标化合物,为黄色油状物,产率为46.6%;
1H NMR(400MHz,CDCl3)δ1.40(9H,s),2.95(1H,d,J=14.4Hz),3.78-3.83(2H,m),4.75(1H,d,J=14.4Hz),7.02-7.06(2H,m),7.19-7.24(1H,m),7.29-7.31(2H,m).
步骤3:3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-4-羟基-3,4-二氢喹啉-1(2H)-羧酸叔丁酯的制备
反应混合物3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(1.2g,3.96mmol)、4-溴-3-羟-3,4-二氢喹啉-1(2H)-羧酸叔丁酯(1.3g,3.96mmol)、K2CO3(1.6g,11.88mmol)在15mL DMF中于90℃搅拌过夜,反应完成以后冷却至室温,向上述反应混合物中加入水(50mL),EA萃取(40mLx3),合并上述有机相用用水和饱和食盐水洗,有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=70:1~60:1)纯化得到504mg目标化合物,为白色固体,产率为23.1%;
1H NMR(400MHz,d6-DMSO)δ1.50(9H,s),3.64(1H,dd,J=13.2Hz,9.2Hz),4.25(1H,dd,J=12.8Hz,4.0Hz),4.41-4.45(1H,m),5.69(1H,d,J=5.2Hz), 5.87(1H,d,J=7.2Hz),6.64(1H,d,J=7.6Hz),6.94(1H,t,J=7.6Hz),7.09-7.22(6H,m),7.40-7.44(2H,m),7.60-7.67(3H,m),8.31(1H,s).
步骤4:3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1,2,3,4-四氢喹啉-4-醇的制备
在室温下,向装有3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-4-羟基-3,4-二氢喹啉-1(2H)-羧酸叔丁酯(445mg,0.81mmol)的100mL圆底烧瓶中加入5mL DCM溶液使其完全溶解,然后再加入60mL饱和HCl的EA溶液,该反应混合物在室温下搅拌过夜,反应完成后真空蒸发至干,再用100mL DCM溶解,此DCM有机相分别用饱和NaHCO3和饱和食盐水洗涤,有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=70:1~65:1)纯化得到216mg目标化合物,为淡黄色固体,产率为59.2%;
1H NMR(400MHz,d6-DMSO)δ3.22(1H,t,J=10.8Hz),3.42-3.47(1H,m),4.46-4.51(1H,m),5.31(1H,d,J=5.2Hz),5.84(1H,d,J=8.4Hz),5.97(1H,d,J=2.4Hz),6.23(1H,d,J=7.6Hz),6.35(1H,t,J=7.6Hz),6.55(1H,J=7.6Hz),6.90(1H,t,J=7.6Hz),7.09-7.19(5H,m),7.40-7.44(2H,m),7.61-7.63(2H,m),8.28(1H,s).
步骤5:1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-4-羟基-3,4-二氢喹啉-1(2H)-基)丙烷-2-烯-1-酮的制备
在氮气保护下,向装有3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1,2,3,4-四氢喹啉-4-醇(300mg,0.67mmol)、Cs2CO3(437mg,1.34mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(61mg,0.67mmol),该反应混合物在室温下搅拌2h,然后慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl 洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=25:1)纯化得到30mg目标化合物,为淡黄色固体,产率为8.9%。
1H NMR(400MHz,d6-DMSO)δ3.84(1H,dd,J=12.8Hz,7.6Hz),4.29-4.33(1H,m),4.50-4.52(1H,m),5.77-5.82(2H,m),5.88(1H,d,J=6.8Hz),6.29(1H,dd,J=16.8Hz,2.0Hz),6.67(1H,dd,J=16.0Hz,10.0Hz),6.76(1H,d,J=7.6Hz),7.06-7.19(7H,m),7.26-7.29(2H,m),7.40-7.44(2H,m),7.62(2H,d,J=8.4Hz),8.29(1H,s).
EM(计算值):504.2;MS(ESI)m/e(M+1H)+:505.2
实施例6
1-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)螺[色满-2,4'-哌啶]-1'-基)丙-2-烯-1-酮的合成步骤如下所示:
Figure PCTCN2016079491-appb-000026
步骤1:4-羰基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯的制备
在室温下,向装有邻羟基苯乙酮(6.8g,49.94mmol)的75mL甲醇溶液中依次加入N-叔丁氧羰基-4-哌啶酮(10.0g,49.94mmol)、四氢吡咯(3.6g,49.94mmol),之后此反应混合物在室温搅拌过夜,反应完成以后用HCl(1mol/L)萃灭,用EA 萃取(100mL x3)合并有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=20:1~10:1)纯化得到11.1g目标化合物,为无色油状物,产率为70.0%;
步骤2:4-羟基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯的制备
在室温下,向装有4-羰基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯(4.8g,15.13mmol)的37.5mL THF和15mL MeOH的混合溶液中分批加入NaBH4(1.1g,30.26mmol),该反应混合物在室温下搅拌1h,然后慢慢滴加饱和NH4Cl淬灭反应,用EA萃取(40mLx3),无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=20:1~3:1)纯化得到3.8g目标化合物,为黄色油状物,产率79.2%;
1H NMR(400MHz,CDCl3)δ1.46(9H,s),1.49-1.70(3H,m),1.78-1.97(4H,m),2.14(1H,d,J=13.6Hz),3.17-3.26(2H,m),3.83-3.86(2H,m),4.88(1H,q,J=6.8Hz),6.85(1H,d,J=8.0Hz),6.96(1H,t,J=7.2Hz),7.20(1H,t,J=7.2Hz),7.43(1H,d,J=7.6Hz).
EM(计算值):319.2;MS(ESI)m/e(M+1H)+:320.3
步骤3:4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(1.5g,4.95mmol)、4-羟基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯(3.2g,9.90mmol)和PPh3(2.3g,7.43mmol)的90mL THF溶液中(0℃)慢慢滴加DIAD(1.5g,7.43mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加水淬灭反应,DCM萃取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=100:1)纯化得到880mg目标化合物,为白色固体,产率为29.1%;EM(计算值):604.3;MS(ESI)m/e(M+1H)+:605.3;
步骤4:3-(4-苯氧基苯基)-1-(螺[色满-2,4'-哌啶]-4-基)-1H-吡唑[3,4-d]嘧啶-4-胺的制备
在室温下,向装有4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯(850mg,1.41mmol)的150mL圆底烧瓶中加入10mL DCM溶液使其完全溶解,然后再加入100mL饱和HCl的EA溶液,该反应混合物在室温下搅拌过夜,反应完成后真空蒸发至干,再用100mL DCM溶解,此DCM有机相分别用饱和NaHCO3和饱和食盐水洗涤,有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=40:1~5:1)纯化得到570mg目标化合物,为淡黄色固体,产率为80.2%;
步骤5:1-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)螺[色满-2,4'-哌啶]-1'-左右)丙-2-烯-1-酮的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1-(螺[色满-2,4'-哌啶]-4-基)-1H-吡唑[3,4-d]嘧啶-4-胺(564mg,1.12mmol)、TEA(226mg,2.24mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(101mg,1.12mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1)纯化得到350mg目标化合物,为白色固体,产率为55.9%。
1H NMR(400MHz,d6-DMSO)δ1.72-1.83(2H,m),1.89-1.99(2H,m),2.38(1H,q,J=6.4Hz),2.68-2.74(1.5H,m),3.00-3.07(0.5H,m),3.58-3.64(1H,m),3.88-3.98(1H,m),4.18-4.27(1H,m),5.69(1H,dd,J=10.4Hz,2.4Hz),6.12(1H,dd,J=16.4Hz,2.0Hz),6.17-6.20(1H,m),6.47(1H,d,J=7.6Hz),6.74-6.78(1H,m),6.79-6.88(1H,m),6.92(1H,d,J=8.0Hz),7.09-7.19(7H,m),7.39-7.44(2H,m), 7.61-7.63(2H,m),8.31(1H,s).
EM(计算值):558.2;MS(ESI)m/e(M+1H)+:559.3
实施例7
1-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-7-氟螺[色满-2,4'-哌啶]-1'-基)丙-2-烯-1-酮的合成步骤如下所示:
Figure PCTCN2016079491-appb-000027
步骤1:7-氟-4-羰基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯的制备
在室温下,向装有4-氟-2-羟基苯乙酮(3.4g,20.76mmol)的75mL甲醇溶液中依次加入N-叔丁氧羰基-4-哌啶酮(4.4g,20.76mmol)、四氢吡咯(1.6g,20.76mmol),之后此反应混合物在室温搅拌过夜,反应完成以后用HCl(1M)萃灭,用EA萃取(100mLx3)合并有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=20:1~10:1)纯化得到6.5g目标化合物,为无色油状物,产率为88.2%;
步骤2:7-氟-4-羟基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯的制备
在室温下,向装有7-氟-4-羰基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯(2.8g,8.35mmol)的25mL THF和10mL MeOH的混合溶液中分批加入NaBH4(629mg,16.64mmol),该反应混合物在室温下搅拌1h,然后慢慢滴加饱和NH4Cl淬灭反 应,用EA萃取(30mLx3),无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=20:1~3:1)纯化得到2.8g目标化合物,为粽色油状物,产率97.2%;
步骤3:4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-7-氟螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(2.0g,6.59mmol)、7-氟-4-羰基螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯(5.6g,16.48mmol)和PPh3(2.7g,10.43mmol)的90mL THF溶液中(0℃)慢慢滴加DIAD(2.0g,10.43mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加水淬灭反应,DCM萃取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=100:1~50:1)纯化得到1.8g目标化合物,为淡黄色固体,产率为43.9%;EM(计算值):622.3;MS(ESI)m/e(M+1H)+:623.3;
步骤4:1-(7-氟螺[色满-2,4'-哌啶]-4-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺的制备
在室温下,向装有4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-7-氟螺[色满-2,4'-哌啶]-1'-羧酸叔丁酯(1.5g,2.41mmol)的150mL圆底烧瓶中加入10mL DCM溶液使其完全溶解,然后再加入100mL饱和HCl的EA溶液,该反应混合物在室温下搅拌过夜,反应完成后真空蒸发至干,再用100mL DCM溶解,此DCM有机相分别用饱和NaHCO3和饱和食盐水洗涤,有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=10:1~5:1)纯化得到824mg目标化合物,为淡黄色固体,产率为65.4%;
步骤5:1-(4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-7-氟螺[色满-2,4'-哌啶]-1'-基)丙烷-2-烯-1-酮的制备
在氮气保护下,向装有1-(7-氟螺[色满-2,4'-哌啶]-4-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(597mg,1.14mmol)、TEA(231mg,2.28mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(103mg,1.14mmol),该反应混合物在室温下搅拌3h,然后向上述反应混合物中慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1~40:1)纯化得到380mg目标化合物,为白色固体,产率为57.8%。
1H NMR(400MHz,d6-DMSO)δ1.71-1.83(2H,m),1.90-1.99(2H,m),2.40(1H,q,J=6.4Hz),2.67-2.72(1H,m),3.01-3.07(0.5H,m),3.24-3.27(0.5H,m),3.57-3.62(1H,m),3.88-3.97(1H,m),4.18-4.26(1H,m),5.69(1H,dd,J=10.4Hz,2.4Hz),6.10-6.18(2H,m),6.49-6.52(1H,m),6.62(1H,ddd,J=11.2Hz,8.8Hz,2.4Hz),6.79-6.91(2H,m),7.09-7.20(6H,m),7.40-7.44(2H,m),7.61-7.63(2H,m),8.31(1H,s).
EM(计算值):622.3;MS(ESI)m/e(M+1H)+:623.3.
实施例8
1-(3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-7-氮杂螺[4.4]壬-7-基)丙-2-烯-1-酮的合成步骤如下所示:
Figure PCTCN2016079491-appb-000028
步骤1:3-烯丙基-3-羟基吡咯-1-羧酸叔丁酯的制备
在氮气保护下,向装有1-叔丁氧碳基-3-吡咯烷酮(5.0g,26.99mmol)、烯丙基溴(8.2g,67.48mmol)的20mL THF溶液中(0℃)加入饱和NH4Cl(28mL),然后在此温度下分批加入锌粉(3.8g,67.48mmol),在加入过程中保持反应温度不超过40℃,该反应混合物在室温下搅拌12h,TLC检测反应完成,然后再慢慢滴加2mol/L H2SO4淬灭反应,EA萃取(40mLx3),合并有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=1:1)纯化得到3.8g目标化合物,为白色固体,产率为62.0%;
1H NMR(400MHz,CDCl3)δ1.46(9H,s),1.84-1.87(2H,m),1.92-1.95(1H,m),2.40(2H,d,J=7.6Hz),3.24-3.39(2H,m),3.46-3.53(2H,m),5.18-5.22(2H,m),5.84-5.90(1H,m).
步骤2:3-(2,3-二溴丙基)-3-羟基吡咯-1-羧酸叔丁酯的制备
在氮气保护下,向装有3-烯丙基-3-羟基吡咯-1-羧酸叔丁酯(1.0g,4.40mmol)的10mL DCM溶液中于-15℃下慢慢滴加Br2(703mg,4.40mmol),之后再室温搅 拌0.5h,TLC检测反应完成以后,慢慢加入饱和Na2S2O3萃灭反应,DCM萃取(30mLx3),合并有机相后用饱和NaHCO3洗涤,无水Na2SO4充分干燥,真空蒸发后至干得到1.0g目标化合物粗品,为黄色油状物,直接用于下一步反应无需进一步纯化;
步骤3:3-溴-1-羰基-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯的制备
在氮气保护下,向装有3-(2,3-二溴丙基)-3-羟基吡咯-1-羧酸叔丁酯的10mLMeOH溶液中于室温下加入固体K2CO3(857mg,6.21mmol),之后再室温搅拌过夜,TLC检测反应完成以后,向上述反应混合物中加入水萃灭反应,DCM萃取(20mLx3),合并有机相后用无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=10:1)纯化得到340mg目标化合物,为淡黄色固体,产率为25.2%;
步骤4:3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯的制备
反应混合物3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(600mg,1.98mmol)、3-溴-1-羰基-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯(600mg,1.98mmol)、Cs2CO3(2.0g,5.94mmol)在20mL DMF中于85℃搅拌过夜,反应完成以后冷却至室温,向上述反应混合物中加入水(40mL),EA萃取(30mLx3),合并上述有机相用用水和饱和食盐水洗,有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=40:1~15:1)纯化得到304mg目标化合物,为白色固体,产率为29.3%;
步骤5:3-(4-苯氧基苯基)-1-(1-氧代-7-氮杂螺[4.4]壬烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺的制备
在室温下,向装有3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯(1.0g,1.89mmol)的500mL圆底烧瓶中加入60mL DCM溶液使其完全溶解,然后再加入200mL饱和HCl的EA溶液,该反应混合物在室温下搅拌过夜,反应完成后真空蒸发至干,再用300mL DCM溶解,此DCM有机相分别用饱和NaHCO3和饱和食盐水洗涤,有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=10:1~5:1)纯化得到650mg目标化合物,为淡黄色固体,产率为80.1%;
步骤6:1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-基)丙烷-2-烯-1-酮的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1-(1-氧代-7-氮杂螺[4.4]壬烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺(300mg,0.70mmol)、TEA(141mg,1.40mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(64mg,0.70mmol),该反应混合物在室温下搅拌3h,然后向上述反应混合物中慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=60:1~40:1)纯化得到102mg目标化合物,为白色固体,产率为30.2%。
1H NMR(400MHz,d6-DMSO)δ1.95-2.19(2H,m),2.53-2.67(2H,m),3.38-3.41(1H,m),3.44-3.51(1H,m),3.73-3.78(1H,m),3.80-3.83(0.5H,m),3.93-3.95(0.5H,m),4.03-4.09(1H,m),4.22-4.28(1H,m),5.54-5.59(1H,m),5.64(1H,ddd,J=12.4Hz,10.0Hz,2.0Hz),6.12(1H,ddd,J=11.6Hz,9.2Hz,2.0Hz),6.49(0.5H,dd,J=16.8Hz,10.0Hz),6.59(0.5H,dd,J=16.8Hz,10.4Hz),7.12-7.21(6H,m),7.44(2H,t,J=8.0Hz),7.69(2H,d,J=8.4Hz),8.26(1H,s).
EM(计算值):682.2;MS(ESI)m/e(M+1H)+:483.3
实施例9
1-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-2-氮杂桥[4.1.0]庚烷-2-基)丙-2-烯-1-酮的合成步骤如下所示:
Figure PCTCN2016079491-appb-000029
步骤1:3-羟基-3,4-二氢吡啶-1(2H)-羧酸苄酯的制备
在-78℃下,向装有3-羟基吡啶(12.0g,0.12mol)、NaHCO3(7.8g,92.85mmol)的20mL无水MeOH溶液中慢慢分批加入NaBH4(12.6g,0.33mol),控制反应温度低于-60℃,之后再于-78℃下向该反应混合物滴加CbzCl(36.4mL,1.9mol),并在此温度下搅拌1.5h,TLC检测反应完成以后,向上述反应混合物中慢慢滴加1mol/L NaOH淬灭反应,EA萃取(50mLx4)合并有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=5:1)纯化得到11.9g目标化合物,为白色固体,产率为41.3%;
1H NMR(400MHz,CDCl3)δ1.99-2.12(2H,m),2.35-2.39(1H,m),3.60-3.71(2H,m),4.13-4.16(1H,m),4.78-4.88(1H,m),5.19(2H,d,J=3.6Hz),6.82-6.95 (1H,m),7.32-7.38(5H,m).
步骤2:4-羟基-2-氮杂双环[4.1.0]庚烷-2-羧酸叔丁酯的制备
在氮气保护下,向装有3-羟基-3,4-二氢吡啶-1(2H)-羧酸苄酯(11.9g,51.01mmol)的1.2-二氯乙烷溶液中(0℃)慢慢滴加Et2Zn(75mL,75.00mmol,1mol/L环已烷)、CH2I2(6mL,75.00mmol),此反应混合物自然升至室温并搅拌6h,TLC检测反应完成以后,再慢慢滴加饱和NaHCO3溶液淬灭反应,DCM萃取(50mLx4)合并有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(PE:EA=3:1~1:1)纯化得到11.0g目标化合物,为白色固体,产率为94.4%;
1H NMR(400MHz,CDCl3)δ0.64-0.77(1H,m),0.84-0.97(1H,m),1.04-1.14(1H,m),1.90-2.01(3H,m),2.86-2.93(2H,m),3.87(1H,dd,J=13.2Hz,4.8Hz),3.97-4.05(1H,m),5.20(2H,s),7.30-7.40(5H,m).
步骤3:4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-2-氮杂双环[4.1.0]庚烷-2-羧酸苄酯的制备
在氮气保护下,向装有3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(B-H)(2.0g,6.59mmol)、4-羟基-2-氮杂双环[4.1.0]庚烷-2-羧酸叔丁酯(2.1g,8.57mmol)和PPh3(2.6g,9.89mmol)的90mL THF溶液中(0℃)慢慢滴加DIAD(2.0g,9.89mmol),该反应混合物在室温下搅拌3h,然后向上述反应混合物中慢慢滴加水淬灭反应,DCM萃取(50mLx3),有机相用无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=50:1~20:1)纯化得到1.1g目标化合物,为白色固体,产率为31.4%;EM(计算值):532.2;MS(ESI)m/e(M+1H)+:533.3;
步骤4:1-(2-氮杂双环[4.1.0]庚烷-4-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4- 胺的制备
在2atm氢气压力下,反应体系4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-2-氮杂双环[4.1.0]庚烷-2-羧酸苄酯(610mg,1.15mmol)、Pd-C(150mg,10%W/W)在20mL MeOH中于室温搅拌过夜,反应完成之后,抽滤除去Pd-C,真空蒸发后通过色谱法(DCM:MeOH=40:1~10:1)纯化得到110mg目标化合物,为白色固体,产率为24.0%;EM(计算值):398.2;MS(ESI)m/e(M+1H)+:399.2;
步骤5:1-(4-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-2-氮杂双环[4.1.0]壬烷-2-基)丙烷-2-烯-1-酮的制备
在氮气保护下,向装有1-(2-氮杂双环[4.1.0]庚烷-4-基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(310mg,0.80mmol)、TEA(162mg,1.61mmol)的20mL DCM溶液中(0℃)慢慢滴加烯丙酰氯(65mg,0.72mmol),该反应混合物在室温下搅拌3h,然后再慢慢滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过色谱法(DCM:MeOH=20:1)纯化得到35mg目标化合物,为白色固体,产率为9.7%。
1H NMR(400MHz,d6-DMSO)δ0.63-0.67(1H,m),1.05-1.11(1H,m),1.54-1.62(1H,m),2.29-2.33(1H,m),2.61-2.71(1H,m),3.02(1H,t,J=11.6Hz),3.09-3.14(1H,m),4.33(1H,dd,J=12.0Hz,3.2Hz),4.57-4.65(1H,m),5.80(1H,dd,J=10.4Hz,2.4Hz),6.23(1H,dd,J=16.4Hz,2.4Hz),7.02(1H,dd,J=16.4Hz,10.4Hz),7.12-7.21(6H,m),7.42-7.46(2H,m),7.67(2H,d,J=8.8Hz),8.24(1H,s).
EM(计算值):452.2;MS(ESI)m/e(M+1H)+:453.2
试验例1:体外BTK抑制激酶活性试验
BTK体外抑制活性试验(试验例1和试验例2)均由上海睿智化学研究有限公司测定。
1:试验材料:
BTK(Carna产品,货号08-180,批号14CBS-0619D);
底物多肽FAM-P2(GL Biochem产品,货号112394,批号P100804-XZ112394);
腺苷三磷酸(Sigma产品,批号A7699-1G,批号987-65-5);
DMSO(Sigma产品,货号D2650,批号474382);
四乙酸二氨基乙烷(Sigma产品,货号E5134,批号60-00-4);
96-孔板(Corning产品,货号3365,批号22008026);
384-孔板(Corning产品,货号3573,批号12608008);
阳性对照物:星孢菌素(Sigma产品,货号S4400-1MG,批号046K4080);
检测仪器:Caliper EZ Reader
2:试验原理:
微流体芯片技术的迁移率检测技术(Mobility-Shift Assay),该技术将毛细管电泳的基本理念应用到微流体环境中,用于实验的底物是带有荧光标记的多肽,在反应体系中酶的作用下,底物转变为产物,其所带的电荷也发生了相应的变化,Mobility-Shift Assay正是利用底物和产物所带电荷的不同,将二者进行分离,并分别进行检测,检测结果由转化率表达出来。
3:试验方法:
(1)配置待测样品:用100%DMSO稀释至反应终浓度的50倍即25umol/L;
(2)稀释:25umol/L为起始浓度,然后以4倍浓度稀释,稀释10个浓度梯度;
(3)阳性对照和阴性对照孔中分别加入100%DMSO;
(4)将配好的10个浓度的化合物分别用1倍激酶缓冲液稀释10倍;其中激酶缓冲液中包含浓度为50mmol/L,pH为7.5的羟乙基哌嗪乙硫磺酸、0.01%的十二烷基聚乙二醇醚、10mmol/L的氯化镁、2mmol/L的二硫苏糖醇;
(5)配制2.5倍酶溶液:将激酶加入1倍激酶缓冲液,形成2.5倍酶溶液;
(6)配制2.5倍的底物溶液:将FAM标记的多肽和ATP加入1倍激酶缓冲液,形成2.5倍底物溶液;
(7)向384孔板中加入酶溶液:384孔反应板中已有5μl的10%DMSO溶解的5倍化合物,然后再加入10μl的2.5倍酶溶液,室温下孵育10分钟;
(8)向384孔板中加入底物溶液:在384孔反应板中加入10μl的2.5倍底物溶液;
(9)激酶反应和终止:28℃下孵育1h,然后加25μl终止液终止反应;其中终止液中包含浓度为100mmol/L,pH为7.5的羟乙基哌嗪乙硫磺酸、0.015%的十二烷基聚乙二醇醚、0.2%的3号表面试剂、20mmol/L的乙二胺四乙酸;
(10)Caliper读取数据Caliper上读取转化率数据;
(11)抑制率计算从Caliper上复制转化率数据。
把转化率转化成抑制率数据,其中max是指DMSO对照的转化率,min是无酶活对照的转化率。
Percent inhibition=(max-conversion)/(max-min)*100.
试验例2:选用不同的细胞株对化合物进行体外细胞增殖抑制活性的测定
1:试验材料:
RPMI1640培养基(INVITROGEN产品,货号61870-127);
胎牛血清(GIBCO产品,货号10099-141);
胰蛋白酶(GIBCO产品,货号25200-072);
发光法细胞活力检测试剂盒(Promega产品,货号G7572);
96孔细胞培养板,黑色(Costar产品,货号3904);
底部白色贴膜(PE产品,货号6005199);
DMSO(Sigma产品,货号D2650);
Paclitaxel(国内供应商)
细胞株:DOHH2、WSU-DLCL2。
2:药物配制方法:
待测药物使用DMSO配制成10mmol/L溶液,震荡溶解后备用。
3:细胞体外培养方法:
选取的两株细胞DOHH2、WSU-DLCL2使用含10%胎牛血清的RPMI1640培养液进行培养,在37℃,5%的CO2的湿润条件下孵育。
4:实验方法:
第一天:细胞铺板
(1)使用胰酶将细胞从细胞培养盘上消化下,使用完全培养基(含10%胎牛血清的RPMI1640培养液)重悬后测定细胞密度;
(2)将细胞稀释成标准密度的细胞溶液;
(3)将调整密度后的细胞溶液以每孔90微升加入96孔细胞实验板中(添加位置为2~12列,第1列补充相应体积的无细胞培养液);
(4)将铺好的细胞培养板放入孵箱,在37℃,5%的CO2湿润条件下孵育24h。
第二天:加化合物
(1)根据实验模板准备200倍浓度的参照化合物和待测试化合物溶液(化合物测试起始浓度为50umol/L,以200倍浓度10mmol/L为起始浓度使用DMSO进行梯度稀释,稀释倍数为3倍,9个浓度点;参照化合物测试起始浓度为1umol/L,以200倍浓度0.2mmol/L为起始浓度使用DMSO进行梯度稀释,稀释倍数为3倍,9个浓度点);
(2)取7微升化合物溶液加入到133微升完全培养基中进行稀释,摇匀10分钟(DMSO终浓度5%);
(3)取10微升稀释后的化合物溶液加入前一天准备好的细胞培养板中(DMSO终浓度0.5%);
(4)加入化合物的细胞培养板重新放回孵箱,在37℃,5%的CO2的湿润条件下孵育72h。
第五天:结果检测
(1)检测试剂在实验前30分钟放置于室温进行平衡;
(2)细胞培养板每孔加入30微升检测试剂、避光条件下摇板10分钟,诱导细胞裂解;
(3)10分钟后将细胞培养板在室温下孵育2分钟以稳定发光信号;
(4)实验板下方平面贴上白色贴膜;
(5)使用Envision读板,读板时时间设定为0.5秒每孔。
细胞抑制率的计算公式:
%抑制率=(最大信号值-化合物信号值)/(最大信号值-最小信号值)×100.
从二甲基亚砜处理72h的细胞得到最大的信号值;从单独的培养基(细胞数为零)得到最小信号值。
使用XLfit软件计算
部分化合物对BTK激酶抑制活性和体外细胞增殖抑制活性结果:
Figure PCTCN2016079491-appb-000030
Figure PCTCN2016079491-appb-000031
Figure PCTCN2016079491-appb-000032
从表中数据可以看出,实施例2、3、4、5、8、9都对BTK活性具有明显的抑制作用,与阳性对照物依鲁替尼作用大致相当,说明将哌啶环替换为一些螺环或桥环等化合物的抑制作用基本没有太大影响,但仍然可以表现出强效的BTK激酶抑制活性。但是在细胞水平上,所测试的两个化合物实施例4和实施例8均表现出比阳性对照物更为强效的细胞增殖抑制活性,尤其是实施例4在DOHH2细胞上的抑制活性提高了差不多8倍。因此,本发明所设计的螺环或桥环化合物可用作BTK抑制剂,具有广阔的抗恶性肿瘤的应用前景。

Claims (9)

  1. 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,其特征在于,包括式(Ⅰ):
    Figure PCTCN2016079491-appb-100001
    其中Y选自下列基团:
    Figure PCTCN2016079491-appb-100002
    其中,R1、R2可以独立地选自氢原子、氟原子、溴原子、碘原子、氨基、氰基、巯基、C1~C5直链或支链烷基;所述烷基上的一个或多个氢原子可以被一个或者多个卤素、氨基、氰基、羟基、硫基取代;
    m1、m2均为C1~C5烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、-S=O、-S(=O)2取代;
    n1、n2均为C0~C5烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、-S=O、-S(=O)2取代;
    G为C、O、S、-S=O、-S(=O)2或NR4;所述R4可以是氢、C1~C6直链或支链烷基、C1~C6直链或支链杂烷基;其中C1~C6直链或支链烷基、C1~C6直链或支链杂烷基上一个或多个氢原子可以进一步被一个或多个氨基、氰基、羟基、硫基、卤素取代。
  2. 根据权利要求1所述的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,其特征在于,包括式(Ⅱ):
    Figure PCTCN2016079491-appb-100003
    其中,R3和R4可以独立地选自氢原子、氟原子、氯原子、溴原子、碘原子、氨基、氰基、巯基、C1~C5直链或支链烷基;所述烷基上的一个或多个氢原子可以被一个或者多个卤素、氨基、氰基、羟基、巯基取代。
  3. 根据权利要求1所述的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,其特征在于,包括式(III):
    Figure PCTCN2016079491-appb-100004
    其中,m1、m2均为C1~C2烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、S=O、S(=O)2取代;
    n1、n2均为C1~C3烷基,所述烷基中的一个或者多个碳原子可被一个或者多个O、S、-C=O、-C=S、S=O、S(=O)2取代;
    G为C、O、S、S=O、S(=O)2或NR4;所述R4可以是氢、C1~C4直链或支链烷基、C1~C4直链或支链杂烷基。
  4. 根据权利要求3所述的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,其特征在于,包括式(IV):
    Figure PCTCN2016079491-appb-100005
    Figure PCTCN2016079491-appb-100006
    其中,R3和R4可以独立地选自卤素原子、C1~C3直链或支链烷基;所述烷基上的一个或多个氢原子可以被一个或者多个卤素、氨基、氰基、羟基、巯基取代。
  5. 根据权利要求1所述的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,其特征在于,选自以下结构:
    Figure PCTCN2016079491-appb-100007
    Figure PCTCN2016079491-appb-100008
    Figure PCTCN2016079491-appb-100009
  6. 根据权利要求5所述的一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂,其特征在于,选自以下结构:
    Figure PCTCN2016079491-appb-100010
  7. 如权利要求6所述的1-(3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺[4.5]癸烷-8-基)丙-2-烯-1-酮的制备方法,其特征在于,包括以下步骤:
    (1)制备4-烯丙基-4羟基哌啶-1-甲酸叔丁酯
    氮气保护下,向含有3.2g镁粉的干乙醚溶液20mL中滴加烯丙基溴,反应引发后继续滴加烯丙基溴,滴加速度控制在维持反应回流状态,两次共加入烯丙基溴13.3g,然后冷却至-15℃,加入含10.0g N-叔丁氧羰基-4-哌啶酮的THF溶液100mL,搅拌反应5h,然后冷却至0℃,再加入饱和NH4Cl萃灭反应,分出有机相,萃取,干燥,真空蒸发后得4-烯丙基-4羟基哌啶-1-甲酸叔丁酯;
    (2)制备3-羟基-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯
    向含有11.3g 4-烯丙基-4羟基哌啶-1-甲酸叔丁酯的100mL叔丁醇和40mL水的混合溶液中加入(11.1g NaIO4,50℃搅拌反应0.5h,然后加入含9.8g Na2S2O5的水溶液30mL,50℃反应7h,室温搅拌48h,最后加水萃灭反应,萃取,干燥,真空蒸发后得3-羟基-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯;
    (3)制备3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯
    氮气保护下,向含1.2g 3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺、1.6g 3-羟基-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯和2.1g PPh3的THF溶液20mL中,0℃下滴加1.6g DIAD,室温下搅拌反应3h,再加水淬灭反应,萃取,充分干燥,真空蒸发得3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯;
    (4)制备3-(4-苯氧基苯基)-1-(1-氧杂-8-氮杂螺[4.5]癸烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺
    室温下,向含有1.5g 3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-8-氮杂螺环[4.5]癸烷-8-甲酸叔丁酯的DCM溶液40mL中滴加5mL TFA,室温下搅拌反应2h,然后滴加NaHCO3至pH大于7,分出有机相,萃取,干燥,真空蒸发得3-(4-苯氧基苯基)-1-(1-氧杂-8-氮杂螺[4.5]癸烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺;
    (5)制备1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-1-氧杂-8-氮杂螺[4.5]癸烷-8-yl)丙烷-2-烯-1-酮
    氮气保护下,向含有751mg 3-(4-苯氧基苯基)-1-(1-氧杂-8-氮杂螺[4.5]癸烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺、344mg TEA的DCM溶液20mL中,0℃下滴加139mg烯丙酰氯,室温下搅拌反应3h,然后再滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后得1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-yl)-1-氧杂-8-氮杂螺[4.5]癸烷-8-yl)丙烷-2-烯-1-酮。
  8. 如权利要求6所述的1-(3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧杂-7-氮杂螺[4.4]壬-7-基)丙-2-烯-1-酮的制备方法,其特征在于,包括以下步骤:
    (1)制备3-烯丙基-3-羟基吡咯-1-羧酸叔丁酯
    氮气保护下,向含有5.0g 1-叔丁氧碳基-3-吡咯烷酮、8.2g烯丙基溴的THF溶液20mL中,0℃下加入28mL饱和NH4Cl,然后再加入3.8g锌粉,在加入过程中保持反应温度不超过40℃,然后在室温下搅拌反应12h,,最后滴加2mol/L H2SO4淬灭反应,EA萃取,合并有机相后用饱和NaCl洗涤,无水Na2SO4干燥,真空蒸发后得3-烯丙基-3-羟基吡咯-1-羧酸叔丁酯;
    (2)制备3-(2,3-二溴丙基)-3-羟基吡咯-1-羧酸叔丁酯
    氮气保护下,向含有1.0g 3-烯丙基-3-羟基吡咯-1-羧酸叔丁酯的DCM溶液10mL中于-15℃下滴加703mg Br2,之后再室温搅拌0.5h,最后加入饱和Na2S2O3萃灭反应,DCM萃取,合并有机相后用饱和NaHCO3洗涤,无水Na2SO4干燥,真空蒸发后得3-(2,3-二溴丙基)-3-羟基吡咯-1-羧酸叔丁酯;
    (3)制备3-溴-1-羰基-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯
    氮气保护下,向含有3-(2,3-二溴丙基)-3-羟基吡咯-1-羧酸叔丁酯的10mL MeOH溶液中于室温下加入857mg K2CO3,之后再室温搅拌过夜,然后加入水萃灭反应,DCM萃取,合并有机相后用无水Na2SO4干燥,真空蒸发后得3-溴-1-羰基-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯;
    (4)制备3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯
    将600mg 3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺、600mg 3-溴-1-羰基-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯、2.0g Cs2CO3在20mL DMF中于85℃搅拌过夜,反应完成后冷却至室温,然后加入水萃灭反应,EA萃取,无水Na2SO4干燥,真空蒸发后得3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯;
    (5)制备3-(4-苯氧基苯基)-1-(1-氧代-7-氮杂螺[4.4]壬烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺
    室温下,向1.0g 3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-羧酸叔丁酯中加入60mL DCM溶液使其完全溶解,然后再加入200mL饱和HCl的EA溶液,该反应混合物在室温下搅拌过夜,反应完成后真空蒸发至干,再用300mL DCM溶解,此DCM有机相分别用饱和NaHCO3和饱和食盐水洗涤,有机相用无水Na2SO4充分干燥,真空蒸发后得3-(4-苯氧基苯基)-1-(1-氧代-7-氮杂螺[4.4]壬烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺;
    (6)制备1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-基)丙烷-2-烯-1-酮
    氮气保护下,向含有300mg 3-(4-苯氧基苯基)-1-(1-氧代-7-氮杂螺[4.4]壬烷-3-基)-1H-吡唑[3,4-d]嘧啶-4-胺、141mg TEA的DCM溶液20mL中,0℃下滴加64mg烯丙酰氯,室温下搅拌反应3h,然后滴加饱和NH4Cl淬灭反应,分出有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后得1-(3-(4-胺基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)-1-氧代-7-氮杂螺[4.4]壬烷-7-基)丙烷-2-烯-1-酮。
  9. 将权利要求1-6任一项所述的具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂用于治疗炎性病症和/或恶性肿瘤。
PCT/CN2016/079491 2016-01-22 2016-04-15 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法 WO2017124657A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610046120.4A CN105732638B (zh) 2016-01-22 2016-01-22 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法
CN201610046120.4 2016-01-22

Publications (1)

Publication Number Publication Date
WO2017124657A1 true WO2017124657A1 (zh) 2017-07-27

Family

ID=56246476

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/079491 WO2017124657A1 (zh) 2016-01-22 2016-04-15 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法

Country Status (2)

Country Link
CN (1) CN105732638B (zh)
WO (1) WO2017124657A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113527332A (zh) * 2020-04-13 2021-10-22 南开大学 具有生物活性的卡波特韦衍生物及其制备方法和应用
CN114163445A (zh) * 2021-12-06 2022-03-11 重庆医科大学 拉罗替尼中间体及其制备方法
CN114736207A (zh) * 2022-04-28 2022-07-12 武汉九州钰民医药科技有限公司 一种小分子her2抑制剂的制备工艺
CN115232192A (zh) * 2022-08-22 2022-10-25 济宁医学院 一种偶氮类乏氧响应型二肽超分子单体及其制备方法和应用

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018009017A1 (en) 2016-07-07 2018-01-11 Daewoong Pharmaceutical Co., Ltd. NOVEL 4-AMINOPYRAZOLO[3,4-d]PYRIMIDINYLAZABICYCLO DERIVATIVES AND PHARMACEUTICAL COMPOSITION COMPRISING THE SAME
US11590167B2 (en) 2016-12-03 2023-02-28 Juno Therapeutic, Inc. Methods and compositions for use of therapeutic T cells in combination with kinase inhibitors
CN106831787B (zh) * 2017-01-20 2018-10-23 成都倍特药业有限公司 用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用
CN107298685A (zh) * 2017-06-29 2017-10-27 上海合全药业股份有限公司 一种8‑(叔丁氧羰基)‑1‑氧杂‑8‑氮杂螺[4.5]癸烷‑2‑羧酸的合成方法
CN108191871B (zh) * 2018-01-02 2020-02-18 成都倍特药业有限公司 新型布鲁顿酪氨酸激酶抑制剂及其制备方法和应用
CN111454268B (zh) * 2019-01-18 2023-09-08 明慧医药(上海)有限公司 作为布鲁顿酪氨酸激酶抑制剂的环状分子
CN110606848A (zh) * 2019-08-27 2019-12-24 药雅科技(上海)有限公司 一种5-氮杂吲哚衍生物Bruton′s酪氨酸激酶抑制剂及其制备方法与用途
CN111471048B (zh) * 2020-04-30 2021-06-15 成都海博为药业有限公司 一种具有含氮桥环、螺环或并环结构的化合物及其用途
CN111533752A (zh) * 2020-05-20 2020-08-14 上海合全药物研发有限公司 一种叔丁基-7-溴-5-氧杂-2-氮杂螺[3.4]辛烷-2-甲酸基酯的制备方法
CN114315820A (zh) * 2021-11-30 2022-04-12 安徽益多康尔医药科技有限公司 N-Boc-9-氮杂双环[3.3.1]壬-3-酮的制备方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014152114A1 (en) * 2013-03-15 2014-09-25 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as btk inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2526933B1 (en) * 2006-09-22 2015-02-25 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
CA2833771C (en) * 2011-06-10 2021-08-03 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
EP2548877A1 (en) * 2011-07-19 2013-01-23 MSD Oss B.V. 4-(5-Membered fused pyridinyl)benzamides as BTK-inhibitors
EP2989106B1 (en) * 2013-04-25 2016-12-14 Beigene, Ltd. Fused heterocyclic compounds as protein kinase inhibitors
US20150141438A1 (en) * 2013-11-15 2015-05-21 Pharmacyclics, Inc. Methods for delaying or preventing the onset of type 1 diabetes
CN105085474B (zh) * 2014-05-07 2018-05-18 北京赛林泰医药技术有限公司 鲁顿酪氨酸激酶抑制剂

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014152114A1 (en) * 2013-03-15 2014-09-25 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as btk inhibitors

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113527332A (zh) * 2020-04-13 2021-10-22 南开大学 具有生物活性的卡波特韦衍生物及其制备方法和应用
CN114163445A (zh) * 2021-12-06 2022-03-11 重庆医科大学 拉罗替尼中间体及其制备方法
CN114163445B (zh) * 2021-12-06 2023-06-20 重庆医科大学 拉罗替尼中间体及其制备方法
CN114736207A (zh) * 2022-04-28 2022-07-12 武汉九州钰民医药科技有限公司 一种小分子her2抑制剂的制备工艺
CN115232192A (zh) * 2022-08-22 2022-10-25 济宁医学院 一种偶氮类乏氧响应型二肽超分子单体及其制备方法和应用

Also Published As

Publication number Publication date
CN105732638B (zh) 2018-01-30
CN105732638A (zh) 2016-07-06

Similar Documents

Publication Publication Date Title
WO2017124657A1 (zh) 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法
ES2963417T3 (es) Derivados de imidazol condensados como moduladores de IL-17
CN109715634B (zh) Menin-mll相互作用的稠合二环抑制剂
US9193733B2 (en) Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
CA2818545C (en) Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
EP2220068B1 (en) 4-pyrazolyl-n-arylpyrimidin-2-amines and 4-pyrazolyl-n-heteroarylpyrimidin-2-amines as janus kinase inhibitors
DK3050882T3 (en) PIPERIDIN-4-YL-AZETIDINE DERIVATIVES AS JAK1 INHIBITORS
ES2551592T3 (es) Pirazolo[1,5-a]piridinas como inhibidores de TRK
US20220281876A1 (en) Degradation of bruton's tyrosine kinase (btk) by conjugation of btk inhibitors with e3 ligase ligand and methods of use
KR20190069529A (ko) 유비퀴틴 특이적 프로테아제 7의 억제제로서 피페리딘 유도체
AU2013347538A1 (en) Purine inhibitors of human phosphatidylinositol 3-kinase delta
KR20230057341A (ko) Jak2 v617f 억제제로서 삼환계 우레아 화합물
US11981683B2 (en) Indazole based compounds and associated methods of use
WO2022134641A1 (zh) 芳香杂环类化合物、药物组合物及其应用
EP4284365A1 (en) Cdk2 inhibitors and methods of using the same
TW202233624A (zh) Btk抑制劑
JP2020525471A (ja) Nik阻害剤としての新規の置換アザインドリン誘導体
AU774397B2 (en) Tricyclic compounds having spiro union
AU2022256725A1 (en) NEW PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS SIGMA LIGANDS
CN113227100B (zh) 噻吩并吡啶酮化合物
CN116113698A (zh) Trpml调节剂
US11951094B2 (en) TLR2 modulator compounds, pharmaceutical compositions and uses thereof
WO2024017372A1 (zh) 一种吲哚酮衍生物及其应用
US20230271942A1 (en) Cdk9 inhibitor and use thereof
WO2023172475A2 (en) Non-aromatic cyclic-pyrimidine analogs as glucosylceramide synthase inhibitors

Legal Events

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

Ref document number: 16885920

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16885920

Country of ref document: EP

Kind code of ref document: A1