WO2020063012A1 - 氨基降茨烷衍生物及其制备方法与应用 - Google Patents

氨基降茨烷衍生物及其制备方法与应用 Download PDF

Info

Publication number
WO2020063012A1
WO2020063012A1 PCT/CN2019/094864 CN2019094864W WO2020063012A1 WO 2020063012 A1 WO2020063012 A1 WO 2020063012A1 CN 2019094864 W CN2019094864 W CN 2019094864W WO 2020063012 A1 WO2020063012 A1 WO 2020063012A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
compound
mmol
ring
Prior art date
Application number
PCT/CN2019/094864
Other languages
English (en)
French (fr)
Inventor
陈荣耀
Original Assignee
南京亘泰医药技术有限公司
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 南京亘泰医药技术有限公司 filed Critical 南京亘泰医药技术有限公司
Priority to US17/279,751 priority Critical patent/US20210332057A1/en
Priority to AU2019348752A priority patent/AU2019348752B2/en
Priority to MX2021003662A priority patent/MX2021003662A/es
Priority to JP2021517486A priority patent/JP7370617B2/ja
Priority to CN201980029247.7A priority patent/CN112135826B/zh
Priority to KR1020217011382A priority patent/KR20210070304A/ko
Priority to BR112021005960-1A priority patent/BR112021005960A2/pt
Priority to CA3114259A priority patent/CA3114259A1/en
Priority to EP19866403.9A priority patent/EP3858833A4/en
Publication of WO2020063012A1 publication Critical patent/WO2020063012A1/zh
Priority to US17/574,716 priority patent/US11420975B2/en

Links

Images

Classifications

    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • the invention belongs to the field of medicine, and particularly relates to an aminonordecane derivative as a Bruton tyrosine kinase inhibitor with high selectivity to a C481S mutant, a pharmaceutical composition thereof, a preparation method thereof, and use thereof in preparing a medicine.
  • BCR B-cell receptor
  • Abnormal BCR-mediated signal transduction can cause deregulated B cell activation and / or the formation of pathogenic autoantibodies, leading to a variety of human diseases, including cancer, autoimmune diseases and xenoimmune diseases, autoimmunity Diseases include lupus erythematosus, chronic lymphocytic lymphoma, diffuse large cell lymphoma, follicular lymphoma, or chronic lymphocytic leukemia.
  • Xenoimmune diseases include inflammatory diseases, asthma, and the like.
  • BTK Bruton tyrosine kinase
  • Btk-deficient mice show a significant improvement in disease progression.
  • Btk-deficient mice are resistant to collagen-induced arthritis (Jansson and Holmdahl, Clin Exp Immunol 1993 94: 459).
  • Selective Btk inhibitors have a clear dose-effect relationship in mouse arthritis models (Pan et al., Chem. Med. Chem. 2007 2: 58-61). Clinical studies on Btk inhibitors for arthritis are ongoing.
  • Ibrutinib (Imbruvica) was the first Bruton tyrosine kinase (BTK) inhibitor to enter the market with great success, with sales of $ 2.6 billion in 2017. However, like many other anticancer drugs, some patients show resistance to the drug.
  • BTK Bruton tyrosine kinase
  • Ibrutinib is pharmacologically active through irreversible covalent binding to the C481 tryptophan residue of BTK kinase.
  • the C481S mutation changes tryptophan It becomes serine and loses its ability to covalently bind to Ibrutinib.
  • BTK (C481S) mutations account for 87% of patients with relapsed chronic lymphoma (CLL) (Woyach et al., JClin Oncol 2017: 35: 1437-1443). Among patients with mantle cell lymphoma (MCL) relapses ⁇ 80 % Is a BTK (C481S) mutation (Chiron et al. Cancer Discovery 2014 (9): 1-14).
  • MCL mantle cell lymphoma
  • the technical problem to be solved by the present invention is to provide a novel, unreported compound that has a highly selective Bruton tyrosine kinase inhibitor for BTK (C481S) mutants, and its pharmaceutically acceptable salts, solvates, Active metabolites, polymorphs, esters, optical isomers or prodrugs, the use of the compounds in pharmaceuticals, and methods of using the compounds of the invention to prevent or treat diseases associated with excessive Btk activity in humans or mammals.
  • C481S Bruton tyrosine kinase inhibitor for BTK
  • the technical solutions adopted by the present invention are:
  • the A ring is selected from one of the following structures:
  • R 5 is selected from hydrogen, halogen, cyano, hydroxy, alkynyl, amino, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, C 1-3 haloalkoxy, C 1 -3 haloalkylamino, C 3-7 cycloalkyl, C 3-7 cycloalkoxy, C 3-7 cycloalkylamino;
  • Ring B is a substituted or unsubstituted aromatic or heteroaryl ring
  • Ring C is a substituted or unsubstituted aromatic or heteroaryl ring
  • L is a single bond, or one of the following structures
  • R 1 is selected from R 3 or one of the following structures,
  • R 3 is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 1-6 alkynyl, substituted or unsubstituted C 1-6 alkenyl, substituted or unsubstituted C 6-10 aryl, substituted or unsubstituted C 1-9 heteroaryl, substituted or unsubstituted C 3-7 cycloalkyl or substituted or unsubstituted C 2-7 heterocycloalkylamino;
  • R 4 is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 6-10 aryl, substituted or unsubstituted C 1-9 heteroaryl, substituted or unsubstituted C 3 -7 cycloalkyl, substituted or unsubstituted C 3-7 heterocycloalkyl;
  • R 2 is selected from H, substituted or unsubstituted C 1-3 alkyl, substituted or unsubstituted C 3-7 cycloalkyl, substituted or unsubstituted C 2-7 heterocycloalkyl, substituted or unsubstituted C 6-10 aryl or substituted or unsubstituted C 1-9 heteroaryl.
  • R 1 , R 2 and the N to which they are attached form a substituted or unsubstituted C 2-7 heterocyclic ring, and R 3 , R 4 and N to which they are attached form or do not form a C 3-7 heterocyclic amino or C 3- 9 heteroaryl ring amino.
  • the R 3 is substituted C 1-6 alkyl, substituted C 1-6 alkynyl group, a substituted C 1-6 alkenyl group, a substituted C 6-10 aryl group, a substituted C 1- 9 Heteroaryl, substituted C 3-7 cycloalkyl, substituted C 2-7 heterocycloalkyl are selected from halogen, cyano, hydroxyl, amino, substituted or unsubstituted acylamino, substituted Or unsubstituted aminoacyl, substituted or unsubstituted C 1-4 alkyl, substituted or unsubstituted C 3-7 cycloalkyl, substituted or unsubstituted C 3-7 cycloalkoxy, substituted or unsubstituted C 1-4 alkylamino, di [substituted or unsubstituted C 1-4 alkyl] amino, substituted or unsubstituted C 3-7 cycloalkylamino, substituted or un
  • the substituent in the 3-7 heterocycloalkyl is selected from the group consisting of halogen, hydroxy, cyano, amino, substituted or unsubstituted C 1-4 alkenyl, substituted or unsubstituted C 3-7 cycloalkyl, substituted or unsubstituted Substituted C 3-7 cycloalkoxy, substituted or unsubstituted C 1-4 alkylamino, di [substituted or unsubstituted C 1-4 alkyl] amino, substituted or unsubstituted C 3-7 ring Alkylamino, substituted or unsubstituted C 3-7 heterocycloalkylamino, substituted or unsubstituted C 1-3 alkoxy, substituted or or
  • the A ring structure is as follows:
  • R 5 is selected from hydrogen, halogen, cyano, hydroxy, alkynyl, amino, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, C 1-3 haloalkoxy, C 1 -3 haloalkylamino, C 3-7 cycloalkyl, C 3-7 cycloalkoxy, C 3-7 cycloalkylamino;
  • the substituent in the substituted aromatic ring or aromatic heterocycle in the B ring is selected from the group consisting of halogen, hydroxy, cyano, amino, C 1-3 alkyl, C 1-3 alkoxy, and halogenated C One or more of 1-3 alkyl, haloC 1-3 alkoxy.
  • the B ring structure is as follows:
  • the substituent in the substituted aromatic ring or aromatic heterocycle in the C ring is selected from the group consisting of halogen, hydroxyl, cyano, amino, C 1-3 alkyl, C 1-3 alkoxy, and halogenated C One or more of 1-3 alkyl, haloC 1-3 alkoxy.
  • the C-ring structure is as follows:
  • R 2 is selected from H
  • R 1 is selected from:
  • R 3 is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 1-6 alkynyl, substituted or unsubstituted C 1-6 alkenyl, substituted or unsubstituted C 6-10 aryl, substituted or unsubstituted C 1-9 heteroaryl, substituted or unsubstituted C 3-7 cycloalkyl, substituted or unsubstituted C 2-7 heterocycloalkylamino.
  • the compound is selected from any one of the following structures:
  • heterologous immune disease autoimmune disease or cancer is associated with excessive Bruton tyrosine kinase activity.
  • the heterogeneous immune disease, autoimmune disease or cancer is associated with abnormal B cell proliferation.
  • the heterogeneous immune disease is an inflammatory disease or asthma.
  • the autoimmune disease is lupus erythematosus, chronic lymphocytic lymphoma, diffuse large cell lymphoma, follicular lymphoma or chronic lymphocytic leukemia.
  • a pharmaceutical composition comprising one or more compounds described in any one of the above.
  • a pharmaceutical formulation comprising a therapeutically effective amount of a compound of any of the above, and a pharmaceutically acceptable excipient.
  • the pharmaceutical preparation is formulated for an administration route selected from oral administration, parenteral administration, oral administration, nasal administration, topical administration, or rectal administration.
  • the pharmaceutical preparation is used for treating a disease or condition related to excessive Bruton tyrosine kinase activity, and comprises administering the pharmaceutical preparation to a human or mammal in need; the excessive Bruton tyrosine kinase activity
  • the related diseases are heterogeneous immune diseases, autoimmune diseases or cancer; the heterogeneous immune diseases are inflammatory diseases, asthma; the autoimmune diseases are lupus erythematosus, chronic lymphocytic lymphoma, diffuse large cells Lymphoma, follicular lymphoma, or chronic lymphocytic leukemia.
  • the invention includes a step of contacting the pharmaceutical preparation with Btk, and the contacting step includes an in vitro or in vivo test.
  • Preparation method 1 of the above compound I includes the following steps: (S1) Suzuki coupling of compound IIIA and boric acid or boric acid ester II to obtain compound IV; (S2) treatment of compound IV with trifluoroacetic acid to remove the benzyloxycarbonyl group and convert to The hydrochloride salt of V; (S3) the compound V is coupled with an organic acid to obtain the compound I described in claim 1;
  • the above-mentioned preparation method 2 of the compound I includes the following steps: (A1) the compound IIIA is treated with trifluoroacetic acid to remove the benzyloxycarbonyl group and converted into the hydrochloride of the compound VI; (A2) the compound VI is coupled with an organic acid to obtain the compound VII; (A3) Suzuki coupling of compound VII with boric acid or borate ester II to obtain compound I as described in claim 1;
  • Preparation method 3 of the above compound I includes the following steps: (B1) compound IIIB and boric acid II are subjected to Chan-Lam-Evans coupling under the catalyst of copper acetate to obtain compound VIII; (B2) compound VIII is treated with trifluoroacetic acid to remove benzyloxy The carbonyl group is converted into the hydrochloride salt of compound IX; (B3) Compound IX is coupled with an organic acid to obtain compound I as claimed in claim 1;
  • R 2 , R 3 , L, A ring, B ring and C ring are as described above.
  • Each of the products obtained by the reactions in Methods 1, 2 and 3 can be obtained by conventional separation techniques, including but not limited to filtration, distillation, crystallization, and chromatographic separation.
  • the starting materials required for the synthesis can be synthesized by yourself or purchased from commercial organizations, such as, but not limited to, Adrich or Sigma. These materials can be characterized using conventional means, such as physical constants and spectral data.
  • the compounds described in the present invention can be synthesized using a synthetic method to obtain a single optical isomer or a mixture of optical isomers.
  • the superscript of the letter indicates the number of the group, and the subscript indicates the number of the atom.
  • R 1 , R 2 , and R 3 represent the 1 to 3 R groups, and the C 1-4 alkyl group contains 1 ⁇ 4 C atom alkyl. The number of C atoms in a substituent is not counted in the main chain.
  • Figure 1 OCI-LY10 transplanted tumor model.
  • FIG. 1 TMD-8 transplanted tumor model.
  • N- (pyridin-2-yl) -4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) benzamide (2g
  • NaIO 4 3.27 g, 18.6 mmol
  • 2N Aqueous HCl (1.65 mL) was added. It was stirred at room temperature for 3 hours.
  • the mixture was diluted with ethyl acetate and washed with brine. It was separated and dried over anhydrous Na 2 SO 4, filtered and concentrated.
  • isopropyl ether (30 mL) was added, stirred for 2 hours, filtered and washed with isopropyl ether (2 x 10 mL) to obtain 180 mg of the desired product A-15 hydrochloride, which was used directly in the next step without purification.
  • the substrate solution was prepared by adding the substrate poly (Glu, Tyr) sodium salt (Sigma Aldrich, St. Louis, MO) to a substrate reaction buffer (20mM Hepes (pH 7.5), 10mM MgCl2, 1mM EGTA, 0.02% Brij35 , 0.02 mg / ml BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT and 1% DMSO) (the final substrate concentration in the reaction was 0.2 uM).
  • the test compound was prepared into a stock solution with a concentration of 10 mM with 100% DMSO, and three-fold serial dilutions of 10 doses were performed in a 384-well circulating olefin copolymer LDV microplate.
  • BTK BTK
  • C481S BTK
  • Test compounds in 100% DMSO were then added to the kinase reaction mixture by acoustic liquid transfer technology (Echo550; nanoliter range) (Labcyte Inc, Sunnyvale, CA) and incubated for 20 minutes at room temperature.
  • 33P-ATP specific activity 10 ⁇ Ci / ⁇ l was added to the reaction mixture to initiate the reaction, followed by incubation at room temperature for 2 hours.
  • the tumor cell line (TMD-8 / OCY-LY10) was suspended in RPMI1640 + FBS10% and cultured in a 37 ° C, 5% CO 2 incubator. Passage at regular intervals and take cells in log phase for plating.
  • IR (%) (1-(RLU compound-RLU blank control) / (RLU vehicle control-RLU blank control)) * 100 %. Calculate the inhibition rate of the compounds at different concentrations in Excel, then use GraphPadPrism software to make the inhibition curve and calculate the IC50.
  • LC-MS / MS Waters Class UPLC Xevo TQD mass spectrometer
  • Z 'value is calculated based on the following formula: 1-[(3 * positive standard deviation + 3 * negative standard deviation) / (mean positive-mean negative)].
  • the C481S mutation reduced Ibrutinib's inhibition of BTK phosphorylation in HEK293 cells from 0.021 ⁇ M to 1.58 ⁇ M, and the compound A-10-10 of the present invention has a strong inhibition of BTK (WT) -transfected HEK293 cells (0.077 ⁇ M ), It has stronger inhibition (0.066 ⁇ M) on HEK293 cells transfected with BTK (C481S).
  • CB17 / SCID female mice with severely immunocompromised functions were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd. and kept in SPF animal rooms.
  • Human-derived OCI-LY10 cells (Shanghai Junrui-UFBN0102) were cultured in monolayer in vitro, and the culture conditions were RPMI 1640 medium plus 10% fetal bovine serum, 100 U / mL penicillin and 100 ⁇ g / mL streptomycin, 37 ° C, 5% CO 2 incubation. Routine processing was performed twice a week. When the cell saturation is 80% -90%, when the number reaches the requirement, the cells are collected and counted.
  • a vehicle 5% DMSO + 20% HP- ⁇ -CD
  • Compounds A-10-10 and Ibrutinib showed extremely strong antitumor activity in the OCI-LY10 transplanted tumor model ( Figure-1).
  • TGI 110%, 110% ; P ⁇ 0.001, p ⁇ 0.001
  • After 28 days of administration all OCI-LY10 transplanted tumors in the 25 mg / kg experimental group also completely disappeared.
  • the TGI value of the control compound Ibrutinib 25 mg / kg group at 28 days was 94% (p ⁇ 0.001).
  • the effect of the test substance on the weight change of tumor-bearing mice is shown in Figure 1.
  • the tumor-bearing mice showed good tolerance to the test drug A-10-10 at all doses, and no significant weight loss in all treatment groups.
  • CB17 / SCID female mice with severely immunocompromised functions were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd. and kept in SPF animal rooms.
  • Human TMD-8 cells (Shanghai Junrui-UFBN1682) were cultured in monolayer in vitro, and the culture conditions were RPMI 1640 medium plus 10% fetal bovine serum, 100 U / mL penicillin and 100 ⁇ g / mL streptomycin, 37 ° C, 5% CO 2 incubation. Routine processing was performed twice a week. When the cell saturation is 80% -90%, when the number reaches the requirement, the cells are collected and counted.
  • Compounds A-10-10 and Ibrutinib showed extremely strong antitumor activity in a TMD-8 transplanted tumor model ( Figure-2).
  • TGI 94%, 104% ; P ⁇ 0.001, p ⁇ 0.001
  • TGI 94%, 104% ; P ⁇ 0.001, p ⁇ 0.001
  • the TGI value of the control compound Ibrutinib 25 mg / kg group was 90% (p ⁇ 0.001).
  • the effect of the test substance on the weight change of tumor-bearing mice is shown in Figure 2.
  • the tumor-bearing mice showed good tolerance to the test drug A-10-10 at all doses, and no significant weight loss in all treatment groups.

Abstract

本发明涉及式I结构的化合物或其药学上可接受的盐、溶剂化物、活性代谢物、多晶型物、酯、光学异构体或前药,包含式I结构的化合物的药物组合物及其作为BTK(C481S)突变体高选择性的布鲁顿酪氨酸激酶抑制剂用于制备预防或治疗异种免疫性疾病、自身免疫性疾病或癌症的药物的用途。

Description

氨基降茨烷衍生物及其制备方法与应用 技术领域
本发明属于医药领域,具体是涉及氨基降茨烷衍生物作为对C481S突变体高选择性的布鲁顿酪氨酸激酶抑制剂、其药物组合物、其制备方法及其在制备药物中的用途。
背景技术
B细胞受体(B-cell receptor,BCR)信号通路在B细胞的成熟,分化和发展中起关键作用。异常的BCR介导的信号转导可引起错误调节的(deregulated)B细胞活化和/或病原性自身抗体的形成,导致多种人类疾病,包括癌症、自身免疫疾病和异种免疫性疾病,自身免疫疾病包括红斑狼疮、慢性淋巴细胞性淋巴瘤、弥漫性大细胞淋巴瘤、滤泡型淋巴瘤或慢性淋巴细胞白血病,异种免疫性疾病包括炎性疾病、哮喘等。
布鲁顿酪氨酸激酶(bruton tyrosine kinase,BTK)是非受体型酪氨酸激酶TEC家族的一员,在BCR信号通路的活化过程中起着关键的作用,是早期B细胞形成以及成熟B细胞激活和存活的关键调节剂(Khan等,Immunity 1995 3:283;Ellmeier等,J Exp Med 2000 192:1611)。Btk在调节B细胞增殖和凋亡发挥重要的作用(Islam和Smith,Immunol Rev 2000 178:49;Davis等,Nature 2010 463:88-94),因此,对Btk的抑制可用于治疗某些B细胞淋巴瘤和白血病(Feldhahn等,J Exp Med 2005 201:1837)。
关于Btk在自身免疫疾病和炎性疾病中的作用的证据已经由Btk-缺陷型小鼠模式得到确认。在系统性红斑狼疮(SLE)的临床前鼠模型中,Btk-缺陷型小鼠显示疾病进展的显著改善。此外,Btk-缺陷型小鼠对胶原蛋白诱惑的关节炎具有抗性(Jansson和Holmdahl,Clin Exp Immunol 1993 94:459)。选择性Btk抑制剂在小鼠关节炎模型中有着明显的量效关系(Pan等,Chem.Med.Chem.2007 2:58-61)。目前Btk抑制剂治疗关节炎的临床研究正在进行中。
依鲁替尼(Ibrutinib,商品名Imbruvica)作为第一个进入市场的布鲁顿酪氨酸激酶(BTK)抑制剂取得极大的成功,2017年年销售达26亿美元。然而与许多其他抗癌药物一样,部分患者对药物表现出耐药性。研究发现,BTK激酶的C481S突变是导致耐药的主要原因,依鲁替尼是通过与BTK激酶的C481色氨酸残基的不可逆共价结合而发生药效作用的,C481S突变将色氨酸变成丝氨酸从而失去与依鲁替尼共价结合的能力。
根据临床统计显示,BTK(C481S)突变占慢性淋巴癌(CLL)复发患者的87%(Woyach等,J Clin Oncol 2017 35:1437-1443),在套细胞淋巴瘤(MCL)复发患者中~80%是 BTK(C481S)突变(Chiron等,Cancer Discovery 2014 4(9):1-14)。开发一种能对BTK(C481S)突变体有效的BTK抑制剂将能克服依鲁替尼因C481S突变而产生的耐药。
发明内容
本发明要解决的技术问题是提供新颖的、未见文献报道的对BTK(C481S)突变体高选择性的布鲁顿酪氨酸激酶抑制剂的化合物,其药学上可接受的盐、溶剂化物、活性代谢物、多晶型物、酯、光学异构体或前药,所述化合物在制药中的用途以及使用本发明化合物预防或治疗人或哺乳动物与过度Btk活性相关疾病的方法。
为解决上述技术问题,本发明采取的技术方案是:
式(I)的化合物
Figure PCTCN2019094864-appb-000001
或其药学上可接受的盐、溶剂化物、活性代谢物、多晶型物、酯、光学异构体或前药,其中,
A环选自以下结构之一:
Figure PCTCN2019094864-appb-000002
R 5选自氢,卤素,氰基,羟基,炔基,氨基,C 1-3烷基,C 1-3烷氧基,C 1-3卤代烷基,C 1-3卤代烷氧基,C 1-3卤代烷氨基,C 3-7环烷基,C 3-7环烷氧基,C 3-7环烷氨基;
B环为取代或非取代的芳环或杂芳环;C环为取代或非取代的芳环或杂芳环;
L为单键、或以下结构之一;
Figure PCTCN2019094864-appb-000003
R 1选自R 3或以下结构之一,
Figure PCTCN2019094864-appb-000004
其中,R 3选自氢、取代或非取代的C 1-6烷基、取代或非取代的C 1-6炔基、取代或非取代的C 1-6烯基、取代或非取代的C 6-10芳基、取代或非取代的C 1-9杂芳基、取代或非取代的C 3-7环烷基或取代或非取代的C 2-7杂环烷氨基;
R 4选自氢、取代或非取代的C 1-6烷基、取代或非取代的C 6-10芳基、取代或非取代的C 1-9杂芳基、取代或非取代的C 3-7环烷基、取代或非取代的C 3-7杂环烷基;
R 2选自H、取代或非取代的C 1-3烷基、取代或非取代的C 3-7环烷基、取代或非取代的C 2-7杂环烷基、取代或非取代的C 6-10芳基或取代或非取代的C 1-9杂芳基。
R 1、R 2以及与之相连的N形成取代或非取代的C 2-7杂环,R 3、R 4以及与之相连的N形成或不形成C 3-7杂环氨基或C 3-9杂芳环氨基。
优选的,所述R 3中的取代的C 1-6烷基、取代的C 1-6炔基、取代的C 1-6烯基、取代的C 6-10芳基、取代的C 1-9杂芳基、取代的C 3-7环烷基、取代的C 2-7杂环烷基中的取代基选自卤素、氰基、羟基、氨基、取代或非取代的酰基胺基、取代或非取代的氨基酰基、取代或非取代的C 1-4烷基、取代或非取代的C 3-7环烷基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 1-4烷基氨基、二[取代或非取代的C 1-4烷基]氨基、取代或非取代的C 3-7环烷氨基、取代或非取代的C 3-7杂环烷氨基、取代或非取代的C 1-3烷氧基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 6-10芳基或取代或非取代的C 3-7杂环烷基中的一个或多个。
优选的,所述R 4中的取代的C 1-6烷基、取代的C 6-10芳基、取代的C 1-9杂芳基、取代的C 3-7环烷基,取代的C 3-7杂环烷基中的取代基选自卤素、羟基、氰基、氨基、取代或非取代的C 1-4烯基、取代或非取代的C 3-7环烷基、取代或非取代的C 3-7环烷氧基、取 代或非取代的C 1-4烷基氨基、二[取代或非取代的C 1-4烷基]氨基、取代或非取代的C 3-7环烷氨基、取代或非取代的C 3-7杂环烷氨基、取代或非取代的C 1-3烷氧基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 6-10芳基或取代或非取代的C 3-7杂环烷基中的一个或多个。
优选的,所述A环结构如下式:
Figure PCTCN2019094864-appb-000005
R 5选自氢,卤素,氰基,羟基,炔基,氨基,C 1-3烷基,C 1-3烷氧基,C 1-3卤代烷基,C 1-3卤代烷氧基,C 1-3卤代烷氨基,C 3-7环烷基,C 3-7环烷氧基,C 3-7环烷氨基;
优选的,所述B环中的取代的芳环或芳杂环中的取代基选自卤素、羟基、氰基、氨基、C 1-3烷基、C 1-3烷氧基、卤代C 1-3烷基、卤代C 1-3烷氧基中的一个或多个。
优选的,所述B环结构如下式:
Figure PCTCN2019094864-appb-000006
优选的,所述C环中的取代的芳环或芳杂环中的取代基选自卤素、羟基、氰基、氨基、C 1-3烷基、C 1-3烷氧基、卤代C 1-3烷基、卤代C 1-3烷氧基中的一个或多个。
优选的,所述C环结构如下式:
Figure PCTCN2019094864-appb-000007
优选的,所述R 2选自H,R 1选自:
Figure PCTCN2019094864-appb-000008
其中,R 3选自氢、取代或非取代的C 1-6烷基、取代或非取代的C 1-6炔基、取代或非取代的C 1-6烯基、取代或非取代的C 6-10芳基、取代或非取代的C 1-9杂芳基、取代或非取代的C 3-7环烷基、取代或非取代的C 2-7杂环烷氨基。
最优选的,所述化合物选自如下任一结构所示:
Figure PCTCN2019094864-appb-000009
Figure PCTCN2019094864-appb-000010
以上任一项所述的化合物在制备预防或治疗异种免疫性疾病、自身免疫性疾病或癌症的药物中的用途。
其中,所述异种免疫性疾病、自身免疫性疾病或癌症与过度布鲁顿酪氨酸激酶活性相关。
其中,所述异种免疫性疾病、自身免疫性疾病或癌症与异常B细胞增殖相关。
更进一步的,所述异种免疫性疾病为炎性疾病或哮喘。
更进一步的,所述自身免疫性疾病为红斑狼疮、慢性淋巴细胞性淋巴瘤、弥漫性大细胞淋巴瘤、滤泡型淋巴瘤或慢性淋巴细胞白血病。
一种药物组合物,包含一种或多种以上任一项所述的化合物。
一种药物制剂,包含治疗有效量的以上任一项所述的化合物,以及在药学上可接受的赋形剂。
所述的药物制剂,其配制用于选自口服施用、肠胃外施用、口腔施用、鼻腔施用、局部施用或直肠施用的施用途径。
所述的药物制剂用于治疗与过度布鲁顿酪氨酸激酶活性相关的疾病或状况,包括向需要的人或哺乳动物施用所述的药物制剂;所述过度布鲁顿酪氨酸激酶活性相关的疾病为异种免疫性疾病、自身免疫性疾病或癌症;所述异种免疫性疾病为炎性疾病、哮喘;所述自身免疫性疾病为红斑狼疮、慢性淋巴细胞性淋巴瘤、弥漫性大细胞淋巴瘤、滤泡型淋巴瘤或慢性淋巴细胞白血病。
本发明包括将所述药物制剂与Btk接触的步骤,所述的接触步骤包括体外或者体内试验。
上述化合物I的制备方法1:包括以下步骤:(S1)化合物IIIA和硼酸或硼酸酯II进行Suzuki偶合得到化合物IV;(S2)化合物IV用三氟乙酸处理脱去苄氧羰基后转化成化合物V的盐酸盐;(S3)化合物V与有机酸偶合得到权利要求1中所述的化合物I;
Figure PCTCN2019094864-appb-000011
其中X=卤素,R 2,R 3,L,A环,B环和C环如前所述。
上述化合物I的制备方法2,包括以下步骤:(A1)化合物IIIA用三氟乙酸处理脱去苄氧羰基后转化成化合物VI的盐酸盐;(A2)化合物VI与有机酸偶合得到化合物VII;(A3)化合物VII与硼酸或硼酸酯II进行Suzuki偶合得到权利要求1中所述的化 合物I;
Figure PCTCN2019094864-appb-000012
其中X=卤素,R 2,R 3,L,A环,B环和C环如前所述。
上述化合物I的制备方法3,包括以下步骤:(B1)化合物IIIB和硼酸II在醋酸铜催化下进行Chan-Lam-Evans偶合得到化合物VIII;(B2)化合物VIII用三氟乙酸处理脱去苄氧羰基后转化成化合物IX的盐酸盐;(B3)化合物IX与有机酸偶合得到权利要求1中所述的化合物I;
Figure PCTCN2019094864-appb-000013
其中R 2,R 3,L,A环,B环和C环如前所述。
方法1、2和3中反应所得的每一个产物可以通过传统分离技术来得到,这种传统技术包括但不限于过滤、蒸馏、结晶、色谱分离等。合成所需要的起始原料可以自己合成或从商业机构购买获得,例如,但不限于,Adrich或Sigma。这些原料可以使用常规手段进行表征,比如物理常数和光谱数据。本发明所描述的化合物可以使用合成方法得到单一的光学异构体或者是光学异构体的混合物。
本发明中字母上标表示基团的标号,下标表示该原子的个数,例如:R 1、R 2、R 3表示第1~3个R基团,C 1-4烷基表示含1~4个C原子的烷基。取代基上C原子数不计算在主链中。
附图说明
图1 OCI-LY10移植肿瘤模型。
图2 TMD-8移植肿瘤模型。
具体实施方式
根据下述实施例,可以更好地理解本发明。然而,本领域的技术人员容易理解,实施例所描述的内容仅用于说明本发明,而不应当也不会限制权利要求书中所详细描述的本发明。
中间体I-5的合成
中间体I-5的合成路线
Figure PCTCN2019094864-appb-000014
4-(苄氧基羰基氨基)双环[2.2.1]庚烷-1-羧酸甲酯I-2
Figure PCTCN2019094864-appb-000015
向4-(甲氧基羰基)双环[2.2.1]庚烷-1-羧酸I-1(3.5g,17.7mmol)和TEA(1.78g,17.7mmol)的甲苯(30mL)溶液中加入DPPA(5.34g,19.5mmol)。将混合物加热至90℃,保持2小时。冷却至室温并加入BnOH(1.9g,17.7mmol)。将反应在90℃下搅拌4天。冷却至室温,用乙酸乙酯稀释并用NaHCO 3水溶液洗涤,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:4的柱色谱法纯化,得到3g所需化合物I-2,收率:56%。
4-(苄氧基羰基氨基)双环[2.2.1]庚烷-1-羧酸I-3
Figure PCTCN2019094864-appb-000016
向4-(苄氧基羰基)双环[2.2.1]庚烷-1-甲酸甲酯I-2(3g,9.9mmol)的溶液中加入NaOH(792mg,19.8mmol),将混合物加热至60℃,保持10小时。浓缩,加入水(50mL),用1N HCl水溶液将pH调节至4。将其用乙酸乙酯(3×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其不经纯化用于下一步,得到2.2g所需化合物I-3,收率:77%。
4-氨基甲酰基双环[2.2.1]庚-1-基氨基甲酸苄酯I-4
Figure PCTCN2019094864-appb-000017
向化合物I-3(2g,6.9mmol)和Et 3N(1g,10mmol)的DCM(20mL)溶液中滴加0℃的氯代异丁基酯(1.36g,10mmol)。将混合物在0℃下搅拌20分钟,滴加NH 4OH(10mL)并将其在室温下搅拌10分钟。将其倒入水(30mL)中,分离有机相,用DCM(2x15mL)萃取水溶液,将合并的有机相用Na 2SO 4干燥,过滤并浓缩。将其通过柱色谱法纯化,用EA/PE(1:1)-EA/MeOH(10:1)洗脱。得到1.7g所需化合物I-4,产率:85%。
4-氨基双环[2.2.1]庚-1-基氨基甲酸苄酯I-5
Figure PCTCN2019094864-appb-000018
将化合物I-4(1.6g,5.55mmol)和羟基甲基磺酰碘苯(2.17g,5.55mmol)在ACN(20mL)中的溶液加热回流1小时,蒸发溶剂并加入1M NaOH(12mL),将其用EA(2x15mL)萃取,将有机相用Na 2SO 4干燥,过滤并浓缩。将其通过柱色谱法纯化,用DCM/MeOH=(10:1)洗脱。得到920mg所需化合物I-5,收率:64%。LC-MS m/z=261.1[M+1] +
中间体硼酸II的合成
中间体II-1的合成路线
Figure PCTCN2019094864-appb-000019
4-溴-N-(吡啶-2-基)苯甲酰胺
Figure PCTCN2019094864-appb-000020
在冰浴中向4-溴苯甲酸(5g,24.8mmol)和吡啶-2-胺(4.68g,49mmol)在吡啶(30mL)中的混合物中滴加POCl 3(11.4g,74mmol)。将悬浮液在室温下搅拌20分钟。将反应倒入水(100mL)中并用乙酸乙酯(3x40mL)萃取。有机相用饱和NaCl水溶液(2x50mL)洗涤。将有机相用无水Na 2SO 4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油=1:9
Figure PCTCN2019094864-appb-000021
1:1进行纯化,得到产物4-溴-N-(吡啶-2-基)苯甲酰胺(3.28g,48%)。LC-MS m/z=277.0[M+1] +
(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酰胺
Figure PCTCN2019094864-appb-000022
将4-溴-N-(吡啶-2-基)苯甲酰胺(2g,7.22mmol),4,4,5,5-四(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1,3,2-二氧杂硼杂环戊烷(2.75g,10.83mmol),PdCl 2(dppf)(527mg,0.72mmol)和KOAc(235mg,2.4mmol)在甲苯(30mL)中的混合物加热至110℃保持6小时。将反应液蒸发并加入水(100mL)。将其用乙酸乙酯(2x40mL)萃取。分离有机相,用无水Na 2SO 4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油醚=1:4
Figure PCTCN2019094864-appb-000023
1:1纯化,得到产物(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酰胺(2g,85%)。
(吡啶-2-基氨基甲酰基)苯基硼酸II-1
Figure PCTCN2019094864-appb-000024
向N-(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酰胺(2g,6.2mmol)在THF:H 2O(24mL:6mL)的混合溶剂中的溶液中加入NaIO 4(3.27g,18.6mmol),并在室温下搅拌30分钟。加入2N HCl水溶液(1.65mL)。将其在室温下搅拌3小时。将混合物用乙酸乙酯稀释,用盐水洗涤。分离并用无水Na 2SO 4干燥,过滤并浓缩。通过使用MeOH/DCM=1:10的柱色谱法纯化得到产物(吡啶-2-基氨基甲酰基)苯基硼酸II-1(1.4g,93%)。LC-MS m/z=243.1[M+1] +
中间体II-2的合成路线
Figure PCTCN2019094864-appb-000025
4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)苯甲酰氯
Figure PCTCN2019094864-appb-000026
向4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酸(10g,40mmol)和1滴DMF的DCM(100mL)溶液中在冰浴下滴加草酰氯(10.2g,80mmol)。将混合物在0℃下搅拌30分钟,然后温热至室温另外3小时。浓缩,不经纯化用于下一步骤。
N-(4-氟吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)苯甲酰胺
Figure PCTCN2019094864-appb-000027
向4-氟吡啶-2-胺(421mg,3.76mmol)在吡啶(3mL)中的溶液中加入4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊-2-基)吡啶-2-基)苯甲酰氯(1g,3.76mmol)的DCM(6mL)溶液中,将悬浮液在0℃下搅拌30分钟。将其倒入水中并用DCM(2×20mL)萃取,有机相用饱和NaCl水溶液洗涤,分离并用无水Na 2SO 4干燥。蒸发并通过柱色谱法用乙酸乙酯/石油=1:9纯化,得到产物(1.04g,81%)。LC-MS m/z=343.2[M+1] +
4-((4-氟吡啶-2-基)氨基甲酰基)苯基硼酸II-2
Figure PCTCN2019094864-appb-000028
向N-(4-氟吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)苯甲酰胺(1.04g,3.04mmol)在THF:H 2O(24mL:6mL)的混合溶剂中的溶液中加入NaIO 4(1.9g,9.12mmol),并在室温下搅拌30分钟。加入HCl水溶液(1.65ml)。将其在室温下搅拌3小时。将混合物用乙酸乙酯稀释,用盐水洗涤。分离并用无水Na 2SO 4干燥,过滤并浓缩。通过柱色谱法用MeOH/DCM=1:10纯化。得到产物II-2(648mg,82%)。LC-MS m/z=261.1[M+1] +
4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸II-3
Figure PCTCN2019094864-appb-000029
以4-(三氟甲基)吡啶-2-胺(609mg,3.76mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊-2-基)吡啶-2-基)苯甲酰氯(1g,3.76mmol)为原料,使用与II-2相同的合成方法得到607mg所需化合物。LC-MS m/z=311.1[M+1] +
4-((4-甲基吡啶-2-基)氨基甲酰基)苯基硼酸II-4
Figure PCTCN2019094864-appb-000030
以4-甲基-吡啶-2-胺(406mg,3.76mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊-2-基)吡啶-2-基)苯甲酰氯(1g,3.76mmol)为原料,使用与II-2相同的合成方法得到589mg所需化合物。LC-MS m/z=257.1[M+1] +
4-((4-氰基吡啶-2-基)氨基甲酰基)苯基硼酸II-5
Figure PCTCN2019094864-appb-000031
以4-氰基-吡啶-2-胺(447mg,3.76mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊-2-基)吡啶-2-基)苯甲酰氯(1g,3.76mmol)为原料,使用与II-2相同的合成方法得到465mg所需化合物。LC-MS m/z=268.0[M+1] +
中间体II-6的合成路线
Figure PCTCN2019094864-appb-000032
4-溴-2-氟-N-(吡啶-2-基)苯甲酰胺
Figure PCTCN2019094864-appb-000033
在冰浴中向4-溴-2-氟苯甲酸(1g,4.56mmol)的DCM(30mL)溶液中滴加草酰氯(1.16g,9.13mmol),然后加入1滴DMF。将混合物在室温下搅拌3小时。将其浓缩并溶于DCM(6mL)中,将该溶液在0℃下加入吡啶-2-胺(428mg,4.56mmol)的吡啶(3ml)溶液中,将悬浮液在0℃下搅拌,30分钟。将其倒入水中并用DCM(2×20mL)萃取,有机相用饱和NaCl水溶液洗涤,分离并用无水Na 2SO 4干燥。蒸发并通过柱色谱法用乙酸乙酯/石油=1:9纯化,得到产物(1.05g,78%)。LC-MS m/z=295.0[M+1] +
(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酰胺
Figure PCTCN2019094864-appb-000034
将4-溴-2-氟-N-(吡啶-2-基)苯甲酰胺(1.05g,3.55mmol),(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1,3,2-二氧杂硼杂环戊烷(1.36g,5.3mmol),PdCl 2(dppf)(260mg,0.36mmol)和KOAc(1.04g,10.65mmol)的甲苯(30mL)溶液加热至110℃保持6小时。将反应物蒸发并加入水(100mL)。将其用乙酸乙酯(2×40mL)萃取。分离有机相,用无水Na 2SO 4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油醚=1:4
Figure PCTCN2019094864-appb-000035
1:1进行纯化, 得到产物(971mg,80%)。
3-氟-4-(吡啶-2-基氨基甲酰基)苯基硼酸II-6
Figure PCTCN2019094864-appb-000036
向2-氟-N-(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烯苯甲酰胺(970mg,2.84mmol)在THF:H 2O(24mL:6mL)的混合溶剂中的溶液中加入NaIO 4(1.8g,8.52mmol),并在室温下搅拌30分钟。HCl水溶液(1.65ml)。将其在室温下搅拌3小时。将混合物用乙酸乙酯稀释,用盐水洗涤。分离并用无水Na 2SO 4干燥,过滤并浓缩。通过柱色谱法用MeOH/DCM=1:10纯化。得到产物II-6(605mg,82%)。LC-MS m/z=261.1[M+1] +
(噻唑-2-基氨基甲酰基)苯基硼酸II-7
Figure PCTCN2019094864-appb-000037
以噻唑-2-胺(376mg,3.76mmol)和4-((4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊-2-基)吡啶-2-基)苯甲酰氯(1g,3.76mmol)为原料,使用与II-2相同的合成方法得到580mg所需化合物II-7。LC-MS m/z=249.1[M+1] +
2-氟-4-(吡啶-2-基氨基甲酰基)苯基硼酸II-8
Figure PCTCN2019094864-appb-000038
以4-溴-3-氟-苯甲酸和吡啶-2-胺为原料,使用与II-1相同的合成方法得到138mg所需化合物II-8。LC-MS m/z=261.0[M+1] +
2-氟-4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸II-9
Figure PCTCN2019094864-appb-000039
以4-溴-3-氟-苯甲酸和4-(三氟甲基)-吡啶-2-胺为原料,使用与II-1相同的合成方法得到130mg所需化合物II-9。LC-MS m/z=329.0[M+1] +
2-甲氧基-4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸II-10
Figure PCTCN2019094864-appb-000040
以4-溴-3-甲氧基苯甲酸(1g,4.36mmol)和4-(三氟甲基)-吡啶-2-胺(706mg,4.36mmol)为原料,使用与II-6相同的合成方法得到440mg所需化合物II-10。LC-MS m/z=341.0[M+1] +
中间体硼酸酯II-11的合成路线
Figure PCTCN2019094864-appb-000041
N-(4-溴苄基)-2-甲氧基苯甲酰胺
Figure PCTCN2019094864-appb-000042
4-溴苄胺(1g,5.38mmol),2-甲氧基苯甲酸(818mg,5.38mmol),HATU(2.45g,6.46mmol)和DIEA(1.39g,10.76mmol)在DMF(20mL)中室温下搅拌2小时。将其倒入水(50mL)中并过滤。将其用水(2×30mL)洗涤并干燥。它无需纯化即可用于下一步。得到所需化合物1.6g,收率:93%。
N-(4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苄基)-2-甲氧基苯甲酰胺II-11
Figure PCTCN2019094864-appb-000043
将N-(4-溴苄基)-2-甲氧基苯甲酰胺(1.6g,5mmol),4,4,5,5-四甲基-2-(4,4,5,5-四甲基-1,3-二氧戊环-2-基)-1,3,2-二氧杂硼杂环戊烷(1.9g,7.5mmol),PdCl 2dppf(365mg,0.5mmol)和KOAc(1.47g,15mmol)的二恶烷(30mL)溶液加热至100℃,保持6小时。浓缩,加入水(100mL),用乙酸乙酯(2×20mL)萃取。分离有机相,用Na 2SO 4干燥,过滤,浓缩,用乙酸乙酯/石油=1:9的柱色谱法纯化。得到1.5g所需化合物II-11,产率:82%。
N-(4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苄基)-5-氟-2-甲氧基苯甲酰胺II-12
Figure PCTCN2019094864-appb-000044
以5-氟-2-甲氧基苯甲酸(915mg,5.38mmol)和4-溴苄胺(1g,5.38mmol)为原料,使用与II-11相同的合成方法得到900mg所需化合物II-12。
N-(4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苄基)-4-氟-2-甲氧基苯甲酰胺II-13
Figure PCTCN2019094864-appb-000045
以4-氟-2-甲氧基苯甲酸(915mg,5.38mmol)和4-溴苄胺(1g,5.38mmol)为原料,使用与II-11相同的合成方法得到1g所需化合物II-13。
中间体A-4的合成路线
Figure PCTCN2019094864-appb-000046
4-(((3-氯吡嗪-2-基)甲基)氨基甲酰基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-1
Figure PCTCN2019094864-appb-000047
4-(苄氧基羰基)双环[2.2.1]庚烷-1-羧酸(2g,6.9mmol),HATU(2.89g,7.6mmol),DIEA(3.56g,27.6mmol)和(3-氯吡嗪-2-基)甲胺的盐酸盐(1.3g,7.24mmol)的DMF(20mL)溶液在室温下搅拌6小时。倒入水(100mL)中并用乙酸乙酯(2×30mL)萃取。用饱和NaCl水溶液洗涤有机相。分离有机相,用Na 2SO 4干燥,过滤并蒸发。用乙酸乙酯/石油醚=1:1~1:0的柱色谱法纯化,得到2g所需化合物A-1,收率:70%。LC-MS m/z=414.9[M+1] +
4-(8-氯咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-2
Figure PCTCN2019094864-appb-000048
向4N-(((3-氯吡嗪-2-基)甲基)氨基甲酰基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-1(2g,4.83mmol)的ACN(30mL)溶液中加入吡啶(381mg,4.83mmol)和PCl 5(4g,19.32mmol),然后将混合物加热至56℃保持1小时。冷却至室温并缓慢倒入100mL冰饱和NaHCO 3水溶液中。保持pH=9,将其用乙酸乙酯(2×30mL)萃取。用饱和NaCl水溶液洗涤有机相。分离有机相,用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到1.5g所需化合物A-2,收率:78%。
4-(8-氯-1-碘咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-3
Figure PCTCN2019094864-appb-000049
将4-(8-氯咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-2(1.5g,3.79mmol)和NIS(1.13g,5.04mmol)的DMF(10mL)混合液在N 2气氛下加热至60℃,保持搅拌10小时。冷却至室温并倒入水(100mL)中并用乙酸乙酯(2×30mL)萃取。用饱和NaCl水溶液洗涤有机相。分离有机相,用Na 2SO 4干燥,过滤并蒸发。用乙酸乙酯/石油醚=2:3的柱色谱法纯化,得到1.52g所需化合物A-3,收率:77%。
4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-4
Figure PCTCN2019094864-appb-000050
向化合物A-3(1.5g,2.87mmol)的IPA(15mL)悬浮液中加入NH 4OH(3mL),将混合物加热至110℃,保持6小时。浓缩并加入20mL饱和NaHCO 3水溶液。用乙酸乙酯(2×30mL)萃取。用饱和NaCl水溶液洗涤有机相。分离有机相,用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚柱色谱纯化=1:1,得到1.1g所需化合物A-4,收率:77%。
中间体B-3的合成路线
Figure PCTCN2019094864-appb-000051
4-(4-氯-7H-吡咯并[2,3-d]嘧啶-7-基)双环[2.2.1]庚-1-基氨基甲酸苄酯B-1
Figure PCTCN2019094864-appb-000052
将2-(4,6-二氯嘧啶-5-基)乙醛(735mg,3.8mmol),4-氨基双环[2.2.1]庚-1-基氨基甲酸苄酯(1g,3.8mmol)和Et 3N(389mg,3.8mmol)的EtOH(20mL)溶液加热至80℃,保持16小时。将其浓缩并加入水(20mL)用乙酸乙酯(2×20mL)萃取,分离有机相,用Na 2SO 4干燥,过滤并蒸发,通过柱色谱用EA/PE=1:4纯化,得到1.25g所需化合物B-1,收率:83%。LC-MS m/z=397.1[M+1] +
4-(4-氯-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)双环[2.2.1]庚-1-基氨基甲酸苄酯B-2
Figure PCTCN2019094864-appb-000053
向化合物B-1(1.25g,3.16mmol)的DMF(10mL)溶液中加入NIS(950mg,4.2mmol),将混合物加热至60℃,保持6小时。将其倒入水(20mL)中并用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发,通过柱色谱法用EA/PE=1:4纯 化,所需得到1.09g化合物,收率:66%。LC-MS m/z=523.1[M+1] +
4-(4-氨基-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)双环[2.2.1]庚-1-基氨基甲酸苄酯B-3
Figure PCTCN2019094864-appb-000054
向4-(4-氯-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)双环苄基的溶液[2.2.1]庚-1-基氨基甲酸酯B-2(1.09g,2.08mmol)的IPA(10mL)溶液中加入NH 4OH(2mL)并将混合物加热至110℃保持6小时,浓缩并倒入NaHCO 3水溶液中,用DCM(2×20mL)萃取,分离有机相,干燥并过滤,将其浓缩并通过柱色谱法用MeOH/DCM=1:20纯化,得到900mg所需化合物B-3,收率:86%
中间体C-4的合成路线
Figure PCTCN2019094864-appb-000055
4-(6-氯-5-硝基嘧啶-4-基氨基)双环[2.2.1]庚-1-基氨基甲酸苄酯C-1
Figure PCTCN2019094864-appb-000056
将4,6-二氯-5-硝基嘧啶(518mg,2.68mmol),4-氨基双环[2.2.1]庚-1-基氨基甲酸苄酯I-5(698mg,2.68mmol)和Et 3N(1.1g,10.72mmol)的DCM(20mL)溶液在室温下搅拌4小时。除去溶剂并将残余物用乙酸乙酯(50mL)处理,用饱和NaCl水溶液洗涤,分离有机相并用Na 2SO 4干燥,过滤并浓缩,通过柱色谱法用EA/PE=1:4纯化。得到633mg所需化合物C-1,收率:57%。
4-(5-氨基-6-氯嘧啶-4-基氨基)双环[2.2.1]庚-1-基氨基甲酸苄酯C-2
Figure PCTCN2019094864-appb-000057
向化合物C-1(400mg,0.96mmol)在混合溶剂(EtOH/H 2O=20mL/4mL)中的溶液中加入Fe粉末(268mg,4.8mmol)和NH 4Cl(254mg,4.8mmol)。将混合物加热至回流1小时。冷却至室温并过滤,用MeOH(10mL)洗涤,浓缩滤液,通过柱色谱法用EA/PE=1:4纯化。得到292mg所需化合物C-2,收率:79%。
4-(6-氯-8-氧代-7,8-二氢嘌呤-9-基)双环[2.2.1]庚-1-基氨基甲酸苄酯C-3
Figure PCTCN2019094864-appb-000058
在0℃下,向化合物C-2(290mg,0.75mmol)的DCM(10mL)溶液中加入Et 3N(166mg,1.5mmol)和三光气(291mg,0.9mmol),然后在0℃下搅拌。持续2小时。将其倒入水(20mL)中,分离有机相,用DCM(2×10mL)萃取水溶液,分离,用Na 2SO 4干燥合并的有机相,过滤并浓缩,将其用于下一步骤不经纯化,得到C-3粗品316mg。
4-(6-氨基-8-氧代-7,8-二氢嘌呤-9-基)双环[2.2.1]庚-1-基氨基甲酸苄酯C-4
Figure PCTCN2019094864-appb-000059
向化合物C-3(310mg,0.75mmol)的IPA(10mL)溶液中加入NH 4OH(2mL),将混合物加热至150℃,保持24小时,浓缩,倒入NaHCO 3水溶液中,用将DCM(2×20mL),分离有机相,干燥并过滤,将其浓缩并通过柱色谱法用MeOH/DCM=1:20纯化,得到100mg所需化合物C-4,收率:34%。LC-MS m/z=395.1[M+1] +
中间体D-5的合成路线
Figure PCTCN2019094864-appb-000060
苄基4-(6-氯-5-甲醛基嘧啶-4-基氨基)双环[2.2.1]庚烷-1-基氨基甲酸D-1
Figure PCTCN2019094864-appb-000061
将I-5(1.47g、5.65mmol)、4,6-二氯嘧啶-5-甲醛(1g、5.65mmol)、三乙胺(1.15g、11.4mmol)在DCM(20mL)中的混合液室温下搅拌过夜。反应液用EA(50mL)稀释,用水洗涤(2x30mL)。有机相用无水Na 2SO 4干燥,过滤和浓缩。残留物用硅胶柱层析,用石油醚/乙酸乙酯(1:1)洗脱,得到所需的产品D-1(1.24g,55%)。LC-MS m/z=401.0[M+1] +
(E)-苄基4-(6-氯-5-(羟基亚氨基)甲基)嘧啶-4-基氨基)双环[2.2.1]庚烷-1-基氨基甲酸D-2
Figure PCTCN2019094864-appb-000062
将D-1(1.2g,3.0mmol),羟胺-O-磺酸(0.41g,3.6mmol)在DCM/ACN(50毫升/50毫升)的混合液在室温下搅拌16h,然后在50℃搅拌6h。冷却后,浓缩至10mL,固体过滤和用ACN(2mL)洗涤,得到所需的产品D-2(1.0g,81%)。LC-MS m/z=416.0[M+1] +
苄基4-(4-氯-1H-吡咯[3,4-d-]嘧啶-1-基)双环[2.2.1]庚烷-1-基氨基甲酸D-3
Figure PCTCN2019094864-appb-000063
向D-2(1.0g,2.41mmol)的DCM(100mL)溶液中加了入DIEA(4mL)。然后滴加TsCl(0.23mL,2.9mmol)。混合物在室温下搅拌3h。反应混合物用水(100mL)处理。有 机相用盐水洗涤(2x30mL),用无水Na 2SO 4干燥,过滤,浓缩。残留物用硅胶柱层析纯化,由乙酸乙酯/石油醚(1:6)洗脱,得到所需的产品D-3(400mg,42%)。LC-MS m/z=398.1[M+1] +
苄基4-(4-氨基-1H-吡咯[3,4-d-]嘧啶-1-基)双环[2.2.1]庚烷-1-基氨基甲酸D-4
Figure PCTCN2019094864-appb-000064
将D-3(400mg、1.0mmol)、氢氧化铵(30%、5mL)在异丙醇(20mL)的混合物在封管中在120℃下搅拌6h。蒸干溶剂,残留物用硅胶柱层析纯化,用PE/EA(2:1)洗脱,得到所需的产品D-4(280mg,73.4%)。
苄基4-(4-氨基-3-碘-1H-吡咯[3,4-d-]嘧啶-1-基)双环[2.2.1]庚烷-1-基氨基甲酸D-5
Figure PCTCN2019094864-appb-000065
将D-4(190mg、0.5mmol)、NIS(400mg、1.78mmol)、HBF 4(50%、19.6mmol、4mL)在ACN(2.5mL)的混合物在封管中加热到85℃搅拌6h。冷却后,用饱和NaHCO 3淬灭,用EA(2x50mL)提取。有机相用无水Na 2SO 4干燥,过滤和浓缩。残留物用硅胶柱层析纯化,由用EA/PE(1:1-1:0)洗脱,得到所需的产品D-5(96mg,38.1%)。LC-MS m/z=505.0[M+1] +
实施例1
化合物A-7-n的合成路线
Figure PCTCN2019094864-appb-000066
3-(4-氨基双环[2.2.1]庚-1-基)-1-碘咪唑并[1,5-a]吡嗪-8-胺A-5
Figure PCTCN2019094864-appb-000067
将化合物A-4(1g,1.99mmol)在TFA/DCM(10mL/10mL)的混合溶剂中的溶液加热至60℃,保持6小时。浓缩并加入DCM(2×20mL),浓缩并溶于DCM(20mL)中,加入5mL HCl的二恶烷溶液并在室温下搅拌10分钟。将其蒸发并加入DCM(2×20mL),将其蒸发并加入DME(20mL)。将其在室温下搅拌30分钟。过滤并用DME(2×10mL)洗涤。所得化合物A-5无需纯化即可用于下一步骤。LC-MS m/z=370.1[M+1] +
N-(4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)丁-2-炔酰胺A-6
Figure PCTCN2019094864-appb-000068
将化合物A-5(1.35g,2.8mmol),DIEA(3.28g,25.2mmol),丁-2-炔酸(235mg,2.8mmol)和HATU(1.06g,2.8mmol)的DMF(20mL)溶液在室温下搅拌30分钟。将其倒入水(30mL)中并用乙酸乙酯(2×30mL)萃取。用饱和NaCl水溶液洗涤有机相。分离有机相,用Na 2SO 4干燥,过滤并蒸发。用乙酸乙酯/石油醚=2:3的柱色谱法纯化,得到800mg所需化合物A-6,收率:65%。
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺A-7-1
Figure PCTCN2019094864-appb-000069
将化合物A-6(30mg,0.069mmol),4-(吡啶-2-基氨基甲酰基)苯硼酸II-1(20mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂的溶液中加热至80℃3小时。浓缩,加入水(20mL),用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到10mg所需化合物A-7-1,收率:30%。LC-MS m/z=506.2[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-氟吡啶吡啶-2-基)苯甲酰胺A-7-2
Figure PCTCN2019094864-appb-000070
将化合物A-6(30mg,0.069mmol),4-((4-氟吡啶-2-基)氨基甲酰基)苯基硼酸II-2(22mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中的的溶液加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到15mg所需化合物A-7-2,收率:34%。LC-MS m/z=524.0[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲 基基)吡啶-2-基)苯甲酰胺A-7-3
Figure PCTCN2019094864-appb-000071
将化合物A-6(30mg,0.069mmol),4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸II-3(26mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中的的溶液中加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到12mg所需化合物A-7-3,收率:31%。LC-MS m/z=574.2[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-甲基吡啶吡啶-2-基)苯甲酰胺A-7-4
Figure PCTCN2019094864-appb-000072
将化合物A-6(30mg,0.069mmol),4-((4-甲基吡啶-2-基)氨基甲酰基)苯基硼酸II-4(22mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中的溶液加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到14mg所需化合物A-7-4,收率:39%。LC-MS m/z=520.2[M+1] +
4-(8-氨基-3(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-氰基吡啶-2-基)苯甲酰胺A-7-5
Figure PCTCN2019094864-appb-000073
将化合物A-6(30mg,0.069mmol),4-((4-氰基吡啶-2-基)氨基甲酰基)苯基硼酸II-5(23mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中的溶液加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到12mg所需化合物A-7-5,收率:33%。LC-MS m/z=531.0[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-2-氟-N-(吡啶-2-基)苯甲酰胺A-7-6
Figure PCTCN2019094864-appb-000074
将化合物A-6(30mg,0.069mmol),3-氟-4-(吡啶-2-基氨基甲酰基)苯基硼酸II-6(22mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中的溶液加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到15mg所需化合物A-7-6, 收率:42%。LC-MS m/z=524.2[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(噻唑-2-基)苯甲酰胺A-7-7
Figure PCTCN2019094864-appb-000075
将化合物A-6(30mg,0.069mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-N-(噻唑-2-基)苯甲酰胺II-7(28mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂的溶液中加热至80℃保持3小时。浓缩,加入水(20mL)。用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸干。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到13mg所需化合物A-7-7,收率:37%。LC-MS m/z=512.0[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-3-氟-N-(吡啶-2-基)苯甲酰胺A-7-8
Figure PCTCN2019094864-appb-000076
将化合物A-6(30mg,0.069mmol),2-氟-4-(吡啶-2-基氨基甲酰基)苯基硼酸II-8(22mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中的溶液加热至80℃达3小时。浓缩,加入水(20mL)。用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸 乙酯/石油醚=1:1的柱色谱法纯化,得到14mg所需化合物A-7-8,收率:39%。LC-MS m/z=524.0[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-3-氟-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-7-9
Figure PCTCN2019094864-appb-000077
将化合物A-6(30mg,0.069mmol),2-氟-4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸II-9(28mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到19mg所需化合物A-7-9,收率:47%。LC-MS m/z=592.0[M+1] +
4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-3-甲氧基-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-7-10
Figure PCTCN2019094864-appb-000078
将化合物A-6(30mg,0.069mmol),2-甲氧基-4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸II-10(29mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂的溶液中加热至80℃,保持3小时。 浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到16mg所需化合物A-7-10,收率:38%。LC-MS m/z=604.0[M+1] +
N-((4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)甲基)-2-甲氧基A-7-11
Figure PCTCN2019094864-appb-000079
将化合物A-6(30mg,0.069mmol),N-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苄基)-2-甲氧基苯甲酰胺II-11(31mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到21mg所需化合物A-7-11,收率:50%。LC-MS m/z=549.3[M+1] +
N-((4-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)甲基)-5-氟-2-甲氧基苯甲酰胺A-7-12
Figure PCTCN2019094864-appb-000080
将化合物A-6(30mg,0.069mmol),N-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2- 基)苄基)-5-氟-2-甲氧基苯甲酰胺II-12(33mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中加热至80℃。持续3个小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用无水Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到24mg所需化合物A-7-12,收率:56%。LC-MS m/z=567.0[M+1] +
N-((-(8-氨基-3-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)甲基)-4-氟-2-甲氧基苯甲酰胺A-7-13
Figure PCTCN2019094864-appb-000081
将化合物A-6(30mg,0.069mmol),N-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苄基)-4-氟-2-甲氧基苯甲酰胺II-13(33mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中加热至80℃。持续3个小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸发。将其用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到26mg所需化合物A-7-13,收率:60%。LC-MS m/z=567.1[M+1]+。
N-(4-(8-氨基-1-(4-苯氧基苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)丁-2-炔酰胺A-7-14
Figure PCTCN2019094864-appb-000082
将化合物A-6(30mg,0.069mmol),4-苯氧基苯硼酸(18mg,0.085mmol),Pd[PPh 3] 4(8mg,0.0069mmol)和Cs 2CO 3(45mg,0.138mmol)在DME/H 2O(1.5mL/0.3mL)的混合溶剂中加热至80℃,保持3小时。浓缩,加入水(20mL)。将其用乙酸乙酯(2×20mL)萃取,分离有机相并用Na 2SO 4干燥,过滤并蒸干。用乙酸乙酯/石油醚=1:1的柱色谱法纯化,得到19mg所需化合物A-7-14,收率:49%。LC-MS m/z=478.0[M+1]+。
化合物A-10-n的合成路线
Figure PCTCN2019094864-appb-000083
4-(1-(4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚烷苄基-1-基氨基甲酸A-8
Figure PCTCN2019094864-appb-000084
4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸苄酯A-4(300mg,0.6mmol),4-(4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯硼酸II-3(229mg,0.738mmol),Pd[PPh 3] 4(69mg,0.06mmol)和Cs 2O 3(239mg,0.738mmol)在DME:H 2O(2.5mL:0.5mL)的混合溶剂中加热到80℃过夜。浓缩,用甲醇/DCM(1:30)柱层析法对其进行纯化。获得265毫克所需化合物A-8,产率:69%。
4-(8-氨基-3-(4-氨基双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-9
Figure PCTCN2019094864-appb-000085
将化合物A-8(265mg,0.42mmol)在DCM/TFA(10mL:10mL)混合溶剂的溶液加热到60℃18小时。蒸干后加入DCM(2x20mL),浓缩。剩余物溶于DCM(30mL)中,加入HCl二恶烷溶液中,蒸干,加入DCM(2X20毫升),浓缩后加入异丙醚(30mL),搅拌2小时,过滤和用异丙醚洗涤(2X10mL),得到196mg所需的产品A-9的盐酸盐,未经纯化直接用于下一步。
(E)-4-(8-氨基-3-(4-(4-甲氧基-丁-2-烯酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-1
Figure PCTCN2019094864-appb-000086
将化合物A-9(15mg,0.024mmol),HATU(9.12mg,0.024mmol),DIEA(16,0.12mmol)和(E)-4-甲氧基丁-2-烯酸(2.8mg,0.024mmol)的DMF(1mL)溶液在室温下搅拌1小时,将其倒入水(5mL)中并用乙酸乙酯(2X5mL)萃取,将有机相用饱和NaCl水溶液洗涤,分离有机相。用无水Na 2SO 4干燥,过滤并蒸干。将其通过柱色谱法纯化,用DCM/MeOH=20:1洗脱,得到5mg产物A-10-1,产率:35%。LC-MS m/z=606.1[M+1]+。
(E)-4-(8-氨基-3-(4-(4-(四氢吡咯-1-基)丁-2-烯酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-2
Figure PCTCN2019094864-appb-000087
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和(E)-4-(四氢吡咯-1-基)-丁-2-烯酸(4mg,0.024mmol)在DMF(1mL)的溶液中在室温下搅拌1小时,倒入水中(5ml),用乙酸乙酯(2x5ml)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-2,产率:38%。LC-MS m/z=645.0[M+1]+。
4-(3-(4-丙烯酰胺双环[2.2.1]庚-1-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-3
Figure PCTCN2019094864-appb-000088
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和丙烯酸(2mg,0.024mmol)在室温下搅拌1小时,倒入水(1ml),用乙酸乙酯(2x5mL)萃取,有机相用饱NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品A-10-3,产率:38%。LC-MS m/z=562.0[M+1]+。
4-(3-(4-乙酰胺基双环[2.2.1]庚-1-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-4
Figure PCTCN2019094864-appb-000089
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和乙酸(1.5mg,0.024mmol)在室温下搅拌1小时,倒入水(1ml),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到4mg产品A-10-4,产率:31%。LC-MS m/z=550.0[M+1]+。
N-(4-(1-(4-(4-(三氟甲基)-吡啶-2-基)-氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-3-甲基环氧丙烷基-3-甲酰胺A-10-5
Figure PCTCN2019094864-appb-000090
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和3-甲基环氧丙烷-3-羧酸(3mg,0.024mmol)的DMF(1ml)的溶液中在室温下搅拌1小时,倒入水(5mL),用乙酸乙酯(2x5mL),有机相洗涤的饱和NaCl水溶液,有机相分离和用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-5,产率:40%。LC-MS m/z=606.1[M+1]+。
4-(8-氨基-3-(4-(2-羟基-2-甲基丙酰胺)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-6
Figure PCTCN2019094864-appb-000091
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-羟-2-甲基丙酸(2.5mg,0.024mmol)在室温下被搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-6,产率:41%。LC-MS m/z=594.1[M+1]+。
4-(8-氨基-3-(4-(2-甲氧基乙酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-7
Figure PCTCN2019094864-appb-000092
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-甲氧基乙酸(2mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品A-10-7,产率:36%。LC-MS m/z=580.1[M+1]+。
4-(8-氨基-3-(4-(3-甲氧基丙酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-8
Figure PCTCN2019094864-appb-000093
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和3-甲氧基丙酸(2.5mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品A-10-8,产率:35%。LC-MS m/z=594.1[M+1]+。
4-(8-氨基-3-(4-(1-羟基环丙烷甲酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-9
Figure PCTCN2019094864-appb-000094
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和1-羟基环丙烷甲酸(2.5mg,0.024mmol)的DMF(1ml)在室温下搅拌1小时,倒入水(5mL),用乙酸乙酯(2X5毫升)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品A-10-9,产率:36%。LC-MS m/z=592.0[M+1]+。
4-(8-氨基-3-(4-(2-吗啉乙酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-10
Figure PCTCN2019094864-appb-000095
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-吗啉乙酸(3.5mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到7mg产品A-10-10,产率:47%。LC-MS m/z=635.0[M+1]+。
N-(4-(1-(4-(4-(三氟甲基)吡-2-基)-氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-四氢呋喃-2-甲酰胺A-10-11
Figure PCTCN2019094864-appb-000096
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和四氢呋喃-2-甲酸(2.8mg,0.024mmol)的DMF(1mL)中在室温下搅拌1小时,倒入水中(5mL),用乙酸乙酯(2X5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品A-10-11,产率:35%。LC-MS m/z=606.0[M+1]+。
4-(8-氨基-3-(4-(1-氰基环丙烷甲酰胺基)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-12
Figure PCTCN2019094864-appb-000097
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和1-氰基环丙烷甲酸(2.7mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯萃取(2X5mL),有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-12,产率:42%。LC-MS m/z=601.3[M+1]+。
4-(8-氨基-3-(4-(2-氰基乙酰胺)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-13
Figure PCTCN2019094864-appb-000098
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-氰基乙酸(2mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-13,产率:44%。LC-MS m/z=575.2[M+1]+。
4-(8-氨基-3-(4-(2-氰基丙酰胺)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺A-10-14
Figure PCTCN2019094864-appb-000099
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-氰基丙酸(2.4mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品A-10-14,产率:36%。LC-MS m/z=589.3[M+1]+。
N-(4-(1-(4-(4-(三氟甲基)吡啶-2-基)-氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-4-氰基四氢-2H-吡喃-4-甲酰胺A-10-15
Figure PCTCN2019094864-appb-000100
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和4-氰基四氢-2H-吡喃-4-羧酸(3.7mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-15,产率:39%。LC-MS m/z=645.0[M+1]+。
1-(4-(1-(4-(4-三氟甲基)吡啶2基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)氨基甲酰基)环丙烷甲酸A-10-16
Figure PCTCN2019094864-appb-000101
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和环丙烷-1,1-二羧酸(3mg,0.024mmol)的DMF(1mL)溶液中在室温下搅拌1小时,倒入水中(5mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-16,产率:40%。LC-MS m/z=555.3[M+1]+。
N-(4-(1-(4-(4-三氟甲基)吡啶2基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)苯甲酰胺A-10-17
Figure PCTCN2019094864-appb-000102
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和苯甲酸(3mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到8mg产品A-10-17,产率:53%。LC-MS m/z=612.2[M+1]+。
N-(4-(1-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-吡啶-2-甲酸胺A-10-18
Figure PCTCN2019094864-appb-000103
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-吡啶甲酸(3mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯萃取(2x5mL),有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到9mg产品A-10-18,产率:60%。LC-MS m/z=613.2[M+1]+。
N-(4-(1-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)烟酰胺A-10-19
Figure PCTCN2019094864-appb-000104
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和烟酸(3mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到8mg产品A-10-19,产量:54%。LC-MS m/z=613.2[M+1]+。
N-(4-(1-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-2-甲氧基苯甲酰胺A-10-20
Figure PCTCN2019094864-appb-000105
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和2-甲氧基苯甲酸(3.6mg,0.024mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤,蒸干。用柱层析法纯化DCM/MeOH=20:1,得到8mg产品A-10-20,产率:52%。LC-MS m/z=642.3[M+1]+。
N-(4-(1-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)呋喃-2-甲酰胺A-10-21
Figure PCTCN2019094864-appb-000106
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和呋喃-2-羧酸(2.7mg,0.024mmol)的DMF(1mL)中在室温下搅拌1小时,倒入水中(5mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-21,产率:43%。LC-MS m/z=602.2[M+1]+。
N-(4-(1-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-8-氨基咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)噻唑-2-甲酰胺A-10-22
Figure PCTCN2019094864-appb-000107
将化合物A-9(15mg,0.024mmol),HTAU(9.12mg,0.024mmol),DIEA(16mg,0.12mmol)和噻唑-2-羧酸(3mg,0.024mmol)的DMF(1mL)中在室温下搅拌1小时,倒入水中(5mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-10-22,产率:40%。LC-MS m/z=619.2[M+1]+。
化合物A-13-n的合成路线
Figure PCTCN2019094864-appb-000108
苄基4-(8-氨基-1-(4-苯氧苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸A-11
Figure PCTCN2019094864-appb-000109
4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸A-4(300mg, 0.6mmol),4-苯氧苯酸(158mg,0.738mmol),Pd[PPh 3] 4(69mg,0.06mmol)和Cs 2O 3(239mg,0.738mmol)在DME:H 2O(2.5mL:0.5mL)的混合溶剂中加热到80℃过夜。浓缩后采用柱层析法纯化,得到268毫克所需化合物A-11,产率:82%。
3-(4-氨基双环[2.2.1]庚-1-基)-1-(4-苯氧苯基)咪唑并[1,5-a]吡嗪-8-胺A-12
Figure PCTCN2019094864-appb-000110
化合物A-11(260mg,0.48mmol)在DCM/TFA(10mL:10mL)混合溶剂中加热到60℃搅拌18小时。蒸干后加入DCM(2x20mL),浓缩。剩余物溶解在DCM(30mL)中,加入HCl二恶烷溶液,蒸干并加入DCM(2x20mL)。浓缩并加入异丙醚(30mL),搅拌2小时,过滤和用异丙醚洗涤(2x10mL),得到200mg所需的产品A-12的盐酸盐,未经纯化直接用于下一步。
N-(4-(8-氨基-1-(4-苯氧苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-2-羟基-2-甲基丙酰胺A-13-1
Figure PCTCN2019094864-appb-000111
将化合物A-12(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和2-羟基-2-丙酸(3mg,0.029mmol)在室温下搅拌1小时,倒入水中(1mL),用乙基乙酯萃取(2x5mL),有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到8mg产品A-13-1, 产率:57%。LC-MS m/z=498.4[M+1]+。
N-(4-(8-氨基-1-(4-苯氧苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-3-甲氧基丙酰胺A-13-2
Figure PCTCN2019094864-appb-000112
将化合物A-12(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和3-甲氧基丙酸(3mg,0.029mmol)在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-13-2,产率:40%。LC-MS m/z=498.7[M+1]+。
N-(4-(8-氨基-1-(4-苯氧苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-3-甲基环氧丙烷-3-甲酰胺A-13-3
Figure PCTCN2019094864-appb-000113
将化合物A-12(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和3-甲基环氧丙烷-3-羧酸(3.4mg,0.029mmol)的DMF(1mL)中在室温下搅拌1小时,倒入水中(5mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到8mg产品A-13-3,产率:53%。LC-MS m/z=510.2[M+1]+。
N-(4-(8-氨基-1-(4-苯氧苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-2-吗啉乙酰胺A-13-4
Figure PCTCN2019094864-appb-000114
将化合物A-12(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和2-吗啉乙酸(4.2mg,0.029mmol)在DMF(1mL)中室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到9mg产品A-13-4,产率:58%。
化合物A-16-n的合成路线
Figure PCTCN2019094864-appb-000115
苄基4-(8-氨基-1-(4-(5-氟-2-甲氧基苯甲酰胺)苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基-氨基甲酸A-14
Figure PCTCN2019094864-appb-000116
将4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基氨基甲酸A-4(300mg,0.6mmol),N-(4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苄)-5-氟-2-甲氧基苯甲酰胺II-12(284mg,0.738mmol),Pd[PPh 3] 4(69mg,0.06mmol)和Cs 2O 3(239mg,0.738mmol)在DME:H 2O(2.5mL:0.5mL)的混合溶剂中加热到80℃搅拌过夜。浓缩后采用柱层析法纯化,得到285毫克所需化合物A-14,产率:75%。
N-(4-(8-氨基-3-(4-氨基双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)甲基)-5-氟-2-甲氧基苯甲酰胺A-15
Figure PCTCN2019094864-appb-000117
化合物A-14(280mg,0.44mmol)在DCM/TFA(10mL:10毫升)混合溶剂的溶液中加热到60℃搅拌18小时。蒸干并加入DCM(2x20mL),浓缩。剩余物溶解在DCM(30mL)中,加入HCl的二恶烷溶液(10mL),蒸干并加入DCM(2x20mL)。浓缩后加入异丙醚(30mL),搅拌2小时,过滤和用异丙醚洗涤(2x10mL),得到180mg所需的产品A-15的盐酸盐,未经纯化直接用于下一步。
N-(4-(8-氨基-3-(4-(2-羟基-2-甲基丙酰胺)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)甲基)-5-氟-2-甲氧基苯甲酰胺A-16-1
Figure PCTCN2019094864-appb-000118
将化合物A-15(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和2-羟-2-丙酸(2.6mg,0.025mmol)在DMF(1mL)中室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯萃取(2x5mL),有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品A-16-1,产率:40%。LC-MS m/z=587.3[M+1]+。
N-(4-(8-氨基-3-(4-(3-甲氧基丙酰胺)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)-5-氟-2-甲氧基苯甲酰胺A-16-2
Figure PCTCN2019094864-appb-000119
将化合物A-15(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和3-甲氧基丙酸(2.6mg,0.025mmol)在DMF(1mL)中室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到8mg产品A-16-2,产率:40%。LC-MS m/z=587.3[M+1]+。
N-(4-(8-氨基-1-(4-(5-氟-2-甲氧基苯甲酰胺)苯基)咪唑并[1,5-a]吡嗪-3-基)双环[2.2.1]庚-1-基)-3-甲基环氧丙烷-3-甲酰胺A-16-3
Figure PCTCN2019094864-appb-000120
将化合物A-15(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和3-甲基环氧丙烷-3-羧酸(2.9mg,0.025mmol)的DMF(1mL)中在室温下搅拌1小时,倒入水中(5mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到10mg产品A-16-3,产率:66%。LC-MS m/z=599.3[M+1]+。
N-(4-(8-氨基-3-(4-(2-吗啉乙酰胺)双环[2.2.1]庚-1-基)咪唑并[1,5-a]吡嗪-1-基)苯基)-5-氟-2-甲氧基苯甲酰胺A-16-4
Figure PCTCN2019094864-appb-000121
将化合物A-15(15mg,0.025mmol),HATU(9.3mg,0.025mmol),DIEA(19mg,0.15mmol)和2-吗啉乙酸(3.6mg,0.025mmol)在DMF(1mL)中室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到9mg产品A-16-4,产量:57%。LC-MS m/z=628.3[M+1]+。
化合物B-6-n的合成路线
Figure PCTCN2019094864-appb-000122
苄基4-(5-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-4-氨基-7H-吡咯[2,3-d]嘧啶-7-基)双环[2.2.1]庚-1-基氨基甲酸B-4
Figure PCTCN2019094864-appb-000123
将化合物B-3(200mg,0.4mmol),4-(4-(三氟甲基)吡啶2-基)氨基甲酰基)苯硼酸II-3(152mg,0.49mmol),Pd[PPh 3] 4(46mg,0.04mmol)和Cs 2CO 3(159mg,0.49mmol)在DME:H 2O(2.5mL:0.5mL)的混合溶剂中加热到80℃过夜。浓缩后剩余物用甲醇/DCM(1:40)柱层析纯化。得到187mg所需化合物B-4,产率:73%。
4-(4-氨基-7-(4-氨基双环[2.2.1]庚-1-基)-7H-吡咯[2,3-d]嘧啶-5-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺B-5
Figure PCTCN2019094864-appb-000124
化合物B-4(187mg,0.29mmol)在DCM/TFA(10mL:10mL)混合溶剂中加热到60℃18小时。蒸干并加入DCM(2x20mL),浓缩。剩余物中再加入DCM(30mL),加 入盐酸二恶烷溶液中(10mL),蒸干,重新加入DCM(2X20毫升),再蒸干。加入异丙醚(30mL),搅拌2小时,过滤和用异丙醚洗涤(2x10mL),得到所需的产品B-5的盐酸盐,未经纯化直接用于下一步反应。
4-(4-氨基-7-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)-7H-吡咯[2,3-d]嘧啶-5-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺B-6-1
Figure PCTCN2019094864-appb-000125
将化合物B-5(19mg,0.031mmol),HATU(12mg,0.031mmol),DIEA(24mg,0.19mmol)和丁-2-炔酸(2.6mg,0.031mmol)的DMF(1mL)溶液在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到11mg产品B-6-1,产率:59%。LC-MS m/z=574.3[M+1]+。
4-(4-氨基-7-(4-(2-羟基-2-甲基丙酰胺)双环[2.2.1]庚-1-基)-7H-吡咯[2,3-d]嘧啶-5-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺B-6-2
Figure PCTCN2019094864-appb-000126
将化合物B-5(19mg,0.031mmol),HATU(12mg,0.031mmol),DIEA(24mg,0.19mmol)和2-羟基-2-甲基丙酸(3.2mg,0.031mmol)的DMF(1mL)溶液在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗, 有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到9mg产品B-6-2,产率:49%。LC-MS m/z=594.2[M+1]+。
4-(4-氨基-7-(4-(3-甲氧基丙酰胺)双环[2.2.1]庚-1-基)-7H-吡咯[2,3-d]嘧啶-5-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺B-6-3
Figure PCTCN2019094864-appb-000127
将化合物B-5(19mg,0.031mmol),HATU(12mg,0.031mmol),DIEA(24mg,0.19mmol)和3-甲氧基丙酸(3.2mg,0.031mmol)的DMF(1mL)溶液在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到12mg产品B-6-3,产率:66%。LC-MS m/z=594.2[M+1]+。
N-(4-(5-(4-(4-(三氟甲基)吡啶2基)氨基甲酰基苯基)-4-氨基-7H-吡咯[2,3-d]嘧啶-7-基)双环[2.2.1]庚-1-基)-3-甲基环氧丙烷-3-甲酰胺B-6-4
Figure PCTCN2019094864-appb-000128
将化合物B-5(19mg,0.031mmol),HATU(12mg,0.031mmol),DIEA(24mg,0.19mmol)和3-甲基环氧丙烷-3-羧酸(3.6mg,0.031mmol)的DMF(1mL)溶液在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1, 得到12mg产品B-6-4,产率:64%。LC-MS m/z=606.3[M+1]+。
4-(4-氨基-7-(4-(2-吗啉乙酰胺)双环[2.2.1]庚-1-基)-7H-吡咯[2,3-d]嘧啶-5-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺B-6-5
Figure PCTCN2019094864-appb-000129
将化合物B-5(19mg,0.031mmol),HATU(12mg,0.031mmol),DIEA(24mg,0.19mmol)和2-吗啉乙酸(4.5mg,0.031mmol)的DMF(1mL)溶液在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到11mg产品B-6-5,产率:58%。LC-MS m/z=635.2[M+1]+。
化合物C-7-n的合成路线
Figure PCTCN2019094864-appb-000130
苄基4-(7-(4-(4-三氟甲基)吡啶-2-基)氨基甲酰基苯基)-6-氨基-8-羰基-7,8-二氢嘌呤-9-基)双环[2.2.1]庚-1-基氨基甲酸C-5
Figure PCTCN2019094864-appb-000131
将化合物C-4(100mg,0.25mmol),4-(4-(三氟甲基)吡啶2基)氨基甲酰基)苯硼酸II-3(160mg,0.5mmol),醋酸铜(60mg,0.3mmol)和Et 3N(30mg,0.3mmol)在DCM(10毫升)中在室温下搅拌24小时。倒入水中(20mL),用乙酸乙酯(2x10mL)萃取。有机相分离,用无水Na 2SO 4干燥,过滤和蒸干,用柱层析法纯化EA/PE=1:1~EA。得到47mg所需的化合物C-5,产率:29%。LC-MS m/z=657.3[M-1] -
4-(6-氨基-9-(4-氨基双环[2.2.1]庚-1-基)-8-羰基-8,9-二氢嘌呤-7-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺C-6
Figure PCTCN2019094864-appb-000132
将化合物C-5(45mg,0.09mmol)在DCM/TFA(10mL:10mL)混合溶剂中加热到60℃18小时。蒸干并且加入DCM(2x20mL),浓缩。剩余物溶解在DCM(30mL)中,加入盐酸二恶烷溶液中(10mL),蒸干后再加DCM(2x20mL),浓缩后加入异丙醚(30mL),搅拌2小时,过滤和用异丙醚(2x10毫升)洗涤,得到所需的产品C-6的盐酸盐,未经纯化直接用于下一步。
4-(6-氨基-9-(4-丁-2-炔酰胺双环[2.2.1]庚-1-基)-8-羰基-8,9-二氢嘌呤-7-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺C-7-1
Figure PCTCN2019094864-appb-000133
将化合物C-6(13mg,0.022mmol),HATU(9mg,0.022mmol),DIEA(16.8mg,0.13mmol)和丁-2-炔酸(1.8mg,0.022mmol)的DMF(1mL)的溶液中在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,有机相分离,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到5mg产品C-7-1,产率:38%。LC-MS m/z=591.2[M+1]+。
4-(6-氨基-9-(4-(2-吗啉乙酰胺)双环[2.2.1]庚-1-基)-8-羰基-8,9-二氢嘌呤-7-基)-N(4-(三氟甲基)吡啶-2-基)苯甲酰胺C-7-2
Figure PCTCN2019094864-appb-000134
将化合物C-6(13mg,0.022mmol),HATU(9mg,0.022mmol),DIEA(16.8mg,0.13mmol)和2-吗啉乙酸(3.2mg,0.022mmol)的DMF(1mL)溶液在室温下搅拌1小时,倒入水中(1mL),用乙酸乙酯(2x5mL)萃取,有机相用饱和NaCl水溶液冲洗,分离有机相,用无水Na 2SO 4干燥,过滤和蒸干。用柱层析法纯化DCM/MeOH=20:1,得到6mg产品C-7-2,产率:42%。LC-MS m/z=652.2[M+1]+。
化合物D-8-n的合成路线
Figure PCTCN2019094864-appb-000135
苄基4-(3-(4-(4-三氟甲基)吡啶2基)氨基甲酰基苯基)-4-氨基-1H-吡咯[3,4-d]嘧啶1-基)双环[2.2.1]庚烷-1-基氨基甲酸D-6
Figure PCTCN2019094864-appb-000136
将D-5(96mg,0.19mmol),II-3(73mg,0.234mmol),Pd[PPh 3] 4(22mg,0.019mmol),Cs 2CO 3(76.4mg,0.234mmol)在DME/H2O(9mL/1mL)的混合液用N 2脱氧1min后在封管中加热到80℃搅拌12h。冷却后,用盐水(20mL)淬火反应,用EA(2x50mL)萃取。有机相用无水Na 2SO 4干燥,过滤和浓缩。残留物用硅胶柱层析纯化,由用PE/EA(1:1)洗脱,得到所需的产品D-6(42mg,34.4%)。LC-MS m/z=643.2[M+1]+。
4-(4-氨基-1-(4-氨基双环[2.2,1]庚烷-1-基)-1H-吡咯[3,4-d]嘧啶-3-基)-N(4-(三氟甲基)吡啶-2-基)苯酰胺D-7
Figure PCTCN2019094864-appb-000137
将D-6(42mg,0.065mmol)在TFA/DCM(1mL/1mL)的混合物在60℃搅拌12h。 冷却后,蒸干溶剂,加入二恶烷(2mL)的HCl溶液,搅拌10min,浓缩。残留物加入异丙醚(10mL),搅拌,再浓缩。重复上述程序两次。固体被过滤和洗与异丙醚,干燥,得到所需的产品D-7的HCl盐(30mg,75%)。
4-(4-氨基-1-(4-丁-2-炔基氨基双环[2.2.1]庚烷-1-基)-1H-吡咯[3,4-d]嘧啶-3-基)-N(4-(三氟甲基)吡啶-2-基)苯酰胺D-8-1
Figure PCTCN2019094864-appb-000138
将D-7(15mg,0.025mmol)、丁-2-炔酸(2mg,0.024mmol)、HATU(9.1mg,0.024mmol)和DIEA(0.041mL,0.241mmol)在DMF(1mL)中的混合液在室温下搅拌4h。反应液用EA(30mL)稀释,用盐水洗涤,有机相用无水Na 2SO 4干燥,过滤,浓缩。残留物用硅胶柱层析纯化,用PE/EA(1:1)洗脱,得到所需的产品D-8-1(12mg,86%)。LC-MS m/z=575.1[M+1]+。
4-(4-氨基-1-(4-(2-吗啉乙酰胺基)双环[2.2.1]庚烷-1-基)-1H-吡咯[3,4-d]嘧啶-3-基)-N(4-(三氟甲基)吡啶-2-基)苯酰胺D-8-2
Figure PCTCN2019094864-appb-000139
将D-7(15mg、0.025mmol)、2-吗啉乙酸(3.5mg、0.024mmol)、HATU(9.1mg、0.024mmol)和DIEA(0.041mL、0.241mmol)在DMF(1mL)的混合液在室温下搅拌4h。反应用EA(30mL)稀释,用盐水洗涤,有机相用无水Na 2SO 4干燥,过滤,浓缩。残留物 用硅胶柱层析纯化,用PE/EA(1:1)洗脱,得到所需的产品D-8-2(8.5mg,56%)。LC-MS m/z=636.3[M+1]+。
1.对BTK(wt)/BTK(C481S)的体外抑制活性(IC50值的测定)
(1)实验方法
底物溶液的配制是将底物聚(Glu,Tyr)钠盐(Sigma Aldrich,St.Louis,MO)加入到底物反应缓冲液(20mM Hepes(pH 7.5),10mM MgCl2,1mM EGTA,0.02%Brij35,0.02mg/ml BSA,0.1mM Na 3VO 4,2mM DTT和1%DMSO)中(最终底物在反应中的浓度是0.2uM)。将测试化合物用100%DMSO配制成10mM浓度的储备液中,并在384孔循环烯烃共聚物LDV微量培养板中进行10次剂量的3倍连续稀释。将BTK(WT)或者BTK(C481S)激酶(重组人源全长蛋白,组氨酸标签,在昆虫细胞中表达,Invitrogen,Carlsbad,CA)加入底物溶液中并轻轻混合(最终BTK在反应中的浓度为8nM)。然后,通过声学液体转移技术(Echo550;纳升范围)(Labcyte Inc,Sunnyvale,CA)将100%DMSO中的测试化合物加入到激酶反应混合物中,并在室温下温育20分钟。将33P-ATP(特定活性10μCi/μl)加入到反应混合物中以引发反应,随后在室温下孵育2小时。取小部分反应液点在P-81离子交换滤纸(Whatman)上。用0.75%磷酸缓冲液洗去滤纸上未结合的磷酸盐(三次)并干燥后,测量留在滤纸上的放射性。激酶活性数据用测试样品中剩余激酶活性与载体(二甲基亚砜)空白反应的百分比来表示。使用Prism(GraphPad Software)软件对获得数据进行曲线拟合来计算IC 50值。
(2)实验结果
本发明所术大部分化合物拥有比第二代BTK抑制剂Acalabrutinib更强的对BTK(WT)野生型酶活性的抑制能力,更为重要的是绝大部分化合物对BTK(C481S)突变体显示出更杰出的抑制活性,有的甚至小于0.1nM。
表1.BTK(wt)/BTK(C481S)酶活性的抑制结果:A≤0.1nM;0.1nM<B≤1nM;1nM<C≤10nM;10nM<D≤50nM;
Figure PCTCN2019094864-appb-000140
Figure PCTCN2019094864-appb-000141
2.体外肿瘤细胞增殖抑制实验
(1)实验方法
·将肿瘤细胞系(TMD-8/OCY-LY10)悬浮于RPMI1640+FBS10%中,在37℃,5%CO 2的培养箱中进行培养。定期传代,取处于对数生长期的细胞用于铺板。
·用台盼兰进行细胞染色并计数活细胞。
·用培养基调节细胞浓度至7000/孔。
·添加90μl细胞悬浮液至96孔板中,在空白对照空中加入不含细胞的培养液。
·将96孔板中的细胞置于37℃,5%CO 2,及100%相对湿度的培养箱中培养过夜。
·制备400X化合物存储板:将待测化合物和参照药物用DMSO溶解,从最高浓度(400uM)3X梯度稀释至最低浓度(0.61uM)。
·10X化合物工作液的配制:在V形底的96孔板中加入78μL细胞培养液,从400X化合物存储板中吸取2μL化合物加入96孔板的细胞培养液中。在溶媒对照和空白对照中加入2μL DMSO。加入化合物或DMSO后用排枪吹打混匀。
·加药:取10μL的10X化合物工作液按表1所示加入到细胞培养板中。在溶媒对照和空白对照中加入10μL DMSO-细胞培养液混合液。DMSO终浓度为0.25%。
·将96孔细胞板放回培养箱中培养72h。
·将CellTiter-Glo缓冲液融化并放置至室温。
·将CellTiter-Glo底物放置至室温。
·在一瓶CellTiter-Glo底物中加入CellTiter-Glo缓冲液以溶解底物,从而配制CellTiter-Glo工作液。
·缓慢涡旋震荡使充分溶解。
·取出细胞培养板放置30分钟使其平衡至室温。
·在每孔中加入50μL(等于每孔中细胞培养液一半体积)的CellTiter-Glo工作液。用铝箔纸包裹细胞板以避光。
·将培养板在轨道摇床上振摇2分钟以诱导细胞裂解。
·培养板在室温放置10分钟以稳定发光信号。
·在2104EnVision读板器上检测发光信号。
·数据分析:用下列公式来计算检测化合物的抑制率(Inhibition rate,IR):IR(%)=(1–(RLU化合物–RLU空白对照)/(RLU溶媒对照–RLU空白对照))*100%。在Excel中计算不同浓度化合物的抑制率,然后用GraphPadPrism软件作抑制曲线图和计算IC50。
(2)实验结果
对B细胞淋巴瘤细胞TMD-8及OCI-LY10的增殖抑制实验中,化合物A-10-10与Ibrutinib表现出很强的肿瘤细胞增殖抑制活性。
表2.化合物A-10-10/Ibrutinib的肿瘤细胞增殖抑制活性结果:
Figure PCTCN2019094864-appb-000142
Figure PCTCN2019094864-appb-000143
3.药代动力学研究
(1)实验方法
雄性SD大鼠,试验前过夜禁食。试验药物混悬于去离子水配制的0.5%甲基纤维素(MC),0.1%的SDS中(w/w/v),悬浊液浓度分别为1mg/mL,以5mL/kg灌胃给药。给药后15分钟,30分钟和1,2,4,6,8及24小时通过动物眼眶静脉丛采集约0.4mL,全血放入肝素抗凝管中,每个时间点采集三只动物并处死,样品的采集在不同个体完成。全血样品将在15分钟内进行离心,离心在4℃,4200转的条件下离心5分钟。所有血浆样品在分析前保存于-80±15℃的冰箱内。样品分析前先建立一个测定化合物的LC-MS/MS(Waters I Class UPLC串联Xevo TQD质谱)测定方法。通过对采集的血浆进行定量分析。动物的血浆浓度-时间数据用WinNonlin(专业版,版本5.2)软件进行分析。非房室模型用于浓度分析。计算受试化合物的药代动力学参数。
成年雄性比格犬(3只),试验前过夜禁食。试验药物混悬于去离子水配制的0.5%甲基纤维素(MC),0.1%的SDS中(w/w/v),悬浊液浓度分别为1mg/mL,以5mL/kg灌胃给药。给药后15分钟,30分钟和1,2,4,6,8及24小时采血。动物通过水合氯醛浅麻醉,用玻璃采血管于前肢静脉采血约0.5mL全血,放于肝素抗凝管中,样品于4℃,4200转的条件下离心5分钟。所有血浆样品在分析前保存于-80±15℃的冰箱内。样品分析前先建立一个测定化合物的LC-MS/MS(Waters I Class UPLC串联Xevo TQD质谱)测定方法。通过对采集的血浆进行定量分析。动物的血浆浓度-时间数据用WinNonlin(专业版,版本5.2)软件进行分析。非房室模型用于浓度分析。计算受试化合物的药代动力学参数。
(2)实验结果
化合物A-10-10,灌胃给药(5mg/kg)。化合物A-10-10表现出良好的吸收,Cmax分别为8323ng/mL和641ng/mL;而化合物A-10-10有着相当高的血液暴露量(AUC0-inf=73318hr*ng/mL和14867hr*ng/mL)。
表3.大鼠/比格犬(雄性)灌胃给药(5mg/kg)药代动力学数据(t1/2-半衰期;Tmax达峰时间;Cmax最大血药浓度;AUC 0-INF指0-inf时间-浓度曲线下面积)
Figure PCTCN2019094864-appb-000144
Figure PCTCN2019094864-appb-000145
4.对BTK(wt)和BTK(C481S)转染细胞HEK293的pBTK抑制实验
Ibrutinib在临床中出现耐药的主要原因是由于BTK激酶发生C481S突变,开发能够有效抑制BTK(C481S)变异的细胞的BTK抑制剂对克服Ibrutinib耐药性有着重要意义。
(1)实验方法
·用全长人源BTK或BTK[C481S]载体瞬时转染HEK293人胚肾细胞。
·将细胞以预先确定的细胞密度分配到96孔板中。
·用100%DMSO将每个待测化合物进行八次3倍系列稀释。
·然后将化合物在组织培养基中稀释至10倍最终测定浓度和5%DMSO。
·将化合物加入到96孔板中的细胞中(在培养基中稀释10倍),最终浓度为1X化合物和0.5%DMSO。对于正(高信号)对照,细胞用0.5%DMSO单独处理。对于负(低信号)对照,细胞用20uM Ibrutinib处理,最终浓度为0.5%DMSO。
·将细胞与化合物在37℃下孵育2小时。
·将细胞裂解,将裂解物转移至ELISA板上,板上先前涂有捕获底物(人源BTK或BTK[C481S])的抗体。
·洗涤板,然后与HRP连接的抗体孵育以检测总的酪氨酸磷酸化。
·将板洗涤,然后加入HRP底物。在450纳米下读取吸光度。
·基于阳性和阴性对照值的吸光度读数计算%抑制值,并根据以下公式:(%INH=((阳性对照-样本)/(阳性对照-阴性对照))*100。
·Z'值基于以下公式计算:1-[(3*阳性标准偏差+3*阴性标准偏差)/(平均阳性-平均阴性)]。
·将%抑制值vs化合物浓度的对数用GraphPad Prism软件作图。
·利用S形剂量反应曲线拟合后测定IC50值。
(2)实验结果
C481S突变使得Ibrutinib对HEK293细胞BTK磷酸化的抑制从0.021μM下降到1.58μM,而本发明的化合物A-10-10除了对BTK(WT)转染的HEK293细胞有很强的抑 制外(0.077μM),对BTK(C481S)转染的HEK293细胞有着更强的抑制(0.066μM)。
表4.BTK(WT)及BTK(C481S)转染的HEK293细胞pBTK抑制值(IC50)
Figure PCTCN2019094864-appb-000146
5.药效学实验1
(1)实验方法
免疫功能严重缺陷CB17/SCID雌性小鼠购自北京维通利华实验动物技术有限公司,饲养于SPF动物房。人源OCI-LY10细胞(上海君瑞-UFBN0102)体外单层培养,培养条件为RPMI 1640培养基中加10%胎牛血清,100U/mL青霉素和100μg/mL链霉素,37℃,5%CO 2孵箱培养。一周两次进行常规处理传代。当细胞饱和度为80%-90%,数量到达要求时,收取细胞,计数。将0.2mL(1×10 7个)OCI-LY10细胞(加基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,在植入一周左右即可测量出肿瘤大小。使用游标卡尺测量肿瘤的大小,并用以下公式计算肿瘤体积:肿瘤体积=(长×宽 2)/2。肿瘤平均体积达到109mm 3时开始分组给药,将小鼠分为4组(每组8个动物),即溶媒(5%DMSO+20%HP-β-CD)对照组,Ibrutinib灌胃给药组(25mg/kg,1次/日)和化合物A-10-10灌胃给药组(25mg/kg,50mg/kg,2次/日)。将Ibrutinib或化合物A-10-10溶解在5%DMSO+20%HP-β-CD中,以10ml/kg灌胃给药,连续给药28天。
(2)实验结果
化合物A-10-10和Ibrutinib在OCI-LY10移植肿瘤模型中显示出极强的抗肿瘤活性(图-1)。化合物A-10-10灌胃给药(剂量为25mg/kg,bid;50mg/kg,bid)能显著抑制弥漫性大B细胞淋巴瘤细胞株OCI-LY10的生长(TGI=110%,110%;p<0.001,p<0.001)。给药21天后,50mg/kg实验组所有OCI-LY10移植瘤完全消失。给药28天后,25mg/kg实验组所有OCI-LY10移植瘤也完全消失。对照化合物Ibrutinib 25mg/kg组在28天的TGI值为94%(p<0.001)。
受试物对荷瘤鼠的体重变化影响如图1。荷瘤鼠对受试药物A-10-10在所有剂量下都显示出良好的耐受性,所有治疗组均无明显体重下降。
6.药效学实验2
(1)实验方法
免疫功能严重缺陷CB17/SCID雌性小鼠购自北京维通利华实验动物技术有限公司,饲养于SPF动物房。人源TMD-8细胞(上海君瑞-UFBN1682)体外单层培养,培养条件为RPMI 1640培养基中加10%胎牛血清,100U/mL青霉素和100μg/mL链霉素,37℃,5%CO 2孵箱培养。一周两次进行常规处理传代。当细胞饱和度为80%-90%,数量到达要求时,收取细胞,计数。将0.2mL(1×107个)TMD-8细胞(加基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,在植入一周左右即可测量出肿瘤大小。使用游标卡尺测量肿瘤的大小,并用以下公式计算肿瘤体积:肿瘤体积=(长×宽 2)/2。肿瘤平均体积达到107mm 3时开始分组给药,将小鼠分为4组(每组8个动物),即溶媒(5%DMSO+20%HP-β-CD)对照组,Ibrutinib灌胃给药组(25mg/kg,1次/日)和化合物A-10-10灌胃给药组(25mg/kg,50mg/kg,2次/日)。将Ibrutinib或化合物A-10-10溶解在5%DMSO+20%HP-β-CD中,以10ml/kg灌胃给药,连续给药27天。
(2)实验结果
化合物A-10-10和Ibrutinib在TMD-8移植肿瘤模型中显示出极强的抗肿瘤活性(图-2)。化合物A-10-10灌胃给药(剂量为25mg/kg,bid;50mg/kg,bid)能显著抑制弥漫性大B细胞淋巴瘤细胞株TMD-8的生长(TGI=94%,104%;p<0.001,p<0.001)。给药27天后,50mg/kg实验组5/8TMD-8移植瘤完全消失。对照化合物Ibrutinib 25mg/kg组的TGI值为90%(p<0.001)。
受试物对荷瘤鼠的体重变化影响如图2。荷瘤鼠对受试药物A-10-10在所有剂量下都显示出良好的耐受性,所有治疗组均无明显体重下降。

Claims (22)

  1. 式I的化合物
    Figure PCTCN2019094864-appb-100001
    或其药学上可接受的盐、溶剂化物、活性代谢物、多晶型物、酯、光学异构体或前药,其中,A环选自以下结构之一:
    Figure PCTCN2019094864-appb-100002
    R 5选自氢,卤素,氰基,羟基,炔基,氨基,C 1-3烷基,C 1-3烷氧基,C 1-3卤代烷基,C 1-3卤代烷氧基,C 1-3卤代烷氨基,C 3-7环烷基,C 3-7环烷氧基,C 3-7环烷氨基;
    B环为取代或非取代的芳环或杂芳环;C环为取代或非取代的芳环或杂芳环;
    L为单键、或以下结构之一:
    Figure PCTCN2019094864-appb-100003
    R 1选自R 3或以下结构之一:
    Figure PCTCN2019094864-appb-100004
    其中,R 3选自氢、取代或非取代的C 1-6烷基、取代或非取代的C 1-6炔基、取代或非取代的C 1-6烯基、取代或非取代的C 6-10芳基、取代或非取代的C 1-9杂芳基、取代或非取代的C 3-7环烷基、取代或非取代的C 2-7杂环烷氨基;
    R 4选自氢、取代或非取代的C 1-6烷基、取代或非取代的C 6-10芳基、取代或非取代的C 1-9杂芳基、取代或非取代的C 3-7环烷基、取代或非取代的C 3-7杂环烷基;
    R 2选自H、取代或非取代的C 1-3烷基、取代或非取代的C 3-7环烷基、取代或非取代的C 2-7杂环烷基、取代或非取代的C 6-10芳基或取代或非取代的C 1-9杂芳基。
  2. 根据权利要求1所述的化合物,其特征在于,R 1、R 2以及与之相连的N形成取代或非取代的C 2-7杂环,R 3、R 4以及与之相连的N形成C 3-7杂环氨基或C 3-9杂芳环氨基。
  3. 根据权利要求1所述的化合物,其特征在于,所述R 3中的取代的C 1-6烷基、取代的C 1-6炔基、取代的C 1-6烯基、取代的C 6-10芳基、取代的C 1-9杂芳基、取代的C 3-7环烷基、取代的C 2-7杂环烷基中的取代基选自卤素、氰基、羟基、氨基、取代或非取代的酰基胺基、取代或非取代的氨基酰基、取代或非取代的C 1-4烷基、取代或非取代的C 3-7环烷基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 1-4烷基氨基、二[取代或非取代的C 1-4烷基]氨基、取代或非取代的C 3-7环烷氨基、取代或非取代的C 3-7杂环烷氨基、取代或非取代的C 1-3烷氧基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 6-10芳基或取代或非取代的C 3-7杂环烷基中的一个或多个。
  4. 根据权利要求1所述的化合物,其特征在于,所述R 4中的取代的C 1-6烷基、取代的C 6-10芳基、取代的C 1-9杂芳基、取代的C 3-7环烷基,取代的C 3-7杂环烷基中的取代基选自卤素、羟基、氰基、氨基、取代或非取代的C 1-4烯基、取代或非取代的C 3-7环烷基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 1-4烷基氨基、二[取代或非取代的C 1-4烷基]氨基、取代或非取代的C 3-7环烷氨基、取代或非取代的C 3-7杂环烷氨基、取代或非取代的C 1-3烷氧基、取代或非取代的C 3-7环烷氧基、取代或非取代的C 6-10芳基或取代或非取代的C 3-7杂环烷基中的一个或多个。
  5. 根据权利要求1所述的化合物,其特征在于,所述B环中的取代的芳环或杂芳环中的取代基选自卤素、羟基、氰基、氨基、C 1-3烷基、C 1-3烷氧基、C 1-3烷氨基、卤代C 1-3烷基、卤代C 1-3烷氧基中的一个或多个。
  6. 根据权利要求1所述的化合物,其特征在于,所述C环中的取代的芳环或杂芳 环中的取代基选自卤素、羟基、氰基、氨基、C 1-3烷基、C 1-3烷氧基、C 1-3烷氨基、卤代C 1-3烷基、卤代C 1-3烷氧基中的一个或多个。
  7. 根据权利要求1所述的化合物,其特征在于,所述A环选自以下结构之一:
    Figure PCTCN2019094864-appb-100005
    R 5选自氢,卤素,氰基,羟基,炔基,氨基,C 1-3烷基,C 1-3烷氧基,C 1-3卤代烷基,C 1-3卤代烷氧基,C 1-3卤代烷氨基,C 3-7环烷基,C 3-7环烷氧基,C 3-7环烷氨基。
  8. 根据权利要求1所述的化合物,其特征在于,所述B环选自以下结构之一:
    Figure PCTCN2019094864-appb-100006
  9. 根据权利要求1所述的化合物,其特征在于,所述C环选自以下结构之一:
    Figure PCTCN2019094864-appb-100007
  10. 根据权利要求1所述的化合物,其特征在于,所述R 1选自以下结构之一:
    Figure PCTCN2019094864-appb-100008
  11. 根据权利要求1所述的化合物,其特征在于,所述R 2选自H,R 1选自:
    Figure PCTCN2019094864-appb-100009
    其中,R 3选自氢、取代或非取代的C 1-6烷基、取代或非取代的C 1-6炔基、取代或非取代的C 1-6烯基、取代或非取代的C 6-10芳基、取代或非取代的C 1-9杂芳基、取代或非取代的C 3-7环烷基、取代或非取代的C 2-7杂环烷氨基。
  12. 根据权利要求1所述的化合物,其特征在于,所述化合物选自如下任一结构所示:
    Figure PCTCN2019094864-appb-100010
    Figure PCTCN2019094864-appb-100011
  13. 权利要求1~12中任一项所述的化合物在制备预防或治疗异种免疫性疾病、自身免疫性疾病或癌症的药物中的用途。
  14. 根据权利要求13所述的用途,其特征在于,所述异种免疫性疾病、自身免疫 性疾病或癌症与过度布鲁顿酪氨酸激酶活性相关。
  15. 根据权利要求13所述的用途,其特征在于,所述异种免疫性疾病、自身免疫性疾病或癌症与异常B细胞增殖相关。
  16. 根据权利要求13所述的用途,其特征在于,所述异种免疫性疾病为炎性疾病或哮喘。
  17. 根据权利要求13所述的用途,其特征在于,所述自身免疫性疾病为红斑狼疮、慢性淋巴细胞性淋巴瘤、弥漫性大细胞淋巴瘤、滤泡型淋巴瘤或慢性淋巴细胞白血病。
  18. 一种药物组合物,包含一种以上如权利要求1~12中任一项所述的化合物。
  19. 一种药物制剂,包含治疗有效量的如权利要求1~12中任一项所述的化合物,以及在药学上可接受的赋形剂。
  20. 权利要求1~12中任一项所述的化合物的制备方法,其特征在于,包括以下步骤:(S1)化合物IIIA和硼酸或硼酸酯II进行Suzuki偶合得到化合物IV;(S2)化合物IV用三氟乙酸处理脱去苄氧羰基后转化成化合物V的盐酸盐;(S3)化合物V与有机酸偶合得到权利要求1中所述的化合物I;
    Figure PCTCN2019094864-appb-100012
    其中X=卤素,R 2,R 3,L,A环,B环和C环如权利要求1~10中所述。
  21. 权利要求1~12中任一项所述的化合物的制备方法,其特征在于,包括以下步骤:(A1)化合物IIIA用三氟乙酸处理脱去苄氧羰基后转化成化合物VI的盐酸盐;(A2)化合物VI与有机酸偶合得到化合物VII;(A3)化合物VII与硼酸或硼酸酯II进行Suzuki偶合得到权利要求1中所述的化合物I;
    Figure PCTCN2019094864-appb-100013
    其中X=卤素,R 2,R 3,L,A环,B环和C环如权利要求1~10中所述。
  22. 权利要求1~12中任一项所述的化合物的制备方法,其特征在于,包括以下步骤:(B1)化合物IIIB和硼酸II在醋酸铜催化下进行Chan-Lam-Evans偶合得到化合物VIII;(B2)化合物VIII用三氟乙酸处理脱去苄氧羰基后转化成化合物IX的盐酸盐;(B3)化合物IX与有机酸偶合得到权利要求1中所述的化合物I;
    Figure PCTCN2019094864-appb-100014
    其中R 2,R 3,L,A环,B环和C环如权利要求1~10中所述。
PCT/CN2019/094864 2018-09-29 2019-07-05 氨基降茨烷衍生物及其制备方法与应用 WO2020063012A1 (zh)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US17/279,751 US20210332057A1 (en) 2018-09-29 2019-07-05 Aminonorbornane derivative and manufacture method therefor and use thereof
AU2019348752A AU2019348752B2 (en) 2018-09-29 2019-07-05 Aminonorbornane derivative and manufacture method therefor and use thereof
MX2021003662A MX2021003662A (es) 2018-09-29 2019-07-05 Derivado de aminonorbornano y metodo de fabricacion del mismo y uso del mismo.
JP2021517486A JP7370617B2 (ja) 2018-09-29 2019-07-05 アミノノルカンファン誘導体及びその製造方法と使用
CN201980029247.7A CN112135826B (zh) 2018-09-29 2019-07-05 氨基降茨烷衍生物及其制备方法与应用
KR1020217011382A KR20210070304A (ko) 2018-09-29 2019-07-05 아미노노보란 유도체 및 이의 제조방법과 응용
BR112021005960-1A BR112021005960A2 (pt) 2018-09-29 2019-07-05 composto, método de fabricação do mesmo, uso do mesmo, composição farmacêutica e formulação farmacêutica
CA3114259A CA3114259A1 (en) 2018-09-29 2019-07-05 Aminonorbornane derivative and manufacture method therefor and use thereof
EP19866403.9A EP3858833A4 (en) 2018-09-29 2019-07-05 AMINONORDECANE DERIVATIVE, METHOD FOR PREPARATION AND USE
US17/574,716 US11420975B2 (en) 2018-09-29 2022-01-13 Substituted imidazo[1,5-a]pyrazines as Bruton's tyrosine kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201811153123.3A CN110964016B (zh) 2018-09-29 2018-09-29 氨基降茨烷衍生物及其制备方法与应用
CN201811153123.3 2018-09-29

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17/279,751 A-371-Of-International US20210332057A1 (en) 2018-09-29 2019-07-05 Aminonorbornane derivative and manufacture method therefor and use thereof
US17/574,716 Continuation US11420975B2 (en) 2018-09-29 2022-01-13 Substituted imidazo[1,5-a]pyrazines as Bruton's tyrosine kinase inhibitors

Publications (1)

Publication Number Publication Date
WO2020063012A1 true WO2020063012A1 (zh) 2020-04-02

Family

ID=69952831

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/094864 WO2020063012A1 (zh) 2018-09-29 2019-07-05 氨基降茨烷衍生物及其制备方法与应用

Country Status (10)

Country Link
US (2) US20210332057A1 (zh)
EP (1) EP3858833A4 (zh)
JP (1) JP7370617B2 (zh)
KR (1) KR20210070304A (zh)
CN (2) CN110964016B (zh)
AU (1) AU2019348752B2 (zh)
BR (1) BR112021005960A2 (zh)
CA (1) CA3114259A1 (zh)
MX (1) MX2021003662A (zh)
WO (1) WO2020063012A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021180107A1 (en) * 2020-03-12 2021-09-16 Fochon Pharmaceuticals, Ltd. Compounds useful as kinase inhibitors
WO2022037649A1 (en) * 2020-08-20 2022-02-24 Beijing Innocare Pharma Tech Co., Ltd. Heterocyclic compounds as btk inhibitors
CN114634512A (zh) * 2020-12-16 2022-06-17 江苏恒瑞医药股份有限公司 作为布鲁顿酪氨酸激酶抑制剂的化合物、其制备方法和医药应用
WO2024088311A1 (zh) * 2022-10-26 2024-05-02 药捷安康(南京)科技股份有限公司 咪唑并吡嗪衍生物的晶型及其制备方法和应用

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115417789B (zh) * 2022-09-03 2023-08-04 郑州大学 一种治疗帕金森氏病的化合物、其制备方法以及复方药物组合物和应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016109222A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Tertiary alcohol imidazopyrazine btk inhibitors
WO2016109223A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Benzamide imidazopyrazine btk inhibitors
CN106831789A (zh) * 2016-12-21 2017-06-13 南京亘泰医药技术有限公司 布鲁顿酪氨酸激酶抑制剂
CN107602564A (zh) * 2017-09-20 2018-01-19 南京亘泰医药技术有限公司 布鲁顿酪氨酸激酶抑制剂

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1675860B1 (en) * 2003-10-15 2011-09-21 OSI Pharmaceuticals, Inc. Imidazo[1,5-a]pyrazines as tyrosine kinase inhibitors
US7514444B2 (en) * 2006-09-22 2009-04-07 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US7718662B1 (en) * 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
EP2548877A1 (en) * 2011-07-19 2013-01-23 MSD Oss B.V. 4-(5-Membered fused pyridinyl)benzamides as BTK-inhibitors
US8501724B1 (en) 2012-01-31 2013-08-06 Pharmacyclics, Inc. Purinone compounds as kinase inhibitors
KR102181915B1 (ko) 2012-11-13 2020-11-23 어레이 바이오파마 인크. Trka 키나아제 억제제로서 n-피롤리디닐, n'-피라졸릴-우레아, 티오우레아, 구아니딘 및 시아노구아니딘 화합물
US9624224B2 (en) * 2013-09-30 2017-04-18 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
EP3174539A4 (en) 2014-08-01 2017-12-13 Pharmacyclics, LLC Inhibitors of bruton's tyrosine kinase
US10611766B2 (en) 2015-09-16 2020-04-07 Loxo Oncology Inc. Pyrazolopyrimidine derivatives as BTK inhibitors for the treatment of cancer
JP7000333B2 (ja) * 2016-03-16 2022-02-10 クラ オンコロジー,インク. メニン-mllの架橋された二環式阻害剤及びその使用方法
CN107459521B (zh) * 2016-06-03 2021-04-16 合肥中科普瑞昇生物医药科技有限公司 Flt3激酶的新型抑制剂及其用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016109222A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Tertiary alcohol imidazopyrazine btk inhibitors
WO2016109223A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Benzamide imidazopyrazine btk inhibitors
CN106831789A (zh) * 2016-12-21 2017-06-13 南京亘泰医药技术有限公司 布鲁顿酪氨酸激酶抑制剂
CN107602564A (zh) * 2017-09-20 2018-01-19 南京亘泰医药技术有限公司 布鲁顿酪氨酸激酶抑制剂

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CHIRON ET AL., CANCER DISCOVERY, vol. 4, no. 9, 2014, pages 1 - 14
DAVIS ET AL., NATURE, vol. 463, 2010, pages 88 - 94
ELLMEIER ET AL., J EXP MED, vol. 192, 2000, pages 1611
FELDHAHN ET AL., J EXP MED, vol. 201, 2005, pages 1837
ISLAMSMITH, IMMUNOL REV, vol. 178, 2000, pages 49
JANSSONHOLMDAHL, CLIN EXP IMMUNOL, vol. 94, 1993, pages 459
KHAN ET AL., IMMUNITY, vol. 3, 1995, pages 283
PAN ET AL., CHEM.MED.CHEM., vol. 2, 2007, pages 58 - 61
WOYACH, J CLIN ONCOL, vol. 35, 2017, pages 1437 - 1443

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021180107A1 (en) * 2020-03-12 2021-09-16 Fochon Pharmaceuticals, Ltd. Compounds useful as kinase inhibitors
WO2022037649A1 (en) * 2020-08-20 2022-02-24 Beijing Innocare Pharma Tech Co., Ltd. Heterocyclic compounds as btk inhibitors
CN114634512A (zh) * 2020-12-16 2022-06-17 江苏恒瑞医药股份有限公司 作为布鲁顿酪氨酸激酶抑制剂的化合物、其制备方法和医药应用
CN114634512B (zh) * 2020-12-16 2023-11-14 江苏恒瑞医药股份有限公司 作为布鲁顿酪氨酸激酶抑制剂的化合物、其制备方法和医药应用
WO2024088311A1 (zh) * 2022-10-26 2024-05-02 药捷安康(南京)科技股份有限公司 咪唑并吡嗪衍生物的晶型及其制备方法和应用

Also Published As

Publication number Publication date
US20210332057A1 (en) 2021-10-28
EP3858833A4 (en) 2022-06-22
EP3858833A1 (en) 2021-08-04
MX2021003662A (es) 2021-05-28
CN110964016A (zh) 2020-04-07
CN112135826A (zh) 2020-12-25
CN110964016B (zh) 2021-05-28
US20220144843A1 (en) 2022-05-12
JP2022501403A (ja) 2022-01-06
JP7370617B2 (ja) 2023-10-30
US11420975B2 (en) 2022-08-23
AU2019348752B2 (en) 2024-03-28
BR112021005960A2 (pt) 2021-07-13
CA3114259A1 (en) 2020-04-02
CN112135826B (zh) 2021-05-28
KR20210070304A (ko) 2021-06-14
AU2019348752A1 (en) 2021-05-06

Similar Documents

Publication Publication Date Title
WO2020063012A1 (zh) 氨基降茨烷衍生物及其制备方法与应用
CN107108583B (zh) 可用作激酶抑制剂的吲哚甲酰胺化合物
TWI662024B (zh) 雜芳基化合物及其用途
WO2020094104A1 (zh) 一类含氮稠杂环类shp2抑制剂化合物、制备方法和用途
KR101530117B1 (ko) 야누스 키나제 억제제 화합물 및 방법
JP2021528469A (ja) T細胞アクティベーターとして有用なナフチリジノン化合物
CN114025756B (zh) 磷脂酰肌醇3-激酶抑制剂
TW201704237A (zh) 適用於治療與kit及pdfgr相關之病症的組合物
TW201811784A (zh) 吡唑并吡啶化合物及其用途
CN115768750A (zh) Gcn2调节剂化合物
KR102345381B1 (ko) 키나제 억제제로서 유용한 카르바졸 카르복스아미드 화합물
JP2023520759A (ja) がん治療のための置換オキソイソインドリン化合物
WO2022121914A1 (zh) 氧代氮环类衍生物调节剂、其制备方法和应用
CN114907341A (zh) 吡啶并嘧啶酮类衍生物及其制备方法和用途
WO2020143763A1 (zh) 卤代烯丙基胺类化合物及其应用
CN112457326B (zh) 一类芳香杂环并内酰胺类化合物、制备方法和用途
JP2020537692A (ja) ヘテロ環式化合物、ヘテロ環式化合物を含む組成物、及びその使用方法
CN113631557A (zh) Jak激酶抑制剂及其制备方法和在医药领域的应用
KR20230043103A (ko) 술폰아마이드 화합물
CN114206874A (zh) 可用作irak4抑制剂的双环杂芳基化合物
JP2022538495A (ja) 化学化合物
WO2023036252A1 (zh) 吡咯并嘧啶类或吡咯并吡啶类衍生物及其医药用途
WO2019056466A1 (zh) 布鲁顿酪氨酸激酶抑制剂
RU2809188C2 (ru) Аминонорборнановое производное и способ его получения, а также его применение
TWI833819B (zh) 用於治療與apj受體活性相關的病狀的化合物及組成物

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: 19866403

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3114259

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021517486

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021005960

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217011382

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2019866403

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2019866403

Country of ref document: EP

Effective date: 20210429

ENP Entry into the national phase

Ref document number: 2019348752

Country of ref document: AU

Date of ref document: 20190705

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021005960

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210326