WO2019056466A1 - 布鲁顿酪氨酸激酶抑制剂 - Google Patents

布鲁顿酪氨酸激酶抑制剂 Download PDF

Info

Publication number
WO2019056466A1
WO2019056466A1 PCT/CN2017/107638 CN2017107638W WO2019056466A1 WO 2019056466 A1 WO2019056466 A1 WO 2019056466A1 CN 2017107638 W CN2017107638 W CN 2017107638W WO 2019056466 A1 WO2019056466 A1 WO 2019056466A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
compound
group
ring
Prior art date
Application number
PCT/CN2017/107638
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
Publication of WO2019056466A1 publication Critical patent/WO2019056466A1/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
    • A61K31/4985Pyrazines 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
    • A61K31/495Heterocyclic 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • 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
    • A61P37/02Immunomodulators
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the invention belongs to the field of medicine, and in particular relates to Bruton's tyrosine kinase inhibitor, a pharmaceutical composition thereof, a preparation method thereof and use thereof in preparing a medicament.
  • the B-cell receptor (BCR) signaling pathway plays a key role in the maturation, differentiation and development of B cells.
  • Abnormal BCR-mediated signal transduction can cause deregulated B cell activation and/or pathogenic autoantibody formation, leading to a variety of human diseases, including cancer, autoimmune diseases, and xenogenic immune diseases, autoimmune
  • the diseases include lupus erythematosus, chronic lymphocytic lymphoma, diffuse large cell lymphoma, follicular lymphoma or chronic lymphocytic leukemia, and xenogeneic diseases include inflammatory diseases, asthma and the like.
  • BTK Bruton tyrosine kinase
  • BCR non-receptor tyrosine kinase TEC family
  • Btk plays an important role in regulating B cell proliferation and apoptosis (Islam and Smith, Immunol Rev 2000 178:49; Davis et al, Nature 2010 463:88-94), therefore, inhibition of Btk can be used to treat certain B cells. Lymphoma and leukemia (Feldhahn et al, J Exp Med 2005 201:1837).
  • CLL Chronic lymphocytic leukemia
  • NDL non-Hodgkin's lymphoma
  • MCL Mantle cell lymphoma
  • the first-line treatment plan for CLL is the FCR program, which is a combination of fludarabine (F), cyclophosphamide (C) and rituximab (R).
  • the program has a certain effect, but no disease.
  • the progression survival rate was 38%, and grade 3 or 4 adverse reactions occurred (Xia Wei et al., Chinese Journal of Blood Medicine 2013 34(2): 182-183).
  • MCL often uses anthracyclines or drugs containing high doses of cytarabine, but most of them are not sensitive to conventional chemotherapy drugs. Although there are many drugs for treatment, the overall survival of patients has not been significantly prolonged (Chandran et al.
  • Ibrutinib (trade name Imbruvica), jointly developed by Pharmacyclics and Jassen, is the first innovative drug for oral Bruton's tyrosine kinase (BTK) inhibitors, due to its remarkable efficacy, Therapeutic drugs for MCL and CLL were approved on November 13, 2013 and February 12, 2014.
  • Btk-deficient mice In preclinical murine models of systemic lupus erythematosus (SLE), Btk-deficient mice showed a significant improvement in disease progression. Furthermore, Btk-deficient mice are resistant to collagen-induced arthritis (Jansson and Holmdahl, Clin Exp Immunol 1993 94:459). Selective Btk inhibitors have a significant dose-effect relationship in the mouse arthritis model (Pan et al, Chem. Med. Chem. 2007 2: 58-61). Several clinical studies have been conducted into the arthritis of Btk inhibitors.
  • the technical problem to be solved by the present invention is to provide a novel, unreported, compound which is a Bruton's tyrosine kinase inhibitor, a pharmaceutically acceptable salt, solvate, active metabolite, polymorph thereof. , esters, optical isomers or prodrugs, the use of said 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.
  • the technical solution adopted by the present invention is:
  • A is selected from CH or N; and ring B is a substituted or unsubstituted aromatic ring Or an aromatic heterocyclic ring; the C ring is a substituted or unsubstituted aromatic heterocyclic ring containing at least one N; the D ring is a substituted or unsubstituted cyclic olefin; and NR 1 R 2 is a substituent at C at any position on the D ring;
  • 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- a 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 3 , R 4 and N attached thereto may or may not form a C 3-7 heterocyclic amino group or a C 3-9 heteroaryl ring amino group;
  • 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 a C 6-10 aryl group or a substituted or unsubstituted C 1-9 heteroaryl group;
  • R 1 , R 2 and the N to which it is attached may or may not form a substituted or unsubstituted C 2-7 heterocyclic ring;
  • the substituted C 1-6 alkyl group in the R 3 is selected from the group consisting of halogen, hydroxy, cyano, amino, substituted or unsubstituted C 1-4 olefin , substituted or unsubstituted C 3-7 cycloalkyl, substituted or unsubstituted C 3-7 cycloalkoxy, substituted or unsubstituted C 1-4 alkylamino, bis[substituted or unsubstituted C 1-4 alkyl]amino, substituted or unsubstituted C 3-7 cycloalkylamino, substituted or unsubstituted C 3-7 heterocyclo
  • the substituted C 1-6 alkyl group in the R 4 a substituted C 6-10 aryl group, a substituted C 1-9 heteroaryl group, a substituted C 3-7 cycloalkyl group, a substituted C
  • the substituent in the 3-7 heterocycloalkyl group is selected from halogen, hydroxy, cyano, amino, substituted or unsubstituted C 1-4 alkenyl, substituted or unsubstituted C 3-7 cycloalkyl, substituted or non.
  • Substituted C 3-7 cycloalkoxy substituted or unsubstituted C 1-4 alkylamino, bis[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 unsubstituted C 3-7 cycloalkoxy, substituted or unsubstituted C One or more of 6-10 aryl or substituted or unsubstituted C3-7 heterocycloalkyl.
  • the B ring is a monocyclic or polycyclic substituted or unsubstituted aromatic or aromatic heterocyclic ring; the aromatic heterocyclic ring contains one or more N atoms.
  • the B-ring structure is as follows:
  • B 1 is selected from N or CR 5 ;
  • B 2 is selected from N or CR 6 ;
  • B 3 is selected from N or CR 7 ;
  • B 4 is selected from N or CR 8 ; wherein R 5 , R 6 , R 7 and R 8 each independently selected from H, halogen, hydroxy, substituted or unsubstituted C 1-3 alkyl or substituted or unsubstituted C 1-3 alkoxy.
  • the R 5 and R 6 together with the carbon atom to which they are attached may form a C 6-10 aryl group or a C 1-9 heteroaryl group.
  • the C ring is a monocyclic or polycyclic substituted or unsubstituted aromatic heterocyclic ring containing at least one N; the hetero atom is selected from N, O or S.
  • E 1 is selected from CH, N, O or S
  • E 2 is selected from CR 9 , N, O or S
  • E 3 is selected from CR 10 , N, O or S
  • E 4 is selected from CH, N, O or key
  • R 9 is selected from H, halogen, cyano, hydroxy, amino, substituted or unsubstituted C 1-4 alkylamino, [substituted or unsubstituted C 1-4 alkyl] 2 amino, substituted or unsubstituted C 3-6 cycloalkylamino, substituted or unsubstituted C 1-4 alkyl, substituted or unsubstituted C 1-3 alkoxy, substituted or unsubstituted C 3-6 cycloalkyl, substituted or unsubstituted C 3-6 cycloalkoxy, C 6-10 aryl, C 1-9 heteroaryl or C 2-7 heterocycloalkyl; said substitution selected from one or more of halogens; R 10 selected From H, halogen, hydroxy, cyano, substituted or unsubstituted C 1-3 alkyl or substituted or unsubstituted C 1-3 alkoxy; R 9 and R 10 may together form a substituted or unsubsti
  • the C ring is selected from one of the following structures:
  • the D ring is selected from one of the following structures:
  • the R 1 is selected from one of the following structures:
  • the xenogeneic immune disease, autoimmune disease or cancer is associated with excessive Bruton's tyrosine kinase activity.
  • the xenogeneic immune disease, autoimmune disease or cancer is associated with abnormal B cell proliferation.
  • the xenogeneic immune disease is an inflammatory disease or asthma.
  • the autoimmune disease is lupus erythematosus, chronic lymphocytic lymphoma, diffuse large and thin Lymphoma, follicular lymphoma or chronic lymphocytic leukemia.
  • a pharmaceutical composition comprising one or more compounds according to any one of the above.
  • a pharmaceutical formulation comprising a therapeutically effective amount of a compound of any of the above, in a pharmaceutically acceptable excipient.
  • the pharmaceutical preparation formulated for administration route selected from the group consisting of oral administration, parenteral administration, buccal administration, nasal administration, topical administration, or rectal administration.
  • Said pharmaceutical preparation for the treatment of a disease or condition associated with excessive Bruton's tyrosine kinase activity comprising administering said pharmaceutical preparation to a human or mammal in need thereof; said excessive Bruton's tyrosine kinase activity
  • the related disease is a heterogeneous immune disease, an autoimmune disease or a cancer
  • the xenogeneic immune disease is an inflammatory disease, asthma
  • the autoimmune disease is lupus erythematosus, chronic lymphocytic lymphoma, diffuse large cell Lymphoma, follicular lymphoma or chronic lymphocytic leukemia.
  • the invention includes the step of contacting the pharmaceutical formulation with Btk, the contacting step comprising an in vitro or in vivo assay.
  • Process 1 for the preparation of the above compound I comprises the steps of: (1a) compound A is treated with hydrobromic acid to remove benzyloxycarbonyl to give compound 1; (2a) compound 1 is coupled with acid or acid chloride to form compound 2; (3a) compound 2 Suzuki coupling with boric acid B to obtain the compound I described in claim 1;
  • R1, R2, B ring, C ring and D ring are as described above.
  • the preparation method 2 of the above compound I comprises the following steps: (1b) compound A and boric acid B are subjected to Suzuki coupling to obtain compound 3; (2b) compound 3 is treated with hydrobromic acid to remove benzyloxycarbonyl group to obtain compound 4; (3b) compound Coupling with an acid or an acid chloride to form the compound I described in claim 1;
  • R 1 , R 2 , B ring, C ring and D ring are as defined above.
  • R 1 , R 2 , D are as defined above, R is hydrogen, cyano, halogen, trifluoromethyl, trifluoromethoxy, C 1-3 alkyl, C 1-3 alkoxy, C 3-7 cycloalkyl, C 3-7 cycloalkoxy, C 2-7 heterocycloalkyl, C 2-7 heterocycloalkoxy.
  • 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, chromatographic separation and the like.
  • the starting materials required for the synthesis can be synthesized by themselves or purchased from commercial establishments 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 herein can be synthesized using synthetic methods to obtain a single optical isomer or a mixture of optical isomers.
  • the letter superscript indicates the number of the group, and the subscript indicates the number of the atoms, for example, R 1 , R 2 , and R 3 represent the first to third R groups, and the C 1-4 alkyl group contains 1 ⁇ 4 C atomic alkyl groups. The number of C atoms on the substituent is not counted in the main chain.
  • Methyl 4-formylbenzoate 16 (20 g, 122 mmol) and diphenylmethylamine 15 (22.3 g, 122 mmol) eluted in ethyl acetate (240 mL) for 4 hr, cooled to room temperature, filtered, ester / petroleum ether (1/1, v / v , 50mL) , dried over Na 2 SO 4, to give the product 17 (29g, 76%).
  • Methyl 4-(3-bromo-8-chloroimidazo[1,5-a]pyrazin-1-yl)benzoate 22 (518 mg, 1.41 mmol), (2,4-dimethoxyphenyl) Methylamine 23 (471 mg, 2.82 mmol) and DIEA (364 mg, 2.82 mmol) were dissolved in dioxane (20 mL) and heated to 145 ° C for 6 hours under N 2 atmosphere. After the reaction was concentrated, the residue was dissolved in DCM and washed with water. The organic phase was separated, dried over Na 2 SO 4, filtered and concentrated. Ethyl acetate/oil Purification by column gave the desired compound M (630 mg, 90%).
  • the synthesis of the compound I-22 was carried out by using the synthesis method 3 as the starting material of the intermediate 5-22. 22mg after purification
  • the substrate solution was prepared by adding the substrate poly(Glu, Tyr) sodium salt (Sigma Aldrich, St. Louis, MO) to the substrate reaction buffer (20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM EGTA, 0.02% Brij35). , 0.02 mg/ml BSA, 0.1 mM Na3VO4, 2 mM DTT and 1% DMSO) (the final substrate concentration in the reaction was 0.2 uM). Test compounds were formulated in 10% DMSO stocks in 100% DMSO and serially diluted 3 times in 10 doses in 384 well circulating olefin copolymer LDV microplates.
  • BTK kinase (recombinant human full-length protein, histidine tag, expressed in insect cells, Invitrogen, Carlsbad, CA) was added to the substrate solution and gently mixed (final BTK concentration in the reaction was 8 nM).
  • Test compounds in 100% DMSO were then added to the kinase reaction mixture by acoustic liquid transfer technique (Echo 550; nanoliter range) (Labcyte Inc, Sunnyvale, CA) and incubated for 20 minutes at room temperature.
  • 33P-ATP specific activity 10 ⁇ Ci/ ⁇ l
  • Most of the compounds of the present invention have similar ability to inhibit BTK enzyme activity as the reference compound Ibrutinib, and are superior to the reference compound ACP-196 in inhibiting BTK enzyme activity.
  • test compound and the reference drug were dissolved in DMSO or PBS as a mother liquid.
  • the IC50 was determined using GraphPad Prism calculation software. The graphical curve was fitted using a nonlinear regression model with a S-type dose response.
  • Tumor cell proliferation inhibitory activity results of some compounds: I ⁇ 100 nM, 100 nM ⁇ II ⁇ 1 ⁇ M, 1 ⁇ M ⁇ III ⁇ 10 ⁇ M, 10 ⁇ M ⁇ IV ⁇ 100 ⁇ M
  • mice Female CD-1 mice, weighing 16-20 grams, were fasted overnight before the test.
  • the test drug was suspended in 0.5% methylcellulose (MC) prepared in deionized water, 0.1% SDS (w/w/v), and the suspension concentration was 1 mg/mL, respectively. 10 mL/kg was administered by intragastric administration. Approximately 0.3 mL was collected from the orbital venous plexus of the animal for 15 minutes, 30 minutes, and 1, 2, 4, 6, 8 and 24 hours after administration. Whole blood was placed in the heparin anticoagulation tube, and three animals were collected at each time point. At the time of sacrifice, the collection of samples was done in different individuals.
  • MC methylcellulose
  • SDS w/w/v
  • the whole blood sample will be centrifuged within 15 minutes, centrifuged at 4 ° C, and centrifuged for 5 minutes at 4200 rpm. All plasma samples were stored in a refrigerator at -80 ⁇ 15 °C prior to analysis.
  • the compound I-3 prepared in Example 3, the reference compounds Ibrutinib and ACP-196 were administered orally (10 mg/kg) under the same conditions.
  • the substrate solution was prepared by adding the substrate poly(Glu, Tyr) sodium salt (Sigma Aldrich, St. Louis, MO) to the substrate reaction buffer (20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM EGTA, 0.02% Brij35). , 0.02 mg/ml BSA, 0.1 mM Na3VO4, 2 mM DTT and 1% DMSO) (the final substrate concentration in the reaction was 0.2 uM). Test compounds were formulated in 10% DMSO stocks in 100% DMSO and serially diluted 3 times in 10 doses in 384 well circulating olefin copolymer LDV microplates.
  • kinase synthetic human full-length protein, histidine tag, expressed in insect cells, Invitrogen, Carlsbad, CA
  • Test compounds in 100% DMSO were then added to the kinase reaction mixture by acoustic liquid transfer technique (Echo 550; nanoliter range) (Labcyte Inc, Sunnyvale, CA) and incubated for 20 minutes at room temperature.
  • 33P-ATP specific activity 10 ⁇ Ci/ ⁇ l
  • a small portion of the reaction solution was spotted on a P-81 ion exchange filter paper (Whatman).
  • the present invention represents that Compound I-3 has a higher selectivity than the reference compound Ibrutinib.
  • the main reason for the emergence of drug resistance in Ibrutinib is that the BTK inhibitor of cells that can effectively inhibit BTK (C481S) mutation plays an important role in overcoming Ibrutinib resistance due to the C481S mutation in BTK kinase.
  • tissue culture medium was then diluted in tissue culture medium to a 10-fold final assay concentration and 5% DMSO.
  • the Z' value is calculated based on the following formula: 1-[(3* positive standard deviation + 3* negative standard deviation) / (mean positive-average negative)].
  • the C481S mutation reduced the inhibition of BTK phosphorylation of HEK293 cells by Ibrutinib from 0.011 ⁇ M to 1.9 ⁇ M, whereas the compound I-3 of the present invention had strong inhibition of BTK (WT) HEK293 cells (0.024 ⁇ M) to BTK. (C481S) also maintained strong inhibition (0.35 ⁇ M).

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选择性布鲁顿酪氨酸激酶的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

布鲁顿酪氨酸激酶抑制剂 技术领域
本发明属于医药领域,具体是涉及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)。
慢性淋巴细胞白血病(chronic lymphocytic leukemia,CLL)是因形态成熟的淋巴细胞不能够正常凋亡,反而在淋巴组织中克隆性增殖而引发的一种慢性血液系统恶性肿瘤(李菲等,中华血液学杂志,2013 34(5):384)。CLL有一定的家族遗传性,属于B细胞非霍奇金淋巴瘤(non-Hodgkin’s lymphoma,NHL)中较常见的一种。套细胞淋巴瘤(mantle cell lymphoma,MCL)是一种罕见的B细胞NHL,约占全部NHL的5%~10%,兼具恶性淋巴瘤的难治愈性及侵袭性(Novero等,Exp Hematol Oncol 2014 3(1):4)。MCL不易诊断,约85%的患者确诊时已处于晚期;且易复发,是远期生存率最低的亚型淋巴瘤。
目前CLL的一线治疗方案是FCR方案,即氟达拉滨(F)、环磷酰胺(C)和利妥昔单抗(R)三者联合治疗的方案,该方案有一定疗效,但无疾病进展生存率为38%,3或4级不良反应时有发生(夏奕等,中华血液杂志2013 34(2):182-183)。MCL常选用蒽环类或含大剂量阿糖胞苷的药物,然而对常规化疗药大多不敏感,虽然已有较多药物用于治疗,但患者的总生存期并没有明显延长(Chandran等,Leuk Lymphoma 2012  53(8):1488-1493);尽管也采用药物与单抗联合化疗的方案治疗MCL,但毒性较大,感染发生率约为14%,3~4级不良反应发生率高达87%(朱军等,中国肿瘤临床杂志2011 38(18):1067-1069)。因此,急待开发更有效安全的药物。由Pharmacyclics公司和杨森制药(Jassen)共同研制的依鲁替尼(Ibrutinib,商品名Imbruvica)作为口服的布鲁顿酪氨酸激酶(BTK)抑制剂类首创新药,因其显著的疗效,分别于2013年11月13日和2014年2月12日批准为MCL和CLL的治疗药物。
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)。目前已有几个Btk抑制剂进入关节炎的临床研究。
发明内容
本发明要解决的技术问题是提供新颖的、未见文献报道的可作为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 PCTCN2017107638-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选自CH或N;B环为取代或非取代的芳环或芳杂环;C环为取代或非取代的含至少一个N的芳杂环;D环为取代或非取代的环烯烃;NR1R2为D环上任意位置的C上的取代基;
R1选自R3或以下结构之一,
Figure PCTCN2017107638-appb-000002
R3选自氢、取代或非取代的C1-6烷基、取代或非取代的C1-6炔基、取代或非取代的C1-6烯基、取代或非取代的C6-10芳基、取代或非取代的C1-9杂芳基、取代或非取代的C3-7环烷基或取代或非取代的C2-7杂环烷氨基;
R4选自氢、取代或非取代的C1-6烷基、取代或非取代的C6-10芳基、取代或非取代的C1-9杂芳基、取代或非取代的C3-7环烷基、取代或非取代的C3-7杂环烷基;
R3、R4以及与之相连的N可形成也可不形成C3-7杂环氨基或C3-9杂芳环氨基;
R2选自H、取代或非取代的C1-3烷基、取代或非取代的C3-7环烷基、取代或非取代的C2-7杂环烷基、取代或非取代的C6-10芳基或取代或非取代的C1-9杂芳基;
R1、R2以及与之相连的N可形成也可不形成取代或非取代的C2-7杂环;
优选的,所述R3中的取代的C1-6烷基、取代的C1-6炔基、取代的C1-6烯基、取代的C6-10芳基、取代的C1-9杂芳基、取代的C3-7环烷基或取代的C2-7杂环烷氨基中的取代基选自卤素、羟基、氰基、氨基、取代或非取代的C1-4烯基、取代或非取代的C3-7环烷基、取代或非取代的C3-7环烷氧基、取代或非取代的C1-4烷基氨基、二[取代或非取代的C1-4烷基]氨基、取代或非取代的C3-7环烷氨基、取代或非取代的C3-7杂环烷氨基、取代或非取代的C1-3烷氧基、取代或非取代的C3-7环烷氧基、取代或非取代的C6-10芳基或取代或非取代的C3-7杂环烷基中的一个或多个。
优选的,所述R4中的取代的C1-6烷基、取代的C6-10芳基、取代的C1-9杂芳基、取代的C3-7环烷基,取代的C3-7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C1-4烯基、取代或非取代的C3-7环烷基、取代或非取代的C3-7环烷氧基、取代或非取代的C1-4烷基氨基、二[取代或非取代的C1-4烷基]氨基、取代或非取代的C3-7 环烷氨基、取代或非取代的C3-7杂环烷氨基、取代或非取代的C1-3烷氧基、取代或非取代的C3-7环烷氧基、取代或非取代的C6-10芳基或取代或非取代的C3-7杂环烷基中的一个或多个。
优选的,所述B环为单环或多环的取代或非取代的芳环或芳杂环;所述芳杂环含一个或多个N原子。
更优选的,所述B环结构如下式:
Figure PCTCN2017107638-appb-000003
其中,B1选自N或CR5;B2选自N或CR6;B3选自N或CR7;B4选自N或CR8;其中,R5、R6、R7和R8各自独立的选自H、卤素、羟基、取代或非取代C1-3烷基或取代或非取代C1-3烷氧基。
所述R5和R6与它们所连接的碳原子可以一起形成C6-10芳基或C1-9杂芳基。
优选的,C环为单环或多环的取代或非取代的含至少一个N的芳杂环;所述杂原子选自N、O或S。
进一步的,所述C环结构如下式:
Figure PCTCN2017107638-appb-000004
其中,E1选自CH、N、O或S;E2选自C-R9、N、O或S;E3选自C-R10、N、O或S;E4选自CH、N、O或键;
其中R9选自H、卤素、氰基、羟基、氨基、取代或非取代的C1-4烷基氨基、[取代或非取代的C1-4烷基]2氨基、取代或非取代的C3-6环烷氨基、取代或非取代的C1-4烷基、取代或非取代的C1-3烷氧基、取代或非取代的C3-6环烷基、取代或非取代的C3-6环烷氧基,C6-10芳基、C1-9杂芳基或C2-7杂环烷基;所述取代选自卤素中的一个或多个;R10选自H、卤素、羟基、氰基、取代或非取代的C1-3烷基或取代或非取代的C1-3烷氧基;R9和R10可以一起形成取代或非取代的C3-7环烯基或取代或非取代的C2-6杂环烯基;所述取代的取代基团选自C1-3烷基、C1-3烷氧基或卤素中的一个或多个。
更优选的,所述C环选自以下结构之一:
Figure PCTCN2017107638-appb-000005
优选的,所述D环选自以下结构之一:
Figure PCTCN2017107638-appb-000006
优选的,所述R1选自以下结构之一:
Figure PCTCN2017107638-appb-000007
最优选的,所述化合物如下图中任一结构所示:
Figure PCTCN2017107638-appb-000008
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:(1a)化合物A用氢溴酸处理脱去苄氧羰基得到化合物1;(2a)化合物1和酸或酰氯偶合生成化合物2;(3a)化合物2和硼酸B进行Suzuki偶合得到权利要求1中所述的化合物I即可;
其中R1,R2,B环,C环和D环如前所述。
Figure PCTCN2017107638-appb-000009
上述化合物I的制备方法2,包括以下步骤:(1b)化合物A和硼酸B进行Suzuki偶合得到化合物3;(2b)化合物3用氢溴酸处理脱去苄氧羰基得到化合物4;(3b)化合物4和酸或酰氯偶合生成权利要求1中所述的化合物I即可;
Figure PCTCN2017107638-appb-000010
其中R1,R2,B环,C环和D环如前所述。
上述化合物I的制备方法3,当B环为苯环,C环为取代或非取代的吡啶环时,包括以下步骤:(1c)化合物M和硼酸酯L进行Suzuki偶合得到化合物5;(2c)化合物5水解氨化得到化合物6;(3c)化合物6与卤代吡啶在钯催化下生成化合物7;(4c)化合物7用TFA脱保护得到化合物8;(5c)化合物8和酸或酰氯偶合生成权利要求1中所述的化合物I即可;
Figure PCTCN2017107638-appb-000011
其中R1,R2,D环如前所述,R为氢,氰基,卤素,三氟甲基,三氟甲氧基,C1-3烷基,C1-3烷氧基,C3-7环烷基,C3-7环烷氧基,C2-7杂环烷基,C2-7杂环烷氧基。
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:R1、R2、R3 表示第1~3个R基团,C1-4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。
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A-1的合成
中间体A-1的合成路线
Figure PCTCN2017107638-appb-000012
4-(苄氧基羰氨基)环己-1-烯羧酸10
Figure PCTCN2017107638-appb-000013
向4-氧代环己基氨基甲酸苄酯9(1g,4mmol)和N-苄基-N,N,N-三乙基氯化铵(10mg,0.044mmol)的CHBr3(15mL)混合液中在冰浴下滴加KOH(1.58g,28mmol)水溶液(15mL)。然后将混合液在室温下搅拌2小时。加入水(20ml),水相用DCM(2×20mL)洗涤,然后用1M HCl水溶液将pH调节至4,用DCM(3x20mL)萃取,干燥,过滤,浓缩得到所需产物10(300mg,27%)。LC-MS m/z=276.1[M+1]+
4-(((3-氯吡嗪-2-基)甲基)氨基甲酰基)环己-3-烯基氨基甲酸苄酯12
Figure PCTCN2017107638-appb-000014
向(3-氯吡嗪-2-基)甲胺盐酸盐11(102mg,0.57mmol)和DIEA(146mg,1.13mmol) 的DCM(10mL)溶液中加入4-(苄氧基羰氨基)环己-1-烯羧酸10(156mg,0.57mmol)和HATU(216mg,0.57mmol)的DCM溶液。然后将混合物室温下搅拌过夜。倒入水(30mL)中,分离有机相,用无水Na2SO4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油醚=1:1洗脱得到化合物12(160mg,70%)。LC-MS m/z=401.2[M+1]+
4-(8-氯咪唑并[1,5-a]吡嗪-3-基)环己-3-烯基氨基甲酸苄酯13
Figure PCTCN2017107638-appb-000015
向4-(((3-氯吡嗪-2-基)甲基)氨基甲酰基)环己-3-烯基氨基甲酸苄酯12(50mg,0.125mmol)和吡啶(10mg,0.125mmol)在乙腈(15mL)的溶液中加入PCl5(104mg,0.5mmol),然后加热至56℃1小时。将其倒入NaHCO3水溶液中,将pH调节至9并用乙酸乙酯(2×20mL)萃取,分离有机相,用Na2SO4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油醚=1:1洗脱得到化合物13(38mg,80%)。LC-MS m/z=383.1[M+1]+
4-(8-氯-1-碘咪唑并[1,5-a]吡嗪-3-基)环己-3-烯基氨基甲酸苄酯14
Figure PCTCN2017107638-appb-000016
向化合物13(2g,5.23mmol)的DMF(2ml)溶液中加入NIS(2.36g,10.46mmol),加热至60℃ 2小时。倒入水(40ml)中,用乙酸乙酯(2×20ml)萃取。有机相用饱和NaHCO3水溶液洗涤。分离有机相,用无水Na2SO4干燥,过滤并浓缩。通过柱色谱法用乙酸乙酯/石油醚=1:6洗脱得到化合物14(800mg,30%)。LC-MS m/z=509.1[M+1]+
4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)环己-3-烯基氨基甲酸苄酯A-1
Figure PCTCN2017107638-appb-000017
化合物14(600mg,1.18mmol)在IPA(30ml)中的溶液中加入氨水(6ml)并加热至120℃5小时。蒸发溶剂并加入1ml饱和NaHCO3水溶液和20ml水。用乙酸乙酯(2×20ml)萃取。分离有机相并用无水Na2SO4干燥,过滤并浓缩。将剩余物通过柱色谱法用MeOH/DCM(1:20)洗脱得到化合物A-1(351mg,61%)。LC-MS m/z=490.0[M+1]+
中间体M的合成
中间体M的合成路线
Figure PCTCN2017107638-appb-000018
(E)-4-((二苯甲基亚氨基)甲基)苯甲酸甲酯17
Figure PCTCN2017107638-appb-000019
4-甲酰基苯甲酸甲酯16(20g,122mmol)和二苯基甲胺15(22.3g,122mmol)在乙酸乙酯(240mL)中回流4小时,冷却至室温,过滤,固体用在乙酸乙酯/石油醚(1/1,v/v,50mL)洗涤,用Na2SO4干燥,得到产物17(29g,76%)。
4-(氨基(3-氯吡嗪-2-基)甲基)苯甲酸甲酯19
Figure PCTCN2017107638-appb-000020
在0℃条件下,向(E)-4-((二苯甲基亚氨基)甲基)苯甲酸甲酯17(20g,60.7mmol)的THF(210mL)溶液中滴加KHMDS(72.6mL,72.6mmol),混合物在该温度下搅拌30分钟,并加入2,3-二氯吡嗪18(10.8g,72.6mmol)的THF(20mL)溶液。反应混合物在0℃下搅拌20分钟,在室温下搅拌40分钟。反应完成后,将混合物用乙酸乙酯和水萃取。有机相用3N HCl水溶液(150mL)处理15分钟。分离后有机层用3N HCl水溶液萃取。合并的水相用EA洗涤后用Na2CO3碱化至pH=9。用乙酸乙酯萃取水溶液,将合并的有机相用Na2SO4干燥,过滤并浓缩。用乙酸乙酯/石油醚
Figure PCTCN2017107638-appb-000021
过柱纯化,得到产物 19(8.4g,50%)。
1H NMR(400MHz,CDCl3):δ3.91(s,3H);5.63(s,1H);7.45-7.47(d,J=8.0Hz,2H);7.99-8.01(d,J=8.0Hz,2H);8.31(d,J=2.0Hz,1H);8.57(d,J=2.4Hz,1H)。LC-MS m/z=278.1[M+1]+
4-((3-氯吡嗪-2-基)(甲酰氨基)甲基)苯甲酸甲酯20
Figure PCTCN2017107638-appb-000022
将4-(氨基(3-氯吡嗪-2-基)甲基)苯甲酸甲酯19(2g,7.22mmol),甲酸(332mg,7.22mmol),HATU(2.74g,7.22mmol)和DIEA(1.86g,14.44mmol)在DMF(20mL)中的混合物在室温下搅拌12小时,倒入水中,用乙酸乙酯(2×20mL)萃取,分离有机相,用Na2SO4干燥,过滤并浓缩,用乙酸乙酯/石油醚
Figure PCTCN2017107638-appb-000023
过柱纯化,得到所需化合物20(985mg,45%)。
1H NMR(400MHz,CDCl3):δ3.91(s,3H);6.70(m,1H);7.45(m,3H);8.01(m,2H);8.31(s,1H);8.41(m,1H);8.57(m,1H)。LC-MS m/z=306.3[M+1]+
4-(8-氯咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯21
Figure PCTCN2017107638-appb-000024
在0℃下,向4-((3-氯吡嗪-2-基)(甲酰氨基)甲基)苯甲酸甲酯20(943mg,3.09mmol)的乙腈(60mL)的溶液中滴加DMF(0.23mL)和POCl3(1.42g,9.27mmol),将其升温至室温并搅拌4小时。将其浓缩并倒入冰NaHCO3水溶液中。用乙酸乙酯萃取(2×30mL),分离有机相,用Na2SO4干燥,过滤并浓缩,用乙酸乙酯/石油醚
Figure PCTCN2017107638-appb-000025
过柱纯化得到所需化合物21(750mg,85%)。
1H NMR(400MHz,CDCl3):δ3.97(s,3H);7.44(m,1H);7.82(m,3H);8.15(m,2H);8.36(s,1H)。LC-MS m/z=288.1[M+1]+
4-(3-溴-8-氯咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯22
Figure PCTCN2017107638-appb-000026
在0℃下,向甲基4-(8-氯咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯21(750mg,2.6mmol)的DMF(20mL)溶液中加入NBS(558mg,3.12mmol),在0℃下搅拌2小时。用乙酸乙酯(2×30mL)萃取,有机相用饱和NaCl水溶液洗涤,分离有机相,用Na2SO4干燥,过滤并浓缩,用乙酸乙酯/石油醚
Figure PCTCN2017107638-appb-000027
过柱纯化得到所需化合物22(762mg,80%)。
1H NMR(400MHz,CDCl3):δ3.97(s,3H);7.54(m,1H);7.80(m,3H);8.15(m,2H)。LC-MS m/z=366.1[M+1]+
4-(8-(2,4-二甲氧基苄基氨基)-3-溴咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯M
Figure PCTCN2017107638-appb-000028
将4-(3-溴-8-氯咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯22(518mg,1.41mmol),(2,4-二甲氧基苯基)甲胺23(471mg,2.82mmol)和DIEA(364mg,2.82mmol)溶解在二恶烷(20mL)中,在密封管中,在N2气氛下加热至145℃6小时。反应完后浓缩,并将剩余物溶于DCM中,用水洗涤。分离有机相,用Na2SO4干燥,过滤并浓缩。用乙酸乙酯/石油
Figure PCTCN2017107638-appb-000029
Figure PCTCN2017107638-appb-000030
过柱纯化得到所需化合物M(630mg,90%)。
1H NMR(400MHz,DMSO):δ3.61(s,3H);3.73(s,3H);3.90(s,3H);5.98(m,1H);6.41(m,1H);6.50(m,1H);7.14(m,1H);7.29(m,1H);7.48(m,1H);7.75(m,2H);8.04(m,2H)。LC-MS m/z=498.3[M+1]+
中间体硼酸的合成
中间体B-1的合成路线
Figure PCTCN2017107638-appb-000031
4-溴-N-(吡啶-2-基)苯甲酰胺
Figure PCTCN2017107638-appb-000032
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POCl3(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(3×40mL)萃取。有机相用饱和NaCl水溶液(2×50mL)洗涤。将有机相用无水Na2SO4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油=
Figure PCTCN2017107638-appb-000033
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 PCTCN2017107638-appb-000034
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),PdCl2(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(2×40mL)萃取。分离有机相,用无水Na2SO4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油醚=
Figure PCTCN2017107638-appb-000035
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B-1
Figure PCTCN2017107638-appb-000036
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:H2O(24mL:6mL)的混合溶剂中的溶液中加入NaIO4(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Na2SO4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(1.4g,93%)。LC-MS m/z=243.1[M+1]+
中间体B-2的合成路线
Figure PCTCN2017107638-appb-000037
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 PCTCN2017107638-appb-000038
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)溶液中加入在冰浴中滴加草酰氯(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 PCTCN2017107638-appb-000039
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Na2SO4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B-2
Figure PCTCN2017107638-appb-000040
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:H2O(24mL:6mL)的混合溶剂中的溶液中加入NaIO4(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Na2SO4干燥,过滤并浓缩。通过柱色谱法用MeOH/DCM=1:10纯化。得到产物(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B-3
Figure PCTCN2017107638-appb-000041
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B-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B-4
Figure PCTCN2017107638-appb-000042
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B-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-乙基吡啶-2-基)氨基甲酰基)苯基硼酸B-5
Figure PCTCN2017107638-appb-000043
以4-乙基-吡啶-2-胺(459mg,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B-2相同的合成方法得到1.06g所需化合物。LC-MS m/z=271.2[M+1]+
4-((4-氰基吡啶-2-基)氨基甲酰基)苯基硼酸B-6
Figure PCTCN2017107638-appb-000044
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B-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B-7的合成路线
Figure PCTCN2017107638-appb-000045
4-溴-2-氟-N-(吡啶-2-基)苯甲酰胺
Figure PCTCN2017107638-appb-000046
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Na2SO4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 PCTCN2017107638-appb-000047
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),PdCl2(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Na2SO4干燥,过滤并蒸发。通过柱色谱法用乙酸乙酯/石油醚=
Figure PCTCN2017107638-appb-000048
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B-7
Figure PCTCN2017107638-appb-000049
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:H2O(24mL:6mL)的混合溶剂中的溶液中加入NaIO4(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Na2SO4干燥,过滤并浓缩。通过柱色谱法用MeOH/DCM=1:10纯化。得到产物(605mg,82%)。LC-MS m/z=261.1[M+1]+
4-((4-甲氧基吡啶-2-基)氨基甲酰基)苯基硼酸B-8
Figure PCTCN2017107638-appb-000050
以4-溴苯甲酸和4-甲氧基-吡啶-2-胺为原料,使用与B-1相同的合成方法得到64mg所需化合物。LC-MS m/z=273.1[M+1]+
(哒嗪-3-基氨基甲酰基)苯基硼酸B-9
Figure PCTCN2017107638-appb-000051
以哒嗪-3-胺(357mg,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B-2相同的合成方法得到425mg所需化合物。LC-MS m/z=244.1[M+1]+
(噻唑-2-基氨基甲酰基)苯基硼酸B-10
Figure PCTCN2017107638-appb-000052
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B-2相同的合成方法得到580mg所需化合物。LC-MS m/z=249.1[M+1]+
2-氟-4-(吡啶-2-基氨基甲酰基)苯基硼酸B-11
Figure PCTCN2017107638-appb-000053
以4-溴-3-氟-苯甲酸和吡啶-2-胺为原料,使用与B-1相同的合成方法得到138mg所需化合物。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B-12
Figure PCTCN2017107638-appb-000054
以4-溴-3-氟-苯甲酸和4-(三氟甲基)-吡啶-2-胺为原料,使用与B-1相同的合成方法得到130mg所需化合物。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B-13
Figure PCTCN2017107638-appb-000055
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B-7相同的合成方法得到440mg所需化合物。LC-MS m/z=341.0[M+1]+
2-甲基-4-((4-(三氟甲基)吡啶-2-基)氨基甲酰基)苯基硼酸B-14
Figure PCTCN2017107638-appb-000056
以4-溴-3-甲基苯甲酸(1g,4.65mmol)和4-(三氟甲基)-吡啶-2-胺(753mg,4.65mmol) 为原料,使用与B-7相同的合成方法得到325mg所需化合物。LC-MS m/z=325.0[M+1]+
中间体硼酸酯L-1a/L-1b的合成路线
Figure PCTCN2017107638-appb-000057
4-(叔丁氧羰基)环戊-1-烯基三氟甲磺酸酯L-1a-1/3-(叔丁氧羰基)环戊-1-烯基三氟甲磺酸酯L-1b-1
Figure PCTCN2017107638-appb-000058
向THF(50mL)的溶剂中加入1M LiHMDS(25mL,25mmol),然后冷却至-78℃,在THF(30mL)中的3-氧代环戊基氨基甲酸叔丁酯(2g,10mmol)在THF在20分钟后滴加N,N-双(三氟甲磺酰基)苯胺(3.92g,11mmol)在THF(10mL)中的溶液。将混合物冷却至室温并搅拌1.5小时,加入水(20mL),加入1M NaOH水溶液(10mL),加入甲苯(20mL),在室温下搅拌30分钟,分离有机相,水层用甲苯(2×20mL)萃取。有机相用饱和的KHCO3水溶液和盐水洗涤。干燥并过滤,浓缩并通过EA-PE=
Figure PCTCN2017107638-appb-000059
的柱色谱纯化。得到混合物L-1a-1/L-1b-1(1.5g,45%)。
3-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)环戊-3-烯基氨基甲酸叔丁酯L-1a/3-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)环戊-2-烯基氨基甲酸叔丁酯L-1b
Figure PCTCN2017107638-appb-000060
将B-1a和B-1b(1.5g,4.5mmol),4,4,5,5-四甲基-2-(4,4,5,5-四甲基-2--1,3,2-二氧杂硼杂环戊烷-2-基)-1,3,2-二氧杂硼杂环戊烷(1.7g,6.8mmol),PdCl2dppf(329mg,0.45mmol) 和KOAc(1.3g,13.5mmol)的二恶烷(30mL)的混合物加热至100℃6小时。加入浓缩物和DCM(40mL)并过滤,用DCM(2×10mL)洗涤,将有机相浓缩并通过EA/PE=
Figure PCTCN2017107638-appb-000061
的柱色谱纯化。得到混合物L-1a/L-1b(600mg,43%)。
5-(叔丁氧羰基)环己-1-烯基三氟甲磺酸酯L-2a-1/3-(叔丁氧羰基)环己-1-烯基三氟甲磺酸酯L-2b-1
Figure PCTCN2017107638-appb-000062
向THF(50mL)的溶剂中加入1M LiHMDS(29mL,29mmol),然后冷却至-78℃,在THF(30mL)中的3-氧代环己基氨基甲酸叔丁酯(2.5g,11.7mmol)在THF在20分钟后滴加N,N-双(三氟甲磺酰基)苯胺(4.61g,12.9mmol)在THF(10mL)中的溶液。将混合物冷却至室温并搅拌1.5小时,加入水(20mL),加入1M NaOH水溶液(10mL),加入甲苯(20mL),在室温下搅拌30分钟,分离有机相,水层用甲苯(2×20mL)萃取。有机相用饱和的KHCO3水溶液和盐水洗涤。干燥并过滤,浓缩并通过EA-PE=
Figure PCTCN2017107638-appb-000063
的柱色谱纯化。得到混合物L-2a-1/L-2b-1(1.6g,40%)。
3-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)环己-3-烯基氨基甲酸叔丁酯L-2a/3-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)环己-2-烯基氨基甲酸叔丁酯L-2b
Figure PCTCN2017107638-appb-000064
将B-3a-1和B-3b-1(1.6g,4.6mmol),4,4,5,5-四甲基-2-(4,4,5,5-四甲基-2--1,3,2-二氧杂硼杂环戊烷-2-基)-1,3,2-二氧杂硼杂环戊烷(1.77g,6.9mmol),PdCl2dppf(336mg,0.46mmol)和KOAc(1.35g,13.8mmol)在二恶烷(30mL)中的混合物加热至100℃6小时。加入浓缩物和DCM(40mL)并过滤,用DCM(2×10mL)洗涤,将有机相浓缩并通过EA/PE=
Figure PCTCN2017107638-appb-000065
的柱色谱纯化。得到1.8g混合物L-2a/L-2b。
实施例1
化合物I-1的合成路线
Figure PCTCN2017107638-appb-000066
4-(8-氨基-1-(4-(吡啶-2-基氨基甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)环己-3-烯基氨基甲酸苄酯24-1
Figure PCTCN2017107638-appb-000067
将中间体A-1(30mg,0.06mmol),4-(吡啶-2-基氨基甲酰基)苯基硼酸B-1(18mg,0.074mmol),Pd(PPh3)4(7mg,0.006mmol)和Cs2CO3(25mg,0.074mmol)在DME:H2O(2.5ml:0.5ml)的混合溶剂中的混合物加热至80℃过夜。将其浓缩并通过柱色谱法纯化,用EA-MeOH/DCM(1:40)洗脱得到化合物24-1(25mg,75%)。
LC-MS m/z=560.2[M+1]+
4-(8-氨基-3-(4-氨基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺25-1
Figure PCTCN2017107638-appb-000068
向化合物24-1(20mg,0.036mmol)的DCM(5ml)溶液中加入HBr(33%,在AcOH中)溶液(2滴)。将悬浮液在室温下搅拌2小时。蒸发溶剂得到粗产品25-1的氢溴酸盐,不经纯化直接用于下一步反应。LC-MS m/z=426.1[M+1]+
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺I-1
Figure PCTCN2017107638-appb-000069
向4-(3-(7-氮杂-双环[2.2.1]庚-1-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺氢溴酸盐25-1(30mg,0.045mmol)的DMF(1mL)溶液中加入HATU(8.5mg,0.023mmol)和丁-2-炔酸(1.9mg,0.023mmol)在DMF中的混合物,然后加入DIEA(29mg,0.225mmol),将反应在室温下搅拌10分钟。加入水(20ml),用乙酸乙酯(2×10ml)萃取。有机相用饱和NaCl水溶液(2×10ml)洗涤。分离有机相并干燥。将其蒸发并通过TLC柱纯化,得到化合物I-1。LC-MS m/z=492.2[M+1]+
实施例2
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-氟吡啶-2- 基)苯甲酰胺I-2
Figure PCTCN2017107638-appb-000070
化合物I-2的合成以中间体A-1和B-2为原料采用I-1的合成方法2合成。纯化后得到化合物I-2。LC-MS m/z=510.2[M+1]+
实施例3
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺I-3
Figure PCTCN2017107638-appb-000071
化合物I-3的合成以中间体A-1和B-3为原料采用I-1的合成方法2合成。纯化后得到化合物I-3。LC-MS m/z=560.2[M+1]+
化合物I-3的合成可以用合成方法3合成
Figure PCTCN2017107638-appb-000072
4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧基羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-3
Figure PCTCN2017107638-appb-000073
4-(8-(2,4-二甲氧基苄基氨基)-3-溴咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯M(721mg,1.45mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂)硼烷-2-基)环己-3-烯基氨基甲酸叔丁酯L-3(562mg,1.74mmol),Pd[PPh3]4(168mg,0.145mmol)和Cs2CO3(706mg,2.17mmol)在二恶烷(30mL)中90℃下搅拌2.5小时。浓缩并溶于DCM(100mL)中,有机相用水洗涤,分离并干燥,浓缩并用乙酸乙酯/石油(1:2)过柱纯化,得到产物5-3(731mg,82%)。
1H NMR(400MHz,CDCl3):δ1.70-1.80(m,1H);2.00-2.20(m,2H);2.60-2.90(m,3H);3.54(s,3H);3.80(s,3H);3.97(s,3H);4.60(m,2H),5.61(m,1H);6.21(m,1H);6.39(m,1H);7.20(m,1H);7.43(m,1H);7.68-7.70(d,J=8.4Hz,2H);8.07-8.09(d,J=8.4Hz,2H)。LC-MS m/z=614.3[M+1]+
4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧基羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酰胺6-3
Figure PCTCN2017107638-appb-000074
向4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧基羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-3(456mg,0.742mmol)的THF:MeOH(10mL:2mL)的混合溶液中加入LiOH(125mg,2.97mmol),将混合物加热至60℃搅拌12小时。浓缩并加入水(20mL),用1M HCl水溶液将pH调节至4。过滤并干燥,得到所需4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸(422mg,95%)。
1H NMR(400MHz,DMSO):δ1.60(m,1H);1.90(m,1H);2.10-2.20(m,1H);2.60-2.80(m,3H);3.58(s,3H);3.72(s,3H);4.48(d,J=5.2Hz,2H);5.79(m,1H);6.23(m,1H);6.41(m,1H),6.49(m,1H);6.92(m,1H);7.11(m,2H);7.72(m,3H);8.01(m,2H);13.0(b,1H)。LC-MS m/z=600.3[M+1]+
向4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸(422mg,0.704mmol)的DCM(30mL)溶液中加入Et3N(86mg,0.845mmol)和氯代甲酸异丁酯(115mg,0.845mmol),然后在0℃下搅拌20分钟,TLC显示原料消失,并滴加氨水(5mL),再搅拌10分钟,倒入水中,将DCM(30mL)将混合物过滤,分离有机相,干燥,浓缩,用乙酸乙酯/石油醚(1:1)
Figure PCTCN2017107638-appb-000075
乙酸乙酯过柱纯化,得到产物6-3(350mg,83%)。
1H NMR(400MHz,DMSO):δ1.60(m,1H);1.90(m,1H);2.20(m,1H);2.60-2.80(m,1H);3.56(s,3H);3.57(m,1H);3.71(s,3H),4.46(d,J=5.6Hz,2H);5.77(m,1H);6.23(m,1H);6.41(m,1H);6.48(m,1H);6.92(m,1H);7.10(m,2H);7.43(m,1H);7.62(m,2H);7.71(m,1H);7.97(m,2H);8.07(m,1H);11.95(s,1H)。LC-MS m/z=599.3[M+1]+
4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺7-3
Figure PCTCN2017107638-appb-000076
将4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧基羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酰胺6-3(340mg,0.568mmol),2-氯(4-三氟甲基)吡啶(123mg,0.681mmol),Pd 2(dba)3(52mg,0.06mmol),Xantphos(65mg,0.11mmol)和Cs2CO3(277mg,0.852mmol)在甲苯(20mL)的混合物加热至120℃4小时,反应结束后浓缩并加入水,用乙酸乙酯萃取(3×20mL),分离有机相,干燥并浓缩,用乙酸乙酯/石油(2:3)
Figure PCTCN2017107638-appb-000077
乙酸乙酯过柱纯化,得到产物7-3(252mg,60%)。
1H NMR(400MHz,DMSO):δ1.60(m,1H);1.90(m,1H);2.20(m,1H);2.60-2.80(m,3H);3.61(s,3H);3.70(s,3H);4.49(d,J=5.6Hz,2H),5.80(m,1H);6.24(m,1H);6.43(m,1H);6.50(m,1H);6.85(m,1H);7.14(m,2H);7.55(m,1H);7.72(m,3H);8.17(m,2H);8.58(m,1H);8.70(m,1H);11.39(m,1H);11.90(m,1H)。LC-MS m/z=744.3[M+1]+
4-(8-氨基-3-(4-氨基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺8-3
Figure PCTCN2017107638-appb-000078
将4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧羰基氨基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺7-3(252mg,0.339mmol)在DCM/TFA(10mL:10mL)加热至60℃18小时。浓缩并加入DCM(2×20mL),再浓缩。将剩余物溶于DCM(30mL)中,加入10mL HCl的二恶烷溶液,蒸发,加入DCM(2×20mL)。 加入浓缩的异丙醚(30mL),将悬浮液搅拌2小时,过滤并用异丙醚(2×10mL)洗涤,得到所需产物8-3的盐酸盐110mg。
1H NMR(400MHz,DMSO):δ1.85(m,1H);2.10(m,1H);2.30-2.80(m,4H);6.36(m,1H);7.15(m,1H);7.57(m,1H);7.81(m,2H),7.94(m,1H);8.23(m,3H);8.56(m,1H);8.71(m,1H)。LC-MS m/z=494.2[M+1]+
4-(8-氨基-3-(4-丁-2-炔酰胺环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺I-3
Figure PCTCN2017107638-appb-000079
向4-(8-氨基-3-(4-氨基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺盐酸盐8-3(50mg)和DIEA(86mg,0.67mmol)的DMF(5mL)溶液中加入丁-2-炔酸(5.6mg,0.067mmol)和HATU(25mg,0.067mmol)在DMF(1mL)中的溶液中。将反应物搅拌20分钟。倒入水(20mL)中并用乙酸乙酯(2×10mL)萃取,有机相用饱和NaCl水溶液洗涤,用Na 2SO4干燥,过滤并蒸发。用MeOH/DCM(1:20)过柱纯化,得到产物(30mg,53%)。
1H NMR(400MHz,DMSO):δ1.65(m,1H);1.80-2.00(m,4H);2.20(m,1H);2.60-2.80(m,3H);3.95(m,1H);6.18(m,2H);6.27(m,1H),7.10(m,1H);7.55(m,1H);7.79(m,3H);8.17(m,2H);8.57-8.61(m,2H);8.69(m,1H);11.35(s,1H)。LC-MS m/z=560.2[M+1]+
实施例4
化合物I-4的合成采用以下合成路线1
Figure PCTCN2017107638-appb-000080
3-(4-氨基环己-1-烯基)-1-碘咪唑并[1,5-a]吡嗪-8-胺24-4
Figure PCTCN2017107638-appb-000081
向化合物A-1(200mg,0.41mmol)在DCM(10mL)中的溶液中加入6滴HBr的AcOH(33%)溶液。将悬浮液在室温下搅拌2小时。将其倒入冰水(20mL)中并将pH调节至9,将混合物用乙酸乙酯(4×15mL)萃取。分离有机相,用无水Na2SO4干燥,过滤并浓缩。将其通过柱色谱法用DCM/MeOH=10:1纯化,得到产物24-4(87mg,60%)。
N-(4-(8-氨基-1-碘咪唑并[1,5-a]吡嗪-3-基)环己-3-烯基)丁-2-炔酰胺25-4
Figure PCTCN2017107638-appb-000082
向化合物24-4(87mg,0.246mmol)的DMF(1mL)中的溶液中加入HATU(93mg,0.246mmol)和丁-2-炔酸(21mg,0.246mmol)在DMF中的混合物,然后加入DIEA(63mg,0.492mmol)。将反应在室温下搅拌30分钟。加入水(20ml),用乙酸乙酯(2×10ml)萃取。有机相用饱和NaCl水溶液(2×10ml)洗涤。分离有机相并干燥。将其蒸发并通过TLC柱纯化,得到产物25-4(65mg,62%)。LC-MS m/z=422.0[M+1]+
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-甲基吡啶 -2-基)苯甲酰胺I-4
Figure PCTCN2017107638-appb-000083
将化合物25-4(30mg,0.07mmol),4-((4-甲基吡啶-2-基)氨基甲酰基)苯基硼酸B-4(22mg,0.086mmol),Pd[PPh3]4(8mg,0.007mmol)和Cs2CO3(28mg,0.086mmol)在DME:H2O(2.5ml:0.5ml)的混合溶剂中的混合物加热至80℃过夜。将其浓缩并通过使用EA-MeOH/DCM(1:40)的柱色谱法纯化得到化合物I-4(19mg,50%)。
LC-MS m/z=506.2[M+1]+
实施例5
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-乙基吡啶-2-基)苯甲酰胺I-5
Figure PCTCN2017107638-appb-000084
化合物I-5的合成以中间体A-1和B-5为原料采用I-1的合成方法2合成。纯化后得到化合物I-5。LC-MS m/z=520.2[M+1]+
实施例6
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-氰基吡啶-2-基)苯甲酰胺I-6
Figure PCTCN2017107638-appb-000085
化合物I-6的合成以中间体25-4和B-6为原料采用I-4的合成方法1合成。纯化后得到化合物I-6。LC-MS m/z=517.2[M+1]+
实施例7
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-2-氟-N-(吡啶-2-基)苯甲酰胺I-7
Figure PCTCN2017107638-appb-000086
将化合物25-4(30mg,0.07mmol)和(吡啶-2-基氨基甲酰基)苯基硼酸B-7(23mg,0.086mmol),Pd[PPh3]4(8mg,0.007mmol)和Cs2CO3(28mg,0.086mmol)在DME:H2O(2.5ml:0.5ml)的混合溶剂中的混合物加热至80℃过夜。将其浓缩并通过使用EA-MeOH/DCM(1:40)的柱色谱法纯化。得到化合物I-7(11mg,28%)。LC-MS m/z=510.3[M+1]+
实施例8
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-甲氧基吡啶-2-基)苯甲酰胺I-8
Figure PCTCN2017107638-appb-000087
化合物I-8的合成以中间体A-1和B-8为原料采用I-1的合成方法2合成。纯化后得到化合物I-8。LC-MS m/z=522.1[M+1]+
实施例9
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(哒嗪-3-基)苯甲酰胺I-9
Figure PCTCN2017107638-appb-000088
化合物I-9的合成以中间体A-1和B-9为原料采用I-1的合成方法2合成。纯化后得到化合物I-9。LC-MS m/z=493.2[M+1]+
实施例10
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(噻唑-2-基)苯甲酰胺I-10
Figure PCTCN2017107638-appb-000089
化合物I-10的合成以中间体A-1和B-10为原料采用I-1的合成方法2合成。纯化后得到化合物I-10。LC-MS m/z=498.2[M+1]+
实施例11
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-3-氟-N-(吡啶-2-基)苯甲酰胺I-11
Figure PCTCN2017107638-appb-000090
化合物I-11的合成以中间体A-1和B-11为原料采用I-1的合成方法2合成。纯化后得到化合物I-11。LC-MS m/z=510.1[M+1]+
实施例12
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-3-氟-N-(4-(三氟甲基)吡啶吡啶-2-基)苯甲酰胺I-12
Figure PCTCN2017107638-appb-000091
化合物I-12的合成以中间体A-1和B-12为原料采用I-1的合成方法2合成。纯化后得到化合物I-12。LC-MS m/z=578.1[M+1]+
实施例13
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-3-甲氧基-N-(4-(三氟甲基)吡啶吡啶-2-基)苯甲酰胺I-13
Figure PCTCN2017107638-appb-000092
化合物I-13的合成以中间体A-1和B-13为原料采用I-1的合成方法2合成。纯化后得到化合物I-13。LC-MS m/z=590.1[M+1]+
实施例14
4-(8-氨基-3-(4-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-3-甲基-N-(4-(三氟甲基)吡啶吡啶-2-基)苯甲酰胺I-14
Figure PCTCN2017107638-appb-000093
化合物I-14的合成以中间体A-1和B-14为原料采用I-1的合成方法2合成。纯化后得到化合物I-14。LC-MS m/z=574.0[M+1]+
实施例15
4-(3-(4-丙烯酰胺基环己-1-烯基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺I-15
Figure PCTCN2017107638-appb-000094
向4-(3-(7-氮杂-双环[2.2.1]庚-1-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺氢溴酸盐25-1(30mg,0.045mmol)的DMF(1mL)溶液中加入HATU(8.5mg,0.023mmol)和丙烯酸(1.9mg,0.023mmol)在DMF中的混合物,然后加入DIEA(29mg,0.225mmol),将反应在室温下搅拌10分钟。加入水(20ml),用乙酸乙酯(2×10ml)萃取。有机相用饱和NaCl水溶液(2×10ml)洗涤。分离有机相并干燥。将其蒸发并通过TLC柱纯化,得到化合物I-15。LC-MS m/z=480.1[M+1]+
实施例16
(E)-4-(8-氨基-3-(4-(4-甲氧基-2-烯酰胺基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶吡啶-2-基)苯甲酰胺I-16
Figure PCTCN2017107638-appb-000095
向化合物25-1(30mg,0.045mmol)的DMF(1mL)溶液中加入HATU(8.5mg,0.023mmol)和(E)-4-甲氧基丁-2-烯酸(1.9mg,0.023mmol)在DMF中的混合物,然后加入DIEA(29mg,0.225mmol),将反应在室温下搅拌10分钟。加入水(20ml),用乙酸乙酯(2×10ml)萃取。有机相用饱和NaCl水溶液(2×10ml)洗涤。分离有机相并干燥。将其蒸发并通过TLC柱纯化,得到化合物I-16。LC-MS m/z=524.1[M+1]+
实施例17
4-(3-(4-乙酰胺基环己-1-烯基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺I-17
Figure PCTCN2017107638-appb-000096
向化合物25-1(30mg,0.045mmol)的DMF(1mL)溶液中加入HATU(8.5mg,0.023mmol)和醋酸(1.9mg,0.023mmol)在DMF中的混合物,然后加入DIEA(29mg,0.225mmol),将反应在室温下搅拌10分钟。加入水(20ml),用乙酸乙酯(2×10ml)萃取。有机相用饱和NaCl水溶液(2×10ml)洗涤。分离有机相并干燥。将其蒸发并通过TLC柱纯化,得到化合物I-17。LC-MS m/z=468.2[M+1]+
实施例18
(E)-4-(8-氨基-3-(4-(4-(二甲基氨基)丁-2-烯酰胺基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1- 基)-N-(吡啶-2-基)苯甲酰胺I-18
Figure PCTCN2017107638-appb-000097
向化合物25-1(30mg,0.045mmol)的DMF(1mL)溶液中加入HATU(8.5mg,0.023mmol)和(E)-4-(二甲基氨基)丁-2-烯酸盐酸盐(1.9mg,0.023mmol)在DMF中的混合物,然后加入DIEA(29mg,0.225mmol),将反应在室温下搅拌10分钟。加入水(20ml),用乙酸乙酯(2×10ml)萃取。有机相用饱和NaCl水溶液(2×10ml)洗涤。分离有机相并干燥。将其蒸发并通过TLC柱纯化,得到化合物I-18。LC-MS m/z=537.3[M+1]+
实施例19及实施例20
甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(3-(叔丁氧基羰基)环戊-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-19/甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧基羰基)环戊-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-20
Figure PCTCN2017107638-appb-000098
4-(8-(2,4-二甲氧基苄基氨基)-3-溴咪唑并[1,5-a]吡咯甲酯的混合物吖嗪-1-基)苯甲酸甲酯M(802mg,1.61mmol),L-1a和L-1b混合物(598mg,1.93mmol),Pd[PPh 3]4(186mg,0.161mmol)和Cs 2CO 3(1.05g,3.22mmol)在二恶烷(30mL)中的混合物加热至100℃5小时。浓缩并溶于DCM(100mL)中,浓缩并用乙酸乙酯/石油=1:4的柱色谱纯化,得到甲 基4-(8-(2,4-二甲氧基苄基氨基)-3-(3-(叔丁氧基羰基)环戊-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-19(479mg,51%)和甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(4-(叔丁氧基羰基)环戊-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-20(395mg,39%)。5-19:LC-MS m/z=600.2[M+1]+。5-20:LC-MS m/z=600.2[M+1]+
4-(8-氨基-3-(3-丁-2-炔酰胺基环戊-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺I-19
Figure PCTCN2017107638-appb-000099
化合物I-19的合成以中间体5-19为原料采用合成方法3合成。纯化后得到19mg化合物I-19。LC-MS m/z=546.2[M+1]+。
4-(8-氨基-3-(4-丁-2-炔酰胺基环戊-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺I-20
Figure PCTCN2017107638-appb-000100
化合物I-20的合成以中间体5-20为原料采用合成方法3合成。纯化后得到22mg化合物I-20。LC-MS m/z=546.2[M+1]+。
实施例21及实施例22
甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(3-(叔丁氧基羰基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-21/甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(5-(叔丁氧基羰基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-22
Figure PCTCN2017107638-appb-000101
4-(8-(2,4-二甲氧基苄基氨基)-3-溴咪唑并[1,5-a]吡咯甲酯的混合物(1g,2mmol),L-2a和L-2b(1.8g,5.57mmol),Pd[PPh3]4(230mg,0.2mmol)和Cs2CO3(1.3g,4mmol)的二恶烷(30mL)溶液加热至100℃5小时。浓缩并溶解于DCM(100mL)中,浓缩并用乙酸乙酯/石油=柱色谱纯化1:4给出产物甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(3-(叔丁氧基羰基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-21(290mg,24%)和甲基4-(8-(2,4-二甲氧基苄基氨基)-3-(5-(叔丁氧基羰基)环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)苯甲酸甲酯5-22(228mg,18%)。5-21:LC-MS m/z=614.2[M+1]+。5-22:LC-MS m/z=614.2[M+1]+
4-(8-氨基-3-(3-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺I-21
Figure PCTCN2017107638-appb-000102
化合物I-21的合成以中间体5-21为原料采用合成方法3合成。纯化后得到18mg化合物I-21。LC-MS m/z=560.2[M+1]+。
4-(8-氨基-3-(5-丁-2-炔酰胺基环己-1-烯基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺I-22
Figure PCTCN2017107638-appb-000103
化合物I-22的合成以中间体5-22为原料采用合成方法3合成。纯化后得到22mg
化合物I-22。LC-MS m/z=560.2[M+1]+。
1.对BTK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 Na3VO4,2mM 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与参照化合物Ibrutinib相似的对BTK酶活性的抑制能力,优于参照化合物ACP-196的BTK酶活性抑制能力。
表1.BTK酶活性的抑制结果:A≤10nM,10nM<B≤100nM
Figure PCTCN2017107638-appb-000104
Figure PCTCN2017107638-appb-000105
Figure PCTCN2017107638-appb-000106
Figure PCTCN2017107638-appb-000107
Figure PCTCN2017107638-appb-000108
2.体外肿瘤细胞增殖抑制实验
(1)实验方法
·收获处于对数生长期的细胞(Pfeiffer细胞源于美国ATCC,DOHH2细胞源于徳国DSMZ)并使用血小板计数器计数细胞。
·用培养基调节细胞浓度。
·添加90μl细胞悬浮液至96孔板中。(测试中使用的细胞密度应根据数据库中生长曲线来确定。或者根据第一次测试的结果来确定最适细胞密度)
·将96孔板中的细胞置于37℃,5%CO2条件下培养过夜。
·第二天,将待测化合物和参照药物用DMSO或PBS溶解,作为母液。
·向96孔板每个孔分别加入10μl药物溶液(10×),每个细胞浓度设置三个复孔。(培养基中DMSO终浓度为0.5%[v/v])
·将96孔板继续培养72小时。
·在室温下平衡细胞板大约30分钟。
·每个孔中添加等量的
Figure PCTCN2017107638-appb-000109
反应物至等量的细胞培养基。(例如,96孔板中添加100μl反应物至100μl含细胞培养基中)
·在定轨摇床上混合2分钟来诱导细胞胞溶作用。
·细胞板在室温下放置10分钟来稳定冷光信号。
·使用多功能酶标仪读取冷光值。
·数据分析:使用GraphPad Prism计算软件来测定IC50。使用带有S型剂量反应的非线性回归模型拟合图形曲线。
(2)实验结果
对B细胞淋巴瘤细胞Pfeiffer及DOHH2的增殖抑制实验中,化合物I-3,I-14与Ibrutinib表现出很强的肿瘤细胞增殖抑制活性,而ACP-196的抑制活性则相对较低。
表2.部分化合物的肿瘤细胞增殖抑制活性结果:I≤100nM,100nM<II≤1μM,1μM<III≤10μM,10μM<IV≤100μM
Figure PCTCN2017107638-appb-000110
3.药代动力学研究
(1)实验方法
雌性CD-1小鼠,体重16-20克,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 10mL/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0.3mL,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。
(2)实验结果
实施例3制备的化合物I-3,参照化合物Ibrutinib和ACP-196在同样条件下灌胃给药(10mg/kg)。化合物I-3表现出良好的吸收,Cmax分别为2709ng/mL,165ng/mL和962ng/mL;而化合物I-3的血液暴露量(AUC0-inf=3197hr*ng/mL)更是远高于参照化合物Ibrutinib(AUC0-inf=228hr*ng/mL)和ACP-196(AUC0-inf=584hr*ng/mL)。
表3.小鼠(雌性)灌胃给药(10mg/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;AUC0-INF指0-inf时间-浓度曲线下面积)
Figure PCTCN2017107638-appb-000111
4.激酶选择性实验
(1)实验方法
对Ibrutinib有较强抑制的十种激酶的体外抑制活性测定(IC50)
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 Na3VO4,2mM 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(最终激酶在反应中的浓度为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
本发明代表化合物I-3拥有比参照化合物Ibrutinib更高的选择性。
表4.激酶活性抑制(IC50)
Figure PCTCN2017107638-appb-000112
[1]Honigberg LA,et al.Proc Natl Acad Sci U S A.2010,107(29),13075-13080.
5.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0.011μM下降到1.9μM,而本发明的化合物I-3除了对BTK(WT)的HEK293细胞有很强的抑制外(0.024μM),对BTK(C481S)也保持较强的抑制(0.35μM)。
表5.BTK(WT)及BTK(C481S)转染的HEK293细胞pBTK抑制值(IC50)
Figure PCTCN2017107638-appb-000113

Claims (23)

  1. 式I的化合物
    Figure PCTCN2017107638-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选自CH或N;B环为取代或非取代的芳环或芳杂环;C环为取代或非取代的含至少一个N的芳杂环;D环为取代或非取代的环烯烃;NR1R2为D环上任意位置的C上的取代基;
    R1选自R3或以下结构之一,
    Figure PCTCN2017107638-appb-100002
    R3选自氢、取代或非取代的C1-6烷基、取代或非取代的C1-6炔基、取代或非取代的C1-6烯基、取代或非取代的C6-10芳基、取代或非取代的C1-9杂芳基、取代或非取代的C3-7环烷基或取代或非取代的C2-7杂环烷氨基;
    R4选自氢、取代或非取代的C1-6烷基、取代或非取代的C6-10芳基、取代或非取代的C1-9杂芳基、取代或非取代的C3-7环烷基、取代或非取代的C3-7杂环烷基;
    R3、R4以及与之相连的N形成或不形成C3-7杂环氨基或C3-9杂芳环氨基;
    R2选自H、取代或非取代的C1-3烷基、取代或非取代的C3-7环烷基、取代或非取代的C2-7杂环烷基、取代或非取代的C6-10芳基或取代或非取代的C1-9杂芳基;
    R1、R2以及与之相连的N形成或不形成取代或非取代的C2-7杂环。
  2. 根据权利要求1所述的化合物,其特征在于,所述R3中的取代的C1-6烷基、取代的C1-6炔基、取代的C1-6烯基、取代的C6-10芳基、取代的C1-9杂芳基、取代的C3-7环烷基或取代的C2-7杂环烷氨基中的取代基选自卤素、羟基、氰基、氨基、取代或非取代的C1-4烯基、取代或非取代的C3-7环烷基、取代或非取代的C3-7环烷氧基、取代或非取代的C1-4烷基氨基、二[取代或非取代的C1-4烷基]氨基、取代或非取代的C3-7环烷氨基、取代或非取代的C3-7杂环烷氨基、取代或非取代的C1-3烷氧基、取代或非取代的C3-7环烷氧基、取代或非取代的C6-10芳基或取代或非取代的C3-7杂环烷基中的一个或多个。
  3. 根据权利要求1所述的化合物,其特征在于,所述R4中的取代的C1-6烷基、取代的C6-10芳基、取代的C1-9杂芳基、取代的C3-7环烷基,取代的C3-7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C1-4烯基、取代或非取代的C3-7环烷基、取代或非取代的C3-7环烷氧基、取代或非取代的C1-4烷基氨基、二[取代或非取代的C1-4烷基]氨基、取代或非取代的C3-7环烷氨基、取代或非取代的C3-7杂环烷氨基、取代或非取代的C1-3烷氧基、取代或非取代的C3-7环烷氧基、取代或非取代的C6-10芳基或取代或非取代的C3-7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B环为单环或多环的取代或非取代的芳环或芳杂环;所述芳杂环含一个或多个N原子。
  5. 根据权利要求4所述的化合物,其特征在于,所述B环结构如下式:
    Figure PCTCN2017107638-appb-100003
    其中,B1选自N或CR5;B2选自N或CR6;B3选自N或CR7;B4选自N或CR8;其中,R5、R6、R7和R8各自独立的选自H、卤素、羟基、取代或非取代C1-3烷基或取代或非取代C1-3烷氧基。
  6. 根据权利要求5所述的化合物,其特征在于,所述R5和R6与它们所连接的碳原子一起形成C6-10芳基或C1-9杂芳基。
  7. 根据权利要求1所述的化合物,其特征在于,C环为单环或多环的取代或非取代的含至少一个N的芳杂环;所述杂原子选自N、O或S。
  8. 根据权利要求7所述的化合物,其特征在于,所述C环结构如下式:
    Figure PCTCN2017107638-appb-100004
    其中,E1选自CH、N、O或S;E2选自C-R9、N、O或S;E3选自C-R10、N、O或S;E4选自CH、N、O或键;
    其中R9选自H、卤素、氰基、羟基、氨基、取代或非取代的C1-4烷基氨基、[取代或非取代的C1-4烷基]2氨基、取代或非取代的C3-6环烷氨基、取代或非取代的C1-4烷基、取代或非取代的C1-3烷氧基、取代或非取代的C3-6环烷基、取代或非取代的C3-6环烷氧基,C6-10芳基、C1-9杂芳基或C2-7杂环烷基;所述取代选自卤素中的一个或多个;
    R10选自H、卤素、羟基、氰基、取代或非取代的C1-3烷基或取代或非取代的C1-3烷氧基。
  9. 根据权利要求8所述的化合物,其特征在于,R9和R10一起形成取代或非取代的C3-7环烯基或取代或非取代的C2-6杂环烯基;所述取代的取代基团选自C1-3烷基、C1-3烷氧基或卤素中的一个或多个。
  10. 根据权利要求8所述的化合物,其特征在于,所述C环选自以下结构之一:
    Figure PCTCN2017107638-appb-100005
  11. 根据权利要求1所述的化合物,其特征在于,所述D环选自以下结构之一:
    Figure PCTCN2017107638-appb-100006
  12. 根据权利要求1所述的化合物,其特征在于,所述R1选自以下结构之一:
    Figure PCTCN2017107638-appb-100007
  13. 根据权利要求1-12所述的化合物,其特征在于,所述化合物如下图中任一结构所示:
    Figure PCTCN2017107638-appb-100008
  14. 权利要求1~13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。
  15. 根据权利要求14所述的制备药物中的用途,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。
  16. 根据权利要求13或14所述的制备药物中的用途,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。
  17. 根据权利要求14所述的制备药物中的用途,其特征在于,所述异种免疫性疾病为炎性疾病或哮喘。
  18. 根据权利要求14所述的制备药物中的用途,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。
  19. 一种药物组合物,包含一种以上如权利要求1~13中任一项所述的化合物。
  20. 一种药物制剂,包含治疗有效量的如权利要求1~13中任一项所述的化合物在药学上可接受的赋形剂。
  21. 权利要求1~13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:(1a)化合物A用氢溴酸处理脱去苄氧羰基得到化合物1;(2a)化合物1和酸或酰氯偶合生成化合物2;(3a)化合物2和硼酸B进行Suzuki偶合得到权利要求1中所述的化合物I即可;
    Figure PCTCN2017107638-appb-100009
    其中R1,R2,B环,C环和D环如权利要求1~13中所述。
  22. 权利要求1~13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:(1b)化合物A和硼酸B进行Suzuki偶合得到化合物3;(2b)化合物3用氢溴酸处理脱去苄氧羰基得到化合物4;(3b)化合物4和酸或酰氯偶合生成权利要求1中所述的化合物I即可;
    Figure PCTCN2017107638-appb-100010
    其中R1,R2,B环,C环和D环如权利要求1~13中所述。
  23. 权利要求1~13中任一项所述的化合物的制备方法,其特征在于,当B环为苯环,C环为取代或非取代的吡啶环时,包括以下步骤:(1c)化合物M和硼酸酯L进行Suzuki偶合得到化合物5;(2c)化合物5水解氨化得到化合物6;(3c)化合物6与卤代吡啶在钯催化下生成化合物7;(4c)化合物7用TFA脱保护得到化合物8;(5c)化合物8和酸或酰氯偶合生成权利要求1中所述的化合物I即可;
    Figure PCTCN2017107638-appb-100011
    其中R1,R2,D环如权利要求1~13中所述,R为氢,氰基,卤素,三氟甲基,三氟甲氧基,C1-3烷基,C1-3烷氧基,C3-7环烷基,C3-7环烷氧基,C2-7杂环烷基,C2-7杂环烷氧基。
PCT/CN2017/107638 2017-09-20 2017-10-25 布鲁顿酪氨酸激酶抑制剂 WO2019056466A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710849859.3 2017-09-20
CN201710849859.3A CN107602564B (zh) 2017-09-20 2017-09-20 布鲁顿酪氨酸激酶抑制剂

Publications (1)

Publication Number Publication Date
WO2019056466A1 true WO2019056466A1 (zh) 2019-03-28

Family

ID=61061204

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/107638 WO2019056466A1 (zh) 2017-09-20 2017-10-25 布鲁顿酪氨酸激酶抑制剂

Country Status (2)

Country Link
CN (1) CN107602564B (zh)
WO (1) WO2019056466A1 (zh)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110964016B (zh) 2018-09-29 2021-05-28 南京药捷安康生物科技有限公司 氨基降茨烷衍生物及其制备方法与应用
CN111377928A (zh) * 2018-12-28 2020-07-07 南京艾德凯腾生物医药有限责任公司 一种制备Acalabrutinib的方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103917545A (zh) * 2011-07-19 2014-07-09 默沙东公司 Btk抑制剂
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

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL295053A (en) * 2007-03-28 2022-09-01 Pharmacyclics Llc Broton tyrosine kinase inhibitors
WO2013010868A1 (en) * 2011-07-19 2013-01-24 Msd Oss B.V. 4 - imidazopyridazin- 1 -yl-benzamides and 4 - imidazotriazin- 1 - yl - benzamides as btk- inhibitors
WO2014113942A1 (en) * 2013-01-23 2014-07-31 Merck Sharp & Dohme Corp. Btk inhibitors
WO2016192074A1 (en) * 2015-06-04 2016-12-08 Merck Sharp & Dohme Corp. Btk inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103917545A (zh) * 2011-07-19 2014-07-09 默沙东公司 Btk抑制剂
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

Also Published As

Publication number Publication date
CN107602564B (zh) 2019-08-30
CN107602564A (zh) 2018-01-19

Similar Documents

Publication Publication Date Title
EP3445357B1 (en) Bifunctional molecules for degradation of egfr and methods of use
WO2018113085A1 (zh) 布鲁顿酪氨酸激酶抑制剂
EP3317273B1 (en) Inhibitors of egfr and methods of use thereof
TWI726030B (zh) 一種btk激酶抑制劑的結晶形式及其製備方法
CN110678459B (zh) 用于治疗癌症的2-杂芳基-3-氧代-2,3-二氢哒嗪-4-甲酰胺
TW201704237A (zh) 適用於治療與kit及pdfgr相關之病症的組合物
JP6409116B2 (ja) プロテインキナーゼ阻害剤としてのアミノピリダジノン化合物
WO2018196677A1 (zh) 氟代烯丙胺衍生物及其用途
TWI753089B (zh) Jak1選擇性抑制劑
EP2964223A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
TW201341385A (zh) 咪唑[1,2-b]噠嗪基化合物、包含其之組成物及其使用方法
WO2019000683A1 (zh) Rho相关蛋白激酶抑制剂、包含其的药物组合物及其制备方法和用途
CN112135826B (zh) 氨基降茨烷衍生物及其制备方法与应用
WO2021078021A1 (zh) 一种小分子化合物
WO2018130124A1 (zh) 作为选择性雌激素受体下调剂的三环类化合物及其应用
CN113365994A (zh) 哒嗪基噻唑甲酰胺类化合物
US20230295119A1 (en) Substituted pyridines for the treatment of inflammatory diseases
CN114907341A (zh) 吡啶并嘧啶酮类衍生物及其制备方法和用途
KR20100015857A (ko) S1p 수용체 조절 활성을 갖는 프탈라진 및 이소퀴놀린 유도체
TW201546060A (zh) 肌旋蛋白相關(tropomyosin-related)之激酶抑制劑
WO2020182159A1 (zh) Jak激酶抑制剂及其制备方法和在医药领域的应用
WO2020156479A1 (zh) 环丙烯并苯并呋喃取代的氮杂芳基化合物、其中间体、制备方法及应用
WO2019056466A1 (zh) 布鲁顿酪氨酸激酶抑制剂
WO2022017494A1 (zh) 一种哒嗪类衍生物自由碱的晶型及其制备方法和应用
WO2020135210A1 (zh) 取代芳基化合物及其制备方法和用途

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: 17926031

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: 17926031

Country of ref document: EP

Kind code of ref document: A1