CN108570048B - Substituted heteroaryl compounds, compositions and uses thereof - Google Patents

Substituted heteroaryl compounds, compositions and uses thereof Download PDF

Info

Publication number
CN108570048B
CN108570048B CN201810159088.XA CN201810159088A CN108570048B CN 108570048 B CN108570048 B CN 108570048B CN 201810159088 A CN201810159088 A CN 201810159088A CN 108570048 B CN108570048 B CN 108570048B
Authority
CN
China
Prior art keywords
group
atoms
compound
cancer
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201810159088.XA
Other languages
Chinese (zh)
Other versions
CN108570048A (en
Inventor
习宁
李晓波
李敏雄
张涛
胡海洋
吴彦君
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangdong HEC Pharmaceutical
Original Assignee
Sunshine Lake Pharma Co Ltd
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 Sunshine Lake Pharma Co Ltd filed Critical Sunshine Lake Pharma Co Ltd
Publication of CN108570048A publication Critical patent/CN108570048A/en
Application granted granted Critical
Publication of CN108570048B publication Critical patent/CN108570048B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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

Abstract

The invention provides a substituted heteroaryl compound, a composition thereof and application thereof. The compound is a compound shown in a formula (I) or a stereoisomer, a tautomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug of the compound shown in the formula (I). The invention also provides pharmaceutical compositions comprising the compounds, which compounds and pharmaceutical compositions can modulate the activity of protein kinases, particularly Aurora kinases and JAK kinases, for the prevention, treatment and alleviation of diseases or disorders mediated by protein kinases, particularly JAK kinase activity.

Description

Substituted heteroaryl compounds, compositions and uses thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a novel substituted heteroaryl compound, pharmaceutically acceptable salts thereof, a pharmaceutical composition containing the compound, and application of the compound and the pharmaceutical composition in preparation of medicines for treating proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, transplant rejection, cancers or other diseases of mammals. More specifically, the compounds of the present invention can modulate the activity of the JAK kinase family (including JAK1, JAK2, JAK3 and TYK2), FLT3 kinase (also known as FLK-2) and Aurora kinases (including Aurora-A, Aurora-B and Aurora-C), and further modulate signal transduction inside and outside cells.
Background
The protein kinase family comprises a large class of structurally related enzymes that control various signal transduction processes within cells, and typically contain a similar 250-300 amino acid catalytic domain that catalyzes the phosphorylation of target protein substrates. Many diseases are reported to be associated with abnormal cellular responses triggered by protein kinase-mediated events. These diseases include benign and malignant proliferative disorders, disorders resulting from inappropriate activation of the immune system, allograft rejection, graft-versus-host disease, autoimmune disorders, inflammatory disorders, bone disorders, metabolic disorders, neurological and neurodegenerative disorders, cancer, cardiovascular disorders, allergy and asthma, alzheimer's disease and hormone-related disorders. Accordingly, there have been a great deal of effort in the field of medicinal chemistry to find protein kinase inhibitors that are effective as therapeutic agents.
Kinases can be classified into families by phosphorylated substrates (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.). Tyrosine phosphorylation is one of the central events that regulates various biological processes such as cell proliferation, migration, differentiation, and survival. Several families of receptor and non-receptor tyrosine kinases control these events: catalyzing the transfer of phosphate from ATP to tyrosine residues of specific cellular protein targets. Currently, motifs generally corresponding to the above-mentioned kinase families have been identified (Hanks et al, FASEB J.,1995,9, 576-596; Knighton et al., Science,1991,253, 407-414; Garcia-Bustos en al. EMBO J.,1994,13: 2352-2361). Examples of kinases in the protein kinase family include, but are not limited to, Aurora, Axl, abl, Akt, bcr-abl, Blk, Brk, Btk, c-Met, c-src, c-fms, CDKl, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK10, cRafl, CSF1R, CSK, EGFR, ErbB2, ErbB3, 4, Erk, Flt-3, Fak, fes, FGFRl, FGFR2, FGFR3, FGFR4, FGFR5, fgrps, fpks, fkn, hk, JAK, IGF-1R, INS-R, KDR, Lck, Lyn, MEK, p38, PKC fr, PIK, PKC 2, tiros, Tie, tre, R2, frzks, R70, and the like.
The Aurora kinase family is a highly related class of serine/threonine kinases that are key regulators of mitosis and are essential for the precise and equal separation (segtion) of genomic material from mother cell to daughter cell. Members of the Aurora kinase family include three classes of related kinases known as Aurora-A, Aurora-B and Aurora-C (also known as Aurora-1, Aurora-2, and Aurora-3). Despite the significant procedural homology, these kinases differ greatly in their localization and function from each other (Richard D. Carvajal, et al, Clin. cancer Res.,2006,12(23): 6869-.
Aurora-A ubiquitously expresses and regulates Cell cycle events that occur from late S phase through M phase, including centrosome maturation (Berdnik D, et al, Curr. Biol.,2002,12:640-647), mitotic entry (Hirota T, et al, Cell,2003,114: 585-598; Dutertre S, et al, J.cell Sci.,2004,117:2523-2531), centrosome separation (Marumoto T, et al, J.biol.Chem.,2003,278:51786-51795), bipolar spindle assembly (Eyers PA, et al, Curr Biol.,2003,13:691-7), chromosomal alignment on plates (umoto T, et al, J.biol.Chem.,2003,278: 786-95; KunitokN, Dev., 85691-7), mitotic alignment on plates (Marumoto T, et al J.Chem., 2003,278: 786-95; Kunitokn, Dev., 8535-5164, Cell division J. 2003,278, and Cell division. Aurora-a protein levels and kinase activity increased from G2 through the M phase, with peak activity in the prometaphase. Once activated, Aurora-a mediates its diverse functions by interacting with various substrates including the centrosomal protein (centrosomin), converting acidic coiled-coil protein, cdc25b, Eg5, and centromeric protein a.
Aurora-B is a chromosomal passenger protein critical for precise chromosomal segregation, cytokinesis (Hauf S, et al., j.cell biol.,2003,161: 281-94; Ditchfield C, et al., j.cell biol.,2003,161: 267-280; Giet R, et al., j.cell biol.,2001,152: 669-682; Goto H, et al., j.biol.chem.,2003,278:8526-8530), protein localization to centromeres and centromeres, correct microtubule-centromery attachment (Murata-Hori M, et al., curr.biol.,2002,12:894-899), and regulation of mitotic checkpoint. Aurora-B localizes first to the chromosome during the promoiety and then to the internal centromere region between sister chromatids during the promoiety and metaphase (zeitin SG, et al, j.cell biol.,2001,155: 1147-1157). Aurora-B is involved in establishing the biological orientation of chromosomes, where sister centromeres are linked to the opposite pole of a bipolar spindle via bi-oriented attachment. Errors in this process, manifested as either a partially oriented ligation (one centromere attached to a microtubule from a bipolar stage) or a co-oriented ligation (two sister centromeres attached to a microtubule from the same stage), if left uncorrected before the start of the later stages, lead to chromosomal instability and aneuploidy. The major role of Aurora-B at the mitotic point is to repair incorrect microtubule-centromere attachment (Hauf S, et al., J.cell biol.,2003,161: 281-. In the case of Aurora-B inactivation, the mitotic checkpoint is impaired, leading to an increased number of aneuploid cells, gene instability and tumorigenesis (Weaver BA, et al, Cancer Cell,2005,8: 7-12).
Aurora-a overexpression is an essential feature of Aurora-a-induced tumorigenesis. In cells with Aurora-A overexpression, mitosis is characterized by the presence of multiple centrosomes and multipolar spindles (Meraldi Pet al, EMBO J.,2002,21: 483-492). Despite the abnormal microtubule-centromere attachment, the cells abrogate the mitotic checkpoint and progress from mid to late stages, causing many chromosome segregation defects. These cells do not undergo cytokinesis and develop additional Cell cycle, polyploidy and progressive chromosomal instability (Anand S, et al, Cancer Cell,2003,3: 51-62).
Aurora overexpression is proved to be closely related to various malignant proliferative diseases, such as rectal cancer, breast cancer, lung cancer, pancreatic cancer, prostate cancer, bladder cancer, head and neck cancer, cervical cancer, ovarian cancer, liver cancer, gastric cancer and the like, and the interest in developing Aurora inhibitors for cancer treatment is stimulated. In normal cells, Aurora-A inhibition causes delayed but not blocked mitotic entry, defects in centrosome separation of unipolar mitotic spindles, and failure of cytokinesis (Marumoto T, et al., J.biol.chem.,2003,278: 51786-. Encouraging antitumor effects of Aurora-a inhibitors are shown in three human pancreatic Cancer cell lines (Panc-i, MIA PaCa-2dnsu.86.86), with growth inhibition in cell culture and nearly total elimination of tumorigenicity in mouse xenografts (Hata T, et al, Cancer res.,2005,65: 2899-.
Aurora-B inhibition causes abnormal centromere-microtubule attachment, failure to achieve chromosomal bio-orientation, cytokinesis failure (Goto H, et al., j.biol.chem.,2003,278: 8526-. Repeated cycles of abnormal mitosis, not including cytokinesis, cause massive polyploidy and ultimately apoptosis (Hauf S, et al, J.cell biol.,2003,161: 281-94; Ditchfield C, et al, J.cell biol.,2003,161: 267-80; Giet R, et al, J.cell biol., 2001; 152: 669-82; Murata-Hori M, Curr.biol.,2002,12: 894-.
Inhibition of Aurora-a or Aurora-B activity in tumor cells causes chromosomal alignment impairment, abrogation of mitotic checkpoints, polyploidy and subsequent cell death. These in vitro effects are better in transformed cells than in non-transformed or non-differentiated cells (Dicchfield C, et al, J.cell biol.,2003,161:267-280), so that the target Aurora can achieve in vivo selectivity for cancer. Although it is expected that it may have some toxicity to rapidly differentiating cells of the hematopoietic and gastrointestinal systems, activity and clinical tolerance in xenograft models still show reasonable therapeutic indices. Several Aurora kinase inhibitors have been developed to date on the premise of preclinical antitumor activity and tumor selectivity potential.
FLT3(Flt3, FMS-related tyrosine kinase 3), also known as FLK-2 (fetal liver kinase 2) and STK-I (human stem cell kinase 1), is a member of the receptor tyrosine kinase (RTK-III) family (Gtirewalt DL et al, Nat. Rev. cancer,2003,3: 650-. FLT3 is a transmembrane protein, composed of four domains, including an extracellular ligand-binding domain composed of five immunoglobulin-like structures, a Transmembrane (TM) domain, a Juxtamembrane (JM) domain, and a cytoplasmic C-terminal Tyrosine Kinase (TK) domain. (Agnes F, et al Gene, l994,145: 283-33288; Scheijen B, et al Oncogene,2002,21: 3314-3333).
The ligand for FLT3 was cloned in 1993 and was shown to be a type I transmembrane protein expressed in hematopoietic bone marrow microenvironment cells including bone marrow fibroblasts and other cells (Lyman SD, et al, Cell,1993,75, 1157-1167). Both membrane bound and soluble forms activate the tyrosine kinase activity of the receptor and stimulate progenitor cell growth in bone marrow and blood. Ligand-receptor binding induces receptor dimerization and activation of the kinase domain; it then autophosphorylates and catalyzes substrate protein phosphorylation of various signal transduction pathways, such as signal transducers and activators of transcription 5 (STAT5), RAS/mitogen-activated protein kinases (RAS/MAPK), phosphoinositide 3-kinase (PI3K), Src homologous and collagen genes (SHC), SH 2-containing myo-inositol 5-phosphatase (SHIP) and cytoplasmic tyrosine phosphatases with 2 Src-homology 2(SH2) domains (SHP2), which play an important role in cell proliferation, differentiation and survival (Dosil M., et al, 1993,13:6572-6585.Zhang S, biochem. Biophys. Res. Commun. l., l. 999,254: 440. Across 445). In addition to hematopoietic cells, the FLT3 gene is also expressed in the placenta, gonads and brain (Maroc N, et al., Oncogene,1993,8: 909-.
FLT3 is also closely related to hematopoietic disorders that precede malignant proliferative disorders, such as thrombocythemia, polycythemia vera, Myelofibrosis (MF), chronic congenital myelofibrosis (IMF), Polycythemia (PV), precancerous myelodysplastic syndrome, hematological malignancies including, but not limited to, leukemia, (non-hodgkin's lymphoma), hodgkin's disease (also known as hodgkin's lymphoma), and myelomas, e.g., Acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), Acute Promyelocytic Leukemia (APL), Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Chronic Neutrophilic Leukemia (CNL). FLT3 was overexpressed at various levels in 70-100% of Acute Myeloid Leukemia (AML) and in a high percentage of Acute Lymphoblastic Leukemia (ALL) (GriffinJD, et al, Haematol J.,2004,5: 188-. In the blast crisis, it is also overexpressed in smaller subtypes of Chronic Myelogenous Leukemia (CML). Studies have shown that B lineage leukemia cells ALL co-express FL with AML frequently, resulting in an autocrine or paracrine signaling cycle of constitutive activation of FLT3 (Zheng R, et al. blood.,2004,103: 267-274). In addition, FLT3 ligand was expressed at high levels in the cell serum of patients with langerhans cell histocytosis and systemic lupus erythematosus, further showing that FLT3 is closely associated with dendritic cell signaling dysregulation in autoimmune diseases.
There is increasing evidence that many types of leukemia and myeloproliferative syndromes have mutations in tyrosine kinases. FLT3 mutation is one of the most frequent somatic alterations in AML, occurring in approximately 1/3 patients. Two types of activating mutations for FLT3 are described in leukemia patients. These include a series of Internal Tandem Duplications (ITDs) occurring in the self-inhibitory proximal membrane domain (Nakao M, et al., Leukemia,1996,10: 1911-. Internal tandem replication (ITD) mutations in the JM domain contribute to about 17-34% of activating mutations of FLT3 in AML. FLT3-ITD was also detected at low frequency in myelodysplastic syndrome (MDS) (Yokota S., et al, Leukemia, l997,11: 1605-. ITDs are always in-frame and are limited to the JM field. However, their length and position vary from patient to patient. These repeat procedures can be used to disrupt the self-inhibitory activity of the JM domain, resulting in constitutive activation of FLT 3. Both the FLT3-ITD and FLT3-Asp835 mutations are involved in FLT3 autophosphorylation and in the phosphorylation of downstream targets (Mizuki M, et al, Blood,2000,96: 3907-.
Currently, the ongoing research for FLT3 inhibitors has reached clinical trials as monotherapy in some or all relapsed or refractory AML patients with FLT3 mutations. Collectively, these data indicate that FLT3 is an attractive therapeutic target for the development of kinase inhibitors for AML and other related diseases.
Janus kinases (JAKs) are a family of intracellular, non-receptor tyrosine kinases that transduce cytokine-mediated signals through the JAK-STAT pathway. The JAK family plays an important role in cytokine-dependent regulation of proliferation and cellular functions involved in immune responses. Cytokines bind to their receptors and cause receptor dimerization, which can promote the cross-phosphorylation of JAKs and also the phosphorylation of specific tyrosine motifs within cytokine receptors. STATs that recognize these phosphorylation motifs are aggregated on receptors and then activated during JAK-dependent tyrosine phosphorylation. Upon activation, STATs dissociate from receptors, dimerize, and translocate to the nucleus, bind to specific DNA sites, and alter transcription.
Currently, there are four known mammalian JAK family members: JAK1(Janus kinase-1), JAK2(Janus kinase-2), JAK3(Janus kinase, leukocytes; JAKL; L-JAK and Janus kinase-3) and TYK2 (protein tyrosine kinase 2). JAK1, JAK2, and TYK2 are ubiquitously expressed, whereas JAK3 is reported to be preferentially expressed in Natural Killer (NK) cells, but not in other T cells ("Biology and design of the JAK/STAT signaling pathways," Growth Factors, April 2012; 30(2): 88).
JAK1 is essential for the signaling of certain type I and type II cytokines. It interacts with the gamma common chain (yc) of the type I cytokine receptor, inducing signaling in the IL-2 receptor family, the IL-4 receptor family, and the gp130 receptor family. It is also important for signaling of type I (IFN-. alpha./beta.) and type II (IFN-. gamma.) interferons, as well as for signaling by members of the IL-10 family of type II cytokine receptors. Genetic and biological studies have shown that JAK1 is functionally and physiologically associated with type I interferons (e.g., IFNalpha), type II interferons (e.g., IFNgamma), IL-2 and IL-6 cytokine receptor complexes. Furthermore, characterization of tissues derived from JAK1 knockout mice demonstrates a key role for this kinase in the IFN, IL-IO, IL-2/IL-4 and IL-6 pathways.
JAK1 expression in cancer cells can cause atrophy of individual cells, potentially allowing them to escape the tumor and metastasize to other parts of the body. The elevated levels of cytokines that transduce signals through JAK1 are implicated in a number of immune and inflammatory diseases. JAK1 or JAK family kinase inhibitors may be useful in the modulation or treatment of these diseases (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, Nat. Rev. mol. cell biol.,3: 651-662). Human monoclonal antibodies targeting the IL-6 pathway (Tocilizumab) were approved by the european commission for the treatment of moderate to severe rheumatoid arthritis (Scheinecker et al, 2009, nat. rev. drug discov.,8: 273-.
JAK2 is involved in signaling by members of the type II cytokine receptor family (e.g., interferon receptors), the GM-CSF receptor family, and the gp130 receptor family. JAK2 signals are activated downstream of the prolactin receptor. Studies have shown that an acquired activation of the JAK2 mutation (JAK2V617F) is prevalent in myeloproliferative diseases such as polycythemia vera, essential thrombocythemia, and idiopathic myelofibrosis. The mutant JAK2 protein is capable of activating downstream signals in the absence of cytokine stimulation, resulting in spontaneous growth and/or hypersensitivity to cytokines, which are thought to play a promoting role in the processes of these diseases. Further mutations or translocations that cause dysfunction of JAK2 can be found in the description of other malignancies (Ihle j.n. and Gilliland d.g., curr. opin. genet.dev.,2007,17: 8; Sayyah j.and Sayeski p.p., curr. oncol. rep.,2009,11: 117). JAK2 inhibitors have been described as having an effect on proliferative diseases (Santos et al, Blood,2010,115: 1131; Barosi g.and rosti.v., curr.opin.hematosol, 2009,16:129, atalah e.and Versotvsek s., exp.rev.anticancer ther.,2009,9: 663).
JAK3 is only associated with the common gamma cytokine receptor chain present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JAK3 is expressed primarily in immune cells and transduces signals through tyrosine phosphorylation activation of interleukin receptors. Since JAK3 is mostly restricted to expression in hematopoietic stem cells, its role in cytokine signaling is more stringent compared to other JAKs. Mutations in JAK3 result in Severe Combined Immunodeficiency (SCID) (O' Shea et al, 2002, Cell,109(suppl.): S121-S131). Based on their role in modulating lymphocytes, targeting JAK3 and JAK 3-mediated pathways has been used to treat immunosuppressive indications (e.g., transplant rejection and rheumatoid Arthritis) (Baslund et al, 2005, Arthritis & rhematic 52: 2686-.
TYK2 is associated with IFN- α, IL-6, IL-10 and IL-12 signaling. Biochemical studies and knockout mice have revealed an important role for TYK2 in immunity. TYK 2-deficient mice are able to grow and reproduce, but exhibit a variety of immunodeficiencies, mainly high sensitivity to infection and defects in tumor surveillance. Conversely, inhibition of TYK2 may improve the ability to fight allergic, autoimmune and inflammatory diseases. In particular, targeting TYK2 appears to be an innovative strategy for the treatment of IL-12-, IL-23-or type I IFN-mediated diseases. Such diseases include, but are not limited to, Rheumatoid arthritis, multiple sclerosis, lupus, psoriasis, psoriatic arthritis, inflammatory bowel disease, uveitis, sarcoidosis, and cancer (Shaw, M.et al, Proc. Natl.Acad.Sci., USA,2003,100, 11594-.
The european commission has recently approved fully human monoclonal antibodies (usekinumab) targeting the p40 subunit common to both IL-12 and IL-23 cytokines for the treatment of moderate to severe plaque psoriasis (Krueger et al, 2007, n.engl.j.med.,356: 580-92; Reich et al, 2009, nat. rev.drug discov.,8: 355-356). In addition, antibodies targeting both the IL-12 and IL-23 pathways were tested in clinical trials for the treatment of Crohn's disease (Mannon et al, N.Engl. J.Med.,2004,351: 2069-79).
In dysregulation, JAK-mediated responses can affect cells positively or negatively, leading to over-activation of malignancies, or immune and hematopoietic deficiencies, respectively, suggesting utility for JAK kinase inhibitors. The JAK/STAT signaling pathway has been implicated in a number of proliferative and Cancer-related processes, including cell cycle progression, apoptosis, angiogenesis, infiltration, Metastasis and immune system evasion (Haura et al, Nature Clinical Practice Oncology,2005,2(6), 315-. In addition, the JAK/STAT signaling pathway plays an important role in hematopoietic stem cell generation and differentiation, dual regulation of pro-and anti-inflammatory, and immune response (O' Sullivan et al, Molecular Immunology,2007,44: 2497).
Thus, the JAK/STAT pathway, and in particular all four members of the JAK family, is thought to play a role in the pathogenesis of asthma responses, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract. The JAK/STAT pathway also plays a role in inflammatory diseases of the eye (diseases)/diseases (conditions) including, but not limited to, iritis, uveitis, scleritis, conjunctivitis and slow hypersensitivity reactions. It may be useful to antagonize a specific cytokine-related disease or a disease associated with a mutation or polymorphism in the JAK/STAT pathway by using various forms of JAK kinases (O' Sullivan et al, mol. Immunol,2007,44: 2497; Murray j., Immunol,2007,178:2623) for cytokines to antagonize different selective JAK kinases within the family.
Rheumatoid Arthritis (RA) is an autoimmune disease characterized by chronic joint inflammation. Rheumatoid arthritis patients taking JAK inhibitors show inhibition of JAK1 and JAK3 module signals elicited by a variety of cytokines that are important for lymphocyte function, including interleukin-2 (IL-2), IL-4, IL-7, IL-9, IL-15, and IL-21 [ Fleischmann, R.et. of "Placebo-controlled trial of tofacitinib mongotherapy in rhematoid arthritis, N.Engl. J.Med.,2012,367,495-507 ]. It is speculated that small molecule Inhibitors that directly inactivate specific JAK isoforms may not only alleviate the clinical symptoms of RA, but may also inhibit the over-regulation of many pro-inflammatory cytokines that contribute to the exacerbation of RA disease ("Inhibitors of JAKs for the treatment of inflammatory disorders: ratiometric and clinical data." clin.invest.,2012,2(1), 39-47).
Sustained activation of STAT3 or STAT5 has been demonstrated to be present in many solid human tumors, including breast, pancreas, prostate, ovary, and liver tumors, as well as in a large number of hematologic tumors, including lymphomas and leukemias. In this regard, inactivation of JAK/STAT signals in hematological tumors can inhibit cell proliferation and/or induce apoptosis. Although STAT3 in tumor cells can be activated by many kinases, JAK2 is still considered to be The most important upstream activator, which can activate STAT3(Mohamad Bassam Sonbol, Belal Firwana, Ahmad Zarzour, Mohammad Morad, Vistal Rana and Ramon V.Tiu "Comprehensive review of JAK inhibitor in myeloprolific neoplasms" Therapeutic Advances in Hematology 2013,4(1), 15-35; Hedvat M, Huszar D, Herma A, Gozgit J M, Schroeder A, Sheehy A, et al, "JAK 36 inhibitor D0 site 2, jar 3 and Cell 487, 3, Cancer Cell 2009). Therefore, inhibition of JAK kinases plays a beneficial role in the treatment of these diseases.
It is clearly known that protein kinase inhibitors have gathered much attention as novel immunosuppressive, anti-inflammatory dual acting drugs and anti-cancer drugs. Therefore, new or improved agents that inhibit protein kinases such as Aurora kinase, FLT3 kinase and JAK kinase have long been needed, which are useful as immunosuppressive agents for organ transplantation, antitumor agents, and also for the prevention and treatment of autoimmune diseases (e.g., multiple sclerosis, psoriasis, rheumatoid arthritis, asthma, type I diabetes, inflammatory bowel disease, crohn's disease, polycythemia vera, essential thrombocythemia, myelofibrosis, autoimmune thyroid disease, alzheimer's disease), diseases involving overactivated inflammatory responses (e.g., eczema), allergies, chronic obstructive pulmonary disease, bronchitis, cancer (e.g., prostate cancer, acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphocytic leukemia, multiple myeloma) and immune responses induced by other therapies (e.g., rash, contact dermatitis or diarrhea), and the like. The compounds, compositions and methods described herein directly address these needs and other objectives.
Disclosure of Invention
The present invention provides a class of compounds that inhibit, modulate and/or regulate the activity of one or more protein kinases, such as JAK kinase, FLT3 kinase and Aurora kinase, for the treatment of proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, transplant rejection and complications thereof. The invention also provides processes for preparing these compounds, methods of using these compounds to treat the above-mentioned disorders in mammals, especially humans, and pharmaceutical compositions containing these compounds. Compared with the existing similar compounds, the compound of the invention has better pharmacological activity, and particularly, the compound of the invention shows better inhibitory activity and optimized kinase selectivity on target kinase. In addition, the compound also has excellent film penetrating property and dissolving property, so the compound has better development prospect.
Specifically, the method comprises the following steps:
in one aspect, the invention relates to a compound of formula (I) or a stereoisomer, a tautomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (I), or a prodrug thereof,
Figure BDA0001582369760000061
wherein, Z, Z 1、A、U1、U2、U3And R1Have the meaning as described in the present invention.
In some embodiments, Z is C7-C12Spirobicycloalkyl radical, C7-C12A fused bicycloalkyl group, a spiroheterobicyclic group of 7 to 12 atoms or a fused heterobicyclic group of 7 to 12 atoms, wherein each C7-C12Spirobicycloalkyl radical, C7-C12Fused bicycloalkyl, 7-12 atom spiroheterobicyclic and 7-12 atom fused heterobicyclic groups are independently optionally substituted with 1, 2, 3, 4 or 5R2Substituted by a group; or Z is C3-C7A monocyclic cycloalkyl group or a monocyclic heterocyclic group of 3 to 7 atoms, wherein each of said C3-C7Monocyclic cycloalkyl and 3-7 atom monocyclic heterocyclyl are independently optionally substituted by 1, 2, 3, 4 or 5R3Substituted by a group;
Z1is H, C1-C12Alkyl radical, C3-C12Cycloalkyl or a heterocyclic group of 3 to 12 atoms, wherein each of said C1-C12Alkyl radical, C3-C12Cycloalkyl and heterocyclyl consisting of 3 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R3Substituted by a group;
when U is turned1When is N, U2Is CH, U3Is N;
when U is turned1When being C, U2Is NH, U3Is N or CH;
each is independently a single bond or a double bond;
a is pyrazolyl optionally substituted by 1, 2 or 3R4Substituted by a group;
R1is H, F, Cl, Br, I, N3、CN、NO2、C1-C12Alkyl radical, C2-C12Alkenyl radical, C 2-C12Alkynyl or- (CR)6R7)nC(=O)NRaRbWherein each of C1-C12Alkyl radical, C2-C12Alkenyl and C2-C12Alkynyl is independently optionally substituted with 1, 2, 3, 4 or 5R8Substituted by a group;
each R2Independently F, Cl, Br, I, NO2、N3CN, oxo (═ O), C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, heteroaryl of 5 to 12 atoms, -C (═ O) CH2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-C(=NRd)NRaRb、-N(Rd)C(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbOr two R2Together with the atoms to which they are attached, form C3-C12Cycloalkyl radical, C6-C12Aryl, heterocyclyl of 3 to 12 atoms or heteroaryl of 5 to 12 atoms, wherein each C is1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, alkynyl,C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R8Substituted by a group;
R3is F, Cl, CN, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R8Substituted by a group;
R4Is F, Cl, CN, C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R8Substituted by a group;
each R5Independently is C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C1-C12Alkoxy radical, C1-C12Alkylamino radical, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (C)1-C4Alkylene group) - (C3-C12Cycloalkyl), - (C)1-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), - (C)1-C4Alkylene group) - (C6-C12Aryl) or- (C)1-C4Alkylene) - (heteroaryl of 5-12 atoms) wherein each of the foregoing is independently optionally substituted with 1, 2, 3, 4 or 5R8Substituted by a group;
each R6And R7Are respectively and independently H, F, Cl, Br, I, NO2、N3、CN、C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl or heteroaryl of 5 to 12 atoms, or R6、R7Together with the carbon atom to which they are attached form C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C 6-C12Aryl or heteroaryl of 5 to 12 atoms, wherein each C is1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and hetero 5-12 atomsAryl is independently optionally substituted with 1, 2, 3, 4 or 5R8Substituted by a group;
each R8Independently F, Cl, Br, I, CN, NO2、N3、OH、NH2、C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C1-C12Haloalkyl, C3-C12Cycloalkyl radical, C6-C12Aryl, heterocyclic group of 3-12 atoms, heteroaryl of 5-12 atoms, C1-C12Aminoalkyl radical, C1-C12Alkylamino radical, C1-C12Alkoxy radical, C1-C12Hydroxyalkyl, -NH (C)0-C4Alkylene group) - (C3-C12Cycloalkyl), -NH (C)0-C4Alkylene group) - (C6-C12Aryl), -NH (C)0-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), -NH (C)0-C4Alkylene) - (heteroaryl of 5-12 atoms), -N [ (C)0-C4Alkylene group) - (C3-C12Cycloalkyl radicals]2、-N[(C0-C4Alkylene group) - (C6-C12Aryl radical)]2、-N[(C0-C4Alkylene) - (heterocyclic radical consisting of 3-12 atoms)]2、-N[(C0-C4Alkylene) - (heteroaryl of 5-12 atoms)]2、-O(C0-C4Alkylene group) - (C3-C12Cycloalkyl), -O (C)0-C4Alkylene group) - (C6-C12Aryl), -O (C)0-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms) or-O (C)0-C4Alkylene) - (heteroaryl of 5-12 atoms);
each Ra、Rb、RcAnd RdAre each independently H, C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C 3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12 atomsConstituent heteroaryl, - (C)1-C4Alkylene group) - (C3-C12Cycloalkyl), - (C)1-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), - (C)1-C4Alkylene group) - (C6-C12Aryl) or- (C)1-C4Alkylene) - (heteroaryl of 5-12 atoms), or Ra、RbAnd the nitrogen atom to which they are attached form a heterocyclic group of 3 to 12 atoms, wherein each C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (C)1-C4Alkylene group) - (C3-C12Cycloalkyl), - (C)1-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), - (C)1-C4Alkylene group) - (C6-C12Aryl) and- (C)1-C4Alkylene) - (heteroaryl of 5-12 atoms) optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from F, Cl, Br, CN, N3、NO2、OH、NH2、C1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Hydroxyalkyl radical, C1-C6Aminoalkyl or C1-C6Substituted with a substituent of alkylamino;
each m is independently 1 or 2; and
each n is independently 0, 1, 2, 3 or 4.
In other embodiments, Z is:
Figure BDA0001582369760000071
Figure BDA0001582369760000081
or a stereoisomer thereof, wherein each X and X1Are each independently-CH2-, -NH-, -S-or-O-; x2is-NH-, -S-or-O-; each substructure represented by the formulae (Z-1) to (Z-54) or a stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R 2Substituted by a group; each substructure represented by (Z-55) to (Z-59) or a stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R3Substituted by a group.
In some further embodiments, Z is:
Figure BDA0001582369760000082
Figure BDA0001582369760000091
or a stereoisomer thereof, wherein each substructure represented by the formulae (Z-61) to (Z-79) or the stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R2Substituted by a group; each substructure represented by the formulae (Z-80) to (Z-95) or a stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R3Substituted by a group.
In some embodiments, Z1Is H, C1-C4Alkyl radical, C3-C6Cycloalkyl or heterocyclyl consisting of 4 to 7 atoms, wherein each of said C1-C4Alkyl radical, C3-C6Cycloalkyl and heterocyclyl consisting of 4 to 7 atoms are independently optionally substituted by 1, 2 or 3R3Substituted by a group.
In another embodiment, Z1Is H, methyl, ethyl, n-propyl, isopropyl or cyclopropyl.
In some embodiments, a is:
Figure BDA0001582369760000092
wherein each of the substructures represented by the formulae (A-1) to (A-3) is independently optionally substituted with 1, 2 or 3R4Substituted by a group.
In some embodiments, R1Is H, F, Cl, Br, I, N3、CN、NO2、C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl or- (CR)6R7)nC(=O)NRaRbWherein each of C1-C6Alkyl radical, C2-C6Alkenyl and C2-C6Alkynyl is independently optionally substituted with 1, 2 or 3R 8Substituted by a group.
In some embodiments, each R is2Independently F, Cl, Br, I, NO2、N3CN, oxo (═ O), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, -C (═ O) CH2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)C(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R8Substituted by a group.
In other embodiments, each R is2Independently F, Cl, Br, CN, oxo (═ O), C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclic group consisting of 4-7 atoms, phenyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isothiazolyl, thiazolyl, or a pharmaceutically acceptable salt thereof,Tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl, triazinyl, -C (═ O) CH2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isothiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl and triazinyl are independently optionally substituted with 1, 2 or 3R 8Substituted by a group.
In some embodiments, R3Is F, Cl, CN, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R8Substituted by a group;
R4is F, Cl, CN, C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R8Substituted by a group.
In other embodiments, R3Is F, Cl, CN, cyclopropyl, phenyl, a heterocyclic group of 4-7 atoms or a heteroaryl group of 5-6 atoms, wherein each of the cyclopropyl, phenyl, the heterocyclic group of 4-7 atoms and the heteroaryl group of 5-6 atoms is independently optionally substituted by 1, 2 or 3R8Substituted by a group;
R4is F, Cl, CN, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, a heterocyclic group consisting of 4 to 7 atoms or a heteroaryl group consisting of 5 to 6 atoms, wherein each of the methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, a heterocyclic group consisting of 4 to 7 atoms and a heteroaryl group consisting of 5 to 6 atoms is independently optionally substituted by 1, 2 or 3R 8Substituted by a group.
In still other embodiments, R3Is F, Cl, CN, cyclopropyl, phenyl, heterocyclic radical composed of 4-7 atoms, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isooxazolylThiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl or triazinyl, wherein each of said cyclopropyl, phenyl, heterocyclyl consisting of 4 to 7 atoms, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isothiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl and triazinyl is independently optionally substituted with 1, 2 or 3R8Substituted by a group;
R4is F, Cl, CN, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, a heterocyclic group consisting of 4 to 7 atoms or a heteroaryl group consisting of 5 to 6 atoms, wherein each of the methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, a heterocyclic group consisting of 4 to 7 atoms and a heteroaryl group consisting of 5 to 6 atoms is independently optionally substituted by 1, 2 or 3R 8Substituted by a group.
In some embodiments, each R is5Independently is C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Alkoxy radical, C1-C4Alkylamino radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C)1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl or- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) wherein each of the foregoing is independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In other embodiments, each R is5Independently methyl, ethyl, n-propyl, isopropyl, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Alkoxy radical, C1-C4Alkylamino radical, C3-C6Cycloalkyl, heterocyclic group consisting of 4-7 atoms, phenyl and 5-6 atomsConstituent heteroaryl, - (C)1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl or- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) wherein each of the foregoing is independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In some embodiments, each R is6And R7Are respectively and independently H, F, Cl, Br, CN, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl or heteroaryl consisting of 5 to 6 atoms, wherein each C is 1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R8Substituted by a group.
In some embodiments, each R is8Independently F, Cl, Br, CN, NO2、OH、NH2、C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C3-C6Cycloalkyl, phenyl, heterocyclyl consisting of 4 to 7 atoms, heteroaryl consisting of 5 to 6 atoms, C1-C6Aminoalkyl radical, C1-C6Alkylamino radical, C1-C6Alkoxy radical, C1-C6Hydroxyalkyl, -NH (C)0-C2Alkylene group) - (C3-C6Cycloalkyl), -NH (C)0-C2Alkylene) -phenyl, -NH (C)0-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), -NH (C)0-C2Alkylene) - (heteroaryl of 5-6 atoms), -N [ (C)0-C2Alkylene group) - (C3-C6Cycloalkyl radicals]2、-N[(C0-C2Alkylene) -phenyl]2、-N[(C0-C2Alkylene) - (heterocyclic radical consisting of 4-7 atoms)]2、-N[(C0-C2Alkylene) - (heteroaryl of 5-6 atoms)]2、-O(C0-C2Alkylene group) - (C3-C6Cycloalkyl), -O (C)0-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms) or-O (C)0-C2Alkylene) - (5-6 atom-constituting heteroaryl).
In some embodiments, wherein each R isa、Rb、RcAnd RdAre each independently H, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C) 1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl or- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) wherein each of C is as defined above1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C)1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl and- (C1-C2Alkylene) - (heteroaryl of 5-6 atoms) optionally substituted with 1, 2 or 3 substituents independently selected from F, Cl, Br, CN, N3、OH、NH2、C1-C3Alkyl radical, C1-C3Haloalkyl, C1-C3Alkoxy radical, C1-C3Hydroxyalkyl radical, C1-C3Aminoalkyl or C1-C3Substituted by alkylamino.
In another aspect, the invention relates to a pharmaceutical composition comprising a compound disclosed herein.
In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable excipient.
In other embodiments, the pharmaceutical composition of the invention further comprises a therapeutic agent selected from the group consisting of chemotherapeutic agents, antiproliferative agents, phosphodiesterase 4(PDE4) inhibitors, beta 2-adrenoceptor agonists, corticosteroids, non-steroidal GR agonists, anticholinergics, antihistamines, anti-inflammatory agents, immunosuppressive agents, immunomodulators, drugs for treating atherosclerosis, drugs for treating pulmonary fibrosis, and combinations thereof.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the manufacture of a medicament for the prevention, treatment, or amelioration of a protein kinase mediated disease in a patient.
In some embodiments, the protein kinase mediated disease of the invention is a JAK-, FLT 3-or Aurora-mediated disease.
In other embodiments, the protein kinase-mediated disease of the invention is a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection.
In other embodiments, the protein kinase mediated disease of the invention is cancer, polycythemia vera, essential thrombocythemia, acute myelogenous leukemia, acute lymphocytic leukemia, myelofibrosis, acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphocytic leukemia, chronic obstructive pulmonary disease, asthma, systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, sjogren's syndrome, psoriasis, type I diabetes, respiratory allergic diseases, sinusitis, eczema, measles, food allergies, insect venom allergies, inflammatory bowel disease, crohn's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection, or cell transplant rejection.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the manufacture of a medicament for modulating the activity of a protein kinase.
In some embodiments, the protein kinase of the invention is a JAK kinase, FLT3 kinase, Aurora kinase, or a combination thereof.
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds encompassed by formula (I).
Biological test results show that the compound provided by the invention can be used as a better protein kinase inhibitor.
Any embodiment of any aspect of the invention may be combined with other embodiments, as long as they do not contradict. Furthermore, in any embodiment of any aspect of the invention, any feature may be applicable to that feature in other embodiments, so long as they do not contradict.
The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting. These and other aspects will be more fully described below.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be referred to as described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and Jerry March, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "subject" as used herein refers to an animal. Typically the animal is a mammal. Subjects, e.g., also primates (e.g., humans, males or females), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, etc. In certain embodiments, the subject is a primate. In other embodiments, the subject is a human.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"chiral" is a molecule having the property of not overlapping its mirror image; and "achiral" refers to a molecule that can overlap with its mirror image.
"enantiomer" refers to two isomers of a compound that are not overlapping but are in mirror image relationship to each other.
"diastereomer" refers to a stereoisomer that has two or more chiral neutrals and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow the general definitions of S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E.and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
Many organic compounds exist in an optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of a molecule with respect to one or more of its chiral centers. The prefixes d and l or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or l indicates that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. A particular stereoisomer is an enantiomer and a mixture of such isomers is referred to as an enantiomeric mixture. A50: 50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process.
Any asymmetric atom (e.g., carbon, etc.) of a compound disclosed herein can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Depending on the choice of starting materials and methods, the compounds of the invention may exist as one of the possible isomers or as mixtures thereof, for example as racemates and mixtures of non-corresponding isomers (depending on the number of asymmetric carbon atoms). Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituents may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have cis or trans configuration.
Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
The racemates of any of the resulting end products or intermediates can be resolved into the optical enantiomers by known methods using methods familiar to those skilled in the art, e.g., by separation of the diastereomeric salts obtained. The racemic product can also be separated by chiral chromatography, e.g., High Performance Liquid Chromatography (HPLC) using a chiral adsorbent. In particular, Enantiomers can be prepared by asymmetric synthesis, for example, see Jacques, et al, Enantiomers, racemes and solutions (Wiley Interscience, New York, 1981); principles of Asymmetric Synthesis (2) nd Ed.Robert E.Gawley,Jeffrey Aubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);Wilen,S.H.Tables of Resolving Agents and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of Notre Dame Press,Notre Dame,IN 1972);Chiral Separation Techniques:A Practical Approach(Subramanian,G.Ed.,Wiley-VCH Verlag GmbH&Co.KGaA,Weinheim,Germany,2007)。
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (low energy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers (valenctautomers) include interconversion by recombination of some of the bonding electrons. A specific example of keto-enol tautomerism is the tautomerism of the pentan-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the tautomerism of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention.
It is understood that the term "optionally substituted" may be used interchangeably with the term "substituted or unsubstituted". In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a particular substituent. "optionally" unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently.
The terms "optional" or "optionally" mean that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl" means that an alkyl may, but need not, be present, and the description includes the scenario where the heterocyclic group is substituted with an alkyl and the scenario where the heterocyclic group is not substituted with an alkyl.
The term "optionally substituted with … …" is used interchangeably with the term "unsubstituted or substituted with … …", i.e., the structure is unsubstituted or substituted with one or more substituents described herein, including but not limited to D, F, Cl, Br, I, N 3、CN、NO2、OH、SH、NH2Oxo (═ O), alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, hydroxyalkyl, alkylthio, alkylamino, aminoalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C (═ O) CH2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-C(=NRd)NRaRb、-N(Rd)C(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbAnd so on. Wherein R is5、R6、R7、Ra、Rb、Rc、RdM and n have the meanings as described in the invention.
In addition, unless otherwise explicitly indicated, the descriptions of the terms "… … independently" and "… … independently" and "… … independently" used in the present invention are interchangeable and should be understood in a broad sense to mean that the specific items expressed between the same symbols do not affect each other in different groups or that the specific items expressed between the same symbols in the same groups do not affect each other.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-C6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group; the term "heterocyclic group consisting of 4 to 7 atoms" means a heterocyclic group consisting of 4 atoms, a heterocyclic group consisting of 5 atoms, a heterocyclic group consisting of 6 atoms or a heterocyclic group consisting of 7 atoms which are independently disclosed.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for that variable recites "alkyl", "heterocyclyl", or "aryl", it is to be understood that the "alkyl", "heterocyclyl", or "aryl" represents an attached alkylene group, heterocyclylene group, or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated, straight or branched chain monovalent hydrocarbon radical containing from 1 to 20 carbon atoms, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 3 carbon atoms. The alkyl group may be optionally substituted with the substituents described herein.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH)3) Ethyl group (Et, -CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl group (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) Tert-butyl (t-Bu, -C (CH)3)3) N-pentyl (-CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH(CH3)2) 3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) N-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2) 2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2) 3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)3) N-heptyl, n-octyl, and the like.
The term "alkylene" refers to a saturated divalent hydrocarbon radical resulting from the removal of two hydrogen atoms from a saturated straight or branched chain hydrocarbon radical. Unless otherwise specifiedIllustratively, the alkylene group contains 1 to 12 carbon atoms. In some embodiments, the alkylene group contains 1 to 6 carbon atoms; in another embodiment, the alkylene group contains 1 to 4 carbon atoms; in other embodiments, the alkylene group contains from 0 to 4 carbon atoms; in still other embodiments, the alkylene group contains from 0 to 3 carbon atoms; in still other embodiments, the alkylene group contains from 0 to 2 carbon atoms. Alkylene having 0 carbon atoms means that alkylene is absent and is directly a single bond. Examples of alkylene groups include, but are not limited to, methylene (-CH) 2-, ethylene (-CH)2CH2-, isopropylidene (-CH (CH)3)CH2-) and the like.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "trans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 8 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. a carbon-carbon sp triple bond. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH) 2C ≡ CH), propynyl (-C ≡ C-CH)3) And so on. The alkynyl group may be optionally substituted with one or more substituents described herein.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein.
Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-PrO, n-propoxy, -OCH)2CH2CH3) 2-propoxy (i-PrO, isopropoxy, -OCH (CH)3)2) 1-butoxy (n-BuO, n-butoxy, -OCH)2CH2CH2CH3) 2-methyl-l-propoxy (i-BuO, isobutoxy, -OCH)2CH(CH3)2) 2-butoxy (s-BuO, sec-butoxy, -OCH (CH)3)CH2CH3) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH)3)3) 1-pentyloxy (n-pentyloxy, -OCH) 2CH2CH2CH2CH3) 2-pentyloxy (-OCH (CH)3)CH2CH2CH3) 3-pentyloxy (-OCH (CH))2CH3)2) 2-methyl-2-butoxy (-OC (CH))3)2CH2CH3) 3-methyl-2-butoxy (-OCH (CH)3)CH(CH3)2) 3-methyl-l-butoxy (-OCH)2CH2CH(CH3)2) 2-methyl-l-butoxy (-OCH)2CH(CH3)CH2CH3) And so on.
The terms "haloalkyl", "haloalkenyl" or "haloalkoxy" denote alkyl, alkenyl or alkoxy groups substituted with one or more halogen atoms, examples of which include, but are not limited to, trifluoromethyl (-CF)3) Trifluoromethoxy (-OCF)3) Difluoroethyl (-CH)2CHF2,-CF2CH3,-CHFCH2F) Trifluoroethyl (-CH)2CF3,-CF2CH2F,-CFHCHF2) And the like.
The terms "hydroxyalkyl" and "hydroxyalkoxy" mean alkyl or alkoxy, as the case may be, substituted with one or more hydroxy groups, where "hydroxyalkyl" and "hydroxyalkyl" may be used interchangeably, and examples include, but are not limited to, hydroxymethyl (-CH)2OH), 2-hydroxyethyl (-CH)2CH2OH), 1-hydroxyethyl (-CH (OH) CH3) 2-hydroxypropan-2-yl (-COH (CH)3)2) 2-hydroxy-2-methylpropyl (-CH)2COH(CH3)2) 3-hydroxypropyl (-CH)2CH2CH2OH), 2-hydroxypropyl (-CH)2CH(OH)CH3) Hydroxy methoxy (-OCH)2OH), and the like.
The term "carbocyclyl" or "carbocycle" denotes a monovalent or multivalent, non-aromatic, saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring system containing 3 to 12 carbon atoms. Carbobicyclic groups include spirocarbocyclic and fused carbocyclic groups, and suitable carbocyclic groups include, but are not limited to, cycloalkyl, cycloalkenyl and cycloalkynyl groups. Examples of carbocyclyl groups further include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 carbon atoms. Bicyclic cycloalkyl groups include spirobicycloalkyl, fused bicycloalkyl and bridged bicycloalkyl groups. In some embodiments, cycloalkyl groups contain 3 to 12 carbon atoms; in other embodiments, cycloalkyl groups contain 3 to 7 carbon atoms; in other embodiments, cycloalkyl groups contain 3 to 6 carbon atoms; in still other embodiments, cycloalkyl is C7-C12Cycloalkyl group containing C7-C12Monocycloalkyl radical, C7-C12Bicycloalkyl (e.g. C)7-C12Spirobicycloalkyl radical, C7-C12Fused bicycloalkyl and C7-C12Bridged bicycloalkyl) or C7-C12A tricycloalkyl group. The cycloalkyl groups may be independently unsubstituted or substituted with one or more substituents described herein. The term "monocyclic cycloalkyl" or "monocycloalkyl" denotes a cycloalkyl group of a monocyclic ring system, wherein the cycloalkyl group has the definitions as described above, and the monocyclic cycloalkyl group can independently be unsubstituted or substituted by one or more substituents described herein.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein and refer to a mono-, bi-or tricyclic ring containing 3 to 12 ring atoms, which is monovalent or multivalent, saturated or partially unsaturated, and which is not aromatic, wherein at least one ring atom is selected from nitrogen, sulfur, and oxygen atoms. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH 2The group may optionally be replaced by-C (═ O) -, the sulfur atom of the ring may optionally be oxidized to S-oxide, and the nitrogen atom of the ring may optionally be oxidized to N-oxygen compound. The heterocyclic group includes a saturated heterocyclic group (heterocycloalkyl group) and a partially unsaturated heterocyclic group. The heterocyclic group has one or more attachment points to the rest of the molecule. Examples of heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, nitrogen monoheterocycloheptanyl, oxazepinyl, oxazepin
Figure BDA0001582369760000151
Radical (e.g. 1, 4-oxa-nitrogen)Hetero compound
Figure BDA0001582369760000152
1, 2-oxaza
Figure BDA0001582369760000153
Alkyl), diazepine
Figure BDA0001582369760000154
Radicals (e.g. 1, 4-diazepine)
Figure BDA0001582369760000155
1, 2-diazepines
Figure BDA0001582369760000156
Basic), dioxa
Figure BDA0001582369760000157
Radicals (e.g. 1, 4-dioxa)
Figure BDA0001582369760000158
1, 2-dioxan
Figure BDA0001582369760000159
Basic), a sulfur aza
Figure BDA00015823697600001510
Radicals (e.g. 1, 4-thiazepine)
Figure BDA00015823697600001511
1, 2-thiaza radical
Figure BDA00015823697600001512
Indolyl), indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1]Hept-5-yl, 2-azaspiro [4.4]Nonanyl, 1, 6-dioxaspiro [4.4 ]]Nonanyl, 2-azaspiro [4.5 ]]Decyl, 8-azaspiro [4.5 ]]Decyl, 7-azaspiro [4.5 ]]Decyl, 3-azaspiro [5.5 ]]Undecyl, 2-azaspiro [5.5]Undecyl, octahydro-1H-isoindolyl, octahydrocyclopenta [ c]Pyrrolyl, indolinyl, 1,2,3, 4-tetra-ethylHydroisoquinolinyl, hexahydrofuro [3,2-b ] s]Furyl and dodecahydroisoquinolinyl, and the like. In heterocyclic radicals of-CH2Examples of-groups substituted by-C (═ O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl and 3, 5-dioxopiperidinyl. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group, 1-dioxothiomorpholinyl group. The heterocyclyl group may be optionally substituted with one or more substituents as described herein. The term "monocyclic heterocyclyl" or "monocyclic heterocycle" denotes a heterocyclyl group of a monocyclic ring system, wherein the heterocyclyl group has the definitions as described above, which monocyclic heterocyclyl group may independently be unsubstituted or substituted by one or more substituents as described herein.
In yet another embodiment, heterocyclyl is a 4-7 atom heterocyclyl and refers to a monovalent or polyvalent, saturated or partially unsaturated, nonaromatic, monocyclic or bicyclic ring containing 4-7 ring atoms wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms. Unless otherwise specified, a heterocyclic group of 4 to 7 atoms may be carbon-based or nitrogen-based, and-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. The 4-7 atom heterocyclyl group has one or more attachment points to the rest of the molecule. Among them, examples of the monocyclic heterocyclic group consisting of 4 to 7 atoms include, but are not limited to: azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, 1, 2-oxazinyl, 1, 2-thiazinyl, hexahydropyridazinyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepanyl, and pyrazolinyl
Figure BDA00015823697600001513
Radical (1, 4-oxaza)
Figure BDA00015823697600001514
1, 2-oxaza
Figure BDA00015823697600001515
Alkyl), diazepine
Figure BDA00015823697600001516
Radical (1, 4-diazepine)
Figure BDA00015823697600001517
1, 2-diazepines
Figure BDA00015823697600001518
Alkyl) and thiazepine
Figure BDA00015823697600001519
Radical (1, 4-thiazepine)
Figure BDA00015823697600001520
1, 2-thiaza radical
Figure BDA00015823697600001521
Base), etc.; examples of bicyclic heterocyclic groups consisting of 4 to 7 atoms include, but are not limited to: 3-azabicyclo [3,2,0 ] s]Heptane, 3-oxobicyclo [3,2,0 ]]Heptane and the like; 4-7 atoms in heterocyclic radical2Examples of-group replacement by-C (═ O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl and 3, 5-dioxopiperidinyl; examples of the sulfur atom in the heterocyclic group consisting of 4 to 7 atoms being oxidized include, but are not limited to, sulfolane group, 1-dioxotetrahydrothiophene, 1-dioxotetrahydrothiopyran, 1-dioxothiomorpholinyl. Said heterocyclyl group of 4 to 7 atoms may be optionally substituted by one or more substituents as described herein.
The terms "fused bicyclic ring" and "fused bicyclic group" are used interchangeably herein and refer to a monovalent or multivalent saturated or partially unsaturated, but non-aromatic, ring system in which the two rings share a single bond. Such systems may contain independent or conjugated unsaturated systems, but the core structure does not contain aromatic or heteroaromatic rings, but aromatic groups may be substituted thereon.
The terms "spirobicyclic group" and "spirobicyclic ring" are used interchangeably herein and refer to a monovalent or polyvalent saturated or partially unsaturated ring system in which one ring is derived from a specific ring carbon atom on the other ring and only one atom is shared by both rings. For example, as depicted in formula a below, a saturated ring system (rings U and U') is referred to as a "fused bicyclic ring", while ring V and ring U share a carbon atom, referred to as a "spirobicyclic ring". Each ring in the fused bicyclic and spirobicyclic groups may be a carbocyclic or heterocyclic group, and each ring is optionally substituted with one or more substituents described herein.
Figure BDA0001582369760000161
The term "heterocycloalkyl" refers to a monovalent or polyvalent saturated monocyclic, bicyclic, or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur, or oxygen atoms. Unless otherwise indicated, heterocycloalkyl can be carbon or nitrogen based, and-CH2The group may optionally be replaced by-C (═ O) -, the sulfur atom of the ring may optionally be oxidized to S-oxide, and the nitrogen atom of the ring may optionally be oxidized to N-oxygen compound. The heterocycloalkyl group can be optionally substituted with one or more substituents described herein.
The term "n-atomic" where n is an integer typically describes the number of ring-forming atoms in a molecule in which the number of ring-forming atoms is n. For example, piperidinyl is a heterocycloalkyl group of 6 atoms, and 1,2,3, 4-tetrahydronaphthyl is a carbocyclyl group of 10 atoms.
The term "unsaturated" as used herein means that the group contains one or more unsaturations.
The term "heteroatom" refers to O, S, N, P and Si, including N, S and any oxidation state form of P; primary, secondary, tertiary amines and quaternary ammonium salt forms; or a form in which a hydrogen on a nitrogen atom in the heterocycle is substituted, for example, N (like N in 3, 4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like NR in N-substituted pyrrolidinyl).
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
The term "azido" or "N3"represents an azide structure. Such groups may be linked to other groups, e.g. to a methyl group to form azidomethane (Men)3) Or linked to a phenyl group to form azidobenzene (PhN)3)。
The term "aryl" denotes monocyclic, bicyclic and tricyclic carbon ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring system is aromatic, wherein each ring system contains 3 to 7 atoms in the ring and one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". Examples of the aryl group may include phenyl, naphthyl and anthracenyl. The aryl group may independently be optionally substituted with one or more substituents described herein.
The term "heteroaryl" denotes monocyclic, bicyclic and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one aromatic ring contains one or more heteroatoms, wherein each ring system contains a ring of 5 to 7 atoms with one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". In one embodiment, heteroaryl is a heteroaryl consisting of 5 to 12 atoms containing 1,2,3 or 4 heteroatoms independently selected from O, S and N. In another embodiment, heteroaryl is a heteroaryl consisting of 5 to 10 atoms containing 1,2,3, or 4 heteroatoms independently selected from O, S, and N. In another embodiment, heteroaryl is 5-6 atom composed of 1,2,3, or 4 heteroatoms independently selected from O, S, and N. The heteroaryl group is optionally substituted with one or more substituents described herein.
Examples of 5-12 atom-composed heteroaryl groups include, but are in no way limited to, the following bicyclic heteroaryl groups: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl, 3-indolyl, 4-indolyl, 5-indolyl, 6-indolyl, 7-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), indazolyl (e.g., 3-indazolyl, 4-indazolyl, 5-indazolyl, 6-indazolyl, 7-indazolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyridyl, pyrazolo [4,3-c ] pyridyl, pyrazolo [3,4-b ] pyridyl, pyrazolo [1,5-a ] pyrimidyl, pyrazolyl, and the like, Imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridinyl, and the like. Examples of heteroaryl groups of 5-12 atoms also include monocyclic heteroaryl groups of 5-6 atoms, examples of which include, but are not limited to, monocyclic rings, furyl (e.g., 2-furyl, 3-furyl), imidazolyl (e.g., 1-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl), isoxazolyl (e.g., 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl), oxazolyl (e.g., 2-oxazolyl, 4-oxazolyl, 5-oxazolyl), pyrrolyl (e.g., 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl), pyridyl (e.g., 2-pyridyl, 3-pyridyl, 4-pyridyl), pyridonyl, pyrimidinyl (e.g., 2-pyrimidinyl, pyrimidinyl), 4-pyrimidinyl, 5-pyrimidinyl), pyrimidinonyl, pyrimidinedionyl, pyridazinyl (e.g., 3-pyridazinyl, 4-pyridazinyl), pyrazinyl (e.g., 2-pyrazinyl, 3-pyrazinyl), thiazolyl (e.g., 2-thiazolyl, 4-thiazolyl, 5-thiazolyl), tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), thienyl (e.g., 2-thienyl, 3-thienyl), pyrazolyl (e.g., 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, 5-pyrazolyl), pyrazolinonyl, isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, pyrazinyl, and 1,3, 5-triazinyl, and the like.
The terms "alkylamino" or "alkylamino" are used interchangeably herein and include "N-alkylamino" and "N, N-dialkylamino" in which the amino groups are each independently substituted with one or two alkyl groups. Wherein, in some embodiments, the alkylamino group is one or two C1-C12The alkyl group is attached to a nitrogen atom to form a lower alkylamino group. In other embodiments, alkylamino is one or two C1-C6The alkyl group is attached to a nitrogen atom to form a lower alkylamino group. In other embodiments, alkylamino is one or two C1-C4The alkyl group is attached to a nitrogen atom to form a lower alkylamino group. In still other embodiments, the alkylamino group is one or two C1-C3The alkyl group is attached to a nitrogen atom to form a lower alkylamino group. Suitable alkylamino groups can be monoalkylamino or dialkylamino, and examples of alkylamino include, but are not limited to, N-methylamino (-NHCH)3) N-ethylamino, N, N-dimethylamino (-NH (CH)3)2) N, N-diethylamino, N-ethylpropyl-2-amino and the like.
The term "arylamino" denotes an amino group substituted with one or two aryl groups, examples of which include, but are not limited to, N-phenylamino. In some embodiments, the aromatic ring on the arylamino group may be further substituted.
The term "aminoalkyl" includes C substituted with one or more amino groups1-C12A straight or branched alkyl group. In some embodiments, aminoalkyl is C substituted with one or more amino groups1-C12An alkyl group; in other embodiments, aminoalkyl is C substituted with one or more amino groups1-C6"lower aminoalkyl", in other embodiments, aminoalkyl is C substituted with one or more amino groups1-C4An alkyl group; in still other embodiments, aminoalkyl is taken from one or more amino groupsSubstituted C1-C3An alkyl group. Examples of aminoalkyl radicals include, but are not limited to, aminomethyl (-CH)2NH2) Aminoethyl (-CH)2CH2NH2,-CH(NH2)CH3) Aminopropyl, aminobutyl and aminohexyl.
As described herein, a ring system formed by drawing a bond connecting a substituent to the center of the ring (as shown in formula b) represents that the substituent may be substituted at any substitutable position on the ring system. For example, the substituent represented by formula b may be substituted at any possible position on the pyrazole ring, as shown in formulae c to f.
Figure BDA0001582369760000171
As described in the present invention, the ring system formed by the two linkages attached to the center of the ring (as shown in formula h) means that both linkages can be attached to the rest of the molecule at any of the available positions on the ring system and that the two attachment points K and K' are interchangeable. Formula h represents that any of the two possible attachment positions on the W ring can be attached to the rest of the molecule.
Figure BDA0001582369760000172
For example, as shown in formula i below, a linkage is attached to the center of the piperidine ring, meaning that the linkage can be attached to the remainder of the molecule at any available position on the piperidine ring. Formula i indicates that the piperidine ring may be attached to the rest of the molecule at positions where it may be attached, as shown in formulae j-m.
Figure BDA0001582369760000173
The term "protecting group" or "PG" refers to a substituent group that blocks or protects a particular functionality when other functional groups in a compound are reacted. For example, "of amino groupsProtecting group "means a substituent attached to an amino group to block or protect the functionality of the amino group in a compound, and suitable amino protecting groups include acetyl, trifluoroacetyl, tert-butoxycarbonyl (BOC ), benzyloxycarbonyl (CBZ ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). Similarly, "hydroxyl protecting group" refers to the functionality of a substituent of a hydroxyl group to block or protect the hydroxyl group, and suitable protecting groups include acetyl and silyl groups. "carboxy protecting group" refers to the functionality of a substituent of a carboxy group to block or protect the carboxy group, and typical carboxy protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General descriptions of protecting groups can be found in the literature: greene, Protective Groups in Organic Synthesis, John Wiley &Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel Delivery Systems, Vol.14 of the A.C.S.Symphosis Series, Edward B.Roche, ed., Bioreproducible Cariers in Drug Design, American Pharmaceutical Association and Pergamon Press,1987, J.Rautio et al, Prodrugs in Design and Clinical Applications, Nature Review Drug Discovery,2008 ,7,255-270,and S.J.Hecker et al.,Prodrugs of Phosphates and Phosphonates,Journal of Medicinal Chemistry,2008,51,2328-2345。
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, description of the scientific acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids formed by reaction with amino groups such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or those obtained by other methods described in the literature above, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, laurylsulfates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates Nicotinate, nitrate, oleate, palmitate, embonate, pectate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, stearate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
The term "treating" or "treatment" as used herein refers, in some embodiments, to ameliorating a disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
As used herein, "inflammatory disease" refers to any disease, disorder or condition of excessive inflammatory symptoms, host tissue damage or loss of tissue function due to excessive or uncontrolled inflammatory response. "inflammatory disease" also refers to a pathological condition mediated by leukocyte influx and/or neutrophil chemotaxis.
As used herein, "inflammation" refers to a local protective response caused by tissue damage or destruction that serves to destroy, dilute, or separate (sequester) harmful substances from damaged tissue. Inflammation is significantly linked to leukocyte influx and/or neutrophil chemotaxis. Inflammation can result from infection by pathogenic organisms and viruses, as well as from non-infectious means, such as trauma or reperfusion following myocardial infarction or stroke, immune and autoimmune responses to foreign antigens. Thus, inflammatory diseases that may be treated with the disclosed compounds include: diseases associated with specific defense system reactions as well as non-specific defense system reactions.
By "specific defense system" is meant that components of the immune system respond to the presence of a particular antigen. Examples of inflammation arising from specific defense system responses include classical responses to foreign antigens, autoimmune diseases, and delayed hypersensitivity responses (mediated by T-cells). Chronic inflammatory diseases, rejection of transplanted solid tissues and organs (such as rejection of kidney and bone marrow transplants), and Graft Versus Host Disease (GVHD) are other examples of specific defense systems against inflammatory reactions.
As used herein, "autoimmune disease" refers to any collection of diseases of tissue damage associated with humoral or cell-mediated responses to the body's own components.
As used herein, "allergy" refers to any symptom of developing an allergy, tissue damage, or loss of tissue function. As used herein, "arthritic disease" refers to any disease characterized by inflammatory injury to the joints attributable to various etiologies. As used herein, "dermatitis" refers to any of a large family of skin diseases characterized by skin inflammation attributable to various etiologies. As used herein, "transplant rejection" refers to any immune response against a transplanted tissue, such as an organ or cell (e.g., bone marrow), characterized by loss of function of the transplanted or surrounding tissue, pain, swelling, leukocytosis, and thrombocytopenia. The therapeutic methods of the invention include methods for treating diseases associated with inflammatory cell activation.
The terms "cancer" and "cancerous" refer to or describe the physiological condition in a patient that is often characterized by uncontrolled cell growth. A "tumor" comprises one or more cancer cells. Examples of cancer include, but are not limited to, carcinoma (carcinoma), lymphoma, blastoma, sarcoma, and leukemia, or lymphoproliferative disorder (lymphoproliferative disorders). More specific examples of such cancers include squamous cell cancer (such as epithelial squamous cell cancer), lung cancer (including small-cell lung cancer, non-small cell lung cancer (NSCLC), adenocarcinoma of the lung and squamous carcinoma of the lung), cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer (including gastrointestinal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer (liver cancer), bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer (kidney or renal cancer), prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal cancer, penile carcinoma, and head and neck cancer.
Description of the Compounds of the invention
The invention discloses a novel compound which can be used as an inhibitor of the activity of protein kinases, particularly the activity of JAK kinase, FLT3 kinase and Aurora kinase. Compounds that are inhibitors of protein kinases are useful in the treatment of diseases associated with inappropriate protein kinase activity, particularly inappropriate JAK kinase, FLT3 kinase and Aurora kinase activity, for example in the treatment and prevention of JAK kinase, FLT3 kinase and Aurora kinase mediated diseases involving signaling pathways. Such diseases include proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, transplant rejection, and complications thereof. In particular, the compounds of the present invention may be used to treat diseases such as cancer, polycythemia vera, essential thrombocythemia, myelofibrosis, myeloid leukemia, acute lymphocytic leukemia, Chronic Myelogenous Leukemia (CML), Chronic Obstructive Pulmonary Disease (COPD), asthma, systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, sjogren's syndrome, psoriasis, type I diabetes, respiratory allergic disease, sinusitis, eczema, measles, food allergy, insect venom allergy, inflammatory bowel disease, crohn's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection, cell transplant rejection, and the like.
In one embodiment, the disclosed compounds may exhibit enhanced inhibitory activity against one or more protein kinases.
In one aspect, the invention relates to a compound of formula (I) or a stereoisomer, a tautomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (I), or a prodrug thereof,
Figure BDA0001582369760000201
wherein, Z, Z1、A、U1、U2、U3And R1Have the meaning as described in the present invention.
In some embodiments, Z is C7-C12Spirobicycloalkyl radical, C7-C12A fused bicycloalkyl group, a spiroheterobicyclic group of 7 to 12 atoms or a fused heterobicyclic group of 7 to 12 atoms, wherein each C7-C12Spirobicycloalkyl radical, C7-C12Fused bicycloalkyl, 7-12 atom spiroheterobicyclic and 7-12 atom fused heterobicyclic groups are independently optionally substituted with 1, 2, 3, 4 or 5R2Substituted by a group; or Z is C3-C7A monocyclic cycloalkyl group or a monocyclic heterocyclic group of 3 to 7 atoms, wherein each of said C3-C7Monocyclic cycloalkyl and 3-7 atom monocyclic heterocyclyl are independently optionally substituted by 1, 2, 3, 4 or 5R3Substituted by a group;
Z1is H, C1-C12Alkyl radical, C3-C12Cycloalkyl or 3-12 atoms, wherein each C is 1-C12Alkyl radical, C3-C12Cycloalkyl and heterocyclyl consisting of 3 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R3Substituted by a group;
when U is turned1When is N, U2Is CH, U3Is N;
when U is turned1Is C, U2Is NH, U3Is N or CH;
each is independently a single bond or a double bond;
a is pyrazolyl optionally substituted by 1, 2 or 3R4Substituted by a group;
R1is H, F, Cl, Br, I, N3、CN、NO2、C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl or- (CR)6R7)nC(=O)NRaRbWherein each of C1-C12Alkyl radical, C2-C12Alkenyl and C2-C12Alkynyl is independently optionally substituted with 1, 2, 3, 4 or 5R8Substituted by a group;
each R2Independently F, Cl, Br, I, NO2、N3CN, oxo (═ O), C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, heteroaryl of 5 to 12 atoms, -C (═ O) CH2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-C(=NRd)NRaRb、-N(Rd)C(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbOr two R2Together with the atoms to which they are attached, form C3-C12Cycloalkyl radical, C6-C12Aryl, heterocyclyl of 3 to 12 atoms or heteroaryl of 5 to 12 atoms, wherein each C is1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R 8Substituted by a group;
R3is F, Cl, CN, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R8Substituted by a group;
R4is F, Cl, CN, C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R8Substituted by a group;
each R5Independently is C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C1-C12Alkoxy radical, C1-C12Alkylamino radical, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (C)1-C4Alkylene group) - (C3-C12Cycloalkyl), - (C)1-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), - (C)1-C4Alkylene group) - (C6-C12Aryl) or- (C)1-C4Alkylene) - (heteroaryl of 5-12 atoms) wherein each of the foregoing is independently optionally substituted with 1, 2, 3, 4 or 5R 8Substituted by a group;
each R6And R7Are respectively and independently H, F, Cl, Br, I, NO2、N3、CN、C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl or heteroaryl of 5 to 12 atoms, or R6、R7And the carbon atom to which they are attachedForm C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl or heteroaryl of 5 to 12 atoms, wherein each C is1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl and heteroaryl of 5 to 12 atoms are independently optionally substituted by 1, 2, 3, 4 or 5R8Substituted by a group;
each R8Independently F, Cl, Br, I, CN, NO2、N3、OH、NH2、C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C1-C12Haloalkyl, C3-C12Cycloalkyl radical, C6-C12Aryl, heterocyclic group of 3-12 atoms, heteroaryl of 5-12 atoms, C1-C12Aminoalkyl radical, C1-C12Alkylamino radical, C1-C12Alkoxy radical, C1-C12Hydroxyalkyl, -NH (C)0-C4Alkylene group) - (C3-C12Cycloalkyl), -NH (C)0-C4Alkylene group) - (C6-C12Aryl), -NH (C)0-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), -NH (C)0-C4Alkylene) - (heteroaryl of 5-12 atoms), -N [ (C)0-C4Alkylene group) - (C3-C12Cycloalkyl radicals]2、-N[(C0-C4Alkylene group) - (C6-C12Aryl radical)]2、-N[(C0-C4Alkylene) - (heterocyclic radical consisting of 3-12 atoms) ]2、-N[(C0-C4Alkylene) - (heteroaryl of 5-12 atoms)]2、-O(C0-C4Alkylene group) - (C3-C12Cycloalkyl), -O (C)0-C4Alkylene group) - (C6-C12Aryl), -O (C)0-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms) or-O (C)0-C4Alkylene) - (heteroaryl of 5-12 atoms);
each Ra、Rb、RcAnd RdAre each independently H, C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (C)1-C4Alkylene group) - (C3-C12Cycloalkyl), - (C)1-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), - (C)1-C4Alkylene group) - (C6-C12Aryl) or- (C)1-C4Alkylene) - (heteroaryl of 5-12 atoms), or Ra、RbAnd the nitrogen atom to which they are attached form a heterocyclic group of 3 to 12 atoms, wherein each C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C12Cycloalkyl, heterocyclic group consisting of 3 to 12 atoms, C6-C12Aryl, 5-12-membered heteroaryl, - (C)1-C4Alkylene group) - (C3-C12Cycloalkyl), - (C)1-C4Alkylene) - (heterocyclic group consisting of 3-12 atoms), - (C)1-C4Alkylene group) - (C6-C12Aryl) and- (C)1-C4Alkylene) - (heteroaryl of 5-12 atoms) optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from F, Cl, Br, CN, N3、NO2、OH、NH2、C1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Hydroxyalkyl radical, C 1-C6Aminoalkyl or C1-C6Substituted with a substituent of alkylamino;
each m is independently 1 or 2; and
each n is independently 0, 1, 2, 3 or 4.
In other embodiments, Z is:
Figure BDA0001582369760000211
Figure BDA0001582369760000221
Figure BDA0001582369760000222
or a stereoisomer thereof, wherein each X and X1Are each independently-CH2-, -NH-, -S-or-O-; x2is-NH-, -S-or-O-; each substructure represented by the formulae (Z-1) to (Z-54) or a stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R2Substituted by a group; each substructure represented by (Z-55) to (Z-59) or a stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R3Substituted by a group.
In some further embodiments, Z is:
Figure BDA0001582369760000223
Figure BDA0001582369760000231
or a stereoisomer thereof, wherein each substructure represented by the formulae (Z-61) to (Z-79) or the stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R2Substituted by a group; each substructure represented by the formulae (Z-80) to (Z-95) or a stereoisomer thereof is independently optionally substituted with 1, 2, 3, 4 or 5R3Substituted by a group.
In some embodiments, Z1Is H, C1-C4Alkyl radical, C3-C6Cycloalkyl or heterocyclyl consisting of 4 to 7 atoms, wherein each of said C1-C4Alkyl radical, C3-C6Cycloalkyl and heterocyclyl consisting of 4 to 7 atoms are independently optionally substituted by 1, 2 or 3R3Substituted by a group.
In another embodiment, Z1Is H, methyl, ethyl, n-butylPropyl, isopropyl or cyclopropyl.
In some embodiments, a is:
Figure BDA0001582369760000232
wherein each of the substructures represented by the formulae (A-1) to (A-3) is independently optionally substituted with 1, 2 or 3R4Substituted by a group.
In some embodiments, R1Is H, F, Cl, Br, I, N3、CN、NO2、C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl or- (CR)6R7)nC(=O)NRaRbWherein each of C1-C6Alkyl radical, C2-C6Alkenyl and C2-C6Alkynyl is independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In some embodiments, each R is2Independently F, Cl, Br, I, NO2、N3CN, oxo (═ O), C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, -C (═ O) CH2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)C(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and 5 to 6 atomsThe constituent heteroaryl groups are independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In other embodiments, each R is2Independently F, Cl, Br, CN, oxo (═ O), C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, a heterocyclic group consisting of 4 to 7 atoms, phenyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isothiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl, triazinyl, -C (═ O) CH 2CN、-(CR6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isothiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl and triazinyl are independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In some embodiments, R3Is F, Cl, CN, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R8Substituted by a group;
R4is F, Cl, CN, C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (CR)6R7)n-ORc、-(CR6R7)n-NRaRb、-C(=O)R5、-OC(=O)R5、-N(Rd)C(=O)R5、-(CR6R7)nC(=O)ORc、-(CR6R7)nC(=O)NRaRb、-N(Rd)S(=O)mR5or-S (═ O)2NRaRbWherein each of C1-C4Alkyl radical, C2-C4Alkenyl radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R 8Substituted by a group.
In other embodiments, R3Is F, Cl, CN, cyclopropyl, phenyl, a heterocyclic group of 4-7 atoms or a heteroaryl group of 5-6 atoms, wherein each of the cyclopropyl, phenyl, the heterocyclic group of 4-7 atoms and the heteroaryl group of 5-6 atoms is independently optionally substituted by 1, 2 or 3R8Substituted by a group;
R4is F, Cl, CN, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, heterocyclic radical composed of 4-7 atoms or composed of 5-6 atomsWherein each of said methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, 4-7 member heterocyclyl and 5-6 member heteroaryl is independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In still other embodiments, R3Is F, Cl, CN, cyclopropyl, phenyl, heterocyclic group consisting of 4-7 atoms, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidonyl, pyridazinyl, thiazolyl, isothiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl or triazinyl, wherein each of the cyclopropyl, phenyl, heterocyclic group consisting of 4-7 atoms, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyridyl, pyridonyl, pyrimidinyl, pyrimidyl, pyridazinyl, thiazolyl, isothiazolyl, tetrazolyl, triazolyl, thienyl, pyrazolyl, oxadiazolyl, thiadiazolyl, pyrazinyl and triazinyl is independently optionally substituted with 1, 2 or 3R 8Substituted by a group;
R4is F, Cl, CN, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, a heterocyclic group consisting of 4 to 7 atoms or a heteroaryl group consisting of 5 to 6 atoms, wherein each of the methyl, ethyl, n-propyl, isopropyl, cyclopropyl, phenyl, a heterocyclic group consisting of 4 to 7 atoms and a heteroaryl group consisting of 5 to 6 atoms is independently optionally substituted by 1, 2 or 3R8Substituted by a group.
In some embodiments, each R is5Independently is C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Alkoxy radical, C1-C4Alkylamino radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C)1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl or- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) wherein each of the foregoing is independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In other embodiments, each R is5Independently methyl, ethyl, n-propyl, isopropyl, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Alkoxy radical, C1-C4Alkylamino radical, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C)1-C2Alkylene group) - (C 3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl or- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) wherein each of the foregoing is independently optionally substituted with 1, 2 or 3R8Substituted by a group.
In some embodiments, each R is6And R7Are respectively and independently H, F, Cl, Br, CN, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl or heteroaryl consisting of 5 to 6 atoms, wherein each C is1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are independently optionally substituted by 1, 2 or 3R8Substituted by a group.
In some embodiments, each R is8Independently F, Cl, Br, CN, NO2、OH、NH2、C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C3-C6Cycloalkyl, phenyl, heterocyclyl consisting of 4 to 7 atoms, heteroaryl consisting of 5 to 6 atoms, C1-C6Aminoalkyl radical, C1-C6Alkylamino radical, C1-C6Alkoxy radical, C1-C6Hydroxyalkyl, -NH (C)0-C2Alkylene group) - (C3-C6Cycloalkyl), -NH (C)0-C2Alkylene) -phenyl, -NH (C)0-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), -NH (C)0-C2Alkylene) - (heteroaryl of 5-6 atoms), -N [ (C) 0-C2Alkylene group) - (C3-C6Cycloalkyl radicals]2、-N[(C0-C2Alkylene) -phenyl]2、-N[(C0-C2Alkylene) - (heterocyclic radical consisting of 4-7 atoms)]2、-N[(C0-C2Alkylene) - (heteroaryl of 5-6 atoms)]2、-O(C0-C2Alkylene group) - (C3-C6Cycloalkyl), -O (C)0-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms) or-O (C)0-C2Alkylene) - (5-6 atom-constituting heteroaryl).
In some embodiments, wherein each R isa、Rb、RcAnd RdAre each independently H, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C)1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene) -phenyl or- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) wherein each of C is as defined above1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl, heterocyclyl consisting of 4 to 7 atoms, phenyl, heteroaryl consisting of 5 to 6 atoms, - (C)1-C2Alkylene group) - (C3-C6Cycloalkyl), - (C)1-C2Alkylene) - (heterocyclic group consisting of 4-7 atoms), - (C)1-C2Alkylene radical)-phenyl and- (C)1-C2Alkylene) - (heteroaryl of 5-6 atoms) optionally substituted with 1, 2 or 3 substituents independently selected from F, Cl, Br, CN, N3、OH、NH2、C1-C3Alkyl radical, C1-C3Haloalkyl, C1-C3Alkoxy radical, C1-C3Hydroxyalkyl radical, C1-C3Aminoalkyl or C 1-C3Substituted by alkylamino.
In some embodiments, the present invention relates to compounds, or stereoisomers, tautomers, nitrogen oxides, solvates, metabolites, pharmaceutically acceptable salts, or prodrugs thereof, of one of the following, but in no way limited to these compounds:
Figure BDA0001582369760000251
Figure BDA0001582369760000261
Figure BDA0001582369760000271
stereoisomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds of formula (I) are included within the scope of the present invention unless otherwise indicated.
The compounds of the present disclosure may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention contemplates that all stereoisomeric forms of the compounds of formula (I), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, and mixtures thereof, such as racemic mixtures, are integral to the invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.
The compounds of formula (I) may exist in different tautomeric forms and all such tautomers, as claimed, are included within the scope of the invention.
The compounds of formula (I) may be present in the form of salts. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith. In another embodiment, the salts need not be pharmaceutically acceptable salts and may be intermediates useful in the preparation and/or purification of compounds of formula (I) and/or in the isolation of enantiomers of compounds of formula (I).
Pharmaceutically acceptable acid addition salts may be formed with inorganic and organic acids, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheophylline, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, Polysilonolactates, propionates, stearates, succinates, sulfosalicylates, tartrates, tosylates and trifluoroacetates.
Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, sulfosalicylic acid, and the like.
Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of groups I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (cholinate), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of Pharmaceutical Salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable Salts.
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms.
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl and125I。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present3H、14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13C. the isotopically enriched compounds can be used for metabolic studies (use) 14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages, which may be attributed to greater metabolic stabilityHigh in power. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is to be considered as a substituent of the compound of formula (I). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D 2O, acetone-d6、DMSO-d6Those solvates of (a).
In another aspect, the invention relates to intermediates for the preparation of compounds of formula (I).
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds of formula (I).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle or combination thereof. In another embodiment, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
In another aspect, the invention relates to a method of treating a disease or disorder modulated by one or more protein kinases, such as JAK kinase, FLT3 kinase and Aurora kinase, comprising administering to a mammal an effective amount of a disclosed compound or pharmaceutical composition of the invention. In some embodiments, the disease or disorder is selected from a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition of the invention disclosed herein for the treatment of a disease or disorder selected from a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the preparation of a medicament for the treatment of a disease or disorder selected from a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the preparation of a medicament for modulating the activity of a protein kinase.
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound disclosed herein, or as listed in the examples; and pharmaceutically acceptable adjuvants, excipients, carriers, vehicles or their combination. The amount of compound in the pharmaceutical compositions disclosed herein is that amount which is effective to detect inhibition of a protein kinase in a biological sample or patient.
It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, if appropriate, in the form of a pharmaceutically acceptable derivative thereof. Some non-limiting embodiments of pharmaceutically acceptable derivatives include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any additional adduct or derivative that upon administration to a patient in need thereof provides, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
The pharmaceutical compositions disclosed herein can be prepared and packaged in bulk (bulk) form, wherein a safe and effective amount of the compound of formula (I) can be extracted and then administered to a patient in the form of a powder or syrup. Alternatively, the pharmaceutical compositions disclosed herein can be prepared and packaged in unit dosage forms, wherein each physically discrete unit contains a safe and effective amount of a compound of formula (I). When prepared in unit dosage form, the disclosed pharmaceutical compositions can generally contain, for example, from 0.5mg to 1g, or from 1mg to 700mg, or from 5mg to 100mg of the disclosed compounds.
The term "pharmaceutically acceptable excipient" as used herein refers to a pharmaceutically acceptable material, mixture or vehicle, which is compatible with the dosage form or pharmaceutical composition to be administered. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when mixed to avoid interactions that would substantially reduce the efficacy of the disclosed compounds and that would result in a pharmaceutical composition that is not pharmaceutically acceptable when administered to a patient. Furthermore, each excipient must be pharmaceutically acceptable, e.g., of sufficiently high purity.
Suitable pharmaceutically acceptable excipients will vary depending on the particular dosage form selected. In addition, pharmaceutically acceptable excipients may be selected for their specific function in the composition. For example, certain pharmaceutically acceptable excipients may be selected that facilitate the production of a uniform dosage form. Certain pharmaceutically acceptable excipients may be selected which may aid in the manufacture of stable dosage forms. Certain pharmaceutically acceptable excipients that facilitate carrying or transporting the disclosed compounds from one organ or portion of the body to another organ or portion of the body when administered to a patient may be selected. Certain pharmaceutically acceptable excipients may be selected that enhance patient compliance.
Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants and buffers. The skilled artisan will recognize that certain pharmaceutically acceptable excipients may provide more than one function, and may provide alternative functions, depending on how much of the excipient is present in the formulation and which other excipients are present in the formulation.
The skilled person is knowledgeable and skilled in the art to enable them to select suitable amounts of suitable pharmaceutically acceptable excipients for use in the present invention. Furthermore, there are a large number of resources available to the skilled person, who describe pharmaceutically acceptable excipients and use them to select suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (The American Pharmaceutical Association and The Pharmaceutical Press).
Various carriers for formulating pharmaceutically acceptable compositions, and well known techniques for their preparation, are disclosed in Remington, The Science and Practice of Pharmacy,21st edition,2005, ed.D.B.Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds.J.Swarbrick and J.C.Boylan, 1988. Annu 1999, Marcel Dekker, New York, The contents of each of which are incorporated herein by reference. Except insofar as any conventional carrier is incompatible with the disclosed compounds, such as by producing any undesirable biological effect or interacting in a deleterious manner with any other ingredient in a pharmaceutically acceptable composition, its use is contemplated as falling within the scope of the present invention.
The pharmaceutical compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some commonly used methods in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company).
Thus, in another aspect, the invention relates to a process for preparing a pharmaceutical composition comprising a compound of the present disclosure and a pharmaceutically acceptable adjuvant, excipient, carrier, vehicle, or combination thereof, which process comprises admixing the ingredients. Pharmaceutical compositions comprising the disclosed compounds may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The compounds disclosed herein are generally formulated in a dosage form suitable for administration to a patient by a desired route. For example, dosage forms include those suitable for the following routes of administration: (1) oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) parenteral administration, such as sterile solutions, suspensions, and reconstituted powders; (3) transdermal administration, such as transdermal patches; (4) rectal administration, e.g., suppositories; (5) inhalation, such as aerosols, solutions, and dry powders; and (6) topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.
In one embodiment, the compounds disclosed herein may be formulated in oral dosage forms. In another embodiment, the compounds disclosed herein may be formulated in an inhalation dosage form. In another embodiment, the compounds disclosed herein can be formulated for nasal administration. In yet another embodiment, the compounds disclosed herein can be formulated for transdermal administration. In yet another embodiment, the compounds disclosed herein may be formulated for topical administration.
The pharmaceutical compositions provided by the present invention may be provided as compressed tablets, milled tablets, chewable lozenges, fast-dissolving tablets, double-compressed tablets, or enteric-coated, sugar-coated or film-coated tablets. Enteric coated tablets are compressed tablets coated with a substance that is resistant to the action of gastric acid but dissolves or disintegrates in the intestine, thereby preventing the active ingredient from contacting the acidic environment of the stomach. Enteric coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalate. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can help to mask unpleasant tastes or odors and prevent oxidation of the tablet. Film-coated tablets are compressed tablets covered with a thin layer or film of a water-soluble substance. Film coatings include, but are not limited to, hydroxyethyl cellulose, sodium carboxymethyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coatings are endowed with the same general characteristics as sugar coatings. A tabletted tablet is a compressed tablet prepared over more than one compression cycle, including a multi-layer tablet, and a press-coated or dry-coated tablet.
Tablet dosage forms may be prepared from the active ingredient in powder, crystalline or granular form, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled release polymers, lubricants, diluents and/or colorants. Flavoring and sweetening agents are particularly useful in forming chewable tablets and lozenges.
The pharmaceutical composition provided by the present invention may be provided in soft or hard capsules, which may be prepared from gelatin, methylcellulose, starch or calcium alginate. The hard gelatin capsules, also known as Dry Fill Capsules (DFC), consist of two segments, one inserted into the other, thus completely encapsulating the active ingredient. Soft Elastic Capsules (SEC) are soft, spherical shells, such as gelatin shells, which are plasticized by the addition of glycerol, sorbitol or similar polyols. The soft gelatin shell may contain a preservative to prevent microbial growth. Suitable preservatives are those as described herein, including methyl and propyl parabens, and sorbic acid. The liquid, semi-solid and solid dosage forms provided by the present invention may be encapsulated in a capsule. Suitable liquid and semi-solid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils or triglycerides. Capsules containing such solutions may be as described in U.S. patent nos.4,328,245; 4,409,239 and 4,410,545. The capsules may also be coated as known to those skilled in the art to improve or maintain dissolution of the active ingredient.
The pharmaceutical compositions provided herein may be provided in liquid and semi-solid dosage forms, including emulsions, solutions, suspensions, elixirs and syrups. Emulsions are two-phase systems in which one liquid is dispersed throughout another in the form of globules, which can be either oil-in-water or water-in-oil. Emulsions may include pharmaceutically acceptable non-aqueous liquids and solvents, emulsifiers and preservatives. Suspensions may include a pharmaceutically acceptable suspending agent and a preservative. The aqueous alcoholic solution may comprise pharmaceutically acceptable acetals, such as di (lower alkyl) acetals of lower alkyl aldehydes, e.g. acetaldehyde diethyl acetal; and water-soluble solvents having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, hydroalcoholic solutions. Syrups are concentrated aqueous solutions of sugars, such as sucrose, and may also contain preservatives. For liquid dosage forms, for example, a solution in polyethylene glycol may be diluted with a sufficient amount of a pharmaceutically acceptable liquid carrier, such as water, for precise and convenient administration.
Other useful liquid and semi-solid dosage forms include, but are not limited to, those comprising the active ingredients provided herein and a secondary mono-or poly-alkylene glycol, including: 1, 2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, where 350, 550, 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further include one or more antioxidants, such as Butylated Hydroxytoluene (BHT), Butylated Hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
Dosage unit formulations for oral administration may be microencapsulated, where appropriate. They may also be prepared as extended or sustained release compositions, for example by coating or embedding the particulate material in a polymer, wax or the like.
The oral pharmaceutical composition provided by the invention can also be provided in the form of liposome, micelle, microsphere or nano system. Micellar dosage forms can be prepared using the methods described in U.S. Pat. No.6,350,458.
The pharmaceutical compositions provided herein can be provided as non-effervescent or effervescent granules and powders for reconstitution into liquid dosage forms. Pharmaceutically acceptable carriers and excipients used in non-effervescent granules or powders may include diluents, sweeteners and wetting agents. Pharmaceutically acceptable carriers and excipients used in effervescent granules or powders may include organic acids and sources of carbon dioxide.
Coloring and flavoring agents may be used in all of the above dosage forms.
The disclosed compounds may also be conjugated to soluble polymers as targeted drug carriers. Such polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol or polyoxyethylene polylysine substituted with palmitoyl residues. In addition, the disclosed compounds may be combined with a class of biodegradable polymers used in achieving controlled release of a drug, such as polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and crosslinked or amphiphilic block copolymers of hydrogels.
The pharmaceutical compositions provided by the present invention may be formulated into immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed release forms.
The pharmaceutical compositions provided by the present invention may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
The pharmaceutical compositions provided by the present invention may be administered parenterally by injection, infusion or implantation for local or systemic administration. Parenteral administration as used herein includes intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration.
The pharmaceutical compositions provided herein can be formulated in any dosage form suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems and solid forms suitable for solution or suspension in a liquid prior to injection. Such dosage forms may be prepared according to conventional methods known to those skilled in The art of pharmaceutical Science (see Remington: The Science and Practice of Pharmacy, supra).
Pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives to inhibit microbial growth, stabilizers, solubility enhancers, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, thickening agents, pH adjusting agents, and inert gases.
Suitable aqueous carriers include, but are not limited to: water, saline, normal saline or Phosphate Buffered Saline (PBS), sodium chloride injection, Ringers injection, isotonic glucose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and the medium chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1, 3-butanediol, liquid polyethylene glycols (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerol, N-methyl-2-pyrrolidone, N-dimethylacetamide, and dimethylsulfoxide.
Suitable antimicrobial agents or preservatives include, but are not limited to, phenol, cresol, mercurial, benzyl alcohol, chlorobutanol, methyl and propyl parabens, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl and propyl parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerol and glucose. Suitable buffers include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcellulose, hydroxypropylmethylcellulose and polyvinylpyrrolidone. Suitable emulsifiers include those described herein, including polyoxyethylene sorbitan monolaurate. Polyoxyethylene sorbitan monooleate 80 and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to, EDTA. Suitable pH adjusters include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl -beta-cyclodextrin, sulfobutyl ether-beta-cyclodextrin and sulfobutyl ether 7-beta-cyclodextrin (b) ((b))
Figure BDA0001582369760000311
CyDex,Lenexa,KS)。
The pharmaceutical compositions provided herein may be formulated for single or multiple dose administration. The single dose formulations are packaged in ampoules, vials or syringes. The multi-dose parenteral formulation must contain a bacteriostatic or fungistatic concentration of the antimicrobial agent. All parenteral formulations must be sterile, as is known and practiced in the art.
In one embodiment, the pharmaceutical composition is provided as a ready-to-use sterile solution. In another embodiment, the pharmaceutical compositions are provided as sterile dried soluble products, including lyophilized powders and subcutaneous injection tablets, which are reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a ready-to-use sterile suspension. In yet another embodiment, the pharmaceutical composition is formulated as a sterile, dry, insoluble product that is reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a sterile emulsion ready for use.
The disclosed pharmaceutical compositions may be formulated into immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed-release forms.
The pharmaceutical composition may be formulated as a suspension, solid, semi-solid, or thixotropic liquid for depot administration for implantation. In one embodiment, the disclosed pharmaceutical compositions are dispersed in a solid internal matrix surrounded by an outer polymeric membrane that is insoluble in body fluids but allows diffusion therethrough of the active ingredient in the pharmaceutical composition.
Suitable internal matrices include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, silicone carbonate copolymers, hydrogels of hydrophilic polymers such as esters of acrylic and methacrylic acid, collagen, crosslinked polyvinyl alcohol, and partially hydrolyzed polyvinyl acetate of the class of copolymers.
Suitable outer polymeric films include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, neoprene, chlorinated polyethylene, polyvinyl chloride, copolymers of chlorinated ethylene and vinyl acetate, vinylidene chloride, ethylene and propylene, ionomers polyethylene terephthalate, butyl rubber chlorohydrin rubber, ethylene/vinyl alcohol copolymers, ethylene/vinyl acetate/vinyl alcohol terpolymers, and ethylene/ethyleneoxyethanol copolymers.
In another aspect, the disclosed pharmaceutical compositions may be formulated in any dosage form suitable for administration to a patient by inhalation, such as a dry powder, aerosol, suspension, or solution composition. In one embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient as a dry powder. In yet another embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient via a nebulizer. Dry powder compositions for delivery to the lung by inhalation typically comprise a finely powdered compound disclosed herein and one or more finely powdered pharmaceutically acceptable excipients. Pharmaceutically acceptable excipients that are particularly suitable for use as dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-and polysaccharides. Fine powders may be prepared, for example, by micronization and milling. Generally, the size-reduced (e.g., micronized) compound may pass through a D of about 1 to 10 microns50Values (e.g., measured by laser diffraction).
Aerosols can be formulated by suspending or dissolving the disclosed compounds in a liquefied propellant. Suitable propellants include chlorinated hydrocarbons, hydrocarbons and other liquefied gases. Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134a), 1-difluoroethane (HFA-152a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane and pentane. Aerosols comprising the compounds disclosed herein are typically administered to a patient via a Metered Dose Inhaler (MDI). Such devices are known to those skilled in the art
The aerosol may contain additional pharmaceutically acceptable excipients that may be used by MDIs, such as surfactants, lubricants, co-solvents, and other excipients, to improve the physical stability of the formulation, to improve valve characteristics, to improve solubility, or to improve taste.
Pharmaceutical compositions suitable for transdermal administration may be prepared as discrete patches intended to remain in intimate contact with the epidermis of the patient for an extended period of time. For example, the active ingredient may be delivered from a patch agent by iontophoresis, as generally described in Pharmaceutical Research,3(6),318 (1986).
Pharmaceutical compositions suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For example, ointments, creams and gels may be formulated with a water or oil base, and suitable thickeners and/or gelling agents and/or solvents. Such bases may include, water, and/or oils such as liquid paraffin and vegetable oils (e.g., peanut oil or castor oil), or solvents such as polyethylene glycol. Thickeners and gelling agents used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycol, lanolin, beeswax, carbopol and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifiers.
Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
Powders for external use may be formed in the presence of any suitable powder base, for example talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base containing one or more dispersing agents, solubilising agents, suspending agents or preservatives.
Topical formulations may be administered by application to the affected area one or more times per day; an occlusive dressing covering the skin is preferably used. Adhesive depot systems allow for continuous or extended administration.
For treatment of the eye, or other organs such as the mouth and skin, the composition may be applied as a topical ointment or cream. When formulated as an ointment, the disclosed compounds may be used with a paraffinic or water soluble ointment base. Alternatively, the disclosed compounds may be formulated as a cream with an oil-in-water cream base or an oil-in-water base.
Use of the Compounds and compositions of the invention
The present invention provides methods of using the disclosed compounds and pharmaceutical compositions for treating, preventing, or ameliorating one or more symptoms of a disease or disorder that is mediated by or otherwise affected by the behavior of one or more protein kinases, such as JAK kinases (including JAK1, JAK2, JAK3, and TYK2 kinases), FLT3 kinase (also known as FLK-2) or Aurora kinases (including Aurora-A, Aurora-B and Aurora C), or diseases or disorders that are mediated or otherwise affected by the behavior of one or more protein kinases, such as JAK kinases (including JAK1, JAK2, JAK3, and TYK2 kinases), FLT3 kinase (also known as FLK-2), or Aurora kinases (including Aurora-A, Aurora-B and Aurora C).
The JAK kinase can be a wild-type and/or a mutation of JAK1, JAK2, JAK3, or TYK2 kinase.
In one embodiment, the present invention provides a class of compounds disclosed herein or pharmaceutical compositions comprising a compound disclosed herein for treating, preventing or ameliorating one or more symptoms of a disease or disorder mediated or otherwise affected by inappropriate JAK1 kinase behavior or a disease or disorder mediated or otherwise affected by inappropriate JAK1 kinase behavior. In another embodiment, the disease, disorder, or one or more symptoms of the disease or disorder is associated with inappropriate JAK2 kinase behavior. In yet another embodiment, the disease, disorder, or one or more symptoms of the disease or disorder are associated with inappropriate JAK3 kinase behavior.
In some embodiments, the invention provides a class of compounds disclosed herein or pharmaceutical compositions comprising a compound disclosed herein for use in treating, preventing or ameliorating one or more symptoms of a disease or disorder mediated or otherwise affected by inappropriate FLT3 kinase behavior or a disease or disorder mediated or otherwise affected by inappropriate FLT3 kinase behavior.
In some embodiments, the invention provides a class of compounds disclosed herein or pharmaceutical compositions comprising a compound disclosed herein for use in treating, preventing, or ameliorating one or more symptoms of a disease or disorder mediated or otherwise affected by inappropriate Aurora-a kinase behavior or a disease or disorder mediated or otherwise affected by inappropriate Aurora-a kinase behavior. In other embodiments, the disease, disorder, or one or more symptoms of the disease or disorder is associated with inappropriate Aurora-B kinase behavior. In some embodiments, the disease, disorder, or one or more symptoms of the disease or disorder are associated with inappropriate Aurora C kinase activity.
By "inappropriate JAK kinase behavior" is meant JAK kinase behavior that occurs in a particular patient that deviates from normal JAK kinase behavior. Inappropriate JAK kinase behavior can be expressed in the form of, for example, abnormal increases in activity, or deviations in the time point and control of JAK kinase behavior. This inappropriate kinase behavior results, for example, from inappropriate or uncontrolled behavior caused by overexpression or mutation of protein kinases. Accordingly, the present invention provides methods of treating these diseases and disorders.
Consistent with the above description, such diseases or disorders include, but are not limited to: myeloproliferative diseases, such as polycythemia vera (PCV), essential thrombocythemia, Idiopathic Myelofibrosis (IMF); leukemias, e.g., myeloid leukemias including Chronic Myeloid Leukemia (CML), imatinib-resistant CML forms, Acute Myeloid Leukemia (AML) and subtypes of AML, acute megakaryoblastic leukemia (AMKL); lymphoproliferative diseases such as Acute Lymphoblastic Leukemia (ALL) and myeloma; cancers include head and neck cancer, prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, brain tumor, pancreatic cancer, and renal cancer; and inflammatory diseases or disorders associated with immune dysfunction, immunodeficiency, immune modulation, autoimmune diseases, tissue transplant rejection, graft-versus-host disease, wound healing, kidney disease, multiple sclerosis, thyroiditis, type I diabetes, sarcoidosis, psoriasis, allergic rhinitis, inflammatory bowel disease including crohn's disease and Ulcerative Colitis (UC), Systemic Lupus Erythematosus (SLE), arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, asthma and Chronic Obstructive Pulmonary Disease (COPD), and dry eye syndrome (or keratoconjunctivitis sicca (KCS)).
In one aspect, the present invention provides a class of compounds disclosed herein or pharmaceutical compositions comprising a compound disclosed herein for use in the prevention and/or treatment of a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection in a mammal (including a human).
In another aspect, the present invention provides a method of treating a mammal suffering from or at risk of suffering from a disease disclosed herein, comprising administering a condition treating effective amount or a condition preventing effective amount of one or more of the pharmaceutical compositions or compounds disclosed herein.
In another aspect, provided herein is a method of treating a mammal suffering from or at risk of suffering from a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection.
In one method of therapeutic aspects, the invention provides methods of treating and/or preventing a mammal susceptible to or suffering from a proliferative disease comprising administering a therapeutically effective amount or a prophylactically effective amount of one or more of the pharmaceutical compositions or compounds disclosed herein.
In another aspect, provided herein is a class of compounds disclosed herein for use in the treatment and/or prevention of a proliferative disease.
In another aspect, provided herein is a class of compounds disclosed herein, or a pharmaceutical composition comprising a compound disclosed herein, for use in the preparation of a medicament for the treatment or prevention of a proliferative disease.
In particular examples, the proliferative disease is selected from cancer (e.g., a solid tumor such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocythemia, myelofibrosis, leukemia (e.g., AML, CML, ALL or CLL), and multiple myeloma.
In another aspect, provided herein is a method of treating and/or preventing a mammal susceptible to or suffering from an autoimmune disease, the method comprising administering a therapeutically effective amount or a prophylactically effective amount of one or more of the pharmaceutical compositions or compounds disclosed herein. .
In another aspect, provided herein is a class of compounds disclosed herein for use in the treatment and/or prevention of autoimmune diseases.
In another aspect, provided herein is a class of compounds disclosed herein, or a pharmaceutical composition comprising a compound disclosed herein, for use in the preparation of a medicament for treating or preventing an autoimmune disease.
In a particular embodiment, the autoimmune disease is selected from COPD, asthma, systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, sjogren's syndrome, psoriasis, type I diabetes and inflammatory bowel disease.
In another aspect, provided herein is a method of treating and/or preventing a mammal susceptible to or suffering from an allergic disease, the method comprising administering a therapeutically effective amount or a prophylactically effective amount of one or more of the pharmaceutical compositions or compounds disclosed herein. In a particular embodiment, the allergic disease is selected from respiratory allergic disease, sinusitis, eczema and measles, food allergy and insect venom allergy.
In another aspect, provided herein is a class of compounds disclosed herein for use in the treatment and/or prevention of allergic diseases.
In another aspect, provided herein is a class of compounds disclosed herein, or a pharmaceutical composition comprising a compound disclosed herein, for use in the preparation of a medicament for the treatment or prevention of an allergic disease.
In a particular embodiment, the allergic disease is selected from respiratory allergic disease, sinusitis, eczema and measles, food allergy and insect venom allergy.
In another aspect, provided herein is a method of treating and/or preventing a mammal susceptible to or suffering from an inflammatory disease, comprising administering a therapeutically effective amount or a prophylactically effective amount of one or more of the pharmaceutical compositions or compounds disclosed herein.
In another aspect, provided herein is a class of compounds disclosed herein for use in the treatment and/or prevention of an inflammatory disease.
In another aspect, provided herein is a class of compounds disclosed herein, or a pharmaceutical composition comprising a compound disclosed herein, for use in the preparation of a medicament for the treatment or prevention of an inflammatory disease.
In particular embodiments, the inflammatory disease is selected from inflammatory bowel disease, crohn's disease, rheumatoid arthritis, juvenile arthritis, and psoriatic arthritis.
In another aspect, provided herein is a method of treating and/or preventing a mammal susceptible to or suffering from transplant rejection comprising administering a therapeutically effective amount or a prophylactically effective amount of one or more of the pharmaceutical compositions or compounds disclosed herein.
In another aspect, provided herein is a class of compounds disclosed herein for use in the treatment and/or prevention of transplant rejection.
In another aspect, provided herein is a class of compounds disclosed herein, or a pharmaceutical composition comprising a compound disclosed herein, for use in the preparation of a medicament for treating or preventing transplant rejection.
In particular examples, the transplant rejection is organ transplant rejection, tissue transplant rejection, and cell transplant rejection.
In another aspect, provided herein is a class of compounds disclosed herein for use as a medicament, particularly for use as a medicament in the treatment and/or prevention of the aforementioned diseases. Also provided is the use of a compound disclosed herein for the manufacture of a medicament for the treatment and/or prevention of the aforementioned diseases.
One particular embodiment of the present methods comprises administering to a subject having inflammation an effective amount of a compound disclosed herein for a time sufficient to reduce the level of inflammation in the subject, and preferably to stop the progression of the inflammation. Particular embodiments of the method comprise administering to a subject suffering from or susceptible to bone rheumatoid arthritis an effective amount of a compound disclosed herein for a time sufficient to reduce or prevent, respectively, inflammation of the joints of said subject, and preferably to stop the progression of said inflammation.
Another particular embodiment of the method comprises administering to a subject having a proliferative disease an effective amount of a compound of the disclosure for a time sufficient to reduce the level of the proliferative disease in the subject and preferably to stop the progression of the proliferative disease. Particular embodiments of the method comprise administering to a subject having cancer an effective amount of a compound disclosed herein for a time sufficient to reduce or prevent, respectively, the signs of cancer in said subject, and preferably to stop the progression of said cancer.
Combination therapy
The compounds of the present invention may be administered as the sole active agent or may be administered in combination with other therapeutic agents, including other compounds that have the same or similar therapeutic activity and are identified as safe and effective for such combination administration.
In one aspect, the invention provides a method of treating, preventing or ameliorating a disease or condition comprising administering a safe and effective amount of a combination comprising a compound of the disclosure and one or more therapeutically active agents. In one embodiment, the combination comprises one or two additional therapeutic agents.
Examples of other therapeutic agents include, but are not limited to: anti-cancer agents, including chemotherapeutic agents and antiproliferative agents; an anti-inflammatory agent; and an immunomodulator or immunosuppressant.
In another aspect, the invention provides products comprising a compound of the invention and at least one other therapeutic agent, formulated for simultaneous, separate or sequential administration in therapy. In some embodiments, the treatment is for a disease or condition mediated by one or more protein kinase, such as JAK kinase, FLT3 kinase, or Aurora kinase activity. The products provided by the combined preparation include compositions comprising a compound disclosed herein and other therapeutic agent in the same pharmaceutical composition, or in different forms, e.g., a kit.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound disclosed herein and one or more additional therapeutic agents. In one embodiment, the pharmaceutical composition may comprise a pharmaceutically acceptable excipient as described above.
In another aspect, the invention provides a kit comprising two or more separate pharmaceutical compositions, wherein at least one pharmaceutical composition comprises a compound disclosed herein. In one embodiment, the kit comprises means for separately holding the compositions, such as a container, a separate bottle, or a separate foil box. An example of such a kit is a blister pack, which is commonly used for packaging tablets, capsules and the like.
The invention also provides the use of a compound of the invention in the treatment of a disease or condition mediated by protein kinase activity, wherein the patient has been previously (e.g. within 24 hours) treated with another therapeutic agent. The invention also provides the use of other therapeutic agents in the treatment of diseases and conditions mediated by protein kinase, such as JAK kinase, FLT3 kinase and Aurora kinase activity, in which a patient has been previously (e.g. within 24 hours) treated with a compound of the invention.
The compounds disclosed herein may be administered as a single active ingredient or as, for example, an adjuvant, co-administered with other therapeutic agents.
In some embodiments, the additional therapeutic agent comprises a chemotherapeutic agent and/or an antiproliferative agent. Known chemotherapeutic agents include, but are not limited to, other therapeutic or anti-cancer drugs that may be used in combination with the compounds of the present invention, surgery, radiation therapy (a few examples are gamma radiation, neutron beam radiation, electron beam radiation, proton therapy, brachytherapy and systemic radioisotope therapy), endocrine therapy, taxanes (taxol, docetaxel, etc.), platinum derivatives (cisplatin), carboplatin), biological response modifiers (interferons, interleukins), tumor necrosis factor (TNF, TRAIL receptor targets), hyperthermia and cryotherapy, to alleviate any adverse effects Reactive agents (such as antiemetics), and other approved chemotherapeutic agents, including, but not limited to, alkylating drugs (mechlorethamine), chlorambucil (chlorambucil), cyclophosphamide (cyclophosphamide), melphalan (melphalan), ifosfamide (ifosfamide)), antimetabolites (methotrexate, pemetrexed (pemetrexed), etc.), purine antagonists and pyrimidine antagonists (6-mercaptopurine (6-mercaptopridine), 5-fluorouracil (5-fluorouracil), cytarabine (cytarabine), gemcitabine (gemcitabine), spindle inhibitors (vinblastine), vincristine (vinchristine), vinorelbine (vinorelbine), podophyllotoxin (etoposide), irinotecan (irinotecan), phytotoxin (phytotoxin), phytotoxin (etoposide), mitomycin (mitomycin), phytotoxin (phytotoxin), phytotoxin, Lomustine (lomustine)), cell division cycle inhibitors (KSP is through mitotic kinesin inhibitors, CENP-E and CDK inhibitors), enzymes (asparaginase), hormones (tamoxifen), leuprolide (leuprolide), flutamide (flutamide), megestrol (megestrol), dexamethasone (dexamethasone), and the like. Anti-angiogenic agents (avastin, etc.). Mabs (belimumab), brentuximab, cetuximab (cetuximab), gemtuzumab (gemtuzumab), ipilimumab (ipilimumab), ofatumumab, panitumumab (panitumumab), ranibizumab (ranibizumab), rituximab (rituximab), tositumomab (tositumomab), trastuzumab (trastuzumab). Kinase inhibitors (imatinib), sunitinib (sunitinib), sorafenib (sorafenib), erlotinib (erlotinib), gefitinib (gefitinib), dasatinib (dasatinib), nilotinib (nilotinib), lapatinib (lapatinib), crizotinib (crizotinib), ruxolitinib, vemurafenib, vandetanib, pazopanib, and the like). Drugs inhibit or activate cancer pathways such as mTOR, the HIF (hypoxia inducible factor) pathway, and others. A broad forum for cancer treatment http://www.nci.nih.gov/The list of oncology drugs approved by FDA is shown inhttp:// www.fda.gov/cder/cancer/druglist-rame.htmAnd merck handbook, tenth2006, eight edition, all of which are incorporated herein by reference.
In still other embodiments, the compounds of the invention may bind to a signal transduction inhibitor. Signal transduction inhibitors target the EGFR family, such as EGFR, HER-2 and HER-4(Raymond et al, Drugs,2000,60 (supply. l), 15-23; Harari et al, Oncogene,2000,19(53),6102-6114) and their respective ligands. Such agents include, but are in no way limited to, antibody therapies such as trastuzumab (trastuzumab), cetuximab (cetuximab), ipilimumab (ipilimumab) and pertuzumab (pertuzumab). Such therapies also include, but are in no way limited to, small molecule kinase inhibitors such as imatinib (imatinib), sunitinib (sunitinib), sorafenib (sorafenib), erlotinib (erlotinib), gefitinib (gefitinib), dasatinib (dasatinib), nilotinib (nilotinib), lapatinib (lapatinib), crizotinib (crizotinib), ruxolitinib, vemurafenib, zonantanib, papanib, afatinib (afatinib), amuvanib, axitinib (axitinib), bosutinib (bosutinib), brivatinib, canertinib, cabozantinib, celandinib, cediranib (cediranib), daraflavicularinib, dasatinib, coritinib, coriertinib, saratinib, coriertinib (coridinib), coriertinib, cori, et al, j.med.chem.2008,51,5522), BZE235, and so forth.
In some embodiments, the compounds disclosed herein may also be co-administered with other drugs. Such other drugs include, immunosuppressive agents, immunomodulatory agents, other anti-inflammatory agents, such as drugs used to treat or prevent acute or chronic rejection of allo-or xenografts, inflammatory, autoimmune diseases; or chemotherapeutic agents, such as malignant cell antiproliferative agents. For example, the compounds disclosed herein may be combined with the following active ingredients: calcineurin inhibitors, such as cyclosporin a or FK 506; mTOR inhibitors, such as rapamycin, 40-O- (2-hydroxyethyl) -rapamycin, CCI779, ABT578, AP23573, TAFA-93, biolimus-7 or biolimus-9; ascomycins with immunosuppressive properties, such as ABT-281, ASM981, etc.; a corticosteroid; cyclophosphamide; azathioprine; methotrexate; leflunomide; mizoribine; mycophenolic acid or salt; mycophenolate mofetil; 15-deoxyspergualin or an immunosuppressive homolog, analog or derivative thereof; PKC inhibitors, such as those described in WO 02/38561 or WO 03/82859, e.g., the compounds of examples 56 or 70; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40, CD45, CD52, CD58, CD80, CD86 or ligands thereof; other immunomodulatory compounds, such as recombinant binding molecules having at least a portion of the extracellular domain of CTLA4 or mutants thereof, such as at least an extracellular portion of CTLA4 linked to a non-CTLA 4 protein sequence or mutants thereof, such as CTLA4Ig (e.g., designated ATCC 68629) or mutants thereof, such as LEA 29Y; adhesion molecule inhibitors, such as LFA-1 antagonists, ICAM-1 or-3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or chemotherapeutic agents, such as paclitaxel, gemcitabine, cisplatin, doxorubicin, or 5-fluorouracil; or an anti-infective agent.
Where the compounds disclosed herein are administered in combination with other immunotherapeutic/immunomodulator, anti-inflammatory, chemotherapeutic or anti-infective therapies, the dosage of the immunosuppressant, immunomodulator, anti-inflammatory, chemotherapeutic or anti-infective compound administered in combination will, of course, vary depending on the type of combination employed, e.g., whether it is a steroid or calcineurin inhibitor, the particular drug employed, the condition being treated, etc.
In one aspect, the invention provides a composition comprising a compound of the disclosure and beta2-a combination of adrenergic receptor agonists. Beta is a2Examples of-adrenoceptor agonists include salmeterol, salbutamol, formoterolSalmeterol, fenoterol, carmoterol, eltanoterol, naminterol, clenbuterol, pirbuterol, flubuterol, reproterol, prometrol, indacaterol, terbutaline, and salts thereof, such as the xinafoate (1-hydroxy-2-naphthoate) salt of salmeterol, the sulfate or free base of salbutamol, or the fumarate salt of formoterol. In one embodiment, the long-acting β is2-adrenergic receptor agonists, e.g. preferably, are compounds which provide effective bronchodilation for 12 hours or more.
β2-the adrenoceptor agonist may form a salt form with a pharmaceutically acceptable acid. The pharmaceutically acceptable acid is selected from the group consisting of sulfuric acid, hydrochloric acid, fumaric acid, hydroxynaphthoic acid (e.g., 1-or 3-hydroxy-2-naphthoic acid), cinnamic acid, substituted cinnamic acids, triphenylacetic acid, sulfamic acid, sulfanilic acid, 3- (1-naphthyl) acrylic acid, benzoic acid, 4-methoxybenzoic acid, 2-or 4-hydroxybenzoic acid, 4-chlorobenzoic acid, and 4-phenylbenzoic acid.
In another aspect, the invention provides a combination comprising a compound of the disclosure and a corticosteroid. Suitable corticosteroids are those that are administered orally and inhaled, and prodrugs thereof that have anti-inflammatory activity. Examples include methylprednisolone, prednisolone (prednisolone), dexamethasone (dexamethasone), fluticasone propionate (fluticasone propionate), S-fluoromethyl 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-17 α - [ (4-methyl-1, 3-thiazole-5-carbonyl) oxy ] -3-oxo-androsta-1, 4-diene-17 β -thiocarboxylate, S-fluoromethyl 6 α,9 α -difluoro-17 α - [ (2-furancarbonyl) oxy ] -11 β -hydroxy-16 α -methyl-3-oxo-androsta-1, 4-diene-17 β -thiocarboxylate (fluticasone furoate), 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-3-oxo-17 α -propionyloxy-androsta-1, 4-diene-17 β -carbothioic acid S- (2-oxo-tetrahydrofuran-3S-yl) ester, 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-3-oxo-17 α - (2,2,3, 3-tetramethylcyclopropylcarbonyl) oxy-androsta-1, 4-diene-17 β -carbothioic acid S-cyanomethyl ester and 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-17 α - (1-ethylcyclopropylcarbonyl) oxy-3-oxo-androsta-1, s-fluoromethyl 4-diene-17 β -thiocarboxylate, beclomethasone esters (such as 17-propionate or 17, 21-dipropionate), budesonide (budesonide), flunisolide (flunisolide), mometasone esters (such as mometasone furoate), triamcinolone acetonide (triamcinolone acetonide), rofleponide (rofleponide), ciclesonide (ciclesonide) (16 α,17- [ [ (R) -cyclohexylmethylene ] bis (oxy) ] -11 β, 21-dihydroxy-pregna-1, 4-diene-3, 20-dione), butocort propionate (butixocort propionate), RPR-106541 and ST-126. Preferred corticosteroids include fluticasone propionate (fluticasone propionate), 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-17 α - [ (4-methyl-1, 3-thiazole-5-carbonyl) oxy ] -3-oxo-androsta-1, 4-diene-17 β -carbothioic acid S-fluoromethyl ester, 6 α,9 α -difluoro-17 α - [ (2-furancarbonyl) oxy ] -11 β -hydroxy-16 α -methyl-3-oxo-androsta-1, 4-diene-17 β -carbothioic acid S-fluoromethyl ester, 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-3-oxo-17 α - (2,2,3, 3-tetramethylcyclopropylcarbonyl) oxy-androsta-1, 4-diene-17 β -carbothioic acid S-cyanomethyl ester and 6 α,9 α -difluoro-11 β -hydroxy-16 α -methyl-17 α - (1-methylcyclopropylcarbonyl) oxy-3-oxo-androsta-1, 4-diene-17 β -carbothioic acid S-fluoromethyl ester. In some embodiments, the corticosteroid is S-fluoromethyl 6 α,9 α -difluoro-17 α - [ (2-furancarbonyl) oxy ] -11 β -hydroxy-16 α -methyl-3-oxo-androsta-1, 4-diene-17 β -thiocarboxylate.
In another aspect, the invention provides a combination comprising a compound of the disclosure and a non-steroidal GR agonist. Non-steroidal compounds with glucocorticoid agonistic activity that are selective for transcriptional inhibition (as compared to transcriptional activation) and are useful in combination therapy include those encompassed by the following patents: WO 03/082827, WO 98/54159, WO 04/005229, WO 04/009017, WO 04/018429, WO 03/104195, WO 03/082787, WO 03/082280, WO 03/059899, WO 03/101932, WO 02/02565, WO 01/16128, WO 00/66590, WO 03/086294, WO 04/026248, WO 03/061651 and WO 03/08277. Further non-steroidal compounds are covered in WO 2006/000401, WO 2006/000398 and WO 2006/015870.
In another aspect, the present invention provides a combination comprising a compound of the present disclosure and non-steroidal anti-inflammatory drugs (NSAID's). Examples of NSAID's include cromolyn sodium, nedocromil sodium (nedocromil sodium), Phosphodiesterase (PDE) inhibitors (e.g., theophylline, PDE4 inhibitors, or mixed PDE3/PDE4 inhibitors), leukotriene antagonists, leukotriene synthesis inhibitors (e.g., montelukast), iNOS inhibitors, trypsin and elastase inhibitors, beta-2 integrin antagonists and adenosine receptor agonists or antagonists (e.g., adenosine 2a receptor agonists), cytokine antagonists (e.g., chemokine receptor antagonists, including CCR3 antagonists), cytokine synthesis inhibitors, or 5-lipoxygenase inhibitors. Among them, iNOS (inducible nitric oxide synthase) inhibitors are preferably administered orally. Examples of iNOS inhibitors include those disclosed in WO93/13055, WO 98/30537, WO 02/50021, WO 95/34534 and WO 99/62875. CCR3 inhibitors include those disclosed in WO 02/26722.
In one embodiment, the invention relates to the use of the disclosed compounds in combination with a phosphodiesterase 4(PDE4) inhibitor, particularly in an inhaled dosage form. The PDE 4-specific inhibitors useful in this aspect of the invention may be any compound known to inhibit the PDE4 enzyme or found to be useful as a PDE4 inhibitor, which are only PDE4 inhibitors and are not compounds that inhibit other members of the PDE family, such as PDE3 and PDE 5. The compounds include cis-4-cyano-4- (3-cyclopentyloxy-4-methoxyphenyl) cyclohexane-1-carboxylic acid, 2-carbomethoxy-4-cyano-4- (3-cyclopropylmethoxy-4-difluoromethoxyphenyl) cyclohexan-1-one and cis- [ 4-cyano-4- (3-cyclopropylmethoxy-4-difluoromethoxyphenyl) cyclohexan-1-ol ]; also included are cis-4-cyano-4- [3- (cyclopropoxy) -4-methoxyphenyl ] cyclohexane-1-carboxylic acid (also known as silox) and salts, esters, prodrugs, or physical forms thereof, which is disclosed in U.S. patent No. 5,552,438, issued 09/03, 1996, which patent and the compounds disclosed therein are incorporated by reference in their entirety.
In another aspect, the present invention provides a combination comprising a compound of the present disclosure and an anticholinergic agent. Examples of anticholinergics are those compounds which act as muscarinic receptor antagonists, in particular those which act as M1 or M3 receptor antagonists, M 1/M3Or M2/M3Dual receptor antagonistsOr M1/M2/M3Compounds which are pan-antagonists of the receptor. Exemplary compounds for inhalation administration include ipratropium (e.g., as bromide, CAS22254-24-6, to
Figure BDA0001582369760000371
Sold under the trade name), oxitropium (e.g., as bromide, CAS 30286-75-0) and tiotropium (e.g., as bromide, CAS 136310-93-5), to
Figure BDA0001582369760000372
Sold under trade name); also of interest are revatoxate (e.g., as the hydrobromide salt, CAS 262586-79-8) and LAS-34273 as disclosed in WO 01/04118. Exemplary compounds for oral administration include pirenzepine (CAS 28797-61-7), darifenacin (CAS 133099-04-4, or its hydrobromide CAS133099-07-7, to
Figure BDA0001582369760000373
Sold as trade name), oxybutynin (CAS 5633-20-5, and
Figure BDA0001582369760000374
sold under the trade name of Tourethrin (CAS 15793-40-5)), tolterodine (CAS 124937-51-5, or its tartrate CAS 124937-52-6, to
Figure BDA0001582369760000375
Sold under the trade name) or otiminium (e.g., as bromide, CAS 26095-59-0, to
Figure BDA0001582369760000376
Sold under the trade name of TrisLoronium chloride (CAS 10405-02-4) and solifenacin (CAS 242478-37-1, or its succinate CAS242478-38-2, Compound YM-905, to
Figure BDA0001582369760000377
Sold under the trade name).
In another aspect, the invention provides a combination comprising a compound disclosed herein and an antagonist of H1. Examples of H1 antagonists include, but are not limited to, amxanthene (amelexanox), sisimizole (astemizole), azatadine (azatadine), azelastine (azelastine), acrivastine (acrivastine), brompheniramine (bropheriramine), cetirizine (cetirizine), levocetirizine (levocetirizine), efletirizine (efletirizine), chlorpheniramine (chlorpheniramine), clemastine (clintine), cyclizine (cyclizine), caristine (carebastine), cyproheptadine (procetyline), carbinoxamine (carbinoxamine), descarboethoxy loratadine (descarboethoxyloratoridine), doxylamine (doxylamine), dimethylhexidipine (mepiquin), clemastine (fenadine (e), clemastine (fenadine), clemastine (e (fenadine), fenadine (fenadine), clemastine (doxylamine), clemastine (fenadine), clemastine (meplate (meperidine), clemastine (e), clemastine (e), clemastine (, Minoxidine (meclizine), norastemizole (norastemizole), olopatadine (olopatadine), piperazines (picumast), pyrilamine (pyrilamine), promethazine (promethazine), terfenadine (terfenadine), tripelennamine (tripelenamine), temastine (temelastine), isobutylazine (trimeprazine), and triprolidine (triprolidine), with cetirizine (cetirizine), levocetirizine (levocetirizine), efletirizine (efletirizine), and fexofenadine (fexofenadine) being preferred. In another embodiment, the invention provides a combination comprising a compound disclosed herein and an antagonist (and/or inverse agonist) of H3. Examples of H3 antagonists include those disclosed in WO 2004/035556 and WO 2006/045416. Other histamine receptor antagonists useful in combination with the disclosed compounds include H4 receptor antagonists (and/or inverse agonists), such as those disclosed in jablonewski et al, j.med.chem.,2003,46: 3957-.
In yet another aspect, the invention provides a pharmaceutical composition comprising a compound of the disclosure, together with a PDE4 inhibitor and beta2-a combination of adrenergic receptor agonists.
In a further aspect, the invention provides a combination comprising a compound disclosed herein, in combination with an anticholinergic and a PDE-4 inhibitor.
The combinations described above may conveniently be presented for use in the form of a pharmaceutical composition and thus a pharmaceutical composition comprising a combination as defined above together with a pharmaceutically acceptable excipient or carrier represents a further aspect of the invention.
The individual compounds of these combinations may be administered sequentially or simultaneously in the form of separate or combined pharmaceutical preparations. In one embodiment, the compound components are administered simultaneously in a combined pharmaceutical formulation. Suitable dosages of known therapeutic agents will be readily understood by those skilled in the art.
Thus, in another aspect, the present invention provides a pharmaceutical composition comprising a compound disclosed herein in combination with another therapeutically active agent.
In some embodiments, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and a chemotherapeutic agent.
In some embodiments, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and an antiproliferative agent.
In one embodiment, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and a phosphodiesterase 4(PDE4) inhibitor.
In another embodiment, the invention provides a pharmaceutical composition comprising a compound of the disclosure in combination with a β 2-adrenoceptor agonist.
In another embodiment, the invention provides a pharmaceutical composition comprising a compound of the disclosure in combination with a corticosteroid.
In another embodiment, the invention provides a pharmaceutical composition comprising a combination of a compound of the present disclosure and a non-steroidal GR agonist.
In another embodiment, the present invention provides a pharmaceutical composition comprising a combination of a compound of the present disclosure and an anticholinergic.
In yet another embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present disclosure in combination with an antihistamine.
In still other embodiments, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and an anti-inflammatory agent.
In still other embodiments, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and an immunomodulator.
In still other embodiments, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and a drug for atherosclerosis.
In still other embodiments, the pharmaceutical compositions provided herein comprise a combination of a compound disclosed herein and a drug for the treatment of pulmonary fibrosis.
In the field of medical oncology, it is common practice to treat cancer patients with a combination of different treatment modalities. In surgical oncology, one or more other co-therapeutic modalities added to the compositions of the invention may be, for example, surgery, radiation therapy, chemotherapy, single transduction inhibitors or modulators (e.g., kinase inhibitors or modulators), and/or monoclonal antibodies.
The compounds disclosed herein may also be advantageously used in combination with other compounds, or in combination with other therapeutic agents, especially antiproliferative agents. Such antiproliferative agents include, but are not limited to, aromatase inhibitors; an antiestrogen; a topoisomerase I inhibitor; a topoisomerase II inhibitor; a microtubule active agent; an alkylating agent; (ii) histone deacetylase inhibitors; compounds that induce a cellular differentiation process; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; an antineoplastic antimetabolite; a platinum compound; compounds that target/reduce protein or lipid kinase activity and other anti-angiogenic compounds; a compound that targets, reduces or inhibits protein or lipid phosphatase activity; gonadorelin agonists; an antiandrogen; methionine aminopeptidase inhibitors; a bisphosphonate; a biological response modifier; an anti-proliferative antibody; heparanase inhibitors; ras oncogenic subtype inhibitors; a telomerase inhibitor; a proteasome inhibitor; agents for treating hematological tumors; compounds that target, decrease or inhibit Flt-3 activity; an Hsp90 inhibitor; temozolomide
Figure BDA0001582369760000381
And calcium folinate.
The term "aromatase inhibitor" as used herein, refers to a compound that inhibits estrogen production, i.e., a compound that inhibits the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to: steroids, in particular atamestane (atamestane), exemestane (exemestane) and formestane (formestane); and, in particular, non-steroids, in particular aminoglutethimide (aminoglutethimide), lotemide (rogethimide), pirglutethimide (pyriglutethimide), trostane (trilostane), testolactone (testolactone), ketoconazole (ketoconazole), fluconazole (vorozole), fadrozole (fadrozole), anastrozole (anastrozole) and letrozole (letrozole). Exemestane can be marketed, e.g. under the trademark Emametan
Figure BDA0001582369760000391
Is administered in the form of (1). Formestane (formestane) is commercially available, e.g. under the trade mark
Figure BDA0001582369760000392
Is administered in the form of (1). Fadrozole is commercially available, e.g., under the trademark fadrozole
Figure BDA0001582369760000393
Is administered in the form of (1). Anastrozole (anastrozole) may be commercially available, for example under the trade mark
Figure BDA0001582369760000394
Figure BDA0001582369760000395
Is administered in the form of (1). Letrozole is commercially available, e.g. under the trade mark
Figure BDA0001582369760000396
Or
Figure BDA0001582369760000397
Is administered in the form of (1). Aminoglutethimide (aminoglutethimide) is commercially available, for example under the trademark Aminoglutethimide
Figure BDA0001582369760000398
Is administered in the form of (1). The combination of the invention comprising aromatase inhibitor chemotherapeutic agents is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
The term "antiestrogen" as used herein, refers to a compound that antagonizes the effects of estrogen at the estrogen receptor level. The term includes, but is not limited to, tamoxifen (tamoxifen), fulvestrant (fulvestrant), raloxifene (raloxifene) and raloxifene hydrochloride (raloxifene hydrochloride). Tamoxifen (tamoxifen) is commercially available, e.g. under the trademark tamoxifen
Figure BDA0001582369760000399
Is administered in the form of (1). Raloxifene hydrochloride is commercially available, for example under the trademark Raloxifene hydrochloride
Figure BDA00015823697600003910
Is administered in the form of (1). Fulvestrant may be in the form of a dosage form as disclosed in US 4,659,516 or may be commercially available, for example under the trade mark fulvestrant
Figure BDA00015823697600003911
Is administered in the form of (1). The present invention includes combinations of antiestrogen chemotherapeutic agents particularly useful for treating estrogen receptor positive tumors, such as breast tumors.
The term "antiandrogen" as used herein refers to any substance capable of inhibiting the biological effects of androgens, including, but not limited to, bicalutamide (trade name)
Figure BDA00015823697600003912
) The dosage form may be prepared according to US patent US 4,636,505.
The term "gonadorelin-type agonist" as used herein includes, but is not limited to abarelix (abarelix), goserelin (gosereli)n) and goserelin acetate. Goserelin is disclosed in US patent 4,100,274 and is commercially available, for example under the trade mark Gosselin
Figure BDA00015823697600003913
Figure BDA00015823697600003914
Is administered in the form of (1). Abarelix (abarelix) can be prepared in a dosage form according to the method disclosed in US patent 5,843,901.
The term "topoisomerase I inhibitor" as used herein includes, but is not limited to, topotecan (topotecan), gimatecan (gimatecan), irinotecan (irinotecan), camptothecin (camptothecan) and analogs thereof, 9-nitrocamptothecin (9-nitrocamptothecin), and the macromolecular camptothecin conjugated compound PNU-166148 (compound a1 in WO 99/17804). Irinotecan can be marketed, e.g. under the trademark irinotecan
Figure BDA00015823697600003915
Is administered in the form of (1). Topotecan is available commercially, e.g., under the trademark TEFLON
Figure BDA00015823697600003916
Is administered in the form of (1).
The term "topoisomerase II inhibitor" as used herein includes, but is not limited to, anthracyclines, such as doxorubicin (doxorubicin), in the form of liposomes, under the trade name Doxorubicin
Figure BDA00015823697600003917
Daunorubicin (daunorubicin); epirubicin (epirubicin); idarubicin (idarubicin); naproxubicin (nemorubicin); anthraquinones mitoxantrone (mitoxantrone) and losoxantrone (losoxantrone); etoposide (etoposide) and teniposide (teniposide), which are podophyllotoxin types. Etoposide can be commercially available, e.g., under the trademark Etoposide
Figure BDA00015823697600003918
Is administered in the form of (1). Teniposide is commercially available, e.g., under the trademark TEOSFor VM 26-
Figure BDA00015823697600003919
Is administered in the form of (1). Doxorubicin is available commercially, e.g., under the trademark doxorubicin
Figure BDA00015823697600003920
Or
Figure BDA00015823697600003921
Figure BDA00015823697600003922
Is administered in the form of (1). Epirubicin is available commercially, e.g. under the trademark epirubicin
Figure BDA00015823697600003923
Is administered in the form of (1). Idarubicin is commercially available, e.g. under the trademark Irpex
Figure BDA00015823697600003924
Is administered in the form of (1). Mitoxantrone is commercially available, e.g. under the trademark Mitraxantrone
Figure BDA00015823697600003925
Is administered in the form of (1).
The term "microtubule active agent" refers to microtubule stabilizing agents, microwave destabilizing agents and microtubule polymerization inhibitors. Including, but not limited to, taxanes such as paclitaxel (paclitaxel) and docetaxel (docetaxel); vinca alkaloids, such as vinblastine (vinblastine), especially vinblastine sulfate, vincristine, especially vinblastine sulfate and vinorelbine (vinorelbine); discodermolides; colchicine; and epothilones and derivatives thereof, such as epothilone B or D or derivatives thereof. Paclitaxel may be commercially available, e.g., under the trademark Taxol
Figure BDA00015823697600003926
Is administered in the form of (1). Docetaxel can be marketed, e.g. under the trademark docetaxel
Figure BDA00015823697600003927
Is administered in the form of (1). Vinblastine sulphate is available commercially, e.g. under the trade name VINBLASTIN
Figure BDA00015823697600003928
Is administered in the form of (1). Vincristine sulfate is commercially available, e.g., under the trademark vinblastine
Figure BDA00015823697600003929
Is administered in the form of (1). Discodermolide is obtainable according to the method disclosed in US patent US 5,010,099. Also included are derivatives of the epothilone class disclosed in WO 98/10121, U.S. Pat. No. 6,194,181, WO 98/25929, WO 98/08849, WO 99/43653, WO 98/22461 and WO 00/31247, with epothilone A and/or B being particularly preferred.
The term "alkylating agent" as used herein includes, but is not limited to, cyclophosphamide (cyclophosphamide), ifosfamide (ifosfamide), melphalan (melphalan) or nitrosourea (nitrosourea, such as BCNU or carmustine). Cyclophosphamide is available commercially, e.g. under the trade mark
Figure BDA0001582369760000401
Figure BDA0001582369760000402
Is administered in the form of (1). Ifosfamide may be commercially available, e.g. under the trade mark ifosfamide
Figure BDA0001582369760000403
Is administered in the form of (1).
The term "histone deacetylase inhibitor" or "HDAC inhibitor" refers to a compound that inhibits histone deacetylase and has antiproliferative activity. Which include the compounds disclosed in WO 02/22577, especially N-hydroxy-3- [4- [ [ (2-hydroxyethyl) [2- (1H-indol-3-yl) ethyl ] -amino ] methyl ] phenyl ] -2E-2-propenamide, N-hydroxy-3- [4- [ [ [2- (2-methyl-1H-indol-3-yl) -ethyl ] -amino ] methyl ] phenyl ] -2E-2-propenamide and pharmaceutically acceptable salts thereof. Particularly included are suberoylanilide hydroxamic acid (SAHA).
The term "antineoplastic antimetabolite"includes, but is not limited to, 5-fluorouracil (5-fluorouracil) or 5-FU; capecitabine (capecitabine); gemcitabine (gemcitabine); DNA demethylating agents such as 5-azacytidine (5-azacytidine) and decitabine (decitabine); methotrexate (methotrexate) and edatrexate (edatrexate); and folic acid antagonists such as pemetrexed (pemetrexed). Capecitabine may be marketed, e.g. under the trademark Capecitabine
Figure BDA0001582369760000404
Is administered in the form of (1). Gemcitabine may be commercially available, e.g. under the trade mark Gemcitabine
Figure BDA0001582369760000405
Figure BDA0001582369760000406
Is administered in the form of (1). The term also includes the monoclonal antibody trastuzumab (trastuzumab), which is commercially available, e.g., under the trademark TETRASTUzumab
Figure BDA0001582369760000407
Is administered in the form of (1).
The term "platinum compound" as used herein includes, but is not limited to, carboplatin (carboplatin), cis-platinum (cis-platinum), cisplatin (cissplatinum) and oxaliplatin (oxaliplatin). Carboplatin can be commercially available, e.g., under the trademark TEFLON
Figure BDA0001582369760000408
Is administered in the form of (1). Oxaliplatin may be marketed, e.g. under the trademark oxaliplatin
Figure BDA0001582369760000409
Is administered in the form of (1).
The term "compound targeting/reducing the activity of a protein or lipid kinase, or the activity of a protein or lipid phosphatase, or other anti-angiogenic compound" as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine inhibitors, or lipid kinase inhibitors, e.g. as described in
a) Compounds that target, decrease or inhibit Platelet Derived Growth Factor Receptor (PDGFR) activity; compounds that target, decrease or inhibit PDGFR activity, particularly compounds that inhibit the PDGF receptor, include N-phenyl-2-pyrimidine-amine derivatives such as imatinib (imatinib), SU101, SU6668, GFB-111, and the like;
b) compounds that target, decrease or inhibit Fibroblast Growth Factor Receptor (FGFR) activity;
c) compounds that target, decrease or inhibit the activity of insulin-like growth factor receptor-1 (IGF-1R); compounds that target, decrease or inhibit IGF-1R activity, particularly compounds that inhibit IGF-1 receptor activity, include those disclosed in patent WO 02/092599;
d) a compound that targets, decreases or inhibits the activity of the Trk receptor tyrosine kinase family;
e) compounds that target, decrease or inhibit the activity of the Axl receptor tyrosine kinase family;
f) compounds that target, decrease or inhibit the activity of the c-Met receptor;
g) compounds that target, decrease or inhibit the activity of Kit/SCFR receptor tyrosine kinases;
h) compounds that target, decrease or inhibit the activity of C-kit receptor tyrosine kinases (part of the PDGFR family); compounds that target, decrease or inhibit the activity of the C-Kit receptor tyrosine kinase family, especially compounds that inhibit the C-Kit receptor, including imatinib (imatinib), and the like;
i) Compounds that target, decrease or inhibit the activity of the c-Abl family and their gene fusion products, such as BCR-Abl kinase; compounds that target, reduce or inhibit c-Abl family members and their gene fusions include N-phenyl-2-pyrimidine-amine derivatives, such as imatinib, PD180970, AG957, NSC 680410, PD173955 from ParkeDavis
j) A compound that targets, reduces or inhibits the activity of a member of the Raf family, a member of the MEK, SRC, JAK, FAK, PDK and Ras/MAPK family, a member of the Pl (3) kinase family, or a member of the Pl (3) kinase-associated kinase family, and/or a member of the cyclin-dependent kinase family (CDK) in protein kinase c (pkc) and silk/threonine kinases; in particular those staurosporine derivatives disclosed in US patent 5,093,330, such as midostaurin (midostaurin); further examples of compounds also include, UCN-01; saffingol (safingol); BAY 43-9006; bryostatin 1; piperacillin (Perifosine); imofosine (llmofosine); RO 318220 and RO 320432; GO 6976; isis 3521; LY333531/LY 379196; isoquinoline compounds, such as those disclosed in WO 00/09495; FTIs; PD 184352; or QAN697 (a P13K inhibitor);
k) Compounds that target, decrease or inhibit the activity of protein tyrosine kinase inhibitors; compounds that target, decrease or inhibit the activity of protein tyrosine kinase inhibitors include imatinib mesylate
Figure BDA00015823697600004010
Or a tyrphostin; the tyrosine phosphorylation inhibitor is preferably low molecular weight (Mr)<1500) A compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the group consisting of benallyldinitriles or S-arylbenpropionitrile or bis-substrate quinolines, further selected from the group consisting of tyrphostin A23/RG-50810, AG 99, tyrphostin AG213, tyrphostin AG 1748, tyrphostin AG 490, tyrphostin B44, tyrphostin B44(+) enantiomer, tyrphostin AG 555, AG 494, tyrphostin AG 556, AG957 and adaphostin (4- { [ (2, 5-dihydroxyphenyl) methyl]Amino } -benzoic acid adamantane esters, NSC 680410, adaphortin); and
I) compounds that target, decrease or inhibit the activity of the epidermal growth factor receptor family (homo-or heterodimers of EGFR, ErbB2, ErbB3, ErbB 4) in receptor tyrosine kinases; compounds which target, reduce or inhibit the epidermal growth factor receptor family are in particular compounds, proteins or antibodies which inhibit members of the EGF receptor family (such as EGF receptor, ErbB2, ErbB3, ErbB4, or substances which bind EGF or EGF-related ligands), in particular compounds, proteins or mabs which are generally or specifically disclosed in the following documents: WO 97/02266 (as example 39), EP 0564409, WO 99/03854, EP 0520722, EP 0566226, EP 0787722, EP 0837063, US 5,747,498, WO 98/10767, WO 97/30034, WO 97/49688 and WO 97/38983, WO 96/30347 (as CP 358774), WO96/33980 (as compound ZD 1839), WO 95/03283 (as compound ZM105180), trastuzumab (herceptin), cetuximab, Iressa, Tarceva, OSI-774, CI-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3, E7.6.3, and the 7H-pyrrolo- [2,3-d ] pyrimidine derivatives disclosed in WO 03/013541.
In addition, anti-angiogenic compounds include compounds with other mechanisms of activity (e.g., not associated with protein or lipid kinase inhibition), such as thalidomide
Figure BDA0001582369760000411
And TNP-470.
The compound that targets, decreases or inhibits protein or lipid kinase activity is a phosphatase-1 inhibitor, a phosphatase 2A inhibitor, a PTEN inhibitor or a CDC25 inhibitor, such as okadaic acid or a derivative thereof.
The compounds that induce the process of cell differentiation are retinoic acid, alpha-, gamma-or delta-tocopherol, alpha-, gamma-or delta-tocotrienol.
The term "cyclooxygenase inhibitors" as used herein includes, but is not limited to, Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acids and derivatives thereof, such as celecoxib
Figure BDA0001582369760000412
Rofecoxib
Figure BDA0001582369760000413
Etoricoxib, valdecoxib, or 5-alkyl-2-arylaminophenylacetic acids, such as 5-methyl-2- (2 '-chloro-6' -fluoroanilino) phenylacetic acid or lumiracoxib
The term "bisphosphonate" as used herein includes, but is not limited to, etidronic acid, clodronic acid, tiludronic acid, pamidronic acid, alendronic acid, ibandronic acid, risedronic acid, and zoledronic acid. Etidronic acid is commercially available, for example under the trade name Etidronic acid
Figure BDA0001582369760000414
Is administered in the form of (1). The chlorophosphonic acids are commercially available, for example under the trade name Chlorophosphonic acid
Figure BDA0001582369760000415
Is administered in the form of (1). Telophosphonic acid is available commercially, for example under the trade name TELUPHOSPHONIC ACID
Figure BDA0001582369760000416
Is administered in the form of (a); pamidronic acid (Pamidronic acid) is commercially available, for example under the trade name adataTM(AREDIATM) Is administered in the form of (a); alendronic acid is available commercially, for example under the trade name
Figure BDA0001582369760000417
Is administered in the form of (a); ibandronic acid is available commercially, for example under the trade name ibandronic acid
Figure BDA0001582369760000418
Is administered in the form of (a); risedronic acid is commercially available, for example under the trade name risedronic acid
Figure BDA0001582369760000419
Figure BDA00015823697600004110
Is administered in the form of (a); zoledronic acid is commercially available, for example under the trade name Zoledronic acid
Figure BDA00015823697600004111
Is administered in the form of (1).
The term "mTOR inhibitor" refers to a compound that inhibits the mammalian target of rapamycin (mTOR), having antiproliferative activity, such as sirolimus (sirolimus,
Figure BDA00015823697600004112
) Everolimus (CERTICAN)TM) CCI-779 and ABT 578.
The term "heparanase inhibitor" as used herein refers to a compound that targets, reduces or inhibits the degradation of heparan sulfate. This term includes, but is not limited to PI-88.
The term "biological response modifier" as used herein refers to lymphokines or interferons, such as interferon gamma.
The term "inhibitor of Ras oncogenic subtype (e.g., H-Ras, K-Ras or N-Ras) as used herein refers to compounds that target, decrease or inhibit Ras oncogenic activity, e.g.," farnesyl transferase inhibitors "such as L-744832, DK8G557 or R115777 (Zarnestra).
The term "telomerase inhibitor" as used herein refers to compounds that target, decrease or inhibit telomerase activity. A compound that targets, reduces or inhibits telomerase activity refers in particular to a compound that inhibits the telomerase receptor, such as, for example, telomerase.
The term "methionine aminopeptidase inhibitor" as used herein refers to a compound that targets, decreases or inhibits the activity of methionine aminopeptidase. Compounds that target, decrease or inhibit methionine aminopeptidase activity include bengamide or derivatives thereof.
The term "proteasome inhibitor" as used herein refers to a compound that targets, decreases or inhibits the activity of the proteasome. Compounds that target, decrease or inhibit proteasome activity include PS-341 and MLN 341.
The term "matrix metalloproteinase inhibitor" or "MMP inhibitor" as used herein includes, but is not limited to, collagen peptide and non-peptide inhibitors, tetracycline derivatives, such as the hydroxamic acid peptide inhibitor batimastat (batimastat) and its oral bioequivalent homolog marimastat (marimastat, BB-2516), primastat (prinomastat, AG3340), metamastat (metastat, NSC 683551), BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ 996.
The term "agent for treating hematological tumors" as used herein includes, but is not limited to, FMS-like tyrosine kinase inhibitors. Compounds that target, decrease or inhibit the activity of FMS-like tyrosine kinase receptor (Flt-3R); interferon, 1-b-D-arabinofuranosyl cytosine (ara-c) and bisufan; and ALK inhibitors, such as compounds that target, decrease, or inhibit anaplastic lymphoma kinase.
Compounds targeting, decreasing or inhibiting the FMS-like tyrosine kinase receptor (Flt-3R) especially refer to compounds, proteins or antibodies inhibiting members of the Flt-3 receptor kinase family, such as PKC412, midostaurin, staurosporine derivatives, SU11248 and MLN 518.
The term "anti-proliferative antibody" as used herein includes, but is not limited to, trastuzumab (HERCEPTIN)TM) trastuzumab-DM 1, erlotinib (TARCEVA)TM) Bevacizumab (AVASTIN)TM) Rituximab (rituximab)
Figure BDA0001582369760000421
PR064553(anti-CD40) and 2C4 antibodies. By antibody is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibody fragments (so long as they have the desired biological activity). For the treatment of Acute Myeloid Leukemia (AML), the disclosed compounds can be used in combination with standard leukemia therapies, especially in combination with therapies for the treatment of AML. In particular, the compounds of the present disclosure may be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs used in the treatment of AML, such as daunorubicin, doxorubicin, Ara-C, VP-16, teniposide, mitoxantrone, idarubicin, carboplatin, and PKC 412.
The compounds disclosed herein may also be advantageously used in combination with other compounds, or in combination with other therapeutic agents, especially other anti-malarial agents. Such antimalarial agents include, but are not limited to, proguanil (proguanil), proguanil (chlorproquine), trimethoprim (trimethoprim), chloroquine (chloroquine), mefloquine (mefloquine), lumefantrine (lumefantrine), atovaquone (atovaquone), pyrimethamine-sulfanilamide (pyrimethamine-sulfadoxine), pyrimethamine-chlorobenzene (pyrimethamine-dapsone), halofantrine (halofantrine), quinine (quinine), quinidine (quinidine), amodiaquine (amodiaquine), amopyroquine (amopyroquine), sulfonamides, artemisinin, arterfine (artelene), artemisia methyl ether, artesunate, primaquine, inhaled NO, L-arginine, predynetriaminol (NO), erythropoietin (rythrone agonist), glitazone, levo-active, and ppa.
The compounds described above, which can be used in combination with the compounds disclosed in the present invention, can be prepared and administered by those skilled in the art according to the methods described in the above documents.
The compounds disclosed herein may also be combined with a therapeutic procedure to enhance the therapeutic effect. For example, hormone therapy or special radiation therapy is administered. The compounds disclosed herein are particularly useful as radiosensitizers, particularly for the treatment of tumors that are weakly sensitive to those radiation treatments.
"combination" means a fixed combination or a kit of parts for the combined administration in the form of a single dosage unit, wherein a compound disclosed in the invention and a combination partner may be administered separately at the same time or may be administered separately within certain time intervals, in particular such that the combination partners show a cooperative, e.g. synergistic, effect. The terms "co-administration" or "co-administration" and the like as used herein are intended to encompass administration of the selected combination partner to a single individual in need thereof (e.g., a patient), and are intended to encompass treatment regimens in which the substances are not necessarily administered by the same route of administration or simultaneously. The term "pharmaceutical combination" as used herein denotes a product obtained by mixing or combining more than one active ingredient and includes both fixed and non-fixed combinations of active ingredients. The term "fixed combination" means that the active ingredients, such as the compounds disclosed herein, and the combination partner are administered to the patient simultaneously, in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g., a compound of the compounds disclosed herein, and the combination partner, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein the mode of administration provides therapeutically effective levels of both compounds in the patient. The latter also applies to cocktail therapies, such as the administration of three or more active ingredients.
Method of treatment
In one embodiment, the presently disclosed methods of treatment comprise administering to a patient in need thereof a safe and effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention. Various embodiments of the present disclosure include methods of treating the above-mentioned diseases by administering to a patient in need thereof a safe and effective amount of a disclosed compound or a pharmaceutical composition comprising a disclosed compound.
In one embodiment, the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds may be administered by any suitable route of administration, including systemic and topical administration. Systemic administration includes oral, parenteral, transdermal and rectal administration. Typical parenteral administration refers to administration by injection or infusion, including intravenous, intramuscular, and subcutaneous injection or infusion. Topical administration includes application to the skin and intraocular, otic, intravaginal, inhalation, and intranasal administration. In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered orally. In another embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered by inhalation. In yet another embodiment, the presently disclosed compounds or compositions comprising the presently disclosed compounds may be administered intranasally.
In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered once or several times at different time intervals over a specified period of time according to a dosing regimen. For example, once, twice, three times or four times daily. In one embodiment, the administration is once daily. In yet another embodiment, the administration is twice daily. The administration may be carried out until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. Suitable dosing regimens for the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds depend on the pharmacokinetic properties of the compound, such as dilution, distribution and half-life, which can be determined by the skilled person. In addition, suitable dosing regimens for the compounds or pharmaceutical compositions comprising the disclosed compounds, including the duration of the regimen, will depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient being treated, the nature of concurrent therapy, the desired therapeutic effect, and other factors within the knowledge and experience of the skilled artisan. Such a skilled artisan will also appreciate that adjustments to the subject's response to the dosage regimen, or the need for changes in the subject's patient over time, may be required.
The compounds disclosed herein may be administered simultaneously, or before or after, one or more other therapeutic agents. The compounds of the invention may be administered separately from the other therapeutic agents, by the same or different routes of administration, or together with them in pharmaceutical compositions.
For an individual of about 50-70kg, the disclosed pharmaceutical compositions and combinations may be in unit dosage form containing from about 1-1000mg, or from about 1-500mg, or from about 1-250mg, or from about 1-150mg, or from about 0.5-100mg, or from about 1-50mg of the active ingredient. The therapeutically effective amount of the compound, pharmaceutical composition or combination thereof will depend on the species, weight, age and condition of the individual, the disease (disorder) or illness (disease) being treated, or the severity thereof. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each active ingredient to prevent, treat or inhibit the progression of the disease (disorder) or condition (disease).
The above cited dose profiles have been demonstrated in vitro and in vivo tests using beneficial mammals (e.g., mice, rats, dogs, monkeys) or isolated organs, tissues and specimens thereof. The compounds disclosed herein are used in vitro in the form of solutions, e.g. aqueous solutions, and also enterally, parenterally, especially intravenously, in vivo, e.g. in the form of suspensions or aqueous solutions.
In one embodiment, a therapeutically effective dose of a compound of the present disclosure is from about 0.1mg to about 2,000mg per day. The pharmaceutical composition thereof should provide a dose of the compound of about 0.1mg to about 2,000 mg. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide from about 1mg to about 2,000mg, from about 10mg to about 1,000mg, from about 20mg to about 500mg, or from about 25mg to about 250mg of the principal active ingredient or a combination of principal ingredients per dosage unit form. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide about 10mg,20mg,25mg,50mg,100mg,250mg,500mg,1000mg or 2000mg of the primary active ingredient.
In addition, the compounds disclosed herein may be administered in the form of a prodrug. In the present invention, a "prodrug" of a disclosed compound is a functional derivative that, when administered to a patient, is ultimately released in vivo. When administering the compounds disclosed herein in the form of a prodrug, one skilled in the art can practice one or more of the following: (a) altering the in vivo onset time of the compound; (b) altering the duration of action of the compound in vivo; (c) altering the in vivo delivery or distribution of the compound; (d) altering the in vivo solubility of the compound; and (e) overcoming side effects or other difficulties faced by the compounds. Typical functional derivatives useful for preparing prodrugs comprise variants of the compounds which are cleaved in vivo either chemically or enzymatically. These variants, which involve the preparation of phosphates, amides, esters, thioesters, carbonates and carbamates, are well known to those skilled in the art.
General synthetic procedure
To illustrate the invention, the following examples are set forth. It is to be understood that the invention is not limited to these embodiments, but is provided as a means of practicing the invention.
In general, the compounds of the present invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
The examples described below, unless otherwise indicated, are all temperatures set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin Haojian Yunyu chemical Co., Ltd, Tianjin Shucheng chemical reagent factory, Wuhan Xin Huayuan scientific and technological development Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaolingyi factory.
The anhydrous tetrahydrofuran, dioxane, toluene and ether are obtained through reflux drying of metal sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, petroleum ether, N-hexane, N, N-dimethylacetamide and N, N-dimethylformamide were used as they were previously dried over anhydrous sodium sulfate.
The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. The glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants.
1HNMR spectra were recorded using a Bruker 300MHz, 400MHz or 600MHz NMR spectrometer.1H NMR Spectrum in CDC13、DMSO-d6、CD3OD or acetone-d6TMS (0ppm) or chloroform (7.26ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (singlets), d (doublets), t (triplets), m (multiplets), br (broad singlets), dd (doublets of doublets), dt (doublets of triplets). Coupling constant in Hertz (Hz)And (4) showing.
The conditions for determining low resolution Mass Spectrometry (MS) data were: agilent 6120 four-stage rod HPLC-M (column model: Zorbax SB-C18, 2.1X 30mm,3.5 micron, 6min, flow rate 0.6 mL/min. mobile phase 5% -95% (CH with 0.1% formic acid)3CN) in (H containing 0.1% formic acid)2O) by electrospray ionization (ESI) at 210nm/254nm, with UV detection.
Pure compounds were detected by UV at 210nm/254nm using Agilent 1260pre-HPLC or Calesep pump 250pre-HPLC (column model: NOVASEP 50/80mm DAC).
The following acronyms are used throughout the invention:
AcOH、HOAc、CH3COOH acetic acid
ACN,MeCN,CH3CN acetonitrile
Boc2Di-tert-butyl O dicarbonate
BOC, Boc tert-butoxycarbonyl
BINAP 1,1 '-binaphthyl-2, 2' -bis (diphenylphosphines)
Bu4NBr tetrabutylammonium bromide
n-BuOH n-butanol
Cbz-Cl benzyl chloroformate
CH2Cl2DCM dichloromethane
CDC13Deuterated chloroform
Cs2CO3Cesium carbonate
DIEA、DIPEA、i-Pr2NEt diisopropylethylamine
DMF dimethyl formamide
DMAP 4-dimethylaminopyridine
DMSO dimethyl sulfoxide
EDC, EDCI 1- (3-dimethylaminopropyl) -3-ethyl-carbodiimide hydrochloride
EDTA ethylene diamine tetraacetic acid
Et3N, TEA Triethylamine
EtOAc, EA ethyl acetate
g
h hours
HATU 2- (7-aza-1H-benzotriazol-1-yl) -1,1,3, 3-tetramethyluronium hexafluorophosphate
HCl hydrochloric acid
HOAT 1-hydroxy-7-azabenzotriazole
K2CO3Potassium carbonate
LDA lithium diisopropylamide
mL, mL
min for
mCPBA m-chloroperoxybenzoic acid
MTBE methyl tert-butyl ether
NaHCO3Sodium bicarbonate
NH4Cl ammonium chloride
M mol/L
Na2CO3Sodium carbonate
Na2SO4Sodium sulfate
NaOH sodium hydroxide
NaH sodium hydride
Na2S2O3Sodium thiosulfate
t-BuONa sodium tert-butoxide
Pd/C Palladium/carbon
Pd(OH)2Palladium hydroxide
Pd(OAc)2Palladium acetate
Pd2(dba)3Tris (dibenzylideneacetone) dipalladium
Pd(dppf)2Cl2[1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
PE Petroleum ether (60-90 ℃ C.)
RT, RT, r.t. Room temperature
Rt Retention time
TsCl tosyl chloride
TFA trifluoroacetic acid
THF tetrahydrofuran
Typical synthetic procedures for preparing the disclosed compounds are shown in schemes 1-5 below. Each Z, Z unless otherwise stated1、R1、R2、R3And R4Having the definitions as described herein; PG (Picture experts group)1And PG2Is a protecting group; p is 0, 1, 2 or 3; q is 0, 1, 2, 3 or 4; d is 0, 1, 2, 3 or 4.
Synthesis scheme 1:
Figure BDA0001582369760000451
Figure BDA0001582369760000461
having the formula(9)Or formula(10)The compounds of the invention of the structures shown can be prepared by general synthetic methods as described in scheme 1, with reference to the examples for specific procedures. Synthesis scheme 1 Synthesis of substituted heteroaryl Compounds under basic conditions(1)Introducing a protecting group to obtain a compound(2). Compound (I)(2)Reacting with LDA and elemental iodine to obtain iodine substituted compound(3). Compound (I)(3)With optionally substituted boron ester derivatives of pyrazoles (4) In the presence of a suitable base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a suitable Pd catalyst (e.g., Pd (OAc))2、Pd(dppf)2Cl2Or Pd2(dba)3Etc.) to produce a compound (A), (B), (C) and (C)5). Compound (A) to (B)5) With optionally substituted heterocyclic compounds(6)In the presence of a base (e.g., triethylamine, diisopropylethylamine), or, in the presence of a suitable base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a Pd catalyst (e.g., Pd (OAc)) 2、Pd(dppf)2Cl2Or Pd2(dba)3Etc.) to produce a compound(7). Compound (I)(7)Sequentially removing two protecting groups in the presence of alkali (such as sodium hydroxide, etc.) and acidic condition (such as trifluoroacetic acid or ethyl acetate solution of hydrogen chloride, etc.) to obtain kinase inhibitor(9). Under appropriate conditions to the compound(9)The introduction of suitable substituent can also obtain another kinase inhibitor(10)
Synthesis scheme 2:
Figure BDA0001582369760000462
having the formula(7)Intermediate compounds of the structures shown can also be prepared by general synthetic methods as depicted in scheme 2, with reference to the examples for specific procedures. In FIG. 2, substituted heteroaryl Compounds: (11) Is oxidized by a suitable oxidizing agent to a compound (12) Followed by an oxide (12) With tetrabutylammonium bromide to obtain bromide (b)13). Under alkaline conditions, to bromide (C)13) Introducing a protecting group to obtain a compound(14)Of a compound(14)Reacting with LDA and elemental iodine to obtain iodine substituted compound(15). Compound (I)(15)With optionally substituted boron ester derivatives of pyrazoles (4) In the presence of a base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a suitable Pd catalyst (e.g., Pd (OAc))2、Pd(dppf)2Cl2Or Pd2(dba)3Etc.) to produce a compound (A), (B), (C) and (C)16). Compound (A) to (B) 16) With optionally substituted heterocyclic compounds(6)In a base such as triethylamine, diisopropylethylamine, or, in a base (such as cesium carbonate, potassium carbonate, sodium carbonate, etc.), and in a suitable Pd catalyst (such as Pd (OAc))2、Pd(dppf)2Cl2Or Pd2(dba)3Etc.) to produce intermediate compounds(7)
Synthesis scheme 3
Figure BDA0001582369760000471
Has the formula (A)21) Or formula (A)22) Compounds of the present invention of the structure shown can be prepared by general synthetic methods as depicted in scheme 3, with reference to the examples for specific procedures. In FIG. 3, the compounds (A), (B), (C)17) And optionally substituted heterocyclic compounds (C:6) Reacting under the action of base (such as triethylamine, diisopropylethylamine, etc.) to produce compound (A)18) Compound (A) to (B)18) And a compound of (A)19) In a reducing agent (e.g. Na)2S2O4Etc.) and polar solvent (such as EtOH, etc.) to obtain compound (A), (B), (C), (20) Then removing the compound (such as trifluoroacetic acid or hydrogen chloride in ethyl acetate) under acidic conditions20) To obtain a protein kinase inhibitor (b)21). Under appropriate conditions to the compound(21)The introduction of suitable substituent can also obtain another kinase inhibitor(22)
Synthesis scheme 4
Figure BDA0001582369760000472
Has the formula (A)26) Or formula (A)27) The disclosed compounds of the present invention of the structure shown can be prepared by the general synthetic methods described in scheme 4, with reference to the examples for specific procedures. In FIG. 4, the compounds (A), (B), (C) 23) And optionally substituted heterocyclic compounds (C:6) Reacting under the action of base (such as triethylamine or diisopropylethylamine, etc.) to produce compound (A)24) Of a compound(24)With optionally substituted boron ester derivatives of pyrazoles (4) In the presence of a suitable base (e.g., cesium carbonate, potassium carbonate or sodium carbonate, etc.), and a suitable Pd catalyst (e.g., Pd (OAc))2、Pd(dppf)2Cl2Or Pd2(dba)3Etc.) to produce a compound (A), (B), (C) and (C)25) Then removing the compound (such as trifluoroacetic acid or hydrogen chloride in ethyl acetate) under acidic conditions25) To obtain a protein kinase inhibitor (b)26). Under appropriate conditions to the compound(26)The introduction of a suitable substituent can also obtain another protein kinase inhibitor(27)
Synthesis scheme 5
Figure BDA0001582369760000481
Has the formula (A)31) Or formula (A)32) The disclosed compounds of the present invention of the structure shown may bePrepared by the general synthetic methods described in FIG. 5, the specific procedures are in reference to the examples. Synthesis scheme 5, Compounds16) With optionally substituted heterocyclic compounds(28)In the presence of a base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a suitable Pd catalyst (e.g., Pd (OAc))2、Pd(dppf)2Cl2Or Pd2(dba)3) Is reacted to generate an intermediate compound(29)Of a compound(29)Sequentially removing two protecting groups in the presence of alkali (such as sodium hydroxide, etc.) and under acidic condition (such as trifluoroacetic acid or ethyl acetate solution of hydrogen chloride, etc.) to obtain protein kinase inhibitor 31). Under appropriate conditions to the compound(31)The introduction of a suitable substituent can also obtain another protein kinase inhibitor(32)
Examples
Example 14- (3-azaspiro [5.5 ]]Undecane-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) -1H- Pyrrolo [2,3-b]Pyridine-5-carboxamides
Figure BDA0001582369760000482
Step 1) 4-chloro-1- (triisopropylsilyl) -1H-pyrrolo [2,3-b]Pyridine compound
To a suspension of sodium hydride (60% [ w/w ] in mineral oil, 2.68g,67.0mmol) in tetrahydrofuran (60mL) at 0 ℃ was added dropwise a solution of 4-chloro-1H-pyrrolo [2,3-b ] pyridine (8.24g,54.0mmol) in tetrahydrofuran (80 mL). After the resulting reaction mixture was stirred at room temperature for 30 minutes, tris (isopropyl) chlorosilane (12.73g,66.0mmol) was further added to the above system. The reaction was stirred at room temperature for 5 hours, and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (100% PE) to give the title compound as a colorless oil (16.68g, 100% yield).
MS(ESI,pos.ion)m/z:309.0[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.16(d,J=5.1Hz,1H),7.35(d,J=3.5Hz,1H),7.08(d,J=5.1Hz,1H),6.68(d,J=3.5Hz,1H),1.88(dq,J=15.0,7.5Hz,3H),1.14(d,J=7.6Hz,18H)。
Step 2) 4-chloro-1- (triisopropylsilyl) -1H-pyrrolo [2,3-b]Pyridine-5-carboxamides
To a solution of 4-chloro-1- (triisopropylsilyl) -1H-pyrrolo [2,3-b ] pyridine (17.55g,56.81mmol) in tetrahydrofuran (120mL) at-78 deg.C was added dropwise a solution of sec-butyllithium in tetrahydrofuran (1.3M,70mL,91.0 mmol). The reaction was continued at-78 ℃ for 1 hour and trimethylsilyl isocyanate (10.15g,83.69mmol) was added to the reaction. After the addition was completed, the reaction mixture was stirred at room temperature for 3.5 hours, and then the reaction was quenched by adding glacial acetic acid (11.92g,198.5mmol) and water (150mL), followed by extraction with ethyl acetate (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) ═ 1/8) to give the title compound as a white solid (13.43g, 67.2% yield).
MS(ESI,pos.ion)m/z:352.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.75(s,1H),7.41(d,J=3.5Hz,1H),6.75(d,J=3.5Hz,1H),6.52(br.s,1H),5.93(br.s,1H),1.86(dq,J=15.0,7.5Hz,3H),1.13(d,J=7.5Hz,18H)。
Step 3) 4-chloro-1H-pyrrolo [2,3-b]Pyridine-5-carboxamides
To a solution of 4-chloro-1- (triisopropylsilyl) -1H-pyrrolo [2,3-b ] pyridine-5-carboxamide (16.53g,46.96mmol) in tetrahydrofuran (100mL) was added dropwise a solution of tetrabutylammonium fluoride in tetrahydrofuran (150mL,1M,150 mmol). The reaction was stirred at room temperature for 2 hours, then quenched by addition of water (100mL) and extracted with dichloromethane (200 mL. times.9). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to a solvent volume of about 100mL, then filtered. The filter cake was washed with dichloromethane (30 mL. times.3), and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/100) to give a white solid, which was combined with the filter cake from the previous filtration to give the title compound as a white solid (7.77g, 84.6% yield).
MS(ESI,pos.ion)m/z:196.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):12.09(br.s,1H),8.29(s,1H),7.89(s,1H),7.65(d,J=3.4Hz,1H),7.61(s,1H),6.56(d,J=3.4Hz,1H)。
Step 4) 4-chloro-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridine-5-carboxamides
To a solution of 4-chloro-1H-pyrrolo [2,3-b ] pyridine-5-carboxamide (7.27g,37.2mmol) in tetrahydrofuran (150mL) at 0 deg.C was added sodium hydride (60% [ w/w ] mass fraction suspended in mineral oil, 1.67g,41.8mmol) portionwise. After the addition was complete, the reaction was maintained at 0 ℃ and stirring was continued for 1 hour. 4-Methylbenzenesulfonyl chloride (7.48g,39.2mmol) was added to the above reaction system in portions. The resulting reaction was stirred at room temperature for 1 hour, then quenched by addition of water (100mL) and extracted with dichloromethane (250 mL. times.3). The combined organic phases were washed with brine (250mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to a solvent volume of about 100mL, then filtered. The filter cake was washed with dichloromethane (90mL) and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 10/1) to give a white solid, which was combined with the filter cake from the previous filtration to give the title compound as a white solid (12.52g, 96.3% yield).
MS(ESI,pos.ion)m/z:350.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):8.41(s,1H),8.09(d,J=4.0Hz,1H),8.01(d,J=8.2Hz,3H),7.79(s,1H),7.44(d,J=8.3Hz,2H),6.93(d,J=4.0Hz,1H),2.36(s,3H)。
Step 5) 4-chloro-2-iodo-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridine-5-carboxamides
To 4-chloro-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] at-78 deg.C]To a solution of pyridine-5-carboxamide (1.00g,2.86mmol) in tetrahydrofuran (20mL) was added dropwise a solution of lithium diisopropylamide in tetrahydrofuran (2M,5mL,10 mmol). After the addition was complete, the reaction was stirred at-78 deg.CAfter 1 hour, a solution of elemental iodine (1.11g,4.37mmol) in tetrahydrofuran (5mL) was added dropwise to the system. The resulting reaction system was stirred at-78 ℃ for 2 hours, then quenched with water (30mL) and extracted with dichloromethane (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to a solvent volume of about 20mL, then filtered. The filter cake was washed with dichloromethane (20 mL. times.3), and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (100% DCM) to give a light yellow solid which was combined with the filter cake from the previous filtration to give the title compound as a light yellow solid (1.12g, 82.4% yield). MS (ESI, pos. ion) M/z 476.0[ M + H ]]+
Step 6) 4-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ]Pyridine (II) Pyridine-5-carboxamides
To 4-chloro-2-iodo-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridine-5-carboxamide (947.2mg,1.99mmol) and 1-methyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazole (460.2mg,2.21mmol) in a mixed solvent of 1, 4-dioxane (40mL) and water (10mL) was added Pd (dppf)2Cl2Dichloromethane complex (327.8mg,0.39mmol) and sodium carbonate (428.1mg,4.04 mmol). The reaction mixture was stirred at 100 ℃ overnight, then quenched by addition of water (100mL) and extracted with dichloromethane (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/80) to give the title compound as a brown solid (804.6mg, 94.0% yield).
MS(ESI,pos.ion)m/z:429.7[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.80(s,1H),7.81(s,1H),7.75(d,J=8.3Hz,2H),7.71(s,1H),7.21(d,J=8.4Hz,2H),6.63(s,1H),6.34(s,1H),5.94(s,1H),4.05(s,3H),2.37(s,3H)。
Step 7)9- ((5-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrole And [2,3-b]Pyridin-4-yl) amino) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To a solution of 4-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine-5-carboxamide (2.53g,5.89mmol) and tert-butyl 3-amino-9-azaspiro [5.5] undecane-9-carboxylate (1.90g,7.08mmol) in n-butanol (20mL) was added DIPEA (1.57g,12.20 mmol). The resulting mixture was warmed to reflux and stirred overnight, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/40) to afford the title compound as a brown solid (1.77g, 45.4% yield).
MS(ESI,pos.ion)m/z:661.8[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):9.25(d,J=7.6Hz,1H),8.39(s,1H),7.79(d,J=8.2Hz,2H),7.70(s,1H),7.61(s,1H),7.21(d,J=8.1Hz,2H),6.48(s,1H),5.70(brs,2H),4.03(s,3H),3.80(m,1H),3.37(m,4H),2.38(s,3H),1.92(m,2H),1.70(m,10H),1.47(s,9H)。
Step 8)9- ((5-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine- 4-yl) amino) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To a solution of tert-butyl 9- ((5-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) -3-azaspiro [5.5] undecane-3-carboxylate (1.77g,2.67mmol) in ethanol (50mL) was added aqueous sodium hydroxide (1M,50mL,50 mmol). The reaction system was stirred at 30 ℃ overnight, and then extracted with methylene chloride (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/80) to give the title compound as a light brown solid (974.8mg, 71.8% yield).
MS(ESI,pos.ion)m/z:508.4[M+H]+
Step 9)4- (3-azaspiro [5.5]]Undecane-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrazo l Pyrrolo [2,3-b]Pyridine-5-carboxamides
To a solution of tert-butyl 9- ((5-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) -3-azaspiro [5.5] undecane-3-carboxylate (974.8mg,1.92mmol) in DCM (10mL) was added a solution of hydrogen chloride in ethyl acetate (10mL,40 mmol). The reaction was stirred at room temperature for 0.5 hour and then concentrated under reduced pressure. The resulting residue was dissolved in water (30mL) and adjusted to pH 10 with saturated aqueous sodium carbonate solution, followed by extraction with dichloromethane (250mL × 3). The combined organic phases were washed with brine (250mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/5) to give the title compound as an off-white solid (652.7mg, 83.4% yield).
MS(ESI,pos.ion)m/z:204.7[(M+2H)/2]+
1H NMR(600MHz,DMSO-d6)δ(ppm):9.05(s,2H),8.39(s,1H),8.24(s,1H),8.00(s,1H),6.77(s,1H),4.10(s,1H),3.89(s,3H),3.01(s,4H),2.00(m,1H),1.91(d,J=3.6Hz,1H),1.66(m,6H),1.51(d,J=8.3Hz,4H)。
Example 24- ((3-acetyl-3-azaspiro [5.5]]Undecane-9-yl) amino) -2- (1-methyl-1H-pyri-dine Oxazol-4-yl) -1H-pyrrolo [2,3-b]Pyridine-5-carboxamides
Figure BDA0001582369760000511
4- (3-Azaspiro [5.5] undecan-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b ] pyridine-5-carboxamide (110.1mg,0.27mmol) and triethylamine (76.4mg,0.76mmol) were suspended in dichloromethane (8mL), and acetic anhydride (34.6mg,0.34mmol) in dichloromethane (2mL) was added to the suspension. The reaction was stirred at room temperature for 20 minutes, then quenched by addition of water (50mL) and extracted with dichloromethane (150 mL. times.6). The combined organic phases were washed with saturated brine (150mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/5) to give the title compound as an off-white solid (64.0mg, 52.7% yield).
MS(ESI,pos.ion)m/z:449.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):9.43(s,1H),8.23(s,1H),7.82(s,1H),7.68(s,1H),6.51(s,1H),5.83(s,2H),4.33(t,J=6.1Hz,1H),4.05(s,1H),3.98(s,3H),3.60(s,2H),3.43(s,2H),2.11(s,3H),1.76(m,2H),1.63(m,6H),1.45(m,4H)。
Example 34- ((3- (2-Cyanoacetyl) -3-azaspiro [ 5.5)]Undecane-9-yl) amino) -2- (1-methyl 1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b]Pyridine-5-carboxamides
Figure BDA0001582369760000512
To a solution of 4- (3-azaspiro [5.5] undecan-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b ] pyridine-5-carboxamide (0.55g,1.30mmol) and 2-cyanoacetic acid (171.0mg,2.01mmol) in dichloromethane (40mL) and DMF (10mL) was added HATU (758.4mg,2.00mmol) and triethylamine (0.35g,3.50 mmol). The reaction was stirred at room temperature for 10 minutes, then quenched by addition of water (50mL) and extracted with dichloromethane (150 mL. times.3). The combined organic phases were washed with saturated brine (150mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/5) to give the title compound as an off-white solid (54.4mg, 8.5% yield).
MS(ESI,pos.ion)m/z:474.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):9.50(s,1H),8.27(s,1H),7.78(s,1H),7.72(s,1H),6.50(s,1H),5.82(s,2H),5.36(s,1H),4.06(s,1H),3.99(s,3H),3.64(dd,J=12.8,9.1Hz,2H),3.51(s,2H),3.45(dd,J=13.4,8.2Hz,2H),2.05(m,4H),1.56(m,8H)。
Example 45-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (3-azaspiro [5.5]Undecane-9-yl) -1H- Pyrrolo [2,3-b]Pyridin-4-amines
Figure BDA0001582369760000521
Step 1) 5-chloro-1H-pyrrolo [2,3-b]Pyridine oxide
To a solution of 5-chloro-1H-pyrrolo [2,3-b ] pyridine (20.00g,131.1mmol) in dichloromethane (500mL) at 0 deg.C was added mCPBA (85% [ w/w ],67.86g,393.2 mmol). The reaction mixture was allowed to warm to room temperature and stirred overnight, then concentrated under reduced pressure. The obtained residue was dissolved in water (250mL), adjusted to pH 8 to 9 with saturated aqueous sodium carbonate solution, and then extracted with dichloromethane (250mL × 3). The combined organic phases were washed with brine (250mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/50) to afford the title compound as a gray solid (14.00g, 63.4% yield).
MS(ESI,pos.ion)m/z:169.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):12.69(s,1H),8.36(d,J=1.4Hz,1H),7.80(d,J=1.4Hz,1H),7.53(d,J=2.8Hz,1H),6.56(d,J=3.1Hz,1H)。
Step 2) 4-bromo-5-chloro-1H-pyrrolo [2,3-b]Pyridine compound
At 0 deg.C, to 5-chloro-1H-pyrrolo [2,3-b ]]Pyridine oxide (5.00g,29.66mmol) and Bu4NBr (19.12g,59.31mmol) in DMF (200mL) was added dropwise to triflic anhydride (20.91g,74.20 mmol). The reaction mixture was allowed to warm to room temperature and stirred for 2 hours, then poured into water (500 mL). The resulting mixture was adjusted to pH 8 to 9 with saturated aqueous sodium bicarbonate solution, and then filtered. The filter cake was washed with water (50mL × 3), collected and dried under vacuum at 60 ℃ overnight to give the title compound as an off-white solid (5.83g, 84.9% yield).
MS(ESI,pos.ion)m/z:231.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):12.24(s,1H),8.30(s,1H),7.68(s,1H),6.45(s,1H)。
Step 3) 4-bromo-5-chloro-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridine compound
To a solution of 4-bromo-5-chloro-1H-pyrrolo [2,3-b ] pyridine (5.83g,25.18mmol) and DMAP (615.0mg,5.03mmol) in dichloromethane (250mL) at 0 deg.C were added triethylamine (5.10g,50.40mmol) and p-toluenesulfonyl chloride (7.20g,37.76 mmol). The reaction was allowed to warm to room temperature and stirred overnight, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) ═ 1/50) to give the title compound as a white solid (7.23g, 74.4% yield).
MS(ESI,pos.ion)m/z:385.0[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.39(s,1H),8.06(d,J=8.4 Hz,2H),7.82(d,J=4.0Hz,1H),7.31(d,J=8.3Hz,2H),6.64(d,J=4.0Hz,1H),2.41(s,3H)。
Step 4) 4-bromo-5-chloro-2-iodo-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridine compound
To a solution of 4-bromo-5-chloro-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (2.53g,6.56mmol) in tetrahydrofuran (20mL) at-78 deg.C was added lithium diisopropylamide (5mL,10mmol) dropwise. The reaction mixture was stirred for 40 minutes while maintaining-78 deg.C, and then a solution of elemental iodine (2.64g,10.4mmol) in tetrahydrofuran (20mL) was added dropwise to the reaction mixture. After the reaction mixture was stirred at-78 ℃ for an additional 1 hour, the reaction was quenched by the addition of saturated aqueous sodium thiosulfate (60 mL). The mixture was extracted with ethyl acetate (100 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (PE/EtOAc (v/v) ═ 50/1) to give the title compound as a light yellow solid (1.24g, 37% yield).
MS(ESI,pos.ion)m/z:513.0[M+H]+
Step 5) 4-bromo-5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3- b]Pyridine compound
To 4-bromo-5-chloro-2-iodo-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ]]Pyridine, 1-methyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazole (491.5mg,2.362mmol), sodium bicarbonate (402.6mg,4.792mmol) and Pd (dppf)2Cl2To a mixture of dichloromethane complex (190.8mg,0.2337mmol) were added 1, 4-dioxane (40mL) and water (3 mL). The reaction system was warmed to 100 ℃, stirred for 3.5 hours under an inert gas atmosphere, and then concentrated under reduced pressure. The resulting residue was diluted with water (80mL) and then extracted with ethyl acetate (100 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (PE/EtOAc (v/v) ═ 2/1) to give the title compound as a pale yellow solid (380mg, 35% yield)
MS(ESI,pos.ion)m/z:465.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.37(s,1H),7.78(s,1H),7.69(d,J=8.2Hz,2H),7.67(s,1H),7.18(d,J=8.2Hz,2H),6.48(s,1H),4.02(s,3H),2.34(s,3H)。
Step 6)9- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3- b]Pyridin-4-yl) amino) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To 4-bromo-5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2, 3-b)]Pyridine (350mg,0.7514mmol), 3-amino-9-azaspiro [5.5 ]Tert-butyl undecane-9-carboxylate (405.6mg,1.511mmol), cesium carbonate (735.2mg,2.256mmol), BINAP (47.3mg,0.076mmol) and Pd (OAc)2(16.4mg,0.0731mmol) to the mixture was added 1, 4-dioxane (20 mL). The reaction system was warmed to 100 ℃, stirred overnight under an inert gas atmosphere, and then concentrated under reduced pressure. The resulting residue was diluted with water (40mL) and extracted with DCM/MeOH (v/v ═ 10/1,60mL × 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 125/1) to give the title compound as a yellow solid (370mg, 75% yield).
MS(ESI,pos.ion)m/z:653.3[M+H]+
1HNMR(400MHz,CDCl3)δ(ppm):8.09(s,1H),7.74(d,J=8.3Hz,2H),7.70(s,1H),7.60(s,1H),7.17(d,J=8.1Hz,2H),6.40(s,1H),4.88(d,J=8.3Hz,1H),3.99(s,3H),3.77-3.66(m,1H),3.45-3.27(m,4H),2.33(s,3H),1.91(dd,J=13.6,3.4Hz,2H),1.71(d,J=14.5Hz,2H),1.53-1.40(m,15H),1.38-1.32(m,2H)。
Step 7)9- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) amino Yl) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To a solution of tert-butyl 9- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) -3-azaspiro [5.5] undecane-3-carboxylate (370mg,0.5664mmol) in ethanol (10mL) was added sodium hydroxide (10mL,10mmol, 1M). The reaction system was heated to 60 ℃ and stirred for 5 hours, after the reaction was completed, water (50mL) was added to quench the reaction. The resulting mixture was extracted with a mixed solvent of DCM/MeOH (v/v ═ 10/1,80mL × 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product as a yellow solid (290 mg). The crude product was used in the next step without purification.
MS(ESI,pos.ion)m/z:499.5[M+H]+
Step 8) 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (3-azaspiro [ 5.5%]Undecane-9-yl) -1H-pyri dine Pyrrolo [2,3-b]Pyridin-4-amines
To 9- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) amino) -3-azaspiro [5.5]To a solution of tert-butyl undecane-3-carboxylate (290mg,0.5812mmol) in ethyl acetate (5mL) was added a solution of hydrogen chloride in ethyl acetate (10mL,20mmol, 2M). The reaction was stirred at room temperature for 1 hour. The reaction mixture was washed with water (50mL × 2), the combined aqueous layers were adjusted to pH 12 with a sodium carbonate solid, and the resulting mixture was extracted with a mixed solvent of dichloromethane and methanol (v/v 10/1,100mL × 4). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue obtained is subjected to preparative thin layer chromatography (DCM/7M NH)3Purified with MeOH (v/v) ═ 10/1) to give the title compound as a yellow solid (190mg, 82% yield).
MS(ESI,pos.ion)m/z:200.3[(M+2H)/2]+
Example 51-9- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl)) -1H-pyrrolo [2,3-b]Pyridin-4-yl) Amino) -3-azaspiro [5.5]Undecane-3-yl) ethanones
Figure BDA0001582369760000541
To a solution of 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (3-azaspiro [5.5] undecan-9-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (80mg,0.2006mmol) in dichloromethane (10mL) was added triethylamine (64.5mg,0.637mmol) and acetic anhydride (21.8mg,0.214 mmol). The reaction system was stirred at room temperature for 30 minutes and then concentrated under reduced pressure. The resulting residue was purified by preparative thin layer chromatography (MeOH/DCM (v/v) ═ 10/1) to afford the title compound as a yellow solid (73.1mg, 82.7% yield).
MS(ESI,pos.ion)m/z:441.3[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):11.29(s,1H),7.92(s,1H),7.81(s,1H),7.71(s,1H),6.44(s,1H),4.96(d,J=8.4Hz,1H),3.98(m,4H),3.59(dd,J=11.7,7.0Hz,2H),3.42(dd,J=11.5,7.4Hz,2H),2.08(d,J=16.2Hz,5H),1.84-1.75(m,4H),1.61-1.53(m,4H),1.50-1.36(m,2H)。
Example 63- (9- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine-4- Yl) amino) -3-azaspiro [5.5]Undec-3-yl) -3-oxopropanenitrile
Figure BDA0001582369760000542
To a mixture of 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (3-azaspiro [5.5] undecan-9-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (100mg,0.2507mmol), 2-cyanoacetic acid (43.7mg,0.514mmol), EDCI (96.3mg,0.502mmol), HOAT (70.4mg,0.517mmol), and triethylamine (131.9mg,1.303mmol) was added dichloromethane (12mL) and DMF (1 mL). The reaction mixture was stirred at room temperature for 1 hour, and the reaction was completed. The mixture was diluted with water (30mL), and the resulting mixture was extracted with a mixed solvent of DCM/MeOH (v/v ═ 10/1,50mL × 4). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by preparative thin layer chromatography (MeOH/DCM (v/v) ═ 1/10) to afford the title compound as a yellow solid (80.2mg, 68.7% yield).
MS(ESI,pos.ion)m/z:466.4[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):11.33(s,1H),7.93(s,1H),7.81(s,1H),7.71(s,1H),6.43(s,1H),4.96(d,J=8.4Hz,1H),3.99(m,4H),3.62(dd,J=11.4,6.3Hz,2H),3.49(s,2H),3.44(dd,J=11.6,8.9Hz,2H),2.08(d,J=10.7Hz,2H),1.76-1.85(m,4H),1.71-1.66(m,1H),1.65-1.60(m,1H),1.54-1.59(m,2H),1.53-1.47(m,2H)。
Example 75-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrole-5-yl) -1H- Pyrrolo [2,3-b]Pyridin-4-amines
Figure BDA0001582369760000543
Step 1)5- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3- b]Pyridin-4-yl) amino) hexahydrocyclopenta [ c) ]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To 4-bromo-5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2, 3-b)]Pyridine (596.9mg,1.28mmol) and 5-aminohexahydrocyclopenta [ c ]]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester (0.57g,2.52mmol) in 1, 4-dioxane (50mL) Pd (OAc) was added2(58.9mg,0.26mmol), BINAP (188.3mg,0.29mmol) and Cs2CO3(867.7mg,2.66 mmol). The reaction mixture was stirred at 100 ℃ overnight and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) ═ 2/1) to give the title compound as a yellow solid (783.2mg, 100% yield).
MS(ESI,pos.ion)m/z:611.5[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.11(s,1H),7.76(d,J=8.3Hz,2H),7.72(s,1H),7.62(s,1H),7.19(d,J=8.2Hz,2H),6.48(s,1H),5.02(d,J=8.2Hz,1H),4.29(m,1H),4.02(s,3H),3.49(m,2H),3.34(d,J=9.8Hz,2H),2.70(m,2H),2.43(dt,J=13.3,6.7Hz,2H),2.36(s,3H),1.48(s,9H),1.45(m,2H)。
Step 2)5- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) amino Radical) hexahydrocyclopenta [ c ] radical]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To a solution of tert-butyl 5- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (751.8mg,1.23mmol) in ethanol (25mL) was added an aqueous solution of sodium hydroxide (1M,25mL,25 mmol). The reaction was warmed to 60 ℃ and stirred for 3.5 hours, then concentrated under reduced pressure. The resulting residue was dissolved in water (50mL), then extracted with dichloromethane (100 mL. times.3), and the combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/50) to give the title compound as a pale yellow solid (495.8mg, 88.2% yield).
MS(ESI,pos.ion)m/z:457.3[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.85(s,1H),7.96(s,1H),7.82(s,1H),7.69(s,1H),6.52(s,1H),5.05(d,J=8.3Hz,1H),4.56(m,1H),4.01(s,3H),3.54(m,2H),3.40(brs,2H),2.78(m,2H),2.56(m,2H),1.55(m,2H),1.50(s,9H)。
Step 3) 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrol-5-yl) -1H-pyri-dine Pyrrolo [2,3-b]Pyridin-4-amines
To a solution of tert-butyl 5- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (471.7mg,1.03mmol) in dichloromethane (10mL) was added a solution of hydrogen chloride in ethyl acetate (10mL,40 mmol). The reaction was stirred at room temperature for 1 hour and then concentrated under reduced pressure. The resulting residue was dissolved in water (30mL) and adjusted to pH 10 with saturated aqueous sodium carbonate solution, followed by extraction with dichloromethane (250mL × 3). The combined organic phases were washed with brine (250mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/10) to give the title compound as an off-white solid (324.6mg, 88.1% yield).
MS(ESI,pos.ion)m/z:357.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):11.65(s,1H),8.13(s,1H),7.93(s,1H),7.77(s,1H),6.71(s,1H),6.62(d,J=9.4Hz,1H),4.45(m,1H),3.87(s,3H),2.98(br.s,1H),2.69(d,J=3.0Hz,4H),2.61(m,2H),2.25(dt,J=13.4,6.9Hz,2H),1.45(dt,J=10.6,5.0Hz,2H)。
Example 83- (5- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine-4- Yl) amino) hexahydrocyclopenta [ c ] s]Pyrrole-2 (1H) -yl) -3-oxopropanenitrile
Figure BDA0001582369760000551
To 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrol-5-yl) -1H-pyrrolo [2,3-b]Pyridine-4-amine (107.4mg,0.30mmol) and 2-cyanoacetic acid (43.0mg,0.51mmol) in dichloromethane (15mL) was added HOAT (87.8mg,0.64mmol), EDCI (121.5mg,0.63mmol) and Et 3N (118.5mg,1.17 mmol). The reaction was stirred at room temperature for 0.5 hour, then quenched by addition of water (30mL) and extracted with dichloromethane (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/30) to give the title compound as an off-white solid (77.6mg, 60.8% yield).
MS(ESI,pos.ion)m/z:424.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.66(s,1H),7.97(s,1H),7.82(s,1H),7.70(s,1H),6.50(s,1H),4.98(d,J=8.1Hz,1H),4.62(m,1H),4.02(s,3H),3.78(m,2H),3.67(m,1H),3.49(m,3H),2.98(m,1H),2.88(m,1H),2.63(m,2H),1.56(m,2H)。
Practice ofExample 91- (5- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine-4- Yl) amino) hexahydrocyclopenta [ c ] s]Pyrrol-2 (1H) -yl) ethanones
Figure BDA0001582369760000561
To a suspension of 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] pyrrol-5-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (128.0mg,0.36mmol) and triethylamine (84.8mg,0.84mmol) suspended in dichloromethane (10mL) was added acetic anhydride (39.6mg,0.39mmol) in dichloromethane (5 mL). The reaction was stirred at room temperature for 15 minutes, and then quenched by addition of water (30 mL). The resulting mixture was extracted with dichloromethane (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/30) to give the title compound as an off-white solid (139.8mg, 97.7% yield).
MS(ESI,pos.ion)m/z:399.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):11.10(s,1H),7.96(s,1H),7.83(s,1H),7.71(s,1H),6.51(s,1H),5.01(d,J=8.2Hz,1H),4.60(m,1H),4.01(s,3H),3.67(m,3H),3.44(dd,J=10.8,3.9Hz,1H),2.87(m,2H),2.60(m,2H),2.10(s,3H),1.56(tt,J=13.0,8.0Hz,2H)。
Example 105-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (3-azaspiro [5.5]Undecane-9- 1H-pyrrolo [2,3-b ] yl]Pyridin-4-amines
Figure BDA0001582369760000562
Step 1)1, 3-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazole
To a solution of 4-bromo-1, 3-dimethyl-1H-pyrazole (5.00g,28.60mmol) in anhydrous 1, 4-dioxane (10)0.0mL) was added 4,4,4',4',5,5,5',5' -octamethyl-2, 2' -bis (1,3, 2-dioxaborolan) (8.71g,34.32mmol), potassium acetate (5.63g,57.41mmol) and Pd (dppf)2Cl2Dichloromethane complex (2.38g,2.86 mmol). The reaction mixture was evacuated of air and purged with nitrogen several times, then warmed to 100 ℃ and stirred overnight. The reaction mixture was concentrated under reduced pressure. The resulting mixture was diluted with water (30mL) and extracted with ethyl acetate (50 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) ═ 1/20 to 1/8) to give the title compound as a brown viscous liquid (3.80g, 60% yield).
MS(ESI,pos.ion)m/z:223.3[M+H]+
Step 2) 4-bromo-5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridine compound
To 4-bromo-5-chloro-2-iodo-1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] ]Pyridine (2.50g,4.89mmol) suspended in a mixed solvent of 1, 4-dioxane (45.0mL) and water (9.0mL) was added to a suspension of 1, 3-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazole (2.18g,9.82mmol), sodium carbonate (1.05g,9.90mmol) and Pd (dppf)2Cl2Dichloromethane complex (0.80g,0.99 mmol). The reaction mixture was evacuated of air and purged with nitrogen several times, then, heated to 100 ℃ and stirred for reaction for 4 hours, and then the mixture was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (100% PE to EtOAc/PE (v/v) ═ 1/4 to 1/2) to afford the title compound as a brown viscous liquid (0.54g, 23% yield).
MS(ESI,pos.ion)m/z:479.0[M+H]+
Step 3)9- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridin-4-yl) amino) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To 4-bromo-5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2, 3-b)]Pyridine (0.54g,1.10mmol) in anhydrous 1,4-Dioxane (15.0mL) solution was added 9-amino-3-azaspiro [ 5.5%]Undecyl-3-carboxylic acid tert-butyl ester (0.62g,2.30mmol), Pd (OAc)2(0.052g,0.23mmol), BINAP (0.14g,0.23mmol), and cesium carbonate (0.75g,2.31 mmol). The reaction mixture was evacuated of air and purged with nitrogen several times, then warmed to reflux and stirred overnight. The reaction mixture was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) ═ 1/4 to 1/2) to give the title compound as a yellow solid (0.46g, 61% yield).
MS(ESI,pos.ion)m/z:667.5M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.12(s,1H),7.67(d,J=8.3Hz,2H),7.50(s,1H),7.15(d,J=8.2Hz,2H),6.33(s,1H),4.89(d,J=8.1Hz,1H),3.93(s,3H),3.77-3.64(m,1H),3.40-3.31(m,4H),2.33(s,3H),2.14(s,3H),1.96-1.87(m,2H),1.76-1.66(m,4H),1.53-1.45(m,4H),1.44(s,9H),1.37-1.31(m,2H)。
Step 4)9- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) Amino) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To a solution of tert-butyl 9- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) -3-azaspiro [5.5] undecane-3-carboxylate (0.46g,0.69mmol) in ethanol (7.0mL) was added aqueous sodium hydroxide (7.0mL,7.0mmol, 1M). The reaction mixture was warmed to 60 ℃ and stirred overnight. The reaction mixture was concentrated under reduced pressure. The resulting residue was diluted with water (10mL), and the resulting solution was extracted with DCM (30mL × 3) and DCM/MeOH ((v/v) ═ 10/1,30mL × 3) in this order. The combined organic phases were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product as a yellow solid (0.33g, 98%). The crude product was used in the next step without purification.
MS(ESI,pos.ion)m/z:513.3[M+H]+
Step 5) 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (3-azaspiro [5.5]Undecane-9-yl) - 1H-pyrrolo [2,3-b]Pyridin-4-amines
To 9- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazole-)4-yl) -1H-pyrrolo [2,3-b]Pyridin-4-yl) amino) -3-azaspiro [5.5]To a solution of tert-butyl undecane-3-carboxylate (0.33g,0.64mmol) in dichloromethane (10mL) was added a solution of hydrogen chloride in ethyl acetate (10.0mL,30.0 mmol). The reaction was stirred at room temperature for 2 hours and then concentrated under reduced pressure. The resulting residue was diluted with DCM (30mL) and saturated aqueous sodium carbonate (30mL) and the resulting mixture was stirred at room temperature for 30 min. After standing, the organic layer was separated, and the aqueous layer was extracted with DCM (50mL × 3) and DCM/MeOH ((v/v) ═ 10/1,50mL × 3) in this order. The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was chromatographed on a silica gel column (DCM/3M NH) 3Purified with MeOH (v/v) ═ 30/1 to 20/1 to 10/1 to give the title compound as a yellow solid (80mg, 30% yield).
MS(ESI,pos.ion)m/z:413.4[M+H]+
Example 113- (9- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine (II) Pyridin-4-yl) amino) -3-azaspiro [5.5]Undec-3-yl) -3-oxopropanenitrile
Figure BDA0001582369760000581
To 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (3-azaspiro [ 5.5%]Undec-9-yl) -1H-pyrrolo [2,3-b]To a suspension of pyridin-4-amine (40mg,0.097mmol) suspended in DCM (10mL) was added 2-cyanoacetic acid (18mg,0.21mmol), EDCI (30mg,0.15mmol), HOAT (12mg,0.15mmol), and triethylamine (31mg,0.31 mmol). The mixture was stirred at room temperature for 30 minutes, and dichloromethane (30mL) was added for dilution, the resulting mixture was washed with water (20 mL. times.2), and the separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was chromatographed on a silica gel column (DCM/3M NH)3Purified with MeOH (v/v) ═ 50/1 to 30/1 to 20/1 to give the title compound as a yellow solid (36mg, 77% yield).
MS(ESI,pos.ion)m/z:480.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):9.93(br,s,1H),7.91(s,1H),7.58(s,1H),6.40(s,1H),4.97(d,J=8.1Hz,1H),4.05-3.96(m,1H),3.92(s,3H),3.66-3.58(m,2H),3.48(d,J=1.9Hz,2H),3.47-3.39(m,2H),2.46(s,3H),2.16-2.06(m,2H),1.87-1.77(m,2H),1.60-1.48(m,4H),1.46-1.37(m,2H),1.29-1.23(m,2H)。
Example 121- (9- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine (II) Pyridin-4-yl) amino) -3-azaspiro [5.5]Undecane-3-yl) ethanones
Figure BDA0001582369760000582
To 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (3-azaspiro [ 5.5% ]Undec-9-yl) -1H-pyrrolo [2,3-b]To a solution of pyridin-4-amine (32mg,0.077mmol) in DCM (5.0mL) was added acetic anhydride (11mg,0.11mmol) and triethylamine (13mg,0.13 mmol). The reaction was stirred at room temperature overnight and then concentrated under reduced pressure. The residue was chromatographed on a silica gel column (DCM/3M NH)3Purified with MeOH (v/v) ═ 50/1 to 30/1 to 20/1 to give the title compound as a yellow solid (28mg, 79% yield).
MS(ESI,pos.ion)m/z:455.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):7.81(s,1H),7.67(d,J=4.0Hz,1H),6.36(s,1H),4.00-3.89(m,1H),3.85(s,3H),3.57-3.49(m,2H),3.43-3.36(m,2H),2.41(s,3H),2.09-2.00(m,5H),1.82-1.70(m,2H),1.61-1.47(m,4H),1.45-1.32(m,4H)。
Example 135-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrole-5- 1H-pyrrolo [2,3-b ] yl]Pyridin-4-amines
Figure BDA0001582369760000583
Step 1)5- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridin-4-yl) amino) hexahydroCyclopentano [ c ]]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To 5-aminohexahydrocyclopenta [ c ]]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester (245.6mg,1.085mmol), 4-bromo-5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ]]Pyridine (250mg,0.5211mmol), cesium carbonate (514.2mg,1.578mmol), BINAP (30.7mg,0.0493mmol) and Pd (OAc)2(10.9mg,0.0486mmol) to the mixture was added 1, 4-dioxane (20 mL). The reaction mixture was warmed to 100 ℃ and stirred overnight under an inert gas atmosphere. The resulting residue was diluted with water (40mL) and extracted with DCM/MeOH (v/v ═ 10/1,60mL × 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (PE/EtOAc (v/v) ═ 1/1) to give the title compound as a yellow solid (260mg, 79% yield).
MS(ESI,pos.ion)m/z:625.5[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.11(s,1H),7.68(d,J=8.3Hz,2H),7.49(s,1H),7.16(d,J=8.1Hz,2H),6.40(s,1H),5.01(d,J=8.0Hz,1H),4.25(s,1H),3.92(s,3H),3.44(d,J=6.9Hz,2H),3.31(d,J=9.3Hz,2H),2.72-2.60(m,2H),2.44-2.35(m,2H),2.33(s,3H),2.15(s,3H),1.49-1.40(m,11H)。
Step 2)5- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) Amino) hexahydrocyclopenta [ c ] s]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To a solution of tert-butyl 5- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (260mg,0.416mmol) in ethanol (10mL) was added sodium hydroxide (10mL,20mmol, 2M). The reaction was warmed to 60 ℃ and stirred overnight, then diluted with water (50 mL). The mixture was extracted with a mixed solvent of DCM/MeOH (v/v ═ 10/1,80mL × 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product as a yellow solid (205 mg). The crude product was used in the next step without purification.
MS(ESI,pos.ion)m/z:471.4[M+H]+
Step 3) 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrole-5-yl) - 1H-pyrrolo [2,3-b]Pyridin-4-amines
To 5- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) amino) hexahydrocyclopenta [ c)]To a solution of tert-butyl pyrrole-2 (1H) -carboxylate (205mg,0.435mmol) in ethyl acetate (5mL) was added a solution of hydrogen chloride in ethyl acetate (10mL,20mmol, 2M). The reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was washed with water (50 mL. times.2). The combined aqueous phases were adjusted to pH 12 with sodium carbonate powder and then extracted with a mixed solvent of DCM/MeOH (v/v 10/1,100mL × 4). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue obtained is subjected to preparative thin layer chromatography (DCM/7M NH) 3Purified with MeOH (v/v) ═ 10/1) to give the title compound as a yellow solid (75mg, 46% yield).
MS(ESI,pos.ion)m/z:371.2[M+H]+
Example 143- (5- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine (II) Pyridin-4-yl) amino) hexahydrocyclopenta [ c)]Pyrrole-2 (1H) -yl) -3-oxopropanenitrile
Figure BDA0001582369760000591
A mixture of 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] pyrrol-5-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (45mg,0.1213mmol), 2-cyanoacetic acid (19.8mg,0.233mmol), EDCI (47.9mg,0.250mmol), HOAT (35.34mg,0.2596mmol) and triethylamine (71.5mg,0.707mmol) was dissolved in a mixed solvent of DCM and DMF (10mL/0.5 mL). The reaction was stirred at room temperature for 20 hours and then diluted with water (20 mL). The resulting mixture was extracted with a mixed solvent of DCM/MeOH (v/v ═ 10/1,20mL × 4). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by preparative thin layer chromatography (DCM/MeOH (v/v) ═ 10/1) to afford the title compound as a light yellow solid (39mg, 73% yield).
MS(ESI,pos.ion)m/z:438.2[M+H]+
1H NMR(400MHz,CDCl3+CD3OD)δ(ppm):7.83(s,1H),7.66(s,1H),6.38(s,1H),4.58-4.49(m,1H),3.86(s,3H),3.74-3.63(m,2H),3.59(dd,J=12.9,4.3Hz,1H),3.46-3.38(m,2H),3.37-3.34(m,1H),2.93-2.86(m,1H),2.86-2.75(m,1H),2.62-2.55(m,2H),2.42(s,3H),1.61-1.48(m,2H)。
Example 151- (5- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine (II) Pyridin-4-yl) amino) hexahydrocyclopenta [ c)]Pyrrol-2 (1H) -yl) ethanones
Figure BDA0001582369760000601
To a solution of 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] pyrrol-5-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (30mg,0.0809mmol) and triethylamine (38.7mg,0.382mmol) in dichloromethane (4mL) was added a solution of acetic anhydride (12.6mg,0.123mmol) in dichloromethane (4 mL). The reaction system was stirred at room temperature for 20 minutes and then concentrated under reduced pressure. The resulting residue was purified by preparative thin layer chromatography (DCM/MeOH (v/v) ═ 10/1) to afford the title compound as a yellow solid (24.9mg, 74.6% yield).
MS(ESI,pos.ion)m/z:413.4[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.85(s,1H),7.88(s,1H),7.60(s,1H),6.44(s,1H),4.99(d,J=7.9Hz,1H),4.63-4.51(m,1H),3.92(s,3H),3.70-3.57(m,3H),3.42(dd,J=10.8,3.8Hz,1H),2.92-2.74(m,2H),2.58(tt,J=14.4,7.3Hz,2H),2.46(s,3H),2.08(s,3H),1.60-1.49(m,2H)。
Example 166- (4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine (II) Pyridin-4-yl) amino) piperidin-1-yl) -3-cyanopyridine
Figure BDA0001582369760000602
Step 1)4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b]Pyridin-4-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
To 4-bromo-5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2, 3-b)]To a solution of pyridine (857mg,1.786mmol), tert-butyl 4-aminopiperidine-1-carboxylate (543mg,2.711mmol) and cesium carbonate (1.77g,5.43mmol) in 1, 4-dioxane (40mL) was added Pd (OAc)2(41mg,0.1826mmol) and BINAP (112.5mg,0.1807 mmol). The reaction mixture was evacuated of air for 2 minutes and refilled with nitrogen. The reaction system was heated to 100 ℃ and stirred for 3 hours, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (PE/EtOAc (v/v) ═ 5/1 to 2/1) to give the title compound as a pale yellow solid (307mg, 28.69% yield).
MS(ESI,pos.ion)m/z:599.5[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.13(s,1H),7.68(d,J=8.3Hz,2H),7.50(s,1H),7.16(d,J=8.2Hz,2H),6.32(s,1H),4.85(d,J=8.3Hz,1H),4.01(d,J=8.2Hz,2H),3.93(s,3H),3.88(dd,J=11.3,7.4Hz,2H),2.94(t,J=11.6Hz,2H),2.34(s,3H),2.14(s,3H),2.00(s,2H),1.45(s,9H)。
Step 2)4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) Amino) piperidine-1-carboxylic acid tert-butyl ester
To a solution of tert-butyl 4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) piperidine-1-carboxylate (300mg,0.501mmol) in ethanol (5mL) was added aqueous sodium hydroxide (5mL,5mmol, 1M). The reaction mixture was warmed to 60 ℃ and stirred overnight. The resulting residue was dissolved in water (10mL), and the resulting solution was extracted with a mixed solvent of DCM/MeOH (v/v ═ 10/1,20mL × 3). The combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/50 to 20/1) to give the title compound as a yellow solid (215mg, 96.48% yield).
MS(ESI,pos.ion)m/z:445.3[M+H]+
Step 3) 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (piperidin-4-yl) -1H-pyrrolo [2,3-b] Pyridin-4-amines
To a solution of tert-butyl 4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) piperidine-1-carboxylate (215mg,0.483mmol) in dichloromethane (12mL) was added a solution of hydrogen chloride in ethyl acetate (5mL,4M,20 mmol). The reaction mixture was stirred at room temperature for 2 hours and then concentrated under reduced pressure. The resulting residue was dissolved in water (20mL), and a saturated aqueous solution of sodium hydrogencarbonate was added thereto to adjust the pH to 8 to 9, followed by extraction with DCM/MeOH (v/v 10/1,100mL × 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was chromatographed on a silica gel column (MeOH/DCM (v/v) ═ 1/10 to 1/5) to give the title compound as a light yellow solid (158mg, 94.84% yield).
MS(ESI,pos.ion)m/z:345.3[M+H]+
Step 4)6- (4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine-4- Yl) amino) piperidin-1-yl) -3-cyanopyridine
To a solution of 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (piperidin-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (53.8mg,0.156mmol) in ethanol (15mL) were added 6-chloropyridine-3-carbonitrile (43.5mg,0.314mmol) and triethylamine (31.6mg,0.312 mmol). The reaction was warmed to 85 ℃ and stirred overnight, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 100/1 to 50/1) to give the title compound as a pale yellow solid (50mg, 71.7% yield).
MS(ESI,pos.ion)m/z:447.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.57(s,1H),8.43(d,J=2.1Hz,1H),7.92(s,1H),7.63(dd,J=9.0,2.3Hz,1H),7.60(s,1H),6.67(d,J=9.0Hz,1H),6.43(s,1H),4.93(d,J=8.4Hz,1H),4.40(d,J=13.9Hz,2H),4.31(dd,J=11.2,7.1Hz,1H),3.93(s,3H),3.33-3.22(m,2H),2.47(s,3H),2.31(d,J=10.1Hz,2H),1.57(dd,J=13.4,6.7Hz,2H)。
Example 174- (4- ((5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine (II) Pyridin-4-yl) amino) piperidin-1-yl) -3-benzonitrile
Figure BDA0001582369760000611
To a solution of 5-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (piperidin-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (144mg,0.418mmol) in dimethyl sulfoxide (15mL) were added 4-fluorobenzonitrile (101.5mg,0.838mmol) and potassium carbonate (115.4mg,0.835 mmol). The reaction mixture was warmed to 100 ℃ and stirred for 20 hours. After completion of the reaction, the mixture was diluted with water (10mL), and the resulting mixture was extracted with a mixed solvent of DCM and MeOH (v/v ═ 10/1,20mL × 3). The combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was chromatographed on a silica gel column (MeOH/DCM (v/v) ═ 1/100 to 1/50) to give the title compound as a yellow solid (25mg, yield 13.42%).
MS(ESI,pos.ion)m/z:446.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.89(s,1H),7.78(s,1H),7.69(s,1H),7.37(d,J=8.9Hz,2H),6.79(d,J=8.9Hz,2H),6.29(s,1H),4.74(d,J=8.4Hz,1H),4.15-4.04(m,1H),3.74(d,J=7.9Hz,5H),3.01(t,J=11.0Hz,2H),2.33(s,3H),2.17(d,J=10.4Hz,2H),1.60(td,J=13.6,3.6Hz,2H)。
Example 186- (4- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridine-4- Yl) amino) piperidin-1-yl) -3-cyanopyridine
Figure BDA0001582369760000621
Step 1)4- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl)-1H-pyrrolo [2,3- b]Pyridin-4-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
To 4-bromo-5-chloro-2- (1-dimethyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2, 3-b)]To a solution of pyridine (205mg,0.4401mmol), tert-butyl 4-aminopiperidine-1-carboxylate (132.5mg,0.662mmol) and cesium carbonate (430.5mg,1.321mmol) in 1, 4-dioxane (40mL) was added Pd (OAc)2(10mg,0.04454mmol) and BINAP (27.5mg,0.0442 mmol). The reaction mixture was evacuated of air for 2 minutes and refilled with nitrogen. The reaction system was heated to 100 ℃ and stirred for 3 hours, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 5/1 to 2/1) to afford the title compound as a yellow solid (162mg, 62.91% yield).
MS(ESI,pos.ion)m/z:585.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.11(s,1H),7.74(d,J=8.3Hz,2H),7.70(s,1H),7.60(s,1H),7.17(d,J=8.2Hz,2H),6.38(s,1H),4.83(d,J=8.4Hz,1H),4.00(s,5H),3.96-3.79(m,2H),2.97(t,J=11.5Hz,2H),2.34(s,3H),2.00(s,2H),1.45(s,9H)。
Step 2)4- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) amino Yl) piperidine-1-carboxylic acid tert-butyl ester
To a solution of tert-butyl 4- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1-p-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) piperidine-1-carboxylate (238mg,0.407mmol) in ethanol (4mL) was added an aqueous solution of sodium hydroxide (4mL,4mmol, 1M). The reaction was stirred at 60 ℃ overnight. The resulting residue was diluted with water (10mL) and extracted with DCM/MeOH (v/v ═ 10/1,20mL × 3). The combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/50 to 20/1) to give the title compound as a yellow solid (164mg, yield 93.57%).
MS(ESI,pos.ion)m/z:431.3[M+H]+
Step 3) 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (piperidin-4-yl) -1H-pyrrolo [2,3-b]Pyridine (II) Pyridin-4-amines
To a solution of tert-butyl 4- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-yl) amino) piperidine-1-carboxylate (164mg,0.3806mmol) in dichloromethane (12mL) was added a solution of hydrogen chloride in ethyl acetate (5mL,4M,20 mmol). The reaction was stirred at room temperature for 2 hours, and then concentrated under reduced pressure. The residue was dissolved in water (20mL), and the mixture was adjusted to pH 8 to 9 with saturated aqueous sodium bicarbonate. The resulting mixture was extracted with a mixed solvent of DCM/MeOH (v/v ═ 10/1,100mL × 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 1/10 to 1/5) to give the title compound as a light yellow solid (112mg, yield 88.96%).
MS(ESI,pos.ion)m/z:331.3[M+H]+
Step 4)6- (4- ((5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -1H-pyrrolo [2, 3-b)]Pyridin-4-yl) Amino) piperidin-1-yl) -3-cyanopyridines
To a solution of 5-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (piperidin-4-yl) -1H-pyrrolo [2,3-b ] pyridin-4-amine (123mg,0.3718mmol) in ethanol (20mL) was added 6-chloropyridine-3-carbonitrile (103mg,0.74341mmol) and triethylamine (75.5mg,0.746 mmol). The resulting reaction was stirred at 85 ℃ overnight and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) ═ 100/1 to 50/1) to give the title compound as a yellow solid (105mg, yield 65.23%).
MS(ESI,pos.ion)m/z:433.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):11.75(s,1H),8.50(d,J=2.2Hz,1H),8.15(s,1H),7.96(s,1H),7.85(dd,J=9.1,2.3Hz,1H),7.81(s,1H),7.00(d,J=9.2Hz,1H),6.75(d,J=1.8Hz,1H),5.53(d,J=9.0Hz,1H),4.46(d,J=13.0Hz,2H),4.40-4.28(m,1H),3.88(s,3H),3.27(d,J=11.6Hz,2H),2.05(d,J=10.8Hz,2H),1.63(dd,J=21.0,10.6Hz,2H)。
Example 196-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrole-5- Yl) -3H-imidazo [4,5-b]Pyridin-7-amines
Figure BDA0001582369760000631
Step 1)4, 5-dichloro-3-nitropyridin-2-amine
To a suspension of 4-chloro-3-nitro-pyridin-2-amine (5.00g,28.85mmol) in acetonitrile (150mL) was added 1-chloropyrrolidine-2, 5-dione (5.39g,40.36 mmol). The reaction mixture was stirred at 80 ℃ overnight and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (pure DCM) to give the crude product, which was further purified by silica gel column chromatography (EA/PE (v/v) ═ 1/5 to 1/3) to give the title compound as a yellow solid (4.26g, 71% yield).
MS(ESI,pos.ion)m/z:207.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.27(s,1H),5.81(s,2H)。
Step 2)5- ((2-amino-5-chloro-3-nitropyridin-4-yl) amino) hexahydrocyclopenta [ c]Pyrrole-2 (1H) - (iv) Tert-butyl formate
To a suspension of 4, 5-dichloro-3-nitropyridin-2-amine (1.50g,7.23mmol) in isopropanol (30.0mL) was added tert-butyl 5-aminohexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (3.27g,14.4mmol) and DIPEA (4.22g,32.7 mmol). The reaction system is heated to reflux and stirred for 24 hours, and after the reaction is finished, the reaction system is decompressed and concentrated. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 100/1) to afford the title compound as a yellow solid (1.62g, 56% yield).
MS(ESI,pos.ion)m/z:341.9[M-56+H]+
1H NMR(600MHz,CDCl3)δ(ppm):8.79(s,1H),7.83(s,1H),6.73(s,2H),4.72–4.62(m,1H),3.54–3.46(m,2H),3.41–3.27(m,2H),2.71–2.63(m,2H),2.53–2.46(m,2H),1.49(s,9H),1.47–1.43(m,2H)。
Step 3)5- ((6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyrazole-7-yl) Amino) hexahydrocyclopenta [ c ] s]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To a suspension of tert-butyl 5- ((2-amino-5-chloro-3-nitropyridin-4-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (0.62g,1.60mmol) and 1, 3-dimethylpyrazole-4-carbaldehyde (0.22g,1.78mmol) in EtOH (30.0mL) was added a fresh aqueous solution of sodium dithionite (6.30mL,6.30mmol,1.0 mol/L). The reaction was heated to 85 ℃ and stirred overnight, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 30/1 to 20/1) to give the title compound as a white solid (0.50g, 68% yield).
MS(ESI,pos.ion)m/z:472.3[M+H]+
1H NMR(600MHz,CDCl3)δ(ppm):7.87(s,1H),7.72(s,1H),5.49–5.39(m,1H),5.03(d,J=8.4Hz,1H),3.92(s,3H),3.58–3.48(m,2H),3.46–3.33(m,2H),2.80–2.72(m,2H),2.69–2.60(m,5H),1.50(s,9H),1.48–1.40(m,2H)。
Step 4) 6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c)]Pyrrole-5-yl)) - 3H-imidazo [4,5-b ]]Pyridin-7-amines
To 5- ((6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyrazol-7-yl) amino) hexahydrocyclopenta [ c]To a suspension of pyrrole-2 (1H) -carboxylic acid tert-butyl ester (0.54g,1.1mmol) in DCM (15.0mL) was added a solution of hydrogen chloride in ethyl acetate (10.0mL,30.0mmol,3 mol/L). The reaction mixture was stirred at room temperature for 1 hour, and then, concentrated under reduced pressure. The resulting residue was taken up in DCM (20mL) and saturated Na 2CO3The aqueous solution (20mL) was diluted and the resulting mixture was stirred at room temperature for 15 minutes. After standing, the organic layer was separated, and the aqueous layer was extracted with DCM (20mL × 3) and DCM/MeOH (v/v) ═ 10/1(20mL × 3) in this order. The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 20/1 to 10/1 to 5/1) to give the title compound as a light yellow solid (0.36g, 85% yield).
MS(ESI,pos.ion)m/z:371.9[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):12.86(s,1H),8.19(s,1H),7.89(s,1H),5.69(d,J=8.7Hz,1H),5.49–5.35(m,1H),3.84(s,3H),3.28–3.20(m,2H),3.08(dd,J=11.6,3.0Hz,2H),2.84–2.71(m,2H),2.51(s,3H),2.47–2.36(m,2H),1.71–1.59(m,2H)。
Example 201- (5- ((6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyridine (II) Pyridin-7-yl) amino) hexahydrocyclopenta [ c)]Pyrrol-2 (1H) -yl) ethanones
Figure BDA0001582369760000641
To 6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] at 0 deg.C]Pyrrol-5-yl)) -3H-imidazo [4,5-b]To a suspension of pyridin-7-amine (0.12g,0.32mmol) and triethylamine (49.87mg,0.49mmol) in dry DCM (15.0mL) was added acetic anhydride (40.14mg,0.39 mmol). The reaction was allowed to warm to room temperature and stirred for 30 min, then diluted with DCM (50mL) and the resulting mixture was washed successively with water (30 mL. times.2), NaHCO3Aqueous solution (30mL) and saturated aqueous NaCl solution (30 mL). The separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with DCM/MeOH (v/v) ═ 30/1 to 15/1 to afford the title compound as a white solid (75mg, 56% yield).
MS(ESI,pos.ion)m/z:413.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):7.95(s,1H),7.81(s,1H),5.55–5.40(m,1H),5.02(d,J=8.7Hz,1H),3.96(s,3H),3.73–3.61(m,3H),3.44(dd,J=10.8,3.7Hz,1H),2.92–2.74(m,2H),2.74–2.66(m,2H),2.64(s,3H),2.11(s,3H),1.48–1.42(m,2H)。
Example 213- (5- ((6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl-3H-imidazo [4, 5-b)]Pyridine- 7-yl) amino) hexahydrocyclopenta [ c ] s]Pyrrole-2 (1H) -yl) -3-oxopropanenitrile
Figure BDA0001582369760000642
To a suspension of 6-chloro-2- (1, 3-dimethyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] pyrrol-5-yl)) -3H-imidazo [4,5-b ] pyridin-7-amine (0.15g,0.40mmol) in dry DCM (20.0mL) was added 2-cyanoacetic acid (0.052g,0.61mmol), EDCl (0.16g,0.81mmol) and HOAT (0.11g,0.81 mmol). The resulting mixture was stirred at room temperature for 30 minutes, then diluted with DCM (50 mL). The resulting mixture was washed with water (30 mL. times.2) and saturated aqueous NaCl solution (30mL), respectively. The separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 30/1 to 15/1) to give the title compound as a white solid (85mg, 48% yield).
MS(ESI,pos.ion)m/z:439.3[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):7.92(s,1H),7.76(s,1H),5.52–5.42(m,1H),4.99(d,J=8.5Hz,1H),3.94(s,3H),3.78–3.61(m,4H),3.45(s,2H),2.96–2.78(m,2H),2.76–2.65(m,2H),2.62(s,3H),1.48–1.40(m,2H)。
Example 226-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopentane [ c)]Pyrrole-5-yl) -3H- Imidazo [4,5-b ]]Pyridin-7-amines
Figure BDA0001582369760000651
Step 1)5- ((6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyridin-7-yl) amino Radical) hexahydrocyclopenta [ c ] radical]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To a suspension of tert-butyl 5- ((2-amino-5-chloro-3-nitropyridin-4-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (1.00g,2.52mmol) and 1-methylpyrazole-4-carbaldehyde (0.31g,2.80mmol) in EtOH (50.0mL) was added a freshly prepared aqueous solution of sodium dithionate (10.0mL,10.0mmol,1 mol/L). The reaction was heated to 85 ℃ and stirred overnight, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 30/1 to 20/1) to give the title compound as a white solid (0.71g, 62% yield).
MS(ESI,pos.ion)m/z:458.3[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.04(s,1H),8.00(s,1H),7.96(s,1H),5.53–5.41(m,1H),5.08(d,J=8.8Hz,1H),4.03(s,3H),3.60–3.52(m,2H),3.46–3.34(m,2H),2.85–2.73(m,2H),2.69–2.59(m,2H),1.51(s,9H),1.48–1.43(m,J=7.5,4.0Hz,2H)。
Step 2) 6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopentane [ c ]]Pyrrol-5-yl) -3H-imidazoles And [4,5-b ]]Pyridin-7-amines
To 5- ((6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyridin-7-yl) amino) hexahydrocyclopentane [ c ]]To a suspension of pyrrole-2 (1H) -carboxylic acid tert-butyl ester (0.62g,1.40mmol) in DCM (15.0mL) was added a solution of hydrogen chloride in EtOAc (15.0mL,45.0mmol,3.0 mol/L). The resulting mixture was stirred at room temperature for 1 hour, and then, concentrated under reduced pressure. The resulting residue was taken up in DCM (20mL) and saturated Na2CO3The aqueous solution (20mL) was diluted and the resulting mixture was stirred at room temperature for 15 minutes. After standing, the organic layer was separated, and the aqueous layer was extracted with DCM (20mL × 3) and DCM/MeOH (v/v) ═ 10/1(20mL × 3) in this order. The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was chromatographed on silica gel (DCM/2 MNH)3Purified with MeOH (v/v) ═ 20/1 to 10/1 to 5/1 to afford the title compound as a white solid (0.32g, 66% yield).
MS(ESI,pos.ion)m/z:358.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.00(s,1H),7.99(s,1H),7.84(s,1H),5.36–5.25(m,1H),3.96(s,3H),2.88–2.82(m,4H),2.75–2.65(m,2H),2.55–2.44(m,2H),1.30–1.23(m,2H)。
Example 233- (5- ((6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyridine-7- Yl) amino) hexahydrocyclopenta [ c ] s]Pyrrole-2 (1H) -yl) -3-oxopropanenitrile
Figure BDA0001582369760000661
To a suspension of 6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] pyrrol-5-yl) -3H-imidazo [4,5-b ] pyridin-7-amine (0.10g,0.28mmol) in dry DCM (10.0mL) were added EDCI (0.11g,0.60mmol), HOAT (0.077g,0.57mmol) and 2-cyanoacetic acid (0.049g,0.58 mmol). The resulting mixture was stirred at room temperature overnight and then diluted with DCM (50 mL). The resulting mixture was washed with water (20mL) and saturated aqueous NaCl solution (20mL), respectively. The separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 20/1 to 15/1) to give the title compound as a pale yellow solid (64mg, 54% yield).
MS(ESI,pos.ion)m/z:425.0[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.07(s,1H),8.04(s,1H),8.01(s,1H),5.57–5.44(m,1H),5.12(d,J=8.5Hz,1H),4.05(s,3H),3.82–3.70(m,2H),3.68–3.62(m,1H),3.52–3.50(m,1H),3.48(d,J=1.9Hz,2H),3.03–2.81(m,2H),2.77–2.66(m,2H),1.54–1.41(m,2H)。
Example 241- (5- ((6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -3H-imidazo [4, 5-b)]Pyridine-7- Yl) amino) hexahydrocyclopentane [ c ]]Pyrrol-2 (1H) -yl) ethanones
Figure BDA0001582369760000662
To a suspension of 6-chloro-2- (1-methyl-1H-pyrazol-4-yl) -N- (octahydrocyclopenta [ c ] pyrrol-5-yl) -3H-imidazo [4,5-b ] pyridin-7-amine (0.090g,0.25mmol) in dry DCM (10.0mL) was added triethylamine (0.040g,0.39mmol) and acetic anhydride (0.032g,0.32 mmol). The reaction was stirred at room temperature for 30 minutes, then diluted with DCM (50mL) and the resulting mixture was washed successively with water (20mL) and saturated aqueous NaCl solution (30 mL). The separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 20/1) to give the title compound as a white solid (86mg, 85% yield).
MS(ESI,pos.ion)m/z:400.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.06(s,1H),8.03(s,1H),7.99(s,1H),5.56–5.44(m,1H),5.05(d,J=8.7Hz,1H),4.05(s,3H),3.74–3.65(m,2H),3.61(dd,J=12.5,4.2Hz,1H),3.45(dd,J=10.8,3.8Hz,1H),2.92–2.79(m,2H),2.74–2.64(m,2H),2.11(s,3H),1.50–1.42(m,2H)。
Example 252- (1-methyl-1H-pyrazol-4-yl) -8- ((octahydrocyclopentane [ c)]Pyrrol-5-yl) amino) imidazoles And [1,2-b ]]Pyridazine-7-carboxamides
Figure BDA0001582369760000671
Step 1) 1-amino-1H-imidazole-2-carboxylic acid ethyl ester
To a suspension of ethyl 1H-imidazole-2-carboxylate (40.01g,285.5mmol) in dry THF (600mL) at 0 deg.C was added sodium hydride (13.82g,345.5mmol, mass fraction 60%). The reaction mixture was stirred at room temperature for 1 hour, then cooled to 0 ℃ and O- (2, 4-dinitrophenyl) hydroxylamine (80.00g,401.8mmol) was added portionwise. The reaction was stirred at room temperature overnight, then diluted in water (600mL), the resulting mixture was extracted with EA (600 mL. times.10), and the separated organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EA/PE (v/v) ═ 1/2 to 1/1 to 2/1) to give the title compound as a brown solid (37.6g, 85% yield).
MS(ESI,pos.ion)m/z:156.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):7.19(s,1H),7.07(s,1H),5.82(s,2H),4.43(q,J=7.1Hz,2H),1.44(t,J=7.1Hz,3H)。
Step 2)1- ((tert-butoxycarbonyl) amino) -1H-imidazole-2-carboxylic acid ethyl ester
To a suspension of ethyl 1-amino-1H-imidazole-2-carboxylate (9.20g,59.3mmol) in DMF (100mL) was added N, N-dimethylpyridin-4-amine (1.45g,11.89mmol), triethylamine (12.40mL,88.90mmol) and di-tert-butyl dicarbonate (15.65g,71.69 mmol). The reaction was warmed to 85 ℃ and stirred overnight, then concentrated under reduced pressure. The resulting residue was subjected to silica gel column chromatography (EA/PE (v/v) ═ 1/4 to 1/2 to 1/1) to give the title compound as a pale yellow solid (11.2g, yield 74%).
MS(ESI,pos.ion)m/z:256.3[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):7.92(s,1H),7.24(s,1H),7.14(d,J=0.7Hz,1H),4.40(q,J=7.1Hz,2H),1.50(s,9H),1.41(t,J=7.1Hz,3H)。
Step 3) 4-bromo-1- ((tert-butoxycarbonyl) amino) -1H-imidazole-2-carboxylic acid ethyl ester
To a suspension of ethyl 1- ((tert-butoxycarbonyl) amino) -1H-imidazole-2-carboxylate (12.50g,48.97mmol) in DMF (100mL) was added 1-bromopyrrolidine-2, 5-dione (8.73g,49.03 mmol). The reaction was stirred at room temperature overnight and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EA/PE (v/v) ═ 1/8 to 1/6 to 1/4) to give the title compound as a pale yellow viscous liquid (12.45g, yield 76%). 2.10g of by-product are simultaneously obtained as a yellow solid.
MS(ESI,pos.ion)m/z:333.8[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.08(s,1H),7.22(s,1H),4.38(q,J=7.1Hz,2H),1.48(s,9H),1.38(t,J=7.1Hz,3H)。
By-products:
5-bromo-1- ((tert-butoxycarbonyl) amino) -1H-imidazole-2-carboxylic acid ethyl ester
MS(ESI,pos.ion)m/z:334.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):7.72(s,1H),7.15(s,1H),4.37(q,J=7.1Hz,2H),1.48(s,9H),1.38(t,J=7.1Hz,3H)。
Step 4) Ethyl 3- (4-bromo-1- ((tert-butoxycarbonyl) amino) -1H-imidazol-2-yl) -3-oxopropanoate
A suspension of 4-bromo-1- ((tert-butoxycarbonyl) amino) -1H-imidazole-2-carboxylic acid ethyl ester (0.35g,1.06mmol) in dry THF (1.0mL) was cooled to 0 deg.C, then a pre-cooled solution of potassium tert-butoxide in tetrahydrofuran (3.20mL,3.20mmol,1mol/L) was added thereto. The reaction mixture was stirred at 0 ℃ for 5 minutes, to which was added previously cooled dry ethyl acetate (0.26mL,2.70mmol) at 0 ℃. The resulting mixture was stirred at 0 ℃ for 15 minutes, then allowed to warm to room temperature and stirred overnight. After the reaction was complete, the reaction was cooled to 0 ℃ and then HCl (1M,1.0mL) was added and the reaction became clear with a pH of 7. The resulting mixture was concentrated under reduced pressure to remove most of the solvent, and the resulting residue was extracted with ethyl acetate (10 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (EA/PE (v/v) ═ 5/1 to 3/1) to give the title compound as a white solid (0.23g, 58% yield).
MS(ESI,pos.ion)m/z:375.8[M+H]+
1H NMR(600MHz,CDCl3)δ(ppm):8.25(br,1H),7.33(s,1H),4.20(q,J=7.1Hz,2H),4.07(s,2H),1.50(s,9H),1.26(t,J=7.1Hz,3H)。
Step 5) 2-bromo-8-oxo-5, 8-dihydroimidazo [1,2-b]Pyridazine-7-carboxylic acid ethyl ester
To a suspension of ethyl 3- (4-bromo-1- ((tert-butoxycarbonyl) amino) -1H-imidazol-2-yl) -3-oxopropanoate (1.60g,4.25mmol) in DCM (15.0mL) was added N, N-dimethylformamide dimethyl acetal (1.57g,13.20 mmol). The reaction was stirred overnight at room temperature and then concentrated under reduced pressure. The residue was used in the next reaction without purification (1.22g, 100% yield).
MS(ESI,pos.ion)m/z:285.8[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.53(s,1H),7.63(s,1H),4.30(q,J=7.1Hz,2H),1.39(t,J=7.1Hz,3H)。
Step 6) 2-bromo-8-oxo-5, 8-dihydroimidazo [1,2-b]Pyridazine-7-carboxylic acid
To a suspension of ethyl 2-bromo-8-oxo-5, 8-dihydroimidazo [1,2-b ] pyridazine-7-carboxylate (1.22g,4.26mmol) in EtOH (15.0mL) was added an aqueous solution of potassium hydroxide (4.30mL,30.10mmol,7 mol/L). The mixture was heated to 100 ℃ for 1 hour and then concentrated under reduced pressure. The resulting residue was diluted with water and then washed with EA (20 mL). The aqueous phase was adjusted to pH 4 with HCl (1M), then filtered, and the filter cake was washed with cold water (5.0mL) and dried under reduced pressure to give the title compound as a pale yellow solid (0.76g, 69% yield).
MS(ESI,neg.ion)m/z:255.9[M-H]-
1H NMR(400MHz,DMSO-d6)δ(ppm):15.94(s,1H),8.27(s,1H),8.02(s,1H)。
Step 7) 2-bromo-8-chloroimidazo [1,2-b]Pyridazine-7-carboxamides
To solid 2-bromo-8-hydroxy-imidazo [1,2-b]To pyridazine-7-carboxylic acid (0.72g,2.80mmol) was added phosphorus oxychloride (15.0mL) and DIPEA (0.50mL,3.00 mmol). The reaction was allowed to react overnight at 120 ℃ under nitrogen, after completion of the reaction, concentrated under reduced pressure, and the resulting residue was diluted with THF (15mL) and stirred for 5 minutes, and concentrated under reduced pressure. This procedure was repeated three times, the resulting residue was diluted with THF (10mL), then cooled to-40 deg.C, and NH was added thereto3A THF solution (20mL,3.0 mol/L). The resulting mixture was stirred at-40 ℃ for 15 minutes, then allowed to warm to room temperature, stirred for 20 minutes, and concentrated under reduced pressure. The resulting residue was diluted with water (20mL) and stirred for 30 min, filtered, and the filter cake was washed with water (10mL) and MTBE (10mL) in that order and dried under reduced pressure to give the title compound as a pale yellow solid (0.39g, 51% yield). MS (ESI, pos. ion) M/z 274.8[ M + H ] ]+
1H NMR(400MHz,DMSO-d6)δ(ppm):8.73(s,1H),8.64(s,1H),8.18(br.s,1H),8.07(br.s,1H)。
Step 8)5- ((2-bromo-7-carbamoylimidazo [1, 2-b)]Pyridazin-8-yl) amino) hexahydrocyclopenta [c]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To a suspension of 2-bromo-8-chloro-imidazo [1,2-b ] pyridazine-7-carboxamide (0.39g,1.40mmol) in N-BuOH (10.0mL) was added 5-aminohexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylic acid tert-butyl ester (0.66g,2.91mmol) and N-methyl-N-isopropyl-propan-2-amine (0.50mL,3.00 mmol). The reaction was heated to reflux for 12 hours and then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 30/1 to 20/1) to afford the title compound as a brown solid (0.58g, 88% yield).
MS(ESI,pos.ion)m/z:464.8[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):10.54(d,J=8.4Hz,1H),8.48(s,1H),8.29(s,1H),7.97(s.br,1H),7.39(s.br,1H),5.40-5.26(m,1H),3.46–3.37(m,2H),3.26–3.17(m,2H),2.72–2.61(m,2H),2.48–2.40(m,2H),1.40(s,9H),1.36–1.26(m,2H)。
Step 9)5- ((7-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) imidazo [1, 2-b)]Pyridazin-8-yl radicals Amino) hexahydrocyclopenta [ c ] s]Pyrrole-2 (1H) -carboxylic acid tert-butyl ester
To a suspension of tert-butyl 5- ((2-bromo-7-carbamoylimidazo [1,2-b ] pyridazin-8-yl) amino) hexahydrocyclopenta [ c ] pyrrole-2 (1H) -carboxylate (0.56g,1.20mmol) in DME (15.0mL) and water (3.0mL) were added 1-methyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazole (0.51g,2.44mmol), 1' -bis (diphenylphosphino) ferrocene (II) palladium dichloride dichloromethane complex (0.20g,0.24mmol) and cesium carbonate (0.79g,2.43 mmol). The reaction mixture was evacuated of air and purged with nitrogen several times, then warmed to reflux and stirred overnight. The reaction mixture was concentrated under reduced pressure. The resulting residue was dissolved in water (20mL), then extracted with DCM (30mL × 5), and the combined organic phases were washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 30/1) to afford the title compound as a brown solid (0.29g, 52% yield).
MS(ESI,pos.ion)m/z:467.4[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.07(d,J=8.2Hz,1H),8.06(s,1H),7.85(s,1H),7.81(s,1H),7.76(s,1H),5.81–5.66(m,3H),3.99(s,3H),3.60–3.50(m,2H),3.44–3.29(m,2H),2.82–2.71(m,2H),2.69–2.58(m,2H),1.53–1.46(m,11H)。
Step 10)2- (1-methyl-1H-pyrazol-4-yl) -8- ((octahydrocyclopenta [ c)]Pyrrol-5-yl) amino) imidazoles And [1,2-b ]]Pyridazine-7-carboxamides
To 5- ((7-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) imidazo [1, 2-b)]Pyridazin-8-yl) amino) hexahydrocyclopenta [ c)]To a suspension of tert-butyl pyrrole-2 (1H) -carboxylate (0.29g,0.62mmol) in DCM (15.0mL) was added a solution of hydrogen chloride in ethyl acetate (15.0mL,45.0mmol,3 mol/L). The reaction mixture was stirred at room temperature for 2 hours, and then concentrated under reduced pressure. The resulting residue was taken up in DCM (20mL) and saturated Na2CO3The aqueous solution (20mL) was diluted and the resulting mixture was stirred at room temperature for 15 minutes. After standing, the organic layer was separated, and the aqueous layer was extracted with DCM (20 mL. times.3). The aqueous layer was concentrated under reduced pressure and the resulting solid was washed with MeOH (100mL), then all organic phases were combined and concentrated under reduced pressure. The residue was chromatographed on silica gel (DCM/3M NH)3Purified with MeOH (v/v) ═ 30/1 to 20/1 to 10/1 to afford the title compound as a light yellow solid (0.17g, 75% yield).
MS(ESI,pos.ion)m/z:367.3[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):10.41–10.27(m,1H),8.39(s,1H),8.20(s,1H),8.07(s,1H),7.98–7.69(m,2H),7.27(br.s,1H),5.46–5.30(m,1H),3.89(s,3H),2.69–2.53(m,6H),2.40(dd,J=12.1,6.1Hz,2H),1.23–1.13(m,2H)。
Example 268- ((2- (2-cyanoacetyl) octahydrocyclopentane [ c)]Pyrrol-5-yl) amino) -2- (1-methyl- 1H-pyrazol-4-yl) imidazo [1,2-b]Pyridazine-7-carboxamides
Figure BDA0001582369760000691
To 2- (1-methyl-1H-pyrazolyl-4-yl) -8- ((octahydrocyclopenta [ c) ]Pyrrol-5-yl) amino) imidazo [1,2-b]To a suspension of pyridazine-7-carboxamide (61mg,0.17mmol) in DCM (1.0mL) were added EDCI (65mg,0.34mmol), HOAT (47mg,0.34mmol), 2-cyanoAcetic acid (29mg,0.34mmol) and Et3N (38mg,0.38 mmol). The resulting mixture was stirred at room temperature for 2 hours, and then concentrated under reduced pressure. The residue was chromatographed on silica gel (DCM/3M NH)3Purified with MeOH (v/v) ═ 50/1 to 30/1 to afford the title compound as a white solid (48mg, 66% yield).
MS(ESI,pos.ion)m/z:434.4[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.11–8.07(m,1H),7.76(s,1H),7.75–7.72(m,1H),7.70(s,1H),5.69–5.60(m,1H),3.89(d,J=1.6Hz,3H),3.73–3.65(m,1H),3.65–3.56(m,2H),3.55–3.51(m,1H),3.51–3.45(m,1H),3.44–3.38(m,2H),2.95–2.85(m,1H),2.83–2.73(m,1H),2.61–2.50(m,2H),1.59–1.46(m,2H)。
Example 278- ((2-Acetyloctahydrocyclopenta [ c)]Pyrrol-5-yl) amino) -2- (1-methyl-1H-pyri-dine Azol-4-yl) imidazo [1,2-b]Pyridazine-7-carboxamides
Figure BDA0001582369760000701
To a suspension of 2- (1-methyl-1H-pyrazolyl-4-yl) -8- ((octahydrocyclopenta [ c ] pyrrol-5-yl) amino) imidazo [1,2-b ] pyridazine-7-carboxamide (65mg,0.18mmol) in DCM (5.0mL) were added acetic anhydride (23mg,0.22mmol) and triethylamine (29mg,0.29 mmol). The reaction was allowed to warm to room temperature and stirred for 30 minutes, then concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 30/1 to 20/1) to give the title compound as a white solid (65mg, 90% yield).
MS(ESI,pos.ion)m/z:408.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.08(s,1H),7.75(s,1H),7.73(s,1H),7.70(s,1H),5.69–5.57(m,1H),3.88(s,3H),3.67–3.61(m,1H),3.59–3.52(m,1H),3.47–3.44(m,1H),3.38–3.33(m,1H),3.29(s,2H),2.84–2.70(m,2H),2.62–2.52(m,2H),2.00(s,3H),1.49–1.36(m,2H)。
Example 288- (3-azaspiro [5.5 ]]Undecane-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) imidazole And [1,2-b ]]Pyridazine-7-carboxamides
Figure BDA0001582369760000702
Step 1)9- ((2-bromo-7-carbamoylimidazo [1, 2-b)]Pyridazin-8-yl) amino) -3-azaspiro [5.5] Undecane-3-carboxylic acid tert-butyl ester
To 2-bromo-8-chloro-imidazo [1, 2-b)]To a suspension of pyridazine-7-carboxamide (0.40g,1.46mmol) in n-BuOH (20.0mL) was added 9-amino-3-azaspiro [ 5.5%]Tert-butyl undecane-3-carboxylate (0.60g,2.23mmol) and DIPEA (0.50mL,3.00 mmol). The resulting mixture was warmed to reflux and stirred overnight, then concentrated under reduced pressure. The residue was chromatographed on silica gel (DCM/3M NH)3Purified with MeOH (v/v) ═ 50/1 to 30/1 to 20/1 to afford the title compound as a yellow solid (0.56g, 76% yield).
MS(ESI,pos.ion)m/z:451.2[M-56+H]+
Step 2)9- ((7-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) imidazo [1, 2-b)]Pyridazin-8-yl radicals Amino) -3-azaspiro [5.5]Undecane-3-carboxylic acid tert-butyl ester
To a suspension of tert-butyl 9- ((2-bromo-7-carbamoylimidazo [1,2-b ] pyridazin-8-yl) amino) -3-azaspiro [5.5] undecane-3-carboxylate (0.53g,1.00mmol0) in DME (15.0mL) and water (3.0mL) were added 1-methyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazole (0.44g,2.12mmol), 1' -bis (diphenylphosphino) ferrocene (II) dichloropalladium dichloromethane complex (0.18g,0.21mmol) and cesium carbonate (0.69g,2.13 mmol). The reaction mixture was evacuated of air and purged with nitrogen several times, then warmed to reflux and stirred overnight. The reaction mixture was concentrated under reduced pressure. The resulting residue was dissolved in water (20mL), then extracted with DCM (30mL × 5), and the combined organic phases were washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) ═ 50/1 to 30/1) to afford the title compound as a brown solid (0.42g, 79% yield).
MS(ESI,pos.ion)m/z:509.4[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):9.99(d,J=8.3Hz,1H),8.03(s,1H),7.84(s,1H),7.77(s,1H),7.72(s,1H),5.67(s,2H),5.28–5.19(m,1H),3.96(s,3H),3.47–3.32(m,4H),2.12–1.98(m,2H),1.75–1.70(m,2H),1.63–1.49(m,4H),1.46(s,9H),1.44–1.37(m,4H)。
Step 3)8- (3-azaspiro [5.5 ]]Undecan-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) imidazo [1,2-b]Pyridazine-7-carboxamides
To 9- ((7-carbamoyl-2- (1-methyl-1H-pyrazol-4-yl) imidazo [1, 2-b)]Pyridazin-8-yl) amino) -3-azaspiro [5.5]Tert-butyl undecane-3-carboxylate (0.42g,0.83mmol) in DCM (15.0mL) was added a solution of hydrogen chloride in ethyl acetate (15.0mL,45.0mmol,3 mol/L). The reaction mixture was stirred at room temperature for 30 minutes, and then concentrated under reduced pressure. The resulting residue was taken up in DCM (20mL) and saturated Na2CO3Aqueous solution (20mL) was diluted, the resulting mixture was stirred at room temperature for 15 minutes, then concentrated under reduced pressure, and the resulting solid was rinsed with MeOH (100 mL). The filter cake was concentrated under reduced pressure and the residue was chromatographed on silica gel (DCM/3M NH)3Purified with MeOH (v/v) ═ 30/1 to 20/1 to 10/1 to afford the title compound as a light yellow solid (0.18g, 53% yield).
MS(ESI,pos.ion)m/z:409.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):10.41(d,J=8.4Hz,1H),8.40(s,1H),8.20(s,1H),8.04(s,1H),8.00–7.74(m,2H),7.27(s,br,1H),5.21–5.09(m,1H),3.89(s,3H),3.45–3.38(m,1H),2.74–2.62(m,3H),1.97–1.87(m,2H),1.73–1.63(m,2H),1.53–1.39(m,4H),1.38–1.28(m,4H)。
Example 298- ((3- (2-cyanoacetyl) -3-azaspiro [ 5.5)]Undecane-9-yl) amino) -2- (1-methyl yl-1H-pyrazol-4-yl) imidazo [1,2-b]Pyridazine-7-carboxamides
Figure BDA0001582369760000711
To 8- (3-azaspiro [5.5 ]]Undecan-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) imidazo [1,2-b]To a suspension of pyridazine-7-carboxamide (0.11g,0.26mmol) in DCM (10.0mL) were added 2-cyanoacetic acid (47mg,0.55mmol), HATU (0.21g,0.54mmol) and triethylamine (0.15mL,1.10 mmol). The resulting mixture was stirred at room temperature for 4 hours, and then concentrated under reduced pressure. The residue was chromatographed on silica gel (DCM/3M NH) 3Purified with MeOH (v/v) ═ 50/1 to 30/1 to 20/1 to give the title compound as a white solid (55mg, 43% yield).
MS(ESI,pos.ion)m/z:476.4[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.01(d,J=8.1Hz,1H),8.08(s,1H),7.78(d,J=3.4 Hz,1H),7.73(s,1H),7.70(d,J=1.5Hz,1H),5.25–5.14(m,1H),3.91(s,3H),3.59–3.51(m,2H),3.51–3.47(m,1H),3.42–3.34(m,2H),3.34–3.31(m,1H),2.08–1.99(m,2H),1.77–1.67(m,2H),1.66–1.35(m,8H)。
Example 308- ((3-acetyl-3-azaspiro [ 5.5)]Undecane-9-yl) amino) -2- (1-methyl-1H-pyri-dine Azol-4-yl) imidazo [1,2-b]Pyridazine-7-carboxamides
Figure BDA0001582369760000712
To 8- (3-azaspiro [5.5 ]]Undecan-9-ylamino) -2- (1-methyl-1H-pyrazol-4-yl) imidazo [1,2-b]To a suspension of pyridazine-7-carboxamide (50mg,0.12mmol) in DCM (10.0mL) were added acetic anhydride (15mg,0.15mmol) and triethylamine (20mg,0.19 mmol). The reaction was allowed to warm to room temperature and stirred for 30 minutes, then concentrated under reduced pressure. The residue was chromatographed on silica gel (DCM/3M NH)3Purified with MeOH (v/v) ═ 50/1 to 30/1 to 20/1 to give the title compound as a white solid (42mg, 75% yield).
MS(ESI,pos.ion)m/z:450.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):10.00(d,J=8.1Hz,1H),8.01(s,1H),7.85(d,J=9.1Hz,1H),7.78(d,J=2.3Hz,1H),7.72(d,J=5.4Hz,1H),5.56(s,2H),5.34–5.20(m,1H),3.97(s,3H),3.63–3.53(m,2H),3.47–3.37(m,2H),2.13–2.03(m,5H),1.82–1.72(m,2H),1.60–1.51(m,4H),1.50–1.40(m,4H)。
Biological assay
The LC/MS/MS system for analysis included an Agilent 1200 series vacuum degassing furnace, a binary injection pump, an orifice plate autosampler, a column oven, an Agilent G6430 three-stage quadrupole mass spectrometer with an electrospray ionization (ESI) source. The quantitative analysis was performed in MRM mode, with the parameters of the MRM transition as shown in table a:
TABLE A
Multiple reaction detection scan 490.2→383.1
Fragmentation voltage 230V
Capillary voltage 55V
Temperature of drying gas 350℃
Atomizer 0.276MPa
Dry air flow rate 10L/min
Analysis 5. mu.L of sample was injected using an Agilent XDB-C18, 2.1X 30mm, 3.5. mu.M column. Analysis conditions were as follows: the mobile phase was 0.1% aqueous formic acid (A) and 0.1% methanolic formic acid (B). The flow rate was 0.4 mL/min. Mobile phase gradients are shown in table B:
TABLE B
Time Gradient of mobile phase B
0.5min 5%
1.0min 95%
2.2min 95%
2.3min 5%
5.0min Terminate
Also used for the analysis was an Agilent 6330 series LC/MS spectrometer equipped with a G1312A binary syringe pump, a G1367A auto sampler and a G1314C UV detector; the LC/MS/MS spectrometer uses an ESI radiation source. The appropriate cation model treatment and MRM conversion for each analyte was performed using standard solutions for optimal analysis. During the analysis a Capcell MP-C18 column was used, with the specifications: 100X 4.6mm I.D., 5. mu.M (Phenomenex, Torrance, California, USA). The mobile phase was 5mM ammonium acetate, 0.1% aqueous methanol (a): 5mM ammonium acetate, 0.1% methanolic acetonitrile solution (B) (70:30, v/v); the flow rate is 0.6 mL/min; the column temperature was kept at room temperature; 20 μ L of sample was injected.
Example A stability in human and rat liver microsomes
Human or rat liver microsomes were incubated in duplicate wells in polypropylene tubes. A typical incubation mixture comprises human or rat liver microsomes (0.5mg protein/mL), the compound of interest (5. mu.M) and a total volume of 200. mu.L of NADPH (1.0mM) potassium phosphate buffer (PBS, 100mM, pH 7.4), and the test compound is dissolved in DMSO and diluted with PBS to give a final DMSO solution concentration of 0.05%. And incubated in a water bath at 37 ℃ in air communication, and after 3 minutes of pre-incubation, protein was added to the mixture and the reaction was started. At various time points (0, 5, 10, 15, 30 and 60 minutes), the reaction was stopped by the addition of the same volume of ice-cold acetonitrile. The samples were stored at-80 ℃ until LC/MS/MS analysis.
The concentration of compound in the human or rat liver microsome incubation mixture was determined by the method of LC/MS. The linear range of concentration ranges is determined for each test compound.
Parallel incubation experiments were incubated at 37 ℃ using denatured microsomes as negative controls, and reactions were terminated at different time points (0,15 and 60 min).
Verapamil (1 μm) was used as a positive control, incubated at 37 ℃ and the reaction terminated at different time points (0, 5, 10, 15, 30 and 60 min). Positive and negative control samples were included in each assay method to ensure integrity of the microsomal incubation system.
In addition, stability data for the compounds of the invention in human or rat liver microsomes can also be obtained from the following assays. Human or rat liver microsomes were incubated in duplicate wells in polypropylene tubes. A typical incubation mixture comprises human or rat liver microsomes (final concentration: 0.5mg protein/mL), the test compound (final concentration: 1.5. mu.M) and a total volume of 30. mu.L of potassium phosphate buffer (containing 1.0mM EDTA,100mM, pH 7.4). The test compound was dissolved in DMSO and diluted with potassium phosphate buffer solution to make the final concentration of DMSO 0.2%. After a pre-incubation time of 10 minutes, 15. mu.L of NADPH (final concentration: 2mM) was added for the enzymatic reaction, and the whole assay was carried out in an incubation tube at 37 ℃. At various time points (0,15, 30 and 60 minutes), the reaction was stopped by the addition of 135. mu.L acetonitrile (containing IS). The supernatant was collected by centrifugation at 4000rpm for 10 minutes to remove protein and analyzed by LC-MS/MS. In the above assay, ketanserin (1 μ M) was selected as a positive control, incubated at 37 ℃ and the reaction terminated at different time points (0,15, 30 and 60 min). Positive and negative control samples were included in each assay method to ensure integrity of the microsomal incubation system.
Data analysis
For each reaction, the concentration of compound (in percent) in human or rat liver microsome incubations was plotted as a percentage of the zero time point to infer intrinsic hepatic clearance CL in vivoint(ref.:Naritomi Y,Terashita S,Kimura S,Suzuki A,Kagayama A,Sugiyama Y.Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans.Drug Metabolism and Disposition 2001,29:1316-1324.)。
The compounds of the invention exhibit suitable stability when incubated in human and rat liver microsomes.
Example B pharmacokinetics of mice, rats, dogs and monkeys after oral or intravenous administration of a quantitative amount of a compound of the present invention Evaluation of
Pharmacokinetic studies of the compounds of the invention in mice, rats, dogs or monkeys were evaluated. The compounds of the invention are administered as aqueous solutions or 2% HPMC + 1% Tween-80 aqueous solution, 5% DMSO + 5% saline solution, 4% MC or capsules. For intravenous administration, animals are given a dose of 1 or 2 mg/kg. For oral doses (p.o.), rats and mice were 5 or 10mg/kg, and dogs and monkeys were 10 mg/kg. Blood (0.3mL) was taken at time points of 0.25, 0.5, 1.0, 2.0, 3.0, 4.0, 6.0, 8.0, 12 and 24 hours and centrifuged at 3,000 or 4,000rpm for 10 minutes. The plasma solutions were collected and stored at-20 ℃ or-70 ℃ until the LC/MS/MS analysis described above was performed.
When the compound provided by the invention is administrated by intravenous injection or oral administration, the compound shows good pharmacokinetic property, good absorption and ideal half-life (T)1/2) And higher oral bioavailability (F).
Example C kinase Activity assay
The utility of the disclosed compounds as protein kinase inhibitors can be evaluated by the following experiments.
General description of kinase assays
Kinase assays by detecting incorporation of gamma-33Myelin Basic Protein (MBP) of P-ATP. Mu.g/ml MBP (Sigma # M-1891) Tris buffered saline (TBS; 50mM Tris pH 8.0,138mM NaCl,2.7mM KCl) was prepared in 60. mu.L/well in high binding white 384 well plates (Greiner). Incubate at 4 ℃ for 24 hours. The plate was then washed 3 times with 100. mu.L TBS. Kinase reaction A total volume of 34. mu.L of kinase buffer (prepared as required, e.g., 5mM Hepes pH7.6,15mM NaCl, 0.01% bovine serum albumin (Sigma # I-5506),10mM MgCl21mM DTT, 0.02% TritonX-100). Compounds were dissolved in DMSO and added to each well at a final concentration of 1% compound in DMSO solution. The assay for each compound was performed in at least two trials. For example, the final concentration of the enzyme is 10nM or 20 nM. Addition of unlabeled ATP (10. mu.M) and gamma- 33P-labelled ATP (2X 10 per well)6cpm, 3000Ci/mmol) was started. The reaction was performed at room temperature with shaking for 1 hour. The 384 well plates were washed with 7 × PBS and 50 μ L of scintillation fluid per well was added. The results were checked with a Wallac Trilux counter. It will be apparent to those skilled in the art that this is only one of many detection methods, and that other methods are possible.
IC inhibited by the test method50And/or suppression constant Ki。IC50Defined as the concentration of compound that inhibits 50% of the enzyme activity under the conditions testedAnd (4) degree. IC was estimated using a dilution factor of 1/2log to generate a curve containing 10 concentration points50Values (e.g., a typical curve is made by compound concentrations of 3. mu.M, 1. mu.M, 0.3. mu.M, 0.1. mu.M, 0.03. mu.M, 0.01. mu.M, 0.003. mu.M, 0.001. mu.M, 0.0003. mu.M, 0. mu.M).
JAK1(h)
JAK1(h) in 20mM Tris/HCl pH 7.5, 0.2mM EDTA, 500. mu. M GEEPLYWSFPAKKK, 10mM magnesium acetate and [ gamma-33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and washed 3 times with 75mM phosphoric acid in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
JAK2(h)
JAK2(h) at 8mM MOPS pH 7.0, 0.2mM EDTA, 100. mu. M KTFCGTPEYLAPEVRREPRILSEEEQEMFRDFDYIADWC, 10mM magnesium acetate and [ gamma-33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and washed 3 times with 75mM phosphoric acid in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
JAK3(h)
JAK3(h) at 8mM MOPS pH 7.0, 0.2mM EDTA, 500. mu. M GGEEEEYFELVKKKK, 10mM magnesium acetate and [ gamma-33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and washed 3 times with 75mM phosphoric acid in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
TYK2(h)
TYK2(h) pH 7.0 at 8mM MOPS, 0.2mM EDTA, 250. mu. M GGMEDIYFEFMGGKKK,10mM magnesium acetate and [ gamma- 33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and washed 3 times with 75mM phosphoric acid in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
FLT3(h)
FLT3(h) pH 7.0 at 8mM MOPS, 0.2mM EDTA, 50. mu. M EAIYAAPFAKKK,10mM magnesium acetate and [ gamma-33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and washed 3 times with 75mM phosphoric acid in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
Aurora-A(h)
Aurora-A (h) at 8mM MOPS pH 7.0,0.2mM EDTA, 200. mu.M LRRASLG (Kemptide),10mM magnesium acetate and [ gamma-33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and washed 3 times with 75mM phosphoric acid in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
Aurora-B(h)
Aurora-B (h) in 8mM MOPS pH 7.0,0.2mM EDTA, 30. mu. M AKRRRLSSLRA,10mM magnesium acetate and [ gamma-33P-ATP](specific activity about 500cpm/pmol, concentration determined as required) in the presence of conditions for incubation. The reaction was started after the addition of the MgATP mixture. After incubation at room temperature for 40 minutes, a 3% phosphoric acid solution was added thereto to terminate the reaction. 10 μ L of the reaction solution was spotted on a P30 filter and treated with 75mM phosphorusThe acid was washed 3 times in 5 minutes and stored in methanol solution immediately before drying and scintillation counting.
The kinase assay of the present invention was performed by Millipore corporation, UK (Millipore UK Ltd, Dundee Technology Park, Dundee DD 21 SW, UK).
Alternatively, the kinase activity of the compound may be measured by KINOMEscanTMDetection, based on the quantitative detection of compounds using an active site-directed competitive binding assay. The test is carried out by combining with three compounds, namely DNA marker enzyme, immobilized ligand and detection compound, and qPCR is carried out by DNA marker to detect the competitive capacity of the compound and the immobilized ligand.
Most experiments were carried out by culturing a kinase-labeled T7 bacteriophage strain in an E.coli host derived from BL21 strain, infecting E.coli in the logarithmic growth phase with T7 bacteriophage, incubating with shaking at 32 ℃ until lysis, centrifuging the lysate to remove cell debris, transferring the remaining kinase to HEK-293 cells, and carrying out qPCR detection with DNA labeling. Streptavidin-coated particles were treated with biotinylated small molecule ligands for 30min at room temperature to generate affinity resins for kinase assays. The ligand particles are blocked by surplus biotin and then passed through blocking Solution (SEABLOCK) TM(Pierce), 1% bovine serum albumin, 0.05% Tween 20,1mM DTT) washes unbound ligand to reduce non-specific binding. By binding buffer (20% SEABLOCK) at 1XTM0.17 x phosphate buffer, 0.05% tween 20,6mM DTT), ligand affinity particles and test compound were subjected to binding reactions, all in 96 well plates, with a final reaction volume of 0.135mL, incubated at room temperature with shaking for 1 hour, washing the affinity particles with washing buffer (1 x phosphate buffer, 0.05% tween 20), resuspended with elution buffer (1 x phosphate buffer, 0.05% tween 20,0.5 μ M non-biotinylated affinity ligand), incubated at room temperature with shaking for 30min, and the kinase concentration in the eluate was determined by qPCR. The kinase activity assay described herein is KINOMEscan by Discovex Rx, Albrae St.Fremont, CA 94538, USATMDepartment, perform the assay. The results of the kinase activity assay are shown in tables 1 and 2.
Table 1 JAK1 kinase assay results for compounds provided by the invention
Figure BDA0001582369760000752
As can be seen from table 1, the compounds of the present invention showed a very good JAK1 inhibitory activity in the kinase assay.
TABLE 2 JAK2, TYK2, JAK3, Aurora-A, Aurora-B and FLT3 kinase assay results for the compounds provided by the invention
Figure BDA0001582369760000751
NT denotes no test
As can be seen from Table 2, the compounds of the present invention showed better kinase inhibitory activity against JAK2, TYK2, JAK3, Aurora-A, Aurora-B and FLT3 in the kinase assay.
Finally, it should be noted that there are other ways of implementing the invention. Accordingly, the embodiments of the present invention will be described by way of illustration, but not limitation to the description of the present invention, and modifications made within the scope of the present invention or equivalents added to the claims are possible. All publications or patents cited herein are incorporated by reference.

Claims (10)

1. A compound which is a compound represented by formula (I) or a pharmaceutically acceptable salt of the compound represented by formula (I),
Figure FDA0002965089640000011
wherein the content of the first and second substances,
z is:
Figure FDA0002965089640000012
wherein each of the substructures represented by formulae (Z-61) and (Z-71) is independently optionally substituted with 1, 2, 3, 4 or 5R2Substituted by a group;
Z1is H, methyl, ethyl, n-propyl or isopropyl;
when U is turned1When is N, U2Is CH, U3Is N;
when U is turned1When being C, U2Is NH, U3Is N or CH;
each is independently a single bond or a double bond;
a is pyrazolyl optionally substituted by 1, 2 or 3R4Substituted by a group;
R1is H, F, Cl, Br, I, C1-C6Alkyl or- (CR) 6R7)nC(=O)NRaRb
Each R2Independently is C1-C4Alkyl, -C (═ O) CH2CN or-C (═ O) R5Wherein, the C is1-C4Alkyl is optionally substituted by 1, 2 or 3R8Substituted by a group;
R4is methyl, ethyl, n-propyl or isopropyl, wherein each of said methyl, ethyl, n-propyl and isopropyl is independently optionally substituted with 1, 2 or 3R8Substituted by a group;
each R5Independently is C1-C4Alkyl, wherein the above groups are optionally substituted by 1, 2 or 3R8Substituted by a group;
each R6And R7Each independently is H or C1-C6Alkyl, wherein, said C1-C6Alkyl is optionally substituted by 1, 2 or 3R8Substituted by a group;
each R8Independently F, Cl, Br, I or CN;
each RaAnd RbEach independently is H or C1-C6Alkyl, wherein, said C1-C6Alkyl is optionally substituted with 1, 2 or 3 substituents independently selected from F, Cl, Br, CN, OH or NH2Substituted with the substituent(s);
each n is independently 0, 1 or 2.
2. The compound of claim 1, wherein a is:
Figure FDA0002965089640000013
wherein each of the substructures represented by the formulae (A-1) to (A-3) is independently optionally substituted with 1, 2 or 3R4Substituted by a group.
3. The compound of claim 1, which is a compound of one of the following:
Figure FDA0002965089640000021
Figure FDA0002965089640000031
Figure FDA0002965089640000032
or a pharmaceutically acceptable salt thereof.
4. A pharmaceutical composition comprising a compound of any one of claims 1-3, further comprising a pharmaceutically acceptable adjuvant.
5. The pharmaceutical composition of claim 4, further comprising a therapeutic agent selected from the group consisting of chemotherapeutic agents, antiproliferative agents, phosphodiesterase 4 inhibitors, beta 2-adrenoreceptor agonists, corticosteroids, non-steroidal GR agonists, anticholinergics, antihistamines, anti-inflammatory agents, immunosuppressive agents, immunomodulatory agents, agents for treating atherosclerosis, agents for treating pulmonary fibrosis, and combinations thereof.
6. Use of a compound according to any one of claims 1 to 3 or a pharmaceutical composition according to any one of claims 4 to 5 in the manufacture of a medicament for the prevention, treatment or alleviation of a protein kinase mediated disease.
7. The use according to claim 6, wherein the protein kinase-mediated disease is a JAK-mediated disease or an Aurora-mediated disease.
8. The use according to claim 6, wherein the protein kinase mediated disease is a proliferative disease, an autoimmune disease, an allergic disease or an inflammatory disease.
9. The use of claim 6, wherein the protein kinase-mediated disease is colorectal cancer, gastric cancer, esophageal cancer, breast cancer, lung cancer, liver cancer, prostate cancer, pancreatic cancer, thyroid cancer, bladder cancer, kidney cancer, brain tumor, head and neck cancer, cancer of the CNS, glioblastoma, non-small cell lung cancer, cervical cancer, testicular tumor, lymphoid cancer, multiple myeloma, small cell lung cancer, neuroblastoma, neuroendocrine cell tumor, medullary thyroid cancer, melanoma, retinoblastoma, uterine cancer, ovarian cancer, primary macroglobulinemia, monocytic leukemia, Sezary syndrome, infectious mononucleosis, colitis, pancreatitis, atherosclerosis, pulmonary fibrosis, polycythemia vera, primary thrombocythemia, myelofibrosis, acute myelocytic leukemia, colon cancer, chronic myelogenous leukemia, acute lymphocytic leukemia, chronic obstructive pulmonary disease, asthma, systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, sjogren's syndrome, psoriasis, type I diabetes, respiratory allergic disease, sinusitis, eczema, measles, food allergy, insect venom allergy, inflammatory bowel disease, crohn's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection, or cell transplant rejection.
10. Use of a compound according to any one of claims 1 to 3 or a pharmaceutical composition according to any one of claims 4 to 5 in the manufacture of a medicament for modulating the activity of a protein kinase, wherein the protein kinase is JAK1 kinase, JAK2 kinase, JAK3 kinase, Aurora-a kinase, Aurora-B kinase, Aurora-C kinase, or a combination thereof.
CN201810159088.XA 2017-03-10 2018-02-26 Substituted heteroaryl compounds, compositions and uses thereof Active CN108570048B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710144188 2017-03-10
CN2017101441880 2017-03-10

Publications (2)

Publication Number Publication Date
CN108570048A CN108570048A (en) 2018-09-25
CN108570048B true CN108570048B (en) 2021-06-08

Family

ID=63576677

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810159088.XA Active CN108570048B (en) 2017-03-10 2018-02-26 Substituted heteroaryl compounds, compositions and uses thereof

Country Status (1)

Country Link
CN (1) CN108570048B (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111138412B (en) * 2018-11-06 2023-09-15 上海奕拓医药科技有限责任公司 Spiro aromatic ring compound and application thereof
WO2020156271A1 (en) * 2019-02-02 2020-08-06 江苏威凯尔医药科技有限公司 Janus kinase (jak) family inhibitor, preparation of same, and applications thereof
CN111518096B (en) * 2019-02-02 2022-02-11 江苏威凯尔医药科技有限公司 Janus kinase JAK family inhibitor and preparation and application thereof
CN111662295B (en) * 2019-03-05 2021-09-10 珠海宇繁生物科技有限责任公司 IRAK4 kinase inhibitor and preparation method thereof
JP7248256B2 (en) * 2019-03-14 2023-03-29 上海華匯拓医薬科技有限公司 JAK Kinase Inhibitors, Preparation Methods Thereof, and Uses Thereof in the Pharmaceutical Field
CN114269736B (en) * 2020-11-26 2024-02-02 科辉智药(深圳)新药研究中心有限公司 Amide compound, pharmaceutical composition and application thereof
CN114591321A (en) * 2020-12-04 2022-06-07 广州费米子科技有限责任公司 Aza-and-cyclic compounds, method for the production thereof and use thereof
CN117800895A (en) * 2022-09-30 2024-04-02 苏州阿尔脉生物科技有限公司 Oxalic acid amine derivative, pharmaceutical composition containing oxalic acid amine derivative and medical application of oxalic acid amine derivative

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101052629A (en) * 2004-08-02 2007-10-10 Osi制药公司 Aryl-amino substituted pyrrolopyrimidine multi-kinase inhibiting compounds
CN101223168A (en) * 2005-07-14 2008-07-16 安斯泰来制药株式会社 Heterocyclic JANUS kinase 3 inhibitors
WO2012125893A1 (en) * 2011-03-17 2012-09-20 Bristol-Myers Squibb Company Pyrrolopyridazine jak3 inhibitors and their use for the treatment of inflammatory and autoimmune diseases
WO2013116291A1 (en) * 2012-01-30 2013-08-08 Cephalon, Inc. Imidazo [4, 5 - b] pyridine derivatives as alk and jak modulators for the treatment of proliferative disorders
WO2014039595A1 (en) * 2012-09-06 2014-03-13 Bristol-Myers Squibb Company Imidazopyridazine jak3 inhibitors and their use for the treatment of inflammatory and autoimmune diseases

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101052629A (en) * 2004-08-02 2007-10-10 Osi制药公司 Aryl-amino substituted pyrrolopyrimidine multi-kinase inhibiting compounds
CN101223168A (en) * 2005-07-14 2008-07-16 安斯泰来制药株式会社 Heterocyclic JANUS kinase 3 inhibitors
WO2012125893A1 (en) * 2011-03-17 2012-09-20 Bristol-Myers Squibb Company Pyrrolopyridazine jak3 inhibitors and their use for the treatment of inflammatory and autoimmune diseases
WO2013116291A1 (en) * 2012-01-30 2013-08-08 Cephalon, Inc. Imidazo [4, 5 - b] pyridine derivatives as alk and jak modulators for the treatment of proliferative disorders
WO2014039595A1 (en) * 2012-09-06 2014-03-13 Bristol-Myers Squibb Company Imidazopyridazine jak3 inhibitors and their use for the treatment of inflammatory and autoimmune diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Aurora Isoform Selectivity: Design and Synthesis of Imidazo[4,5-b]pyridine Derivatives as Highly Selective Inhibitors of Aurora-A Kinase in Cells;Vassilios Bavetsias 等;《Journal of Medicinal Chemistry》;20131106;第56卷(第22期);9122-9135 *

Also Published As

Publication number Publication date
CN108570048A (en) 2018-09-25

Similar Documents

Publication Publication Date Title
CN108570048B (en) Substituted heteroaryl compounds, compositions and uses thereof
US9399637B2 (en) Substituted heteroaryl compounds and methods of use
US10059689B2 (en) Substituted heteroaryl compounds and methods of use
CN109776522B (en) Substituted heteroaryl compounds, compositions and uses thereof
US10266521B2 (en) Substituted heteroaryl compounds and methods of use
US20170081338A1 (en) Substituted heteroaryl compounds and methods of use
US10683297B2 (en) Substituted heteroaryl compounds and methods of use
WO2018169700A1 (en) Substituted heteroaryl compounds and methods of use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20210419

Address after: No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Applicant after: SUNSHINE LAKE PHARMA Co.,Ltd.

Address before: 523808 Guangdong city of Dongguan province Hubei Songshan Industrial Park Industrial Road No. 1

Applicant before: SUNSHINE LAKE PHARMA Co.,Ltd.

Applicant before: CALITOR SCIENCES, LLC

GR01 Patent grant
GR01 Patent grant
CP01 Change in the name or title of a patent holder
CP01 Change in the name or title of a patent holder

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee before: SUNSHINE LAKE PHARMA Co.,Ltd.