CN111689991B - Substituted heteroaryl compounds, compositions and uses thereof - Google Patents

Substituted heteroaryl compounds, compositions and uses thereof Download PDF

Info

Publication number
CN111689991B
CN111689991B CN202010157262.4A CN202010157262A CN111689991B CN 111689991 B CN111689991 B CN 111689991B CN 202010157262 A CN202010157262 A CN 202010157262A CN 111689991 B CN111689991 B CN 111689991B
Authority
CN
China
Prior art keywords
alkyl
mmol
phenyl
disease
compound
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
CN202010157262.4A
Other languages
Chinese (zh)
Other versions
CN111689991A (en
Inventor
请求不公布姓名
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to PCT/CN2020/078990 priority Critical patent/WO2020182188A1/en
Publication of CN111689991A publication Critical patent/CN111689991A/en
Application granted granted Critical
Publication of CN111689991B publication Critical patent/CN111689991B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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 belongs to the field of medicaments, and in particular relates to a substituted heteroaryl compound shown as a formula (I) or a stereoisomer, a tautomer, a nitrogen oxide, a solvate, a metabolite or a pharmaceutically acceptable salt thereof, and pharmaceutical compositions comprising the same, and methods of using the same and pharmaceutical compositions thereof for the treatment of proliferative, autoimmune, allergic, and inflammatory disorders in mammals,Use in medicine for inflammatory diseases, transplant rejection, cancer or other diseases. The compounds provided by the invention exhibit excellent inhibitory activity against target kinases and optimized kinase selectivity. In addition, the compound provided by the invention also has excellent membrane permeation property and excellent pharmacokinetics property in animals, so the compound provided by the invention has very good development prospect.

Description

Substituted heteroaryl compounds, compositions and uses thereof
Technical Field
The invention belongs to the field of medicaments, and in particular relates to a novel substituted heteroaryl compound, pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the compound, and application of the compound and the pharmaceutical composition thereof in preparing medicaments 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 modulate the activity of the TAM kinase family (including Axl, mer and Tyro-3), the Trk kinase family (tropomyosin receptor kinase, including TrkA, trkB and TrkC), etc., and thus modulate signal transduction inside and outside the cell.
Background
The protein kinase family contains a broad class of structurally related enzymes that control various signal transduction processes within cells, catalyzing the phosphorylation of target protein substrates. Many diseases are associated with abnormal cellular responses triggered by protein kinase mediated events. These diseases include benign and malignant proliferative diseases, diseases resulting from inappropriate activation of the immune system, allograft rejection, graft-versus-host disease, autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, alzheimer's disease and hormone-related diseases. Accordingly, protein kinase inhibitors have been developed in the pharmaceutical field that are effective in treating diseases.
Kinases can be divided into families (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.) by phosphorylated substrates. Tyrosine phosphorylation is one of the central events regulating various biological processes such as cell proliferation, migration, differentiation and survival. Multiple families of receptor and non-receptor tyrosine kinases control tyrosine groups that catalyze the transfer of phosphate from ATP to specific cellular protein targets. Currently, the general corresponding motifs of the respective kinase families described above have been identified (Hanks et al, FASEB J.,1995,9,576-596;Knighton et.al, science,1991,253,407-414; garcia-Buston en. 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, erbB4, erk, flt-3, fak, fes, FGFRl, FGFR2, FGFR3, FGFR4, FGFR5, fgr, fyn, AXL, IGF-1R, INS-R, KDR, lck, lyn, MEK, mer, p38, PDGFR, PIK, PKC, PYK2, ros, tie, tie-2, TRK, yes, tyro3, and Zap70 et al (Robinson, D.R.; wu, Y.M.; lin, S.F. the protein tyrosine kinase family of the human genome oncogene 2000,19,5548-5557).
Cancers (and other hyperproliferative diseases) are characterized by uncontrolled cell proliferation. The activity of many protein kinases is increased in human tumors compared to normal tissues, and this increased activity may be due to a number of factors including increased kinase levels, mutated expression of coactivators or inhibitory proteins.
Axl is a receptor tyrosine kinase (ligand: growth inhibition specific protein 6, gas 6) that is unique in having two immunoglobulin-like repeat units in tandem and two fibronectin type III repeat units, are common features of cell adhesion molecules. Axl kinase belongs to the TAM family of receptor tyrosine kinases, and binding to ligand Gas6 activates the tyrosine kinase activity of Axl, thereby activating its downstream signal transduction pathways involved in the processes of growth, migration, aggregation, and apoptosis of cells (Rothlin, C.V.; ghosh, S.; zuniga, E.I.; oldstone, M.B.A.; lemke, G.TAM receptors are pleiotropic inhibitors of the innate immune response.cell 2007,131,1124-1136). Axl and Tyro-3 have the most similar gene structure, whereas Axl and Mer have the most similar tyrosine kinase domain amino acid sequence. Like other Receptor Tyrosine Kinases (RTKs), the TAM family of structures comprises an extracellular domain, a transmembrane domain, and a conserved intracellular kinase domain. The extracellular domain contains two immunoglobulin-like (Ig) domains and two fibronectin domain iii (FN iii) repeats, which are binding sites for endogenous ligands; the conserved amino acid sequence KW (I/L) A (I/L) ES located in the kinase domain is a structural feature unique to the TAM family. TAM family members have a common ligand, growth inhibitory specific protein 6 (Gas 6), which binds to all TAM receptor tyrosine kinases, with Axl binding to Gas6 being the strongest and 3-10 times stronger than the weakest Mer (Zago' rska A, et al, diversity of TAM receptor tyrosine kinase function. Nature Immunology 2014, 15:920-928;Nagata K,Ohashi K,Nakano T,et al.Identification of the Product of Growth Arrest-specific Gene 6as a Common Ligand for Axl,Sky,and Mer Receptor Tyrosine Kinases.Journal of Biological Chemistry,1996,271 (47): 30022-30027.).
Axl kinase is overexpressed or activated in a variety of cancers, including ovarian cancer, melanoma, renal cell carcinoma, uterine leiomyomas, endometrial cancer, thyroid cancer, gastric cancer, breast cancer, NSCLC, CML, AML, colon cancer, prostate cancer, various lymphomas, and esophageal cancer. Recent studies have shown that Axl overexpression is particularly severe in cancer cells that develop resistance after chemotherapy and treatment with receptor Tyrosine Kinase Inhibitors (TKI), one of the important causes of development of resistance (Zhang, z.; lee, j.c.; lin, l.; et al Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet.2012,44,852-860). Currently, the emergence of resistance in Cancer patients remains a difficulty in the course of Cancer treatment, and inhibition of Axl activity can enhance chemotherapy sensitivity and delay the onset of resistance, so Axl protooncogene is an attractive and valuable target for the discovery and development of new therapeutic agents (Feneyroles C, spenlinhauer A, guiet L, et al Axl Kinase as a Key Target for Oncology: focus on Small Molecule inhibitors. Mol Cancer Ther 2014,13 (9): 2141-2148). Based on implications in a variety of human malignancies, there is a need to design specific and selective inhibitors to treat cancer and other conditions mediated by and/or associated with Axl kinase. The present invention fulfills these needs and provides other related advantages.
Trk, tropomyosin receptor kinase, is a product of the proto-oncogene Trk, produced by fusion of tropomyosin and tyrosine kinase, and is a member of the Receptor Tyrosine Kinase (RTK) family. Trk receptors are classified as TrkA, trkB and TrkC, with the corresponding ligands being NGF, BDNF and NT-3, respectively, and the other ligand of TrkB being NT4/5, NT-3 also acting on TrkA and TrkB. NTs family members signal transduction primarily through autophosphorylation of Trk receptors, producing an effect (Huang, E.J.; reichardt, L.F.TRK receptors: roles in neuronal signal transduction. Annu. Rev. Biochem.2003,72, 609-642).
The NTRK gene fusions involved in NTRK1, NTRK2 or NTRK3 (encoding the neurotrophin receptors TrkA, trkB and TrkC, respectively) are oncogenic drivers of various adult and pediatric tumor types. The TRK fusion protein has ligand independent kinase activity and is capable of activating downstream signaling pathways associated therewith, stimulating cell growth and survival, and eliciting cancers that are mutually exclusive to other oncogenic drivers in humans. The data indicate that inhibition of TRK fusion proteins (e.g., larotentiib or entrectiniib) can treat NTRK fusion positive cancer patients. ( E.cocco, e.; scaltri, m.; drilon, A., NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol.2018, 15 (12): 731-747. )
Immunotherapy is a therapeutic approach that refers to the artificially enhanced or suppressed immune function of the body against a lowered or elevated immune state of the body to achieve the goal of treating a disease. There are many methods of immunotherapy, which are suitable for the treatment of a variety of diseases. Immunotherapy of tumors aims at activating the human immune system, killing cancer cells and tumor tissues by means of autoimmune functions. Unlike previous surgery, chemotherapy, radiation therapy and targeted therapies, immunotherapy targets not tumor cells and tissues, but the human immune system itself.
Protein kinase inhibitors have gained much attention as novel immunomodulating, anti-inflammatory and anti-cancer agents. Thus, new or improved agents that inhibit protein kinases such as Axl kinase, and Trk kinase are useful as immunomodulators, antineoplastic agents, analgesics, anti-organ fibrosis agents, and in 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, primary thrombocythemia, myelofibrosis, autoimmune thyroiditis, alzheimer's disease), diseases involving hyperactive inflammatory responses (e.g., eczema), allergies, chronic obstructive pulmonary disease, bronchitis, fibrosis, cancers (e.g., gastric cancer, liver cancer, lung cancer, colorectal cancer, prostate cancer, acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphocytic leukemia, multiple myeloma) and other treatment-induced immune responses (e.g., rash, contact dermatitis or diarrhea), chronic pain and acute pain, or diseases in which the pain is associated with cancer, bone fracture, tumor metastasis, bone pain, osteomyelitis, osteoarthritis, chronic pain in the bladder, chronic pain in the joints, backbones, pain, chronic pain, the bladder, chronic pain, the joint or the like.
The compounds, compositions, and methods described herein directly address these needs and other objects. In particular, the invention provides a class of compounds that inhibit, modulate and/or regulate the activity of one or more protein kinases, such as TAM kinase (Axl, mer and Tyro 3), and Trk kinase, for use in the treatment of proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, pain, fibrosis, transplant rejection, and complications thereof. Compared with the existing similar compounds, the compound has better pharmacological activity, and particularly, the compound has excellent inhibition activity and optimized kinase selectivity on target kinase. In addition, the compound of the invention has excellent membrane permeation property and excellent pharmacokinetics property in animals, so the compound of the invention has very good development prospect.
Disclosure of Invention
Definition of terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structural and chemical formulas. The invention is intended to cover all 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 of one or more of the incorporated references, patents and similar materials differing from or contradictory to the present application (including but not limited to defined terms, term application, described techniques, etc.), the present application controls.
The articles "a," "an," and "the" are intended to include "at least one" or "one or more" unless the context clearly dictates otherwise or otherwise. Thus, as used herein, the articles refer to articles of manufacture that include one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., more than one component is contemplated as being employed or used in embodiments of the described embodiments.
"stereoisomers" refer to compounds having the same chemical structure but different arrangements of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformational isomers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can be interconverted by a low energy barrier (low energy barrier). If tautomerism is possible (e.g., in solution), chemical equilibrium of the tautomers can be achieved. For example, proton tautomers (also known as proton transfer tautomers (prototropic tautomer)) include interconversions by proton transfer, such as keto-enol isomerisation and imine-enamine isomerisation. Valence tautomers (valance tautomers) include interconversions by recombination of some of the bond-forming electrons. Specific examples of keto-enol tautomerism are tautomerism of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one tautomer. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the interconversion 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, for example, for compounds of the general formula of the invention, or as specific examples, subclasses within the examples, and classes of compounds encompassed by the invention.
It is to be 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 specific substituent. "optionally" unless otherwise indicated, an optional substituent 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, then the substituents may be the same or different at each position.
The term "optionally substituted with … …" may be 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 H, D, oxo (=o), F, cl, br, -OH, -CN, -NO 2 、-NR c R d 、-C(=O)R 9 、-OC(=O)R 9 、-C(=O)OR 9a 、-S(=O) 0-2 R 9 、-OS(=O) 1-2 R 9 、-S(=O) 1-2 OR 9a 、-N(R 10a )C(=O)R 10 、-C(=O)NR 10a R 10 、-OC(=O)NR 10a R 10 、-N(R 10a )S(=O) 1-2 R 10 、-S(=O) 1-2 NR 10a R 10 、-N(R 10a )C(=O)NR 10a R 10 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Aminoalkyl, cyano-substituted C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylamino, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each of the C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl and C 1-6 Substitution of groups of alkoxy groups, and the like. Wherein R is c 、R d 、R 9 、R 9a 、R 10 And R is 10a Has the meaning as described in the present invention.
In the various parts of the present specification, substituents of the presently disclosed compounds are disclosed in terms of the type or scope of groups. It is specifically noted that the present invention includes each individual subcombination of the individual members of these group classes and ranges. For example, the term "C 1 -C 6 Alkyl "means in particular methyl, ethyl, C independently disclosed 3 Alkyl, C 4 Alkyl, C 5 Alkyl and C 6 An alkyl group.
The term "alkyl" or "alkyl group" as used herein means a saturated, straight or branched, monovalent hydrocarbon group 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 to 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 one or more substituents described herein.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH 3 ) Ethyl (Et, -CH) 2 CH 3 ) N-propyl (n-Pr, -CH) 2 CH 2 CH 3 ) Isopropyl (i-Pr, -CH (CH) 3 ) 2 ) N-butyl (n-Bu, -CH) 2 CH 2 CH 2 CH 3 ) Isobutyl (i-Bu, -CH) 2 CH(CH 3 ) 2 ) Sec-butyl (s-Bu, -CH (CH) 3 )CH 2 CH 3 ) Tert-butyl (t-Bu, -C (CH) 3 ) 3 ) N-pentyl (-CH) 2 CH 2 CH 2 CH 2 CH 3 ) 2-pentyl (-CH (CH) 3 )CH 2 CH 2 CH 3 ) 3-pentyl (-CH (CH) 2 CH 3 ) 2 ) 2-methyl-2-butyl (-C (CH) 3 ) 2 CH 2 CH 3 ) 3-methyl-2-butyl (-CH (CH) 3 )CH(CH 3 ) 2 ) 3-methyl-1-butyl (-CH) 2 CH 2 CH(CH 3 ) 2 ) 2-methyl-1-butyl (-CH) 2 CH(CH 3 )CH 2 CH 3 ) N-hexyl (-CH) 2 CH 2 CH 2 CH 2 CH 2 CH 3 ) 2-hexyl (-CH (CH) 3 )CH 2 CH 2 CH 2 CH 3 ) 3-hexyl (-CH (CH) 2 CH 3 )(CH 2 CH 2 CH 3 ) 2-methyl-2-pentyl (-C (CH) 3 ) 2 CH 2 CH 2 CH 3 ) 3-methyl-2-pentyl (-CH (CH) 3 )CH(CH 3 )CH 2 CH 3 ) 4-methyl-2-pentyl (-CH (CH) 3 )CH 2 CH(CH 3 ) 2 ) 3-methyl-3-pentyl (-C (CH) 3 )(CH 2 CH 3 ) 2 ) 2-methyl-3-pentyl (-CH (CH) 2 CH 3 )CH(CH 3 ) 2 ) 2, 3-dimethyl-2-butyl (-C (CH) 3 ) 2 CH(CH 3 ) 2 ) 3, 3-dimethyl-2-butyl (-CH (CH) 3 )C(CH 3 ) 3 ) N-heptyl, n-octyl, and the like.
The term "haloalkyl" or "haloalkoxy" means that the alkyl or alkoxy groups are substituted with one or more halogen atoms, examples of which include, but are not limited to, trifluoromethyl (-CF) 3 ) Trifluoromethoxy (-OCF) 3 ) Difluoroethyl (-CH) 2 CHF 2 ,-CF 2 CH 3 ,-CHFCH 2 F) Trifluoroethyl group (-CH) 2 CF 3 ,-CF 2 CH 2 F,-CFHCHF 2 ) Etc.
The terms "hydroxyalkyl" and "hydroxyalkoxy" denote alkyl or alkoxy groups, optionally substituted with one or more hydroxy groups, wherein "hydroxyalkyl" and "hydroxyalkyl" may be used interchangeably, examples of which include, but are not limited to, hydroxymethyl (-CH) 2 OH), 2-hydroxyethyl (-CH 2 CH 2 OH), 1-hydroxyethyl (-CH (OH) CH 3 ) 2-hydroxy-prop-2-yl (-COH (CH) 3 ) 2 ) 2-hydroxy-2-methylpropyl (-CH) 2 COH(CH 3 ) 2 ) 3-hydroxypropyl (-CH) 2 CH 2 CH 2 OH), 2-hydroxypropyl (-CH) 2 CH(OH)CH 3 ) Hydroxy methoxy (-OCH) 2 OH), and the like.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing 3 to 12 carbon atoms. Bicyclic cycloalkyl groups include spirobicycloalkyl, fused bicycloalkyl and bridged bicycloalkyl. In some embodiments, cycloalkyl groups comprise 3 to 12 carbon atoms; in other embodiments, cycloalkyl groups comprise 3 to 10 carbon atoms; in other embodiments, cycloalkyl groups comprise 3 to 8 carbon atoms; in other embodiments, cycloalkyl groups comprise 3 to 7 carbon atoms; in other embodiments, cycloalkyl groups comprise 3 to 6 carbon atoms; in some embodiments, cycloalkyl is C 7 -C 12 Cycloalkyl group comprising C 7 -C 12 Monocycloalkyl, C 7 -C 12 Bicycloalkyl radicals (e.g. C 7 -C 12 Spirobicycloalkyl, C 7 -C 12 Condensed bicycloalkyl and C 7 -C 12 Bridged bicycloalkyl) or C 7 -C 12 Tricycloalkyl groups. The cycloalkyl groups may independently be unsubstituted or substituted with one or more substituents described herein. The term "monocyclic cycloalkyl" or "monocyclic alkyl" means cycloalkyl of a monocyclic system, wherein the cycloalkyl has the definition as previously described, and the monocyclic cycloalkyl groups may independently be unsubstituted or substituted with one or more substituents described herein. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-enyl, 1-cyclopentyl-2-enyl, 1-cyclopentyl-3-enyl, cyclohexyl, 1-cyclohexyl-1-enyl, 1-cyclohexyl-2-enyl, 1-cyclohexyl-3-enyl, cyclohexanedienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
The term "cycloalkylalkyl" includes cycloalkyl-substituted alkyl groups. In some embodiments, a cycloalkylalkyl group refers to a "lower cycloalkylalkyl" group, i.e., the cycloalkyl group is attached to C 1-6 Is present. In other embodiments, cycloalkylalkyl groups refer to C-containing groups 1-3 "phenylalkylene" of alkyl groups of (2). Specific examples thereof include, but are not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, cyclopentylethyl, cyclohexylethyl, and the like. Cycloalkyl groups on cycloalkylalkyl groups may be further substituted with one or more substituents described herein.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein to refer to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic, monocyclic, bicyclic or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms. In some embodiments, the heterocyclyl or heterocycle contains from 4 to 12 ring atoms. In some embodiments, the heterocyclyl or heterocycle contains 5-12 cyclic precursorsAnd (5) a seed. In some embodiments, the heterocyclyl or heterocycle contains 5-8 ring atoms. In some embodiments, the heterocyclyl or heterocycle contains 5-7 ring atoms. Unless otherwise indicated, a heterocyclic group may be a carbon or nitrogen group, and-CH 2 The group may optionally be replaced by-C (=o) -the sulphur atom of the ring may optionally be oxidised to S-oxide and the nitrogen atom of the ring may optionally be oxidised to N-oxide. The heterocyclic group includes saturated heterocyclic groups (heterocycloalkyl groups) and partially unsaturated heterocyclic groups. The heterocyclic group has one or more points of attachment to the remainder of the molecule. Examples of heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxacyclopentyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiazalkyl, homopiperazinyl, homopiperidinyl, oxepinyl, thietanyl, azepane, oxaaza
Figure BDA0002404519330000041
Radical (e.g., 1, 4-oxazal->
Figure BDA0002404519330000042
Radical, 1, 2-oxaaza +.>
Figure BDA0002404519330000043
Radical), diaza->
Figure BDA0002404519330000044
Radical (e.g., 1, 4-diaza->
Figure BDA0002404519330000045
Radical, 1, 2-diaza- >
Figure BDA0002404519330000046
Base groupDioxa->
Figure BDA0002404519330000047
Radical (e.g., 1, 4-dioxa->
Figure BDA0002404519330000048
Radical, 1, 2-dioxa->
Figure BDA0002404519330000049
Radical), thiazepine->
Figure BDA00024045193300000410
Radicals (e.g. 1, 4-thiazal->
Figure BDA00024045193300000411
Radical, 1, 2-thiaaza +.>
Figure BDA00024045193300000412
Group), indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1 ]]Hept-5-yl, 2-azaspiro [4.4 ]]Nonylalkyl, 1, 6-dioxaspiro [4.4 ]]Nonylalkyl, 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-tetrahydroisoquinolinyl, hexahydrofuro [3,2-b ]]Furyl and dodecahydroisoquinolinyl, and the like. Examples of the substitution of the-CH 2-group in the heterocyclic group by-C (=o) -include, but are not limited to, 2-oxo-pyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidonyl, and 3, 5-dioxopiperidyl. Examples of sulfur atoms in the heterocyclic group that are oxidized include, but are not limited to, sulfolane, 1-dioxothiomorpholino. The heterocyclyl group may be optionally substituted with one or more substituents described herein.
In one embodiment, the heterocyclic group is a heterocyclic group consisting of 4 to 7 atoms, meaning a monovalent or polyvalent, saturated or partially unsaturated, non-aromatic, monocyclic or bicyclic ring comprising 4 to 7 ring atoms, wherein at least one ring atom is selected from nitrogen Sulfur and oxygen atoms. Unless otherwise indicated, a heterocyclic group consisting of 4 to 7 atoms may be a carbon group or a nitrogen group, and-CH 2 The group may optionally be replaced by-C (=o) -. The sulfur atom of the ring may optionally be oxidized to an S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxide. The heterocyclic group consisting of 4 to 7 atoms has one or more points of attachment to the remainder of the molecule. Examples of monocyclic heterocyclic groups of 4 to 7 atoms include, but are not limited to: azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaalkyl, 1, 2-oxazinyl, 1, 2-thiazinyl, hexahydropyridazinyl, homopiperazinyl, homopiperidinyl, oxaheptanyl, thietanyl, oxaazepinyl
Figure BDA0002404519330000051
Radical (1, 4-oxaaza +.>
Figure BDA0002404519330000052
Radical, 1, 2-oxaaza +.>
Figure BDA0002404519330000053
Radical), diaza- >
Figure BDA0002404519330000054
Radical (1, 4-diaza->
Figure BDA0002404519330000055
Radical, 1, 2-diaza->
Figure BDA0002404519330000056
Radical) and thiazepine->
Figure BDA0002404519330000057
The base (1,4-thiazal->
Figure BDA0002404519330000058
Radical, 1, 2-thiaaza +.>
Figure BDA0002404519330000059
Base), etc.; examples of 4-7 atom bicyclic heterocyclyls include, but are not limited to: 3-azabicyclo [3,2,0 ]]Heptane, 3-oxo-bicyclo [3,2,0 ]]Heptane and the like; examples of the substitution of the-CH 2-group by-C (=O) -in the heterocyclic group consisting of 4 to 7 atoms include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidonyl and 3, 5-dioxopiperidyl; 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. The 4-7 atom heterocyclyl group may be optionally substituted with one or more substituents described herein.
The term "heterocyclylalkyl" includes heterocyclyl-substituted alkyl groups in which both heterocyclyl and alkyl have the meaning described herein, examples of which include, but are not limited to, tetrahydrofuranylmethyl, pyrrol-2-ylmethyl, morpholin-4-ylethyl, piperazin-4-ylethyl, piperidin-4-ylethyl, and the like.
The term "aryl" means a monocyclic, bicyclic and tricyclic carbocyclic ring system 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 a ring of 3 to 7 atoms, and one or more points of attachment are attached to the remainder of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". Examples of aryl groups may include phenyl, naphthyl and anthracenyl. The aryl groups may independently be optionally substituted with one or more substituents described herein.
The term "arylalkyl" or "aralkyl" includes aryl substituted alkyl groups. In some embodiments, an arylalkyl group refers to a "lower arylalkyl" group, i.e., the aryl group is attached to C 1-6 Is present. In other embodiments, an arylalkyl group means a C-containing group 1-3 "phenylalkylene" of alkyl groups of (2). Specific examples thereof include, but are not limited to, benzyl, diphenylmethyl, phenethyl, and the like. Aryl groups on arylalkyl groups may be further substituted with one or more substituents described herein.
The term "heteroaryl" means 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 and has one or more points of attachment to the remainder of the molecule. The term "heteroaryl" may be used interchangeably with the term "heteroaromatic ring" or "heteroaromatic compound". In one embodiment, heteroaryl is a 5-12 atom heteroaryl group comprising 1,2,3 or 4 heteroatoms independently selected from O, S and N. In another embodiment, heteroaryl is a 5-10 atom composition heteroaryl comprising 1,2,3, or 4 heteroatoms independently selected from O, S, and N. In another embodiment, heteroaryl is a 5-6 atom composition heteroaryl comprising 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 heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1, 3-dithiotriazinyl, 1, 3-dithio, 3-triazolyl, 1, 3-triazolyl; the following bicyclic rings are also included, but are in no way limited to: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl), imidazo [1,2-a ] pyridinyl, pyrazolo [1,5-a ] pyrimidinyl, 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.
The term "heteroarylalkyl" means an alkyl group substituted with one or more heteroaryl groups, where both heteroaryl and alkyl groups have the meaning described herein, examples of which include, but are not limited to, pyridine-2-methyl, imidazole-2-methyl, furan-2-ethyl, indole-3-methyl, and the like.
The term "halogen" refers to F, cl, br or I.
As used herein, "pharmaceutically acceptable salts" refers to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as in the literature: S.M. Berge et al describe pharmaceutically acceptable salts in detail in J.pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid forming salts include, but are not limited to, inorganic acid salts formed by reaction with amino groups such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, and organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonate, or by other methods described in the literature such as ion exchange. Other pharmaceutically acceptable salts include adipic acid salts, alginates, ascorbates, aspartic acid salts, benzenesulfonates, benzoic acid salts, bisulfate salts, borates, butyric acid salts, camphoric acid salts, camphorsulfonic acid salts, cyclopentylpropionates, digluconate, dodecylsulfate, ethanesulfonate, formate salts, fumaric acid salts, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, caproate, hydroiodic acid salts, 2-hydroxy-ethanesulfonate, lactobionic aldehyde Salts of acids, lactates, laurates, laurylsulfates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanates, p-toluenesulfonates, undecanoates, valerates, and the like. Salts obtained by suitable bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl group 4 Is a salt of (a). The present invention also contemplates quaternary ammonium salts formed from any compound containing a group of N. The water-soluble or oil-soluble or dispersible product may be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. The pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and counter-ion forming amine cations, such as halides, hydroxides, carboxylates, sulphates, phosphates, nitrates, C 1-8 Sulfonate and aromatic sulfonate.
"solvate" according to the present invention refers to an association of one or more solvent molecules with a compound according to the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethylsulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that are water.
Description of the Compounds of the invention
The present invention discloses a novel class of compounds useful as inhibitors of protein kinase activity, particularly TAM family kinases (including TYRO3, AXL and MER), and NTRK family kinases (including NTRKA, NTRKB and NTRKC) activity. Compounds that are inhibitors of protein kinases are useful in the treatment of diseases related to inappropriate protein kinase activity, particularly inappropriate TAM kinase, and NTRK kinase activity. Compared with the existing similar compounds, the compound has better pharmacological activity, and particularly, the compound has excellent inhibition activity and optimized kinase selectivity on target kinase. In addition, the compound of the invention has excellent membrane permeation property and excellent pharmacokinetics property in animals, so the compound of the invention has very good development prospect.
The disclosed compounds may exhibit potent inhibitory activity against one or more protein kinases. In one aspect, the invention relates to a compound having the structure of formula (I):
Figure BDA0002404519330000061
or a stereoisomer, tautomer, nitroxide, solvate, metabolite, or pharmaceutically acceptable salt thereof;
Wherein, the liquid crystal display device comprises a liquid crystal display device,
U 1 and U 2 Each independently is N or-C (R) a )-;
R 1 、R 2 And R is 4 H, C each independently of the other 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Aminoalkyl, cyano-substituted C 1-6 Alkyl, C 3-10 Cycloalkyl, C 3-10 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each of the C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Aminoalkyl, cyano-substituted C 1-6 Alkyl, C 3-10 Cycloalkyl, C 3-10 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1,2,3 or 4R 11 Substitution;
each R is a 、R 3 、R 5 、R 6 、R 7 And R is 8 Each independently is H, D, F,Cl、Br、-OH、-CN、-NO 2 、-NR c R d 、C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Aminoalkyl, cyano-substituted C 1-6 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each of the C 1-6 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1,2,3 or 4R 12 Substitution;
or R is 2 And R is 3 Optionally forming a heterocyclic ring of 4 to 12 atoms together with the carbon and nitrogen atoms to which it is attached, wherein the heterocyclic ring of 4 to 12 atoms is optionally substituted with 0, 1, 2, 3, 4 or 5R 13 Substitution;
each R is 11 、R 12 And R is 13 H, D, oxo (=o), F, cl, br, -OH, -CN, -NO 2 、-NR c R d 、-C(=O)R 9 、-OC(=O)R 9 、-C(=O)OR 9a 、-S(=O) 0-2 R 9 、-OS(=O) 1-2 R 9 、-S(=O) 1-2 OR 9a 、-N(R 10a )C(=O)R 10 、-C(=O)NR 10a R 10 、-OC(=O)NR 10a R 10 、-N(R 10a )S(=O) 1-2 R 10 、-S(=O) 1-2 NR 10a R 10 、-N(R 10a )C(=O)NR 10a R 10 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Aminoalkyl, cyano-substituted C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylamino, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each of the C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl and C 1-6 Substitution of the alkoxy group;
each R is c 、R d 、R 9 、R 9a 、R 10 And R is 10a H, D, C each independently of the other 1-6 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each of the C 1-6 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1,2,3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl and C 1-6 Substitution of the alkoxy group; and
n is 0, 1, or 2.
In some embodiments, the method comprises, among other things,
R 2 is H, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl, or C 2-7 Heterocyclyl C 1-4 An alkyl group; wherein each of the C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl and C 2-7 Heterocyclyl C 1-4 Alkyl is independently optionally substituted with 0, 1,2,3 or 4R 12 Substitution;
R 3 h, D, -CN, C 1-4 Alkyl, wherein each of the above C 1-4 Alkyl is optionally substituted with 0, 1,2,3 or 4R 11 Substitution;
or R is 2 And R is 3 Optionally forming a heterocyclic ring of 5 to 12 atoms together with the carbon and nitrogen atoms to which it is attached, wherein the heterocyclic ring of 5 to 12 atoms is optionally substituted with 0, 1,2,3, 4 or 5R 13 And (3) substitution.
In other embodiments, the compounds of the present invention have the structure of formula (II):
Figure BDA0002404519330000071
wherein, the liquid crystal display device comprises a liquid crystal display device,
X 1 is O, S, -N (R) 13a )-、-C(=O)-、-(CH 2 ) t1 -、-X 2 -(CH 2 ) t1 -, a part of or- (CH) 2 ) t1 -X 2 -(CH 2 ) t2 -;
X 2 Is O, S, -N (R) 13a ) -, or-C (=o) -;
each R is 13a H, D, oxo (=o), F, cl, br, -OH, -CN, -NO 2 Amino, C 1-6 Alkyl group,C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl, C 1-9 Heteroaryl C 1-6 Alkyl, -C (=o) R 9 、-C(=O)OR 9a 、-S(=O) 0-2 R 9 、-S(=O) 1-2 OR 9a 、-S(=O) 1-2 NR 10a R 10 or-C (=O) NR 10a R 10 Wherein each of the above C 1-6 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-6 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-6 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-6 Alkyl, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1,2,3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl and C 1-6 Substitution of the alkoxy group;
each t1 and t2 is independently 0, 1,2, or 3; and
m is 0, 1,2, 4, or 5.
In some embodiments, wherein X 1 Is O, S, -N (R) 13a )-、-C(=O)-、-(CH 2 ) t1 -、-X 2 -(CH 2 ) t1 -, a part of or- (CH) 2 ) t1 -X 2 -(CH 2 ) t2 -; and X 2 Is O, S, or-N (R) 13a )-。
In some embodiments, R 13 Each independently is H, D, oxo (=O), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-4 Alkyl, C 2-4 Alkenyl, C 2-4 Alkynyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Alkylamino, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-4 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-4 Alkyl, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-4 An alkyl group; wherein each of the C 1-4 Alkyl, C 2-4 Alkenyl, C 2-4 Alkynyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Alkylamino, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-4 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-4 Alkyl, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-4 Alkyl is independently optionally substituted with 0, 1, 2, 3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-4 Alkyl and C 1-4 The groups of the alkoxy groups are substituted.
In some embodiments, each R 13a H, D, oxo (=o), F, cl, br, -OH, -CN, -NO 2 Amino, C 1-4 Alkyl, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-4 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-4 Alkyl, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-4 An alkyl group; wherein each of the C 1-4 Alkyl C 3-8 Cycloalkyl, C 3-8 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl, C 2-7 Heterocyclyl C 1-4 Alkyl, C 6-12 Aryl, C 6-12 Aryl C 1-4 Alkyl, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-4 Alkyl is independently optionally substituted with 0, 1,2,3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl and C 1-6 The groups of the alkoxy groups are substituted.
In some embodiments, each t1 and t2 is independently 0, 1,2, or 3; and m is 0, 1,2, or 4.
In some embodiments, wherein R 1 Is C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 3-8 Cycloalkyl, phenyl, or C 1-9 Heteroaryl; wherein each of the C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 3-8 Cycloalkyl, phenyl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1,2,3 or 4R 11 And (3) substitution.
In some embodiments, the method comprises, among other things,
U 1 and U 2 Each independently is N or-C (R) a )-;
Each R is a And R is 8 H, D, F, cl, br, -OH, -CN, -NO, respectively and independently 2 、-NH 2 Or C 1-4 An alkyl group; wherein each of the C 1-4 Alkyl is independently optionally substituted with 0, 1,2,3 or 4R 12 Substitution; and
n is 0, 1, or 2.
In some embodiments, wherein R 4 H, D is methyl, ethyl, propyl, isopropyl, butyl, C 1-4 Haloalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 3-6 Cycloalkyl, C 3-6 Cycloalkyl C 1-4 Alkyl, C 2-7 Heterocyclyl, or C 2-7 Heterocyclyl C 1-4 An alkyl group.
In some embodiments, wherein R 5 Is H, D, -NR c R d 、C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, or cyano-substituted C 1-4 An alkyl group.
In some embodiments, wherein R 6 And R is 7 H, D, F, cl, br, -OH, -N, respectively and independentlyR c R d 、-CN、-NO 2 、C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, or cyano-substituted C 1-4 An alkyl group.
In some embodiments, wherein each R 11 、R 12 And R is 13 H, D, oxo (=o), F, cl, br, -OH, -CN, -NO 2 、-NR c R d 、C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 1-4 Alkoxy, or C 1-4 An alkylamino group; wherein each of the C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Aminoalkyl, cyano-substituted C 1-4 Alkyl, C 1-4 Alkoxy and C 1-4 Alkylamino groups are independently optionally substituted with 0, 1, 2, 3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 And C 1-4 The groups of the alkoxy groups are substituted.
In some embodiments, wherein each R c 、R d 、R 9 、R 9a 、R 10 And R is 10a H, D, C each independently of the other 1-4 Alkyl, C 3-6 Cycloalkyl, or C 2-7 A heterocyclic group; wherein each of the C 1-4 Alkyl C 3-6 Cycloalkyl and C 2-7 Heterocyclyl is independently optionally substituted with 0, 1, 2, 3 or 4 groups independently selected from H, D, oxo (=o), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl and C 1-6 The groups of the alkoxy groups are substituted.
In some embodiments, the compounds of the present invention are compounds having one of the following structures:
Figure BDA0002404519330000091
or a stereoisomer, tautomer, nitroxide, solvate, metabolite or pharmaceutically acceptable salt thereof.
Stereoisomers, tautomers, solvates, metabolites or pharmaceutically acceptable salts of the compounds of formulas (I) and (II) are included within the scope of the invention unless otherwise indicated.
The disclosed compounds may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention is intended to encompass all stereoisomeric forms of the compounds of formula (I) or (II), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, as well as mixtures thereof, such as racemic mixtures, as part of the present invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not indicated, then all stereoisomers of that structure are contemplated as being within the present invention and are included as presently disclosed compounds. When stereochemistry is indicated by the solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of that structure are so defined and defined.
The compounds of formula (I) or (II) 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 salt is not necessarily a pharmaceutically acceptable salt, and may be an intermediate for preparing and/or purifying a compound of formula (I) or (II) and/or for separating enantiomers of the compound of formula (I) or (II).
In another aspect, the present invention relates to intermediates for preparing compounds of formula (I) and (II).
In another aspect, the present invention relates to methods for the preparation, isolation and purification of compounds of formula (I) and (II).
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable adjuvant, diluent or carrier, or combination thereof. In another embodiment, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel or spray form.
In some embodiments, the pharmaceutical compositions of the invention further comprise an additional therapeutic agent.
In a further aspect, the invention relates to the use of a compound according to the invention or a pharmaceutical composition according to the invention in the manufacture of a medicament for the prophylaxis or treatment of one or more Axl and/or Trk protein kinase mediated diseases and/or conditions.
In some embodiments, the disease and/or disorder is selected from a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease, or transplant rejection.
In some embodiments, the disease and/or disorder is selected from the group consisting of treating and preventing TAM kinase, and NTRK kinase mediated diseases involving signaling pathways. Such diseases include proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, graft rejection, and complications thereof. In particular, the compounds of the invention may be used to treat diseases such as cancer, polycythemia vera, primary thrombocythemia, myelofibrosis, myelogenous leukemia, acute lymphoblastic 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 diseases, 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.
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound of the present disclosure, or a compound listed in the examples; and pharmaceutically acceptable adjuvants, diluents, carriers, vehicles or combinations thereof. The amount of a compound in the pharmaceutical compositions disclosed herein is an amount effective to detect inhibition of protein kinase in a biological sample or patient.
It will also be appreciated that certain compounds of the invention may exist in free form for use in therapy or, if appropriate, in the form of pharmaceutically acceptable derivatives thereof. Some non-limiting embodiments of pharmaceutically acceptable derivatives include pharmaceutically acceptable salts, esters, salts of such esters, or any other adduct or derivative that provides, directly or indirectly, a compound of the invention or a metabolite or residue thereof upon administration to a patient in need thereof.
Various carriers for use in formulating pharmaceutically acceptable compositions, and well-known techniques for their preparation, are disclosed in documents such as 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,1988-1999,Marcel Dekker,New York, the contents of each of which are incorporated herein by reference. It is within the scope of the present invention to contemplate its use in addition to any common carrier that is incompatible with the disclosed compounds of the present invention, such as by producing any undesirable biological effect, or by interacting in a deleterious manner with any other component of the pharmaceutically acceptable composition.
The pharmaceutical compositions provided herein may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
Use of the compounds and compositions of the invention
The present invention provides methods of treating, preventing, or ameliorating one or more symptoms of a disease or disorder mediated or otherwise affected by the behavior of one or more protein kinases, such as TAM family kinases (including Tyro3, AXL and MER kinases), or NTRK family kinases (NTRKA, NTRKB and NTRKC), or by one or more protein kinases, such as TAM family kinases (including Tyro3, AXL and MER kinases), or NTRK family kinases (NTRKA, NTRKB and NTRKC), using the disclosed compounds and pharmaceutical compositions.
TAM family kinases (including Tyro3, AXL and MER kinases), or NTRK family kinases (NTRKA, NTRKB and NTRKC) may be wild type and/or mutant of the included kinases.
In some embodiments, the invention provides a class of compounds disclosed herein or pharmaceutical compositions comprising the compounds disclosed herein for treating, preventing, or ameliorating one or more symptoms of a disease or disorder mediated or otherwise affected by inappropriate AXL kinase activity or a disease or disorder mediated or otherwise affected by inappropriate AXL kinase activity. In another embodiment, the disease, disorder, or one or more symptoms of the disease or disorder is associated with inappropriate MER kinase behavior. In yet another embodiment, the disease, disorder, or one or more symptoms of the disease or disorder is associated with inappropriate TYRO3 kinase activity.
In some embodiments, the invention provides A class of compounds disclosed herein or pharmaceutical compositions comprising the compounds disclosed herein for treating, preventing, or ameliorating one or more symptoms of A disease or disorder mediated or otherwise affected by inappropriate NTRK-A kinase activity. In other embodiments, the disease, disorder, or one or more symptoms of the disease or disorder is associated with inappropriate NTRK-B kinase activity. In some embodiments, the disease, disorder, or one or more symptoms of the disease or disorder is associated with inappropriate NTRK-C kinase activity.
By "inappropriate AXL kinase behavior" is meant AXL kinase behavior that deviates from normal AXL kinase behavior that occurs in a particular patient. Inappropriate AXL kinase behavior can take the form of, for example, an abnormal increase in activity, or deviations in time points and control of AXL kinase behavior. Such inappropriate kinase behavior results from inappropriate or uncontrolled behavior, for example, due to over-expression 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), idiopathic thrombocythemia, idiopathic Myelofibrosis (IMF); leukemias, for example, myeloid leukemia 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; the cancer includes head and neck cancer, prostatic cancer, breast cancer, ovarian cancer, melanoma, lung cancer, brain tumor, pancreatic cancer, urothelial cancer, liver cancer, gastric cancer, renal cancer, etc.; and inflammatory diseases or disorders associated with immune dysfunction, immunodeficiency, immunomodulation, autoimmune diseases, tissue graft 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 invention provides a class of compounds disclosed herein or pharmaceutical compositions comprising the compounds disclosed herein for use in the prevention and/or treatment of proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, or transplant rejection in a mammal, including a human.
In another aspect, the invention provides a method of treating a mammal suffering from or at risk of suffering from a disorder disclosed herein, the method comprising administering one or more pharmaceutical compositions or compounds disclosed herein in an amount effective to treat the disorder or in an amount effective to prevent the disorder.
In particular examples, the proliferative disease is selected from cancer (e.g., colon cancer, glioblastoma, endometrial cancer, liver cancer, lung cancer, melanoma, kidney cancer, thyroid cancer, lymphoma, lymphoproliferative disorders, small cell lung cancer, squamous cell lung cancer, glioma, breast cancer, prostate cancer, ovarian cancer, cervical cancer, etc., hematological malignancies such as Acute Myelogenous Leukemia (AML), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic Myelogenous Leukemia (CML), T-cell acute lymphoblastic leukemia (T-ALL), B-cell acute lymphoblastic leukemia (B-ALL), non-hodgkin's lymphoma (NHL), B-cell lymphoma, polycythemia vera, primary thrombocythemia, myelofibrosis, multiple myeloma, etc.
In another aspect, the invention provides 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 particular embodiments, 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, the invention provides 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 presently disclosed pharmaceutical compositions or compounds. In particular embodiments, the allergic disease is selected from respiratory allergic diseases, sinusitis, eczema and measles, food allergy, and insect venom allergy.
In particular embodiments, the allergic disease is selected from respiratory allergic diseases, sinusitis, eczema and measles, food allergy, and insect venom allergy.
In another aspect, the invention provides a method of treating and/or preventing a mammal susceptible to or suffering from an inflammatory disease, the method comprising administering a therapeutically effective amount or a prophylactically effective amount of one or more of the presently disclosed pharmaceutical compositions or compounds.
In another aspect, the present invention provides a class of compounds of the present disclosure for use as a medicament, in particular as a medicament for the treatment and/or prophylaxis of the diseases described herein. Also provided are medicaments for the treatment and/or prophylaxis of the diseases according to the invention using the compounds disclosed herein.
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 determined to be safe and effective for such combination administration.
In one aspect, the invention provides a method of treating, preventing or ameliorating a disease or disorder comprising administering a safe and effective amount of a combination comprising a compound of the present 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: anticancer agents, including chemotherapeutic agents and antiproliferative agents; an anti-inflammatory agent; and an immunomodulator or immunosuppressant.
In another aspect, the invention provides a product comprising a compound of the invention and at least one other therapeutic agent, which can be prepared as a combination for simultaneous, separate or sequential administration in therapy. In some embodiments, the treatment is directed to treatment of a disease or condition mediated by the activity of one or more protein kinases, such as AXL kinase, or NTRK kinase. The co-preparation provides products including compositions comprising the disclosed compounds and other therapeutic agents in the same pharmaceutical composition, or in different forms, e.g., kits.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the present disclosure and one or more additional therapeutic agents. In one embodiment, the pharmaceutical composition may comprise pharmaceutically acceptable excipients as described above.
In another aspect, the invention provides a kit comprising two or more separate pharmaceutical compositions, wherein at least one of the pharmaceutical compositions comprises a compound of the present disclosure. In one embodiment, the kit comprises means for separately holding the composition, such as a container, a separate bottle or a separate foil cartridge. Examples of such kits are blister packs, which are commonly used for packaging tablets, capsules and the like.
The disclosed compounds 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 therapies or anticancer drugs that may be used in combination with the compounds of the present invention, surgery, radiation therapy (a few examples such as gamma radiation, neutron beam radiation therapy, electron beam radiation therapy, proton therapy, brachytherapy and systemic radioisotope therapy), endocrine therapy, taxanes (taxol), docetaxel (taxotere) and the like, platinum derivatives (cisplatin), carboplatin (carboplatin), biological response modifiers (interferons, interleukins), tumor necrosis factor (TNF, TRAIL receptor targets), hyperthermia and cryotherapy, agents that mitigate any adverse reaction (such as antiemetics), and other approved chemotherapeutic agents including, but not limited to, alkylating agents (mechlorethamine), chlorambucil (chlorambucil), cyclophosphamide (cyclophosphamide), horse flange (mexiletine), ifomide (ifoline), ifosfamide (epothilone), antimetabolite (methotrexate), antimetabolite (6-toxin), toxin (6-5-toxin), toxin (5-toxin), and the like Irinotecan (irinotecan), topotecan (topotecan)), antibiotics (doxorubicin), bleomycin (bleomycin), mitomycin (mitomycin), nitrosoureas (carmustine), lomustine (lomustine)), inhibitors of the cell division cycle (KSP via mitotic kinesin inhibitors, CENP-E and CDK inhibitors), enzymes (asparaginase), hormones (tamoxifen), leuprolide (leuprolide), flutamide (flutamide), megestrol (megestrol), dexamethasone (dexamethamate), and the like. Anti-angiogenic agents (avastin) and the like). Monoclonal antibodies (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), ruxotinib (ruxolitinib), vemurafenib (vemurafenib), vandetanib (vanretanib), pazopanib (pazopanib), and the like. Drugs inhibit or activate cancer pathways such as mTOR, HIF (hypoxia inducible factor) pathways, and others.
The compounds disclosed herein may also be combined with other therapeutic procedures to enhance therapeutic efficacy. For example, hormonal therapy or specific radiation therapy is administered. The compounds disclosed herein are particularly useful as radiosensitizers, particularly in the treatment of tumors that are weakly sensitive to those radiation treatments.
"combination" means a kit of parts for fixed combination or for combined administration in a single dosage unit form, wherein the presently disclosed compounds and the combination partners may be administered independently at the same time or may be administered separately at certain time intervals, in particular such that the combination partners exhibit a co-operation, e.g. a 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 (e.g., a patient) in need thereof, and are intended to include treatment regimens in which the substances are not necessarily administered by the same route of administration or simultaneously.
Therapeutic method
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. Embodiments of the present disclosure include methods of treating the diseases mentioned herein by administering to a patient in need thereof a safe and effective amount of a disclosed compound of the present disclosure or a pharmaceutical composition comprising a disclosed compound of the present disclosure.
In embodiments thereof, the presently disclosed compounds or pharmaceutical compositions comprising the presently disclosed compounds may be administered at one time or, depending on the dosing regimen, at several times at different time intervals over a specified period of time. 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 performed until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. Suitable dosing regimens for the presently disclosed compounds, or pharmaceutical compositions comprising the presently disclosed compounds, depend on the pharmacokinetic properties of the compounds, such as dilution, distribution, and half-life, which can be determined by the skilled artisan. Furthermore, suitable dosing regimens for the presently disclosed compounds or pharmaceutical compositions comprising the presently disclosed compounds, including the duration of time for which the regimen is practiced, depend on the disease being treated, the severity of the disease 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 the like, among other factors within the knowledge and experience of the skilled artisan. Such a skilled artisan will also appreciate that adjustment of the regimen may be required for the individual patient's response to the regimen, or as the individual patient needs to change over time.
The presently disclosed compounds may be administered simultaneously with, or before or after, one or more other therapeutic agents. The compounds of the present invention may be administered separately from other therapeutic agents by the same or different routes of administration, or in pharmaceutical compositions therewith.
General synthetic scheme
For the purpose of illustrating the invention, examples are set forth below. It is to be understood that the invention is not limited to these examples but provides a method of practicing the invention.
In general, the compounds of the invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I) or (II), unless otherwise indicated. The following reaction schemes and examples are provided to further illustrate the present invention.
The examples described below are given unless otherwise indicated that all temperatures are given 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. Typical reagents are purchased from Shanzhou chemical plant, guangzhou chemical plant, tianjin Fuchen chemical plant, wuhan Xinhua Yuan technology development Co., ltd, and Qingdao ocean chemical plant.
Anhydrous tetrahydrofuran, dioxane, toluene and diethyl ether are obtained by reflux drying of metallic sodium. The anhydrous methylene chloride and chloroform are obtained by reflux drying of calcium hydride. Ethyl acetate, petroleum ether, N-hexane, N-dimethylacetamide and N, N-dimethylformamide were dried over anhydrous sodium sulfate in advance for use.
The following reaction is typically carried out under nitrogen or argon pressure or with a dry tube (unless otherwise indicated) over anhydrous solvent, the reaction flask is capped with a suitable rubber stopper and the substrate is injected through a syringe. The glassware was all dried.
The chromatographic column is a silica gel column. Silica gel (300-400 mesh) was purchased from Qingdao ocean chemical plant.
1 H NMR spectra were recorded using a Bruker 300MHz, 400MHz or 600MHz nuclear magnetic resonance spectrometer. 1 H NMR Spectroscopy with CDC1 3 、DMSO-d 6 、CD 3 OD or acetone-d 6 TMS (0 ppm) or chloroform (7.26 ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (single, singlet), d (doublet ), t (triplet), m (multiplet), br (broad), br (broadened singlet, broad singlet), dd (doublet of doublets, doublet), dt (doublet of triplets, doublet). Coupling constants are expressed in hertz (Hz).
The measurement conditions for low resolution Mass Spectrometry (MS) data are: agilent 6120 four-stage HPLC-M (column type: zorbax SB-C18, 2.1X130 mm,3.5 μm, 6min, flow rate 0.6mL/min. Mobile phase: 5% -95% (CH containing 0.1% formic acid) 3 CN) at (containingH of 0.1% formic acid 2 O), using electrospray ionization (ESI), at 210nm/254nm, using UV detection.
The pure compounds were detected by UV at 210nm/254nm using Agilent 1260pre-HPLC or Calesep pump 250pre-HPLC (column model: NOVASEP 50/80mm DAC).
Typical synthetic procedures for preparing the disclosed compounds are shown in the following synthetic schemes 1-2. Unless otherwise indicated, each R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、U 1 、U 2 And n has the definition as described herein; PG 1 And PG 2 Is a protecting group.
Synthesis scheme 1:
Figure BDA0002404519330000141
having the formula(6)The compounds of the present invention of the structure shown can be prepared by the general synthetic method described in scheme 1, and reference is made to the examples for specific procedures. In FIG. 1, a boron ester derivative is produced under alkaline conditions(1)In a suitable base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a suitable Pd catalyst (e.g., pd (OAc) 2 、Pd(dppf) 2 Cl 2 Or Pd (or) 2 (dba) 3 Etc.) with a substituted heteroaryl compound(2)The coupling reaction is carried out to obtain the compound (3). Removal of protecting groups PG 1 The aromatic amine derivative is obtained(4). Carboxylic acid derivatives(5)With compounds in the presence of condensing agents, e.g. EDCI or HATU(4)Condensing to obtain target kinase inhibitor(6)
Carboxylic acid derivatives(5)Can be described in literature (see, e.g., "Practical synthesis of bicyclic pyrazol-5-one derivatives", "Xuejin Feng, michael a. Xi, yanjun Wu, xiaovang Wang, ning Xi Tetrahedron lett.2017,58,46-49;Facile synthesis of bicyclic1-arylpyrazol-5-ones", "Wu, y; wang, k; li, z; bai, x;, xi, n. Tetrahedron lett.2014,55, 142-147)The synthesis method is adopted.
Synthesis scheme 2:
Figure BDA0002404519330000142
having the formula(6)The compounds of the present invention of the structure shown can also be prepared by the general synthetic methods described in scheme 2, for specific procedures, reference being made to the examples. In scheme 2, aryl or heteroaryl compounds7)With compounds in the presence of condensing agents, e.g. EDCI or HATU(5)Condensation to give compounds(8). Boron ester derivatives under alkaline conditions(10)In a suitable base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a suitable Pd catalyst (e.g., pd (OAc) 2 、Pd(dppf) 2 Cl 2 Or Pd (or) 2 (dba) 3 Etc.) with a substituted heteroaryl compound(8)The coupling reaction is carried out to obtain the compound (9). Boron ester derivatives under alkaline conditions(9)In a suitable base (e.g., cesium carbonate, potassium carbonate, sodium carbonate, etc.), and a suitable Pd catalyst (e.g., pd (OAc) 2 、Pd(dppf) 2 Cl 2 Or Pd (or) 2 (dba) 3 Etc.) with substituted heteroaryl derivatives(2)The target kinase inhibitor is obtained through the coupling reaction(6)
Examples
Example 1N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
Figure BDA0002404519330000143
Step 1) N- (4-bromophenyl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
Containing 2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ]]Pyridine-3-carboxylic acid (650 mg,65%,1.636 mmol), triethanolamine (0.7 mL,5 mmol), 4-bromoaniline (282 mg,1.639 mmol) and HATU (627mg, 1.636 mmol)The mixture of dichloromethane (20 mL) was stirred at room temperature for 23 hours. To the reaction was added water (20 mL), extracted with dichloromethane (30 mL x 2), the organic phases were combined and concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (EtOAc/MeOH (v/v) =100/1) to give the title compound (386.8 mg, 57.35%) as a yellow solid. MS (ESI, pos.ion) m/z 414.2[ M+H ]] +1 H NMR(400MHz,CDCl 3 )δ(ppm):10.61(s,1H),7.54(dd,J=16.0,8.3Hz,4H),7.45(t,J=7.4Hz,1H),7.42-7.31(m,4H),3.55(t,J=5.9Hz,2H),3.38(t,J=6.4Hz,2H),2.12-2.02(m,2H),1.96-1.86(m,2H)。
Step 2) 2-oxo-1-phenyl-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -1,2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
To a composition containing N- (4-bromophenyl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ]]To a solution of pyridine-3-carboxamide (3836 mg,0.936 mmol), bis-pinacolatyldiboron (268 mg,1.056 mmol) and 1,1' -bis (diphenylphosphine) ferrocene palladium (II) dichloride (104 mg,0.142 mmol) in 1, 4-dioxane (8 mL) was added potassium acetate (235 mg, 2.399mmol). The reaction solution was heated to reflux under nitrogen protection, stirred for 7.5 hours, 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 (368.5 mg, 85.70%) as a white solid. MS (ESI, pos.ion) m/z 460.2[ M+H ]] +1 H NMR(400MHz,CDCl 3 )δ(ppm):10.67(s,1H),7.75(d,J=8.4Hz,2H),7.68(d,J=8.5Hz,2H),7.53(t,J=7.6Hz,2H),7.44(t,J=7.4Hz,2H),7.35(d,J=7.3Hz,2H),3.55(t,J=5.9Hz,2H),3.40(t,J=6.4Hz,2H),2.07(dt,J=11.6,5.8Hz,3H),1.91(m,3H),1.33(s,13H)。
Step 3) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
To a compound containing 2-oxo-1-phenyl-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl)]-1,2,4,5,6, 7-hexahydropyrazolo [1,5-a ]]Pyridine-3-carboxamide (365 mg,0.80 mmol) and 5-bromo-7-methyl-7H-pyrrolo [2,3-d]To a solution of pyrimidin-4-amine (182 mg,0.80 mmol) in 1, 4-dioxane (8 mL) was added 1,1' -bis(diphenylphosphine) ferrocene Palladium (II) dichloride dichloromethane complex (134 mg,0.161 mmol) and cesium carbonate (522 mg,1.602 mmol). The reaction solution was heated to 110℃under nitrogen and stirred for 15 hours, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) =10/1) to give a yellow solid, which was purified by anion separation column to give the title compound (14.7 mg, 3.46%) as a white solid. MS (ESI, pos.ion) m/z 480.4[ M+H ] ] + 。HRMS(ESI + ) Calculated value C 27 H 26 N 7 O 2 [M+H] + 480.21, found 480.2155. 1 H NMR(400MHz,CDCl 3 )δ(ppm):10.66(s,1H),8.33(s,1H),7.75(d,J=8.5Hz,2H),7.55(t,J=7.6Hz,2H),7.45(t,J=7.5Hz,1H),7.39(dd,J=14.3,7.9Hz,4H),5.11(s,2H),3.83(s,3H),3.57(t,J=5.9Hz,2H),3.42(t,J=6.4Hz,3H),2.13-2.05(m,3H),1.97-1.89(m,3H)。
Example 2N- (5- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) pyridin-2-yl) -2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide
Figure BDA0002404519330000151
Step 1) 2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carbonyl chloride
Oxalyl chloride (0.6 mL,7 mmol) was added to a solution of tetrahydrofuran (10 mL) containing 2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxylic acid (480 mg,1.845 mmol). The reaction was heated to 60 ℃ and stirred for 4h, concentrated under reduced pressure to give the title compound (515 mg, 100.2%) as a yellow solid.
Step 2) N- (5-bromopyridin-2-yl) -2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide
After stirring a solution of 5-bromopyridin-2-amine (320 mg,1.849 mmol) and triethylamine (0.8 mL,6 mmol) in methylene chloride (20 mL) at room temperature for 10 minutes, 2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] was added to the mixture][1,4]Oxazine-3-carbonyl chloride (515 mg,1.848 mmol). Stirring the reaction solution at room temperatureAfter stirring for 24 hours, water (20 mL) was added dropwise for dilution and extracted with dichloromethane (50 mL). The organic phase was separated and concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) =100/1), and the collected yellow solid was recrystallized from a mixed solution of dichloromethane/methanol/petroleum ether (6 mL/0.5mL/12 mL) to give the title compound (380 mg, 49.53%) as a white solid. MS (ESI, pos.ion) m/z 415.0[ M+H ] ] +1 H NMR(400MHz,CDCl 3 )δ(ppm):10.58(s,1H),8.13(s,1H),7.99(d,J=8.8Hz,1H),7.57(dd,J=8.8,2.0Hz,1H),7.37(t,J=7.5Hz,2H),7.30(d,J=6.8Hz,1H),7.21(d,J=7.6Hz,2H),5.04(s,2H),3.97(t,J=4.8Hz,2H),3.50(t,J=4.7Hz,2H)。
Step 3) 2-oxo-1-phenyl-N- (5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridin-2-yl) -2,4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide
To a solution of N- (5-bromopyridin-2-yl) -2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide (380 mg,0.915 mmol), dipinacol diboron (260 mg,1.024 mmol) and tetrakis (triphenylphosphine) palladium (0) (151 mg,0.130 mmol) in 1, 4-dioxane (10 mL) was added potassium acetate (274 mg,2.833 mmol). The reaction was heated to reflux under nitrogen for 5 hours and concentrated under reduced pressure to give the title compound (425 mg, 100.4%) as a white solid.
Step 4) N- (5- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) pyridin-2-yl) -2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide
To a catalyst containing 2-oxo-1-phenyl-N- (5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridin-2-yl) -2,4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ]][1,4]Oxazine-3-carboxamide (425 mg,0.919 mmol) and 5-bromo-7-methyl-7H-pyrrolo [2,3-d]To a mixture of pyrimidine-4-amine (235 mg,1.035 mmol) 1, 4-dioxane (10 mL) and water (5 mL) was added tetrakis (triphenylphosphine) palladium (0) (215 mg,0.186 mmol) and potassium carbonate (390 mg,2.794 mmol). The reaction solution was heated to 105℃under nitrogen and stirred for 20 hours, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) =50/1) to give a yellow solid, which was purified by anion separation column The title compound (73 mg, 16.46%) was obtained as a white solid after conversion. MS (ESI, pos.ion) m/z 483.1[ M+H ]] + 。HRMS(ESI + ) Calculated value C 25 H 22 N 8 O 3 ([M+H] + ) 483.18, found 483.1886. 1 H NMR(400MHz,DMSO-d 6 )δ(ppm):10.77(s,1H),8.37(d,J=1.7Hz,1H),8.27(d,J=8.5Hz,1H),8.16(s,1H),7.84(dd,J=8.5,2.1Hz,1H),7.64-7.58(m,2H),7.56-7.50(m,3H),7.37(s,1H),6.18(s,2H),5.13(s,2H),4.12-4.08(m,2H),3.74(s,3H),3.70(t,J=4.5Hz,2H)。 13 C NMR(151MHz,CDCl 3 )δ(ppm):163.48,161.98,156.86,155.43,152.09,151.22,151.04,147.73,137.79,132.95,129.63,128.80,126.57,125.68,124.08,113.97,112.34,101.16,98.26,47.39,31.24,23.76,22.58,19.69。
Example 3N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Figure BDA0002404519330000161
Step 1) N- (4-bromophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Will contain 2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b]A solution of pyrazole-3-carboxylic acid (700 mg,2.87mmol, 100%), 4-bromoaniline (490 mg,2.87 mmol) and HATU (1.15 g,2.87 mmol) in dichloromethane (20 mL) was stirred at room temperature for 4 hours. The reaction was quenched with water (30 mL), extracted with dichloromethane (50 mL x 3), the organic phases combined and washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound as a white solid (10200 mg, 89.4%). MS (ESI, pos.ion) m/z 398.1[ M+H ]] +
Step 2) 2-oxo-1-phenyl-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2,4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To a composition containing N- (4-bromophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ]Pyrazole-3-carboxamide (10200 mg,2.56 mmol), bis-methyl-amideBippinacol diboron (733 mg,2.88 mmol) and Pd (dppf) Cl 2 To a solution of (28mg, 0.39 mmol) in 1, 4-dioxane (20 mL) was added potassium acetate (630 mg,6.42 mmol). The reaction solution was heated to reflux under nitrogen protection, stirred for 7 hours, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (DCM/MeOH (v/v) =20/1) to give the title compound (900 mg, 78.9%) as a white solid. MS (ESI, pos.ion) m/z 446.3[ M+H ]] +
Step 3) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To a catalyst containing 2-oxo-1-phenyl-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2,4,5, 6-tetrahydro-1H-pyrrolo [1,2-b]Pyrazole-3-carboxamide (600 mg,1.34 mmol) and 5-bromo-7-methyl-7H-pyrrolo [2, 3-d)]Pd (dppf) Cl was added to a mixture of pyrimidine-4-amine (343 mg,1.51 mmol) in 1, 4-dioxane (20 mL) and water (2 mL) 2 (II) (200 mg, 0.267 mmol) and cesium carbonate (876 mg,2.69 mmol). The reaction solution was heated to reflux under nitrogen protection, stirred for 7 hours, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) =1/20) to give the title compound (120 mg, 19.13%) as a white solid.
MS(ESI,pos.ion)m/z:466.1[M+H] + 。HRMS(ESI + ) Calculated value C 26 H 23 N 7 O 2 [M+H] + 466.1913, found 466.1983. 1 H NMR(400MHz,CDCl 3 )δ(ppm):10.27(s,1H),8.36(s,1H),7.79(d,J=8.4Hz,2H),7.54(d,J=7.6Hz,2H),7.45(d,J=8.4Hz,4H),6.94(s,1H),5.14(s,2H),3.86(s,3H),3.75(s,2H),3.36(s,2H),2.56(s,2H),1.28(s,2H)。 13 C NMR(101MHz,DMSO-d 6 )(ppm):165.82,161.80,160.73,157.62,151.98,151.61,150.89,137.81,134.86,129.99,129.79,129.39,127.77,125.57,124.59,123.87,119.91,115.22,100.35,98.84,97.02,49.94,31.18,26.01,22.26。
Example 4N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide
Figure BDA0002404519330000171
Step 1) (tert-butyl 4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) carbamate
To a mixture of 1, 4-dioxane (200 mL) and water (50 mL) containing (4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -phenyl) carbamic acid tert-butyl ester (16.88 g,52.88 mmol), 5-bromo-7-methyl-pyrrolo [2,3-d ] pyrimidin-4-amine (10 g,44.04 mmol) was added tetrakis (triphenylphosphine) palladium (0) (5.2 g,4.50 mmol) and cesium carbonate (36 g,110 mmol). The reaction mixture was heated to 105℃under nitrogen and stirred for 24.5 hours, water (200 mL) and ethyl acetate (100 mL) were added and stirred for 10min, the aqueous phase was separated, extracted with ethyl acetate (250 mL. Times.3), the organic phases were combined and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EtOAc (v/v) =1/1) to give the title compound (11.63 g, 77.8%) as a yellow solid.
Step 2) 5- (4-aminophenyl) -7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-4-amine
A solution of tert-butyl (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) carbamate (4.5 g,13 mmol) and concentrated hydrochloric acid (13 mL, 12M) in methanol (75 mL) was heated to 60℃and stirred for 5 hours. The reaction solution was concentrated under reduced pressure, and methylene chloride (100 mL) was added thereto, followed by basification with saturated sodium hydrogencarbonate solution (100 mL). The aqueous phase was separated and extracted with dichloromethane (200 mL), the organic phases were combined and washed with water (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (2.58 g, 81%) as a yellow solid.
Step 3) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ] [1,4] oxazine-3-carboxamide
To a compound containing 2-oxo-1-phenyl-2, 4,6, 7-tetrahydro-1H-pyrazolo [5,1-c ]][1,4]Oxazine-3-carboxylic acid (100 mg,0.384 mmol) and 5- (4-aminophenyl) -7-methyl-7H-pyrrolo [2,3-d ]]To a solution of pyrimidin-4-amine (90 mg, 0.356 mmol) in N, N-dimethylformamide (4 mL) were added EDCI (101 mg,0.516 mmol) and HOAT (112 mg,0.806 mmol). Will be reversedThe reaction solution was heated to 55℃and stirred for 6 hours, and concentrated under reduced pressure. The residue was washed with water (10 mL) to give the title compound (65.6 mg, 32.0%) as a brown solid. MS (ESI, pos.ion) m/z 482.1[ M+H ]] + 。HRMS(ESI + ) Calculated value C 26 H 24 N 7 O 3 [M+H] + 482.19, found 482.1928. 1 H NMR(400MHz,DMSO-d 6 )δ(ppm):10.48(s,1H),8.12(s,1H),7.74(d,J=8.6Hz,2H),7.64-7.57(m,2H),7.56-7.48(m,6H),7.15(s,2H),6.64(s,1H),5.13(s,2H),4.10(t,J=4.8Hz,2H),3.73-3.68(m,5H)。 13 C NMR(151MHz,DMSO-d 6 )δ(ppm):162.07,161.09,151.56,149.67,138.83,137.15,132.36,129.98,129.74,129.63,129.32,127.63,126.95,121.18,119.54,102.86,98.57,96.04,63.56,63.23,45.70,30.18。
Example 5N- (4- (4-amino-7-cyclopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Figure BDA0002404519330000172
Step 1) 4-chloro-7-cyclopropyl-5-iodo-7H-pyrrolo [2,3-d ] pyrimidine
To a compound containing 4-chloro-5-iodo-7H-pyrrolo [2,3-d]To a solution of pyrimidine (5.00 g,17.89 mmol) in DCE (300 ml) was added cyclopropylboronic acid (3.07 g,35.78 mmol), copper acetate (3.57 g,17.89 mmol), sodium carbonate (3.79 g,35.78 mmol) and 2,2' -bipyridine (2.79 g,17.89 mmol). The reaction was heated to reflux and stirred for 1 hour, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) =1/10) to give the title compound (2.30 g, 40.2%) as a white solid. MS (ESI, pos.ion) m/z 320.0[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.64(s,1H),7.93(s,1H),3.65-3.55(m,1H),1.07(dd,J=10.2,3.2Hz,4H)。
Step 2) (tert-butyl 4- (4-chloro-7-cyclopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) carbamate
To a compound containing 4-chloro-7-cyclopropyl-5-iodo-7H-pyrrolo [2,3-d]Pyrimidine (2.30 g,7.20 mmol) 1, 4-dioxaneTo a mixture of ring (75 mL) and water (15 mL) was added tert-butyl (4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) carbamate (2.30 g,7.20 mmol), 1' -bis (diphenylphosphine) ferrocene palladium (II) dichloride (0.53 g,0.72 mmol) and sodium carbonate (2.29 g,21.60 mmol). The reaction was heated to reflux and stirred for 8 hours, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (EtOAc/PE (v/v) =1/3) to give the title compound (2.40 g, 86.6%) as a white solid. MS (ESI, pos.ion) m/z 385.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δ(ppm):9.42(s,1H),8.66(s,1H),7.70(s,1H),7.51(d,J=8.4Hz,2H),7.39(d,J=8.5Hz,2H),3.70-3.60(m,1H),1.14(s,2H),1.10(d,J=2.4Hz,2H),1.07(s,9H)。
Step 3) 5- (4-aminophenyl) -7-cyclopropyl-7H-pyrrolo [2,3-d ] pyrimidin-4-amine
In an autoclave, a mixture containing (4- (4-chloro-7-cyclopropyl-7H-pyrrolo [2, 3-d)]To a solution of t-butyl pyrimidyl-5-yl) phenyl carbamate (1.50 g,3.90 mmol) in 1, 4-dioxane (10 ml) was added aqueous ammonia (50 ml,25% in water). The reaction was heated to 130 ℃ and stirred for 24 hours, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (MeOH/DCM (v/v) =1/30) to give the title compound (0.70 g, 68.0%) as a yellow solid. MS (ESI, pos.ion) m/z 266.2[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.12(s,1H),7.10(d,J=8.2Hz,2H),7.03(s,1H),6.65(d,J=8.2Hz,2H),5.98(s,2H),5.19(s,2H),3.58-3.49(m,1H),1.04-0.97(m,4H)。
Step 4) N- (4- (4-amino-7-cyclopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To a compound containing 5- (4-aminophenyl) -7-cyclopropyl-7H-pyrrolo [2,3-d]To a solution of pyrimidin-4-amine (0.25 g,0.94 mmol) in dichloromethane (30 ml) was added 2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ]]Pyrazole-3-carboxylic acid (0.23 g,0.94 mmol), EDCI (0.36 g,1.89 mmol) and HOAT (0.26 g,1.89 mmol). The reaction mixture was heated to reflux and stirred overnight, quenched with water (50 ml) and extracted with dichloromethane (200 ml x 3). The organic phases were combined, washed with saturated sodium bicarbonate solution (50 mL) and brine (100ml x 2), 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 (0.30 g, 65.0%) as a yellow solid. MS (ESI, pos.ion) m/z 492.2[ M+H ]] + 。HRMS(ESI + ) Calculated value C 28 H 26 N 7 O 2 [M+H] + 492.2148, found 492.2140. 1 H NMR(400MHz,DMSO-d 6 )δ(ppm):10.26(s,1H),8.18(s,1H),7.72(d,J=8.4Hz,2H),7.54(dd,J=14.5,7.2Hz,4H),7.42(d,J=8.3Hz,3H),7.25(s,1H),6.17(s,2H),3.81(t,J=6.9Hz,2H),3.59-3.48(m,1H),3.18(t,J=7.3Hz,2H),2.47-2.38(m,2H),1.04(dd,J=7.9,6.1Hz,4H)。 13 C NMR(151MHz,DMSO-d 6 )δ(ppm):165.79,161.79,160.73,157.30,151.85,151.60,151.57,151.55,137.87,134.83,129.81,129.74,129.47,129.34,127.78,123.84,123.02,121.21,119.81,115.27,100.76,96.98,60.23,49.95,27.12,26.02,22.26,6.50。
Example 6N- (5- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) pyridin-2-yl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
Figure BDA0002404519330000181
Step 1) N- (5-bromopyridin-2-yl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
To 2-oxo-1-phenyl-4, 5,6, 7-tetrahydropyrazolo [1,5-a ]]To a mixture of pyridine-3-carboxylic acid (1.6 g,6.2 mmol) in DCM (50 mL) was added 1-hydroxy 7-azabenzotriazole (170 mg,1.22 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrobromide (1.6 g,8.2 mmol) and 5-bromopyridine-2-amine (1.2 g,6.7 mmol). The mixture was heated to 45℃and stirred for 21h. TLC showed no starting material present, then water (20 mL) was added dropwise to the mixture, and the resulting mixture was extracted twice with DCM (50 mL) and concentrated in vacuo to give a yellow solid. The residue was purified by flash column chromatography (PE: etoac=1:1-0:1) to give N- (5-bromo-2-pyridinyl) -2-oxo-1-phenyl-4, 5,6, 7-tetrahydropyrazolo [1,5-a ]]Pyridine-3-carboxamide is yellowSolid (1.35 g,3.27mmol, 53% yield). MS (ESI, pos.ion) m/z 412.9[ M+H ]] +
Step 2) 2-oxo-1-phenyl-N- (5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridin-2-yl) -1,2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
To N- (5-bromo-2-pyridinyl) -2-oxo-1-phenyl-4, 5,6, 7-tetrahydropyrazolo [1,5-a ]]Pyridine-3-carboxamide (1.35 g,3.27 mmol), 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (0.95 g,3.7 mmol) and Pd (dppf) 2 Cl 2 To a suspension of (0.4 g,0.5 mmol) was added a solution of KOAc (0.8 g,8 mmol) in 1, 4-dioxane (30 mL). The reaction mixture was taken up in N 2 Reflux stirring is carried out for 5h under atmosphere. TLC showed no starting material present and concentrated in vacuo to give the title compound (1.5 g,3.27mmol, 100.0% yield).
Step 3) N- (5- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) pyridin-2-yl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrrolo [1,5-a ] pyridine-3-carboxamide
To 2-oxo-1-phenyl-N- [5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridine]-4,5,6, 7-tetrahydropyrazolo [1,5-a ]]Pyridine-3-carboxamide (1.5 g,3.3 mmol) and 5-bromo-7-methyl-pyrrolo [2, 3-d)]Pyrimidin-4-amine (1.0 g,4.4 mmol), H 2 To a solution of 1, 4-dioxane (10 mL) of O (5 mL) was added 1,1' -bis (diphenylphosphino) ferrocene-palladium (II) dichloride dichloromethane complex (0.66 g,0.79 mmol) and Cs 2 CO 3 (2.6 g,8.0 mmol). The mixture is put under N 2 Stirring overnight at 105℃under an atmosphere, and concentrating in vacuo. The residue was purified by flash column chromatography (DCM: meoh=50:1-10:1) to give a yellow solid (240 mg). Purifying the solid with an anion separation column to obtain N- (5- (4-amino-7-methyl-7H-pyrrolo [2, 3-d)]Pyrimidin-5-yl) pyridin-2-yl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ]Pyridine-3-carboxamide is a white solid (6.9 mg,0.014mmol, yield 0.44%). MS (ESI, pos.ion) m/z 481.4[ M+H ]]+。 1 H NMR(600MHz,DMSO)δ11.84(s,1H),8.62(d,J=8.3Hz,1H),8.48(s,1H),8.15(dd,J=7.9,1.4Hz,1H),7.51–7.48(m,1H),7.26(d,J=8.7Hz,2H),7.25–7.21(m,1H),6.92(d,J=8.7Hz,2H),5.41(s,2H),3.72(s,4H),2.55(s,3H)。 13 C NMR(151MHz,CDCl 3 )δ163.48,161.98,156.86,155.43,152.09,151.22,151.04,147.73,137.79,132.95,129.63,128.80,126.57,125.68,124.08,113.97,112.34,101.16,98.26,47.39,31.24,23.76,22.58,19.69。
Example 7N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) -3-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Figure BDA0002404519330000191
Step 1) N- (4-bromo-3-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To 4-bromo-3-fluoroaniline (200 mg,1.0 mmol) in CH 2 Cl 2 To the solution (6 mL) was added 2-oxo-1-phenyl-5, 6-dihydro-4H-pyrrolo [1,2-b]Pyrazole-3-carboxylic acid (274 mg,1.1 mmol), EDCI (240 mg,12 mmol) and HOAT (28 mg,0.2 mmol). After stirring at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give the title compound as a yellow solid (260 mg, 61%). MS (ESI, pos.ion) M/z 416.0 (M+1). 1 H NMR(400MHz,CDCl 3 )δ10.31(s,1H),7.84-7.81(dd,J=10.9,2.3Hz,1H),7.56-7.52(t,J=7.8Hz,2H),7.47-7.39(m,4H),7.19-7.17(dd,J=8.6,1.9Hz,1H),3.76-3.73(t,J=6.9Hz,2H),3.34-3.31(t,J=7.4Hz,2H),2.59-2.52(m,2H)。
Step 2) N- (3-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To N- (4-bromo-3-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b]To a solution of pyrazole-3-carboxamide (260 mg,0.62 mmol) in dioxane (6 mL) was added 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (178 mg,0.69 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (52mg,0.063mmol) And potassium acetate (161 mg,1.6 mmol). The reaction was degassed with nitrogen and after stirring the mixture under reflux overnight the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give the title compound as a yellow solid (210 mg, 73%). MS (ESI, pos.ion) M/z 464.2 (M+1).
Step 3) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) -3-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To N- (3-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b]Pyrazole-3-carboxamide (210 mg,0.45 mmol) dioxane (6 mL)/H 2 To a solution of O (1 mL) was added 5-bromo-7-methyl-pyrrolo [2, 3-d)]Pyrimidine-4-amine (108 mg,0.45 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (37 mg,0.045 mmol) and cesium carbonate (301 mg,0.91 mmol). The reaction was degassed with nitrogen and after stirring the mixture under reflux overnight the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=500/1) to give the title compound as a yellow solid (46 mg, 21%). MS (ESI, pos.ion) M/z 484.2 (M+1). 1 H NMR(400MHz,DMSO)δ10.38(s,1H),8.15(s,1H),7.88-7.85(d,J=12.8Hz,1H),7.60-7.52(m,4H),7.45-7.41(t,J=7.3Hz,1H),7.36-7.35(m,2H),7.30(s,1H),6.03(s,2H),3.85-2.81(t,J=6.9Hz,2H),3.75(s,3H),3.20-3.17(t,J=6.8Hz,2H),2.46-2.43(m,2H)。
Example 8N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) -2-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Figure BDA0002404519330000201
Step 1) 2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carbonyl chloride
To 2-oxo-1-phenyl-5, 6-dihydro-4H-pyrrolo [1,2-b]Pyrazole-3-carboxylic acid (200 mg,0.82 mmol) CH 2 Cl 2 DMF (1 mg,0.014 mmol) and oxalyl were added to a solution (5 mL)Chlorine (0.1 mL,1 mmol). After stirring at room temperature for 4 hours, the reaction mixture was concentrated in vacuo, which was used in the next step without further purification.
Step 2) N- (4-bromo-2-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To 4-bromo-2-fluoroaniline (175 mg,0.9 mmol) in CH 2 Cl 2 To a solution of (3 mL) was added triethylamine (0.25 mL,1.8 mmol) and the reaction mixture was stirred at 0deg.C. Adding 2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b to the reaction mixture]CH of pyrazole-3-carbonyl chloride (215 mg,0.82 mmol) 2 Cl 2 (2 mL) solution. After stirring overnight at room temperature, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give the title compound as a yellow solid (0.15 g,44%, two steps). MS (ESI, pos.ion) M/z 416.0 (M+1). 1 H NMR(400MHz,CDCl 3 )δ10.47(s,1H),8.46-8.42(t,J=8.6Hz,1H),7.55-7.51(t,J=7.8Hz,2H),7.45-7.43(d,J=7.6Hz,2H),7.41-7.37(t,J=7.4Hz,1H),7.30-7.25(m,2H),3.76-3.73(t,J=6.9Hz,2H),3.35-3.31(t,J=7.4Hz,2H),2.59-2.51(m,2H)。
Step 3) N- (2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To N- (4-bromo-2-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b]To a solution of pyrazole-3-carboxamide (150 mg,0.36 mmol) in dioxane (6 mL) was added 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (187 mg,0.72 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (30 mg,0.036 mmol) and potassium acetate (93 mg,0.9 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give the title compound as a yellow solid (135 mg, 81%). MS (ESI, pos.ion) M/z 464.3 (M+1). 1 H NMR(400MHz,CDCl 3 )δ10.59-10.58(d,J=2.3Hz,1H),8.57-8.53(t,J=7.8Hz,1H),7.59-7.54(dd,J=11.6,6.5Hz,2H),7.52-7.50(d,J=8.0Hz,2H),7.45-7.44(d,J=7.6Hz,2H),7.40-7.36(t,J=7.3Hz,1H),3.76-3.72(t,J=6.9Hz,2H),3.36-3.32(t,J=7.4Hz,2H),2.58-2.51(m,2H),1.35(s,12H)。
Step 4) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) -2-fluorophenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To N- (2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b]Pyrazole-3-carboxamide (135 mg,0.29 mmol) dioxane (6 mL)/H 2 To a solution of O (1 mL) was added 5-bromo-7-methyl-pyrrolo [2,3-d]Pyrimidine-4-amine (70 mg,0.29 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (24 mg,0.029 mmol) and cesium carbonate (194 mg,0.58 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=500/1) to give the title compound as a yellow solid (46 mg, 33%). MS (ESI, pos.ion) M/z 484.2 (M+1). 1 H NMR(400MHz,DMSO)δ10.51-10.50(d,J=1.8Hz,1H),8.53-8.49(t,J=8.4Hz,1H),8.16(s,1H),7.59-7.52(m,4H),7.45-7.41(t,J=7.0Hz,1H),7.35-7.32(m,2H),7.27-7.25(d,J=8.5Hz,1H),6.18(s,2H),3.84-3.81(t,J=6.8Hz,2H),3.74(s,3H),3.20-3.17(t,J=7.2Hz,2H),2.48-2.41(m,2H)。
Example 9N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) -2-fluorophenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
Figure BDA0002404519330000211
Step 1) 1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carbonyl chloride
To 1, 5-dimethyl-3-oxo-2-phenyl-pyrazole-4-carboxylic acid (300 mg,1.3 mmol) in CH 2 Cl 2 To a solution of (6 mL) was added DMF (1 mg,0.014 mmol) and oxalyl chloride (0.2 mL,2 mmol). After stirring at room temperature for 4 hours, the reaction mixture was concentrated in vacuo, which was done without further purificationWhich can be used for the next step.
Step 2) N- (4-bromo-2-fluorophenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
To CH of 4-bromo-2-fluoroaniline (274 mg,1.4 mmol) 2 Cl 2 To a solution of (4 mL) was added triethylamine (0.36 mL,2.6 mmol) and the reaction mixture was stirred at 0deg.C. To the reaction mixture was added 1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carbonyl chloride (322 mg,1.3 mmol) in CH 2 Cl 2 (2 mL) solution. After stirring overnight at room temperature, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give the title compound as a yellow solid (0.44 g,85%, two steps). MS (ESI, pos.ion) M/z 404.0 (M+1). 1 H NMR(400MHz,CDCl3)δ10.98(s,1H),8.46-8.42(t,J=8.7Hz,1H),7.58-7.54(t,J=7.6Hz,2H),7.49-7.46(t,J=7.4Hz,1H),7.39-3.37(d,J=7.4Hz,2H),7.28-7.25(m,2H),3.37(s,3H),2.80(s,3H)。
Step 3) N- (2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
To a solution of N- (4-bromo-2-fluorophenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide (440 mg,1.1 mmol) in dioxane (6 mL) was added 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (564 mg,2.2 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (90 mg,0.11 mmol) and potassium acetate (281mg, 2.7 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give the title compound as a yellow solid (410 mg, 83%). MS (ESI, pos.ion) M/z 452.3 (M+1). 1 H NMR(600MHz,CDCl 3 )δ11.10(s,1H),8.57-8.54(t,J=7.8Hz,1H),7.58-7.53(m,4H),7.48-7.45(t,J=7.5Hz,1H),7.39-7.37(d,J=7.4Hz,2H),3.37(s,3H),2.81(s,3H),1.35(s,12H)。
Step 4) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) -2-fluorophenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
To N- (2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide (410 mg,0.91 mmol) dioxane (8 mL)/H 2 To a solution of O (1.5 mL) was added 5-bromo-7-methyl-pyrrolo [2, 3-d)]Pyrimidine-4-amine (217 mg,0.91 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (75 mg,0.091 mmol) and cesium carbonate (604 mg,1.8 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=500/1) to give the title compound as a yellow solid (78 mg, 18%). MS (ESI, pos.ion) M/z 472.1 (M+1). 1 H NMR(400MHz,DMSO)δ11.05(s,1H),8.54-8.50(t,J=8.4Hz,1H),8.16(s,1H),7.62-7.59(t,J=7.5Hz,2H),7.55-7.51(m,1H),7.46-7.44(d,J=7.6Hz,2H),7.35-7.30(m,2H),7.26-7.24(d,J=8.3Hz,1H),6.17(s,2H),3.74(s,3H),3.38(s,3H),2.73(s,3H)。
Example 10N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
Figure BDA0002404519330000212
Step 1) N- (4-bromophenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
To 4-bromoaniline (500 mg,2.9 mmol) in CH 2 Cl 2 To a solution (15 mL) was added 1, 5-dimethyl-3-oxo-2-phenyl-pyrazole-4-carboxylic acid (739 mg,3.2 mmol), EDCI (675 mg,3.5 mmol) and HOAT (80 mg,0.58 mmol). After stirring at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give the title compound as a yellow solid (340 mg, 31%). MS (ESI, pos.ion) M/z 386.0 (M+1). 1 H NMR(400MHz,CDCl 3 )δ10.74(s,1H),7.59-7.55(m,4H),7.50-7.47(t,J=7.4Hz,1H),7.43-7.41(d,J=8.8Hz,2H),7.38-7.36(d,J=7.7Hz,2H),3.37(s,3H),2.80(s,3H)。
Step 2) 1, 5-dimethyl-3-oxo-2-phenyl-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2, 3-dihydro-1H-pyrazole-4-carboxamide
To a solution of N- (4-bromophenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide (340 mg,0.88 mmol) in dioxane (6 mL) was added 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (250 mg,0.97 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (73 mg,0.088 mmol) and potassium acetate (227 mg,2.2 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give the title compound as a yellow solid (280 mg, 73%). MS (ESI, pos.ion) M/z 434.3 (M+1). 1 H NMR(400MHz,CDCl3)δ10.81(s,1H),7.79-7.77(d,J=8.3Hz,2H),7.71-7.69(d,J=8.3Hz,2H),7.58-7.54(t,J=7.7Hz,2H),7.49-7.46(t,J=7.4Hz,1H),7.38-7.37(d,J=7.5Hz,2H),3.36(s,3H),2.81(s,3H),1.35(s,12H)。
Step 3) N- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazole-4-carboxamide
To 1, 5-dimethyl-3-oxo-2-phenyl-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -2, 3-dihydro-1H-pyrazole-4-carboxamide (280 mg,0.65 mmol) dioxane (6 mL)/H 2 To a solution of O (1 mL) was added 5-bromo-7-methyl-pyrrolo [2,3-d]Pyrimidine-4-amine (154 mg,0.64 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (53 mg,0.064 mmol) and cesium carbonate (430 mg,1.3 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at reflux overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=500/1) to give the title compound as a yellow solid (68 mg, 23%). MS (ESI, pos.ion) M/z 454.3 (M+1). 1 H NMR(400MHz,DMSO)δ10.82(s,1H),8.15(s,1H),7.71-7.69(d,J=8.5Hz,2H),7.62-7.58(t,J=7.6Hz,2H),7.54-7.50(t,J=7.4Hz,1H),7.46-7.44(d,J=7.4Hz,2H),7.41-7.39(d,J=8.4Hz,2H),7.28(s,1H),6.06(s,2H),3.74(s,3H),3.37(s,3H),2.72(s,3H)。
Example 11N- (4- (4-amino-7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-1, 2,4,5,6, 7-hexahydropyrazolo [1,5-a ] pyridine-3-carboxamide
Figure BDA0002404519330000221
To 5- (4-aminophenyl) -7-isopropyl-7H-pyrrolo [2,3-d]Pyrimidin-4-amine (100 mg,0.37 mmol) CH 2 Cl 2 To the (2 mL) solution was added 2-oxo-1-phenyl-4, 5,6, 7-tetrahydropyrazolo [1,5-a ]]Pyridine-3-carboxylic acid (116 mg,0.45 mmol), EDCI (110 mg,0.56 mmol) and HOAT (10 mg,0.072 mmol). After stirring at reflux for 8h, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=100/0) to give the title compound as a yellow solid (97 mg, 51%). MS (ESI, pos.ion) M/z 508.4 (M+1). 1 H NMR(400MHz,DMSO)δ10.68(s,1H),8.13(s,1H),7.71-7.69(d,J=8.4Hz,2H),7.61-7.57(t,J=7.5Hz,2H),7.53-7.51(d,J=7.2Hz,1H),7.48-7.46(d,J=7.4Hz,2H),7.43-7.41(m,3H),6.05(s,2H),5.02-4.92(m,1H),3.59-3.56(t,J=5.7Hz,2H),3.24-3.21(t,J=6.2Hz,2H),1.99-1.98(m,2H),1.84-1.81(m,2H),1.47-1.45(d,J=6.7Hz,6H)。
Example 12N- (4- (4-amino-7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Figure BDA0002404519330000231
Step 1) 4-chloro-5-iodo-7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidine
To 4-chloro-5-iodo-7H-pyrrolo [2,3-d]To a solution of pyrimidine (2.0 g,7.0 mmol) in THF (35 mL) was added potassium tert-butoxide (1.23 g,10.7 mmol) and the reaction mixture was stirred for 0.5h. To the reaction mixture was added 2-iodopropane (0.9 mL,9.0 mmol). After stirring overnight at 70 ℃, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate) Ester=3/1) to give the title compound as a yellow solid (1.42 g, 63%). MS (ESI, pos.ion) M/z 322.1 (M+1). 1 H NMR(400MHz,CDCl3)δ8.61(s,1H),7.47(s,1H),5.18-5.08(m,1H),1.54-1.52(d,J=6.8Hz,6H)。
Step 2) 4- (4-chloro-7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) aniline
To 4-chloro-5-iodo-7-isopropyl-7H-pyrrolo [2,3-d]Pyrimidine (1.31 g,4.07 mmol) dioxane (20 mL)/H 2 To a solution of O (5 mL) was added 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) aniline (0.91 g,4.1 mmol), pd (dppf) Cl 2 .CH 2 Cl 2 (0.34 g,0.41 mmol) and sodium carbonate (0.87 g,8.2 mmol). The reaction was degassed with nitrogen. After the mixture was stirred at 90 ℃ overnight, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give the title compound as a yellow solid (0.68 g, 58%). MS (ESI, pos.ion) M/z 287.2 (M+1). 1 H NMR(400MHz,DMSO)δ8.60(s,1H),7.80(s,1H),7.19-7.17(d,J=8.3Hz,2H),6.63-6.61(d,J=8.3Hz,2H),5.14(s,2H),5.11-5.06(m,1H),1.52-1.50(d,J=6.7Hz,6H)。
Step 3) 5- (4-aminophenyl) -7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidin-4-amine
To 4- (4-chloro-7-isopropyl-7H-pyrrolo [2, 3-d)]To a solution of pyrimidin-5-yl) aniline (0.68 g,2.4 mmol) in dioxane (20 mL) was added ammonium hydroxide (20 mL). After stirring the mixture in a closed tube at 130 ℃ for 24 hours, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=100/1) to give the title compound as a yellow solid (0.30 g, 47%). MS (ESI, pos.ion) M/z 268.2 (M+1). 1 H NMR(400MHz,DMSO)δ8.09(s,1H),7.23(s,1H),7.13-7.11(d,J=8.2Hz,2H),6.67-6.65(d,J=8.2Hz,2H),5.97(s,2H),5.16(s,2H),4.99-4.89(m,1H),1.45-1.43(d,J=6.7Hz,6H)。
Step 4) N- (4- (4-amino-7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
To 5- (4-aminophenyl) -7-isopropyl-7H-pyrrolo [2,3-d]Pyrimidin-4-amine (100 mg,0.37mmol) CH 2 Cl 2 To the (2 mL) solution was added 2-oxo-1-phenyl-5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole-3-carboxylic acid (110 mg,0.45 mmol), EDCI (110 mg,0.56 mmol) and HOAT (10 mg,0.072 mmol). After stirring the mixture under reflux for 9h, the reaction mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (dichloromethane/methanol=100/0) to give the title compound as a yellow solid (94 mg, 51%). MS (ESI, pos.ion) M/z 494.2 (M+1). 1 H NMR(400MHz,DMSO)δ10.26(s,1H),8.13(s,1H),7.74-7.72(d,J=8.3Hz,2H),7.59-7.51(m,4H),7.44-7.41(m,4H),6.06(s,2H),5.00-4.92(td,J=13.3,6.6Hz,1H),3.82-3.79(t,J=6.8Hz,2H),3.20-3.17(t,J=5.8Hz,3H),2.46-2.40(m,2H),1.47-1.46(d,J=6.7Hz,6H)。
Example 13N- (4- (4-amino-7- (cyclopropylmethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
Figure BDA0002404519330000232
Step 1) 4-chloro-7- (cyclopropylmethyl) -5-iodo-7H-pyrrolo [2,3-d ] pyrimidine
4-chloro-5-iodo-7H-pyrrolo [2,3-d]Pyrimidine (12.00 g,42.94 mmol), KOH (4.82 g,85.9 mmol) in DMSO (60 mL) was reacted at 53 ℃. 2-Bromoethanol (12 mL) was then added dropwise to the mixture, and the reaction mixture was maintained for 24 hours. The mixture was poured into water (150 mL) and filtered in vacuo to give the title compound as a yellow oil (11.53 g, 83.00%). MS (ESI, pos.ion) m/z 323.90[ M+H ] ] +1 H NMR(400MHz,DMSO)δ8.63(s,1H),7.98(s,1H),4.93(t,J=5.4Hz,1H),4.31(t,J=5.4Hz,2H),3.75(q,J=5.4Hz,2H)。
Step 2) (4- (4-chloro-7- (cyclopropylmethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) carbamic acid tert-butyl ester
(4- (4-chloro-7- (cyclopropylmethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-5-yl) phenyl tert-butyl carbamate (2.66 g,7.97 mmol), N- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl]Tert-butyl carbamate (3.03 g,9.49 mm)ol),H 2 O(7.5mL),Pd(dppf)Cl 2 (0.85 g,0.79 mmol) and Na 2 CO 3 A mixture of (2.12 g,20.0 mmol) of 1, 4-dioxane (35 mL) was stirred at 93℃for 12h and then concentrated in vacuo. The residue was purified by flash column chromatography (PE: ea=5:1) to give the title compound as a yellow solid (1.37 mg, 43.1%). MS (ESI, pos.ion) m/z 399.10[ M+H ]] +
Step 3) (4- (4-amino-7- (cyclopropylmethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) carbamic acid tert-butyl ester
(4- (4-amino-7- (cyclopropylmethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-5-yl) phenyl-carbamic acid tert-butyl ester (1.37 g,3.43 mmol) and NH 4 A mixture of OH (28%, 40 mL) and 1, 4-dioxane (10 mL) was stirred at 120deg.C for 37h and then concentrated in vacuo. The residue was purified by flash column chromatography (DCM: meoh=30:1) to give the title compound as a yellow solid (675.6 mg, 51.8%). MS (ESI, pos.ion) m/z 280.20[ M+H ] ] +
Step 4) 5- (4-aminophenyl) -7- (cyclopropylmethyl) -7H-pyrrolo [2,3-d ] pyrimidin-4-amine
5- (4-aminophenyl) -7- (cyclopropylmethyl) -7H-pyrrolo [2,3-d]A mixture of pyrimidin-4-amine (675.6 mg,1.78 mmol) and HCl (37%, 5 mL) in methanol (10 mL) was stirred at 60℃for 5h and then concentrated in vacuo. Vacuum filtration gave the title compound as a yellow solid (312.5 mg, 62.85%). MS (ESI, pos.ion) m/z 280.20[ M+H ]] +
Step 5) N- (4- (4-amino-7- (cyclopropylmethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -2-oxo-1-phenyl-2, 4,5, 6-tetrahydro-1H-pyrrolo [1,2-b ] pyrazole-3-carboxamide
2-oxo-1-phenyl-5, 6-dihydro-4H-pyrrolo [1,2-b]Pyrazole-3-carboxylic acid (139.6 mg,0.57 mmol), 5- (4-aminophenyl) -7- (cyclopropylmethyl) pyrrolo [2, 3-d)]A mixture of pyrimidine-4-amine (159.9 mg,0.5725 mmol), HOAT (24.3 mg,0.18 mmol) and EDCI (198.5 mg,1.03 mmol) in DCM (10 mL) was stirred at 45℃for 12h. The mixture was concentrated in vacuo to give the title compound as a yellow solid (103.5 g, 35.76%). 1 H NMR(400MHz,DMSO)δ10.26(s,1H),8.14(s,1H),7.73(d,J=8.4Hz,2H),7.63–7.49(m,4H),7.46–7.33(m,4H),6.09(s,2H),4.02(d,J=7.1Hz,2H),3.80(t,J=6.8Hz,2H),3.18(dd,J=9.5,4.9Hz,2H),2.47–2.38(m,2H),0.92–0.77(m,1H),0.57–0.46(m,2H),0.43(d,J=4.0Hz,2H)。 13 C NMR(151MHz,DMSO)δ165.80,161.80,160.73,157.70,151.98,150.57,137.78,134.84,130.06,129.80,129.41,127.77,123.84,123.57,119.87,115.24,100.33,97.01,49.95,48.51,26.02,22.26,12.08,4.13。MS(ESI,pos.ion)m/z:503.20[M+H] +
Examples 14 to 17 and example 20 were prepared by a similar method to example 2, and examples 18 to 19 were prepared by a similar method to example 10, see for details table E:
Table E
Figure BDA0002404519330000241
/>
Figure BDA0002404519330000251
Biological assay
The LC/MS/MS system for analysis comprises an Agilent 1200 series vacuum degassing furnace, a binary injection pump, an orifice plate automatic sampler, a column oven and an Agilent G6430 three-stage quadrupole mass spectrometer of an Electrified Spray Ionization (ESI) source. Quantitative analysis was performed in MRM mode, and parameters of MRM conversion are shown in table a:
table A
Multiple reaction detection scan 490.2→383.1
Fracture voltage 230V
Capillary voltage 55V
Drying gas temperature 350℃
Atomizer 0.276MPa
Drying air flow rate 10L/min
Analysis Using an Agilent XDB-C18, 2.1X130 mm, 3.5. Mu.M column, 5. Mu.L of sample was injected. Analysis conditions: the mobile phase was 0.1% formic acid in water (A) and 0.1% formic acid in methanol (B). The flow rate was 0.4mL/min. The mobile phase gradient is 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 Termination of
Also for analysis was an Agilent 6330 series LC/MS spectrometer equipped with a G1312A binary syringe pump, a G1367A autosampler and a G1314C UV detector; LC/MS spectrometers employ ESI radiation sources. The appropriate cation model treatment and MRM transformations were performed for each analyte using standard solutions for optimal analysis. Capcell MP-C18 column was used during the analysis, with the following 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 0.6mL/min; the column temperature is kept at room temperature; 20. Mu.L of sample was injected.
Example a stability in human and rat liver microsomes
Human or rat liver microsomes were placed in polypropylene tubes, double Kong Fuyo. Typical incubation mixtures include human or rat liver microsomes (0.5 mg protein/mL), target compound (5 μm) and NADPH (1.0 mM) potassium phosphate buffer (PBS, 100mM, ph 7.4) in a total volume of 200 μl, test compounds were 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, 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 terminated by adding the same volume of ice-cold acetonitrile. The samples were stored at-80℃until LC/MS/MS analysis was performed.
The concentration of the compound in the human or rat liver microsome incubation mixture was determined by LC/MS method. The linear range of concentration ranges is determined by each test compound.
Parallel incubation experiments were performed using denatured microsomes as negative control, incubated at 37 ℃ and the reactions terminated at different time points (0, 15 and 60 minutes).
Verapamil (1 μm) served as a positive control, incubated at 37 ℃, and the reactions were terminated at different time points (0, 5, 10, 15, 30 and 60 minutes). Positive and negative control samples were included in each assay to ensure the integrity of the microsomal hatching system.
Furthermore, stability data of the compounds of the present invention in human or rat liver microsomes can also be obtained from the following test. Human or rat liver microsomes were placed in polypropylene tubes, double Kong Fuyo. Typical incubation mixtures include human or rat liver microsomes (final concentration: 0.5mg protein/mL), test compound (final concentration: 1.5. Mu.M) and a total volume of 30. Mu.L potassium phosphate buffer (containing 1.0mM EDTA,100mM,pH 7.4). Test compounds were dissolved in DMSO and diluted with potassium phosphate buffer to a final DMSO concentration of 0.2%. After 10 minutes of pre-incubation, 15. Mu.L of NADPH (final concentration: 2 mM) was added for enzymatic reaction, and the whole experiment was performed in an incubation tube at 37 ℃. At various time points (0, 15, 30 and 60 minutes) the reaction was terminated by the addition of 135. Mu.L of acetonitrile (containing IS). Centrifugation at 4000rpm for 10 minutes, removal of protein, collection of supernatant, analysis with LC-MS/MS. In the above experiments, ketanserin (1 μm) was selected as a positive control, incubated at 37 ℃, and the reactions were terminated at different time points (0, 15, 30 and 60 minutes). Positive and negative control samples were included in each assay to ensure the integrity of the microsomal hatching system.
Data analysis
For each reaction, the concentration of compound (in percent) in human or rat liver microsome incubation was plotted as a percentage of the relative zero time point to infer in vivo hepatic intrinsic clearance CL int (see, e.g., naritomi Y, tershita 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 present invention exhibit suitable stability when incubated in human and rat liver microsomes.
TABLE 1 liver microparticle stability in humans and rats
Figure BDA0002404519330000271
EXAMPLE B pharmacokinetic of mice, rats, dogs and monkeys after oral or intravenous administration of the compounds of the invention Evaluation
Pharmacokinetic studies of the compounds of the invention in mice, rats, dogs or monkeys were evaluated. The compounds of the present invention are administered as aqueous solutions or 2% HPMC+1% Tween-80 in water, 5% DMSO+5% saline solution, 4% MC or in the form of 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 10mg/kg. Blood (0.3 mL) was taken at time points 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 LC/MS/MS analysis as described above was performed.
When the compound provided by the invention is administrated by intravenous injection or oral administration, the compound of the invention shows good pharmacokinetic property, is well absorbed and has ideal half-life (T) 1/2 ) And higher oral bioavailability (F).
TABLE 2 rat PK data
Figure BDA0002404519330000272
Figure BDA0002404519330000281
TABLE 3 mouse PK data
Figure BDA0002404519330000282
TABLE 4 canine PK data
Figure BDA0002404519330000283
TABLE 5 monkey PK data
Figure BDA0002404519330000284
EXAMPLE C kinase Activity assay
The utility of the disclosed compounds as protein kinase inhibitors can be evaluated by the following experiments.
Kinase assay by detection of incorporation of gamma- 33 P-ATP Myelin Basic Protein (MBP). A20. Mu.g/ml solution of MBP (Sigma #M-1891) in Tris buffer (TBS; 50mM Tris pH=8.0, 138mM NaCl,2.7mM KCl) was prepared, coated with a highly binding white 384 well plate (Greiner), 60. Mu.L per well. Incubation was carried out at 4℃for 24 hours. The plate was then washed 3 times with 100. Mu.L TBS. Kinase reaction in a total volume of 34. Mu.L kinase buffer (formulated as needed, e.g., 5mM Hepes pH 7.6, 15mM NaCl,0.01% bovine serum albumin (Sigma #I-5506), 10mM MgCl 2 1mM DTT,0.02%TritonX-100). Compounds were dissolved in DMSO and added to each well with a final concentration of 1% compound in DMSO solution. The assay for each compound was performed at least twice. For example, the final concentration of enzyme is 10nM or 20nM. Addition of unlabeled ATP (10. Mu.M) and gamma-) 33 P-labeled ATP (2X 10 per well) 6 cpm,3000 Ci/mmol) was started. The reaction was run with shaking at room temperature for 1 hour. 384-well plates were washed with 7 XPBS and 50. Mu.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 just one of many detection methods, as well as others.
IC with suppressed test method 50 And/or inhibition constant K i 。IC 50 Defined as the concentration of the compound that inhibited 50% of the enzyme activity under the assay conditions. IC was estimated by making a curve containing 10 concentration points using a dilution factor of 1/2log 50 Values (e.g., a typical curve is made by the following compound concentrations: 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), or 10. Mu.M, 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. Mu.M).
AXL(h)
AXL (h) at 8mM MOPS ph= 7.0,0.2mM EDTA,250 μ M KKSRGDYMTMQIG,10mM magnesium acetate and [ γ ] 33 P-ATP]Incubation was performed in the presence (specific activity and concentration as determined by demand). The reaction was started after the addition of the MgATP mixture. After incubation for 40 minutes at room temperature, the reaction was terminated by adding a phosphoric acid solution thereto to a concentration of 0.5%. mu.L of the reaction solution was spotted on a P30 filter, and the mixture was washed with 0.425% phosphoric acid solution 4 times and methanol 1 time for 4 minutes. After drying, the measurement was performed by a scintillation counter.
TrkA(h)
TrkA (h) at 8mM MOPS ph= 7.0,0.2mM EDTA,250 μ M KKKSPGEYVNIEFG,10mM magnesium acetate and [ γ ] 33 P-ATP]Incubation was performed in the presence (specific activity and concentration as determined by demand). The reaction was started after the addition of the MgATP mixture. After incubation for 40 minutes at room temperature, the reaction was terminated by adding a phosphoric acid solution thereto to a concentration of 0.5%. mu.L of the reaction solution was spotted on a P30 filter, and the mixture was washed with 0.425% phosphoric acid solution 4 times and methanol 1 time for 4 minutes. After drying, the measurement was performed by a scintillation counter.
TrkB(h)
TrkB (h) at 8mM MOPS pH= 7.0,0.2mM EDTA,0.1mg/mL poly (Glu, tyr) 4:1,10mM magnesium acetate and [ gamma ] 33 P-ATP]Incubation was performed in the presence (specific activity and concentration as determined by demand). The reaction was started after the addition of the MgATP mixture. After incubation for 40 minutes at room temperature, the reaction was terminated by adding a phosphoric acid solution thereto to a concentration of 0.5%. mu.L of the reaction solution was spotted on the A filter, and the mixture was washed 4 times with 0.425% phosphoric acid solution and 1 time with methanol for 4 minutes. After drying, useAnd (5) measuring by a scintillation counter.
TrkC(h)
TrkB (h) at 8mM MOPS pH= 7.0,0.2mM EDTA,500 μ M GEEPLYWSFPAKKK,10mM magnesium acetate and [ γ ] 33 P-ATP]Incubation was performed in the presence (specific activity and concentration as determined by demand). The reaction was started after the addition of the MgATP mixture. After incubation for 40 minutes at room temperature, the reaction was terminated by adding a phosphoric acid solution thereto to a concentration of 0.5%. mu.L of the reaction solution was spotted on the A filter, and the mixture was washed 4 times with 0.425% phosphoric acid solution and 1 time with methanol for 4 minutes. After drying, the measurement was performed by a scintillation counter.
TABLE 6 results of Axl (h), trkA (h), trkB and TrkC kinase experiments for the compounds provided by the invention
Figure BDA0002404519330000291
Figure BDA0002404519330000292
Figure BDA0002404519330000301
/>
As can be seen from Table 6, the compounds of the present invention showed good AXL (h), trkA (h), trkB and TrkC kinase inhibitory activities in the kinase assay.
EXAMPLE D cell Activity assay
This was accomplished by examining the inhibition of the growth of both Ba/F3 AXL cell line and Ba/F3 parental cell line strains by both compounds. Harvesting cells in the logarithmic growth phase, counting the cells by adopting a platelet counter, and detecting the cell activity by using a trypan blue exclusion method to ensure that the cell activity is more than 90%; regulating the cell concentration; add 90. Mu.L of cell suspension to 96-well plates, respectively; cells in 96-well plates were placed at 37℃in 5% CO 2 Incubated overnight at 95% humidity. Preparing 10 times of medicinal solution with maximum concentration of 100 μm and 9 concentratesDegree, 3.16 times dilution, adding 10 μl of drug solution into each well of 96-well plate inoculated with cells, and setting three compound wells for each drug concentration; cells in the dosed 96-well plates were placed at 37℃with 5% CO 2 The culture was continued for 72 hours at 95% humidity, after which CTG analysis was performed. The CTG reagent was thawed and the cell plates were equilibrated to room temperature for 30 minutes, an equal volume of CTG solution was added to each well, the cells were lysed by shaking on an orbital shaker for 5 minutes, and the cell plates were left at room temperature for 20 minutes to stabilize the luminescence signal and read for luminescence values. Analysis of data using GraphPad Prism 5.0 software, fitting data to derive dose-response curves using nonlinear S-curve regression, and calculating IC therefrom 50 Value, cell viability (%) = (Lum) Drug to be tested -Lum Culture broth control )/(Lum Cell control -Lum Culture broth control )×100%。
TABLE 7 inhibition of growth of two Ba/F3 strains by the compounds provided by the invention
Figure BDA0002404519330000302
Example E cytochrome P450 enzyme Induction experiment
Cytochrome P450 induction assays can identify the potential of test compounds to induce CYP1A2, CYP2B6 or CYP3A4 in human hepatocytes.
Experimental System
Inducer and substrate:
suitable CYP enzyme positive control (inducer), negative control (flumazenil in general) and probe substrate (for enzyme activity assay only, no probe substrate is needed if mRNA analysis is only performed) are selected. Common CYP enzyme tools are shown in table 8 below, and other types of tools can be selected according to the purpose of the reference.
Table 8 common CYP enzyme tool medicine
Figure BDA0002404519330000303
Hepatocyte donor:
the inductivity was evaluated using hepatocytes of three donors.
Cytotoxicity test:
in the induction experiment, the influence of the tested reagent on the activity of liver cells is detected in parallel, and CCK-8, LDH, MTT, neutral red and the like can be adopted.
Experimental procedure
Hepatocyte resuscitation/passaging, plating:
resuscitates the frozen liver cells or passages the liver cells reaching the requirements, and inoculates the frozen liver cells on a proper cell culture plate according to a certain density. The method is operated according to the culture requirement of a required culture model (such as monolayer cells, sandwich culture, 3D culture and the like) so as to meet the requirement of cell growth.
Inducer incubation and adding a dosing solution containing a positive/negative control drug or a test drug freshly prepared with an incubation solution and preheated to the corresponding wells, incubating for 2-3 days continuously and changing the solution every day, and operating the multiple wells in parallel for each compound. The actual concentration of the test agent may be sampled and analyzed at various time points of the last day of incubation of the test agent, if desired.
Substrate incubation and CCK-8 incubation:
after the last administration, the prepared and preheated substrate working solution is added into the culture plate for incubation for a certain time. The cytotoxic assay plate was incubated with CCK-8 solution for a period of time.
Cytotoxicity and enzyme activity assays:
the measurement of absorbance was performed using a microplate reader or a spectrophotometer to calculate cytotoxicity. The substrate working solution was incubated and analyzed after sample treatment to calculate the enzyme activity.
Extraction of mRNA, reverse transcription and fluorescent quantitative PCR:
the above experimental procedure was performed according to the instructions of the relevant kit.
Data processing
The enzyme induction experiment calculates the induction times of mRNA level, enzyme activity and the induction activity of each relative positive control medicine; the ratio of cell viability to the blank after administration of each compound was calculated for the cytotoxicity test.
Fold induction calculation of mRNA levels:
subtracting the Ct value of the internal reference (such as 18 s) from the Ct value of the target gene to obtain the delta Ct value of the target gene, and calculating the relative expression difference (induction multiple) of mRNA (messenger ribonucleic acid), namely, the following formula is used for judging the induction condition of the mRNA expression level.
Delta ct=ct-target gene-Ct 18s
ΔΔct = Δct-control/sample- Δct-vehicle control relative expression difference = 2- ΔΔct.
Calculation of relative enzyme activity:
the specific metabolites of each CYP-subunit probe substrate were detected using established assays, and enzyme activity induction was calculated by comparing the differences in metabolites of the test group and the vehicle control group. The relative positive control activity was calculated according to the following formula:
relative positive control activity (%) = (test group sample activity-vehicle group sample activity)/(positive control group sample activity-vehicle group sample activity) ×100.
Evaluation criteria:
the experimental system is considered normal by the factor of 2 times or more of the induction of the enzyme activity of the positive control drug and the factor of 4 times or more of the induction of the mRNA level, and the standard of the experimental system can be properly reduced when the induction capacity of individual sub-enzymes of some cells is weaker. The subject group was considered to be hepatotoxic when cell viability was < 70%. The test agent was considered to have an induction risk at a fold induction of ≡2 fold with respect to the positive control agent ≡20% of the mRNA level. Meanwhile, in order to reduce false negatives, if either of the mRNA level induction multiple of ∈4×and the enzyme activity induction multiple of ∈2×and the enzyme activity induction relative to the positive control drug ∈20% is satisfied, it is considered that there is an induction risk.
The results of the enzyme induction experiments at 10 μm concentration for some of the compounds of the present invention are shown in table 9:
TABLE 9 results of enzyme-induced experiments with the compounds of the invention
Figure BDA0002404519330000311
Finally, it should be noted that there are other ways to implement the invention. Accordingly, the embodiments of the present invention are to be construed as illustrative, not restrictive of the invention, but may be modified and equivalents added to the scope of the invention as defined by the appended claims. All publications or patents cited herein are incorporated by reference.

Claims (7)

1. A compound having a structure represented by formula (I) or formula (II):
Figure QLYQS_1
(I),
Figure QLYQS_2
(II),
or a tautomer, or pharmaceutically acceptable salt thereof;
wherein, the liquid crystal display device comprises a liquid crystal display device,
U 1 is-CH-;
U 2 is N or-CH-;
R 1 is phenyl; the phenyl groups are independently optionally substituted with 0 or 1R 11 Substitution;
R 11 f, cl or Br;
R 2 is C 1-6 Alkyl, C 1-6 Hydroxyalkyl, or C 2-7 Heterocyclyl C 1-6 Alkyl, wherein said C 2-7 The heterocyclic group is tetrahydrofuranyl;
R 4 is C 1-6 Alkyl, C 3-10 Cycloalkyl, C 3-10 Cycloalkyl C 1-6 An alkyl group;
R 3 is C 1-6 An alkyl group;
R 5 is-NH 2
Each R is 6 And R is 7 Each independently is H or D;
R 8 f, cl or Br;
n is 0 or 1;
X 1 is- (CH) 2 ) t1 -、-X 2 -(CH 2 ) t1 -; the- (CH) 2 ) t1 -wherein t1 is 1 or 2, said-X 2 -(CH 2 ) t1 -wherein t1 is 1;
X 2 Is O or S;
m is 0.
2. The compound according to claim 1, wherein,
R 4 is methyl, ethyl, propyl, butyl, C 3-6 Cycloalkyl or C 3-6 Cycloalkyl C 1-4 An alkyl group.
3. The compound of claim 1, which is a compound having one of the following structures:
Figure QLYQS_5
(1),/>
Figure QLYQS_4
(2),/>
Figure QLYQS_17
(3),/>
Figure QLYQS_9
(4),/>
Figure QLYQS_22
(5),/>
Figure QLYQS_11
(6),/>
Figure QLYQS_15
(7),/>
Figure QLYQS_10
(8),/>
Figure QLYQS_21
(9),/>
Figure QLYQS_3
(10),/>
Figure QLYQS_16
(11),/>
Figure QLYQS_7
(12),/>
Figure QLYQS_19
(13),/>
Figure QLYQS_12
(14),/>
Figure QLYQS_14
(15),/>
Figure QLYQS_13
(16),/>
Figure QLYQS_18
(17),/>
Figure QLYQS_8
(18),/>
Figure QLYQS_20
(19) Or->
Figure QLYQS_6
(20),
Or a tautomer, or a pharmaceutically acceptable salt thereof.
4. A pharmaceutical composition comprising a compound of any one of claims 1-3, or a tautomer thereof, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable adjuvant, diluent, or carrier, or combination of same.
5. Use of a compound according to any one of claims 1 to 3 or a pharmaceutical composition according to claim 4 in the manufacture of a medicament for the prevention or treatment of one or more Axl and/or Trk protein kinase mediated diseases and/or conditions selected from a proliferative disease, an autoimmune disease or an inflammatory disease.
6. The use according to claim 5, wherein the disease and/or condition is selected from cancer, polycythemia vera, essential thrombocythemia, myelofibrosis, chronic obstructive pulmonary disease, asthma, systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, sjogren's syndrome, psoriasis, eczema, measles, inflammatory bowel disease, crohn's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis.
7. The use according to claim 5, wherein the disease and/or condition is selected from acute myelogenous leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia.
CN202010157262.4A 2019-03-13 2020-03-09 Substituted heteroaryl compounds, compositions and uses thereof Active CN111689991B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2020/078990 WO2020182188A1 (en) 2019-03-13 2020-03-12 Substituted heteroaryl compound, composition of same, and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019101867577 2019-03-13
CN201910186757 2019-03-13

Publications (2)

Publication Number Publication Date
CN111689991A CN111689991A (en) 2020-09-22
CN111689991B true CN111689991B (en) 2023-06-09

Family

ID=72476265

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010157262.4A Active CN111689991B (en) 2019-03-13 2020-03-09 Substituted heteroaryl compounds, compositions and uses thereof

Country Status (1)

Country Link
CN (1) CN111689991B (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021233236A1 (en) * 2020-05-18 2021-11-25 北京范恩柯尔生物科技有限公司 Substituted heteroaryl compound, and composition and use thereof
CN113912628B (en) * 2020-07-10 2023-05-02 北京范恩柯尔生物科技有限公司 Triazine compound, composition and application thereof
CN116583287A (en) * 2021-02-05 2023-08-11 四川科伦博泰生物医药股份有限公司 Pyridazinone heterocyclic compound, preparation method and application thereof
WO2023016296A1 (en) * 2021-08-12 2023-02-16 四川科伦博泰生物医药股份有限公司 Heterocyclic compound and preparation method therefor and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108250200A (en) * 2016-12-28 2018-07-06 中国科学院上海药物研究所 A kind of compound and its preparation and application with Axl inhibitory activity
CN109348715A (en) * 2016-03-28 2019-02-15 因赛特公司 Pyrrolotriazine compounds as TAM inhibitor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109348715A (en) * 2016-03-28 2019-02-15 因赛特公司 Pyrrolotriazine compounds as TAM inhibitor
CN108250200A (en) * 2016-12-28 2018-07-06 中国科学院上海药物研究所 A kind of compound and its preparation and application with Axl inhibitory activity

Also Published As

Publication number Publication date
CN111689991A (en) 2020-09-22

Similar Documents

Publication Publication Date Title
US10934302B1 (en) SHP2 phosphatase inhibitors and methods of use thereof
CN111689991B (en) Substituted heteroaryl compounds, compositions and uses thereof
TWI753892B (en) Pyrrolotriazine compounds as tam inhibitors
AU2009210517B2 (en) Pyrido [4, 3-D] pyrimidinone derivatives as kinase inhibitors
KR101083177B1 (en) Triazolo-pyridazine protein kinase modulators
KR101871436B1 (en) Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
CN106928219B (en) Nitrogen-containing fused heterocyclic compound, preparation method, intermediate, composition and application
JP2018150358A (en) Tank-binding kinase inhibitor compounds
TW202128685A (en) Bicyclic heterocycles as fgfr inhibitors
JP2019516759A (en) Pyrazolopyridine derivatives for the treatment of cancer
JP2023513580A (en) Novel PRMT5 inhibitor
WO2016086200A1 (en) 4,5,6,7-tetrahydro-1 h-pyrazolo[4,3-c]pyridin-3-amine compounds as cbp and/or ep300 inhibitors
EP3818063A1 (en) Fused pyrazine derivatives as a2a / a2b inhibitors
JP2021533179A (en) Pyrazine compounds and their use
CN113912628B (en) Triazine compound, composition and application thereof
JP7399968B2 (en) Pyrazolo[4,3-D]pyrimidine compounds as ALK2 and/or FGFR modulators
EP3464267B1 (en) New phenylpyrazolylacetamide compounds and derivatives as cdk8/cdk19 inhibitors
WO2020182188A1 (en) Substituted heteroaryl compound, composition of same, and uses thereof
JP2021503479A (en) Substituted heteroaryl compound and usage
CN113683629B (en) Substituted heteroaryl compounds, compositions and uses thereof
JP2023534837A (en) Indole compounds as androgen receptor modulators
CN115850301A (en) Pyrazolone compound and composition and application thereof
CN116507627A (en) Substituted 1H-pyrazolo [4,3-C ] pyridines and derivatives as EGFR inhibitors
EP3691642A1 (en) New [1,6]naphthyridine compounds and derivatives as cdk8/cdk19 inhibitors

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
GR01 Patent grant
GR01 Patent grant