CN115260202A - Cyclic compounds as JAK2 inhibitors and uses thereof - Google Patents

Cyclic compounds as JAK2 inhibitors and uses thereof Download PDF

Info

Publication number
CN115260202A
CN115260202A CN202110476658.XA CN202110476658A CN115260202A CN 115260202 A CN115260202 A CN 115260202A CN 202110476658 A CN202110476658 A CN 202110476658A CN 115260202 A CN115260202 A CN 115260202A
Authority
CN
China
Prior art keywords
cancer
group
compound
pharmaceutically acceptable
alkyl
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.)
Pending
Application number
CN202110476658.XA
Other languages
Chinese (zh)
Inventor
李洪林
赵振江
刁妍妍
朱丽丽
刘丹丹
葛欢
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.)
East China University of Science and Technology
Original Assignee
East China University of Science and Technology
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 East China University of Science and Technology filed Critical East China University of Science and Technology
Priority to CN202110476658.XA priority Critical patent/CN115260202A/en
Publication of CN115260202A publication Critical patent/CN115260202A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • 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
    • A61P19/00Drugs for skeletal disorders
    • 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
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Abstract

The invention relates to a cyclic compound serving as a JAK2 inhibitor and application thereof. In particular, the invention relates to a compound shown in the formula I and application of the compound in treating JAK2 mediated related diseases and preparing drugs for treating JAK2 mediated related diseases.

Description

Cyclic compounds as JAK2 inhibitors and uses thereof
Technical Field
The present invention relates to the field of medicinal chemistry; in particular, the present invention relates to novel JAK2 inhibitors and uses thereof.
Background
Janus kinase (JAK) is an unresponsive tyrosine protein kinase, a relatively large protein of the tyrosine kinase family (120-130 kDa). The JAK family has four members: JAK1, JAK2, TYK2 and JAK3. The first three are widely expressed in vivo, whereas JAK3 is expressed only in myeloid and lymphoid cells. All members of the JAK family contain 7 homologous domains of different lengths, with the tyrosine kinase domain JH1 and the pseudokinase domain JH2 located in the carboxy-terminal portion. The pseudokinase domain JH2 modulates kinase domain activity and produces inhibition. Domains JH3-JH4 are similar in structure to SH2 domains. The JH4-JH7 domain is located at the amino terminus of the kinase, which is important for binding of JAKs kinase to cytokine receptors on the cell surface and for maintaining receptor expression.
JAK kinases play a pivotal role in the signaling of many cytokine receptors, primarily through the JAK-STAT signaling pathway to regulate the signaling process. JAK-STAT refers to the JAK kinase-cell signal transduction and transcriptional activator pathway, which operates downstream of over 50 cytokines and growth factors, and is considered an important communication node of the immune system. The JAK-STAS signaling process generally involves, first, the binding of cytokines to specific receptors on the cell membrane, inducing receptor-JAK complex dimerization, and the activation of JAK kinases. Activated JAKs then phosphorylate tyrosine residues in the cytoplasmic domain of cytokine receptors to provide docking sites for STATs. Specific STATs bind to cytokine receptors through their SH2 domain and are phosphorylated on tyrosine residues by JAKs, resulting in the formation of homo-or heterodimers via SH 2-phosphate interactions. Dimeric STATs are then transferred from the cytoplasm to the nucleus, where they regulate gene expression by binding to specific DNA.
JAKs play a key role in both innate and adaptive immunity as well as hematopoiesis, making them attractive targets for many therapeutic indications. Therefore, since its discovery in the early 1990 s, JAKs have been attracting attention as targets of new drugs. Currently, there are a variety of chemical entities in the clinic, covering a variety of indications, including myeloproliferative diseases and a variety of inflammatory diseases.
JAK2 signaling plays a key role in the pathogenesis of myeloproliferative diseases, with a point mutation in the JAK2 pseudokinase domain V617F being found in about 90% of patients with polycythemia vera, and in about 50% of patients with primary myelofibrosis and primary thrombocythemia. This mutation results in continuous cytokine-independent activation of JAK2 and activates downstream signaling pathways. This mutation was also found in the pathogenesis of Ph negative chronic myelogenous leukemia, chronic myelogenous monocytic leukemia, megakaryocytic acute myelogenous leukemia, and juvenile myelogenous monocytic leukemia (10-20%). Other mutations in the JAK2 pseudokinase domain, including the point mutation of Arg683, have been detected in approximately 20% of down syndrome-associated acute lymphocytic and acute myelogenous leukemias. Therefore, JAK2 has become a hot target in the field, and JAK2 inhibitors have good application prospects in inflammation, autoimmune diseases, myelofibrosis and various hematologic malignancies.
The JAK small-molecule inhibitors which are marketed up to now are mostly pan-inhibitors, and lack of sufficient selectivity for JAK family members, which causes possible adverse reactions in the clinical application process. For example, JAK2 inhibitors, while inhibiting JAK3, can cause immunodeficiency. Therefore, research and development of a novel structure with high selectivity on JAK2 have important clinical significance and application value.
Disclosure of Invention
The invention aims to provide a novel large-ring JAK inhibitor which has a selective inhibition effect on JAK2, so that the novel large-ring JAK inhibitor has clinical significance and application prospects.
In a first aspect of the present invention, there is provided a compound represented by the following formula I, or a stereoisomer or an optical isomer, a pharmaceutically acceptable salt, a prodrug or a solvate thereof,
Figure BDA0003047622440000021
in the formula (I), the compound is shown in the specification,
x is selected from N and CH;
R1selected from the group consisting of: hydrogen, halogen, C1-C10Alkyl, hydroxy, C1-C10Alkoxy, nitro, amino, acetylamino, C1-C3Alkyl formyl, hydroxy acetyl, C1-C3An alkoxyformyl group; n is selected from 0,1,2 and 3;
R2selected from the group consisting of: hydrogen, halogen, C1-C10Alkyl radical, C2-C10An acyl group;
R3selected from the group consisting of: hydrogen, halogen, hydroxy, amino, C1-C10Alkyl radical, C1-C10Alkoxy, or substituted by one or more C1-C10Alkyl or C1-C10Alkoxy-substituted 4-to 6-membered nitrogen-or oxygen-containing heterocycles;
R4selected from the group consisting of: hydrogen, substituted or unsubstituted C1-C10Alkyl, substituted or unsubstituted C1-C10Alkyl- (4-6 membered nitrogen or oxygen containing heterocycle);
X1is- (L)m-, wherein said L is selected from the group consisting of: -CHR-, -C (O) -, -NR-, -O-, -CH = CH-,
Figure BDA0003047622440000031
r is H or C1-C4An alkyl group; m is 2,3, 4, 5, 6, 7, 8, 9, 10 or 11;
unless otherwise specified, the substitution refers to the substitution of one or more hydrogen atoms on the group with a substituent selected from the group consisting of: halogen, C1-C10An alkyl group.
In another preferred embodiment, X is1Is a chain structure selected from the group consisting of:
Figure BDA0003047622440000032
wherein R is5Is C1-C3An alkyl group;
l, m, o and r are each independently selected from 0,1 or 2;
p and q are each independently selected from 0,1,2,3;
in another preferred embodiment, the compound of formula I has the structure shown in formula III below:
Figure BDA0003047622440000033
in the formula
R2、R3、R4、X1Is defined as in claim 1.
In another preferred embodiment, the compound of formula I has the structure shown in formula III below:
Figure BDA0003047622440000034
wherein, X1Is selected from
Figure BDA0003047622440000035
R4Selected from the group consisting of: hydrogen, methyl, or a structural unit selected from the group consisting of:
Figure BDA0003047622440000041
Figure BDA0003047622440000042
wherein x is 1,2 or 3.
In another preferred embodiment, R is5Selected from the group consisting of: hydrogen, methyl, ethyl, C1-C10Alkyl-substituted 4-6 membered nitrogen or oxygen containing heterocycles.
In another preferred embodiment, the compound of formula I has the structure shown in formula IV below:
Figure BDA0003047622440000043
R4selected from:
Figure BDA0003047622440000044
X1selected from the group consisting of:
Figure BDA0003047622440000045
in another preferred embodiment, the compound is selected from the group consisting of:
Figure BDA0003047622440000051
in a second aspect of the present invention, there is provided a pharmaceutical composition comprising a compound according to the first aspect of the present invention, or a stereoisomer or an optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, and a pharmaceutically acceptable carrier or excipient.
In a third aspect of the present invention, there is provided a use of the compound of the first aspect of the present invention, or a stereoisomer or an optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, for the preparation of a medicament for the prevention or treatment of a JAK 2-mediated disease; and/or for the preparation of a JAK2 inhibitor.
In another preferred embodiment, the JAK 2-mediated disease is myelodysplastic syndrome (MDS), eosinophilia, a tumor, an inflammatory disease, or an infection caused by a bacterium, virus, or fungus;
preferably, the tumor is selected from the group consisting of: myeloproliferative carcinoma (MPN), melanoma, lung cancer, kidney cancer, ovarian cancer, prostate cancer, breast cancer, colon cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, uterine cancer, rectal cancer, anal cancer, stomach cancer, testicular cancer, carcinoma of the fallopian tubes, endometrial cancer, cervical cancer, vaginal cancer, vulval cancer, hodgkin's disease, non-hodgkin's lymphoma, esophageal cancer, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, pediatric solid tumors, lymphocytic lymphoma, cancer of the bladder, cancer of the kidney or ureter, carcinoma of the renal pelvis, central Nervous System (CNS) tumors, primary CNS lymphoma, tumor angiogenesis, spinal axis tumors, brain stem glioma, pituitary adenoma, kaposi's sarcoma, epidermoid carcinoma, squamous cell carcinoma, T-cell lymphoma; and/or
The inflammatory disease is selected from the group consisting of: rheumatoid arthritis, ankylosing spondylitis, autoimmune hemolytic anemia, arthritis, myasthenia gravis, systemic lupus erythematosus, pernicious anemia, polymyositis; and/or
The virus is selected from the group consisting of: hepatitis viruses (type a, type b and type c), herpes viruses, influenza viruses, adenoviruses, coronaviruses, measles viruses, dengue viruses, polio viruses, rabies viruses; and/or
The bacteria are selected from the group consisting of: chlamydia, rickettsia, mycobacteria, staphylococci, pneumococci, cholera, tetanus; and/or
The fungus is selected from the group consisting of: candida, aspergillus, dermatitides.
In a fourth aspect of the present invention, there is provided a JAK2 inhibitor comprising a compound according to the first aspect of the present invention or a stereoisomer or an optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, or a pharmaceutical composition according to the second aspect of the present invention.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be repeated herein, depending on the space.
Drawings
FIG. 1 is a dendogram of the kinase spectrum of compound L-4;
FIG. 2 is a kinase selectivity spectrum of compound L-4;
FIG. 3 is a graph of the effect of L-4 on ulcerative colitis in mice; (a) a graph of the change in body weight of the mice; (B) DAI change profile in mice; (C) the effect of L13 on the ratio of colon mass to length.
Detailed Description
The inventor finds the potential application of the aminopyrimidine macrocyclic structure as a JAK inhibitor through extensive and intensive research, finally finds a series of novel macrocyclic compounds capable of selectively inhibiting JAK2 kinase, and provides a brand new material basis for the development of immune inflammation and antitumor drugs. On this basis, the inventors have completed the present invention.
Definition of terms
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosed invention belongs. For the purpose of understanding the present invention, the following definitions will be made for terms related to the present invention, but the scope of the present invention is not limited to these specific definitions.
As used herein, "JAK2" refers to Janus kinase 2, an intracytoplasmic non-receptor type soluble protein tyrosine kinase. JAK-STAT is a Janus kinase-cell signal transduction and transcriptional activator pathway, and is a hot spot in the current cytokine research field.
Herein, "alkyl" refers to a straight or branched chain saturated group consisting of carbon atoms and hydrogen atoms. For example, "C1-C10Alkyl "refers to a saturated branched or straight chain alkyl group having a carbon chain length of 1 to 10 carbon atoms, preferably 1 to 6 carbon atoms, more preferably 1 to 3 carbon atoms. Examples of alkyl groups include, but are not limited to: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, heptyl, pentyl, and the like.
Herein, "alkoxy" refers to an oxy group substituted with an alkyl group. In a particular embodiment, alkoxy as used herein is an alkoxy group of 1 to 10 carbon atoms in length, more preferably 1 to 4 carbon atoms in length. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, and the like. In further embodiments, the alkoxy group may be a substituted alkoxy group, for example, a halogen substituted alkoxy group. In particular embodiments, halogen substituted C is preferred1-C3An alkoxy group.
Herein, "halogen" refers to fluorine, chlorine, bromine and iodine. In a preferred embodiment, halogen is fluorine or chlorine.
As used herein, "halo" refers to fluoro, chloro, bromo, and iodo.
Herein, "substituted or unsubstituted" or "optionally substituted" means that the substituent modified by the term may be optionally substituted with 1 to 5 (e.g., 1,2,3, 4, or 5) substituents selected from: halogen, C1-4 aldehyde group, C1-6 straight or branched alkyl group, halogen-substituted C1-6 straight or branched alkyl group (e.g., trifluoromethyl), C1-6 alkoxy group, halogen-substituted C1-6 alkoxy group (e.g., trifluoromethoxy), cyano, nitro, amino, hydroxyl, hydroxymethyl, carboxyl, ethoxyformyl, N (CH 3), and C1-4 acyl group.
Active ingredient
As used herein, "compound of the invention" refers to a compound of formula (I), and also includes various crystalline forms, pharmaceutically acceptable salts, hydrates or solvates of the compound of formula (I),
Figure BDA0003047622440000071
in the formula I, R1、R2、R3、R4N and X1As described above.
Based on the teachings of the present invention and the general knowledge in the art, one skilled in the art will appreciate that various groups in the compounds of the present invention can be further substituted to provide derivatives that have the same or similar activity as the specifically disclosed compounds of the present invention. Each group in the compounds of the present invention may be substituted with various substituents which are conventional in the art, as long as such substitution does not violate the rules of chemical synthesis or the rules of valency.
The term "substituted" as used herein means that one or more hydrogen atoms on a particular group are replaced with a particular substituent. The specific substituents may be those described above, or may be those specified in the examples or those conventional in the art. Therefore, in the present invention, the substituents in the general formula may also each independently be the corresponding group in the specific compounds in the examples; that is, the present invention includes both combinations of the respective substituents in the above general formulae and combinations of partial substituents shown in the general formulae with other specific substituents appearing in the examples. Preparing compounds having such combinations of substituents and testing the resulting compounds for activity is readily accomplished by those skilled in the art based on routine skill in the art.
The term "pharmaceutically acceptable salt" as used herein refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, etc., organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, phenylmethanesulfonic acid, benzenesulfonic acid, etc.; and acidic amino acids such as aspartic acid and glutamic acid.
Unless otherwise specified, the structural formulae depicted herein are intended to include all isomeric forms (e.g., enantiomers, diastereomers and geometric isomers (or conformational isomers)): for example, the R and S configuration containing asymmetric center, the (Z) and (E) isomers of double bond, etc. Thus, individual stereochemical isomers of the compounds of the present invention or mixtures of enantiomers, diastereomers or geometric isomers (or conformers) thereof are within the scope of the present invention.
As used herein, the term "tautomer" means that structural isomers having different energies may exceed the low energy barrier and thus be converted to each other. For example, proton tautomers (i.e., proton transmutations) include interconversion by proton shift, such as 1H-indazoles and 2H-indazoles. Valence tautomers include interconversion by recombination of some of the bonding electrons.
As used herein, the term "solvate" refers to a compound of the present invention coordinated to solvent molecules to form a complex in a specified ratio.
As used herein, the term "hydrate" refers to a complex formed by the coordination of a compound of the present invention with water.
Pharmaceutical compositions and methods of administration
Since the compound of the present invention has an excellent inhibitory activity against JAK kinases, the compound of the present invention and various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates thereof, and a pharmaceutical composition containing the compound of the present invention as a main active ingredient can be used for the prevention and/or treatment (stabilization, alleviation or cure) of JAK kinase-associated diseases.
The pharmaceutical compositions of the present invention comprise a safe and effective amount of a compound of the present invention in combination with a pharmaceutically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 10-200mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g. stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g. soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g. propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (e.g. propylene glycol, glycerol, mannitol, sorbitol, etc.)
Figure BDA0003047622440000091
) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, parenteral (intravenous, intramuscular or subcutaneous).
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) Fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethyl cellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia; (c) humectants, for example, glycerol; (d) Disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary amine compounds; (g) Wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, especially cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
When administered in combination, the pharmaceutical composition further comprises one or more (2, 3, 4, or more) other pharmaceutically acceptable compounds. One or more of the other pharmaceutically acceptable compounds may be administered simultaneously, separately or sequentially with a compound of the invention.
When using pharmaceutical compositions, a safe and effective amount of a compound of the present invention is administered to a mammal (e.g., a human) in need of treatment, wherein the administration is a pharmaceutically acceptable and effective dose, and the daily dose for a human of 60kg body weight is usually 1 to 2000mg, preferably 20 to 500mg. Of course, the particular dosage will also take into account such factors as the route of administration, the health of the patient, and the like, which are within the skill of the skilled practitioner.
Preparation method
The compounds of the present invention may be prepared according to conventional routes or methods, or may be obtained according to the methods or routes described herein. Such as route 1.
Scheme 1:
Figure BDA0003047622440000111
synthetic route to the 14-membered ringa
The synthetic route for the 14-membered macrocycles is as above.
aReagents and conditions: (a) Cs2CO3,Xantphos,Pd2(dba)3,dioxane,reflux,5h,45%;(b)NaOH,H2O,MeOH,40℃,1h,78%;(c)Cs2CO3,DMF,100℃,overnight,50.6%;(d)NaBH3CN,MeOH,r.t.,12h,70%;(e)HATU,DIPEA,DMF,r.t.,1h,54%;(f)10%Pd/C,H2,MeOH,40℃,1h,46.7%;(g)Cs2CO3,Xantphos,Pd2(dba)3,DMF,140℃,6h,56%.
Scheme 2:
Figure BDA0003047622440000121
synthetic route to 19-membered rings
The synthetic route for the 14-membered ring macrocycles is as above.
aReagents and conditions: (a) NaOH, bu4NHSO4,H2O,CH2Cl2,r.t.,3h,40%;(b)NaH,DMF,0℃,1h,53%;(c)Cs2CO3,Xantphos,Pd2(dba)3,DMF,80℃,10h,60%;(d)SnCl2,CH2Cl2,MeOH,r.t.,18h,41.7%;(e)TsOH·H2O,n-butanol,105℃,2h,74.4%.
The main advantages of the invention are:
1. the compound has a novel structure and an excellent JAK kinase inhibitor effect;
2. the compounds of the invention are more selective for JAK2 inhibition.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are by weight.
The test materials and reagents used in the following examples are commercially available without specific reference.
Herein, "reflux" means reflux.
EXAMPLE 1 preparation of Compound L-1
1) 1) Synthesis of methyl 3- ((2-chloro-5-methylpyrimidin-4-yl) amino) benzoate
Figure BDA0003047622440000131
2-chloro-4-amino-5-methylpyrimidine (500.00mg, 3.4826mmol) was added to the reaction flask followed by methyl 3-bromobenzoate (898.73mg, 4.1792mmol), cs2CO3(3.40g,10.4352mmol)、Xantphos(403.00mg,0.6965mmol)、Pd2(dba)3(319.00mg,0.3484mmol),N2After the displacement, an anhydrous 1, 4-dioxane solution was added and heated under reflux for about 5 hours. After completion of the reaction, filtration and extraction with dichloromethane were carried out, and the product was separated by silica gel column chromatography (PE: EA = 5.
1H-NMR(400MHz,DMSO)δ9.05(s,1H),8.26(s,1H),8.09(s,1H),8.03(d,J=8.1,1.2Hz,1H),7.69(d,J=7.8Hz,1H),7.52(t,J=7.9Hz,1H),3.87(s,3H),2.19(s,3H).LC-MS(m/z):278.10/280.10(M+H)+.
2) Synthesis of 3- ((2-chloro-5-methylpyrimidin-4-yl) aminobenzoic acid
Figure BDA0003047622440000132
Methyl 3- ((2-chloro-5-methylpyrimidin-4-yl) amino) benzoate (100mg, 0.3600mmol) was dissolved in methanol (3 mL), and an aqueous solution (1 mL) of NaOH (345.7mg, 8.6425mmol) was added, and the mixture was stirred at 40 ℃ for about 40min. After the reaction was complete, water was added for dilution, methanol was dried by spinning, and 1M HCl was added to adjust the pH to 2. Extraction with ethyl acetate, combination of the organic phases and spin-drying gave 74.06mg (78%) of 3- ((2-chloro-5-methylpyrimidin-4-yl) amino) benzoic acid as a brown solid.
1H-NMR(400MHz,CD3OD)δ8.24(s,1H),7.99–7.92(m,2H),7.80(d,J=7.7Hz,1H),7.46(t,J=7.9Hz,1H),2.21(s,3H).LC-MS(m/z):264.10/266.10(M+H)+.
3) Synthesis of 5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) benzaldehyde
Figure BDA0003047622440000141
2-hydroxy-5-nitrobenzaldehyde (500.00mg, 2.9919mmol), chloroethylpyrrolidine hydrochloride (712.40mg, 4.1886mmol) and Cs2CO3(1.462g, 4.4879mmol) was added to the reaction flask, then anhydrous DMF (8 mL) was added thereto and dissolved, and the mixture was stirred overnight with warming to 100 ℃. And monitoring the reaction progress by TLC, cooling after the reaction is finished, pouring the reaction liquid into ice water, extracting by dichloromethane, washing by brine, combining organic phases and concentrating. The product was isolated by silica gel column chromatography (DCM: meOH = 30) to give 400mg (50.6%) of 5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) benzaldehyde as a yellow sticky solid.
1H-NMR(400MHz,DMSO)δ10.34(s,1H),8.49(dd,J=9.2,2.9Hz,1H),8.41(d,J=2.9Hz,1H),7.50(d,J=9.2Hz,1H),4.41(t,J=5.4Hz,2H),2.95(t,J=5.2Hz,2H),2.60(s,4H),1.70(s,4H).LC-MS(m/z):265.20(M+H)+.
4) Synthesis of N-methyl-1- (5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) phenyl) methylamine
Figure BDA0003047622440000142
5-Nitro-2- (2- (pyrrolidin-1-yl) ethoxy) benzaldehyde (316.00mg, 1.1970mmol) was added to a reaction flask, and methanol (5 mL) was added to dissolve, followed by the sequential addition of methylamine hydrochloride (162.00mg, 2.3993mmol), and NaBH3CN(90.26mg,1.4363mmol),N2And (4) replacing, and reacting at room temperature for about 12h. After completion of the reaction, extracted with ethyl acetate and washed with brine. The organic phases were combined and concentrated. The product was isolated by silica gel column chromatography (DCM: meOH = 10) to give N-methyl-1- (5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) phenyl) methylamine 234.05mg (70%) as a yellow viscous solid.
1H-NMR(400MHz,DMSO)δ8.18(s,1H),8.11(d,J=8.9Hz,1H),7.18(d,J=9.0Hz,1H),4.21(t,J=5.5Hz,2H),3.63(s,2H),2.81(t,J=5.5Hz,2H),2.50–2.41(m,4H),2.25(s,3H),1.69–1.62(m,4H).LC-MS(m/z):280.20(M+H)+.
5) Synthesis of 3- ((2-chloro-5-methylpyrimidin-4-yl) amino) N-methyl-N- (5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) benzyl) benzamide
Figure BDA0003047622440000151
3- ((2-chloro-5-methylpyrimidin-4-yl) amino) benzoic acid (34.00mg, 0.1289mmol), N-methyl-1- (5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) phenyl) methylamine (43.17mg, 0.1547mmol) and DIPEA (166.60mg, 1.2891mmol) were dissolved in DMF and HATU (68.60mg, 0.1804mmol) was added and allowed to react at room temperature for about 1h. After the reaction was complete, ethyl acetate was extracted, followed by saturated NaHCO3Aqueous solution, brine washing. The organic phases were combined and concentrated. The product was isolated by silica gel column chromatography (DCM: meOH = 30) to give 36.55mg (54%) of 3- ((2-chloro-5-methylpyrimidin-4-yl) amino) N-methyl-N- (5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) benzyl) carboxamide as a yellow solid.
1H-NMR(400MHz,CD3OD)δ7.88(s,1H),7.82(s,1H),7.76–7.67(m,2H),7.48(d,J=7.9Hz,1H),7.40(t,J=7.8Hz,1H),7.28(d,J=7.4Hz,1H),7.24(d,J=7.7Hz,1H),6.84(d,J=8.5Hz,1H),4.62(s,2H),3.97(t,J=5.5Hz,2H),3.03(s,3H),2.74(t,J=5.5Hz,2H),2.54–2.46(m,4H),2.03(s,3H),1.76–1.69(m,4H).LC-MS(m/z):525.20/527.20(M+H)+.
6) Synthesis of 3- ((2-chloro-5-methylpyrimidin-4-yl) amino) N-methyl-N- (5-amino-2- (2- (pyrrolidin-1-yl) ethoxy) benzyl) benzamide
Figure BDA0003047622440000152
3- ((2-chloro-5-methylpyrimidin-4-yl) amino) N-methyl-N- (5-nitro-2- (2- (pyrrolidin-1-yl) ethoxy) benzyl) benzamide (68.00mg, 0.1295 mmol) was dissolved in methanol (2 mL) and 10% Pd/C (13.60mg, 20%), H2After the displacement, the reaction was carried out at 40 ℃ for about 30min. The reaction is completedAfter that, celite was filtered, spin-dried, and the product was isolated by silica gel column chromatography (DCM: meOH = 30.
1H-NMR(400MHz,CD3OD)δ7.88(s,1H),7.82(s,1H),7.76–7.67(m,2H),7.48(d,J=7.9Hz,1H),7.40(t,J=7.8Hz,1H),7.28(d,J=7.4Hz,1H),7.24(d,J=7.7Hz,1H),6.84(d,J=8.5Hz,1H),4.76(s,2H),4.62(s,2H),3.97(t,J=5.5Hz,2H),3.03(s,3H),2.74(t,J=5.5Hz,2H),2.54–2.46(m,4H),2.03(s,3H),1.76–1.69(m,4H).LC-MS(m/z):495.20/497.20(M+H)+.
7)357-dimethyl-14Synthesis of- (2- (pyrrolidin-1-yl) ethoxy) -2,4, 7-triaza-3 (2, 4) -pyrimidin-1, 5 (1, 3) -dibenzocyclooctan-6-one (L-1)
Figure BDA0003047622440000161
3- ((2-chloro-5-methylpyrimidin-4-yl) amino) N-methyl-N- (5-amino-2- (2- (pyrrolidin-1-yl) ethoxy) benzyl) benzamide (50.00mg, 0.1010mmol) was added to the reaction flask, followed by Cs2CO3(131.60mg,0.4039mmol),Xantphos(16.36mg,0.0283mmol),Pd2(dba)3(18.50mg,0.0202mmol),N2After displacement, anhydrous DMF solution (1.5 mL) was added and the reaction was heated at 140 ℃ for about 6h. After completion of the reaction, filtration and extraction with dichloromethane, the product was isolated by silica gel column chromatography (DCM: meOH = 20.
1H-NMR(400MHz,DMSO)δ9.11(s,1H),8.83(d,J=2.3Hz,1H),8.72(s,1H),7.89(s,1H),7.65(s,1H),7.42(t,J=7.8Hz,1H),7.25(d,J=7.6Hz,1H),7.06(d,J=7.3Hz,1H),7.04–6.97(m,1H),6.90(d,J=8.9Hz,1H),4.27(s,2H),4.03(t,2H),2.94(s,3H),2.81(t,2H),2.57–2.52(m,4H),2.11(s,3H),1.74–1.65(m,4H).13C NMR(151MHz,DMSO)δ170.58,159.24,157.89,156.77,150.08,139.64,137.56,135.26,128.83,124.70,124.48,122.75,120.15,119.67,118.25,112.92,104.37,67.48,54.50,54.11,47.74,32.20,23.27,13.45.HRMS(ESI):(m/z):[M+H]+calcd for C26H31N6O2 459.2508;found 459.2507.
EXAMPLE 2 preparation of Compound L-2
Figure BDA0003047622440000171
Synthesis of L-2 was carried out in the same manner as for L-1, to obtain 23mg (56%) of a pale yellow solid.
1H-NMR(400MHz,DMSO)δ9.19(s,1H),8.85(d,J=5.3Hz,2H),7.94(s,1H),7.53(s,1H),7.12(d,J=10.7Hz,1H),7.03(dd,J=8.8,2.6Hz,1H),6.96(d,J=8.5Hz,1H),6.91(d,J=8.9Hz,1H),4.27(s,2H),4.03(t,J=5.4Hz,2H),2.93(s,3H),2.79(s,2H),2.50–2.46(m,4H),2.12(s,3H),1.73–1.64(m,4H).13C-NMR(151MHz,DMSO)δ169.19,161.06,158.94,157.85,157.19,150.18,141.68,138.97,135.13,124.52,120.20,119.78,118.34,112.93,109.32,107.00,104.63,67.55,54.52,54.13,48.67,32.26,23.29,13.39.HRMS(ESI):(m/z):[M+H]+calcd for C26H30FN6O2 477.2414;found 477.2416.
EXAMPLE 3 preparation of Compound L-3
Figure BDA0003047622440000172
Synthesis of L-3 by the method used for the synthesis of L-1 gave 18mg (56%) of a pale yellow solid.
1H-NMR(400MHz,DMSO)δ9.12(s,1H),8.53(d,J=5.9Hz,2H),7.91(s,1H),7.66(d,J=6.7Hz,1H),7.42–7.34(m,1H),7.21(s,1H),7.02(d,J=6.6Hz,1H),6.90(d,J=8.7Hz,1H),4.26(s,2H),4.02(t,J=5.5Hz,2H),2.94(s,4H),2.79(t,2H),2.50–2.48(m,4H),2.11(s,3H),1.75–1.62(m,5H).13C-NMR(151MHz,DMSO)δ169.81,159.26,157.88,157.03,154.37,150.16,135.21,133.48,128.81,126.73,124.65,122.76,119.40,118.48,115.98,112.99,104.31,67.56,54.52,54.12,48.66,32.43,23.30,13.35.HRMS(ESI):(m/z):[M+H]+calcd for C26H30FN6O2 477.2414;found 477.2415.
EXAMPLE 4 preparation of Compound L-4
Synthesis of 1- ((4-bromo-2-butenyloxy) methyl) -3-bromobenzene
Figure BDA0003047622440000181
3-bromobenzyl alcohol (200.00mg, 1.0693mmol), 1, 4-dibromo-2-butene (206mg, 0.9631mmol) and Bu4NHSO4(36mg, 0.1060 mmol) was dissolved in methylene chloride (4 mL), and a saturated aqueous solution of sodium hydroxide (342mg, 8.5500 mmol) was added dropwise thereto and reacted at room temperature for 3 hours. After the reaction was monitored by TLC, dichloromethane was extracted, and the organic phases were combined, dried over anhydrous sodium sulfate and concentrated. The product was isolated by silica gel column chromatography (PE: EA = 100) to give 136.00mg (40%) of a colorless oily liquid.
1H-NMR(400MHz,DMSO)δ7.52–7.46(m,2H),7.36–7.30(m,2H),6.02–5.86(m,2H),4.47(s,2H),4.15(dd,J=4.5,1.7Hz,2H),4.02(d,J=3.1Hz,2H).
2) Synthesis of 1- (pyrrolidin-1-yl) ethoxy) - (2- ((4- (3-bromobenzyloxy) butyl-2-en-1-yl) oxy) methyl) -4-nitrobenzene
Figure BDA0003047622440000182
(5-Nitro-2- (2- (pyrrolidin-1-yl) ethoxy) phenyl) ethanol (100mg, 0.3755mmol), 60% NaH (18.78mg, 0.4694mmol) was added to the reaction flask, N2After displacement, anhydrous DMF (4 mL) was added and stirred at 0 ℃ for 40min. 1- ((4-bromo-2-butenyloxy) methyl) -3-bromobenzene (156.23mg, 0.4882mmol) was added dropwise and reacted at 0 ℃ for about 1h. After the reaction was completed, the reaction solution was poured into ice water, extracted with dichloromethane, and the organic phases were combined and concentrated. The product was isolated by silica gel column chromatography (DCM: meOH = 50) to give 100.58mg (53%) of 1- (pyrrolidin-1-yl) ethoxy) - (2- ((4- (3-bromobenzyloxy) butyl-2-en-1-yl) oxy) methyl) -4-nitrobenzene as a yellow solid.
1H-NMR(400MHz,DMSO)δ8.18(d,J=7.3Hz,2H),7.58–7.40(m,2H),7.38–7.26(m,2H),7.22(d,J=9.7Hz,1H),5.87(d,J=18.6Hz,2H),4.50(s,2H),4.47(s,2H),4.24(t,J=5.6Hz,2H),4.10(s,2H),4.02(s,2H),2.81(t,J=5.6Hz,2H),2.50(s,4H),1.65(s,4H).LC-MS(m/z):506.05(M+H)+.
3) Synthesis of 2-chloro-5-methyl-N- (3- (((4- ((5-nitro-2- (2- (pyridin-1-yl) ethoxy) oxy) butyl) -2-en-1-yl) oxy) methyl) phenyl) pyrimidin-4-amine
Figure BDA0003047622440000191
1- (pyrrolidin-1-yl) ethoxy) - (2- ((4- (3-bromobenzyloxy) butyl-2-en-1-yl) oxy) methyl) -4-nitrobenzene (500.00mg, 0.9893mmol), 2-chloro-5-methyl-4-aminopyrimidine (118mg, 0.8219mmol), cs2CO3(967mg,2.9679mmol)、Xantphos(114mg,0.1970mmol)、Pd2(dba)3(90mg, 0.0983mmol) was added to a reaction flask, N2Protection, anhydrous DMF (10 mL) was added and the reaction was carried out at 80 ℃ for about 10h. After the reaction is finished, filtering by using kieselguhr, extracting by using EA (ethylene-acetic acid) for three times, combining organic phases, drying by using anhydrous sodium sulfate and performing spin drying. The product was isolated by wet-loading on silica gel column chromatography (DCM: meOH = 50).
1H-NMR(400MHz,DMSO)δ8.90(s,1H),8.18(d,J=7.4Hz,2H),8.03(s,1H),7.60(s,2H),7.38–7.29(m,1H),7.22(d,J=9.2Hz,2H),7.06(d,J=7.2Hz,1H),5.93–5.82(m,2H),4.49(d,J=7.7Hz,4H),4.23(t,J=5.4Hz,2H),4.11(s,2H),4.05(s,2H),2.80(t,J=5.4Hz,2H),2.50(s,4H),2.16(s,3H),1.64(s,4H).
4) Synthesis of 2-chloro-5-methyl-N- (3- (((4- ((5-amino-2- (2- (pyridin-1-yl) ethoxy) oxy) butyl) -2-en-1-yl) oxy) methyl) phenyl) pyrimidin-4-amine
Figure BDA0003047622440000192
2-chloro-5-methyl-N- (3- (((4- ((5-nitro-2- (2- (pyridin-1-yl) ethoxy) oxy) butyl) -2-en-1-yl) oxy) methyl) phenyl) pyrimidin-4-amine (100mg, 0.1760mmol) was dissolved in a mixed solution of DCM: meOH =1 (4 mL), and SnCl was added under ice bath2(133.49mg, 0.7040mmol), moved to room temperature for about 18h, and monitored by TLC for progress of the reaction. After the reaction is finished, the solvent is dried by spinning, a small amount of dichloromethane is added for dissolution, and saturated Na is slowly added2CO3The aqueous solution was adjusted to pH 8 and then filtered through a sand core funnel, the filtrate was extracted three times with dichloromethane, the filter cake was washed with methanol to no product, after combining the organic phases, dried over anhydrous sodium sulfate, spun dry and purified by silica gel column chromatography (DCM: meOH = 30) to give 39.5g (41.7%) of a yellow solid.
1H-NMR(400MHz,DMSO)δ8.91(s,1H),8.04(s,1H),7.62(d,J=8.2Hz,1H),7.59(s,1H),7.34(t,J=7.8Hz,1H),7.06(d,J=7.5Hz,1H),6.68(d,J=8.6Hz,1H),6.60(d,J=2.5Hz,1H),6.43(dd,J=8.5,2.7Hz,1H),5.88-5.79(m,2H),4.63(s,2H),4.48(s,2H),4.36(s,2H),4.03(s,2H),4.00(s,2H),3.90(t,J=5.9Hz,2H),2.69(t,J=5.9Hz,2H),2.49-2.42(m,4H),2.17(s,3H),1.67-1.59(m,4H).LC-MS(m/z):539.25/541.20(M+H)+.
5)35-methyl-14Synthesis of- (2- (pyrrolidin-1-yl) ethoxy) -7, 12-dioxa-2, 4-diaza-3 (2, 4) -pyrimidine-1, 5 (1, 3) -dibenzocyclotridecan-9-ene (L-4)
Figure BDA0003047622440000201
2-chloro-5-methyl-N- (3- (((4- ((5-amino-2- (2- (pyridin-1-yl) ethoxy) oxy) butyl) -2-en-1-yl) oxy) methyl) phenyl) pyrimidin-4-amine (750mg, 1.3938mmol) was dissolved in N-butanol (15 mL), warmed to 105 ℃ and added TsOH. HH.in one portion2O (530.26mg, 2.7876mmol) was reacted at 105 ℃ for about 2 hours. After the reaction was completed, saturated Na was used2CO3The aqueous solution was adjusted to pH 8-9, extracted three times with dichloromethane, and the organic phase was washed with brine, dried over anhydrous sodium sulfate, spun-dried, and purified by silica gel column chromatography (DCM: meOH = 20.
1H-NMR(400MHz,DMSO)δ8.64(s,1H),8.30(s,1H),7.85(s,1H),7.61(s,1H),7.55(s,1H),7.28(d,J=7.7Hz,1H),7.19-7.07(m,2H),6.98(d,J=7.2Hz,1H),6.79(d,J=8.7Hz,1H),5.56-5.36(m,2H),4.30(s,2H),4.14(s,2H),4.02-3.93(m,4H),3.84(d,J=3.7Hz,2H),2.76(t,J=5.6Hz,2H),2.58-2.51(m,4H),2.08(s,3H),1.73-1.64(m,4H).13C-NMR(151MHz,DMSO)δ169.81,159.26,157.88,157.03,154.37,150.16,135.21,133.48,128.81,126.73,124.65,122.76,119.40,118.48,115.98,112.99,104.31,67.56,54.52,54.12,48.66,32.43,23.30,13.35.HRMS(ESI):(m/z):[M+H]+calcd for C29H36N5O3502.2818;found 502.2820.
EXAMPLE 5 preparation of Compound L-5
Figure BDA0003047622440000211
Synthesis of L-5 by the method of L-4 gave 121mg (54%) of a yellow solid.
1H-NMR(400MHz,DMSO)δ8.77(s,1H),8.50(s,1H),7.89(s,1H),7.63(d,J=2.5Hz,1H),7.43(s,1H),7.18(d,J=11.5Hz,1H),7.10(dd,J=8.7,2.5Hz,1H),6.82(d,J=8.8Hz,1H),6.77(d,J=9.1Hz,1H),5.58–5.31(m,2H),4.31(s,2H),4.18(s,2H),4.03–3.94(m,4H),3.84(d,J=4.8Hz,2H),2.77(t,J=5.8Hz,2H),2.53(s,4H),2.09(s,3H),1.68(s,4H).13C-NMR(151MHz,DMSO)δ162.68,161.09,158.76,156.84,151.35,141.98,141.45,133.81,129.98,129.48,126.74,122.08,120.92,116.94,112.22,108.50,108.34,105.01,71.38,70.55,69.49,67.84,66.60,54.56,54.25,23.31,13.51.HRMS(ESI):(m/z):[M+H]+calcd for C29H35FN5O3 520.2724;found 520.2727.
EXAMPLE 6 preparation of Compound L-6
Figure BDA0003047622440000212
Synthesis of L-6 by the method of L-4 gave 21mg (54%) of a yellow solid.
1H-NMR(400MHz,DMSO)δ8.71(s,1H),8.23(s,1H),7.82(s,1H),7.49–7.42(m,2H),7.34(s,1H),7.25(d,J=10.1Hz,1H),7.20(d,J=4.9Hz,1H),6.65(d,J=9.0Hz,1H),5.85–5.45(m,2H),4.43(s,2H),4.26(s,2H),4.01–3.94(m,4H),3.93(s,2H),2.74(t,J=5.5Hz,2H),2.50(s,5H),2.08(s,3H),1.67(s,4H).13C-NMR(151MHz,DMSO)δ159.97,158.49,155.64,155.22,149.60,134.99,133.51,130.38,129.58,127.22,126.86,126.57,125.70,119.98,118.51,115.73,111.89,104.90,70.50,69.85,69.51,67.31,60.99,53.78,53.00,22.78,13.95.HRMS(ESI):(m/z):[M+H]+calcd for C29H35FN5O3 520.2724;found 520.2725.
EXAMPLE 7 preparation of Compound L-7
Figure BDA0003047622440000221
Synthesis of L-7 was carried out in the same manner as in the synthesis of L-4 to obtain 47mg (54%) of a yellow solid.
1H NMR(400MHz,DMSO)δ8.65(s,1H),8.32(s,1H),7.85(s,1H),7.61(s,1H),7.55(d,J=2.1Hz,1H),7.28(d,J=7.9Hz,1H),7.18-7.10(m,2H),6.98(d,J=7.4Hz,1H),6.79(d,J=8.8Hz,1H),5.57-5.34(m,2H),4.30(s,2H),4.13(s,2H),3.96(d,J=5.3Hz,2H),3.82(d,J=4.5Hz,2H),3.71(s,3H),2.09(s,3H).13C NMR(151MHz,DMSO)δ159.14,158.76,156.32,152.13,139.82,139.74,133.83,129.96,129.46,127.90,126.28,122.24,121.88,121.55,120.88,110.99,104.79,71.59,70.29,69.05,66.52,55.58,13.59.HRMS(ESI):(m/z):[M+H]+calcd for C24H27N4O3 419.2083;found 419.2082.
EXAMPLE 8 molecular level Activity assay for JAK2 inhibitors
JAK2 kinase inhibitory activity assay:
the experimental principle is as follows:
JAK2 catalyzes the transfer of one phosphate group of Adenosine Triphosphate (ATP) to a polypeptide substrate, which is labeled with two fluorescent groups, coumarin and fluorescein. Based on fluorescence energy resonance transfer (FRET) method, JAK2 catalyzes ATP to react to cause two fluorophores to approach, a donor (coumarin) is excited at 400nM, part of the energy is released, the emission wavelength is 445nM, and the other part of the energy is transferred to fluoroscein, the emission wavelength is 520nM. Different compounds inhibit JAK2 to different extents, resulting in different extents of substrate phosphorylation, and thus the inhibition rates of different compounds were calculated by determining the ratio of the percentage of enzyme-catalyzed substrate phosphorylation.
The experimental method comprises the following steps:
adding 2.5. Mu.L of test compound, 5. Mu.L of kinase/peptide substrate mixture, 2.5. Mu.L of ATP solution into a 384-well plate, shaking 10. Mu.L of reaction system for 30s, mixing uniformly, and incubating at room temperature for 1h; adding 5 mu L of proteolytic enzyme, oscillating a reaction system of 15 mu L for 30s, mixing uniformly, and incubating for 1h at room temperature; adding 5 mu L of stopping reagent, oscillating the reaction system with the total volume of 20 mu L for 30s, mixing uniformly, and detecting a fluorescent signal by using a microplate reader, wherein the excitation wavelength is 400nm, and the emission wavelengths are 445nm and 520nm respectively. The inhibition of the compounds was determined at 7 concentration gradients and the IC of each compound was calculated by Origin8.0 fitting of the curve50The value is obtained. Positive control is carried out in the experimental process to confirm the feasibility of the reaction system, and each experiment is carried out in three parallels. Fedratinib is used as a positive control in the experimental process, and at least three replicates are arranged in each experiment.
TABLE 1 Activity of Compounds on JAK2 in vitro enzyme assays
Figure BDA0003047622440000231
Figure BDA0003047622440000232
CCK-8 cell proliferation assay:
the experimental principle is as follows:
WST-8 (chemical name: 2- (2-Methoxy-4-nitrophenyl) -3- (4-nitrophenyl) -5- (2, 4-disulfonic acid phenyl) -2H-tetrazole monosodium salt), which is reduced by a dehydrogenase in the cell to a yellow Formazan product (Formazan dye) with high water solubility under the action of the electron carrier 1-Methoxy-5-methylphenazinium dimethyl sulfate (1-Methoxy PMS). The amount of formazan produced was proportional to the number of living cells. Cell viability can therefore be calculated indirectly by measuring the light absorption at 450 nm. Half inhibition rate of the compound on H3122, SET2 and HEL cell proliferation is determined by CCK-8 method.
The experimental method comprises the following steps:
culturing cells using RPIM medium containing 10% FBS, adding 100. Mu.L per well, inoculating 3500 cells into 96-well plate, incubating at 37 ℃,5% CO2Culturing in an incubator for 24h, adding medicines with different concentration gradients respectively when the cell confluency reaches 50-70%, continuously incubating for 72h, adding 10 μ L of CCK-8 into each hole, shaking and uniformly mixing, incubating at 37 ℃ for 2h, and finally measuring the light absorption value of each hole under the wavelength of 450 nm. Finally, calculating IC by origin software50The value is obtained.
Note: HEL, SET-2, baF3 cells were purchased from ATCC (American type culture collection), Z' -LYTETM kinase assay platform from Invitrogen, and Cell Counting Kit-8 from Bycyan sky.
TABLE 2 antiproliferative activity of the compounds on cells
Figure BDA0003047622440000241
Kinase profiling test:
the kinase selectivity profile was performed by Discovex corporation. The compound L-4 is screened by kinase with the concentration of 100nM, and the screened targets comprise various wild type kinases of human, mutants, pathogens, liposomes and 468. The screening results are expressed as "percent control" (see fig. 1), where a smaller number indicates a stronger hit, i.e. stronger binding to the target. In the kinase spectrum dendrogram, the kinases found to have binding activity are marked with red circles, the larger the red circle the higher the binding affinity.
The kinase selectivity spectrum of the compound L-4 is shown in figure 2, and the result shows that the compound L-4 has better selectivity on JAK2 kinase.
Mouse in vivo UC model:
the experimental animals were purchased from Shanghai Sphere-BiKai experimental animals Co., ltd, and were BALB/c male mice, and the weight was 20-22g. Dextran sulfate sodium salt (DSS) was purchased from saint assist organisms. SASP is used as a positive drug. The mice were modeled for 1-7 days, and on day 8, they were again divided into four groups for intragastric administration, once a day: model tap water, control tap water, SASP500mg/kg, L13 mg/kg. The colon is taken on day 15 for length measurement and 1-2 cm of the colon is fixed with formaldehyde.
The effect of L-4 on ulcerative colitis in mice is shown in FIG. 3, and FIG. 3 (A) shows the body weight change of mice, which was not different from those in the model-making group and SASP group after administration. FIG. 3 (B) shows a graph of the change in mouse DAI, with a significant improvement in the mouse DAI score relative to the building block after administration, comparable to the SASP group; FIG. 3 (C) shows the effect of compound L-4 on the ratio of the length of colon mass, and the results show that the index is significantly improved relative to the building block in mice after administration, comparable to the SASP group.
All documents mentioned in this application are incorporated by reference in this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

1. A compound shown as the following formula I, or a stereoisomer or an optical isomer, a pharmaceutically acceptable salt, a prodrug or a solvate thereof,
Figure FDA0003047622430000011
in the formula (I), the compound is shown in the specification,
x is selected from N and CH;
R1selected from the group consisting of: hydrogen, halogen, C1-C10Alkyl, hydroxy, C1-C10Alkoxy, nitro, amino, acetylamino, C1-C3Alkyl formyl, hydroxy acetyl, C1-C3An alkoxyformyl group; n is selected from 0,1,2 and 3;
R2selected from the group consisting of: hydrogen, halogen, C1-C10Alkyl radical, C2-C10An acyl group;
R3selected from the group consisting of: hydrogen, halogen, hydroxy, amino, C1-C10Alkyl radical, C1-C10Alkoxy, or substituted by one or more C1-C10Alkyl or C1-C10Alkoxy-substituted 4-to 6-membered nitrogen-or oxygen-containing heterocycles;
R4selected from the group consisting of: hydrogen, substituted or unsubstituted C1-C10Alkyl, substituted or unsubstituted C1-C10Alkyl- (4-to 6-membered nitrogen-or oxygen-containing heterocycle);
X1is- (L)m-, wherein said L is selected from the group consisting of: -CHR-, -C (O) -, -NR-, -O-, -CH = CH-,
Figure FDA0003047622430000012
r is H or C1-C4An alkyl group; m is 2,3, 4, 5, 6, 7, 8, 9, 10 or 11;
unless otherwise specified, the substitution refers to the substitution of one or more hydrogen atoms on the group with a substituent selected from the group consisting of: halogen, C1-C10An alkyl group.
2. The compound of claim 1, or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, wherein X is1Is a chain structure selected from the group consisting of:
Figure FDA0003047622430000013
wherein R is5Is C1-C3An alkyl group;
l, m, o and r are each independently selected from 0,1 or 2;
p and q are each independently selected from 0,1,2,3.
3. The compound of claim 1, or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, wherein the compound of formula I has the structure shown in formula III below:
Figure FDA0003047622430000021
in the formula
R2、R3、R4、X1Is as defined in claim 1.
4. The compound of claim 2, or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt, prodrug, or solvate thereof, wherein the compound of formula I has the structure shown in formula III below:
Figure FDA0003047622430000022
wherein, X1Is selected from
Figure FDA0003047622430000023
R4Selected from the group consisting of: hydrogen, methyl, or a structural unit selected from the group consisting of:
Figure FDA0003047622430000025
wherein x is 1,2 or 3.
5. The compound of claim 3, or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt, prodrug, or solvate thereof, wherein the compound of formula I has the structure shown in formula IV below:
Figure FDA0003047622430000031
R4selected from:
Figure FDA0003047622430000032
X1selected from the group consisting of:
Figure FDA0003047622430000033
6. the compound of any one of claims 1-4, or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, wherein the compound is selected from the group consisting of:
Figure FDA0003047622430000034
7. a pharmaceutical composition comprising a compound of any one of claims 1-6, or a stereoisomer or an optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, and a pharmaceutically acceptable carrier or excipient.
8. Use of a compound of any one of claims 1-7, or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, for the manufacture of a medicament for the prevention or treatment of a JAK 2-mediated disease; and/or for the preparation of a JAK2 inhibitor.
9. The use according to claim 8, wherein the JAK 2-mediated disease is myelodysplastic syndrome (MDS), eosinophilia, tumor, inflammatory disease, or infection by bacteria, viruses, or fungi;
preferably, the tumor is selected from the group consisting of: myeloproliferative carcinoma (MPN), melanoma, lung cancer, kidney cancer, ovarian cancer, prostate cancer, breast cancer, colon cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, uterine cancer, rectal cancer, anal cancer, stomach cancer, testicular cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, hodgkin's disease, non-hodgkin's lymphoma, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, pediatric solid tumors, lymphocytic lymphoma, cancer of the bladder, cancer of the kidney or ureter, cancer of the renal pelvis, tumors of the Central Nervous System (CNS), primary CNS lymphoma, tumor angiogenesis, spinal axis tumors, brain stem glioma, pituitary adenoma, kaposi's sarcoma, epidermoid carcinoma, T-cell lymphoma; and/or
The inflammatory disease is selected from the group consisting of: rheumatoid arthritis, ankylosing spondylitis, autoimmune hemolytic anemia, arthritis, myasthenia gravis, systemic lupus erythematosus, pernicious anemia, polymyositis; and/or
The virus is selected from the group consisting of: hepatitis viruses (type a, type b and type c), herpes viruses, influenza viruses, adenoviruses, coronaviruses, measles viruses, dengue viruses, polio viruses, rabies viruses; and/or
The bacteria are selected from the group consisting of: chlamydia, rickettsia, mycobacteria, staphylococci, pneumococci, cholera, tetanus; and/or
The fungus is selected from the group consisting of: candida, aspergillus, dermatitis bud yeast.
10. A JAK2 inhibitor comprising a compound of any one of claims 1 to 6, or a stereoisomer or an optical isomer thereof, or a pharmaceutically acceptable salt, prodrug or solvate thereof, or a pharmaceutical composition of claim 7.
CN202110476658.XA 2021-04-29 2021-04-29 Cyclic compounds as JAK2 inhibitors and uses thereof Pending CN115260202A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110476658.XA CN115260202A (en) 2021-04-29 2021-04-29 Cyclic compounds as JAK2 inhibitors and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110476658.XA CN115260202A (en) 2021-04-29 2021-04-29 Cyclic compounds as JAK2 inhibitors and uses thereof

Publications (1)

Publication Number Publication Date
CN115260202A true CN115260202A (en) 2022-11-01

Family

ID=83745842

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110476658.XA Pending CN115260202A (en) 2021-04-29 2021-04-29 Cyclic compounds as JAK2 inhibitors and uses thereof

Country Status (1)

Country Link
CN (1) CN115260202A (en)

Similar Documents

Publication Publication Date Title
CN109422726B (en) Blocking agent of CD47/SIRP alpha and application thereof
JP3649395B2 (en) Fused heteroaryl derivatives
JP6606278B2 (en) 2-Arylaminopyridine, pyrimidine or triazine derivatives and their production and use
CN112119070B (en) Selective JAK2 inhibitors and uses thereof
WO2013170770A1 (en) Acetylene derivatives having antitumor activity
WO2013170671A1 (en) Pteridine ketone derivative and applications thereof as egfr, blk, and flt3 inhibitor
AU2016272057A1 (en) Use of pteridinone derivative serving as EGFR inhibitor
CN107245075B (en) 2,4, 6-trisubstituted pyrido [3,4-d ] pyrimidine compound and salt and application thereof
CN114656482A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN105732615A (en) CDK kinase inhibitor
CN112321604A (en) Macrocyclic JAK2 inhibitor and application thereof
JP2020023441A (en) Novel compound useful for egfr inhibition and tumor treatment
CN112867717A (en) Compounds useful as kinase inhibitors and uses thereof
CN108329274B (en) Bruton's tyrosine kinase inhibitors
CN107793363B (en) Substituted arylamino aromatic heterocyclic compound and application thereof as antitumor drug
JP3810017B2 (en) Fused heteroaryl derivatives
CN108456214B (en) Quinazoline compound containing oxazole or imidazole structure and application thereof
CN115260202A (en) Cyclic compounds as JAK2 inhibitors and uses thereof
CN105461708A (en) Quinazoline tyrosine kinase inhibitor, and preparation method and application thereof
CN111138417B (en) Triazole modified 5-fluoro-2, 4-pyrimidinediamine compound and application thereof
CN109438279B (en) Small molecule compound for overcoming EGFR drug-resistant mutation and preparation method and application thereof
WO2017049711A1 (en) Quinoline derivative, and pharmaceutical composition, preparation method and use thereof
CN111217821B (en) Preparation method of series dioxane quinazoline derivatives
CN115894486B (en) Hydrogenated pyridoquinazoline compound, composition and application thereof
CN113135938B (en) Substituted macrocyclic tyrosine kinase inhibitors and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication