WO2017092523A1 - Composé de pyrimidine condensée, composition le comprenant et son utilisation - Google Patents

Composé de pyrimidine condensée, composition le comprenant et son utilisation Download PDF

Info

Publication number
WO2017092523A1
WO2017092523A1 PCT/CN2016/103368 CN2016103368W WO2017092523A1 WO 2017092523 A1 WO2017092523 A1 WO 2017092523A1 CN 2016103368 W CN2016103368 W CN 2016103368W WO 2017092523 A1 WO2017092523 A1 WO 2017092523A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
formula
hydrogen
disease
Prior art date
Application number
PCT/CN2016/103368
Other languages
English (en)
Chinese (zh)
Inventor
王义汉
李焕银
Original Assignee
深圳市塔吉瑞生物医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳市塔吉瑞生物医药有限公司 filed Critical 深圳市塔吉瑞生物医药有限公司
Priority to CN201680056599.8A priority Critical patent/CN108137614B/zh
Publication of WO2017092523A1 publication Critical patent/WO2017092523A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the invention belongs to the technical field of medicine, and in particular relates to a fused pyrimidine compound and a composition comprising the same and use thereof.
  • Protein tyrosine kinases play an important role in cell regulation, and abnormal expression or mutations have been observed in cancer cells or autoimmune diseases. Protein tyrosine kinases are enzymes that catalyze the transport of phosphate groups from ATP to tyrosine located on a protein substrate. Many growth factor receptor proteins function as tyrosine kinases to transmit cellular signals. The interaction between growth factors and their receptors usually controls cell growth, but abnormal signaling caused by mutation or overexpression of any of the receptors often induces a variety of cancers or autoimmune diseases (eg rheumatoid joints) inflammation).
  • autoimmune diseases eg rheumatoid joints
  • EGFR tyrosine kinase inhibitor is a molecularly targeted drug targeting EGFR, which binds to the EGFR tyrosine kinase catalytic domain binding site on the cell surface by competitively binding to ATP, blocking signaling into cells. Further delivery inhibits tumor cell growth and induces apoptosis.
  • EGFR-TKI such as erlotinib and gefitinib have been widely used in clinical practice.
  • EGFR inhibitors such as gefitinib and erlotinib have achieved remarkable results in EGFR-mutant advanced non-small cell lung cancer (NSCLC).
  • NSCLC EGFR-mutant advanced non-small cell lung cancer
  • BTK Bruton's tyrosine kinase
  • abnormal signaling can induce dysregulated B cell proliferation and differentiation, leading to all kinds of lymphoma, including various acute or chronic lymphocytic leukemia; and can lead to the formation of autoantibodies, causing inflammatory diseases, autoimmune Disease and/or immune mediated disease.
  • T cells play a role in signaling through the activation of various kinases between cells, such as the Janus kinase, which are transmitted by antigen presenting cells to downstream effectors via T cell receptors on the cell surface. At this time, they secrete various interleukins or interferon- ⁇ to activate various white blood cells as well as B cells.
  • Protein kinases involved in T cell signaling are Janus kinase (JAK) (eg, JAK1, JAK2, JAK3, and TYK2), IL-2 inducible T cell kinase (ITK), and TEC family kinases (eg, resting lymphocyte kinase, Resting) Lymphocyte Kinase, RLK).
  • JAK3 inhibitors can be used to treat rheumatoid arthritis, psoriasis, allergic dermatitis, lupus, multiple sclerosis, complications of type I diabetes and diabetes, cancer, asthma, autoimmune thyroid disease, ulcerative colitis Crohn's disease, Alzheimer's disease, leukemia and other indications in which immunosuppression is beneficial (eg organ transplantation or xenograft).
  • the present invention discloses a fused pyrimidine compound, a composition comprising the same, and a use thereof, which have protein kinase inhibitory activity and have better pharmacodynamic/pharmacokinetic properties.
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or solvent compound thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 and R 20 are each independently selected from hydrogen, deuterium, halogen or trifluoromethyl;
  • X is selected from O, NH, S or SO 2 ;
  • Y is selected from the group consisting of hydrogen, deuterium, halogen, optionally substituted C1-6 alkyl one or more times, and optionally one or more deuterated C1-6 alkoxy groups;
  • Z is selected from the group consisting of hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 .
  • the compound of formula (I) contains at least one ruthenium atom, more preferably three ruthenium atoms, more preferably six ruthenium atoms, more preferably eight ruthenium atoms.
  • the cerium isotope content of cerium in the deuterated position is at least 0.015%, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than the natural strontium isotope content.
  • the ground is greater than 95%, more preferably greater than 99%.
  • the strontium isotope content of each of the R 9 , R 16 , R 17 , R 18 , R 19 and R 20 is at least 5%, preferably greater than 10%, more preferably greater than 15%, and even more preferably greater than 20%.
  • the ground is greater than 95%, more preferably greater than 99%.
  • the sixteen places contain sputum, and more preferably seventeen contain sputum, more preferably eighteen contain sputum, more preferably nineteen contain sputum, and more preferably twenty contain
  • the compound of the formula (I) contains at least one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, ten Three, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, twenty-one, twenty-two, twenty-three, twenty-four Helium atom.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are each independently hydrazine or hydrogen.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are deuterium.
  • R 9 , R 10 and R 11 are each independently hydrazine or hydrogen.
  • R 9 , R 10 , R 11 are deuterium.
  • R 12 and R 13 are each independently hydrazine or hydrogen.
  • R 12 and R 13 are deuterium.
  • R 14 , R 15 , R 16 and R 17 are each independently hydrazine or hydrogen.
  • R 14 , R 15 , R 16 , R 17 are deuterium.
  • R 18 , R 19 and R 20 are each independently hydrazine or hydrogen.
  • R 18 , R 19 , R 20 are deuterium.
  • X is selected from the group consisting of O, NH, S or SO 2 .
  • X is O.
  • X is NH
  • Y is selected from hydrogen, deuterium, halogen, optionally substituted by one or more C 1-6 alkyl deuterated and optionally one or more deuterated C 1-6 alkoxy .
  • Y is a hydrogen atom
  • Y is a fluorine atom.
  • Y is three deuterated methoxy group (-OCD 3).
  • Z is selected from the group consisting of hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 .
  • Z is methyl three deuterated (CD 3).
  • the present invention also discloses a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a fused pyrimidine compound as described above, or a polymorph thereof, a pharmaceutically acceptable salt, a former A drug, stereoisomer, isotope variant, hydrate or solvate.
  • the present invention also discloses a process for the preparation of a pharmaceutical composition as described above, comprising the steps of: pharmaceutically acceptable excipients with a fused pyrimidine compound as described above, or polymorph thereof
  • pharmaceutically acceptable salts, prodrugs, stereoisomers, isotopic variations, hydrates or solvates are combined to form a pharmaceutical composition.
  • the pharmaceutical composition is an injection, a sachet, a tablet, a pill, a powder or a granule.
  • the pharmaceutical composition further comprises an additional therapeutic agent, which is cancer, cardiovascular disease, inflammation, infection, immune disease, cell proliferative disease, viral disease. , a metabolic disease, or a drug for organ transplantation.
  • an additional therapeutic agent which is cancer, cardiovascular disease, inflammation, infection, immune disease, cell proliferative disease, viral disease. , a metabolic disease, or a drug for organ transplantation.
  • the invention provides a compound of the first aspect of the invention, or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or solvent thereof Use of the compound for the preparation of a medicament for the treatment and/or prevention of a disease associated with a protein kinase.
  • the present invention provides a method of treating and/or preventing a protein kinase-associated disease in a subject,
  • the method comprises administering to the subject a fused pyrimidine compound of formula (I), a polymorph, a pharmaceutically acceptable salt, a prodrug, a stereoisomer, an isotope variant, a hydrate or a solvent compound, Or a pharmaceutical composition thereof.
  • the present invention provides a polypyrimidine compound of the formula (I), a polymorph, a pharmaceutically acceptable salt, a prodrug, a stereoisomer, an isotopic variation, a hydrate or a solvent compound, or A pharmaceutical composition thereof for use in the treatment and/or prevention of a disease associated with a protein kinase.
  • the compound or pharmaceutical composition is for treating and/or preventing a disease: cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplantation, viral disease, cardiovascular disease Or metabolic disease.
  • the cancer includes, but is not limited to, lung cancer, head and neck cancer, breast cancer, prostate cancer, esophageal cancer, rectal cancer, colon cancer, nasopharyngeal cancer, uterine cancer, pancreatic cancer, lymphoma, blood cancer , osteosarcoma, melanoma, kidney cancer, stomach cancer, liver cancer, bladder cancer, thyroid cancer or colorectal cancer.
  • the immune disease or inflammation includes, but is not limited to, rheumatoid arthritis, osteoarthritis, rheumatoid spondylitis, gout, asthma, bronchitis, rhinitis, chronic obstructive pulmonary disease, pouch Sexual fibrosis.
  • the cell proliferative disorder refers to lung cancer, head and neck cancer, breast cancer, prostate cancer, esophageal cancer, rectal cancer, colon cancer, nasopharyngeal cancer, uterine cancer, pancreatic cancer, lymphoma, blood cancer. , osteosarcoma, melanoma, kidney cancer, stomach cancer, liver cancer, bladder cancer, thyroid cancer or colorectal cancer.
  • the cancer is non-small cell lung cancer.
  • the protein kinase is selected from the group consisting of: epidermal growth factor receptor (EGFR) tyrosine kinase or a mutant thereof, Bruton's tyrosine kinase (BTK), Janus kinase 3 (JAK3), interleukin -2 Inducible T cell kinase (ITK), resting lymphocyte kinase (RLK) and bone marrow tyrosine kinase (BMX).
  • EGFR epidermal growth factor receptor
  • BTK Bruton's tyrosine kinase
  • JAK3 Janus kinase 3
  • ITK interleukin -2 Inducible T cell kinase
  • RTK resting lymphocyte kinase
  • BMX bone marrow tyrosine kinase
  • the present invention provides a kit comprising: a first container comprising a fused pyrimidine compound of formula (I), a polymorph, a pharmaceutically acceptable salt, a prodrug, a stereoisomer thereof An isotope variant, hydrate or solvent compound; and, optionally, a second container containing other therapeutic agents; and optionally a third container containing for diluting or suspending the compound and/or other treatment A pharmaceutically acceptable excipient for the drug.
  • the invention also discloses the use of a fused pyrimidine compound as described above for the preparation of a pharmaceutical composition for inhibiting protein kinases.
  • a pharmaceutical composition for inhibiting protein kinases Preferably, it is used to prepare a pharmaceutical composition that inhibits EGFR kinase.
  • the compound of the formula (I) of the present invention is selectively and effectively inhibited mainly in an abnormality.
  • Bruton tyrosine kinase (BTK), Janus kinase 3 (JAK3), interleukin-2 inducible T cell kinase (ITK), resting lymphocyte kinase (RLK) expressed in activated B lymphocytes and/or T lymphocytes And bone marrow tyrosine kinase (BMX).
  • the compound of formula (I) of the present invention can treat or prevent abnormally activated B lymphocytes A cancer, a tumor, an inflammatory disease, an autoimmune disease, or an immune-mediated disease caused by cells, T lymphocytes, or both. Accordingly, the present invention also provides a pharmaceutical composition for treating and/or preventing cancer, a tumor, an inflammatory disease, an autoimmune disease or an immune-mediated disease, comprising the compound of the formula (I) of the present invention or Polymorphic forms, pharmaceutically acceptable salts, prodrugs, stereoisomers, isotopic variations, hydrates or solvates are included as active ingredients.
  • inflammatory diseases may include, but are not limited to, arthritis, rheumatoid Arthritis, spondylarthritis, gouty arthritis, osteoarthritis, juvenile arthritis, other arthritic conditions, lupus, systemic lupus erythematosus (SLE), skin-related diseases, psoriasis, eczema, dermatitis, Allergic dermatitis, pain, lung disease, lung inflammation, adult respiratory distress syndrome (ARDS), pulmonary sarcoidosis, chronic pulmonary inflammatory disease, chronic obstructive pulmonary disease (COPD), cardiovascular disease, atherosclerosis , myocardial infarction, congestive heart failure, myocardial ischemia-reperfusion injury, inflammatory bowel disease, Crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, Sjogren's syndrome, autoimmune thyroid disease, urticaria , multiple sclerosis,
  • a compound of the formula (I) of the present invention or a polymorphic form thereof, a pharmaceutically acceptable salt, a prodrug, a stereoisomer, an isotope variant, a hydrate or a solvate thereof, is used for the treatment of inflammation
  • a compound of the formula (I) of the present invention, or a polymorphic form, a pharmaceutically acceptable salt, a prodrug, a stereoisomer thereof, when administered in combination with a therapeutic agent for a sexual disease, an autoimmune disease, and an immune-mediated disease Isotope variants, hydrates or solvates provide enhanced therapeutic effects.
  • Representative examples of other therapeutic agents for treating inflammatory diseases, autoimmune diseases, and immune-mediated diseases may include, but are not limited to, steroidal drugs (eg, prednisone, hydrogenated prednisone, methylhydrogenated waves) Nixon, cortisone, hydroxycortisone, betamethasone, dexamethasone, etc.), methotrexate, leflunomide, anti-TNF ⁇ agents (eg, etanercept, infliximab, Ada Limonizumab, etc., calcineurin inhibitors (eg, tacrolimus, pimecrolimus, etc.) and antihistamines (eg, diphenhydramine, hydroxyzine, loratadine, iba Statin, ketotifen, cetirizine, levocetirizine, fexofenadine, etc., and at least one therapeutic agent selected from the group consisting of may be included in the pharmaceutical composition of the present invention.
  • steroidal drugs
  • the invention also includes isotopically labeled compounds (also referred to as "isotopic variants"), equivalent to the original compounds disclosed herein.
  • isotopes which may be listed as compounds of the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, respectively. , 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • isotopically-labeled compounds of the present invention such as the radioisotopes of 3 H and 14 C, are also among them, useful in tissue distribution experiments of drugs and substrates. ⁇ , ie 3 H and carbon-14, ie 14 C, are easier to prepare and detect and are preferred in isotopes. In addition, heavier isotopic substitutions such as guanidine, or 2 H, are preferred in certain therapies due to their good metabolic stability, such as increased half-life or reduced dosage in vivo, and therefore may be preferred in certain circumstances. Isotopically labeled compounds can be prepared in a conventional manner by substituting a readily available isotopically labeled reagent with a non-isotopic reagent using the protocol of the examples.
  • the beneficial effects of the present invention are as follows: First, the fused pyrimidine compound adopting the technical scheme of the present invention has excellent inhibition to protein kinase. Second, the metabolism of the compound in the organism is improved, and the compound has better pharmacokinetic parameters. characteristic. In this case, the dosage can be changed and a long-acting preparation can be formed to improve the applicability. Third, the drug concentration of the compound in the animal is increased, and the drug efficacy is improved. Fourth, certain metabolites are inhibited and the safety of the compounds is increased.
  • C 1 -C 6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1 -C 6 , C 1 -C 5 , C 1 -C 4 , C 1 - C 3 , C 1 -C 2 , C 2 -C 6 , C 2 -C 5 , C 2 -C 4 , C 2 -C 3 , C 3 -C 6 , C 3 -C 5 , C 3 -C 4 C 4 -C 6 , C 4 -C 5 and C 5 -C 6 alkyl.
  • Halogen means fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
  • C 1 -C 6 alkyl refers to a straight or branched saturated hydrocarbon group having from 1 to 6 carbon atoms, also referred to herein as “lower alkyl.” In some embodiments, a C 1 -C 4 alkyl group is particularly preferred.
  • alkyl group examples include, but are not limited to, methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), uncle Butyl (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl (C 5 ), 3-methyl-2-butyl (C 5 ), tert-amyl (C 5 ) and n-hexyl (C 6 ).
  • each alkyl group is independently optionally substituted, that is, unsubstituted (“unsubstituted alkyl") or substituted with one or more substituents ("substituted alkyl”); for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent.
  • the alkyl group is unsubstituted C 1 -C 6 alkyl (e.g., -CH 3).
  • the alkyl group is a substituted C 1 -C 6 alkyl.
  • C 1 -C 6 alkoxy refers to the group -OR wherein R is a substituted or unsubstituted C 1 -C 6 alkyl group. In some embodiments, a C 1 -C 4 alkoxy group is particularly preferred. Specific alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentyloxy, N-Hexyloxy and 1,2-dimethylbutoxy.
  • polymorph refers to a different arrangement of chemical drug molecules, generally expressed as the presence of a pharmaceutical material in a solid state.
  • a drug may exist in a plurality of crystalline forms, and different crystal forms of the same drug may have different dissolution and absorption in the body, thereby affecting the dissolution and release of the formulation.
  • pharmaceutically acceptable salt means that, within the scope of sound medical judgment, it is suitable for contact with tissues of humans and lower animals without excessive toxicity, irritation, allergies, etc., and with reasonable benefits/dangers. Those salts that are proportionate.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al., pharmaceutically acceptable salts as described in detail in J. Pharmaceutical Sciences (1977) 66: 1-19.
  • Pharmaceutically acceptable salts of the compounds of the invention include those derived from suitable inorganic and organic acids and inorganic and organic bases.
  • non-toxic acid addition salts examples include salts with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or salts with organic acids such as acetic acid, oxalic acid, Maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Also included are salts formed using conventional methods in the art, for example, ion exchange methods.
  • adipic acid salts alginate, ascorbate, aspartate, besylate, benzoate, disulfate, borate, butyrate, camphor Acid salt, camphor sulfonate, citrate, cyclopentanoate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate, gluconate, glycerol Phosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate , malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate Salt, pectin
  • Pharmaceutically acceptable salts derived from suitable bases include the alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like.
  • other pharmaceutically acceptable salts include non-toxic ammonium salts, quaternary ammonium salts and amine cations formed with counterions, counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, Nitrate, lower alkyl sulfonate and aryl sulfonate.
  • Subjects for administration include, but are not limited to, humans (ie, males or females of any age group, eg, pediatric subjects (eg, infants, children, adolescents) or adult subjects (eg, young Adults, middle-aged adults or older adults) and/or non-human animals, for example, mammals, for example, primates (eg, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep , goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "person,” “patient,” and “subject” are used interchangeably herein.
  • treating includes the effect that occurs when a subject has a particular disease, disorder or condition that reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition.
  • prevention includes the effect that occurs before a subject begins to have a particular disease, disorder or condition.
  • the compounds of the invention may include one or more asymmetric centers, and thus may exist in a variety of "stereoisomer" forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the invention may be in the form of individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of a mixture of stereoisomers, A racemic mixture and a mixture rich in one or more stereoisomers are included.
  • the isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of a chiral salt; or preferred isomers can be passed Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • prodrugs are also included within the context of the present invention.
  • the term "prodrug” as used herein refers to a compound which is converted in vivo to an active form thereof having a medical effect by, for example, hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, ACSSymposium Series, Vol. 14, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: Solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each This article is incorporated herein by reference.
  • a prodrug is any covalently bonded carrier which, when administered to a patient, releases the compound of the invention in vivo.
  • Prodrugs are typically prepared by modifying functional groups that cleave the prodrug in vivo to yield the parent compound.
  • Prodrugs include, for example, a compound of the invention wherein a hydroxy, amino or thiol group is bonded to any group which, when administered to a patient, can be cleaved to form a hydroxy, amino or thiol group.
  • representative examples of prodrugs include, but are not limited to, covalent derivatives of the compounds of the invention formed by the hydroxyl, amino or thiol functional groups thereof with acetic acid, formic acid or benzoic acid.
  • an ester such as a methyl ester, an ethyl ester or the like can be used.
  • the ester itself may be active and/or may hydrolyze under conditions in humans.
  • Suitable pharmaceutically acceptable in vivo hydrolysable ester package Those which are easily decomposed in the human body to release the parent acid or its salt.
  • “Pharmaceutically acceptable excipient” as used in the present invention refers to a non-toxic carrier, adjuvant or vehicle which does not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants, or vehicles that can be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (eg, human serum albumin) ), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based material, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxyprop
  • the present invention relates to a fused pyrimidine compound of the formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotope variant, hydrate or solvent compound thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 and R 20 are each independently selected from hydrogen, deuterium, halogen or trifluoromethyl;
  • X is selected from O, NH, S or SO 2 ;
  • Y is selected from hydrogen, deuterium, halogen, optionally substituted by one or more C 1-6 alkyl deuterated and optionally one or more deuterated C 1-6 alkoxy;
  • Z is selected from the group consisting of hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 ;
  • the above fused pyrimidine compound contains at least one ruthenium atom.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 and R 20 are each independently selected from hydrogen, deuterium, halogen or trifluoromethyl" including R 1 being selected from hydrogen, deuterium, halogen or trifluoromethyl, R 2 Selected from hydrogen, hydrazine, halogen or trifluoromethyl, R 3 is selected from hydrogen, deuterium, halogen or trifluoromethyl, and so on, until R 20 is selected from the group consisting of hydrogen, hydrazine, halogen or trifluoromethyl .
  • R 1 is hydrogen, R 1 is deuterium, R 1 is halogen (F, Cl, Br or I) or R 1 is trifluoromethyl
  • R 2 is hydrogen, R 2 is deuterium, and R 2 is Halogen (F, Cl, Br or I) or R 2 is trifluoromethyl
  • R 3 is hydrogen, R 3 is deuterium, R 3 is halogen (F, Cl, Br or I) or R 3 is trifluoromethyl
  • R 20 is hydrogen, R 20 is deuterium, R 20 is halogen (F, Cl, Br or I) or R 20 is a trifluoromethyl group.
  • X is selected from O, NH, S, or SO 2
  • X is O
  • X is NH
  • X is S
  • X 2 is SO aspect
  • Y is selected from the group consisting of hydrogen, deuterium, halogen, optionally substituted C1-6 alkyl one or more times, and optionally one or more deuterated C1-6 alkoxy groups.
  • including Y is hydrogen, Y is hydrazine, Y is halogen, Y is optionally substituted by one or more C 1-6 alkyl groups, and Y is optionally one or more deuterated C 1-6 alkoxy groups.
  • a halogen including Y is hydrogen, Y is deuterium, Y is F, Cl, Br or I, and Y is not deuterated or is once, twice, three times, four times, five times, six times or more.
  • C 1-6 alkyl groups eg methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, isobutyl, n-pentyl, 3-pentyl
  • Base pentyl, neopentyl, 3-methyl-2-butyl, tert-amyl and n-hexyl, etc.
  • Y is not deuterated or is once, twice, three times, four times, five times, six
  • One or more deuterated C 1-6 alkoxy groups eg, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, A technical solution of n-pentyloxy, n-hexyloxy and 1,2-dimethylbutoxy groups.
  • Z is selected from the group consisting of hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 ′′ includes Z as hydrogen (H), Z as ⁇ (D), Z as methyl, Z as CH 2 D, Z is CHD 2 , Z is CD 3 , Z is CH 2 CH 3 , Z is CHDCH 3 , Z is CHDCH 2 D, Z is CHDCHD 2 , Z is CHDCD 3 , Z is CD 2 CH 3 , Z is CD 2 CH 2 D, Z is a technical solution of CD 2 CHD 2 and Z is CD 2 CD 3 .
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or A solvent compound wherein X is O or NH, Y is hydrogen, deuterium, halogen, optionally substituted by one or more C 1-6 alkoxy groups, and R 1 - R 20 and Z are as defined above.
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or a solvent compound, wherein X is O or NH, Y is hydrogen, deuterium, halogen, optionally substituted by one or more C 1-6 alkoxy groups, and R 1 - R 20 are each independently selected from hydrogen or hydrazine, and Z is selected from methyl or CD 3 .
  • X is O or NH
  • Y is hydrogen, deuterium, halogen, optionally substituted by one or more C 1-6 alkoxy groups
  • R 1 - R 20 are each independently selected from hydrogen or hydrazine
  • Z is selected from methyl or CD 3 .
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or a solvent compound wherein X is O or NH, Y is hydrogen, hydrazine, F, methoxy or a tertiary deuterated methoxy group (-OCD 3 ), and R 1 to R 20 are each independently selected from hydrogen or hydrazine, and Z is selected from methyl or CD 3 .
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or a solvent compound wherein X is O or NH, Y is hydrogen, hydrazine, F, methoxy or triple deuterated methoxy (-OCD 3 ), and R 1 to R 8 are each independently selected from hydrogen or hydrazine, R 9- R 20 is hydrogen and Z is selected from methyl or CD 3 .
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or A solvent compound wherein X is O, Y is hydrogen or deuterium, and R 1 - R 20 and Z are as defined above.
  • the invention relates to a fused pyrimidine compound of formula (I), or a polymorph, pharmaceutically acceptable salt, prodrug, stereoisomer, isotopic variation, hydrate or A solvent compound wherein X is O, Y is hydrogen or deuterium, R 1 - R 8 are each independently selected from hydrogen or deuterium, R 9 - R 20 are hydrogen, and Z is selected from methyl or CD 3 .
  • the fused pyrimidine compound is any of the following structures, or a pharmaceutically acceptable salt thereof, but is not limited to the following structures:
  • formulation examples illustrate representative pharmaceutical compositions that can be prepared in accordance with the present invention.
  • the invention is not limited to the following pharmaceutical compositions.
  • Exemplary Formulation 1 - Tablet The compound of the present invention in dry powder form can be mixed with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 0.3-30 mg tablets (each tablet contains 0.1-10 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 2 - Tablet The compound of the present invention in dry powder form can be mixed with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is formed into a 30-90 mg tablet (each tablet contains 10-30 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 3 - Tablet The compound of the present invention in dry powder form can be mixed with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 90-150 mg tablets (30-50 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 4-Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is formed into a 150-240 mg tablet (each tablet contains 50-80 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 5 - Tablets The compound of the invention in dry powder form can be mixed with the dried gel binder in a weight ratio of about 1:2. Hehe. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 240-270 mg tablets (each tablet contains 80-90 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 6-Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into a 270-450 mg tablet (each tablet contains 90-150 mg of active compound) in a tablet press.
  • Exemplary Formulation 7-Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 450-900 mg tablets (each tablet contains 150-300 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 8 - Capsules The compound of the invention in dry powder form can be combined with the starch diluent in a weight ratio of about 1:1. The mixture was filled into 250 mg capsules (each capsule containing 125 mg of active compound).
  • Exemplary Formulation 9-Liquid The compound of the present invention (125 mg) can be mixed with sucrose (1.75 g) and xanthan gum (4 mg), and the resulting mixture can be blended, passed through a No. 10 mesh U.S. sieve, and then It was mixed with an aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11:89, 50 mg) prepared in advance. Sodium benzoate (10 mg), flavor and color are diluted with water and added with stirring. Then, sufficient water can be added to give a total volume of 5 mL.
  • Exemplary Formulation 10 - Injection The compound of the invention may be dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of about 5 mg/mL.
  • the pharmaceutical composition provided by the present invention can be administered by a variety of routes including, but not limited to, oral administration, parenteral administration, inhalation administration, topical administration, rectal administration, nasal administration, oral administration, vaginal administration.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intra-articular administration, intra-arterial administration, intrasynovial administration, intrasternal administration. , intracerebroventricular administration, intralesional administration, and intracranial injection or infusion techniques.
  • an effective amount of a compound provided herein is administered.
  • the amount of compound actually administered can be determined by the physician. .
  • the compound provided herein is administered to a subject at risk of developing the condition, typically based on a physician's recommendation and administered under the supervision of a physician, at the dosage level as described above.
  • Subjects at risk of developing a particular condition typically include subjects with a family history of the condition, or those subjects that are particularly susceptible to developing the condition by genetic testing or screening.
  • long-term administration can also be administered chronically.
  • Long-term administration refers to administration of a compound or a pharmaceutical composition thereof for a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or can be continuously administered indefinitely, For example, the rest of the subject.
  • chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, for example, within a therapeutic window.
  • a pharmaceutical composition of the present invention can be further delivered using various methods of administration.
  • a pharmaceutical composition can be administered by bolus injection, for example, to rapidly increase the concentration of a compound in the blood to an effective level.
  • Bolus dose depends on the active group
  • the target systemic level of the fraction for example, the intramuscular or subcutaneous bolus dose allows for a slow release of the active component, while the bolus delivered directly to the vein (eg, by IV IV drip) enables more rapid delivery, resulting in an active group
  • the concentration in the blood is rapidly increased to an effective level.
  • the pharmaceutical composition can be administered in a continuous infusion form, for example, by IV intravenous drip to provide a steady state concentration of the active ingredient in the subject's body.
  • a bolus dose of the pharmaceutical composition can be administered first, followed by continued infusion.
  • Oral compositions can be in the form of a bulk liquid solution or suspension or bulk powder. More generally, however, the composition is provided in unit dosage form for ease of precise dosing.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active ingredient suitable to produce the desired therapeutic effect with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of the liquid compositions, or pills, tablets, capsules and the like in the case of solid compositions.
  • the compound will generally be a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being useful for forming the desired form of administration.
  • a carrier or excipient and a processing aid is provided in unit dosage form for ease of precise dosing.
  • a representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides from about 0.01 to about 20 mg/kg of a compound of the invention, each preferably providing from about 0.1 to about 10 mg/kg, especially from about 1 to about 5 mg/kg.
  • a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably about 0.1. To about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the injection dose level ranges from about 1 mg/kg/hr to at least 10 mg/kg/hr from about 1 to about 120 hours, especially 24 to 96 hours.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more can also be administered.
  • the maximum total dose cannot exceed about 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous vehicles as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • the solid form may include, for example, any of the following components, or a compound having similar properties: a binder, for example, microcrystalline cellulose, tragacanth or gelatin; an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silica; a sweetener such as sucrose or saccharin; or a flavoring agent such as mint, water Methyl salicylate or orange flavoring.
  • a binder for example, microcrystalline cellulose, tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel or corn star
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound will typically be a minor component, often from about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient.
  • the active component When formulated as an ointment, the active component is typically combined with a paraffin or water miscible ointment base.
  • the active ingredient can be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art and generally include other ingredients for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope of the invention.
  • transdermal administration can be accomplished using a reservoir or a porous membrane type, or a patch of a plurality of solid matrices.
  • compositions for oral administration, injection or topical administration are merely representative.
  • Other materials, as well as processing techniques and the like, are set forth in Section 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
  • the compounds of the invention may also be administered in sustained release form or from a sustained release delivery system.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention further relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation comprises water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are alpha-, beta- and gamma-cyclodextrins consisting of 6, 7 and 8 alpha-1,4-linked glucose units, respectively, optionally including one on the attached sugar moiety. Or a plurality of substituents including, but not limited to, methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substituted.
  • the cyclodextrin is a sulfoalkyl ether beta-cyclodextrin, eg, sulfobutylether beta-cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation comprises hexapropyl- ⁇ -cyclodextrin (eg, 10-50% in water).
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C).
  • the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
  • N-(3-methoxy-4-nitrophenyl)piperazine-1-carboxylic acid tert-butyl ester (reduced reaction) was sequentially added to a 50 mL one-necked flask under nitrogen atmosphere, and stirred at room temperature for 3 hours. The product was obtained as a yellow oily product.
  • LC-MS (APCI): m / z 238.2 (M + 1) +.
  • N-(4-nitrophenyl)piperazine-1-carboxylic acid tert-butyl ester (resources) was sequentially added to a 50 mL one-necked flask, and stirred at room temperature for 3 hours to obtain a yellow oily product.
  • N-(4-nitrophenyl)piperazine was sequentially added to a 50 mL one-necked flask, and stirred at room temperature for 3 hours to obtain a yellow oily product.
  • N-(4-nitrophenyl)piperazine (2.1 g, 9 mmol), acetonitrile (20 mL), Cs 2 CO 3 (5.85 g, 18 mmol) and p-toluenesulfonic acid d3-methyl
  • the ester (2.55 g, 13.5 mmol) was added to a single-necked flask, and the mixture was refluxed for 3 hr, cooled to room temperature, 50 mL of water was added, and extracted with dichloromethane. The organic phase was collected and dried to give a white solid product N-(4- Nitrophenyl)-4-d3-methyl-piperazine.
  • N-(4-Nitrophenyl)-4-d3-methyl-piperazine (0.75 g, 3 mmol)
  • iron powder (1 g, 18 mmol)
  • NH 4 Cl 160 mg, 3 mmol
  • the temperature of the reaction solution was returned to room temperature, the ethanol was spun, water was added, and the mixture was extracted with dichloromethane. The organic phase was collected and concentrated to give a yellow oily product 4- (4) -d3-methylpiperazin-1-yl)aniline.
  • the difference is that the target compound N-(3-(2-(4-(4-d3-methyl-2,2,3,3, 5,5,6,6-d8-piperazin-1-yl)phenylamino)thieno[3,2-d]pyrimidin-4-yloxy)phenyl)acrylamide (formula (3)).
  • the compounds obtained in the above examples were subjected to biological evaluation to determine their biological activities.
  • anti-proliferative activity in some of these compounds was screened in human A431 skin cancer cells and human NCI-H1975 and HCC827 lung cancer cell lines, and the activity was demonstrated to be in the range of ⁇ 20 nM.
  • the cytotoxic or growth inhibitory effects of the compounds on the tumor cells of interest were evaluated.
  • Test compounds were dissolved in DMSO to make a 20 mM stock solution. The solution was diluted in DMSO to a final concentration of 100 times the dilution. Dilute to 10 times the final concentration of the dilution solution with the buffer.
  • EGFR and EGFR [T790M/L858R] kinase assay After buffer preparation, the enzyme was mixed with different concentrations of pre-diluted compounds for 10 minutes, each double well. The corresponding substrate and ATP were added and reacted at room temperature for 20 minutes (in which a negative positive control was set). After the reaction is completed, the detection reagent is added, and after incubation at room temperature for 30 minutes, the machine is detected and data is collected. Data analysis and mapping according to Graphpad 5.0 software.
  • EGFR [d746-750] Kinase Assay: After the buffer was prepared, the mixed solution of the enzyme and the antibody was mixed with the different concentrations of the compound prepared by pre-dilution for 10 minutes, and the concentration was doubled. Kinase tracer 199 was added and incubated for 60 minutes at room temperature (where a negative positive control was set). After the reaction is completed, the machine is tested, data is collected, and analysis and mapping are performed.
  • A represents IC 50 ⁇ 1 nM
  • B represents 1 nM ⁇ IC 50 ⁇ 10 nM
  • C represents 10 nM ⁇ IC 50 ⁇ 100 nM
  • D represents 100 nM ⁇ IC 50 ⁇ 300 nM
  • E represents 300 nM ⁇ IC 50 ⁇ 1000 nM
  • F represents IC 50 > 1000 nM.
  • the compounds of the present invention exhibited relatively low inhibitory activity against WT EGFR associated with adverse effects (IC50 greater than 100 nM, more preferably greater than 300 nM, more preferably greater than 1000 nM), whereas EGFR L858R/T790M mutants ( It is resistant to commercially available EGFR inhibitors) and exhibits excellent inhibitory activity (IC50 is less than or equal to 100 nM, more preferably less than or equal to 10 nM, more preferably less than or equal to 1 nM, and selectivity multiples are greater than 20).
  • the compound of the present invention is an effective safe drug which can be used for an EGFR-mediated disease such as NSCLC, since it exhibits potent excellent inhibitory activity against EGFR mutants and has weak inhibitory activity against WT EGFR expressed in normal cells.
  • the inhibitory activities of the compounds of the present invention obtained in Examples 3 to 13 on BTK and JAK3 kinase were measured, respectively.
  • the test results are shown in Table 2 below.
  • Example number BTK IC 50 (nM) JAK3 IC 50 (nM) Example number BTK IC 50 (nM) JAK3 IC 50 (nM)
  • Example 3 ⁇ 100 ⁇ 100
  • Example 9 ⁇ 100 ⁇ 100
  • Example 4 ⁇ 100 ⁇ 100
  • Example 10 ⁇ 100 ⁇ 100
  • Example 5 ⁇ 100 ⁇ 100
  • Example 11 ⁇ 100 ⁇ 100
  • Example 6 ⁇ 100 ⁇ 100
  • Example 12 ⁇ 100 ⁇ 100
  • Example 7 ⁇ 100 ⁇ 100
  • Example 13 ⁇ 100 ⁇ 100
  • Example 8 ⁇ 100 ⁇ 100
  • the experimental results show that the compound of the present invention exhibits excellent inhibitory activity against BTK and JAK kinase (IC 50 ⁇ 100 nM).
  • the anti-proliferative activity of the compound of the present invention against two tumor cells cultured in vitro was examined by the MTS method.
  • the experimental results show that the compound of the present invention has an inhibitory effect on the in vitro proliferation of cancer cells cultured in vitro; wherein the inhibition of proliferation of lung cancer cells in vitro is stronger than that of skin cancer cells in vitro.
  • Cell lines skin cancer A431 (purchased from the American Standard Collection of Biological Products (ATCC)); lung cancer cells NCI-H1975 (purchased from the American Standard Collection of Biological Products (ATCC)) and HCC827 (purchased from the US Standard Collection of Biological Products) (ATCC)); both were cultured in RPMI1640 medium containing 10% fetal bovine serum, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin.
  • ATCC American Standard Collection of Biological Products
  • ATCC827 purchased from the US Standard Collection of Biological Products
  • Reagents and consumables RPMI-1640 (GIBCO, catalog number A10491-01); fetal bovine serum (GIBCO, catalog number 10099141); 0.25% trypsin-EDTA (GIBCO, catalog number 25200); penicillin-streptomycin; GIBCO, Cat. No. 15140-122); DMSO (Sigma, Cat. No. D2650); MTS Test Kit (Promega, Cat. No. G3581), 96-well plate (Coming, Cat. No. 3365).
  • test compound preparation The test compound was dissolved in DMSO to prepare a 20 mM mother liquor and stored at -20 °C. Dilute 3 times with a gradient of DMSO and the like, and dilute 10 times. The drug medium was diluted 4 times with the drug.
  • MTS cell viability assay 0.25% trypsin-EDTA digested logarithmic growth phase cells, inoculated with 150 ⁇ l in 96-well plates at an optimized density, and added to the medium 4 times after dilution for 4 hours, 50 ⁇ l/well (generally 10 Concentrations: 100, 33.3, 11.1, 3.70, 1.23, 0.412, 0.137, 0.0457, 0.0152, 0.00508 ⁇ M). A well of the same volume of 0.5% DMSO was added as a control. After the cells were cultured for 72 hours, MTS was assayed for cell viability.
  • HM61713 is a novel third-generation, orally active, irreversible, selective tyrosine kinase inhibitor (TKI) against epidermal growth factor receptor mutations.
  • TKI selective tyrosine kinase inhibitor
  • Example 12 >2000 ⁇ 20 50-60
  • Example 13 >2000 ⁇ 20 50-60 HM61713 >2000 ⁇ 20 50-60
  • the compounds of the present invention showed extremely low inhibitory activity against wild-type cells (A431) associated with adverse reactions (IC50 greater than 2000 nM), and showed excellent inhibition against EGFR L858R/T790M mutant cells (H1975). Activity (IC50 less than 20 nM).
  • the compounds of the invention are useful in the treatment of diseases mediated by EGFR.
  • Microsomal experiments human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate buffer Agent (pH 7.4).
  • phosphate buffer 100 mM, pH 7.4.
  • the pH of the solution was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
  • NADPH regeneration system containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride was prepared and placed on wet ice before use.
  • Formulation stop solution acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 ⁇ L of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 ⁇ L of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. 25057.5 ⁇ L of phosphate buffer (pH 7.4) was taken into a 50 mL centrifuge tube, and 812.5 ⁇ L of SD rat liver microsomes were added and mixed to obtain a liver microsome dilution having a protein concentration of 0.625 mg/mL.
  • the corresponding compound had a reaction concentration of 1 ⁇ M and a protein concentration of 0.5 mg/mL.
  • 100 ⁇ L of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper, and the reaction was terminated by vortexing for 3 min.
  • the plate was centrifuged at 5000 x g for 10 min at 4 °C.
  • 100 ⁇ L of the supernatant was taken into a 96-well plate to which 100 ⁇ L of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
  • Rats were fed a standard diet and given water. Fasting began 16 hours before the test.
  • the drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the eyelids at a time point of 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after administration.
  • Rats were briefly anesthetized after inhalation of ether, and 300 ⁇ L of blood samples were collected from the eyelids in test tubes. There was 30 ⁇ L of 1% heparin salt solution in the test tube. The tubes were dried overnight at 60 ° C before use. After the blood sample collection was completed at the last time point, the rats were anesthetized with ether and sacrificed.
  • Plasma samples were centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate plasma from red blood cells. Pipette 100 ⁇ L of plasma into a clean plastic centrifuge tube, indicating the name and time of the compound. Plasma was stored at -80 °C prior to analysis. The concentration of the compound of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.
  • Example 4 The experimental results of Example 4 and Example 6 are shown in Table 5 below, and the control compound is N-(3-(2-(2-methoxy-4-(4-methylpiperazin-1-yl)phenyl) Amino)thieno[3,2-d]pyrimidin-4-ylamino)phenyl)acrylamide (Compound A), the drug peak concentration Cmax is higher than the control compound A relative to the control compound, and further, Example 4 The area under the curve of the drug (AUC) was comparable to that of Compound A, and the area under the curve of the drug of Example 6 (AUC) was increased to 625.1 ng/mL*h, indicating that the compound of the present invention can be better absorbed in vivo.
  • the control compound is N-(3-(2-(2-methoxy-4-(4-methylpiperazin-1-yl)phenyl) Amino)thieno[3,2-d]pyrimidin-4-ylamino)phenyl)acrylamide (Compound A)
  • PK parameters Compound A PO data
  • Example 4 PO data
  • Example 6 PO data T max (h) 8.00 1.33 8.00 C max (ng/mL) 6.6 16.3 59.4 AUC (ng/mL*h) 76.3 78.0 625.1 MRT INF pred (h) ND ND ND T 1/2 (h) ND ND ND
  • Example 11 The results of the experiment of Example 11 are shown in Table 6 below.
  • the half-life of Example 11 in the present invention was prolonged by two times (4.54 hours and 9.83 hours, respectively) with respect to the control compound HM61713, and the metabolic stability was remarkably improved.
  • the compound of the present invention was The area under the curve (AUC) increased from 1101.4 ng/mL*h to 1505 ng/mL*h, indicating that the compound of the present invention can be better absorbed in vivo.

Abstract

La présente invention concerne un composé de pyrimidine condensée représenté par la formule (I), sa préparation et son utilisation. En particulier, la présente invention concerne un composé de pyrimidine condensée représenté par la formule (I) ou une forme cristalline, un sel pharmaceutiquement acceptable, un promédicament, un stéréoisomère, un variant isotopique, un hydrate ou un solvate de celui-ci, ainsi qu'une composition pharmaceutique le comprenant et son utilisation. Le composé de pyrimidine condensée et la composition comprenant le composé décrit dans la présente invention présentent un excellent effet d'inhibition contre les protéine kinases, et présentent des caractéristiques de paramètres pharmacocinétiques améliorées, qui permettent une plus grande concentration médicamenteuse de ces composés dans le corps d'un animal et améliorent l'efficacité et la tolérance des médicaments. FORMULE (I)
PCT/CN2016/103368 2015-12-02 2016-10-26 Composé de pyrimidine condensée, composition le comprenant et son utilisation WO2017092523A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201680056599.8A CN108137614B (zh) 2015-12-02 2016-10-26 一种稠合嘧啶化合物及包含该化合物的组合物及其用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510872331.9 2015-12-02
CN201510872331 2015-12-02

Publications (1)

Publication Number Publication Date
WO2017092523A1 true WO2017092523A1 (fr) 2017-06-08

Family

ID=58796232

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/103368 WO2017092523A1 (fr) 2015-12-02 2016-10-26 Composé de pyrimidine condensée, composition le comprenant et son utilisation

Country Status (2)

Country Link
CN (1) CN108137614B (fr)
WO (1) WO2017092523A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107954918A (zh) * 2017-11-30 2018-04-24 郑州泰基鸿诺医药股份有限公司 一种n-氘代甲基吲哚类化合物的合成方法
EP3356372A4 (fr) * 2015-10-30 2019-03-27 Hanmi Pharm. Co., Ltd. Nouveau procédé de préparation d'un composé thiénopyrimidine et intermédiaire utilisé à cet égard

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102947316A (zh) * 2010-06-23 2013-02-27 韩美科学株式会社 用于抑制酪氨酸激酶活性的新型稠合嘧啶衍生物

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3425676B2 (ja) * 1992-10-26 2003-07-14 武田薬品工業株式会社 縮合ピリミジン誘導体、その製造法および用途
DE10148883A1 (de) * 2001-10-04 2003-04-10 Merck Patent Gmbh Pyrimidinderivate
US9745321B2 (en) * 2013-09-30 2017-08-29 Shanghai Yingli Pharmaceutical Co., Ltd Fused pyrimidine compound, intermediate, preparation method therefor, and composition and application thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102947316A (zh) * 2010-06-23 2013-02-27 韩美科学株式会社 用于抑制酪氨酸激酶活性的新型稠合嘧啶衍生物

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KATSNELSON, A.;: "Heavy Drugs Draw Heavy Interest from Pharma Backers", NATURE MEDICINE, vol. 19, no. 6, 30 June 2013 (2013-06-30), pages 656, XP055387255 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3356372A4 (fr) * 2015-10-30 2019-03-27 Hanmi Pharm. Co., Ltd. Nouveau procédé de préparation d'un composé thiénopyrimidine et intermédiaire utilisé à cet égard
US10392403B2 (en) * 2015-10-30 2019-08-27 Hanmi Pharm. Co., Ltd. Process for preparing thienopyrimidine compound and intermediates used therein
CN107954918A (zh) * 2017-11-30 2018-04-24 郑州泰基鸿诺医药股份有限公司 一种n-氘代甲基吲哚类化合物的合成方法

Also Published As

Publication number Publication date
CN108137614B (zh) 2021-01-05
CN108137614A (zh) 2018-06-08

Similar Documents

Publication Publication Date Title
EP3345907B1 (fr) Composées pyrrolo[3,4-d]pyrimidines ou leurs sel
TWI752580B (zh) Kras突變蛋白抑制劑
EP3170825B1 (fr) Pyrrolopyridines utiles en tant qu'inhibiteurs de protéines kinases
KR102534962B1 (ko) 8,9-디하이드로이미다졸[1,2-a]피리미도[5,4-e]피리미딘-5(6H)-케톤계 화합물
JP5629324B2 (ja) ピロロ[2,3−d]ピリミジン化合物
JP5739446B2 (ja) ピロロ[2,3−d]ピリミジン化合物
CN110062754B (zh) 作为选择性Janus激酶抑制剂的氨基吡唑类化合物
RU2633694C2 (ru) Дейтерированный фениламинопиримидин и фармацевтическая композиция, содержащая такое соединение
CN110343090B (zh) 喹唑啉衍生物盐型晶型及制备方法和应用
WO2018086446A1 (fr) Composé quinazoline substitué ayant une capacité de pénétration de barrière hémato-encéphalique
WO2019154091A1 (fr) Composé de diaminopyrimidine substituée
WO2015058661A1 (fr) Inhibiteur de bcr-abl kinase et application correspondante
EP3653622B1 (fr) Composé aminopyrimidine, composition le comprenant et utilisation associée
WO2019034153A1 (fr) Composé chimique, composition pharmaceutique de celui-ci, utilisation et application correspondantes
CN110903283B (zh) 一种取代的喹唑啉类化合物、包含该化合物的药物组合物和该化合物的用途
EP4006031A1 (fr) Inhibiteur de jak2 sélectif de dihydro-pyrrolo-pyrimidine
WO2017190637A1 (fr) Composé de pyrimidine condensé pour inhiber l'activité de la protéine tyrosine kinase
CN110357858B (zh) 具有穿过血脑屏障能力的5取代二氟哌啶化合物
WO2017092523A1 (fr) Composé de pyrimidine condensée, composition le comprenant et son utilisation
WO2023109751A1 (fr) Dérivé de pyrimidine ou de pyridine et son utilisation médicinale
WO2019001307A1 (fr) Composé amide, composition le contenant, et utilisation associée
EP3932913A1 (fr) Inhibiteur de kinase pan-raf de type nouveau et son utilisation
CA3225872A1 (fr) Azetidinyl pyrimidines et utilisations associees
WO2019196619A1 (fr) Forme cristalline sous forme de sel dérivée de quinazoline, son procédé de préparation et son utilisation
EP2854780B1 (fr) Inhibiteurs du virus respiratoire syncytial

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16869833

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16869833

Country of ref document: EP

Kind code of ref document: A1