WO2019228330A1 - Substituted benzo[d]imidazole compound and pharmaceutical composition thereof - Google Patents

Substituted benzo[d]imidazole compound and pharmaceutical composition thereof Download PDF

Info

Publication number
WO2019228330A1
WO2019228330A1 PCT/CN2019/088749 CN2019088749W WO2019228330A1 WO 2019228330 A1 WO2019228330 A1 WO 2019228330A1 CN 2019088749 W CN2019088749 W CN 2019088749W WO 2019228330 A1 WO2019228330 A1 WO 2019228330A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
hydrogen
cancer
mmol
deuterium
Prior art date
Application number
PCT/CN2019/088749
Other languages
French (fr)
Chinese (zh)
Inventor
王义汉
任兴业
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
Publication of WO2019228330A1 publication Critical patent/WO2019228330A1/en

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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention belongs to the technical field of medicine, and particularly relates to a substituted benzo [d] imidazole compound, a pharmaceutical composition containing the compound, and use thereof. More specifically, the present invention relates to certain deuterated N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5-((4- (4-fluoro-1-isopropyl-2-methyl-1H-benzo [d] imidazol-6-yl) pyrimidin-2-yl) amino) phenyl) acrylamide, these deuterated compounds and The composition can be used to treat EGFR-related diseases, and these deuterium-substituted compounds have more excellent pharmacokinetic properties.
  • the epidermal growth factor receptor (ie, EGFR, ErbB-1 or HER1) is a member of the ErbB receptor family.
  • the ErbB receptor family includes four closely related members of the receptor tyrosine kinase: EGFR (ErbB-1), Her2 / c-neu (ErbB-2), Her3 (ErbB-3) and Her4 (ErbB-4).
  • EGFR is a cell surface receptor for members of the epidermal growth factor family (EGF family) of extracellular protein ligands. Mutations that affect EGFR expression or activity may cause cancer. EGFR has been reported to be dysregulated in most solid tumors such as lung, breast, and brain tumors. It is estimated that 30% of epithelial cancers are associated with mutations, amplifications or disorders of EGFR or family members.
  • gefitinib and erlotinib Treatment methods based on the inhibition of EGFR by antibody drugs or small molecule inhibitor drugs such as gefitinib and erlotinib have been developed. In the case of non-small cell lung cancer (NSCLC), gefitinib and erlotinib are beneficial for 10% to 40% of patients. However, after a period of treatment, acquired resistance to gefitinib or erlotinib has become a major clinical problem. Studies have confirmed that one of the main causes of drug resistance is due to a new mutation in T790M, which is the "gatekeeper" of EGFR. researchers then developed inhibitors targeting T790M, such as BIBW2992, and showed advantages in clinical trials.
  • T790M which is the "gatekeeper" of EGFR.
  • WO2018019204 discloses a compound T having the following structure, and the chemical name is N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5-((4- ( 4-fluoro-1-isopropyl-2-methyl-1H-benzo [d] imidazol-6-yl) pyrimidin-2-yl) amino) phenyl) acrylamide, compound T not only effectively inhibits the T790M mutation Moreover, it is highly selective for the T790M mutation compared to the wild type.
  • ADME absorption, distribution, metabolism, and / or excretion
  • the present invention discloses a new deuterium-substituted benzo [d] imidazole compound, as well as a composition and use thereof, which has better EGFR kinase inhibitory activity, and is resistant to drug-resistant mutations T790M, L858R, and Both of them are highly selective, have lower side effects, and better pharmacokinetic properties, and can be used to treat, prevent, and alleviate EGFR kinase-mediated diseases.
  • Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from hydrogen or deuterium;
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • the additional condition is that the above compounds contain at least one deuterium atom
  • the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient.
  • a compound of the invention is provided in the pharmaceutical composition in an effective amount.
  • a compound of the invention is provided in a therapeutically effective amount.
  • a compound of the invention is provided in a prophylactically effective amount.
  • the present invention provides a method for preparing a pharmaceutical composition as described above, comprising the steps of: mixing a pharmaceutically acceptable excipient with a compound of the present invention to form a pharmaceutical composition.
  • the present invention provides treatment of EGFR-caused cancers (including cancers caused by EGFR mutations, eg, cancers with T790M mutation, L858R mutation, and L858R / T790M double mutation) -related disorders in a subject in need thereof.
  • the cancer caused by the EGFR is selected from the group consisting of: non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, Gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma, etc.
  • the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In a specific embodiment, the compound is administered chronically.
  • deuterated refers to the replacement of one or more hydrogens in a compound or group with deuterium; deuteration may be mono-, di-, poly- or fully substituted.
  • deuteration may be mono-, di-, poly- or fully substituted.
  • deuteration may be mono-, di-, poly- or fully substituted.
  • deuteration may be mono-, di-, poly- or fully substituted.
  • deuterated and “one or more deuterated” are used interchangeably.
  • non-deuterated compound refers to a compound containing a deuterium atomic proportion not higher than the natural deuterium isotope content (0.015%).
  • pharmaceutically acceptable salt means, within the scope of sound medical judgment, suitable for contact with human and lower animal tissues without excessive toxicity, irritation, allergies, etc., and with reasonable benefits / dangers Proportion of those salts.
  • 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 salts derived from suitable inorganic and organic acids and bases.
  • the compounds of the invention may be in amorphous or crystalline form.
  • the compounds of the invention may exist in one or more crystalline forms. Accordingly, the invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • crystalline form refers to different arrangements of chemical drug molecules, which generally appear as the existing form of the drug substance in a solid state.
  • a drug can exist in multiple crystalline substance states, and different crystal forms of the same drug may have different dissolution and absorption in the body, which will affect the dissolution and release of the preparation.
  • crystalline form refers to different arrangements of chemical drug molecules, which generally appear as the existing form of the drug substance in a solid state.
  • a drug can exist in multiple crystalline substance states, and different crystal forms of the same drug may have different dissolution and absorption in the body, which will affect the dissolution and release of the preparation.
  • the term "subject” includes, but is not limited to: a human (ie, a male or female of any age group, for example, a pediatric subject (eg, infant, child, adolescent) or an adult subject (eg, Young adults, middle-aged adults or older adults)) and / or non-human animals, for example, mammals, for example, primates (for example, 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.
  • treatment includes the effect of a subject having a specific disease, disorder, or condition, which reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder, or condition. Or development of a condition ("therapeutic treatment”), and also includes effects that occur before a subject begins to suffer from a particular disease, disorder, or disease (“prophylactic treatment”).
  • an "effective amount" of a compound refers to an amount sufficient to elicit a biological response of interest.
  • the effective amount of a compound of the present invention can vary depending on factors such as the biological objective, the pharmacokinetics of the compound, the disease to be treated, the mode of administration, and the age of the subject. Health conditions and symptoms. Effective amounts include therapeutically and prophylactically effective amounts.
  • a "therapeutically effective amount" of a compound as used herein is an amount sufficient to provide a therapeutic benefit during the treatment of a disease, disorder, or condition, or one or more of which is associated with the disease, disorder, or condition. Symptoms are delayed or minimized.
  • a therapeutically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other therapies that provides a therapeutic benefit in the treatment of a disease, disorder, or condition.
  • the term "therapeutically effective amount” may include an amount that improves the overall treatment, reduces or avoids the symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
  • a prophylactically effective amount of a compound used herein is an amount sufficient to prevent a disease, disorder, or condition, or an amount sufficient to prevent one or more symptoms associated with a disease, disorder, or condition, or to prevent a disease , The number of recurrences of a disorder or condition.
  • a prophylactically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other agents that provides a preventative benefit in the prevention of a disease, disorder, or condition.
  • the term “prophylactically effective amount” may include an amount that improves overall prevention, or an amount that enhances the preventive efficacy of other preventive agents.
  • Combination and related terms refer to the simultaneous or sequential administration of a therapeutic agent of the invention.
  • a compound of the invention can be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously in a single unit dosage form with another therapeutic agent.
  • the present invention provides a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof:
  • Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from hydrogen or deuterium;
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • the additional condition is that the above compound contains at least one deuterium atom.
  • the deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably Ground is greater than 95%, and more preferably greater than 99%.
  • Y 1, Y 2, Y 3, Y 4, Y 5, Y 6, Y 7, Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen or trifluoromethyl
  • Y 1 is selected from hydrogen, deuterium, halogen, or trifluoromethyl
  • Y 2 is selected from hydrogen, deuterium, halogen, or trifluoromethyl
  • Y 3 is selected from hydrogen, deuterium, halogen, or trifluoromethyl
  • Y 9 is selected from the group consisting of hydrogen, deuterium, halogen or trifluoromethyl.
  • Y 1 is hydrogen, Y 1 is deuterium, Y 1 is halogen (F, Cl, Br, or I) or Y 1 is trifluoromethyl
  • Y 2 is hydrogen, Y 2 is deuterium, and Y 2 is Halogen (F, Cl, Br or I) or Y 2 is trifluoromethyl
  • Y 3 is hydrogen, Y 3 is deuterium, Y 3 is halogen (F, Cl, Br or I) or Y 3 is trifluoromethyl
  • Y 9 is hydrogen, Y 9 is deuterium, Y 9 is halogen (F, Cl, Br or I) or Y 9 is trifluoromethyl.
  • R 1, R 2, R 3, R 4 and R 5 are each independently selected from hydrogen or deuterium” includes R 1 is selected hydrogen or deuterium, R 2 is selected from hydrogen or deuterium, R 3 is selected from hydrogen or deuterium, and so on until R 5 is selected from hydrogen or deuterium. More particularly, R 1 is hydrogen comprising, R 1 is deuterium, R 2 is hydrogen, R 2 is deuterium, R 3 is hydrogen, R 3 is deuterium, and so on, until R 5 is hydrogen, deuterium, R 5 is Technical solutions.
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D" including X 1 X 3 is selected from CH 3 , CD 3 , CHD 2 or CH 2 D, X 2 is selected from CH 3 , CD 3 , CHD 2 or CH 2 D, X 3 is selected from CH 3 , CD 3 , CHD 2 or CH 2 D, and By analogy, until X 7 is selected from the technical schemes of CH 3 , CD 3 , CHD 2 or CH 2 D.
  • X 1 is CH 3
  • X 1 is CD 3
  • X 1 is CHD 2 or X 1 is CH 2 D
  • X 2 is CH 3
  • X 2 is CD 3
  • X 2 is CHD 2 or X 2 Is CH 2 D
  • X 3 is CH 3
  • X 3 is CD 3
  • X 3 is CHD 2 or X 3 is CH 2 D
  • X 7 is CHD 2 or X 7 is a technical solution of CH 2 D.
  • the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof, wherein Y 1 to Y 9 are each independently selected from hydrogen or deuterium, and R 1 to R 5 and X 1 to X 7 are as defined above, with the proviso that the above compound contains at least one deuterium atom.
  • the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof, wherein Y 3 to Y 9 are hydrogen, Y 1 and Y 2 are each independently selected from hydrogen or deuterium, R 1 to R 5 are each independently selected from hydrogen or deuterium, and X 1 to X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D.
  • Y 3 to Y 9 are hydrogen
  • Y 1 and Y 2 are each independently selected from hydrogen or deuterium
  • R 1 to R 5 are each independently selected from hydrogen or deuterium
  • X 1 to X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D.
  • Y 1 is hydrogen
  • X 1 and X 2 are the same.
  • X 1 and X 2 are each independently selected from CH 3 or CD 3 .
  • X 3 and X 4 are each independently selected from CH 3 or CD 3 .
  • X 6 and X 7 are each independently selected from CH 3 , CD 3 or CHD 2 .
  • X 5 is CH 3 .
  • R 2 , R 3 , R 4 and R 5 are hydrogen.
  • R 1 is deuterium
  • R 1 is hydrogen
  • Y 2 is deuterium
  • Y 2 is hydrogen
  • the compound has any one of the following structures, or a pharmaceutically acceptable salt thereof, but is not limited to the following structures:
  • the compounds of the invention may include one or more asymmetric centers, and thus may exist in multiple stereoisomeric forms, for example, enantiomeric and / or diastereomeric forms.
  • the compounds of the invention may be individual enantiomers, diastereomers or geometric isomers (such as cis and trans isomers), or may be in the form of a mixture of stereoisomers, This includes racemic mixtures and mixtures rich in one or more stereoisomers.
  • Isomers can be separated from a mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and formation and crystallization of chiral salts; or preferred isomers can be obtained by Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • organic compounds can form complexes with solvents that react in the solvent or precipitate or crystallize from the solvent. These complexes are called “solvates”. When the solvent is water, the complex is called a "hydrate”. The invention encompasses all solvates of the compounds of the invention.
  • solvate refers to the form of a compound or a salt thereof in combination with a solvent, usually formed by a solvolysis reaction. This physical association may include hydrogen bonding.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be able to be separated, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid.
  • the "solvate” includes a solvate in a solution state and a separable solvate. Representative solvates include hydrates, ethanolates, and methanolates.
  • hydrate refers to a compound that is combined with water. Generally, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, for example, hydrates of the compounds may be used on behalf of the general formula Rx H 2 O, where R is the compound, and x is a number greater than 0.
  • a given compound can form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, for example, hemihydrate (R0.5H 2 O )) and a multi-hydrate (x is greater than 1, e.g., dihydrate (R2H 2 O) and hexahydrate (R6H 2 O)).
  • the compounds of the invention may be in an amorphous or crystalline form (polymorphic form).
  • the compounds of the invention may exist in one or more crystalline forms. Accordingly, the invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • polymorph refers to a crystalline form (or a salt, hydrate, or solvate) of a compound in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, photoelectric properties, stability, and solubility. Recrystallization solvents, crystallization rates, storage temperatures, and other factors can lead to the predominance of a crystalline form.
  • Various polymorphs of the compounds can be prepared by crystallization under different conditions.
  • the present invention also includes isotopically-labeled compounds, which are equivalent to those described in formula (I), but one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number commonly found in nature.
  • isotopes that can be introduced into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl.
  • Compounds of the present invention containing the above-mentioned isotopes and / or other isotopes of other atoms, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or said prodrugs are all within the scope of the present invention.
  • Certain isotopically-labeled compounds of the invention, such as those incorporating radioisotopes such as 3 H and 14 C, can be used for drug and / or substrate tissue distribution assays. Thallium, i.e. 3 H and carbon-14, i.e. 14 C isotopes, are particularly preferred because they are easy to prepare and detect.
  • An isotope-labeled compound of the formula (I) of the present invention and a prodrug thereof can generally be prepared in such a manner that, when performing the processes disclosed in the following schemes and / or examples and preparation examples, non-isotope-labeled reagents are replaced with readily available isotopically-labeled reagents Labeled reagent.
  • prodrugs are also included in the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted into its active form with a medical effect in vivo by, for example, hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs 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, or drug, delivery: solubility, limitation, overcome, and use of prodrugs", Advanced Drug Delivery Reviews (1996) 19 (2) 115-130, each article This article is for reference.
  • a prodrug is any covalently bonded compound of the invention, and when such a prodrug is administered to a patient, it releases the parent compound in vivo.
  • Prodrugs are usually prepared by modifying functional groups, and the modification is performed in a manner such that the modification can be performed by routine manipulation or cleavage in vivo to produce the parent compound.
  • Prodrugs include, for example, compounds of the invention in which a hydroxyl, amino, or thiol group is bonded to an arbitrary group, and when administered to a patient, can cleave to form a hydroxyl, amino, or thiol group.
  • prodrugs include, but are not limited to, acetate, amide, formate / amide, and benzoate / amide derivatives of hydroxyl, thiol, and amino functional groups of the compound of formula (I).
  • a carboxylic acid -COOH
  • an ester such as methyl ester, ethyl ester, or the like can be used.
  • the esters themselves can be active and / or can be hydrolyzed under conditions in the human body.
  • Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those groups that readily break down in the human body to release the parent acid or its salt.
  • the compounds of the invention can be prepared using known organic synthesis techniques, and can be synthesized according to any of a number of possible synthetic pathways, such as those in the schemes below.
  • the reaction for preparing the compound of the present invention can be performed in a suitable solvent, and those skilled in the art of organic synthesis can easily select a solvent.
  • Suitable solvents may be substantially non-reactive with the starting material (reactant), intermediate, or product at the temperature at which the reaction is performed (eg, a temperature in the range of the solvent freezing temperature to the solvent boiling temperature).
  • a given reaction may be performed in one solvent or a mixture of more than one solvent.
  • the skilled person can select a solvent for a specific reaction step depending on the specific reaction step.
  • the preparation of the compounds of the invention may involve the protection and removal of different chemical groups. Those skilled in the art can easily determine whether protection and removal of protection are needed and the selection of an appropriate protecting group.
  • the chemical properties of protecting groups can be found, for example, in Wuts and Greene, Protective Groups, Organic Synthesis, 4th Edition, John Wiley & Sons: New Jersey, (2006), which is incorporated herein by reference in its entirety.
  • the reaction can be monitored according to any suitable method known in the art.
  • spectroscopic means such as nuclear magnetic resonance (NMR) spectroscopy (e.g., 1 H or 13 C), infrared (IR) spectroscopy, spectrophotometry (e.g., UV-visible light), mass spectrometry (MS)), or by chromatography Methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC) to monitor product formation.
  • NMR nuclear magnetic resonance
  • IR infrared
  • MS mass spectrometry
  • HPLC high performance liquid chromatography
  • TLC thin layer chromatography
  • compositions preparations and kits
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of an active ingredient.
  • the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient.
  • the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
  • a pharmaceutically acceptable excipient for use in the present invention refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compounds 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 (such as human serum albumin ), Buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substance, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene-embe
  • kits eg, pharmaceutical packaging.
  • the provided kits can include a compound of the invention, other therapeutic agents, and first and second containers (e.g., vials, ampoules, bottles, syringes, and / or dispersible packaging or other Suitable container).
  • the provided kit may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and / or other therapeutic agent.
  • a compound of the invention and other therapeutic agents are provided in a first container and a second container to form a unit dosage form.
  • parenteral administration as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intra-articular, intra-arterial, intra-synovial, and sternal administration , Cerebrospinal spinal membrane 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 .
  • a compound provided herein is administered to a subject at risk of developing the condition, typically based on and under the supervision of a physician, at a dosage level as described above.
  • Subjects at risk for developing a particular disorder typically include subjects with a family history of the disorder, or those who are determined by genetic testing or screening to be particularly sensitive to the development of the disorder.
  • the pharmaceutical compositions provided herein can also be administered chronically ("long-term administration").
  • Long-term administration refers to administration of a compound or a pharmaceutical composition thereof over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or the administration can be continued indefinitely, For example, the remainder of a subject's life.
  • chronic administration is intended to provide a constant level of the compound in the blood over a long period of time, for example, within a therapeutic window.
  • the pharmaceutical composition may be administered by bolus, for example, to rapidly increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic level of the active ingredient, for example, an intramuscular or subcutaneous bolus dose allows for a slow release of the active ingredient, whereas a bolus delivered directly to a vein (e.g., by IV intravenous drip) can be more Rapid delivery allows the concentration of the active ingredient in the blood to rise quickly to an effective level.
  • the pharmaceutical composition may be administered in the form of a continuous infusion, for example, by IV infusion, to provide a steady state concentration of the active ingredient in the subject's body.
  • the bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
  • Oral compositions can take the form of a liquid solution or suspension in bulk or a powder in bulk. However, more generally, to facilitate accurate dosing, the composition is provided in unit dosage form.
  • unit dosage form refers to a physically discrete unit suitable as a unit dose for human patients and other mammals, each unit containing a predetermined number of active substances and suitable pharmaceutical excipients suitable for producing the desired therapeutic effect.
  • Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or pills, tablets, capsules, etc. in the case of solid compositions.
  • the compound is usually a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), and the remainder is each useful for forming a desired administration form A carrier or excipient and processing aid.
  • a representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides about 0.01 to about 20 mg / kg of a compound of the invention, and preferred doses each provide about 0.1 to about 10 mg / kg, especially about 1 to about 5 mg / kg.
  • transdermal doses are usually selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, preferably about 0.1 To about 10% by weight, and more preferably about 0.5 to about 15% by weight.
  • the injection dose level ranges from about 0.1 mg / kg / hour to at least 10 mg / kg / hour.
  • a preloaded bolus of about 0.1 mg / kg to about 10 mg / kg or more can also be given.
  • the maximum total dose cannot exceed about 2 g / day.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous vehicles and 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 compounds having similar properties: a binder, such as microcrystalline cellulose, tragacanth, or gelatin; an excipient, such as starch or lactose, a disintegrant, For example, alginic acid, Primogel, or corn starch; lubricants, such as magnesium stearate; glidants, such as colloidal silicon dioxide; sweeteners, such as sucrose or saccharin; or flavoring agents, such as mint, water Methyl salicylate or orange flavor.
  • a binder such as microcrystalline cellulose, tragacanth, or gelatin
  • an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel, or corn starch
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • Transdermal compositions are typically formulated as a topical ointment or cream containing an active ingredient.
  • the active ingredient When formulated as an ointment, the active ingredient is typically combined with a paraffin or a water-miscible ointment base.
  • the active ingredient may 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 components for enhancing the 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 achieved using a reservoir or porous membrane type, or a variety of solid matrix patches.
  • compositions for oral administration, injection or topical administration are merely representative.
  • Other materials and processing techniques are described in Section 8 of Remington's Pharmaceuticals, Science, 17th Edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
  • the compounds of the invention may also be administered in a sustained release form or from a sustained release delivery system.
  • sustained-release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention also relates to a pharmaceutically acceptable formulation of a compound of the invention.
  • the formulation comprises water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are ⁇ -, ⁇ -, and ⁇ -cyclodextrin consisting of 6, 7, and 8 ⁇ -1,4-linked glucose units, respectively, which optionally include one on the linked sugar moiety Or more substituents, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substituted.
  • the cyclodextrin is a sulfoalkyl ether ⁇ -cyclodextrin, for example, a sulfobutyl ether ⁇ -cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation includes hexapropyl- ⁇ -cyclodextrin (eg, 10-50% in water).
  • the present invention provides a method for inhibiting a protein tyrosine kinase (such as EGFR kinase) or treating a disease (such as cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplant, viral disease, cardiovascular disease or Method of metabolic disease), which comprises the step of administering to a subject in need of treatment a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystalline form, prodrug thereof Or an isotope derivative, or a pharmaceutical composition according to the invention.
  • a protein tyrosine kinase such as EGFR kinase
  • a disease such as cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplant, viral disease, cardiovascular disease or Method of metabolic disease
  • the compounds of the invention are useful for treating cancer caused by EGFR.
  • the compounds are useful in the treatment of cancers caused by EGFR expressing EGFR mutants and in the treatment of cancers of EGFR refractory to RTKI therapy (eg, erlotinib or gefitinib).
  • the compounds of the present invention are inhibitors of at least one mutant of EGFR and are therefore suitable for use in treatment with one or more EGFR mutants (e.g., deletion mutations, activation mutations, resistance mutations, or combinations thereof, specific examples including T790M mutations, L858R mutation and L858R / T790M double mutation) activity related to one or more disorders.
  • EGFR mutants e.g., deletion mutations, activation mutations, resistance mutations, or combinations thereof, specific examples including T790M mutations, L858R mutation and L858R / T790M double mutation
  • the present invention provides a method for treating a mutant EGFR-mediated disorder, which comprises administering to a patient in need thereof a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, A solvate, a hydrate, a crystalline form, a prodrug or an isotope derivative, or a step of administering a pharmaceutical composition according to the present invention.
  • Cancers treatable by the compounds of the present invention include, but are not limited to, non-small cell lung cancer (NSCLS), small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, Gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma and other hyperproliferative diseases.
  • NSCS non-small cell lung cancer
  • small cell lung cancer small cell lung cancer
  • lung adenocarcinoma lung squamous cell carcinoma
  • pancreatic cancer breast cancer
  • prostate cancer liver cancer
  • skin cancer epithelial cell cancer
  • Gastrointestinal stromal tumor Gastrointestinal stromal tumor
  • leukemia histiocytic lymphoma
  • nasopharyngeal carcinoma other hyperproliferative diseases.
  • the compounds of the present invention are also useful for maintaining the
  • an effective amount of a compound of the present invention is usually administered in a single or multiple doses at an average daily dose of 0.01 to 50 mg of the compound per kg of the patient's body weight, preferably 0.1 to 25 mg of the compound per kg of the patient's body weight.
  • the compound of the present invention can be administered to the patient in need of such treatment at a daily dose range of about 1 mg to about 3500 mg per patient, preferably 10 mg to 1000 mg.
  • the daily dose per patient may be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000mg.
  • Administration can be one or more times daily, weekly (or several days apart), or on an intermittent schedule.
  • the compound can be administered one or more times per day on an weekly basis (e.g., every Monday), indefinitely or for several weeks, such as 4-10 weeks.
  • the compound can be administered daily for several days (e.g., 2-10 days), and then the compound is not administered for several days (e.g., 1-30 days), the cycle is repeated indefinitely, or a given number of times, such as 4-10 Cycles.
  • a compound of the invention may be administered daily for 5 days, then intermittently for 9 days, and then daily for 5 days, then intermittently for 9 days, and so on, the cycle may be repeated indefinitely or a total of 4-10 times.
  • EGFR-TKI eg, erlotinib or gefitinib
  • the components of the combination therapy can be administered at their monotherapy dose levels and schedules.
  • erlotinib has been administered orally at 150 mg per day for the treatment of non-small cell lung cancer, and it has been administered orally at 100 mg per day for pancreatic cancer.
  • gefitinib has been administered orally at 250 mg per day for the treatment of non-small cell lung cancer.
  • EGFR-TKI eg, erlotinib or gefitinib
  • the dosage level of one or both of its components is reduced compared to when used alone.
  • each reaction is usually performed in an inert solvent at room temperature to reflux temperature (for example, 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.
  • the gray solid (220 mg, 0.48 mmol) obtained above was dispersed uniformly by adding acetonitrile (5 mL), and then N 1 , N 1 , N 2 -trimethylethane-1,2-diamine (59 mg, 0.57 mmol), and potassium carbonate were added. (132 mg, 0.96 mmol), heated to reflux at 90 ° C for 2 hours. The reaction solution was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness to obtain Compound 5 as a red oil (126 mg).
  • the red oily compound 5 (126 mg) was dissolved in a mixed solution of ethanol-water (4 mL + 1 mL), and reduced iron powder (106 mg) and ammonium chloride (34 mg) were added, and the mixture was heated under reflux at 90 ° C for 5 hours to react. The reaction was cooled to room temperature, the insoluble solid was removed by filtration, and the filtrate was evaporated to dryness. The obtained oil was dissolved by adding 10 mL of DCM, 5 mL of a saturated aqueous sodium hydrogen carbonate solution was added, and the solution was cooled in an ice bath.
  • step 1 in Example 1 is replaced by the following steps:
  • Trifluoroacetic acid (10 mL) was added to a solution of compound 21 (1.70 g, 2.79 mmol) in dichloromethane (20 mL), and the reaction was stirred in a greenhouse for 1 hr.
  • the reaction solution was concentrated under reduced pressure to remove the reaction solution, and the pH was adjusted to be alkaline with a saturated sodium bicarbonate solution.
  • Water (30 mL) was added and stirred overnight.
  • the solid was filtered, the solid was washed with water, and then washed with cold ethanol (3 mL x 2), and dried under vacuum at 55 ° C to obtain 1.1 g of a red solid, yield: 77.44%.
  • LC-MS (APCI): m / z 509.2 (M + 1) + .
  • test compound was dissolved in DMSO to prepare a 20 mM mother liquor. Then, a 3-fold dilution in DMSO was performed ten times. When adding medicine, dilute it with buffer 10 times.
  • Detection of WT EGFR and EGFR [L858R / T790M] kinase In 5x kinase buffer A, mix WT EGFR or EGFR [L858R / T790M] kinase with a compound of different concentration prepared in a pre-diluted form for 10 minutes, and duplicate the wells at each concentration . Add the corresponding substrate and ATP, and react at room temperature for 20 minutes (wherein a negative positive control is set: a negative is a blank control and a positive is erlotinib). After the reaction, add detection reagents (reagents in the HTRF Kinase TK kit), and incubate for 30 minutes at room temperature.
  • detection reagents reagents in the HTRF Kinase TK kit
  • the compound of the present invention was tested in the above kinase inhibition experiment, and it was found that the compound of the present invention has a potent activity on EGFR [L858R / T790M] and an excellent selectivity over WT EGFR compared to the non-deuterated compound T.
  • the results for representative example compounds are summarized in Table 1 below.
  • the anti-proliferative activity of the compound of the present invention on three tumor cells cultured in vitro was detected 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 inhibitory effect on the proliferation of lung cancer cells in vitro is stronger than the inhibitory effect on the proliferation of skin cancer cells in vitro.
  • Skin cancer cells A431 purchased from the American Standard Biological Collection (ATCC)
  • lung cancer cells NCI-H1975 purchased from the American Standard Biological Collection (ATCC)
  • HCC827 purchased from the American Standard Biological Collection (ATCC))
  • All were cultured in RPMI1640 medium containing 10% fetal bovine serum, 100 U / ml penicillin, and 100 ⁇ g / ml streptomycin.
  • RPMI-1640 (GIBCO, catalog number A10491-01); fetal bovine serum (GIBCO, catalog number 10099141); 0.25% trypsin-EDTA (GIBCO, catalog number 25200); penicillin-streptomycin, liquid (GIBCO, catalog number 15140-122); DMSO (Sigma, catalog number D2650); MTS test kit (Promega, catalog number G3581), 96-well plate (Corning, catalog number 3365).
  • test compound preparation The test compound was dissolved in DMSO to prepare a 20 mM mother liquor, and stored at -20 ° C. When using, dilute with DMSO and other gradients 3 times, 10 times. At the time of dosing, it was diluted 4 times with cell culture medium RPMI-1640.
  • MTS cell viability test 0.25% trypsin-EDTA digested cells in logarithmic growth phase, inoculated 150 ⁇ l in a 96-well plate according to the optimized density, 24 hours after adding the medium to dilute the compound 4 times, 50 ⁇ l / well Concentration: 100, 33.3, 11.1, 3.70, 1.23, 0.412, 0.137, 0.0457, 0.0152, 0.00508 ⁇ M). As a control, the same volume of 0.5% DMSO was added. After the cells were cultured for 72 hours, MTS was used to detect cell viability.
  • adherent cells discard the culture medium, and add a mixed solution containing 20 ⁇ L MTS and 100 ⁇ L culture medium to each well. Put it in the incubator and continue to incubate for 1-4 hours to detect OD490, and use the OD650 value as a reference. Using GraphPad Prism software to make dose-response curves and calculate IC 50.
  • the compound of the present invention was tested in the above cytotoxicity experiment, and it was found that the compound of the present invention has strong activity on lung cancer cells NCI-H1975 and HCC827 and an excellent choice better than skin cancer cell A431 compared with the non-deuterated compound T. Sex.
  • the results of representative example compounds are summarized in Table 2 below.
  • Microsomal experiments human liver microsomes: 0.5mg / mL, Xenotech; rat liver microsomes: 0.5mg / mL, Xenotech; coenzyme (NADPH / NADH): 1mM, Sigma Life Science; magnesium chloride: 5mM, 100mM phosphate buffered Agent (pH 7.4).
  • Preparation of the stock solution A certain amount of powder of the example compound and the reference compound was precisely weighed, and dissolved to 5 mM with DMSO, respectively.
  • phosphate buffer solution 100 mM, pH 7.4.
  • NADPH regeneration system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U / mL G-6-PD, 3.3mM magnesium chloride), and place on wet ice before use.
  • stop solution an acetonitrile solution containing 50ng / mL propranolol hydrochloride and 200ng / mL tolbutamide (internal standard). Take 25057.5 ⁇ L of phosphate buffer solution (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.
  • phosphate buffer solution pH 7.4
  • Determination of metabolic stability 300 ⁇ L of pre-cooled stop solution was added to each well of a 96-well deep well plate, and placed on ice as a stop plate. Place the 96-well incubation plate and NADPH regeneration system in a 37 ° C water bath, shake at 100 rpm, and pre-incubate for 5 minutes. Take 80 ⁇ L of the incubation solution from each well of the incubation plate and add it to the termination plate, mix well, and add 20 ⁇ L of the NADPH regeneration system solution as a 0min sample. Add 80 ⁇ L of NADPH regeneration system solution to each well of the incubation plate, start the reaction, and start timing.
  • the reaction concentration of the corresponding compound was 1 ⁇ M, and the protein concentration was 0.5 mg / mL.
  • 100 ⁇ L of each reaction solution was taken, added to the stop plate, and vortexed for 3 minutes to stop the reaction.
  • the stop plate was centrifuged at 5000 ⁇ g for 10 min at 4 ° C. Take 100 ⁇ L of the supernatant into a 96-well plate pre-added with 100 ⁇ L of distilled water, mix well, and use LC-MS / MS for sample analysis.
  • the compounds of the present invention and their non-deuterated compounds were tested and compared simultaneously to evaluate their metabolic stability in human and rat liver microsomes.
  • Undeuterated compound T was used as a reference.
  • the compounds of the present invention can significantly improve metabolic stability by comparison with the undeuterated compound T.
  • Table 3 The results for representative example compounds are summarized in Table 3 below.
  • Rats were raised on a standard diet and given water. Fasting began 16 hours before the test.
  • the drug was dissolved with PEG400 and dimethyl sulfoxide.
  • Orbital blood was collected at 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 anesthetized briefly after inhaling ether, and 300 ⁇ L of blood samples were collected in test tubes from the orbit.
  • the test tube contained 30 ⁇ L of a 1% heparin salt solution. Before use, test tubes were dried at 60 ° C overnight. After blood samples were collected at the last time point, rats were sacrificed after ether anesthesia.
  • the blood sample was centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate the plasma from the red blood cells. Pipette 100 ⁇ L of plasma into a clean plastic centrifuge tube with the name and time point of the compound. Plasma was stored at -80 ° C before analysis. LC-MS / MS was used to determine the concentration of a compound of the invention in plasma. Pharmacokinetic parameters were calculated based on the blood drug concentration of each animal at different time points.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Transplantation (AREA)
  • Virology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Communicable Diseases (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides a substituted benzo[d]imidazole compound and a pharmaceutical composition thereof, and uses thereof; the benzo[d]imidazole compound is a compound represented by the formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer, or isotopic variant of the described compound. The compound and composition of the present invention can be used for methods for treating illnesses related to cancer caused by EGFR (comprising cancer caused by EGFR mutations, for example, cancer having a T790M mutation, an L858R mutation, and a L858R/T790M double mutation).

Description

取代的苯并[d]咪唑类化合物及其药物组合物Substituted benzo [d] imidazole compounds and pharmaceutical compositions thereof 技术领域Technical field
本发明属于医药技术领域,尤其涉及一种取代的苯并[d]咪唑类化合物及包含该化合物的药物组合物及其用途。更具体而言,本发明涉及某些氘取代的N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺,这些氘取代的化合物及其组合物可用于治疗与EGFR相关的疾病,且这些氘取代的化合物具有更优良的药代动力学性质。The invention belongs to the technical field of medicine, and particularly relates to a substituted benzo [d] imidazole compound, a pharmaceutical composition containing the compound, and use thereof. More specifically, the present invention relates to certain deuterated N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5-((4- (4-fluoro-1-isopropyl-2-methyl-1H-benzo [d] imidazol-6-yl) pyrimidin-2-yl) amino) phenyl) acrylamide, these deuterated compounds and The composition can be used to treat EGFR-related diseases, and these deuterium-substituted compounds have more excellent pharmacokinetic properties.
背景技术Background technique
表皮生长因子受体(即EGFR、ErbB-1或HER1)是ErbB受体家族的成员之一,ErbB受体家族包括四种密切相关的受体酪氨酸激酶成员:EGFR(ErbB-1)、Her2/c-neu(ErbB-2)、Her3(ErbB-3)和Her4(ErbB-4)。EGFR是胞外蛋白配体的表皮生长因子家族(EGF家族)成员的细胞表面受体。影响EGFR表达或活性的突变可能导致癌症。据报道,在大多数实体瘤如肺癌、乳腺癌和脑瘤中,EGFR处于失调状态。据估计,30%的上皮癌与EGFR或家族成员的突变、扩增或失调有关联。The epidermal growth factor receptor (ie, EGFR, ErbB-1 or HER1) is a member of the ErbB receptor family. The ErbB receptor family includes four closely related members of the receptor tyrosine kinase: EGFR (ErbB-1), Her2 / c-neu (ErbB-2), Her3 (ErbB-3) and Her4 (ErbB-4). EGFR is a cell surface receptor for members of the epidermal growth factor family (EGF family) of extracellular protein ligands. Mutations that affect EGFR expression or activity may cause cancer. EGFR has been reported to be dysregulated in most solid tumors such as lung, breast, and brain tumors. It is estimated that 30% of epithelial cancers are associated with mutations, amplifications or disorders of EGFR or family members.
已经研发出基于通过抗体药或小分子抑制剂药物(例如吉非替尼和厄洛替尼)抑制EGFR的治疗方法。在非小细胞肺癌(NSCLC)的情况下,吉非替尼和厄洛替尼对10%~40%的病人有益处。然而,治疗一段时间后,对吉非替尼或厄洛替尼的获得性耐药性成为主要的临床问题。研究证实,产生耐药性的一个主要原因是由于T790M的新突变,其为EGFR的“门卫”。然后,研发人员又研发了针对T790M的抑制剂,如BIBW2992,并在临床试验中表现出优势。但是,这些以EGFR的T790M突变为靶标的抑制剂对野生型EGFR也具有相当的抑制活性,这导致的严重的毒副作用限制了其临床应用。所以,有必要进一步研发出更多仅靶向突变型而非野生型的EGFR的选择性抑制剂的有效类型。Treatment methods based on the inhibition of EGFR by antibody drugs or small molecule inhibitor drugs such as gefitinib and erlotinib have been developed. In the case of non-small cell lung cancer (NSCLC), gefitinib and erlotinib are beneficial for 10% to 40% of patients. However, after a period of treatment, acquired resistance to gefitinib or erlotinib has become a major clinical problem. Studies have confirmed that one of the main causes of drug resistance is due to a new mutation in T790M, which is the "gatekeeper" of EGFR. Researchers then developed inhibitors targeting T790M, such as BIBW2992, and showed advantages in clinical trials. However, these inhibitors targeting the T790M mutation of EGFR also have considerable inhibitory activity against wild-type EGFR, which results in serious toxic side effects that limit its clinical application. Therefore, it is necessary to further develop more effective types of selective inhibitors of EGFR that target only mutant rather than wild type.
WO2018019204公开了如下结构的化合物T,化学名称为N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺,化合物T不仅能有效抑制T790M突变,而且相对于野生型而言对T790M突变具有高选择性。WO2018019204 discloses a compound T having the following structure, and the chemical name is N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5-((4- ( 4-fluoro-1-isopropyl-2-methyl-1H-benzo [d] imidazol-6-yl) pyrimidin-2-yl) amino) phenyl) acrylamide, compound T not only effectively inhibits the T790M mutation Moreover, it is highly selective for the T790M mutation compared to the wild type.
Figure PCTCN2019088749-appb-000001
Figure PCTCN2019088749-appb-000001
已知较差的吸收、分布、代谢和/或排泄(ADME)性质是导致许多候选药物临床试验失败的主要原因。当前上市的许多药物也由于较差的ADME性质限制了它们的应用范围。药物的快速代谢会导致许多本来可以高效治疗疾病的药物由于过快的从体内代谢清除掉而难以成药。频繁或高剂量服药虽然有可能解决药物快速清除的问题,但该方法会带来诸如病人依从性差、高剂量服药引起的副作用及治疗成本上升等问题。另外,快速代谢的药物也可能会使患者暴露于不良的毒性或反应性代谢物中。It is known that poor absorption, distribution, metabolism, and / or excretion (ADME) properties are the main reasons for the failure of clinical trials for many drug candidates. Many drugs currently on the market also limit their scope of application due to poor ADME properties. The rapid metabolism of drugs will cause many drugs that could be highly effective in treating diseases to be difficult to make because they are metabolized from the body too quickly. Although frequent or high-dose medication may solve the problem of rapid drug removal, this method will bring problems such as poor patient compliance, side effects caused by high-dose medication, and rising treatment costs. In addition, rapidly metabolizing drugs may expose patients to adverse toxic or reactive metabolites.
因此发现具有治疗EGFR相关的疾病且具有很好的口服生物利用度且有成药性的新型化合物还是具有挑战性的工作。因此,本领域仍需开发对适用作治疗剂的突变EGFR介导疾病具有选择性抑制活性和/或更好地药效学/药代动力学的化合物,本发明提供了这样的化合物。Therefore, it is still a challenging task to find new compounds that have the ability to treat EGFR-related diseases, have good oral bioavailability, and are druggable. Therefore, there remains a need in the art to develop compounds that have selective inhibitory activity and / or better pharmacodynamics / pharmacokinetics of mutant EGFR-mediated diseases suitable for use as therapeutic agents, and the present invention provides such compounds.
发明概述Summary of invention
针对以上技术问题,本发明公开了一种新的氘取代的苯并[d]咪唑类化合物及其组合物和用途,其具有更好的EGFR激酶抑制活性,以及对于耐药突变T790M、L858R及其二者的高选择性,同时具有更低的副作用、更好地药代动力学性能,可用于治疗、预防以及缓解EGFR激酶介导的疾病。In view of the above technical problems, the present invention discloses a new deuterium-substituted benzo [d] imidazole compound, as well as a composition and use thereof, which has better EGFR kinase inhibitory activity, and is resistant to drug-resistant mutations T790M, L858R, and Both of them are highly selective, have lower side effects, and better pharmacokinetic properties, and can be used to treat, prevent, and alleviate EGFR kinase-mediated diseases.
对此,本发明采用以下技术方案:In this regard, the present invention adopts the following technical solutions:
本发明的第一方面中,提供了式(I)化合物:In a first aspect of the invention, a compound of formula (I) is provided:
Figure PCTCN2019088749-appb-000002
Figure PCTCN2019088749-appb-000002
其中,among them,
Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7、Y 8和Y 9各自独立地选自氢、氘、卤素或三氟甲基; Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
R 1、R 2、R 3、R 4和R 5各自独立地选自氢或氘; R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from hydrogen or deuterium;
X 1、X 2、X 3、X 4、X 5、X 6和X 7各自独立地选自CH 3、CD 3、CHD 2或CH 2D; X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
附加条件是,上述化合物至少含有一个氘原子;The additional condition is that the above compounds contain at least one deuterium atom;
或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体。Or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, polymorph, stereoisomer or isotope variant thereof.
在另一方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物。在具体实施方案中,本发明化合物以有效量提供在所述药物组合物中。在具体实施方案中,本发明化合物以治疗有效量提供。在具体实施方案中,本发明化合物以预防有效量提供。In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient. In a specific embodiment, a compound of the invention is provided in the pharmaceutical composition in an effective amount. In a specific embodiment, a compound of the invention is provided in a therapeutically effective amount. In a specific embodiment, a compound of the invention is provided in a prophylactically effective amount.
在另一方面,本发明提供了一种如上所述的药物组合物的制备方法,包括以下步骤:将药学上可接受的赋形剂与本发明化合物进行混合,从而形成药物组合物。In another aspect, the present invention provides a method for preparing a pharmaceutical composition as described above, comprising the steps of: mixing a pharmaceutically acceptable excipient with a compound of the present invention to form a pharmaceutical composition.
在另一个方面,本发明提供了在需要其的受试者中治疗EGFR导致的癌症(包括EGFR突变导致的癌症,例如,带有T790M突变、L858R突变和L858R/T790M双突变的癌症)相关病症的方法,所述方法包括:给予受试者有效量的本发明化合物。在具体实施方案中,所述EGFR导致的癌症选自:非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、前列腺癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌等。在具体实施方案中,口服、皮下、静脉内或肌肉内给药所述化合物。在具体实施方案中,长期给药所述化合物。In another aspect, the present invention provides treatment of EGFR-caused cancers (including cancers caused by EGFR mutations, eg, cancers with T790M mutation, L858R mutation, and L858R / T790M double mutation) -related disorders in a subject in need thereof. A method of: administering to a subject an effective amount of a compound of the invention. In a specific embodiment, the cancer caused by the EGFR is selected from the group consisting of: non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, Gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma, etc. In specific embodiments, the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In a specific embodiment, the compound is administered chronically.
由随后的具体实施方式、实施例和权利要求,本发明的其它目的和优点将对于本领域技术人员显而易见。Other objects and advantages of the invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
定义definition
本文中,如无特别说明,“氘代”指化合物或基团中的一个或多个氢被氘所取代;氘代可以是一取代、二取代、多取代或全取代。术语“一个或多个氘代的”与“一次或多次氘代”可互换使用。As used herein, unless otherwise specified, "deuterated" refers to the replacement of one or more hydrogens in a compound or group with deuterium; deuteration may be mono-, di-, poly- or fully substituted. The terms "one or more deuterated" and "one or more deuterated" are used interchangeably.
本文中,如无特别说明,“非氘代的化合物”是指含氘原子比例不高于天然氘同位素含量(0.015%)的化合物。Herein, unless otherwise specified, "non-deuterated compound" refers to a compound containing a deuterium atomic proportion not higher than the natural deuterium isotope content (0.015%).
术语“药学上可接受的盐”是指,在可靠的医学判断范围内,适合与人和低等动物的组织接触而没有过度毒性、刺激性、变态反应等等,并且与合理的益处/危险比例相称的那些盐。药学上可接受的盐在本领域是众所周知的。例如,Berge等人在J.Pharmaceutical Sciences(1977)66:1-19中详细描述的药学上可接受的盐。本发明化合物的药学上可接受的盐包括衍生自合适无机和有机酸和碱的盐。The term "pharmaceutically acceptable salt" means, within the scope of sound medical judgment, suitable for contact with human and lower animal tissues without excessive toxicity, irritation, allergies, etc., and with reasonable benefits / dangers Proportion of those salts. 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 salts derived from suitable inorganic and organic acids and bases.
本发明化合物可以是无定形或结晶形式。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“晶型”是指化学药物分子的不同排列方式,一般表现为药物原料在固体状态下的存在形式。一种药物可以多种晶型物质状态存在,同一种药物的不同晶型,在体内的溶解和吸收可能不同,从而会对制剂的溶出和释放产生影响。The compounds of the invention may be in amorphous or crystalline form. In addition, the compounds of the invention may exist in one or more crystalline forms. Accordingly, the invention includes within its scope all amorphous or crystalline forms of the compounds of the invention. The term "crystalline form" refers to different arrangements of chemical drug molecules, which generally appear as the existing form of the drug substance in a solid state. A drug can exist in multiple crystalline substance states, and different crystal forms of the same drug may have different dissolution and absorption in the body, which will affect the dissolution and release of the preparation.
术语“晶型”是指化学药物分子的不同排列方式,一般表现为药物原料在固体状态下的存在形式。一种药物可以多种晶型物质状态存在,同一种药物的不同晶型,在体内的溶解和吸收可能不同,从而会对制剂的溶出和释放产生影响。The term "crystalline form" refers to different arrangements of chemical drug molecules, which generally appear as the existing form of the drug substance in a solid state. A drug can exist in multiple crystalline substance states, and different crystal forms of the same drug may have different dissolution and absorption in the body, which will affect the dissolution and release of the preparation.
如本文所用,术语“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在另一些实施方案中,受试者是非人动物。As used herein, the term "subject" includes, but is not limited to: a human (ie, a male or female of any age group, for example, a pediatric subject (eg, infant, child, adolescent) or an adult subject (eg, Young adults, middle-aged adults or older adults)) and / or non-human animals, for example, mammals, for example, primates (for example, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses , Sheep, goats, rodents, cats and / or dogs. In some embodiments, the subject is a human. In other embodiments, the subject is a non-human animal.
“疾病”、“障碍”和“病症”在本文中可以互换地使用。"Disease", "disorder" and "disorder" are used interchangeably herein.
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括受试者开始患有具体疾病、障碍或疾病之前发生的作用(“预防性治疗”)。As used herein, unless otherwise stated, the term "treatment" includes the effect of a subject having a specific disease, disorder, or condition, which reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder, or condition. Or development of a condition ("therapeutic treatment"), and also includes effects that occur before a subject begins to suffer from a particular disease, disorder, or disease ("prophylactic treatment").
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药物动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗和预防性治疗有效量。Generally, an "effective amount" of a compound refers to an amount sufficient to elicit a biological response of interest. As will be understood by one of ordinary skill in the art, the effective amount of a compound of the present invention can vary depending on factors such as the biological objective, the pharmacokinetics of the compound, the disease to be treated, the mode of administration, and the age of the subject. Health conditions and symptoms. Effective amounts include therapeutically and prophylactically effective amounts.
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗有益处的数量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化。化合物的治疗有效量是指单独使用或与其他疗法联用的治疗剂的数量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其他治疗剂的治疗效能的数量。Unless stated otherwise, a "therapeutically effective amount" of a compound as used herein is an amount sufficient to provide a therapeutic benefit during the treatment of a disease, disorder, or condition, or one or more of which is associated with the disease, disorder, or condition. Symptoms are delayed or minimized. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other therapies that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" may include an amount that improves the overall treatment, reduces or avoids the symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的数量,或足以预防与疾病、障碍或病症有关的一或多种症状的数量,或防止疾病、障碍或病症复发的数量。化合物的预防有效量是指单独使用或与其它药剂联用的治疗剂的数量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的数量,或增强其它预防药剂的预防效能的数量。Unless otherwise stated, a "prophylactically effective amount" of a compound used herein is an amount sufficient to prevent a disease, disorder, or condition, or an amount sufficient to prevent one or more symptoms associated with a disease, disorder, or condition, or to prevent a disease , The number of recurrences of a disorder or condition. A prophylactically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other agents that provides a preventative benefit in the prevention of a disease, disorder, or condition. The term "prophylactically effective amount" may include an amount that improves overall prevention, or an amount that enhances the preventive efficacy of other preventive agents.
“组合”以及相关术语是指同时或依次给药本发明的治疗剂。例如,本发明化合物可以与另一治疗剂以分开的单位剂型同时或依次给药,或与另一治疗剂一起呈单一单位剂型同时给药。"Combination" and related terms refer to the simultaneous or sequential administration of a therapeutic agent of the invention. For example, a compound of the invention can be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously in a single unit dosage form with another therapeutic agent.
具体实施方式Detailed ways
化合物Compound
本发明提供式(I)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体:The present invention provides a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof:
Figure PCTCN2019088749-appb-000003
Figure PCTCN2019088749-appb-000003
其中,among them,
Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7、Y 8和Y 9各自独立地选自氢、氘、卤素或三氟甲基; Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
R 1、R 2、R 3、R 4和R 5各自独立地选自氢或氘; R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from hydrogen or deuterium;
X 1、X 2、X 3、X 4、X 5、X 6和X 7各自独立地选自CH 3、CD 3、CHD 2或CH 2D; X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
附加条件是,上述化合物至少含有一个氘原子。The additional condition is that the above compound contains at least one deuterium atom.
作为本发明的优选实施方案,氘在氘代位置的氘同位素含量至少是大于天然氘同位素含量0.015%,较佳地大于30%,更佳地大于50%,更佳地大于75%,更佳地大于95%,更佳地大于99%。As a preferred embodiment of the present invention, the deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably Ground is greater than 95%, and more preferably greater than 99%.
在具体实施方案中,“Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7、Y 8和Y 9各自独立地选自氢、氘、卤素或三氟甲基”包括Y 1选自氢、氘、卤素或三氟甲基,Y 2选自氢、氘、卤素或三氟甲基,Y 3选自氢、氘、卤素或三氟甲基,以此类推,直至Y 9选自氢、氘、卤素或三氟甲基的技术方案。更具体地,包括Y 1为氢、Y 1为氘、Y 1为卤素(F、Cl、Br或I)或Y 1为三氟甲基,Y 2为氢、Y 2为氘、Y 2为卤素(F、Cl、Br或I)或Y 2为三氟甲基,Y 3为氢、Y 3为氘、Y 3为卤素(F、Cl、Br或I)或Y 3为三氟甲基,以此类推,直至Y 9为氢、Y 9为氘、Y 9为卤素(F、Cl、Br或I)或Y 9为三氟甲基的技术方案。 In a specific embodiment, "Y 1, Y 2, Y 3, Y 4, Y 5, Y 6, Y 7, Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen or trifluoromethyl" comprising Y 1 is selected from hydrogen, deuterium, halogen, or trifluoromethyl, Y 2 is selected from hydrogen, deuterium, halogen, or trifluoromethyl, Y 3 is selected from hydrogen, deuterium, halogen, or trifluoromethyl, and so on, until Y 9 is selected from the group consisting of hydrogen, deuterium, halogen or trifluoromethyl. More specifically, Y 1 is hydrogen, Y 1 is deuterium, Y 1 is halogen (F, Cl, Br, or I) or Y 1 is trifluoromethyl, Y 2 is hydrogen, Y 2 is deuterium, and Y 2 is Halogen (F, Cl, Br or I) or Y 2 is trifluoromethyl, Y 3 is hydrogen, Y 3 is deuterium, Y 3 is halogen (F, Cl, Br or I) or Y 3 is trifluoromethyl And so on, until Y 9 is hydrogen, Y 9 is deuterium, Y 9 is halogen (F, Cl, Br or I) or Y 9 is trifluoromethyl.
在另一具体实施方案中,“R 1、R 2、R 3、R 4和R 5各自独立地选自氢或氘”包括R 1选自氢或氘,R 2选自氢或氘,R 3选自氢或氘,以此类推,直至R 5选自氢或氘的技术方案。更具体地,包括R 1为氢、R 1为氘,R 2为氢、R 2为氘,R 3为氢、R 3为氘,以此类推,直至R 5为氢、R 5为氘的技术方案。 In another specific embodiment, "R 1, R 2, R 3, R 4 and R 5 are each independently selected from hydrogen or deuterium" includes R 1 is selected hydrogen or deuterium, R 2 is selected from hydrogen or deuterium, R 3 is selected from hydrogen or deuterium, and so on until R 5 is selected from hydrogen or deuterium. More particularly, R 1 is hydrogen comprising, R 1 is deuterium, R 2 is hydrogen, R 2 is deuterium, R 3 is hydrogen, R 3 is deuterium, and so on, until R 5 is hydrogen, deuterium, R 5 is Technical solutions.
在另一具体实施方案中,“X 1、X 2、X 3、X 4、X 5、X 6和X 7各自独立地选自CH 3、CD 3、CHD 2或CH 2D”包括X 1选自CH 3、CD 3、CHD 2或CH 2D,X 2选自CH 3、CD 3、CHD 2或CH 2D,X 3选自CH 3、CD 3、CHD 2或CH 2D,以此类推,直至X 7选自CH 3、CD 3、CHD 2或CH 2D的技术方案。更具体地,包括X 1为CH 3、X 1为CD 3、X 1为CHD 2或X 1为CH 2D,X 2为CH 3、X 2为CD 3、X 2为CHD 2或X 2为CH 2D,X 3为CH 3、X 3为CD 3、X 3为CHD 2或X 3为CH 2D,以此类推,直至X 7为CH 3、X 7为CD 3、X 7为CHD 2或X 7为CH 2D的技术方案。 In another specific embodiment, "X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D" including X 1 X 3 is selected from CH 3 , CD 3 , CHD 2 or CH 2 D, X 2 is selected from CH 3 , CD 3 , CHD 2 or CH 2 D, X 3 is selected from CH 3 , CD 3 , CHD 2 or CH 2 D, and By analogy, until X 7 is selected from the technical schemes of CH 3 , CD 3 , CHD 2 or CH 2 D. More specifically, X 1 is CH 3 , X 1 is CD 3 , X 1 is CHD 2 or X 1 is CH 2 D, X 2 is CH 3 , X 2 is CD 3 , and X 2 is CHD 2 or X 2 Is CH 2 D, X 3 is CH 3 , X 3 is CD 3 , X 3 is CHD 2 or X 3 is CH 2 D, and so on, until X 7 is CH 3 , X 7 is CD 3 , and X 7 is CHD 2 or X 7 is a technical solution of CH 2 D.
在优选地实施方案中,本发明涉及一种式(I)的化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体,其中,Y 1-Y 9各自独立地选自氢或氘,R 1-R 5和X 1-X 7如上所定义,附加条件是上述化合物至少含有一个氘原子。 In a preferred embodiment, the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof, wherein Y 1 to Y 9 are each independently selected from hydrogen or deuterium, and R 1 to R 5 and X 1 to X 7 are as defined above, with the proviso that the above compound contains at least one deuterium atom.
在优选地实施方案中,本发明涉及一种式(I)的化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体,其中,Y 3-Y 9为氢,Y 1和Y 2各自独立地选自氢或氘,R 1-R 5各自独立地选自氢或氘,X 1-X 7各自独立地选自CH 3、CD 3、CHD 2或CH 2D。 In a preferred embodiment, the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof, wherein Y 3 to Y 9 are hydrogen, Y 1 and Y 2 are each independently selected from hydrogen or deuterium, R 1 to R 5 are each independently selected from hydrogen or deuterium, and X 1 to X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D.
在优选地实施方案中,Y 1为氢。 In a preferred embodiment, Y 1 is hydrogen.
在优选地实施方案中,X 1和X 2是相同的。 In a preferred embodiment, X 1 and X 2 are the same.
在优选地实施方案中,X 1和X 2各自独立的选自CH 3或CD 3In a preferred embodiment, X 1 and X 2 are each independently selected from CH 3 or CD 3 .
在优选地实施方案中,X 3和X 4各自独立地选自CH 3或CD 3In a preferred embodiment, X 3 and X 4 are each independently selected from CH 3 or CD 3 .
在优选地实施方案中,X 6和X 7各自独立地选自CH 3、CD 3或CHD 2In a preferred embodiment, X 6 and X 7 are each independently selected from CH 3 , CD 3 or CHD 2 .
在优选地实施方案中,X 5是CH 3In a preferred embodiment, X 5 is CH 3 .
在优选地实施方案中,R 2、R 3、R 4和R 5是氢。 In a preferred embodiment, R 2 , R 3 , R 4 and R 5 are hydrogen.
在优选地实施方案中,R 1是氘。 In a preferred embodiment, R 1 is deuterium.
在优选地实施方案中,R 1是氢。 In a preferred embodiment, R 1 is hydrogen.
在优选地实施方案中,Y 2是氘。 In a preferred embodiment, Y 2 is deuterium.
在优选地实施方案中,Y 2是氢。 In a preferred embodiment, Y 2 is hydrogen.
作为本发明的优选实施方案,所述化合物为如下任一结构,或其药学上可接受的盐,但不限于下列结构:As a preferred embodiment of the present invention, the compound has any one of the following structures, or a pharmaceutically acceptable salt thereof, but is not limited to the following structures:
Figure PCTCN2019088749-appb-000004
Figure PCTCN2019088749-appb-000004
Figure PCTCN2019088749-appb-000005
Figure PCTCN2019088749-appb-000005
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。The compounds of the invention may include one or more asymmetric centers, and thus may exist in multiple stereoisomeric forms, for example, enantiomeric and / or diastereomeric forms. For example, the compounds of the invention may be individual enantiomers, diastereomers or geometric isomers (such as cis and trans isomers), or may be in the form of a mixture of stereoisomers, This includes racemic mixtures and mixtures rich in one or more stereoisomers. Isomers can be separated from a mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and formation and crystallization of chiral salts; or preferred isomers can be obtained by Prepared by asymmetric synthesis.
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。Those skilled in the art will understand that organic compounds can form complexes with solvents that react in the solvent or precipitate or crystallize from the solvent. These complexes are called "solvates". When the solvent is water, the complex is called a "hydrate". The invention encompasses all solvates of the compounds of the invention.
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理缔合可包括氢键键合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、乙醚等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂合物包括药学上可接受的溶剂合物且进一步包括化学计量的溶剂合物和非化学计量的溶剂合物。在一些情况下,所述溶剂合物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂合物”包括溶液状态的溶剂合物和可分离的溶剂合物。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。The term "solvate" refers to the form of a compound or a salt thereof in combination with a solvent, usually formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like. The compounds described herein can be prepared, for example, in crystalline form, and can be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be able to be separated, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. The "solvate" includes a solvate in a solution state and a separable solvate. Representative solvates include hydrates, ethanolates, and methanolates.
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式Rx H 2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为 1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R0.5H 2O))和多水合物(x为大于1的数,例如,二水合物(R2H 2O)和六水合物(R6H 2O))。 The term "hydrate" refers to a compound that is combined with water. Generally, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, for example, hydrates of the compounds may be used on behalf of the general formula Rx H 2 O, where R is the compound, and x is a number greater than 0. A given compound can form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, for example, hemihydrate (R0.5H 2 O )) and a multi-hydrate (x is greater than 1, e.g., dihydrate (R2H 2 O) and hexahydrate (R6H 2 O)).
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。The compounds of the invention may be in an amorphous or crystalline form (polymorphic form). In addition, the compounds of the invention may exist in one or more crystalline forms. Accordingly, the invention includes within its scope all amorphous or crystalline forms of the compounds of the invention. The term "polymorph" refers to a crystalline form (or a salt, hydrate, or solvate) of a compound in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, photoelectric properties, stability, and solubility. Recrystallization solvents, crystallization rates, storage temperatures, and other factors can lead to the predominance of a crystalline form. Various polymorphs of the compounds can be prepared by crystallization under different conditions.
本发明还包括同位素标记的化合物,它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如 3H和 14C)的那些可用于药物和/或底物组织分布测定。氚、即 3H和碳-14、即 14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即 2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(I)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。 The present invention also includes isotopically-labeled compounds, which are equivalent to those described in formula (I), but one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number commonly found in nature. Examples of isotopes that can be introduced into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl. Compounds of the present invention containing the above-mentioned isotopes and / or other isotopes of other atoms, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or said prodrugs are all within the scope of the present invention. Certain isotopically-labeled compounds of the invention, such as those incorporating radioisotopes such as 3 H and 14 C, can be used for drug and / or substrate tissue distribution assays. Thallium, i.e. 3 H and carbon-14, i.e. 14 C isotopes, are particularly preferred because they are easy to prepare and detect. Furthermore, replacement with heavier isotopes, such as deuterium, ie, 2 H, may be preferable in some cases because higher metabolic stability can provide therapeutic benefits, such as extending half-life in the body or reducing dosage requirements. An isotope-labeled compound of the formula (I) of the present invention and a prodrug thereof can generally be prepared in such a manner that, when performing the processes disclosed in the following schemes and / or examples and preparation examples, non-isotope-labeled reagents are replaced with readily available isotopically-labeled reagents Labeled reagent.
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。In addition, prodrugs are also included in the context of the present invention. The term "prodrug" as used herein refers to a compound that is converted into its active form with a medical effect in vivo by, for example, hydrolysis in blood. Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs 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, or drug, delivery: solubility, limitation, overcome, and use of prodrugs", Advanced Drug Delivery Reviews (1996) 19 (2) 115-130, each article This article is for reference.
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(I) 化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。A prodrug is any covalently bonded compound of the invention, and when such a prodrug is administered to a patient, it releases the parent compound in vivo. Prodrugs are usually prepared by modifying functional groups, and the modification is performed in a manner such that the modification can be performed by routine manipulation or cleavage in vivo to produce the parent compound. Prodrugs include, for example, compounds of the invention in which a hydroxyl, amino, or thiol group is bonded to an arbitrary group, and when administered to a patient, can cleave to form a hydroxyl, amino, or thiol group. Thus, representative examples of prodrugs include, but are not limited to, acetate, amide, formate / amide, and benzoate / amide derivatives of hydroxyl, thiol, and amino functional groups of the compound of formula (I). In the case of a carboxylic acid (-COOH), an ester such as methyl ester, ethyl ester, or the like can be used. The esters themselves can be active and / or can be hydrolyzed under conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those groups that readily break down in the human body to release the parent acid or its salt.
合成synthesis
本发明化合物(包括其盐)可使用已知有机合成技术来制备,且可按照多种可能合成途径中的任一种(诸如下文方案中的那些)来合成。用于制备本发明化合物的反应可在合适的溶剂中进行,有机合成领域的技术人员可容易地选择溶剂。合适的溶剂可在进行反应的温度(例如,在溶剂结冻温度至溶剂沸点温度范围内的温度)下与起始物质(反应物)、中间体或产物实质上不反应。既定反应可在一种溶剂或一种以上溶剂的混合物中进行。技术人员可依据具体反应步骤来选择用于具体反应步骤的溶剂。The compounds of the invention, including their salts, can be prepared using known organic synthesis techniques, and can be synthesized according to any of a number of possible synthetic pathways, such as those in the schemes below. The reaction for preparing the compound of the present invention can be performed in a suitable solvent, and those skilled in the art of organic synthesis can easily select a solvent. Suitable solvents may be substantially non-reactive with the starting material (reactant), intermediate, or product at the temperature at which the reaction is performed (eg, a temperature in the range of the solvent freezing temperature to the solvent boiling temperature). A given reaction may be performed in one solvent or a mixture of more than one solvent. The skilled person can select a solvent for a specific reaction step depending on the specific reaction step.
本发明化合物的制备可涉及不同化学基团的保护和去除保护。本领域技术人员可容易地判定是否需要保护和去除保护以及适当保护基的选择。保护基的化学性质可参见例如Wuts和Greene,Protective Groups in Organic Synthesis,第4版,John Wiley&Sons:New Jersey,(2006),其通过引用整体并入本文中。The preparation of the compounds of the invention may involve the protection and removal of different chemical groups. Those skilled in the art can easily determine whether protection and removal of protection are needed and the selection of an appropriate protecting group. The chemical properties of protecting groups can be found, for example, in Wuts and Greene, Protective Groups, Organic Synthesis, 4th Edition, John Wiley & Sons: New Jersey, (2006), which is incorporated herein by reference in its entirety.
可按照本领域已知任何合适的方法来监测反应。例如,可通过光谱手段(诸如核磁共振(NMR)光谱法(例如 1H或 13C)、红外(IR)光谱法、分光光度法(例如,UV-可见光)、质谱(MS))或通过色谱方法(诸如高效液相色谱法(HPLC)或薄层色谱法(TLC))来监测产物形成。 The reaction can be monitored according to any suitable method known in the art. For example, spectroscopic means (such as nuclear magnetic resonance (NMR) spectroscopy (e.g., 1 H or 13 C), infrared (IR) spectroscopy, spectrophotometry (e.g., UV-visible light), mass spectrometry (MS)), or by chromatography Methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC) to monitor product formation.
药物组合物、制剂和试剂盒Pharmaceutical compositions, preparations and kits
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的活性组分。在一些实施方案中,所述药物组合物包含治疗有效量的活性组分。在一些实施方案中,所述药物组合物包含预防有效量的活性组分。In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
用于本发明的药学上可接受的赋形剂是指不会破坏一起配制的化合物的药理学活性的无毒载剂、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括但不限于,离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷 酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。A pharmaceutically acceptable excipient for use in the present invention refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compounds 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 (such as human serum albumin ), Buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substance, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene-embedded Polymer, polyethylene glycol and lanolin.
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。The invention also includes kits (eg, pharmaceutical packaging). The provided kits can include a compound of the invention, other therapeutic agents, and first and second containers (e.g., vials, ampoules, bottles, syringes, and / or dispersible packaging or other Suitable container). In some embodiments, the provided kit may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and / or other therapeutic agent. In some embodiments, a compound of the invention and other therapeutic agents are provided in a first container and a second container to form a unit dosage form.
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。The pharmaceutical composition provided by the present invention can be administered by many routes, including but not limited to: oral administration, parenteral administration, inhalation administration, topical administration, rectal administration, nasal administration, oral administration, vaginal administration Medication, implantation or other means of administration. For example, parenteral administration as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intra-articular, intra-arterial, intra-synovial, and sternal administration , Cerebrospinal spinal membrane administration, intralesional administration, and intracranial injection or infusion techniques.
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。Generally, an effective amount of a compound provided herein is administered. Depending on the circumstances, including the condition being treated, the route of administration chosen, the compound actually administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, etc., the amount of compound actually administered can be determined by the physician .
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。When used to prevent a condition described in the present invention, a compound provided herein is administered to a subject at risk of developing the condition, typically based on and under the supervision of a physician, at a dosage level as described above. Subjects at risk for developing a particular disorder typically include subjects with a family history of the disorder, or those who are determined by genetic testing or screening to be particularly sensitive to the development of the disorder.
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。The pharmaceutical compositions provided herein can also be administered chronically ("long-term administration"). Long-term administration refers to administration of a compound or a pharmaceutical composition thereof over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or the administration can be continued indefinitely, For example, the remainder of a subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a long period of time, for example, within a therapeutic window.
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度快速提高至有效水平。推注剂量取决于活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注(例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药 物组合物,而后持续输液。Various methods of administration can be used to further deliver the pharmaceutical composition of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered by bolus, for example, to rapidly increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the target systemic level of the active ingredient, for example, an intramuscular or subcutaneous bolus dose allows for a slow release of the active ingredient, whereas a bolus delivered directly to a vein (e.g., by IV intravenous drip) can be more Rapid delivery allows the concentration of the active ingredient in the blood to rise quickly to an effective level. In other embodiments, the pharmaceutical composition may be administered in the form of a continuous infusion, for example, by IV infusion, to provide a steady state concentration of the active ingredient in the subject's body. Furthermore, in other embodiments, the bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。Oral compositions can take the form of a liquid solution or suspension in bulk or a powder in bulk. However, more generally, to facilitate accurate dosing, the composition is provided in unit dosage form. The term "unit dosage form" refers to a physically discrete unit suitable as a unit dose for human patients and other mammals, each unit containing a predetermined number of active substances and suitable pharmaceutical excipients suitable for producing the desired therapeutic effect. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or pills, tablets, capsules, etc. in the case of solid compositions. In such a composition, the compound is usually a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), and the remainder is each useful for forming a desired administration form A carrier or excipient and processing aid.
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。For oral doses, a representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses. Using these dosage modes of administration, each dose provides about 0.01 to about 20 mg / kg of a compound of the invention, and preferred doses each provide about 0.1 to about 10 mg / kg, especially about 1 to about 5 mg / kg.
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。In order to provide a blood level similar to, or lower than, the injected dose, transdermal doses are usually selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, preferably about 0.1 To about 10% by weight, and more preferably about 0.5 to about 15% by weight.
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。From about 1 to about 120 hours, especially 24 to 96 hours, the injection dose level ranges from about 0.1 mg / kg / hour to at least 10 mg / kg / hour. To obtain a sufficient steady state level, a preloaded bolus of about 0.1 mg / kg to about 10 mg / kg or more can also be given. For human patients from 40 to 80 kg, the maximum total dose cannot exceed about 2 g / day.
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous vehicles and 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 compounds having similar properties: a binder, such as microcrystalline cellulose, tragacanth, or gelatin; an excipient, such as starch or lactose, a disintegrant, For example, alginic acid, Primogel, or corn starch; lubricants, such as magnesium stearate; glidants, such as colloidal silicon dioxide; sweeteners, such as sucrose or saccharin; or flavoring agents, such as mint, water Methyl salicylate or orange flavor.
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art. As mentioned earlier, in this composition, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。Transdermal compositions are typically formulated as a topical ointment or cream containing an active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffin or a water-miscible ointment base. Alternatively, the active ingredient may 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 components for enhancing the stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope of the invention.
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。The compounds of the invention may also be administered via a transdermal device. Therefore, transdermal administration can be achieved using a reservoir or porous membrane type, or a variety of solid matrix patches.
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington's Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。The above components of the composition for oral administration, injection or topical administration are merely representative. Other materials and processing techniques are described in Section 8 of Remington's Pharmaceuticals, Science, 17th Edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材料的描述可在Remington's Pharmaceutical Sciences中找到。The compounds of the invention may also be administered in a sustained release form or from a sustained release delivery system. A description of representative sustained-release materials can be found in Remington's Pharmaceutical Sciences.
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。The invention also relates to a pharmaceutically acceptable formulation of a compound of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α-, β-, and γ-cyclodextrin consisting of 6, 7, and 8 α-1,4-linked glucose units, respectively, which optionally include one on the linked sugar moiety Or more substituents, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substituted. In some embodiments, the cyclodextrin is a sulfoalkyl ether β-cyclodextrin, for example, a sulfobutyl ether β-cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645. In some embodiments, the formulation includes hexapropyl-β-cyclodextrin (eg, 10-50% in water).
适应症Indication
本发明提供了一种抑制蛋白酪氨酸激酶(如EGFR激酶)的方法或治疗疾病(如癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病)的方法,它包括步骤:给需要治疗的受试者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物。The present invention provides a method for inhibiting a protein tyrosine kinase (such as EGFR kinase) or treating a disease (such as cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplant, viral disease, cardiovascular disease or Method of metabolic disease), which comprises the step of administering to a subject in need of treatment a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystalline form, prodrug thereof Or an isotope derivative, or a pharmaceutical composition according to the invention.
本发明化合物可用于治疗EGFR导致的癌症。尤其是,所述化合物可用于治疗表达EGFR突变体的EGFR导致的癌症和用于治疗对RTKI疗法(例如,厄洛替尼或吉非替尼)难治的EGFR导致的癌症。The compounds of the invention are useful for treating cancer caused by EGFR. In particular, the compounds are useful in the treatment of cancers caused by EGFR expressing EGFR mutants and in the treatment of cancers of EGFR refractory to RTKI therapy (eg, erlotinib or gefitinib).
本发明化合物是EGFR的至少一种突变体的抑制剂并且因此适用于治疗与一种或一种以上EGFR突变体(例如缺失突变、活化突变、抗性突变或其组合,具体实例包括T790M突变、L858R突变和L858R/T790M双突变)的活性相关的一种或一种以上病症。因此,在具体实施方案中,本发明提供一种治疗突变EGFR介导的病症的方法,其包含向有需要的患者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物的步骤。The compounds of the present invention are inhibitors of at least one mutant of EGFR and are therefore suitable for use in treatment with one or more EGFR mutants (e.g., deletion mutations, activation mutations, resistance mutations, or combinations thereof, specific examples including T790M mutations, L858R mutation and L858R / T790M double mutation) activity related to one or more disorders. Therefore, in a specific embodiment, the present invention provides a method for treating a mutant EGFR-mediated disorder, which comprises administering to a patient in need thereof a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, A solvate, a hydrate, a crystalline form, a prodrug or an isotope derivative, or a step of administering a pharmaceutical composition according to the present invention.
本发明化合物可治疗的癌症包括但不限于:非小细胞肺癌(NSCLS)、小细胞肺癌、肺腺癌、肺 鳞癌、胰腺癌、乳腺癌、前列腺癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌等过度增殖性疾病。此外,本发明化合物也可用于在需要此类治疗的患者中起到预防癌症复发的维持作用。Cancers treatable by the compounds of the present invention include, but are not limited to, non-small cell lung cancer (NSCLS), small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, Gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma and other hyperproliferative diseases. In addition, the compounds of the present invention are also useful for maintaining the prevention of cancer recurrence in patients in need of such treatment.
本发明化合物的有效量通常在平均日剂量为0.01mg至50mg化合物/千克患者体重,优选0.1mg至25mg化合物/千克患者体重,以单次或多次给药。通常,本发明化合物可向该有此治疗需要的患者以每位患者约1mg至约3500mg的日剂量范围给药,优选10mg至1000mg。例如,每位患者的日剂量可为10、20、30、40、50、60、70、80、90、100、150、200、250、300、350、400、500、600、700、800、900或1000mg。可每天、每周(或间隔数天)或以间歇时间表,给药一次或多次。例如,可在每周的基础上(例如每周一),每天给予所述化合物一次或多次,不定地或持续几周,例如4-10周。或者,可每天给药持续几天(例如2-10天),然后几天(例如1-30天)不给药所述化合物,不定地重复该循环或重复给定的次数,例如4-10个循环。例如,本发明化合物可每天给药持续5天,然后间断9天,然后再每天给药持续5天,然后间断9天,以此类推,不定地重复该循环或共重复4-10次。An effective amount of a compound of the present invention is usually administered in a single or multiple doses at an average daily dose of 0.01 to 50 mg of the compound per kg of the patient's body weight, preferably 0.1 to 25 mg of the compound per kg of the patient's body weight. Generally, the compound of the present invention can be administered to the patient in need of such treatment at a daily dose range of about 1 mg to about 3500 mg per patient, preferably 10 mg to 1000 mg. For example, the daily dose per patient may be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000mg. Administration can be one or more times daily, weekly (or several days apart), or on an intermittent schedule. For example, the compound can be administered one or more times per day on an weekly basis (e.g., every Monday), indefinitely or for several weeks, such as 4-10 weeks. Alternatively, the compound can be administered daily for several days (e.g., 2-10 days), and then the compound is not administered for several days (e.g., 1-30 days), the cycle is repeated indefinitely, or a given number of times, such as 4-10 Cycles. For example, a compound of the invention may be administered daily for 5 days, then intermittently for 9 days, and then daily for 5 days, then intermittently for 9 days, and so on, the cycle may be repeated indefinitely or a total of 4-10 times.
当EGFR-TKI(例如,厄洛替尼或吉非替尼)与本发明化合物组合使用时,该组合疗法的各个成分可以以它们单一疗法的剂量水平和方案给药。例如,厄洛替尼,对于治疗非小细胞肺癌,已经以每天150mg口服给药,对于胰腺癌,已经以每天100mg口服给药。在另一实例中,吉非替尼对于治疗非小细胞肺癌已经以每天250mg口服给药。When EGFR-TKI (eg, erlotinib or gefitinib) is used in combination with a compound of the invention, the components of the combination therapy can be administered at their monotherapy dose levels and schedules. For example, erlotinib has been administered orally at 150 mg per day for the treatment of non-small cell lung cancer, and it has been administered orally at 100 mg per day for pancreatic cancer. In another example, gefitinib has been administered orally at 250 mg per day for the treatment of non-small cell lung cancer.
优选地,当EGFR-TKI(例如,厄洛替尼或吉非替尼)与本发明化合物组合使用,其一种或两种成分的剂量水平相比于单独使用时降低。Preferably, when EGFR-TKI (eg, erlotinib or gefitinib) is used in combination with a compound of the invention, the dosage level of one or both of its components is reduced compared to when used alone.
实施例Examples
下面结合具体实施例,作进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则份数和百分比为重量份和重量百分比。The present invention will be further described below with reference to specific embodiments. It should be understood that these examples are only used to illustrate the present invention and not to limit the scope of the present invention. The experimental methods without specific conditions in the following examples are generally based on conventional conditions or conditions recommended by the manufacturer. Unless stated otherwise, parts and percentages are parts by weight and percent by weight.
通常,在制备流程中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~80℃)下进行。反应时间通常为0.1-60小时,优选地为0.5-24小时。Generally, in the preparation process, each reaction is usually performed in an inert solvent at room temperature to reflux temperature (for example, 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.
本文所用的缩写具有以下含义:The abbreviations used in this article have the following meanings:
APCIAPCI 大气压力化学游离法Atmospheric pressure chemical dissociation
TEATEA 三乙胺Triethylamine
B 2pin 2 B 2 pin 2 联硼酸频那醇酯Pinacol diborate
pTSApTSA 对甲苯磺酸p-Toluenesulfonic acid
PEPE 石油醚Petroleum ether
EAEA 乙酸乙酯Ethyl acetate
DMSODMSO 二甲亚砜Dimethyl sulfoxide
DMFDMF N,N-二甲基乙酰胺N, N-dimethylacetamide
DCMDCM 二氯甲烷Dichloromethane
XantPhosXantPhos 4,5-双二苯基膦-9,9-二甲基氧杂蒽4,5-bisdiphenylphosphine-9,9-dimethylxanthene
DIPEADIPEA N,N-二异丙基乙胺N, N-diisopropylethylamine
TFATFA 三氟乙酸Trifluoroacetate
实施例1 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-2-甲基-1-(丙-2-基Example 1 N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -5-((4- (4-fluoro-2-methyl-1- (propane-2 -base -1,1,1,3,3,3-d 6)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物I-1)的制备。 -1,1,1,3,3,3-d 6 ) -1H-benzo [d] imidazole-6-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide ( Preparation of compound I-1).
Figure PCTCN2019088749-appb-000006
Figure PCTCN2019088749-appb-000006
采用以下路线进行合成:Use the following route for synthesis:
Figure PCTCN2019088749-appb-000007
Figure PCTCN2019088749-appb-000007
步骤1 化合物1的合成Step 1 Synthesis of Compound 1
取氘代丙酮(5.00g,78.0mmol)溶于1,2-二氯乙烷(100mL),加入对甲氧基苄胺(10.7g,78.0mmol),室温搅拌2小时。缓慢加入NaBH 3CN,室温搅拌过夜。过滤,取滤液浓缩,柱层析(PE/EA,V/V,5/1)得化合物69为无色油状液体8.05g。 Deuterated acetone (5.00 g, 78.0 mmol) was dissolved in 1,2-dichloroethane (100 mL), p-methoxybenzylamine (10.7 g, 78.0 mmol) was added, and the mixture was stirred at room temperature for 2 hours. Add NaBH 3 CN slowly and stir overnight at room temperature. Filtration, concentration of the filtrate, and column chromatography (PE / EA, V / V, 5/1) gave compound 69 as a colorless oily liquid 8.05 g.
步骤2 化合物2的合成Step 2 Synthesis of Compound 2
将化合物1(8.05g,43.4mmol)溶于二氯甲烷(80mL),加入95mL饱和碳酸氢钠溶液,冰水浴下滴加乙酰氯(5.1g,65.14mmol)的二氯甲烷(15mL)溶液,自然升温搅拌过夜,分液,水相用二氯甲烷萃取(30mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得到棕色油状液体(9.842g)。取部分上述棕色油状产物(4.00g,17.6mmol)置于单口烧瓶,加入10mL水分散,加入10mL浓盐酸,加热回流5小时。冷却至室温,乙酸乙酯萃取(10mL×3),有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得到无色液体(0.85g)。Compound 1 (8.05 g, 43.4 mmol) was dissolved in dichloromethane (80 mL), 95 mL of a saturated sodium bicarbonate solution was added, and a solution of acetyl chloride (5.1 g, 65.14 mmol) in dichloromethane (15 mL) was added dropwise under an ice water bath. The mixture was stirred with natural heating overnight, and the layers were separated. The aqueous phase was extracted with dichloromethane (30 mL × 3). The organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated by filtration to obtain a brown oily liquid (9.842 g). A part of the above brown oily product (4.00 g, 17.6 mmol) was placed in a single-necked flask, 10 mL of water was added to disperse, 10 mL of concentrated hydrochloric acid was added, and the mixture was heated under reflux for 5 hours. Cooled to room temperature, extracted with ethyl acetate (10 mL × 3), washed the organic phase with saturated brine, dried over anhydrous sodium sulfate, and concentrated by filtration to obtain a colorless liquid (0.85 g).
步骤3 化合物3的合成Step 3 Synthesis of Compound 3
取化合物2(850mg,7.93mmol)溶于甲苯(10mL),加入化合物4-溴-2,6-二氟苯胺(825mg,3.97mmol),一次性加入三氯氧磷(1.13mL,12.1mmol)和TEA(1.64mL,11.9mmol),加热回流5小时。冷却至室温,加入EA(30mL)稀释,过滤浓缩,得棕色液体917mg。取棕色液体(917mg,3.03mmol)溶于THF(15mL),室温下加入tBuOK(680mg,6.06mmol),加热回流过夜。冷却至室温,浓缩反应液,加入EA(5mL)和饱和食盐水(5mL)分液,水相用EA萃取(3mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得淡黄色固体(658mg,2.37mmol)。上述淡黄色固体置于单口烧 瓶,依次加入联硼酸频哪醇酯(904mg,3.56mmol)、三环己基磷(113mg,0.04mmol)、醋酸钯(59mg,0.26mol)、醋酸钾(699mg,7.12mmol),氮气置换三次,在氮气保护下加入无水DMSO(11mL),90℃加热搅拌反应45分钟。冷却反应至室温,倒入50mL水中,EA-PE(v/v,1/1,10mL*2)萃取,合并有机相蒸干得到粗品直接用于下一步。Take compound 2 (850 mg, 7.93 mmol) in toluene (10 mL), add compound 4-bromo-2,6-difluoroaniline (825 mg, 3.97 mmol), and add phosphorus oxychloride (1.13 mL, 12.1 mmol) in one portion. And TEA (1.64 mL, 11.9 mmol), heated to reflux for 5 hours. It was cooled to room temperature, diluted with EA (30 mL), filtered and concentrated to obtain 917 mg of a brown liquid. A brown liquid (917 mg, 3.03 mmol) was dissolved in THF (15 mL), tBuOK (680 mg, 6.06 mmol) was added at room temperature, and the mixture was heated under reflux overnight. Cool to room temperature, concentrate the reaction solution, add EA (5mL) and saturated brine (5mL) to separate the layers, extract the aqueous phase with EA (3mL × 3), combine the organic phases and wash with saturated brine, dry over anhydrous sodium sulfate, and filter Concentrated to give a pale yellow solid (658 mg, 2.37 mmol). The light yellow solid was placed in a single-necked flask, and pinacol diborate (904 mg, 3.56 mmol), tricyclohexyl phosphorus (113 mg, 0.04 mmol), palladium acetate (59 mg, 0.26 mol), and potassium acetate (699 mg, 7.12) were added in this order. mmol), replaced with nitrogen three times, and added anhydrous DMSO (11 mL) under nitrogen protection, and heated and stirred at 90 ° C for 45 minutes. The reaction was cooled to room temperature, poured into 50 mL of water, and extracted with EA-PE (v / v, 1/1, 10 mL * 2). The organic phases were combined and evaporated to dryness to obtain the crude product, which was directly used in the next step.
步骤4 化合物4的合成Step 4 Synthesis of Compound 4
将上述粗品化合物3(485mg,0.57mmol),2,4-二氯嘧啶(85mg,0.57mmol),Pd(PPh 3) 2Cl 2(24mg,0.034mmol),2M的碳酸钠溶液(1.1mL)溶于DMF(5mL),氮气置换三次,60℃加热搅拌2小时。冷却至室温,反应液倒入20mL水中,EA萃取(5mL×3),合并EA并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析(PE/EA,V/V,2/1)得化合物4为淡黄色固体(262mg)。 The above crude compound 3 (485 mg, 0.57 mmol), 2,4-dichloropyrimidine (85 mg, 0.57 mmol), Pd (PPh 3 ) 2 Cl 2 (24 mg, 0.034 mmol), 2M sodium carbonate solution (1.1 mL) Dissolved in DMF (5mL), replaced with nitrogen three times, and heated and stirred at 60 ° C for 2 hours. After cooling to room temperature, the reaction solution was poured into 20 mL of water and extracted with EA (5 mL × 3). The EA was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, and column chromatography (PE / EA, V / V, 2 / 1) Compound 4 was obtained as a pale yellow solid (262 mg).
步骤5 化合物5的合成Step 5 Synthesis of Compound 5
取化合物4(262mg,0.84mmol),4-氟-2-甲氧基-5-硝基苯胺(188mg,1.01mmol)、pTSA(641mg,3.37mmol)于氮气保护下加入到单口反应瓶中,之后加入无水二氧六环(10mL),氮气置换三次,100℃加热搅拌反应24小时。减压蒸干溶剂,制备级薄层色谱纯化(PE/EA,V/V,1/1)得220mg灰色固体。以上所得灰色固体(220mg,0.48mmol)加入乙腈(5mL)分散均匀,先后加入N 1,N 1,N 2-三甲基乙烷-1,2-二胺(59mg,0.57mmol)、碳酸钾(132mg,0.96mmol),90℃加热回流反应2小时。将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得化合物5为红色油状物(126mg)。 Take compound 4 (262 mg, 0.84 mmol), 4-fluoro-2-methoxy-5-nitroaniline (188 mg, 1.01 mmol) and pTSA (641 mg, 3.37 mmol) into a single-mouth reaction flask under the protection of nitrogen. After that, anhydrous dioxane (10 mL) was added, and nitrogen was substituted three times. The reaction was heated and stirred at 100 ° C for 24 hours. The solvent was evaporated to dryness under reduced pressure and purified by preparative thin layer chromatography (PE / EA, V / V, 1/1) to obtain 220 mg of a gray solid. The gray solid (220 mg, 0.48 mmol) obtained above was dispersed uniformly by adding acetonitrile (5 mL), and then N 1 , N 1 , N 2 -trimethylethane-1,2-diamine (59 mg, 0.57 mmol), and potassium carbonate were added. (132 mg, 0.96 mmol), heated to reflux at 90 ° C for 2 hours. The reaction solution was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness to obtain Compound 5 as a red oil (126 mg).
步骤6 化合物I-1的合成Step 6 Synthesis of Compound I-1
将红色油状物化合物5(126mg)溶于乙醇-水(4mL+1mL)的混合溶液中,加入还原铁粉(106mg)和氯化铵(34mg),90℃加热回流反应5小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入10mL DCM溶解,加入饱和碳酸氢钠水溶液5mL,冰浴冷却。向以上两相体系滴加0.15M的丙烯酰氯二氯甲烷(2.5mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,20/1)得化合物I-1为白色固体(59mg)。 1H NMR(400MHz,CDCl 3)δ9.65–9.37(m,2H),8.51(d,J=5.3Hz,1H),8.03(d,J=1.3Hz,1H),7.81–7.71(m,1H),7.67(s,1H),7.14(dd,J=5.3,2.4Hz,1H),6.72(s,1H),6.54–6.07(m,2H),5.90–5.40(m,1H),4.72(s,1H),3.90(s,3H),3.20(t,J=6.0Hz,2H),2.89(d,J=16.1Hz,2H),2.71(s,3H),2.68(s,3H),2.66(s,6H)。 The red oily compound 5 (126 mg) was dissolved in a mixed solution of ethanol-water (4 mL + 1 mL), and reduced iron powder (106 mg) and ammonium chloride (34 mg) were added, and the mixture was heated under reflux at 90 ° C for 5 hours to react. The reaction was cooled to room temperature, the insoluble solid was removed by filtration, and the filtrate was evaporated to dryness. The obtained oil was dissolved by adding 10 mL of DCM, 5 mL of a saturated aqueous sodium hydrogen carbonate solution was added, and the solution was cooled in an ice bath. To the above two-phase system, 0.15 M of acryloyl chloride methylene chloride (2.5 mL) was added dropwise, and the reaction was carried out under an ice bath for 30 minutes. The layers were separated and the organic phase was evaporated to dryness. Purification by preparative thin layer chromatography (DCM / MeOH, V / V, 20/1) gave compound I-1 as a white solid (59 mg). 1 H NMR (400MHz, CDCl 3 ) δ 9.65–9.37 (m, 2H), 8.51 (d, J = 5.3Hz, 1H), 8.03 (d, J = 1.3Hz, 1H), 7.81–7.71 (m, 1H), 7.67 (s, 1H), 7.14 (dd, J = 5.3, 2.4Hz, 1H), 6.72 (s, 1H), 6.54-6.07 (m, 2H), 5.90-5.40 (m, 1H), 4.72 (s, 1H), 3.90 (s, 3H), 3.20 (t, J = 6.0 Hz, 2H), 2.89 (d, J = 16.1 Hz, 2H), 2.71 (s, 3H), 2.68 (s, 3H) , 2.66 (s, 6H).
实施例2 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-2-甲基-1-(丙-2-基-2-d)-1H-苯并Example 2 N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -5-((4- (4-fluoro-2-methyl-1- (propane-2 -Yl-2-d) -1H-benzo [d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物I-2)的制备。[d] Preparation of imidazole-6-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-2).
Figure PCTCN2019088749-appb-000008
Figure PCTCN2019088749-appb-000008
根据实施例1中所述的合成方法,用以下步骤代替实施例1中的步骤1:According to the synthesis method described in Example 1, step 1 in Example 1 is replaced by the following steps:
取丙酮(3.2g,56mmol),对甲氧基苄胺(5.1g,37mmol)加入到甲苯(50mL)中溶解,在130℃油浴中加热回流分水3小时,冷却至室温,补加3.0g丙酮继续回流分水4小时。冷却反应至室温,减压蒸去大部分甲苯,加入四氢呋喃30mL,MeOD 10mL,冰浴冷却,分批加入氘代硼氢化钠0.8g室温搅拌反应过夜。减压蒸干溶剂,加水20mL,DCM 20mL分液。有机相与饱和碳酸氢钠水溶液(30mL)混合搅拌分散均匀冰浴冷却,滴加乙酰氯(2.89g,37mmol),滴加完毕继续反应1小时。分液,有机相蒸干柱层析(PE/EA,v/v,8/1,0.5%TEA)得到化合物N-(4-甲氧基苯基)丙-2-d-2-胺3.04g。Take acetone (3.2g, 56mmol), add p-methoxybenzylamine (5.1g, 37mmol) to toluene (50mL) to dissolve it, heat and reflux in a 130 ° C oil bath for 3 hours, cool to room temperature, add 3.0 g of acetone continued to reflux for 4 hours. The reaction was cooled to room temperature, most of the toluene was distilled off under reduced pressure, 30 mL of tetrahydrofuran and 10 mL of MeOD were added, and the solution was cooled in an ice bath. 0.8 g of sodium deuterated borohydride was added in portions and the reaction was stirred at room temperature overnight. The solvent was evaporated to dryness under reduced pressure, 20 mL of water was added, and 20 mL of DCM was used for liquid separation. The organic phase was mixed with a saturated sodium bicarbonate aqueous solution (30 mL), stirred and dispersed uniformly, and cooled in an ice bath, and acetyl chloride (2.89 g, 37 mmol) was added dropwise. The reaction was continued for 1 hour after the dropwise addition. Separate the liquid and evaporate the organic phase through column chromatography (PE / EA, v / v, 8/1, 0.5% TEA) to obtain the compound N- (4-methoxyphenyl) propan-2-d-2-amine 3.04 g.
用上述步骤得到的化合物N-(4-甲氧基苯基)丙-2-d-2-胺替代实施例1中的步骤2的化合物1,再重复实施例1中的步骤2-6,制备得到化合物I-2,化合物I-2为灰白色固体(70mg)。 1H NMR(400MHz,CDCl 3)δ9.88(s,1H),9.63(s,1H),8.52(d,J=5.3Hz,1H),8.03(d,J=1.3Hz,1H),7.79(d,J=11.4Hz,1H),7.67(s,1H),7.13(d,J=5.3Hz,1H),6.78(s,1H),6.46(s,2H),5.69(d,J=11.8Hz,1H),3.89(s,3H),2.97(s,2H),2.70(s,3H),2.67(s,3H),2.46(s,2H),2.36(s,6H),1.65(s,6H). Use the compound N- (4-methoxyphenyl) propan-2-d-2-amine obtained in the above step to replace the compound 1 in step 2 in Example 1, and then repeat steps 2-6 in Example 1, Compound I-2 was prepared. Compound I-2 was an off-white solid (70 mg). 1 H NMR (400MHz, CDCl 3 ) δ 9.88 (s, 1H), 9.63 (s, 1H), 8.52 (d, J = 5.3 Hz, 1H), 8.03 (d, J = 1.3 Hz, 1H), 7.79 (d, J = 11.4 Hz, 1H), 7.67 (s, 1H), 7.13 (d, J = 5.3 Hz, 1H), 6.78 (s, 1H), 6.46 (s, 2H), 5.69 (d, J = 11.8Hz, 1H), 3.89 (s, 3H), 2.97 (s, 2H), 2.70 (s, 3H), 2.67 (s, 3H), 2.46 (s, 2H), 2.36 (s, 6H), 1.65 ( s, 6H).
实施例3 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-(甲基-d 3)-1H-苯并[d] Example 3 N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -5-((4- (4-fluoro-1-isopropyl-2- (methyl -d 3 ) -1H-benzo [d] 咪唑-6-基-5-d)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物I-3)的制备。Preparation of imidazole-6-yl-5-d) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-3).
Figure PCTCN2019088749-appb-000009
Figure PCTCN2019088749-appb-000009
采用以下路线进行合成:Use the following route for synthesis:
Figure PCTCN2019088749-appb-000010
Figure PCTCN2019088749-appb-000010
步骤1 化合物7的合成Step 1 Synthesis of Compound 7
室温下将NaOD(40%的重水溶液,催化量)滴加入到化合物6(1.50g,5.55mmol)的重水(50mL)和氘代甲醇-d 4(1mL)的混合溶剂中,反应在140℃下封管反应过夜。冷却至室温,用二氯甲烷(50mL x 3)萃取,合并有机相,饱和食盐水(50mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到1.45g白色固体,收率:97.0%。LC-MS(APCI):m/z=275.0(M+1)。 1H NMR(500MHz,DMSO-d 6)δ7.73(s,1H),4.87–4.60(m,1H),1.51(d,J=6.9Hz,6H). NaOD (40% heavy aqueous solution, catalytic amount) was added dropwise to a mixed solvent of compound 6 (1.50 g, 5.55 mmol) in heavy water (50 mL) and deuterated methanol-d 4 (1 mL) at room temperature, and the reaction was performed at 140 ° C. The tube was sealed and allowed to react overnight. Cool to room temperature, extract with dichloromethane (50 mL x 3), combine the organic phases, wash with saturated brine (50 mL), dry over anhydrous sodium sulfate, remove the solvent, and concentrate the column for separation (eluent: petroleum ether / ethyl acetate Ester (v / v) = 3: 1), 1.45 g of a white solid was obtained in a yield of 97.0%. LC-MS (APCI): m / z = 275.0 (M + 1). 1 H NMR (500MHz, DMSO-d 6 ) δ7.73 (s, 1H), 4.87-4.60 (m, 1H), 1.51 (d, J = 6.9Hz, 6H).
步骤2 化合物8的合成Step 2 Synthesis of Compound 8
氮气保护下将醋酸钯(200mg)和三环己基磷(400mg)加入到化合物7(2.00g,7.30mmol),联硼酸频那醇酯(2.80g,10.95mmol)和醋酸钾(2.10g,21.90mmol)的无水二甲亚砜(20mL)溶液中,反应在氮气保护下100℃反应2小时。冷却至室温,硅藻土过滤,乙酸乙酯洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到1.90g白色固体。收率:80.8%。LC-MS(APCI):m/z=323.2(M+1) +Palladium acetate (200 mg) and tricyclohexyl phosphorus (400 mg) were added to compound 7 (2.00 g, 7.30 mmol), pinacol diborate (2.80 g, 10.95 mmol), and potassium acetate (2.10 g, 21.90) under nitrogen. In a solution of anhydrous dimethyl sulfoxide (20 mL), the reaction was carried out at 100 ° C. for 2 hours under a nitrogen atmosphere. Cooled to room temperature, filtered through celite, washed the filter cake with ethyl acetate, washed the filtrate with saturated brine, dried over anhydrous sodium sulfate, concentrated the filtrate under reduced pressure, and concentrated the solution for column separation (eluent: petroleum ether / ethyl acetate (v / v) = 3: 1), 1.90 g of a white solid was obtained. Yield: 80.8%. LC-MS (APCI): m / z = 323.2 (M + 1) + .
步骤3 化合物9的合成Step 3 Synthesis of Compound 9
氮气保护下将乙腈(18mL)和水(6mL)加入到化合物8(1.90g,5.88mmol)和2,4-二氯嘧啶(1.10g,7.06mmol)、碳酸钠(1.60g,14.70mmol)、双三苯基磷二氯化钯(190mg)的混合物中,反应在氮气保护下,80℃搅拌反应2小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼, 用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1)得到1.26g白色固体。收率为69.6%。LC-MS(APCI):m/z=309.1(M+1) +Acetonitrile (18 mL) and water (6 mL) were added to compound 8 (1.90 g, 5.88 mmol) and 2,4-dichloropyrimidine (1.10 g, 7.06 mmol), sodium carbonate (1.60 g, 14.70 mmol), In a mixture of bistriphenylphosphonium palladium dichloride (190 mg), the reaction was stirred at 80 ° C. for 2 hours under the protection of nitrogen. Cooled to room temperature, filtered through celite, washed the filter cake with dichloromethane, washed the filtrate with saturated brine, dried over anhydrous sodium sulfate, concentrated the filtrate under reduced pressure, and concentrated the solution for column separation (eluent: petroleum ether / ethyl acetate Ester (v / v) = 3: 1) gave 1.26 g of a white solid. The yield was 69.6%. LC-MS (APCI): m / z = 309.1 (M + 1) + .
步骤4 化合物11的合成Step 4 Synthesis of Compound 11
氮气保护下,将Pd(OAc) 2(120mg)和Xantphos(200mg)加入到化合物9(1.26g,4.09mmol),化合物10(900mg,3.41mmol)和碳酸铯(2.70g,8.40mmol)的无水DMF(20mL)中,反应液氮气保护下100℃下反应过夜,冷却至室温,加水淬灭反应,乙酸乙酯(50mL x 3)萃取,合并有机相,饱和食盐水(50mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到1.40g黄色固体。LC-MS(APCI):m/z=541.2(M+1) +Under nitrogen, Pd (OAc) 2 (120 mg) and Xantphos (200 mg) were added to compound 9 (1.26 g, 4.09 mmol), compound 10 (900 mg, 3.41 mmol), and cesium carbonate (2.70 g, 8.40 mmol). In water DMF (20mL), the reaction solution was reacted at 100 ° C overnight under the protection of nitrogen, cooled to room temperature, quenched by adding water, extracted with ethyl acetate (50mL x 3), combined with organic phases, washed with saturated brine (50mL), and anhydrous. After drying over sodium sulfate, the solvent was removed, and the concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 10: 1) to obtain 1.40 g of a yellow solid. LC-MS (APCI): m / z = 541.2 (M + 1) + .
步骤5 化合物12的合成Step 5 Synthesis of Compound 12
将还原铁粉(870mg,15.60mmol)和氯化铵(416mg,7.80mmol)加入到化合物11的乙醇和水(16mL/4mL)混合溶液中,反应回流反应2hrs,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1),得到600mg黄色固体,两步收率34.5%。LC-MS(APCI):m/z=511.2(M+1) +Reduced iron powder (870 mg, 15.60 mmol) and ammonium chloride (416 mg, 7.80 mmol) were added to a mixed solution of compound 11 in ethanol and water (16 mL / 4 mL). The reaction was refluxed for 2 hrs, filtered through Celite, and the filtrate was decompressed It was concentrated, and the concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 8: 1) to obtain 600 mg of a yellow solid with a yield of 34.5% in two steps. LC-MS (APCI): m / z = 511.2 (M + 1) + .
步骤6 化合物I-3的合成Step 6 Synthesis of Compound I-3
氮气保护下,将化合物12(600mg,1.10mmol)溶于30mL无水二氯甲烷中,逐滴滴加DIPEA(0.46mL,2.80mmol),滴加完成后将反应体系冷却至-10℃,在该温度下缓慢加入丙烯酰氯(100mg,1.10mmol),搅拌1.0hrs。加水,二氯甲烷(50mL x 3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1)得到350mg,收率为56.3%。HPLC:95.92%。LC-MS(APCI):m/z=565.2(M+1) +1H NMR(400MHz,DMSO-d6)δ10.03(s,1H),8.91(s,1H),8.49(d,J=5.3Hz,1H),8.22(d,J=3.0Hz,2H),7.57(d,J=5.3Hz,1H),7.02(s,1H),6.51(br,1H),6.29–6.22(m,1H),5.76–5.72(m,1H),4.87–4.78(m,1H),3.86(s,3H),3.04–2.88(m,2H),2.69(s,3H),2.63–2.56(m,2H),2.27(s,6H),1.58(d,J=6.9Hz,6H). Under nitrogen protection, compound 12 (600 mg, 1.10 mmol) was dissolved in 30 mL of anhydrous dichloromethane, and DIPEA (0.46 mL, 2.80 mmol) was added dropwise. After the dropwise addition was completed, the reaction system was cooled to -10 ° C. Acryloyl chloride (100 mg, 1.10 mmol) was slowly added at this temperature and stirred for 1.0 hrs. Water was added, and dichloromethane (50 mL x 3) was extracted. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the filtrate was concentrated under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 10: 1) 350 mg was obtained with a yield of 56.3%. HPLC: 95.92%. LC-MS (APCI): m / z = 565.2 (M + 1) + , 1 H NMR (400MHz, DMSO-d6) δ 10.03 (s, 1H), 8.91 (s, 1H), 8.49 (d, J = 5.3Hz, 1H), 8.22 (d, J = 3.0Hz, 2H), 7.57 (d, J = 5.3Hz, 1H), 7.02 (s, 1H), 6.51 (br, 1H), 6.29–6.22 (m , 1H), 5.76--5.72 (m, 1H), 4.87--4.78 (m, 1H), 3.86 (s, 3H), 3.04--2.88 (m, 2H), 2.69 (s, 3H), 2.63--2.56 (m , 2H), 2.27 (s, 6H), 1.58 (d, J = 6.9Hz, 6H).
实施例4 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-(甲基-d 3)-1H-苯并[d] Example 4 N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -5-((4- (4-fluoro-1-isopropyl-2- (methyl -d 3 ) -1H-benzo [d] 咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物I-4)的制备。Preparation of imidazole-6-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-4).
Figure PCTCN2019088749-appb-000011
Figure PCTCN2019088749-appb-000011
根据实施例3中所述的合成方法,用以下步骤代替实施例3中的步骤1:According to the synthesis method described in Example 3, the following steps are used to replace Step 1 in Example 3:
将5-溴-N 1-异丙基苯-1,2-二胺(3.00m,13.10mmol)加入到冰醋酸-d 4(15mL)中,反应在回流反应2hrs。冷却至室温,减压除去醋酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50ml x 3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1)得到2.68g棕红色油状化合物6-溴-1-异丙基-2-(甲基-d 3)-1H-苯并[d]咪唑,收率为:80.22%,LC-MS(APCI):m/z=256.1(M+1) +5-Bromo-N 1 -isopropylbenzene-1,2-diamine (3.00m, 13.10mmol) was added to glacial acetic acid-d 4 (15mL), and the reaction was carried out at reflux for 2hrs. Cool to room temperature, remove acetic acid under reduced pressure, adjust the residue to pH 7 with saturated sodium bicarbonate solution, extract with dichloromethane (50ml x 3), wash the combined organic layers with saturated brine, dry over anhydrous sodium sulfate, and concentrate the filtrate under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 20: 1) to obtain 2.68 g of a 6-bromo-1-isopropyl-2- (methyl-d) compound as a brown-red oil. 3 ) -1H-benzo [d] imidazole, yield: 80.22%, LC-MS (APCI): m / z = 256.1 (M + 1) + .
用上述步骤得到的化合物6-溴-1-异丙基-2-(甲基-d 3)-1H-苯并[d]咪唑替代实施例3中步骤2中的化合物7,再重复实施例3中的步骤2-6,制备得到100mg化合物I-4,收率为68.3%。HPLC:95.63%。LC-MS(APCI):m/z=564.0(M+1) +1H NMR(400MHz,CDCl 3)δ9.95(s,1H),9.64(s,1H),8.54(d,J=5.2Hz,1H),8.05(s,1H),7.81(d,J=11.1Hz,1H),7.68(s,1H),7.15(d,J=5.2Hz,1H),6.79(s,1H),6.48(s,2H),5.75–5.67(m,1H),4.84–4.72(m,1H),3.91(s,3H),3.03–2.93(m,2H),2.72(s,3H),2.51–2.42(m,2H),2.38(s,6H),1.68(d,J=7.0Hz,6H). 1H NMR(500MHz,DMSO-d 6):δ9.97(s,1H),8.89(s,1H),8.46(s,1H),8.25–8.08(m,2H),7.89(d,J=12.8Hz,1H),7.63–7.44(m,1H),6.99(s,1H),6.68–6.36(m,1H),6.23(d,J=16.5Hz,1H),5.72(d,J=8.5Hz,1H),4.87–4.72(m,1H),3.84(s,3H),3.03–2.88(m,2H),2.66(s,3H),2.61–2.51(m,2H),2.33(s,6H),1.56(d,J=5.8Hz,6H). Use the compound 6-bromo-1-isopropyl-2- (methyl-d 3 ) -1H-benzo [d] imidazole obtained in the above step to replace the compound 7 in step 2 in Example 3, and repeat the example. Step 2-6 in 3, 100 mg of compound I-4 was prepared, and the yield was 68.3%. HPLC: 95.63%. LC-MS (APCI): m / z = 564.0 (M + 1) + , 1 H NMR (400MHz, CDCl 3 ) δ9.95 (s, 1H), 9.64 (s, 1H), 8.54 (d, J = 5.2Hz, 1H), 8.05 (s, 1H), 7.81 (d, J = 11.1Hz, 1H), 7.68 (s, 1H), 7.15 (d, J = 5.2Hz, 1H), 6.79 (s, 1H) , 6.48 (s, 2H), 5.75--5.67 (m, 1H), 4.84--4.72 (m, 1H), 3.91 (s, 3H), 3.03--2.93 (m, 2H), 2.72 (s, 3H), 2.51 --2.42 (m, 2H), 2.38 (s, 6H), 1.68 (d, J = 7.0Hz, 6H). 1 H NMR (500MHz, DMSO-d 6 ): δ9.97 (s, 1H), 8.89 ( s, 1H), 8.46 (s, 1H), 8.25–8.08 (m, 2H), 7.89 (d, J = 12.8 Hz, 1H), 7.63--7.44 (m, 1H), 6.99 (s, 1H), 6.68 --6.36 (m, 1H), 6.23 (d, J = 16.5Hz, 1H), 5.72 (d, J = 8.5Hz, 1H), 4.87--4.72 (m, 1H), 3.84 (s, 3H), 3.03-- 2.88 (m, 2H), 2.66 (s, 3H), 2.61-2.51 (m, 2H), 2.33 (s, 6H), 1.56 (d, J = 5.8Hz, 6H).
实施例5 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪Example 5 N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -5-((4- (4-fluoro-1-isopropyl-2-methyl- 1H-benzo [d] imid 唑-6-基-5-d)嘧啶-2-基)氨基)-4-(甲氧基-d 3)苯基)丙烯酰胺(化合物I-5)的制备。 Preparation of azole-6-yl-5-d) pyrimidin-2-yl) amino) -4- (methoxy-d 3 ) phenyl) acrylamide (Compound I-5).
Figure PCTCN2019088749-appb-000012
Figure PCTCN2019088749-appb-000012
采用以下路线进行合成:Use the following route for synthesis:
Figure PCTCN2019088749-appb-000013
Figure PCTCN2019088749-appb-000013
步骤1 化合物14的合成Step 1 Synthesis of Compound 14
室温下,将pTSA·H 2O(342mg 1.80mmol)加入到化合物13(450mg,1.50mmol)和4-氟-2-(甲氧基-d 3)-5-硝基苯胺(312mg,1.65mmol)的2-戊醇(10mL)溶液中,反应在是115℃下反应10hrs,冷却至室温,固体过滤,用CH 3CN(3mL)洗涤固体。固体加入到10mL水中的,用浓氨水调pH至碱性,搅拌2.5hrs。固体过滤,用H 2O(30mL)洗涤。真空70℃干燥的600mg土灰色固体。收率:87.33%。LC-MS(APCI)=458.0(M+1) +At room temperature, pTSA · H 2 O (342 mg 1.80 mmol) was added to compound 13 (450 mg, 1.50 mmol) and 4-fluoro-2- (methoxy-d 3 ) -5-nitroaniline (312 mg, 1.65 mmol In a solution of 2-pentanol (10 mL), the reaction was performed at 115 ° C. for 10 hrs, cooled to room temperature, the solid was filtered, and the solid was washed with CH 3 CN (3 mL). Add the solid to 10mL of water, adjust the pH to alkaline with concentrated ammonia water, and stir for 2.5hrs. The solid was filtered, washed with H 2 O (30mL). 600 mg earthy gray solid dried under vacuum at 70 ° C. Yield: 87.33%. LC-MS (APCI) = 458.0 (M + 1) + .
步骤2 化合物15的合成Step 2 Synthesis of Compound 15
室温下,将K 2CO 3(365mg,2.62mmol)加入到化合物14(600mg,1.31mmol)和N 1,N 1,N 2-三甲基乙烷-1,2-二胺(161mg,1.58mmol)的CH 3CN(15mL)溶液中,反应升温至回流搅拌5hrs,减压除去反应液得红色固体。固体用H 2O(30mL)和DCM(30mL x 3)萃取,有机层用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩得到600mg棕红色固体,收率:84.82%。LC-MS(APCI)=540.0(M+1) +K 2 CO 3 (365 mg, 2.62 mmol) was added to compound 14 (600 mg, 1.31 mmol) and N 1 , N 1 , N 2 -trimethylethane-1,2-diamine (161 mg, 1.58) at room temperature. In a CH 3 CN (15 mL) solution, the reaction was heated to reflux and stirred for 5 hrs. The reaction solution was removed under reduced pressure to obtain a red solid. The solid was extracted with H 2 O (30 mL) and DCM (30 mL x 3). The organic layer was washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain 600 mg of a brown-red solid. Yield: 84.82%. LC-MS (APCI) = 540.0 (M + 1) + .
步骤3 化合物16的合成Step 3 Synthesis of Compound 16
室温下将Pd/C(5g)加入到化合物15(600mg,1.11mmol)的甲醇(30mL)溶液中,氢气置换空气三次,反应在是室温下反应过夜。硅藻土过滤,用MeOH洗涤滤饼,滤液减压浓缩得到450mg黄色固体,收率:79.65%。LC-MS(APCI)=510.2(M+1) +Pd / C (5 g) was added to a solution of compound 15 (600 mg, 1.11 mmol) in methanol (30 mL) at room temperature, and the air was replaced with hydrogen three times. The reaction was allowed to react at room temperature overnight. Filter through celite, wash the filter cake with MeOH, and concentrate the filtrate under reduced pressure to obtain 450 mg of a yellow solid. Yield: 79.65%. LC-MS (APCI) = 510.2 (M + 1) + .
步骤4 化合物I-5的合成Step 4 Synthesis of Compound I-5
氮气保护下,将化合物15(450mg,0.88mmol)溶于30mL无水二氯甲烷中,逐滴滴加DIPEA(0.35mL,1.76mmol),滴加完成后将反应体系冷却至-10℃,在该温度下缓慢加入丙烯酰氯(1.42 mL,0.88mmol,0.618mmol/mL),搅拌1.0hrs。加水,二氯甲烷(50mL x 3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1)得到300mg,收率为60.4%。HPLC:96.28%。LC-MS(APCI):m/z=564.3(M+1) +1H NMR(300MHz,DMSO-d 6)δ10.06(s,1H),8.94(s,1H),8.46(d,J=3.5Hz,1H),8.20(s,2H),7.90(d,J=11.8Hz,1H),7.54(s,1H),6.97(s,1H),6.51(s,1H),6.22(d,J=16.8Hz,1H),5.71(d,J=11.2Hz,1H),4.87–4.69(m,1H),2.98–2.83(m,2H),2.65(s,3H),2.58(s,3H),2.46–2.34(m,2H),2.26(s,6H),1.53(d,J=5.5Hz,6H). Under nitrogen protection, compound 15 (450 mg, 0.88 mmol) was dissolved in 30 mL of anhydrous dichloromethane, and DIPEA (0.35 mL, 1.76 mmol) was added dropwise. After the dropwise addition was completed, the reaction system was cooled to -10 ° C. Acryloyl chloride (1.42 mL, 0.88 mmol, 0.618 mmol / mL) was slowly added at this temperature, and stirred for 1.0 hrs. Water was added, and dichloromethane (50 mL x 3) was extracted. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the filtrate was concentrated under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 10: 1) 300 mg was obtained with a yield of 60.4%. HPLC: 96.28%. LC-MS (APCI): m / z = 564.3 (M + 1) + , 1 H NMR (300MHz, DMSO-d 6 ) δ 10.06 (s, 1H), 8.94 (s, 1H), 8.46 (d, J = 3.5Hz, 1H), 8.20 (s, 2H), 7.90 (d, J = 11Hz, 1H), 7.54 (s, 1H), 6.97 (s, 1H), 6.51 (s, 1H), 6.22 ( d, J = 16.8 Hz, 1H), 5.71 (d, J = 11.2 Hz, 1H), 4.87–4.69 (m, 1H), 2.98–2.83 (m, 2H), 2.65 (s, 3H), 2.58 (s , 3H), 2.46--2.34 (m, 2H), 2.26 (s, 6H), 1.53 (d, J = 5.5Hz, 6H).
实施例6 N-(5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基Example 6 N- (5-((4- (4-fluoro-1-isopropyl-2-methyl-1H-benzo [d] imidazol-6-yl) pyrimidin-2-yl) amino)- 4-methoxy -2-(甲基(2-(甲基(甲基-d 3)氨基)乙基)氨基)苯基)丙烯酰胺(化合物I-6)的制备。 Preparation of 2- (methyl (2- (methyl (methyl-d 3 ) amino) ethyl) amino) phenyl) acrylamide (Compound I-6).
Figure PCTCN2019088749-appb-000014
Figure PCTCN2019088749-appb-000014
采用以下路线进行合成:Use the following route for synthesis:
Figure PCTCN2019088749-appb-000015
Figure PCTCN2019088749-appb-000015
步骤1 化合物18的合成Step 1 Synthesis of Compound 18
室温下,将K 2CO 3(1.52g,11.0mmol)加入到化合物17(2.50g,5.50mmol)和N 1,N 1,N 2-三甲基乙烷-1,2-二胺(600mg,6.60mmol)的CH 3CN(40mL)溶液中,反应升温至回流搅拌过夜,减压除去反应液得红色固体。固体用H 2O(100mL)打浆搅拌3hrs,固体过滤,再用冷乙醇(3mL x 2)洗涤固体,真空下55℃干燥得到红色固体(2.5g),收率:87.01%。LC-MS(APCI):m/z=523.3(M+1) +K 2 CO 3 (1.52 g, 11.0 mmol) was added to compound 17 (2.50 g, 5.50 mmol) and N 1 , N 1 , N 2 -trimethylethane-1,2-diamine (600 mg) at room temperature. , 6.60 mmol) in a solution of CH 3 CN (40 mL), the reaction was heated to reflux and stirred overnight, and the reaction solution was removed under reduced pressure to obtain a red solid. The solid was slurried with H 2 O (100 mL) and stirred for 3 hrs. The solid was filtered, and the solid was washed with cold ethanol (3 mL x 2) and dried under vacuum at 55 ° C to obtain a red solid (2.5 g). Yield: 87.01%. LC-MS (APCI): m / z = 523.3 (M + 1) + .
步骤2 化合物19的合成Step 2 Synthesis of Compound 19
室温下,将K 2CO 3(276mg,2.00mmol)加入到化合物18(523mg,1.0mmol)和TsO-CD 3(208mg,1.10mmol)的CH 3CN(40mL)溶液中,反应升温至50℃下搅拌过夜,减压除去反应液得红色固体。固体用H 2O(100mL)打浆搅拌3hrs,固体过滤,再用冷乙醇(2mL x 2)洗涤固体,真空下55℃干燥得到红色固体(200mg),收率:37.01%。LC-MS(APCI):m/z=540.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ8.72(s,1H),8.52(d,J=5.3Hz,1H),8.32(s,1H),8.22(s,1H),7.83(d,J=12.2Hz,1H),7.61(d,J=5.3Hz,1H),6.85(s,1H),4.89–4.79(m,1H),3.96(s,3H),3.30–3.26(m,2H),2.84(s,3H),2.62(s,3H),2.61–2.55(m,2H),2.25(s,3H),1.58(d,J=6.9Hz,6H). K 2 CO 3 (276 mg, 2.00 mmol) was added to a solution of compound 18 (523 mg, 1.0 mmol) and TsO-CD 3 (208 mg, 1.10 mmol) in CH 3 CN (40 mL) at room temperature, and the reaction was warmed to 50 ° C. After stirring overnight, the reaction solution was removed under reduced pressure to obtain a red solid. The solid was slurried with H 2 O (100 mL) and stirred for 3 hrs. The solid was filtered, and the solid was washed with cold ethanol (2 mL x 2) and dried under vacuum at 55 ° C. to obtain a red solid (200 mg). Yield: 37.01%. LC-MS (APCI): m / z = 540.3 (M + 1) + ; 1 H NMR (400MHz, DMSO-d 6 ) δ 8.72 (s, 1H), 8.52 (d, J = 5.3Hz, 1H) , 8.32 (s, 1H), 8.22 (s, 1H), 7.83 (d, J = 12.2 Hz, 1H), 7.61 (d, J = 5.3 Hz, 1H), 6.85 (s, 1H), 4.89--4.79 ( m, 1H), 3.96 (s, 3H), 3.30--3.26 (m, 2H), 2.84 (s, 3H), 2.62 (s, 3H), 2.61--2.55 (m, 2H), 2.25 (s, 3H) 1.58 (d, J = 6.9 Hz, 6H).
步骤3 化合物20的合成Step 3 Synthesis of Compound 20
室温下将Pd/C(50mg)加入到化合物19(200mg)的甲醇(30mL)溶液中,氢气置换空气三次,反应在是室温下反应过夜。硅藻土过滤,用二氯甲烷洗涤滤饼,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12)得到120mg,收率为65%。LC-MS(APCI):m/z=510.2(M+1) +Pd / C (50 mg) was added to a solution of compound 19 (200 mg) in methanol (30 mL) at room temperature, and the air was replaced with hydrogen three times. The reaction was allowed to react at room temperature overnight. The celite was filtered, the filter cake was washed with dichloromethane, the filtrate was concentrated under reduced pressure, and the concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 12) to obtain 120 mg with a yield of 65%. LC-MS (APCI): m / z = 510.2 (M + 1) + .
步骤4 化合物I-6的合成Step 4 Synthesis of Compound I-6
氮气保护下,将化合物20(120mg,0.23mmol)溶于30mL无水二氯甲烷中,逐滴滴加DIPEA(0.08mL,0.5mmol),滴加完成后将反应体系冷却至-20℃,在该温度下缓慢加入丙烯酰氯(0.37mL,0.23mmol,0.618mmol/mL),搅拌1.0hrs。加水,二氯甲烷(30mL x 3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=9%)得到75mg,收率为%。HPLC:96.55%。LC-MS(APCI):m/z=564.3(M+1) +Under nitrogen protection, compound 20 (120 mg, 0.23 mmol) was dissolved in 30 mL of anhydrous dichloromethane, and DIPEA (0.08 mL, 0.5 mmol) was added dropwise. After the dropwise addition was completed, the reaction system was cooled to -20 ° C. Acryloyl chloride (0.37 mL, 0.23 mmol, 0.618 mmol / mL) was slowly added at this temperature, and stirred for 1.0 hrs. Water was added, and dichloromethane (30 mL x 3) was extracted. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the filtrate was concentrated under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 9%) 75 mg was obtained in a yield of%. HPLC: 96.55%. LC-MS (APCI): m / z = 564.3 (M + 1) + .
实施例7 N-(2-((2-(双(甲基-d 2)氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d] Example 7 N- (2-((2- (bis (methyl-d 2) amino) ethyl) (methyl) amino) -5-((4- (4-fluoro-1-isopropyl- 2-methyl-1H-benzo [d] 咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物I-7)的制备。Preparation of imidazole-6-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound 1-7).
Figure PCTCN2019088749-appb-000016
Figure PCTCN2019088749-appb-000016
采用以下路线进行合成:Use the following route for synthesis:
Figure PCTCN2019088749-appb-000017
Figure PCTCN2019088749-appb-000017
步骤1 化合物21的合成Step 1 Synthesis of Compound 21
室温下,将K 2CO 3(920mg,6.60mmol)加入到化合物17(1.50g,3.30mmol)和叔丁基(2-(甲氨基)乙基)氨基甲酸酯(700mg,3.96mmol)的CH 3CN(40mL)溶液中,反应升温至回流搅拌过夜,减压除去反应液得红色固体。固体用H 2O(100mL)打浆搅拌3hrs,固体过滤,再用冷乙醇(3mL x 2)洗涤固体,真空下55℃干燥得到红色固体(2.5g),收率:87.01%。LC-MS(APCI):m/z=609.2(M+1) +K 2 CO 3 (920 mg, 6.60 mmol) was added to compound 17 (1.50 g, 3.30 mmol) and tert-butyl (2- (methylamino) ethyl) carbamate (700 mg, 3.96 mmol) at room temperature. In CH 3 CN (40 mL) solution, the reaction was heated to reflux and stirred overnight. The reaction solution was removed under reduced pressure to obtain a red solid. The solid was slurried with H 2 O (100 mL) and stirred for 3 hrs. The solid was filtered, and the solid was washed with cold ethanol (3 mL x 2) and dried under vacuum at 55 ° C. to obtain a red solid (2.5 g). Yield: 87.01%. LC-MS (APCI): m / z = 609.2 (M + 1) + .
步骤2 化合物22的合成Step 2 Synthesis of Compound 22
将三氟乙酸(10mL)加入到化合物21(1.70g,2.79mmol)的二氯甲烷(20mL)溶液中,反应在是温室下搅拌1hr。减压浓缩除去反应液,再用饱和碳酸氢钠溶液调pH值为碱性,加水(30mL)搅拌过夜。固体过滤,用水洗涤固体,再用冷乙醇(3mL x 2)洗涤,真空下55℃干燥得到红色固体1.1g,收率:77.44%。LC-MS(APCI):m/z=509.2(M+1) +Trifluoroacetic acid (10 mL) was added to a solution of compound 21 (1.70 g, 2.79 mmol) in dichloromethane (20 mL), and the reaction was stirred in a greenhouse for 1 hr. The reaction solution was concentrated under reduced pressure to remove the reaction solution, and the pH was adjusted to be alkaline with a saturated sodium bicarbonate solution. Water (30 mL) was added and stirred overnight. The solid was filtered, the solid was washed with water, and then washed with cold ethanol (3 mL x 2), and dried under vacuum at 55 ° C to obtain 1.1 g of a red solid, yield: 77.44%. LC-MS (APCI): m / z = 509.2 (M + 1) + .
步骤3 化合物23的合成Step 3 Synthesis of Compound 23
冰浴下,将氘代甲醛(1mL,20%w)加入到化合物22(600mg,1.18mmol)的氘代氯仿(15mL)和氘代甲醇(5mL)的混合物中,反应在冰浴下继续搅拌30min,加入三乙酰基硼氢化钠(525mg,2.48mmol),反应在室温下过夜。用饱和碳酸氢钠溶液淬灭反应,减压除去甲醇,水层用二氯甲烷(50mL x3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8%)得到220mg,收率为34.49%。LC-MS(APCI):m/z=541.2(M+1)+。In an ice bath, deuterated formaldehyde (1 mL, 20% w) was added to a mixture of deuterated chloroform (15 mL) and deuterated methanol (5 mL) of compound 22 (600 mg, 1.18 mmol), and the reaction was continued stirring in the ice bath For 30 min, sodium triacetylborohydride (525 mg, 2.48 mmol) was added and the reaction was allowed to proceed at room temperature overnight. The reaction was quenched with a saturated sodium bicarbonate solution, and the methanol was removed under reduced pressure. The aqueous layer was extracted with dichloromethane (50 mL × 3). The organic layers were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. Column separation (eluent: dichloromethane / methanol (v / v) = 8%) gave 220 mg with a yield of 34.49%. LC-MS (APCI): m / z = 541.2 (M + 1) +.
步骤4 化合物24的合成Step 4 Synthesis of Compound 24
室温下将Pd/C(50mg)加入到化合物23(220mg,0.40mmol)的甲醇(30mL)溶液中,氢气置换空气三次,反应在是室温下反应1.5hrs。硅藻土过滤,用二氯甲烷洗涤滤饼,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=14%)得到180mg,收率为86.62%。LC-MS(APCI):m/z=511.2(M+1) +Pd / C (50 mg) was added to a solution of compound 23 (220 mg, 0.40 mmol) in methanol (30 mL) at room temperature, and the air was replaced with hydrogen three times. The reaction was performed at room temperature for 1.5 hrs. Filter through diatomaceous earth, wash the filter cake with dichloromethane, and concentrate the filtrate under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 14%) to obtain 180 mg with a yield of 86.62%. . LC-MS (APCI): m / z = 511.2 (M + 1) + .
步骤5 化合物I-7的合成Step 5 Synthesis of Compound I-7
氮气保护下,将化合物24(180mg,0.35mmol)溶于30mL无水二氯甲烷中,逐滴滴加DIPEA(0.11mL,0.71mmol),滴加完成后将反应体系冷却至-20℃,在该温度下缓慢加入丙烯酰氯(0.57mL,0.35mmol,0.618mmol/mL),搅拌1.0hrs。加水,二氯甲烷(30mL x 3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=9%)得到145mg,收率为72.85%。HPLC:95.36%。LC-MS(APCI):m/z=565.4(M+1) +Under nitrogen protection, compound 24 (180 mg, 0.35 mmol) was dissolved in 30 mL of anhydrous dichloromethane, and DIPEA (0.11 mL, 0.71 mmol) was added dropwise. After the dropwise addition was completed, the reaction system was cooled to -20 ° C. Acryloyl chloride (0.57 mL, 0.35 mmol, 0.618 mmol / mL) was slowly added at this temperature, and stirred for 1.0 hrs. Water was added, and dichloromethane (30 mL x 3) was extracted. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the filtrate was concentrated under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 9%) yielded 145 mg with a yield of 72.85%. HPLC: 95.36%. LC-MS (APCI): m / z = 565.4 (M + 1) + .
实施例8  N-(2-((2-(双(甲基-d 3)氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d] 咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物I-8)的制备。 Example 8 N- (2-((2- (bis (methyl-d 3 ) amino) ethyl) (methyl) amino) -5-((4- (4-fluoro-1-isopropyl- Preparation of 2-methyl-1H-benzo [d] imidazol-6-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-8).
Figure PCTCN2019088749-appb-000018
Figure PCTCN2019088749-appb-000018
采用以下路线进行合成:Use the following route for synthesis:
Figure PCTCN2019088749-appb-000019
Figure PCTCN2019088749-appb-000019
步骤1 化合物25的合成Step 1 Synthesis of Compound 25
冰浴下,将氘代甲醛(1mL,20%w)加入到化合物22(400mg,0.78mmol)的氘代氯仿(15mL)和氘代甲醇(10mL)的混合物中,反应在冰浴下继续搅拌30min。加入到氰基硼氘化钠(104mg,1.57mmol),反应在室温下过夜。用饱和碳酸氢钠溶液淬灭反应,减压除去甲醇,水层用二氯甲烷(50mL x3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8%)得到180mg,收率为42.17%。LC-MS(APCI):m/z=543.2(M+1) +In an ice bath, deuterated formaldehyde (1 mL, 20% w) was added to a mixture of deuterated chloroform (15 mL) and deuterated methanol (10 mL) of compound 22 (400 mg, 0.78 mmol). The reaction was continued to stir in the ice bath 30min. Added to sodium cyanoborodeuteride (104 mg, 1.57 mmol) and reacted at room temperature overnight. The reaction was quenched with a saturated sodium bicarbonate solution, and the methanol was removed under reduced pressure. The aqueous layer was extracted with dichloromethane (50 mL x 3). The organic layers were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. Column separation (eluent: dichloromethane / methanol (v / v) = 8%) gave 180 mg with a yield of 42.17%. LC-MS (APCI): m / z = 543.2 (M + 1) + .
步骤2 化合物26的合成Step 2 Synthesis of Compound 26
室温下将Pd/C(50mg)加入到化合物25(180mg,0.33mmol)的甲醇(30mL)溶液中,氢气置换空气三次,反应在是室温下反应1.5hrs。硅藻土过滤,用二氯甲烷洗涤滤饼,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=14%)得到140mg,收率为82.33%。LC-MS(APCI):m/z=513.2(M+1) +Pd / C (50 mg) was added to a solution of compound 25 (180 mg, 0.33 mmol) in methanol (30 mL) at room temperature, hydrogen was used to replace the air three times, and the reaction was performed at room temperature for 1.5 hrs. Filtered with celite, washed the filter cake with dichloromethane, and concentrated the filtrate under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 14%) to obtain 140 mg with a yield of 82.33%. . LC-MS (APCI): m / z = 513.2 (M + 1) + .
步骤3 化合物I-8的合成Step 3 Synthesis of Compound I-8
氮气保护下,将化合物26(140mg,0.26mmol)溶于30mL无水二氯甲烷中,逐滴滴加DIPEA(0.10mL,0.54mmol),滴加完成后将反应体系冷却至-20℃,在该温度下缓慢加入丙烯酰氯(0.42mL,0.26mmol,0.618mmol/mL),搅拌1.0hrs。加水,二氯甲烷(30mL x 3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=9%)得到50mg,收率为33.50%。HPLC:99.24%。LC-MS(APCI):m/z=567.5(M+1) +1H NMR(300MHz,DMSO-d 6):δ10.04(s,1H),8.91(s,1H),8.49(d,J=5.2Hz,1H),8.22(d,J=1.5Hz,2H),7.92(d,J =12.5Hz,1H),7.57(d,J=5.3Hz,1H),7.02(s,1H),6.68–6.45(m,1H),6.26(dd,J=17.0,1.9Hz,1H),5.88–5.67(m,1H),4.83(dt,J=14.0,6.9Hz,1H),3.86(s,3H),3.05–2.91(m,2H),2.69(s,3H),2.62(s,3H),2.52–2.50(m,2H),1.58(d,J=6.9Hz,6H). Under the protection of nitrogen, compound 26 (140 mg, 0.26 mmol) was dissolved in 30 mL of anhydrous dichloromethane, and DIPEA (0.10 mL, 0.54 mmol) was added dropwise. After the dropwise addition was completed, the reaction system was cooled to -20 ° C. Acryloyl chloride (0.42 mL, 0.26 mmol, 0.618 mmol / mL) was slowly added at this temperature, and stirred for 1.0 hrs. Water was added, and dichloromethane (30 mL x 3) was extracted. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the filtrate was concentrated under reduced pressure. The concentrated solution was subjected to column separation (eluent: dichloromethane / methanol (v / v) = 9%) 50 mg was obtained with a yield of 33.50%. HPLC: 99.24%. LC-MS (APCI): m / z = 567.5 (M + 1) + ; 1 H NMR (300MHz, DMSO-d 6 ): δ 10.04 (s, 1H), 8.91 (s, 1H), 8.49 (d , J = 5.2 Hz, 1H), 8.22 (d, J = 1.5 Hz, 2H), 7.92 (d, J = 12.5 Hz, 1H), 7.57 (d, J = 5.3 Hz, 1H), 7.02 (s, 1H ), 6.68--6.45 (m, 1H), 6.26 (dd, J = 17.0, 1.9 Hz, 1H), 5.88--5.67 (m, 1H), 4.83 (dt, J = 14.0, 6.9 Hz, 1H), 3.86 ( s, 3H), 3.05--2.91 (m, 2H), 2.69 (s, 3H), 2.62 (s, 3H), 2.52--2.50 (m, 2H), 1.58 (d, J = 6.9Hz, 6H).
生物活性测试。Biological activity test.
(1)激酶抑制作用(1) Kinase inhibition
试剂和耗材:Reagents and consumables:
WT EGFR(Carna,目录号08-115),EGFR[L858R](Carna,目录号08-502),EGFR[L858R/T790M](Carna,目录号08-510),ATP(Sigma,目录号A7699-1G),DMSO(Sigma,目录号D2650),96孔板(Corning,目录号3365),384孔板(Greiner,目录号784076),HTRF Kinase TK试剂盒(Cisbio,目录号62TK0PEJ),厄洛替尼(Selleckchem,目录号S7787),EGFR[d746-750](Life Technologies,目录号PV6178),5x激酶缓冲液A(Life Technologies,目录号PV3186),激酶示踪剂199(Life Technologies,目录号PV5830),
Figure PCTCN2019088749-appb-000020
Eu-anti-GST抗体(Life Technologies,目录号PV5594)。
WT EGFR (Carna, Catalog No. 08-115), EGFR [L858R] (Carna, Catalog No. 08-502), EGFR [L858R / T790M] (Carna, Catalog No. 08-510), ATP (Sigma, Catalog No. A7699- 1G), DMSO (Sigma, catalog number D2650), 96-well plate (Corning, catalog number 3365), 384-well plate (Greiner, catalog number 784076), HTRF Kinase TK kit (Cisbio, catalog number 62TK0PEJ), erlotine (Selleckchem, catalog number S7787), EGFR [d746-750] (Life Technologies, catalog number PV6178), 5x kinase buffer A (Life Technologies, catalog number PV3186), kinase tracer 199 (Life Technologies, catalog number PV5830) ),
Figure PCTCN2019088749-appb-000020
Eu-anti-GST antibody (Life Technologies, catalog number PV5594).
具体实验方法:Specific experimental methods:
化合物配制:将受试化合物溶于DMSO配成20mM母液。然后,在DMSO中等梯度3倍稀释,稀释十次。加药时再用缓冲液稀释10倍。Compound formulation: The test compound was dissolved in DMSO to prepare a 20 mM mother liquor. Then, a 3-fold dilution in DMSO was performed ten times. When adding medicine, dilute it with buffer 10 times.
WT EGFR及EGFR[L858R/T790M]激酶检测:在5x激酶缓冲液A中,将WT EGFR或EGFR[L858R/T790M]激酶与预先稀释配制的不同浓度的化合物混合10分钟,每个浓度双复孔。加入对应底物及ATP,室温反应20分钟(其中设置阴阳性对照:阴性为空白对照,阳性为厄洛替尼)。反应完毕加入检测试剂(HTRF Kinase TK试剂盒内的试剂),室温孵育30分钟后,通过Evnvision酶标仪检测,测定在各浓度的本发明化合物存在下的酶活力,并计算不同浓度的化合物对酶活力的抑制活性,之后根据四参数方程,根据Graphpad 5.0软件对不同浓度化合物下酶活力的抑制活性进行拟合,计算出IC 50值。 Detection of WT EGFR and EGFR [L858R / T790M] kinase: In 5x kinase buffer A, mix WT EGFR or EGFR [L858R / T790M] kinase with a compound of different concentration prepared in a pre-diluted form for 10 minutes, and duplicate the wells at each concentration . Add the corresponding substrate and ATP, and react at room temperature for 20 minutes (wherein a negative positive control is set: a negative is a blank control and a positive is erlotinib). After the reaction, add detection reagents (reagents in the HTRF Kinase TK kit), and incubate for 30 minutes at room temperature. Then use an Evnvision microplate reader to measure the enzyme activity in the presence of each concentration of the compound of the invention, and calculate the compound concentration of different concentrations. Inhibition activity of enzyme activity, then according to the four-parameter equation, Graphpad 5.0 software was used to fit the inhibitory activity of enzyme activity at different concentrations of compounds to calculate the IC 50 value.
在上述激酶抑制实验中测试了本发明化合物,发现与非氘代化合物T相比,本发明化合物对EGFR[L858R/T790M]具有强效的活性以及优于WT EGFR的优异选择性。代表性的实施例化合物的结果归纳于下表1中。The compound of the present invention was tested in the above kinase inhibition experiment, and it was found that the compound of the present invention has a potent activity on EGFR [L858R / T790M] and an excellent selectivity over WT EGFR compared to the non-deuterated compound T. The results for representative example compounds are summarized in Table 1 below.
表1:Table 1:
 Zh EGFR(WT)EGFR (WT) L858R/T790ML858R / T790M
化合物TCompound T 1~51 ~ 5 <1<1
化合物I-1Compound I-1 1~51 ~ 5 <1<1
化合物I-2Compound I-2 1~51 ~ 5 <1<1
化合物I-3Compound I-3 1~51 ~ 5 <1<1
化合物I-4Compound I-4 1~51 ~ 5 <1<1
化合物I-5Compound I-5 1~51 ~ 5 <1<1
化合物I-6Compound I-6 1~51 ~ 5 <1<1
化合物I-7Compound I-7 1~51 ~ 5 <1<1
化合物I-8Compound I-8 1~51 ~ 5 <1<1
(2)细胞毒性实验(2) Cytotoxicity test
采用MTS方法检测了本发明化合物对体外培养的3株肿瘤细胞的体外抗增殖活性。实验结果表明本发明化合物对体外培养的癌细胞的体外增殖具有抑制作用;其中对肺癌细胞的体外增殖的抑制作用比皮肤癌细胞的体外增殖的抑制作用强。The anti-proliferative activity of the compound of the present invention on three tumor cells cultured in vitro was detected 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 inhibitory effect on the proliferation of lung cancer cells in vitro is stronger than the inhibitory effect on the proliferation of skin cancer cells in vitro.
细胞系:Cell line:
皮肤癌细胞A431(购自美国标准生物品收藏中心(ATCC));肺癌细胞NCI-H1975(购自美国标准生物品收藏中心(ATCC))和HCC827(购自美国标准生物品收藏中心(ATCC));均用含10%胎牛血清、100U/ml青霉素、100μg/ml链霉素的RPMI1640培养基培养。Skin cancer cells A431 (purchased from the American Standard Biological Collection (ATCC)); lung cancer cells NCI-H1975 (purchased from the American Standard Biological Collection (ATCC)) and HCC827 (purchased from the American Standard Biological Collection (ATCC)) ); All were cultured in RPMI1640 medium containing 10% fetal bovine serum, 100 U / ml penicillin, and 100 μg / ml streptomycin.
试剂和耗材:Reagents and consumables:
RPMI-1640(GIBCO,目录号A10491-01);胎牛血清(GIBCO,目录号10099141);0.25%胰蛋白酶-EDTA(GIBCO,目录号25200);青霉素-链霉素,液体(GIBCO,目录号15140-122);DMSO(Sigma,目录号D2650);MTS测试试剂盒(Promega,目录号G3581),96孔板(Corning,目录号3365)。RPMI-1640 (GIBCO, catalog number A10491-01); fetal bovine serum (GIBCO, catalog number 10099141); 0.25% trypsin-EDTA (GIBCO, catalog number 25200); penicillin-streptomycin, liquid (GIBCO, catalog number 15140-122); DMSO (Sigma, catalog number D2650); MTS test kit (Promega, catalog number G3581), 96-well plate (Corning, catalog number 3365).
具体实验方法:Specific experimental methods:
化合物配制:受试化合物溶于DMSO配成20mM母液,-20℃保存。使用时用DMSO等梯度3倍稀释,稀释10次。加药时再用细胞培养基RPMI-1640稀释4倍。Compound preparation: The test compound was dissolved in DMSO to prepare a 20 mM mother liquor, and stored at -20 ° C. When using, dilute with DMSO and other gradients 3 times, 10 times. At the time of dosing, it was diluted 4 times with cell culture medium RPMI-1640.
MTS细胞活力检测:0.25%胰蛋白酶-EDTA消化对数生长期细胞,按已优化的密度接种150μl于96孔板,24小时后加入培养基稀释4倍的化合物,50μl/孔(一般选择十个浓度:100、33.3、11.1、3.70、1.23、0.412、0.137、0.0457、0.0152、0.00508μM)。以加入同样体积的0.5%DMSO的孔作为对照。细胞继续培养72小时后,MTS检测细胞活力。MTS cell viability test: 0.25% trypsin-EDTA digested cells in logarithmic growth phase, inoculated 150 μl in a 96-well plate according to the optimized density, 24 hours after adding the medium to dilute the compound 4 times, 50 μl / well Concentration: 100, 33.3, 11.1, 3.70, 1.23, 0.412, 0.137, 0.0457, 0.0152, 0.00508 μM). As a control, the same volume of 0.5% DMSO was added. After the cells were cultured for 72 hours, MTS was used to detect cell viability.
具体操作:贴壁细胞,弃去培养基,每孔加入含20μL MTS和100μL培养基的混合液。放入培 养箱继续培养1-4小时后检测OD490,以OD650值作为参考。用GraphPad Prism软件制作量效曲线并计算IC 50Specific operation: adherent cells, discard the culture medium, and add a mixed solution containing 20 μL MTS and 100 μL culture medium to each well. Put it in the incubator and continue to incubate for 1-4 hours to detect OD490, and use the OD650 value as a reference. Using GraphPad Prism software to make dose-response curves and calculate IC 50.
在上述细胞毒性实验中测试了本发明化合物,发现发现与非氘代的化合物T相比,本发明化合物对肺癌细胞NCI-H1975和HCC827具有强效的活性以及优于皮肤癌细胞A431的优异选择性。代表性的实施例化合物的结果归纳于下表2中。The compound of the present invention was tested in the above cytotoxicity experiment, and it was found that the compound of the present invention has strong activity on lung cancer cells NCI-H1975 and HCC827 and an excellent choice better than skin cancer cell A431 compared with the non-deuterated compound T. Sex. The results of representative example compounds are summarized in Table 2 below.
表2:Table 2:
Figure PCTCN2019088749-appb-000021
Figure PCTCN2019088749-appb-000021
(3)代谢稳定性评价(3) Evaluation of metabolic stability
微粒体实验:人肝微粒体:0.5mg/mL,Xenotech;大鼠肝微粒体:0.5mg/mL,Xenotech;辅酶(NADPH/NADH):1mM,Sigma Life Science;氯化镁:5mM,100mM磷酸盐缓冲剂(pH为7.4)。Microsomal experiments: human liver microsomes: 0.5mg / mL, Xenotech; rat liver microsomes: 0.5mg / mL, Xenotech; coenzyme (NADPH / NADH): 1mM, Sigma Life Science; magnesium chloride: 5mM, 100mM phosphate buffered Agent (pH 7.4).
储备液的配制:精密称取一定量的实施例化合物和对照品化合物的粉末,并用DMSO分别溶解至5mM。Preparation of the stock solution: A certain amount of powder of the example compound and the reference compound was precisely weighed, and dissolved to 5 mM with DMSO, respectively.
磷酸盐缓冲液(100mM,pH7.4)的配制:取预先配好的150mL的0.5M磷酸二氢钾和700mL的0.5M磷酸氢二钾溶液混合,再用0.5M磷酸氢二钾溶液调节混合液pH值至7.4,使用前用超纯水稀释5倍,加入氯化镁,得到磷酸盐缓冲液(100mM),其中含100mM磷酸钾,3.3mM氯化镁,pH为7.4。Preparation of phosphate buffer solution (100 mM, pH 7.4): Take the pre-mixed 150 mL of 0.5 M potassium dihydrogen phosphate and 700 mL of 0.5 M potassium dihydrogen phosphate solution and mix with 0.5 M dipotassium phosphate solution The pH value of the solution was 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer solution (100 mM), which contained 100 mM potassium phosphate, 3.3 mM magnesium chloride, and had a pH of 7.4.
配制NADPH再生系统溶液(含有6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D,3.3mM氯化镁),使用前置于湿冰上。Prepare NADPH regeneration system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U / mL G-6-PD, 3.3mM magnesium chloride), and place on wet ice before use.
配制终止液:含有50ng/mL盐酸普萘洛尔和200ng/mL甲苯磺丁脲(内标)的乙腈溶液。取25057.5μL磷酸盐缓冲液(pH7.4)至50mL离心管中,分别加入812.5μL人肝微粒体,混匀,得到蛋白浓度为0.625mg/mL的肝微粒体稀释液。取25057.5μL磷酸盐缓冲液(pH7.4)至50mL离心管中,分别加入812.5μL SD大鼠肝微粒体,混匀,得到蛋白浓度为0.625mg/mL的肝微粒体稀释液。Preparation of stop solution: an acetonitrile solution containing 50ng / mL propranolol hydrochloride and 200ng / mL tolbutamide (internal standard). Take 25057.5 μL of phosphate buffer solution (pH 7.4) into a 50 mL centrifuge tube, add 812.5 μL of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg / mL. Take 25057.5 μL phosphate buffer solution (pH 7.4) into a 50 mL centrifuge tube, add 812.5 μL SD rat liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg / mL.
样品的孵育:用含70%乙腈的水溶液将相应化合物的储备液分别稀释至0.25mM,作为工作液,备用。分别取398μL的人肝微粒体或者大鼠肝微粒体稀释液加入96孔孵育板中(N=2),分别加入2μL0.25mM的的工作液中,混匀。Sample incubation: Dilute the stock solution of the corresponding compound to 0.25 mM with 70% acetonitrile in water, and use it as a working solution. 398 μL of human liver microsomes or rat liver microsomes dilutions were added to 96-well incubation plates (N = 2), 2 μL of 0.25 mM working solution were added and mixed.
代谢稳定性的测定:在96孔深孔板的每孔中加入300μL预冷的终止液,并置于冰上,作为终止板。将96孔孵育板和NADPH再生系统置于37℃水浴箱中,100转/分钟震荡,预孵5min。从孵育板每孔取出80μL孵育液加入终止板,混匀,补充20μL NADPH再生系统溶液,作为0min样品。再向孵育板每孔加入80μL的NADPH再生系统溶液,启动反应,开始计时。相应化合物的反应浓度为1μM,蛋白浓度为0.5mg/mL。分别于反应10、30、90min时,各取100μL反应液,加入终止板中,涡旋3min终止反应。将终止板于5000×g,4℃条件下离心10min。取100μL上清液至预先加入100μL蒸馏水的96孔板中,混匀,采用LC-MS/MS进行样品分析。Determination of metabolic stability: 300 μL of pre-cooled stop solution was added to each well of a 96-well deep well plate, and placed on ice as a stop plate. Place the 96-well incubation plate and NADPH regeneration system in a 37 ° C water bath, shake at 100 rpm, and pre-incubate for 5 minutes. Take 80 μL of the incubation solution from each well of the incubation plate and add it to the termination plate, mix well, and add 20 μL of the NADPH regeneration system solution as a 0min sample. Add 80 μL of NADPH regeneration system solution to each well of the incubation plate, start the reaction, and start timing. The reaction concentration of the corresponding compound was 1 μM, and the protein concentration was 0.5 mg / mL. When the reaction was 10, 30, and 90 minutes, 100 μL of each reaction solution was taken, added to the stop plate, and vortexed for 3 minutes to stop the reaction. The stop plate was centrifuged at 5000 × g for 10 min at 4 ° C. Take 100 μL of the supernatant into a 96-well plate pre-added with 100 μL of distilled water, mix well, and use LC-MS / MS for sample analysis.
数据分析:通过LC-MS/MS系统检测相应化合物及内标的峰面积,计算化合物与内标峰面积比值。通过化合物剩余量的百分率的自然对数与时间作图测得斜率,并根据以下公式计算t 1/2和CL int,其中V/M即等于1/蛋白浓度。 Data analysis: The peak areas of the corresponding compounds and internal standard were detected by LC-MS / MS system, and the ratio of compound to internal standard peak area was calculated. The slope was measured by plotting the natural logarithm of the percentage of the remaining compound against time, and calculated t 1/2 and CL int according to the following formula, where V / M is equal to 1 / protein concentration.
Figure PCTCN2019088749-appb-000022
t 1/2(min);CL int(μL/min/mg)。
Figure PCTCN2019088749-appb-000022
t 1/2 (min); CL int (μL / min / mg).
对本发明化合物及其没有氘代的化合物同时测验比较,评价其在人和大鼠肝微粒体的代谢稳定性。采用未经氘代的化合物T作为对照品。在人和大鼠肝微粒体实验中,通过与未经氘代的化合物T对照,本发明化合物可以明显改善代谢稳定性。代表性的实施例化合物的结果归纳于下表3中。The compounds of the present invention and their non-deuterated compounds were tested and compared simultaneously to evaluate their metabolic stability in human and rat liver microsomes. Undeuterated compound T was used as a reference. In human and rat liver microsome experiments, the compounds of the present invention can significantly improve metabolic stability by comparison with the undeuterated compound T. The results for representative example compounds are summarized in Table 3 below.
表3:table 3:
Figure PCTCN2019088749-appb-000023
Figure PCTCN2019088749-appb-000023
Figure PCTCN2019088749-appb-000024
Figure PCTCN2019088749-appb-000024
(4)大鼠药代动力学实验(4) Pharmacokinetic experiments in rats
6只雄性Sprague-Dawley大鼠,7-8周龄,体重约210g,分成2组,每组3只,经静脉或口服单个剂量的化合物(口服10mg/kg),比较其药代动力学差异。Six male Sprague-Dawley rats, 7-8 weeks old, weighing about 210 g, were divided into 2 groups of 3 rats each by intravenous or oral administration of a single dose of the compound (oral 10 mg / kg), and their pharmacokinetic differences were compared. .
大鼠采用标准饲料饲养,给予水。试验前16小时开始禁食。药物用PEG400和二甲亚砜溶解。眼眶采血,采血的时间点为给药后0.083小时,0.25小时、0.5小时、1小时、2小时、4小时、6小时、8小时、12小时和24小时。Rats were raised on a standard diet and given water. Fasting began 16 hours before the test. The drug was dissolved with PEG400 and dimethyl sulfoxide. Orbital blood was collected at 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.
大鼠吸入乙醚后短暂麻醉,眼眶采集300μL血样于试管。试管内有30μL 1%肝素盐溶液。使用前,试管于60℃烘干过夜。在最后一个时间点血样采集完成之后,大鼠乙醚麻醉后处死。Rats were anesthetized briefly after inhaling ether, and 300 μL of blood samples were collected in test tubes from the orbit. The test tube contained 30 μL of a 1% heparin salt solution. Before use, test tubes were dried at 60 ° C overnight. After blood samples were collected at the last time point, rats were sacrificed after ether anesthesia.
血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血样在4℃5000rpm离心5分钟,将血浆与红细胞分离。用移液器吸出100μL血浆到干净的塑料离心管中,标明化合物的名称和时间点。血浆在进行分析前保存在-80℃。用LC-MS/MS测定血浆中本发明化合物的浓度。药代动力学参数基于每只动物在不同时间点的血药浓度进计算。Immediately after the blood sample is collected, gently invert the test tube at least 5 times to ensure that it is fully mixed and placed on ice. The blood sample was centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate the plasma from the red blood cells. Pipette 100 μL of plasma into a clean plastic centrifuge tube with the name and time point of the compound. Plasma was stored at -80 ° C before analysis. LC-MS / MS was used to determine the concentration of a compound of the invention in plasma. Pharmacokinetic parameters were calculated based on the blood drug concentration of each animal at different time points.
实验表明,本发明化合物在动物体内具有更好的药代动力学性质,因此具有更好的药效学和治辽效果。Experiments show that the compounds of the present invention have better pharmacokinetic properties in animals, and therefore have better pharmacodynamics and therapeutic effects.
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。The above content is a further detailed description of the present invention in combination with specific preferred embodiments, and it cannot be considered that the specific implementation of the present invention is limited to these descriptions. For those of ordinary skill in the technical field to which the present invention pertains, without deviating from the concept of the present invention, several simple deductions or replacements can be made, which should all be regarded as belonging to the protection scope of the present invention.

Claims (16)

  1. 一种式(I)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体:A compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof:
    Figure PCTCN2019088749-appb-100001
    Figure PCTCN2019088749-appb-100001
    其中,among them,
    Y 1、Y 2、Y 3、Y 4、Y 5、Y 6、Y 7、Y 8和Y 9各自独立地选自氢、氘、卤素或三氟甲基; Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
    R 1、R 2、R 3、R 4和R 5各自独立地选自氢或氘; R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from hydrogen or deuterium;
    X 1、X 2、X 3、X 4、X 5、X 6和X 7各自独立地选自CH 3、CD 3、CHD 2或CH 2D; X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
    附加条件是,上述化合物至少含有一个氘原子。The additional condition is that the above compound contains at least one deuterium atom.
  2. 根据权利要求1所述的化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体,其中,The compound according to claim 1, or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof, wherein,
    Y 1、Y 2、R 1、R 2、R 3、R 4和R 5各自独立地选自氢或氘; Y 1 , Y 2 , R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from hydrogen or deuterium;
    X 1、X 2、X 3、X 4、X 5、X 6和X 7各自独立地选自CH 3、CD 3、CHD 2或CH 2D; X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are each independently selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
    Y 3、Y 4、Y 5、Y 6、Y 7、Y 8和Y 9为氢; Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are hydrogen;
    附加条件是,上述化合物至少含有一个氘原子。The additional condition is that the above compound contains at least one deuterium atom.
  3. 根据权利要求1-2中任一项所述的化合物,其中,Y 1为氢。 The compound according to any one of claims 1-2, wherein Y 1 is hydrogen.
  4. 根据权利要求1-3中任一项所述的化合物,其中,R 2、R 3、R 4和R 5为氢。 The compound according to any one of claims 1-3, wherein R 2 , R 3 , R 4 and R 5 are hydrogen.
  5. 根据权利要求1-4中任一项所述的化合物,其中,X 5为CH 3The compound according to any one of claims 1-4, wherein X 5 is CH 3 .
  6. 根据权利要求1-5中任一项所述的化合物,其中,Y 2为氢。 The compound according to any one of claims 1 to 5, wherein Y 2 is hydrogen.
  7. 根据权利要求1-6中任一项所述的化合物,其中,R 1为氢。 The compound according to any one of claims 1-6, wherein R 1 is hydrogen.
  8. 根据权利要求1-7中任一项所述的化合物,其中,X 1和X 2为CD 3The compound according to any one of claims 1 to 7, wherein X 1 and X 2 are CD 3 .
  9. 根据权利要求1-8中任一项所述的化合物,其中,X 3为CD 3The compound according to any one of claims 1 to 8, wherein X 3 is CD 3 .
  10. 根据权利要求1-9中任一项所述的化合物,其中,X 6和X 7各自独立地选自CD 3、CD 2H或CH 3The compound according to any one of claims 1-9, wherein X 6 and X 7 are each independently selected from CD 3 , CD 2 H, or CH 3 .
  11. 根据权利要求1-10中任一项所述的化合物,其中,X 4为CD 3The compound according to any one of claims 1 to 10, wherein X 4 is CD 3 .
  12. 根据权利要求1-10中任一项所述的化合物,其中所述化合物可选自如下任一结构:The compound according to any one of claims 1-10, wherein the compound can be selected from any one of the following structures:
    Figure PCTCN2019088749-appb-100002
    Figure PCTCN2019088749-appb-100002
    Figure PCTCN2019088749-appb-100003
    Figure PCTCN2019088749-appb-100003
  13. 一种药物组合物,其含有药学上可接受的赋形剂和权利要求1-12任一项所述的式(I)的化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体。A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of formula (I) according to any one of claims 1-12, or a pharmaceutically acceptable salt, prodrug, hydrate or Solvent compounds, crystalline forms, stereoisomers, or isotopic variations.
  14. 权利要求1-15任一项所述的式(I)的化合物或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体,或权利要求13任一项所述的药物组合物在制备治疗由EGFR介导的疾病的药物中的用途。The compound of formula (I) or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, crystalline form, stereoisomer or isotope variant thereof according to any one of claims 1-15, or claim Use of the pharmaceutical composition according to any one of the claims 13 to 13 for preparing a medicament for treating a disease mediated by EGFR.
  15. 根据权利要求14所述的用途,其中所述的EGFR介导的疾病选自癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病。The use according to claim 14, wherein the EGFR-mediated disease is selected from cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplant, viral disease, cardiovascular disease or metabolic disease.
  16. 根据权利要求14所述的用途,所述癌症选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、前列腺癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌和鼻咽癌。The use according to claim 14, the cancer is selected from the group consisting of non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cancer, stomach Intestinal stromal tumor, leukemia, histiocytic lymphoma, and nasopharyngeal carcinoma.
PCT/CN2019/088749 2018-05-29 2019-05-28 Substituted benzo[d]imidazole compound and pharmaceutical composition thereof WO2019228330A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810527410 2018-05-29
CN201810527410.X 2018-05-29

Publications (1)

Publication Number Publication Date
WO2019228330A1 true WO2019228330A1 (en) 2019-12-05

Family

ID=67371773

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/088749 WO2019228330A1 (en) 2018-05-29 2019-05-28 Substituted benzo[d]imidazole compound and pharmaceutical composition thereof

Country Status (2)

Country Link
CN (1) CN110066272B (en)
WO (1) WO2019228330A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020151742A1 (en) * 2016-07-26 2020-07-30 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113493419A (en) * 2020-03-18 2021-10-12 南京药石科技股份有限公司 EGFR tyrosine kinase inhibitor and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018019204A1 (en) * 2016-07-26 2018-02-01 深圳市塔吉瑞生物医药有限公司 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2959194A1 (en) * 2014-10-11 2016-04-14 Shanghai Hansoh Biomedical Co., Ltd. 4-substituted-2-(n-(5-substituted allyl amide)phenyl)amino)pyrimidine compound, its use as an egfr inhibitor, and preparation and application thereof
CN107827875B (en) * 2017-09-25 2021-07-09 文韬创新药物研究(北京)有限责任公司 Application of benzimidazole derivative as cyclin-dependent kinase 4/6 inhibitor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018019204A1 (en) * 2016-07-26 2018-02-01 深圳市塔吉瑞生物医药有限公司 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020151742A1 (en) * 2016-07-26 2020-07-30 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
US11111233B2 (en) 2016-07-26 2021-09-07 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Also Published As

Publication number Publication date
CN110066272B (en) 2022-09-13
CN110066272A (en) 2019-07-30

Similar Documents

Publication Publication Date Title
JP6948659B1 (en) Pyridadinyl thiaazole carboxamide compound
TW202200562A (en) Quinoxaline dione derivative as irreversible inhibitor of KRAS G12C mutant protein
CN111285886B (en) Substituted pyrazolo [1,5-a ] pyridine compounds, compositions comprising the same and uses thereof
CN109970745B (en) Substituted pyrrolotriazine compound, pharmaceutical composition and application thereof
CN110759908B (en) N-benzenesulfonyl benzamide compound for inhibiting Bcl-2 protein and composition and application thereof
JP2021073246A (en) Maleate salt of b-raf kinase, crystal form, adjustment method and use thereof
CN109851638B (en) Substituted diaminopyrimidine compounds
EP1741714A1 (en) Heterocyclic compound and anti-malignant-tumor agent containing the same as active ingredient
WO2019201131A1 (en) Di(hetero)aryl macrocyclic compound for inhibiting protein kinase activity
EP3476841A1 (en) Crystals of aniline pyrimidine compound serving as egfr inhibitor
WO2023134266A1 (en) 2-piperidyl or 2-pyrazolyl substituted pyrimidine compound serving as egfr inhibitor
WO2019120121A1 (en) Diphenylaminopyrimidine compound for inhibiting kinase activity
WO2019228330A1 (en) Substituted benzo[d]imidazole compound and pharmaceutical composition thereof
JP2022504765A (en) Dihydroimidazolipyrazinone compound, composition containing the compound and its use
WO2019134573A1 (en) Method for preparing deuterated diphenylaminopyrimidine compound and crystal form thereof
TW202214603A (en) Crystal form of pyridazine derivative free base, preparation method, and application thereof
JP2023036957A (en) Diarylpyrazole compound, composition comprising the compound, and use thereof
WO2023030335A1 (en) Compound as tyk2/jak1 pseudokinase domain inhibitor, and synthesis and use methods
WO2022242712A1 (en) Substituted macrocyclic compound, composition comprising compound, and use thereof
WO2019001307A1 (en) Amide compound, composition containing same, and use thereof
EP4155304A1 (en) Compound used as ret kinase inhibitor and application thereof
WO2021057796A1 (en) Substituted fused tricyclic derivative, and composition and use thereof
WO2023093859A1 (en) Salt of axl kinase inhibitor, preparation method therefor and use thereof
WO2022161447A1 (en) Dicarboxamide compound, preparation method therefor, and pharmaceutical use thereof
CN113354630A (en) 5, 6-dihydrobenzo [ h ] quinazoline compound and application thereof

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: 19812065

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: 19812065

Country of ref document: EP

Kind code of ref document: A1