WO2024088379A1 - Procédé de traitement ou de prévention d'une tumeur - Google Patents

Procédé de traitement ou de prévention d'une tumeur Download PDF

Info

Publication number
WO2024088379A1
WO2024088379A1 PCT/CN2023/127066 CN2023127066W WO2024088379A1 WO 2024088379 A1 WO2024088379 A1 WO 2024088379A1 CN 2023127066 W CN2023127066 W CN 2023127066W WO 2024088379 A1 WO2024088379 A1 WO 2024088379A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
phenyl
pyrimidin
benzo
oxy
Prior art date
Application number
PCT/CN2023/127066
Other languages
English (en)
Chinese (zh)
Inventor
黄湧
蒋晨然
陈学明
陈杰安
刘运
安源
吴冠林
Original Assignee
香港科技大学
深圳湾实验室坪山生物医药研发转化中心
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 香港科技大学, 深圳湾实验室坪山生物医药研发转化中心 filed Critical 香港科技大学
Publication of WO2024088379A1 publication Critical patent/WO2024088379A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present disclosure relates generally to the field of medicinal chemistry, and more particularly, to methods for treating and preventing tumors.
  • Drugs for treating cancer mainly include cytotoxic drugs and molecular targeted drugs.
  • Cytotoxic drugs that is, traditional chemotherapy drugs, mainly inhibit tumor growth by killing rapidly dividing tumor cells.
  • cytotoxic drugs can also harm cells that divide rapidly under normal circumstances, such as bone marrow, gastrointestinal tract and hair follicle cells, leading to common side effects in chemotherapy: bone marrow suppression, mucositis, hair loss, etc.
  • Targeted drug therapy achieves treatment by interfering with specific protein targets that affect tumor cell proliferation.
  • An excellent protein or enzyme target has tumor cell-specific mutations or other genetic changes that are not present in normal tissue cells, thereby achieving specific cell selectivity.
  • targeted drugs often have defects such as drug resistance and target molecule-related toxic side effects, resulting in many drug candidates failing in clinical trials despite showing good effects in animal models.
  • the combined use of targeted inhibitors and cytotoxic drugs can improve drug efficacy through two different mechanisms of action, thereby achieving a better efficacy/toxicity window by reducing the dosage.
  • Anaplastic lymphoma kinase is a transmembrane protein tyrosine kinase that belongs to the insulin receptor kinase subfamily. It was first discovered in anaplastic large cell lymphoma (ALCL) with a nucleophosmin (NPM)-ALK fusion form, a T-cell non-Hodgkin lymphoma that is often associated with chromosomal abnormalities. Since then, many cancers have been found to be associated with different forms of ALK fusion.
  • NSCLC non-small cell lung cancer
  • EML4-ALK inflammatory myofibroblastic tumor
  • DLBCL diffuse large B-cell lymphoma
  • amplification of the ALK gene and mutations of the wild-type ALK protein have been reported in various tumors.
  • ALK anaplastic lymphoma kinase
  • the present disclosure relates to a method for treating or preventing tumors, comprising administering to an individual in need thereof a therapeutically or prophylactically effective amount of a compound represented by general formula (I), or a pharmaceutically acceptable salt thereof:
  • R1 is selected from halogen, optionally substituted aryl or optionally substituted heteroaryl;
  • R2 is selected from an optionally substituted hydrocarbon group, an optionally substituted aryl group or an optionally substituted cycloalkyl group;
  • X and Y are each independently selected from O, S or N.
  • the present disclosure relates to the use of a compound represented by general formula (I), or a pharmaceutically acceptable salt thereof, in the preparation of a drug for treating or preventing a tumor:
  • R1 is selected from halogen, optionally substituted aryl or optionally substituted heteroaryl;
  • R2 is selected from an optionally substituted hydrocarbon group, an optionally substituted aryl group or an optionally substituted cycloalkyl group;
  • X and Y are each independently selected from O, S or N.
  • FIG1 shows the interaction pattern between the compound YH-I-001 of the present disclosure and ALK
  • FIG2 shows the blood concentration-time curve of the compound YH-I-001 of the present disclosure in ICR mice after intravenous administration at a dose of 1 mg ⁇ kg -1 ;
  • FIG3 shows the blood concentration-time curve of the compound YH-I-001 of the present disclosure in ICR mice after intravenous administration at a dose of 5 mg ⁇ kg -1 ;
  • FIG4 shows a graph showing the changes in tumor volume of KELLY tumor-bearing mice.
  • FIG. 5 shows the body weight changes of KELLY tumor-bearing mice.
  • the pharmaceutical composition mentioned containing "a compound or a pharmaceutically acceptable salt thereof represented by the general formula (I)” includes one compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof, or two or more compounds represented by the general formula (I) or pharmaceutically acceptable salts thereof.
  • C1 - C4 alkyl describes an alkyl group as defined below having a total of 1 to 4 carbon atoms
  • C3 - C10 cycloalkyl describes a cycloalkyl group as defined below having a total of 3 to 10 carbon atoms.
  • the total number of carbons in the shorthand notation does not include carbons that may be present in substituents of the group being described.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • hydroxyl refers to an -OH group.
  • amino refers to a -NH2 group.
  • cyano refers to a -CN group.
  • hydrocarbyl refers to an aliphatic hydrocarbon group.
  • the hydrocarbyl portion may be a "saturated hydrocarbyl” group, meaning that it does not contain any alkene or alkyne moieties.
  • the hydrocarbyl portion may also be an "unsaturated hydrocarbyl” portion, meaning that it contains at least one alkene or alkyne moiety.
  • alkene portion refers to a straight or branched hydrocarbon chain group consisting of two to eight carbon atoms and at least one carbon-carbon double bond, and connected to the rest of the molecule by a single bond, such as vinyl, prop-1-enyl, but-1-enyl, pent-1-enyl, pent-1,4-dienyl, etc.
  • alkyne portion refers to a straight or branched hydrocarbon chain group consisting of two to eight carbon atoms and at least one carbon-carbon triple bond, and connected to the rest of the molecule by a single bond.
  • the hydrocarbyl portion whether saturated or unsaturated, may be branched or Straight chain.
  • the hydrocarbyl group can have 1 to 8 carbon atoms (each time it appears in this disclosure, a numerical range such as “1 to 8” refers to each integer in the given range; e.g., "1 to 8” means that the hydrocarbyl group can consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, etc., up to and including 8 carbon atoms, although this definition also covers the occurrence of the term "hydrocarbyl" without specifying a numerical range).
  • the hydrocarbyl group may be optionally substituted, i.e., substituted or unsubstituted.
  • the substituent groups are individually and independently selected from one or more of the following groups: cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, hydrocarbyl, aryloxy, mercapto, hydrocarbyl, arylthio, cyano, halo, carbonyl, thiocarbonyl, O-carbamoyl, N-carbamoyl, O-thiocarbamoyl, N-thiocarbamoyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxyl, O-carboxyl, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, -NR'R" (R' and
  • Typical hydrocarbyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl, vinyl, propenyl, butenyl, ethynyl, propynyl and butynyl.
  • substituents may be substituted by one of the substituents described above.
  • C 1 -C 4 hydrocarbyl refers to a hydrocarbyl group as defined above containing from one to four carbon atoms.
  • the C 1 -C 4 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • C 1 -C 4 hydrocarbyl may be a C 1 -C 4 alkyl group.
  • the C 1 -C 4 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • C 1 -C 6 hydrocarbyl refers to a hydrocarbyl group as defined above containing from one to six carbon atoms.
  • the C 1 -C 6 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 1 -C 6 hydrocarbyl” may be a C 1 -C 6 alkyl group.
  • the C 1 -C 6 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • C 1 -C 12 hydrocarbyl refers to a hydrocarbyl group as defined above containing from one to twelve carbon atoms.
  • the C 1 -C 12 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 1 -C 12 hydrocarbyl” may be a C 1 -C 12 alkyl group.
  • the C 1 -C 12 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • C 2 -C 6 hydrocarbyl refers to a hydrocarbyl group as defined above containing two to six carbon atoms.
  • the C 2 -C 6 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 2 -C 6 hydrocarbyl” may be a C 2 -C 6 alkyl group.
  • the C 2 -C 6 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 2 -C 6 alkyl” may be a C 2 -C 6 alkenyl group.
  • the C 2 -C 6 alkenyl group may be optionally substituted as defined for an alkyl group.
  • a "C 2 -C 6 alkyl” may be a C 2 -C 6 alkynyl group.
  • a C 2 -C 6 alkynyl group may be optionally substituted as defined for an alkyl group.
  • C 3 -C 6 hydrocarbyl refers to a hydrocarbyl group as defined above containing three to six carbon atoms.
  • the C 3 -C 6 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 3 -C 6 hydrocarbyl” may be a C 3 -C 6 alkyl group.
  • the C 3 -C 6 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 3 -C 6 alkyl” may be a C 3 -C 6 alkenyl group.
  • the C 3 -C 6 alkenyl group may be optionally substituted as defined for an alkyl group.
  • a "C 3 -C 6 alkyl” may be a C 3 -C 6 alkynyl group.
  • a C 3 -C 6 alkynyl group may be optionally substituted as defined for an alkyl group.
  • C 3 -C 12 hydrocarbyl refers to a hydrocarbyl group as defined above containing three to twelve carbon atoms.
  • the C 3 -C 12 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 3 -C 12 hydrocarbyl” may be a C 3 -C 12 alkyl group.
  • the C 3 -C 12 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 3 -C 12 alkyl” may be a C 3 -C 12 alkenyl group.
  • the C 3 -C 12 alkenyl group may be optionally substituted as defined for an alkyl group.
  • a "C 3 -C 12 alkyl” may be a C 3 -C 12 alkynyl group.
  • the C 3 -C 12 alkynyl group may be optionally substituted as defined for an alkyl group.
  • C 6 -C 12 hydrocarbyl refers to a hydrocarbyl group as defined above containing six to twelve carbon atoms.
  • the C 6 -C 12 hydrocarbyl group may be arbitrarily selected from the group defined above. generation.
  • a "C 6 -C 12 hydrocarbyl” may be a C 6 -C 12 alkyl group.
  • the C 6 -C 12 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 6 -C 12 alkyl” may be a C 6 -C 12 alkenyl group.
  • the C 6 -C 12 alkenyl group may be optionally substituted as defined for an alkyl group.
  • a "C 6 -C 12 alkyl” may be a C 6 -C 12 alkynyl group.
  • the C 6 -C 12 alkynyl group may be optionally substituted as defined for an alkyl group.
  • C7 - C12 hydrocarbyl refers to a hydrocarbyl group as defined above containing seven to twelve carbon atoms.
  • the C7 - C12 hydrocarbyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 7 -C 12 hydrocarbyl” may be a C 7 -C 12 alkyl group.
  • the C 7 -C 12 alkyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 7 -C 12 hydrocarbyl” may be a C 7 -C 12 alkenyl group.
  • the C 7 -C 12 alkenyl group may be optionally substituted as defined for a hydrocarbyl group.
  • a "C 7 -C 12 alkyl” may be a C 7 -C 12 alkynyl group.
  • the C 7 -C 12 alkynyl group may be optionally substituted as defined for an alkyl group.
  • alkyloxy refers to the general formula -O-alkyl, wherein alkyl is as defined in the present disclosure.
  • alkyloxy include, but are not limited to, methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentoxy, and tert-pentoxy.
  • aryl refers to a carbocyclic ring (full carbon) or two or more fused rings (rings sharing two adjacent carbon atoms) with a completely delocalized Pi electron system.
  • Aryl groups include, but are not limited to, fluorenyl, phenyl, and naphthyl. Aryl groups, for example, may have five to twelve carbon atoms. The aryl groups of the present disclosure may be substituted or unsubstituted.
  • the hydrogen atoms are substituted by one or more groups independently selected from the group consisting of hydrocarbon, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, protected hydroxy, hydrocarbonoxy, aryloxy, mercapto, hydrocarbonthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, O-carbamoyl, N-carbamoyl, O-thiocarbamoyl, N-thiocarbamoyl, C-amido, N-amido, S-sulfonylamino, N-sulfonylamino, C-carboxyl, protected C-carboxyl, O-carboxyl, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, -NR'R"(R' and R" are hydrocarbon groups as defined in the group consisting
  • heteroaryl refers to a 5- to 18-membered aromatic ring group consisting of one to seventeen carbon atoms and one to ten heteroatoms selected from nitrogen, oxygen and sulfur.
  • the heteroaryl group may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include a fused or bridged ring system; and the nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atom may be optionally quaternized.
  • heteroaryl groups include, but are not limited to, azepine, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepanyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyrone, benzofuranyl, benzofuranone, benzothiophenyl, benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzo
  • the heteroaryl groups of the present disclosure may be substituted or
  • the hydrogen atoms are replaced by one or more groups independently selected from the group consisting of hydrocarbon, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, protected hydroxy, hydrocarbonoxy, aryloxy, mercapto, hydrocarbonthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, O-carbamoyl, N-carbamoyl, O-thiocarbamoyl, N-thiocarbamoyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, -NR'R" (R' and R" are hydrocarbon groups as defined in the present disclosure) or protected amino
  • cycloalkyl refers to a cycloalkyl group consisting only of carbon and hydrogen atoms and having three to four carbon atoms.
  • cycloalkyl is intended to include cycloalkyl as defined above optionally substituted by one or more substituents selected from the group consisting of cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkyloxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, O-carbamoyl, N-carbamoyl, O-thiocarbamoyl, N-thiocarbamoyl, C-acylamino, N-acylamino, S-sulfinylamino, N-sulfinylamino, C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, -NR'R"(R
  • C 3 -C 6 cycloalkyl refers to a cycloalkyl group as defined above having three to six carbon atoms.
  • the C 3 -C 6 cycloalkyl group may be optionally substituted as defined for the cycloalkyl group above.
  • C 3 -C 10 cycloalkyl refers to a cycloalkyl group as defined above having three to ten carbon atoms.
  • the C 3 -C 10 cycloalkyl group may be optionally substituted as defined for the cycloalkyl group above.
  • C 3 -C 12 cycloalkyl refers to a cycloalkyl group as defined above having three to twelve carbon atoms.
  • the C 3 -C 12 cycloalkyl group may be optionally substituted as defined for the cycloalkyl group above.
  • heterocycloalkyl refers to a stable three- to twelve-membered non-aromatic ring group consisting of carbon atoms and one to five heteroatoms selected from nitrogen, oxygen and sulfur.
  • heterocyclyl groups include, but are not limited to, dioxolane, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuranyl,
  • the term "compound of the present disclosure or a pharmaceutically acceptable salt thereof” refers to The invention relates to compounds represented by the general formula (I) and their pharmaceutically acceptable salts, as well as any specific compounds falling within the general formula (I) and their pharmaceutically acceptable salts.
  • mammal refers to animals including, for example, dogs, cats, cows, sheep, horses, and humans, etc. In certain embodiments, the mammal includes humans.
  • the term "patient” refers to animals (e.g., humans), companion animals (e.g., dogs, cats, or horses), and livestock (e.g., cattle, pigs, and sheep).
  • the patient is a mammal, including males and females.
  • the patient is a human.
  • the term "pharmaceutically acceptable” refers to the carrier, vehicle, diluent, excipient and/or salt which must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the terms “optional” or “arbitrarily” mean that the subsequently described event or circumstance may or may not occur, and the description includes instances where the event or circumstance occurs and instances where it does not occur.
  • the term "pharmaceutically acceptable excipients” includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent or emulsifier approved by the U.S. Food and Drug Administration for use in humans or animals, and various forms of carriers that have no side effects on the composition of the pharmaceutical composition.
  • carrier is defined as a compound that facilitates the introduction of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • pharmaceutically acceptable salt includes “acceptable acid addition salts” and “acceptable base addition salts”.
  • the term "acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases and are biologically or otherwise suitable and are formed using inorganic or organic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzenecarboxylic acid, 4-acetamidobenzenecarboxylic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethan
  • the term "acceptable base addition salts” refers to those salts which retain the biological effectiveness and properties of the free acids, which are biologically or otherwise suitable. These salts are prepared by adding inorganic or organic bases to the free acids. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts, and the like. In certain embodiments, the inorganic salts are ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benzylamine, phenylethylenediamine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like.
  • the organic base is isooprop
  • the term "pharmaceutical composition” refers to a preparation of the compounds described in the present disclosure and a medium generally accepted in the art for delivering the bioactive compound to mammals such as humans.
  • a medium includes all pharmaceutically acceptable carriers, diluents or excipients.
  • the term "therapeutically effective amount” refers to the amount of a compound or combination of compounds that improves, attenuates or eliminates a particular disease or condition and the symptoms of a particular disease or condition, or avoids or delays the onset of a particular disease or condition or the symptoms of a particular disease or condition.
  • the amount of the compound described in the present disclosure that constitutes a “therapeutically effective amount” will vary, but those skilled in the art can determine the amount of the compound described in the present disclosure according to routine based on their own knowledge and the present disclosure.
  • treating encompasses treating a relevant disease or condition in a mammal, such as a human, that suffers from the relevant disease or condition, and includes:
  • preventing refers to preventing the onset, recurrence, or spread of a disease or disease state, or one or more symptoms thereof.
  • the term "prophylactically effective amount” refers to an amount of a compound or combination of compounds sufficient to prevent a disease or disease state, or to prevent its recurrence or spread. Depending on the compound, the disease state and its severity, and the age, weight, etc. of the mammal to be treated, the amount of the compound described in the present disclosure that constitutes a “prophylactically effective amount” will vary, but those skilled in the art can determine the amount of the compound described in the present disclosure according to routine based on their own knowledge and the present disclosure.
  • disease and “disease state” may be used interchangeably, or may be distinct in that a particular disease or disease state may have no known causative agent (and therefore cannot be explained etiologically) and therefore is not recognized as a disease, but rather is considered an undesirable disease state or condition in which clinicians have identified a more or less specific set of symptoms.
  • topoisomerase refers to an enzyme that corrects the number of DNA strands by severing the phosphodiester bonds in one or both strands of DNA followed by rewinding and sealing.
  • the present disclosure relates to a method for treating or preventing tumors, comprising administering to an individual in need thereof a therapeutically or preventively effective amount of a compound represented by general formula (I), and a pharmaceutically acceptable salt thereof:
  • R1 is selected from halogen, optionally substituted aryl or optionally substituted heteroaryl;
  • R2 is selected from an optionally substituted hydrocarbon group, an optionally substituted aryl group or an optionally substituted cycloalkyl group;
  • X and Y are each independently selected from O, S or N.
  • R is selected from halogen, unsubstituted aryl, optionally substituted alkyl substituted aryl, alkoxy substituted aryl, halogen substituted aryl, cyano substituted aryl, optionally substituted amino substituted aryl, sulfonamido substituted aryl, unsubstituted heteroaryl, optionally substituted alkyl substituted heteroaryl, alkoxy substituted heteroaryl, cyano substituted heteroaryl, optionally substituted amino substituted heteroaryl or heterocycloalkyl substituted heteroaryl.
  • R is selected from halogen, unsubstituted phenyl, optionally substituted alkyl-substituted phenyl, optionally substituted heterocycloalkyl-substituted optionally substituted alkyl-substituted phenyl, alkoxy-substituted phenyl, halogen-substituted phenyl, cyano-substituted phenyl, optionally substituted amino-substituted phenyl, sulfonamido-substituted phenyl, unsubstituted pyridyl, unsubstituted thienyl, unsubstituted pyrimidinyl, unsubstituted pyrazolyl, optionally substituted alkyl-substituted pyrazolyl, amino-substituted alkyl-substituted pyrazolyl, alkyl-substituted amino-
  • R is selected from halogen, unsubstituted aryl, alkyl-substituted aryl, halogen-substituted alkyl-substituted aryl, alkyl-substituted heterocycloalkyl-substituted alkyl-substituted aryl, alkyl-substituted heterocycloalkyl-substituted oxoalkyl-substituted aryl, alkoxy-substituted aryl, halogen-substituted aryl, cyano-substituted aryl, alkyl-substituted amino-substituted aryl, optionally substituted heterocycloalkyl-substituted amino-substituted aryl, sulfonamido-substituted phenyl, unsubstituted heteroaryl, optionally substituted alkyl-substi
  • R 1 is selected from iodo, phenyl, tolyl, isopropylphenyl, trifluoromethylphenyl, 4-(4-methylpiperazin-1-yl)methylphenyl, 4-(4-methylpiperazin-1-yl)oxymethylphenyl, methoxyphenyl, isopropoxyphenyl, bromophenyl, cyano-substituted phenyl, dimethylamino phenyl, 2-methyl-4-piperidinyl substituted amino substituted phenyl, 2-methyl-4-(N-methylpiperidinyl)-amino substituted phenyl, 2-methyl-4-tetrahydropyrrolyl substituted amino substituted phenyl, 2-methyl-4-(N-methyltetrahydropyrrolyl)-amino substituted phenyl, 2-methyl-4-sulfonamido substituted phenyl, pyridyl, thienyl, pyr
  • R is selected from:
  • R 2 is selected from optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, or optionally substituted aryl and heterocycloalkyl.
  • R 2 is selected from optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, or optionally substituted benzoheterocycloalkyl.
  • R 2 is selected from unsubstituted alkyl, halogen-substituted aryl, optionally substituted alkyl-substituted aryl, optionally substituted alkoxy-substituted aryl, unsubstituted cycloalkyl, unsubstituted benzoheterocycloalkyl, or halogen-substituted benzoheterocycloalkyl.
  • R2 is selected from unsubstituted alkyl, halogen-substituted phenyl, optionally substituted alkyl-substituted phenyl, optionally substituted alkoxy-substituted phenyl, optionally substituted pyridyl, optionally substituted pyrrolyl, optionally substituted pyrazolyl, optionally substituted thiazolyl, optionally substituted oxazolyl, optionally substituted thienyl, unsubstituted cycloalkyl, unsubstituted benzo[1,3]dioxolanyl, or halogen-substituted benzo[1,3]dioxolanyl.
  • R2 is selected from hexyl, 3,4-dichlorophenyl, 4-chlorophenyl, 4-isopropylphenyl, 3,5-trifluoromethylphenyl, 4-trifluoromethylphenyl, 4-trifluoromethoxyphenyl, cyclopentanyl, or cyclohexanyl.
  • R2 is selected from:
  • X is N and Y is O.
  • X is N and Y is S.
  • X is O and Y is N.
  • X is O and Y is S.
  • the present disclosure relates to compounds, and pharmaceutically acceptable salts thereof, wherein the compound is selected from:
  • the compounds of the present disclosure have potent inhibitory activity against ALK.
  • the compounds of the present disclosure are resistant to drug resistance of ALK mutants.
  • the compounds of the present disclosure have excellent stability.
  • the compounds of the present disclosure have excellent pharmaceutical efficacy.
  • the methods of treating or preventing tumors disclosed herein further comprise administering another active agent to the individual.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, and other active agents are administered to a subject simultaneously, sequentially, overlappingly, concomitantly, intermittently, continuously, synchronously or any combination thereof.
  • illustrative examples of other active agents include, but are not limited to, topoisomerase inhibitors.
  • topoisomerase inhibitors that can be used in the present disclosure include, but are not limited to, topoisomerase I inhibitors and topoisomerase II inhibitors.
  • topoisomerase inhibitors that can be used in the present disclosure include, but are not limited to, topotecan, irinotecan, belotecan, aclarubicin, doxorubicin, epirubicin, edamycin, etoposide, and mitoxantrone.
  • the tumor that can be used in the methods of treating or preventing tumors disclosed herein is mediated by tyrosine kinase (TK).
  • TK tyrosine kinase
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound represented by the general formula (I) of the present disclosure or a pharmaceutically acceptable salt thereof, or a compound of the present disclosure or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises a compound of formula (I) of the present disclosure: or a pharmaceutically acceptable salt thereof, or a compound of the present disclosure or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the compound represented by the general formula (I) of the present invention or its pharmaceutically acceptable salt, or the compound of the present invention or its pharmaceutically acceptable salt for treating or preventing tyrosine kinase (TK)-mediated diseases or disease states when administered to mammals can be administered by the gastrointestinal route or the parenteral route.
  • the compound represented by the general formula (I) of the present invention or its pharmaceutically acceptable salt, or the compound of the present invention or its pharmaceutically acceptable salt for tyrosine kinase (TK)-mediated diseases or disease states when administered to mammals can be an oral route.
  • the compound represented by the general formula (I) of the present invention or its pharmaceutically acceptable salt, or the compound of the present invention or its pharmaceutically acceptable salt for tyrosine kinase (TK)-mediated diseases or disease states when administered to mammals can be administered via the rectal route.
  • the compounds described in the present disclosure can be obtained in any suitable form such as tablets, capsules, powders, oral solutions, suspensions, rectal gels, rectal foams, rectal enemas or rectal suppositories, etc.
  • suitable form such as tablets, capsules, powders, oral solutions, suspensions, rectal gels, rectal foams, rectal enemas or rectal suppositories, etc.
  • the tablets include, but are not limited to, plain tablets, sugar-coated tablets, and film-coated tablets.
  • Examples of pharmaceutically acceptable excipients that can be used in the pharmaceutical compositions of the present disclosure include, but are not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent or emulsifier approved by the U.S. Food and Drug Administration for use in humans or animals, and various forms of carriers that have no side effects on the composition of the pharmaceutical composition.
  • compositions of the present disclosure can be administered by any method that achieves its intended purpose.
  • administration can be performed by oral, parenteral, topical, enteral, intravenous, intramuscular, inhalation, nasal, intraarticular, intraspinal, transtracheal, ocular, subcutaneous, intraperitoneal, transdermal or buccal routes.
  • the route of administration can be parenteral, oral, and rectal routes.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • Suitable dosage forms include, but are not limited to, capsules, tablets, pills, dragees, semisolid preparations, powders, granules, suppositories, ointments, creams, lotions, inhalants, injections, poultices, gels, tapes, eye drops, solutions, syrups, aerosols, suspensions, emulsions, which can be prepared according to methods known in the art.
  • Particularly suitable for oral administration are ordinary tablets (plain tablets), sugar-coated tablets, film-coated tablets, pills, capsules, powders, granules, syrups, juices or drops, suitable for rectal administration are suppositories, suitable for parenteral administration are solutions, can also be oil-based solutions or aqueous solutions, in addition there are suspensions, emulsions or implants, suitable for local use are ointments, creams or powders.
  • the product in the present disclosure can also be lyophilized, and the generated lyophilized material is used for example to prepare injections.
  • the given preparation can be sterilized and/or contain an assistant, such as a wetting agent, a preservative, a stabilizer and/or a wetting agent, an emulsifier, a salt for changing the osmotic pressure, a buffer substance, a dye, a flavoring agent and/or numerous other active ingredients, such as one or more vitamins.
  • an assistant such as a wetting agent, a preservative, a stabilizer and/or a wetting agent, an emulsifier, a salt for changing the osmotic pressure, a buffer substance, a dye, a flavoring agent and/or numerous other active ingredients, such as one or more vitamins.
  • the pharmaceutical compositions of the present disclosure are prepared as tablets, solutions, granules, patches, ointments, capsules, aerosols or suppositories for parenteral, transdermal, mucosal, nasal, buccal, sublingual or oral use.
  • Preservatives Preservatives, stabilizers, dyes, sweeteners, aromatics, spices, etc. can be provided in the pharmaceutical composition.
  • sodium benzoate, ascorbic acid and esters of p-hydroxybenzoic acid can be added as preservatives.
  • antioxidants and suspending agents can be used.
  • alcohols, esters, sulfated aliphatic alcohols, etc. can be used as surfactants; sucrose, glucose, lactose, starch, crystalline cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium aluminate methyl silicate, synthetic aluminum silicate, calcium carbonate, calcium bicarbonate, calcium hydrogen phosphate, hydroxymethylcellulose calcium, etc. can be used as excipients; magnesium stearate, talc, hardened oil, etc.
  • lubricants can be used as lubricants; coconut oil, olive oil, sesame oil, peanut oil, soybean can be used as suspending agents or lubricants; cellulose acetate phthalate, which is a derivative of sugars such as cellulose or sugar, or methyl acetate-methacrylate copolymer, which is a derivative of polyethylene, can be used as suspending agents; and plasticizers such as phthalates can be used as suspending agents.
  • Suitable routes of administration may include, for example, oral administration, rectal administration, transmembrane administration, parenteral delivery, topical administration or enteral administration; parenteral delivery includes intramuscular injection, subcutaneous injection, Intravenous injection, intramedullary injection and intrathecal injection, direct intraventricular injection, intraperitoneal injection, intranasal injection or intraocular injection.
  • the compound can also be extended and/or timed, pulsed at a predetermined rate in a sustained or controlled release dosage form including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, etc.
  • compositions of the present disclosure can be produced in a known manner, for example, by conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting operations.
  • the pharmaceutical composition used can be prepared in a conventional manner using one or more physiologically acceptable carriers comprising excipients and adjuvants, which are conducive to processing the active compound into a pharmaceutically available preparation.
  • suitable preparations depend on the selected route of administration. Any known technology, carrier and excipient can be used as suitable and understood in the art.
  • Injection can be prepared into the following conventional forms: as a solution or suspension, a solid dosage form suitable for making a solution or suspension before injection, or as an emulsion.
  • Suitable excipients are, for example, water, saline, glucose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, etc.
  • the injection pharmaceutical composition can contain a small amount of non-toxic adjuvants, such as wetting agents, pH buffers, etc.
  • Physiologically suitable buffers include, but are not limited to, Hank's solution, Ringer's solution, or physiological saline buffer. If necessary, absorption enhancing preparations (such as liposomes) can be used.
  • the compounds can be easily formulated by combining the active compounds with pharmaceutically acceptable carriers known in the art.
  • Such carriers enable the compounds of the present disclosure to be formulated as tablets, pills, lozenges, capsules, liquids, gels, syrups, pastes, suspensions, solutions, powders, etc., for oral ingestion by the patient to be treated.
  • Pharmaceutical preparations for oral administration can be obtained by mixing the active compound with a solid excipient, optionally grinding the resulting mixture and processing the granular mixture, if necessary, after adding suitable auxiliary agents to obtain tablets or lozenge cores.
  • Suitable excipients are in particular fillers such as sugars, including lactose, sucrose, mannitol or sorbitol; cellulose preparations such as corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • disintegrants such as cross-linked polyvinylpyrrolidone, agar or alginic acid or sea salts such as sodium alginate may be added. Alginate.
  • the lozenge core is suitably coated.
  • concentrated sugar solution can be used, and this sugar solution can optionally include gum arabic, talcum, polyvinyl pyrrolidone, carbopol gel (carbopol gel), polyethylene glycol and/or titanium dioxide, lacquer solution and suitable organic solvent or solvent mixture.
  • dye or pigment can be added to tablet or lozenge coating.
  • concentrated sugar solution can be used, and this sugar solution can optionally include gum arabic, talcum, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol and/or titanium dioxide, lacquer solution and suitable organic solvent or solvent mixture.
  • compositions that can be used for oral administration include push-fit capsules made of gelatin, and soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • Push-fit capsules can contain the active ingredient mixed with a filler such as lactose, a binder such as starch and/or a lubricant such as talc or magnesium stearate and optionally a stabilizer.
  • the active ingredient can be dissolved or suspended in a suitable liquid, such as a fatty oil, liquid paraffin or liquid polyethylene glycol.
  • a stabilizer can be added. All formulations for oral administration should be in a dosage suitable for such administration.
  • the pharmaceutical compositions of the present disclosure may contain 0.1%-95% of a compound of the present disclosure or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical compositions of the present disclosure may contain 1%-70% of a compound of the present disclosure or a pharmaceutically acceptable salt thereof.
  • composition or formulation to be administered will contain an amount of a compound of the present disclosure, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, in an amount effective to treat the disease/condition in the subject being treated.
  • At least one compound of the present invention or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising at least one compound of the present invention or a pharmaceutically acceptable salt thereof can be administered to a patient by any method suitable for systemic and/or local delivery of the compound of the present invention or a pharmaceutically acceptable salt thereof.
  • Non-limiting examples of administration methods include (a) administration by an oral route, including administration in the form of capsules, tablets, granules, sprays, syrups or other such forms; (b) administration by a non-oral route, such as rectal, vaginal, intraurethral, intraocular, nasal or into the ear, the administration includes administration in the form of aqueous suspensions, oily preparations, etc., or in the form of drops, sprays, suppositories, ointments, ointments, etc.; (c) administration by subcutaneous injection, intraperitoneal injection, intravenous injection, intramuscular injection, intradermal injection, intraorbital injection, intracapsular injection, intraspinal injection, intrasternal injection, etc., including delivery by infusion pump; (d) local administration such as injection directly in the kidney area or the heart area, for example, by reservoir implantation; and (e) local administration; as deemed appropriate by those skilled in the art, the compound described in the present disclosure is in contact with living
  • compositions suitable for administration include compositions containing an effective amount of active ingredients to achieve their desired effects.
  • the dosage required for the therapeutically effective amount of the pharmaceutical compositions described in the present disclosure depends on the route of administration, the type of animal being treated, including humans, and the physical characteristics of the particular animal being considered. The dosage can be adjusted to achieve the desired effect, but this will depend on the following factors: body weight, diet, simultaneous drug therapy, and other factors recognized by technicians in other medical fields. More specifically, a therapeutically effective amount refers to the amount of a compound that effectively prevents, alleviates or improves symptoms of a disease, or prolongs the life span of an individual receiving treatment. The actual ability of those skilled in the art can well determine the therapeutically effective amount, particularly in accordance with the detailed disclosure provided by the present disclosure.
  • the dosage and specific mode of administration for in vivo administration will vary depending on the age, weight and type of mammal being treated, the specific compound used and the specific purpose of the compounds used.
  • Those skilled in the art can achieve the purpose of determining effective dosage levels, i.e., the dosage levels necessary for determining the desired effect, using conventional pharmacological methods.
  • human clinical applications of the product are initiated at lower dosage levels, with the dosage level increasing until the desired effect is achieved.
  • acceptable in vitro studies can be used to establish effective dosages and routes of administration of the compositions identified by the present method.
  • the potential product is started at a higher dose level and the dose is reduced until the desired effect is no longer achieved or the adverse side effects disappear.
  • the dose range can be wide, depending on the desired effect and the therapeutic indication.
  • the dose can be about 10 ⁇ g/kg body
  • the dosage may be about 100 ⁇ g/kg to 300 mg/kg body weight.
  • the dosage may be based on and calculated in accordance with the patient's body surface area.
  • each physician can select the exact formulation, route of administration and dosage of the pharmaceutical composition described in the present disclosure according to the patient's condition.
  • the dosage range of the composition administered to the patient can be about 0.5 mg/kg to 1000 mg/kg of the patient's body weight.
  • the dosage can be given once or twice or more during one day or several days.
  • the human dosage of the compound has been established due to at least some conditions, the present disclosure will use those same dosages, or a dosage range of about 0.1% to 500% of the determined human dosage, and in certain embodiments, a dosage range of 25% to 250% of the determined human dosage.
  • a suitable human dosage can be inferred from the median value of the half effective dose or the infectious dose, or other suitable values from in vitro or in vivo studies, as quantified in toxicity studies and efficacy studies in animals.
  • the attending physician will know how and when to terminate, interrupt or adjust the administration. On the contrary, if the clinical response is insufficient (excluding toxicity), the attending physician will also know to adjust the treatment to a higher level.
  • the size of the dosage in the treatment of the disease concerned will vary with the severity of the disease state to be treated and the change of the route of administration. For example, the severity of the disease state can be evaluated in part by standard prognostic evaluation methods.
  • the dosage and possible dosage frequency will also vary according to the age, weight, and response of the individual patient. A scheme comparable to the above-mentioned discussion scheme can be used in veterinary medicine.
  • the daily dosing regimen for adult patients is, for example, an oral dose of 0.1 mg to 2000 mg of each active ingredient, in certain embodiments 1 mg to 2000 mg of each active ingredient, such as 5 mg to 1500 mg of each active ingredient.
  • the intravenous, subcutaneous or intramuscular dose of each active ingredient used is 0.01 mg to 1000 mg, in certain embodiments 0.1 mg to 1000 mg, such as 1 mg to 800 mg.
  • the dose can be calculated as a free base.
  • the composition is administered 1 to 4 times daily.
  • composition described in the present disclosure can be administered by continuous intravenous infusion, in certain embodiments at a dose of up to 2000 mg of each active ingredient per day.
  • a dose of up to 2000 mg of each active ingredient per day it is necessary to administer the compounds described in the present disclosure in an amount exceeding or far exceeding the above dosage range.
  • the compounds are administered during continuous treatment, such as one or several weeks, or several months or years.
  • Dosage and dosing interval can be adjusted individually to provide the plasma level of the active part that is enough to maintain the adjustment effect or minimum effective concentration (MEC).
  • MEC adjustment effect or minimum effective concentration
  • the MEC of each compound is different, but MEC can be assessed from in vitro data.
  • the required dosage reaching MEC depends on individual characteristics and route of administration.
  • HPLC high performance liquid chromatography
  • the MEC value also enables determination of dosing intervals.
  • Compositions should be administered using a regimen that maintains plasma levels above the MEC for 10-90% of the time, in certain embodiments 30-90% of the time, and in certain embodiments 50-90% of the time.
  • the effective local concentration of the drug is independent of plasma concentration.
  • composition administered will, of course, be dependent upon the individual being treated, on the individual's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • the efficacy and toxicity of the compounds described in the present disclosure can be evaluated using known methods.
  • the toxicology of a particular compound or a subset of such compounds that share certain chemical moieties can be established by determining the toxicity of a cell line in vitro, such as a mammalian cell line and in some embodiments a human cell line.
  • the results of such studies can generally predict toxicity in an animal such as a mammal, or more specifically, in a human.
  • the toxicity of a particular compound in an animal model such as a mouse, rat, rabbit or monkey can be determined using known methods.
  • the efficacy of a particular compound can be determined using several recognized methods, such as in vitro methods, animal models or human clinical trials.
  • compositions may be placed in a pack or dispenser device which
  • the device may include one or more unit dosage forms containing active ingredients.
  • the packaging may, for example, include metal or plastic foil, such as a blister pack.
  • the packaging or dispensing device may carry instructions for administration.
  • the packaging or dispensing device may also carry precautions associated with the container, which are prescribed by a government agency that manages drug production, use or sales, and which reflect that the drug form has been approved by the agency for human or veterinary administration. Such precautions, for example, may be labels for prescription drugs approved by the State Food and Drug Administration or the U.S. Food and Drug Administration, or approved product instructions.
  • Compositions comprising compounds of the present disclosure, stereoisomers thereof, or pharmaceutically acceptable salts thereof, which may also be prepared in suitable containers and placed in compatible pharmaceutical carriers, and labeled for use in the treatment of specified disease states.
  • the present disclosure relates to a method for preparing a compound represented by general formula (Ia),
  • X1 and Y1 are each independently selected from amino, thiol or hydroxyl;
  • X and Y are each independently selected from O, S or N;
  • R2 is selected from an optionally substituted hydrocarbon group, an optionally substituted aryl group or an optionally substituted cycloalkyl group.
  • reaction to prepare the compound represented by formula (Ia) is carried out in the presence of a base and a strong oxidant.
  • illustrative examples of bases that can be used in the present disclosure include, but are not limited to, alkali metal salts, organic bases, or any mixture thereof.
  • alkali metal salts that can be used in the present disclosure include, but are not limited to, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, lithium alkoxide, sodium alkoxide, potassium alkoxide, or any mixture thereof.
  • illustrative examples of alcohols that can be used in the present disclosure include, but are not limited to, methanol, ethanol, or tert-butanol.
  • DIPEA N,N-diisopropylethylamine
  • DABCO 1,4-diazabicyclo[2.2.2]octane
  • illustrative examples of strong oxidizing agents that can be used in the present disclosure include, but are not limited to, periodates, dichromates, pyridinium chlorochromates, or any mixture thereof.
  • periodate salts that can be used in the present disclosure include, but are not limited to, sodium periodate, potassium periodate, or any mixture thereof.
  • dichromates that can be used in the present disclosure include, but are not limited to, potassium dichromate, sodium dichromate, or any mixture thereof.
  • the present disclosure relates to a method for preparing a compound represented by general formula (Ib),
  • R1 is selected from halogen, optionally substituted aryl or optionally substituted heteroaryl;
  • B is selected from boric acid or boric acid esters.
  • reaction to prepare the compound represented by formula (Ib) is carried out in the presence of a palladium catalyst and a base.
  • illustrative examples of palladium catalysts that can be used in the present disclosure include, but are not limited to, palladium, Pd(PPh 3 ) 4 , Pd(PPh 3 ) 2 Cl 2 , Pd(OAc) 2 , Pd(dppf)Cl 2 , or any mixture thereof.
  • illustrative examples of bases that can be used in the present disclosure include, but are not limited to, alkali metal salts, organic bases, or any mixture thereof.
  • alkali metal salts that can be used in the present disclosure include, but are not limited to, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, lithium alkoxide, sodium alkoxide, Potassium alcoholate or any mixture thereof.
  • illustrative examples of alcohols that can be used in the present disclosure include, but are not limited to, methanol, ethanol, or tert-butanol.
  • DIPEA N,N-diisopropylethylamine
  • DABCO 1,4-diazabicyclo[2.2.2]octane
  • borate esters that can be used in the present disclosure include, but are not limited to, bis(pinacolato)diboron (Bpin), Bcat, or any mixture thereof.
  • the present disclosure relates to a method for preparing a compound represented by general formula (Id),
  • the method for preparing the compound represented by formula (Ic) is carried out in the presence of a base.
  • illustrative examples of bases that can be used in the present disclosure include, but are not limited to, alkali metal salts, organic bases, or any mixture thereof.
  • alkali metal salts that can be used in the present disclosure include, but are not limited to, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, lithium alkoxide, sodium alkoxide, potassium alkoxide, or any mixture thereof.
  • illustrative examples of alcohols that can be used in the present disclosure include, but are not limited to, methanol, ethanol, or tert-butanol.
  • DIPEA N,N-diisopropylethylamine
  • DABCO 1,4-diazabicyclo[2.2.2]octane
  • the compound represented by the general formula (Ia) is reacted in the presence of boron bromide to obtain the compound represented by the general formula (Ib).
  • the present disclosure relates to a method for inhibiting tyrosine kinase (TK), which comprises contacting tyrosine kinase with an inhibitory effective amount of a compound represented by the general formula (I) of the present disclosure or a pharmaceutically acceptable salt thereof, a compound of the present disclosure or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure.
  • TK tyrosine kinase
  • TKs tyrosine kinases
  • ALK anaplastic lymphoma kinase
  • ROS1 oncogene receptor tyrosine kinase ROS1 oncogene receptor tyrosine kinase
  • EGFR epidermal growth factor receptor
  • PDGF platelet growth factor receptor
  • ABL fibroblast growth factor receptor
  • IRAK interleukin receptor-associated kinase
  • FLT human tyrosine kinase receptor
  • BRAF V-raf mouse sarcoma viral oncogene homolog B
  • KDR vascular endothelial growth factor receptor 2
  • VGFR rearranged kinase by transfection
  • RET Bruton's tyrosine kinase
  • B-lymphoid tyrosine kinase B-lymphoid tyrosine kinase
  • the present disclosure relates to a method for treating or preventing diseases or disease states mediated by tyrosine kinase (TK), which comprises administering to an individual in need of the method a therapeutically or preventively effective amount of a compound represented by the general formula (I) of the present disclosure or a pharmaceutically acceptable salt thereof, a compound of the present disclosure or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure.
  • TK tyrosine kinase
  • illustrative examples of individuals that can be used in the methods of treating or preventing tyrosine kinase (TK)-mediated diseases or disease states disclosed herein include, but are not limited to, mammals.
  • the subject is a human.
  • TKs tyrosine kinases
  • ALK anaplastic lymphoma kinase
  • ROS1 oncogene receptor tyrosine kinase ROS1 oncogene receptor tyrosine kinase
  • EGFR epidermal growth factor receptor
  • PD-1 platelet-derived growth factor receptor
  • EGFR epidermal growth factor receptor
  • illustrative examples of diseases or disease states that can be used in the methods of treating or preventing tyrosine kinase (TK)-mediated diseases or disease states of the present disclosure include, but are not limited to, lymphoma, blastoma, sarcoma, neuroendocrine tumors, carcinoid tumors, gastrinoma, islet cell carcinoma, mesothelioma, schwannoma, acoustic neuroma, meningioma, adenocarcinoma, melanoma, leukemia, lymphoid malignancies, lung cancer, squamous cell carcinoma of the lung, peritoneal cancer, gastric cancer, intestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, breast cancer, colon cancer, rectal cancer, colorectal cancer, uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, anal cancer,
  • a method for treating or preventing a disease or disease state mediated by tyrosine kinase comprises administering 1 mg-10 g of a compound of the present disclosure or a pharmaceutically acceptable salt thereof to an individual in need of the method.
  • a method for treating or preventing a disease or condition mediated by tyrosine kinase comprises administering 10 mg to 3000 mg of a compound of the present disclosure or a pharmaceutically acceptable salt thereof to an individual in need of the method.
  • a method for treating or preventing a disease or condition mediated by tyrosine kinase comprises administering 100 mg to 1000 mg of a compound of the present disclosure or a pharmaceutically acceptable salt thereof to an individual in need of said method.
  • a method of treating or preventing a disease or condition mediated by tyrosine kinase comprises administering to an individual in need thereof 100 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, 500 mg, 550 mg, 650 mg, 700 mg, 750 mg, 800 mg, 850 mg, 900 mg or 1000 mg of a compound of the present disclosure. or a pharmaceutically acceptable salt thereof.
  • the methods of treating or preventing tyrosine kinase (TK)-mediated diseases or disease states disclosed herein further comprise administering other active agents to the individual.
  • TK tyrosine kinase
  • examples of active agents that can be used in the methods of treating or preventing tyrosine kinase (TK)-mediated diseases or disease states of the present disclosure include, but are not limited to, nitrogen mustards, aziridines, methylmelamines, alkyl sulfonates, nitrosoureas, triazenes, folic acid analogs, pyrimidine analogs, purine analogs, vinca alkaloids, epipodophyllotoxins, antibiotics, topoisomerase inhibitors, anticancer vaccines, acivicin, aclarubicin, acodazole hydrochloride, aclonine, adolesin, aldesleukin, ambromycin, amenthranol acetate, aminoglutethimide, amsacrine, anastrozole, anthramycin, asparaginase, trilinomycin, azacitidine, azatepa, azomycin, batimastat, be
  • the present disclosure relates to compounds represented by the general formula (I) of the present disclosure and pharmaceutically acceptable salts thereof for inhibiting tyrosine kinase (TK).
  • the present disclosure relates to compounds represented by the general formula (I) of the present disclosure and their pharmaceutically acceptable salts for treating or preventing diseases or disease states mediated by tyrosine kinase (TK).
  • TK tyrosine kinase
  • the present disclosure relates to the compounds represented by the general formula (I) of the present disclosure and their pharmaceutically acceptable salts or the use of the compounds of the present disclosure and their pharmaceutically acceptable salts in the preparation of drugs for inhibiting tyrosine kinase (TK).
  • TK tyrosine kinase
  • the present disclosure relates to the compounds represented by the general formula (I) of the present disclosure and their pharmaceutically acceptable salts, or the compounds of the present disclosure and their pharmaceutically acceptable salts in the preparation of drugs for treating or preventing diseases or disease states mediated by tyrosine kinase (TK).
  • TK tyrosine kinase
  • reagents and equipment used in the examples of the present disclosure are conventional and commercially available. For example:
  • Step a 4-chloro-6-iodo-7H-pyrrolo[2,3-d]pyrimidine (5.0 g, 17.9 mmol) was added to NaH (1.08 g, 27 mmol, 60%) was added to the mixture in THF (60 mL), and the mixture was stirred at 0°C for 30 minutes. SEMCl (3.58 g, 3.8 mL, 8.64 mmol) was added to the reaction mixture, which was warmed to room temperature and stirred for 2 hours, and quenched by saturated NH 4 C solution.
  • Step b To a mixture of 2-amino-4-methoxyphenol (1.39 g, 10 mmol) in THF (100 mL) was added phenyl p-chloroisothiocyanate (2.54 g, 15 mmol) and K 2 CO 3 (2.764 g, 20 mmol). The reaction was stirred at room temperature overnight. NaIO 4 solution (427.78 mg in 100 mL H 2 O, 20 mol%) was poured into the reaction. TLC showed that 2-amino-4-methoxyphenol was completely consumed. Extracted with EA. Concentrated and purified by silica gel column to give the desired intermediate (2.1976 g, 80%).
  • Step c To a suspension of the product from step b (2.1976 g, 8 mmol) in DCM (80 mL) was added BBr 3 ⁇ DCM (2M, 20 mL, 5 equivalents). The reaction was stirred at room temperature. The reaction was quenched with MeOH until TLC showed no starting material remaining. Concentration and purification by silica gel column gave the desired product F2 (1.88 g, 90%).
  • Step e Pd(dppf)Cl 2 (408.3 mg, 0.5 mmol), K 2 CO 3 (6.91 mg, 50 mmol) and pinacol borane (5.06 g, 20 mmol) were added to a solution of the product of step d (2.83 g, 10 mmol) in DMF (50 mL). The nitrogen atmosphere was replaced 3 times, and then stirred at 100° C. for 4 hours. The mixture was extracted with EA, washed with brine, dried over sodium sulfate, concentrated and purified by silica gel column to give the desired product F3 (2.805 g, 85%) as a yellow oil.
  • Step f A 100 mL round-bottom flask was charged with compound F1 (1.9632 g, 4.8 mmol), Pd(PPh 3 ) 2 Cl 2 (280.7 mg, 0.4 mmol), K 2 CO 3 (1.66 g, 12 mmol) and compound F3 (1.346 g, 4.0 mmol) in dioxane/H 2 O (36 mL/4 mL, 0.1 M). Nitrogen was replaced 3 times, and then stirred at 100° C. overnight. Concentration and purification by silica gel column gave the desired intermediate (1.34 g, 69%) as a yellow solid.
  • Step g Compound F2 (0.343 g, 1.32 mmol) and Cs 2 CO 3 (0.643 g, 1.974 mmol) were added to a solution of the product obtained in the previous step (0.32 g, 0.658 mmol) in NMP (3 mL, 0.2 M). The reaction was stirred at 120°C for 4 hours. Extracted with EA, washed with saturated NaCl solution and dried over Na 2 SO 4. Purified with silica gel column to give the desired product (288.8 mg, 62%) as a brown solid.
  • Step h DCM/TFA (4mL/4mL, 0.1M) was added to the product in step g (288.8mg, 0.406mmol). The reaction was stirred at room temperature. After 1 hour, TLC showed that no raw material remained. Concentration removed most of the DCM and TFA. Et2O was added to obtain the TFA salt of the product. Extracted with EA, the pH was adjusted to 12 to make the compound a free amine. The crude product was further purified by silica gel column to obtain the title compound YH-I-001 (153.7mg, 65%) as a white solid.
  • Step i To a solution of compound F1 (0.82 g, 2 mmol) in NMP (4 mL, 0.2 M) was added compound F2 (0.51 g, 2 mmol) and Cs 2 CO 3 (1.629 g, 5 mmol). The reaction was stirred at 120° C. for 4 hours. Extracted with EA, washed with saturated NaCl solution and dried over Na 2 SO 4. Purified with silica gel column to give the desired product (509.8 mg, 40%) as a white solid.
  • Step j To a mixture of 3,5-dimethylpyrrolopinacol (1.11 g, 5.0 mmol) in THF (15 mL) was added NaH (0.30 g, 7.5 mmol, 60%), and the mixture was stirred at 0°C for 30 minutes. SEMCl (2.49 g, 2.66 mL, 6.0 mmol) was added to the reaction mixture, which was warmed to room temperature and stirred for 2 hours, and quenched by saturated NH 4 Cl solution.
  • Step k To a mixture of 3,5-dimethyl-4-bromopyrrole (1.75 g, 10.0 mmol) in DMF (33 mL) was added NaH (0.60 g, 15 mmol, 60%), and the mixture was stirred at 0 ° C for 30 minutes. 4-Sulfonate-Boc piperidine (3.35 g, 12 mmol, 5.32 mL) was added to the reaction mixture, which was warmed to room temperature and stirred for 2 hours, and quenched by saturated NH4Cl solution.
  • Step 1 Pd(OAc) 2 (11.2 mg, 0.05 mmol), KOAc (408.3 mg, 2 mmol) and pinacol borane (0.506 g, 2 mmol) were added to a solution of the product of step k (179.14 mg, 0.5 mmol) in DMA (2 mL). Nitrogen was replaced 3 times, and then stirred at 100°C for 4 hours. The mixture was extracted with EA, washed with brine, dried over anhydrous sodium sulfate, concentrated and purified by silica gel column to obtain The desired intermediate (0.108 g, 53%) was obtained as a tan oil.
  • test compound Dilute 1 volume of protein culture solution 4 times, and add 50 ⁇ M dithiothreitol as culture solution for standby use. Add different concentrations of the test compound to the 384-well plate (10 ⁇ M as the starting concentration, 3-fold dilution, 10 concentrations, duplicate wells). Seal the 384-well plate to be tested and centrifuge the plate at 1000g for 1 minute. After mixing 2 times ALK protein (test protein) with 1 times protein culture solution, take 2.5 ⁇ L of protein mixture and add it to each well plate, centrifuge the plate at 1000g for 30s, and react at room temperature for 10min. At the same time, mix 2 times substrate and ATP with 1 times protein culture solution and shake well.
  • ALK protein test protein
  • Discard the culture medium add 150 ⁇ L of dimethyl sulfoxide, and shake for 15 minutes.
  • the compound incubation time for Kelly, SH-SY5Y, TGW, and NCIH2228 cells is 72 hours, and the compound incubation time for SUDHL-1, SK-N-SH, and KP-N-RT-BM-1 cells is 120 hours.
  • the compounds disclosed in the present invention were tested for pharmacokinetic properties, and the test operation was as follows: the compound to be tested was dissolved and prepared into a storage solution of the required concentration for standby use. Then, male healthy mice were divided into groups of 3 mice per group. The mice were fasted for one night, allowed to drink water freely, and then fed and administered 4 hours later. The dosage was as shown in the following table. Whole blood samples were collected through the orbital vein at designated time points (0.083h, 0.25h, 0.5h, 1h, 2h, 4h, 8h, 24h), and 30 ⁇ L of the sample was flowed into the sodium heparin anticoagulation tube and centrifuged at 4000rcf/4°C for 5 minutes before taking the upper plasma sample. Finally, LC-MS/MS was used to collect data on the samples, and after data processing, the pharmacokinetic parameters of the compound to be tested were obtained.
  • Tables 5 and 6 show the blood concentration-time data of the compound YH-I-001 of the present disclosure.
  • FIG2 and FIG3 show the blood concentration-time curves of the compound YH-I-001 disclosed in the present invention.
  • KELLY (brain, neuroblastoma, purchased from ECACC, Cat No.: 92110411, Lot No.14A023) cells were cultured in vitro in monolayers under the conditions of RPMI-1640 medium with 2mM Glutamine, 10% heat-inactivated fetal bovine serum, and cultured at 37°C without CO2 . Digestion and passage were performed with trypsin-EDTA twice a week.
  • the experimental index is to examine whether tumor growth can be inhibited, delayed or cured.
  • the tumor diameter is measured with a vernier caliper twice a week.
  • the anti-tumor efficacy of the compound is evaluated by T/C (%).
  • T/C% TRTV/CRTV ⁇ 100% (TRTV: RTV of the treatment group; CRTV: RTV of the negative control group).
  • V 1 is the tumor volume measured at the time of cage administration (i.e.
  • TGI tumor volume measured on the tth day.
  • TGI (%) (1-(TV treatment-Dn -TV treatment-D1 )/(TV Control-Dn -TV Control-D1 )) ⁇ 100%
  • TV Control tumor volume of the control group
  • TV Treatment tumor volume of the treatment group.
  • TW C tumor weight of control group
  • TWT tumor weight of treatment group.
  • TGI tumor inhibition rate
  • relational terms such as first and second, etc. are used merely to distinguish one entity or operation from another entity or operation, but do not necessarily require or imply any such actual relationship or order between these entities or operations.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

L'invention concerne un procédé de traitement ou de prévention d'une tumeur. Le procédé comprend : l'administration, à un individu nécessitant un tel procédé, d'une quantité thérapeutiquement ou prophylactiquement efficace d'un composé représenté par la formule générale (I), ou d'un sel pharmaceutiquement acceptable de celui-ci : formule générale (I), R1, R2, X et Y étant tels que définis dans la présente invention.
PCT/CN2023/127066 2022-10-27 2023-10-27 Procédé de traitement ou de prévention d'une tumeur WO2024088379A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211329128 2022-10-27
CN202211329128.3 2022-10-27

Publications (1)

Publication Number Publication Date
WO2024088379A1 true WO2024088379A1 (fr) 2024-05-02

Family

ID=90830130

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/127066 WO2024088379A1 (fr) 2022-10-27 2023-10-27 Procédé de traitement ou de prévention d'une tumeur

Country Status (1)

Country Link
WO (1) WO2024088379A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004085425A1 (fr) * 2003-03-21 2004-10-07 Amgen Inc Azoles fusionnes tels que benzimidazoles, benzoxazoles et benzothiazles 2,5-disubstitues comme inhibiteurs de kinase
WO2006017443A2 (fr) * 2004-08-02 2006-02-16 Osi Pharmaceuticals, Inc. Composes a base de pyrrolopyrimidine a substitution arylamine inhibiteurs de kinases multiples
WO2008016192A2 (fr) * 2006-08-04 2008-02-07 Takeda Pharmaceutical Company Limited Dérivé hétérocyclique fusionné et son procédé d'utilisation
US20130096136A1 (en) * 2011-09-30 2013-04-18 Methylgene Inc. Inhibitors of Protein Tyrosine Kinase Activity

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004085425A1 (fr) * 2003-03-21 2004-10-07 Amgen Inc Azoles fusionnes tels que benzimidazoles, benzoxazoles et benzothiazles 2,5-disubstitues comme inhibiteurs de kinase
WO2006017443A2 (fr) * 2004-08-02 2006-02-16 Osi Pharmaceuticals, Inc. Composes a base de pyrrolopyrimidine a substitution arylamine inhibiteurs de kinases multiples
WO2008016192A2 (fr) * 2006-08-04 2008-02-07 Takeda Pharmaceutical Company Limited Dérivé hétérocyclique fusionné et son procédé d'utilisation
US20130096136A1 (en) * 2011-09-30 2013-04-18 Methylgene Inc. Inhibitors of Protein Tyrosine Kinase Activity

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MICHELE H POTASHMAN, JAMES BREADY, ANGELA COXON, THOMAS M DEMELFI, LUCIAN DIPIETRO, NICHOLAS DOERR, DANIEL ELBAUM, JUAN ESTRADA, P: "Design, synthesis, and evaluation of orally active benzimidazoles and benzoxazoles as vascular endothelial growth factor-2 receptor tyrosine kinase inhibitors", JOURNAL OF MEDICINAL CHEMISTRY, UNITED STATES, 6 September 2007 (2007-09-06), United States , pages 4351 - 4373, XP055135231, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pubmed/17696416> DOI: 10.1021/jm070034i *
RUAN, ZHIHUA ET AL.: "Pseudo Receptor Probes: A Novel Pseudo Receptor-based QSAR Method and Application into Studies on a New Kind of Selective Vascular Endothelial Growth Factor-2 Receptor Inhibitors", CHEMOMETRICS AND INTELLIGENT LABORATORY SYSTEMS, vol. 92, no. 2, 8 March 2008 (2008-03-08), pages 157 - 168, XP022678586, DOI: 10.1016/j.chemolab.2008.02.007 *

Similar Documents

Publication Publication Date Title
TWI627173B (zh) 作為NIK抑制劑的新穎3-(1H-吡唑-4-基)-1H-吡咯并[2,3-c]吡啶衍生物
CN102625807B (zh) 4-取代的吡啶-3-基-甲酰胺化合物和使用方法
CN110621316A (zh) 用ehmt2抑制剂进行的组合疗法
US20190031662A1 (en) Aurora kinase inhibitors for inhibiting mitotic progression
KR20210075992A (ko) Sting 작동 화합물
US20230145003A1 (en) Compounds and uses thereof
US10815225B2 (en) Antiproliferation compounds and uses thereof
US11939331B2 (en) Tricyclic heterocycles as FGFR inhibitors
KR20170129757A (ko) Fgfr4 억제제로서의 포르밀화 n-헤테로시클릭 유도체
US20200071326A1 (en) Tam kinase inhibitors
US20230002384A1 (en) Tricyclic heterocycles as fgfr inhibitors
US20230114765A1 (en) Tricyclic compounds as inhibitors of kras
US20240140931A1 (en) Tricyclic compounds and uses thereof
CN114437116A (zh) 杂环化合物及其制备方法、药物组合物和应用
WO2024088379A1 (fr) Procédé de traitement ou de prévention d&#39;une tumeur
WO2024088377A1 (fr) Analogue de pyrimidopyrrole
US20220389033A1 (en) Hetero-bicyclic inhibitors of kras
CN115368378A (zh) 取代的大环化合物及包含该化合物的组合物及其用途
JP2021534114A (ja) 併用療法
JP2022507514A (ja) Erkインヒビター及びその使用
CN113993871B (zh) 含有5-氮杂螺庚烷的btk抑制剂
WO2024040131A1 (fr) Inhibiteurs de pyridopyrimidine kras
WO2024006726A2 (fr) Composés utilisés en tant qu&#39;inhibiteurs d&#39;axl
US20220242849A1 (en) Wdr5 inhibitors and modulators
WO2023174406A1 (fr) Inhibiteur sous forme de dérivé hétérocyclique contenant de l&#39;azote, son procédé de préparation et son utilisation