WO2020200069A1 - 吡咯并杂环类衍生物、其制备方法及其在医药上的应用 - Google Patents

吡咯并杂环类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2020200069A1
WO2020200069A1 PCT/CN2020/081591 CN2020081591W WO2020200069A1 WO 2020200069 A1 WO2020200069 A1 WO 2020200069A1 CN 2020081591 W CN2020081591 W CN 2020081591W WO 2020200069 A1 WO2020200069 A1 WO 2020200069A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
general formula
alkyl
compound represented
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2020/081591
Other languages
English (en)
French (fr)
Inventor
李心
蔡国栋
杨芳
贺峰
陶维康
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
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 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to KR1020217033781A priority Critical patent/KR20210146956A/ko
Priority to CA3135070A priority patent/CA3135070A1/en
Priority to MX2021011823A priority patent/MX2021011823A/es
Priority to CN202080019049.5A priority patent/CN113544131B/zh
Priority to AU2020251621A priority patent/AU2020251621A1/en
Priority to BR112021018924A priority patent/BR112021018924A2/pt
Priority to JP2021557719A priority patent/JP2022528083A/ja
Priority to US17/600,043 priority patent/US20220185818A1/en
Priority to EP20783525.7A priority patent/EP3950690A4/en
Publication of WO2020200069A1 publication Critical patent/WO2020200069A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • 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
    • 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 belongs to the field of medicine, and relates to a pyrrolo heterocyclic derivative, a preparation method thereof, and application in medicine.
  • the present disclosure relates to the pyrrolo heterocyclic derivatives represented by the general formula (I), their preparation methods and pharmaceutical compositions containing the derivatives, and their use as ERK inhibitors to treat ERK-mediated diseases and disorders or Inhibit the use of MAPK-ERK signaling pathway.
  • Mitogen-activated protein kinase plays an extremely important role in the signal transduction pathway, and extracellular signal regulated kinase (ERK) is a member of the MAPK family.
  • ERK extracellular signal regulated kinase
  • RAS-RAF-MEK-ERK the exogenous stimulating signal is transmitted to the ERK, and the activated ERK transfers into the nucleus to regulate the activity of transcription factors, thereby regulating the biological functions of cell proliferation, differentiation and apoptosis, or through phosphorylation
  • the cytoskeleton components in the cytoplasm are involved in the regulation of cell morphology and the redistribution of the cytoskeleton.
  • B-RAF/MEK inhibitors not only inhibit tumor growth, but also regulate the immune microenvironment of tumors.
  • B-RAF/MEK inhibitors can enhance the expression of tumor-specific antigens, improve the recognition and killing of tumors by antigen-specific T cells, and promote the migration and infiltration of immune cells.
  • the expression of PD-L1 in tumor tissues is enhanced.
  • checkpoint molecule antibodies such as PD-1 antibody, CTLA4 antibody
  • BioMed Valley Discoveries' BVD-523 is in the second clinical phase
  • Merck's MK-8353 and Astex's Astex-029 are in the first clinical phase.
  • Related patents are WO1999061440A1, WO2001056557A2, WO2001056993A2, WO2001057022A2, WO2002022601A1, WO2012118850A1, WO2013018733A1, WO2014179154A2, WO2015103133A1, WO2016192063A1, WO2017180817A1, WO2018049127A1.
  • R 1 is selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, aminoalkyl and nitro, and the alkyl is optionally NR 7 R 8 , alkoxy, halogen, cyano, nitro, hydroxy, and hydroxyalkyl substituted by one or more substituents;
  • R 2 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino and nitro;
  • R 3 is selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclic, aryl and heteroaryl
  • the alkyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group are optionally further selected from alkyl, alkoxy, oxo, halogen, amino, cyano, nitro, and hydroxy. , Hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl substituted by one or more substituents;
  • R 4 is selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino and nitro;
  • R 5 is the same or different, each independently selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocycle Group, aryl and heteroaryl;
  • R 6 is selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino and nitro;
  • R 7 and R 8 are the same or different, and are each independently selected from a hydrogen atom, an alkyl group, a hydroxyalkyl group and a halogenated alkyl group;
  • n is selected from 0, 1, 2, 3, 4 or 5;
  • n is selected from 0, 1, 2 or 3;
  • z is selected from 0, 1, 2, 3 or 4;
  • Q is selected from 0, 1, or 2.
  • the compound represented by general formula (I) is a compound represented by general formula (I-P):
  • R 1 to R 6 , m, n, z, and Q are as defined in the general formula (I).
  • the compound represented by general formula (I) is a compound represented by general formula (II):
  • R 1 to R 6 , m, z, and Q are as defined in the general formula (I).
  • the compound represented by general formula (I) is a compound represented by general formula (II-P):
  • R 1 to R 6 , m, z, and Q are as defined in the general formula (I).
  • R 1 is selected from a hydrogen atom, a C 1-6 alkyl group, a hydroxyl group, an amino C 1-6 alkyl group, a C 1-6 alkylamino C 1-6 alkyl group and a C 1-6 hydroxyalkyl group;
  • R 1 is selected from hydrogen atom, methyl group, hydroxymethyl group, aminomethyl group and methylaminomethyl group.
  • the compound represented by general formula (I) is a compound represented by general formula (III):
  • p is selected from 0, 1, 2 or 3, preferably 1;
  • R 2 , R 3 , R 5 , R 6 , m, n, z, and Q are as defined in the general formula (I).
  • the compound represented by general formula (I) is a compound represented by general formula (III-P):
  • R 2 , R 3 , R 5 , R 6 , m, n, z, p, and Q are as defined in the general formula (III).
  • the compound represented by the general formula (I) or its stereoisomers, tautomers, mesoisomers, racemates, enantiomers , Diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof wherein said R 3 is heteroaryl, and said heteroaryl is optionally further selected from alkyl, alkoxy, One or more substituents of oxo, halogen, amino, cyano, nitro, hydroxy and hydroxyalkyl; preferably, R 3 is a 5-10 membered heteroaryl group, said 5-10 The membered heteroaryl group is optionally further selected from C 1-6 alkyl, C 1-6 alkoxy, oxo, halogen, amino, cyano, nitro, hydroxyl and C 1-6 hydroxyalkyl One or more substituents are substituted; more preferably, R 3 is pyrazolyl, and the pyrazolyl is optionally substituted with C 1-6 alkyl, preferably
  • Typical compounds of the present disclosure include but are not limited to:
  • R w is a hydroxyl protecting group
  • R 2 , R 3 , R 5 , R 6 , m, n, p, Q, and z are as defined in the general formula (III).
  • Another aspect of the present disclosure relates to a compound represented by the general formula (III-PA):
  • R w is a hydroxyl protecting group
  • R 2 , R 3 , R 5 , R 6 , m, n, p, Q, and z are as defined in the general formula (III).
  • Typical compounds of general formula (IIIA) of the present disclosure include but are not limited to:
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (III), the method comprising the following steps:
  • the hydroxy protecting group R w is preferably TBS
  • R 2 , R 3 , R 5 , R 6 , m, n, p, Q, and z are as defined in the general formula (III).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (III-P), the method comprising the following steps:
  • the hydroxy protecting group R w is preferably TBS
  • R 2 , R 3 , R 5 , R 6 , m, n, p, Q, and z are as defined in the general formula (III).
  • Another aspect of the present disclosure relates to a pharmaceutical composition containing a therapeutically effective amount of a compound represented by general formula (I) or general formula (II) or general formula (III) of the present disclosure or its stereoisomers Conformers, tautomers, mesoisomers, racemates, enantiomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, and one or more pharmaceuticals Acceptable carrier, diluent or excipient.
  • a pharmaceutical composition containing a therapeutically effective amount of a compound represented by general formula (I) or general formula (II) or general formula (III) of the present disclosure or its stereoisomers Conformers, tautomers, mesoisomers, racemates, enantiomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, and one or more pharmaceuticals Acceptable carrier, diluent or excipient.
  • the present disclosure further relates to compounds represented by general formula (I) or general formula (II) or general formula (III) or their stereoisomers, tautomers, mesoisomers, racemates, enantiomers Use of isomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them, in the preparation of drugs for inhibiting ERK.
  • the present disclosure further relates to compounds represented by general formula (I) or general formula (II) or general formula (III) or their stereoisomers, tautomers, mesoisomers, racemates, enantiomers Use of isomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them in the preparation of drugs for the treatment or prevention of cancer, inflammation, or other proliferative diseases ,
  • the cancer is selected from melanoma, liver cancer, kidney cancer, lung cancer (such as non-small cell lung cancer or small cell lung cancer), nasopharyngeal cancer, colorectal cancer , Pancreatic cancer, cervical cancer, ovarian cancer, breast cancer, bladder cancer, prostate cancer, leukemia, head and neck squamous cell carcinoma, cervical cancer, thyroid cancer, lymphoma, sarcoma, neuroblastoma, brain tumor, myeloma ( Such as multiple mye
  • the present disclosure also relates to a method for inhibiting ERK, which comprises administering to a desired patient a therapeutically effective amount of a compound represented by general formula (I) or general formula (II) or general formula (III) or its stereoisomers, mutual Tautomers, mesosomes, racemates, enantiomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them.
  • a method for inhibiting ERK which comprises administering to a desired patient a therapeutically effective amount of a compound represented by general formula (I) or general formula (II) or general formula (III) or its stereoisomers, mutual Tautomers, mesosomes, racemates, enantiomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them.
  • the present disclosure also relates to a method for treating or preventing ERK-mediated diseases, which comprises administering to a patient a therapeutically effective amount of a compound represented by general formula (I) or general formula (II) or general formula (III) or Stereoisomers, tautomers, mesoisomers, racemates, enantiomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or drugs containing them combination.
  • the present disclosure also relates to a method for treating or preventing cancer, inflammation, or other proliferative diseases, preferably a method for treating cancer, which comprises administering to a patient a therapeutically effective amount of general formula (I) or general formula (II) or general The compound represented by formula (III) or its stereoisomers, tautomers, mesoisomers, racemates, enantiomers, diastereomers, or mixtures thereof or A pharmaceutically acceptable salt, or a pharmaceutical composition containing the same; wherein the cancer is selected from melanoma, liver cancer, kidney cancer, lung cancer (such as non-small cell lung cancer or small cell lung cancer), nasopharyngeal cancer, colorectal cancer, pancreas Cancer, cervical cancer, ovarian cancer, breast cancer, bladder cancer, prostate cancer, leukemia, head and neck squamous cell carcinoma, cervical cancer, thyroid cancer, lymphoma, sarcoma, neuroblastoma, brain tumor, myeloma (such as
  • the present disclosure further relates to a compound represented by general formula (I) or general formula (II) or general formula (III) or its stereoisomers, tautomers, mesosomes, racemates, Enantiomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them, which are used as drugs.
  • the present disclosure also relates to compounds represented by general formula (I) or general formula (II) or general formula (III) or their stereoisomers, tautomers, mesosomes, racemates, enantiomers Isomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them, which are used as ERK inhibitors.
  • the present disclosure also relates to compounds represented by general formula (I) or general formula (II) or general formula (III) or their stereoisomers, tautomers, mesosomes, racemates, enantiomers Isomers, diastereomers, or mixtures thereof or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them, which are used to treat or prevent ERK-mediated diseases.
  • the present disclosure also relates to compounds represented by general formula (I) or general formula (II) or general formula (III) or their stereoisomers, tautomers, mesosomes, racemates, enantiomers Isomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them, which are used for the treatment or prevention of cancer, inflammation, or other proliferative diseases, preferably for treatment Or prevent cancer; wherein the cancer is selected from melanoma, liver cancer, kidney cancer, lung cancer (such as non-small cell lung cancer or small cell lung cancer), nasopharyngeal cancer, colorectal cancer, pancreatic cancer, cervical cancer, ovarian cancer, breast cancer Cancer, bladder cancer, prostate cancer, leukemia, head and neck squamous cell carcinoma, cervical cancer, thyroid cancer, lymphoma, sarcoma, neuroblastoma, brain tumor, myeloma (such as multiple myeloma), astrocytoma
  • the active compound can be prepared in a form suitable for administration by any appropriate route, and the active compound is preferably in a unit dose form, or a form in which the patient can self-administer in a single dose.
  • the expression of the unit dose of the compound or composition of the present disclosure can be tablet, capsule, cachet, bottled syrup, powder, granule, lozenge, suppository, regenerated powder or liquid preparation.
  • the dosage of the compound or composition used in the treatment methods of the present disclosure will generally vary with the severity of the disease, the weight of the patient, and the relative efficacy of the compound.
  • a suitable unit dose can be 0.1-1000 mg.
  • the pharmaceutical composition of the present disclosure may contain one or more auxiliary materials selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients Wait.
  • auxiliary materials selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients Wait.
  • the composition may contain 0.1 to 99% by weight of the active compound.
  • the pharmaceutical composition containing the active ingredient may be in a form suitable for oral administration, such as tablets, dragees, lozenges, water or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or Elixirs.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions. Such compositions can contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents and preservatives, In order to provide pleasing and delicious medicinal preparations.
  • the tablet contains the active ingredient and non-toxic pharmaceutically acceptable excipients suitable for the preparation of tablets for mixing.
  • Aqueous suspensions contain the active substance and excipients suitable for the preparation of aqueous suspensions for mixing.
  • the aqueous suspension may also contain one or more preservatives such as ethyl paraben or n-propyl paraben, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents. Flavoring agent.
  • Oil suspensions can be formulated by suspending active ingredients in vegetable oils. Oil suspensions may contain thickeners. The above-mentioned sweeteners and flavoring agents can be added to provide a palatable preparation.
  • dispersible powders and granules suitable for preparing aqueous suspensions can be provided with active ingredients and dispersing or wetting agents for mixing, suspending agents or one or more preservatives. Suitable dispersing or wetting agents and suspending agents can illustrate the above examples. Other excipients such as sweetening agents, flavoring agents and coloring agents may also be added. These compositions are preserved by adding antioxidants such as ascorbic acid.
  • composition of the present disclosure may also be in the form of an oil-in-water emulsion.
  • the pharmaceutical composition may be in the form of a sterile injectable aqueous solution.
  • Acceptable solvents or solvents that can be used are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injection preparation may be a sterile oil-in-water injection microemulsion in which the active ingredient is dissolved in an oil phase.
  • the active ingredient is dissolved in a mixture of soybean oil and lecithin.
  • the oil solution is added to the mixture of water and glycerin to form a microemulsion.
  • the injection or microemulsion can be injected into the patient's bloodstream by local large injections.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM. 5400 intravenous pump.
  • the pharmaceutical composition may be in the form of a sterile injection water or oil suspension for intramuscular and subcutaneous administration.
  • the suspension can be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents mentioned above.
  • the sterile injection preparation may also be a sterile injection solution or suspension prepared in a parenterally acceptable non-toxic diluent or solvent.
  • sterile fixed oil can be conveniently used as a solvent or suspension medium.
  • the compounds of the present disclosure can be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid in the rectum and thus will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, polyethylene glycols of various molecular weights and mixtures of fatty acid esters of polyethylene glycol.
  • the dosage of the drug depends on many factors, including but not limited to the following factors: the activity of the specific compound used, the age of the patient, the weight of the patient, the health of the patient, and the behavior of the patient , The patient’s diet, time of administration, mode of administration, rate of excretion, combination of drugs, etc.; in addition, the best mode of treatment such as the mode of treatment, the daily dosage of compound (I) or the amount of pharmaceutically acceptable salt
  • the type can be verified according to the traditional treatment plan.
  • alkyl refers to a saturated aliphatic hydrocarbon group, which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 12 carbon atoms, more preferably containing 1 to 6 carbons Atom of the alkyl group.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 , 2-Dimethylpropyl, 2,2-Dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 -Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -Methylhexyl, 3-methylhexyl, 4-methylhe
  • a lower alkyl group containing 1 to 6 carbon atoms non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Group, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Group, 2,3-dimethylbutyl, etc.
  • Alkyl groups may be substituted or unsubstituted. When substituted, substituents may be substituted at any available attachment point.
  • the substituents are preferably one or more of the following groups, which are independently selected from alkanes Group, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkane
  • One or more substituents of oxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio and oxo are substituted.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent.
  • the cycloalkyl ring contains 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 8 Carbon atoms, most preferably containing 3 to 6 (e.g. 3, 4, 5 or 6) carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene Cycloalkyl group, cyclooctyl group, etc. are preferred; polycyclic cycloalkyl group includes spiro ring, fused ring and bridged ring cycloalkyl group.
  • spirocycloalkyl refers to a polycyclic group that shares one carbon atom (called a spiro atom) between 5- to 20-membered monocyclic rings. It may contain one or more double bonds, but none of the rings have complete conjugate ⁇ electronic system. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan.
  • the spirocycloalkyl group is classified into a single spirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a single spirocycloalkyl group and a bispirocycloalkyl group. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered monospirocycloalkyl.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to a 5- to 20-membered all-carbon polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ electron system. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan. According to the number of constituent rings, it can be classified into bicyclic, tricyclic, tetracyclic or polycyclic condensed cycloalkyls, preferably bicyclic or tricyclic, and more preferably 5-membered/5-membered or 5-membered/6-membered bicyclic alkyl.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to a 5- to 20-membered, all-carbon polycyclic group with any two rings sharing two carbon atoms that are not directly connected. It may contain one or more double bonds, but no ring has complete Conjugated ⁇ electron system. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan. It can be classified into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyls according to the number of constituent rings, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring includes the above-mentioned cycloalkyl groups (such as monocyclic, fused ring, spiro ring and bridged cycloalkyl) fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the ring is connected to the parent structure
  • the ring together is a cycloalkyl group, and non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, etc.; preferably phenylcyclopentyl, tetrahydronaphthyl.
  • Cycloalkyl groups may be optionally substituted or unsubstituted.
  • the substituents are preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthio and oxo groups are substituted by one or more substituents.
  • alkoxy refers to -O- (alkyl) and -O- (unsubstituted cycloalkyl), where alkyl or cycloalkyl is as defined above.
  • alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from hydrogen atom, halogen, alkyl, alkoxy, haloalkyl , Hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl substituted by one or more substituents.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, which contains 3 to 20 ring atoms, one or more of which are selected from nitrogen, oxygen, sulfur, S( O) or S(O) 2 heteroatoms, but not including the ring part of -OO-, -OS- or -SS-, and the remaining ring atoms are carbon. It preferably contains 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; preferably contains 3 to 8 ring atoms, of which 1 to 3 are heteroatoms; preferably contains 3 to 6 ring atoms, of which 1 to 3 Are heteroatoms.
  • Non-limiting examples of monocyclic heterocyclic groups include azetidinyl, pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydrofuranyl Hydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, etc., preferably tetrahydropyranyl, piperidinyl, pyrrolidinyl.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclic group refers to a polycyclic heterocyclic group sharing one atom (called a spiro atom) between 5- to 20-membered monocyclic rings, in which one or more ring atoms are selected from nitrogen, oxygen, sulfur, and S (O) or S(O) 2 , the remaining ring atoms are carbon. It can contain one or more double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 yuan, more preferably 7 to 11 yuan.
  • the spiro heterocyclic group is classified into a single spiro heterocyclic group, a dispiro heterocyclic group or a polyspiro heterocyclic group, preferably a single spiro heterocyclic group and a dispiro heterocyclic group. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered monospiro heterocyclic group.
  • spiroheterocyclic groups include:
  • fused heterocyclic group refers to a 5- to 20-membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system.
  • One or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system, where one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the rest of the ring
  • the atom is carbon. It is preferably 6 to 14 yuan, more preferably 7 to 11 yuan.
  • fused heterocyclic groups include:
  • bridged heterocyclic group refers to a 5- to 14-membered polycyclic heterocyclic group with any two rings sharing two atoms that are not directly connected. It may contain one or more double bonds, but none of the rings has a complete common A conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer of 0 to 2), and the remaining ring atoms are carbon. It is preferably 6 to 14 yuan, more preferably 7 to 11 yuan.
  • bridged heterocyclic groups include:
  • the heterocyclic ring includes the above heterocyclic groups (such as monocyclic, fused ring, spiro ring and bridged heterocyclic group) fused to an aryl, heteroaryl or cycloalkyl ring, wherein the parent structure is connected to The ring together is a heterocyclic group, non-limiting examples of which include:
  • the heterocyclic group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthio and oxo groups are substituted by one or more substituents.
  • aryl refers to a 6 to 20-membered all-carbon monocyclic or fused polycyclic (that is, rings sharing adjacent pairs of carbon atoms) group with a conjugated ⁇ -electron system, preferably 6 to 10 members, more preferably 6 members, such as phenyl and naphthyl.
  • the aryl ring includes the above-mentioned aryl group fused to a heteroaryl, heterocyclic or cycloalkyl ring, wherein the ring connected to the parent structure is an aryl ring, and non-limiting examples thereof include:
  • Aryl groups may be substituted or unsubstituted.
  • the substituents are preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, and heterocycle
  • One or more substituents in the alkylthio group are substituted.
  • heteroaryl refers to a heteroaromatic system containing 1 to 4 heteroatoms and 5 to 20 ring atoms, where the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • Heteroaryl groups are preferably 5 to 10 members, containing 1 to 3 heteroatoms; more preferably 5 or 6 members, containing 1 to 3 heteroatoms; non-limiting examples are pyrazolyl, imidazolyl, furanyl, Thienyl, thiazolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc.
  • the heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring connected to the parent structure is a heteroaryl ring, non-limiting examples of which include:
  • the heteroaryl group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio And one or more substituents in the heterocycloalkylthio group.
  • cycloalkyloxy refers to -O-cycloalkyl, where cycloalkyl is as defined above.
  • haloalkyl refers to an alkyl substituted by halogen, where alkyl is as defined above.
  • haloalkoxy refers to an alkoxy substituted by halogen, wherein alkoxy is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group, where the alkyl group is as defined above.
  • aminoalkyl refers to an alkyl group substituted with an amino group, wherein the alkyl group is as defined above.
  • alkylaminoalkyl refers to an alkyl group substituted with an alkylamino group, where the alkyl group is as defined above.
  • hydroxy refers to the -OH group.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • amino refers to -NH 2 .
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • aldehyde group refers to -C(O)H.
  • carboxylate group refers to -C(O)O(alkyl) or -C(O)O(cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may but need not be present, and the description includes the case where the heterocyclic group is substituted by an alkyl group and the case where the heterocyclic group is not substituted by an alkyl group .
  • Substituted refers to one or more hydrogen atoms in the group, preferably at most 5, more preferably 1 to 3 hydrogen atoms independently of each other by a corresponding number of substituents, wherein each substituent has an independent (I.e. the substituents can be the same or different). It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art can determine (by experiment or theory) possible or impossible substitutions without too much effort. For example, an amino group or a hydroxyl group with free hydrogen may be unstable when combined with a carbon atom with an unsaturated (eg, olefinic) bond.
  • “Pharmaceutical composition” means a mixture containing one or more of the compounds described herein or their physiologically/pharmaceutically acceptable salts or prodrugs and other chemical components, and other components such as physiological/pharmaceutically acceptable carriers And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, which is beneficial to the absorption of the active ingredients and thus the biological activity.
  • “Pharmaceutically acceptable salt” refers to the salt of the compound of the present disclosure. Such salt is safe and effective when used in the body of a mammal, and has due biological activity.
  • the compounds of the present disclosure may also include isotopic derivatives thereof.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • 18 F-fluorine label 18 F isotope
  • 11 C-, 13 C-, or 14 C-rich Compounds in which collective carbons ( 11 C-, 13 C-, or 14 C-carbon labels; 11 C-, 13 C-, or 14 C-isotopes) replace carbon atoms are within the scope of the present disclosure.
  • Such compounds can be used, for example, as analytical tools or probes in biological assays, or as tracers for in vivo diagnostic imaging of diseases, or as tracers for pharmacodynamics, pharmacokinetics, or receptor studies.
  • Deuterated compounds can generally retain activity comparable to that of non-deuterated compounds, and when deuterated at certain specific sites, they can achieve better metabolic stability, thereby obtaining certain therapeutic advantages (such as increased in vivo half-life or reduced dosage requirements) ).
  • the term "therapeutically effective amount” refers to a sufficient amount of a drug or agent that is non-toxic but can achieve the desired effect.
  • the determination of the effective amount varies from person to person, and depends on the age and general conditions of the recipient, as well as the specific active substance. The appropriate effective amount in a case can be determined by those skilled in the art according to routine experiments.
  • the preparation method of the form or its pharmaceutically acceptable salt includes the following steps:
  • the hydroxy protecting group R w is preferably TBS
  • R 2 , R 3 , R 5 , R 6 , m, n, p, Q, and z are as defined in the general formula (III).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (III-P), the method comprising:
  • the hydroxyl deprotection group R w is preferably TBS
  • R 2 , R 3 , R 5 , R 6 , m, n, p, Q, and z are as defined in the general formula (III).
  • Reagents that provide acidic conditions include, but are not limited to, hydrogen chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-toluenesulfonic acid, Me 3 SiCl, and TMSOT f ; preferably trifluoroacetic acid.
  • the reagents that provide alkaline conditions include organic bases and inorganic bases.
  • the organic bases include, but are not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, and lithium diisopropylamide.
  • the inorganic bases include but are not limited to sodium hydride, potassium phosphate, sodium carbonate, sodium acetate, potassium acetate, potassium carbonate or cesium carbonate, sodium hydroxide, hydrogen Lithium oxide and potassium hydroxide.
  • the deprotection reaction for removing the hydroxyl protecting group is well known in the art, and the hydroxyl protecting group is, for example, the protecting group described in Protecting Group in Organic Synthesis by T. Greene et al. It is generally preferred to use tetrahydropyran-2-yl and tert-butyldisilyl hydroxy protecting groups; tert-butyldimethylsilane (TBS) is preferred.
  • Hydroxyl protecting reagents include, but are not limited to: methoxymethyl ether, 2-methoxyethoxymethyl ether, tetrahydropyran ether, benzyl ether, p-methoxybenzyl ether, trimethylsilyl ether, trimethylsilyl ether Ethyl silyl ether, triisopropylsilyl ether, tert-butyldimethylsilyl ether, tert-butyldimethylsilyl chloride, triphenylmethylsilyl ether, acetate, Substituted acetate, pivaloate, benzoate, methanesulfonate and p-toluenesulfonate; preferably tert-butyldimethylchlorosilane (TBSCl).
  • TBSCl tert-butyldimethylchlorosilane
  • the above reaction is preferably carried out in a solvent.
  • the solvents used include but are not limited to: acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 ,4-Dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof; dichloromethane is preferred.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • NMR was measured with Bruker AVANCE-400 nuclear magnetic instrument, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), and the internal standard was four Methylsilane (TMS).
  • MS uses Agilent 1200/1290 DAD-6110/6120 Quadrupole MS LC/MS (manufacturer: Agilent, MS model: 6110/6120 Quadrupole MS). waters ACQuity UPLC-QD/SQD (manufacturer: waters, MS model: waters ACQuity Qda Detector/waters SQ Detector) THERMO Ultimate 3000-Q Exactive (manufacturer: THERMO, MS model: THERMO Q Exactive).
  • HPLC High performance liquid chromatography analysis uses Agilent HPLC 1200DAD, Agilent HPLC 1200VWD and Waters HPLC e2695-2489 high pressure liquid chromatograph.
  • HPLC preparation uses Waters 2545-2767, Waters 2767-SQ Detecor2, Shimadzu LC-20AP and Gilson GX-281 preparative chromatographs.
  • CombiFlash rapid preparation instrument uses Combiflash Rf200 (TELEDYNE ISCO).
  • the thin layer chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate, the size of the silica gel plate used in thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm, and the size of thin layer chromatography separation and purification products is 0.4mm ⁇ 0.5mm.
  • the silica gel column chromatography generally uses Yantai Huanghai silica gel 200-300 mesh silica gel as the carrier.
  • the known starting materials of the present disclosure can be synthesized by or according to methods known in the art, or can be purchased from ABCR GmbH & Co.KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Companies such as Darui Chemicals.
  • reaction can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • the argon atmosphere or nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon with a volume of about 1L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon with a volume of about 1L.
  • the pressure hydrogenation reaction uses Parr 3916EKX hydrogenator and Qinglan QL-500 hydrogen generator or HC2-SS hydrogenator.
  • the hydrogenation reaction is usually evacuated, filled with hydrogen, and repeated three times.
  • the microwave reaction uses the CEM Discover-S 908860 microwave reactor.
  • the solution refers to an aqueous solution.
  • reaction temperature is room temperature, which is 20°C to 30°C.
  • the monitoring of the reaction progress in the examples adopts thin-layer chromatography (TLC).
  • the developing reagent used in the reaction, the eluent system of column chromatography used in the purification of the compound and the developing reagent system of thin-layer chromatography include: A: Dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of triethylamine and Adjust with alkaline or acidic reagents such as acetic acid.
  • compound 1d (360mg, 0.75mmol) was dissolved in 50mL 1,4-dioxane, and 4,4,4',4',5,5,5',5'-octamethyl was added sequentially Group-2,2'-bis(1,3,2-dioxaborolane) (189mg, 0.76mmol), potassium acetate (219mg, 2.23mmol) and [1,1'-bis(diphenyl Phosphono)ferrocene]palladium dichloride (109g, 0.15mmol), stirred at 90°C for 2 hours. After cooling, filtering through celite, the filtrate was concentrated, and the residue was purified by column chromatography with eluent system C to obtain the title compound 1e (100 mg), yield: 25%.
  • N-(1-methyl-1H-pyrazol-5-yl)carboxamide 1j (270mg, 2.15mmol, using the well-known method "Bioorganic and Medicinal Chemistry, 1997, 5(3), 557-567" was prepared and Obtained), dissolved in N,N-dimethylformamide, added sodium hydride (60%, 250mg, 6.5mmol) at 0°C, stirred for 0.5 hours, added compound 1i (445mg, 2.15mmol), and continued the reaction 2. hour.
  • compound 6d (300mg, 0.61mmol) was dissolved in 50mL dioxane, and 4,4,4',4',5,5,5',5'-octamethyl-2 ,2'-bis(1,3,2-dioxaborolane) (254mg, 0.92mmol), potassium acetate (181mg, 1.84mmol) and [1,1'-bis(diphenylphosphino) Ferrocene] palladium dichloride (90 mg, 1.23 mmol), stirred at 90°C for 2 hours. After cooling, filtering through diatomaceous earth, the filtrate was concentrated, and the residue was purified by column chromatography with eluent system C to obtain the title compound 6e (120 mg), yield: 36%.
  • compound 1f 50mg, 0.22mmol
  • compound 6e 120mg, 0.22mmol
  • [1,1'-bis(diphenylphosphino)ferrocene] palladium dichloride 33mg, 0.04 mmol
  • cesium carbonate 146 mg, 0.45 mmol
  • compound 9g (220mg, 0.45mmol) was dissolved in 3mL 1,4-dioxane, and 4,4,4',4',5,5,5',5'-octamethyl Group-2,2'-bis(1,3,2-dioxaborolane) (173mg, 0.68mmol), potassium acetate (178mg, 1.81mmol) and [1,1'-bis(diphenyl Phosphono)ferrocene]palladium dichloride (33mg, 0.05mmol), stirred at 90°C for 4 hours. Cooled, filtered through Celite, and concentrated the filtrate to obtain the title crude compound 9h (240 mg). The product was directly used in the next reaction without purification.
  • compound 1f (100mg, 0.45mmol) was dissolved in 6mL dioxane, and 9h (241mg, 0.45mmol), cesium carbonate (291mg, 0.89mmol) and [1,1'-bis( Diphenylphosphino)ferrocene]palladium dichloride (65mg, 0.088mmol), stirred at 85°C in microwave for 1.5 hours. After cooling, filtering through diatomaceous earth, the filtrate was concentrated, and the residue was purified by column chromatography with eluent system A to obtain the title compound 9i (100 mg), yield: 37.7%.
  • compound 15d (7g, 15.5mmol) was dissolved in 100mL dioxane, and 4,4,4',4',5,5,5',5'-octamethyl-2 ,2'-bis(1,3,2-dioxaborolane) (5.9g, 23.3mmol), potassium acetate (3.1g, 31.1mmol) and [1,1'-bis(diphenylphosphine) Yl)ferrocene]palladium dichloride (2.3g, 2.1mmol), stirred at 90°C for 2 hours. After cooling, filtering through diatomaceous earth, the filtrate was concentrated, and the residue was purified by column chromatography with eluent system C to obtain the title compound 15e (4g), yield: 51.7%.
  • compound 2,4-dichloro-5-methylpyrimidine 17a (76.3 mg, 0.47 mmol, Shanghai Bi De Pharmaceutical Technology Co., Ltd.), compound 2e (220 mg, 0.43 mmol), [1,1' -Bis(diphenylphosphino)ferrocene]palladium dichloride (62.3mg, 0.08mmol), cesium carbonate (277.3mg, 0.85mmol) mixture is suspended in 10mL 1,4-dioxane and 2mL water, Heat to 80°C and stir to react for 14 hours.
  • compound 18c (100mg, 0.31mmol) was dissolved in 10mL dioxane, and 4,4,4',4',5,5,5',5'-octamethyl-2 ,2'-bis(1,3,2-dioxaborolane) (95mg, 0.37mmol), potassium acetate (60mg, 0.61mmol) and [1,1'-bis(diphenylphosphino) Ferrocene] Palladium dichloride (22mg, 30umol), stirred at 90°C for 2 hours. After cooling, filtering through Celite, the filtrate was concentrated, and the residue was purified by column chromatography with eluent system C to obtain the title compound 18d (28 mg), yield: 24.5%.
  • Test example 1 ERK1 enzyme activity test
  • Purpose of this experiment was to detect the ability of compounds to inhibit the activity of ERK1 and evaluated in vitro activity of compound 50 according to the size of the IC.
  • This experiment uses the ADP-Glo TM Kinase Assay Kit. Under the action of the enzyme, the substrate is phosphorylated and ADP is produced at the same time. The ADP-Glo reagent is added to remove the unreacted ATP in the reaction system. The kinase detection reagent ( Kinase detection reagent) detects the ADP produced by the reaction. In the presence of the compound, the inhibition rate of the compound is calculated by measuring the signal value.
  • Enzyme and substrate configuration ERK1 (1879-KS-010, R&D) and substrate (AS-61777, anaspec) are respectively configured in buffer (40mM Tris, 20mM MgCl2, 0.1mg/ml BSA, 50uM DTT) 0.75ng/ul and 100 ⁇ 0, then the enzyme solution and the substrate solution are prepared into a mixed solution at a volume ratio of 2:1, and set aside. Dilute ATP to 300uM with buffer, dissolve the compound with DMSO, prepare a stock solution with an initial concentration of 20mM, and then prepare the compound with Bravo (SGC120TH34702, Agilent Technologies).
  • buffer 40mM Tris, 20mM MgCl2, 0.1mg/ml BSA, 50uM DTT
  • each well of the 384-well plate add a 3 ⁇ plate of enzyme and substrate mixture solution, 1 and the bottom of different concentrations of compound (the initial concentration is 50uM, 4-fold dilution), incubate at 30°C for 10 minutes, and finally each well Add 1 ⁇ m of 300uM ATP solution and incubate at 30°C for 2 hours. Then add 5uL of ADP-Glo, incubate at 30°C for 40 minutes, then add 10uL of Kinase Detection buffer, and incubate at 30°C for 40 minutes. Take out the 384-well plate and place it in a microplate reader (BMG labtech, PHERAstar FS), and measure the chemiluminescence with the microplate reader.
  • a microplate reader BMG labtech, PHERAstar FS
  • Table 1 The IC 50 value of the compounds of the present disclosure on the inhibition of ERK1 enzyme activity
  • the compound of the present disclosure has a significant inhibitory effect on ERK1 enzyme activity.
  • Test example 2 ERK2 enzyme activity test
  • Purpose of this experiment was to detect the ability of compounds to inhibit the activity of ERK2 in vitro activity of compounds evaluated according to the size of the IC 50.
  • This experiment uses the ADP-Glo TM Kinase Assay Kit. Under the action of enzyme, the substrate is phosphorylated and ADP is produced at the same time. ADP-Glo reagent is added to remove unreacted ATP in the reaction system. Kinase detection reagent ( Kinase detection reagent) detects the ADP produced by the reaction. In the presence of the compound, the inhibition rate of the compound is calculated by measuring the signal value.
  • Enzyme and substrate configuration ERK2 (1230-KS-010, R&D) and substrate (custom peptide, Gil Biochemical) are configured in buffer (40mM Tris, 20mM MgCl 2 , 0.1mg/ml BSA, 50uM DTT) 0.75ng/ul and 1500ng, then the enzyme solution and the substrate solution are prepared into a mixed solution at a volume ratio of 2:1, and set aside. Dilute ATP to 500uM with buffer, dissolve the compound with DMSO, prepare a stock solution with an initial concentration of 20mM, and then prepare the compound with Bravo (SGC120TH34702, Agilent Technologies).
  • buffer 40mM Tris, 20mM MgCl 2 , 0.1mg/ml BSA, 50uM DTT
  • each well of the 384-well plate add 3 plates of each enzyme and substrate mixture solution, 1 ⁇ and different concentrations of compounds (the initial concentration is 50uM, 4-fold dilution), incubate at 30°C for 10 minutes, and finally each well Add 1 ⁇ m of 500uM ATP solution and incubate at 30°C for 2 hours. Then add 5uL of ADP-Glo and incubate at 30°C for 40 minutes, then add 10uL of Kinase detection buffer and incubate at 30°C for 40 minutes. Take out the 384-well plate and place it in a microplate reader (BMG labtech, PHERAstar FS), and measure the chemiluminescence with a microplate reader.
  • a microplate reader BMG labtech, PHERAstar FS
  • Table 2 The IC 50 value of the compounds of the present disclosure for inhibition of ERK2 enzyme activity
  • Test Example 3 Inhibition test of compound on Colo205 tumor cell proliferation in vitro
  • the purpose of this experiment is to test the compound's inhibitory activity on the proliferation of Colo205 cells (CCL-222, ATCC) in vitro.
  • the cells were treated in vitro with different concentrations of compounds, and after 3 days of culture, CTG ( Luminescent Cell Viability Assay, Promega, NO: G7573) added to the reagent cell is detected, the evaluation of the compound according to the in vitro activity of IC 50 values.
  • Digest Colo205 resuspend after centrifugation, mix the single cell suspension, adjust the density of viable cells to 5.0 ⁇ 10 4 cells/ml with cell culture medium (RPMI1640+2% FBS), add 95 ⁇ l/well to 96-well cell culture plate . Only add 100ul medium to the peripheral wells of the 96-well plate. The culture plate was cultured in an incubator for 24 hours (37°C, 5% CO 2 ).
  • the compound was dissolved in DMSO to prepare a stock solution with an initial concentration of 20 mM.
  • the initial concentration of the small molecule compound is 2mM, diluted 4 times, 9 points of dilution, and the 10th point is DMSO.
  • Take another 96-well plate add 90ul of cell culture medium (RPMI1640+2% FBS) to each well, then add 10ul of different concentrations of the test sample to each well, mix well, and then add 5 ⁇ of the same different concentration to the cell culture plate
  • the samples to be tested, each sample has two duplicate holes.
  • the culture plate was incubated in an incubator for 3 days (37°C, 5% CO 2 ).
  • Test Example 4 Mouse pharmacokinetic test of the compound of the present disclosure
  • mice Using mice as the test animals, the LC/MS/MS method was used to determine the concentration of the drug in plasma at different times after the mice were intragastrically administered the compounds of Example 3, Example 10, Example 15 and Example 20. To study the pharmacokinetic behavior of the compound of the present disclosure in mice and evaluate its pharmacokinetic characteristics.
  • Example 3 The compounds of Example 3, Example 10, Example 15 and Example 20.
  • mice were fasted overnight and then administered by gavage.
  • the dosage was 2mg/kg, and the dosage was 0.2ml/10g.
  • mice were given intragastrically, and 0.1ml of blood was collected at 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 11.0, 24.0 hours before and after the administration, placed in a heparinized test tube, and centrifuged at 3500 rpm for 10 After minutes, the plasma was separated and stored at -20°C.
  • the compound of the present disclosure has better pharmacokinetic absorption and has pharmacokinetic advantages.

Abstract

本申请提供了吡咯并杂环类衍生物、其制备方法及其在医药上的应用。具体而言,本申请提供了一种通式(I)所示的新的吡咯并杂环类衍生物、其制备方法及含有该衍生物的药物组合物以及其作为治疗剂,特别是作为ERK抑制剂的用途,其中通式(I)的各取代基与说明书中的定义相同。

Description

吡咯并杂环类衍生物、其制备方法及其在医药上的应用 技术领域
本公开属于医药领域,涉及一种吡咯并杂环类衍生物、其制备方法及其在医药上的应用。特别地,本公开涉及通式(I)所示的吡咯并杂环类衍生物、其制备方法及含有该衍生物的药物组合物,以及其作为ERK抑制剂治疗ERK介导的疾病及病症或抑制MAPK-ERK信号通路的用途。
背景技术
正常细胞的增殖、分化、代谢、凋亡受到体内细胞信号转导通路的严格调节。丝裂原活化蛋白激酶(mitogen-activated protein kinase,MAPK)在信号转导通路中起着极为重要的作用,胞外信号调节激酶(extracellular signal regulated kinase,ERK)是MAPK家族的一员。通过RAS-RAF-MEK-ERK步骤,外源的刺激信号被传递给ERK,活化后的ERK转移进入细胞核,调节转录因子活性,从而调节细胞的增殖分化凋亡等生物学功能,或者通过磷酸化胞浆中细胞骨架成分参与细胞形态的调节及细胞骨架的重新分布。
RAS和RAF基因突变造成MAPK-ERK信号通路的持续激活,促使细胞恶性转化、异常增殖,最终产生肿瘤(Roberts PJ等,Oncogene,2007,26(22),3291-3310)。MEK抑制剂跟B-RAF抑制剂联用可以进一步提高B-RAF抑制剂抑制肿瘤生长的效果,可以显著提高携带BRAFV600E和V600K突变的黑色素瘤病人的无病进展期和总体生存率(Frederick DT等,Clinical Cancer Research,2013.19(5),1225-1231)。虽然B-RAF/MEK抑制剂联用可以起到抑制肿瘤的效果,但是他们的疗效是短暂的,在2-18个月内绝大多数患者会产生耐药,肿瘤会进一步恶化。B-RAF/MEK抑制剂耐药性的产生机制非常复杂,大多与ERK信号通路的重新激活有着直接关系(Smalley I等,Cancer Discovery,2018,8(2),140-142)。所以,开发新的ERK抑制剂,不仅对MAPK信号通路产生突变的病人有效,对于B-RAF/MEK抑制剂产生耐药的病人也同样有效。
B-RAF/MEK抑制剂在抑制肿瘤生长的同时,对肿瘤的免疫微环境起到了调控作用。B-RAF/MEK抑制剂可以增强肿瘤特异性抗原的表达,提高抗原特异性T细胞对肿瘤的识别和杀伤,促进免疫细胞的迁移和浸润。动物模型中,经过B-RAF/MEK抑制剂处理后,肿瘤组织中PD-L1表达增强,与检查点(checkpoint)分子的抗体(例如PD-1抗体、CTLA4抗体)联用时,更显示出优于B-RAF/MEK抑制剂单用时的抑制肿瘤生长的效果(Boni A等,Cancer Research,2010,70(13),5213-5219)。研究表明,ERK抑制剂与B-RAF/MEK抑制剂类似,与检查点抗体联用可以起到调节肿瘤微环境的作用,提高细胞毒性T细胞的功能,达到抑制肿瘤生长的效果。
目前已有多个化合物被开发。其中BioMed Valley Discoveries公司的BVD-523在临床二期,默克公司的MK-8353以及Astex的Astex-029在临床一期。相关的专利有WO1999061440A1、WO2001056557A2、WO2001056993A2、WO2001057022A2、WO2002022601A1、WO2012118850A1、WO2013018733A1、WO2014179154A2、WO2015103133A1、WO2016192063A1、WO2017180817A1、WO2018049127A1。
发明内容
本公开的目的在于提供一种通式(I)所示的化合物:
Figure PCTCN2020081591-appb-000001
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中:
R 1选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、氨基烷基和硝基,所述的烷基任选地被NR 7R 8、烷氧基、卤素、氰基、硝基、羟基、羟烷基中的一个或多个取代基所取代;
R 2相同或不同,各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基和硝基;
R 3选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基,所述的烷基、环烷基、杂环基、芳基和杂芳基任选进一步被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R 4选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基和硝基;
R 5相同或不同,各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
R 6选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷 基、氰基、氨基和硝基;
R 7和R 8相同或不同,各自独立地选自氢原子、烷基、羟烷基和卤代烷基;
m选自0、1、2、3、4或5;
n选自0、1、2或3;
z选自0、1、2、3或4;
Q选自0、1或2。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 1选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、氨基烷基和硝基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,其为通式(I-P)所示的化合物:
Figure PCTCN2020081591-appb-000002
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中,R 1~R 6、m、n、z、Q如通式(I)中所定义。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 4为氢原子。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的n为1或2。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,其为通式(II)所示的化合物:
Figure PCTCN2020081591-appb-000003
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中,R 1~R 6、m、z、Q如通式(I)中所定义。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,其为通式(II-P)所示的化合物:
Figure PCTCN2020081591-appb-000004
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中,R 1~R 6、m、z、Q如通式(I)中所定义。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 1选自氢原子、烷基、羟基、氨基烷基、烷基氨基烷基和羟烷基;优选地,R 1选自氢原子、C 1-6烷基、羟基、氨基C 1-6烷基、C 1-6烷基氨基C 1-6烷基和C 1-6羟烷基;更优选地,R 1选自氢原子、甲基、羟甲基、氨基甲基和甲基氨基甲基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 1选自氢原子、烷基、羟基、氨基烷基和羟烷基;优选地,R 1选自氢原子、C 1-6烷基、羟基、氨基C 1-6烷基和C 1-6羟烷基;更优选地,R 1选自氢原子、甲基、羟甲基和氨基甲基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,其为通式(III)所示的化合物:
Figure PCTCN2020081591-appb-000005
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中:
p选自0、1、2或3,优选为1;
R 2、R 3、R 5、R 6、m、n、z、Q如通式(I)中所定义。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,其为通式(III-P)所示的化合物:
Figure PCTCN2020081591-appb-000006
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中:
R 2、R 3、R 5、R 6、m、n、z、p、Q如通式(III)中所定义。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 2选自氢原子、卤素和烷基;优选地,R 2选自氢原子、卤素和C 1-6烷基;更优选地,R 2为C 1-6烷基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 3为烷基、羟烷基、环烷基、杂环基和杂芳基,所述的烷基、环烷基、杂环基和杂芳基任选进一步被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和羟烷基中的一个或多个取代基所取代;优选地,R 3为C 1-6烷基、C 1-6羟烷基、C 3-6环烷基、3-8元杂环基和5-10元杂芳基, 所述的C 1-6烷基、C 3-6环烷基、3-8元杂环基和5-10元杂芳基任选进一步被选自C 1-6烷基、C 1-6烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和C 1-6羟烷基中的一个或多个取代基所取代。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 3为杂芳基,所述的杂芳基任选进一步被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和羟烷基中的一个或多个取代基所取代;优选地,R 3为5-10元杂芳基,所述的5-10元杂芳基任选进一步被选自C 1-6烷基、C 1-6烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和C 1-6羟烷基中的一个或多个取代基所取代;更优选地,R 3为吡唑基,所述的吡唑基任选地被C 1-6烷基,优选甲基取代。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 5选自氢原子、烷基、烷氧基和卤素;优选地,R 5选自氢原子、C 1-6烷基、C 1-6烷氧基和卤素。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 6为氢原子。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中m为1或2。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中z为0或1。
本公开的典型化合物包括但不限于:
Figure PCTCN2020081591-appb-000007
Figure PCTCN2020081591-appb-000008
Figure PCTCN2020081591-appb-000009
Figure PCTCN2020081591-appb-000010
Figure PCTCN2020081591-appb-000011
Figure PCTCN2020081591-appb-000012
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐。
本公开的另一方面涉及一种通式(IIIA)所示的化合物:
Figure PCTCN2020081591-appb-000013
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中:
R w为羟基保护基;
R 2、R 3、R 5、R 6、m、n、p、Q和z如通式(III)中所定义。
本公开的另一方面涉及一种通式(III-PA)所示的化合物:
Figure PCTCN2020081591-appb-000014
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中:
R w为羟基保护基;
R 2、R 3、R 5、R 6、m、n、p、Q和z如通式(III)中所定义。
本公开通式(IIIA)的典型化合物包括但不限于:
Figure PCTCN2020081591-appb-000015
Figure PCTCN2020081591-appb-000016
Figure PCTCN2020081591-appb-000017
Figure PCTCN2020081591-appb-000018
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐。
本公开的另一方面涉及一种制备通式(III)所示的化合物的方法,该方法包括以下步骤:
Figure PCTCN2020081591-appb-000019
通式(IIIA)化合物在酸性条件下,脱去羟基保护基R w,得到通式(III)的化合物,
其中:
所述的羟基保护基R w优选TBS;
R 2、R 3、R 5、R 6、m、n、p、Q和z如通式(III)中所定义。
本公开的另一方面涉及一种制备通式(III-P)所示的化合物的方法,该方法包括以下步骤:
Figure PCTCN2020081591-appb-000020
通式(III-PA)化合物在酸性条件下,脱去羟基保护基R w,得到通式(III-P)的化合物,
其中:
所述的羟基保护基R w优选TBS;
R 2、R 3、R 5、R 6、m、n、p、Q和z如通式(III)中所定义。
本公开的另一方面涉及一种药物组合物,所述药物组合物含有治疗有效量的本公开通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
本公开进一步涉及通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物,在制备用于抑制ERK的药物中的用途。
本公开进一步涉及通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或 其混合物形式或其可药用盐,或包含其的药物组合物在制备用于治疗或预防癌症、炎症、或其它增殖性疾病的药物中的用途,优选在制备用于治疗或预防癌症的药物中的用途;所述的癌症选自黑色素瘤、肝癌、肾癌、肺癌(如非小细胞肺癌或小细胞肺癌)、鼻咽癌、结肠直肠癌、胰腺癌、宫颈癌、卵巢癌、乳腺癌、膀胱癌、前列腺癌、白血病、头颈鳞状细胞癌、子宫颈癌、甲状腺癌、淋巴瘤、肉瘤、成神经细胞瘤、脑瘤、骨髓瘤(如多发性骨髓瘤)和星形细胞瘤、胶质瘤。
本公开还涉及一种抑制ERK的方法,其包括给予所需患者治疗有效量的通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物。
本公开还涉及一种治疗或预防ERK介导的疾病的方法,其包括给予所需患者治疗有效量的通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物。
本公开还涉及一种治疗或预防癌症、炎症、或其它增殖性疾病的方法,优选治疗癌症的方法,其包括给予所需患者治疗有效量的通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物;其中所述的癌症选自黑色素瘤、肝癌、肾癌、肺癌(如非小细胞肺癌或小细胞肺癌)、鼻咽癌、结肠直肠癌、胰腺癌、宫颈癌、卵巢癌、乳腺癌、膀胱癌、前列腺癌、白血病、头颈鳞状细胞癌、子宫颈癌、甲状腺癌、淋巴瘤、肉瘤、成神经细胞瘤、脑瘤、骨髓瘤(如多发性骨髓瘤)、星形细胞瘤和胶质瘤。
本公开进一步涉及一种通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物,其用作药物。
本公开还涉及通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物,其用作ERK抑制剂。
本公开还涉及通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物,其用于治疗或预防ERK介导的疾病。
本公开还涉及通式(I)或通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用盐,或包含其的药物组合物,其用于治疗或预防癌症、炎症、或其它增殖性疾病,优选用于治疗或预防癌症;其中所述的癌症选自黑色素瘤、 肝癌、肾癌、肺癌(如非小细胞肺癌或小细胞肺癌)、鼻咽癌、结肠直肠癌、胰腺癌、宫颈癌、卵巢癌、乳腺癌、膀胱癌、前列腺癌、白血病、头颈鳞状细胞癌、子宫颈癌、甲状腺癌、淋巴瘤、肉瘤、成神经细胞瘤、脑瘤、骨髓瘤(如多发性骨髓瘤),星形细胞瘤和胶质瘤。
可将活性化合物制成适合于通过任何适当途径给药的形式,活性化合物优选是以单位剂量的方式,或者是以患者可以以单剂自我给药的方式。本公开化合物或组合物的单位剂量的表达方式可以是片剂、胶囊、扁囊剂、瓶装药水、粉末剂、颗粒剂、锭剂、栓剂、再生粉末剂或液体制剂。
本公开治疗方法中所用化合物或组合物的剂量通常将随疾病的严重性、患者的体重和化合物的相对功效而改变。不过,作为一般性指导,合适的单位剂量可以是0.1~1000mg。
本公开的药物组合物除活性化合物外,可含有一种或多种辅料,所述辅料选自以下成分:填充剂(稀释剂)、粘合剂、润湿剂、崩解剂或赋形剂等。根据给药方法的不同,组合物可含有0.1至99重量%的活性化合物。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊、或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。
水混悬液含有活性物质和用于混合的适宜制备水混悬液的赋形剂。水混悬液也可以含有一种或多种防腐剂例如尼泊金乙酯或尼泊金正丙酯、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂。
油混悬液可通过使活性成分悬浮于植物油中配制而成。油混悬液可含有增稠剂。可加入上述的甜味剂和矫味剂,以提供可口的制剂。
通过加入水可使适用于制备水混悬液的可分散粉末和颗粒提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂可说明上述的例子。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
本公开的药物组合物也可以是水包油乳剂的形式。
药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳。例如将活性成分溶于大豆油和卵磷脂的混合物中。然后将油溶液加入水和甘油的混合物中处理形成微乳。可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本公开化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。这种装 置的实例是Deltec CADD-PLUS.TM.5400型静脉注射泵。
药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在肠胃外可接受的无毒稀释剂或溶剂中制备的无菌注射溶液或混悬液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。
可按用于直肠给药的栓剂形式给予本公开化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。此类物质包括可可脂、甘油明胶、氢化植物油、各种分子量的聚乙二醇和聚乙二醇的脂肪酸酯的混合物。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行为、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合等;另外,最佳的治疗方式如治疗的模式、通式化合物(I)的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的, 当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基和氧代基中的一个或多个取代基所取代。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至8个碳原子,最优选包含3至6个(例如3、4、5或6)碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等,优选环烷基;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2020081591-appb-000021
术语“稠环烷基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2020081591-appb-000022
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更优选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2020081591-appb-000023
所述环烷基环包括上述环烷基(例如单环、稠环、螺环和桥环环烷基)稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等;优选苯基并环戊基、四氢萘基。
环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基和氧代基中的一个或多个取代基所取代。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基或环烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧、硫、S(O)或S(O) 2的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;优选包含3至8个环原子,其中1~3个是杂原子;优选包含3至6个环原子,其中1~3个是杂原子。单环杂环基的非限制性实例包括氮杂环丁基、吡咯烷基、咪唑烷基、四氢呋喃基、四氢吡喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等,优选四氢吡喃基、哌啶基、吡咯烷基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧、硫、S(O)或S(O) 2,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至11元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2020081591-appb-000024
术语“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至11元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2020081591-appb-000025
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至11元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更优选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2020081591-appb-000026
所述杂环基环包括上述杂环基(例如单环、稠环、螺环和桥环杂环基)稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2020081591-appb-000027
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基和氧代基中的一个或多个取代基所取代。
术语“芳基”指具有共轭的π电子体系的6至20元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,更优选6元,例如苯基和萘基。所述芳基环包括上述芳基稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2020081591-appb-000028
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基和杂环烷硫基中的一个或多个取代基所取代。
术语“杂芳基”指包含1至4个杂原子、5至20个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至3个杂原子;非限制性实例如吡唑基、咪唑基、呋喃基、噻吩基、噻唑基、噁唑基、吡咯基、三唑基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2020081591-appb-000029
Figure PCTCN2020081591-appb-000030
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基和杂环烷硫基中的一个或多个取代基所取代。
术语“环烷基氧基”指-O-环烷基,其中环烷基如上所定义。
术语“卤代烷基”指被卤素取代的烷基,其中烷基如上所定义。
术语“卤代烷氧基”指被卤素取代的烷氧基,其中烷氧基如上所定义。
术语“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
术语“氨基烷基”指被氨基取代的烷基,其中烷基如上所定义。
术语“烷基氨基烷基”指被烷基氨基取代的烷基,其中烷基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
术语“醛基”指-C(O)H。
术语“羧基”指-C(O)OH。
术语“羧酸酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基、环烷基如上所定义。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代,其中每个取代基都有独立的选项(即取代基可以相同,也可以不同)。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本公开化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
本公开的化合物还可包含其同位素衍生物。术语“同位素衍生物”指结构不同仅在于存在一种或多种同位素富集原子的化合物。例如,具有本公开的结构,除了用“氘”或“氚”代替氢,或者用 18F-氟标记( 18F同位素)代替氟,或者用 11C-、 13C-、或者 14C-富集的碳( 11C-、 13C-、或者 14C-碳标记; 11C-、 13C-、或者 14C-同位素)代替碳原子的化合物处于本公开的范围内。这样的化合物可用作例如生物学测定中的分析工具或探针,或者可以用作疾病的体内诊断成像示踪剂,或者作为药效学、药动学或受体研究的示踪剂。氘代物通常可以保留与未氘代的化合物相当的活性,并且当氘代在某些特定位点时可以取得更好的代谢稳定性,从而获得某些治疗优势(如体内半衰期增加或剂量需求减少)。
针对药物或药理学活性剂而言,术语“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
本公开化合物的合成方法
为了完成本公开的目的,本公开采用如下技术方案:
方案一
本公开通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2020081591-appb-000031
通式(IIIA)化合物在酸性条件下,在溶剂中,脱去羟基保护基R w,得到通式(III)的化合物,
其中:
所述的羟基保护基R w优选TBS;
R 2、R 3、R 5、R 6、m、n、p、Q和z如通式(III)中所定义。
方案二
本公开的另一方面涉及一种制备通式(III-P)所示的化合物的方法,该方法包括:
Figure PCTCN2020081591-appb-000032
通式(III-PA)化合物在酸性条件下,在溶剂中,脱去羟基保护基R w,得到通式(III-P)的化合物,
其中:
所述的羟基脱保护基R w优选TBS;
R 2、R 3、R 5、R 6、m、n、p、Q和z如通式(III)中所定义。
提供酸性的条件的试剂包括但不限于氯化氢、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸、Me 3SiCl和TMSOT f;优选三氟乙酸。
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、醋酸钾、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、醋酸钠、醋酸钾、碳酸钾或碳酸铯、氢氧化钠、氢氧化锂和氢氧化钾。
脱除羟基保护基的脱保护反应是本领域公知的,所述羟基保护基是例如T.Greene等在Protecting Group in Organic Synthesis中所描述的保护基。通常优选使用四氢吡喃-2-基和叔丁基二甲硅烷基羟基保护基;优选叔丁基二甲基硅烷(TBS)。
羟基保护试剂包括但不限于:甲氧基甲醚、2-甲氧基乙氧基甲醚、四氢吡喃醚、苄醚、对甲氧基苄醚、三甲基甲硅烷基醚、三乙基甲硅烷基醚、三异丙基甲硅烷基醚、叔丁基二甲基甲硅烷基醚、叔丁基二甲基氯硅烷、三苯基甲基甲硅烷基醚、乙酸酯、取代的乙酸酯、新戊酸酯(pivaloate)、苯甲酸酯、甲基磺酸酯和对甲苯磺酸酯;优选叔丁基二甲基氯硅烷(TBSCl)。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、正丁醇、甲苯、四氢呋喃、二氯甲烷、石油醚、乙酸乙酯、正己烷、二甲基亚砜、1,4-二氧六环、乙二醇二甲醚、水或N,N-二甲基甲酰胺及其混合物;优选二氯甲烷。
在以上说明书中提出了本公开一种或多种实施方式的细节。虽然可使用与本文所述类似或相同的任何方法和材料来实施或测试本公开,但是以下描述优选的方法和材料。通过说明书和权利要求书,本公开的其他特点、目的和优点将是显而易见的。在说明书和权利要求书中,除非上下文中有清楚的另外指明,单数形式包括复数指代物的情况。除非另有定义,本文使用的所有技术和科学术语都具有本公开所属领域普通技术人员所理解的一般含义。说明书中引用的所有专利和 出版物都通过引用纳入。提出以下实施例是为了更全面地说明本公开的优选实施方式。这些实施例不应以任何方式理解为限制本公开的范围,本公开的范围由权利要求书限定。
具体实施方式
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10 -6(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6)、氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS)。
MS的测定用Agilent 1200/1290 DAD-6110/6120 Quadrupole MS液质联用仪(生产商:Agilent,MS型号:6110/6120 Quadrupole MS)。waters ACQuity UPLC-QD/SQD(生产商:waters,MS型号:waters ACQuity Qda Detector/waters SQ Detector)THERMO Ultimate 3000-Q Exactive(生产商:THERMO,MS型号:THERMO Q Exactive)。
高效液相色谱法(HPLC)分析使用Agilent HPLC 1200DAD、Agilent HPLC 1200VWD和Waters HPLC e2695-2489高压液相色谱仪。
手性HPLC分析测定使用Agilent 1260 DAD高效液相色谱仪。
高效液相制备使用Waters 2545-2767、Waters 2767-SQ Detecor2、Shimadzu LC-20AP和Gilson GX-281制备型色谱仪。
手性制备使用Shimadzu LC-20AP制备型色谱仪。
CombiFlash快速制备仪使用Combiflash Rf200(TELEDYNE ISCO)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
硅胶柱色谱法一般使用烟台黄海硅胶200~300目硅胶为载体。
激酶平均抑制率及IC 50值的测定用NovoStar酶标仪(德国BMG公司)。
本公开的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH & Co.KG、Acros Organics、Aldrich Chemical Company、韶远化学科技(Accela ChemBio Inc)、达瑞化学品等公司。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂,纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷/甲醇体系,B:正己烷/乙酸乙酯体系,C:石油醚/乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1
(S)-2-(1-(3-氟-5-甲氧基苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮1
Figure PCTCN2020081591-appb-000033
第一步
(S)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氟-5-甲氧基苯基)乙-1-胺1a
将(S)-2-氨基-2-(3-氟-5-甲氧基苯基)乙醇1k(2g,10.8mmol,上海皓鸿生物医药科技有限公司)、咪唑(1.47g,21.6mmol)溶于80mL二氯甲烷中,冰浴下加入叔丁基二甲基氯硅烷(TBSCl,2.44g,16.19mmol),搅拌反应14小时。加水,二氯甲烷萃取(80mL×3)。合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物1a(2.0g),产率:61.8%。
MS m/z(ESI):300.2[M+1]。
第二步
(S)-N-((4-溴-1H-吡咯-2-基)甲基)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氟-5-甲氧基苯基)乙胺1c
将(S)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氟-5-甲氧基苯基)乙-1-胺1a(516mg,1.72mmol)、化合物4-溴-1H-吡咯-2-甲醛1b(300mg,1.72mmol,上海毕得医药有限公司)搅拌反应3小时。加5mL甲醇稀释,降温至0℃,加入硼氢化钠(65mg,1.72mmol)搅拌反应2小时。加水,反应液减压浓缩。加水,乙酸乙酯萃取(10mL×2)。合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物1c(500mg),产率:63%。
MS m/z(ESI):457.1[M+1]。
第三步
(S)-6-溴-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氟-5-甲氧基苯基)乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮1d
将化合物1c(400mg,0.9mmol)溶于40mL四氢呋喃中,冰浴下加入N,N'-羰基二咪唑(219mg,1.36mmol)搅拌0.5小时,加入氢化钠(60%,69mg,1.8mmol),搅拌反应14小时。加饱和氯化铵。反应液减压浓缩,用柱层析色谱法以洗脱剂体系C纯化得到标题化合物1d(400mg),产率:94%。
MS m/z(ESI):483.2[M+1]。
第四步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氟-5-甲氧基苯基)乙基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H吡咯并[1,2-c]咪唑-3(2H)-酮1e
在氩气氛下,将化合物1d(360mg,0.75mmol)溶于50mL1,4-二氧六环中,依次加入4,4,4’,4’,5,5,5’,5’-八甲基-2,2’-二(1,3,2-二氧硼杂环戊烷)(189mg,0.76mmol)、乙酸钾(219mg,2.23mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(109g,0.15mmol),在90℃搅拌2小时。冷却,通过硅藻土过滤,将滤液浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物1e(100mg),产率:25%。
MS m/z(ESI):531.4[M+1]。
第五步
4-氯-5-甲基-2-(甲基磺酰基)嘧啶1i
将4-氯-5-甲基-2-(甲硫基)嘧啶1h(500mg,2.86mmol,上海毕得医药有限公司)溶于10mL的二氯甲烷中,加入间氯过氧苯甲酸(1.270g,6.3mmol),搅拌反应2小时。饱和硫代硫酸钠溶液洗涤,饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品标题化合物1i(445mg),产物不经纯化,直接用于下步反应。
MS m/z(ESI):207.2[M+1]。
第六步
4-氯-5-甲基-N-(1-甲基-1H-吡唑-5-基)嘧啶-2-胺1f
将N-(1-甲基-1H-吡唑-5-基)甲酰胺1j(270mg,2.15mmol,采用公知的方法“Bioorganic and Medicinal Chemistry,1997,5(3),557-567”制备而得),溶于N,N-二甲基甲酰胺中,于0℃加入氢化钠(60%,250mg,6.5mmol),搅拌反应0.5小时,加入化合物1i(445mg,2.15mmol),继续反应2小时。加水20mL,乙酸乙酯萃取(20mL×3),合并有机相减压浓缩,用薄层色谱法以展开剂体系C纯化所得残余物,得到标题化合物1f(240mg),产率:49.7%。
MS m/z(ESI):224.3[M+1]。
第七步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氟-5-甲氧基苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮1g
在氩气氛下,将化合物1e(100mg,0.19mmol)、化合物1f(42mg,0.19mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(28mg,0.04mmol)、碳酸铯(123mg,0.4mmol)混合物悬浮于30mL 1,4-二氧六环和4mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(20mL×3),合并有机相,减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物1g(80mg),产率:72%。
MS m/z(ESI):592.1[M+1]。
第八步
(S)-2-(1-(3-氟-5-甲氧基苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮1
将化合物1g(80mg,0.14mmol)溶于20mL二氯甲烷中,滴加5mL三氟乙酸,加毕,搅拌反应4小时。用饱和碳酸氢钠调pH至7,二氯甲烷萃取(20mL×2),合并有机相,减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物1(18mg),产率:28%。
MS m/z(ESI):478.3[M+1]。
1H NMR(400MHz,CD 3OD):δ8.21(s,1H),7.71(s,1H),7.43(d,1H),6.79-6.68(m,4H),6.32(d,1H),5.23-5.22(m,1H),4.64(d,1H),4.39-4.35(m,1H),4.18-4.08(m,2H),3.81(s,3H),3.75(s,3H),2.40(s,3H)。
实施例2
(S)-6-(5-氯-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-2-(1-(3-氯苯基)-2-羟乙基)-1H吡咯并[1,2-c]咪唑-3(2H)-酮2
Figure PCTCN2020081591-appb-000034
第一步
(S)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙胺2b
将(S)-2-氨基-2-(3-氯苯基)乙醇2a(4g,23.3mmol,上海毕得医药科技有限公司)/咪唑(3.2g,46.6mmol)溶于80mL二氯甲烷中,冰浴下加入叔丁基二甲基氯硅烷(5.2g,35mmol),搅拌反应14小时。加水,二氯甲烷萃取(80mL×3)。合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物2b(6.5g),产率:97%。
MS m/z(ESI):286.1[M+1]。
第二步
(S)-N-((4-溴-1H-吡咯-2-基)甲基)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙胺 2c
将化合物1b(2.37g,13.62mmol)、化合物2b(3.9g,13.64mmol)搅拌反应3小时。加100mL甲醇稀释,降温至0℃,加入硼氢化钠(516mg,13.64mmol)搅拌反应2小时。加水,反应液减压浓缩。加水,乙酸乙酯萃取(40mL×3)。合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物2c(4.8g),产率:79%。
MS m/z(ESI):444.2[M+1]。
第三步
(S)-6-溴-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮2d
将化合物2c(4.8g,10.81mmol)溶于100mL四氢呋喃中,冰浴下加入N,N'-羰基二咪唑(2.45g,15.11mmol)搅拌0.5小时,加入氢化钠(60%,621mg,16.22umol)室温搅拌反应14小时。加饱和氯化铵。反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物2d(4.0g),产率:78%。
MS m/z(ESI):469.1[M+1]。
第四步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮2e
在氩气氛下,将化合物2d(4.0g,8.51mmol)溶于50mL 1,4-二氧六环中,依次加入4,4,4’,4’,5,5,5’,5’-八甲基-2,2’-二(1,3,2-二氧硼杂环戊烷)(3.24g,12.76mmol)、乙酸钾(3.34g,34.04mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(1.24g,1.70mmol),在90℃搅拌2小时。冷却,通过硅藻土过滤,将滤液浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物2e(2.0g),产率:45%。
MS m/z(ESI):517.2[M+1]。
第五步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(2,5-二氯嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮2g
在氩气氛下,将化合物2e(430mg,0.83mmol)、2,4,5-三氯嘧啶2f(183mg,0.99mmol,上海毕得医药有限公司)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(60mg,0.08mmol)、碳酸钠(175mg,1.65mmol)混合物悬浮于2mL1,4-二氧六环和1mL水中,氩气置换,微波85℃搅拌反应1.5小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(10mL×3),合并有机相浓缩,残余物用薄层色谱法的展开剂体系C纯化得到标题化合物2g(200mg),产率:44%。
MS m/z(ESI):537.1[M+1]。
第六步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(5-氯-2-((1-甲基-1H-吡 唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮2i
在氩气氛下,将化合物2g(100mg,0.18mmol)溶于1mL 1,4-二氧六环,加入三(二亚苄基丙酮)二钯(25mg,27.30umol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(32mg,55.30umol)、碳酸铯(121mg,0.37mmol)、1-甲基-1H-吡唑-5-胺2h(36mg,0.37mmol,上海毕得医药有限公司),微波100℃搅拌反应1小时。冷却,通过硅藻土过滤,滤液浓缩,残余物用薄层色谱法的展开剂体系A纯化得到标题化合物2i(50mg),产率:44%。
MS m/z(ESI):597.9[M+1]。
第七步
(S)-6-(5-氯-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-2-(1-(3-氯苯基)-2-羟乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮2
将化合物2i(50mg,83.52umol)溶于3mL二氯甲烷中,滴加1mL三氟乙酸,加毕,搅拌反应3小时,用饱和碳酸氢钠调pH至7,二氯甲烷(10mL×2),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物2(16mg),产率:39%。
MS m/z(ESI):484.0[M+1]。
1H NMR(400MHz,CD 3OD):δ8.35(s,1H),8.08(s,1H),7.49-7.42(m,2H),7.36(m,3H),6.83(s,1H),6.36(d,1H),5.24(dd,1H),4.64(d,1H),4.34(d,1H),4.25-4.15(m,1H),4.12-4.04(m,1H),3.76(s,3H)。
实施例3
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮3
Figure PCTCN2020081591-appb-000035
第一步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮3a
在氩气氛下,将化合物1f(800mg,3.57mmol)、化合物2e(2.03g,3.93mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(392mg,0.54mmol)、碳酸铯(2.33g,7.15mmol)混合物悬浮于30mL 1,4-二氧六环和4mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(40mL×3),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物3a(1.2g),产率:58%。
MS m/z(ESI):578.3[M+1]。
第二步
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮3
将化合物3a(1.2g,2.07mmol)溶于20mL二氯甲烷中,滴加5mL三氟乙酸,加毕,搅拌反应4小时。用饱和碳酸氢钠调pH至7,二氯甲烷萃取(20mL×2),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物3(600mg),产率:62%。
MS m/z(ESI):464.2[M+1]。
1H NMR(400MHz,CD 3OD):δ8.21(s,1H),7.71(s,1H),7.46(s,2H),7.42-7.30(m,3H),6.75(s,1H),6.36(s,1H),5.26(dd,1H),4.66(d,1H),4.36(d,1H),4.26-4.16(m,1H),4.14-4.02(m,1H),3.76(s,3H),2.39(s,3H)。
实施例4
(S)-6-(5-氯-2-((四氢-2H-吡喃-4-基)氨基)嘧啶-4-基)-2-(1-(3-氯苯基)-2-羟乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮4
Figure PCTCN2020081591-appb-000036
采用实施例2中的合成路线,将第六步原料化合物2h替换为化合物四氢-2H-吡喃-4-胺(上海毕得医药有限公司),制得化合物5(12mg)。
MS m/z(ESI):488.0[M+1]。
1H NMR(400MHz,CDCl 3):δ8.24(s,1H),8.13(s,1H),7.44-7.33(m,3H), 7.27(s,1H),6.82(s,1H),5.17(dd,1H),5.10(d,1H),4.49(d,1H),4.38-4.18(m,3H),4.04(d,1H),4.02(d,1H),3.66-3.50(m,2H),2.09(d,2H),1.3-1.28(m,2H)。
实施例5
6-(5-氯-2-(((S)-1-羟基丙基-2-基)氨基)嘧啶-4-基)-2-((S)-1-(3-氯苯基)-2-羟乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮5
Figure PCTCN2020081591-appb-000037
第一步
2-((S)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(5-氯-2-(((S)-1-羟基丙基-2-基)氨基)嘧啶-4-基)-1,2-二氢-3H-吡咯并[1,2-c]咪唑-3-酮5b
将化合物2g(30mg,55.77umol)、(S)-2-氨基丙烷-1-醇5a(21mg,279.59umol,上海毕得医药有限公司)溶于1mL四氢呋喃,微波110℃搅拌反应1小时。反应液减压浓缩,得到粗品标题化合物5b(32mg),直接用于下一步反应。
MS m/z(ESI):576.2[M+1]。
第二步
6-(5-氯-2-(((S)-1-羟基丙基-2-基)氨基)嘧啶-4-基)-2-((S)-1-(3-氯苯基)-2-羟乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮5
将化合物5b(30mg,52.03umol)溶于3mL二氯甲烷中,滴加1mL三氟乙酸,加毕,搅拌反应3小时,用饱和碳酸氢钠调pH至7,二氯甲烷萃取(5mL×2),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物5(10.8mg),产率:44%。
MS m/z(ESI):462.0[M+1]。
1H NMR(400MHz,CD 3OD):δ8.24(s,1H),8.15(s,1H),7.46(s,1H),7.40-7.34(m,3H),6.93(s,1H),5.26(dd,1H),4.67(d,1H),4.37(d,1H),4.24-4.06(m,3H),3.67-3.56(m,2H),1.27(d,3H)。
实施例6
(S)-2-(1-(3-氯-4-氟苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮6
Figure PCTCN2020081591-appb-000038
第一步
(S)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯-4-氟苯基)乙胺6b
将(S)-2-氨基-2-(3-氯-4-氟苯基)乙-1-醇盐酸盐6a(250mg,1.1mmol,上海毕得医药科技有限公司)溶于10mL二氯甲烷中,加入咪唑(225.84mg,3.3174mmol),降温至0℃,加入叔丁基二甲基氯硅烷(250mg,1.7mmol),搅拌反应14小时。反应液中加入20mL水,二氯甲烷萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物6b(290mg),产率: 86%。
MS m/z(ESI):304.1[M+1]。
第二步
(S)-N-((4-溴-1H-吡咯-2-基)甲基)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯-4-氟苯基)乙胺6c
将化合物6b(290mg,0.95mmol)、化合物1b(166.05mg,0.95mmol)搅拌反应3小时。加5mL甲醇,降温至0℃,加入硼氢化钠(36mg,0.95mmol)搅拌反应2小时。加水,反应液减压浓缩,加水,乙酸乙酯萃取。合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物6c(300mg),产率:68%。
MS m/z(ESI):459.1[M-1]。
第三步
(S)-6-溴-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯-4-氟苯基)乙基)-1H-吡咯并[1,2-c]-3(2H)-酮6d
将化合物6c(300mg,0.65mmol)溶于40mL四氢呋喃中,冰浴下加入N,N'-羰基二咪唑(147mg,0.91mmol),搅拌反应0.5小时,加入氢化钠(60%,37mg,0.97mmol),室温搅拌反应14小时。加饱和氯化铵,反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物6d(300mg),产率:94%。
MS m/z(ESI):487.2[M+1]。
第四步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯-4-氟苯基)乙基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮6e
在氩气氛下,将化合物6d(300mg,0.61mmol)溶于50mL二氧六环中,依次加入4,4,4’,4’,5,5,5’,5’-八甲基-2,2’-二(1,3,2-二氧硼杂环戊烷)(254mg,0.92mmol)、乙酸钾(181mg,1.84mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(90mg,1.23mmol),在90℃搅拌2小时。冷却,通过硅藻土过滤,将滤液浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物6e(120mg),产率:36%。
MS m/z(ESI):534.1[M+1]。
第五步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯-4-氟苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1,2-二氢-3H-吡咯并[1,2-c]咪唑-3-酮6f
在氩气氛下,将化合物1f(50mg,0.22mmol)、化合物6e(120mg,0.22mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(33mg,0.04mmol)、碳酸铯(146mg,0.45mmol)混合物悬浮于30mL1,4-二氧六环和6mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(15mL×2),合并有机相减 压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物6f(100mg),产率:74%。
MS m/z(ESI):596.1[M+1]。
第六步
(S)-2-(1-(3-氯-4-氟苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮6
将化合物6f(100mg,0.17mmol)溶于3mL二氯甲烷中,滴加1mL三氟乙酸,加毕,搅拌反应4小时。用饱和碳酸氢钠溶液调pH至7,二氯甲烷萃取(15mL×2),合并有机相减压浓缩,残余物用薄层色谱法的展开剂体系A纯化得到标题化合物6(15mg),产率:18%。
MS m/z(ESI):482.2[M+1]。
1H NMR(400MHz,CD 3OD):δ8.19(s,1H),7.55(s,1H),7.52-7.44(m,2H),7.41(d,1H),7.22-7.13(m,1H),6.54(s,1H),6.14(d,1H),5.14(dd,1H),4.42(d,1H),4.32(dd,1H),4.23-4.12(m,2H),3.78(s,3H),2.33(s,3H)。
实施例7
6-(5-氯-2-((3-氟-4-羟基环戊基)氨基)嘧啶-4-基)-2-((S)-1-(3-氯苯基)-2-羟乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮7
Figure PCTCN2020081591-appb-000039
第一步
(3-氟-4-羟基环戊基)氨基甲酸苄酯7b
将(6-氧杂二环[3.1.0]己烷-3-基)氨基甲酸苄酯7a(1g,4.28mmol,采用公知方法“Tetrahedron,56(2000)9633-9640”制备而得)和氟化氢吡啶络合物(1.06g,6.41mmol,纯度60%)溶于1,2-二氯乙烷(5mL)中,搅拌4小时。反应液用饱和碳酸氢钠水溶液洗涤,饱和氯化钠洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余 物用薄层层析以展开剂体系C纯化得到化合物7b(500mg),产率:46%。
MS m/z(ESI):253.6[M+1]。
第二步
4-氨基-2-氟环戊醇7c
在氢气氛下,将化合物7b(150mg,592.25umol)和10%钯碳加氢催化剂(湿)(30mg,281.90umol)溶于5mL甲醇。反应搅拌16小时,过滤,滤液减压浓缩,得粗品化合物7c(150mg),直接用于下一步反应。
第三步
6-(5-氯-2-((3-氟-4-羟基环戊基)氨基)嘧啶-4-基)-2-((S)-1-(3-氯苯基)-2-羟乙基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮7
采用实施例2中合成路线,将第六步原料化合物2h替换为粗品化合物7c,制得化合物7(5mg)。
MS m/z(ESI):506.1[M+1]。
1H NMR(400MHz,CDCl 3):δ8.18(s,1H),8.07(d,1H),7.36(d,3H),7.27(d,1H),6.79(s,1H),5.62(d,1H),5.19(td,1H),4.96(dd,1H),4.50(d,2H),4.38-4.15(m,4H),2.60-2.48(m,1H),2.46-2.29(m,2H),1.85(d,2H)。
实施例8
6-(5-氯-2-(((R-1-羟基丙基-2-基)氨基)嘧啶-4-基)-2-((S)-1-(3-氯苯基)-2-羟乙基)-1,2-二氢-3H-吡咯并[1,2-c]咪唑-3-酮8
Figure PCTCN2020081591-appb-000040
采用实施例5中合成路线,将第一步原料化合物5a替换为(R)-2-氨基丙烷-1-醇,制得化合物8(17mg)。
MS m/z(ESI):462.0[M+1]。
1H NMR(400MHz,CD 3OD):δ8.23(s,1H),8.15(s,1H),7.45(s,1H),7.42-7.30(m,3H),6.92(s,1H),5.25(dd,1H),4.67(d,1H),4.36(d,1H),4.24-4.04(m,3H),3.62(t,2H),1.27(d,3H)。
实施例9
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-3,4-二氢吡咯并[1,2-c]嘧啶-1(2H)-酮9
Figure PCTCN2020081591-appb-000041
第一步
4-溴-2-(2-甲氧基乙烯基)-1-对甲苯磺酰基-1H-吡咯9c
将(甲氧基甲基)三苯基氯化鏻9b(8.36g,24.37mmol)溶于150mL四氢呋喃中,降温至0℃,加入叔丁醇钾(2.74g,24.37mmol),搅拌反应20分钟,加入4-溴-1-对甲苯磺酰基-1H-吡咯-2-甲醛9a(4.0g,612.19mmol,采用公知的方法“Journal of Porphyrins and Phthalocyanines,2009,13(10),1098-1110”制备而得),搅拌过夜。加水,反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物9c(4.0g),产率:92%。
MS m/z(ESI):356.0[M+1]。
第二步
2-(4-溴-1-对甲苯磺酰基-1H-吡咯-2-基)乙醛9d
将化合物9c(3g,8.42mmol)溶于10mL四氢呋喃中,加入10mL浓盐酸,搅拌反应3小时。加饱和碳酸氢钠调节pH至7。反应液乙酸乙酯萃取(50mL×2),有机相浓缩,得到标题粗品化合物9d(2.88g),产物不经纯化,直接用于下一步反应。
MS m/z(ESI):342.2[M+1]。
第三步
(S)-N-(2-(4-溴-1-对甲苯磺酰基-1H-吡咯-2-基)乙基)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙烷-1-胺9e
将粗品化合物9d(2.88g,8.42mmol)和化合物2b(2g,6.99mmol)溶于20mL甲醇,搅拌反应1小时,加入硼氢化钠(396mg,10.48mmol),室温搅拌反应14小时。加水淬灭,反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物9e(2.2g),产率:51.4%。
MS m/z(ESI):611.1[M+1]。
第四步
(S)-N-(2-(4-溴-1H-吡咯-2-基)乙基)-2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙烷-1-胺9f
将化合物9e(500mg,0.82mmol)溶于5mL四氢呋喃中,于0℃加入甲醇钠甲醇溶液(441mg,8.16mmol,50%),搅拌4小时。加2N盐酸调节pH至7,反应液乙酸乙酯萃取(10mL×2),有机相减压浓缩,残余物用薄层色谱法以展开剂体系C纯化得到标题化合物9f(280mg),产率:74.8%。
MS m/z(ESI):459.1[M+1]。
第五步
(S)-6-溴-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-3,4-二氢吡咯并[1,2-c]嘧啶-1(2H)-酮9g
将化合物9f(280mg,0.61mmol)溶于10mL四氢呋喃中,加入N,N'-羰基二咪唑(198mg,1.22mmol),搅拌30分钟,加入氢化钠(60%,47mg,1.23mmol),搅拌反应14小时。加水,反应液减压浓缩,用薄层色谱法的展开剂体系C纯化得到标题化合物9g(220mg),产率:74%。
MS m/z(ESI):483.1[M+1]。
第六步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-3,4-二氢吡咯并[1,2-c]嘧啶-1(2H)-酮9h
在氩气氛下,将化合物9g(220mg,0.45mmol)溶于3mL1,4-二氧六环中,依次加入4,4,4’,4’,5,5,5’,5’-八甲基-2,2’-二(1,3,2-二氧硼杂环戊烷)(173mg,0.68mmol)、醋酸钾(178mg,1.81mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(33mg,0.05mmol),在90℃搅拌4小时。冷却,通过硅藻土过滤,将滤液浓缩,得到标题粗品化合物9h(240mg),产物不经纯化,直接用于下一步反应。
MS m/z(ESI):530.9[M+1]。
第七步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H- 吡唑-5-基)氨基)嘧啶-4-基)-3,4-二氢吡咯并[1,2-c]嘧啶-1(2H)-酮9i
在氩气氛下,将化合物1f(100mg,0.45mmol)溶于6mL二氧六环中,依次加入9h(241mg,0.45mmol)、碳酸铯(291mg,0.89mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(65mg,0.088mmol),微波85℃搅拌1.5小时。冷却,通过硅藻土过滤,将滤液浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物9i(100mg),产率:37.7%。
MS m/z(ESI):592.2[M+1]。
第八步
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-3,4-二氢吡咯并[1,2-c]嘧啶-1(2H)-酮9
将化合物9i(100mg,0.17mmol)溶于3mL二氯甲烷中,滴加1mL三氟乙酸,加毕,搅拌反应4小时。用饱和碳酸氢钠调pH至7,二氯甲烷萃取(50mL×2),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物9(22mg),产率:27%。
MS m/z(ESI):478.2[M+1]。
1H NMR(400MHz,CD 3OD):δ8.18(s,1H),7.88(s,1H),7.46-7.43(m,2H),7.39-7.34(m,3H),6.62(s,1H),6.31(s,1H),5.72-5.69(m,1H),4.15-4.14(d,2H),3.74(s,3H),3.66-3.60(m,1H),3.35(m,1H),3.04-3.00(m,1H),2.99-2.88(m,1H),2.38(s,3H)。
实施例10
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1,2-二氢-3H-吡咯并[1,2-c]咪唑-3-酮10
Figure PCTCN2020081591-appb-000042
第一步
4-氯-N-(1-甲基-1H-吡唑-5-基)嘧啶-2-胺10b
将N-(1-甲基-1H-吡唑-5-基)甲酰胺1j(324.82mg,2.60mmol)溶于15mL N,N-二甲基甲酰胺中,于0℃加入氢化钠(60%,311.47mg,7.79mmol),搅拌反应0.5小时,加入化合物10a(500mg,2.60mmol),继续反应2小时。加水20mL,乙酸乙酯萃取(20mL×3),合并有机相减压浓缩,残余物用薄层色谱法以展开剂体系C纯化所得残余物,得到标题化合物10b(270mg),产率:49.6%。
MS m/z(ESI):210.3[M+1]。
第二步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1,2-二氢-3H-吡咯并[1,2-c]咪唑-3(2H)-酮10c
在氩气氛下,将化合物2e(98.6mg,0.19mmol)、4-氯-N-(1-甲基-1H-吡唑-5-基)嘧啶-2-胺10b、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(28mg,0.02mmol)、碳酸铯(124mg,0.2mmol)混合物悬浮于20mL 1,4-二氧六环和4mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(20mL×3),合并有机相,减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物10c(100mg),产率:92%。
MS m/z(ESI):564.3[M+1]。
第三步
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1,2-二氢-3H-吡咯并[1,2-c]咪唑-3-酮10
将化合物10c(100mg,0.17mmol)溶于20mL二氯甲烷中,滴加1mL三氟乙酸,加毕,搅拌反应4小时。用饱和碳酸氢钠调pH至7,二氯甲烷萃取(20mL×2),合并有机相,减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物10(15mg),产率:18%。
MS m/z(ESI):450.1[M+1]。
1H NMR(400MHz,CDCl 3):δ8.33(d,1H),7.72(s,1H),7.48(d,1H),7.41-7.33(m,3H),7.28-7.24(m,1H),7.18(s,1H),6.92(d,1H),6.51(s,1H),6.32(d,1H),5.17(dd,1H),4.46(d,1H),4.32(dd,1H),4.27-4.17(m,3H),3.82(s,3H)。
实施例11、12、13
(S)-2-(2-氨基-1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮11
(R)-2-(1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮12
(S)-2-(1-(3-氯苯基)-2-(甲氨基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶 -4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮13
Figure PCTCN2020081591-appb-000043
第一步
(S)-2-(3-氯苯基)-2-(6-(5-甲基-2-(1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-3-氧代-1H-吡咯并[1,2-c]咪唑-2(3H)-基)乙基)甲磺酸酯11a
将化合物3(12.0mg,0.026mmol)溶于10mL二氯甲烷中,依次加入三乙胺(8.0mg,0.079mmol)、甲磺酰氯(6.0mg,0.052mmol),搅拌反应30分钟。加少量水,减压浓缩得到标题化合物11a(14mg),产率:99%。
MS m/z(ESI):542.0[M+1]。
第二步
(S)-2-(2-叠氮基-1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮11b
将化合物11a(14mg,0.026mmol)溶于7mL N,N-二甲基甲酰胺中,加入叠氮钠(8.4mg,0.130mmol),加毕,70℃搅拌反应3小时。反应液中加水,二氯甲烷萃取(10mL×2),合并有机相,饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到化合物11b(12mg),产率:95%。
MS m/z(ESI):489.0[M+1]。
第三步
(S)-2-(2-氨基-1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮11
(R)-2-(1-(3-氯苯基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮12
(S)-2-(1-(3-氯苯基)-2-(甲氨基)乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮13
将化合物11b(17mg,0.035mmol)溶于5mL甲醇中,加入钯碳(42mg,0.35mmol,10%钯),氢气置换三次,搅拌反应30分钟。过滤,减压浓缩,高效液相 制备纯化得到化合物11(1.0mg),产率:6%,化合物12(1.0mg),产率:6%,化合物13(1.0mg),产率:6%。
化合物11
MS m/z(ESI):463.2[M+1]。
1H NMR(400MHz,CD 3OD):δ8.23(s,1H),7.75(s,1H),7.60(s,1H),7.52(s,1H),7.47-7.42(m,3H),6.74(s,1H),6.33(s,1H),5.37-5.34(dd,1H),4.56-4.52(d,1H),4.36(d,1H),4.26-4.16(m,1H),4.14-4.02(m,1H),3.76(s,3H),2.39(s,3H)。
化合物12
MS m/z(ESI):448.1[M+1]。
1H NMR(400MHz,CD 3OD):δ8.23(s,1H),7.74(s,1H),7.60(s,1H),7.53(s,1H),7.45-7.34(m,3H),6.75(s,1H),6.42(s,1H),5.45-5.40(dd,1H),4.58-4.54(d,1H),4.20-4.16(d,1H),3.77(s,3H),2.41(s,3H),1.74-1.72(d,3H)。
化合物13
MS m/z(ESI):477.2[M+1]。
1H NMR(400MHz,CDCl 3)δ8.15(s,1H),7.93(s,1H),7.58(s,1H),7.46(s,1H),7.42-7.27(m,4H),6.61(s,1H),6.28(s,1H),5.39(d,1H),4.41(d,1H),4.21(d,1H),3.81(s,3H),3.68-3.52(m,2H),2.35(s,3H),2.07-2.01(m,3H)。
实施例14
(S)-2-(1-(3-氟-4-氯苯基)-2-羟乙基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮14
Figure PCTCN2020081591-appb-000044
采用实施例6中合成路线,将第一步原料化合物6a替换为(S)-2-氨基-2-(3-氟-4-氯苯基)乙-1-醇,制得化合物14(15mg)。
MS m/z(ESI):482.2[M+1]。
1H NMR(400MHz,CDCl 3)δ8.19(s,1H),7.61-7.48(m,2H),7.47-7.36(m,2H),7.22(d,1H),7.13(d,1H),6.56(s,1H),6.15(s,1H),5.15(dd,1H),4.42(d,1H),4.32(dd,1H),4.26-4.13(m,2H),3.78(s,3H),2.33(s,3H)。
实施例15
(S)-2-(2-羟基-1-(间甲苯基)乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮15
Figure PCTCN2020081591-appb-000045
第一步
(S)-2-((叔丁基二甲基硅烷基)氧基)-1-(间甲苯基)乙胺15b
将(S)-2-氨基-2-(间甲苯基)乙-1-醇15a(3g,19.8mmol,上海毕得医药科技有限公司)溶于100mL二氯甲烷中,加入咪唑(4g,58.7mol),降温至0℃,加入叔丁基二甲基氯硅烷(3.9g,25.9mmol),搅拌反应14小时。反应液中加入100mL水,二氯甲烷萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物15b(5g),产率:95%。
MS m/z(ESI):266.2[M+1]。
第二步
(S)-N-((4-溴-1H-吡咯-2-基)甲基)-2-((叔丁基二甲基硅烷基)氧基)-1-(间甲苯基)乙胺15c
将化合物15b(5g,19.5mmol)、化合物1b(3.4g,19.5mmol)搅拌反应3小时。加50mL甲醇,降温至0℃,加入硼氢化钠(800mg,21.1mmol)搅拌反应2小时。加水,反应液减压浓缩,加水,乙酸乙酯萃取。合并有机相,用饱和氯化钠溶液 洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物15c(7.3g),产率:88%。
MS m/z(ESI):423.1[M+1]。
第三步
(S)-6-溴-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(间甲苯基)乙基)-1H-吡咯并[1,2-c]-3(2H)-酮15d
将化合物15c(7.3g,17.2mmol)溶于150mL四氢呋喃中,冰浴下加入N,N'-羰基二咪唑(8.4g,51.7mmol),搅拌反应0.5小时,加入氢化钠(60%,2g,51.7mmol),室温搅拌反应14小时。加饱和氯化铵,反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物15d(7g),产率:90%。
MS m/z(ESI):450.1[M+1]。
第四步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(间甲苯基)乙基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮15e
在氩气氛下,将化合物15d(7g,15.5mmol)溶于100mL二氧六环中,依次加入4,4,4’,4’,5,5,5’,5’-八甲基-2,2’-二(1,3,2-二氧硼杂环戊烷)(5.9g,23.3mmol)、乙酸钾(3.1g,31.1mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(2.3g,2.1mmol),在90℃搅拌2小时。冷却,通过硅藻土过滤,将滤液浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物15e(4g),产率:51.7%。
MS m/z(ESI):497.2[M+1]。
第五步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(间甲苯基)乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮15f
在氩气氛下,将化合物10b(550mg,2.62mmol)、化合物15e(1.56g,3.15mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(1.7g,0.26mmol)、碳酸铯(1.7g,5.2mmol)混合物悬浮于30mL1,4-二氧六环和6mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(15mL×2),合并有机相减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物15f(1.4g),产率:98%。
MS m/z(ESI):544.2[M+1]。
第六步
(S)-2-(2-羟基-1-(间甲基苯基)乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮15
将化合物15f(1.3g,2.4mmol)溶于10mL二氯甲烷中,滴加3mL三氟乙酸,加毕,搅拌反应4小时。用饱和碳酸氢钠溶液调pH至7,二氯甲烷萃取(30mL×2),合并有机相减压浓缩,用薄层色谱法的展开剂体系A纯化得到标题化合物15(500mg),产率:48.6%。
MS m/z(ESI):430.1[M+1]。
1H NMR(400MHz,CD 3OD)δ8.29(d,1H),7.80(s,1H),7.43(d,1H),7.32-7.24(m,1H),7.21(s,1H),7.19-7.12(m,2H),7.10(d,1H),6.63(s,1H),6.31(d,1H),5.23(dd,1H),4.61(d,1H),4.29(d,1H),4.19(dd,1H),4.11-3.99(m,1H),3.74(s,3H),2.34(s,3H)。
实施例16
(S)-2-(1-(3-氯-4-氟苯基)-2-羟乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮16
Figure PCTCN2020081591-appb-000046
采用实施例6中合成路线,将第五步原料化合物1f替换为10b,制得化合物16(20mg)。
MS m/z(ESI):468.0[M+1]。
1H NMR(400MHz,CDCl 3)δ8.31(d,1H),7.69(s,1H),7.51-7.42(m,2H),7.33(s,1H),7.26-7.22(m,1H),7.21-7.13(m,1H),6.90(d,1H),6.49(s,1H),6.30(s,1H),5.14(dd,1H),4.45(d,1H),4.35-4.24(m,1H),4.24-4.13(m,2H),3.80(s,3H)。
实施例17
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(2-(异丙基氨基)-5-甲基嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮17
Figure PCTCN2020081591-appb-000047
Figure PCTCN2020081591-appb-000048
第一步
(S)-2-(2-((叔丁基二甲基硅烷基)氧基)-1-(3-氯苯基)乙基)-6-(2-氯-5-甲基嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮17b
在氩气氛下,将化合物2,4-二氯-5-甲基嘧啶17a(76.3mg,0.47mmol,上海毕得医药科技有限公司)、化合物2e(220mg,0.43mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(62.3mg,0.08mmol)、碳酸铯(277.3mg,0.85mmol)混合物悬浮于10mL 1,4-二氧六环和2mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(10mL×3),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物17b(100mg),产率:45.4%。
MS m/z(ESI):517.1[M+1]。
第二步
(S)-2-(2-((叔丁基二甲基硅)氧基)-1-(3-氯苯基)乙基)-6-(2-(异丙基氨基)-5-甲基嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮17d
将化合物17b(80mg,154.5umol)、异丙基胺17c(91.4mg,1.5mmol,上海毕得医药有限公司)溶于2mL N,N-二甲基乙酰胺,微波150℃搅拌反应6小时。反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物17d(20mg),产率:24%。
第三步
(S)-2-(1-(3-氯苯基)-2-羟乙基)-6-(2-(异丙基氨基)-5-甲基嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮17
将化合物17d(20mg,37umol)溶于5mL二氯甲烷中,滴加0.5mL三氟乙酸,加毕,搅拌反应1小时。用饱和碳酸氢钠调pH至7,二氯甲烷萃取(20mL×2),合并有机相浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物17(5mg),产率:32%。
MS m/z(ESI):426.2[M+1]。
1H NMR(400MHz,CDCl 3)δ8.08(s,1H),7.61(s,1H),7.34(d,3H),7.25(d,2H),6.70(s,1H),5.13(dd,1H),4.45(d,1H),4.36-4.25(m,2H),4.23-4.14(m,2H),2.30(s,3H),1.26(d,6H)。
实施例18
2-(3,4-二氟苄基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并 [1,2-c]咪唑-3(2H)-酮18
Figure PCTCN2020081591-appb-000049
第一步
1-(4-溴-1H-吡咯-2-基)-N-(3,4-二氟苄基)甲酰胺18b
将化合物(3,4-二氟苯基)甲胺18a(863mg,6.0mmol)、化合物1b(1g,5.7mmol)搅拌反应3小时。加20mL甲醇,降温至0℃,加入硼氢化钠(361mg,5.7vmmol)搅拌反应2小时。加水,反应液减压浓缩,加水,乙酸乙酯萃取。合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物18b(1.7g),产率:98.2%。
MS m/z(ESI):301.0[M+1]。
第二步
6-溴-2-(3,4-二氟苄基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮18c
将化合物18b(0.5g,1.7mmol)溶于50mL四氢呋喃中,冰浴下加入N,N'-羰基二咪唑(286mg,2.0mmol),搅拌反应0.5小时,加入氢化钠(60%,15mg,0.63mmol),室温搅拌反应14小时。加饱和氯化铵溶液,反应液减压浓缩,残余物用柱层析色谱法以洗脱剂体系C纯化得到标题化合物18c(220mg),产率:40.5%。
MS m/z(ESI):327.0[M+1]。
第三步
2-(3,4-二氟苄基)-6-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮18d
在氩气氛下,将化合物18c(100mg,0.31mmol)溶于10mL二氧六环中,依次加入4,4,4’,4’,5,5,5’,5’-八甲基-2,2’-二(1,3,2-二氧硼杂环戊烷)(95mg,0.37mmol)、乙酸钾(60mg,0.61mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(22mg,30umol),在90℃搅拌2小时。冷却,通过硅藻土过滤,将滤液浓缩,残余物用柱层析色谱 法以洗脱剂体系C纯化得到标题化合物18d(28mg),产率:24.5%。
MS m/z(ESI):375.0[M+1]。
第四步
2-(3,4-二氟苄基)-6-(5-甲基-2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮18
在氩气氛下,将化合物18d(27mg,72umol)、化合物1f(15mg,67umol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(6mg,8.2umol)、碳酸钾(18mg,130umol)混合物悬浮于5mL 1,4-二氧六环和1mL水中,加热至80℃搅拌反应14小时。冷却,通过硅藻土过滤,收集滤液,乙酸乙酯萃取(10mL×2),合并有机相减压浓缩,残余物用柱层析色谱法以洗脱剂体系A纯化得到标题化合物18(10mg),产率:34%。
MS m/z(ESI):435.9[M+1]。
1H NMR(400MHz,CD 3OD)δ8.23(s,1H),7.80(s,1H),7.63-7.54(m,1H),7.36-7.22(m,2H),7.19(s,1H),6.75(s,1H),6.48(d,1H),4.69(s,2H),4.41(s,2H),3.80(s,3H),2.42(s,3H)。
实施例19
(S)-2-(1-(3-氟苯基)-2-羟乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮19
Figure PCTCN2020081591-appb-000050
采用实施例15中合成路线,将第一步原料化合物(S)-2-氨基-2-(3-甲基苯基)乙-1-醇15a替换为(S)-2-氨基-2-(3-氟苯基)乙-1-醇,制得化合物19(50mg)。
MS m/z(ESI):434.1[M+1]。
1H NMR(400MHz,CD 3OD)δ8.29(d,1H),7.81(s,1H),7.51-7.34(m,2H),7.28-7.15(m,2H),7.14-6.99(m,2H),6.65(s,1H),6.32(d,1H),5.26(dd,1H),4.64(d,1H),4.34(d,1H),4.26-4.14(m,1H),4.13-4.03(m,1H),3.75(s,3H)。
实施例20
(S)-2-(1-(3-氟-4-氯苯基)-2-羟乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮20
Figure PCTCN2020081591-appb-000051
采用实施例14中合成路线,将第五步原料化合物1f替换为10b,制得化合物20(50mg)。
MS m/z(ESI):468.1[M+1]。
1H NMR(400MHz,CD 3OD)δ8.29(d,1H),7.81(s,1H),7.49(t,1H),7.44(d,1H),7.35(dd,1H),7.22(d,1H),7.10(d,1H),6.65(s,1H),6.32(d,1H),5.24(dd,1H),4.64(d,1H),4.36(d,1H),4.21-4.13(m,1H),4.11-4.03(m,1H),3.75(s,3H)。
实施例21
(S)-2-(1-(4-氯苯基)-2-羟乙基)-6-(2-((1-甲基-1H-吡唑-5-基)氨基)嘧啶-4-基)-1H-吡咯并[1,2-c]咪唑-3(2H)-酮21
Figure PCTCN2020081591-appb-000052
采用实施例15中合成路线,将第一步原料化合物15a替换为(S)-2-氨基-2-(4-氯苯基)乙-1-醇,制得化合物21(10mg)。
MS m/z(ESI):450.1[M+1]。
1H NMR(400MHz,CDCl 3)δ8.33(d,1H),7.75(s,1H),7.48(d,1H),7.42-7.35(m,2H),7.31(d,2H),6.94(d,1H),6.54(s,1H),6.35(s,1H),5.14(dd,1H),4.44(d,1H),4.30(dd,1H),4.25-4.14(m,2H),3.82(s,3H)。
生物学评价
测试例1:ERK1酶学活性测试
一、测试目的
本实验的目的是为了检测化合物对ERK1酶活性的抑制能力,根据IC 50大小评价化合物的体外活性。本实验使用ADP-Glo TM激酶测试试剂盒(Kinase Assay Kit),在酶的作用下,底物被磷酸化同时产生ADP,加入ADP-Glo试剂除去反应体系中 未反应的ATP,激酶检测试剂(Kinase detection reagent)检测反应产生的ADP。在化合物存在的条件下,通过测量信号值算出化合物的抑制率。
二、实验方法
酶和底物配置:将ERK1(1879-KS-010,R&D)和底物(AS-61777,anaspec)在缓冲液(40mM Tris,20mM MgCl2,0.1mg/ml BSA,50uM DTT)中分别配置成0.75ng/ul和100μ0,然后将酶溶液和底物溶液以2:1的体积比配制成混合溶液,待用。用缓冲液将ATP稀释至300uM,用DMSO溶解化合物,配制成初始浓度为20mM的存储液,然后用Bravo(SGC120TH34702,Agilent Technologies)配制化合物。最后在384孔板每孔中依次加入3μ板酶和底物的混合液溶液,1和底不同浓度的化合物(起始浓度为50uM,4倍稀释),30℃孵育10分钟,最后每孔再加入1μ钟300uM的ATP溶液,在30℃孵育2小时。然后加入5uL的ADP-Glo,30℃孵育40分钟,接着加入10uL的激酶检测缓冲液(Kinase detection buffer),30℃孵育40分钟。取出384孔板,置于酶标仪(BMG labtech,PHERAstar FS)中,用酶标仪测定化学发光。
三、数据分析
用Microsoft Excel,Graphpad Prism 5对数据进行处理分析。得出化合物的IC 50值,结果见下表1。
表1 本公开化合物对ERK1酶活性抑制的IC 50
实施例编号 IC 50(nM)
1 9
3 3
5 10
6 14
9 12
10 5
13 2
15 5
16 70
19 24
20 30
21 67
结论:本公开化合物对ERK1酶活性有明显的抑制作用。
测试例2:ERK2酶学活性测试
一、测试目的
本实验的目的是为了检测化合物对ERK2酶活性的抑制能力,根据IC 50大小评价化合物的体外活性。本实验使用ADP-Glo TM激酶测试试剂盒(Kinase Assay Kit), 在酶的作用下,底物被磷酸化同时产生ADP,加入ADP-Glo试剂除去反应体系中未反应的ATP,激酶检测试剂(Kinase detection reagent)检测反应产生的ADP。在化合物存在的条件下,通过测量信号值算出化合物的抑制率。
二、实验方法
酶和底物配置:将ERK2(1230-KS-010,R&D)和底物(定制多肽,吉尔生化)在缓冲液(40mM Tris,20mM MgCl 2,0.1mg/ml BSA,50uM DTT)中配置成0.75ng/ul和1500ng,然后将酶溶液和底物溶液以2:1的体积比配制成混合溶液,待用。用缓冲液将ATP稀释至500uM,用DMSO溶解化合物,配制成初始浓度为20mM的存储液,然后用Bravo(SGC120TH34702,Agilent Technologies)配制化合物。最后在384孔板每孔中依次加入3板每酶和底物的混合液溶液,1μ和不同浓度的化合物(起始浓度为50uM,4倍稀释),30℃孵育10分钟,最后每孔再加入1μ钟500uM的ATP溶液,在30℃孵育2小时。然后加入5uL的ADP-Glo,30℃孵育40分钟,接着加入10uL的激酶检测缓冲液(Kinase detection buffer),30℃孵育40分钟。取出384孔板,置于酶标仪(BMG labtech,PHERAstar FS)中,用酶标仪测定化学发光。
三、数据分析
用Microsoft Excel,Graphpad Prism 5对数据进行处理分析。得出化合物的IC 50值,结果见下表2。
表2 本公开化合物对ERK2酶活性抑制的IC 50
实施例编号 IC 50(nM)
1 2
2 3
3 2
4 6
5 7
6 10
7 63
8 39
9 6
10 7
11 18
12 45
13 2
14 5
15 5
16 44
18 16
19 20
20 18
21 45
结论:本公开化合物对ERK2酶活性有明显的抑制作用。
测试例3:化合物对Colo205肿瘤细胞体外增殖抑制测试
一、测试目的
本实验的目的是为了检测化合物对Colo205细胞(CCL-222,ATCC)体外增殖的抑制活性。以不同浓度的化合物体外处理细胞,经3天培养后,采用CTG(
Figure PCTCN2020081591-appb-000053
Luminescent Cell Viability Assay,Promega,货号:G7573)试剂对细胞的增值进行检测,根据IC 50值评价该化合物的体外活性。
二、实验方法
下面以对Colo205细胞体外增殖抑制测试方法为例,说明本公开中测试本公开化合物对肿瘤细胞体外增殖抑制活性测试的方法。本方法同样适用于,但不限于对其它肿瘤细胞进行体外增殖抑制活性测试。
将Colo205消化,离心后重悬,单细胞悬液混匀,用细胞培养液(RPMI1640+2%FBS)调整活细胞密度至5.0×10 4cells/ml,以95μl/孔加入96孔细胞培养板。96孔板外周孔只加入100ul培养基。将培养板在培养箱培养24小时(37℃,5%CO 2)。
用DMSO溶解化合物,配制成初始浓度为20mM的存储液。小分子化合物的起始浓度为2mM,4倍稀释,稀释9个点,第10个点为DMSO。另取一块96孔板,每孔加入90ul的细胞培养液(RPMI1640+2%FBS),然后每孔加入10ul不同浓度的待测样品,混匀,接着向细胞培养板中加入5μ同的不同浓度的待测样品,每个样品两复孔。将培养板在培养箱孵育3天(37℃,5%CO 2)。取出96孔细胞培养板,向每孔加入50μ0CTG溶液,室温孵育10分钟。于酶标仪(BMG labtech,PHERAstar FS)中,用酶标仪测定化学发光。
三、数据分析
用Microsoft Excel,Graphpad Prism 5对数据进行处理分析。实施例结果见下表3。
表3 本公开化合物对Colo205肿瘤细胞体外增殖抑制的IC 50
实施例编号 IC 50(nM)
1 90
2 32
3 26
4 82
5 81
6 74
7 158
9 77
10 62
13 99
14 92
15 55
16 180
19 101
20 68
21 120
药代动力学评价
测试例4、本公开化合物的小鼠药代动力学测试
1、摘要
以小鼠为受试动物,应用LC/MS/MS法测定了小鼠灌胃给予实施例3、实施例10、实施例15和实施例20化合物后,不同时刻血浆中的药物浓度。研究本公开化合物在小鼠体内的药代动力学行为,评价其药动学特征。
2、试验方案
2.1试验药品
实施例3、实施例10、实施例15和实施例20化合物。
2.2试验动物
C57小鼠36只,雌性,平均分为4组,购自上海杰思捷实验动物有限公司,动物生产许可证号:SCXK(沪)2013-0006。
2.3药物配制
称取一定量化合物,加5%体积的DMSO和5%吐温80使其溶解,然后加90%生理盐水配置成0.1mg/ml无色澄明溶液。
2.4给药
C57小鼠禁食过夜后灌胃给药,给药剂量均为2mg/kg,给药体积均为0.2ml/10g。
3、操作
小鼠灌胃给药,于给药前及给药后0.25、0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时采血0.1ml,置于肝素化试管中,3500转/分钟离心10分钟后,分离血浆,于-20℃保存。
测定不同浓度的药物注射给药后小鼠血浆中的待测化合物含量:取给药后各时刻的小鼠血浆25μl,加入内标溶液喜树碱(中国生物制品检定所)50μl(100ng/mL),乙腈200μl,涡旋混合5分钟,离心10分钟(4000转/分钟),血浆样品取上清液4μl进行LC/MS/MS分析。
4、药代动力学参数结果
本公开化合物的药代动力学参数如下:
Figure PCTCN2020081591-appb-000054
结论:本公开化合物的药代吸收较好,具有药代动力学优势。

Claims (21)

  1. 一种通式(I)所示的化合物:
    Figure PCTCN2020081591-appb-100001
    或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
    其中:
    R 1选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、氨基烷基和硝基,其中所述烷基任选地被NR 7R 8、烷氧基、卤素、氰基、硝基、羟基、羟烷基中的一个或多个取代基所取代;
    R 2相同或不同,各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基和硝基;
    R 3选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基,所述的烷基、环烷基、杂环基、芳基和杂芳基任选进一步被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R 4选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基和硝基;
    R 5相同或不同,各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
    R 6选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基和硝基;
    R 7和R 8相同或不同,各自独立地选自氢原子、烷基、羟烷基和卤代烷基;
    m选自0、1、2、3、4或5;
    n选自0、1、2或3;
    z选自0、1、2、3或4;
    Q选自0、1或2。
  2. 根据权利要求1所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 1选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、氨基烷基和硝基。
  3. 根据权利要求1或2所述的通式(I)所示的化合物,其为通式(I-P)所示的化合物:
    Figure PCTCN2020081591-appb-100002
    或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
    其中,R 1~R 6、m、n、z、Q如权利要求1或2中所定义。
  4. 根据权利要求1至3中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 4为氢原子。
  5. 根据权利要求1至4中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的n为1或2。
  6. 根据权利要求1或2所述的通式(I)所示的化合物,其为通式(II)或通式(II-P)所示的化合物:
    Figure PCTCN2020081591-appb-100003
    或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
    其中,R 1~R 6、m、z、Q如权利要求1或2中所定义。
  7. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 1选自氢原子、烷基、羟基、氨基烷基、烷基氨基烷基和羟烷基;优选地,R 1选自氢原子、C 1-6烷基、羟基、氨基C 1-6烷基、C 1-6烷基氨基C 1-6烷基和C 1-6羟烷基;更优选地,R 1选自氢原子、甲基、羟甲基、氨基甲基和甲基氨基甲基。
  8. 根据权利要求1至7中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 1选自氢原子、烷基、羟基、氨基烷基和羟烷基;优选地,R 1选自氢原子、C 1-6烷基、羟基、氨基C 1-6烷基和C 1-6羟烷基;更优选地,R 1选自氢原子、甲基、羟甲基和氨基甲基。
  9. 根据权利要求1或2所述的通式(I)所示的化合物,其为通式(III)或通式(III-P)所示的化合物:
    Figure PCTCN2020081591-appb-100004
    或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映 异构体、或其混合物形式或其可药用的盐,
    其中:
    p选自0、1、2或3,优选为1;
    R 2、R 3、R 5、R 6、m、n、z、Q如权利要求1或2中所定义。
  10. 根据权利要求1至9中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 2选自氢原子、卤素和烷基;优选地,R 2选自氢原子、卤素和C 1-6烷基;更优选地,R 2为C 1-6烷基。
  11. 根据权利要求1至10中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 3为烷基、羟烷基、环烷基、杂环基和杂芳基,所述的烷基、环烷基、杂环基和杂芳基任选进一步被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和羟烷基中的一个或多个取代基所取代;优选地,R 3为C 1-6烷基、C 1-6羟烷基、C 3-6环烷基、3-8元杂环基和5-10元杂芳基,所述的C 1-6烷基、C 3-6环烷基、3-8元杂环基和5-10元杂芳基任选进一步被选自C 1-6烷基、C 1-6烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和C 1-6羟烷基中的一个或多个取代基所取代。
  12. 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 3为杂芳基,所述的杂芳基任选进一步被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和羟烷基中的一个或多个取代基所取代;优选地,R 3为5-10元杂芳基,所述的5-10元杂芳基任选进一步被选自C 1-6烷基、C 1-6烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基和C 1-6羟烷基中的一个或多个取代基所取代;更优选地,R 3为吡唑基,所述的吡唑基任选地被C 1-6烷基,优选甲基取代。
  13. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 5选自氢原子、烷基、烷氧基和卤素;优选地,R 5选自氢原子、C 1-6烷基、C 1-6烷氧基和卤素。
  14. 根据权利要求1至13中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 6为氢原子。
  15. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其选自:
    Figure PCTCN2020081591-appb-100005
    Figure PCTCN2020081591-appb-100006
  16. 一种通式(IIIA)或通式(III-PA)所示的化合物:
    Figure PCTCN2020081591-appb-100007
    或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
    其中:
    R w为羟基保护基;
    R 2、R 3、R 5、R 6、m、n、p、Q和z如权利要求9中所定义。
  17. 根据权利要求16所述的通式(IIIA)或通式(III-PA)所示的化合物,其选自:
    Figure PCTCN2020081591-appb-100008
    Figure PCTCN2020081591-appb-100009
  18. 一种制备根据权利要求9所述的通式(III)或通式(III-P)所示的化合物的方法,该方法包括以下步骤:
    Figure PCTCN2020081591-appb-100010
    通式(IIIA)或通式(III-PA)化合物在酸性条件下,脱去羟基保护基R w,得到通式(III)或通式(III-P)的化合物;
    其中:
    所述的羟基保护基R w优选TBS;
    R 2、R 3、R 5、R 6、m、n、p、Q和z如权利要求9中所定义。
  19. 一种药物组合物,所述药物组合物含有治疗有效量的根据权利要求1至15中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  20. 根据权利要求1至15中任一项所述的通式(I)所示的化合物或其立体异 构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐或根据权利要求19所述的药物组合物,在制备用于抑制ERK的药物中的用途。
  21. 根据权利要求1至15中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐或根据权利要求19所述的药物组合物在制备用于治疗或预防癌症、炎症、或其它增殖性疾病的药物中的用途,优选癌症;所述的癌症选自黑色素瘤、肝癌、肾癌、肺癌、鼻咽癌、结肠直肠癌、结肠癌、直肠癌、胰腺癌、宫颈癌、卵巢癌、乳腺癌、膀胱癌、前列腺癌、白血病、头颈鳞状细胞癌、子宫颈癌、甲状腺癌、淋巴瘤、肉瘤、成神经细胞瘤、脑瘤、骨髓瘤,星形细胞瘤和胶质瘤。
PCT/CN2020/081591 2019-03-29 2020-03-27 吡咯并杂环类衍生物、其制备方法及其在医药上的应用 WO2020200069A1 (zh)

Priority Applications (9)

Application Number Priority Date Filing Date Title
KR1020217033781A KR20210146956A (ko) 2019-03-29 2020-03-27 피롤로헤테로고리형 유도체, 이의 제조 방법, 및 의약에서 이의 응용
CA3135070A CA3135070A1 (en) 2019-03-29 2020-03-27 Pyrroloheterocyclic derivative, preparation method therefor, and application thereof in medicine
MX2021011823A MX2021011823A (es) 2019-03-29 2020-03-27 Derivado pirroloheterociclico, metodo de preparacion del mismo y su aplicacion en medicina.
CN202080019049.5A CN113544131B (zh) 2019-03-29 2020-03-27 吡咯并杂环类衍生物、其制备方法及其在医药上的应用
AU2020251621A AU2020251621A1 (en) 2019-03-29 2020-03-27 Pyrroloheterocyclic derivative, preparation method therefor, and application thereof in medicine
BR112021018924A BR112021018924A2 (pt) 2019-03-29 2020-03-27 Derivado de heterocíclico pirrol, método de preparação do mesmo e aplicação do mesmo em medicamento
JP2021557719A JP2022528083A (ja) 2019-03-29 2020-03-27 ピロロ複素環式誘導体、この誘導体のための調製方法及び医学におけるこの誘導体の用途
US17/600,043 US20220185818A1 (en) 2019-03-29 2020-03-27 Pyrroloheterocyclic derivative, preparation method therefor, and application thereof in medicine
EP20783525.7A EP3950690A4 (en) 2019-03-29 2020-03-27 PYRROLOHETEROCYCLIC DERIVATIVE, PROCESS FOR PRODUCTION THEREOF AND APPLICATION THEREOF IN MEDICINE

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201910247297.4 2019-03-29
CN201910247297 2019-03-29
CN202010194720.1 2020-03-19
CN202010194720 2020-03-19

Publications (1)

Publication Number Publication Date
WO2020200069A1 true WO2020200069A1 (zh) 2020-10-08

Family

ID=72664469

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/081591 WO2020200069A1 (zh) 2019-03-29 2020-03-27 吡咯并杂环类衍生物、其制备方法及其在医药上的应用

Country Status (11)

Country Link
US (1) US20220185818A1 (zh)
EP (1) EP3950690A4 (zh)
JP (1) JP2022528083A (zh)
KR (1) KR20210146956A (zh)
CN (1) CN113544131B (zh)
AU (1) AU2020251621A1 (zh)
BR (1) BR112021018924A2 (zh)
CA (1) CA3135070A1 (zh)
MX (1) MX2021011823A (zh)
TW (1) TW202102505A (zh)
WO (1) WO2020200069A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021110169A1 (zh) * 2019-12-06 2021-06-10 南京明德新药研发有限公司 作为erk抑制剂的噻唑并内酰胺类化合物及其应用
WO2022068860A1 (zh) * 2020-09-29 2022-04-07 江苏恒瑞医药股份有限公司 一种吡咯并杂环类衍生物的晶型及其制备方法
WO2022253186A1 (zh) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 一种二甲基取代的噻唑并吡咯酮类化合物的晶型及其制备方法
WO2022268065A1 (en) * 2021-06-22 2022-12-29 Fochon Biosciences, Ltd. Compounds as erk inhibitors
WO2023185869A1 (zh) * 2022-03-28 2023-10-05 江苏恒瑞医药股份有限公司 一种吡咯并杂环类衍生物的富马酸盐的晶型及其制备方法

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114315837B (zh) * 2020-09-29 2023-06-16 江苏恒瑞医药股份有限公司 一种erk抑制剂的结晶形式及其制备方法

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999061440A1 (en) 1998-05-26 1999-12-02 Smithkline Beecham Corporation Novel substituted imidazole compounds
WO2001057022A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of erk
WO2001056993A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of erk
WO2001056557A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of erk
WO2002022601A1 (en) 2000-09-15 2002-03-21 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
WO2012118850A1 (en) 2011-02-28 2012-09-07 Array Biopharma Inc. Serine/threonine kinase inhibitors
JP2013010719A (ja) * 2011-06-30 2013-01-17 Dainippon Sumitomo Pharma Co Ltd ベンズイミダゾロンおよびオキシインドール誘導体ならびにそれらの医薬用途
WO2013018733A1 (ja) 2011-07-29 2013-02-07 富士フイルム株式会社 1,5-ナフチリジン誘導体又はその塩
WO2013130660A1 (en) * 2012-02-28 2013-09-06 Amgen Inc. Amides as pim inhibitors
WO2014179154A2 (en) 2013-04-30 2014-11-06 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2015103133A1 (en) 2013-12-30 2015-07-09 Genentech, Inc. Serine/threonine kinase inhibitors
WO2016192063A1 (en) 2015-06-03 2016-12-08 Changzhou Jiekai Pharmatech Co. Ltd Heterocyclic compounds as erk inhibitors
CN107108648A (zh) * 2014-12-22 2017-08-29 伊莱利利公司 作为ERK抑制剂的噻吩并[2,3‑c]吡咯‑4‑酮衍生物
WO2017180817A1 (en) 2016-04-15 2017-10-19 Musc Foundation For Research Development Treatment of septicemia and ards with erk inhibitors
WO2018049127A1 (en) 2016-09-08 2018-03-15 Sabila Biosciences Llc 1,2-dithiolane compounds useful in neuroprotection, autoimmune and cancer diseases and conditions
CN107922387A (zh) * 2015-06-15 2018-04-17 阿沙纳生物科学公司 Erk1与erk2的杂环抑制剂及其在癌症治疗中的应用

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999061440A1 (en) 1998-05-26 1999-12-02 Smithkline Beecham Corporation Novel substituted imidazole compounds
WO2001057022A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of erk
WO2001056993A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of erk
WO2001056557A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of erk
WO2002022601A1 (en) 2000-09-15 2002-03-21 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
WO2012118850A1 (en) 2011-02-28 2012-09-07 Array Biopharma Inc. Serine/threonine kinase inhibitors
JP2013010719A (ja) * 2011-06-30 2013-01-17 Dainippon Sumitomo Pharma Co Ltd ベンズイミダゾロンおよびオキシインドール誘導体ならびにそれらの医薬用途
WO2013018733A1 (ja) 2011-07-29 2013-02-07 富士フイルム株式会社 1,5-ナフチリジン誘導体又はその塩
WO2013130660A1 (en) * 2012-02-28 2013-09-06 Amgen Inc. Amides as pim inhibitors
WO2014179154A2 (en) 2013-04-30 2014-11-06 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2015103133A1 (en) 2013-12-30 2015-07-09 Genentech, Inc. Serine/threonine kinase inhibitors
CN107108648A (zh) * 2014-12-22 2017-08-29 伊莱利利公司 作为ERK抑制剂的噻吩并[2,3‑c]吡咯‑4‑酮衍生物
WO2016192063A1 (en) 2015-06-03 2016-12-08 Changzhou Jiekai Pharmatech Co. Ltd Heterocyclic compounds as erk inhibitors
CN107849046A (zh) * 2015-06-03 2018-03-27 常州捷凯医药科技有限公司 作为erk抑制剂的杂环化合物
CN107922387A (zh) * 2015-06-15 2018-04-17 阿沙纳生物科学公司 Erk1与erk2的杂环抑制剂及其在癌症治疗中的应用
WO2017180817A1 (en) 2016-04-15 2017-10-19 Musc Foundation For Research Development Treatment of septicemia and ards with erk inhibitors
WO2018049127A1 (en) 2016-09-08 2018-03-15 Sabila Biosciences Llc 1,2-dithiolane compounds useful in neuroprotection, autoimmune and cancer diseases and conditions

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BIOORGANIC AND MEDICINAL CHEMISTRY, vol. 5, no. 3, 1997, pages 557 - 567
BONI A ET AL., CANCER RESEARCH, vol. 70, no. 13, 2010, pages 5213 - 5219
FREDERICK DT ET AL., CLINICAL CANCER RESEARCH, vol. 19, no. 5, 2013, pages 1225 - 1231
JOURNAL OF PORPHYRINS AND PHTHALOCYANINES, vol. 13, no. 10, 2009, pages 1098 - 1110
ROBERTS PJ ET AL., ONCOGENE, vol. 26, no. 22, 2007, pages 3291 - 3310
See also references of EP3950690A4
SMALLEY I ET AL., CANCER DISCOVERY, vol. 8, no. 2, 2018, pages 140 - 142
T. GREENE, PROTECTING GROUP IN ORGANIC SYNTHESIS
TETRAHEDRON, vol. 56, 2000, pages 9633 - 9640

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021110169A1 (zh) * 2019-12-06 2021-06-10 南京明德新药研发有限公司 作为erk抑制剂的噻唑并内酰胺类化合物及其应用
WO2022068860A1 (zh) * 2020-09-29 2022-04-07 江苏恒瑞医药股份有限公司 一种吡咯并杂环类衍生物的晶型及其制备方法
WO2022253186A1 (zh) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 一种二甲基取代的噻唑并吡咯酮类化合物的晶型及其制备方法
WO2022268065A1 (en) * 2021-06-22 2022-12-29 Fochon Biosciences, Ltd. Compounds as erk inhibitors
WO2023185869A1 (zh) * 2022-03-28 2023-10-05 江苏恒瑞医药股份有限公司 一种吡咯并杂环类衍生物的富马酸盐的晶型及其制备方法

Also Published As

Publication number Publication date
EP3950690A4 (en) 2022-12-07
JP2022528083A (ja) 2022-06-08
MX2021011823A (es) 2021-10-22
CN113544131B (zh) 2024-03-15
EP3950690A1 (en) 2022-02-09
BR112021018924A2 (pt) 2022-02-01
KR20210146956A (ko) 2021-12-06
TW202102505A (zh) 2021-01-16
AU2020251621A1 (en) 2021-11-04
US20220185818A1 (en) 2022-06-16
CN113544131A (zh) 2021-10-22
CA3135070A1 (en) 2020-10-08

Similar Documents

Publication Publication Date Title
WO2020200069A1 (zh) 吡咯并杂环类衍生物、其制备方法及其在医药上的应用
TWI772291B (zh) 取代的吡咯並嘧啶類cdk抑制劑、包含其藥物組合物及其用途
JP2019530670A (ja) Hpk1調節薬としてのピラゾロピリジン誘導体及びがんの治療のためのその用法
WO2018045956A1 (zh) 苯并咪唑类化合物激酶抑制剂及其制备方法和应用
WO2021249519A1 (zh) 吡啶酮并嘧啶类衍生物、其制备方法及其在医药上的应用
JP2021523168A (ja) がん幹細胞を標的化するがん治療
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
WO2023274397A1 (zh) Cdk2抑制剂及其制备方法和用途
WO2020238776A1 (zh) 取代的稠合双环类衍生物、其制备方法及其在医药上的应用
WO2020192750A1 (zh) 噻吩并杂环类衍生物、其制备方法及其在医药上的应用
WO2020125513A1 (zh) 作为cdk抑制剂的大环化合物、其制备方法及其在医药上的应用
WO2020011220A1 (zh) 杂芳基类衍生物、其制备方法及其在医药上的应用
WO2020221209A1 (zh) 一种cd73抑制剂,其制备方法和应用
CN107428762A (zh) 酞嗪酮衍生物、其制备方法及用途
JP2022502484A (ja) キナーゼ阻害活性を有する芳香族複素環式化合物
WO2023072297A1 (zh) 含氮的四环化合物、其制备方法及其在医药上的应用
CN115594695A (zh) 大环类化合物、其制备方法及其在医药上的应用
CN112996783B (zh) 2-氨基嘧啶类衍生物、其制备方法及其在医药上的应用
CN113912608B (zh) 嘧啶并嘧啶酮类衍生物、其制备方法及其在医药上的应用
WO2022199676A1 (zh) 稠合四环类化合物、其制备方法及其在医药上的应用
WO2021143821A1 (zh) 稠合杂芳基类衍生物、其制备方法及其在医药上的应用
WO2022166810A1 (zh) 稠合的氮杂三环类衍生物、其制备方法及其在医药上的应用
CN114907406A (zh) 嘧啶衍生物、其制备方法及其在医药上的应用
WO2022161447A1 (zh) 二甲酰胺类化合物、其制备方法及其在医药上的应用
WO2023109540A1 (zh) 具有akt激酶抑制活性的杂环化合物及其制备方法和医药用途

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: 20783525

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3135070

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021557719

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021018924

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217033781

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020783525

Country of ref document: EP

Effective date: 20211029

ENP Entry into the national phase

Ref document number: 2020251621

Country of ref document: AU

Date of ref document: 20200327

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112021018924

Country of ref document: BR

Free format text: APRESENTAR A TRADUCAO SIMPLES DA FOLHA DE ROSTO DA CERTIDAO DE DEPOSITO DAS PRIORIDADES CN 201910247297.4 DE 29/03/2019 E CN 202010194720.1 DE 19/03/2020 OU DECLARACAO CONTENDO, OBRIGATORIAMENTE, TODOS OS DADOS IDENTIFICADORES DESTAS (DEPOSITANTE(S), INVENTOR(ES), NUMERO DE REGISTRO, DATA DE DEPOSITO E TITULO), CONFORME O ART. 15 DA PORTARIA 39/2021.

ENP Entry into the national phase

Ref document number: 112021018924

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210922