WO2024008088A1 - 作为trk抑制剂和/或ret抑制剂的化合物及其用途 - Google Patents

作为trk抑制剂和/或ret抑制剂的化合物及其用途 Download PDF

Info

Publication number
WO2024008088A1
WO2024008088A1 PCT/CN2023/105745 CN2023105745W WO2024008088A1 WO 2024008088 A1 WO2024008088 A1 WO 2024008088A1 CN 2023105745 W CN2023105745 W CN 2023105745W WO 2024008088 A1 WO2024008088 A1 WO 2024008088A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
ethyl
mdi
indazol
cycloalkyl
Prior art date
Application number
PCT/CN2023/105745
Other languages
English (en)
French (fr)
Inventor
路良
黄海
张龙争
赵赛赛
张霁旋
Original Assignee
河南迈英诺医药科技有限公司
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 河南迈英诺医药科技有限公司 filed Critical 河南迈英诺医药科技有限公司
Publication of WO2024008088A1 publication Critical patent/WO2024008088A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention provides the use of a class of compounds for TRK kinase and/or RET kinase.
  • the invention also relates to compositions comprising said compounds, and the use of said compounds and said compositions for the preparation of medicaments for the treatment and/or prevention of diseases or conditions associated with TRK and/or RET.
  • Protein kinases are a family of enzymes that catalyze the phosphorylation of specific residues in proteins and are broadly classified into tyrosine and serine/threonine kinases. Inappropriate kinase activity due to mutation, overexpression or inappropriate regulation, dysregulation or dysregulation, and overproduction or underproduction of growth factors or cytokines is implicated in many diseases, including but not limited to cancer, cardiovascular disease, Allergies, asthma and other respiratory diseases, autoimmune diseases, inflammatory diseases, bone diseases, metabolic disorders and neurological and neurodegenerative conditions (such as Alzheimer's disease).
  • diseases including but not limited to cancer, cardiovascular disease, Allergies, asthma and other respiratory diseases, autoimmune diseases, inflammatory diseases, bone diseases, metabolic disorders and neurological and neurodegenerative conditions (such as Alzheimer's disease).
  • TRK kinases are high-affinity receptor tyrosine kinases activated by a group of soluble growth factors called neurotrophic factors (NTs).
  • the TRK family has three members: TRKA, TRKB and TRKC.
  • TRKA is activated by nerve growth factor (NGF)
  • TRKB is activated by brain-derived nerve growth factor (BDNF) and NT-4/5
  • TRKC is activated by NT3.
  • TRK is widely expressed in neuronal tissues and is involved in the maintenance, signaling and survival of neuronal cells.
  • Inhibitors of the TRK/neurotrophin pathway have been shown to be effective in many preclinical animal models of pain. NGF secreted by tumor cells and tumor-invasive macrophages has also been shown to directly stimulate peripherally located pain fibers, so inhibitors of TrkA and/or other Trk kinases may provide an effective treatment for chronic pain states and cancer-related pain. It has also been reported that overexpression, activation, amplification and/or mutation of Trk kinase is associated with many cancers, such as neuroblastoma, colorectal cancer, melanoma, gastric cancer, lung cancer, breast cancer, etc.
  • Vitrakvi also known as larotrectinib, larotrectinib, LOXO101
  • Loxo Oncology for the treatment of patients with neurotrophic tyrosine kinase (NTRK) genes
  • NRRK neurotrophic tyrosine kinase
  • RET kinase is also a very important drug target.
  • Selpercatinib Retevmo
  • Loxo Oncology the FDA approved the RET kinase inhibitor Pralsetinib (Pralsetinib) developed by Blueprint Medicines for the treatment of adult patients with RET fusion-positive metastatic non-small cell lung cancer.
  • TRK or RET inhibitors have been approved for marketing, and a large number of TRK or RET inhibitors are in the clinical research stage, these TRK/RET inhibitors are not satisfactory in terms of efficacy or safety. Therefore, there is always a need for TRK inhibitors or RET inhibitors with better efficacy and/or fewer side effects.
  • One purpose of the present invention is to provide a new class of TRK inhibitors that can replace existing TRK inhibitors, thereby providing more options for the treatment of TRK-related diseases.
  • a further object of the present invention is to provide a new type of TRK inhibitor with better efficacy and/or better safety than existing TRK inhibitors.
  • Another object of the present invention is to provide a new class of RET inhibitors that can replace existing RET inhibitors, thereby providing more options for the treatment of RET-related diseases.
  • a further object of the present invention is to provide a new class of RET inhibitors with better efficacy and/or better safety than existing RET inhibitors.
  • Another object of the present invention is to treat diseases related to TRK and/or RET such as pruritus, psoriasis, atopic dermatitis, acne, vitiligo, alopecia areata, asthma, rhinitis, hemorrhoids, cervicitis, pneumonia, cancer (tumor), etc.
  • RET diseases related to TRK and/or RET
  • Chinese invention patent CN111606908B (equivalent to US Patent Publication US2022/0073524A1) discloses a class of compounds that serve as inhibitors of JAK.
  • JAK is the abbreviation of English Janus kinase, which is a cytoplasmic tyrosine kinase that transduces cytokine signals from membrane receptors to STAT transcription factors.
  • JAK is a very important drug target, which is involved in many important biological processes such as cell proliferation, differentiation, apoptosis and immune regulation.
  • JAK inhibitors developed for this target are mainly used to treat hematological diseases, tumors, rheumatoid arthritis and other drugs.
  • the JAK protein family includes 4 members: JAK1, JAK2, JAK3 and TYK2. According to CN111606908B, the compound disclosed therein has high inhibitory activity against JAK1, JAK2, JAK3, and TYK2, and is a highly efficient JAK inhibitor.
  • the inventor unexpectedly found that the compound disclosed in CN111606908B also has very high inhibitory activity against TRK kinase, and has very good inhibitory activity against TRKA, TRKB, and TRKC, so it is a class of TRK inhibitors.
  • the inventor unexpectedly found that the compound disclosed in CN111606908B also has very high inhibitory activity against RET kinase.
  • the compounds described in the present application can provide better efficacy than existing drugs in the treatment of many diseases, and can treat acne, a disease caused by other JAK inhibitors. Side reactions (Int.J.Dermatol.2021 Aug 22.doi:10.1111/ijd.15853.PMID:34423443).
  • the compounds described herein have been found to be particularly suitable for the treatment of autoimmune diseases, especially cutaneous autoimmune diseases.
  • the compounds involved in the present application have effects on itching, psoriasis, atopic dermatitis, skin side effects caused by EGFR inhibitors (such as those diseases listed in CN112933095A), acne, vitiligo, alopecia areata, Therapeutic effect on diseases such as asthma, rhinitis, hemorrhoids, cervicitis, pneumonia, slow wound healing (caused by diabetes), diabetes, and complications of diabetes (such as diabetic foot).
  • EGFR inhibitors such as those diseases listed in CN112933095A
  • acne vitiligo
  • alopecia areata
  • Therapeutic effect on diseases such as asthma, rhinitis, hemorrhoids, cervicitis, pneumonia, slow wound healing (caused by diabetes), diabetes, and complications of diabetes (such as diabetic foot).
  • CN111606908B of the present application has a very good therapeutic effect on diabetic foot and chronic wound healing including bedsores.
  • Diabetic foot is a common complication of diabetes, affecting nearly 400 million people worldwide. About 30% of patients end up with amputation due to difficulty in healing the wound.
  • Recent studies have shown that wound healing and skin repair require lipolysis and conversion of adipocytes into myofibroblasts, and patients with diabetic foot lack this ability (Cell Stem Cell 26,1–16, June 4, 2020).
  • JAK/TRK dual inhibitors preferably pan-JAK/pan-TRK dual inhibitors
  • JAK/TRK/RET multiple inhibitors preferably pan-JAK/pan-TRK/RET multiple inhibitors
  • the invention provides compounds of formula (G):
  • TRK isotope labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite as TRK
  • inhibitor drugs and/or RET inhibitor drugs or use in the preparation of TRK inhibitor drugs and/or RET inhibitor drugs, wherein:
  • X 1 is N or CR 14 ;
  • X 2 is N or CR 15 ;
  • X 3 is N or CR 16 ;
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -SR 12 , -OR 12 , -CN, halogen, -NO 2 , -SF 5 , -SC 1-4 Alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic group, C 5-11 bicyclic alkyl, 5-11 membered bicyclic heteroalkyl, and R 13 is replaced by 0, 1, 2, 3 or 4 R 1 Substituted; wherein R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkyne Base, C 3-7 cycloalkyl, C 3-7
  • isotopically labeled compounds of the compounds of formula (G) above are used.
  • isotopically labeled compounds of the compound of formula (G) are used, wherein each H is independently optionally substituted with D.
  • X 1 is N. In some preferred embodiments of the invention, in formula (G), X2 is N. In some preferred embodiments of the invention, in formula (G), X3 is N. In some preferred embodiments of the present invention, in formula (G), X 1 is CR 14 , X 2 is N or CR 15 , and X 3 is CR 16 . In some preferred embodiments of the present invention, in formula (G), X 1 is CR 14 , X 2 is CR 15 , and X 3 is CR 16 .
  • X 1 is CR 14
  • X 2 is CR 15
  • X 3 is CR 16
  • R 14 , R 15 , and R 16 are each independently selected from H , -OH, -CN, halogen, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl.
  • X 1 is CR 14
  • X 2 is N
  • X 3 is CR 16
  • R 14 and R 16 are each independently selected from H, -OH, -CN, halogen, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl.
  • X 1 , X 2 , and X 3 are the same.
  • X 1 , X 2 , and X 3 are all CH.
  • X 1 , X 2 , and X 3 are all N.
  • X 1 is C(CH 3 ), and X 2 and X 3 are both CH.
  • X 2 is C(CH 3 ), and X 1 and X 3 are both CH.
  • X 3 is C(CH 3 ), and X 1 and X 2 are both CH.
  • X 1 is N, and X 2 and X 3 are both CH.
  • X 2 is N, and X 1 and X 3 are both CH. In some preferred embodiments of the present invention, in formula (G), X 3 is N, and X 1 and X 2 are both CH.
  • isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 1 , X 2 , X 3 are the same . In some more preferred embodiments of the invention, isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 1 , X 2 , X 3 are all CH . In some more preferred embodiments of the invention, isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 1 , X 2 , X 3 are all N .
  • isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 1 is C(CH 3 ), X 2 , X 3 are all CH. In some more preferred embodiments of the invention, isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted with D, and X2 is C( CH3 ), X1 , X 3 are all CH.
  • isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted with D, and X3 is C( CH3 ), X1 , X 2 are all CH.
  • isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 1 is N, X 2 , X 3 are all for CH.
  • isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 2 is N, X 1 , X 3 are both for CH.
  • isotopically labeled compounds of the compound of formula (G) are used, wherein all H are each independently optionally substituted by D, and X 3 is N, X 1 , X 2 are both for CH.
  • X 1 , X 2 , and X 3 are all CH, and L is CH 2 .
  • X 1 , X 2 , and X 3 are all CH, and L is a connecting bond.
  • X 1 , X 2 , and X 3 are all N, and L is CH 2 .
  • X 1 , X 2 , and X 3 are all N, and L is a connecting bond.
  • X 1 , X 2 , and X 3 are all CR 14 , wherein R 14 is selected from -OH, -CN, halogen, C 1-6 alkane group, C 1-6 alkoxy group, C 3-7 cycloalkyl group, 3-7 membered heterocycloalkyl group, L is CH 2 .
  • X 1 , X 2 , and X 3 are all CR 14 , wherein R 14 is selected from -OH, -CN, halogen, C 1-6 alkane base, C 1-6 alkoxy group, C 3-7 cycloalkyl group, 3-7 membered heterocycloalkyl group, L is the connecting bond.
  • X 1 is C(CH 3 ), X 2 and X 3 are both CH, and L is CH 2 .
  • X 1 is C(CH 3 ), X 2 and X 3 are both CH, and L is a connecting bond.
  • X 2 is C(CH 3 ), X 1 and X 3 are both CH, and L is CH 2 .
  • X 2 is C(CH 3 ), X 1 and X 3 are both CH, and L is a connecting bond.
  • X 3 is C(CH 3 ), X 1 and X 2 are both CH, and L is CH 2 .
  • X 3 is C(CH 3 ), X 1 and X 2 are both CH, and L is a connecting bond.
  • X 1 is N
  • X 2 and X 3 are both CH
  • X 1 is N
  • X 2 and X 3 are both CH
  • X 1 is N
  • X 2 and X 3 are both CH
  • L is CH 2 .
  • X 1 is N
  • X 2 and X 3 are both CH
  • L is a connecting bond.
  • X 2 is N
  • X 1 and X 3 are both CH
  • X 2 is N
  • X 1 and X 3 are both CH
  • X 2 is N, X 1 and X 3 are both CH, and L is CH 2 .
  • X 2 is N
  • X 1 and X 3 are both CH
  • L is a connecting bond.
  • X 3 is N, X 1 and X 2 are both CH, and L is CH 2 .
  • X 3 is N, X 1 and X 2 are both CH, and L is a connecting bond.
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN, Halogen, -NO 2 , -SF 5 , -SC 1-4 alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5 -7-membered heteroaryl, C 7-11 bicyclic aryl, 7-11-membered bicyclic heteroaryl, 11-15-membered tricyclic, C 5-11 bicycloalkyl, 5-11-membered bicyclic heteroalkyl, and R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl , C 5-7 aryl , 5-7 membered heteroaryl, and optionally substituted by one or more -
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN, Halogen, -NO 2 , -SF 5 , -SC 1-4 alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5 -7-membered heteroaryl, C 7-11 bicyclic aryl, 7-11-membered bicyclic heteroaryl, 11-15-membered tricyclic group, and R 17 and R 18 are as defined above (wherein R 13 is optionally 1, 2, 3 or 4 R 1 replaced).
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, C 1-6 alkyl or C 3 -7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, and R 17 and R 18 are as defined above (wherein R 13 is optionally replaced by 1 , 2, 3 or 4 R 1 replaced).
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy, C 1-6 alkyl or C 3-7 Cycloalkyl, 4-6 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, and R 17 and R 18 are as defined above (wherein R 13 is optionally replaced by 1, 2 or 3 R 1 replaced).
  • R 13 is -N(R 17 )(R 18 ), C 1-3 alkoxy, C 3-6 cycloalkyl, 4-6 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl or C 1-4 alkyl, and R 17 and R 18 are as defined above (wherein R 13 is optionally replaced by 1, 2 or 3 R 1 replaced).
  • R 13 is -N(H)(C 1-3 alkyl), -N(H)(3-6 membered cycloalkyl), - N(H)(4-6 membered heterocycloalkyl), -N(C 1-3 alkyl)(C 1-3 alkyl), C 1-3 alkoxy, C 3-6 cycloalkyl, 4-6 membered azacycloalkyl or oxaheterocycloalkyl, phenyl, 5-6 membered azaaryl or C 1-4 alkyl; or R 13 is -N (R 17 ) (R 18 ), And R 17 , R 18 and the N atoms connected to them together form a 4-10 membered ring (where R 13 is optionally replaced by 1, 2 or 3 R 1 ).
  • R 13 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methyl, ethyl, propyl, butyl, methyl Oxygen, ethoxy, propoxy, -N(H)(CH 3 ), -N(H)(CH 2 CH 3 ), -N(H)(CH 2 CH 2 OH), -N(H )(CH 2 CH 2 CN), -N(CH 3 )(CH 3 ), -N(H)(cyclopropyl), -N(H)(cyclobutyl), -N(H)(tetrahydrofuryl) ), pyrazinyl, pyridazinyl, pyrrolidinyl, pyrazolyl, piperidinyl, phenyl, azetidinyl, morpholinyl, piperazinyl or tetrahydro
  • R 13 is cyclopropyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is cyclobutyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is cyclopentyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is cyclohexyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is methyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is ethyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is propyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is butyl.
  • R 13 is pyrazinyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is pyridazinyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is pyrrolidinyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is pyrazolyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is piperidinyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is phenyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is azetidinyl.
  • R 13 is morpholinyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is piperazinyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is tetrahydropyranyl. In some particularly preferred embodiments of the invention, in formula (G), R 13 is methoxy. In some particularly preferred embodiments of the invention, in formula (G), R 13 is ethoxy. In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(H)(CH 3 ). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(H)(CH 2 CH 3 ).
  • R 13 is -N(H)(CH 2 CH 2 OH). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(H)(CH 2 CH 2 CN). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(CH 3 )(CH 3 ). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(H)(cyclopropyl). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(H)(cyclobutyl). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(H)(tetrahydrofuryl). In some particularly preferred embodiments of the invention, in formula (G), R 13 is -N(R 17 )(R 18 ), and R 17 , R 18 and the N atoms connected to them together form a 7-membered ring.
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl base, C 3-7 heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, and optionally substituted by one or more -OH, -CN, -SH, halogen, -NO 2 , -SF 5 superseded.
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 heterocycle Alkyl, optionally substituted by one or more -OH, -CN, -SH, halogen, -NO2 , -SF5 .
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 heterocycle Alkyl, optionally substituted by one or more -OH, -CN.
  • R 17 and R 18 are each independently selected from H, methyl, ethyl, propyl, 3-membered cycloalkyl, 4-membered cycloalkyl, 5-membered cycloalkyl, 5-membered heterocycloalkyl, 6-membered heterocycloalkyl, and optionally substituted by one or more -OH, -CN.
  • R 17 , R 18 and the N atoms connected to them together form a 4-10 membered ring.
  • R 17 , R 18 and the N atoms connected to them together form a 7-membered ring.
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH , -SH, -CN, halogen, -NO 2 , -SF 5 or -SC 1-4 alkyl, and R 13 is replaced by 0, 1, 2, 3 or 4 R 1 ; wherein R 17 and R 18 are each Independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl, C 5-7 aryl, 5-7 membered hetero Aryl, and optionally substituted by one or more -OH, -CN, -SH, halogen, -NO 2 , -SF 5 ; or R 17 , R 18 and the N atoms connected to them together form 3-14 Yuan ring.
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy
  • R 17 , R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl, and optionally replaced by one or more -OH , -CN, -SH, halogen, -NO 2 , -SF 5 substitution; or R 17 , R 18 and the N atoms connected to them together form a 3-10 membered ring.
  • R 13 is methoxy, ethoxy, propoxy, -N(H)(CH 3 ) , -N(H)(CH 2 CH 3 ), -N(H)(CH 2 CH 2 OH), -N(H)(CH 2 CH 2 CN), -N(CH 3 )(CH 3 ), -N(H)(cyclopropyl), -N(H)(cyclobutyl), -N(H)(tetrahydrofuryl); or R 13 is -N(R 17 )(R 18 ), and R 17 , R 18 and the N atoms connected to them together form a 7-membered ring.
  • R 13 is -N(H)(tetrahydrofuranyl).
  • R 13 is -N(R 17 )(R 18 ), and R 17 , R 18 and connected to them The N atoms together form a 7-membered ring.
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from H, halogen, -OH, -NO 2 , -CN, - SF 5 , -SH, -SC 1-4 alkyl, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkynyl , C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, wherein -SC 1-4 alkyl, C 1-6 alkyl, C 3-7 cycloalkyl, 4-10 membered heterocycle
  • Each alkyl group is optionally substituted with 1, 2 or 3 substituents each independently selected from the group consisting of: halogen, -OH, -NH2 , -NH( CH3 ), -N( CH3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from halogen, C 1-6 alkyl and C 3-6 cycloalkyl group, wherein each of the C 1-6 alkyl and C 3-6 cycloalkyl groups is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, -NH 2. -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from halogen, C 1-6 alkyl, wherein said C 1- 6
  • the alkyl group is optionally substituted with 1, 2 or 3 substituents each independently selected from the following groups: halogen, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 1 or 2, and R 2 is selected from halogen and C 1-6 alkyl.
  • the number of R 2 is 1 or 2, and R 2 is selected from fluorine, chlorine, bromine, methyl, ethyl, n-propyl, iso Propyl, n-butyl, isobutyl. In some preferred embodiments of the present invention, in formula (G), the number of R 2 is 1 or 2, and R 2 is selected from fluorine, chlorine, methyl, ethyl, n-propyl, isopropyl . In some preferred embodiments of the present invention, in formula (G), the number of R 2 is 1 or 2, and R 2 is selected from fluorine, methyl, and ethyl.
  • the number of R 2 is 1 or 2, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (G), the number of R 2 is 1, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (G), the number of R 2 is 2, and R 2 is selected from fluorine and ethyl. In some particularly preferred embodiments of the present invention, in formula (G), there are two R 2 , which are fluorine and ethyl respectively. In some particularly preferred embodiments of the invention, in formula (G), 1 R2 is present and R2 is ethyl.
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1 , and each R 1 is independently selected from H, halogen, -OH, - NO 2 , -CN, -SF 5 , -SH, -SC 1-4 alkyl, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy, C 3-7 cycloalkyl, 3-10 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, wherein The -SC 1-4 alkyl, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy groups are optionally substituted by 1, 2, or 3 Or 4 R 3 substituted, and wherein the C 3-7 cycloalkyl, 3-10 membered heterocycloalkyl, C
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1 , and each R 1 is independently selected from H, halogen, -OH, - NO 2 , -CN, -SF 5 , -SH, -SC 1-4 alkyl, C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl base, 5-7 membered heteroaryl, wherein the -SC 1-4 alkyl, C 1-8 alkyl is optionally substituted by 1, 2, 3 or 4 R 3 , and wherein the C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl are optionally substituted by 1, 2, 3 or 4 R 4s .
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1s , and each R 1 is independently selected from halogen, -OH, -CN, C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, wherein the C 1-8 alkyl is optional is substituted by 1, 2, 3 or 4 R 3 , and wherein the C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered hetero Aryl groups are optionally substituted with 1, 2, 3 or 4 R4 .
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1s , and each R 1 is independently selected from halogen, -OH, -CN, C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, wherein the C 1-8 alkyl is optionally substituted by 1, 2 or 3 R 3 , and Wherein the C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl are optionally substituted by 1, 2 or 3 R 4s .
  • R 13 is replaced by 0 or 1 R 1 , and each R 1 is independently selected from halogen, -OH, -CN, C 1-6 alkane base, C 3-7 cycloalkyl, 5-7 membered heterocycloalkyl, wherein the C 1-6 alkyl is optionally substituted by 1, 2 or 3 R 3 , and wherein the C 3 -7 cycloalkyl and 5-7 membered heterocycloalkyl are optionally substituted by 1, 2 or 3 R4 .
  • R 13 is replaced by 0 or 1 R 1 , and each R 1 is independently selected from halogen, -OH, -CN, C 1-4 alkane base, C 3-6 cycloalkyl, 5-7 membered heterocycloalkyl, wherein the C 1-4 alkyl is optionally substituted by 1 or 2 R 3 , and wherein the C 3-6 cycloalkyl Alkyl, 5-7 membered heterocycloalkyl is optionally substituted by 1, 2 or 3 R4 .
  • R 13 is substituted by 0 or 1 R 1 , and each R 1 is independently selected from methyl, ethyl, hydroxyl, -CN, Piperidinyl, morpholinyl, piperazinyl, cyclopropyl, wherein piperidinyl, morpholinyl, piperazinyl are optionally substituted by 1, 2, 3 or 4 C 1-3 alkyl groups .
  • R 13 is substituted by 0 or 1 R 1 , and each R 1 is independently selected from methyl, ethyl, hydroxyl, -CN, Piperidinyl, morpholinyl, 1-methylpiperazinyl, cyclopropyl.
  • R1 is absent.
  • R1 is 1-methylpiperazinyl.
  • R1 is methyl.
  • R1 is ethyl.
  • R1 is piperidinyl. In some particularly preferred embodiments of the invention, in formula (G), R1 is morpholinyl. In some particularly preferred embodiments of the invention, in formula (G), R1 is hydroxyl. In some particularly preferred embodiments of the invention, in formula (G), R1 is -CN. In some particularly preferred embodiments of the invention, in formula (G), R1 is cyclopropyl.
  • the invention provides compounds of formula (G'):
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -SR 12 , -OR 12 , -CN, halogen, -NO 2 , -SF 5 , -SC 1-4 Alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic group, C 5-11 bicyclic alkyl, 5-11 membered bicyclic heteroalkyl, and R 13 is replaced by 0, 1, 2, 3 or 4 R 1 Substituted; wherein R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkyne Base, C 3-7 cycloalkyl, C 3-7
  • R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 are each independently H or selected from the following group: C 1-6 alkyl, C 1-4 haloalkyl, C 3-7 cycloalkyl, 4-14 membered heterocycloalkyl, C 6-10 aryl, 5-10 membered heteroaryl, (C 3-7 cycloalkyl)-C 1-4 alkyl-, ( 4-10 membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl)-C 1-4 alkyl- and (5-10 membered heteroaryl)-C 1-4 alkyl -, wherein each option within this group is optionally substituted with 1, 2, 3 or 4 substituents each independently selected from the group: halogen, -CF 3 , -OH, -NH 2 , - NH(CH 3 ), -N(CH 3 ) 2 , -CN, oxo, C 1-4
  • isotopically labeled compounds of the compound of formula (G') above are used.
  • isotopically labeled compounds of the compound of formula (G') are used, wherein each H is independently optionally substituted with D.
  • X is N. In some more preferred embodiments of the invention, in formula (G), X is CH.
  • isotopically labeled compounds of the compound of formula (G') are used, wherein each H is independently optionally substituted with D and X is N. In some more preferred embodiments of the invention, isotopically labeled compounds of the compound of formula (G') are used, wherein each H is independently optionally substituted with D and X is CH.
  • in formula (G') is CH2 .
  • in formula (G') is a connecting bond.
  • X is CH and L is CH2 .
  • X is CH and L is a connecting bond.
  • X is N and L is CH2 .
  • X is N and L is a connecting bond.
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN , Halogen, -NO 2 , -SF 5 , -SC 1-4 alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic, C 5-11 bicycloalkyl, 5-11 membered bicyclic heteroalkyl,
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl, C 5-7 aryl radical, 5-7 membered heteroaryl, and optionally substituted by one or more -OH, -
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN , Halogen, -NO 2 , -SF 5 , -SC 1-4 alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic group, and R 17 and R 18 are as defined above (where R 13 is optional replaced by 1, 2, 3 or 4 R 1 ).
  • R 13 is H, -N(R 17 ) (R 18 ), C 1-6 alkoxy, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, and R 17 and R 18 are as defined above (wherein R 13 is optionally 1 1, 2, 3 or 4 R replaced by 1 ).
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy, C 1-6 alkyl or C 3- 7- cycloalkyl, 4-6-membered heterocycloalkyl, phenyl, 5-6-membered heteroaryl, and R 17 and R 18 are as defined above (where R 13 is optionally replaced by 1, 2 or 3 replaced by R 1 ).
  • R 13 is -N(R 17 )(R 18 ), C 1-3 alkoxy, C 3-6 cycloalkyl, 4- 6-membered heterocycloalkyl, phenyl, 5-6-membered heteroaryl or C 1-4 alkyl, and R 17 and R 18 are as defined above (where R 13 is optionally replaced by 1, 2 or 3 replaced by R 1 ).
  • R 13 is -N(H) (C 1-3 alkyl), -N (H) (3-6 membered cycloalkyl), -N(H)(4-6 membered heterocycloalkyl), -N(C 1-3 alkyl)(C 1-3 alkyl), C 1-3 alkoxy, C 3-6 cycloalkyl , 4-6 membered azacycloalkyl or oxaheterocycloalkyl, phenyl, 5-6 membered azaaryl or C 1-4 alkyl; or R 13 is -N (R 17 ) (R 18 ) , and R 17 , R 18 and the N atoms connected to them together form a 4-10 membered ring (where R 13 is optionally replaced by 1, 2 or 3 R 1 ).
  • R 13 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methyl, ethyl, propyl, butyl, Methoxy, ethoxy, propoxy, -N(H)(CH 3 ), -N(H)(CH 2 CH 3 ), -N(H)(CH 2 CH 2 OH), -N( H)(CH 2 CH 2 CN), -N(CH 3 )(CH 3 ), -N(H)(cyclopropyl), -N(H)(cyclobutyl), -N(H)(tetrahydrofuran base), pyrazinyl, pyridazinyl, pyrrolidinyl, pyrazolyl, piperidinyl, phenyl, azetidinyl, morpholinyl, piperazinyl or tetrahydropyranyl
  • R 13 is cyclopropyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is cyclobutyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is cyclopentyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is cyclohexyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is methyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is ethyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is propyl.
  • R 13 is butyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is pyrazinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is pyridazinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is pyrrolidinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is pyrazolyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is piperidinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is phenyl.
  • R 13 is azetidinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is morpholinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is piperazinyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is tetrahydropyranyl. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is methoxy. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is ethoxy. In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(H)(CH 3 ).
  • R 13 is -N(H)(CH 2 CH 3 ). In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(H)(CH 2 CH 2 OH). In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(H)(CH 2 CH 2 CN). In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(CH 3 )(CH 3 ). In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(H)(cyclopropyl).
  • R 13 is -N(H)(cyclobutyl). In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(H)(tetrahydrofuryl). In some particularly preferred embodiments of the invention, in formula (G'), R 13 is -N(R 17 )(R 18 ), and R 17 , R 18 and the N atoms connected to them together form 7 Yuan ring.
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 ring Alkyl, C 3-7 heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, optionally substituted by one or more -OH, -CN, -SH, halogen, -NO 2 , -SF 5 replaced.
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 hetero Cycloalkyl, optionally substituted by one or more -OH, -CN, -SH, halogen, -NO2 , -SF5 .
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 hetero Cycloalkyl, optionally substituted by one or more -OH, -CN.
  • R 17 and R 18 are each independently selected from H, methyl, ethyl, propyl, 3-membered cycloalkyl, 4-membered cycloalkyl , 5-membered cycloalkyl, 5-membered heterocycloalkyl, 6-membered heterocycloalkyl, and optionally substituted by one or more -OH, -CN.
  • R 17 , R 18 and the N atoms connected to them together form a 4-10 membered ring.
  • R 17 , R 18 and the N atoms connected to them together form a 7-membered ring.
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy, - OH, -SH, -CN, halogen, -NO 2 , -SF 5 or -SC 1-4 alkyl, and R 13 is replaced by 0, 1, 2, 3 or 4 R 1 ; wherein R 17 , R 18 Each is independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl, C 5-7 aryl, 5-7 yuan Heteroaryl, and optionally substituted by one or more -OH, -CN, -SH, halogen, -NO 2 , -SF 5 ; or R 17 , R 18 and the N atoms connected to them together form 3- 14-membered ring.
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl, and are optionally replaced by one or more -OH, -CN , -SH, halogen, -NO 2 , -SF 5 substitution; or R 17 , R 18 and the N atoms connected to them together form a 3-10 membered ring.
  • R 13 is methoxy, ethoxy, propoxy, -N(H)(CH 3 ), -N(H)(CH 2 CH 3 ), -N(H)(CH 2 CH 2 OH), -N(H)(CH 2 CH 2 CN), -N(CH 3 )(CH 3 ) , -N(H)(cyclopropyl), -N(H)(cyclobutyl), -N(H)(tetrahydrofuryl); or R 13 is -N(R 17 )(R 18 ), and R 17 , R 18 and the N atoms connected to them together form a 7-membered ring.
  • R 13 is -N(H)(tetrahydrofuryl).
  • R 13 is -N(R 17 )(R 18 )
  • R 17 , R 18 and their combinations The connected N atoms together form a 7-membered ring.
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from H, halogen, -OH, -NO 2 , -CN, -SF 5 , -SH, -SC 1-4 alkyl, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkyne base, C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, wherein -SC 1-4 alkyl, C 1-6 alkyl, C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl Each cycloalkyl group is optionally substituted with 1, 2 or 3 substituents each independently selected from the group consisting of: halogen, -OH, -NH2 , -NH( CH3 ), -N( CH3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from halogen, C 1-6 alkyl and C 3-6 ring Alkyl, wherein each of the C 1-6 alkyl and C 3-6 cycloalkyl is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, - NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy .
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from halogen, C 1-6 alkyl, wherein said C 1 -6 Alkyl is optionally substituted with 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 1 or 2, and R 2 is selected from halogen and C 1-6 alkyl.
  • the number of R 2 is 1 or 2, and R 2 is selected from fluorine, chlorine, bromine, methyl, ethyl, n-propyl, Isopropyl, n-butyl, isobutyl. In some preferred embodiments of the present invention, in formula (G'), the number of R 2 is 1 or 2, and R 2 is selected from fluorine, chlorine, methyl, ethyl, n-propyl, isopropyl base. In some preferred embodiments of the present invention, in formula (G'), the number of R 2 is 1 or 2, and R 2 is selected from fluorine, methyl, and ethyl.
  • the number of R 2 is 1 or 2, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (G'), the number of R 2 is 1, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (G'), the number of R 2 is 2, and R 2 is selected from fluorine and ethyl. In some particularly preferred embodiments of the present invention, in formula (G'), there are two R 2 , which are fluorine and ethyl respectively. In some particularly preferred embodiments of the invention, in formula (G'), 1 R 2 is present and R 2 is ethyl.
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1 , and each R 1 is independently selected from H, halogen, -OH, -NO 2 , -CN, -SF 5 , -SH, -SC 1-4 alkyl, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy , C 3-7 cycloalkyl, 3-10 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, Wherein the -SC 1-4 alkyl, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy groups are optionally replaced by 1, 2, 3 One or four R 3 are substituted, and wherein the C 3-7 cycloalkyl, 3-10 membered heterocycloalkyl,
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1 , and each R 1 is independently selected from H, halogen, -OH, -NO 2 , -CN, -SF 5 , -SH, -SC 1-4 alkyl, C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 Aryl, 5-7 membered heteroaryl, wherein the -SC 1-4 alkyl, C 1-8 alkyl is optionally substituted by 1, 2, 3 or 4 R 3 , and wherein the The C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, and 5-7 membered heteroaryl are optionally substituted by 1, 2, 3 or 4 R 4 .
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1 , and each R 1 is independently selected from halogen, -OH, -CN , C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, wherein the C 1-8 alkyl is any Optionally substituted by 1, 2, 3 or 4 R 3 , and wherein the C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered Heteroaryl groups are optionally substituted with 1, 2, 3 or 4 R4 .
  • R 13 is replaced by 0, 1, 2, 3 or 4 R 1 , and each R 1 is independently selected from halogen, -OH, -CN , C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, wherein the C 1-8 alkyl is optionally substituted by 1, 2 or 3 R 3 , And wherein the C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl are optionally substituted by 1, 2 or 3 R 4s .
  • R 13 is replaced by 0 or 1 R 1 , and each R 1 is independently selected from halogen, -OH, -CN, C 1-6 Alkyl, C 3-7 cycloalkyl, 5-7 membered heterocycloalkyl, wherein the C 1-6 alkyl is optionally substituted by 1, 2 or 3 R 3 , and wherein the C 3-7 cycloalkyl and 5-7 membered heterocycloalkyl are optionally substituted by 1, 2 or 3 R4 .
  • R 13 is replaced by 0 or 1 R 1 , and each R 1 is independently selected from halogen, -OH, -CN, C 1-4 Alkyl, C 3-6 cycloalkyl, 5-7 membered heterocycloalkyl, wherein the C 1-4 alkyl is optionally substituted by 1 or 2 R 3 , and wherein the C 3-6 Cycloalkyl and 5-7 membered heterocycloalkyl are optionally substituted by 1, 2 or 3 R4 .
  • R 13 is substituted by 0 or 1 R 1 , and each R 1 is independently selected from methyl, ethyl, hydroxyl, -CN , piperidinyl, morpholinyl, piperazinyl, cyclopropyl, wherein piperidinyl, morpholinyl, piperazinyl is optionally replaced by 1, 2, 3 or 4 C 1-3 alkyl replace.
  • R 13 is substituted by 0 or 1 R 1 , and each R 1 is independently selected from methyl, ethyl, hydroxyl, -CN , piperidinyl, morpholinyl, 1-methylpiperazinyl, cyclopropyl.
  • R1 is absent.
  • R1 is 1-methylpiperazinyl.
  • R1 is methyl.
  • R1 is ethyl.
  • R1 is piperidinyl. In some particularly preferred embodiments of the invention, in formula (G'), R1 is morpholinyl. In some particularly preferred embodiments of the invention, in formula (G'), R1 is hydroxyl. In some particularly preferred embodiments of the invention, in formula (G'), R1 is -CN. In some particularly preferred embodiments of the invention, in formula (G'), R1 is cyclopropyl.
  • Ring A is C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic hetero Aryl, 11-15 membered tricyclic group, C 5-11 bicycloalkyl or 5-11 membered bicyclic heteroalkyl, which may be optionally substituted by R 1 , and the definitions of L, R 1 , R 2 , X As described above for compounds of formula (G').
  • TRK isotope labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite as TRK
  • inhibitor drugs and/or RET inhibitor drugs or use in the preparation of TRK inhibitor drugs and/or RET inhibitor drugs, wherein:
  • X is CH or N
  • Ring A is C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl Base, 11-15 membered tricyclic group;
  • R 1 is 0, 1, 2, 3 or 4, and R 1 is selected from H, halogen, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1- 8 alkoxy, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic Heteroaryl, wherein the C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy groups are optionally replaced by 1, 2, 3 or 4 R 3 is substituted, and wherein the C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7- The 11-membered bicyclic heteroaryl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 3 is selected from halogen, cyano, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -N(R 5 )(R 6 ), -CON(R 7 )(R 8 ) or 3- 7-membered heterocycloalkyl, wherein the 3-7-membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 4 is selected from halogen, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -NH 2 , -NHCH 3 or -N(CH 3 ) 2 ;
  • R 5 , R 6 , R 7 and R 8 are each independently hydrogen or C 1-4 alkyl
  • R 9 is selected from H, C 1-4 alkyl, C 1-4 haloalkyl or C 3-7 cycloalkyl;
  • R 10 is H or selected from the following group: C 1-4 alkyl, C 1-4 haloalkyl, C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, C 6-10 aryl, 5 -10-membered heteroaryl, (C 3-7 cycloalkyl)-C 1-4 alkyl-, (4-10-membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl base)-C 1-4 alkyl- and (5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally replaced by 1, 2, 3 or 4 each Substituted with substituents independently selected from the following groups: -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 3-7 cycloalkyl group, C 1-4 hydroxyalkyl group, -SC 1-4 alkyl group, -C
  • R 11 is selected from H, C 1-4 alkyl and C 3-7 cycloalkyl
  • R 12 is selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4- to 14-membered heterocycloalkyl, C 6-10 aryl, 5-10 membered heteroaryl, (C 3-7 Cycloalkyl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl)-C 1-4 alkyl- and ( 5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -CF 3 , -CN, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , oxo, -SC 1-4 alkyl, C 1-4 alkyl, C 1- 4 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloal
  • X is CH. In other embodiments of the invention, in formula (I), X is N.
  • X is CH and L is CH2 .
  • X is CH and L is a connecting bond.
  • X is N and L is CH2 .
  • X is N and L is a connecting bond.
  • ring A is C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl group (wherein ring A is optionally substituted by 1, 2, 3 or 4 R 1 ).
  • ring A is C 5-6 cycloalkyl, 5-6 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl (wherein ring A is optionally replaced by 1, 2, 3 or 4 R1 ).
  • ring A is 5-6 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl (wherein ring A is optionally replaced by 1, 2, 3 or 4 R 1 replaced).
  • ring A is a 5-6 membered azacycloalkyl, phenyl, 5-6 membered azaaryl (wherein ring A is optionally replaced by 1 1, 2, 3 or 4 R replaced by 1 ).
  • ring A is pyrazinyl, pyrazolyl, piperidinyl or phenyl (wherein ring A is optionally replaced by 1, 2, 3 or 4 R replaced by 1 ).
  • Ring A is pyrazinyl.
  • Ring A is pyrazolyl.
  • Ring A is piperidinyl.
  • ring A is phenyl.
  • R 1 is absent or R 1 is selected from C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1- 8 alkoxy, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, wherein the C 1-8 alkyl, C 2- 8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy is optionally substituted by 1, 2, 3 or 4 R 3 , and wherein the C 3-7 cycloalkyl, 3 -7-membered heterocycloalkyl, C 5-7 aryl, 5-7-membered heteroaryl are optionally substituted by 1, 2, 3 or 4 R 4s .
  • R 1 is absent or R 1 is selected from C 1-8 alkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, wherein the C 1-8 alkyl is optionally substituted by 1, 2, 3 or 4 R 3 , and wherein the C 3- 7- cycloalkyl, 3-7-membered heterocycloalkyl, C 5-7 aryl, 5-7-membered heteroaryl are optionally substituted by 1, 2, 3 or 4 R 4s .
  • R 1 is absent or R 1 is selected from C 1-8 alkyl, 3-7 membered heterocycloalkyl, wherein said C 1-8 alkyl The group is optionally substituted by 1, 2, 3 or 4 R3, and wherein the 3-7 membered heterocycloalkyl group is optionally substituted by 1, 2, 3 or 4 R4 .
  • R 1 is absent or R 1 is selected from C 1-6 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-6 alkyl The group is optionally substituted by 1, 2, 3 or 4 R3 , and wherein the 5-7 membered heterocycloalkyl group is optionally substituted by 1, 2, 3 or 4 R4 .
  • R 1 is absent or R 1 is selected from C 1-6 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-6 alkyl radical is optionally substituted by 1 or 2 R3 , and wherein the 5-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 C1-3 alkyl.
  • R 1 is absent or R 1 is selected from C 1-4 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-4 alkyl radical is optionally substituted by 1 or 2 R3 , and wherein the 5-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 C1-3 alkyl.
  • R 1 is absent or R 1 is selected from methyl, piperidyl, morpholinyl, piperazinyl, wherein piperidyl, morpholinyl, Piperazinyl is optionally substituted with 1, 2, 3 or 4 C 1-3 alkyl groups.
  • R 1 is absent or R 1 is selected from methyl, piperidinyl, morpholinyl, 1-methylpiperazinyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is absent. In some particularly preferred embodiments of the invention, in formula (I), R1 is 1-methylpiperazinyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is methyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is piperidinyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is morpholinyl.
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from halogen, C 1-6 alkyl and C 3-6 cycloalkyl group, wherein each of the C 1-6 alkyl and C 3-6 cycloalkyl groups is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, -NH 2. -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 1, 2 or 3, and R 2 is selected from halogen, C 1-6 alkyl, wherein said C 1- 6
  • the alkyl group is optionally substituted with 1, 2 or 3 substituents each independently selected from the following groups: halogen, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 1 or 2, and R 2 is selected from fluorine, chlorine, bromine, methyl, ethyl, n-propyl, iso Propyl, n-butyl, isobutyl. In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 1 or 2, and R 2 is selected from fluorine, chlorine, methyl, ethyl, n-propyl, isopropyl . In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 1 or 2, and R 2 is selected from fluorine, methyl, and ethyl.
  • the number of R 2 is 1 or 2, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 1, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 2, and R 2 is selected from fluorine and ethyl. In some particularly preferred embodiments of the present invention, in formula (I), there are two R 2 , which are fluorine and ethyl respectively. In some particularly preferred embodiments of the invention, in formula (I), 1 R2 is present and R2 is ethyl.
  • TRK isotope labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite as TRK
  • inhibitor drugs and/or RET inhibitor drugs or use in the preparation of TRK inhibitor drugs and/or RET inhibitor drugs, wherein:
  • X is CH
  • Ring A is a 5-7 membered heteroaryl group, C 5-7 aryl group;
  • R 1 is 0, 1, 2, 3 or 4, and R 1 is selected from C 1-8 alkyl, 3-7 membered heterocycloalkyl, wherein the C 1-8 alkyl is optionally Substituted by 1, 2, 3 or 4 R 3 , and wherein the 3-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 3 is selected from halogen, cyano, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -N(R 5 )(R 6 ), -CON(R 7 )(R 8 ) or 3- 7-membered heterocycloalkyl, wherein the 3-7-membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 4 is selected from halogen, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -NH 2 , -NHCH 3 or -N(CH 3 ) 2 ;
  • R 5 , R 6 , R 7 and R 8 are each independently hydrogen or C 1-4 alkyl
  • R 9 is selected from H, C 1-4 alkyl, C 1-4 haloalkyl or C 3-7 cycloalkyl;
  • R 10 is H or selected from the following group: C 1-4 alkyl, C 1-4 haloalkyl, C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, C 6-10 aryl, 5 -10-membered heteroaryl, (C 3-7 cycloalkyl)-C 1-4 alkyl-, (4-10-membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl base)-C 1-4 alkyl- and (5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally replaced by 1, 2, 3 or 4 each Substituted with substituents independently selected from the following groups: -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 3-7 cycloalkyl group, C 1-4 hydroxyalkyl group, -SC 1-4 alkyl group, -C
  • R 11 is selected from H, C 1-4 alkyl and C 3-7 cycloalkyl
  • R 12 is selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4- to 14-membered heterocycloalkyl, C 6-10 aryl, 5-10 membered heteroaryl, (C 3-7 Cycloalkyl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl)-C 1-4 alkyl- and ( 5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -CF 3 , -CN, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , oxo, -SC 1-4 alkyl, C 1-4 alkyl, C 1- 4 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloal
  • ring A is a 5-6 membered heteroaryl or phenyl group (wherein ring A is optionally replaced by 1, 2, 3 or 4 R 1 replaced).
  • Ring A is pyrazinyl, pyrazolyl or phenyl (wherein Ring A is optionally replaced by 1, 2, 3 or 4 R 1 replaced).
  • Ring A is pyrazinyl.
  • Ring A is pyrazolyl.
  • ring A is phenyl.
  • R 1 is absent or R 1 is selected from C 1-8 alkyl, 3-7 membered heterocycloalkyl, wherein said C 1-8 alkyl The group is optionally substituted by 1, 2, 3 or 4 R3, and wherein the 3-7 membered heterocycloalkyl group is optionally substituted by 1, 2, 3 or 4 R4 .
  • R 1 is absent or R 1 is selected from C 1-6 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-6 alkyl The group is optionally substituted by 1, 2, 3 or 4 R3 , and wherein the 5-7 membered heterocycloalkyl group is optionally substituted by 1, 2, 3 or 4 R4 .
  • R 1 is absent or R 1 is selected from C 1-6 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-6 alkyl radical is optionally substituted by 1 or 2 R3 , and wherein the 5-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 C1-3 alkyl.
  • R 1 is absent or R 1 is selected from C 1-4 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-4 alkyl radical is optionally substituted by 1 or 2 R3 , and wherein the 5-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 C1-3 alkyl.
  • R 1 is absent or R 1 is selected from methyl, piperidinyl, morpholinyl, wherein piperidinyl or morpholinyl is optionally replaced by 1 Substituted with one, 2, 3 or 4 C 1-3 alkyl groups.
  • R 1 is absent or R 1 is selected from methyl, piperidinyl, morpholinyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is absent. In some particularly preferred embodiments of the invention, in formula (I), R1 is methyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is piperidinyl. In some particularly preferred embodiments of the invention, in formula (I), R1 is morpholinyl.
  • the number of R 2 is 1 or 2, and R 2 is selected from halogen, C 1-6 alkyl and C 3-6 cycloalkyl, Each of the C 1-6 alkyl and C 3-6 cycloalkyl groups is optionally substituted by 1, 2 or 3 substituents each independently selected from the following groups: halogen, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 1 or 2, and R 2 is selected from halogen, C 1-6 alkyl, wherein the C 1-6 alkyl The group is optionally substituted with 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN , C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • the number of R 2 is 2, and R 2 is selected from fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl , n-butyl, isobutyl. In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 2, and R 2 is selected from fluorine, chlorine, methyl, ethyl, n-propyl, isopropyl. In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 2, and R 2 is selected from fluorine, methyl, and ethyl.
  • the number of R 2 is 2, and R 2 is selected from fluorine and ethyl. In some preferred embodiments of the present invention, in formula (I), the number of R 2 is 2, and R 2 is selected from fluorine and ethyl. In some particularly preferred embodiments of the present invention, in formula (I), there are two R 2 , which are fluorine and ethyl respectively.
  • the compounds used as TRK inhibitor drugs and/or RET inhibitor drugs are selected from:
  • the compounds used are selected from:
  • the "compound represented by formula (G)” or “compound of formula (G)” or “compound of the present invention” mentioned later also covers any optical isomer, geometric form of the compound of formula (G) Isomers, tautomers or mixtures of isomers; the later-mentioned “compounds represented by formula (G')” or “compounds of formula (G')” or “compounds of the present invention” are also covered Any optical isomer, geometric isomer, tautomer or mixture of isomers of the compound of formula (G'); the “compound represented by formula (I)” or “the compound represented by formula (I)” mentioned below )” or “compounds of the invention” also encompass any optical isomer, geometric isomer, tautomer or mixture of isomers of the compound of formula (I).
  • optical isomer means that when a compound has one or more chiral centers, each chiral center can exist in R configuration or S configuration, and the various isomers formed thereby are optical isomers. conformation. Optical isomers include all diastereoisomers, enantiomers, meso, racemates or mixtures thereof. Optical isomers can be separated, for example, by chiral chromatography columns or by chiral synthesis.
  • geometric isomer means that when a double bond is present in a compound, the compound may exist as cis isomer, trans isomer, E isomer and Z isomer. Geometric isomers include cis isomers, trans isomers, E isomers, Z isomers or mixtures thereof.
  • tautomer refers to an isomer resulting from the rapid movement of an atom between two positions in a molecule. Those skilled in the art can understand that tautomers can transform into each other and may reach an equilibrium state and coexist in a certain state.
  • the "compound represented by formula (G)” described herein also encompasses any tautomer of the compound represented by formula (G); the “compound represented by formula (G')” described herein also encompasses formula (G') Any tautomer of the compound; the “compound represented by formula (I)” mentioned herein also encompasses any tautomer of the compound of formula (I).
  • the present invention includes all pharmaceutically acceptable isotopically labeled compounds of the compound of formula (G) in which one or more atoms are replaced by atoms having the same atomic number but a different atomic mass or mass number as atoms typically found in nature. replaced.
  • the present invention includes all pharmaceutically acceptable isotopically labeled compounds of the formula (G') in which one or more atoms are substituted with the same atomic number but a different atomic mass or mass number as atoms typically found in nature. replaced by atoms.
  • the present invention includes all pharmaceutically acceptable isotopically labeled compounds of the compounds of formula (I) in which one or more atoms are replaced by atoms having the same atomic number but a different atomic mass or mass number than atoms normally found in nature. replaced.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen such as 2 H(D) and 3 H(T), isotopes of carbon such as 11 C, 13 C and 14 C, and isotopes of chlorine such as 36 Cl, isotopes of fluorine such as 18 F, isotopes of iodine such as 123 I and 125 I, isotopes of nitrogen such as 13 N and 15 N, isotopes of oxygen such as 15 O, 17 O and 18 O, and sulfur isotope, Such as 35S .
  • isotopes of hydrogen such as 2 H(D) and 3 H(T)
  • isotopes of carbon such as 11 C, 13 C and 14 C
  • isotopes of chlorine such as 36 Cl
  • isotopes of fluorine such as 18 F
  • isotopes of iodine such as 123 I and 125 I
  • isotopes of nitrogen such as 13 N and 15 N
  • the radioactive isotopes deuterium (i.e. 2H ) and carbon-14 (i.e. 14C ) are particularly useful for this purpose given their ease of introduction and convenience of detection means.
  • substitution with heavier isotopes such as deuterium may provide certain therapeutic benefits resulting from greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements).
  • positron emitting isotopes such as 11 C, 18 F, 15 O, and 13 N
  • PET Positron Emission Topography
  • Isotopically labeled compounds of formula (G) can generally be prepared by conventional techniques known to those skilled in the art or by using a suitable isotopically labeled reagent in place of a previously used non-labeled reagent similar to that described in the Examples and preparations attached hereto. method to prepare.
  • Isotopically labeled compounds of formula (G') can generally be prepared by conventional techniques known to those skilled in the art or by using a suitable isotopically labeled reagent in place of a previously used non-labeled reagent similar to that described in the examples and preparations appended hereto. method to prepare.
  • Isotopically labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by using a suitable isotopically labeled reagent in place of a previously used non-labeled reagent, similarly as described in the Examples and preparations attached hereto. method to prepare.
  • the compound of formula (G) may exist in the form of a pharmaceutically acceptable salt, such as an acid addition salt and/or a base addition salt of the compound of formula (G).
  • pharmaceutically acceptable salts as used herein include acid addition salts or base addition salts that may occur in compounds of formula (G).
  • the compound of formula (G') may exist in the form of a pharmaceutically acceptable salt, such as an acid addition salt and/or a base addition salt of the compound of formula (G').
  • pharmaceutically acceptable salts as used herein include acid addition salts or base addition salts that may occur in compounds of formula (G').
  • the compounds of formula (I) may exist in the form of pharmaceutically acceptable salts, such as acid addition salts and/or base addition salts of compounds of formula (I).
  • pharmaceutically acceptable salts such as acid addition salts and/or base addition salts of compounds of formula (I).
  • pharmaceutically acceptable salts include acid addition salts or base addition salts that may occur in compounds of formula (I).
  • Pharmaceutically acceptable salts of the compound of formula (G), the compound of formula (G') and the compound of formula (I) include acid addition salts and base addition salts thereof. Suitable acid addition salts are formed from acids that form nontoxic salts. Examples include, but are not limited to: acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate , camphor sulfonate, citrate, cyclohexylamine sulfonate, ethylene disulfonate, formate, fumarate, grape heptonate, gluconate, glucuronate, hexanate Fluorophosphate, 2-(4-hydroxybenzyl)benzoate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, 2-isethionate, lactate , malate, maleate, malonate, methanesulf
  • Suitable base addition salts are formed from bases that form nontoxic salts. Examples include, but are not limited to: aluminum, arginine, calcium, choline, diethylamine, diethanolamine, glycine, lysine, magnesium, meglumine, ethanolamine, potassium, sodium, tromethamine, and zinc salts. Half-salts of acids and bases may also be formed, such as hemisulfate and hemicalcium salts.
  • suitable salts see Handbook of Pharmaceutical Salts: Properties, Selection and Use by Stahl and Wermuth (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds described herein are known to those skilled in the art.
  • Certain compounds of the invention may exist in unsolvated as well as solvated forms, including hydrated forms.
  • compounds of formula (G), compounds of formula (G') and compounds of formula (I), whether present in solvated or unsolvated form are included within the scope of the present invention.
  • Certain compounds of the present invention may exist in different crystalline or amorphous forms. No matter in which form they exist, compounds of formula (G), compounds of formula (G') and compounds of formula (I) are included in the present invention. within the range.
  • pharmaceutically acceptable means that the corresponding compound, carrier or molecule is suitable for administration to humans.
  • the term refers to a product certified for use in mammals, preferably humans, by a regulatory agency such as CFDA (China), EMEA (Europe), FDA (USA), or any other national regulatory agency.
  • Prodrugs refer to derivatives that are converted into compounds of the present invention through reactions with enzymes, gastric acid, etc. under physiological conditions in vivo, such as through oxidation, reduction, hydrolysis, etc., respectively catalyzed by enzymes.
  • Metal refers to all molecules derived from any compound of the invention in a cell or organism, preferably a human.
  • hydroxy refers to -OH.
  • halogen refers to -F, -Cl, -Br, or -I.
  • cyano refers to -CN.
  • each substituent is selected independently of each other, that is, these substituents may be the same or different.
  • these R 1's may be the same or different.
  • these R 2's may be the same or different.
  • R 9 and R 10 in R 1 and R 2 can be selected independently, that is, R 9 and R 2 in R 1 R 9 in R 1 may be the same or different, R 10 in R 1 and R 2 may be the same or different.
  • R 9 and R 10 among the two R 1s can be selected independently, that is, the first R 9 in R 1 and R 9 in the second R 1 may be the same or different, and R 10 in the first R 1 and R 10 in the second R 1 may be the same or different.
  • substituted means that one or more (preferably 1 to 5, more preferably 1 to 3) hydrogen atoms in a group are independently replaced by a corresponding number of substituents.
  • the term “optionally” or “optionally” means that the event it describes may or may not occur.
  • a group that is “optionally substituted” means that the group may be unsubstituted or substituted.
  • heteroatom represents oxygen (O), nitrogen (N), or S(O) m (where m can be 0, 1, or 2, i.e., a sulfur atom S, or a sulfoxide group SO, or sulfonyl S(O) 2 ).
  • alkyl refers to saturated aliphatic hydrocarbons, including straight and branched chains. In some embodiments, an alkyl group has 1-8, or 1-6, or 1-3 carbon atoms.
  • C 1-8 alkyl refers to a straight or branched chain radical of atoms having 1 to 8 carbon atoms.
  • C 1-8 alkyl includes in its definition the terms “C 1-6 alkyl", “C 1 -C 3 alkyl” and "C 1 -C 4 alkyl”.
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, 3-pentyl, iso Pentyl, neopentyl, (R)-2-methylbutyl, (S)-2-methylbutyl, 3-methylbutyl, 2,3-dimethylpropyl, 2,3- Dimethylbutyl, hexyl, etc.
  • Alkyl groups may be optionally substituted with one or more (eg, 1 to 5) suitable substituents.
  • alkenyl refers to an aliphatic hydrocarbon having at least one carbon-carbon double bond, including straight and branched chains having at least one carbon-carbon double bond.
  • alkenyl groups have 2-8 carbon atoms, 2-6 carbon atoms, 3-6 carbon atoms, or 2-4 carbon atoms.
  • C 2-8 alkenyl refers to a straight or branched chain unsaturated radical of 2 to 8 carbon atoms (having at least one carbon-carbon double bond). The double bond may or may not be the point of attachment to another group.
  • Alkenyl groups include, but are not limited to, vinyl, 1-propenyl, 2-propenyl, 2-methyl-2-propenyl, butenyl, pentenyl, 3-hexenyl, and the like. Alkenyl groups may be optionally substituted with one or more (eg, 1 to 5) suitable substituents. When a compound of formula (I) contains an alkenyl group, the alkenyl group may be present in pure E form, pure Z form, or any mixture thereof.
  • alkynyl refers to an aliphatic hydrocarbon having at least one carbon-carbon triple bond, including straight and branched chains having at least one carbon-carbon triple bond.
  • an alkynyl group has 2-8 carbon atoms, 2-6 carbon atoms, 3-6 carbon atoms, or 2-4 carbon atoms.
  • C 2-8 alkynyl refers to a straight or branched chain unsaturated radical of 2 to 8 carbon atoms (having at least one carbon-carbon triple bond). The triple bond may or may not be the point of attachment to another group.
  • Alkynyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, 2-methyl-2-propynyl, butynyl, pentynyl, 3-hexynyl, and the like. Alkynyl groups may be optionally substituted with one or more (eg, 1 to 5) suitable substituents.
  • C 3-7 cycloalkyl refers to a cycloalkyl group having 3 to 7 ring-forming carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloalkyl Hexenyl, cycloheptyl. Cycloalkyl groups may be optionally substituted with one or more suitable substituents.
  • n-membered heterocycloalkyl refers to a cycloalkyl group having m ring-forming carbon atoms and (n-m) ring-forming heteroatoms selected from the group consisting of O, S, and N.
  • 3-7 membered heterocycloalkyl groups include, but are not limited to, oxetane, thietane, azetidine, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, tetrahydropyran, tetrahydrothiopyran , piperidine, morpholine, piperazine, oxepane, thiepane, azepane.
  • Heterocycloalkyl groups may be optionally substituted with one or more suitable substituents.
  • C 5-7 aryl refers to an aryl group having an aromatic ring containing 5 to 7 carbon atoms, preferably phenyl.
  • n-membered heteroaryl refers to a heteroaryl group having m aromatic ring-forming carbon atoms and (n-m) aromatic ring-forming heteroatoms selected from O, S and N.
  • 5-7 membered heteroaryl groups include, but are not limited to, pyrazine, pyrazole, pyrrole, furan, thiophene, thiazole, and pyridine.
  • Heteroaryl groups may be optionally substituted with one or more suitable substituents.
  • C 7-11 bicyclic aryl refers to a bicyclic aryl group having 7-11 carbon atoms, such as naphthalene, indene, and the like. Bicyclic aryl groups may be optionally substituted with one or more suitable substituents.
  • n-membered bicyclic heteroaryl refers to a bicyclic heteroaryl group having m carbon atoms forming an aromatic bicyclic ring and (n-m) heteroatoms forming an aromatic bicyclic ring, the heteroatoms being selected from From O, S and N.
  • 7-11 membered bicyclic heteroaryl groups include, but are not limited to, quinoline, isoquinoline, benzothiazole, etc.
  • Bicyclic heteroaryl groups may be optionally substituted with one or more suitable substituents.
  • 11-15 membered tricyclyl includes, but is not limited to, acridine and the like.
  • the 11-15 membered tricyclic group may be optionally substituted with one or more suitable substituents.
  • haloalkyl refers to an alkyl group having one or more halogen substituents (up to a perhaloalkyl group, i.e., each hydrogen atom of the alkyl group is replaced by a halogen atom) .
  • C 1-6 haloalkyl refers to a C 1-6 alkyl group having one or more halogen substituents (up to a perhaloalkyl group, i.e., each hydrogen atom of the alkyl group is replaced by a halogen atoms replaced).
  • C 1-4 haloalkyl refers to a C 1-4 alkyl group having one or more halogen substituents (up to a perhaloalkyl group, i.e., each hydrogen atom of the alkyl group is substituted by a halogen atom);
  • C 1-3 haloalkyl refers to a C 1-3 alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., each of the alkyl groups hydrogen atoms are all replaced by halogen atoms);
  • C 1-2 haloalkyl refers to a C 1-2 alkyl group (i.e., methyl or ethyl) having one or more halogen substituents (up to Perhaloalkyl, i.e., each hydrogen atom of the alkyl group is replaced by a halogen atom).
  • C 1 haloalkyl refers to a methyl group having 1, 2 or 3 halogen substituents.
  • haloalkyl groups include: CF3 , C2F5 , CHF2 , CH2F , CH2CF3 , CH2Cl , and the like.
  • alkoxy refers to an alkyl group with a single bond attached to an oxygen atom. The point of attachment of the alkoxy group to the molecule is through the oxygen atom. Alkoxy groups may be described as alkyl-O-.
  • C 1-6 alkoxy refers to a straight or branched chain alkoxy group containing 1 to 6 carbon atoms.
  • C 1-6 alkoxy includes in its definition the term "C 1-3 alkoxy”.
  • Alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, hexyloxy, and the like. Alkoxy groups may be optionally substituted with one or more suitable substituents.
  • the numerical range related to the number of substituents, the number of carbon atoms, and the number of ring atoms represents an enumeration of all integers within the range, and the range is only used as a simplified expression. For example:
  • 3-12 membered ring means 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 membered ring;
  • 3-14 membered ring means 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 membered ring;
  • 3-8 membered ring means 3, 4, 5, 6, 7 or 8 membered ring
  • “1-12 carbon atoms” or C 1-12 ” means 1 (C 1 ), 2 (C 2 ), 3 (C 3 ), 4 (C 4 ), 5 (C 5 ), 6 (C 6 ), 7 (C 7 ), 8 (C 8 ), 9 (C 9 ), 10 (C 10 ), 11 (C 11 ), or 12 carbon atoms (C 12 ) ;
  • 1-6 carbon atoms or “C 1-6” means 1 (C 1 ), 2 (C 2 ), 3 (C 3 ), 4 (C 4 ), 5 (C 5 ) or 6 carbon atoms (C 6 );
  • 1-4 carbon atoms or “C 1-4 ” means 1 (C 1 ), 2 (C 2 ), 3 (C 3 ) or 4 carbon atoms (C 4 );
  • C 3-8 means 3 (C 3 ), 4 (C 4 ), 5 (C 5 ), 6 (C 6 ), 7 (C 7 ) or 8 carbon atoms (C 8 ) ;
  • 3-8 ring atoms means 3, 4, 5, 6, 7 or 8 ring atoms.
  • the numerical range related to the number of substituents, the number of carbon atoms, and the number of ring atoms also covers any subrange thereof, and each subrange is also deemed to be disclosed herein.
  • the compound of formula (G), compound of formula (G') or compound of formula (I) used in the present invention is a known compound and can be synthesized by a variety of methods familiar to those skilled in the field of organic synthesis, such as Chinese invention patents The method disclosed in CN111606908B.
  • the compound of formula (G), the compound of formula (G') or the compound of formula (I) used in the present invention can be used as a "TRK inhibitor and/or RET inhibitor", which means that they can either be TRK inhibitors can also be TRK inhibitors or TRK/RET dual inhibitors.
  • TRK inhibitor and/or RET inhibitor which means that they can either be TRK inhibitors can also be TRK inhibitors or TRK/RET dual inhibitors.
  • these compounds can also be used as JAK inhibitors.
  • the compound of formula (G), the compound of formula (G') or the compound of formula (I) used in the present invention can be used as JAK/TRK/RET multiple inhibitors work, that is, they can be: JAK/TRK dual inhibitors, JAK/RET dual inhibitors, TRK/RET dual inhibitors, or JAK/TRK/RET triple inhibitors. Because multiple inhibitors inhibit multiple disease targets simultaneously, the compounds used in the present invention are more effective than traditional single-target inhibitors in treating certain diseases (as shown in the examples below). Therefore, the compound of formula (G), the compound of formula (G') or the compound of formula (I) used in the present invention preferably functions as a JAK/TRK/RET multiple inhibitor.
  • the compound of formula (G), the compound of formula (G') or the compound of formula (I) used in the present invention functions as a JAK/TRK dual inhibitor; among them, some particularly preferred compounds They can function as Pan-JAK/Pan-TRK dual inhibitors (i.e., simultaneously inhibit all members of the JAK kinase family and the TRK kinase family), thus making them useful in the treatment of autoimmune diseases, skin diseases such as pruritus, and diseases such as diabetic foot. There is an extremely outstanding performance in it.
  • the present invention provides a TRK inhibitor pharmaceutical composition, which contains a compound of formula (G), a compound of formula (G') or a compound of formula (I) or an isotope thereof as described above Labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite, and one or Various pharmaceutically acceptable carriers, adjuvants or excipients.
  • a TRK inhibitor pharmaceutical composition which contains a compound of formula (G), a compound of formula (G') or a compound of formula (I) or an isotope thereof as described above Labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite, and one or Various pharmaceutically acceptable carriers, adjuvants or excipients.
  • the present invention provides a RET inhibitor pharmaceutical composition, which contains a compound of formula (G), a compound of formula (G') or a compound of formula (I) or an isotope thereof as described above Labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite, and one or Various pharmaceutically acceptable carriers, adjuvants or excipients.
  • a compound of formula (G) a compound of formula (G') or a compound of formula (I) or an isotope thereof as described above Labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite, and one or Various pharmaceutically acceptable carriers, adjuvants or excipients.
  • the pharmaceutical composition of the present invention can be formulated as needed for oral, topical (including but not limited to topical application, spraying, etc.), parenteral (including subcutaneous, intramuscular, cortical and intravenous) administration, bronchial administration or nasal administration. dosage form.
  • the pharmaceutical composition of the present invention is formulated into a dosage form (preparation) suitable for oral or external use. More preferably, the pharmaceutical composition of the present invention is formulated into a dosage form (preparation) suitable for oral administration.
  • the preparation may be in the form of tablets, as a powder or granules in hard gel capsules, or in the form of lozenges or lozenges.
  • Solid carriers may include conventional excipients such as binders, fillers, tableting lubricants, disintegrating agents, wetting agents, and the like.
  • the tablets may be film-coated if desired by conventional techniques.
  • a liquid carrier the preparations may be in the form of a syrup, lotion, ointment, soft gel capsule, sterile vehicle for injection, aqueous or non-aqueous liquid suspension, or may be formulated with water or other appropriate Carrier-restored dry product.
  • Liquid preparations may contain conventional additives such as suspending agents, emulsifiers, wetting agents, non-aqueous vehicles (including edible oils), preservatives, and flavoring and/or coloring agents.
  • the carrier will consist at least in large part of sterile water, although saline solutions, dextrose solutions, and the like may also be used.
  • injectable suspensions may also be used, in which case conventional suspending agents may be used.
  • Conventional preservatives, buffering reagents, etc. may also be added to parenteral dosage forms.
  • Pharmaceutical compositions are prepared by conventional techniques suitable for the desired formulation containing an appropriate amount of the active ingredient (i.e. a compound of formula (G), a compound of formula (G') or a compound of formula (I) of the invention).
  • compositions suitable for parenteral injection may include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions and sterile powders for sterile injectable solutions or dispersions.
  • suitable aqueous and non-aqueous carriers, diluents, solvents include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, etc.), suitable mixtures thereof, vegetable oils (e.g., olive oil), and injectables Organic esters (eg, ethyl oleate).
  • compositions may also contain various excipients such as preservatives, wetting agents, emulsifying agents and dispersing agents. Inhibition of the action of microorganisms can be ensured by various antibacterial and antifungal agents (for example, parabens, chlorobutanol, phenol, sorbic acid, etc.). Isotonic agents such as sugar, sodium chloride, etc. may also be included. Prolonged absorption of injectable pharmaceutical dosage forms can be brought about by the use of agents that delay absorption (eg, aluminum monostearate and gels).
  • agents that delay absorption eg, aluminum monostearate and gels.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one inert excipient (or carrier) (for example, sodium citrate or dicalcium phosphate), which may also include: (a) fillers or admixtures (for example, starch, lactose, sucrose, glucose, mannitol and silicic acid); (b) binders (e.g.
  • ком ⁇ онентs e.g., glycerol
  • Disintegrants e.g., agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain synthetic silicates, sodium carbonate
  • Solutions Retarder for example, paraffin
  • Absorption enhancer for example, quaternary ammonium compound
  • Wetting agent for example, cetyl alcohol and glycerol monostearate
  • Adsorption agents e.g., kaolin and bentonite
  • lubricants e.g., talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate
  • Solid compositions of a similar type may also be used as fillers in soft-filled and hard-filled gel capsules using excipients such as lactose and high molecular weight polyethylene glycols.
  • Solid dosage forms eg, tablets, dragees, capsules, pills, and granules
  • coatings and shells eg, enteric coatings and others known in the art. They may contain opacifying agents, and they may be compositions which release the active compound or active compounds in a certain part of the intestinal tract in a delayed manner. Examples of useful embedding compositions are polymeric substances and waxes.
  • the active ingredient can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, dispersions, syrups, and elixirs.
  • liquid dosage forms may contain inert diluents (e.g., water or other solvents), solubilizers, and emulsifiers commonly used in the art (e.g., ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzene Methanol, benzyl benzoate, propylene glycol, 1,3-butanediol, dimethylformamide), oil (specifically, cottonseed oil, peanut oil, corn oil, olive oil, castor oil, sesame oil), propylene glycol Triol, tetrahydrofuranol, fatty acid esters of polyethylene glycol and sorbitan or mixtures of these substances, etc.
  • inert diluents e.g., water or other solvents
  • solubilizers emul
  • compositions may also include, for example, wetting agents, emulsifying and suspending agents, perfuming, flavoring and perfuming agents.
  • Suspensions may contain, in addition to the active compound, suspending agents, for example ethoxylated isostearyl alcohols, polyoxyethylene sorbitol, sorbitan esters, microcrystalline fibers, aluminum metahydroxide, bentonite, agar-agar and Gum tragacanth or mixtures of these substances, etc.
  • suspending agents for example ethoxylated isostearyl alcohols, polyoxyethylene sorbitol, sorbitan esters, microcrystalline fibers, aluminum metahydroxide, bentonite, agar-agar and Gum tragacanth or mixtures of these substances, etc.
  • Dosage forms for topical administration of the compounds of this invention include ointments, powders, sprays and inhalants.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any desired preservatives, buffers or propellants.
  • Ophthalmic formulations, ophthalmic ointments, powders and solutions are also included within the scope of the present invention.
  • the compounds of the present invention may be formulated for external use in the form of water-in-oil (W/O) or oil-in-water (O/W) emulsions, multiple emulsions, such as water-in-oil-in-water (W/O/W) or oil-in-oil emulsions.
  • W/O water-in-oil
  • O/W oil-in-water
  • Topical dosage forms of the compounds of the present invention may contain additives and formulation auxiliaries such as emulsifiers, thickeners, gelling agents, water fixatives, spreading agents, stabilizers, dyes, perfumes and preservatives.
  • emulsifiers include stearic acid, triethanolamine and PEG-40-stearate.
  • Suitable thickeners include glyceryl monostearate and PEG600.
  • Suitable preservatives include propylparaben and chlorocresol.
  • Suitable spreading agents include dimethicone and polydimethylcyclosiloxane.
  • Suitable water fixatives include polyethylene glycol, preferably polyethylene glycol 600.
  • the topical dosage forms of the compounds of the present invention may include ointments, lotions, gels, emulsions, microemulsions, sprays, skin patches, etc., which can be applied topically to treat atopic dermatitis, skin side effects caused by EGFR inhibitors, Acne, eczema, psoriasis, scleroderma, itching, vitiligo, hair loss and other skin diseases.
  • the topical dosage form of the compound of the present invention is an ointment, which can be applied topically to treat atopic dermatitis, skin side effects caused by EGFR inhibitors, acne, eczema, psoriasis, scleroderma, pruritus, vitiligo, Skin diseases such as hair loss.
  • the amount of the compound of formula (G), the compound of formula (G') or the compound of formula (I) in pharmaceutical compositions and dosage forms can be appropriately determined by those skilled in the art as needed, for example, the compound of formula (G), the compound of formula ( The compound of G') or the compound of formula (I) may be present in a pharmaceutical composition or dosage form in a therapeutically effective amount.
  • the present invention provides a method of treating a disease or condition associated with TRK or RET, the method comprising adding a therapeutically effective amount of a compound of formula (G) as described above, formula (G' ) compound or compound of formula (I) or isotopically labeled compound thereof, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its
  • the prodrug, or metabolite thereof, or a composition as described above is administered to a patient in need thereof.
  • the patient is preferably a mammal, more preferably a human patient.
  • the route of administration may be oral, topical (including but not limited to topical application, spraying, etc.), parenteral (including subcutaneous, intramuscular, cortical and intravenous) administration, bronchial administration or nasal administration, etc.
  • administration by oral administration or external application is preferable; administration by oral administration is more preferable.
  • TRK or RET-related diseases or conditions include, but are not limited to:
  • Autoimmune diseases or conditions including autoimmune conditions of a single organ or single cell type, such as Hashimoto's thyroiditis, autoimmune hemolytic anemia, autoimmune atrophic gastritis with pernicious anemia, autoimmune encephalomyelitis, autoimmune Orchitis, Goodpasture's disease, autoimmune thrombocytopenia, sympathetic ophthalmia, myasthenia gravis, Graves' disease, primary biliary cirrhosis, chronic invasive hepatitis, ulcerative colitis and membranous glomerulopathies, those involving systemic autoimmune disorders (e.g., systemic lupus erythematosus, rheumatoid arthritis, Sjogren's syndrome, Reiter's syndrome, polymyositis-dermatomyositis, systemic sclerosis, polyarteritis nodosa, multiple sclerosis, and bullous pemphigoid) and other O-cell (humoral)
  • Cancer or tumors including gastrointestinal/gastrointestinal cancer, colorectal cancer, liver cancer, skin cancer (including mastocytoma and squamous cell carcinoma), breast and breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia (including Acute myeloid leukemia and chronic myeloid leukemia), kidney cancer, lung cancer, myeloma, bone cancer, bladder cancer, brain cancer, melanoma (including oral and metastatic melanoma), Kaposi's sarcoma, myeloma (including multiple myeloma), myeloproliferative disorders, proliferative diabetic retinopathy, or angiogenesis-related disorders (including solid tumors);
  • Diabetes including type 1 diabetes or complications of diabetes
  • Eye diseases, disorders, or conditions including autoimmune diseases of the eye, keratoconjunctivitis, vernal conjunctivitis, uveitis (including uveitis associated with Behcet's disease and lenticular uveitis), keratitis, herpetic Keratitis, keratoconus, corneal epithelial dystrophy, leukoplakia, ocular pemphigus, Moren's ulcer, scleritis, Grave's ophthalmopathy, Vogt-Koyanagi-Harada syndrome, keratoconjunctivitis sicca (dry eye) , blisters, iridocyclitis, sarcoidosis, endocrine eye disease, sympathetic ophthalmia, allergic conjunctivitis or ocular neovascularization;
  • Intestinal inflammation allergies or conditions, including Crohn's disease and/or ulcerative colitis, inflammatory bowel disease, celiac disease, proctitis, eosinophilic gastroenteritis or mastocytosis;
  • Neurodegenerative diseases including motor neuron disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, neurodegenerative diseases caused by cerebral ischemia or traumatic injury, stroke, glutamate neurotoxicity, or Hypoxia; ischemia/reperfusion injury from stroke, myocardial ischemia, renal ischemia, heart attack, cardiac hypertrophy, atherosclerosis and arteriosclerosis, organ hypoxia, or platelet aggregation;
  • Skin diseases, conditions or disorders including atopic dermatitis, skin side effects caused by EGFR inhibitors, acne, eczema, psoriasis, scleroderma, pruritus or other itchy conditions, vitiligo, and hair loss (alopecia areata);
  • Allergies including mammalian allergic dermatitis (including equine allergic diseases such as bite allergy), summer eczema, Culex mosquito bite syndrome (sweet itch), emphysema, inflammatory airway disease, recurrent Airway obstruction, airway hyperresponsiveness, or chronic obstructive pulmonary disease;
  • mammalian allergic dermatitis including equine allergic diseases such as bite allergy), summer eczema, Culex mosquito bite syndrome (sweet itch), emphysema, inflammatory airway disease, recurrent Airway obstruction, airway hyperresponsiveness, or chronic obstructive pulmonary disease;
  • Asthma and other obstructive airway diseases including chronic or refractory asthma, advanced asthma, bronchitis, bronchial asthma, allergic asthma, intrinsic asthma, exogenous asthma or dust asthma;
  • Transplant rejection including islet transplant rejection, bone marrow transplant rejection, graft-versus-host disease, organ and cell transplant rejection (e.g., bone marrow, cartilage, cornea, heart, intervertebral disc, islet, kidney, limb, liver, lung, muscle, myoblast , nerve, pancreas, skin, small intestine or trachea) or xenograft.
  • organ and cell transplant rejection e.g., bone marrow, cartilage, cornea, heart, intervertebral disc, islet, kidney, limb, liver, lung, muscle, myoblast , nerve, pancreas, skin, small intestine or trachea
  • the compounds of formula (G), compounds of formula (G') or compounds of formula (I) described herein are particularly suitable for the treatment of diseases or conditions selected from the following: arthritis, autoimmune diseases or conditions, Cancer or tumors, diabetes, slow wound healing due to diabetes, eye disease, disorder or condition, intestinal inflammation, allergies or conditions, neurodegenerative diseases, skin diseases, conditions or conditions, allergies, asthma and other obstructive airway diseases , transplant rejection.
  • the compound of formula (G), the compound of formula (G') or the compound of formula (I) as described above is most preferably used to treat pruritus, psoriasis, atopic dermatitis, and acne.
  • vitiligo, alopecia areata, asthma, rhinitis, hemorrhoids, cervicitis, pneumonia and other diseases.
  • these compounds can also be used to treat allergic conjunctivitis, cancer (tumor), slow wound healing caused by diabetes, diabetic foot and other diseases.
  • a compound of formula (G), a compound of formula (G') or a compound of formula (I) as described above can be used as a JAK/pan-TRK inhibitor, especially a pan-JAK/pan-TRK inhibitor.
  • TRK inhibitors or pan-JAK/pan-TRK/RET inhibitors can be used as a JAK/pan-TRK inhibitor, especially a pan-JAK/pan-TRK inhibitor.
  • a compound of formula (G), a compound of formula (G') or a compound of formula (I) as described above can be used to treat autoimmune diseases, chronic wound healing, and complications of diabetes.
  • Diseases e.g. selected from: arthritis, intestinal inflammation, allergies or conditions, neurodegenerative diseases, skin diseases, conditions or conditions, allergies, asthma and other obstructive airway diseases, transplant rejection, pressure ulcers, wound healing due to diabetes Slowness, eye disease, disorder or condition, etc.
  • a compound of formula (G), a compound of formula (G') or a compound of formula (I) as described above can be used to treat pruritus, psoriasis, atopy Dermatitis, skin side effects caused by EGFR inhibitors, acne, vitiligo, alopecia areata, asthma, rhinitis, hemorrhoids, cervicitis, pneumonia, bedsores, slow wound healing caused by diabetes, diabetic foot, diabetic retinopathy, etc.
  • the present invention provides a formula as described above that is used as a TRK inhibitor drug and/or a RET inhibitor drug, or as a drug for treating diseases or conditions related to TRK or RET.
  • the definitions and preferences of the substituents in the compound of formula (G), the compound of formula (G') or the compound of formula (I) are as described above, and the diseases or conditions related to TRK or RET are also as described above. .
  • the present invention provides the following embodiments:
  • X 1 is N or CR 14 ;
  • X 2 is N or CR 15 ;
  • X 3 is N or CR 16 ;
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -SR 12 , -OR 12 , -CN, halogen, -NO 2 , -SF 5 , -SC 1-4 Alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic group, C 5-11 bicyclic alkyl, 5-11 membered bicyclic heteroalkyl, and R 13 is replaced by 0, 1, 2, 3 or 4 R 1 Substituted; wherein R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkyne base, C 3-7 cycloalkyl, C 3-7
  • R 14 , R 15 , R 16 are selected from H, -OH, -SH, -CN, halogen, -NO 2 , C 1-6 alkyl.
  • R 14 , R 15 , R 16 are selected from H, -OH, C 1-6 alkyl .
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN , Halogen, -NO 2 , -SF 5 , -SC 1-4 alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic, C 5-11 bicycloalkyl, 5-11 membered bicyclic heteroalkyl, And R 13 is substituted by 0, 1, 2, 3 or 4 R 1 ; wherein R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 Cycloalkyl, C 3-7 heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl,
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN , Halogen, -NO 2 , -SF 5 , -SC 1-4 alkyl, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl, 11-15 membered tricyclic group, and R 13 is replaced by 0, 1, 2, 3 or 4 R1 replaced.
  • R 13 is H, -N(R 17 )(R 18 ), C 1-6 alkoxy, C 1-6 alkyl or C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, and R 13 is replaced by 0, 1, 2, 3 or 4 R 1 replaced.
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy, C 1-6 alkyl or C 3- 7- cycloalkyl, 4-6-membered heterocycloalkyl, phenyl, 5-6-membered heteroaryl, and R 13 is substituted by 0, 1, 2 or 3 R 1s .
  • R 17 and R 18 are each independently selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, C 3-7 hetero Cycloalkyl, optionally substituted by one or more -OH, -CN, -SH, halogen, -NO2 , -SF5 .
  • R 13 is -N(R 17 )(R 18 ), C 1-6 alkoxy, -OH, -SH, -CN, halogen, -NO 2 , -SF 5 or -SC 1-4 alkyl, and R 13 is replaced by 0, 1, 2 , 3 or 4 R 1 ; wherein R 17 and R 18 are each independent is selected from H, C 1-6 alkyl, C 1-6 alkoxy, C 3-7 cycloalkyl, C 3-7 heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl group, and optionally substituted by one or more -OH, -CN, -SH, halogen, -NO 2 , -SF 5 ; or R 17 , R 18 and the N atoms connected to them together form 3-14 yuan ring.
  • R2 is selected from H, halogen, -OH, -NO2 , -CN, - SF 5 , -SH, -SC 1-4 alkyl, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkynyl , C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, wherein -SC 1-4 alkyl, C 1-6 alkyl, C 3-7 cycloalkyl, 4-10 membered heterocycle
  • Each alkyl group is optionally substituted with 1, 2 or 3 substituents each independently selected from the group consisting of: halogen, -OH, -NH2 , -NH( CH3 ), -N( CH3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy
  • R2 is selected from halogen, C1-6 alkyl and C3-6 cycloalkyl group, wherein each of the C 1-6 alkyl and C 3-6 cycloalkyl groups is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, -NH 2. -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • R 13 is substituted by 0 or 1 R 1 and R 1 is selected from halogen, -OH, -CN, C 1-6 alkyl, 5-7 membered heterocycloalkyl, C 3-7 cycloalkyl, wherein the C 1-6 alkyl is optionally substituted by 1, 2 or 3 R 3 , and wherein the 5-7 membered Heterocycloalkyl, C 3-7 cycloalkyl is optionally substituted by 1, 2, 3 or 4 C 1-3 alkyl.
  • X is CH or N
  • Ring A is C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic heteroaryl Base, 11-15 membered tricyclic group;
  • R 1 is 0, 1, 2, 3 or 4, and R 1 is selected from H, halogen, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1- 8 alkoxy, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7-11 membered bicyclic Heteroaryl, wherein the C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkoxy groups are optionally replaced by 1, 2, 3 or 4 R 3 is substituted, and wherein the C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered heteroaryl, C 7-11 bicyclic aryl, 7- The 11-membered bicyclic heteroaryl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 3 is selected from halogen, cyano, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -N(R 5 )(R 6 ), -CON(R 7 )(R 8 ) or 3- 7-membered heterocycloalkyl, wherein the 3-7-membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 4 is selected from halogen, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -NH 2 , -NHCH 3 or -N(CH 3 ) 2 ;
  • R 5 , R 6 , R 7 and R 8 are each independently hydrogen or C 1-4 alkyl
  • R 9 is selected from H, C 1-4 alkyl, C 1-4 haloalkyl or C 3-7 cycloalkyl;
  • R 10 is H or selected from the following group: C 1-4 alkyl, C 1-4 haloalkyl, C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, C 6-10 aryl, 5 -10-membered heteroaryl, (C 3-7 cycloalkyl)-C 1-4 alkyl-, (4-10-membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl base)-C 1-4 alkyl- and (5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally replaced by 1, 2, 3 or 4 each Substituted with substituents independently selected from the following groups: -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 3-7 cycloalkyl group, C 1-4 hydroxyalkyl group, -SC 1-4 alkyl group, -C
  • R 11 is selected from H, C 1-4 alkyl and C 3-7 cycloalkyl
  • R 12 is selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4- to 14-membered heterocycloalkyl, C 6-10 aryl, 5-10 membered heteroaryl, (C 3-7 Cycloalkyl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl)-C 1-4 alkyl- and ( 5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -CF 3 , -CN, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , oxo, -SC 1-4 alkyl, C 1-4 alkyl, C 1- 4 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloal
  • Ring A is C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 5-7 aryl, 5-7 membered hetero Aryl.
  • Ring A is 5-6 membered heteroaryl or phenyl.
  • R 1 is selected from C 1-6 alkyl, 5-7 membered heterocycloalkyl, wherein The C 1-6 alkyl group is optionally substituted by 1 or 2 R 3 , and the 5-7 membered heterocycloalkyl group is optionally substituted by 1, 2, 3 or 4 C 1-3 Alkyl substitution.
  • R2 is selected from halogen, C1-6 alkyl and C3-6 cycloalkyl, wherein Each of the C 1-6 alkyl and C 3-6 cycloalkyl groups is optionally substituted by 1, 2 or 3 substituents each independently selected from the following groups: halogen, -OH, -NH 2 , - NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • X is CH
  • Ring A is a 5-7 membered heteroaryl group, C 5-7 aryl group;
  • R 1 is 0, 1, 2, 3 or 4, and R 1 is selected from C 1-8 alkyl, 3-7 membered heterocycloalkyl, wherein the C 1-8 alkyl is optionally Substituted by 1, 2, 3 or 4 R 3 , and wherein the 3-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 3 is selected from halogen, cyano, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -N(R 5 )(R 6 ), -CON(R 7 )(R 8 ) or 3- 7-membered heterocycloalkyl, wherein the 3-7-membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 R 4 ;
  • R 4 is selected from halogen, C 1-3 alkyl, hydroxyl, C 1-6 alkoxy, -NH 2 , -NHCH 3 or -N(CH 3 ) 2 ;
  • R 5 , R 6 , R 7 and R 8 are each independently hydrogen or C 1-4 alkyl
  • R 9 is selected from H, C 1-4 alkyl, C 1-4 haloalkyl or C 3-7 cycloalkyl;
  • R 10 is H or selected from the following group: C 1-4 alkyl, C 1-4 haloalkyl, C 3-7 cycloalkyl, 4-10 membered heterocycloalkyl, C 6-10 aryl, 5 -10-membered heteroaryl, (C 3-7 cycloalkyl)-C 1-4 alkyl-, (4-10-membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl base)-C 1-4 alkyl- and (5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally replaced by 1, 2, 3 or 4 each Substituted with substituents independently selected from the following groups: -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 3-7 cycloalkyl group, C 1-4 hydroxyalkyl group, -SC 1-4 alkyl group, -C
  • R 11 is selected from H, C 1-4 alkyl and C 3-7 cycloalkyl
  • R 12 is selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4- to 14-membered heterocycloalkyl, C 6-10 aryl, 5-10 membered heteroaryl, (C 3-7 Cycloalkyl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, (C 6-10 aryl)-C 1-4 alkyl- and ( 5-10 membered heteroaryl)-C 1-4 alkyl-, wherein each option within this group is optionally substituted by 1, 2 or 3 substituents each independently selected from the following group: halogen, -CF 3 , -CN, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , oxo, -SC 1-4 alkyl, C 1-4 alkyl, C 1- 4 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloal
  • R 1 is selected from C 1-6 alkyl, 5-7 membered heterocycloalkyl, wherein said C 1-6 alkyl radical is optionally substituted by 1 or 2 R3 , and wherein the 5-7 membered heterocycloalkyl is optionally substituted by 1, 2, 3 or 4 C1-3 alkyl.
  • R 2 is selected from halogen, C 1-6 alkyl and C 3-6 cycloalkyl, wherein said C 1-6 Alkyl and C 3-6 cycloalkyl are each optionally substituted with 1, 2 or 3 substituents each independently selected from the following group: halogen, -OH, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy.
  • isotopically labeled compounds optical isomers, geometric isomers, tautomers or isomer mixtures, or pharmaceutically acceptable salts, prodrugs, or metabolites of any of the above compounds.
  • isotopically labeled compounds optical isomers, geometric isomers, tautomers or isomer mixtures, or pharmaceutically acceptable salts, prodrugs, or metabolites of any of the above compounds.
  • JAK/TRK dual inhibitor preferably as a pan-JAK/pan-TRK dual inhibitor
  • JAK/TRK /RET multiple inhibitor preferably used as pan-JAK/pan-TRK/RET multiple inhibitor
  • a TRK inhibitor and/or RET inhibitor pharmaceutical composition which contains a compound as defined in any one of embodiments 1-35 or an isotope-labeled compound thereof, or an optical isomer or geometric isomer thereof isomers, tautomers or isomer mixtures, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof, and one or more pharmaceutically acceptable carriers, adjuvants or excipients form agent.
  • composition of embodiment 37 formulated as an oral dosage form or a topical dosage form suitable for topical administration.
  • composition according to embodiment 37 which is formulated as a JAK/TRK dual inhibitor drug (preferably used as a pan-JAK/pan-TRK dual inhibitor drug) or a JAK/TRK/RET multiple inhibitor Drugs (preferably formulated as pan-JAK/pan-TRK/RET multiple inhibitor drugs).
  • the disease or disorder associated with TRK and/or RET is selected from the group consisting of arthritis, autoimmune diseases or disorders, cancer or tumors, diabetes and diabetic complications, (diabetes-induced ) Slow healing of wounds, eye diseases, conditions or conditions, intestinal inflammation, allergies or conditions, neurodegenerative diseases, skin diseases, conditions or conditions, allergies, asthma and other obstructive airway diseases, transplant rejection.
  • the disease or disorder associated with TRK and/or RET is selected from the group consisting of pruritus, psoriasis, atopic dermatitis, skin side effects caused by EGFR inhibitors, acne, vitiligo, Alopecia areata, asthma, rhinitis, hemorrhoids, cervicitis, pneumonia, slow wound healing caused by diabetes, diabetic foot, diabetic retinopathy, cancer (tumor), and bedsores.
  • Figure 1 shows the IL-5 Elisa detection results of lung wash fluid of the ovalbumin-induced mouse asthma model. Among them, data are expressed as mean ⁇ SEM (*p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001vs. vehicle control group, One-way ANOVA, Bonferroni's Multiple Comparison Test).
  • Figure 2 shows the results of counting inflammatory cells in lung wash fluid of the ovalbumin-induced mouse asthma model.
  • the data are expressed as mean ⁇ SEM (*p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001 vs. vehicle control group, One-way ANOVA, Bonferroni's Multiple Comparison Test).
  • Figure 3 shows the serum IL-5 Elisa detection results of the ovalbumin-induced mouse asthma model. The data are expressed as mean ⁇ SEM (*p ⁇ 0.05;**p ⁇ 0.01;***p ⁇ 0.001 vs. vehicle control group, One-way ANOVA, Bonferroni's Multiple Comparison Test).
  • Figure 4 shows representative pictures of pathological staining of lung tissue in an ovalbumin-induced mouse asthma model.
  • Figure 5 shows the pathological scoring results of lung tissue in the ovalbumin-induced mouse asthma model.
  • the data are expressed as mean ⁇ SEM (*p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001 vs. vehicle control group, One-way ANOVA, Bonferroni's Multiple Comparison Test).
  • Figure 6 shows the drug intervention scoring results of male mice in the allergic rhinitis mouse model (*, p ⁇ 0.05).
  • Figure 7 shows the drug intervention scoring results of female mice in the allergic rhinitis mouse model (*, p ⁇ 0.05).
  • Figure 8 shows the results of HE staining of sections of mice of different genders in the allergic rhinitis mouse model.
  • Figure 9 shows the detection of IL-4 content in the serum of male mice in the allergic rhinitis mouse model (*, p ⁇ 0.05).
  • Figure 10 shows the detection of IL-4 content in the serum of female mice in the allergic rhinitis mouse model (*, p ⁇ 0.05).
  • Figure 11 shows the HE staining results of lung tissue of each group in the chronic obstructive pulmonary disease rat animal model.
  • Figure 12 shows changes in IL- ⁇ expression in serum and lavage fluid in the chronic obstructive pulmonary disease rat animal model.
  • Figure 13 shows the effect of external hemorrhoid modeling in rats and the effect of positive control drugs.
  • Figure 14 shows the HE staining scores of pathological sections of each group in the rat external hemorrhoid model.
  • Figure 15 shows the detection of TNF- ⁇ expression levels in samples from each group in the rat external hemorrhoid model.
  • Figure 16 shows the effect of drugs on vulvar inflammation in rats with phenol-induced cervicitis.
  • Figure 17 shows the effect of drugs on erythema symptoms in rats with phenol-induced cervicitis.
  • Figure 18 shows the effect of drugs on edema symptoms in rats with phenol-induced cervicitis.
  • Figure 19 shows the effect of drugs on secretory symptoms in rats with phenol-induced cervicitis.
  • Figure 20 shows the effect of drugs on cervical index in rats with phenol-induced cervicitis.
  • Figure 21 shows the vaginal HE staining results of rats with phenol-induced cervicitis.
  • Figure 22 shows the scoring results of mouse skin redness, bleeding, eruption and desquamation in the atopic dermatitis/pruritus mouse model.
  • Figure 23 shows the area under the curve (AUC) scores for redness, bleeding, eruption and desquamation of mouse skin in atopic dermatitis/pruritus mouse model.
  • Figure 24 shows the results of HE staining experiments on atopic dermatitis/pruritus mouse model ( ⁇ 400).
  • Figure 25 shows the results of toluidine blue staining experiment on atopic dermatitis/pruritus mouse model ( ⁇ 400).
  • Figure 26 shows the effect of drugs on IL-4 in the serum of atopic dermatitis/pruritus mice (* compared with the normal group, p ⁇ 0.05).
  • Figure 27 shows the effect of drugs on IL-13 in the serum of atopic dermatitis/pruritus mice (* compared with the normal group, p ⁇ 0.05).
  • Figure 28 shows the effect of drugs on IFN- ⁇ in the serum of atopic dermatitis/pruritus mice (* compared with the normal group, p ⁇ 0.05).
  • Figure 29 shows the effect of drugs on TNF- ⁇ in the serum of atopic dermatitis/pruritus mice (* compared with the normal group, p ⁇ 0.05).
  • Figure 30 shows the effect of drugs on the skin depigmentation area score of vitiligo mice (medication site, skin depigmentation site).
  • Figure 31 shows the effect of the drug on the fur depigmentation area score of vitiligo mice (non-medicated parts, namely the ears, non-medicated parts of the trunk, and tail).
  • Figure 32 shows the results of HE staining of vitiligo mice.
  • Figure 33 shows the effect of the drug on serum TNF- ⁇ in vitiligo mice (* compared with the blank control group, p ⁇ 0.01, # compared with the model group, p ⁇ 0.01).
  • Figure 34 shows the effect of the drug on serum IL-6 in vitiligo mice (* compared with the blank control group, p ⁇ 0.01, # compared with the model group, p ⁇ 0.01).
  • Figure 35 shows a comparison of the area under the hair growth score curve in mice with alopecia areata.
  • Figure 36 shows hair growth in alopecia areata mice (day 21).
  • Figure 37 shows hair growth in female alopecia areata mice.
  • Figure 38 shows hair growth in male alopecia areata mice.
  • Figure 39 shows skin thickness scores in imiquimod-induced psoriasis mice.
  • Figure 40 shows clinical scores of imiquimod-induced psoriasis mice.
  • Figure 41 shows the AUC of skin thickness score of imiquimod-induced psoriasis mice (*p ⁇ 0.05, **p ⁇ 0.01, ****p ⁇ 0.0001 vs. vehicle control group, One-way ANOVA ).
  • Figure 42 shows the AUC of clinical scores in imiquimod-induced psoriasis mice (***p ⁇ 0.001, ****p ⁇ 0.0001 vs. vehicle control group, One-way ANOVA).
  • Figure 43 shows the spleen weight of imiquimod-induced psoriasis mice at the end of the experiment (***p ⁇ 0.001, ****p ⁇ 0.0001 vs. vehicle control group, One-way ANOVA).
  • Figure 44 shows the pathological scoring results of psoriasis mice (****p ⁇ 0.0001 vs. solvent control group, One-way ANOVA).
  • Figure 45 shows the detection results of cytokine TNF- ⁇ in psoriasis mice (*p ⁇ 0.05 vs. vehicle control group, One-way ANOVA).
  • Figure 46 shows the effect of drugs on the weight of acne rabbit ear pieces (##, compared with the blank control, p ⁇ 0.01, * compared with the model (vehicle) group, p ⁇ 0.05).
  • Figure 47 shows the histopathological picture of acne rabbit ear.
  • Figure 48 shows the effect of drugs on the diameter of sebaceous glands in ear tissue (##, compared with the blank control, p ⁇ 0.01, * compared with the model (vehicle) group, p ⁇ 0.05).
  • Figure 49 shows the effect of drugs on the hair follicle area of acne rabbit ear tissue (##, compared with the blank control, p ⁇ 0.01, # compared with the blank control, p ⁇ 0.05, ** compared with the model (vehicle) group, p ⁇ 0.01, *Compared with model (vehicle) group, p ⁇ 0.05).
  • Figure 50 shows the effect of the drug on serum IL-1 ⁇ of acne rabbits (##, compared with the blank control, p ⁇ 0.01, ** compared with the model (vehicle) group, p ⁇ 0.01).
  • Figure 51 shows the effect of the drug on serum IL-6 in acne rabbits (##, compared with the blank control, p ⁇ 0.01, ** compared with the model (vehicle) group, p ⁇ 0.01).
  • Figure 52 shows the effect of drugs on serum DHT in acne rabbits.
  • Figure 53 shows the experimental process and experimental results of the effects of compounds on adipocyte lipolysis in vitro.
  • Figure 54 shows the results of HE staining of mouse non-wound skin and wound tissue.
  • Figure 55 shows experimental results of compounds promoting epithelial healing in wounds.
  • Example 1 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazol-5(1H)-yl)(5-(piperidin-1-yl)pyrazin-2-yl)methanone (MDI-2)
  • Example 1 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5(1H)-yl)(5-morpholinopyrazin-2-yl)methanone (MDI-201)
  • Synthetic intermediate MDI-201-3 (2-(6-bromo1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazol-3-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)-4,6-dihydropyrrolo[3,4-d]imidazol-5(1H)-yl)(5-morpholinepyrazine- 2-yl)methanone
  • intermediate MDI-201-3 (43.0mg, 0.06mmol), (2-((5-ethyl-2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2) -Dioxaboran-2-yl)phenoxy)methoxy)ethyl)trimethylsilane (27.1 mg, 0.07mmol), Pd(dppf)Cl 2 (4.2mg, 0.006mmol) and potassium phosphate (36.2 mg, 0.17 mmol) was dissolved in 1,4-dioxane (10 ml) and water (2 ml), replaced with nitrogen three times, heated to 100 degrees, reacted overnight, cooled to room temperature, added water, and washed with ethyl acetate The ester was extracted twice, the organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified on a silica gel column to obtain intermediate MDI-201-4 with a yield of 40.9%.
  • Dissolve intermediate MDI-201-4 (22.0 mg, 0.02 mmol) in methanol (4 ml), add concentrated hydrochloric acid (2 ml), heat to 50 degrees, react for 6 hours, concentrate, dissolve the solid with 1 ml of methanol, and add 2 ml of Concentrate ammonia solution, concentrate, add ammonia solution three times with methanol, and purify with a preparation plate to obtain 8.0 mg of the final product, with a yield of 61.9%.
  • Example 3 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrrolo [3,4-d]imidazol-5(1H)-yl)(1-methyl-1H-pyrazol-4-yl)methanone (MDI-202)
  • Example 4 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)pyrrolo[3,4-d]imidazole- 5(1H ,4H,6H)-yl)(1-methylpiperidin-4-yl)methanone (MDI-203)
  • MDI-203 can also be named 5-ethyl-2-fluoro-4- ⁇ 3-[5-(1-methylpiperidine-4-carbonyl)-1H,4H,5H,6H-pyrrolo[3 ,4-d]imidazol-2-yl]-1H-indazol-6-yl ⁇ phenol was synthesized according to the method shown in Example 4 of the Chinese invention patent CN111606908B.
  • the hydrogen nuclear magnetic resonance spectrum data of the obtained product is as follows:
  • Example 5 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)pyrrolo[3,4-d]imidazole -5(1H ,4H,6H)-yl)(5-(4-methylpiperazin-1-yl)pyrazin-2-yl)methanone (MDI-204)
  • MDI-204 can also be named 5-ethyl-2-fluoro-4- ⁇ 3-[5-(4-methylpiperazine-1-carbonyl)-1H,4H,5H,6H-pyrrolo[3 ,4-d]imidazol-2-yl]-1H-indazol-6-yl ⁇ phenol was synthesized according to the method shown in Example 5 of Chinese invention patent CN111606908B.
  • the hydrogen nuclear magnetic resonance spectrum data of the obtained product is as follows:
  • Example 6 (2-(6-(2-ethyl-4-hydroxyphenyl)-1H-indazol-3-yl)pyrrolo[3,4-d]imidazole- 5(1H,4H,6H )-yl)(5-(4-methylpiperazin-1-yl)pyrazin-2-yl)methanone (MDI-205)
  • MDI-205 is also named 3-ethyl-4- ⁇ 3-[5-(4-methylpiperazine-1-carbonyl)-1H,4H,5H,6H-pyrrolo[3,4-d ]imidazol-2-yl]-1H-indazol-6-yl ⁇ phenol was synthesized according to the method shown in Example 6 of Chinese invention patent CN111606908B.
  • the hydrogen nuclear magnetic resonance spectrum data of the obtained product is as follows:
  • Example 7 5-ethyl-2-fluoro-4-(3-(5-(phenylsulfonyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazole-2- (MDI-206)-1H-indazol-6-yl)phenol (MDI-206)
  • Example 8 5-ethyl-2-fluoro-4-(3-(5-(pyrazin-2ylmethyl)-1,4,5,6-tetrahydropyrro[3,4-d] Imidazol -2-yl)-1H-indazol-6-yl)phenol (MDI-207)
  • Example 9 4-(3-(5-(cyclopropylmethyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl)-1H- indazole -6-yl)-5-ethyl-2-fluorophenol (MDI-208)
  • Synthetic intermediate MDI-1233-1 (2-(6-bromo1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazol-3-yl)-1-( (2-(Trimethylsilyl)ethoxy)methyl)pyrrolo[3,4-d]imidazol-5(1H,4H,6H)-yl)(cyclopropyl)methanone
  • Synthetic intermediate MDI-1233-2 cyclopropyl(2-(6-(2-ethyl-5-fluoro-4-((2-(trimethylsilyl)ethoxy)methoxy)benzene base)1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazol-3-yl)-1-((2-(trimethylsilyl)ethoxy) Methyl)pyrrolo[3,4-d]imidazole-5(1H,4H,6H)-yl)methanone
  • the intermediate MDI-1233-1 (50.5mg, 0.08mmol), (2-((5-ethyl-2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2) -Dioxaboran-2-yl)phenoxy)methoxy)ethyl)trimethylsilane (34.8 mg, 0.1 mmol), Pd(dppf)Cl 2 (5.9 mg, 0.008 mmol) and potassium phosphate (50.9 mg, 0.24 mmol) was dissolved in 1,4-dioxane (10 ml) and water (2 ml), replaced with nitrogen three times, heated to 100 degrees, reacted overnight, cooled to room temperature, added water, and washed with ethyl acetate The ester was extracted twice, the organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified on a silica gel column to obtain intermediate MDI-1233-2 with a yield of 76.1%.
  • Example 11 4-(3-(5-(cyclobutylmethyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl)-1H- indazole-6 -yl)-5-ethyl-2-fluorophenol (MDI-210)
  • Example 12 cyclobutyl(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)pyrrolo [3,4-d]imidazole- 5(1H,4H,6H)-yl)methanone (MDI-211)
  • Example 13 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)pyrrolo[3,4-d] imidazole -5( 1H,4H,6H)-yl)(3-hydroxycyclobutyl)methanone (MDI-213)
  • Example 14 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-pyrrolo[3,4-d] imidazole -5 -(1H,4H,6H)-yl)(pyridazin-4-yl)methanone (MDI-214)
  • Example 15 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-pyrrolo[3,4-d] imidazole -5 -(1H,4H,6H)-yl)(pyridazin-3-yl)methanone (MDI-215)
  • Example 17 5-ethyl-2-fluoro-4-(3-(5-(4-hydroxycyclohexyl)-1,4,5,6-tetrahydropyrrolo[3,4-d] imidazole- 2-yl)-1H-indazol-6-yl)phenol (MDI-217)
  • Example 18 4-(3-(5-(cyclopropylsulfonyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl)-1H- indazole- 6-yl)-5-ethyl-2-fluorophenol (MDI-218)
  • Synthetic intermediate MDI-218-1 6-bromo-3-(5-(cyclopropylsulfonyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1,4, 5,6-Tetrahydropyrrolo[3,4-d]imidazol-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole
  • Synthetic intermediate MDI-218-2 3-(5-cyclopropylsulfonyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1,4,5,6-tetrakis Hydropyrrolo[3,4-d]imidazol-2-yl)-6-(2-ethyl-5-fluoro-4-((2-(trimethylsilyl)ethoxy)methoxy) Phenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole
  • the intermediate MDI-218-1 (36.0mg, 0.05mmol), (2-((5-ethyl-2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2) -Dioxaboran-2-yl)phenoxy)methoxy)ethyl)trimethylsilane (25.5 mg, 0.06 mmol), Pd(dppf)Cl 2 (3.9 mg, 0.005 mmol) and potassium phosphate (34.2 mg, 0.16 mmol) was dissolved in 1,4-dioxane (6 ml) and water (1 ml), replaced with nitrogen three times, heated to 100 degrees, reacted overnight, cooled to room temperature, added water, and washed with ethyl acetate The ester was extracted twice, the organic phases were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified on a silica gel column to obtain intermediate MDI-218-2 with a yield of 70.0%.
  • Synthetic compound MDI-218 4-(3-(5-(cyclopropylsulfonyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl)-1H-indole Azol-6-yl)-5-ethyl-2-fluorophenol
  • Dissolve intermediate MDI-218-2 (36.0 mg, 0.04 mmol) in methanol (4 ml), add concentrated hydrochloric acid (2 ml), heat to 50 degrees, react for 6 hours, concentrate, dissolve the solid with 1 ml of methanol, and use hydrogen carbonate Adjust the pH of the sodium solution to 8-9, extract 4 times with dichloromethane, combine the organic phases, dry with anhydrous sodium sulfate, and purify with a preparative plate to obtain 16 mg of the final product with a yield of 81.4%.
  • Example 19 4-(3-(5-(cyclobutylsulfonyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl) -1H-indazole -6-yl)-5-ethyl-2-fluorophenol (MDI-219)
  • Example 20 4-(3-(5-(cyclopentylsulfonyl)-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl) -1H-indazole -6-yl)-5-ethyl-2-fluorophenol (MDI-220)
  • Example 21 5-ethyl-2-fluoro-4-(3-(5-((1-methyl-1H-pyrazol-4-yl)methyl)-1,4,5,6-tetrakis Hydropyrrolo [3,4-d]imidazol-2-yl)-1H-indazol-6-yl)phenol (MDI-221)
  • Example 22 4-(3-(5-cyclopentyl-1,4,5,6-tetrahydropyrrolo[3,4-d]imidazol-2-yl)-1H-indazole -6- (MDI-224)-5-ethyl-2-fluorophenol
  • Example 24 1-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3, 4-d]imidazole-5-(1H)-yl)ethan-1-one (MDI-226)
  • Example 25 1-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3, 4-d]imidazole-5-(1H)-yl)propan-1-one (MDI-227)
  • Example 26 1-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3, 4-d]imidazole-5-(1H)-yl)-2-methylpropan-1-one (MDI-228)
  • Example 27 2-cyclopropyl-1-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6- di Hydropyrrolo[3,4-d]imidazol-5-(1H)-yl)ethan-1-one (MDI-229)
  • Example 28 1-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3, 4-d]imidazole-5-(1H)-yl)-3-methylbutan-1-one (MDI-230)
  • Example 29 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5-(1H)-yl)(pyrrolidin-1-yl)methanone (MDI-231)
  • Synthetic intermediate MDI-231-1 (2-(6-(4-(benzyloxy)-2-ethyl-5-fluorophenyl)-1-((2-(trimethylsilyl)) Ethoxy)methyl)-1H-indazol-3-yl)-1-((2--(trimethylsilyl)ethoxy)methyl)-4,6-dihydropyrro[ 3,4-d]imidazol-5-(1H)-yl)(pyrrolidin-1-yl)methanone
  • Example 31 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5-(1H)-yl)(piperidin-1-yl)methanone (MDI-233)
  • Example 32 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5-(1H)-yl)(morpholino)methanone (MDI-234)
  • Example 33 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5-(1H)-yl)(4-methylpiperazin-1-yl)methanone (MDI-235)
  • Example 34 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5-(1H)-yl)(4-ethylpiperazin-1-yl)methanone (MDI-236)
  • Example 35 Cyclopropyl(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-pyrazolo[4,3-b]pyridin-3- yl)-4 ,6-Dihydropyrro[3,4-d]imidazol-5(1H)-yl)methanone (MDI-237)
  • Example 36 cyclopropyl(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-4-methyl-1H-indazol-3-yl) -4,6-di Hydropyrrolo[3,4-d]imidazole-5(1H)-yl)methanone (MDI-239)
  • Example 38 Cyclopropyl(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-pyrazolo[4,3-c]pyridin-3- yl)-4 ,6-Dihydropyrro[3,4-d]imidazol-5(1H)-yl)methanone (MDI-242)
  • Example 41 (2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4- d]imidazole-5(1H)-yl)(4-hydroxypiperidin-1-yl)methanone (MDI-245)
  • Dissolve MDI-245-1 (116 mg, 0.14 mmol) in 20 ml methanol, add 20 mg palladium carbon, replace hydrogen, react at 40 degrees for 1 hour, after the reaction is completed, filter, and the filtrate is concentrated.
  • the concentrate is dissolved in 12 ml methanol, and 6 ml concentrated Hydrochloric acid, react at 50 degrees for 7 hours, concentrate, add methanol with hydrochloric acid 3 times, concentrate, dissolve in 8 ml of methanol, add 0.8 ml of ammonia, concentrate, and purify with a preparation plate to obtain 30 mg of the final product, with a yield of 44.4%.
  • Example 42 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-N-methyl-4,6-dihydropyrro [ 3,4-d]imidazole-5-(1H)-carboxamide (MDI-246)
  • Example 43 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-N-ethyl-4,6-dihydropyrro [ 3,4-d]imidazole-5-(1H)-carboxamide (MDI-247)
  • Example 44 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-N-(2-hydroxyethyl) -4,6- Dihydropyrro[3,4-d]imidazole-5(1H)-carboxamide (MDI-248)
  • Dissolve MDI-248-1 (44 mg, 0.06 mmol) in 10 ml of methanol, add 8 mg of palladium on carbon, replace the hydrogen, and react at 40 degrees for 1 hour. After the reaction is completed, filter, and the filtrate is concentrated. The concentrate is dissolved in 6 ml of methanol, and 3 ml of concentrated Hydrochloric acid, react at 50 degrees for 7 hours, concentrate, add methanol with hydrochloric acid 3 times, concentrate, dissolve in 5 ml of methanol, add 0.5 ml of ammonia, concentrate, and purify with a preparation plate to obtain 14 mg of the final product, with a yield of 56.6%.
  • Example 46 1-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-1,4,5,6-tetrahydropyridine Chro[3,4-d]imidazole-5-carbonyl)pyrrolidine-3-carbonitrile (MDI-250)
  • Example 47 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-N-(tetrahydrofuran-3-yl )-4,6- Dihydropyrro[3,4-d]imidazole-5(1H)-carboxamide (MDI-251)
  • Example 48 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4-d ] Imidazole-5(1H)-carboxylic acid methyl ester (MDI-252)
  • Example 49 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3,4-d ] Imidazole-5(1H)-carboxylic acid ethyl ester (MDI-253)
  • Dissolve MDI-253-1 (33 mg, 0.04 mmol) in 10 ml of ethanol, add 6 mg of palladium carbon, replace the hydrogen, and react at 40 degrees for 1 hour. After the reaction is completed, filter, and the filtrate is concentrated. The concentrate is dissolved in 6 ml of ethanol, and 3 ml of concentrated Hydrochloric acid, react at 50 degrees for 7 hours, concentrate, add ethanol with hydrochloric acid 3 times, concentrate, dissolve in 5 ml of ethanol, add 0.5 ml of ammonia, concentrate, and purify with a preparation plate to obtain 10 mg of the final product, with a yield of 54.8%.
  • Example 50 (S)-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-pyrazolo[3,4-b]pyridin-3- yl)- 4,6-Dihydropyrrolo[3,4-d]imidazol-5(1H)-yl)(3-hydroxypyrrolidin-1-yl)methanone (MDI- 255)
  • Example 51 3-(2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6-dihydropyrro [3, 4-d]imidazole-5(1H)-yl)-3-oxopropionitrile (MDI-256)
  • Example 52 2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-N,N-dimethyl-4,6- dihydro Pyrro[3,4-d]imidazole-5(1H)-carboxamide (MDI-257)
  • the synthesis process is similar to the synthesis method of intermediate MDI-246-1 in Chinese invention patent CN111606908B, except that dimethylamine hydrochloride is used instead of methylamine hydrochloride.
  • Example 53 N-(2-cyanoethyl)-2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl) -4,6 -Dihydropyrro[3,4-d]imidazole-5(1H)-carboxamide (MDI-258)
  • Example 54 N-cyclopropyl-2-(6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-1H-indazol-3-yl)-4,6- dihydropyrrolo [3,4-d]imidazole-5(1H)-carboxamide (MDI-259)
  • Example 58 (R)-6-(2-ethyl-5-fluoro-4-hydroxyphenyl)-3-(5-prolyl-1,4,5,6-tetrahydropyrrolo [3 ,4-d]imidazol-2-yl)-1H-indazole (MDI-263)
  • Compound inhibition rate (%inh) 100%-(compound-positive control)/(negative control-positive control)*100%
  • Example 60 Test of inhibitory activity of compounds against RET kinase
  • Compound inhibition rate (%inh) 100%-(compound-positive control)/(negative control-positive control)*100%
  • Example 61 Therapeutic effect of compounds on ovalbumin-induced mouse asthma model
  • Ovalbumin (OVA) Sigma-A5243
  • Interleukin 5 (IL-5) Elisa kit Beinlay-DRE30011
  • PBS Phosphate buffer saline
  • mice aged 7-8 weeks were purchased from Changzhou Cavins Experimental Animal Co., Ltd., animal certificate number: 202005500.
  • mice were kept in the animal room of Shanghai Institute of Materia Medica, Chinese Academy of Sciences, SPF grade, temperature 22-24°C, humidity 40%-70%, light 7:00-19:00, 3 mice per cage.
  • Food and Water Ad libitum.
  • ⁇ Immune preparation configuration Weigh an appropriate amount of OVA and add an appropriate amount of PBS to make the final concentration 0.1mg/ml. Add an equal volume of aluminum adjuvant, mix well, place at 4°C, and prepare immediately.
  • ⁇ Preparation of sensitizer (5% OVA): Weigh an appropriate amount of OVA and add an appropriate amount of PBS to make the final concentration 5%. Mix well and set aside at 4°C until ready to use.
  • ⁇ Vehicle preparation Add 200ul Tween 80 to every 100ml of normal saline and mix well. Set aside at 4 degrees.
  • ⁇ Preparation of solution of compound MDI-1288 Weigh an appropriate amount of MDI-1288 and solvent, and wait until it is dissolved to make the final concentration 1.2 mg/ml. Mix well, set aside at 4 degrees Celsius, and prepare once every three days.
  • ⁇ Preparation of solution of compound MDI-1228 Weigh an appropriate amount of MDI-1228 and solvent, and wait until it is dissolved to make the final concentration 1.2 mg/ml. Mix well, set aside at 4 degrees Celsius, and prepare once every three days.
  • ⁇ Preparation of solution of compound MDI-1233 Weigh appropriate amounts of MDI-1233 and solvent, wait until it is dissolved, and make the final concentrations 0.4, 1.2 and 4 mg/ml. Mix well, set aside at 4 degrees Celsius, and prepare once every three days.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Diabetes (AREA)
  • Dermatology (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Endocrinology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Psychology (AREA)
  • Cardiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Psychiatry (AREA)
  • Vascular Medicine (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)
  • Otolaryngology (AREA)
  • Hospice & Palliative Care (AREA)
  • Reproductive Health (AREA)
  • Ophthalmology & Optometry (AREA)

Abstract

公开了一类作为TRK抑制剂药物或RET抑制剂药物的化合物及其用途。具体地,公开了如式(G)所示的化合物、或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物在制备TRK抑制剂或RET抑制剂药物中的用途。

Description

作为TRK抑制剂和/或RET抑制剂的化合物及其用途 技术领域
本发明提供了一类化合物用于TRK激酶和/或RET激酶的用途。本发明还涉及包含所述化合物的组合物,以及所述化合物和所述组合物在制备用于治疗和/或预防与TRK和/或RET相关的疾病或病症的药物中的用途。
背景技术
蛋白质激酶是催化蛋白质中特定残基磷酸化的酶家族,并广义地分类为酪氨酸和丝氨酸/苏氨酸激酶。由于突变、过度表达或不适当调节、调节异常或失调,以及生长因子或细胞因子的过度产生或产生不足所导致的不适当的激酶活性涉及许多疾病,其包括但不限于癌症、心血管疾病、变态反应、哮喘和其它呼吸疾病、自身免疫病、炎症疾病、骨病、代谢紊乱及神经病症和神经变性病症(例如阿尔茨海默病)。不适当的激酶活性触发多种生物细胞反应,所述生物细胞反应与涉及上述和相关疾病的细胞生长、细胞分化、细胞功能、存活、凋亡和细胞运动性有关。因此,蛋白质激酶已成为一类重要的作为治疗性介入的靶点的酶。
TRK激酶为由一组可溶性生长因子(称为神经营养因子(NT))活化的高亲和力受体酪氨酸激酶。TRK家族具有三个成员:TRKA、TRKB和TRKC。其中,TRKA由神经生长因子(NGF)活化,TRKB由脑源性神经生长因子(BDNF)和NT-4/5活化,TRKC由NT3活化。TRK在神经元组织中广泛表达并且涉及神经元细胞的维持、信号传导和存活。
已证实TRK/神经营养因子途径的抑制剂在许多疼痛的临床前动物模型中是有效的。还已显示由肿瘤细胞和肿瘤侵入巨噬细胞分泌的NGF直接刺激位于周围疼痛纤维,所以TrkA和/或其他Trk激酶的抑制剂可提供一种用于慢性疼痛状态和癌症相关疼痛的有效治疗。还有报道,Trk激酶的过度表达、活化、扩增和/或突变与许多癌症有关,例如成神经细胞癌、结肠直肠癌、黑色素瘤、胃癌、肺癌、乳腺癌等。2018年美国FDA正式批准有史以来第一款口服TRK抑制剂,即Loxo Oncology公司生产的Vitrakvi(亦称larotrectinib、拉罗替尼、LOXO101),用于治疗具有神经营养酪氨酸激酶(NTRK)基因融合的实体瘤患者,其已被证实对多种癌症具有极高的治疗有效率。还发现TRK抑制剂还可以用于治疗炎性病症和自身免疫性病症。
此外,RET激酶也是一类非常重要的药物靶点。2020年5月FDA批准了全球首个RET激酶抑制剂,Loxo Oncology公司开发的塞尔帕替尼(Selpercatinib,Retevmo),用于治疗非小细胞肺癌、甲状腺髓样癌、以及其他类型的甲状腺癌。之后,2020年9月,FDA批准了Blueprint Medicines开发的RET激酶抑制剂普拉替尼(Pralsetinib),用于RET融合阳性转移性非小细胞肺癌成人患者的治疗。
虽然目前已有一些TRK或RET抑制剂获批准上市,并且还有大量TRK或RET抑制剂处于临床研究阶段,但这些TRK/RET抑制剂在疗效或安全性方面并不尽如人意。因此,始终存在对疗效更好和/或副作用更少的TRK抑制剂或RET抑制剂的需求。
发明内容
本发明的一个目的是提供一类替代现有TRK抑制剂的新型TRK抑制剂,从而为TRK相关疾病的治疗提供更多的选择。
本发明的进一步目的是提供一类比现有TRK抑制剂功效更好和/或安全性更佳的新型TRK抑制剂。
本发明的另一个目的是提供一类替代现有RET抑制剂的新型RET抑制剂,从而为RET相关疾病的治疗提供更多的选择。
本发明的进一步目的是提供一类比现有RET抑制剂功效更好和/或安全性更佳的新型RET抑制剂。
本发明的另一个目的是为瘙痒、银屑病、特应性皮炎、痤疮、白癜风、斑秃、哮喘、鼻炎、痔疮、宫颈炎、肺炎、癌症(肿瘤)等与TRK和/或RET相关的疾病提供替代疗法或更有效的疗法,从而为这些疾病的治疗提供更多的选择。中国发明专利CN111606908B(等同于美国专利公开US2022/0073524A1)公开了一类作为范JAK抑制剂的化合物。JAK是英文Janus kinase的缩写,是转导细胞因子信号从膜受体到STAT转录因子的细胞质酪氨酸激酶。JAK是一类非常重要的药物靶点,其涉及细胞的增殖、分化、凋亡以及免疫调节等许多重要的生物学过程。针对这一靶点而研发的JAK抑制剂主要用于治疗血液系统疾病、肿瘤、类风湿性关节炎等药物。JAK蛋白家族共包括4个成员:JAK1、JAK2、JAK3以及TYK2。根据CN111606908B所述,其中公开的化合物对于JAK1、JAK2、JAK3、TYK2均具有较高的抑制活性,是一种高效的范JAK抑制剂。
发明人在研究过程中出人意料地发现:CN111606908B公开的化合物对于TRK激酶也具有非常高的抑制活性,并且对于TRKA、TRKB、TRKC都有非常好的抑制活性,因此是一类范TRK抑制剂。
此外,发明人还出人意料地发现:CN111606908B公开的化合物对于RET激酶也具有非常高的抑制活性。
已有报道,Sienna Pharm公司开发的Pegcantratinib(SNA-125)能同时抑制JAK3和TRKA,但是对于JAK激酶和TRK激酶家族中的其它成员抑制效果较差,并且在银屑病的临床实验中最终未能成功。就发明人所知,目前还没有报道过同时对JAK激酶和TRK激酶具有广谱高效抑制活性的化合物,本申请所述化合物是首次报道的高效范JAK/范TRK抑制剂。
由于对JAK、TRK、RET的双重或多重广谱抑制活性,本申请所述化合物对许多疾病的治疗能够提供比现有药物更好的疗效,并且能够治疗痤疮这一由其他JAK抑制剂引起的副反应(Int.J.Dermatol.2021 Aug 22.doi:10.1111/ijd.15853.PMID:34423443)。已发现本申请所述化合物尤其适合治疗自身免疫性疾病,尤其是皮肤自免疫疾病。此外,通过动物模型,已经证实了本申请所涉及化合物对于瘙痒、银屑病、特应性皮炎、EGFR抑制剂引起的皮肤副作用(例如CN112933095A中列出的那些疾病)、痤疮、白癜风、斑秃、哮喘、鼻炎、痔疮、宫颈炎、肺炎、(糖尿病导致的)伤口愈合缓慢、糖尿病以及糖尿病并发症(如糖尿病足)等疾病的治疗效果。
出人意料地,发明人还发现本申请的CN111606908B公开的化合物对于糖尿病足以及包括褥疮在内的慢性伤口愈合具有非常好的治疗效果。糖尿病足作为一种常见的糖尿病并发症,困扰着全球接近4亿人口。由于伤口难以愈合,30%左右的患者最终会截肢。目前对糖尿病足的治疗尚缺乏有效的药物。最近的研究表明,伤口的愈合以及皮肤的修复需要脂肪细胞发生脂解并转化为肌成纤维细胞,而糖尿病足的患者正是缺乏这样的能力(Cell Stem Cell 26,1–16,June 4,2020)。发明人出人意料地发现CN111606908B公开的化合物能在体外有效促进脂肪细胞的脂解,但其他的JAK抑制剂Tofacitinib就完全没有类似效果,目前也尚无报道指出JAK抑制剂能够促进脂肪细胞的脂解。发明人也发现CN111606908B公开的化合物能在体内大幅增加糖尿病动物伤口处肌成纤维细胞的数量,并促进糖尿病动物伤口的愈合。鉴于常规JAK抑制剂并没有促进脂肪细胞脂解的功效,发明人认为CN111606908B公开的化合物在糖尿病足动物中展现出的疗效源自这些化合物泛JAK/泛TRK的双重抑制活性,或者泛JAK/泛TRK/RET的多重抑制活性。发明人据此认为:JAK/TRK双重抑制剂(优选泛JAK/泛TRK的双重抑制剂),或者JAK/TRK/RET多重抑制剂(优选泛JAK/泛TRK/RET的多重抑制剂)会对包括但不限于糖尿病足、褥疮在内的慢性伤口愈合产生较好的效果。
在第一个方面,本发明提供了式(G)的化合物:
或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物作为TRK抑制剂药物和/或RET抑制剂药物的用途,或者在制备TRK抑制剂药物和/或RET抑制剂药物中的用途,其中:
L为C=O、O=S=O、CH2或连接键;且
X1为N或CR14;且
X2为N或CR15;且
X3为N或CR16;且
R14、R15、R16各自独立地选自H、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C2-8烯基、C2-8炔基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基任选地被1个、2个或3个选自卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、-N(C1-4烷基)(C(=O)C1-4烷基)、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基的取代基取代;且
R13为H、-N(R17)(R18)、C1-6烷氧基、-SR12、-OR12、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基、C7- 11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且任选地被一个或多个各自独立选自以下群组的取代基所取代:-OH、-CN、-SH、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、 C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;或者R17、R18以及与它们相连的N原子共同形成3-14元环;且
R2的个数为0、1、2、3或4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且
R1选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;且
R3、R4各自独立地选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1- 6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R5)(R6)、-N(R11)(C(=O)R12)、-CON(R7)(R8)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-6烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且R5、R6、R7、R8、R9、R10、R11、R12各自独立地是H或选自以下群组:C1- 6烷基、C1-4卤代烷基、C3-7环烷基、4-14元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CF3、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、氧代、C1-4烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
在本发明的一些优选实施方式中,使用了上述式(G)的化合物的同位素标记化合物。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代。
在本发明的一些优选实施方式中,在式(G)中,X1为N。在本发明的一些优选实施方式中,在式(G)中,X2为N。在本发明的一些优选实施方式中,在式(G)中,X3为N。在本发明的一些优选实施方式中,在式(G)中,X1为CR14、X2为N或CR15、X3为CR16。在本发明的一些优选实施方式中,在式(G)中,X1为CR14、X2为CR15、X3为CR16。在本发明的一些优选实施方式中,在式(G)中,X1为CR14、X2为CR15、X3为CR16,且R14、R15、R16各自独立地选自H、-OH、-CN、卤素、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基。在本发明的一些优选实施方式中,在式(G)中,X1为CR14、X2为N、X3为CR16,且R14、R16各自独立地选自H、-OH、-CN、卤素、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基。在本发明的一些优选实施方式中,在式(G)中,X1、X2、X3是相同的。在本发明的一些优选实施方式中,在式(G)中,X1、X2、X3均为CH。在本发明的一些优选实施方式中,在式(G)中,X1、X2、X3均为N。在本发明的一些优选实施方式中,在式(G)中,X1为C(CH3),X2、X3均为CH。在本发明的一些优选实施方式中,在式(G)中,X2为C(CH3),X1、X3均为CH。在本发明的一些优选实施方式中,在式(G)中,X3为C(CH3),X1、X2均为CH。在本发明的一些优选实施方式中,在式(G)中,X1为N,X2、X3均为CH。在本发明的一些优选实施方式中,在式(G)中,X2为N,X1、X3均为CH。在本发明的一些优选实施方式中,在式(G)中,X3为N,X1、X2均为CH。
在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X1、X2、X3是相同的。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X1、X2、X3均为CH。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X1、X2、X3均为N。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X1为C(CH3),X2、X3均为CH。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X2为C(CH3),X1、X3均为CH。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X3为C(CH3),X1、X2均为CH。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X1为N,X2、X3均为CH。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X2为N,X1、X3均为CH。在本发明的一些更优选的实施方式中,使用了式(G)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,并且X3为N,X1、X2均为CH。
在本发明的一些优选实施方式中,在式(G)中,L为C=O、O=S=O或CH2。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O。在本发明的一些特别优选的实施方式中,在式(G)中,L为O=S=O。在本发明的一些特别优选的实施方式中,在式(G)中,L为CH2。在本发明的另一些实施方式中,在式(G)中,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为N,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为N,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为N,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为N,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CR14,其中R14选自-OH、-CN、卤素、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CR14,其中R14选自-OH、-CN、卤素、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CR14,其中R14选自-OH、-CN、卤素、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X1、X2、X3均为CR14,其中R14选自-OH、-CN、卤素、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X1为C(CH3),X2、X3均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1为C(CH3),X2、X3均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1为C(CH3),X2、X3均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X1为C(CH3),X2、X3均为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X2为C(CH3),X1、X3均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X2为C(CH3),X1、X3均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X2为C(CH3),X1、X3均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X2为C(CH3),X1、X3均为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X3为C(CH3),X1、X2均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X3为C(CH3),X1、X2均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X3为C(CH3),X1、X2均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X3为C(CH3),X1、X2均为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X1为N,X2、X3均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1为N,X2、X3均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X1为N,X2、X3均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X1为N,X2、X3均为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X2为N,X1、X3均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X2为N,X1、X3均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X2为N,X1、X3均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X2为N,X1、X3均为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G)中,X3为N,X1、X2均为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X3为N,X1、X2均为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G)中,X3为N,X1、X2均为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G)中,X3为N,X1、X2均为CH,L为连接键。
在本发明的一些优选实施方式中,在式(G)中,R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3- 7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代(其中R13任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(G)中,R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基,且R17、R18如上文所限定(其中R13任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(G)中,R13为H、-N(R17)(R18)、C1- 6烷氧基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,且R17、R18如上文所限定(其中R13任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(G)中,R13为-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、4-6元杂环烷基、苯基、5-6元杂芳基,且R17、R18如上文所限定(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些优选实施方式中,在式(G)中,R13为-N(R17)(R18)、C1-3烷氧基、C3-6环烷基、4-6元杂环烷基、苯基、5-6元杂芳基或者C1-4烷基,且R17、R18如上文所限定(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些优选实施方式中,在式(G)中,R13为-N(H)(C1-3烷基)、-N(H)(3-6元环烷基)、-N(H)(4-6元杂环烷基)、-N(C1-3烷基)(C1-3烷基)、C1-3烷氧基、C3-6环烷基、4-6元氮杂环烷基或氧杂环烷基、苯基、5-6元氮杂芳基或者C1-4烷基;或者R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成4-10元环(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为环丙基、环丁基、环戊基、环己基、甲基、乙基、丙基、丁基、甲氧基、乙氧基、丙氧基、-N(H)(CH3)、-N(H)(CH2CH3)、-N(H)(CH2CH2OH)、-N(H)(CH2CH2CN)、-N(CH3)(CH3)、-N(H)(环丙基)、-N(H)(环丁基)、-N(H)(四氢呋喃基)、吡嗪基、哒嗪基、吡咯烷基、吡唑基、哌啶基、苯基、氮杂环丁基、吗啉基、哌嗪基或四氢吡喃基;或者R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为环丙基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为环丁基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为环戊基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为环己基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为甲基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为乙基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为丙基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为丁基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为吡嗪基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为哒嗪基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为吡咯烷基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为吡唑基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为哌啶基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为苯基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为氮杂环丁基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为吗啉基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为哌嗪基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为四氢吡喃基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为甲氧基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为乙氧基。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(CH3)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(CH2CH3)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(CH2CH2OH)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(CH2CH2CN)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(CH3)(CH3)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(环丙基)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(环丁基)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(H)(四氢呋喃基)。在本发明的一些特别优选的实施方式中,在式(G)中,R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环。
在本发明的一些优选实施方式中,在式(G)中,R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。在本发明的一些优选实施方式中,在式(G)中,R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。在本发明的一些优选实施方式中,在式(G)中,R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN取代。在本发明的一些优选实施方式中,在式(G)中,R17、R18各自独立地选自H、甲基、乙基、丙基、3元环烷基、4元环烷基、5元环烷基、5元杂环烷基、6元杂环烷基,且任选地被一个或多个-OH、-CN取代。在本发明的一些优选实施方式中,在式(G)中,R17、R18以及与它们相连的N原子共同形成4-10元环。在本发明的一些优选实施方式中,在式(G)中,R17、R18以及与它们相连的N原子共同形成7元环。
在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5或-S-C1-4烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代;或者R17、R18以及与它们相连的N原子共同形成3-14元环。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(R17)(R18)、C1-6烷氧基;其中R17、R18各自独立地选自H、C1-6烷基、C3- 7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代;或者R17、R18以及与它们相连的N原子共同形成3-10元环。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为甲氧基、乙氧基、丙氧基、-N(H)(CH3)、-N(H)(CH2CH3)、-N(H)(CH2CH2OH)、-N(H)(CH2CH2CN)、-N(CH3)(CH3)、-N(H)(环丙基)、-N(H)(环丁基)、-N(H)(四氢呋喃基);或者R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为甲氧基。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为乙氧基。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(CH3)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(CH2CH3)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(CH2CH2OH)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(CH2CH2CN)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(CH3)(CH3)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(环丙基)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(环丁基)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(H)(四氢呋喃基)。在本发明的一些特别优选的实施方式中,在式(G)中,L为C=O,且R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环。
在本发明的一些优选实施方式中,在式(G)中,R2的个数为1、2或3个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基,其中所述-S-C1-4烷基、C1-6烷基、C3-7环烷基、4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1、2或3个,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1、2或3个,并且R2选自卤素、C1-6烷基,其中所述C1-6烷基任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1个或2个,并且R2选自卤素、C1-6烷基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1或2个,并且R2选自氟、氯、溴、甲基、乙基、正丙基、异丙基、正丁基、异丁基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1或2个,并且R2选自氟、氯、甲基、乙基、正丙基、异丙基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1或2个,并且R2选自氟、甲基、乙基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1或2个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为1个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(G)中,R2的个数为2个,并且R2选自氟、乙基。在本发明的一些特别优选的实施方式中,在式(G)中,存在2个R2,分别为氟、乙基。在本发明的一些特别优选的实施方式中,在式(G)中,存在1个R2,并且R2为乙基。
在本发明的一些优选实施方式中,在式(G)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基,其中所述-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(G)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,其中所述-S-C1-4烷基、C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(G)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-8烷基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(G)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-8烷基、C3-7环烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个或3个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基任选地被1个、2个或3个R4取代。在本发明的一些优选实施方式中,在式(G)中,R13被0个或1个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-6烷基、C3-7环烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1个、2个或3个R3取代,并且其中所述C3-7环烷基、5-7元杂环烷基任选地被1个、2个或3个R4取代。在本发明的一些优选实施方式中,在式(G)中,R13被0个或1个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-4烷基、C3-6环烷基、5-7元杂环烷基,其中所述C1-4烷基任选地被1个或2个R3取代,并且其中所述C3-6环烷基、5-7元杂环烷基任选地被1个、2个或3个R4取代。在本发明的一些特别优选的实施方式中,在式(G)中,R13被0个或1个R1取代,且各个R1独立地选自甲基、乙基、羟基、-CN、哌啶基、吗啉基、哌嗪基、环丙基,其中哌啶基、吗啉基、哌嗪基任选地被1个、2个、3个或4个C1- 3烷基取代。在本发明的一些特别优选的实施方式中,在式(G)中,R13被0个或1个R1取代,且各个R1独立地选自甲基、乙基、羟基、-CN、哌啶基、吗啉基、1-甲基哌嗪基、环丙基。在本发明的一些特别优选的实施方式中,在式(G)中,R1不存在。在本发明的一些特别优选的实施方式中,在式(G)中,R1为1-甲基哌嗪基。在本发明的一些特别优选的实施方式中,在式(G)中,R1为甲基。在本发明的一些特别优选的实施方式中,在式(G)中,R1为乙基。在本发明的一些特别优选的实施方式中,在式(G)中,R1为哌啶基。在本发明的一些特别优选的实施方式中,在式(G)中,R1为吗啉基。在本发明的一些特别优选的实施方式中,在式(G)中,R1为羟基。在本发明的一些特别优选的实施方式中,在式(G)中,R1为-CN。在本发明的一些特别优选的实施方式中,在式(G)中,R1为环丙基。
在以上各种优选实施方式中分别提到的各个取代基的优选选项可以相互组合,其各种组合形式都在本发明的范围内。
在式(G)的化合物中,当X1、X2、X3相同时,式(G)的化合物也可表示为以下式(G’)的化合物:
其中X为N或CR14,并且R14、R13、R1、L、R2的定义如式(G)的化合物中所述。
在一个优选的实施方式中,本发明提供了式(G’)的化合物:
或其同位素标记化合物、或其光学异构体、几何异构`体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物作为TRK抑制剂和/或RET抑制剂的用途,或者在制备TRK抑制剂药物和/或RET抑制剂药物中的用途,其中:
其中X为CH或N,
L为C=O、O=S=O、CH2或连接键;
R13为H、-N(R17)(R18)、C1-6烷氧基、-SR12、-OR12、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基、C7- 11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且任选地被一个或多个各自独立选自以下群组的取代基所取代:-OH、-CN、-SH、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷 氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;或者R17、R18以及与它们相连的N原子共同形成3-14元环;且
R2的个数为0、1、2、3或4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且
R1选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;且
R3、R4各自独立地选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1- 6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R5)(R6)、-N(R11)(C(=O)R12)、-CON(R7)(R8)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-6烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且
R5、R6、R7、R8、R9、R10、R11、R12各自独立地是H或选自以下群组:C1- 6烷基、C1-4卤代烷基、C3-7环烷基、4-14元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CF3、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、氧代、C1-4烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
在本发明的一些优选实施方式中,使用了上述式(G’)的化合物的同位素标记化合物。在本发明的一些更优选的实施方式中,使用了式(G’)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代。
在本发明的一些优选实施方式中,在式(G)中,X为N。在本发明的一些更优选的实施方式中,在式(G)中,X为CH。
在本发明的一些更优选的实施方式中,使用了式(G’)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,X为N。在本发明的一些更优选的实施方式中,使用了式(G’)的化合物的同位素标记化合物,其中所有H各自独立地任选地被D取代,X为CH。
在本发明的一些优选实施方式中,在式(G’)中,L为C=O、O=S=O或CH2。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O。在本发明的一些特别优选的实施方式中,在式(G’)中,L为O=S=O。在本发明的一些特别优选的实施方式中,在式(G’)中,L为CH2。在本发明的另一些实施方式中,在式(G’)中,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G’)中,X为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G’)中,X为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G’)中,X为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(G’)中,X为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(G’)中,X为N,L为C=O。
在本发明的一些特别优选的实施方式中,在式(G’)中,X为N,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(G’)中,X为N,L为CH2
在本发明的一些特别优选的实施方式中,在式(G’)中,X为N,L为连接键。
在本发明的一些优选实施方式中,在式(G’)中,R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3- 7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代(其中R13任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(G’)中,R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基,且R17、R18如上文所限定(其中R13任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(G’)中,R13为H、-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,且R17、R18如上文所限定(其中R13任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(G’)中,R13为-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、4-6元杂环烷基、苯基、5-6元杂芳基,且R17、R18如上文所限定(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些优选实施方式中,在式(G’)中,R13为-N(R17)(R18)、C1-3烷氧基、C3-6环烷基、4-6元杂环烷基、苯基、5-6元杂芳基或者C1-4烷基,且R17、R18如上文所限定(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些优选实施方式中,在式(G’)中,R13为-N(H)(C1-3烷基)、-N(H)(3-6元环烷基)、-N(H)(4-6元杂环烷基)、-N(C1-3烷基)(C1-3烷基)、C1-3烷氧基、C3-6环烷基、4-6元氮杂环烷基或氧杂环烷基、苯基、5-6元氮杂芳基或者C1-4烷基;或者R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成4-10元环(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为环丙基、环丁基、环戊基、环己基、甲基、乙基、丙基、丁基、甲氧基、乙氧基、丙氧基、-N(H)(CH3)、-N(H)(CH2CH3)、-N(H)(CH2CH2OH)、-N(H)(CH2CH2CN)、-N(CH3)(CH3)、-N(H)(环丙基)、-N(H)(环丁基)、-N(H)(四氢呋喃基)、吡嗪基、哒嗪基、吡咯烷基、吡唑基、哌啶基、苯基、氮杂环丁基、吗啉基、哌嗪基或四氢吡喃基;或者R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环(其中R13任选地被1个、2个或3个R1所取代)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为环丙基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为环丁基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为环戊基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为环己基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为甲基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为乙基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为丙基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为丁基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为吡嗪基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为哒嗪基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为吡咯烷基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为吡唑基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为哌啶基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为苯基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为氮杂环丁基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为吗啉基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为哌嗪基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为四氢吡喃基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为甲氧基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为乙氧基。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(CH3)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(CH2CH3)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(CH2CH2OH)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(CH2CH2CN)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(CH3)(CH3)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(环丙基)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(环丁基)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(H)(四氢呋喃基)。在本发明的一些特别优选的实施方式中,在式(G’)中,R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环。
在本发明的一些优选实施方式中,在式(G’)中,R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。在本发明的一些优选实施方式中,在式(G’)中,R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。在本发明的一些优选实施方式中,在式(G’)中,R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN取代。在本发明的一些优选实施方式中,在式(G’)中,R17、R18各自独立地选自H、甲基、乙基、丙基、3元环烷基、4元环烷基、5元环烷基、5元杂环烷基、6元杂环烷基,且任选地被一个或多个-OH、-CN取代。在本发明的一些优选实施方式中,在式(G’)中,R17、R18以及与它们相连的N原子共同形成4-10元环。在本发明的一些优选实施方式中,在式(G’)中,R17、R18以及与它们相连的N原子共同形成7元环。
在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5或-S-C1-4烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代;或者R17、R18以及与它们相连的N原子共同形成3-14元环。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(R17)(R18)、C1-6烷氧基;其中R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代;或者R17、R18以及与它们相连的N原子共同形成3-10元环。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为甲氧基、乙氧基、丙氧基、-N(H)(CH3)、-N(H)(CH2CH3)、-N(H)(CH2CH2OH)、-N(H)(CH2CH2CN)、-N(CH3)(CH3)、-N(H)(环丙基)、-N(H)(环丁基)、-N(H)(四氢呋喃基);或者R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为甲氧基。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为乙氧基。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(CH3)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(CH2CH3)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(CH2CH2OH)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(CH2CH2CN)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(CH3)(CH3)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(环丙基)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(环丁基)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(H)(四氢呋喃基)。在本发明的一些特别优选的实施方式中,在式(G’)中,L为C=O,且R13为-N(R17)(R18),且R17、R18以及与它们相连的N原子共同形成7元环。
在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1、2或3个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基,其中所述-S-C1-4烷基、C1-6烷基、C3-7环烷基、4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1、2或3个,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1、2或3个,并且R2选自卤素、C1-6烷基,其中所述C1-6烷基任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1个或2个,并且R2选自卤素、C1-6烷基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1或2个,并且R2选自氟、氯、溴、甲基、乙基、正丙基、异丙基、正丁基、异丁基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1或2个,并且R2选自氟、氯、甲基、乙基、正丙基、异丙基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1或2个,并且R2选自氟、甲基、乙基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1或2个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为1个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(G’)中,R2的个数为2个,并且R2选自氟、乙基。在本发明的一些特别优选的实施方式中,在式(G’)中,存在2个R2,分别为氟、乙基。在本发明的一些特别优选的实施方式中,在式(G’)中,存在1个R2,并且R2为乙基。
在本发明的一些优选实施方式中,在式(G’)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基,其中所述-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(G’)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,其中所述-S-C1-4烷基、C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(G’)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-8烷基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(G’)中,R13被0、1、2、3或4个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-8烷基、C3-7环烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个或3个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基任选地被1个、2个或3个R4取代。在本发明的一些优选实施方式中,在式(G’)中,R13被0个或1个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-6烷基、C3-7环烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1个、2个或3个R3取代,并且其中所述C3-7环烷基、5-7元杂环烷基任选地被1个、2个或3个R4取代。在本发明的一些优选实施方式中,在式(G’)中,R13被0个或1个R1取代,且各个R1独立地选自卤素、-OH、-CN、C1-4烷基、C3-6环烷基、5-7元杂环烷基,其中所述C1-4烷基任选地被1个或2个R3取代,并且其中所述C3-6环烷基、5-7元杂环烷基任选地被1个、2个或3个R4取代。在本发明的一些特别优选的实施方式中,在式(G’)中,R13被0个或1个R1取代,且各个R1独立地选自甲基、乙基、羟基、-CN、哌啶基、吗啉基、哌嗪基、环丙基,其中哌啶基、吗啉基、哌嗪基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些特别优选的实施方式中,在式(G’)中,R13被0个或1个R1取代,且各个R1独立地选自甲基、乙基、羟基、-CN、哌啶基、吗啉基、1-甲基哌嗪基、环丙基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1不存在。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为1-甲基哌嗪基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为甲基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为乙基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为哌啶基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为吗啉基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为羟基。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为-CN。在本发明的一些特别优选的实施方式中,在式(G’)中,R1为环丙基。
在以上各种优选实施方式中分别提到的各个取代基的优选选项可以相互组合,其各种组合形式都在本发明的范围内。
在式(G’)的化合物中,当R13为环时,式(G’)的化合物也可表示为下式(I)的化合物:
其中环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基或5-11元双环杂烷基,其可任选地被R1取代,并且L、R1、R2、X的定义如上文关于式(G’)的化合物所述。
特别地,本发明提供了式(I)的化合物:
或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物作为TRK抑制剂药物和/或RET抑制剂药物的用途,或者在制备TRK抑制剂药物和/或RET抑制剂药物中的用途,其中:
L为C=O、O=S=O、CH2或连接键;
X为CH或N;
环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基;
R1的个数为0、1、2、3或4个,并且R1选自H、卤素、C1-8烷基、C2- 8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基,其中所述C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;
R2的个数为0、1、2、3、4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基以及4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12
R3选自卤素、氰基、C1-3烷基、羟基、C1-6烷氧基、-N(R5)(R6)、-CON(R7)(R8)或3-7元杂环烷基,其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
R4选自卤素、C1-3烷基、羟基、C1-6烷氧基、-NH2、-NHCH3或-N(CH3)2
R5、R6、R7、R8各自独立地为氢或C1-4烷基;
R9选自H、C1-4烷基、C1-4卤代烷基或C3-7环烷基;
R10是H或选自以下群组:C1-4烷基、C1-4卤代烷基、C3-7环烷基、4-10元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3- 7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基;
R11选自H、C1-4烷基以及C3-7环烷基;
R12选自C1-6烷基、C3-7环烷基、4-至14-元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CF3、-CN、-OH、-NH2、-NH(CH3)、-N(CH3)2、氧代、-S-C1-4烷基、C1-4烷基、C1-4卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4烷氧基以及C1-4卤代烷氧基。
在本发明的一些优选实施方式中,在式(I)中,L为C=O、O=S=O或CH2。在本发明的一些特别优选的实施方式中,在式(I)中,L为C=O。在本发明的一些特别优选的实施方式中,在式(I)中,L为O=S=O。在本发明的一些特别优选的实施方式中,在式(I)中,L为CH2。在本发明的另一些实施方式中,在式(I)中,L为连接键。
在本发明的一些特别优选的实施方式中,在式(I)中,X为CH。在本发明的另一些实施方式中,在式(I)中,X为N。
在本发明的一些特别优选的实施方式中,在式(I)中,X为CH,L为C=O。
在本发明的一些特别优选的实施方式中,在式(I)中,X为CH,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(I)中,X为CH,L为CH2
在本发明的一些特别优选的实施方式中,在式(I)中,X为CH,L为连接键。
在本发明的一些特别优选的实施方式中,在式(I)中,X为N,L为C=O。
在本发明的一些特别优选的实施方式中,在式(I)中,X为N,L为O=S=O。
在本发明的一些特别优选的实施方式中,在式(I)中,X为N,L为CH2
在本发明的一些特别优选的实施方式中,在式(I)中,X为N,L为连接键。
在本发明的一些优选实施方式中,在式(I)中,环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(I)中,环A为C5-6环烷基、5-6元杂环烷基、苯基、5-6元杂芳基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(I)中,环A为5-6元杂环烷基、苯基、5-6元杂芳基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(I)中,环A为5-6元氮杂环烷基、苯基、5-6元氮杂芳基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(I)中,环A为吡嗪基、吡唑基、哌啶基或苯基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些特别优选的实施方式中,在式(I)中,环A为吡嗪基。在本发明的一些特别优选的实施方式中,在式(I)中,环A为吡唑基。在本发明的一些特别优选的实施方式中,在式(I)中,环A为哌啶基。在本发明的一些特别优选的实施方式中,在式(I)中,环A为苯基。
在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,其中所述C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-8烷基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-8烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1个、2个、3个或4个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-4烷基、5-7元杂环烷基,其中所述C1-4烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自甲基、哌啶基、吗啉基、哌嗪基,其中哌啶基、吗啉基、哌嗪基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自甲基、哌啶基、吗啉基、1-甲基哌嗪基。在本发明的一些特别优选的实施方式中,在式(I)中,R1不存在。在本发明的一些特别优选的实施方式中,在式(I)中,R1为1-甲基哌嗪基。在本发明的一些特别优选的实施方式中,在式(I)中,R1为甲基。在本发明的一些特别优选的实施方式中,在式(I)中,R1为哌啶基。在本发明的一些特别优选的实施方式中,在式(I)中,R1为吗啉基。
在本发明的一些优选实施方式中,在式(I)中,R2的个数为1、2或3个,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1、2或3个,并且R2选自卤素、C1-6烷基,其中所述C1-6烷基任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、 C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1或2个,并且R2选自氟、氯、溴、甲基、乙基、正丙基、异丙基、正丁基、异丁基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1或2个,并且R2选自氟、氯、甲基、乙基、正丙基、异丙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1或2个,并且R2选自氟、甲基、乙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1或2个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为2个,并且R2选自氟、乙基。在本发明的一些特别优选的实施方式中,在式(I)中,存在2个R2,分别为氟、乙基。在本发明的一些特别优选的实施方式中,在式(I)中,存在1个R2,并且R2为乙基。
在以上各种优选实施方式中分别提到的各个取代基的优选选项可以相互组合,其各种组合形式都在本发明的范围内。
特别地,本发明提供了式(I)的化合物:
或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物作为TRK抑制剂药物和/或RET抑制剂药物的用途,或者在制备TRK抑制剂药物和/或RET抑制剂药物中的用途,其中:
L为C=O或O=S=O;
X为CH;
环A为5-7元杂芳基、C5-7芳基;
R1的个数为0、1、2、3或4个,并且R1选自C1-8烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
R2的个数为1、2或3个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基以及4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12
R3选自卤素、氰基、C1-3烷基、羟基、C1-6烷氧基、-N(R5)(R6)、-CON(R7)(R8)或3-7元杂环烷基,其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
R4选自卤素、C1-3烷基、羟基、C1-6烷氧基、-NH2、-NHCH3或-N(CH3)2
R5、R6、R7、R8各自独立地为氢或C1-4烷基;
R9选自H、C1-4烷基、C1-4卤代烷基或C3-7环烷基;
R10是H或选自以下群组:C1-4烷基、C1-4卤代烷基、C3-7环烷基、4-10元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3- 7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基;
R11选自H、C1-4烷基以及C3-7环烷基;
R12选自C1-6烷基、C3-7环烷基、4-至14-元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CF3、-CN、-OH、-NH2、-NH(CH3)、-N(CH3)2、氧代、-S-C1-4烷基、C1-4烷基、C1-4卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4烷氧基以及C1-4卤代烷氧基。
在本发明的一些优选实施方式中,在式(I)中,L为O=S=O。在本发明的一些优选实施方式中,在式(I)中,L为C=O。
在本发明的一些优选实施方式中,在式(I)中,环A为5-6元杂芳基或苯基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些优选实施方式中,在式(I)中,环A为吡嗪基、吡唑基或苯基(其中环A任选地被1个、2个、3个或4个R1所取代)。在本发明的一些特别优选的实施方式中,在式(I)中,环A为吡嗪基。在本发明的一些特别优选的实施方式中,在式(I)中,环A为吡唑基。在本发明的一些特别优选的实施方式中,在式(I)中,环A为苯基。
在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-8烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1个、2个、3个或4个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个R4取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自C1-4烷基、5-7元杂环烷基,其中所述C1-4烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自甲基、哌啶基、吗啉基,其中哌啶基、吗啉基任选地被1个、2个、3个或4个C1-3烷基取代。在本发明的一些优选实施方式中,在式(I)中,R1不存在或R1选自甲基、哌啶基、吗啉基。在本发明的一些特别优选的实施方式中,在式(I)中,R1不存在。在本发明的一些特别优选的实施方式中,在式(I)中,R1为甲基。在本发明的一些特别优选的实施方式中,在式(I)中,R1为哌啶基。在本发明的一些特别优选的实施方式中,在式(I)中,R1为吗啉基。
在本发明的一些优选实施方式中,在式(I)中,R2的个数为1或2个,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为1或2个,并且R2选自卤素、C1-6烷基,其中所述C1-6烷基任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为2个,并且R2选自氟、氯、溴、甲基、乙基、正丙基、异丙基、正丁基、异丁基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为2个,并且R2选自氟、氯、甲基、乙基、正丙基、异丙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为2个,并且R2选自氟、甲基、乙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为2个,并且R2选自氟、乙基。在本发明的一些优选实施方式中,在式(I)中,R2的个数为2个,并且R2选自氟、乙基。在本发明的一些特别优选的实施方式中,在式(I)中,存在2个R2,分别为氟、乙基。
在以上各种优选实施方式中分别提到的各个取代基的优选选项可以相互组合,其各种组合形式都在本发明的范围内。
在本发明的优选的实施方式中,用作作为TRK抑制剂药物和/或RET抑制剂药物的化合物选自:
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-(哌啶-1-基)吡嗪-2-基)甲酮(MDI-2);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(1-甲基-1H-吡唑-4-基)甲酮(MDI-202);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(1-甲基哌啶-4-基)甲酮(MDI-203);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-204);
(2-(6-(2-乙基-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-205);
5-乙基-2-氟-4-(3-(5-(苯磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-206);
5-乙基-2-氟-4-(3-(5-(吡嗪-2基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-207);
4-(3-(5-(环丙基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-208);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233);
4-(3-(5-(环丁基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-210);
环丁基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-211);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(3-羟基环丁基)甲酮(MDI-213);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪唑-5-(1H,4H,6H)-基)(哒嗪-4-基)甲酮(MDI-214);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪唑-5-(1H,4H,6H)-基)(哒嗪-3-基)甲酮(MDI-215);
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228);
5-乙基-2-氟-4-(3-(5-(4-羟基环己基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-217);
4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218);
4-(3-(5-(环丁基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-219);
4-(3-(5-(环戊基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-220);
5-乙基-2-氟-4-(3-(5-((1-甲基-1H-吡唑-4-基)甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-221);
4-(3-(5-(环戊烷基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-224);
5-乙基-2-氟-4-(3-(5-(四氢-2H-吡喃-4-基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-225);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)乙-1-酮(MDI-226);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)丙-1-酮(MDI-227);
(1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)-2-甲基丙-1-酮)(MDI-228);
2-环丙基-1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)基)乙-1-酮(MDI-229);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)-3-甲基丁-1-酮(MDI-230);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231);
N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-b]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-237);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-239);
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-240);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-c]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-242);
(R)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-243);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-甲基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-246);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-5-羰基)吡咯烷-3-腈(MDI-250);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(四氢呋喃-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-251);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸甲酯(MDI-252);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253);
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[3,4-b]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-255);
3-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)-3-氧丙腈(MDI-256);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257);
N-(2-氰基乙基)-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-258);
N-环丙基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-259);
N-环丁基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-260);
(S)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-262);
(R)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-263)。
在本发明的最优选的实施方式中,所使用的化合物选自:
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201)
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233)
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228)
4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218)
N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257)
为了简明起见,后文所述“式(G)所示的化合物”或“式(G)的化合物”或“本发明的化合物”也涵盖式(G)的化合物的任意光学异构体、几何异构体、互变异构体或异构体的混合物;后文所述“式(G’)所示的化合物”或“式(G’)的化合物”或“本发明的化合物”也涵盖式(G’)的化合物的任意光学异构体、几何异构体、互变异构体或异构体的混合物;后文所述“式(I)所示的化合物”或“式(I)的化合物”或“本发明的化合物”也涵盖式(I)的化合物的任意光学异构体、几何异构体、互变异构体或异构体的混合物。
术语“光学异构体”意指,当化合物具有一个或更多个手性中心时,每个手性中心可以存在R构型或S构型,由此构成的各种异构体为光学异构体。光学异构体包括所有的非对映异构体、对映异构体、内消旋体、外消旋体或其混合物形式。例如,通过手性色谱柱或通过手性合成可以分离光学异构体。
术语“几何异构体”意指,当化合物中存在双键时,该化合物可以存在顺式异构体、反式异构体、E型异构体和Z型异构体。几何异构体包括顺式异构体、反式异构体、E型异构体、Z型异构体或其混合物形式。
术语“互变异构体”指因分子中某一原子在两个位置迅速移动而产生的异构体。本领域技术人员可以理解:互变异构体之间可以互相转变,在在某一状态下可能会达到一种平衡状态而共存。本文所述“式(G)所示的化合物”也涵盖式(G)的化合物的任意互变异构体;本文所述“式(G’)所示的化合物”也涵盖式(G’)的化合物的任意互变异构体;本文所述“式(I)所示的化合物”也涵盖式(I)的化合物的任意互变异构体。
除非另有指明,本文提到“式(G)所示的化合物”或“式(G)的化合物”或“本发明的化合物”时也涵盖该化合物中任一个原子被其同位素原子代替而得到的同位素标记化合物;本文提到“式(G’)所示的化合物”或“式(G’)的化合物”或“本发明的化合物”时也涵盖该化合物中任一个原子被其同位素原子代替而得到的同位素标记化合物;本文提到“式(I)所示的化合物”或“式(I)的化合物”或“本发明的化合物”时也涵盖该化合物中任一个原子被其同位素原子代替而得到的同位素标记化合物。
本发明包括式(G)的化合物的所有药学上可接受的同位素标记化合物,其中,一个或者多个原子被具有与通常在自然界中所发现的原子相同原子序数但是不同原子质量或者质量数的原子所替换。本发明包括式(G’)的化合物的所有药学上可接受的同位素标记化合物,其中,一个或者多个原子被具有与通常在自然界中所发现的原子相同原子序数但是不同原子质量或者质量数的原子所替换。本发明包括式(I)的化合物的所有药学上可接受的同位素标记化合物,其中,一个或者多个原子被具有与通常在自然界中所发现的原子相同原子序数但是不同原子质量或者质量数的原子所替换。
适用于包含在本发明的化合物中的同位素的实例包括氢的同位素,诸如2H(D)和3H(T),碳的同位素,诸如11C、13C和14C,氯的同位素,诸如36Cl,氟的同位素,诸如18F,碘的同位素,诸如123I和125I,氮的同位素,诸如13N和15N,氧的同位素,诸如15O、17O和18O,以及硫的同位素, 诸如35S。
式(G)的某些同位素标记化合物(例如包含放射性同位素的那些)可用于药物和/或底物组织分布研究;式(G’)的某些同位素标记化合物(例如包含放射性同位素的那些)可用于药物和/或底物组织分布研究;式(I)的某些同位素标记化合物(例如包含放射性同位素的那些)可用于药物和/或底物组织分布研究。考虑到引入的容易性和检测手段的方便性,放射性同位素氘(即2H)和碳-14(即14C)对于该目的是特别有用的。
利用诸如氘(即2H)的较重同位素进行取代可以提供某些治疗方面的好处并且因此在某些情况下可能是优选的,所述治疗方面的好处是由更大的代谢作用稳定性(例如,增长的体内半衰期或者减小的剂量要求)带来的。
利用正电子放射同位素(诸如11C、18F、15O和13N)进行取代可以用于正电子放射受体图像(Positron Emission Topography(PET))研究,用于检测底物受体占用状态。
式(G)的同位素标记化合物一般可以通过本领域技术人员已知的常规技术或者通过使用合适的同位素标记试剂代替先前使用的非标记试剂以类似于在本文所附的实例和制备中所描述的方法,来进行制备。式(G’)的同位素标记化合物一般可以通过本领域技术人员已知的常规技术或者通过使用合适的同位素标记试剂代替先前使用的非标记试剂以类似于在本文所附的实例和制备中所描述的方法,来进行制备。式(I)的同位素标记化合物一般可以通过本领域技术人员已知的常规技术或者通过使用合适的同位素标记试剂代替先前使用的非标记试剂以类似于在本文所附的实例和制备中所描述的方法,来进行制备。
式(G)的化合物可以药学上可接受的盐的形式存在,比如,式(G)的化合物的酸加成盐和/或碱加成盐。除非另有指明,否则本文所用的“药学上可接受的盐”包括可出现于式(G)的化合物内的酸加成盐或碱加成盐。式(G’)的化合物可以药学上可接受的盐的形式存在,比如,式(G’)的化合物的酸加成盐和/或碱加成盐。除非另有指明,否则本文所用的“药学上可接受的盐”包括可出现于式(G’)的化合物内的酸加成盐或碱加成盐。式(I)的化合物可以药学上可接受的盐的形式存在,比如,式(I)的化合物的酸加成盐和/或碱加成盐。除非另有指明,否则本文所用的“药学上可接受的盐”包括可出现于式(I)的化合物内的酸加成盐或碱加成盐。
式(G)的化合物、式(G’)的化合物和式(I)的化合物的药学上可接受的盐类包括其酸加成盐和碱加成盐。适当的酸加成盐是由形成无毒性盐的酸所形成的。其实例包括但不限于:乙酸盐、己二酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、碳酸氢盐/碳酸盐、硫酸氢盐/硫酸盐、硼酸盐、樟脑磺酸盐、柠檬酸盐、环己胺磺酸盐、乙二磺酸盐、甲酸盐、反丁烯二酸盐、葡萄庚糖酸盐、葡萄糖酸盐、葡萄糖醛酸盐、六氟磷酸盐、2-(4-羟苄基)苯甲酸盐、氢氯化物/氯化物、氢溴化物/溴化物、氢碘化物/碘化物、2-羟乙磺酸盐、乳酸盐、苹果酸盐、顺丁烯二酸盐、丙二酸盐、甲磺酸盐、甲基硫酸盐、萘酸盐、2-萘磺酸盐、烟碱酸盐、硝酸盐、乳清酸盐、草酸盐、十六酸盐、磷酸盐/磷酸氢盐/磷酸二氢盐、焦谷氨酸盐、葡萄糖二酸盐、硬脂酸盐、水杨酸盐、单宁酸盐、酒石酸盐、甲苯磺酸盐和三氟乙酸盐。适当的碱加成盐是由形成无毒性盐的碱所形成的。其实例包括但不限于:铝、精氨酸、钙、胆碱、二乙胺、二乙醇胺、甘氨酸、赖氨酸、镁、葡甲胺、乙醇胺、钾、钠、氨丁三醇和锌盐。还可形成酸和碱的半盐,例如半硫酸盐和半钙盐。关于合适的盐的综述,参见Handbook of Pharmaceutical Salts:Properties,Selection and Use by Stahl and Wermuth(Wiley-VCH,2002)。用于制备本文中所述的化合物的药学上可接受的盐的方法是本领域技术人员已知的。
本发明的某些化合物可以以非溶剂化形式以及溶剂化形式(包括水合形式)存在。一般而言,式(G)的化合物、式(G’)的化合物以及式(I)的化合物,无论以溶剂化形式存在或以未溶剂化形式存在,其都包括在本发明的范围内。
本发明的某些化合物可以不同晶型或不定型形式存在,无论以何种形式存在,式(G)的化合物、式(G’)的化合物以及式(I)的化合物都包括在本发明的范围内。
为了避免歧义,下面对本文中所使用的术语给出定义。除非另有说明,本文所用术语的含义如下。
术语“药学上可接受的”是指相应的化合物、载体或分子适于给予人。优选地,该术语是指由管理机构例如CFDA(中国)、EMEA(欧洲)、FDA(美国)等任意国家管理机构认证的用于哺乳动物优选人。
“前体药”是指通过与酶、胃酸等在生理条件下在活体内例如通过各自在酶催化下进行的氧化、还原、水解等反应转化为本发明化合物的衍生物。
“代谢物”是指在细胞或有机体优选人中源自本发明任意化合物的所有分子。
术语“羟基”是指-OH。
术语“卤素”或“卤“是指-F,-Cl,-Br,或-I。
术语“氰基”是指-CN。
在本发明中,当某类取代基存在多个时,每个取代基是彼此独立地选择的,即这些取代基可以相同也可以不同。例如,当存在2个、3个或4个R1时,这些R1可以相同也可以不同。例如,当存在2个、3个或4个R2时,这些R2可以相同也可以不同。例如,当R1和R2均为-N(R9)(R10)时,R1和R2中的R9和R10可以独立地选择,即,R1中的R9与R2中的R9可以相同也可以不同,R1中的R10与R2中的R10可以相同也可以不同。例如,当存在2个R1,这2个R1均为-N(R9)(R10)时,这2个R1中的R9和R10可以独立地选择,即,第一个R1中的R9与第二个R1中的R9可以相同也可以不同,第一个R1中的R10与第二个R1中的R10可以相同也可以不同。以上说明适用于R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18
在本文中使用时,术语“被取代”是指基团中的一个或多个(优选1至5个,更优选1至3个)氢原子独立地被相应数目的取代基所代替。
在本文中使用时,术语“独立地”是指当取代基的个数超过一个时,这些取代基可以相同也可以不同。
在本文中使用时,术语“任选”或“任选地”表示其所描述的事件可以发生或不发生。例如,一个基团“任选地被取代”表示:该基团可以是未被取代的,也可以是被取代的。
在本文中使用时,术语“杂原子”代表氧(O)、氮(N)、或S(O)m(其中m可以是0、1或2,即硫原子S、或亚砜基SO、或磺酰基S(O)2)。
在本文中使用时,术语“烷基”是指饱和的脂族烃,包括直链及支链。在一些实施方式中,烷基基团具有1-8个、或1-6个、或1-3个碳原子。例如,术语“C1-8烷基”是指具有1-8个碳原子的直链或支链原子团。术语“C1-8烷基”在其定义中包括术语“C1-6烷基”、“C1-C3烷基”和“C1-C4烷基”。烷基的实例包括但不限于甲基、乙基、丙基、异丙基、丁基、异丁基、仲丁基、叔丁基、戊基、2-戊基、3-戊基、异戊基、新戊基、(R)-2-甲基丁基、(S)-2-甲基丁基、3-甲基丁基、2,3-二甲基丙基、2,3-二甲基丁基、己基等。烷基基团可任选地被一或多个(例如,1至5个)适当的取代基所取代。
在本文中使用时,术语“烯基”是指具有至少一个碳-碳双键的脂族烃,包括具有至少一个碳-碳双键的直链及支链。在一些实施方式中,烯基基团具有2-8个碳原子、2-6个碳原子、3-6个碳原子、或2-4个碳原子。例如,术语“C2-8烯基”是指具有2-8个碳原子的直链或支链的不饱和原子团(具有至少一个碳-碳双键)。所述双键可以是或者可以不是另一基团的连接点。烯基包括但不限于乙烯基、1-丙烯基、2-丙烯基、2-甲基-2-丙烯基、丁烯基、戊烯基、3-己烯基等。烯基基团可任选地被一或多个(例如,1至5个)适当的取代基所取代。当式(I)的化合物含有烯基基团时,该烯基基团可以纯E形式、纯Z形式、或其任何混合物存在。
在本文中使用时,术语“炔基”是指具有至少一个碳-碳三键的脂族烃,包括具有至少一个碳-碳三键的直链及支链。在一些实施方式中,炔基基团具有2-8个碳原子、2-6个碳原子、3-6个碳原子、或2-4个碳原子。例如,术语“C2-8炔基”是指具有2-8个碳原子的直链或支链的不饱和原子团(具有至少一个碳-碳三键)。所述三键可以是或者可以不是另一基团的连接点。 炔基包括但不限于乙炔基、1-丙炔基、2-丙炔基、2-甲基-2-丙炔基、丁炔基、戊炔基、3-己炔基等。炔基基团可任选地被一或多个(例如,1至5个)适当的取代基所取代。
在本文中使用时,术语“C3-7环烷基”是指具有3-7个形成环的碳原子的环烷基,例如环丙基、环丁基、环戊基、环己基、环己烯基、环庚基。环烷基可任选地被一或多个适当的取代基所取代。
在本文中使用时,术语“n元杂环烷基”是指具有m个形成环的碳原子和(n-m)个形成环的杂原子的环烷基,所述杂原子选自O、S及N。例如,3-7元杂环烷基包括但不限于氧杂环丁烷、硫杂环丁烷、氮杂环丁烷、四氢呋喃、四氢噻吩、吡咯烷、四氢吡喃、四氢噻喃、哌啶、吗啉、哌嗪、氧杂环庚烷、硫杂环庚烷、氮杂环庚烷。杂环烷基可任选地被一或多个适当的取代基所取代。
在本文中使用时,术语“C5-7芳基”是指具有含5-7个碳原子的芳环的芳基,优选为苯基。
在本文中使用时,术语“n元杂芳基”是指具有m个形成芳环的碳原子和(n-m)个形成芳环的杂原子的杂芳基,所述杂原子选自O、S及N。例如,5-7元杂芳基包括但不限于吡嗪、吡唑、吡咯、呋喃、噻吩、噻唑、吡啶。杂芳基可任选地被一或多个适当的取代基所取代。
在本文中使用时,术语“C7-11双环芳基”是指具有7-11个碳原子的双环芳基,例如萘、茚等。双环芳基可任选地被一或多个适当的取代基所取代。
在本文中使用时,术语“n元双环杂芳基”是指具有m个形成芳族双环的碳原子和(n-m)个形成芳族双环的杂原子的双环杂芳基,所述杂原子选自O、S及N。例如,7-11元双环杂芳基包括但不限于喹啉、异喹啉、苯并噻唑等。双环杂芳基可任选地被一或多个适当的取代基所取代。
在本文中使用时,术语“11-15元三环基”包括但不限于吖啶等。11-15元三环基可任选地被一或多个适当的取代基所取代。
在本文中使用时,术语“卤代烷基”是指具有一或多个卤素取代基的烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代)。例如,术语“C1-6卤代烷基”是指具有一或多个卤素取代基的C1-6烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代)。另举一例,术语“C1-4卤代烷基”是指具有一或多个卤素取代基的C1-4烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代);术语“C1-3卤代烷基”是指具有一或多个卤素取代基的C1-3烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代);且术语“C1-2卤代烷基”是指具有一或多个卤素取代基的C1-2烷基基团(即,甲基或乙基)(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代)。再另举一例,术语“C1卤代烷基”是指具有1、2或3个卤素取代基的甲基基团。卤代烷基基团的例子包括:CF3、C2F5、CHF2、CH2F、CH2CF3、CH2Cl等。
在本文中使用时,术语“烷氧基”是指单键连接至氧原子的烷基。烷氧基与分子的连接点是通过氧原子。烷氧基可被描述为烷基-O-。术语“C1-6烷氧基”是指包含1-6个碳原子的直链或支链的烷氧基。术语“C1-6烷氧基”在其定义中包括术语“C1-3烷氧基”。烷氧基包括但不限于甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、己氧基等。烷氧基可任选地被一或多个适当的取代基所取代。
本文中,与取代基个数、碳原子个数、环原子个数相关的数目范围表示该范围内所有整数的逐个列举,而范围仅是作为一种简化的表示法。例如:
“1-4个取代基”表示1、2、3或4个取代基;
“1-3个取代基”表示1、2或3取代基;
“3-12元环”表示3、4、5、6、7、8、9、10、11或12元环;
“3-14元环”表示3、4、5、6、7、8、9、10、11、12、13或14元环;
“3-8元环”表示3、4、5、6、7或8元环;
“1-12个碳原子”或C1-12”表示1个(C1)、2个(C2)、3个(C3)、4个(C4)、5个(C5)、6个(C6)、7个(C7)、8个(C8)、9个(C9)、10个(C10)、11个(C11)或12个碳原子(C12);
“1-6个碳原子”或“C1-6”表示1个(C1)、2个(C2)、3个(C3)、4个(C4)、5个(C5)或6个碳原子(C6);
“1-4个碳原子”或“C1-4”表示1个(C1)、2个(C2)、3个(C3)或4个碳原子(C4);
“2-6个碳原子”或“C2-6”表示2个(C2)、3个(C3)、4个(C4)、5个(C5)或6个碳原子(C6);
“C3-8”表示3个(C3)、4个(C4)、5个(C5)、6个(C6)、7个(C7)或8个碳原子(C8);
“3-8个环原子”表示3个、4个、5个、6个、7个或8个环原子。
因此,与取代基个数、碳原子个数、环原子个数相关的数目范围也涵盖其任意一个子范围,且每一个子范围也视为被本文公开。
本发明使用的式(G)的化合物、式(G’)的化合物或式(I)的化合物是已知化合物,可以用有机合成领域的技术人员所熟悉的多种方法合成,例如中国发明专利CN111606908B中所公开的方法。
本领域技术人员可以理解,本发明使用的式(G)的化合物、式(G’)的化合物或式(I)的化合物可以作为“TRK抑制剂和/或RET抑制剂”表示它们既可以是TRK抑制剂,也可以是TRK抑制剂,还可以是TRK/RET双重抑制剂。同时,根据中国发明专利CN111606908B中所公的内容,这些化合物还可以作为JAK抑制剂,因此本发明使用的式(G)的化合物、式(G’)的化合物或式(I)的化合物可以作为JAK/TRK/RET多重抑制剂发挥作用,即它们可以是:JAK/TRK双重抑制剂、JAK/RET双重抑制剂、TRK/RET双重抑制剂、或JAK/TRK/RET三重抑制剂。因为多重抑制剂同时对多个疾病靶点进行抑制,这使得本发明所使用的化合物在治疗某些疾病时效果比传统的单靶点抑制剂效果更佳(如后文实施例所示)。因此,本发明使用的式(G)的化合物、式(G’)的化合物或式(I)的化合物优选作为JAK/TRK/RET多重抑制剂发挥作用。在特别优选的实施方式中,本发明使用的式(G)的化合物、式(G’)的化合物或式(I)的化合物作为JAK/TRK双重抑制剂发挥作用;其中,一些特别优选的化合物可以作为Pan-JAK/Pan-TRK双重抑制剂(即同时抑制JAK激酶家族和TRK激酶家族的所有成员)发挥作用,因此使得它们在自免疫疾病、瘙痒症等皮肤疾病以及糖尿病足等疾病的治疗中有极其出色的表现。
在第二个方面,本发明提供了一种TRK抑制剂药物组合物,其含有如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,以及一种或多种药学上可接受的载体、佐剂或赋形剂。
在第三个方面,本发明提供了一种RET抑制剂药物组合物,其含有如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,以及一种或多种药学上可接受的载体、佐剂或赋形剂。
本发明的药物组合物可根据需要配制成适用于口服、外用(包括但不限于外敷、喷涂等)、胃肠外(包括皮下、肌肉、皮层和静脉)施予、支气管施予或鼻施予的剂型。其中,优选地,本发明的药物组合物被配制成适用于口服或外用的剂型(制剂)。更优选地,本发明的药物组合物被配制成适用于口服的剂型(制剂)。
如果使用固体载剂,则该制剂可以成片,以粉末或颗粒形式置于硬质凝胶胶囊中,或以糖锭或锭剂形式。固体载剂可以包括常规的赋形剂,诸如粘合剂、填料、成片润滑剂、崩解剂、润湿剂等等。如果需要可以通过常规技术膜包衣该片剂。如果使用液体载剂,则该制剂可以是糖浆、乳液、膏剂、软凝胶胶囊、用于注射的无菌载体、水性或非水性液体悬浮液形式的,或者可以是在使用前用水或其他适当载体复原的干品。液体制剂可以包含常规添加剂,诸如悬浮剂、乳化剂、润湿剂、非水性载体(包括可食用油)、防腐剂以及香味剂和/或着色剂。为了胃肠外施予,通常载体至少大部分包括无菌水,但也可以使用盐水溶液、葡萄糖溶液等。也可以使用可注射悬浮液,在这种情况下,可以使用常规悬浮剂。常规防腐剂、缓冲试剂等也可以添加到胃肠外剂型中。药物组合物通过对包含适量的活性成分(即本发明的式(G)的化合物、式(G’)的化合物或式(I)的化合物)的所需制剂合适的常规技术制备。
适于非肠道注射的组合物可以包括生理学上可接受无菌水性或非水性溶液、分散液、悬浮液或乳液和用于无菌可注射溶液或分散液的无菌粉末。合适的水性和非水性载体、稀释剂、溶剂的实例包括水、乙醇、多元醇(丙二醇、聚乙二醇、丙三醇等)、其合适的混合物、植物油(例如,橄榄油)和可注射有机酯(例如,油酸乙酯)。
这些组合物还可以包含各种赋形剂,例如,防腐剂、润湿剂、乳化剂和分散剂。可以通过各种抗菌剂和抗真菌剂(例如,对羟基苯甲酸酯、氯丁醇、苯酚、山梨酸等)来确保对微生物的作用的抑制。还可以包括等渗剂,例如,糖、氯化钠等。可以通过使用延迟吸收试剂(例如,单硬脂酸铝和凝胶)来延长可注射药学剂型的吸收。
用于口服的固体剂型包括胶囊、药片、药丸、粉末和颗粒。在这种固体剂型中,将活性化合物与至少一种惰性赋型剂(或载体)(例如,柠檬酸钠或磷酸二钙)混合,其中还可以包括:(a)填料或混合剂(例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸);(b)粘结剂(例如,羧基甲基纤维素、褐藻酸酯、凝胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯树胶);(c)保湿剂(例如,丙三醇);(d)崩解剂(例如,琼脂-琼脂、碳酸钙、马铃薯或木薯淀粉、褐藻酸、某些合成的硅酸酯、碳酸钠);(e)溶液阻滞剂(例如,石蜡);(f)吸收促进剂(例如,季铵化合物);(g)润湿剂(例如,十六烷醇和单硬脂酸丙三醇酯);(h)吸附剂(例如,高岭土和斑脱土)和(i)润滑剂(例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、月桂基硫酸钠)或其混合物混合。
类似类型的固体组合物还可以在使用例如乳糖以及高分子量聚乙二醇等作为赋型剂的软填充和硬填充凝胶胶囊中作为填料。
固体剂型(例如,药片、糖衣丸、胶囊、药丸和颗粒)可以采用涂层和外壳(例如,肠道涂层和本领域已知的其它)来制备。它们可以包含遮光剂,它们还可以是以延迟方式在肠道的某一部分中释放活性化合物或各种活性化合物的组合物。可用的包埋组合物的实例是聚合物质和蜡。活性组分还可以以微胶囊化形式,如果适当的话,可以具有一种或更多种上述赋型剂。
用于口服的液体剂型包括药学上可接受乳液、溶液、分散液、糖浆和酏剂。除了活性化合物以外,液体剂型可以包含本领域中通常所用的惰性稀释剂(例如,水或其它溶剂)、增溶剂和乳化剂(例如,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、苯甲醇、苯甲酸苯甲酯、丙二醇、1,3丁二醇、二甲基甲酰铵)、油(具体为,棉花子油、落花生油、玉米油、橄榄油、蓖麻油、芝麻油)、丙三醇、四氢呋喃醇、聚乙二醇和山梨聚糖的脂肪酸酯或这些物质的混合物等。
除了这些惰性稀释剂,组合物还可以包括,例如,润湿剂、乳化和悬浮剂、香化剂、调味剂和加香剂。
除了活性化合物,悬浮液可以包含悬浮剂,例如乙氧基化异十八烷醇、聚氧化乙烯山梨醇、山梨聚糖酯、微晶纤维、偏氢氧化铝、斑脱土、琼脂-琼脂和黄芪胶或这些物质的混合物等。
本发明的化合物的局部给药用剂型包括膏剂、粉末、喷雾和吸入剂。该活性组分在无菌条件下与生理学上可接受载体和任何所需要的防腐剂、缓冲剂或推进剂混合。眼用配方、眼药膏、粉末和溶液也包括在本发明的范围内。
本发明的化合物的外用剂型可以呈油包水(W/O)或水包油(O/W)乳液的形式,多乳液形式,如水包油包水(W/O/W)形式或油包水包油(O/W/O)乳液形式,或者以水分散体或脂分散体、凝胶或气溶胶形式制得。
本发明的化合物的外用剂型可以包含添加剂和制剂助剂,如乳化剂、增稠剂、胶凝剂、水固定剂、铺展剂、稳定剂、染料、香料和防腐剂。合适的乳化剂包括硬脂酸、三乙醇胺和PEG-40-硬脂酸酯。合适的增稠剂包括单硬脂酸甘油酯和PEG600。合适的防腐剂包括对羟苯甲酸丙酯和氯甲酚。适的铺展剂包括二甲聚硅氧烷和聚二甲基环硅氧烷。合适的水固定剂包括聚乙二醇,优选聚乙二醇600。
本发明的化合物的外用剂型可以包括膏剂、洗剂、凝胶、乳液、微乳剂、喷雾剂、皮肤贴剂等,其可局部施用,以治疗特应性皮炎、EGFR抑制剂引起的皮肤副作用、痤疮、湿疹、银屑病、硬皮病、瘙痒、白癜风、脱发等皮肤疾病。特别地,本发明的化合物的外用剂型是膏剂,其可局部外敷施用,以治疗特应性皮炎、EGFR抑制剂引起的皮肤副作用、痤疮、湿疹、银屑病、硬皮病、瘙痒、白癜风、脱发等皮肤疾病。
在药物组合物和剂型中式(G)的化合物、式(G’)的化合物或式(I)的化合物的量可以由本领域技术人员根据需要适当地确定,例如式(G)的化合物、式(G’)的化合物或式(I)的化合物可以治疗有效量存在于药物组合物或剂型中。
在第四个方面,本发明提供了一种治疗与TRK或RET相关的疾病或病症的方法,所述方法包括将治疗有效量的如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,或如上文所述的组合物给予有需要的患者。其中,所述患者优选是哺乳动物,更优选是人类患者。其中给药途径可以是口服、外用(包括但不限于外敷、喷涂等)、胃肠外(包括皮下、肌肉、皮层和静脉)施予、支气管施予或鼻施予等。其中,优选地通过口服或外用施予;更优选地通过口服施予。
在本文中,“与TRK或RET相关的疾病或病症”包括但不限于:
关节炎,包括类风湿性关节炎、幼年型关节炎和银屑病关节炎;
自身免疫疾病或病症,包括单一器官或单一细胞类型自身免疫病症,例如桥本甲状腺炎、自身性免疫溶血性贫血、恶性贫血的自身免疫性萎缩性胃炎、自身免疫性脑脊髓炎、自身免疫性睾丸炎、古德帕斯彻病、自身免疫性血小板减少症、交感性眼炎、重症肌无力、格雷夫斯病、原发性胆汁性肝硬变、慢性侵袭性肝炎、溃疡性结肠炎和膜性肾小球病、涉及全身性自身免疫病症的那些(例如全身性红斑狼疮、类风湿性关节炎、干燥综合征、莱特尔综合征、多肌炎-皮肌炎、系统性硬化病、结节性多动脉炎、多发性硬化和大疱性类天疱疮)以及其它O-细胞(体液)型或T细胞型自身免疫病(包括寇甘综合征)、强直性脊椎炎、Wegener’s氏肉芽肿、自身免疫性脱发(斑秃)、I型糖尿病或幼年发病型糖尿病或甲状腺炎;
癌症或肿瘤,包括消化道/胃肠道癌、结直肠癌、肝癌、皮肤癌(包括肥大细胞瘤和鳞状细胞癌)、乳房和乳腺癌、卵巢癌、前列腺癌、淋巴瘤、白血病(包括急性髓性白血病和慢性髓性白血病)、肾癌、肺癌、肌癌、骨癌、膀胱癌、脑癌、黑色素瘤(包含口腔和转移性黑色素瘤)、卡波西肉瘤、骨髓瘤(包括多发性骨髓瘤)、骨髓增殖性病症、增生型糖尿病视网膜病变或与血管生成相关的病症(包括实体瘤);
糖尿病,包括I型糖尿病或糖尿病并发症;
眼疾病、病症或病况,包括眼的自身免疫病、角膜结膜炎、春季结膜炎、葡萄膜炎(包括与贝切特病相关的葡萄膜炎和晶状体性葡萄膜炎)、角膜炎、疱疹性角膜炎、圆锥形角膜炎、角膜上皮营养障碍、角膜白斑、眼天疱疮、莫伦溃疡、巩膜炎、格雷夫眼病、Vogt-Koyanagi-Harada综合征、干燥性角结膜炎(干眼症)、水疱、虹膜睫状体炎、结节病、内分泌性眼病、交感性眼炎、变应性结膜炎或眼部新生血管形成;
肠炎症、变态反应或病况,包括克罗恩氏病和/或溃疡性结肠炎、炎性肠病、乳糜泻、直肠炎、嗜酸细胞性胃肠炎或肥大细胞增多症;
神经变性疾病,包括运动神经元病、阿尔茨海默病、帕金森病、肌萎缩侧索硬化、亨廷顿病、脑缺血或创伤性损伤引发的神经变性疾病、卒中、谷氨酸神经毒性或缺氧;卒中的缺血/再灌注损伤、心肌缺血、肾缺血、心脏病发作、心脏肥大、动脉粥样硬化和动脉硬化、器官缺氧或血小板聚集;
皮肤疾病、病况或病症,包括特应性皮炎、EGFR抑制剂引起的皮肤副作用、痤疮、湿疹、银屑病、硬皮病、瘙痒或其它瘙痒病况、白癜风、脱发(斑秃);
变态反应,包括哺乳动物的变应性皮炎(包括马变应性疾病,例如叮咬过敏)、夏季湿疹、库蚊叮痒综合症(sweet itch)、肺气肿、炎症性气道疾病、复发性气道阻塞、气道反应过度或慢性阻塞性肺疾病;
哮喘和其它阻塞性气道疾病,包括慢性或顽固型哮喘、晚期哮喘、支气管炎、支气管性哮喘、变应性哮喘、内源性哮喘、外源性哮喘或粉尘性哮喘;
移植排斥,包括胰岛移植排斥、骨髓移植排斥、移植物抗宿主病、器官和细胞移植排斥(例如骨髓、软骨、角膜、心脏、椎间盘、胰岛、肾、四肢、肝、肺、肌肉、成肌细胞、神经、胰脏、皮肤、小肠或气管)或者异种移植。
已经发现,本申请所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物特别适合于治疗选自以下的疾病或病症:关节炎,自身免疫疾病或病症,癌症或肿瘤,糖尿病,糖尿病引起的伤口愈合缓慢、眼疾病、病症或病况,肠炎症、变态反应或病况,神经变性疾病,皮肤疾病、病况或病症,变态反应,哮喘和其它阻塞性气道疾病,移植排斥。
通过动物模型实验已发现,如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物最优选用于治疗瘙痒、银屑病、特应性皮炎、痤疮、白癜风、斑秃、哮喘、鼻炎、痔疮、宫颈炎、肺炎等疾病。此外,这些化合物还可以用于治疗过敏性结膜炎、癌症(肿瘤)、糖尿病引起的伤口愈合缓慢、糖尿病足等疾病。
在一些特别优选的实施方式中,如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物可以作为JAK/泛TRK抑制剂,特别是泛JAK/泛TRK抑制剂或泛JAK/泛TRK/RET抑制剂。
在一些特别优选的实施方式中,如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物可以用于治疗自身免疫疾病、慢性伤口愈合、以及糖尿病并发症,例如选自:关节炎,肠炎症、变态反应或病况,神经变性疾病,皮肤疾病、病况或病症,变态反应,哮喘和其它阻塞性气道疾病,移植排斥,褥疮,糖尿病引起的伤口愈合缓慢、眼疾病、病症或病况等。在一些最优选的实施方式中,如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物可以用于治疗选自瘙痒、银屑病、特应性皮炎、EGFR抑制剂引起的皮肤副作用、痤疮、白癜风、斑秃、哮喘、鼻炎、痔疮、宫颈炎、肺炎、褥疮、糖尿病引起的伤口愈合缓慢、糖尿病足、糖尿病视网膜病变等的疾病或病症。
在第五个方面,本发明提供了一种被用作TRK抑制剂药物和/或RET抑制剂药物、或者被用作治疗与TRK或RET相关的疾病或病症的药物的如上文所述的式(G)的化合物、式(G’)的化合物或式(I)的化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物。其中,式(G)的化合物、式(G’)的化合物或式(I)的化合物中取代基定义和优选项如前文所述,其中与TRK或RET相关的疾病或病症亦如前文所述。
优选地,本发明提供了以下实施方案:
1.(G)的化合物:
或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物在制备TRK抑制剂药物和/或RET抑制剂药物中的用途,其中:
L为C=O、O=S=O、CH2或连接键;且
X1为N或CR14;且
X2为N或CR15;且
X3为N或CR16;且
R14、R15、R16各自独立地选自H、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C2-8烯基、C2-8炔基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基任选地被1个、2个或3个选自卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、 C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、-N(C1-4烷基)(C(=O)C1-4烷基)、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基的取代基取代;且
R13为H、-N(R17)(R18)、C1-6烷氧基、-SR12、-OR12、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,C7- 11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且任选地被一个或多个各自独立选自以下群组的取代基所取代:-OH、-CN、-SH、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;或者R17、R18以及与它们相连的N原子共同形成3-14元环;且
R2的个数为0、1、2、3或4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且
R1选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;且
R3、R4各自独立地选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1- 6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R5)(R6)、-N(R11)(C(=O)R12)、-CON(R7)(R8)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-6烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且R5、R6、R7、R8、R9、R10、R11、R12各自独立地是H或选自以下群组:C1- 6烷基、C1-4卤代烷基、C3-7环烷基、4-14元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CF3、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、氧代、C1-4烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
2.根据实施方案1所述的用途,其中所有H各自独立地任选地被D取代。
3.根据实施方案1或2所述的用途,其中X1、X2、X3其中仅一个为N。
4.根据实施方案1或2所述的用途,其中X1、X2、X3其中仅两个为N。
5.根据实施方案1或2所述的用途,其中X1、X2、X3是相同的。
6.根据实施方案5所述的用途,其中X1为CR14、X2为CR15、X3为CR16,且R14、R15、R16是相同的。
7.根据实施方案6所述的用途,其中R14、R15、R16选自H、-OH、-SH、-CN、卤素、-NO2、C1-6烷基。
8.根据实施方案7所述的用途,其中R14、R15、R16选自H、-OH、C1- 6烷基。
9.根据实施方案7所述的用途,其中X1、X2、X3均为CH。
10.根据实施方案5所述的用途,其中X1、X2、X3均为N。
11.根据实施方案1-10中任一项所述的用途,其中L为C=O、O=S=O或CH2
12.根据实施方案1-10中任一项所述的用途,其中R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。
13.根据实施方案1-10中任一项所述的用途,其中R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基,且R13被0个、1个、2个、3个或4个R1取代。
14.根据实施方案1-10中任一项所述的用途,其中R13为H、-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,且R13被0个、1个、2个、3个或4个R1取代。
15.根据实施方案1-10中任一项所述的用途,其中R13为-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、4-6元杂环烷基、苯基、5-6元杂芳基,且R13被0个、1个、2个或3个R1取代。
16.根据实施方案1-10中任一项所述的用途,其中R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。
17.根据实施方案1-10中任一项所述的用途,其中R17、R18以及与它们相连的N原子共同形成4-10元环。
18.根据实施方案1-10中任一项所述的用途,其中L为C=O,且R13为-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5或-S-C1- 4烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代;或者R17、R18以及与它们相连的N原子共同形成3-14元环。
19.根据实施方案1-10中任一项所述的用途,其中存在1个、2个或3个R2,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基,其中所述-S-C1-4烷基、C1-6烷基、C3-7环烷基、4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
20.根据实施方案1-10中任一项所述的用途,其中存在1个、2个或3个R2,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
21.根据实施方案15所述的用途,其中存在1个或2个R2,并且R2选自卤素、C1-6烷基。
22.根据实施方案1-10中任一项所述的用途,其中R13被0个或1个R1取代,并且R1选自卤素、-OH、-CN、C1-6烷基、5-7元杂环烷基、C3-7环烷基,其中所述C1-6烷基任选地被1个、2个或3个R3取代,并且其中所述5-7元杂环烷基、C3-7环烷基任选地被1个、2个、3个或4个C1-3烷基取代。
23.根据实施方案1所述的用途,其中所述化合物为式(I)的化合物:
或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中:
L为C=O、O=S=O、CH2或连接键;
X为CH或N;
环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基;
R1的个数为0、1、2、3或4个,并且R1选自H、卤素、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基,其中所述C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;
R2的个数为0、1、2、3或4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基以及4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12
R3选自卤素、氰基、C1-3烷基、羟基、C1-6烷氧基、-N(R5)(R6)、-CON(R7)(R8)或3-7元杂环烷基,其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
R4选自卤素、C1-3烷基、羟基、C1-6烷氧基、-NH2、-NHCH3或-N(CH3)2
R5、R6、R7、R8各自独立地为氢或C1-4烷基;
R9选自H、C1-4烷基、C1-4卤代烷基或C3-7环烷基;
R10是H或选自以下群组:C1-4烷基、C1-4卤代烷基、C3-7环烷基、4-10元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基;
R11选自H、C1-4烷基以及C3-7环烷基;
R12选自C1-6烷基、C3-7环烷基、4-至14-元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CF3、-CN、-OH、-NH2、-NH(CH3)、-N(CH3)2、氧代、-S-C1-4烷基、C1-4烷基、C1-4卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4烷氧基以及C1-4卤代烷氧基。
24.根据实施方案23所述的用途,其中L为C=O、O=S=O或CH2
25.根据实施方案23所述的用途,其中X为CH。
26.根据实施方案23-25中任一项所述的用途,其中环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基。
27.根据实施方案23-25中任一项所述的用途,其中环A为5-6元杂芳基或苯基。
28.根据实施方案23-25中任一项所述的用途,其中存在0个或1个R1,并且R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。
29.根据实施方案23-25中任一项所述的用途,其中存在1个或2个R2,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3- 6环烷基各任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
30.根据实施方案23-25中任一项所述的用途,其中:
L为C=O或O=S=O;
X为CH;
环A为5-7元杂芳基、C5-7芳基;
R1的个数为0、1、2、3或4个,并且R1选自C1-8烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
R2的个数为1、2或3个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基以及4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12
R3选自卤素、氰基、C1-3烷基、羟基、C1-6烷氧基、-N(R5)(R6)、-CON(R7)(R8)或3-7元杂环烷基,其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
R4选自卤素、C1-3烷基、羟基、C1-6烷氧基、-NH2、-NHCH3或-N(CH3)2
R5、R6、R7、R8各自独立地为氢或C1-4烷基;
R9选自H、C1-4烷基、C1-4卤代烷基或C3-7环烷基;
R10是H或选自以下群组:C1-4烷基、C1-4卤代烷基、C3-7环烷基、4-10元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基;
R11选自H、C1-4烷基以及C3-7环烷基;
R12选自C1-6烷基、C3-7环烷基、4-至14-元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CF3、-CN、-OH、-NH2、-NH(CH3)、-N(CH3)2、氧代、-S-C1-4烷基、C1-4烷基、C1-4卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4烷氧基以及C1-4卤代烷氧基。
31.根据实施方案30所述的用途,其中环A为5-6元杂芳基或苯基。
32.根据实施方案30所述的用途,其中存在0个或1个R1,并且R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。
33.根据实施方案30所述的用途,其中存在1个或2个R2,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
34.根据实施方案1所述的用途,其中所述化合物选自:
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-(哌啶-1-基)吡嗪-2-基)甲酮(MDI-2);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(1-甲基-1H-吡唑-4-基)甲酮(MDI-202);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(1-甲基哌啶-4-基)甲酮(MDI-203);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-204);
(2-(6-(2-乙基-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-205);
5-乙基-2-氟-4-(3-(5-(苯磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-206);
5-乙基-2-氟-4-(3-(5-(吡嗪-2基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-207);
4-(3-(5-(环丙基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-208);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233);
4-(3-(5-(环丁基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-210);
环丁基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-211);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(3-羟基环丁基)甲酮(MDI-213);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪唑-5-(1H,4H,6H)-基)(哒嗪-4-基)甲酮(MDI-214);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪唑-5-(1H,4H,6H)-基)(哒嗪-3-基)甲酮(MDI-215);
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228);
5-乙基-2-氟-4-(3-(5-(4-羟基环己基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-217);
4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218);
4-(3-(5-(环丁基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-219);
4-(3-(5-(环戊基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-220);
5-乙基-2-氟-4-(3-(5-((1-甲基-1H-吡唑-4-基)甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-221);
4-(3-(5-(环戊烷基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-224);
5-乙基-2-氟-4-(3-(5-(四氢-2H-吡喃-4-基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-225);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)乙-1-酮(MDI-226);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)丙-1-酮(MDI-227);
(1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)-2-甲基丙-1-酮)(MDI-228);
2-环丙基-1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)基)乙-1-酮(MDI-229);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)-3-甲基丁-1-酮(MDI-230);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231);
N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-b]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-237);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-239);
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-240);
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-c]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-242);
(R)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-243);
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-甲基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-246);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248);
1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-5-羰基)吡咯烷-3-腈(MDI-250);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(四氢呋喃-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-251);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸甲酯(MDI-252);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253);
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[3,4-b]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-255);
3-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)-3-氧丙腈(MDI-256);
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257);
N-(2-氰基乙基)-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-258);
N-环丙基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-259);
N-环丁基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-260);
(S)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-262);
(R)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-263);
或以上任一化合物的同位素标记化合物、光学异构体、几何异构体、互变异构体或异构体混合物、或药学上可接受的盐、前体药、或代谢物。
35.根据实施方案1所述的用途,其中所述化合物选自:
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201)
环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233)
(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228)
4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218)
N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢 吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236)
(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253)
2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257)
或以上任一化合物的同位素标记化合物、光学异构体、几何异构体、互变异构体或异构体混合物、或药学上可接受的盐、前体药、或代谢物。
36.根据实施方案1-35中任意一项所述的用途,其中所述化合物被用作JAK/TRK双重抑制剂(优选被用作pan-JAK/pan-TRK双重抑制剂)或JAK/TRK/RET多重抑制剂(优选被用作pan-JAK/pan-TRK/RET多重抑制剂)。
37.一种TRK抑制剂和/或RET抑制剂药物组合物,其包含如实施方案1-35中任意一项中所定义的化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,以及一种或多种药学上可接受的载体、佐剂或赋形剂。
38.根据实施方案37所述的药物组合物,其被配制成口服剂型或适于局部施用的外用剂型。
39.根据实施方案37所述的药物组合物,其被配制成JAK/TRK双重抑制剂药物(优选被用作pan-JAK/pan-TRK双重抑制剂药物)或JAK/TRK/RET多重抑制剂药物(优选被被配制成pan-JAK/pan-TRK/RET多重抑制剂药物)。
40.根据实施方案1-35中任意一项中所定义化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物或者根据实施方案37或38或39所述的组合物在制备用于治疗和/或预防与TRK和/或RET相关的疾病或病症的药物中的用途。
41.根据实施方案40所述的用途,其中所述与TRK和/或RET相关的疾病或病症选自关节炎,自身免疫疾病或病症,癌症或肿瘤,糖尿病及糖尿病并发症,(糖尿病引起的)伤口愈合缓慢、眼疾病、病症或病况,肠炎症、变态反应或病况,神经变性疾病,皮肤疾病、病况或病症,变态反应,哮喘和其它阻塞性气道疾病,移植排斥。
42.根据实施方案40所述的用途,其中所述与TRK和/或RET相关的疾病或病症选自瘙痒、银屑病、特应性皮炎、EGFR抑制剂引起的皮肤副作用、痤疮、白癜风、斑秃、哮喘、鼻炎、痔疮、宫颈炎、肺炎、糖尿病引起的伤口愈合缓慢、糖尿病足、糖尿病视网膜病变、癌症(肿瘤)、褥疮。
下面结合附图和具体实施例对本发明做进一步的说明和描述。
附图说明
图1示出了卵清蛋白诱导的小鼠哮喘模型的肺盥洗液IL-5 Elisa检测结果。其中,数据以均数±标准误(Mean±SEM)表示(*p<0.05;**p<0.01;***p<0.001vs.溶媒对照组,One-way ANOVA,Bonferroni's Multiple Comparison Test)。
图2示出了卵清蛋白诱导的小鼠哮喘模型的肺盥洗液炎症细胞计数结果。其中数据以均数±标准误(Mean±SEM)表示(*p<0.05;**p<0.01;***p<0.001vs.溶媒对照组,One-way ANOVA,Bonferroni's Multiple Comparison Test)。
图3示出了卵清蛋白诱导的小鼠哮喘模型的血清IL-5 Elisa检测结果。 其中数据以均数±标准误(Mean±SEM)表示(*p<0.05;**p<0.01;***p<0.001vs.溶媒对照组,One-way ANOVA,Bonferroni's Multiple Comparison Test)。
图4示出了卵清蛋白诱导的小鼠哮喘模型的肺组织病理染色代表图片。
图5示出了卵清蛋白诱导的小鼠哮喘模型的肺组织病理评分结果。其中数据以均数±标准误(Mean±SEM)表示(*p<0.05;**p<0.01;***p<0.001vs.溶媒对照组,One-way ANOVA,Bonferroni's Multiple Comparison Test)。
图6示出了过敏性鼻炎小鼠模型中雄鼠药物干预打分结果(*,p<0.05)。
图7示出了过敏性鼻炎小鼠模型中雌鼠药物干预打分结果(*,p<0.05)。
图8示出了过敏性鼻炎小鼠模型中不同性别小鼠各组切片HE染色结果。
图9示出了过敏性鼻炎小鼠模型中雄性小鼠血清中IL-4含量检测(*,p<0.05)。
图10示出了过敏性鼻炎小鼠模型中雌性小鼠血清中IL-4含量检测(*,p<0.05)。
图11示出了慢性阻塞性肺疾病大鼠动物模型中各组的肺组织HE染色结果。
图12示出了慢性阻塞性肺疾病大鼠动物模型中血清和灌洗液中的IL-β表达量变化。
图13示出了大鼠外痔造模效果和阳性对照药物效果。
图14示出了对大鼠外痔模型中各组病理切片HE染色打分情况。
图15示出了对大鼠外痔模型中各组样品中TNF-α表达水平检测。
图16示出了药物对苯酚诱导的宫颈炎大鼠外阴炎症的影响。
图17示出了药物对苯酚诱导的宫颈炎大鼠红斑症状的影响。
图18示出了药物对苯酚诱导的宫颈炎大鼠水肿症状的影响。
图19示出了药物对苯酚诱导的宫颈炎大鼠分泌物症状的影响。
图20示出了药物对苯酚诱导的宫颈炎大鼠的宫颈指数的影响。
图21示出了苯酚诱导的宫颈炎大鼠的阴道HE染色结果。
图22示出了特应性皮炎/皮肤瘙痒症小鼠模型中小鼠皮肤发红、出血、喷发和脱屑情况的评分结果。
图23示出了特应性皮炎/皮肤瘙痒症小鼠模型中小鼠皮肤发红、出血、喷发和脱屑情况评分曲线下面积(AUC)。
图24示出了特应性皮炎/皮肤瘙痒症小鼠模型HE染色实验结果(×400)。
图25示出了特应性皮炎/皮肤瘙痒症小鼠模型甲苯胺蓝染色实验结果(×400)。
图26示出了药物对特应性皮炎/皮肤瘙痒症小鼠血清中IL-4的影响(*与正常组比较,p<0.05)。
图27示出了药物对特应性皮炎/皮肤瘙痒症小鼠血清中IL-13的影响(*与正常组比较,p<0.05)。
图28示出了药物对特应性皮炎/皮肤瘙痒症小鼠血清中IFN-γ的影响(*与正常组比较,p<0.05)。
图29示出了药物对特应性皮炎/皮肤瘙痒症小鼠血清中TNF-α的影响(*与正常组比较,p<0.05)。
图30示出了药物对白癜风小鼠皮毛脱色面积评分影响(用药部位,皮毛脱色部位)。
图31示出了药物对白癜风小鼠皮毛脱色面积评分影响(非用药部位即耳部,躯干非涂药部位,尾巴)。
图32示出了白癜风小鼠HE染色结果。
图33示出了药物对白癜风小鼠血清TNF-α影响(*与空白对照组比较,p<0.01,#与模型组比较,p<0.01)。
图34示出了药物对白癜风小鼠血清IL-6影响(*与空白对照组比较,p<0.01,#与模型组比较,p<0.01)。
图35示出了斑秃小鼠毛发生长评分曲线下面积比较。
图36示出了斑秃小鼠毛发生长情况(第21天)。
图37示出了雌性斑秃小鼠毛发生长情况。
图38示出了雄性斑秃小鼠毛发生长情况。
图39示出了咪喹莫特诱导的银屑病小鼠皮肤厚度评分。
图40示出了咪喹莫特诱导的银屑病小鼠临床评分。
图41示出了咪喹莫特诱导的银屑病小鼠皮肤厚度评分的AUC(*p<0.05,**p<0.01,****p<0.0001vs.溶媒对照组,One-way ANOVA)。
图42示出了咪喹莫特诱导的银屑病小鼠临床评分的AUC(***p<0.001,****p<0.0001vs.溶媒对照组,One-way ANOVA)。
图43示出了咪喹莫特诱导的银屑病小鼠在实验终点的脾脏重量(***p<0.001,****p<0.0001vs.溶媒对照组,One-way ANOVA)。
图44示出了银屑病小鼠的病理评分结果(****p<0.0001vs.溶剂对照组,One-way ANOVA)。
图45示出了银屑病小鼠的细胞因子TNF-α的检测结果(*p<0.05vs.溶媒对照组,One-way ANOVA)。
图46示出了药物对痤疮兔子耳片重量的影响(##,与空白对照比较,p<0.01,*与模型(溶媒)组比较,p<0.05)。
图47示出了痤疮兔子耳组织病理图。
图48示出了药物对耳组织皮脂腺直径的影响(##,与空白对照比较,p<0.01,*与模型(溶媒)组比较,p<0.05)。
图49示出了药物对痤疮兔子耳组织毛囊面积的影响(##,与空白对照比较,p<0.01,#与空白对照比较,p<0.05,**与模型(溶媒)组比较,p<0.01,*与模型(溶媒)组比较,p<0.05)。
图50示出了药物对痤疮兔子血清IL-1α的影响(##,与空白对照比较,p<0.01,**与模型(溶媒)组比较,p<0.01)。
图51示出了药物对痤疮兔子血清IL-6的影响(##,与空白对照比较,p<0.01,**与模型(溶媒)组比较,p<0.01)。
图52示出了药物对痤疮兔子血清DHT的影响。
图53示出了化合物对体外脂肪细胞脂解的影响的实验过程和实验结果。
图54示出了小鼠非伤口处皮肤和伤口组织的HE染色结果。
图55示出了化合物促进伤口中上皮愈合的实验结果。
实施例
为了使本发明的发明目的、技术方案和有益技术效果更加清晰,以下结合实施例进一步详细描述本发明的实施方式。但是,应当理解的是,本发明的实施例仅仅是为了解释本发明,并非为了限制本发明,且本发明的实施例并不局限于说明书中给出的实施例。实施例中未注明具体来源的试剂或仪器均为化学或生物实验室的常规试剂或仪器;实施例中未注明具体实验条件或操作条件的操作按本领域技术人员熟知的常规条件进行,或按材料供应商或仪器生产商推荐的条件进行。
实施例1:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5(1H)-基)(5-(哌啶-1-基)吡嗪-2-基)甲酮(MDI-2)
按照中国发明专利CN111606908B的实施例1所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.67(s,1H),8.28(dd,J=8.0Hz,J=4.0Hz,1H),8.21(s,1H),7.40(s,1H),7.18(dd,J=8.0Hz,J=4.0Hz,1H),6.96-6.89(m,2H),5.14(s,2H),4.82(s,2H),3.76-3.73(m,4H),2.58(dd,J=12.0Hz,J=8.0Hz,2H),1.76-1.66(m,6H),1.10(t,J=8.0Hz,3H).
实施例1:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201)
按照中国发明专利CN111606908B的实施例2所示方法进行合成,具体如下。
MDI-201的合成路线:
合成方法:
合成中间体MDI-201-1:5-吗啉吡嗪-2-甲酸甲酯
将5-氯-吡嗪-2-甲酸甲酯(1.5g,8.7mmol)溶于10ml DMF中,加入N,N-二异丙基乙胺(3.0ml,17.4mmol)和吗啉(0.91g,10.4mmol),在室温下搅拌过夜,在剧烈搅拌下,加入水,有固体析出,过滤,用水洗滤饼,干燥,得到中间体MDI-201-1,收率72.2%。
合成中间体MDI-201-2:5-吗啉吡嗪-2-甲酸
将中间体MDI-201-1(1.4g,6.27mmol)溶于20ml四氢呋喃和20ml水,加入氢氧化锂(0.32g,7.53mmol),室温反应4小时,减压浓缩掉四氢呋喃,用1N HCl调pH=4,有固体析出,过滤,用水洗滤饼,干燥得中间体MDI-201-2,收率99.1%。
1H NMR(400MHz,CDCl3)δ8.92(s,1H),8.04(s,1H),3.88-3.86(m,4H),3.80-3.77(m,4H).
合成中间体MDI-201-3:(2-(6-溴1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮
将中间体MDI-201-2(27.4mg,0.13mmol)和N,N-二异丙基乙胺(46.0mg,0.36mmol)溶于DMF中,加入HATU(67.8mg,0.18mmol),室温反应10分钟。中间体2-(6-溴1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酸叔丁酯(80mg,0.12mmol)溶于5ml二氯甲烷中,加入1ml三氟乙酸,室温搅拌30分钟,浓缩,用二氯甲烷带3次三氟乙酸,溶于DMF中然后缓慢将其加入前面的反应液中,室温反应过夜,加入水淬灭反应,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-201-3,收率47.8%。
1H NMR(400MHz,CDCl3)δ8.91(d,J=8.0Hz,1H),8.44-8.36(m,1H),8.10(d,J=8.0Hz,1H),7.80(s,1H),7.46-7.41(m,1H),5.96(s,2H),5.74(d,J=4.0Hz,2H),5.27(s,1H),5.19(s,1H),5.00(s,1H),4.92(s,1H),3.90-3.88(m,4H),3.75-3.72(m,4H),3.64-3.58(m,4H),0.96-0.89(m,4H),0.03(s,9H),0.02(s,9H).
合成中间体MDI-201-4:(2-(6-(2-乙基-5-氟-4-((2-(三甲基硅基)乙氧基)甲基)羟基苯基)1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮
将中间体MDI-201-3(43.0mg,0.06mmol),(2-((5-乙基-2-氟-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯氧基)甲氧基)乙基)三甲基硅烷(27.1mg,0.07mmol),Pd(dppf)Cl2(4.2mg,0.006mmol)和磷酸钾(36.2mg,0.17mmol)溶于1,4-二氧六环(10ml)和水(2ml)中,氮气置换3次,加热到100度,反应过夜,冷却到室温,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-201-4,收率40.9%。
1H NMR(400MHz,CDCl3)δ8.91(dd,J=4.0Hz,J=4.0Hz,1H),8.52(dd,J=8.0Hz,J=16.0Hz,1H),8.10(dd,J=8.0Hz,J=4.0Hz,1H),7.49(s,1H),7.27(s,1H),7.20(d,J=8.0Hz,1H),7.06(d,J=12.0Hz,1H),6.00(s,2H),5.79(d,J=4.0Hz,2H),5.35(s,2H),5.29(s,1H),5.20(s,1H),5.02(s,1H),4.94(s,1H),3.91-3.86(m,6H),3.76-3.72(m,4H),3.65-3.61(m,4H),2.58(t,J=8.0Hz,2H),1.10-1.03(m,3H),0.95-0.91(m,6H),0.06(s,9H),0.04(s,9H),0.03(s,9H).
合成MDI-201:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮
将中间体MDI-201-4(22.0mg,0.02mmol)溶于甲醇(4ml)中,加入浓盐酸(2ml),加热到50度,反应6小时,浓缩,固体用1ml甲醇溶解,用加入2ml浓氨水,浓缩,用甲醇带3次氨水,制备板纯化,得到最终产物8.0mg,收率61.9%。
1H NMR(400MHz,DMSO-d6)δ13.35(s,1H),9.89(s,1H),8.66(d,J=4.0Hz,1H),8.38-8.33(m,2H),7.42(s,1H),7.15(d,J=8.0Hz,1H),7.06(d,J=12.0Hz,1H),6.95(d,J=8.0Hz,1H),5.05(s,2H),4.72(s,2H),3.76-3.74(m,4H),3.71-3.68(m,4H),2.52(dd,J=12.0Hz,J=4.0Hz,2H),1.05(t,J=8.0Hz,3H).
实施例3:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5(1H)-基)(1-甲基-1H-吡唑-4-基)甲酮(MDI-202)
按照中国发明专利CN111606908B的实施例3所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,DMSO-d6)δ13.33(s,1H),12.87(s,1H),9.89(s,1H),8.35(d,J=8.0Hz,2H),7.94(s,1H),7.42(s,1H),7.15(d,J=8.0Hz,1H),7.06(d,J=12.0Hz,1H),6.95(d,J=12.0Hz,1H),4.89(s,2H),4.67(s,2H),3.92(s,3H),2.51-2.48(m,2H),1.05(t,J=8.0Hz,3H).
实施例4:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑 -5(1H,4H,6H)-基)(1-甲基哌啶-4-基)甲酮(MDI-203)
MDI-203也可被命名为5-乙基-2-氟-4-{3-[5-(1-甲基哌啶-4-羰基)-1H,4H,5H,6H-吡咯并[3,4-d]咪唑-2-基]-1H-吲唑-6-基}苯酚,按照中国发明专利CN111606908B的实施例4所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,DMSO-d6)δ13.35(s,1H),9.87(s,1H),9.24(s,1H), 8.32(d,J=8.0Hz,1H),7.42(s,1H),7.22(d,J=8.0Hz,1H),7.03(d,J=12.0Hz,1H),6.96(d,J=12.0Hz,1H),4.80(s,2H),4.48(s,2H),3.04-3.01(m,2H),2.79(s,3H),2.55-2.51(m,2H),2.05-1.99(m,3H),1.85-1.78(m,2H),1.01-0.98(m,3H).两个H的信号被水峰(δ=3.37)所掩盖。
实施例5:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑 -5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-204)
MDI-204也可被命名为5-乙基-2-氟-4-{3-[5-(4-甲基哌嗪-1-羰基)-1H,4H,5H,6H-吡咯并[3,4-d]咪唑-2-基]-1H-吲唑-6-基}苯酚,按照中国发明专利CN111606908B的实施例5所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,DMSO-d6)δ13.29(s,1H),12.79(d,J=16.0Hz,1H),9.85(s,1H),8.62(s,1H),8.36(s,1H),8.34-8.30(m,1H),7.40(s,1H),7.14-7.10(m,1H),7.03(d,J=12.0Hz,1H),6.92(d,J=12.0Hz,1H),5.08-4.65(m,4H),2.55-2.49(m,6H),2.24(s,3H),2.03-1.97(m,4H),1.04-1.02(m,3H).
实施例6:(2-(6-(2-乙基-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑- 5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-205)
MDI-205也可被命名为3-乙基-4-{3-[5-(4-甲基哌嗪-1-羰基)-1H,4H,5H,6H-吡咯并[3,4-d]咪唑-2-基]-1H-吲唑-6-基}苯酚,按照中国发明专利CN111606908B的实施例6所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.72(d,J=4.0Hz,1H),8.28(dd,J=4.0Hz,J=8.0Hz,2H),7.40(s,1H),7.18(dd,J=4.0Hz,J=8.0Hz,1H),7.09(d,J=8.0Hz,1H),6.80(d,J=4.0Hz,1H),6.72-6.69(m,1H),5.17(s,2H),4.85(s,2H),3.83-3.81(m,4H),2.67-2.64(m,4H),2.60(dd,J=4.0Hz,J=8.0Hz,2H),2.43(s,3H),1.10(t,J=8.0Hz,3H).
实施例7:5-乙基-2-氟-4-(3-(5-(苯磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2- 基)-1H-吲唑-6-基)苯酚(MDI-206)
按照中国发明专利CN111606908B的实施例7所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.22(d,J=8.0Hz,1H),7.98-7.96(m,2H),7.69-7.65(m,3H),7.41(s,1H),7.16(d,J=8.0Hz,1H),6.96-6.89(m,2H),4.61-4.52(m,4H),2.57(dd,J=16.0Hz,J=8.0Hz,2H),1.08(t,J=8.0Hz,3H).
实施例8:5-乙基-2-氟-4-(3-(5-(吡嗪-2基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪 唑-2-基)-1H-吲唑-6-基)苯酚(MDI-207)
按照中国发明专利CN111606908B的实施例8所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.80(d,J=4.0Hz,1H),8.65(dd,J=4.0Hz,J=4.0Hz,1H),8.56(d,J=4.0Hz,1H),8.26(dd,J=4.0Hz,J=4.0Hz,1H),7.41(d,J=4.0Hz,1H),7.17(dd,J=12.0Hz,J=4.0Hz,1H),6.91-6.89(m,2H),4.30(s,2H),4.07(s,4H),2.56(dd,J=8.0Hz,J=16.0Hz,2H),1.07(t,J=8.0Hz,3H).
实施例9:4-(3-(5-(环丙基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲 唑-6-基)-5-乙基-2-氟苯酚(MDI-208)
按照中国发明专利CN111606908B的实施例9所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.27(dd,J=4.0Hz,J=8.0Hz,1H),7.42(s,1H),7.17(dd,J=4.0Hz,J=8.0Hz,1H),6.97-6.89(m,2H),4.02(s,4H),2.80(d,J=8.0Hz,2H),2.59-2.53(m,2H),1.10(m,4H),0.66-0.61(m,2H),0.30-0.27(m,2H).
实施例10:环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并 [3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233)
按照中国发明专利CN111606908B的实施例10所示方法进行合成(在CN111606908B中,该化合物编号为MDI-209,在本申请中编号为MDI-1233),具体如下。
MDI-1233的合成路线:
合成方法:
合成中间体MDI-1233-1:(2-(6-溴1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(环丙基)甲酮
将中间体2-(6-溴1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酸叔丁酯(80mg,0.12mmol)溶于5ml二氯甲烷中,加入1ml三氟乙酸,室温搅拌30分钟,浓缩,用二氯甲烷带3次三氟乙酸,溶于5ml DCM中,然后向体系中加入三乙胺(24.3mg,0.24mmol),降温至0℃,缓慢滴加环丙基甲酰氯(18.8mg,0.18mmol),滴加完成后升至室温反应1-2h,停止反应,向体系中加水淬灭,分液,有机相用硫酸钠干燥,浓缩柱层析得到化合物MDI-1233-1,收率45%。
1H NMR(400MHz,CDCl3)δ8.36(dd,J=17.8Hz,J=8.6Hz,1H),7.80-7.79(m,1H),7.41(d,J=8.6Hz,1H),5.97-5.92(m,2H),5.71(d,J=2.4Hz,2H),4.96-4.66(m,4H),3.62-3.54(m,4H),1.78-1.67(m,1H),1.10–1.07(m,2H),0.94–0.84(m,6H),-0.05(s,9H),-0.08(s,9H).
合成中间体MDI-1233-2:环丙基(2-(6-(2-乙基-5-氟-4-((2-(三甲基硅基)乙氧基)甲氧基)苯基)1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮
将中间体MDI-1233-1(50.5mg,0.08mmol),(2-((5-乙基-2-氟-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯氧基)甲氧基)乙基)三甲基硅烷(34.8mg,0.1mmol),Pd(dppf)Cl2(5.9mg,0.008mmol)和磷酸钾(50.9mg,0.24mmol)溶于1,4-二氧六环(10ml)和水(2ml)中,氮气置换3次,加热到100度,反应过夜,冷却到室温,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-1233-2,收率76.1%。
1H NMR(400MHz,CDCl3)δ8.50-8.43(m,1H),7.46-7.45(m,1H),7.25-7.22(m,1H),7.16(d,J=8.0Hz,1H),7.02(d,J=12.0Hz,1H),5.99-5.94(m,2H),5.76(s,2H),5.32(s,2H),4.98-4.67(m,4H),3.88-3.84(m,2H),3.64-3.55(m,4H),2.57-2.51(m,2H),1.79-1.68(m,1H),1.07-1.02(m,6H),0.95-0.87(m,5H),0.03(s,9H),-0.06--0.08(m,18H).
合成化合物MDI-1233:环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮
将中间体MDI-1233-2(50mg,0.06mmol)溶于甲醇(4ml)中,加入浓盐酸(2ml),加热到50度,反应6小时,浓缩,固体用1ml甲醇溶解,用加入2ml浓氨水,浓缩,用甲醇带3次氨水,浓缩制备分离,得到最终产物10.0mg,收率38.1%。
1H NMR(400MHz,MeOD-d4)δ8.28(d,J=8.0Hz,1H),7.43(s,1H),7.18(dd,J=8.4Hz,J=1.4Hz,1H),6.98(d,J=12.0Hz,1H),6.92(d,J=12.0Hz,1H),4.95(s,2H),4.65(s,2H),2.59-2.53(m,2H),1.98-1.89(m,1H),1.08(t,J=8.0Hz,3H),1.02-1.00(m,2H),0.98-0.92(m,2H).
实施例11:4-(3-(5-(环丁基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H- 吲唑-6-基)-5-乙基-2-氟苯酚(MDI-210)
按照中国发明专利CN111606908B的实施例11所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.27(dd,J=4.0Hz,J=8.0Hz,1H),7.42(s,1H),7.17(dd,J=4.0Hz,J=8.0Hz,1H),6.97-6.89(m,2H),3.98(s,4H),3.00(d,J=8.0Hz,2H),2.72-2.68(m,1H),2.59-2.53(m,2H),2.21-2.18(m,2H),1.89-1.85(m,4H),1.07(t,J=8.0Hz,3H).
实施例12:环丁基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并 [3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-211)
按照中国发明专利CN111606908B的实施例12所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.27(d,J=8.0Hz,1H),7.43(s,1H),,7.17(dd,J=8.4,1.4Hz,1H),6.93(dd,J=20.0Hz,J=12.0Hz,2H),4.68-4.63(m,4H),3.54-3.46(m,1H),2.57-2.53(m,2H),2.43-2.26(m,4H),2.16-2.04(m,1H),1.98-1.89(m,1H),1.08(t,J=8.0Hz,3H).
实施例13:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪 唑-5(1H,4H,6H)-基)(3-羟基环丁基)甲酮(MDI-213)
按照中国发明专利CN111606908B的实施例13所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.28(dd,J=4.0Hz,J=8.0Hz,1H),7.43-7.42(m,1H),7.19(dd,J=4.0Hz,J=8.0Hz,1H),6.97-6.89(m,2H),4.72-4.62(m,4H),4.23-4.20(m,1H),2.95-2.91(m,1H),2.63-2.53(m,4H),2.26-2.18(m,2H),1.10(t,J=8.0Hz,3H).
实施例14:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪 唑-5-(1H,4H,6H)-基)(哒嗪-4-基)甲酮(MDI-214)
按照中国发明专利CN111606908B的实施例14所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ9.48(dd,J=2.3,J=1.3Hz,1H),9.42(dd,J=5.2,J=1.3Hz,1H),8.26(s,1H),8.02(dd,J=5.3,J=2.2Hz,1H),7.43(d,J=1.1Hz,1H),7.17(d,J=8.2Hz,1H),6.93(dd,J=19.7,J=10.4Hz,2H),4.90(s,2H),4.73(s,2H),2.55(q,J=7.5Hz,2H),1.08(t,J=7.5Hz,3H).
实施例15:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪 唑-5-(1H,4H,6H)-基)(哒嗪-3-基)甲酮(MDI-215)
按照中国发明专利CN111606908B的实施例15所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,DMSO-d6)δ13.31(s,1H),12.83(d,J=33.0Hz,1H),9.85(s,1H),9.39(dd,J=5.0Hz,J=1.7Hz,1H),8.37–8.31(m,1H),8.07(s,1H),7.92(dd,J=8.5Hz,J=5.0Hz,1H),7.40(s,1H),7.13(d,J=8.1Hz,1H),7.03(d,J=11.9Hz,1H),6.92(d,J=9.1Hz,1H),4.84–4.45(m,4H),2.49(q,J=7.5Hz,2H),1.02(t,J=7.5Hz,3H).
实施例16:(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯 并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228)
按照中国发明专利CN111606908B的实施例16所示方法进行合成(在CN111606908B中,该化合物编号为MDI-216,在本申请中编号为MDI-1228),具体过程如下。
MDI-1228的合成路线:
合成方法:
合成中间体MDI-1228-1:6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基硅基)乙氧基)甲基)-3-(1-((2-(三甲基硅基)乙氧基)甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑
将2-(6-溴1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲哚-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酸叔丁酯(500mg,0.75mmol),2-(4-(苄氧基)-2-乙基-5-氟苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(401mg,1.13mmol),Pd(dppf)Cl2(75mg,0.075mmol)和磷酸钾(495mg,2.25mmol)溶于1,4-二氧六环(30ml)和水(6ml)中,氮气置换3次,加热到100度,反应16h,冷却到室温,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,纯化所得产品溶于25ml二氯甲烷,滴加5ml三氟乙酸,室温搅拌30分钟,浓缩,用二氯甲烷带3次三氟乙酸,浓缩,硅胶柱纯化,得中间体MDI-1228-1 210mg,收率39.2%。
1H NMR(400MHz,CDCl3)δ8.48(d,J=8.3Hz,1H),7.52(d,J=7.4Hz,1H),7.49–7.37(m,5H),7.25(d,J=8.4Hz,1H),7.23-6.96(m,2H),5.93(s,2H),5.77(s,2H),5.23(s,2H),4.21(d,J=35.1Hz,4H),3.66–3.52(m,4H),2.54(q,J=7.6Hz,2H),1.05(t,J=7.5Hz,3H),0.95–0.89(m,4H),0.02(s,9H),-0.05(d,J=3.4Hz,9H).
合成中间体MDI-1228-2:(S)-(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-(苄氧基)吡咯烷-1-基)甲酮
将三光气(25.8mg,0.09mmol)溶于5ml四氢呋喃中,0度滴加中间体MDI-1228-1(80mg,0.09mmol)的四氢呋喃(5ml)溶液,室温搅拌10分钟,加入(S)-3-(苄氧基)吡咯烷(31.9mg,0.18mmol)的四氢呋喃溶液,室温搅拌5分钟,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-1228-2 71mg,收率86.1%。
1H NMR(400MHz,CDCl3)δ8.46(d,J=8.3Hz,1H),7.53–7.51(m,2H),7.47–7.42(m,3H),7.39–7.29(m,6H),7.24(dd,J=8.4Hz,J=4.0Hz,1H),7.07–6.97(m,2H),5.95(s,2H),5.77(s,2H),5.23(s,2H),4.92–4.88(m,2H), 4.76–4.69(m,2H),4.60(s,2H),4.23(s,1H),3.76–3.70(m,2H),3.66–3.58(m,6H),2.54(q,J=7.5Hz,2H),2.15–2.13(m,1H),2.06–2.02(m,1H),1.05(t,J=7.5Hz,3H),0.95–0.91(m,4H),-0.01–-0.11(m,18H).
合成化合物MDI-1228:(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮
将中间体MDI-1228-2(83mg,0.11mmol)溶于甲醇(10ml)中,加入10mgPd/C,滴加浓盐酸(5ml),加热到50度,反应6小时,过滤,浓缩,加甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水,浓缩,制备板纯化,得到最终产物8mg,收率15.2%。
1H NMR(400MHz,MeOD-d4)δ8.27(d,J=8.4Hz,1H),7.43(d,J=1.0Hz,1H),7.17(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.81–4.61(m,4H),4.46–4.44(m,1H),3.79–3.69(m,2H),3.50–3.43(m,2H),2.56(q,J=7.5Hz,2H),2.09–1.99(m,2H),1.08(t,J=7.5Hz,3H).
实施例17:5-乙基-2-氟-4-(3-(5-(4-羟基环己基)-1,4,5,6-四氢吡咯并[3,4-d] 咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-217)
按照中国发明专利CN111606908B的实施例17所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.27(dd,J=4.0Hz,J=8.0Hz,1H),7.43-7.42(m,1H),7.19(dd,J=4.0Hz,J=8.0Hz,1H),6.96-6.88(m,2H),3.98(s,4H),3.93(m,1H),2.74-2.72(m,1H),2.58(q,J=8.0Hz,2H),2.04-2.05(m,1H), 1.90-1.80(m,5H),1.64-1.62(m,2H),1.10(t,J=8.0Hz,J=16.0Hz,3H).
实施例18:4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H- 吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218)
按照中国发明专利CN111606908B的实施例18所示方法进行合成,具体如下。
MDI-218的合成路线:
合成方法:
合成中间体MDI-218-1:6-溴-3-(5-(环丙磺酰基)-1-((2-(三甲基硅基)乙氧基)甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑
将2-(6-溴1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基硅基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酸叔丁酯(100mg,0.15mmol)溶于5ml二氯甲烷中,加入1ml三氟乙酸,室温搅拌30分钟,浓缩,用碳酸氢钠淬灭,用二氯甲烷萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩。把得到的化合物溶于5ml DCM和Et3N(0.08ml,0.59mmol)中,冷却到0度,缓慢加入环丙基磺酰氯(22.4mg,0.16mmol),室温反应2小时,加入水淬灭反应,用DCM萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-218-1,收率36.0%。
1H NMR(400MHz,CDCl3)δ8.37(d,J=8.0Hz,1H),7.80(d,J=4.0Hz,1H),7.43(d,J=8.0Hz,1H),5.91(s,2H),5.73(s,2H),4.75-4.74(m,2H),4.66-4.65(m,2H),3.63-3.58(m,4H),2.50-2.44(m,1H),1.33-1.31(m,2H),1.06-1.02(m,2H),0.96-0.91(m,4H),0.00--0.05(m,18H).
合成中间体MDI-218-2:3-(5-环丙磺酰基)-1-((2-(三甲基硅基)乙氧基)甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-6-(2-乙基-5-氟-4-((2-(三甲基硅基)乙氧基)甲氧基)苯基)-1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑
将中间体MDI-218-1(36.0mg,0.05mmol),(2-((5-乙基-2-氟-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯氧基)甲氧基)乙基)三甲基硅烷(25.5mg,0.06mmol),Pd(dppf)Cl2(3.9mg,0.005mmol)和磷酸钾(34.2mg,0.16mmol)溶于1,4-二氧六环(6ml)和水(1ml)中,氮气置换3次,加热到100度,反应过夜,冷却到室温,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-218-2,收率70.0%。
1H NMR(400MHz,CDCl3)δ8.47(d,J=8.0Hz,1H),7.48(s,1H),7.26(d,J=7.9Hz,1H),7.18(d,J=8.0Hz,1H),7.04(d,J=12.0Hz,1H),5.95(s,2H),5.78(s,2H),5.34(s,2H),4.76(s,2H),4.68(s,2H),3.88(t,J=8.0Hz,2H),3.68-3.57(m,4H),2.56(q,J=7.6Hz,2H),2.24(t,J=7.7Hz,1H),1.12-0.86(m,13H),-0.01--0.06(m,27H).
合成化合物MDI-218:4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚
将中间体MDI-218-2(36.0mg,0.04mmol)溶于甲醇(4ml)中,加入浓盐酸(2ml),加热到50度,反应6小时,浓缩,固体用1ml甲醇溶解,用碳酸氢钠溶液调pH=8-9,用二氯甲烷萃取4次,合并有机相,用无水硫酸钠干燥,制备板纯化,得到最终产物16mg,收率81.4%。
1H NMR(400MHz,MeOD-d4)δ8.27(d,J=8.0Hz,1H),7.43(s,1H),7.18(dd,J=8.0Hz,J=4.0Hz,1H),6.93(dd,J=20.0Hz,J=12.0Hz,2H),4.65(s,4H),2.76-2.69(m,1H),2.60-2.51(m,2H),1.20-1.18(m,2H),1.10-1.06(m,5H).
实施例19:4-(3-(5-(环丁基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)- 1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-219)
按照中国发明专利CN111606908B的实施例19所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.26(d,J=8.0Hz,1H),7.43(s,1H),7.17(dd,J=8.4Hz,J=1.4Hz,1H),6.93(dd,J=20.0Hz,J=12.0Hz,2H),4.60(s,4H),4.26-4.18(m,1H),2.68-2.52(m,4H),2.40-2.31(m,2H),2.13-2.02(m,2H),1.08(t,J=7.5Hz,3H).
实施例20:4-(3-(5-(环戊基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)- 1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-220)
按照中国发明专利CN111606908B的实施例20所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD-d4)δ8.27(dd,J=8.0Hz,J=4.0Hz,1H),7.43(s,1H),7.17(dd,J=8.0Hz,J=1.4Hz,1H),6.93(dd,J=20.0Hz,J=12.0Hz,2H),4.65(s,4H),3.91-3.83(m,1H),2.58-2.52(m,2H),2.13–2.03(m,4H),1.89-1.78(m,2H),1.75-1.64(m,2H),1.08(t,J=8.0Hz,3H).
实施例21:5-乙基-2-氟-4-(3-(5-((1-甲基-1H-吡唑-4-基)甲基)-1,4,5,6-四氢吡 咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-221)
按照中国发明专利CN111606908B的实施例21所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,MeOD-d4)δ8.25(d,J=8.0Hz,1H),7.68(s,1H),7.56(s,1H),7.42(s,1H),7.16(dd,J=8.4Hz,J=1.4Hz,1H),6.93(dd,J=20.0Hz,J=12.0Hz,2H),4.03-3.96(m,6H),3.92(s,3H),2.58-2.53(m,2H),1.08(t,J=8.0Hz,3H).
实施例22:4-(3-(5-环戊烷基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑 -6-基)-5-乙基-2-氟苯酚(MDI-224)
按照中国发明专利CN111606908B的实施例22所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,MeOD-d4)δ8.26(d,J=8.0Hz,1H),7.42(s,1H),7.17(d,J=8.0Hz,1H),6.93(dd,J=20.0Hz,J=12.0Hz,2H),4.05-3.94(m,4H),3.27-3.25(m,1H),2.59-2.54(m,2H),2.08-2.01(m,2H),1.87-1.79(m,2H),1.73-1.56(m,4H),1.08(t,J=8.0Hz,3H).
实施例23:5-乙基-2-氟-4-(3-(5-(四氢-2H-吡喃-4-基)-1,4,5,6-四氢吡咯并 [3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-225)
按照中国发明专利CN111606908B的实施例23所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,MeOD-d4)δ8.26(dd,J=12.0Hz,J=4.0Hz,1H),7.42(s,1H),7.16(dd,J=8.0Hz,J=4.0Hz,1H),6.93(dd,J=20.0Hz,J=12.0 Hz,2H),4.07-3.99(m,6H),3.52-3.49(m,2H),2.95-2.90(m,1H),2.58-2.53(m,2H),2.00-1.97(m,2H),1.69-1.59(m,2H),1.08(t,J=8.0Hz,3H).
实施例24:1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯 并[3,4-d]咪唑-5-(1H)-基)乙-1-酮(MDI-226)
按照中国发明专利CN111606908B的实施例24所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,甲醇-d4)δ8.28(d,J=8Hz,1H),7.43(s,1H),7.18(d,J=8Hz,1H),6.94(dd,J=22,10Hz,2H),4.79(s,2H),4.65(s,2H),2.59-2.53(m,2H),2.23(s,3H),1.08(t,J=7.5Hz,3H).
实施例25:1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯 并[3,4-d]咪唑-5-(1H)-基)丙-1-酮(MDI-227)
按照中国发明专利CN111606908B的实施例25所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,DMSO-d6)δ13.29(s,1H),12.80(s,1H),9.85(s,1H),8.33(d,J=8Hz,1H),7.40(s,1H),7.12(d,J=8Hz,1H),7.03(d,J=12Hz, 1H),6.92(d,J=12Hz,1H),4.73-4.58(m,2H),4.50-4.42(m,2H),2.50-2.47(m,2H),2.43-2.37(m,2H),1.08-1.01(m,6H).
实施例26:1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯 并[3,4-d]咪唑-5-(1H)-基)-2-甲基丙-1-酮(MDI-228)
按照中国发明专利CN111606908B的实施例26所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,MeOD-d4)δ8.26(d,J=8.0Hz,1H),7.43(s,1H),7.17(d,J=8Hz,1H),6.93(dd,J=20,12Hz,2H),4.83-4.59(m,4H),2.94-2.90(m,1H),2.58-2.52(m,2H),1.22(d,J=8.0Hz,6H),1.08(t,J=8.0Hz,3H).
实施例27:2-环丙基-1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6- 二氢吡咯并[3,4-d]咪唑-5-(1H)-基)乙-1-酮(MDI-229)
按照中国发明专利CN111606908B的实施例27所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,MeOD-d4)δ8.29(d,J=8.0Hz,1H),7.43(s,1H),7.19-7.17(m,1H),6.98-6.90(m,2H),4.73-4.61(m,4H),2.59-2.53(m,2H),2.46(d,J=8.0Hz,2H),1.17(m,1H),1.08(t,J=8.0Hz,3H),0.64-0.59(m,2H),0.30-0.26(m,2H).
实施例28:1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯 并[3,4-d]咪唑-5-(1H)-基)-3-甲基丁-1-酮(MDI-230)
按照中国发明专利CN111606908B的实施例28所示方法进行合成,所得产物的核磁共振氢谱如下:
1H NMR(400MHz,MeOD-d4)δ8.29–8.26(m,1H),7.43(s,1H),7.19–7.16(m,1H),6.97–6.89(m,2H),4.75–4.70(m,4H),2.56(q,J=7.5Hz,2H),2.36(m,2H),2.29–2.20(m,1H),1.10–1.05(m,9H).
实施例29:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231)
按照中国发明专利CN111606908B的实施例29所示方法进行合成,具体如下。
MDI-231的合成路线:
合成方法:
合成中间体MDI-231-1:(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2--(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮
将三光气(64.4mg,0.21mmol)溶于15ml二氯甲烷中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(150mg,0.21mmol)的二氯甲烷(5ml)溶液,加入三乙胺(63.6mg,0.63mmol),室温搅拌5分钟,加入吡咯烷(29.8mg,0.42mmol)的二氯甲烷溶液,室温搅拌10分钟,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-231-1 140mg,收率82.4%。
1H NMR(400MHz,CDCl3)δ8.48(d,J=8Hz,1H),7.53-7.38(m,6H),7.22(d,J=8Hz,1H),7.03(d,J=12Hz,1H),6.95(d,J=8Hz,1H),5.96(s,2H),5.77(s,2H),5.23(s,2H),4.81(s,2H),4.67(s,2H),3.66-3.59(m,4H),3.53-3.51(m,4H),2.56-2.52(m,2H),1.93-1.88(m,4H),1.03(t,J=8Hz,3H),0.93-0.87(m,4H),-0.05--0.09(m,18H).
合成化合物MDI-231:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮
将中间体MDI-231-1(140mg,0.173mmol)溶于甲醇(6ml)中,加入15mgPd/C,滴加浓盐酸(3ml),加热到50度,反应6小时,过滤,浓缩,加甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水,浓缩,制备板纯化,得到最终产物21mg,收率26.3%。
1H NMR(400MHz,DMSO-d6)δ13.25(s,1H),12.69(s,1H),9.83(s,1H),8.31(d,J=8Hz,1H),7.39(s,1H),7.11(d,J=8Hz,1H),7.02(d,J=12Hz,1H),6.91(d,J=12Hz,1H),4.57-4.56(m,2H),4.49-4.48(m,2H),3.32-3.31(m,4H),2.48-2.44(m,2H),1.85-1.79(m,4H),1.02(t,J=7Hz,3H).
实施例30:N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288)
MDI-1288的合成路线:
具体合成方法:
合成化合物MDI-1288-2:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1-(四氢-2H-吡喃-2-基)-1H-吲唑-3-基)-1-((2-三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基氮杂环丁烷-1-基)甲酮
将MDI-1288-1(根据合成MDI-231-1的方法,使用合适的起始原料制得,51.00g,66.49mmol)溶于500ml四氢呋喃中,加入7g钯碳,置换氢气,40度反应16小时,反应完毕,过滤,滤液浓缩,得MDI-1288-I07共44.10g,收率97.8%。
合成化合物MDI-1288:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基氮杂环丁烷-1-基)甲酮
将MDI-1288-I07(44.10g,65.01mmol)溶于240ml甲醇,加入120ml浓盐酸,50度反应过夜。反应完毕,反应液过滤,滤饼50度干燥箱干燥,得29.8g。用150ml甲醇打浆0.5h后得到27.2g盐酸盐粗品,然后经重结晶提纯。
1H NMR(400MHz,DMSO-d6)δ14.25(s,1H),9.90(s,1H),8.43(d,J=8.4Hz,1H),7.56(s,1H),7.27(dd,J=8.5,1.4Hz,1H),7.04(d,J=11.8Hz,1H),6.97(d,J=9.1Hz,1H),6.71(s,1H),4.59(s,4H),3.83–3.78(m,1H),3.68(dd,J=11.2,4.2Hz,1H),3.54(dd,J=11.2,6.0Hz,1H),3.28–3.15(m,2H),2.48(q,J=7.5Hz,2H),1.02(t,J=7.5Hz,3H).
实施例31:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233)
按照中国发明专利CN111606908B的实施例31所示方法进行合成,具体如下。
MDI-233的合成路线:
合成方法:
合成中间体MDI-233-1:(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮
将三光气(54.1mg,0.182mmol)溶于5ml四氢呋喃中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(130mg,0.182mmol)的四氢呋喃(5ml)溶液,加入三乙胺(55.2mg,0.550mmol),室温搅拌5分钟,加入哌啶盐酸盐(44.4mg,0.364mmol)的四氢呋喃溶液,室温搅拌10分钟,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-233-1 100mg,收率66.6%。
1H NMR(400MHz,CDCl3)δ8.44(d,J=8.3Hz,1H),7.50–7.48(m,2H),7.44–7.35(m,4H),7.23–7.20(m,1H),7.04–6.94(m,2H),5.93(s,2H),5.74(s,2H),5.20(s,2H),4.69(d,J=54.8Hz,4H),3.64–3.56(m,4H),3.31(s,4H),2.54(q,J=7.5Hz,2H),1.64(s,6H),1.03(t,J=7.5Hz,3H),0.93–0.86(m,4H),-0.07(d,J=2.7Hz,18H).
合成化合物MDI-233:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮
将中间体MDI-233-1(100mg,0.121mmol)溶于甲醇(6ml)中,加入10mg10%Pd/C,滴加浓盐酸(3ml),加热到50度,反应6小时,过滤,浓缩, 加甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水,浓缩,制备板纯化,得到最终产物33mg,收率57.3%。
1H NMR(400MHz,MeOD-d4)δ8.25(d,J=8.4Hz,1H),7.40(s,1H),7.16–7.14(m,1H),6.95–6.87(m,2H),4.83–4.65(m,4H),3.35–3.33(m,4H),2.54(q,J=7.5Hz,2H),1.67–1.65(m,6H),1.06(t,J=7.5Hz,3H).
实施例32:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234)
按照中国发明专利CN111606908B的实施例32所示方法进行合成,具体如下。
MDI-234的合成路线:
合成方法:
合成中间体MDI-234-1:(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮
将三光气(54.1mg,0.182mmol)溶于5ml四氢呋喃中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(130mg,0.182mmol)的四氢呋喃(5ml)溶液,加入三乙胺(55.1mg,0.546mmol),室温搅拌5分钟,加入吗啉(31.7mg,0.364mmol)的四氢呋喃溶液,室温搅拌10分钟,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-234-1120mg,收率79.7%。
1H NMR(400MHz,CDCl3)δ8.45(d,J=8.3Hz,1H),7.53–7.51(m,2H),7.47–7.35(m,4H),7.26–7.23(m,1H),7.06–6.97(m,2H),5.96(s,2H),5.77(s,2H),5.23(s,2H),4.68(d,J=54.8Hz,4H),3.80–3.78(m,3H),3.67–3.59(m,4H),3.43–3.40(m,3H),3.27–3.21(m,6H),2.54(q,J=7.5Hz,2H),1.05(t,J=7.5Hz,3H),0.96–0.89(m,4H),-0.04(d,J=2.7Hz,18H).
合成化合物MDI-234:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮
将中间体MDI-234-1(120mg,0.145mmol)溶于甲醇(6ml)中,加入12mg10%Pd/C,滴加浓盐酸(3ml),加热到50度,反应6小时,过滤,浓缩,加甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水,浓缩,制备板纯化,得到最终产物42mg,收率60.9%。
1H NMR(400MHz,MeOD-d4)δ8.28(d,J=8.4Hz,1H),7.43(s,1H),7.19–7.16(m,1H),6.97–6.90(m,2H),4.71–4.66(m,4H),3.78–3.75(m,4H),3.41-3.39(m,4H),2.54(q,J=7.5Hz,2H),1.06(t,J=7.5Hz,3H).
实施例33:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235)
按照中国发明专利CN111606908B的实施例33所示方法进行合成,具体如下。
MDI-235的合成路线:
合成方法:
合成中间体MDI-235-1:(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮
将三光气(8.3mg,0.028mmol)溶于5ml四氢呋喃中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(20mg,0.028mmol)的四氢呋喃(5ml)溶液,加入三乙胺(8.5mg,0.084mmol),室温搅拌5分钟,加入1-甲基哌嗪(5.60mg,0.056mmol)的四氢呋喃溶液,室温搅拌10分钟,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-235-1 20mg,收率85.1%。
1H NMR(400MHz,CDCl3)δ8.44(d,J=8.3Hz,1H),7.50–7.48(m,2H),7.44–7.33(m,4H),7.23–7.21(m,1H),7.04–6.94(m,2H),5.93(s,2H),5.75(s,2H),5.20(s,2H),4.70(d,J=54.8Hz,4H),3.64–3.56(m,4H),3.43–3.41 (m,4H),2.56–2.49(m,6H),2.34(s,3H),1.03(t,J=7.5Hz,3H),0.93–0.86(m,4H),-0.07(d,J=2.7Hz,18H).
合成化合物MDI-235:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮
将中间体MDI-235-1(20mg,0.024mmol)溶于甲醇(6ml)中,加入5mgPd/C,滴加浓盐酸(3ml),加热到50度,反应6小时,过滤,浓缩,加甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水,浓缩,制备板纯化,得到最终产物3mg,收率14.8%。
1H NMR(400MHz,MeOD-d4)δ8.25(d,J=8.4Hz,1H),7.40(s,1H),7.16–7.14(m,1H),6.95–6.87(m,2H),4.83–4.66(m,4H),3.44–3.41(m,4H),2.56–2.51(m,6H),2.35(s,3H),1.06(t,J=7.5Hz,3H).
实施例34:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236)
按照中国发明专利CN111606908B的实施例34所示方法进行合成,具体如下。
MDI-236的合成路线:
合成方法:
合成中间体MDI-236-1:(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2--(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮
将三光气(54.07mg,0.182mmol)溶于15ml二氯甲烷中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(130mg,0.182mmol)的二氯甲烷(5ml)溶液,加入三乙胺(55.2mg,0.55mmol),搅拌5分钟,加入乙基哌嗪(41.5mg,0.364mmol)的二氯甲烷溶液,室温搅拌10分钟,加入水,用乙酸乙酯萃取2次,合并有机相,用水和饱和食盐水洗,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-236-1 100mg,收率64.3%。
1H NMR(400MHz,CDCl3)δ8.44(d,J=8Hz,1H),8.28(s,1H),7.50-7.33(m,5H),7.22(d,J=8Hz,1H),7.03(d,J=12Hz,1H),6.95(d,J=8Hz,1H),5.93(s,2H),5.74(s,2H),5.20(s,2H),4.77(s,2H),4.63(s,2H),3.63-3.61(m,4H),3.43-3.42(m,4H),2.53-2.46(m,8H),1.03(t,J=6Hz,3H),0.93-0.86(m,7H),-0.06--0.08(m,18H).
合成化合物MDI-236:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮
将中间体MDI-236-1(100mg,0.117mmol)溶于甲醇(10ml)中,加入10mgPd/C,滴加浓盐酸(5ml),加热到50度,反应6小时,过滤,浓缩,加甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水,浓缩,制备板纯化,得到最终产物21mg,收率35.6%。
1H NMR(400MHz,MeOD-d4)δ8.27(d,J=8Hz,1H),7.43(s,1H),7.17(d,J=8Hz,1H),6.96(d,J=12Hz,1H),6.91(d,J=8Hz,1H),4.75-4.60(m,4H),3.48-3.44(m,4H),2.61-2.48(m,8H),1.17(t,J=8Hz,3H),1.08(t,J=8Hz,3H).
实施例35:环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-b]吡啶-3- 基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-237)
按照中国发明专利CN111606908B的实施例35所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,DMSO)δ13.60(s,1H),12.60-12.48(m,1H),10.02(s,1H),8.53(d,J=1.6Hz,1H),7.95(s,1H),7.16(d,J=11.8Hz,1H),6.98(d,J=9.1Hz,1H),4.91-4.41(m,4H),2.51-2.47(m,2H),1.96-1.84(m,1H),1.03(t,J=8.0Hz,3H),0.87-0.80(m,4H).LC-MS m/z(ESI)[M+H]+针对C23H22FN6O2的计算值为:433.2;测量值为:433.2.
实施例36:环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)- 4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-239)
按照中国发明专利CN111606908B的实施例36所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ7.27(s,1H),6.95-6.88(m,3H),4.96(s,2H),4.66(s,2H),2.63(s,3H),2.55(q,J=7.5Hz,2H),1.98-1.92(m,1H),1.07(t,J=7.5Hz,3H),1.04-0.92(m,4H).
实施例37:(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6- 二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-240)
按照中国发明专利CN111606908B的实施例37所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ7.27(s,1H),6.95-6.88(m,3H),4.85-4.82(m,2H),4.62-4.59(m,2H),4.46-4.45(m,1H),3.79-3.69(m,2H),3.64-3.57(m,1H),3.46-3.42(m,1H),2.61(s,3H),2.56(q,J=7.5Hz,2H),2.09-1.98(m,2H),1.07(t,J=7.5Hz,3H).
实施例38:环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-c]吡啶-3- 基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-242)
按照中国发明专利CN111606908B的实施例38所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ9.61(d,J=1.0Hz,1H),7.77(d,J=1.1Hz,1H),7.16(d,J=11.6Hz,1H),6.93(d,J=8.8Hz,1H),5.07-4.88(m,2H),4.68-4.62(m,2H),2.69-2.64(m,2H),1.98-1.89(m,1H),1.09-1.05(m,3H),1.01-0.98(m,2H),0.96-0.94(m,2H).
实施例39:(R)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡 咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-243)
按照中国发明专利CN111606908B的实施例39所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(d,J=1.0Hz, 1H),7.17(d,J=8.4Hz,1H),6.98–6.90(m,2H),4.82–4.60(m,4H),4.47–4.45(m,1H),3.79–3.70(m,2H),3.60–3.57(m,1H),3.46–3.43(m,1H),2.56(q,J=7.5Hz,2H),2.09–1.98(m,2H),1.08(t,J=7.5Hz,3H).
实施例41:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245)
按照中国发明专利CN111606908B的实施例41所示方法进行合成,具体如下。
MDI-245的合成路线:
合成方法:
合成中间体MDI-245-1:(2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-羟基哌啶-1-基)甲酮
将三光气(54.1mg,0.18mmol)溶于10ml干燥二氯甲烷中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(130mg,0.18mmol)的二氯甲烷(5ml)溶液,加完后缓慢滴加干燥三乙胺(185mg,1.8mmol)室温搅拌10分钟,TLC监测原料消失,加入哌啶-4-醇(36.9mg,0.36mmol)的二氯甲烷(5ml)溶液,室温搅拌20分钟,加水淬灭,用二氯甲烷萃取2次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-245-1 116mg,收率75.7%。
1H NMR(400MHz,CDCl3)δ8.46(d,J=8.3Hz,1H),7.53–7.51(m,2H),7.47–7.35(m,4H),7.25(d,J=8.4Hz,1H),7.06-6.97(m,2H),5.96(s,2H),5.75(s,2H),5.37(s,2H),4.79–4.66(m,4H),3.95–3.92(m,1H),3.75–3.72(m,2H),3.66-3.52(m,4H),3.12–3.07(m,2H),2.54(q,J=7.6Hz,2H),2.02–1.91(m,2H),1.68–1.63(m,2H),1.06(t,J=7.5Hz,3H),0.99–0.89(m,4H),0.02(s,9H),-0.05(d,J=3.4Hz,9H).
合成化合物MDI-245:(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-羟基哌啶-1-基)甲酮
将MDI-245-1(116mg,0.14mmol)溶于20ml甲醇中,加入20mg钯碳,置换氢气,40度反应1小时,反应完毕,过滤,滤液浓缩,浓缩物溶于12ml甲醇,加入6ml浓盐酸,50度反应7小时,浓缩,加甲醇带3次盐酸,浓缩,溶于8ml甲醇,加0.8ml氨水,浓缩,制备板纯化,得到最终产物30mg,收率44.4%。
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.72–4.65(m,4H),3.88–3.82(m,1H),3.76–3.73(m,2H),3.13–3.06(m,2H),2.56(q,J=7.5Hz,2H),1.97–1.95(m,2H),1.63–1.55(m,2H),1.08(t,J=7.5Hz,3H).
实施例42:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-甲基-4,6-二氢 吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-246)
按照中国发明专利CN111606908B的实施例42所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.56(s,4H),2.84(s,3H),2.56(q,J=7.5Hz,2H),1.08(t,J=7.5Hz,3H).
实施例43:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢 吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247)
按照中国发明专利CN111606908B的实施例43所示方法进行合成,具体如下。
MDI-247的合成路线:
合成方法:
合成中间体MDI-247-1:2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-N-乙基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺
将三光气(22.9mg,0.08mmol)溶于6ml干燥二氯甲烷中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(55mg,0.08mmol)的二氯甲烷(5ml)溶液,加完后缓慢滴加干燥三乙胺(78.0mg,0.8mmol)室温搅拌10分钟,TLC监测原料消失,加入乙胺盐酸盐(12.6mg,0.16mmol),室温搅拌2小时,加水淬灭,用二氯甲烷萃取2次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-247-1 47mg,收率77.7%。
1H NMR(400MHz,CDCl3)δ8.46(d,J=8.3Hz,1H),7.53–7.51(m,2H),7.47–7.35(m,4H),7.25(d,J=8.4Hz,1H),7.07–6.97(m,2H),5.96(s,2H),5.78(s,2H),5.23(s,2H),4.72–4.54(m,4H),3.65–3.58(m,4H),3.45–3.38(m,2H),2.54(q,J=7.6Hz,2H),1.18–1.14(m,3H),1.05(t,J=7.5Hz,3H),0.95–0.89(m,4H),0.02(s,9H),-0.05(d,J=3.4Hz,9H).
合成化合物MDI-247:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺
将MDI-247-1(47mg,0.06mmol)溶于10ml甲醇中,加入8mg钯碳,置换氢气,40度反应1小时,反应完毕,过滤,滤液浓缩,浓缩物溶于6ml甲醇,加入3ml浓盐酸,50度反应7小时,浓缩,加甲醇带3次盐酸,浓缩,溶于5ml甲醇,加0.5ml氨水,浓缩,制备板纯化,得到最终产物11mg,收率42.4%。
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.98–6.90(m,2H),4.57(s,4H),3.38–3.28(m,2H),2.56(q,J=7.5Hz,2H),1.21(t,J=7.2Hz,3H),1.08(t,J=7.5Hz,3H).
实施例44:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)- 4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248)
按照中国发明专利CN111606908B的实施例44所示方法进行合成,具体如下。
MDI-248的合成路线:
合成方法:
合成中间体MDI-248-1:2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-N-(2-羟基乙基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺
将三光气(22.9mg,0.08mmol)溶于6ml干燥二氯甲烷中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(55mg,0.08mmol)的二氯甲烷(5ml)溶液,加完后缓慢滴加干燥三乙胺(78.0mg,0.8mmol)室温搅拌10分钟,TLC监测原料消失,加入乙醇胺(9.4mg,0.16mmol)的二氯甲烷(5ml)溶液,室温搅拌1小时,加水淬灭,用二氯甲烷萃取2次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-248-1 44mg,收率71.3%。
1H NMR(400MHz,CDCl3)δ8.46(d,J=8.3Hz,1H),7.73–7.51(m,2H),7.48–7.35(m,4H),7.27–7.24(m,1H),7.07–6.97(m,2H),5.96(s,2H),5.78(s,2H),5.23(s,2H),4.73–4.57(m,4H),3.65–3.53(m,6H),3.33–3.29(m,2H),2.54(q,J=7.6Hz,2H),1.05(t,J=7.5Hz,3H),0.95–0.89(m,4H),0.02(s,9H),-0.05(s,9H).
合成化合物MDI-248:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺
将MDI-248-1(44mg,0.06mmol)溶于10ml甲醇中,加入8mg钯碳,置换氢气,40度反应1小时,反应完毕,过滤,滤液浓缩,浓缩物溶于6ml甲醇,加入3ml浓盐酸,50度反应7小时,浓缩,加甲醇带3次盐酸,浓缩,溶于5ml甲醇,加0.5ml氨水,浓缩,制备板纯化,得到最终产物14mg,收率56.6%。
1H NMR(400MHz,MeOD)δ8.28(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.59(s,4H),3.68(t,J=5.8Hz,2H),3.40(t,J=5.8Hz,2H),2.56(q,J=7.5Hz,2H),1.08(t,J=7.5Hz,3H)
实施例46:1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-1,4,5,6-四氢吡 咯并[3,4-d]咪唑-5-羰基)吡咯烷-3-腈(MDI-250)
按照中国发明专利CN111606908B的实施例46所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(s,1H),7.18 (d,J=8.4Hz,1H),6.98–6.90(m,2H),4.70(s,4H),3.89–3.85(m,1H),3.78–3.76(m,1H),3.70–3.59(m,2H),3.23–3.18(m,1H),2.56(q,J=7.5Hz,2H),2.42–2.19(m,2H),1.08(t,J=7.5Hz,3H).
实施例47:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(四氢呋喃-3- 基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-251)
按照中国发明专利CN111606908B的实施例47所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.28(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.63(s,4H),4.45–4.40(m,1H),4.03–3.94(m,2H),3.87–3.81(m,1H),3.71–3.68(m,1H),2.56(q,J=7.5Hz,2H),2.32–1.87(m,2H),1.08(t,J=7.5Hz,3H).
实施例48:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5(1H)-羧酸甲酯(MDI-252)
按照中国发明专利CN111606908B的实施例48所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.28(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.61(s,4H),3.83(s,3H),2.56(q,J=7.5Hz,2H),1.08(t,J=7.5Hz,3H).
实施例49:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并 [3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253)
按照中国发明专利CN111606908B的实施例49所示方法进行合成,具体如下。
MDI-253的合成路线:
合成方法:
合成中间体MDI-253-1:2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯
将三光气(20.1mg,0.07mmol)溶于5ml干燥二氯甲烷中,0度滴加中间体6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-3-(1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(48mg,0.07mmol)的二氯甲烷(5ml)溶液,加完后缓慢滴加干燥三乙胺(68.1mg,0.67mmol)室温搅拌10分钟,TLC监测原料消失,反应液浓缩,溶于10ml乙醇,加入DMAP(8.2mg,0.07mmol),80度反应4小时,反应液浓缩加水,用二氯甲烷萃取2次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,硅胶柱纯化,得中间体MDI-253-1 33mg,收率62.5%。
1H NMR(400MHz,CDCl3)δ8.50–8.45(m,1H),7.53–7.51(m,2H),7.47–7.37(m,4H),7.25(d,J=8.4Hz,1H),7.07–6.96(m,2H),5.96(s,2H),5.77(s,2H),5.23(s,2H),4.72–4.59(m,4H),4.29–4.25(m,2H),3.65–3.58(m,4H),2.54(q,J=7.6Hz,2H),1.38–1.34(m,3H),1.05(t,J=7.5Hz,3H),0.95–0.89(m,4H),0.02(s,9H),-0.05(s,9H).
合成化合物MDI-253:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯
将MDI-253-1(33mg,0.04mmol)溶于10ml乙醇中,加入6mg钯碳,置换氢气,40度反应1小时,反应完毕,过滤,滤液浓缩,浓缩物溶于6ml乙醇,加入3ml浓盐酸,50度反应7小时,浓缩,加乙醇带3次盐酸,浓缩,溶于5ml乙醇,加0.5ml氨水,浓缩,制备板纯化,得到最终产物10mg,收率54.8%。
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),6.97–6.90(m,2H),4.60(s,4H),4.26(q,J=7.1Hz,2H),2.57(q,J=7.5Hz,2H),1.36(t,J=7.1Hz,3H),1.08(t,J=7.5Hz,3H).
实施例50:(S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[3,4-b]吡啶-3- 基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI- 255)
按照中国发明专利CN111606908B的实施例50所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,DMSO)δ13.61(s,1H),10.22(s,1H),8.78(d,J=8.0Hz,1H),7.71(d,J=8.0Hz,1H),7.34(d,J=12.0Hz,1H),6.97(d,J=8.0Hz,1H),4.93(d,J=4.0Hz,1H),4.75-4.42(m,4H),4.30-4.27(m,1H),3.58-3.53(m,2H),3.41-3.40(m,1H),3.26-3.23(m,1H),2.73-2.71(m,2H),2.01-1.79(m,2H),1.09(t,J=8.0Hz,3H).
实施例51:3-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯 并[3,4-d]咪唑-5(1H)-基)-3-氧丙腈(MDI-256)
按照中国发明专利CN111606908B的实施例51所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.27(d,J=8.0Hz,1H),7.43(s,1H),7.18(d,J=8.0Hz,1H),6.97(dd,J=8.0Hz,J=20.0Hz,2H),4.77-4.70(m,4H), 3.62(s,2H),2.59-2.53(m,2H),1.09(t,J=8.0Hz,3H).
实施例52:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6- 二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257)
按照中国发明专利CN111606908B的实施例52所示方法进行合成,具体如下。
MDI-257的合成路线:
合成方法:
合成中间体MDI-257-1:2-(6-(4-(苄氧基)-2-乙基-5-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吲唑-3-基)-N,N-二甲基-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺
合成过程类似中国发明专利CN111606908B中的中间体MDI-246-1的合成方法,用二甲胺盐酸盐替代甲胺盐酸盐。
合成化合物MDI-257:2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺
将中间体MDI-257-1(41mg,0.05mmol)溶于甲醇(6ml)中,加入8mg10%Pd/C,氢气置换3次,加热到40度,反应1小时,过滤,浓缩,加甲醇4ml和浓盐酸1ml,加热到50度,反应6小时,浓缩,用甲醇带3次盐酸,浓缩,溶于甲醇,加1ml氨水中和,浓缩,制备板纯化,得到最终产物8mg,收率35.2%。
1H NMR(400MHz,DMSO)δ13.28(s,1H),9.85(s,1H),8.32(d,J=8.0Hz,1H),7.40(s,1H),7.13(d,J=8.0Hz,1H),7.03(d,J=12.0Hz,1H),6.93(d,J=12.0Hz,1H),4.54-4.53(m,4H),2.85(s,6H),2.50-2.46(m,2H),1.04(t,J=8.0Hz,3H).
实施例53:N-(2-氰基乙基)-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)- 4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-258)
按照中国发明专利CN111606908B的实施例53所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.25(d,J=8.4Hz,1H),7.41(s,1H),7.16(d,J=8.4Hz,1H),6.91(dd,J=20.8,10.3Hz,2H),4.61-4.54(m,4H),3.55-3.50(m,2H),2.66-2.51(m,4H),1.06(t,J=7.5Hz,3H).
实施例54:N-环丙基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二 氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-259)
按照中国发明专利CN111606908B的实施例54所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.25(d,J=8.4Hz,1H),7.43(s,1H),7.16(dd,J=8.4,1.4Hz,1H),6.91(dd,J=20.6,10.4Hz,2H),4.66-4.48(m,4H),2.68-2.62(m,1H),2.59-2.53(m,2H),1.08(t,J=7.5Hz,3H),0.76-0.71(m,2H),0.60-0.56(m,2H).
实施例55:N-环丁基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二 氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-260)
按照中国发明专利CN111606908B的实施例55所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,1H),7.43(s,1H),7.18(d,J=8.4Hz,1H),7.01-6.85(m,2H),4.57(s,4H),4.35-4.31(m,1H),2.59-2.53(m,2H),2.36-2.30(m,2H),2.11-2.04(m,2H),1.76-1.69(m,2H),1.08(t,J=7.5Hz,3H).
实施例57:(S)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯 并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-262)
按照中国发明专利CN111606908B的实施例57所示方法进行合成,所得产物的核磁共振氢谱数据如下:。
1H NMR(400MHz,MeOD)δ8.28(d,J=8.0Hz,1H),7.44(s,1H),7.18(d,J=8.5Hz,1H),6.96(d,J=11.7Hz,1H),6.91(d,J=8.9Hz,1H),4.80-4.64(m,4H),4.09-4.05(m,1H),3.26-3.22(m,2H),2.59-2.53(m,2H),2.06-1.86(m,4H),1.08(t,J=8.0Hz,3H).LC-MS m/z(ESI)[M+H]+针对C25H26FN6O2的计算值为:461.2;测量值为:461.2.
实施例58:(R)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯 并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-263)
按照中国发明专利CN111606908B的实施例58所示方法进行合成,所得产物的核磁共振氢谱数据如下:
1H NMR(400MHz,MeOD)δ8.27(d,J=8.0Hz,1H),7.44(s,1H),7.18(d,J=8.4Hz,1H),6.96(d,J=12.2Hz,1H),6.91(d,J=8.8Hz,1H),4.82-4.60(m,4H),4.21-4.15(m,1H),3.33-3.23(m,1H),3.08-2.99(m,1H),2.59-2.53(m,2H),2.08-1.86(m,4H),1.08(t,J=8.0Hz,3H).
实施例59:化合物对于TRKA、TRKB、TRKC的抑制活性测试
1.实验材料和仪器
2.实验方法
2.1制备1X激酶反应缓冲液:
1倍体积的5X激酶反应缓冲液和4倍体积的水;5mM MgCl2;1mM DTT。
2.2激酶和底物准备:
2.5X底物混合物配制
2.3化合物的抑制活性测试过程:
1)在稀释板中用DMSO对化合物进行4倍稀释,化合物起始浓度为1000nM。
2)将化合物50倍稀释到1X激酶反应缓冲液中,在振荡器上震荡20分钟。
3)用1X的酶反应缓冲液配制准备2X TRKA。
4)向反应板中每孔加入2μl TRKA激酶(步骤3中配制)。
5)向每孔加入1μl在缓冲液中稀释好的化合物,用封板膜封住板子1000g离心30秒,室温放置10分钟。
6)用1X的酶反应缓冲液配制4x ATP&sub混合液,向反应板中加入1μl 4x ATP&sub混合液。
7)用封板膜封住板子1000g离心30秒,室温反应60分钟。
8)转移4μL ADP-Glo到384反应板中1000rpm/min,离心1min,25℃孵育40min。
9)转移8μL Detection溶液到384反应板中1000rpm/min,离心1min,25℃孵育40min。
10)使用Biotek多功能读板机读取RLU(Relative luminescence unit)信号。信号强度用于表征激酶的活性程度。
3.数据分析
3.1抑制率计算如下:
化合物抑制率(%inh)=100%-(化合物-阳性对照)/(阴性对照-阳性对照)*100%
阴性对照:DMSO
阳性对照:LOXO101
3.2计算IC50并绘制化合物的抑制曲线:
利用以下非线性拟合公式来得到化合物的IC50(半数抑制浓度):用Graphpad 6.0软件进行数据分析。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*Hill Slope))
X:化合物浓度log值
Y:抑制率(%inh)
3.3质检
一位实验员整理数据后,由另一位实验员再次进行检查,以确保数据分析的准确性。
实验数据确保:Z factor>0.5;S/B>2;阳性对照IC50在历次平均值3倍以内。
3.化合物活性测试结果
实施例60:化合物对于RET激酶的抑制活性测试
1.实验材料和仪器
2.实验方法
2.1制备1X激酶反应缓冲液:
1倍体积的5X激酶反应缓冲液和4倍体积的水;5mM MgCl2;1mM DTT。
2.2反应条件:
2.3化合物的抑制活性测试过程:
1)在稀释板中用DMSO对化合物进行4倍稀释,化合物起始浓度为1uM。
2)将化合物40倍稀释到1X激酶反应缓冲液中,在振荡器上震荡20分钟。
3)用1X的酶反应缓冲液配制准备2.5X激酶。
4)向反应板中每孔加入2μl激酶(步骤3中配制)。
5)向每孔加入1μl在缓冲液中稀释好的化合物,用封板膜封住板子1000g离心30秒,室温放置10分钟。
6)用1X的酶反应缓冲液配制2.5x Tk-substrate-biotin和ATP混合液,向反应板中加入2μl Tk-substrate-biotin/ATP混合液。
7)用封板膜封住板子1000g离心30秒,室温反应30分钟。
8)用HTRF检测缓冲液配制5X Sa-XL 665(250nM)。
9)每孔加入5μl XL665和5μl Tk-antibody-Cryptate,1000g离心30秒,室温反应1h。
10)用Biotek读615nm(Cryptate)和665nm(XL665)的荧光信号。
3.数据分析
3.1计算每孔的比率(Ratio_665/615nm),然后按如下计算抑制率:
化合物抑制率(%inh)=100%-(化合物-阳性对照)/(阴性对照-阳性对照)*100%
阳性对照:Pralsetinib 1000nM
阴性对照:0.5%DMSO
3.2计算IC50并绘制化合物的抑制曲线:
利用以下非线性拟合公式来得到化合物的IC50(半数抑制浓度):用Graphpad 6.0软件进行数据分析。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*Hill Slope))
X:化合物浓度log值
Y:抑制率(%inh)
3.3质检
一位实验员整理数据后,由另一位实验员再次进行检查,以确保数据分析的准确性。
实验数据确保:Z factor>0.5;S/B>2;阳性对照IC50在历次平均值3倍以内。
3.化合物活性测试结果
实施例61:化合物对卵清蛋白诱导的小鼠哮喘模型的治疗效果
一、材料和方法
实验材料和试剂
卵清蛋白(OVA):Sigma-A5243
铝佐剂:Sigma-239186
Diff Quick:国药集团,100092680
苏木素:鸿泉生物,HQ60002
伊红:鸿泉生物,HQ60001
白介素5(IL-5)Elisa试剂盒:贝因莱-DRE30011
IgE Elisa试剂盒:贝因莱-DRE30653
磷酸盐缓冲液(PBS):Solarbio-P1022
吐温80:Solarbio-LA7760
实验仪器
电子天平:五鑫衡器,型号MAX-A3003
酶标仪:芬兰(Labsystems Multiskan MS),型号352
洗板机:芬兰(Thermo Labsystems),型号AC8
离心机:微量高速离心机,型号TGL-16
移液器:吉尔森P型移液器(Pipetman),型号F123601
培养箱:隔水式恒温培养箱,型号BPN-190RHP
显微镜:OLYMPUS,型号CX41
恒温烘箱:上海恒一科学仪器,型号DHG-9140
石蜡切片机:徕克,型号SQ2125
摊片机:徕克,型号PPTHK-21B
水浴锅:Leica,型号HI1210
雾化器:江苏鱼跃医疗,型号403M
实验动物
7-8周龄雌性Balb/c小鼠购自常州卡文斯实验动物有限公司,动物合格证号:202005500。
小鼠饲养于上海中科院药物所动物房,SPF级,温度22-24℃,湿度40%-70%,光照7:00-19:00,每笼3只。食物和水:自由饮食。
动物分组
动物到达后,经过7天的适应,根据体重对动物进行随机分组,实验分为8组,每组8只,详见下表。

a为0.2%吐温80/生理盐水。BID-每天两次,QD-每天一次,NA-无。
试剂配制
●免疫制剂配置:称取适量OVA,加入适量PBS,使其终浓度为0.1mg/ml。加入等体积的铝佐剂,混匀,4度放置,现配现用。
●致敏剂的配置(5%OVA):称取适量OVA,加入适量PBS,使其终浓度为5%。混匀,4度放置备用,现配现用。
●溶媒(Vehicle)配制:每100毫升生理盐水加入200ul吐温80,混匀。4度放置备用。
●化合物MDI-1288溶液配制:称取适量MDI-1288,溶媒,待其溶解,使其终浓度为1.2mg/ml。混匀,4度放置备用,三天配制一次。
●化合物MDI-1228溶液配制:称取适量MDI-1228,溶媒,待其溶解,使其终浓度为1.2mg/ml。混匀,4度放置备用,三天配制一次。
●化合物MDI-1233溶液配制:称取适量MDI-1233,溶媒,待其溶解,使其终浓度为0.4、1.2和4mg/ml。混匀,4度放置备用,三天配制一次。
模型建立
免疫:第0、7、14和21天,第一组腹腔注射200ul PBS,第2-8组腹腔注射200ul(10ug)OVA/铝佐剂乳剂。
致敏:第28-32天,第一组动物在雾化盒内PBS雾化处理20min,第2-8组动物在雾化盒内5%OVA雾化处理20min,然后取出。时间为每天中午12:30。
给药:每天给药两次,分别为早上9点和下午17点,滴鼻给药。地塞米松(Dex)灌胃给药,每天给药一次,早上9点。
评价指标
血清Ig E:动物经二氧化碳安乐死后眼眶采血,收集血浆,-40度保存,用于检测血浆Ig E。
肺泡盥洗液(BALF)细胞计数:称取适量胎牛血清溶于PBS,使其终浓度为1%。眼眶采血后,打开气管,1ml注射器针头换成小鼠灌胃针,注射器吸取0.8ml盥洗液,盥洗肺部,共三次。收集盥洗液,1000转/分离心10分钟,收集上清,-40度保存。细胞加1ml PBS重悬,细胞计数。取100ul离心涂片,Diff quick染色,记录嗜酸粒细胞、嗜碱粒细胞、淋巴细胞和巨噬细胞的数量。
肺盥洗液IL-5:收集肺盥洗液,检测IL-5。
肺组织病理:取肺组织甲醛固定,24小时后包埋切片,HE染色,观察炎症细胞浸润并评分。病理评分标准为:
A)0分:无淋巴细浸润;
B)1分:微量淋巴细胞浸润;
C)2分:少量淋巴细胞浸润;
D)3分:中度淋巴细胞浸润;
E)4分:重度淋巴细胞浸润;
评分结束,计算并分析。
二、实验结果
肺盥洗液IL-5
实验结果见图1。正常组动物肺盥洗液IL-5表达水平较低。和正常组比较,溶媒组动物肺盥洗液IL-5表达水平显著升高(p<0.001)。和模型组(即溶媒组)比较,阳性对照地塞米松能显著降低IL-5的表达(p<0.001);化合物MDI-1228和MDI-1288也能显著降低IL-5的表达(p<0.001);和阳性对照比较,化合物MDI-1228和MDI-1288降低IL-5表达能力较弱。和模型组比较,化合物MDI-1233三个剂量组均能降低IL-5的表达,3mpk和10mpk组有显著性差异(p<0.001);和化合物MDI-1228和MDI-1288比较,化合物MDI-1233 3mpk降低IL-5能力和化合物MDI-1228、MDI-1288基本一致;和阳性对照地塞米松比较,化合物MDI-1233 10mpk降低IL-5能力和阳性对照地塞米松基本一致。肺盥洗液IL-5检测数据表明MDI-1233各给药组能降低IL-5的表达水平,且有剂量效应。
肺盥洗液炎症细胞计数
实验结果见图2。正常组动物肺盥洗液炎症细胞数量较低。和正常组比较,溶媒组动物肺盥洗液炎症细胞数量显著升高(p<0.001)。和模型组(即溶媒组)比较,阳性对照地塞米松能显著降低炎症细胞数量(p<0.001);化合物MDI-1228和MDI-1288也能显著降低炎症细胞数量(p<0.001);和阳性对照比较,化合物MDI-1228和MDI-1288降低炎症细胞数量能力较弱。和模型组比较,化合物MDI-1233三个剂量组均能降低炎症细胞数量,3mpk和10mpk组有显著性差异(p<0.001);和化合物MDI-1228和MDI-1288比较,化合物MDI-1233 3mpk降低炎症细胞数量和化合物MDI-1228、MDI-1288基本一致;和阳性对照地塞米松比较,化合物MDI-1233 10mpk降低炎症细胞数量和阳性对照地塞米松基本一致。肺盥洗液炎症细胞数量检测数据表明MDI-1233各给药组能降低炎症细胞数量,且有剂量效应。
血清IgE
实验结果见图3。正常组动物血清IgE表达水平较低。和正常组比较,溶媒组动物血清IgE表达水平显著升高(p<0.001)。和模型组比较,阳性对照地塞米松能显著降低IgE的表达(p<0.001);化合物MDI-1228和MDI-1288也能显著降低IgE的表达(p<0.001);和阳性对照比较,化合物MDI-1228和MDI-1288降低IgE表达能力较弱。和模型组比较,化合物MDI-1233三个剂量组均能降低IgE的表达,3mpk和10mpk组有显著性差异(p<0.001);和化合物MDI-1228和MDI-1288比较,化合物MDI-1233 3mpk降低IgE能力和化合物MDI-1228、MDI-1288基本一致;和阳性对照地塞米松比较,化合物MDI-1233 10mpk降低IgE能力和阳性对照地塞米松基本一致。血清IgE检测数据表明MDI-1233各给药组能降低IgE的表达水平,且有剂量效应。
病理检测
实验结果见图4。正常组动物病理染色无异常。和正常组比较,溶媒组动物肺组织炎症细胞浸润显著升高(p<0.001)。和模型组比较,阳性对照地塞米松能显著降低肺组织炎症细胞浸润(p<0.001);化合物MDI-1228和MDI-1288也能显著降低肺组织炎症细胞浸润(p<0.001);和阳性对照比较,化合物MDI-1228和MDI-1288降低肺组织炎症细胞浸润能力较弱。和模型组比较,化合物MDI-1233三个剂量组均能降低肺组织炎症细胞浸润,3mpk和10mpk组有显著性差异(p<0.001);和化合物MDI-1228和MDI-1288比较,化合物MDI-1233 3mpk降低肺组织炎症细胞浸润和化合物MDI-1228、MDI-1288基本一致;和阳性对照地塞米松比较,化合物MDI-1233 10mpk降低肺组织炎症细胞浸润和阳性对照地塞米松基本一致。病理结果表明MDI-1233各给药组能降低肺组织炎症细胞浸润,且有剂量效应。
肺组织病理评分实验结果见图5。结果表明化合物MDI-1233对哮喘动物模型具有较好的治疗效果,且有剂量依赖效应;MDI-1228和MDI-1288比较也有一定的治疗效果。
实施例62:测试化合物对小鼠过敏性鼻炎的抑制效果
一、材料和方法
主要试剂
(1)0.2%Tween 80的配制:取TW80 1ml+499ml生理盐水
(2)0.05mg/ml卵清蛋白混悬液(致敏)的配制:
1)取OVA 10mg+10ml生理盐水
2)上述10mg/10ml的卵清蛋白液)2ml+38ml生理盐水摇匀后加入200mg氢氧化铝
(3)MDI-1233的配制:
1)1mg/kg MDI-1233:C=(1mg/kg×25g×10-3kg)÷(20×10-3ml)=1.25mg/ml,取10mg/kg MDI-1233的混悬液0.3ml+2.7ml0.2%TW80摇匀
2)3mg/kg MDI-1233:C=(3mg/kg×25g×10-3kg)÷(20×10-3ml)=3.75mg/ml,取10mg/kg MDI-1233的混悬液0.9ml+2.1ml0.2%TW80摇匀
3)10mg/kg MDI-1233:C=(10mg/kg×25g×10-3kg)÷(20×10-3ml)=12.5mg/ml,取56.25mg MDI-1233+4.5ml 0.2%TW80摇匀
(4)MDI-1228的配制:
C=(3mg/kg×25g×10-3kg)÷(20×10-3ml)=3.75mg/ml
称取MDI-1228 11.25mg+3.54ml生理盐水摇匀
(5)MDI-1288的配制:
C=(3mg/kg×25g×10-3kg)÷(20×10-3ml)=3.75mg/ml
称取MDI-1288 11.2mg+2.98ml生理盐水摇匀
(6)10%OVA的配制:取12mg OVA+1.2ml生理盐水摇匀
仪器设备
动物
42-49日龄BALB/C小鼠(厂家:辽宁长生生物科技股份有限公司,许可证号:SCXK(疗)2015-0001)雌性和雄性,饲养条件:常规饲料饲养,温度20~26℃,湿度40%~70%。动物分组和造模
1.空白对照组(不治疗)
2.过敏性鼻炎组(溶媒处理:含0.2%Tween 80的生理盐水)
3.过敏性鼻炎+化合物MDI-1233:1mg/kg
4.过敏性鼻炎+化合物MDI-1233:3mg/kg
5.过敏性鼻炎+化合物MDI-1233:10mg/kg
6.过敏性鼻炎+化合物MDI-1228:3mg/kg
7.过敏性鼻炎+化合物MDI-1288:3mg/kg
8.过敏性鼻炎+阳性对照组(丙酸氟替卡松,葛兰素史克,E88D)
制备小鼠AR模型是采用卵清蛋白(ovalbumin,OVA)致敏及激发BALB/c小鼠,模拟自然接种接触过敏源造成的急性症状。对实验动物称重和雌雄分开。在第1天,第5天,第9天,第12天进行致敏,第13天开始连续7天,每天进行一次激发,最后一次激发后观察小鼠行为并进行评分。致敏方法为腹腔注射50μg/ml的OVA(Sigma,A5503-1G)混悬液(10ml生理盐水溶解500μg卵清蛋白和50mg AL(OH)3(CAS 21645-51-2),pH 7.4),每只小鼠腹腔注射200μl。激发方法为配制pH 7.4的OVA(10mg/ml)生理盐水溶液后,每只小鼠每侧鼻孔用微量注射器滴入10μl,观察30分钟内小鼠挠鼻、打喷嚏、流鼻涕的次数。第12天使用10%OVA滴鼻激发1次(对照组使用PBS),按照下表进行评分,采用叠加法计算总分,总分大于5分为造模成功,判断造模成功后进行后续实验。
给药方法
于造模第13天起进行化合物给药和激发。化合物给药方法为根据分组要求按照小鼠体重换算化合物滴鼻量,用含0.2%Tween 80的生理盐水溶解化合物MDI-1233、MDI-1228、MDI-1288到特定浓度。在激发前30min给药,每天2次给药。阳性对照治疗为于OVA滴鼻刺激前30min滴鼻1次(10ul/侧),每天2次治疗。在药物第二次给药30min后使用10%OVA激发,于每次激发后观察并记录30min内小鼠鼻涕量、喷嚏及挠鼻的次数并打分。对照组用生理盐水滴鼻。
样品收集
鼻炎造模完成后评分稳定后,每组在第20天评分结束后行麻醉处死。采集样本,取外周血清(采集全血后常温下静置1h后离心取上层清液)进行ELISA检测;取鼻腔粘膜进行HE染色(用10%的福尔马林固定);鼻腔冲洗后取上层清液做ELISA检测。
病理检测
(1)HE染色:取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ15min、Ⅱ15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60min。石蜡包埋,切片。将石蜡切片进行展片、烤片,然后进行脱蜡、水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
(2)ELISA检测:试剂盒采用双抗体一步夹心法酶联免疫吸附试验(ELISA)。往预先包被小鼠待检指标捕获抗体的包被微孔中,依次加入标本、标准品、HRP标记的检测抗体,经过温育并彻底洗涤。用底物TMB显色,TMB在过氧化物酶的催化下转化成蓝色,并在酸的作用下转化成最终的黄色。颜色的深浅和样品中的待检物呈正相关。用酶标仪在450nm波长下测定吸光度(OD450值),计算样品浓度。
二、实验结果
动物造模结果
动物造模分为预实验和正式实验。其中预实验以OVA滴鼻激发后的行为学观察打分计算阳性率)和病理切片HE染色为判断鼻炎模型造模是否成功。预实验总共用10只小鼠,在第15天滴鼻激发后检测阳性率仅为30%。第19天和第20天的阳性率分别为90%和80%。
同时,为了验证造模的可靠性,将打分为阳性的小鼠鼻黏膜进行了病理切片和HE染色。结果表明空白对照组鼻黏膜纤毛较密集,而模型组中鼻黏膜上的粘膜纤毛已经消失,炎症明显。
药物干预鼻炎模型后结果
在正式实验中,共分为8组进行。除空白组外,其余各组均进行鼻炎造模。其中化合物MDI-1233的剂量分别是1mg/kg、3mg/kg、10mg/kg,化合物MDI-1228剂量为3mg/kg,MDI-1288剂量为3mg/kg,阳性对照药物为丙酸氟替卡松。经过7天的治疗期,分别在计划时间内对动物的行为学进行打分并统计(图6、7)。在雄鼠中使用化合物干预后,鼻炎打分的阳性率呈下降趋势。鼻炎阳性率较模型组有显著降低(p<0.05)。在雌鼠中使用化合物干预后,鼻炎阳性率也呈现显著性降低(p<0.05)。统计分析表明化合物干预组和阳性对照药物组对抑制其实验动物的鼻炎行为均有显著性效果。综合结果表明MDI-1233在给药剂量为10mg/Kg时,通过分析行为学打分证明在雄鼠鼻炎模型上具有抑制作用;化合物MDI-1228不能显著抑制雄性小鼠的过敏性反应;MDI-1288在给药剂量为3mg/Kg时,通过分析行为学打分证明在雄和雌鼠鼻炎模型上均具有抑制作用。
病理切片分析
通过化合物干预后,各组均进行了取样和切片。经过HE染色后,通过随机视野观察和记录来分析鼻黏膜病理变化情况,结果见图8。结果表明空白组雄和雌鼻黏膜结构整齐,纤毛密集,杯状细胞明显,无明显炎症细胞浸润(图8A、8H);鼻炎组纤毛消失,鼻黏膜结构紊乱,黏膜下组织伴有水肿、出血症状,有大量炎症细胞浸润(图8B、8I);化合物MDI-1288干预组,鼻黏膜结构基本正常,纤毛结构明显,仍有部分样本伴有鼻黏膜下出血及纤毛脱落,杯状细胞明显,炎症细胞减少(图8C、8J); 化合物MDI-1233治疗药物浓度1mg/kg和3mg/kg组,鼻黏膜下仍伴有出血,炎症细胞浸润,鼻黏膜纤毛脱落,杯状细胞不明显,水肿症状有所改善(图8D、8E、8K和8L);而在化合物10mg/kg治疗组,鼻黏膜结构基本清晰,部分纤毛成团簇状或倒毛状,仍有部分纤毛脱落,水肿症状明显改善,杯状细胞有所呈现,伴有炎症细胞浸润(图8F、8M);阳性对照药物组鼻黏膜下伴有少量出血,水肿症状不明显,纤毛成团簇状,少量脱落,杯状细胞明显,仍有炎症细胞浸润(图8G、8N)。从切片结果整体判断,雄鼠和雌鼠鼻炎组造模结果达到病理指标,主要体现在鼻黏膜纤毛脱落和炎性细胞浸润。经过化合物干预后,鼻黏膜的病理症状逐渐出现改善。鼻黏膜结构基本正常,纤毛结构明显,仍有部分样本伴有鼻黏膜下出血及纤毛脱落,杯状细胞明显,炎症细胞减少。阳性对照药物丙酸氟替卡松也同样能抑制鼻炎进程,促进鼻黏膜上的纤毛修复。从病理切片特征判断,MDI-1233以10mg/kg的剂量预处理组的鼻黏膜纤毛修复效果较显著,纤毛较清晰,但仍有炎性细胞浸润。然而以MDI-1288干预鼻炎模型后,其治疗效果和阳性对照药物结果相近。阳性对照药物丙酸氟替卡松也同样能抑制鼻炎进程,促进鼻黏膜上的纤毛修复。
ELISA检测
通过病理切片检测各组实验动物的鼻黏膜和纤毛损伤修复情况的同时,也通过ELISA对受试动物的血清和鼻腔灌洗液进行了检测。主要检测指标分别为炎性因子IL-4和IL-17、Ⅱ型干扰素IFN-γ和免疫球蛋白IgE。在血清中,IL-4表达量的变化在雄和雌两组中呈现较一致的趋势(图9、10)。当鼻炎造模组中,检测到IL-4的血清中含量较空白对照组升高约2倍。通过阳性对照药物的治疗后,其含量回落到和空白组相近。在待测药物组中,IL-4血清中含量出现了显著下降的趋势,呈现出药物剂量相关性。
实施例63:测试化合物对大鼠慢性阻塞性肺疾病动物模型的治疗效果
一、材料和方法
实验动物
健康6-8周龄SD大鼠(厂家:湖南斯莱克景达有限公司,许可证号:SCXK(湘)2019-0004),雌雄各半,饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)。
动物分组和造模
(1)空白对照组(7只)
(2)COPD组+溶媒组(溶媒:0.2%Tween 80/saline buffer)(8只)
(3)COPD+阳性对照药物(噻托溴铵)组(8只)
(4)COPD+化合物MDI-1233(1mg/kg)剂量组(8只)
(5)COPD+化合物MDI-1233(3mg/kg)剂量组(8只)
(6)COPD+化合物MDI-1233(10mg/kg)剂量组(8只)
实验环境:温度20℃-26℃,湿度40%-70%
大鼠适应性饲养7天,制作慢性阻塞性肺疾病模型,并使用不同药物进行治疗。
造模:第1、14天将大鼠以10%水合氯醛腹腔注射麻醉后,通过气管插管向大鼠气管内注入LPS 200μg/200μL,完毕后将大鼠直立旋转10~20s,使LPS均匀分布于肺部。第2~13天、15~28天每天在自制有机玻璃密闭熏烟箱(80cm×60cm×50cm),持续吸入香烟烟雾30min,15支/天。
动物给药
各组均从烟熏后第29天起开始滴鼻给药,各组连续给药,2次/d,共30d。
方法:空白对照组(不治疗)不做任何处理;COPD组(生理盐水)组造模后生理盐水滴鼻;化合物MDI-1233组造模后给予对应剂量的化合物;阳性对照药物组给予0.1mg/kg/d剂量滴鼻,1天2次。
期间,每周称量1次大鼠体重,观察大鼠饮食、咳嗽、活动性等一般情况。
动物取样
治疗结束后,10%水合氯醛腹腔注射麻醉后,切开胸部,暴露气管及肺部,从心脏取血约5mL,3500r/min,离心10min,取血清-20℃保存。结扎右主支气管,于隆突上用套管针穿刺至左肺,以2mL生理盐水灌洗左肺,再缓慢回抽,每次回收液体约1.5mL,如此反复灌洗3次。混匀灌洗液,4℃,1000r/min,离心10min,取上清液-20℃保存。右肺中叶浸泡于4%多聚甲醛液中固定72h,常规石蜡包埋。HE染色观察各组大鼠肺部病理情况。
HE染色
切取动物肺部组织,固定于10%中性福尔马林缓冲溶液中,脱水,石蜡包埋,组织切片HE染色观察组织病理形态变化。HE染色所需药瓶和使用仪器有:苏木素染液(AR11800-1,BOSTER);伊红染色液(AR11800-2,BOSTER);超净高级封片胶(YZB,BASO);Scott蓝化液(G1865,Solarbio)等。药品冷藏柜(BYC-310,山东博科生物);显微镜(CX41OLYMPUS);切片机(BQ-318D,伯纳);电热鼓风干燥箱(DHG-9070A,恒科学仪器有限公司)。取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ15min、Ⅱ15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60分钟。石蜡包埋,切片。将石蜡切片进行烤片,然后脱蜡,水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
ELISA检测
ELISA检测血清和肺泡灌洗液中炎症因子TNF-α、IL-1β、IL-6、IL-8的含量。
主要仪器设备
二、实验结果
大鼠慢性阻塞性肺炎造模
通过在第1、14天通过气管插管向大鼠气管内注入LPS,第2~13天、15~28天每天在自制有机玻璃密闭熏烟箱,持续吸入香烟烟雾30min进行慢性阻塞性肺疾病造模,通过肺部组织染色可见模型组支气管壁有大量炎性细胞附着,气管黏膜上皮坏死、脱落、杂乱,杯状细胞及腺体显著增生,表明模型成功。
HE染色
30天给药后,光学显微镜下观察各组大鼠肺组织形态及炎症细胞浸润情况(如图11所示)。正常对照组大鼠支气管管壁未见增厚,未见明显炎性细胞浸润,肺泡结构完整,纤毛未见脱落;COPD模型组大鼠支气管壁及血管周围均有大量及多种炎性细胞浸润,上皮细胞大量脱落,纤毛倒伏、气管壁断裂、增厚明显,管腔狭窄,肺泡断裂融合;阳性对照药物组大鼠气管腔可见局部轻度变形,管壁轻度增厚,上皮细胞少量脱落,部分纤毛倒伏,脱落不明显,部分肺泡融合,可见炎性细胞浸润明显减少;MDI-1233药物给药组大鼠气管腔可见变形及炎症细胞浸润相对COPD模型组减轻,随着药物浓度增加,管腔可见变形减轻,炎症细胞逐渐减少,其中MDI-1233药物高剂量组效果最好,但是相较于阳性对照药物组稍差。
ELISA检测
基于机体发生COPD时的氧化应激反应及炎症反应等致病机制,实验主要选取COPD病变时炎症趋化因子等指标来评价MDI-1233药物对模型小鼠COPD时肺部炎症水平和氧化应激程度的作用。COPD是一种慢性炎症性疾病,炎症的刺激使巨噬细胞、中性粒细胞等激活,使IL-1β的分泌增加。由图12可以看出,与空白对照组相比,模型组经溶媒处理后,其血清和肺泡灌洗液中IL-1β含量仍增高。阳性对照药物处理后能下调其表达量。不同剂量的MDI-1233也能下调其表达量,且和药物浓度成一定的剂量依赖关系(图12),与染色观察结果一致。
三、结论
以上结果表明MDI-1233对大鼠COPD模型具有剂量依赖型的治疗效果。
实施例64:测试化合物对大鼠外痔的缓解和治疗效果
一、材料和方法
实验动物
健康雄性6周龄SD大鼠(厂家:湖南斯莱克景达有限公司,许可证号:SCXK(湘)2019-0004),雄性,饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)。
动物分组和造模
(1)空白对照组(7只)
(2)痔疮模型组(10只)
(3)痔疮模型+阳性对照药物(马应龙痔疮膏)组(8只)
(4)痔疮模型组+溶媒处理(凝胶赋形剂)(8只)
(5)痔疮模型+0.1%MDI-1228凝胶制剂组(8只)
(6)痔疮模型+0.3%MDI-1228凝胶制剂组(8只)
(7)痔疮模型+1%MDI-1228凝胶制剂组(8只)
(8)痔疮模型+3%MDI-1228凝胶制剂组(8只)
实验环境:温度20℃-26℃,湿度40%-70%
除正常对照外,所有组均采用首先消毒肛周皮肤,配制75%乙酸溶液并于各大鼠肛门外周皮下散点注射0.05ml。24小时后观察,可见白色溃疡面、肛周肿胀并有炎性渗出,表明造模成功。
MDI-1228凝胶的制备
1%MDI-1228凝胶配方和制备方法如下:
步骤一:称取处方量水,加入10%量的丙二醇,搅拌均匀(搅拌5~15分钟)。
步骤二,称取MDI-1228甲磺酸盐,剩余量丙二醇至步骤一得到溶液中,搅拌均匀(搅拌30~60分钟)。
步骤三,将卡波姆加入步骤二溶液中,搅拌12~18小时。充分溶胀。
步骤四,加入三乙醇胺(约处方量1.4%后需要细微调节),边加边搅拌,调节pH至6.5左右。
0.5%以及3%MDI-1228凝胶的配方根据MDI-1228的量进行相应比例的调整,并使用相同工艺制得。
动物给药
对照组和模型组不做任何处理(不治疗),诱发模型成功24h后开始给药,第3)组涂抹马应龙麝香痔疮膏(7.5g/Kg),分3次涂抹,10min/次;第4)组在肛周涂抹空白凝胶制剂(1g/Kg);第5)-8)组在肛周涂抹不同浓度MDI-1228化合物凝胶制剂(1g/Kg),每日涂抹2次,10min/次再以保鲜膜覆盖、胶布固定,防止因外敷药物脱落而影响药效,6h后清除敷药,生理盐水清洗用药局部,连续11天。
肛周溃疡评分
于给药第3,5,7,9,11天观察溃疡局部愈合情况,评定溃疡积分(评定标准:有溃疡渗液1分;少许溃疡渗液2分;有焦痂,基本愈合3分;完全愈合4分)。
动物取样
于给药第11天将动物通过吸入过量二氧化碳处死后,取肛周直肠做HE染色,取血清做ELISA检测。
HE染色
切取动物直肠组织,固定于10%中性福尔马林缓冲溶液中,脱水,石蜡包埋,组织切片HE染色观察直肠组织病理形态变化。HE染色所需药瓶和使用仪器有:苏木素染液(AR11800-1,BOSTER);伊红染色液(AR11800-2,BOSTER);超净高级封片胶(YZB,BASO);Scott蓝化液(G1865,Solarbio)等。药品冷藏柜(BYC-310,山东博科生物);显微镜(CX41 OLYMPUS);切片机(BQ-318D,伯纳);电热鼓风干燥箱(DHG-9070A,恒科学仪器有限公司)。取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ 15min、Ⅱ 15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60分钟。石蜡包埋,切片。将石蜡切片进行烤片,然后脱蜡,水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
ELISA检测
试剂盒采用双抗体一步夹心法酶联免疫吸附试验(ELISA)。往预先包被大鼠待检指标捕获抗体的包被微孔中,依次加入标本、标准品、HRP标记的检测抗体,经过温育并彻底洗涤。用底物TMB显色,TMB在过氧化物酶的催化下转化成蓝色,并在酸的作用下转化成最终的黄色。颜色的深浅和样品中的待检物呈正相关。用酶标仪在450nm波长下测定吸光度(OD450值),计算样品浓度。
组织病理染色观察
对大鼠直肠组织病理变化进行分级,“-”直肠黏膜腺上皮排列整齐未见其他病理改变,记0分;“+”直肠黏膜腺上皮排列整齐,偶见炎性细胞浸润,记1分;“++”直肠黏膜上皮增生、排列紊乱,黏膜下见大量炎症细胞浸润,记3分;“+++”直肠黏膜缺损、坏死,表面附着炎性渗出,炎细胞浸润,大量中性粒细胞浸润,结缔组织增生,记5分。
二、实验结果
大鼠外痔造模和阳性对照药物效果的确认
利用乙酸注射的方法,成功建立了大鼠外痔模型,可见肛周有溃疡出现,严重的有液体渗出。利用阳性对照药物马应龙痔疮膏治疗之后通过病理切片检测,可以看到外痔造模后直肠溃疡部分出现组织结构破坏、有凝血块并伴发炎症反应,阳性对照药物治疗后溃疡部位修复,炎症减弱(图13)。证明外痔造模和阳性对照药物治疗的操作均正确。待测药物的治疗 效果
大鼠外痔造模成功后,利用凝胶赋形剂和含有不同浓度MDI-1228的凝胶进行治疗并观察其愈合情况做打分评价。结果表明空白组无自发性外痔,为满分。模型组从第三天开始的愈合速度低于其余有治疗干预组。阳性对照药物治疗效果从第七天后与模型组相比即有显著性差异。表明外痔造模和阳性对照药物治疗测试体系正常。用于制备MDI-1228的空白凝胶赋形剂在第九天后与模型组对比,表现出对溃疡面有促进愈合的作用,但其效果显著低于含各剂量MDI-1228的凝胶治疗组。在各剂量MDI-1228的凝胶治疗组中,1%浓度组在第7天即表现出显著治疗效果。3%浓度组在第9天和第11天治疗效果接近。
HE染色
动物造模并给药后,取动物肛周直肠组织做病理切片和HE染色,对各组样品的病理特征进行打分。结果表明,模型组的病理特征显著,而阳性对照药物治疗后有显著缓解。凝胶赋形剂对促进外痔溃疡愈合有一定效果。而0.3%以上浓度的MDI-1228治疗效果更显著。其中以3%浓度的MDI-1228效果最好(图14)(*:与对照组比较,p<0.05;#:与赋形剂组比较,p<0.05)。
ELISA检测
除了对病理样品的分析,还对各组动物的外周血中的炎症相关因子指标IL-1β和IL-6,肿瘤坏死因子TNF-α和血液中NO含量做了检测。结果表明外痔造模后,大鼠血液中TNF-α表达量上调,阳性对照药物治疗后其表达量显著下降。外痔组经过赋形剂涂抹后,TNF-α表达量与模型组没有显著下调。而不同含量的MDI-1228涂抹均能下调TNF-α表达量,其下调量与药物剂量成线性关系(见图15,*:与对照组比较,p<0.05;#:与赋形剂比较,p<0.05)。
三、结论
综合以上实验结果可知:不同浓度的MDI-1228凝胶制剂能有效缓解和治疗大鼠外痔。
实施例65:化合物MDI-1228对苯酚诱导的大鼠宫颈炎模型的治疗效
一、材料和方法
实验动物
6-8周的SD大鼠(180-220g)(厂家:湖南斯莱克景达有限公司,许可证号:SCXK(湘)2019-0004))雌性,饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)。
主要试剂的配制
25%苯酚胶浆:取5ml 50摄氏度液态苯酚+15ml蒸馏水+8g阿拉伯胶在水浴中充分搅拌
溶媒的配制(20%DMA+65%PEG400+15%H2O):取10ml DMA+32.5ml PEG400+7.5ml H2O
MDI1228(4.5mg/ml):第1次,称取MDI-1228 57.7mg+6.1ml DMA溶解,加19.7ml PEG400,再加4.5ml水,得无色澄清溶液;第2次,称取MDI-1228 50.9mg+2.3mlDMA溶解,加7.4ml PEG400,再加1.7ml水,得无色澄清溶液
MDI-1228(15mg/ml):第1次,称取MDI-1228 186.5mg+2.5ml DMA溶解,加8.1ml PEG400,再加1.8ml水,得无色澄清溶液;第2次,称取MDI-1228 157.2mg+2.1ml DMA溶解,加6.8ml PEG400,再加1.6ml水,得无色澄清溶液
30mg/kg,45mg/ml DMI-1228的配制:0.67ml/kg,第1次,称取MDI-1228 362.4mg+1.6ml DMA溶解,加5.2ml PEG400,再加1.2ml水,得淡黄色澄清溶液。第2次,称取MDI-1228 368.0mg+1.6ml DMA溶解,加5.3ml PEG400,再加1.2ml水,得淡黄色澄清溶液
阳性对照药物洁尔阴的配制:(恩威药业,国药准字:Z10930008)(1:5稀释)0.67ml/kg。第1次,取4.0ml洁尔阴+16.0ml水,第2次,取4.0ml洁尔阴+16.0ml水
动物分组和造模
空白对照组(不治疗)(N=5+2只)
宫颈炎模型组(溶媒处理:20%DMA+65%PEG400+15%H2O)(N=8+2只)
宫颈炎模型+阳性对照药物组(洁尔阴洗液(1:5稀释),2次/d,连续7d)(N=8只)
宫颈炎模型+低剂量MDI-1228化合物洗剂组(剂量:3mg/kg/次,2次/d,连续7d)(N=8只)
宫颈炎模型+中剂量MDI-1228化合物洗剂组(剂量:10mg/kg/次,2次/d,连续7d)(N=8只)
宫颈炎模型+高剂量MDI-1228化合物洗剂组(剂量:30mg/kg/次,2次/d,连续7d)(N=8只)
实验环境:温度20℃-26℃,湿度40%-70%。
造模:
SD大鼠适应性饲养一周,异氟烷麻醉后,将大鼠四肢固定于操作台上,保持仰卧位,1mL注射器钝圆针头置入大鼠阴道内约1cm,缓慢注入0.1mL 25%苯酚胶浆,立即用无菌棉球堵塞阴道口,然后使大鼠保持头低尾高体位20min。每2d注射1次,连续3次。观察大鼠阴道口,出现白色分泌物且阴道充血、红肿为造模成功。
动物给药
造模成功的大鼠随机分为6组,各组动物按照分组用药,空白组(不治疗)不做任何处理,阳性对照组给予洁尔阴洗液(1:5稀释),每天灌洗阴道2次,连续给药7天;给予低、中、高剂量的MDI-1228化合物洗剂灌洗阴道2次,连续给药7天;模型组给予等量溶媒处理,处理方式和时间同供试药物组。
检测指标
临床症状观察:连续观察大鼠的一般情况(活动、饮食、精神状态)以及用药前后称大鼠体重,每天观察并记录大鼠外阴局部及分泌物变化, 大鼠阴道口是否干燥,外阴及阴道口粘膜有无充血、红肿,阴道有无流液血,并按照下表评分。
脏器重量及其脏器指数计算:末次给药治疗后,次日立即处死大鼠,取出子宫,剔除脂肪组织及系膜等,称量宫颈(子宫阴道部开口至双子宫分叉处)及肝脏、脾脏的重量,计算脏器指数。
病理检测:ELISA检测,血清4个指标IL-4、IL-6、IL-1β、TNF-α;ELISA检测血浆CP;生化检测宫颈组织的MDA、T-SOD。
组织病理学检测方法
HE染色:切取动物阴道、宫颈、子宫组织,4%多聚甲醛固定,脱水,石蜡包埋,组织切片HE染色观察组织病理形态变化。HE染色所需药瓶和使用仪器有:苏木素染液(AR11800-1,BOSTER);伊红染色液(AR11800-2,BOSTER);超净高级封片胶(YZB,BASO);Scott蓝化液(G1865,Solarbio)等。药品冷藏柜(BYC-310,山东博科生物);显微镜(CX41OLYMPUS);切片机(BQ-318D,伯纳);电热鼓风干燥箱(DHG-9070A,恒科学仪器有限公司)。取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ15min、Ⅱ15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60分钟。石蜡包埋,切片。将石蜡切片进行烤片,然后脱蜡,水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s, 流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
ELISA检测:所使用的试剂盒信息如下:大鼠肿瘤坏死因子α(TNF-α)试剂盒(MM-0180R1,酶免),大鼠白细胞介素β(IL-1β)试剂盒(MM-0047R1,酶免),大鼠白细胞介素4(IL-4)试剂盒(MM-0191R1,酶免),大鼠白细胞介素6(IL-6)试剂盒(MM-0190R1,酶免),大鼠铜蓝蛋白(CP)试剂盒(MM-21238R1,酶免)。
生化检测:所使用的试剂盒信息如下:MDA检测试剂盒(MDA,E-BC-K025-M,Elabscience),总超氧化物歧化酶检测试剂盒(T-SOD,E-BC-K019-M,Elabscience)。
主要仪器设备
二、实验结果
大鼠一般症状的观察
制备宫颈炎模型过程中,大鼠表现出易怒和竖毛,外阴口可见明显的红斑和水肿,并伴有大量黄色或白色甚至血丝的黏性分泌物。经药物治疗后,各给药组大鼠上述症状均比模型组得到不同程度的改善,其中MDI-1228(30mg/kg)治疗能显著地降低宫颈炎综合分值。通过对连续打分曲线的面积计算也表明,MDI-1228(30mg/kg)能显著降低外阴炎症、红斑、水肿和分泌物的评分幅度分别为23.42%、29.99%、19.61%和21.05%(图16-图19)。
药物对脏器指数的影响
苯酚刺激造模后,宫颈炎模型动物的宫颈指数均值略升高,经过药物干预后,MDI-1228(30mg/kg)组中的宫颈指数出现明显的下调(图20)。
HE染色结果
观测结果见图21。模型(溶媒)组在阴道、宫颈、子宫三个组织部位均有大量炎症细胞浸润,且阴道粘膜有不同程度的变薄或增厚的现象,个别样本出现粘膜层坏死脱落,宫颈可见血管内充血和间质出血,纤维增生,子宫内膜脱落,结构紊乱;阳性对照药物组可见炎症细胞明显减少,阴道粘膜层及子宫内膜结构趋于完整,个别样本宫颈仍可见明显炎症细胞浸润和少量纤维化现象;MDI-1228药物治疗组中,与模型(溶媒)组相比较,不同药物浓度均有一定的治疗效果,但其中以MDI-1228(30mg/kg)组治疗效果最佳,相较而言,MDI-1228(3mg/kg)组整个组织结构炎症细胞浸润仍明显,但各部位组织结构有一定程度上的好转,MDI-1228(10mg/kg)组和MDI-1228(30mg/kg)组治疗效果相差并无太大差别,两组中炎症细胞明显减少,但从组织的整体结构上观察,MDI-1228(10mg/kg)组有个别样本阴道和宫颈仍存在粘膜结构损坏和纤维化的现象,但MDI-1228(30mg/kg)高剂量组整体恢复症状都较好。
三、结论
MDI-1228各剂量组可以改善宫颈炎大鼠红斑、水肿、分泌物状况,外观炎症评分降低,可以降低炎症因子IL-1β、IL-4、IL-6、TNF-α含量,起到治疗宫颈炎的作用。
实施例66:化合物凝胶对小鼠特应性皮炎/皮肤瘙痒症的治疗效果
一、材料和方法
实验动物
健康的C57BL/6小鼠,6-8周龄,重20g左右(厂家:湖南斯莱克景达有限公司,许可证号:SCXK(湘)2019-0004),雌雄各半,饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)。
主要试剂配制
1)尘螨的配制(Derf,Lot:361863,GREER):取一瓶尘螨(含蛋白4.51mg),加入4.51ml的PBS混匀至尘螨完全溶解,配置成1mg/ml的母液,每次用PBS稀释成100μg/ml的尘螨蛋白液。
2)SEB的配制(Lot:92815B,Toxin Technology):取一瓶SEB,加入10ml的PBS,配置成100μg/ml的母液,每次用PBS稀释成10μg/ml的SEB。
3)托法替尼配制:取10mg的托法替尼(CAS#477600-75-2,lot#79614,MCE),加入40μl的二甲基乙酰胺促溶(CAS#127-19-5,源叶生物),再加入200μl的生理盐水稀释至41.67mg/ml。
4)空白凝胶,0.5%,1%以及3%MDI-1228凝胶根据实施例64中描述的工艺路线制得。
动物分组和造模
空白对照组(不做处理)(N=5+3)
空白皮炎模型组(不做处理)(N=8+3)
皮炎模型+空白赋形剂组(溶媒处理:空白凝胶制剂,剂量:1g/kg,2次/天/只)(N=8)
皮炎模型+阳性对照药物组(托法替尼,10mg/kg)(N=8)
皮炎模型+MDI-1228(0.5%)(剂量:0.7g/kg/只凝胶,2次/天/只)(N=8)
皮炎模型+MDI-1228(1%)(剂量:1g/kg/只凝胶,2次/天/只)(N=8)
皮炎模型+MDI-1228(3%)(剂量:1g/kg/只凝胶,2次/天/只)(N=8)
实验环境:温度20℃-26℃,湿度40%-70%。
造模:
小鼠适应性喂养7天,异氟烷麻醉后,背部脱毛,形成3cm2左右脱毛区域,并在除毛后的皮肤上轻刮造成皮损。生理盐水小鼠清洗背部,用消毒棉签轻擦干背部。将无菌化妆棉剪为1.5×1.5cm大小的方块,将100μL Derf及50μL SEB滴于化妆棉上,将化妆棉使用胶布固定于小鼠脱毛区域,并使用胶布将其缠在小鼠腰腹位置。第一次过敏源刺激后第4天,将敷药装置解除,休息1-2天后再重复给药一次(若皮损不严重或毛发再生,则在给药前再用脱毛膏进行一次皮损)。前两次给药为第一轮刺激。第9天后再进行第二轮给药(共两次),步骤同前。一共利用过敏原刺激四轮(共8次)。造模第40天,皮损评分大于等于5分的为造模成功小鼠。
皮损评分及搔抓实验:对小鼠背部造模区域的四个参数:发红,出血,喷发和脱屑进行评价,评级按以下严重性等级:0-无症状,1-轻度,2-中级,3-严重。造模第40天时,对小鼠背部特定部位(自拟定并标记)进行derf和SEB混合溶液的喷施,喷施剂量与造模剂量同,并进行30-60min行为学观察,主要观察指标为小鼠在造模及喷施部位的抓挠次数,判定标准为小鼠抓挠身上除头面外其他部位为有效,以小鼠开始抓挠到抓挠用爪重新触地或舔舐抓挠用爪计为一次,最后合并统计每次抓挠的总时长。同时还记录分析小鼠从过敏源刺激开始到开始抓挠的时间间隔,做为抓挠潜伏期指标。
模型验证:造模第40天,评分大于等于5分的为造模成功小鼠,取正常组和模型组小鼠各3只,行HE染色及甲苯胺蓝染色进行病理特征判断验证模型制备成功性,3只正常组小鼠血液用于实验2的ELISA检测。
动物给药
具体操作如下:造模第40天(治疗第0天)按照以上分组开始对小鼠进行涂抹治疗14天,每天涂抹2次(9点和17点,间隔6h以上)。在第1、4、7、14天对小鼠进行皮肤损伤程度评分(方法同上);并在17点前进行搔抓实验并进行行为学变化记录(方法同上)。
注:为了防止动物之间有舔舐药物的现象。动物涂完凝胶后,单独放置在无垫料的独立笼内活动一段时间,让凝胶自然风干1小时,然后再放入饲养笼正常喂养。禁单独喂养,容易出现抑郁;禁用纱布包裹涂抹部位,会吸附走药剂。
检测指标
小鼠在治疗第14天,行为学变化记录结束后,立即心脏采血并取皮肤组织:
ELISA检测炎症指标:取血清,ELISA检测IL-4、IL-13、TNF-α、IFN-γ的含量。检测方法严格按照ELISA检测试剂盒说明书进行。
病理检测:取皮肤组织进行HE染色和甲苯胺蓝染色,观察表皮厚度和肥大细胞数量、毛细血管通透性、水肿以及炎性成分渗出等差异。取皮肤组织,免疫组化检测TSLP表达情况。
组织病理学检测方法
HE染色:HE染色所使用的试剂和仪器有苏木素染液(AR11800-1,BOSTER)、伊红染色液(AR11800-2,BOSTER)、超净高级封片胶(YZB,BASO)和Scott蓝化液(G1865,Solarbio)等。设备主要有药品冷藏柜(BYC-310,山东博科生物)、显微镜(CX41OLYMPUS)、切片机(BQ-318D,伯纳)和电热鼓风干燥箱(DHG-9070A,恒科学仪器有限公司)。实验流程主要为先切取动物皮肤组织,经4%多聚甲醛固定、脱水、石蜡包埋和切片。最后组织切片经HE染色利用显微观察皮肤组织病理形态变化。具体操作步骤为先将样品置于4%多聚甲醛常温固定4小时,取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水后再用纯酒精和二甲苯等量混合的溶液处理15min,再经二甲苯溶液透化两次,每次各15min,至样品透明为止。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各60min。石蜡包埋后,将样品按照预先选择的切面方向进行切片。将石蜡切片进行烤片、脱蜡和水化。将水化后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
主要仪器设备
二、实验结果
药物对小鼠皮肤发红,出血,喷发和脱屑情况(皮损评分)的影响
在整个实验过程中,模型组小鼠皮肤出现红肿、出血、皮疹和鳞屑。模型组皮损评分明显高于正常组,说明造模成功(表1)。造模成功后,未经任何干预的模型组皮损评分在第0、4、7、14天均呈逐渐下降趋势,表明小鼠皮肤具有一定的自愈能力。空白凝胶组的第14天评分低于模型组,说明空白凝胶干预后对皮肤的修复也有一定的促进作用。但从临床评分来看,阳性对照药物的治疗效果明显优于空白凝胶。此外,含有不同浓度MDI-1228的凝胶也显示出与阳性对照药物相似的治疗效果。从皮炎评分的AUC看,与模型组比较,各组的AUC都有下降,其中MDI-1228(3%)剂量组降低最明显(23.75%)(表1、图22和图23)。
表1.药物对小鼠皮肤发红,出血,喷发和脱屑情况(皮损评分)的影响

药物对小鼠挠抓次数和挠抓时间的影响
同时,还对小鼠在治疗过程中的行为学做了记录和分析。分别从抓挠次数和总抓挠时长两个参数做效果评价。在挠抓次数上,对比第1天和第14天的数据,MDI-1228高剂量组基本没有变化,其余各组数据均有增高,这也表明受试动物对皮肤刺激表现出主动应激反应。第14天的时候,MDI-1228高剂量组的抓挠次数也低于模型组和其他治疗组。在抓挠总时长上,除空白凝胶组外,第14天的抓挠时间总体上比第1天的降低,MDI-1228高剂量药物组挠抓时长减少幅度更大。这些数据显示MDI-1228高剂量组在行为学分析上表现出有一定治疗效果,阳性对照药物托法替尼组在行为学分析上的效果不显著(表2)。
表2.药物对小鼠挠抓次数和挠抓时间的影响
HE染色和甲苯胺蓝染
如图24、25所示,与正常组相比,模型组有不同程度的角化过度或角化不全现象,真皮层出现增厚,结构紊乱,有大量炎症细胞浸润,毛囊受损及数量减少,肥大细胞明显增多(甲苯胺蓝染色)。空白赋形剂组和阳性对照药物组对上述病理现象均有有轻微治疗效果,较模型组表皮层损伤相对减轻,肥大细胞个别样本减少,但炎症细胞浸润仍明显。0.5%MDI-1228药物组表皮结构较模型组平整,仍存在角质层增厚的现象,伴有炎症细胞浸润,但肥大细胞和炎症细胞较模型组减少。1%MDI-1228药物组和3%MDI-1228药物组治疗效果相对明显,炎症细胞和肥大细胞较模型组明显减少,组织结构完整清晰,表皮未见明显损伤,但两组之间无明显区别。
药物对小鼠血清中IL-4、IL-13、TNF-α、IFN-γ含量的影响
特异性皮炎是一个由Th1/Th2免疫反应引发的双相性炎性皮肤病,Th1/Th2细胞因子之间存在拮抗关系,IFN-γ主要是Th1分泌的细胞因子,IL-4及IL-13主要是Th2分泌的细胞因子,Th1/Th2失衡可以导致过敏性疾病,Th1/Th2平衡状态对于保持机体正常的免疫有重要意义。结果(图26-28)显示,与正常组比较,模型组IL-4及IL-13均升高,IFN-γ降低。与模型组比较,MDI-1228药物组IL-4,IL-13表达水平明显降低,IFN-γ表达水平明显升高,提示MDI-1228可以降低机体的过敏反应,调节Th1、Th2细胞系统平衡。
当机体受到过敏原刺激后可分泌大量的IL-1β,导致TNF-α的分泌增加,参与AD的发生、发展。TNF-α是一种多向性的促炎因子,促进炎性反应的发生。结果(图29)显示,与正常组比较,各组TNF-α均升高,且模型组升高最多,与模型组比较,MDI-1228高、中、低剂量药物组均可降低TNF-α值。
三、结论
MDI-1228凝胶制剂在治疗因尘螨提取物和金黄色葡萄球菌肠毒素B诱导小鼠特应性皮炎/皮肤瘙痒症上显现出显著的抑制炎症的效果,且在皮肤外观和病理上显示出有改善的效果,在抓挠行为学上,也呈现出好转趋势。
实施例67:化合物对小鼠白癜风模型的治疗效果
一、材料和方法
实验动物
健康C57BL/6小鼠,雌雄各半(厂家:湖南斯莱克景达实验动物有限公司,许可证号:SCXK(湘)2019-0004)。饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)
动物分组和造模
(1)正常对照组(5只,不做处理)
(2)白癜风模型组(8只,不做处理)
(3)白癜风模型+溶媒处理组(8只,空白凝胶制剂,剂量:1.5g/kg,每天2次)
(4)白癜风模型+阳性对照药物处理组(8只,0.03%他克莫司,每天2次,50mg/次)
(5)白癜风模型+MDI-1228(0.5%)化合物凝胶制剂组(8只,1g/kg,每天2次)
(6)白癜风模型+MDI-1228(1%)化合物凝胶制剂组(8只,1.5g/kg,每天2次)
(7)白癜风模型+MDI-1228(3%)化合物凝胶制剂组(8只,1.5g/kg,每天2次)
实验环境:温度20℃-26℃,湿度40%-70%
主要试剂:
空白凝胶,0.5%,1%以及3%MDI-1228凝胶根据实施例64中描述的工艺路线制得。
莫诺本宗脱色膏(厂家:南京新妆颜);
他克莫司软膏(厂家:安斯泰来制药,批准文号:国药准字J20140147)。
造模步骤:小鼠适应性饲养7天后,制作白癜风模型:脱去小鼠背部2cm2黑色毛,用刮勺稍用力涂药。正常对照组涂抹凡士林50mg每日一次;模型组涂抹40%莫诺本宗乳膏50mg每日一次,连续给药93天。
模型验证:取空白组和模型组各1只小鼠做模型验证。皮毛脱色评估标准:每日肉眼观察皮毛脱色,并进行积分记录。将脱色部位分为用药部位和非用药部位,每个部位总分为5分,每只小鼠共10分。小鼠皮毛脱色评分标准:0分为皮毛无脱色,1分为皮毛脱色面积0~10%,2分为皮毛脱色面积11%~25%,3分为皮毛面积26%~50%,4分为皮毛脱色面积51%~75%,5分为皮毛脱色面积76%~100%。
注:为了防止动物之间有舔舐药物的现象。动物涂完凝胶后,单独放置在无垫料的独立笼内活动一段时间,让凝胶自然风干(预计1小时内),然后再放入饲养笼正常喂养。禁单独喂养,容易出现抑郁;禁用纱布包裹涂抹部位,会吸附走药剂。
给药后小鼠白癜风疗效观察
治疗给药35天后,开始15天内肉眼观察治疗小鼠白癜风疗效,每3天观察一次(共5次)并拍照。疗效评判标准:将脱色部位分为用药部位和非用药部位,每个部位总分为5分,每只小鼠共10分。小鼠皮毛脱色评分标准:0分为皮毛无脱色,1分为皮毛脱色面积0~10%,2分为皮毛脱色面积11%~25%,3分为皮毛面积26%~50%,4分为皮毛脱色面积51%~75%,5分为皮毛脱色面积76%~100%。
病理检测
(1)HE染色:取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ15min、Ⅱ15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60min。石蜡包埋,切片。将石蜡切片进行展片、烤片,然后进行脱蜡、水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗, 伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
(2)ELISA检测:试剂盒采用双抗体一步夹心法酶联免疫吸附试验(ELISA)。往预先包被小鼠待检指标捕获抗体的包被微孔中,依次加入标本、标准品、HRP标记的检测抗体,经过温育并彻底洗涤。用底物TMB显色,TMB在过氧化物酶的催化下转化成蓝色,并在酸的作用下转化成最终的黄色。颜色的深浅和样品中的待检物呈正相关。用酶标仪在450nm波长下测定吸光度(OD450值),计算样品浓度。所使用的试剂盒信息如下:小鼠白细胞介素6(IL-6)试剂盒(MM-0163M1,酶免),小鼠肿瘤坏死因子α(TNF-α)试剂盒(MM-0132M1,酶免)。
主要仪器设备
二、实验结果
给药后小鼠白癜风疗效观察
外用莫诺苯宗乳膏后首先在用药部位出现斑点状脱色,再在非用药部位如耳部和尾部等也出现脱色。主要是因为莫诺苯宗能激活T淋巴细胞自体免疫反应而在色素沉积部位和非暴露区出现脱色。模型组小鼠用药部位和非用药部位的脱色面积评分分别为3.43±0.20,1.57±0.20。与模型组比较, MDI-1228给药组均能降低脱色面积评分,可以改善白癜风小鼠皮毛脱色的现象,差异具有显著性(p<0.01)。在皮毛脱色评分的曲线下面积(AUC)比较中,MDI-1228药物组与阳性对照药物组相当。与模型组比较,MDI-1228(3%)凝胶制剂组评分AUC降低最多,效果最好(图30、31)。
HE染色
结果见图32。与正常组相比,模型组毛囊数量有所减少,及毛囊内黑色素聚集现象减少,个别样本出明显的表皮变薄,棘细胞有空泡变性的现象,且炎症细胞浸润明显,两性比较之下,雌性更为明显;阳性对照药物组毛囊数量及黑色素颗粒明显增加,表皮增厚,炎症细胞有所较少;用药组中,MDI-1228(1%)和MDI-1228(3%)组效果最为明显,两组毛囊数量明显增多,黑色素颗粒聚集明显,总体治疗效果无明显差异,但雌性鼠总体炎症数量大于雄性鼠,MDI-1228(1%)组雄性鼠炎症细胞浸润改善最为明显。
ELISA检测
与正常空白对照比较,模型组TNF-α含量升高,与模型组比较,阳性对照药物组和MDI-1228药物组的含量均降低,且药物组减少TNF-α含量的趋势呈剂量依赖关系,MDI-1228(3%)药物组降低TNF-α含量最显著(p<0.01),见图33。
与正常空白对照比较,各组血清IL-6含量均升高,模型组和空白凝胶组IL-6含量升高最显著(p<0.01),与模型组比较,阳性对照药物组和MDI-1228药物组的含量均降低,且药物组减少IL-6含量的趋势呈剂量依赖关系,MDI-1228(3%)药物组降低IL-6含量最显著(p<0.01),见图34。
三、结论
在药物诱导的小鼠白癜风模型上,经过不同浓度MDI-1228药物治疗后,各给药组的皮毛脱色面积评分均降低,毛囊数量明显增多,黑色素颗粒聚集明显,血清中TNF-α,IL-6含量降低,说明MDI-1228药物能改善小鼠白癜风小鼠皮毛脱色的现象,具有明显治疗效果。
实施例68:MDI-1228化合物凝胶制剂及MDI-1228化合物对小鼠斑秃的 治疗效果
一、材料和方法
实验动物
健康C3H/HeJ小鼠(6周龄)72只,雌雄各半(厂家:北京维通利华实验动物有限公司,许可证号:SCXK(京)2016-0011),饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)。
动物分组和造模
空白对照组(不做处理)(N=5)
空白斑秃模型组(不做处理)(N=8)
斑秃模型+空白赋形剂组(溶媒处理:空白凝胶制剂,剂量:1.5g/kg次/只,2次/天/只)(N=8)
斑秃模型+阳性对照药物组(复方倍他米松乳膏(糖皮质激素),每天2次,50mg/次/只,)(N=8)
斑秃模型+0.5%MDI-1228化合物凝胶制剂组(剂量:1.0g/kg次/只,2次/天/只)(N=8)
斑秃模型+1%MDI-1228化合物凝胶制剂组(剂量:1.5g/kg次/只,2次/天/只)(N=8)
斑秃模型+3%MDI-1228化合物凝胶制剂组(剂量:1.5g/kg次/只,2次/天/只)(N=8)
空白斑秃模型(溶媒处理:5%DMSO+75%PEG300+20%EtOH)(N=8)
造模:在动物项部涂敷咪喹莫特乳膏(无需脱毛),每周3次,连续3周。对照组仅用不含咪喹莫特的空白赋形剂。每次先用清洁的医用棉签蘸少许咪喹莫特约0.03g均匀涂抹,范围约为1cm×1cm,要求每次涂抹前均使用生理盐水清洗涂抹部位。操作中动作尽量轻柔,涂抹均匀即可,避免反复摩擦导致皮肤划伤或毛发机械性脱落。约20天后,涂抹咪喹莫特乳膏的部位附近形成面积大于1cm×1cm的脱发斑,此时斑秃病理模型方为制备成功。造模期间,记录小鼠毛发脱落情况。
主要试剂及其配制
空白凝胶,0.5%,1%以及3%MDI-1228凝胶根据实施例64中描述的工艺路线制得。
咪喹莫特(厂家:四川明欣药业,批准文号:国药准字H20030129);
复方倍他米松新霉素乳膏(厂家:联邦制药,批准文号:国药准字1120040492)。
动物给药
空白对照组(不治疗)不做任何处理;药物组使用对应剂量的复方倍他米松乳膏、空白凝胶制剂、MDI-1228化合物凝胶制剂进行皮肤涂抹,每天2次(9点和17点,间隔6h以上)。注:为了防止动物之间有舔舐药物的现象。动物涂完凝胶后,单独放置在无垫料的独立笼内活动一段时间,让凝胶自然风干(预计1小时内),然后再放入饲养笼正常喂养。禁单独喂养,容易出现抑郁;禁用纱布包裹涂抹部位,会吸附走药剂。
临床症状评分
治疗期间观察小鼠脱毛部位局部变化,详细记录其毛发生长及不良反应,并按照下表打分。
HE染色
HE染色所需药瓶和使用仪器有:苏木素染液(AR11800-1,BOSTER);伊红染色液(AR11800-2,BOSTER);超净高级封片胶(YZB,BASO);Scott蓝化液(G1865,Solarbio)等。药品冷藏柜(BYC-310,山东博科生物);显微镜(CX41 OLYMPUS);切片机(BQ-318D,伯纳);电热鼓风干燥箱(DHG-9070A,恒科学仪器有限公司)。取出组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ15min、Ⅱ15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60分钟。石蜡包埋,切片。
将石蜡切片进行烤片,然后脱蜡,水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
二、实验结果
临床症状评分
给予造模药物咪喹莫特后,小鼠毛发散乱,排列紊乱,形成明显的斑秃样病损。在不同时间点的毛发生长评分线性图中,随着给药时间的推移,含不同剂量的MDI-1228凝胶均能持续地促进受试动物的毛发生长。阳性对照药物复方倍他米松新霉素乳膏在前2周治疗中能促进毛发生长,但是从第三周开始逐渐出现了明显的副作用,该组动物皮肤出现了萎缩变薄,毛细血管扩张,有潮红现象,毛发生长速度也出现了回落。通过计算各组毛发评分的曲线下面积(AUC),和空白凝胶组进行比较,结果显示不同浓度的MDI-1228凝胶均能显著促进斑秃动物的毛发生长(图35)。经过21天药物治疗后,模型组小鼠毛发生长缓慢,评分低。各剂量MDI-1228药物治疗组毛发生长评分均有不同程度的升高,与模型组比较,评分均显著性升高,其中以MDI-1228(3%)效果最好(图36)。
HE染色
小鼠皮肤样品经过HE染色后,进行病理检测和分析。正常小鼠的毛囊结构发育良好。斑秃模型组小鼠皮肤表皮出现增生,真皮内成纤维细胞浸润增多,毛囊出现集束状,部分毛囊口可见大量角质栓伸入其内。毛囊萎缩和空泡化,毛乳头上移。空白凝胶组和溶媒组未见显著变化,毛囊大多与模型组一样,处于退行期或休止期。MDI-1228治疗组中,不同浓度的药物均有一定的治疗效果,三个浓度组中存在结构完整的生长期毛囊,其中以雌性MDI-1228(1%)组和雄性MDI-1228(3%)组较为明显,但从毛囊总体数量及毛囊的恢复期观察,雌性MDI-1228(3%)组和雄性MDI-1228(1%)组毛囊数量较多(图37和图38)。
三、实验结论
以上试验结果表明MDI-1228能在一定程度上改善小鼠斑秃症状,有助于毛发的生长。
实施例69:化合物在咪喹莫特诱导的银屑病模型的体内药效
一、实验材料与设备
实验材料
5%咪喹莫特乳膏,Aldara,3M Pharmaceuticals
受试化合物:
受试化合物为MDI-1228的甲磺酸盐(MDI-1228_mesylate),其制备方法如下:
1)取5.00g化合物MDI-1228加入到三口瓶中,加入100mL甲醇升温至50~55℃搅拌,体系变得粘稠,又加入50mL甲醇,继续搅拌5h。过滤,滤饼用20mL甲醇淋洗,45℃下真空干燥8h,最终得到3.7g的晶体,其为MDI-1228的水合物。
2)称取约200mg的MDI-1228水合物晶体至20mL玻璃小瓶中;
3)加入54mg甲磺酸至20mL玻璃小瓶中,并加入10mL EtOAc;
4)25℃悬浮搅拌约3天后得到乳白色悬浊液,室温减压抽滤得到固体,将固体转移至室温真空干燥3小时得到白色粉末状样品,即为MDI-1228甲磺酸盐,其纯度大于99%。
溶媒:5%DMSO:75%PEG300:20%EtOH
受试化合物溶液配制方法:称取化合物,溶于DMSO,再加入(PEG300:乙醇=3.75:1)的溶液配置成最高剂量(30MPK,母液)。使用时用溶媒稀释母液来配制低浓度化合物溶液。
对照化合物:VecticalTM(Calcitriol,即骨化三醇乳膏),Ointment,Galderma
实验仪器
剃毛器:Codos CP-5000.
化合物称量天平:Sartorius,CPA225D
动物称量天平:常州天之平电子天平,YH2000
实验动物及饲养环境
实验方法
试剂准备
咪喹莫特乳膏:除正常组以外的所有组别小鼠,每只称量62.5mg,每天称取。
溶媒:量取5%DMSO,加入75%PEG300,最后加入20%EtOH后混匀使用,4℃保存备用。
MDI-1228甲磺酸盐:称取化合物,溶于DMSO,再加入(PEG300:乙醇=3.75:1)的溶液配置成最高剂量(30MPK,母液)。用溶媒稀释母液配制低浓度化合物。化合物每3天配制一次,4度保存。
小鼠剃毛
实验开始前,小鼠背部剃毛,通过使用模具保证面积一致,约2cm*3cm。
分组和给药安排
实验开始前一天,依据体重将80只小鼠随机分成6组,保证每组体重平均值一致。组内体重过重或者过轻,以及剃毛后发现背部皮肤发黑、皱褶过大的小鼠都被剔除,不纳入本次实验。最终入组55只小鼠,分组详细信息见下表:
咪喹莫特致敏和受试药物给药上午9:00在第2至第6组小鼠背部剃毛区皮肤上进行涂抹给药,
下午4:30在第2、第4、第5和第6组小鼠背部剃毛区皮肤上进行涂抹给药,
为诱导第2至第6组小鼠出现银屑病样症状,下午1:00在小鼠背部剃毛区皮肤上涂抹咪喹莫特乳膏,持续7天,正常组小鼠剃毛后不做任何处理。
模型评分
每天上午记录小鼠体重,并对背部皮肤炎症严重程度进行评分。临床评分分为三个指标,包括红斑、结痂和增厚,各项指标均为0-3分。病理 分析
将小鼠皮肤样品收集于固定液中,然后进行石蜡包埋、切片及HE染色。病理医生对染色结果进行评分。具体银屑病皮肤病理评分细则见下表所示。
银屑病皮肤病理评分标准
细胞因子检测
取动物背部皮肤速冻、匀浆并收集上清,后续通过超敏试剂盒进行细胞因子检测(TNF-α)。
统计学处理
实验数据应用平均数±标准误表示(Mean±SEM),临床评分和体重用双因素方差分析(Two way ANOVA),AUC,细胞因子和病理结果用单因素方差分析(One way ANOVA),认为p<0.05有显著性差异。
二、实验结果与讨论
临床评分
本实验评价了化合物MDI-1228甲磺酸盐在咪喹莫特诱导的小鼠银屑病模型中对临床评分的改善作用。溶媒对照组的平均临床评分逐渐升高,至第4天达到7.8分(皮肤厚度达到2.4分),提示咪喹莫特诱导的小鼠银屑病模型的成功建立(图39-图42、表3、表4)。MDI-1228甲磺酸盐在3,10和30mg/kg三个剂量下,对小鼠银屑病临床评分均有抑制作用,其中30mg/kg MDI-1228甲磺酸盐组最显著。针对皮肤厚度,30mg/kg MDI-1228甲磺酸盐组从第3天开始与溶媒组有显著性差异,并持续到实验结束,效果与阳性药相近(p<0.0001)。
通过分析每组每只动物的皮肤厚度评分曲线,计算曲线下面积AUC,通过组间AUC平均值,计算各给药组相对于溶媒对照组的抑制率,结果如图39。MDI-1228甲磺酸盐在3,10和30mg/kg给药组抑制率分别为30.2%,25.1%和58.1%。与溶媒对照组相比,3mg/kg剂量组的p值小于0.05;10mg/kg剂量组的p值小于0.01;30mg/kg剂量组的p值小于0.0001。阳性药骨化三醇组抑制率为64.2%。
通过分析每组每只动物的临床评分曲线,计算曲线下面积AUC,通过组间AUC平均值,计算各给药组相对于溶媒对照组的抑制率,结果如图40。MDI-1228甲磺酸盐在3,10和30mg/kg给药组抑制率分别为10.7%,12.0%和29.8%,呈剂量依赖性。与溶媒对照组相比,3,10mg/kg剂量组的p值小于0.05;30mg/kg剂量组的p值小于0.0001。阳性药骨化三醇组抑制率为47.3%
实验终点收集小鼠脾脏称重,该模型中脾脏重量与疾病严重程度正相关。如图43所示,30mg/kg MDI-1228甲磺酸盐组显著抑制了脾脏的增重(p<0.05)。
表3.咪喹莫特诱导的银屑病小鼠皮肤厚度评分数据统计学分析
表4.咪喹莫特诱导的银屑病小鼠临床评分数据统计学分析
病理结果
将小鼠皮肤样品收集于固定液中,然后进行石蜡包埋、切片及HE染色。病理医生对染色结果进行评分。银屑病皮肤病理评分结果如图44所示,MDI-1228甲磺酸盐在3,10和30mg/kg给药条件下显著抑制了银屑病样皮炎中角化不全、表皮突长度、棘层增厚和淋巴细胞浸润的现象,病理总分的抑制率分别为59.7%,51.6%和50.8%。从组织病理学上,骨化三醇呈现出缓解的趋势。
TNF-α检测结果
取动物背部皮肤速冻、匀浆并收集上清,后续通过超敏试剂盒进行细胞因子TNF-α的检测。细胞因子检测结果如图45所示。与病理结果相对应,MDI-1228甲磺酸盐在3,10和30mg/kg给药条件下显著抑制了皮肤样品中TNFa的浓度(*p<0.05)。
三、实验结论
受试化合物MDI-1228甲磺酸盐3、10和30mg/kg剂量组对银屑病均有明显改善作用;受试化合物MDI-1228甲磺酸盐3、10和30mg/kg剂量组对银屑病模型皮肤样品中细胞因子TNF-α有显著抑制作用。
实施例70:化合物对兔痤疮模型的治疗效果
一、材料和方法
实验动物
新西兰家兔,1.7-2.2kg(厂家:赣州市畜牧研究所,许可证号:SCXK(赣)2018-0009))雄性,饲养条件(普通饲料饲养,温度20~26℃,湿度40%~70%)
主要试剂和药物配制
1)2%煤焦油溶液的配制:
取49ml玉米油、1ml煤焦油、充分震荡混匀;
2)哥伦比亚血琼脂平板:
取3.9g哥伦比亚血琼脂基础培养基粉末溶于100ml蒸馏水中,121℃高压灭菌20min,冷却至50-55℃,加入5ml无菌脱纤维羊血混合均匀倒平板备用。BHI液体培养基:取3.85g BHI溶于100ml蒸馏水中,121℃高压灭菌20min,冷却后放冰箱备用。
3)痤疮丙酸杆菌Propionibacterium acnes(P.acnes ATCC11827):
菌种先在哥伦比亚血琼脂培养板上活化,然后接种至到BHI培养基中在厌氧条件下(10%H2、10%CO2和80%N2)培养,37℃培养48h。利用离心法收集培养菌,悬浮于PBS中,所得悬浮液为6×108CFU/ml。
4)溶媒的配制(5%DMSO+75%PEG300+20%EtOH):
取1ml DMSO+4ml EtOH+15ml PEG300充分混匀得无色澄清溶液。
5)20mg/ml MDI-1228(1mg/kg)的配制:
称取MDI-1228 0.0817g+0.21ml DMSO溶解,加0.82ml EtOH,再加3.08ml PEG300,得浅白色乳浊液。
6)60mg/ml MDI-1228(3mg/kg)的配制:
称取MDI-128 0.2634g+0.22ml DMSO溶解,加0.88ml EtOH,再加3.29ml PEG300,得白色乳浊液。
7)180mg/ml MDI-1228(9mg/kg)的配制:
称取MDI-1228 0.5788g+0.16ml DMSO溶解,加0.64ml EtOH,再加2.41ml PEG300,得白色乳浊液。
8)60mg/ml MDI-1288(3mg/kg)的配制:
称取MDI-1288 0.2595g+0.22ml DMSO溶解,加0.86ml EtOH,再加3.24ml PEG300,得黄色澄清溶液。
动物分组和造模
空白对照组(不做处理)(N=5+2)
模型组(溶媒处理:5%DMSO+75%PEG300+20%EtoH,2次/天/只,涂抹给药,给药2周)(N=8+2)
模型+阳性对照组(1%克林霉素凝胶,0.5g/kg,1次/天/只,涂抹给药, 给药2周)(N=8)
模型+低剂量MDI-1228化合物组(剂量:1mg/kg/次,2次/天/只,涂抹给药,给药2周)(N=8)
模型+中剂量MDI-1228化合物组(剂量:3mg/kg/次,2次/天/只,涂抹给药,给药2周)(N=8)
模型+高剂量MDI-1228化合物组(剂量:9mg/kg/次,2次/天/只,涂抹给药,给药2周)(N=8)
模型+MDI-1288化合物组(剂量:3mg/kg/次,2次/天/只,涂抹给药,给药2周)(N=8)
造模:兔子适应性实验7天,在兔右耳内侧耳管出口处2cm×2cm范围,每日涂2%煤焦油溶液1次,每次0.5~1mL,连续26天。痤疮丙酸杆菌菌种直接接种于生梭孢菌培养基,置于厌氧袋中,37℃培养48h后,使用生理盐水调整痤疮丙酸杆菌为6×108CFU/ml,备用。第8天兔耳皮内3-4位点注射痤疮丙酸杆菌,50μl每只,隔日注射1次,共注射5次,最后一次注射后第9天随机分组开始治疗。
动物给药
兔子按照分组用药,空白组不做任何处理,阳性对照组给予克林霉素凝胶,每天涂抹1次,连续14天。给予低、中、高剂量的MDI-1228化合物和MDI-1288涂抹耳朵每天两次,连续14天给药。模型组给予等量溶媒处理,处理方式和时间同供试药物组。
检测指标
末次给药24h后,使用2.8cm打孔器在涂药部位打孔制备耳片,并测量其重量;取造模部位耳朵4%多聚甲醛固定后,相同位置连续切片(3张),进行HE染色,进行病理组织学分析,同时测定3个毛囊面积和3个皮脂腺直径,计算平均值;心脏取血,离心取血清按ELISA试剂盒要求测定血清中白细胞介素-1α(IL-1α)、白细胞介素-6(IL-6)和二氢睾酮(DHT)含量。
组织病理学检测
HE染色:
HE染色所需药瓶和使用仪器有:苏木素染液(AR11800-1,BOSTER);伊红染色液(AR11800-2,BOSTER);超净高级封片胶(YZB,BASO);Scott蓝化液(G1865,Solarbio)等。
设备主要有药品冷藏柜(BYC-310,山东博科生物);显微镜(CX41OLYMPUS);切片机(BQ-318D,伯纳);电热鼓风干燥箱(DHG-9070A,恒科学仪器有限公司)。
实验流程主要为切取动物耳片组织,4%多聚甲醛固定,脱水,石蜡包埋,组织切片HE染色观察组织病理形态变化。具体操作步骤为先将样品置于4%多聚甲醛常温固定4小时,取出耳片组织流水冲洗数小时,经70%、80%、90%各级乙醇溶液脱水,纯酒精、二甲苯等量混合液15min,二甲苯Ⅰ15min、Ⅱ15min(至透明为止)。放入二甲苯和石蜡各半的混合液15min,再放入石蜡Ⅰ、石蜡Ⅱ透蜡各50-60分钟。石蜡包埋,切片。将石蜡切片进行烤片,然后脱蜡,水化。将已入蒸馏水后的切片放入苏木精水溶液中染色3min,盐酸乙醇分化液分化15s,稍水洗,返蓝液返蓝15s,流水冲洗,伊红染色3min,流水冲洗,脱水,透明,封片,镜检。
ELISA检测:
试剂盒采用双抗体一步夹心法酶联免疫吸附试验(ELISA)。在预先包被有能特异识别和捕获待测细胞因子抗体的微孔板中,依次加入标本、标准品、HRP标记的检测抗体,经过温育并彻底洗涤。用底物TMB显色,TMB在过氧化物酶的催化下转化成蓝色,并在酸的作用下转化成最终的黄色。颜色的深浅和样品中的待检物呈正相关。用酶标仪在450nm波长下测定吸光度(OD450值),计算样品浓度。
所使用的试剂盒信息如下:兔白介素1α(IL-1α)试剂盒(MM-8267601,酶免),兔白细胞介素6(IL-6)试剂盒(MM-030201,酶免),兔双氢睾酮(DHT)试剂盒(MM-8267901,酶免)。
主要仪器设备
二、实验结果
1.兔子一般症状观察和药物对兔耳片重量的影响
一般观察,正常组兔子耳廓菲薄,柔软,毛细血管清晰,内侧可见清晰细胞毛囊和纤细毛发,模型组耳朵明显增厚,毛孔粗大,粗糙。兔子耳片重量检测中,与空白对照组比较,模型组耳片重量显著性升高,阳性对照药物克林霉素组未见降低耳片重量,3mg/kg MDI-1228、9mg/kg MDI-1228和3mg/kg MDI-1288组可以降低耳片重量,减轻兔耳的增厚情况(图46)。
2.HE染色结果
模型(溶媒)组可见角质层增厚,表皮层凹凸不平,结构紊乱,毛孔粗大,皮脂腺腺泡增多,真皮层有出血及炎症细胞浸润情况。阳性对照药物克林霉素组仍可见大量角质增生,表皮痤疮疮面减少,但毛孔粗大的现象未得到改善。其他治疗组中,从数据分析上看,3mg/kg MDI-1228中剂量组治疗效果最佳,相较其他组而言,中剂量组毛孔收缩效果较为明显,表皮痤疮疮面愈合度较高,角质层角化程度相对较低(图47)。通过测量和计算各切片的皮脂腺直径和毛囊面积,结果表明各组的皮脂腺直径变化不显著(图48),但是毛囊面积有显著变化。模型组的毛囊面积显著增加,克林霉素治疗未能改善此症状,但是3mg/kg MDI-1228和9mg/Kg MDI-1228能显著减少毛囊的扩大(图49)。
3.药物对兔血清IL-1α、IL-6和DHT含量的影响
DHT可与皮脂腺细胞内受体结合,刺激皮脂腺细胞的增生和分泌,毛囊皮脂腺角化过度而使排泄皮脂的通道变窄导致的厌氧环境促进了丙酸杆菌繁殖,形成了皮脂增生-排脂受阻-细菌感染为轴心的痤疮发病机制。丙酸杆菌和toll样受体结合,引发一系列的炎症反应,诱导IL-1α和IL-6的产生进一步引起毛囊的炎症损伤。该模型中可见异常的睾酮升高,睾酮的大量增加而导致DHT合成的增加。
各组兔子IL-1α,IL-6和DHT检测,与空白对照组比较,模型(溶媒)组IL-1α、LI-6和DHT含量均显著升高,3mg/kg MDI-1228、9mg/kg MDI-1228和3mg/kg MDI-1288组与模型(溶媒)组比较,IL-1α和DHT含量显著性减少(图50和图52)。1mg/kg MDI-1228低剂量组有降低3个指标趋势(图51)。
三、结论
以上实验表明在煤焦油联合丙酸杆菌致兔痤疮模型中,MDI-1288和MDI-1228具有降低炎症因子表达、改善痤疮的作用。
实施例72化合物对体外脂肪细胞脂解的效果
将分离提取的新生C57BL/6小鼠皮肤成纤维细胞(具体操作步骤见文献:Cell Reports(2023)/doi:10.1016/j.celrep.2023.112647),铺板种植于培养皿中,待细胞密度最大达到接触抑制时,加入分化培养基(DMEM+10%FB+1x抗生素/具体配方见文献:Cell Reports(2023)/doi:10.1016/j.celrep.2023.112647),诱导新生脂肪细胞生成,随后加入胰岛素,促脂肪细胞成熟,之后分别加入实验药物(MDI-1228或者Tofacitinib), Control是加入等量DMSO的空白对照,用Leica倒置相差显微镜观察脂肪细胞的转分化变化。实验流程和计划如图53A所示。从细胞相差图片(图53B)可见,在已经分化的成熟脂肪细胞中加入MDI-1228(5μM)能促进脂肪细胞脂解为小的脂滴,而加入另一个JAK抑制剂Tofacitinib(5μM)则不能促进脂解。
实施例73化合物对糖尿病足动物模型的治疗效果
1.高脂诱导肥胖模型建立
从集萃药康订购2个月正常野生C57BL/6雄鼠20只,10只为一组,分为两组,一组喂食正常饲料(standard diet,SD),一组喂食60%高脂饲料(high fat diet,HFD)诱导小鼠肥胖,两组在同样饲养环境下喂食约6个月。
另订购2月龄年轻雄鼠5只,与SD组(8个月龄,10只)和HFD组(8个月龄,10只)进行后续的伤口造模和比较。
2.小鼠伤口造模+给药处理
2.1.小鼠伤口造模:
使用异氟醚麻醉小鼠,使用剃毛器除去背部毛发,裸露出背部皮肤,先使用记号笔和直尺在背部两侧各画出1.0cm x 0.5cm大小的长方形区域,用消毒的手术剪把该区域皮肤剪掉,勿伤及皮下组织。待小鼠清醒后,将其小心放回笼中,在受伤以后的指定时间间隔点处死动物并收集创面组织用于后续实验和分析。
2.2.给药处理:
小鼠背部左右两侧造伤口后,按照下表进行分组和给药(其中Control表示DMSO+溶剂的空白药物对照)。给药时,每12小时给予1228凝胶(0.5%或1%)一次,每次给药剂量50mg,每天(每24小时)对伤口进行拍照记录并称重,直到Day 7(第7天)收样,具体操作细节见后面“3收样和分析”所述。
2.3.观察(每天记录伤口面积变化)
小鼠麻醉后,置于操作台上,背部舒展,进行小鼠全背部和伤口局部的高清照片拍摄(含标尺),该数据用于后期伤口图片展示和伤口大小的定量比较。并需要测量小鼠体重,以便分析药物对全身性代谢的影响。
3.收样和分析
3.1.收样操作步骤
在小鼠背部造模后,伤口修复的第七天处死小鼠,并收集创面组织用于后续实验和分析,分别包括伤口中心组织、和远端非创口对照皮肤,组织包埋于OCT经冷冻切片用于后续HE或多重染色分析等实验。
3.2.冰冻组织和切片
取小鼠皮肤组织放入装有OCT的包埋盒(O.C.T.Compound,Catalog#4583,Sakura Finetek USA Inc))中,平放于干冰上迅速冷冻,待OCT完全冷冻后转移包埋盒至-20℃冰箱保存。切片前先预冷冰冻切片机,使温度降至-25℃,将冷冻的组织块上样本夹固定,先设置切片厚度为50um将组织修平,然后设置切片厚度为16um,调整防卷板开始正式切片,将展平的组织切片附贴在载玻片上,在显微镜下观察切片是否完整。切好的片子在室温晾置2-3h,再放入-20℃冰箱储存。
3.3.HE染色
首先将切片从-20℃拿出,并且在室温下解冻,晾干冷凝水10-20分钟直到完全干燥,然后用500-1000微升冷冻的4%多聚甲醛(4%多聚甲醛需要用10毫升16%PFA-多聚甲醛和30毫升冷冻的PBS溶液稀释,并且保存在4℃环境下)室温下在切片上固定组织10分钟,再用dH2O(单次蒸馏水)洗一次切片(孵育<1分钟),洗完后加入苏木精溶液,孵育55-60秒,用流动的自来水快速洗3次直到没有蓝色染料出现为止。再加入伊红溶液,孵育25-30秒,用流动的自来水快速洗3次,再用70%乙醇洗一遍。使用100%乙醇浸泡脱水5分钟,再置于二甲苯中脱水5分钟。最后用中性树脂封片,用镊子夹住盖玻片平稳盖下,并且值得注意的是小心不要在切片上挤出气泡。在通风橱中放置一段时间后即可用显微镜观察结果并拍照记录。
实验结果见图54。
图54A:将各组小鼠的非伤口处皮肤和伤口组织剪下,包埋于OCT中,进行冷冻切片,组织PFA固定后用于苏木精-伊红(HE)染色。图中展示了组织染色后全貌,黄色虚线框为待放大区域,伤口边缘的表皮迁移舌结构用黄色勾勒出。
图54B:图54A中黄框区域的放大图,白色空心结构初步判断为脂肪组织,标尺为400微米。
从图54A可见:HFD组的伤口上方的表皮细胞迁移舌相距最远,2MSD组(即SD-2M组)的伤口最小,而且伤口中实质细胞最多,而MDI-1228处理组,相比于对照组,伤口中的空白区域(初步判断为脂肪细胞)更少,类成纤维细胞更多。进一步的将伤口中心局部区域放大后进行观察(图54B),可见:MDI-1228处理后相比对照组,伤口中心的脂肪细胞大小有显著减少,这暗示抑制剂MDI-1228可能促进了脂肪细胞脂解并向pAd或myoFB转分化。
另外,通过Bodipy(绿色标记脂滴)/PHA(红色标记肌动蛋白)和细胞核(DAPI染色,蓝色)染色实验进一步发现了:MDI-1228处理组的伤口中心绿色脂滴面积明显减小,小脂滴数量增加;这进一步确认了MDI-1228可能促进了伤口中脂肪细胞的脂解以及向pAd/myoFB的转分化。
3.4.DAPI染色
为了进一步明确药物对上皮细胞向伤口中心迁移的改变作用,伤口组织进行Bodipy(绿色标记脂滴)/PHA(红色标记肌动蛋白)染色,细胞核用DAPI(蓝色)染色标记,该染色能清晰区分皮肤不同层面的组织结构。Bodipy采购自ThermoFisher Scientific(Catalog D3922);PHA采购自Cytoskeleton Inc(Catalog PHDH1-A);含的DAPI染料的封片剂采购自ThermoFisher Scientific(Catalog P36931),具体染色方法参考文献Cell Reports(2023)/doi:10.1016/j.celrep.2023.112647。
Bodipy/PHA染色过程如下:首先将切片从-20℃取出,在室温下解冻干燥10分钟,用Immunopen玻片标记笔标记好玻片的顶部和底部,再用1毫升4%多聚甲醛溶液室温下固定10分钟,用PBS溶液洗玻片5分钟,重复两次,用标记笔在组织切片区域画圆框出。然后将组织载玻片用稀释的Bodipy(脂滴染料)和Rhodamine Phalloidin(肌动蛋白染料)溶液在室温下合适湿度黑暗环境下于PBS缓冲液中孵育1小时,再用PBS溶液洗玻片10分钟,重复三次。最后用含DAPI的延长金抗褪色固定试剂(ProLongTM Gold Antifade Mountant with DAPI)封定染色切片,这个过程能将细胞核用DAPI(蓝色)染上,最后将封定好的切片储存在4℃环境下。随后使用共聚焦显微镜观察和拍摄。
实验结果见图55。图55中,伤口两边向伤口中心迁移的上皮(migrating epithelium tongue)用黄色点线标记,两边上皮中心的距离用红色双箭头标记,并定量其长度。
图55(A):各组小鼠(SD,standard diet;HFD,high fat diet;;ctrl=空白凝胶;1228=1%MDI-1228凝胶)的伤口皮肤通过Bodipy+PHA+DAPI染色后,伤口两边正在往中心迁移的上皮通过黄色点线标记,伤口中心未有上皮迁移的距离(伤口未愈合距离)用红色双箭头线标记,并定量。
图55(B):图55(A)图中伤口中心未愈合距离定量柱图,纵坐标单位为毫米(mm),组间显著性差异(One way Anova)的p value标注在图上,每组=3~4个伤口
伤口愈合过程中,两边的上皮会被激活形成migrating tongue,并往伤口中心迁移,当两边migrating tongue在伤口中心交汇时,伤口再上皮化完成(Adv Wound Care(New Rochelle).2014 Jul 1;3(7):445-464.),因此通过测量migrating tongue的距离,可以更加精准地定量伤口再上皮化的情况。发明人通过对不同组间migrating tongue的距离进行定量测量,发现高脂(HFD)小鼠的上皮愈合较正常饮食(SD)组较慢,并且给HFD组涂抹1%MDI-1228后能有效促进上皮愈合(图55)。
结论
以上实验表明在小鼠糖尿病足模型中,MDI-1288能够明显的促进脂解,显著增加肌成纤维细胞的比例,有效的促进伤口愈合。
虽然前面已经阐明并描述了本发明的特定实施方式,但并不意味着这些实施方式阐明了并描述了本发明的所有可能形式。更确切地,用在本说明书中的文字仅仅是描述性的文字并非限制性的。对于本领域技术人员明显的是,在不脱离本公开的一般范围的情况下,可以进行各种其他改变和修改。因此,在所附权利要求中,旨在包括在本发明范围内的所有这些改变和修改。

Claims (42)

  1. (G)的化合物:
    或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物在制备TRK抑制剂和/或RET抑制剂药物中的用途,其中:
    L为C=O、O=S=O、CH2或连接键;且
    X1为N或CR14;且
    X2为N或CR15;且
    X3为N或CR16;且
    R14、R15、R16各自独立地选自H、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C2-8烯基、C2-8炔基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C3-7环烷基、3-7元杂环烷基任选地被1个、2个或3个选自卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、-N(C1-4烷基)(C(=O)C1-4烷基)、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基的取代基取代;且
    R13为H、-N(R17)(R18)、C1-6烷氧基、-SR12、-OR12、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、 5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,C7- 11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且任选地被一个或多个各自独立选自以下群组的取代基所取代:-OH、-CN、-SH、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;或者R17、R18以及与它们相连的N原子共同形成3-14元环;且
    R2的个数为0、1、2、3或4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且
    R1选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述-S-C1-4烷基、C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;且
    R3、R4各自独立地选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1- 6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、-N(R5)(R6)、-N(R11)(C(=O)R12)、-CON(R7)(R8)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基、3-10元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、以及7-11元双环杂芳基各自任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-6烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12;且R5、R6、R7、R8、R9、R10、R11、R12各自独立地是H或选自以下群组:C1- 6烷基、C1-4卤代烷基、C3-7环烷基、4-14元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:卤素、-CF3、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、氧代、C1-4烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  2. 根据权利要求1所述的用途,其中所有H各自独立地任选地被D取代。
  3. 根据权利要求1或2所述的用途,其中X1、X2、X3其中仅一个为N。
  4. 根据权利要求1或2所述的用途,其中X1、X2、X3其中仅两个为N。
  5. 根据权利要求1或2所述的用途,其中X1、X2、X3是相同的。
  6. 根据权利要求5所述的用途,其中X1为CR14、X2为CR15、X3为CR16,且R14、R15、R16是相同的。
  7. 根据权利要求6所述的用途,其中R14、R15、R16选自H、-OH、-SH、-CN、卤素、-NO2、C1-6烷基。
  8. 根据权利要求7所述的用途,其中R14、R15、R16选自H、-OH、C1- 6烷基。
  9. 根据权利要求7所述的用途,其中X1、X2、X3均为CH。
  10. 根据权利要求5所述的用途,其中X1、X2、X3均为N。
  11. 根据权利要求1-10中任一项所述的用途,其中L为C=O、O=S=O或CH2
  12. 根据权利要求1-10中任一项所述的用途,其中R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基、C5-11双环烷基、5-11元双环杂烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。
  13. 根据权利要求1-10中任一项所述的用途,其中R13为H、-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5、-S-C1-4烷基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基,且R13被0个、1个、2个、3个或4个R1取代。
  14. 根据权利要求1-10中任一项所述的用途,其中R13为H、-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基,且R13被0个、1个、2个、3个或4个R1取代。
  15. 根据权利要求1-10中任一项所述的用途,其中R13为-N(R17)(R18)、C1-6烷氧基、C1-6烷基或者C3-7环烷基、4-6元杂环烷基、苯基、5-6元杂芳基,且R13被0个、1个、2个或3个R1取代。
  16. 根据权利要求1-10中任一项所述的用途,其中R17、R18各自独立地选自H、C1-6烷基、C3-7环烷基、C3-7杂环烷基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代。
  17. 根据权利要求1-10中任一项所述的用途,其中R17、R18以及与它们相连的N原子共同形成4-10元环。
  18. 根据权利要求1-10中任一项所述的用途,其中L为C=O,且R13为-N(R17)(R18)、C1-6烷氧基、-OH、-SH、-CN、卤素、-NO2、-SF5或-S-C1-4烷基,且R13被0、1、2、3或4个R1取代;其中R17、R18各自独立地选自H、C1-6烷基、C1-6烷氧基、C3-7环烷基、C3-7杂环烷基、C5-7芳基、5-7元杂芳基,且任选地被一个或多个-OH、-CN、-SH、卤素、-NO2、-SF5取代;或者R17、R18以及与它们相连的N原子共同形成3-14元环。
  19. 根据权利要求1-10中任一项所述的用途,其中存在1个、2个或3个R2,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、-SH、-S-C1-4烷基、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基,其中所述-S-C1-4烷基、C1-6烷基、C3-7环烷基、4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  20. 根据权利要求1-10中任一项所述的用途,其中存在1个、2个或3个R2,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  21. 根据权利要求15所述的用途,其中存在1个或2个R2,并且R2选自卤素、C1-6烷基。
  22. 根据权利要求1-10中任一项所述的用途,其中R13被0个或1个R1取代,并且R1选自卤素、-OH、-CN、C1-6烷基、5-7元杂环烷基、C3-7环烷基,其中所述C1-6烷基任选地被1个、2个或3个R3取代,并且其中所述5-7元杂环烷基、C3-7环烷基任选地被1个、2个、3个或4个C1-3烷基取代。
  23. 根据权利要求1所述的用途,其中所述化合物为式(I)的化合物:
    或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中:
    L为C=O、O=S=O、CH2或连接键;
    X为CH或N;
    环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基、11-15元三环基;
    R1的个数为0、1、2、3或4个,并且R1选自H、卤素、C1-8烷基、C2- 8烯基、C2-8炔基、C1-8烷氧基、C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基,其中所述C1-8烷基、C2-8烯基、C2-8炔基、C1-8烷氧基任选地被1个、2个、3个或4个R3取代,并且其中所述C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基、C7-11双环芳基、7-11元双环杂芳基任选地被1个、2个、3个或4个R4取代;
    R2的个数为0、1、2、3或4个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基以及4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12
    R3选自卤素、氰基、C1-3烷基、羟基、C1-6烷氧基、-N(R5)(R6)、-CON(R7)(R8)或3-7元杂环烷基,其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
    R4选自卤素、C1-3烷基、羟基、C1-6烷氧基、-NH2、-NHCH3或-N(CH3)2
    R5、R6、R7、R8各自独立地为氢或C1-4烷基;
    R9选自H、C1-4烷基、C1-4卤代烷基或C3-7环烷基;
    R10是H或选自以下群组:C1-4烷基、C1-4卤代烷基、C3-7环烷基、4-10元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3- 7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基;
    R11选自H、C1-4烷基以及C3-7环烷基;
    R12选自C1-6烷基、C3-7环烷基、4-至14-元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CF3、-CN、-OH、-NH2、-NH(CH3)、-N(CH3)2、氧代、-S-C1-4烷基、C1-4烷基、C1-4卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  24. 根据权利要求23所述的用途,其中L为C=O、O=S=O或CH2
  25. 根据权利要求23所述的用途,其中X为CH。
  26. 根据权利要求23-25中任一项所述的用途,其中环A为C3-7环烷基、3-7元杂环烷基、C5-7芳基、5-7元杂芳基。
  27. 根据权利要求23-25中任一项所述的用途,其中环A为5-6元杂芳基或苯基。
  28. 根据权利要求23-25中任一项所述的用途,其中存在0个或1个R1,并且R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。
  29. 根据权利要求23-25中任一项所述的用途,其中存在1个或2个R2,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  30. 根据权利要求23-25中任一项所述的用途,其中:
    L为C=O或O=S=O;
    X为CH;
    环A为5-7元杂芳基、C5-7芳基;
    R1的个数为0、1、2、3或4个,并且R1选自C1-8烷基、3-7元杂环烷基,其中所述C1-8烷基任选地被1个、2个、3个或4个R3取代,并且其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
    R2的个数为1、2或3个,并且R2选自H、卤素、-OH、-NO2、-CN、-SF5、C1-6烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-7环烷基、4-10元杂环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-N(R9)(R10)、-C(=O)-R12、-C(=O)-OR12、-OC(=O)R12、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12,其中所述C1-6烷基、C3-7环烷基以及4-10元杂环烷基各自任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CN、-OH、C1-4烷基、C1-4烷氧基、C1-4卤代烷基、C1-4卤代烷氧基、C3-6环烷基、-N(R9)(R10)、-N(R11)(C(=O)R12)、-C(=O)-OR12、-C(=O)H、-C(=O)R12、-C(=O)-N(R9)(R10)、-N(R11)(S(=O)2R12)、-S(=O)2-N(R9)(R10)、-SR12及-OR12
    R3选自卤素、氰基、C1-3烷基、羟基、C1-6烷氧基、-N(R5)(R6)、-CON(R7)(R8)或3-7元杂环烷基,其中所述3-7元杂环烷基任选地被1个、2个、3个或4个R4取代;
    R4选自卤素、C1-3烷基、羟基、C1-6烷氧基、-NH2、-NHCH3或-N(CH3)2
    R5、R6、R7、R8各自独立地为氢或C1-4烷基;
    R9选自H、C1-4烷基、C1-4卤代烷基或C3-7环烷基;
    R10是H或选自以下群组:C1-4烷基、C1-4卤代烷基、C3-7环烷基、4-10元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2、3或4个各自独立选自以下群组的取代基所取代:-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C3-7环烷基、C1-4羟烷基、-S-C1-4烷基、-C(=O)H、-C(=O)-C1-4烷基、-C(=O)-O-C1-4烷基、-C(=O)-NH2、-C(=O)-N(C1-4烷基)2、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基;
    R11选自H、C1-4烷基以及C3-7环烷基;
    R12选自C1-6烷基、C3-7环烷基、4-至14-元杂环烷基、C6-10芳基、5-10元杂芳基、(C3-7环烷基)-C1-4烷基-、(4-10元杂环烷基)-C1-4烷基-、(C6-10芳基)-C1-4烷基-以及(5-10元杂芳基)-C1-4烷基-,其中该群组内的各个选项任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-CF3、-CN、-OH、-NH2、-NH(CH3)、-N(CH3)2、氧代、-S-C1-4烷基、C1-4烷基、C1-4卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  31. 根据权利要求30所述的用途,其中环A为5-6元杂芳基或苯基。
  32. 根据权利要求30所述的用途,其中存在0个或1个R1,并且R1选自C1-6烷基、5-7元杂环烷基,其中所述C1-6烷基任选地被1或2个R3取代,并且其中所述5-7元杂环烷基任选地被1个、2个、3个或4个C1-3烷基取代。
  33. 根据权利要求30所述的用途,其中存在1个或2个R2,并且R2选自卤素、C1-6烷基以及C3-6环烷基,其中所述C1-6烷基及C3-6环烷基各任选地被1、2或3个各自独立选自以下群组的取代基所取代:卤素、-OH、-NH2、-NH(CH3)、-N(CH3)2、-CN、C1-4烷基、C1-4卤代烷基、C1-4烷氧基以及C1-4卤代烷氧基。
  34. 根据权利要求1所述的用途,其中所述化合物选自:
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-(哌啶-1-基)吡嗪-2-基)甲酮(MDI-2);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(1-甲基-1H-吡唑-4-基)甲酮(MDI-202);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(1-甲基哌啶-4-基)甲酮(MDI-203);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-204);
    (2-(6-(2-乙基-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(5-(4-甲基哌嗪-1-基)吡嗪-2-基)甲酮(MDI-205);
    5-乙基-2-氟-4-(3-(5-(苯磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-206);
    5-乙基-2-氟-4-(3-(5-(吡嗪-2基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-207);
    4-(3-(5-(环丙基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-208);
    环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233);
    4-(3-(5-(环丁基甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-210);
    环丁基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-211);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)(3-羟基环丁基)甲酮(MDI-213);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪唑-5-(1H,4H,6H)-基)(哒嗪-4-基)甲酮(MDI-214);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-吡咯并[3,4-d]咪唑-5-(1H,4H,6H)-基)(哒嗪-3-基)甲酮(MDI-215);
    (S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228);
    5-乙基-2-氟-4-(3-(5-(4-羟基环己基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-217);
    4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218);
    4-(3-(5-(环丁基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-219);
    4-(3-(5-(环戊基磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-220);
    5-乙基-2-氟-4-(3-(5-((1-甲基-1H-吡唑-4-基)甲基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-221);
    4-(3-(5-(环戊烷基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-224);
    5-乙基-2-氟-4-(3-(5-(四氢-2H-吡喃-4-基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)苯酚(MDI-225);
    1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)乙-1-酮(MDI-226);
    1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)丙-1-酮(MDI-227);
    (1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)-2-甲基丙-1-酮)(MDI-228);
    2-环丙基-1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)基)乙-1-酮(MDI-229);
    1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)-3-甲基丁-1-酮(MDI-230);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231);
    N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236);
    环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-b]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-237);
    环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-239);
    (S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-4-甲基-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-240);
    环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[4,3-c]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)甲酮(MDI-242);
    (R)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-243);
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-甲基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-246);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248);
    1-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-5-羰基)吡咯烷-3-腈(MDI-250);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(四氢呋喃-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-251);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸甲酯(MDI-252);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253);
    (S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吡唑并[3,4-b]吡啶-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-255);
    3-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)-3-氧丙腈(MDI-256);
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257);
    N-(2-氰基乙基)-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-258);
    N-环丙基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-259);
    N-环丁基-2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-260);
    (S)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-262);
    (R)-6-(2-乙基-5-氟-4-羟基苯基)-3-(5-脯氨酰基-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑(MDI-263);
    或以上任一化合物的同位素标记化合物、光学异构体、几何异构体、互变异构体或异构体混合物、或药学上可接受的盐、前体药、或代谢物。
  35. 根据权利要求1所述的用途,其中所述化合物选自:
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(5-吗啉吡嗪-2-基)甲酮(MDI-201)
    环丙基(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)吡咯并[3,4-d]咪唑-5(1H,4H,6H)-基)甲酮(MDI-1233)
    (S)-(2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(3-羟基吡咯烷-1-基)甲酮(MDI-1228)
    4-(3-(5-(环丙磺酰基)-1,4,5,6-四氢吡咯并[3,4-d]咪唑-2-基)-1H-吲唑-6-基)-5-乙基-2-氟苯酚(MDI-218)
    N-(3-氯-2-羟丙基)-2-(6-(2-乙基-5-氟-4-羟苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-1288)
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吡咯烷-1-基)甲酮(MDI-231)
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(哌啶-1-基)甲酮(MDI-233)
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(吗啉代)甲酮(MDI-234)
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-甲基哌嗪-1-基)甲酮(MDI-235)
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-基)(4-乙基哌嗪-1-基)甲酮(MDI-236)
    (2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-基)(4-羟基哌啶-1-基)甲酮(MDI-245)
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-乙基-4,6-二氢吡咯并[3,4-d]咪唑-5-(1H)-甲酰胺(MDI-247)
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N-(2-羟基乙基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-甲酰胺(MDI-248)
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酸乙酯(MDI-253)
    2-(6-(2-乙基-5-氟-4-羟基苯基)-1H-吲唑-3-基)-N,N-二甲基-4,6-二氢吡咯并[3,4-d]咪唑-5(1H)-羧酰胺(MDI-257)
    或以上任一化合物的同位素标记化合物、光学异构体、几何异构体、互变异构体或异构体混合物、或药学上可接受的盐、前体药、或代谢物。
  36. 根据权利要求1-35中任意一项所述的用途,其中所述化合物被用作JAK/TRK双重抑制剂(优选被用作pan-JAK/pan-TRK双重抑制剂)或JAK/TRK/RET多重抑制剂(优选被用作pan-JAK/pan-TRK/RET多重抑制剂)。
  37. 一种TRK抑制剂和/或RET抑制剂药物组合物,其包含如权利要求1-35中任意一项中所定义的化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,以及一种或多种药学上可接受的载体、佐剂或赋形剂。
  38. 根据权利要求37所述的药物组合物,其被配制成口服剂型或适于局部施用的外用剂型。
  39. 根据权利要求37所述的药物组合物,其被配制成JAK/TRK双重抑制剂药物(优选被用作pan-JAK/pan-TRK双重抑制剂药物)或JAK/TRK/RET多重抑制剂药物(优选被被配制成pan-JAK/pan-TRK/RET多重抑制剂药物)。
  40. 根据权利要求1-35中任意一项中所定义化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物或者根据权利要求37或38或39所述的组合物在制备用于治疗和/或预防与TRK和/或RET相关的疾病或病症的药物中的用途。
  41. 根据权利要求40所述的用途,其中所述与TRK和/或RET相关的疾病或病症选自关节炎,自身免疫疾病或病症,癌症或肿瘤,糖尿病及糖尿病并发症,(糖尿病引起的)伤口愈合缓慢、眼疾病、病症或病况,肠炎症、变态反应或病况,神经变性疾病,皮肤疾病、病况或病症,变态反应,哮喘和其它阻塞性气道疾病,移植排斥。
  42. 根据权利要求40所述的用途,其中所述与TRK和/或RET相关的疾病或病症选自瘙痒、银屑病、特应性皮炎、EGFR抑制剂引起的皮肤副作用、痤疮、白癜风、斑秃、哮喘、鼻炎、痔疮、宫颈炎、肺炎、糖尿病引起的伤口愈合缓慢、糖尿病足、糖尿病视网膜病变、癌症(肿瘤)、褥疮。
PCT/CN2023/105745 2022-07-04 2023-07-04 作为trk抑制剂和/或ret抑制剂的化合物及其用途 WO2024008088A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210777651 2022-07-04
CN202210777651.6 2022-07-04

Publications (1)

Publication Number Publication Date
WO2024008088A1 true WO2024008088A1 (zh) 2024-01-11

Family

ID=89365663

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/105745 WO2024008088A1 (zh) 2022-07-04 2023-07-04 作为trk抑制剂和/或ret抑制剂的化合物及其用途

Country Status (2)

Country Link
CN (1) CN117343063A (zh)
WO (1) WO2024008088A1 (zh)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103717599A (zh) * 2011-07-27 2014-04-09 辉瑞有限公司 吲唑
WO2017077283A1 (en) * 2015-11-03 2017-05-11 Topivert Pharma Limited 4,5,6,7-tetrahydro-1h-imidazo[4,5-c]pyridine and 1,4,5,6,7,8-hexahydroimidazo[4,5-d]azepine derivatives as janus kinase inhibitors
WO2017077288A1 (en) * 2015-11-03 2017-05-11 Topivert Pharma Limited 4,5,6,7-tetrahydro-1h-imidazo[4,5-c]pyridine and 1,4,5,6,7,8-hexahydroimidazo[4,5-d]azepine derivatives as janus kinase inhibitors
CN111606908A (zh) * 2019-02-25 2020-09-01 河南迈英诺医药科技有限公司 Jak抑制剂化合物及其用途
CN113121509A (zh) * 2019-12-30 2021-07-16 路良 Jak抑制剂化合物及其用途
CN114075200A (zh) * 2020-08-14 2022-02-22 河南迈英诺医药科技有限公司 用于治疗重症肺炎的jak抑制剂化合物
CN114524818A (zh) * 2021-01-15 2022-05-24 深圳康溯医药科技有限公司 Fgfr抑制剂化合物及其用途

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103717599A (zh) * 2011-07-27 2014-04-09 辉瑞有限公司 吲唑
WO2017077283A1 (en) * 2015-11-03 2017-05-11 Topivert Pharma Limited 4,5,6,7-tetrahydro-1h-imidazo[4,5-c]pyridine and 1,4,5,6,7,8-hexahydroimidazo[4,5-d]azepine derivatives as janus kinase inhibitors
WO2017077288A1 (en) * 2015-11-03 2017-05-11 Topivert Pharma Limited 4,5,6,7-tetrahydro-1h-imidazo[4,5-c]pyridine and 1,4,5,6,7,8-hexahydroimidazo[4,5-d]azepine derivatives as janus kinase inhibitors
CN111606908A (zh) * 2019-02-25 2020-09-01 河南迈英诺医药科技有限公司 Jak抑制剂化合物及其用途
CN113121509A (zh) * 2019-12-30 2021-07-16 路良 Jak抑制剂化合物及其用途
CN114075200A (zh) * 2020-08-14 2022-02-22 河南迈英诺医药科技有限公司 用于治疗重症肺炎的jak抑制剂化合物
CN114524818A (zh) * 2021-01-15 2022-05-24 深圳康溯医药科技有限公司 Fgfr抑制剂化合物及其用途

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"Modern Tumor Immuno-Targeted Therapy", 30 November 2010, SOUTHEAST UNIVERSITY PRESS, CN, article FANG, LIANGHUA: "Section 3, Multi-Target Small Molecule Targeted Medicine", pages: 316 - 329, XP009551738 *

Also Published As

Publication number Publication date
CN117343063A (zh) 2024-01-05

Similar Documents

Publication Publication Date Title
CN103153994B (zh) 双环杂芳基激酶抑制剂及使用方法
CN115192577B (zh) Kras突变蛋白抑制剂
JP7023243B2 (ja) イソキノリン-3イル-カルボキサミドならびにその調製および使用の方法
CN109963842B (zh) 苯并咪唑类化合物激酶抑制剂及其制备方法和应用
JP6609631B2 (ja) 縮合環ヘテロアリール化合物及びtrk抑制剤としての用途
CA3054455C (en) Fgfr inhibitor and application thereof
WO2020239077A1 (zh) 含氮杂环类衍生物调节剂、其制备方法和应用
TWI626050B (zh) 吡唑並[1,5-a]嘧啶類之醫藥組成物與用途
KR102634308B1 (ko) 피롤로피리미딘 화합물
MX2012009541A (es) Derivados de ciclobutano y metilciclobutano como inhibidores de janus cinasa.
EP2569293A1 (en) Pyrazolopyridines as inhibitors of the kinase lrrk2
CA2818545A1 (en) Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
TW201609692A (zh) 取代吡唑及其用途
JP2005089334A (ja) 8−ヒドロキシアデニン化合物
TW201623307A (zh) 作爲酪胺酸激酶抑制劑之經取代的乙炔基雜雙環化合物
WO2018127195A1 (zh) 取代的稠合杂芳基化合物作为激酶抑制剂及其应用
JP7198820B2 (ja) 置換アゼチジンジヒドロチエノピリミジンおよびホスホジエステラーゼ阻害剤としてのそれらの使用
CN114728941A (zh) 作为alk5抑制剂的经取代的1,5-萘啶或喹啉
WO2023160727A1 (zh) 吲唑类化合物用于治疗银屑病的用途
CN109937202A (zh) 作为pde2抑制剂的[1,2,4]三唑并[1,5-a]嘧啶化合物
CN109890824A (zh) 作为pde2抑制剂的[1,2,4]三唑并[1,5-a]嘧啶化合物
KR102559624B1 (ko) Fgfr4 억제제로서 사용된 융합된 환 유도체
EP4186907A1 (en) Jak inhibitor compound for treating severe pneumonia
WO2024008088A1 (zh) 作为trk抑制剂和/或ret抑制剂的化合物及其用途
CN104822658A (zh) 作为多种激酶抑制剂的稠合三环酰胺类化合物

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: 23834855

Country of ref document: EP

Kind code of ref document: A1