WO2024032774A1 - 用作电压-门控钠通道抑制剂的化合物 - Google Patents

用作电压-门控钠通道抑制剂的化合物 Download PDF

Info

Publication number
WO2024032774A1
WO2024032774A1 PCT/CN2023/112584 CN2023112584W WO2024032774A1 WO 2024032774 A1 WO2024032774 A1 WO 2024032774A1 CN 2023112584 W CN2023112584 W CN 2023112584W WO 2024032774 A1 WO2024032774 A1 WO 2024032774A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
haloalkyl
optionally substituted
deuteriums
Prior art date
Application number
PCT/CN2023/112584
Other languages
English (en)
French (fr)
Inventor
邓代国
钟文和
卢杰联
雷曾荣
Original Assignee
广州费米子科技有限责任公司
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 广州费米子科技有限责任公司 filed Critical 广州费米子科技有限责任公司
Publication of WO2024032774A1 publication Critical patent/WO2024032774A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • 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
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • 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
    • 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
    • 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
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • 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
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • 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
    • A61P27/16Otologicals
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/02Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having nitrogen atoms of carboxamide groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • C07C233/11Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having nitrogen atoms of carboxamide groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals with carbon atoms of carboxamide groups bound to carbon atoms of an unsaturated carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C381/00Compounds containing carbon and sulfur and having functional groups not covered by groups C07C301/00 - C07C337/00
    • C07C381/10Compounds containing sulfur atoms doubly-bound to nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/52Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring condensed with a ring other than six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/04Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings with only hydrogen atoms, halogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D237/20Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/08Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated

Definitions

  • the present invention relates to a compound used as a voltage-gated sodium channel inhibitor and its preparation method, pharmaceutical composition and application.
  • the compound has the structural characteristics of formula (I).
  • Nav Nav1.1-Nav1.9. Based on whether they can be effectively inhibited by nanomolar tetrodotoxin (TTX), Nav is divided into TTX-sensitive (TTX-S) and TTX-insensitive. Sensitive type (TTX-R), and the tissue expression of different subtypes is extremely different.
  • TTX-S nanomolar tetrodotoxin
  • TTX-R Sensitive type
  • Nav1.1, Nav1.2, Nav1.3 are TTX-S type
  • Nav1.4 Nav1.6 and Nav1.7 are TTX-S type, among which Nav1.1, Nav1.2 and Nav1.3 are in the central nervous system (CNS)
  • CNS central nervous system
  • Nav1.4 exists in large quantities in skeletal muscles
  • Nav1.6 and Nav1.7 mainly exist in large quantities in the central nervous system.
  • Nav1.5, Nav1.8 and Nav1.9 are TTX-R types, among which Nav1.5 mainly exists in cardiomyocytes, and Nav1.8 and Nav1.9 exist in the dorsal root ganglion (PNS-DRG) of the peripheral nervous system.
  • PNS-DRG dorsal root ganglion
  • Nav inhibitors have been proven to be effective.
  • the local anesthetic lidocaine relieves pain by inhibiting Nav.
  • Non-selective Nav inhibitors such as lamotrigine, lacosamide, and mexiletine have been successfully used to treat chronic pain.
  • Nav inhibitors currently used clinically lack subtype selectivity and can inhibit sodium ion channels expressed in the heart and central nervous system. The therapeutic window is narrow and the scope of application is limited.
  • Nav1.8 is mainly distributed in the peripheral nervous system and is limited to pain-sensing neurons, making it a highly selective target for pain treatment. Selectively inhibiting Nav1.8 has good prospects for reducing potential toxic side effects.
  • Nav1.8 is a subtype of voltage-gated sodium channels (VGSC/Nav).
  • NaV1.8 participates in the conduction and transmission process of nociceptive action potential. Because of its dependence on depolarization voltage, NaV1.8 is the main mediator in the rising stage of action potential. At the same time, NaV1.8 can quickly recover from inactivation, and it also has Contributes to repeated high frequency emissions. By inhibiting the activity of NaV1.8, the propagation of pain can be blocked, and because NaV1.8 is mainly expressed in neurons that transmit pain signals in the dorsal root ganglia (DRG) of the peripheral nervous system, selective inhibition of Nav1.8 can also Can effectively reduce potential toxic side effects.
  • DDG dorsal root ganglia
  • the present invention provides a compound useful as a voltage-gated sodium channel inhibitor. It has the activity of selectively inhibiting Nav1.8 and is therefore used to treat a variety of diseases mediated by it, especially various types of pain.
  • the present invention provides compounds of general formula (I), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, and pro-forms thereof.
  • Drug or isotopic variant :
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, Solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, and pharmaceutically acceptable excipients; preferably, they also contain other therapeutic agents.
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, as well as mixtures thereof, or the use of pharmaceutical compositions containing the same for the preparation of medicaments for use as voltage-gated sodium channel inhibitors.
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, as well as mixtures thereof, or the use of pharmaceutical compositions containing the same in the preparation of drugs for use as sodium ion channel 1.8 (NaV1.8) inhibitors.
  • NaV1.8 sodium ion channel 1.8
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, and mixtures thereof, or pharmaceutical compositions containing the same, for use as voltage-gated sodium channel inhibitors.
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, as well as mixtures thereof, or pharmaceutical compositions containing the same, for use as sodium ion channel 1.8 (NaV1.8) inhibitors.
  • NaV1.8 sodium ion channel 1.8
  • the invention provides a method of inhibiting voltage-gated sodium channels in a subject, comprising administering to said subject a compound as defined in the invention or a pharmaceutically acceptable version thereof Salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, or pharmaceutical compositions containing the same .
  • the invention provides a method of inhibiting sodium ion channel 1.8 (NaV1.8) in a subject, comprising administering to the subject a compound as defined in the invention or a pharmaceutical agent thereof.
  • a method of inhibiting sodium ion channel 1.8 (NaV1.8) in a subject comprising administering to the subject a compound as defined in the invention or a pharmaceutical agent thereof.
  • Voltage-gated sodium channel inhibitors are used to treat a disease selected from the group consisting of acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headache, trigeminal neuralgia, herpes Neuralgia, general neuralgia, epilepsy or epileptic conditions, neurodegenerative diseases, psychiatric disorders such as anxiety and depression, bipolar disorder, myotonia, cardiac arrhythmias, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis , irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, post-herpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, headache, neck pain, severe or intractable pain, injury Pain, penetrating pain, post-surgical pain, cancer pain, stroke, cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, stress or exercise-induced angina, heart palpitations, high blood pressure, migraine, or
  • the method relates to a disease selected from the group consisting of radicular pain, sciatica, back pain, headache, neck pain, intractable pain, acute pain, post-surgical pain, back pain, tinnitus, or cancer pain.
  • the elements involved in the groups and compounds of the present invention include carbon, hydrogen, oxygen, sulfur, nitrogen or halogen, including their isotopic conditions. condition. Furthermore, the elements carbon, hydrogen, oxygen, sulfur or nitrogen involved in the groups and compounds of the present invention are optionally further replaced by one or more of their corresponding isotopes, wherein the isotopes of carbon include 12 C, 13 C and 14 C.
  • Isotopes of hydrogen include protium (H), deuterium (D, also called heavy hydrogen), and tritium (T, also called superheavy hydrogen).
  • Isotopes of oxygen include 16 O, 17 O, and 18 O.
  • Isotopes of sulfur include 32 S, 33 S, 34 S and 36 S, nitrogen isotopes include 14 N and 15 N, fluorine isotopes include 19 F, chlorine isotopes include 35 Cl, 36 Cl and 37 Cl, bromine isotopes include 79 Br and 81 Br .
  • alkyl refers to a saturated linear or branched aliphatic hydrocarbon group, specifically a saturated hydrocarbon containing primary (normal) carbon atoms, secondary carbon atoms, tertiary carbon atoms, quaternary carbon atoms or combinations thereof. Phrases containing this term, for example, "C 1-6 alkyl” refer to alkyl groups containing 1 to 6 carbon atoms, and each occurrence may be independently C 1 alkyl, C 2 alkyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, C 6 alkyl.
  • an alkyl group containing 1 to 20 carbon atoms may be included, preferably an alkyl group containing 1 to 10 carbon atoms, more preferably an alkyl group containing 1 to 6 carbon atoms or 1 to 4 carbon atoms. of lower alkyl.
  • Non-limiting examples of alkyl groups include: methyl, ethyl, 1-propyl, 2-propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1-butyl (n- Bu, n-butyl, -CH 2 CH 2 CH 2 CH 3 ), 2-methyl-prop-1-yl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2 -Butyl (s-Bu, sec-butyl, -CH(CH 3 )CH 2 CH 3 ), 2-methyl-prop-2-yl (t-Bu, tert-butyl, -C(CH 3 ) 3 ), 1-pentyl (n-pentyl, -CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH 2 CH 3 ) 2 ), 2-methyl-but-2-yl (-C(CH 3 )
  • the alkyl group can be substituted or unsubstituted.
  • alkenyl is an alkyl group as defined herein that contains at least one carbon-carbon double bond.
  • the alkenyl group contains 2 to 20 carbon atoms, preferably 2 to 12 carbon atoms, further preferably 2 to 8 carbon atoms, even more preferably 2 to 6 carbon atoms.
  • alkenyl groups include substituted or unsubstituted vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3-nonenyl or 4-decenel, etc.
  • Alkynyl as defined herein is an alkyl group containing at least one carbon-carbon triple bond.
  • the alkynyl group contains 2 to 20 carbon atoms, preferably 2 to 12 carbon atoms, further preferably 2 to 8 carbon atoms, and even more preferably 2 to 6 carbon atoms.
  • Cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic carbon-containing group.
  • the cycloalkyl group is a 3- to 6-membered (C 3-6 ) monocyclic ring, a 3 to 8-membered (C 3-8 ) monocyclic ring, a 3 to 10-membered (C 3-10 ) monocyclic ring.
  • Carbon rings include bridged rings or spiro rings.
  • Non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclopentenyl, cyclohexadienyl, cycloheptatrienyl, and the like .
  • Heterocyclyl or “heterocycle” refers to a substituted or unsubstituted saturated or partially unsaturated monocyclic or polycyclic cyclic group containing heteroatoms selected from N, O and S.
  • the heterocyclyl group may be a 3- to 8-membered monocyclic ring, a 4- to 12-membered bicyclic ring, or a 10- to 15-membered tricyclic ring system.
  • heterocyclic or bicyclic heterocyclyl group It is preferably a 3 to 10-membered monocyclic or bicyclic heterocyclyl group, more preferably a 5 to 8-membered monocyclic or bicyclic heterocyclyl group, and contains at least one, preferably 1 to 4, 1 to 3 or 1 to 2 heteroatoms selected from N, O or S. If present, heteroatoms N or S in the heterocycle can be oxidized to various oxidation states to form, for example, N-oxides. Heterocycles can be connected to other parts of the molecule through heteroatoms or carbon atoms. Heterocyclic rings include bridged rings or spiro rings.
  • Non-limiting examples of monocyclic heterocycles include ethylene oxide, azetidinyl, oxetanyl, azetidinyl, 1,3-dioxolanyl, dihydrofuranyl , tetrahydrofuranyl, tetrahydropyrrolyl, tetrahydroimidazolyl, tetrahydrothiazolyl, dithiolanyl, oxanyl, thiacyclohexyl, azepanyl, azepanyl, diaza Cycloheptyl, morpholinyl, thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, dihydropyridyl, tetrahydrothiophenyl, sulfur-oxidized tetrahydrothiophenyl, etc.
  • Non-limiting examples of bicyclic heterocyclyl groups include benzodihydrofuran, tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolyl, octahydrocyclopentapyrrolyl, octahydropyrrolopyrrole base, octahydropentadienyl, etc.
  • Aryl refers to a substituted or unsubstituted all-carbon monocyclic or condensed polycyclic unsaturated group with a conjugated ⁇ electron system.
  • the aryl group is a 6 to 14 membered aromatic ring, preferably a 6 to 10 membered aromatic ring.
  • Non-limiting examples thereof include phenyl or naphthyl; the aryl group may be fused with a heteroaryl, heterocyclyl or cycloalkyl group, and the point of attachment to the moiety of the molecule is on the aryl group.
  • Non-limiting examples of aryl groups include benzene.
  • Heteroaryl refers to a monocyclic or fused polycyclic unsaturated group that is substituted or unsubstituted and contains at least one heteroatom selected from N, O and S.
  • the heteroaryl group is a 5 to 15 membered heteroaryl ring, a 5 to 14 membered heteroaryl ring, or preferably a 5 to 10 membered heteroaryl ring, or more preferably a 5 to 6 membered heteroaryl group, wherein The number of heteroatoms is 1 to 4, preferably 1 to 3, more preferably 1 to 2.
  • heteroaryl groups include pyrrolyl, furyl, thienyl, N-alkylpyrrolyl, imidazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzofuran, benzimidazole, benzopyridine or pyrrolopyridine etc.
  • Halogen refers to F, Cl, Br or I. "Halo” means replacing one or more hydrogen atoms in a molecule or group with a halogen selected from F, Cl, Br or I.
  • linking substituents are described.
  • the Markush variables listed for that group should be understood as referring to the linking group.
  • the alkyl group represents a linked alkylene group, for example, the group "-C 1 -C 3 haloalkyl”
  • the alkyl group in should be understood as an alkylene group.
  • “Pharmaceutically acceptable salts” refer to salts of pharmaceutically acceptable non-toxic acids or bases, including salts with inorganic acids or inorganic bases or salts with organic acids and organic bases.
  • Salts derived from inorganic bases include, but are not limited to, metal salts formed with Al, Ca, Li, Mg, K, Na and Zn; salts derived from organic bases include, but are not limited to, salts formed with primary, secondary or tertiary amines Salt.
  • the primary, secondary or tertiary amines include naturally occurring substituted or unsubstituted amines, cyclic amines and basic ion exchange resins such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine , tripropylamine, diethanolamine, ethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, caffeine, procaine, choline, betaine, Penicillin, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purine, piperazine, piperidine, N-ethylpiperidine or polyamine resin; derived from inorganic Salts of acids and organic acids include, but are not limited to, salts with the following acids: sulfuric acid, phosphoric acid, nitric acid, hydrobromic acid, hydrochloric acid, formic acid, ace
  • solvate can also be called “solvent compound” or “solvate”, which refers to a compound containing solvent molecules, where the solvent molecules can include coordination bonds, covalent bonds, van der Waals forces, ionic bonds, hydrogen Combined with compound molecules through bonds and other means.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, etc.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates.
  • the solvate will be able to isolate, for example, when one or more solvent molecules are incorporated into the crystal lattice of the crystalline solid.
  • “Solvate” includes solvates in solution and isolatable solvates. Representative solvates include hydrates, ethanolates, and methoxides.
  • hydrate refers to a compound combined with water. Typically, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined.
  • a hydrate of a compound may be represented, for example, by the general formula R.xH2O , where R is the compound and x is a number greater than zero.
  • a given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1), for example, hemihydrate (R ⁇ 0.5H 2 O)) and polyhydrates (x is a number greater than 1, for example, dihydrate (R ⁇ 2H 2 O) and hexahydrate (R ⁇ 6H 2 O)).
  • monohydrate x is 1
  • lower hydrate x is a number greater than 0 and less than 1
  • hemihydrate R ⁇ 0.5H 2 O
  • polyhydrates x is a number greater than 1, for example, dihydrate (R ⁇ 2H 2 O) and hexahydrate (R ⁇ 6H 2 O)
  • prodrug refers to any compound that when administered to an organism produces a drug, ie, an active ingredient, as a result of a spontaneous chemical reaction, an enzyme-catalyzed chemical reaction, a photolytic and/or a metabolic chemical reaction.
  • Prodrugs are thus covalently modified analogs or potential forms of therapeutically active compounds. Suitable examples include, but are not limited to: carboxylic acid esters, carbonic acid esters, phosphate esters, nitrate esters, sulfate esters, sulfone esters, sulfoxide esters, amino compounds, carbamates, azo compounds, phosphoramides, glucosides , ether, acetal and other forms.
  • the present invention also includes isotopically labeled compounds (isotopic variants) which are equivalent to those of general formula or specific compounds described herein, but with one or more atoms having an atomic mass or mass number different from that commonly seen in nature. Replaced by number of atoms.
  • isotopes to be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 respectively. O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl, preferably 2 H (ie deuterium, D).
  • isotopically labeled compounds of the present invention and their prodrugs can generally be prepared by substituting readily available isotopically labeled reagents for non-isotopically labeled reagents when performing the processes disclosed in the following schemes and/or examples and preparation examples.
  • the compounds of the present invention contain one or more asymmetric centers and thus may exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the present invention may be individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, Includes racemic mixtures and mixtures enriched in one or more stereoisomers.
  • the isomers may be separated from the mixture by methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or the preferred isomers may be separated by Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • aryl optionally substituted by alkyl means that alkyl may but need not be present, and the term includes both cases where aryl is substituted with alkyl and cases where aryl is not substituted by alkyl.
  • “Pharmaceutically acceptable excipient” refers to a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
  • pharmaceutically acceptable excipient includes buffers compatible with pharmaceutical administration, sterile water for injection, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic agents And absorption delaying agents and the like. Each excipient must be “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the patient.
  • Suitable examples include, but are not limited to: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch, potato starch and substituted or unsubstituted ⁇ -cyclodextrin; (3) cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) Powdered tragacanth; (5) Malt; (6) Gelatin; (7) Talc; (8) Fu excipients, such as cocoa butter and suppository wax; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols Alcohols, such as glycerol, sorbitol, mannitol and polyethylene glycol; (12) Esters, such as ethyl oleate and ethyl laurate; (13) Agar; (14) Bu
  • polymorph refers to a crystalline form of a compound (or a salt, hydrate or solvate thereof) in a specific crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms often have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors can lead to the dominance of one crystalline form. Various polymorphs of the compounds can be prepared by crystallization under different conditions.
  • the present invention uses traditional methods such as mass spectrometry and nuclear magnetism to identify compounds.
  • Each step and condition can refer to the conventional operating steps and conditions in the art.
  • this invention employs standard nomenclature and standard laboratory procedures and techniques of analytical chemistry, synthetic organic chemistry, and optics. In some cases, standard techniques are used for chemical synthesis, chemical analysis, and performance testing of light-emitting devices.
  • the dosage form and administration mode of the compound of the present invention or its composition are not particularly limited.
  • Representative modes of administration include, but are not limited to, oral, intratumoral, rectal, parenteral (intravenous, intraperitoneal, intramuscular or subcutaneous) injection and/or topical administration.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) filler Materials or compatibilizers, such as starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, such as hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and arabic Gum; (c) humectants, such as glycerol; (d) disintegrants, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, complex silicates and sodium carbonate; (e) solvents, such as paraffin ; (f) absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glyceryl
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • liquid dosage forms may contain inert diluents (such as water or other solvents), solubilizers and emulsifiers conventionally employed in the art. Specific examples are, for example, ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide and oils, in particular cottonseed oil, peanut oil, corn germ oil, olive oil oil, castor oil and sesame oil or a mixture of these substances.
  • compositions may also contain adjuvants such as wetting agents, suspending agents, sweetening, flavoring and perfuming agents.
  • the suspension may contain a suspending agent.
  • ethoxylated isostearyl alcohol polyoxyethylene sorbitol and sorbitan esters
  • microcrystalline cellulose aluminum methoxide and agar or mixtures thereof.
  • compositions for parenteral injection may contain physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions, or emulsions, as well as sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous or non-aqueous carriers, diluents, solvents or excipients are selected from water, ethanol and polyols, or suitable mixtures thereof.
  • Dosage forms for topical administration include ointments, powders, patches, sprays, and inhalants. It consists of the active ingredient mixed under sterile conditions with a pharmaceutically acceptable carrier and a preservative, buffer and/or propellant as may be required.
  • the present invention relates to the following embodiments.
  • the present invention relates to compounds of general formula (I'), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof substance, polymorph, prodrug or isotopic variant:
  • X is N or CR x ;
  • Y is N or CR y ;
  • Z is N or CR z ;
  • W is N or CR w ;
  • G is N or CR g ;
  • Rx , Ry and Rz are each independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, - OR s1 and -L 1 -R 1 , where at least one of R x , Ry and R z is -L 1 -R 1 , said group optionally substituted with one or more deuteriums until fully deuterated ;
  • Rw and Rg are each independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl and -OR s1 , The group is optionally substituted with one or more deuteriums, until fully deuterated;
  • -L 1 - is -(CR a R b ) m -, and if the chemical valence allows, any methylene unit in -(CR a R b ) m - is optionally and independently replaced by -NR'-, -O-, -S- and -C(O)-substitute and/or R a and R b together with the carbon atom to which they are attached form a C 3-8 cycloalkyl or 3-10 membered heterocyclyl group;
  • A is selected from C 3-8 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, which is optionally substituted by s R 2 substituents;
  • R' is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and C 1-6 haloalkyl, which group is optionally substituted with one or more deuterium, until Completely deuterated;
  • R” is -L 3 -B
  • B is selected from C 3-8 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, which is optionally substituted by t R 5 substituents;
  • -L 3 - is -(CR e R f ) p -, and if the chemical valence allows, any methylene unit in -(CR e R f ) p - is optionally and independently replaced by -NR'-, -O-, -S- and -C(O)-substitute and/or R e and R f together with the carbon atom to which they are attached form a C 3-8 cycloalkyl or 3-10 membered heterocyclyl group;
  • R 2 , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1- 6 Haloalkyl, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the group is optionally substituted by one or more deuteriums until fully deuterated;
  • R 4 and R 4 ' are each independently selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and C 1-6 haloalkyl, which group is optionally replaced by a Or multiple deuterium substitutions, until complete deuterium substitution;
  • R 5 is each independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the groups are optionally replaced by One or more deuterium substitutions, up to complete deuterium substitution;
  • R 6 is selected from the group consisting of H, C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R a , R b , R c , R d , Re and R f are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1 and R s2 are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, which groups are optionally substituted by one or more deuteriums until completely deuterated;
  • n 0, 1, 2, 3, 4, 5 or 6;
  • n 0, 1, 2, 3, 4, 5 or 6;
  • p 0, 1, 2, 3, 4, 5 or 6;
  • t is 1, 2, 3, 4 or 5;
  • s is 1, 2, 3, 4, 5, 6, 7 or 8;
  • q 1 or 2.
  • the present invention relates to compounds of general formula (I), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof , polymorph, prodrug or isotopic variant:
  • X is N or CR x ;
  • Y is N or CR y ;
  • Z is N or CR z ;
  • W is N or CR w ;
  • G is N or CR g ;
  • Rx , Ry and Rz are each independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, - OR s1 and -L 1 -R 1 , where at least one of R x , Ry and R z is -L 1 -R 1 , said group optionally substituted with one or more deuteriums until fully deuterated ;
  • Rw and Rg are each independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl and -OR s1 , The group is optionally substituted with one or more deuteriums, until fully deuterated;
  • -L 1 - is -(CR a R b ) m -, and if the chemical valence allows, any methylene unit in -(CR a R b ) m - is optionally and independently replaced by -NR'-, -O-, -S- and -C(O)-substitute and/or R a and R b together with the carbon atom to which they are attached form a C 3-8 cycloalkyl or 3-10 membered heterocyclyl group;
  • A is selected from C 3-8 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, which is optionally substituted by s R 2 substituents;
  • R' is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and C 1-6 haloalkyl, which group is optionally substituted with one or more deuterium, until Completely deuterated;
  • R” is -L 3 -B
  • B is selected from C 3-8 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, which is optionally substituted by t R 5 substituents;
  • -L 3 - is -(CR e R f ) p -, and if the chemical valence allows, any methylene unit in -(CR e R f ) p - is optionally and independently replaced by -NR'-, -O-, -S- and -C(O)-substitute and/or R e and R f together with the carbon atom to which they are attached form a C 3-8 cycloalkyl or 3-10 membered heterocyclyl group;
  • R 2 , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1- 6 Haloalkyl, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the group is optionally substituted by one or more deuteriums until fully deuterated;
  • R 4 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and C 1-6 haloalkyl, which group is optionally substituted with one or more deuterium, until Completely deuterated;
  • R 5 is each independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the groups are optionally replaced by One or more deuterium substitutions, up to complete deuterium substitution;
  • R 6 is selected from the group consisting of H, C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R a , R b , R c , R d , Re and R f are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1 and R s2 are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, which groups are optionally substituted by one or more deuteriums until completely deuterated;
  • n 0, 1, 2, 3, 4, 5 or 6;
  • n 0, 1, 2, 3, 4, 5 or 6;
  • p 0, 1, 2, 3, 4, 5 or 6;
  • t is 1, 2, 3, 4 or 5;
  • s is 1, 2, 3, 4, 5, 6, 7 or 8;
  • q 1 or 2.
  • the present invention relates to the compound of formula (I) or (I') above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate thereof substance, hydrate, polymorph, prodrug or isotopic variant of formula (II),
  • the present invention relates to the compound of formula (I) or formula (II) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate thereof substance, hydrate, polymorph, prodrug or isotopic variant, wherein -L 2 - is -O-.
  • the present invention relates to the compound of formula (I) or formula (II) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate thereof substance, hydrate, polymorph, prodrug or isotopic variant, wherein -L2- is a bond.
  • the present invention relates to the above-mentioned compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, Prodrug or isotopic variant, wherein A is selected from phenyl, C 3-6 cycloalkyl, 5-6 membered heteroaryl and 5-10 membered heterocyclyl.
  • the present invention relates to the above-mentioned compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, Prodrug or isotopic variant, wherein A is selected from cyclopentyl, cyclohexyl, azepanyl, oxanyl, thiacyclohexyl, azepanyl, diazepanyl, octahydropentyl Dienopyrrolyl, octahydropyrrolopyrrolyl, octahydropentacyclopentadienyl, phenyl, pyrrolyl, furyl, thienyl, pyridyl, pyrimidinyl and pyridazinyl.
  • the present invention relates to the above-mentioned compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, Prodrug or isotopic variant, where m is 0.
  • the present invention relates to the above-mentioned compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, Prodrugs or isotopic variants, where:
  • n 1, 2, 3, 4 or 5
  • Any methylene unit in -(CR a R b ) m - is optionally and independently replaced by -O-, -S- and -C(O)-; and/or R a and R b are different from the ones they replace.
  • the connected carbon atoms together form C 3-6 cycloalkyl and 3-5 membered heterocyclyl;
  • R a is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R b is selected from H, C 1-6 alkyl and C 1-6 haloalkyl.
  • the present invention relates to the compound of formula (I) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of formula (III),
  • the present invention relates to the compound of formula (I) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of formula (IV)
  • X is N or CR x ;
  • Y is N or CR y ;
  • R x and R y are each independently selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted with one or more deuteriums until fully deuterated;
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R” is -L 3 -B
  • B is a 5-7 membered heterocyclyl group, which is optionally substituted by t R 5 substituents;
  • -L 3 - is -(CR e R f ) p -, and if the chemical valence allows, any methylene unit in -(CR e R f ) p - is optionally and independently replaced by -O-, - S-and-C(O)-substitution;
  • R 2a , R 2b , R 2c , R 2d , R 2e , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkene group, C 2-6 alkynyl group, C 1-6 haloalkyl group, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O) NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the groups optionally substituted by one or more deuteriums until completely deuterated;
  • R 5 is each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally substituted with one or more deuteriums until completely deuterated;
  • R e and R f are independently selected from H, D, C 1-6 alkyl and C 1-6 haloalkyl, which groups are optionally substituted with one or more deuteriums until fully deuterated;
  • R s1 and R s2 are independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, and the group is optionally substituted by one or more deuterium until completely deuterated;
  • p 0, 1, 2 or 3;
  • t 1, 2 or 3.
  • the present invention relates to the compound of formula (IV) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R” is -L 3 -B
  • B is a 5-6 membered heterocyclyl group, which is optionally substituted by t R 5 substituents;
  • R 2a , R 2b , R 2c , R 2d , R 2e , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, The group is optionally substituted with one or more deuteriums, until fully deuterated;
  • R 5 is each independently selected from H, D, C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally substituted with one or more deuteriums until completely deuterated;
  • R e and R f are independently selected from H, D, C 1-6 alkyl and C 1-6 haloalkyl, which groups are optionally substituted with one or more deuteriums until completely deuterated;
  • p 0, 1, 2 or 3;
  • t 1, 2 or 3;
  • -L 3 -B is selected from
  • the present invention relates to the compound of formula (IV) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N
  • R x is H or D
  • R’ is H
  • R” is -L 3 -B
  • B is a 5-6 membered heterocyclyl group, preferably azepanyl or azepanyl;
  • -L 3 - is -(CR e R f ) p -, p is 0 or 1;
  • R 2a is C 1-6 alkyl, preferably methyl
  • R 2b is H or D
  • R 2c is halogen, preferably F
  • R 2d is H or D
  • R 2e is H or D
  • R 3a is H or D
  • R 3b is C 1-6 haloalkyl, preferably -CF 3 ;
  • R 3c is halogen, preferably Cl
  • R 3d is H or D
  • R e is selected from H, D and C 1-6 alkyl
  • R f is selected from H, D and C 1-6 alkyl
  • -L 3 -B is selected from
  • the present invention relates to the compound of formula (I) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of formula (V)
  • the present invention relates to the compound of formula (V) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is N or CR x ;
  • Y is N or CR y ;
  • R x and R y are each independently selected from H, D, halogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl and -OR s1 , The group is optionally substituted with one or more deuteriums, until fully deuterated;
  • -L 2 - is a bond, -O-, -S- or -NH-;
  • A is selected from phenyl, C 3-8 cycloalkyl, 5-6 membered heteroaryl and 5-10 membered heterocyclyl, which is optionally substituted by s R 2 substituents;
  • any methylene unit in -(CR a R b ) m - is optionally and independently replaced by -O-, -S- and -C(O)-;
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R 2 , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1- 6 Haloalkyl, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the group is optionally substituted by one or more deuteriums until fully deuterated;
  • R 4 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R a and R b are selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R a and R b together with the carbon atoms to which they are connected form C 3-6 cycloalkyl and 3-5 membered heterocyclyl;
  • R s1 and R s2 are independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • n 0, 1, 2, 3 or 4;
  • s is 1, 2, 3, 4, 5, 6, 7 or 8.
  • the present invention relates to the compound of formula (IV) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x and R y are each independently selected from H, D, halogen and C 1-6 alkyl, which group is optionally substituted with one or more deuteriums until fully deuterated;
  • A is selected from And 5-8 membered heterocyclyl, which is optionally substituted by s R 2 substituents;
  • any methylene unit in -(CR a R b ) m - is optionally and independently replaced by -O-;
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R 2 is each independently selected from H, D, halogen, C 1-6 alkyl, C 1-6 haloalkyl and -OR s1 , which group is optionally substituted with one or more deuteriums until fully deuterated ;
  • R 2a is selected from H, D, halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR s1a and -SR s1a , which group is optionally substituted by one or more deuteriums until completely deuterated generation;
  • R 2b is H or D
  • R 2c is H, D, halogen, -OR s1c or -SR s1c ;
  • R 2d is H or D
  • R 2e is H or D
  • R 3a is H or D
  • R 3b is C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3c is halogen
  • R 3d is H or D
  • R 4 is selected from C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R a and R b are selected from H, D, halogen and C 1-6 alkyl, which groups are optionally substituted by one or more deuteriums until completely deuterated;
  • R a and R b together with the carbon atom to which they are attached form a C 3-6 cycloalkyl group
  • Each R s1 is independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1a is selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1c is selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • n 1, 2 or 3;
  • s 1 or 2.
  • the present invention relates to the compound of formula (IV) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x is H
  • R y is halogen
  • A is selected from and azepanyl, which is substituted by s R 2 ;
  • R’ is H
  • R 2 is each independently selected from H and halogen is preferably F, and said group is optionally substituted by one or more deuteriums until fully deuterated;
  • R 2a is H or C 1-6 alkyl, preferably methyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 2b is H
  • R 2c is halogen or -OR s1c ;
  • R 2d is H
  • R 2e is H
  • R 3a is H
  • R 3b is C 1-6 haloalkyl, preferably -CF 3 ;
  • R 3c is halogen, preferably Cl
  • R 3d is H
  • R 4 is C 1-6 alkyl, preferably methyl
  • R s1c is H, C 1-6 alkyl or C 1-6 haloalkyl, preferably -CF 3 ;
  • s 1 or 2.
  • the present invention relates to the compound of formula (I) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of formula (VI):
  • the present invention relates to the compound of formula (VI) above, or a pharmaceutically acceptable salt, enantiomer, non-enantiomer thereof Enantiomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, of which:
  • X is N or CR x ;
  • Y is N or CR y ;
  • R x and R y are each independently selected from H, D, halogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl and -OR s1 , The group is optionally substituted with one or more deuteriums, until fully deuterated;
  • -L 2 - is a bond, -O-, -S- or -NH-;
  • A is selected from phenyl, C 3-8 cycloalkyl, 5-10 membered heteroaryl and 5-10 membered heterocyclyl, which is optionally substituted by s R 2 substituents;
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R 2 , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1- 6 Haloalkyl, OR s1 , SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the group is optionally substituted by one or more deuteriums, until fully deuterated;
  • R 4 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R s1 and R s2 are independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, and the group is optionally substituted by one or more deuterium until completely deuterated;
  • s is 1, 2, 3, 4, 5, 6, 7 or 8.
  • the present invention relates to the above-mentioned compound of formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • A is selected from cyclopentyl, cyclohexyl, azepanyl, oxanyl, thiacyclohexyl, azepanyl, diazepanyl, octahydropentapyrrolyl, octahydrogen Pyrrolopyrrolyl, octahydropentadienyl, phenyl, pyrrolyl, furyl, thienyl, pyridyl, pyrimidinyl and pyridazinyl, preferably, A and its substituents are selected from the following:
  • R 2a , R 2b , R 2c and R 2d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 Haloalkyl, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O)NR s1 R s2 , -SOR s1 and -SO 2 R s1 , said group is optionally substituted by one or more deuteriums, until fully deuterated.
  • the present invention relates to the above-mentioned compound of formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • A is selected from cyclopentyl and cyclohexyl, preferably cyclohexyl.
  • the present invention relates to the above-mentioned compound of formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • A is selected from the group consisting of azepanyl, diazepanyl, octahydropentapyrrolyl, octahydropyrrolopyrrolyl and octahydropentapyrrolyl.
  • A is nitrogen-containing In the case of a heterocyclic ring, it is connected to the phenyl group through a nitrogen atom.
  • the present invention relates to the compound of formula (I) above, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of formula (VII)
  • X is N or CR x ;
  • Y is N or CR y ;
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R 2a , R 2b , R 2c , R 2d , R 2e , R 3a , R 3b , R 3c and R 3d are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 2-6 alkene group, C 2-6 alkynyl group, C 1-6 haloalkyl group, -OR s1 , -SR s1 , NR s1 R s2 , -C(O)R s1 , -C(O)OR s1 , -C(O) NR s1 R s2 , -SOR s1 and -SO 2 R s1 , the groups optionally substituted by one or more deuteriums until completely deuterated;
  • R 4 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R s1 and R s2 are independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which groups are optionally substituted with one or more deuteriums until fully deuterated.
  • the present invention relates to the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is N or CR x ;
  • Y is N or CR y ;
  • R x and R y are each independently selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted with one or more deuteriums until fully deuterated;
  • R' is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R 2a is selected from H, D, halogen, -OR s1a , -SR s1a , NR s1a R s2a , C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally replaced by one or more deuterium Substitution, until complete deuteration;
  • R 2b is H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 2c is selected from H, D, halogen, -OR s1c , -SR s1c , NR s1c R s2c , C 1-6 alkyl and C 1-6 haloalkyl, which group is optionally replaced by one or more deuterium Substitution, until complete deuteration;
  • R 2d is H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 2e is H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3a is H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3b is selected from H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R 3c is selected from H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3d is H, D, halogen, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 4 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl, the group is optionally substituted by one or more deuterium until completely deuterated;
  • R s1a , R s2a , R s1c and R s2c are each independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted with one or more deuterium, up to complete deuterium generation.
  • the present invention relates to the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x and R y are independently selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted with one or more deuteriums until fully deuterated;
  • R' is H, C 1-6 alkyl or C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 2a is -OR s1a , -SR s1a , NR s1a R s2a , C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 2b is H, D or halogen
  • R 2c is H, D, halogen, -OR s1c , -SR s1c or NR s1c R s2c , said group optionally substituted with one or more deuteriums until fully deuterated;
  • R 2d is H, D or halogen
  • R 2e is H, D or halogen
  • R 3a is H, D or halogen
  • R 3b is H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3c is H, D or halogen
  • R 3d is H, D or halogen
  • R 4 is H, C 1-6 alkyl or C 1-6 haloalkyl, the group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1a , R s2a , R s1c and R s2c are each independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted with one or more deuterium, up to complete deuterium generation;
  • the proviso is that when Y is N, R 2a is not C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention relates to the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x and R y are independently selected from H, D or halogen
  • R' is H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 2a is -OR s1a , -SR s1a , C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 2b is H, D or halogen
  • R 2c is H, D, halogen, -OR s1c or -SR s1c , said group optionally substituted with one or more deuteriums until fully deuterated;
  • R 2d is H or D
  • R 2e is H or D
  • R 3a is H or D
  • R 3b is C 1-6 haloalkyl
  • R 3c is halogen
  • R 3d is H or D
  • R 4 is C 1-6 alkyl or C 1-6 haloalkyl
  • R s1a and R s1c are independently selected from C 1-6 alkyl or C 1-6 haloalkyl;
  • the proviso is that when Y is N, R 2a is not C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention relates to the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x and R y are independently selected from H, D or halogen
  • R’ is H
  • R 2a is -OR s1a , -SR s1a or C 1-6 alkyl, which group is optionally substituted with one or more deuteriums until completely deuterated;
  • R 2b is H or D
  • R 2c is halogen, -OR s1c or -SR s1c , said group optionally substituted with one or more deuteriums until fully deuterated;
  • R 2d is H or D
  • R 2e is H or D
  • R 3a is H or D
  • R 3b is C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3c is halogen
  • R 3d is H or D
  • R 4 is C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1a and R s1c are each independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, which groups are optionally substituted with one or more deuteriums until fully deuterated.
  • the present invention relates to the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x is H or D
  • R y is H, D or halogen
  • R’ is H
  • R 2a is -OR s1a or -SR s1a , said group optionally substituted with one or more deuteriums until fully deuterated;
  • R 2b is H or D
  • R 2c is halogen, -OR s1c or -SR s1c ;
  • R 2d is H or D
  • R 2e is H or D
  • R 3a is H or D
  • R 3b is C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R 3c is halogen
  • R 3d is H or D
  • R 4 is C 1-6 alkyl or C 1-6 haloalkyl, which group is optionally substituted by one or more deuteriums until completely deuterated;
  • R s1a and R s1c are independently selected from C 1-6 alkyl or C 1-6 haloalkyl, which groups are optionally substituted with one or more deuteriums until fully deuterated.
  • the present invention relates to the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants of which:
  • X is CR x ;
  • Y is N or CR y ;
  • R x is H
  • R y is halogen, preferably F
  • R’ is H
  • R 2a is -OR s1a , the group is optionally substituted by one or more deuteriums until completely deuterated, preferably -OCD 3 ;
  • R 2b is H
  • R 2c is halogen or -OR s1c , preferably F or -OCF 3 ;
  • R 2d is H
  • R 2e is H
  • R 3a is H
  • R 3b is C 1-6 haloalkyl, preferably -CF 3 ;
  • R 3c is halogen, preferably Cl
  • R 3d is H
  • R 4 is C 1-6 alkyl
  • R s1a is C 1-6 alkyl
  • R s1c is C 1-6 haloalkyl.
  • the compound is selected from the following structures:
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate thereof , solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, and pharmaceutically acceptable excipients; preferably, they also contain other therapeutic agents.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, Solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, and pharmaceutically acceptable excipients; preferably, they also contain other therapeutic agents.
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, as well as mixtures thereof, or the use of pharmaceutical compositions containing the same for the preparation of medicaments for use as voltage-gated sodium channel inhibitors.
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, as well as mixtures thereof, or the use of pharmaceutical compositions containing the same in the preparation of drugs for use as sodium ion channel 1.8 (NaV1.8) inhibitors.
  • NaV1.8 sodium ion channel 1.8
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, and mixtures thereof, or pharmaceutical compositions containing the same, for use as voltage-gated sodium channel inhibitors.
  • the invention provides a compound as defined in the invention or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , polymorphs, prodrugs or isotopic variants, as well as mixtures thereof, or pharmaceutical compositions containing the same, for use as sodium ion channel 1.8 (NaV1.8) inhibitors.
  • NaV1.8 sodium ion channel 1.8
  • the invention provides a method of inhibiting voltage-gated sodium channels in a subject, comprising administering to said subject a compound as defined in the invention or a pharmaceutically acceptable version thereof Salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, or pharmaceutical compositions containing the same .
  • the invention provides a method of inhibiting sodium ion channel 1.8 (NaV1.8) in a subject, comprising administering to the subject a compound as defined in the invention or a pharmaceutical agent thereof.
  • a method of inhibiting sodium ion channel 1.8 (NaV1.8) in a subject comprising administering to the subject a compound as defined in the invention or a pharmaceutical agent thereof.
  • Voltage-gated sodium channel inhibitors are used to treat a disease selected from the group consisting of acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headache, trigeminal neuralgia, herpes Neuralgia, general neuralgia, epilepsy or epileptic conditions, neurodegenerative diseases, psychiatric disorders such as anxiety and depression, bipolar disorder, myotonia, cardiac arrhythmias, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis , irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, post-herpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, headache, neck pain, severe or intractable pain, injury Pain, penetrating pain, post-surgical pain, cancer pain, stroke, cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, stress or exercise-induced angina, heart palpitations, high blood pressure, migraine, or
  • the method relates to a disease selected from the group consisting of radicular pain, sciatica, back pain, headache, neck pain, intractable pain, acute pain, post-surgical pain, back pain, tinnitus, or cancer pain.
  • Step 4 N-(2-(benzylthio)pyridin-4-yl)-5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl) Benzamide
  • Step 5 4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine-2-sulfonyl chloride
  • Step 6 (2-((4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine)-2- Sulfonamido)ethyl)(methyl)carbamate tert-butyl ester
  • Step 7 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(2-(methylamino))ethyl)sulfamoyl)pyridine-4 -yl)-4-(trifluoromethyl)benzamide (compound 1)
  • Step 1 (R)-3-((4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)-benzamido)pyridine )-2-Sulfonylamino)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 2 (R)-5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(pyrrolidin-3-yl)sulfamoyl)pyridine- 4-yl)-4-(trifluoromethyl)benzamide (compound 2)
  • Step 1 (S)-3-((4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine) -2-Sulfonylamino)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 2 (S)-5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(pyrrolidin-3-yl)sulfamoyl)pyridine- 4-yl)-4-(trifluoromethyl)benzamide (compound 3)
  • Step 1 4-((4-(5-chloro-2-(4fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine)-2-sulfonyl Amino)piperidine-1-carboxylic acid tert-butyl ester
  • 4-aminopiperidine-1-carbamic acid tert-butyl ester (76.55mg, 0.38mmol) was dissolved in pyridine (2mL), cooled to 0°C, and 4-(5-chloro-2-( 4-Fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine-2-sulfonyl chloride (200 mg, 0.38 mmol) was dissolved in dichloromethane (4 mL) and added dropwise to In the reaction solution, the reaction was maintained at this temperature and stirred for 2 hours.
  • Step 2 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(piperidin-4-yl)sulfamoyl)pyridin-4-yl) -4-(Trifluoromethyl)benzamide (Compound 4)
  • Step 1 4-(((4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine)-2- Sulfonylamino)methyl)piperidine-1-carboxylic acid tert-butyl ester
  • Step 2 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(piperidin-4-ylmethyl)sulfamoyl)pyridine-4- methyl)-4-(trifluoromethyl)benzamide (compound 5)
  • Step 1 (R)-3-(((4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine )-2-Sulfonylamino)methyl)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 2 (R)-5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(pyrrolidin-3-ylmethyl)sulfamoyl) Pyridin-4-yl)-4-(trifluoromethyl)benzamide (compound 6)
  • Step 1 (S)-3-(((4-(5-chloro-2-(4-fluoro-2-methylphenoxy)-4-(trifluoromethyl)benzamido)pyridine )-2-Sulfonylamino)methyl)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 2 (S)-5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(N-(pyrrolidin-3-ylmethyl)sulfamoyl) Pyridin-4-yl)-4-(trifluoromethyl)benzamide (Compound 7)
  • reaction solution was concentrated under reduced pressure to obtain a crude product; the crude product was separated and purified by high-performance liquid chromatography (elution system: formic acid, water, acetonitrile) to obtain the title compound (27.16 mg, yield: 28.1%, white solid) .
  • reaction solution was concentrated under reduced pressure to obtain a crude product, which was separated and purified by high-performance liquid chromatography (elution system: formic acid, water, acetonitrile) to obtain the title compound (27.36 mg, yield: 25.5%, white solid).
  • Step 4 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(S-methylimidosulfonyl)pyridin-4-yl)-4-( Trifluoromethyl)benzamide (compound 13)
  • Step 3 5-chloro-2-(4-fluoro-2-(methoxy-d 3 )phenoxy)-N-(2-(methylthio)pyridin-4-yl)-4-( Trifluoromethyl)benzamide
  • Step 4 5-chloro-2-(4-fluoro-2-(methoxy-d 3 )phenoxy)-N-(2-(S-methylimidosulfonyl)pyridine-4- methyl)-4-(trifluoromethyl)benzamide (compound 14)
  • Compound 14 was separated by a chiral preparation column (chromatographic column: 250*25mm, 10 ⁇ m; mobile phase: A-Supercritical CO 2 ; B-MEOH (+0.1% 7.0mol/l Ammonia in MEOH), 50% B ratio elution, flow rate: 100mL/min, column temperature: room temperature) separation Single configurations of compound 14E1 (shorter retention time) and compound 14E2 (longer retention time) were obtained.
  • Step 1 5-chloro-2-(2-(methoxy-d 3 )-4-(trifluoromethoxy)phenoxy)-N-(2-(methylthio)pyridine-4- methyl)-4-(trifluoromethyl)benzamide
  • Step 2 5-chloro-2-(2-(methoxy-d 3 )-4-(trifluoromethoxy)phenoxy)-N-(2-(S-methyl iminosulfonate) Acyl)pyridin-4-yl)-4-(trifluoromethyl)benzamide (compound 15)
  • Step 1 5-chloro-N-(2-(methylthio)pyridin-4-yl)-2-(4-(trifluoromethoxy)phenoxy)-4-(trifluoromethyl) Benzamide
  • Step 2 5-chloro-N-(2-(S-methylimidosulfonyl)pyridin-4-yl)-2-(4-(trifluoromethoxy)phenoxy)-4- (Trifluoromethyl)benzamide (compound 16)
  • Step 1 5-Chloro-2-(4,4-difluoroazepan-1-yl)-N-(2-(methylthio)pyridin-4-yl)-4-trifluoromethyl )benzamide
  • Compound 17 was separated by a chiral preparation column (chromatographic column: 250*25mm, 10 ⁇ m; mobile phase: A-Supercritical CO 2 ; B-MEOH (+0.1% 7.0mol/l Ammonia in MEOH), 25% B ratio elution, flow rate: 70mL/min, column temperature: room temperature) separation Single configurations of compound 17E1 (shorter retention time) and compound 17E2 (longer retention time) were obtained.
  • Step 1 5-chloro-2-((3aR,6aS)-5,5-difluorohexahydrocyclopenta[c]pyrrole-2(1H)-yl)-N-(2-(methyl Thio)pyridin-4-yl)-4-(trifluoromethyl)benzamide
  • Step 2 5-chloro-2-((3aR,6aS)-5,5-difluorohexahydrocyclopenta[c]pyrrole-2(1H)-yl)-N-(2-(S -methylimidosulfonyl)pyridin-4-yl)-4-(trifluoromethyl)benzamide (compound 18)
  • Step 1 5-chloro-2-((4,4-difluorocyclohexyl)oxy)-N-(2-(methylthio)pyridin-4-yl)-4-(trifluoromethyl) Benzamide
  • Step 3 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(6-(methylthio)pyridazin-4-yl)-4-(trifluoromethyl) Benzamide
  • Step 4 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(6-(S-methylimidosulfonyl)pyridazin-4-yl)-4- (Trifluoromethyl)benzamide (Compound 20)
  • Step 1 5-Chloro-N-(6-(methylthio)pyridazin-4-yl)-2-(4-(trifluoromethoxy)phenoxy)-4-(trifluoromethyl )benzamide
  • the first step 5-chloro-2-(4,4-difluoroazepan-1-yl)-N-(6-(methylthio)pyridazin-4-yl)-4-(trifluoro Methyl)benzamide
  • Step 3 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(4-fluoro-3-(methylthio)phenyl)-4-(trifluoromethyl) Benzamide
  • Step 4 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(4-fluoro-3-(S-methylimidosulfonyl)phenyl)-4- (Trifluoromethyl)benzamide (Compound 23)
  • Compound 23 was separated by a chiral preparation column (chromatographic column: 250*25mm, 10 ⁇ m; mobile phase: A-Supercritical CO2; B-MEOH (+0.1% 7.0mol/l Ammonia in MEOH), 30% B ratio elution, flow rate: 70mL/min, column temperature: room temperature) separated Single configuration compound 23E1 (shorter retention time) and compound 23E2 (longer retention time).
  • Step 1 5-chloro-2-(4-fluoro-2-(methoxy-d3)phenoxy)-N-(4-fluoro-3-(methylthio)phenyl)-4-( Trifluoromethyl)benzamide
  • Step 2 5-chloro-2-(4-fluoro-2-(methoxy-d 3 )phenoxy)-N-(4-fluoro-3-(S-methyliminosulfonyl) Phenyl)-4-(trifluoromethyl)benzamide (compound 24)
  • Step 1 5-chloro-N-(4-fluoro-3-(methylthio)phenyl)-2-(2-(methoxy-d 3 )-4-(trifluoromethoxy)benzene Oxy)-4-(trifluoromethyl)benzamide
  • Step 2 5-chloro-N-(4-fluoro-3-(S-methylimidosulfonyl)phenyl)-2-(2-(methoxy-d 3 )-4-(tri Fluoromethoxy)phenoxy)-4-(trifluoromethyl)benzamide (compound 25)
  • Step 1 5-chloro-N-(4-fluoro-3-(methylthio)phenyl)-2-(4-(trifluoromethoxy)phenoxy)-4-(trifluoromethyl) )benzamide
  • the first step 5-chloro-2-(4,4-difluoroazepan-1-yl)-N-(4-fluoro-3-(methylthio)phenyl)-4-(tris Fluoromethyl)benzamide
  • Compound 27 was separated by a chiral preparation column (chromatographic column: 250*25mm, 10 ⁇ m; mobile phase: A-Supercritical CO 2 ; B-MEOH (+0.1% 7.0mol/l Ammonia in MEOH), 45% B ratio elution, flow rate: 70mL/min, column temperature: room temperature) separation Single configurations of compound 27E1 (shorter retention time) and compound 27E2 (longer retention time) were obtained.
  • Step 1 5-chloro-2-((3aR,6aS)-5,5-difluorohexahydrocyclopenta[c]pyrrole-2(1H)-yl)-N-(4-fluoro- 3-(Methylthio)phenyl)-4-(trifluoromethyl)benzamide
  • Step 2 5-chloro-2-((3aR,6aS)-5,5-difluorohexahydrocyclopenta[c]pyrrole-2(1H)-yl)-N-(4-fluoro- 3-(S-methylimidosulfonyl)phenyl)-4-(trifluoromethyl)benzamide (compound 28)
  • Step 1 5-chloro-2-((4,4-difluorocyclohexyl)oxy)-N-(4-fluoro-3-(methylthio)phenyl)-4-(trifluoromethyl) )benzamide
  • Step 2 5-chloro-2-((4,4-difluorocyclohexyl)oxy)-N-(4-fluoro-3-(S-methylimidosulfonyl)phenyl)-4 -(Trifluoromethyl)benzamide (compound 29)
  • Step 5 N-(((4-bromopyridin-2-yl)methyl)(methyl)(oxo)- ⁇ 6 -sulfylidene)-2,2,2-trifluoroacetamide
  • Step 6 ((4-bromopyridin-2-yl)methyl)(imino)(methyl)- ⁇ 6 -sulfoxide
  • Step 7 (((4-bromopyridin-2-yl)methyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamate benzyl ester
  • Step 8 (1-(4-bromopyridin-2-yl)cyclopropyl)(imino)(methyl)- ⁇ 6 -sulfoxide
  • Step 9 ((1-(4-bromopyridin-2-yl)cyclopropyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamate benzyl ester
  • Step 10 ((1-(4-((diphenylmethylene)amino)pyridin-2-yl)cyclopropyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamic acid Benzyl ester
  • Step 11 ((1-(4-Aminopyridin-2-yl)cyclopropyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamate benzyl ester
  • Step 12 ((1-(4-(5-chloro-2-fluoro-4-(trifluoromethyl)benzoylamino)pyridin-2-yl)cyclopropyl)(methyl)(oxygen Generation)- ⁇ 6 -sulfanylidene) carbamate benzyl ester
  • Step 14 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(1-(S-methylimidosulfonyl)cyclopropyl)pyridine- 4-yl)-4-(trifluoromethyl)benzamide (compound 30)
  • Step 1 ((1-(4-(5-chloro-2-(4-(trifluoromethoxy)phenoxy))-4-(trifluoromethyl)benzoylamino)pyridine-2- (methyl)cyclopropyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamate benzyl ester
  • Step 2 5-chloro-N-(2-(1-(S-methylimidosulfonyl)cyclopropyl)pyridin-4-yl)-2-(4-(trifluoromethoxy) Phenoxy)-4-(trifluoromethyl)benzamide (Compound 31)
  • Step 1 ((1-(4-(5-chloro-2-(4,4-difluoroazepan-1-yl)-4-(trifluoromethyl)benzoylamino)pyridine- 2-yl)cyclopropyl)(methyl)(oxo)- ⁇ 6 -sulfylidene)carbamic acid benzyl ester
  • Step 2 5-chloro-2-(4,4-difluoroazepan-1-yl)-N-(2-(1-(S-methylimidosulfonyl)cyclopropyl) Pyridin-4-yl)-4-(trifluoromethyl)benzamide (compound 32)
  • Step 2 (2-((4-bromopyridin-2-yl)oxy)ethyl)(imino)(methyl)- ⁇ 6 -sulfoxide
  • Step 3 ((2-((4-bromopyridin-2-yl)oxy)ethyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamate benzyl ester
  • Step 4 ((2-((4-((diphenylmethylene)amino)pyridin-2-yl)oxy)ethyl)(methyl)(oxo)- ⁇ 6 - Benzyl sulfide) carbamate
  • Step 5 ((2-((4-Aminopyridin-2-yl)oxy)ethyl)(methyl)(oxo)- ⁇ 6 -sulfanylidene)carbamate benzyl ester
  • Step 6 ((2-((4-(5-chloro-2-fluoro-4-(trifluoromethyl)benzamide)pyridin-2-yl)oxy)ethyl)(methyl)( Oxo)- ⁇ 6 -thio)carbamate benzyl ester
  • Step 7 ((2-((4-(5-chloro-2-(4-fluoro-2-methylphenoxy))-4-(trifluoromethyl)benzamide)pyridin-2-yl )oxy)ethyl)(methyl)(oxo)- ⁇ 6 -sulfylidene)carbamate benzyl ester
  • Step 8 5-chloro-2-(4-fluoro-2-methylphenoxy)-N-(2-(2-(S-methylimidosulfonyl)ethoxy)pyridine-4 -yl)-4-(trifluoromethyl)benzamide (compound 33)
  • the first step 2-(4-acetyl-1,4-diazepan-1-yl)-5-chloro-N-(2-methylthio)pyridin-4-yl)-4- Trifluoromethylbenzamide
  • Step 2 4-acetyl-1,4-diazacyclopent-5-chloro-N-(4-fluoro-3--S-methanesulfonylimidophenyl)-4-trifluoromethyl Benzamide (compound 35)
  • Step 1 4-(4-chloro-2-((2-(methylthio)pyridin-4-yl)carbamoyl)-5-(trifluoromethyl)phenyl)-1,4-di Aza-1-carboxylic acid tert-butyl ester
  • Step 3 5-chloro-2-(4-(2,2-difluoroethyl)-1,4-diaza-1-yl)-N-(2-(methylthio)pyridine-4 -yl)-4-(trifluoromethyl)benzamide
  • Step 4 5-chloro-2-(4-(2,2-difluoroethyl)-1,4-diaza-1-yl)-N-(2-(S-methylsulfonyl) Amino)pyridin-4-yl)-4-(trifluoromethyl)benzamide
  • the first step 5-chloro-N-2-methylthiopyridin-4-yl-2-(2,2,2-trifluoroethyl)-1,4-diazacyclopentan-1-yl )-4-Trifluoromethylbenzamide
  • Step 2 5-chloro-N-(2-methanesulfonylimido)pyridin-4-yl)-2-(4-(2,2,2-trifluoroethyl)-1,4-di Azocyclopentan-1-yl)-4-trifluoromethylbenzamide (compound 37)
  • Step 2 (4-aminopyridin-2-yl)imino)dimethyl- ⁇ 6 -sulfonamide
  • Step 3 5-chloro-N-(2-((dimethyl(oxo)- ⁇ 6 -sulfonamiylidene)amino)pyridin-4-yl)-2-fluoro-4-(trifluoromethyl )benzamide
  • Step 4 5-chloro-N-(2-((dimethyl(oxo)- ⁇ 6 -thio)amino)pyridin-4-yl)-2-(4-fluoro-2-methyl Phenoxy)-4-(trifluoromethyl)benzamide (compound 38)
  • reaction was stopped, the reaction solution was filtered, and then purified by high-performance liquid chromatography (elution system: ammonium bicarbonate, water, acetonitrile) to obtain the title compound (38.25 mg, yield: 22.9%, white solid).
  • Test Example A Blocking activity of the compound of the present invention on sodium ion channel 1.8 (NaV1.8)
  • microelectrode drawing instrument to draw the capillary glass tube into recording electrodes.
  • drug administration is started. Each drug concentration is activated for 5 minutes (or the current reaches stability) before the next concentration is detected. Two concentrations of each test compound are detected.
  • the voltage stimulation protocol for whole-cell patch clamp recording of Nav1.8 sodium current is as follows: after the whole-cell seal is formed, the cell voltage is clamped at -120mV for 30ms. The clamping voltage is depolarized to 0mV for 50ms, and then the voltage returns to -50mV (refer to the half-deactivation voltage of the IV test for the specific voltage) and maintained for 8s. Then the cell membrane potential returned to -120mV, maintained for 20ms, then depolarized to 0mV, maintained for 50ms, and finally returned to the clamping voltage -120mV, maintained for 30ms. Data collection is repeated every 20 seconds. Observe the effect of the drug on the sodium current peak. The test data was collected by the EPC-10 amplifier (HEKA) and stored in the PatchMaster (HEKA) software.
  • HEKA EPC-10 amplifier
  • HEKA PatchMaster
  • Test Example B Pharmacokinetic evaluation of the compound of the present invention by intravenous injection or intragastric administration in rats
  • the purpose of this test example is to evaluate the pharmacokinetic properties of the compound of the present invention.
  • the inventors performed a pharmacokinetic evaluation of the compounds of the invention in rats. Among them, animal information is detailed in Table 2.
  • Table 2 Information table of test animals of the present invention
  • the compound of the present invention was administered to the test animals in the form of 5% DMSO + 5% tween80 + 90% physiological saline solution.
  • the animals fasted for 12 hours before administration and drank water freely.
  • the dosage is 1 mg/kg; for intragastric administration, the dosage is 10 mg/kg.
  • blood was collected from the veins at the following time points (the blood volume was about 0.3 mL): 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 8.0 and 24 hours.
  • EDTA-K2 was pre-added to the blood collection tube as an anticoagulant, and the blood samples were Centrifuge at low temperature for 6 minutes at 6800 g, collect plasma, and store at -80°C.
  • Sample preparation for LC-MS/MS determination 20 ⁇ L plasma sample was subjected to protein precipitation with 400 ⁇ L methanol containing 100 ng/mL IS (IS is tolbutamide). The mixture was vortexed for 1 min and then centrifuged at 18000 g for 7 min. Transfer 400 ⁇ L of supernatant to a 96-well plate. 2 ⁇ L of the supernatant was subjected to LC-MS/MS analysis. The analysis results show that the compounds of the present invention have good pharmacokinetic properties in rats.
  • IS IS is tolbutamide
  • Test Example C Pharmacokinetic evaluation of the compound of the present invention by intravenous or intragastric administration in mice
  • the purpose of this test example is to evaluate the pharmacokinetic properties of the compound of the present invention.
  • mice The inventors performed a pharmacokinetic evaluation of the compounds of the invention in mice. Among them, detailed animal information is shown in Table 3.
  • Table 3 Information table of test animals of the present invention
  • the compound of the present invention was administered to the test animals in the form of 5% DMSO + 5% tween80 + 90% physiological saline solvent.
  • the animals fasted for 12 hours before administration and drank water freely.
  • the dosage is 1 mg/kg; for intragastric administration, the dosage is 10 mg/kg.
  • blood was collected from the veins at the following time points (the blood volume was about 0.3 mL): 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 8.0 and 24 hours.
  • EDTA-K2 was pre-added to the blood collection tube as an anticoagulant, and the blood samples were Centrifuge at low temperature for 6 minutes at 6800 g, collect plasma, and store at -80°C.
  • Sample preparation for LC-MS/MS determination 15 ⁇ L plasma sample was subjected to protein precipitation with 300 ⁇ L methanol containing 100 ng/mL IS (IS is tolbutamide). The mixture was vortexed for 1 min and then centrifuged at 18000 g for 10 min. Transfer 300 ⁇ L of supernatant to a 96-well plate. 2 ⁇ L of the supernatant was subjected to LC-MS/MS analysis. The analysis results show that the compounds of the present invention have good pharmacokinetic properties in mice.
  • IS IS is tolbutamide

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Rheumatology (AREA)
  • Psychology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Anesthesiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Transplantation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Pyridine Compounds (AREA)

Abstract

提供了一种用作电压-门控钠通道抑制剂的化合物及其制备方法、药物组合物和应用。所述化合物具有式(I)结构特征。

Description

用作电压-门控钠通道抑制剂的化合物 技术领域
本发明涉及一种用作电压-门控钠通道抑制剂的化合物及其制备方法、药物组合物和应用。所述化合物具有式(I)结构特征。
背景技术
人类Nav主要有9种亚型,分别为Nav1.1-Nav1.9,根据能否被纳摩尔河豚毒素(tetrodotoxin,TTX)有效抑制,Nav被分为TTX敏感型(TTX-S)和TTX不敏感型(TTX-R),且不同亚型的组织表达极其不同。
Nav1.1、Nav1.2、Nav1.3为、Nav1.4、Nav1.6和Nav1.7为TTX-S型,其中Nav1.1、Nav1.2和Nav1.3在中枢神经***(CNS)中大量表达,Nav1.4在骨骼肌中大量存在,Nav1.6和Nav1.7主要在中枢神经***中大量存在。
Nav1.5、Nav1.8和Nav1.9为TTX-R型,其中Nav1.5主要存在于心肌细胞中,Nav1.8、Nav1.9存在于外周神经***背根神经节(PNS-DRG)。
表1 Nav各亚型信息
研究表明,离子通道改变是炎症或神经病理损伤后外周敏化、中枢敏化及脱抑制作用的分子基础,也是疼痛发生的重要分子机制。目前Nav抑制剂已被证明有效,如局部***利多卡因通过抑制Nav来止痛,非选择性的Nav抑制剂例如拉莫三嗪、拉科酰胺、美西律已经成功地用于治疗慢性疼痛。但现有临床中使用的Nav抑制剂由于缺乏亚型选择性,能够抑制表达在心脏和中枢神经***中的钠离子通道,治疗窗口较窄,应用范围受到限制。
高选择性Nav抑制剂为电压门控钠离子通道研发重点方向之一,Nav1.8因主要分布在外周神经***、局限于感受疼痛的神经元,是疼痛治疗的高选择性作用靶点,所以选择性地抑制Nav1.8对降低潜在毒副作用上具有较好的前景。
Nav1.8是电压门控钠离子通道(voltage-gated sodium channels,VGSC/Nav)的一种亚型。
NaV1.8参与伤害感受动作电位的传导和传递过程,NaV1.8因其对去极化电压依赖性,是动作电位上升阶段的主要介质,同时NaV1.8因能迅速从失活中恢复,也有助于重复的高频发射。通过抑制NaV1.8的活性,能够阻断痛觉的传播,且因NaV1.8主要表达在外周神经***背根神经节(DRG)中传递痛觉信号的神经元中,选择性地抑制Nav1.8也可以有效地减少潜在毒副作用。
目前需要活性更优的作为电压-门控钠通道抑制剂尤其是选择性地抑制Nav1.8的药物,从而在疾病的治疗中提供治疗益处。
发明内容
基于此,本发明提供了一种用作电压-门控钠通道抑制剂的化合物。其具有选择性抑制Nav1.8的活性,并因此用于治疗多种由其介导的疾病,尤其是各种疼痛。
本发明提供通式(I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体:
其中,各取代基如本发明中所定义。
在一个实施方案中,本发明提供药物组合物,其包含如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物在制备用作电压-门控钠通道抑制剂的药物中的用途。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物在制备用作钠离子通道1.8(NaV1.8)抑制剂的药物中的用途。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物,其用作电压-门控钠通道抑制剂。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物,其用作钠离子通道1.8(NaV1.8)抑制剂。
在一个实施方案中,本发明提供一种在受试者中抑制电压-门控钠通道的方法,包括向所述受试者给药如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物。
在一个实施方案中,本发明提供一种在受试者中抑制钠离子通道1.8(NaV1.8)的方法,包括向所述受试者给药如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物。
如本发明所述的电压-门控钠通道抑制剂用于治疗选自以下的疾病:急性、慢性、神经性、或炎性疼痛、关节炎、偏头痛、丛集性头痛、三叉神经痛、疱疹性神经痛、一般性神经痛、癫痫或癫痫病症、神经变性疾病、精神障碍如焦虑和抑郁、双相性精神障碍、肌強直、心律失常、运动障碍、神经内分泌障碍、共济失调、多发性硬化、肠易激综合征、失禁、内脏痛、骨关节炎痛、疱瘆后神经痛、糖尿病性神经病、神经根痛、坐骨神经痛、背痛、头痛、颈痛、严重或顽固性疼痛、伤害性疼痛、贯穿性疼痛、手术后疼痛、癌症疼痛、中风、脑缺血、外伤性脑损伤、肌萎缩性侧索硬化、应激或运动诱发的心绞痛、心悸、高血压、偏头痛或异常的胃肠活动。
在一些实施方案中,本方法涉及的疾病选自神经根痛、坐骨神经痛、背痛、头痛、颈痛、顽固性疼痛、急性疼痛、手术后疼痛、背痛、耳鸣或癌症疼痛。
具体实施方式
定义
以下结合具体实施例对本发明的化合物及其制备方法、药物组合物和应用作进一步详细的说明。本发明可以以许多不同的形式来实现,并不限于本文所描述的实施方式。相反地,提供这些实施方式的目的是使人们对本发明公开内容理解更加透彻全面。
除非另有定义,本文使用的所有技术和科学术语具有本领域技术人员通常理解的含义。本文在说明书中所使用的术语只是为了描述具体的实施方案的目的,不旨在于限制本发明。本文所使用的术语“和/或”意指一个或多个相关的所列项目中的任意一种和所有项目的组合。
除非有相反的陈述,在说明书和权利要求书中使用的术语具有下述含义。
本发明所述基团和化合物中所涉及的元素包括碳、氢、氧、硫、氮或卤素均包括它们的同位素情 况。进一步,本发明所述基团和化合物中所涉及的元素碳、氢、氧、硫或氮任选进一步被一个或多个它们对应的同位素所替代,其中碳的同位素包括12C、13C和14C,氢的同位素包括氕(H)、氘(D,又叫重氢)、氚(T,又叫超重氢),氧的同位素包括16O、17O和18O,硫的同位素包括32S、33S、34S和36S,氮的同位素包括14N和15N,氟的同位素包括19F,氯的同位素包括35Cl、36Cl和37Cl,溴的同位素包括79Br和81Br。
术语“烷基”是指饱和的直链或支链脂肪族烃基团,具体为包含伯(正)碳原子、仲碳原子、叔碳原子、季碳原子或其组合的饱和烃。包含该术语的短语,例如,“C1-6烷基”是指包含1至6个碳原子的烷基,每次出现时,可以互相独立地为C1烷基、C2烷基、C3烷基、C4烷基、C5烷基、C6烷基。在其中一个实施方案中,可以包括含有1至20个碳原子的烷基,优选含有1至10个碳原子的烷基,更优选的是含有1至6个碳原子或1至4个碳原子的低级烷基。烷基的非限制性实例包括:甲基、乙基、1-丙基、2-丙基(i-Pr、i-丙基、-CH(CH3)2)、1-丁基(n-Bu、n-丁基、-CH2CH2CH2CH3)、2-甲基-丙-1-基(i-Bu、i-丁基、-CH2CH(CH3)2)、2-丁基(s-Bu、仲丁基、-CH(CH3)CH2CH3)、2-甲基-丙-2-基(t-Bu、叔丁基、-C(CH3)3)、1-戊基(n-戊基、-CH2CH2CH2CH2CH3)、2-戊基(-CH(CH3)CH2CH2CH3)、3-戊基(-CH(CH2CH3)2)、2-甲基-丁-2-基(-C(CH3)2CH2CH3)、3-甲基-丁-2-基(-CH(CH3)CH(CH3)2)、3-甲基-丁-1-基(-CH2CH2CH(CH3)2)、2-甲基-丁-1-基(-CH2CH(CH3)CH2CH3)、1-己基(-CH2CH2CH2CH2CH2CH3)、己-2-基(-CH(CH3)CH2CH2CH2CH3)、己-3-基(-CH(CH2CH3)(CH2CH2CH3))、2-甲基-戊-2-基(-C(CH3)2CH2CH2CH3)、3-甲基-戊-2-基(-CH(CH3)CH(CH3)CH2CH3)、4-甲基-戊-2-基(-CH(CH3)CH2CH(CH3)2)、3-甲基-戊-3-基(-C(CH3)(CH2CH3)2)、2-甲基-戊-3-基(-CH(CH2CH3)CH(CH3)2)、2,3-二甲基-丁-2-基(-C(CH3)2CH(CH3)2)、3,3-二甲基-丁-2-基(-CH(CH3)C(CH3)3、辛基(-(CH2)7CH3)和正壬基,及其各种支链异构体。所述烷基可以是取代的或未取代的,当烷基被取代时,除非另有定义,取代基优选为1至5个,并且取代基独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷氧基、羟基、硝基、氰基和氨基。
“烯基”是本发明定义的烷基中包含至少一个碳-碳双键。在其中一个实例中,所述烯基含有2至20个碳原子,优选2至12个碳原子,进一步优选2至8个碳原子,更进一步优选为2至6个碳原子。烯基的非限定实例包括取代或未取代的乙烯基、2-丙烯基、3-丁烯基、2-丁烯基、4-戊烯基、3-戊烯基、2-己烯基、3-己烯基、2-庚烯基、3-庚烯基、4-庚烯基、3-辛烯基、3-壬烯基或4-癸烯基等。当烯基被取代时,除非另有定义,取代基优选为1至5个,并且取代基独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷氧基、羟基、硝基、氰基和氨基。
“炔基”是本发明定义的烷基中包含至少一个碳-碳叁键。在其中一个实例中,所述炔基含有2至20个碳原子,优选2至12个碳原子,进一步优选2至8个碳原子,更进一步优选2-6个碳原子。炔基的非限定实例包括取代或未取代的乙炔基、1-丙炔基、2-丙炔基、1-丁炔基、2-丁炔基、3-丁炔基、4-戊炔基、3-戊炔基、2-己炔基、3-己炔基、3-丁炔基、2-庚炔基、3-庚炔基、4-庚炔基、3-辛炔基、3-壬炔基或4-癸炔基等,当炔基被取代时,除非另有定义,取代基优选为1至5个,并且取代基独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷氧基、羟基、硝基、氰基和氨基。
“环烷基”是指饱和或者部分不饱和的单环或多环环状含碳基团。在其中一个实施方案中,环烷基是3至6元(C3-6)的单环、3至8元(C3-8)的单环、3至10元(C3-10)的单环、4至12元双环(C4-12)或者10至15元(C10-15)三环***。碳环包括桥环或者螺环。环烷基的非限制性实例包括环丙基、环丁基、环戊基、环己基、环己烯基、环庚基、环戊烯基、环己二烯基、环庚三烯基等。环烷基可以任选被取代。当环烷基被取代时,除非另有定义,取代基优选为1至5个,并且所述取代基独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷氧基、羟基、硝基、氰基和氨基。
“杂环基”或“杂环”是指取代的或未取代的饱和或者部分不饱和的含杂原子的单环或多环环状基团,所述杂原子选自N、O和S。在其中一个实施方案中,所述杂环基可以是3至8元的单环、4至12元双环或者10至15元三环***。优选为3至10元的单环或双环杂环基,更优选为5至8元的单环或双环杂环基,且包含至少一个,优选1至4个、1至3个或1至2个选自N、O或S的杂原子。如果存在,杂环中的杂原子N或S可被氧化成各种氧化态形成例如N-氧化物。杂环可以通过杂原子或者碳原子与分子的其它部分相连。杂环包括桥环或者螺环。单环杂环的非限制性实例包括,环氧乙烷、氮杂环丙基、氧杂环丁烷基、氮杂环丁烷基、1,3-二氧戊环基、二氢呋喃基、四氢呋喃基、四氢吡咯基、四氢咪唑基、四氢噻唑基、二硫杂戊环基、氧杂环己基、硫杂环己基、氮杂环己基、氮杂环庚基、二氮杂环庚基、吗啉基、硫吗啉基、二氢吡喃基、四氢吡喃基、二氢吡啶基、四氢噻吩基、硫氧化的四氢噻吩基等。双环杂环基的非限制性实例包括苯并二氢呋喃、四氢喹啉基、四氢异喹啉基、二氢吲哚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基、八氢并环戊二烯基等。当杂环基被取代时,除非另有定义,取代基优选为1至5个并且独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷 氧基、羟基、硝基、氰基和氨基。
“芳基”是指取代的或未取代全碳单环或稠和多环不饱和基团,具有共轭的π电子体系。在其中一个实施方案中,芳基为6至14元芳香环,优选6至10元芳香环。其非限定性实例包括苯基或萘基;所述芳基可以与杂芳基、杂环基或环烷基稠和,且与分子其与部分连接的部位在芳基上。芳基的非限定性实例包括苯。当芳基被取代时,除非另有定义,取代基优选为1至5个,并且取代基独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷氧基、羟基、硝基、氰基和氨基。
“杂芳基”是指取代或未取代的且含有至少一个选自N、O和S杂原子的单环或稠和多环不饱和基团。在其中一个实施方案中,杂芳基为5至15元杂芳环、5至14元杂芳基,或优选5至10元杂芳环,或更优选为5至6元杂芳基,其中杂原子的数量为1至4个,优选为1至3个,更优选为1至2个。杂芳基的非限制性实例包括吡咯基、呋喃基、噻吩基、N-烷基吡咯基、咪唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、苯并呋喃、苯并咪唑、苯并吡啶或吡咯并吡啶等。当杂芳基被取代时,除非另有定义,取代基优选为1至5个,并且取代基独立地选自F、Cl、Br、I、=O、烷基、烯基、炔基、烷氧基、羟基、硝基、氰基和氨基。
“卤素”是指F、Cl、Br或I。“卤代”是指用选自F、Cl、Br或I的卤素替换分子或基团中的一个或多个氢原子。
在本发明的各部分,描述了连接取代基。当该结构清楚地需要连接基团时,针对该基团所列举的马库什变量应理解为连接基团。例如,如果该结构需要连接基团并且针对该变量的马库什基团定义列举了“烷基”或“芳基”,则应该理解,该“烷基”或“芳基”分别代表连接的亚烷基基团或亚芳基基团。在一些具体的结构中,当烷基基团清楚地表示为连接基团时,则该烷基基团代表连接的亚烷基基团,例如,基团“-C1-C3卤代烷基”中的烷基应当理解为亚烷基。
“药学上可接受的盐”是指药学上可接受的无毒酸或碱的盐,包括与无机酸或无机碱形成的盐或与有机酸和有机碱形成的盐。衍生自无机碱的盐包括但不限于与Al、Ca、Li、Mg、K、Na和Zn形成的金属盐;衍生自有机碱的盐包括但不限于与伯胺、仲胺或叔胺形成的盐。所述伯胺、仲胺或叔胺包括天然存在的取代或未取代的胺、环胺和碱性离子交换树脂,例如氨、异丙基胺、三甲基胺、二乙胺、三乙胺、三丙基胺、二乙醇胺、乙醇胺、二甲基乙醇胺、2-二甲基氨基乙醇、2-二乙基氨基乙醇、二环己基胺、咖啡碱、普鲁卡因、胆碱、甜菜碱、苯明青霉素、乙二胺、葡萄糖胺、甲基葡糖胺、可可碱、三乙醇胺、氨丁三醇、嘌呤、哌嗪、哌啶、N-乙基哌啶或聚胺树脂;衍生自无机酸和有机酸的盐包括但不限于与以下酸形成的盐:硫酸、磷酸、硝酸、氢溴酸、盐酸、甲酸、乙酸、丙酸、苯磺酸、苯甲酸、苯乙酸、水杨酸、褐藻酸、氨茴酸、樟脑酸、柠檬酸、乙烯磺酸、蚁酸、富马酸、糠酸、葡萄糖酸、葡萄糖醛酸、谷氨酸、乙醇酸、羟乙磺酸、乳酸、马来酸、苹果酸、扁桃酸、粘液酸、双羟萘酸、泛酸、硬脂酸、琥珀酸、磺胺酸、酒石酸、对甲苯磺酸、丙二酸、2-羟基丙酸、草酸、羟乙酸、葡萄糖醛酸、半乳糖醛酸、枸橼酸、赖氨酸、精氨酸、门冬氨酸、肉桂酸、对甲苯磺酸、甲磺酸、乙磺酸或三氟甲磺酸等。
术语“溶剂合物”也可以称为“溶剂化合物”、“溶剂化物”,指的是含有溶剂分子的化合物,其中溶剂分子可以以包括配位键、共价键、范德华力、离子键、氢键等方式与化合物分子相结合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、***等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂化物包括药学上可接受的溶剂化物且进一步包括化学计量的溶剂化物和非化学计量的溶剂化物。在一些情况下,所述溶剂化物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂化物”包括溶液状态的溶剂化物和可分离的溶剂化物。代表性的溶剂化物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R·x H2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R·0.5H2O))和多水合物(x为大于1的数,例如,二水合物(R·2H2O)和六水合物(R·6H2O))。
术语“前药”是指当被施用至生物体时由于自发化学反应、酶催化的化学反应、光解和/或代谢化学反应而产生药物,即活性成分的任何化合物。前药因此是治疗活性化合物的共价改性的类似物或潜在形式。合适的实例包括但不限于:化合物的羧酸酯、碳酸酯、磷酸酯、硝酸酯、硫酸酯、砜酯、亚砜酯、氨基化合物、氨基甲酸盐、偶氮化合物、磷酰胺、葡萄糖苷、醚、乙缩醛等形式。
本发明还包括同位素标记的化合物(同位素变体),它们等同于本申请所述的那些通式或具体化合物,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可 以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F和36Cl,优选为2H(即氘,D)。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如3H和14C)的那些可用于药物和/或底物组织分布测定。氚、即3H和碳-14、即14C同位素是特别优选的,因为它们容易制备和检测。另外,被较重的同位素例如氘(即2H)取代,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下是优选的。同位素标记的本发明化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
本发明化合物包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
"任选的"或"任选地"意味着随后所描述的事件或环境可以但不必发生,包括该事件或环境发生或不发生的场合。例如,“芳基任选被烷基取代”意味着烷基可以但不必须存在,该术语包括芳基被烷基取代的情形和芳基不被烷基取代的情形。
“药学上可接受的赋形剂”指药学上可接受的材料、组合物或媒剂,例如液体或固体填充剂、稀释剂、赋形剂、溶剂或囊封材料。如本文所用,术语“药学上可接受的赋形剂”包括与药物施用相容的缓冲剂、注射用无菌水、溶剂、分散介质、包衣、抗细菌剂及抗真菌剂、等渗剂及吸收延迟剂及诸如此类。在与配制物中其他成分兼容且对患者无害的意义上,每种赋形剂必须为“药学上可接受的”。合适的实例包括但不限于:(1)糖,例如乳糖、葡萄糖及蔗糖;(2)淀粉,例如玉米淀粉、马铃薯淀粉及经取代或未经取代的β-环糊精;(3)纤维素及其衍生物,例如羧甲基纤维素钠、乙基纤维素及乙酸纤维素;(4)粉状黄蓍胶;(5)麦芽;(6)明胶;(7)滑石;(8)赋形剂,例如可可脂及栓剂蜡;(9)油类,例如花生油、棉籽油、红花油、芝麻油、橄榄油、玉米油及大豆油;(10)二醇,例如丙二醇;(11)多元醇,例如甘油、山梨醇、甘露醇及聚乙二醇;(12)酯类,例如油酸乙酯及月桂酸乙酯;(13)琼脂;(14)缓冲剂,例如氢氧化镁及氢氧化铝;(15)海藻酸;(16)无热原水;(17)等渗盐水;(18)林格氏溶液;(19)乙醇;(20)磷酸盐缓冲液;及(21)药物配制物中所采用的其他无毒兼容物质。
术语“多晶型”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂化物)。所有的多晶型物具有相同的元素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。
除非另有规定,本文使用的所有技术术语和科学术语具有要求保护主题所属领域的标准含义。倘若对于某术语存在多个定义,则以本文定义为准。应该理解,在本发明中使用的单数形式,如“一种”,包括复数指代,除非另有规定。
此外,术语“包括”、“包含”是开放性限定并非封闭式,即包括本发明所指明的内容,但并不排除其他方面的内容。
除非另有说明,本发明采用质谱、核磁等传统方法鉴定化合物,各步骤和条件可参照本领域常规的操作步骤和条件。
除非另有指明,本发明采用分析化学、有机合成化学和光学的标准命名及标准实验室步骤和技术。在某些情况下,标准技术被用于化学合成、化学分析、发光器件性能检测。
另外,需要说明的是,除非以其他方式明确指出,在本发明中所采用的描述方式“…分别独立地”应做广义理解,是指所描述的各个个体之间是相互独立的,可以独立地为相同或不同的具体基团。更详细地,描述方式“…分别独立地”既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响;也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
本发明的化合物或其组合物的剂型和施用方式没有特别限制。
代表性的施用方式包括但并不限于:口服、瘤内、直肠、肠胃外(静脉内、腹膜内、肌内或皮下)注射和/或局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填 料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和***胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、复合硅酸盐和碳酸钠;(e)溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。
用于口服给药的液体剂型包括药学上可接受的乳液剂、溶液剂、悬浮剂、糖浆剂或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂(如水或其它溶剂)、增溶剂和乳化剂。具体实例为例如,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油类,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物。除了惰性稀释剂外,组合物也可包含助剂,如润湿剂、悬浮剂、甜味剂、矫味剂和香料。例如,悬浮液可包含悬浮剂。具体实例为例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或它们的混合物。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散剂、悬浮液或乳液,以及用于重新溶解成无菌的可注射溶液或分散剂的无菌粉末。适宜的含水或非水载体、稀释剂、溶剂或赋形剂选自水、乙醇和多元醇,或者它们适宜的混合物。
用于局部给药的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。由活性成分在无菌条件下与药学上可接受的载体及防腐剂、缓冲剂和/或必要时可能需要的推进剂一起混合而成。
本发明涉及如下实施方案。
在一个实施方案中,本发明涉及通式(I’)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体:
其中:
X为N或CRx
Y为N或CRy
Z为N或CRz
W为N或CRw
G为N或CRg
条件是X、Y和Z中至多两者同时为N;
Rx、Ry和Rz各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1和-L1-R1,其中Rx、Ry和Rz中至少一者为-L1-R1,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rw和Rg各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
-L1-为-(CRaRb)m-,在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Ra和Rb与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
R1为-S(O)qNR’R”、-S(O)(=NR’)R4或-N=S(O)R4R4’;
A选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经s个R2取代基取代;
-L2-选自键、-O-、-NR’-、-S-、-C(O)-和-(CRcRd)n-,在化合价允许的情况下,-(CRcRd)n-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Rc和Rd与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
R’选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R”为-L3-B;
B选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经t个R5取代基取代;
-L3-为-(CReRf)p-,在化合价允许的情况下,-(CReRf)p-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Re和Rf与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R4和R4’各自独立地选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R5各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R6选自选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Ra、Rb、Rc、Rd、Re和Rf独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1和Rs2独立地选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
m为0、1、2、3、4、5或6;
n为0、1、2、3、4、5或6;
p为0、1、2、3、4、5或6;
t为1、2、3、4或5;
s为1、2、3、4、5、6、7或8;
q为1或2。
在一个实施方案中,本发明涉及通式(I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体:
其中:
X为N或CRx
Y为N或CRy
Z为N或CRz
W为N或CRw
G为N或CRg
条件是X、Y和Z中至多两者同时为N;
Rx、Ry和Rz各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1和-L1-R1,其中Rx、Ry和Rz中至少一者为-L1-R1,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rw和Rg各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
-L1-为-(CRaRb)m-,在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Ra和Rb与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
R1为-S(O)qNR’R”或-S(O)(=NR’)R4
A选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经s个R2取代基取代;
-L2-选自键、-O-、-NR’-、-S-、-C(O)-和-(CRcRd)n-,在化合价允许的情况下,-(CRcRd)n-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Rc和Rd与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
R’选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R”为-L3-B;
B选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经t个R5取代基取代;
-L3-为-(CReRf)p-,在化合价允许的情况下,-(CReRf)p-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Re和Rf与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R4选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R5各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R6选自选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Ra、Rb、Rc、Rd、Re和Rf独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1和Rs2独立地选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
m为0、1、2、3、4、5或6;
n为0、1、2、3、4、5或6;
p为0、1、2、3、4、5或6;
t为1、2、3、4或5;
s为1、2、3、4、5、6、7或8;
q为1或2。
在一个实施方案中,本发明涉及上述式(I)或(I’)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(II),
在一个实施方案中,本发明涉及上述式(I)或式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中-L2-为-O-。
在一个实施方案中,本发明涉及上述式(I)或式(II)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中-L2-为键。
在一个实施方案中,本发明涉及上述化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中A选自苯基、C3-6环烷基、5-6元杂芳基和5-10元杂环基。
在一个实施方案中,本发明涉及上述化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中A选自环戊基、环己基、氮杂环己基、氧杂环己基、硫杂环己基、氮杂环庚基、二氮杂环庚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基、八氢并环戊二烯基、苯基、吡咯基、呋喃基、噻吩基、吡啶基、嘧啶基和哒嗪基。
在一个实施方案中,本发明涉及上述化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中m为0。
在一个实施方案中,本发明涉及上述化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
m为1、2、3、4或5,
-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-O-、-S-和-C(O)-替代;和/或Ra和Rb与它们所连接的碳原子一起形成C3-6环烷基和3-5元杂环基;
Ra选自H、C1-6烷基和C1-6卤代烷基;
Rb选自H、C1-6烷基和C1-6卤代烷基。
在一个实施方案中,本发明涉及上述式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(III),
在一个实施方案中,本发明涉及上述式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(IV)
其中:
X为N或CRx
Y为N或CRy
Rx和Ry各自独立地选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R”为-L3-B;
B为5-7元杂环基,其任选经t个R5取代基取代;
-L3-为-(CReRf)p-,在化合价允许的情况下,-(CReRf)p-中的任意一个亚甲基单元任选且独立地被-O-、-S-和-C(O)-替代;
R2a、R2b、R2c、R2d、R2e、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R5各自独立地选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Re和Rf独立地选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1和Rs2独立地选H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
p为0、1、2或3;
t为1、2或3。
在一个实施方案中,本发明涉及上述式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N;
Rx选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R”为-L3-B;
B为5-6元杂环基,其任选经t个R5取代基取代;
-L3-为-(CReRf)p-;
R2a、R2b、R2c、R2d、R2e、R3a、R3b、R3c和R3d独立地选自H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R5各自独立地选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Re和Rf独立地选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
p为0、1、2或3;
t为1、2或3;
优选地,-L3-B选自
在一个实施方案中,本发明涉及上述式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N;
Rx为H或D;
R’为H;
R”为-L3-B;
B为5-6元杂环基,优选为氮杂环戊基或氮杂环己基;
-L3-为-(CReRf)p-,p为0或1;
R2a为C1-6烷基优选为甲基;
R2b为H或D;
R2c为卤素优选为F;
R2d为H或D;
R2e为H或D;
R3a为H或D;
R3b为C1-6卤代烷基优选为-CF3
R3c为卤素优选为了Cl;
R3d为H或D;
Re选自H、D和C1-6烷基;
Rf选自H、D和C1-6烷基;
优选地,-L3-B选自
在一个实施方案中,本发明涉及上述式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(V)
在一个实施方案中,本发明涉及上述式(V)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为N或CRx
Y为N或CRy
Rx和Ry各自独立地选自H、D、卤素、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
-L2-为键、-O-、-S-或-NH-;
A选自苯基、C3-8环烷基、5-6元杂芳基和5-10元杂环基,其任选经s个R2取代基取代;
在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-O-、-S-和-C(O)-替代;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Ra和Rb选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
或者Ra和Rb与它们所连接的碳原子一起形成C3-6环烷基和3-5元杂环基;
Rs1和Rs2独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
m为0、1、2、3或4;
s为1、2、3、4、5、6、7或8。
在一个实施方案中,本发明涉及上述式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx和Ry各自独立地选自H、D、卤素和C1-6烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
A选自和5-8元杂环基,其任选经s个R2取代基取代;
在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-O-替代;
-L2-为键或-O-;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2各自独立地选自H、D、卤素、C1-6烷基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2a选自H、D、卤素、C1-6烷基、C1-6卤代烷基、-ORs1a和-SRs1a,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H或D;
R2c为H、D、卤素、-ORs1c或-SRs1c
R2d为H或D;
R2e为H或D;
R3a为H或D;
R3b为C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3c为卤素;
R3d为H或D;
R4选自C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Ra和Rb选自H、D、卤素和C1-6烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
或者Ra和Rb与它们所连接的碳原子一起形成C3-6环烷基;
Rs1各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1a选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1c选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
m为1、2或3;
s为1或2。
在一个实施方案中,本发明涉及上述式(IV)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx为H;
Ry为卤素;
A选自和氮杂环庚烷基,其经s个R2取代;
-(CRaRb)m-选自-CH2-、-O-CH2-CH2-、
-L2-为键或-O-;
R’为H;
R2各自独立地选自H和卤素优选为F,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2a为H或C1-6烷基优选为甲基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H;
R2c为卤素或-ORs1c
R2d为H;
R2e为H;
R3a为H;
R3b为C1-6卤代烷基优选为-CF3
R3c为卤素优选为Cl;
R3d为H;
R4为C1-6烷基优选为甲基;
Rs1c为H、C1-6烷基或C1-6卤代烷基,优选为-CF3
s为1或2。
在一个实施方案中,本发明涉及上述式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(VI):
在一个实施方案中,本发明涉及上述式(VI)化合物,或其药学上可接受的盐、对映异构体、非对 映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为N或CRx
Y为N或CRy
Rx和Ry各自独立地选自H、D、卤素、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
-L2-为键、-O-、-S-或-NH-;
A选自苯基、C3-8环烷基、5-10元杂芳基和5-10元杂环基,其任选经s个R2取代基取代;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、ORs1、SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1和Rs2独立地选H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
s为1、2、3、4、5、6、7或8。
在一个实施方案中,本发明涉及上述式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
A选自环戊基、环己基、氮杂环己基、氧杂环己基、硫杂环己基、氮杂环庚基、二氮杂环庚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基、八氢并环戊二烯基、苯基、吡咯基、呋喃基、噻吩基、吡啶基、嘧啶基和哒嗪基,优选地,A与其上的取代基选自以下:
其中,R2a、R2b、R2c和R2d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代。
在一个实施方案中,本发明涉及上述式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
-L2-为-O-;
A选自环戊基和环己基,优选为环己基。
在一个实施方案中,本发明涉及上述式(VI)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
-L2-为键;
A选自氮杂环庚基、二氮杂环庚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基和八氢并环戊二烯基,优选地,在A为含氮杂环的情况下通过氮原子与苯基相连。
在一个实施方案中,本发明涉及上述式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(VII)
其中:
X为N或CRx
Y为N或CRy
Rx和Ry各自独立地选自H、D、卤素、C1-6烷基、C2-6烯基、C2-6炔基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2a、R2b、R2c、R2d、R2e、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1和Rs2独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
在一个实施方案中,本发明涉及上述式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为N或CRx
Y为N或CRy
Rx和Ry各自独立地选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2a选自H、D、卤素、-ORs1a、-SRs1a、NRs1aRs2a、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2c选自H、D、卤素、-ORs1c、-SRs1c、NRs1cRs2c、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2d为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2e为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3a为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3b选自H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3c选自H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3d为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1a、Rs2a、Rs1c和Rs2c各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
在一个实施方案中,本发明涉及上述式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx和Ry独立地选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R’为H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2a为-ORs1a、-SRs1a、NRs1aRs2a、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H、D或卤素;
R2c为H、D、卤素、-ORs1c、-SRs1c或NRs1cRs2c,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2d为H、D或卤素;
R2e为H、D或卤素;
R3a为H、D或卤素;
R3b为H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3c为H、D或卤素;
R3d为H、D或卤素;
R4为H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1a、Rs2a、Rs1c和Rs2c各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
条件是当Y为N时,R2a不为C1-6烷基或C1-6卤代烷基。
在一个实施方案中,本发明涉及上述式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx和Ry独立地选自H、D或卤素;
R’为H、C1-6烷基或C1-6卤代烷基;
R2a为-ORs1a、-SRs1a、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H、D或卤素;
R2c为H、D、卤素、-ORs1c或-SRs1c,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2d为H或D;
R2e为H或D;
R3a为H或D;
R3b为C1-6卤代烷基;
R3c为卤素;
R3d为H或D;
R4为C1-6烷基或C1-6卤代烷基;
Rs1a和Rs1c独立地选自C1-6烷基或C1-6卤代烷基;
条件是当Y为N时,R2a不为C1-6烷基或C1-6卤代烷基。
在一个实施方案中,本发明涉及上述式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx和Ry独立地选自H、D或卤素;
R’为H;
R2a为-ORs1a、-SRs1a或C1-6烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H或D;
R2c为卤素、-ORs1c或-SRs1c,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2d为H或D;
R2e为H或D;
R3a为H或D;
R3b为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3c为卤素;
R3d为H或D;
R4为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1a和Rs1c各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
在一个实施方案中,本发明涉及上述式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx为H或D;
Ry为H、D或卤素;
R’为H;
R2a为-ORs1a或-SRs1a,所述基团任选地被一个或多个氘取代,直至完全氘代;
R2b为H或D;
R2c为卤素、-ORs1c或-SRs1c
R2d为H或D;
R2e为H或D;
R3a为H或D;
R3b为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
R3c为卤素;
R3d为H或D;
R4为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
Rs1a和Rs1c独立地选自C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
在一个实施方案中,本发明涉及上述式(VII)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
X为CRx
Y为N或CRy
Rx为H;
Ry为卤素优选为F;
R’为H;
R2a为-ORs1a,所述基团任选地被一个或多个氘取代,直至完全氘代,优选为-OCD3
R2b为H;
R2c为卤素或-ORs1c,优选为F或-OCF3
R2d为H;
R2e为H;
R3a为H;
R3b为C1-6卤代烷基优选为-CF3
R3c为卤素,优选为Cl;
R3d为H;
R4为C1-6烷基;
Rs1a为C1-6烷基;
Rs1c为C1-6卤代烷基。
在一个具体的实施方案中,所述的化合物选自如下结构:



在一个具体的实施方案中,本发明涉及药物组合物,其包含如本发明所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
在一个实施方案中,本发明提供药物组合物,其包含如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物在制备用作电压-门控钠通道抑制剂的药物中的用途。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物在制备用作钠离子通道1.8(NaV1.8)抑制剂的药物中的用途。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物,其用作电压-门控钠通道抑制剂。
在一个实施方案中,本发明提供如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物,其用作钠离子通道1.8(NaV1.8)抑制剂。
在一个实施方案中,本发明提供一种在受试者中抑制电压-门控钠通道的方法,包括向所述受试者给药如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物。
在一个实施方案中,本发明提供一种在受试者中抑制钠离子通道1.8(NaV1.8)的方法,包括向所述受试者给药如本发明中所定义的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物。
如本发明所述的电压-门控钠通道抑制剂用于治疗选自以下的疾病:急性、慢性、神经性、或炎性疼痛、关节炎、偏头痛、丛集性头痛、三叉神经痛、疱疹性神经痛、一般性神经痛、癫痫或癫痫病症、神经变性疾病、精神障碍如焦虑和抑郁、双相性精神障碍、肌強直、心律失常、运动障碍、神经内分泌障碍、共济失调、多发性硬化、肠易激综合征、失禁、内脏痛、骨关节炎痛、疱瘆后神经痛、糖尿病性神经病、神经根痛、坐骨神经痛、背痛、头痛、颈痛、严重或顽固性疼痛、伤害性疼痛、贯穿性疼痛、手术后疼痛、癌症疼痛、中风、脑缺血、外伤性脑损伤、肌萎缩性侧索硬化、应激或运动诱发的心绞痛、心悸、高血压、偏头痛或异常的胃肠活动。
在一些实施方案中,本方法涉及的疾病选自神经根痛、坐骨神经痛、背痛、头痛、颈痛、顽固性疼痛、急性疼痛、手术后疼痛、背痛、耳鸣或癌症疼痛。
实施例
本文所用的材料或试剂为可购买到的或由本领域通常已知的合成方法制备。
实施例1
5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(2-(甲氨基)乙基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物1)
第一步:(2-(苄基硫基)吡啶-4-基)氨基甲酸叔丁酯
将2-(苄基硫基)-4-溴吡啶(3.7g,13.214mmol)溶解在超干二氧六环(37mL)中,加入氨基甲酸叔丁酯(3.1g,26.429mmol,),碳酸铯(12.9g,39.643mmol),三(二亚苄基丙酮)二钯(Pd2dba3)(1.2g,1.321mmol)和(9,9-二甲基-9H-呫吨-4,5-二基)双(二苯基膦)(XantPhos)(1.5g,2.643mmol),氮气置换,油浴100℃搅拌16小时。停止反应,将反应液真空旋干,加二氯乙烷(40ml)溶解,用装有硅藻土的砂芯漏斗抽滤,滤饼用二氯乙烷(80ml)淋洗,收集滤液旋干拌样,得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=10:1)分离纯化得到标题化合物(3.7g,收率:77.6%,无色油状物)。
MS(ESI):m/z 317.2[M+H]+
第二步:2-(苄基硫基)吡啶-4-胺
将(2-(苄基硫基)吡啶-4-基)氨基甲酸叔丁酯(1g,3.16mmol)溶解在二氯甲烷溶液(15mL)中,冰浴0℃下滴加三氟乙酸(5mL),室温下搅拌1小时。停止反应,反应液加饱和碳酸氢钠溶液(10ml)淬灭,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(10ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到标题化合物(680mg,无色油状物),直接投下一步反应。
MS(ESI):m/z 217.1[M+H]+
第三步:N-(2-(苄基硫基)吡啶-4-基)-5-氯-2-氟-4-(三氟甲基)苯甲酰胺
将2-(苄基硫基)吡啶-4-胺(680mg,3.148mmol)溶解在二氯亚砜(1mL)中,氮气置换三次后,80℃下搅拌2小时,旋干备用。将5-氯-2-氟-4-(三氟甲基)苯甲酸(763.6mg,3.148mmol)溶于二氯甲烷(8mL),冰浴0℃下滴加N,N-二异丙基乙胺(1.63g,12.592mmol),将开始时旋干备用的反应液溶于二氯甲烷(1mL),冰浴0℃下滴加到上述反应液中,室温搅拌16小时。停止反应,将反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=5:1)分离纯化得到标题化合物(370mg,收率:26.7%,无色油状物)。
MS(ESI):m/z 441.1[M+H]+
第四步:N-(2-(苄基硫基)吡啶-4-基)-5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺
将N-(2-(苄基硫基)吡啶-4-基)-5-氯-2-氟-4-(三氟甲基)苯甲酰胺(330mg,0.749mmol)溶解在N,N-二甲基甲酰胺(3.3mL)中,加入4-氟-2-甲基苯酚(94.3mg,0.749mmol),碳酸铯(487.8mg,1.497mmol),100℃油浴中搅拌反应1小时。停止反应,反应液加水(5ml)淬灭,水相用乙酸乙酯(10ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=5:1)分离纯化得到标题化合物(343mg,收率:83.9%,无色油状物)。
MS(ESI):m/z 547.0[M+H]+
第五步:4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯
将N-(2-(苄基硫基)吡啶-4-基)-5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺(100mg,0.183mmol)置于三口瓶,氮气置换,向瓶内加入(冰乙酸:水:二氯甲烷=7:1:2)(2mL),加入1,3-二氯-5,5-二甲基咪唑烷-2,4-二酮(108mg,0.548mmol),室温下搅拌3小时。停止反应,反应液加水(3ml)淬灭,水相用二氯甲烷(5ml×3)萃取,合并有机相,经饱和食盐水(3ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到标题化合物(80mg,粗品,淡黄色固体)。
MS(ESI):m/z 523.1[M+H]+
第六步:(2-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰胺基)乙基)(甲基)氨基甲酸叔丁基酯
将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(120mg,0.229mmol)溶于二氯甲烷(1ml)后,在0℃下滴加进(2-氨基乙基)(甲基)氨基甲酸叔丁基酯(39.9mg,0.229mmol)的吡啶(1mL)溶液,室温下搅拌2小时。停止反应,反应液加饱和碳酸氢钠溶液(3ml)淬灭,水相用乙酸乙酯(5ml×3)萃取,合并有机相,经饱和食盐水(2ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(45mg,收率:29.8%,淡黄色固体)。
MS(ESI):m/z 661.2[M+H]+
第七步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(2-(甲氨基))乙基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物1)
将(2-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰胺基)乙基)(甲基)氨基甲酸叔丁基酯(59mg,0.089mmol)溶解在二氯甲烷(3mL)中,冰浴0℃下加入三氟乙酸(1mL),室温搅拌1小时。停止反应,反应液真空旋除溶剂得到粗品;粗品经反相制备色谱(洗脱体系:氨水,水,乙腈)纯化得到标题化合物(18.7mg,收率:37.5%,棕黄色固体)。
MS(ESI):m/z 561.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.41(s,1H),8.62(d,J=5.4Hz,1H),8.26(d,J=1.4Hz,1H),8.12(s,1H),7.77(d,J=5.3Hz,1H),7.23–7.17(m,1H),7.10(dd,J=7.1,4.6Hz,3H),2.96(t,J=6.5Hz,2H),2.47(t,J=6.5Hz,2H),2.17(s,3H),2.16(s,3H).
实施例2
(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物2)
第一步:(R)-3-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)-苯甲酰胺基)吡啶)-2-磺酰氨基)吡咯烷-1-甲酸叔丁酯
将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(300mg,0.57mmol)和(R)-3-氨基吡咯烷-1-甲酸叔丁酯(107mg,0.57mmol)溶解在吡啶(2mL)和二氯甲烷(4mL)的混合溶液中,在0℃下反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的盐酸溶液调至中性,乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤后滤液减压旋干经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(222mg,收率:58%,黄色固体)。
MS(ESI):m/z 673.2[M+H]+
第二步:(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物2)
将(R)-3-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)-苯甲酰胺基)吡啶)-2-磺酰氨基)吡咯烷-1-甲酸叔丁酯(210mg,0.312mmol)用三氟乙酸(2mL)和二氯甲烷(6mL)的混合溶剂溶解,室温下反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥。过滤后滤液减压旋干,经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(122.14mg,收率:68%,白色固体)。
MS(ESI):m/z 573.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.40(s,1H),8.65(d,J=5.4Hz,1H),8.34(d,J=1.7Hz,1H),8.12(s,1H),7.80(dd,J=5.4,1.9Hz,1H),7.21(dd,J=9.2,2.3Hz,1H),7.15–7.06(m,3H),4.05–3.96(m,1H),3.22(dd,J=11.8,6.6Hz,1H),3.15(dd,J=15.2,7.8Hz,1H),3.10–3.05(m,1H),2.96(dd,J=11.8,5.4Hz,1H),2.17(s,3H),2.00–1.93(m,1H),1.81–1.74(m,1H).
实施例3
(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物3)
第一步:(S)-3-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)吡咯烷-1-甲酸叔丁酯
将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(300mg,0.57mmol)和(S)-3-氨基吡咯烷-1-甲酸叔丁酯(107mg,0.57mmol)溶解在吡啶(2mL)和二氯甲烷(4mL)的混合溶液中,在0℃下反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的盐酸溶液调至中性,乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析 (硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(210mg,收率:56%,黄色固体)。
MS(ESI):m/z 673.3[M+H]+
第二步:(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物3)
将(S)-3-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)吡咯烷-1-甲酸叔丁酯(210mg,0.312mmol)用三氟乙酸(2mL)和二氯甲烷(6mL)的混合溶剂溶解,室温反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥。经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(71.26mg,收率:40.0%,白色固体)。
MS(ESI):m/z 572.9[M+H]+
1H NMR(400MHz,DMSO-d6)δ8.64(d,J=5.4Hz,1H),8.33(s,1H),8.31(d,J=1.8Hz,1H),8.13(s,1H),7.81(dd,J=5.4,2.0Hz,1H),7.21(dd,J=9.2,2.7Hz,1H),7.14–7.05(m,3H),3.96–3.86(m,1H),3.07–2.97(m,2H),2.92(d,J=6.6Hz,1H),2.77(dd,J=11.6,4.9Hz,1H),2.17(s,3H),1.92–1.87(m,1H),1.67–1.62(m,1H).
实施例4
5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(哌啶-4-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物4)
第一步:4-((4-(5-氯-2-(4氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)哌啶-1-甲酸叔丁酯
室温搅拌条件下,将4-氨基哌啶-1-氨基甲酸叔丁酯(76.55mg,0.38mmol)溶解在吡啶(2mL)中,降温至0℃,将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(200mg,0.38mmol)溶于二氯甲烷(4mL)并滴加至反应液中,反应维持在此温度下搅拌反应2小时。停止反应,将反应液倒入水(10mL)中,并用乙酸乙酯(10mL×3)萃取。合并萃取液,用1N HCl(5mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品。粗品柱层析(硅胶,乙酸乙酯:石油醚=1:1)分离纯化得到标题化合物(120mg,收率:45.7%,白色固体)。
MS(ESI):m/z 687.2[M+H]+
第二步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(哌啶-4-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物4)
室温搅拌条件下,将4-((4-(5-氯-2-(4氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)哌啶-1-甲酸叔丁酯(120mg,0.17mmol)置入25ml单口瓶中,并溶解在二氯甲烷(3mL)中;加入三氟乙酸(1mL),恢复至室温,反应在室温下继续搅拌1小时。停止反应,将反应液减压浓缩,得到粗品,粗品经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)分离纯化得到标题化合物(87.96mg,收率:85.8%,白色固体)。
MS(ESI):m/z 587.2[M+H]+
实施例5
5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(哌啶-4-基甲基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物5)
第一步:4-(((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)甲基)哌啶-1-甲酸叔丁酯
在0℃下将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(200mg,0.38mmol)和4-(氨基甲基)哌啶-1-甲酸叔丁酯(82mg,0.38mmol)依次加入到吡啶(2mL)和二氯甲烷(4mL)的混合溶液中,反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的盐酸溶液调至中性,乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(110mg,收率:41.2%,黄色固体)。
MS(ESI):m/z 701.1[M+H]+
第二步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(哌啶-4-基甲基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物5)
将4-(((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)甲基)哌啶-1-甲酸叔丁酯(100mg,0.143mmol)用三氟乙酸(1mL)和二氯甲烷(3mL)的混合溶剂溶解,室温反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥。过滤后滤液减压旋干,经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(40.99mg,收率:48%,白色固体)。
MS(ESI):m/z 601.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.39(s,1H),8.63(d,J=5.4Hz,1H),8.29(d,J=1.7Hz,1H),8.12(s,1H),7.77(dd,J=5.4,1.9Hz,1H),7.21(dd,J=9.2,2.3Hz,1H),7.14–7.06(m,3H),3.19(d,J=12.0Hz,2H),2.81(d,J=6.6Hz,2H),2.74(t,J=11.8Hz,2H),2.16(s,3H),1.76(d,J=13.3Hz,2H),1.63(brs,1H),1.21(dd,J=23.0,10.9Hz,2H).
实施例6
(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基甲基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物6)
第一步:(R)-3-(((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)甲基)吡咯烷-1-甲酸叔丁酯
在0℃下将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(300mg,0.57mmol)和(S)-3-(氨基甲基)吡咯烷-1-甲酸叔丁酯(115mg,0.57mmol)溶解在吡啶(2mL)和二氯甲烷(4mL)的混合溶液中,反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的盐酸溶液调至中性,乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(256mg,收率:65.4%,黄色固体)。
MS(ESI):m/z 687.3[M+H]+
第二步:(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基甲基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物6)
将(R)-3-(((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)甲基)吡咯烷-1-甲酸叔丁酯(240mg,0.35mmol)用三氟乙酸(2mL)和二氯甲烷(6mL)的混合溶剂溶解,室温下搅拌反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥。过滤后滤液减压旋干,经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(112.26mg,收率:54.6%,白色固体)。
MS(ESI):m/z 587.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.31(s,1H),8.64(d,J=5.5Hz,1H),8.31(d,J=1.7Hz,1H),8.11(s,1H),7.77(dd,J=5.4,1.9Hz,1H),7.21(dd,J=9.2,2.4Hz,1H),7.14–7.06(m,3H),3.23–3.09(m,2H),3.08–3.02(m,1H),2.98(d,J=7.0Hz,2H),2.81(dd,J=11.6,7.7Hz,1H),2.36(dt,J=15.3,7.6Hz,1H),2.16(s,3H),1.97–1.92(m,1H),1.61–1.56(m,1H).
实施例7
(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基甲基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物7)
第一步:(S)-3-(((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)甲基)吡咯烷-1-甲酸叔丁酯
在0℃下将4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶-2-磺酰氯(300mg,0.57mmol)和(R)-3-(氨基甲基)吡咯烷-1-甲酸叔丁酯(115mg,0.57mmol)溶解在吡啶(2mL)和二氯甲烷(4mL)的混合溶液中,反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的盐酸溶液调至中性,乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(256mg,收率:65.4%,黄色固体)。
MS(ESI):m/z 687.2[M+H]+
第二步:(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基甲基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物7)
将(S)-3-(((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺基)吡啶)-2-磺酰氨基)甲基)吡咯烷-1-甲酸叔丁酯(240mg,0.35mmol)用三氟乙酸(2mL)和二氯甲烷(6mL)的混合溶剂溶解,室温反应2小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥。过滤后滤液减压旋干,经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(63.06mg,收率:30.7%,白色固体)。
MS(ESI):m/z 586.9[M+H]+
1H NMR(400MHz,DMSO-d6)δ8.63(d,J=5.5Hz,1H),8.36(s,1H),8.29(d,J=1.7Hz,1H),8.12(s,1H),7.79(dd,J=5.4,1.9Hz,1H),7.21(dd,J=9.2,2.6Hz,1H),7.15–7.05(m,3H),3.12(dd,J=11.3,7.8Hz,1H),3.09–3.03(m,1H),2.99(d,J=7.8Hz,1H),2.95(d,J=7.1Hz,2H),2.74(dd,J=11.3,7.3Hz,1H),2.31(dd,J=14.7,7.3Hz,1H),2.16(s,3H),1.88(dt,J=13.0,6.5Hz,1H),1.53(dq,J=15.4,7.8Hz,1H).
实施例8
(S)-5-氯-N-(2-(N-(1-乙基吡咯烷-3-基)氨磺酰基)吡啶-4-基)-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基))苯甲酰胺(化合物8)
将(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(102mg,0.178mmol)用四氢呋喃(1.5mL)溶解,加入三乙胺(54mg,0.535mmol)和碘乙烷(28mg,0.178mmol),密封后油浴70℃反应16小时。停止反应,向反应液加1N的稀盐酸调至偏酸性,用乙酸乙酯(30mL×3)萃取,合并有机相经饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,二氯甲烷:甲醇=10:1)分离纯化,而后由高效液相色谱制备(洗脱体系:氨水,水,乙腈)纯化,得到标题化合物(21.24mg,收率:19.9%,白色固体)
MS(ESI):m/z 601.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.33(s,1H),8.63(d,J=5.4Hz,1H),8.27(d,J=1.8Hz,1H),8.13(s,1H),8.07(d,J=6.8Hz,1H),7.78(dd,J=5.4,1.9Hz,1H),7.20(dd,J=9.1,2.2Hz,1H),7.14–7.05(m,3H),3.77(brs,1H),2.59(dd,J=9.3,7.3Hz,1H),2.42–2.25(m,4H),2.22–2.14(m,4H),1.94–1.85(m,1H),1.52(dq,J=8.0,5.9Hz,1H),0.93(t,J=7.2Hz,3H).
实施例9
(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(1-丙基吡咯烷-3-基)磺酰氨)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物9)
室温条件下,将(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(80mg,0.14mmol)和丙醛(24.33mg,0.42mmol)溶解在二氯甲烷(2mL)中,加入醋酸(25.13mg,0.42mmol),反应在室温下搅拌2小时;然后加入三乙酰氧基硼氢化钠(NaBH(OAc)3)(88.77mg,0.42mmol),反应继续在室温下搅拌2小时。反应完毕后,将反应液减压浓缩,得到粗品;粗品经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)分离纯化得到标题化合物(27.16mg,收率:28.1%,白色固体)。
MS(ESI):m/z 615.2[M+H]+
实施例10
(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(1-(2-氟乙基)吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物10)
将(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(70mg,0.12mmol)溶解在N,N-二甲基甲酰胺(1mL)中,加入1-氟-2-碘乙烷(21.25mg,0.12mmol),碳酸钾(33.7mg,0.24mmol),油浴50℃反应16小时。停止反应,向反应液中加水(10mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤后滤液减压旋干,经高效液相色谱制备(洗脱体系:氨水,水,乙腈)纯化得到标题化合物(28.51mg,收率:37.7%,白色固体)
MS(ESI):m/z 618.9[M]+
1H NMR(400MHz,DMSO-d6)δ11.33(s,1H),8.63(d,J=5.4Hz,1H),8.27(s,1H),8.15–8.07(m,2H),7.78(dd,J=5.4,1.7Hz,1H),7.23–7.17(m,1H),7.13–7.09(m,3H),4.44(dt,J=44.0,4.9Hz,2H),3.79(dd,J=14.4,6.6Hz,1H),2.72–2.58(m,5H),2.34–2.26(m,1H),2.16(s,3H),1.98–1.83(m,1H),1.58–1.51(m,1H).
实施例11
(R)-5-氯-N-(2-(N-(1-乙基吡咯烷-3-基)氨磺酰基)吡啶-4-基)-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基))苯甲酰胺(化合物11)
将(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(80mg,0.14mmol)用四氢呋喃(2.0mL)溶解,加入三乙胺(42.38mg,0.42mmol)和碘乙烷(21.78mg,0.14mmol),密封后油浴70℃反应16小时。停止反应,向反应液加1N的稀盐酸调至偏酸性,用乙酸乙酯(30mL×3)萃取,合并有机相经饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥。经柱层析(硅胶,二氯甲烷:甲醇=10:1)分离纯化,而后由高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化,得到标题化合物(44.72mg,收率:42.6%,白色固体)
MS(ESI):m/z 601.1[M+H]+
实施例12
(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(1-丙基吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物12)
室温条件下,将(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(N-(吡咯烷-3-基)氨磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(80mg,0.14mmol)和丙醛(24.32mg,0.42mmol)溶解在二氯甲烷(2mL)中,加入醋酸(25.13mg,0.42mmol),反应在室温下搅拌2小时;然后加入三乙酰氧基硼氢化钠(88.77mg,0.42mmol),反应继续在室温下搅拌2小时。反应完毕后,将反应液减压浓缩,得到粗品,经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)分离纯化得到标题化合物(27.36mg,收率:25.5%,白色固体)。
MS(ESI):m/z 615.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.35(s,1H),8.64–8.62(m,1H),8.28–8.27(m,1H),8.15–8.08(m,2H),7.79–7.77(m,1H),7.21–7.19(m,1H),7.13–7.05(m,3H),3.83–3.76(m,1H),2.69–2.64(m,1H),2.46–2.26(m,5H),2.16(s,3H),1.95–1.86(m,1H),1.59–1.50(m,1H),1.40–1.31(m,2H),0.83–0.78(m,3H).
实施例13
5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物13)
第一步:2-(甲硫基)吡啶-4-胺
将2-氯吡啶-4-胺(1g,7.8mmol)溶解在NMP(10mL)中,加入甲硫醇钠(1.12g,15.6mmol),微波200摄氏度反应0.5小时。停止反应,将反应液加水(30ml)淬灭,水相用氯仿/异丙醇(v:v=3:1)(40ml×3)萃取,合并有机相,经无水硫酸钠干燥,旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(900mg,粗品),直接投下一步反应。
MS(ESI):m/z 141.2[M+H]+
第二步:5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将2-(甲硫基)吡啶-4-胺(400mg,2.85mmol)溶解在吡啶(6mL)中,加入5-氯-2-氟-4-(三氟甲基)苯甲酸(691mg,2.85mmol),0℃下滴加三氯氧磷(1.31g,8.55mmol),继续搅拌反应2小时。停止反应,将反应液缓慢滴加到冰水(30ml)中,用1N盐酸调pH至5~6,水相用乙酸乙酯(30ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=4:1)分离纯化得到标题化合物(720mg,收率:69.2%,无色油状物)。
MS(ESI):m/z 364.9[M+H]+
第三步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(200mg,0.55mmol)溶解在DMF(2mL)中,加入4-氟-2-甲基苯酚(138mg,1.097mmol),加入碳酸铯(357mg,1.097mmol),油浴100℃下搅拌反应2小时。反应液加水(10ml)淬灭,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=4:1)分离纯化得到标题化合物(273mg,收率:92.5%,白色固体)。
MS(ESI):m/z 471.1[M+H]+
第四步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物13)
将5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(250mg,0.53mmol)溶解在甲醇(3mL)中,加入碳酸铵(153mg,1.59mmoL)和二乙酸碘苯(683mg,2.12mmoL),室温搅拌1小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离得产物粗品,粗品用DMF溶解送反相制备色谱(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(75.41mg,收率:28.2%,白色固体)。
MS(ESI):m/z 502.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ8.61(d,J=5.5Hz,1H),8.41(s,1H),8.38(s,1H),8.10(s,1H),7.77(d,J=5.3Hz,1H),7.20(d,J=10.0Hz,1H),7.14–7.04(m,3H),3.12(s,3H),2.15(s,3H).
实施例14
5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物14)
第一步:1-(苄氧基)-4-氟-2-(甲氧基-d3)苯
将2-(苄氧基)-5-氟苯酚(400mg,1.83mmol)溶解在DMF(4mL)中,加入碳酸钾(506mg,3.66mmol),0℃下加入氘代碘甲烷(319mg,2.20mmol),封管室温搅拌反应16小时。停止反应,将反应液加水(10ml)淬灭,水相用乙酸乙酯(15mL×3)萃取,合并有机相,经无水硫酸钠干燥,过滤后滤液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=30:1)分离纯化得标题化合物(340mg,白色固体,收率:79.1%)。
1H NMR(400MHz,CDCl3)δ7.44–7.29(m,5H),6.80(dd,J=8.8,5.5Hz,1H),6.65(dd,J=10.2,2.9Hz,1H),6.52(td,J=8.5,2.9Hz,1H),5.10(s,2H).
第二步:4-氟-2-(甲氧基-d3)苯酚
将1-(苄氧基)-4-氟-2-(甲氧基-d3)苯(300mg,1.275mmol)溶解在甲醇(9mL)中,加入 Pd(OH)2(60mg),室温搅拌16h。停止反应,将反应液过滤,滤饼用甲醇(9mL×3)淋洗,滤液旋干得到标题化合物(136mg,收率:73.5%,褐色油状物)。
第三步:5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将4-氟-2-(甲氧基-d3)苯酚(50mg,0.34mmol)溶解在DMF(1mL)中,加入5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(62.8mg,0.17mmol),加入碳酸钾(47.5mg,0.34mmol),封管100℃搅拌反应2小时。停止反应,反应液加水(10ml)淬灭,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(100mg,收率:97.7%,白色固体)。
MS(ESI):m/z 490.1[M+H]+
第四步:5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物14)
将5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(85mg,0.17mmol)溶解在甲醇(1mL)中,加入碳酸铵(50mg,0.52mmoL),加入二乙酸碘苯(224mg,0.70mmoL),室温搅拌0.5小时。反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离得产物粗品,粗品经反相制备色谱(洗脱体系:甲酸,水,乙腈)纯化得到5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(FZ008-114)(47.93mg,收率:52.9%,白色固体)。
MS(ESI):m/z 521.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.31(s,1H),8.63(d,J=5.4Hz,1H),8.40(s,1H),8.07(s,1H),7.82(d,J=3.6Hz,1H),7.29(dd,J=8.8,5.8Hz,1H),7.14(dd,J=10.6,2.8Hz,1H),7.00(s,1H),6.85(td,J=8.6,2.9Hz,1H),4.37(s,1H),3.14(s,3H).
化合物14手性异构体拆分:
(S)-5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
(R)-5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
化合物14经手性制备分离柱(色谱柱:250*25mm,10μm;流动相:A-Supercritical CO2;B-MEOH(+0.1%7.0mol/l Ammonia in MEOH),50%B比例洗脱,流速:100mL/min,柱温:室温)分离得到单一构型化合物14E1(较短保留时间)和化合物14E2(较长保留时间)。
实施例15
5-氯-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物15)
第一步:5-氯-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(78mg,0.21mmol)溶解在DMF(1mL)中,加入2-(甲氧基-d3)-4-(三氟甲氧基)苯酚(90mg,0.43mmol),加入碳酸钾(59mg,0.43mmol),油浴100℃下搅拌反应2小时。停止反应,反应液加水(10ml)淬灭,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=3:1)分离纯化得到标题化合物(100mg,收率:84.7%,白色固体)。
MS(ESI):m/z 556.1[M+H]+
第二步:5-氯-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物15)
将5-氯-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(88mg,0.16mmol)溶解在甲醇(1mL)中,加入碳酸铵(45.6mg,0.48mmoL),加入二乙酸碘苯(204mg,0.63mmoL),室温搅拌反应0.5小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离得产物粗品,粗品经反相制备色谱(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(57.30mg,收率:61.3%,白色固体)。
MS(ESI):m/z 587.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.31(s,1H),8.62(d,J=5.5Hz,1H),8.38(s,1H),8.09(s,1H),7.80(dd,J=5.4,1.8Hz,1H),7.30(d,J=8.8Hz,1H),7.23–7.12(m,2H),7.00(d,J=8.8Hz,1H),3.14(s,3H).
实施例16
5-氯-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物16)
第一步:5-氯-N-(2-(甲硫基)吡啶-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(70mg,0.192mmol)和4-(三氟甲氧基)苯酚(68.5mg,0.385mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸钾(54mg,0.385mmol),油浴70℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(120mg,收率:93.02%,白色固体)。
MS(ESI):m/z 523.2[M+H]+
第二步:5-氯-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物16)
将5-氯-N-(2-(甲硫基)吡啶-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(100mg,0.192mmol)用甲醇(2mL)溶解,加入碳酸铵(55.3mg,0.575mmol)、二乙酸碘苯(247mg,0.767mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:9)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(68.69mg,收率:64.8%,白色固体)。
MS(ESI):m/z 554.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.34(s,1H),8.60(d,J=5.5Hz,1H),8.34(d,J=1.9Hz,1H),8.15(s,1H),7.76(dd,J=5.4,2.0Hz,1H),7.52(s,1H),7.40(d,J=8.4Hz,2H),7.24–7.20(m,2H),4.36(s,1H),3.13(s,3H).
实施例17
5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物17)
第一步:5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(甲硫基)吡啶-4-基)-4-三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(100mg,0.274mmol)和4,4-二氟氮杂环丁烷盐酸盐(234mg,1.371mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸铯(625.3mg,1.92mmol),油浴100℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液减压旋干经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(125mg,收率:92.6%,白色固体)。
MS(ESI):m/z 480.1[M+H]+
第二步:5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物17)
将5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(甲硫基)吡啶-4-基)-4-三氟甲基)苯甲酰胺(65mg,0.136mmol)用甲醇(2mL)溶解,加入碳酸铵(40mg,0.41mmol)、二乙酸碘苯(175mg,0.543mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:10)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(35.02mg,收率:50.49%,白色固体)。
MS(ESI):m/z 511.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.24(s,1H),8.63(d,J=5.3Hz,1H),8.41(s,1H),7.84(d,J=4.7Hz,1H),7.75(s,1H),7.35(s,1H),4.37(s,1H),3.39–3.32(m,4H),3.15(s,3H),2.24(brs,2H),2.07–2.04(m,2H),1.79(brs,2H).
化合物17手性异构体拆分:
(S)-5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
(R)-5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
化合物17经手性制备分离柱(色谱柱:250*25mm,10μm;流动相:A-Supercritical CO2;B-MEOH(+0.1%7.0mol/l Ammonia in MEOH),25%B比例洗脱,流速:70mL/min,柱温:室温)分离得到单一构型化合物17E1(较短保留时间)和化合物17E2(较长保留时间)。
实施例18
5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物18)
第一步:5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(100mg,0.27mmol)溶解在DMF(3mL)中,加入(3aR,6aS)-5,5-二氟八氢环戊二烯并[c]吡咯盐酸盐(98.8mg,0.54mmol)和碳酸铯(536mg,1.64mmol),氮气置换,100℃反应12小时。停止反应,反应液加水(15mL),水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(10ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=9:1)分离纯化得到标题化合物(100mg,收率:74.6%,白色固体)。
MS(ESI):m/z 492.1[M+H]+
第二步:5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物18)
将5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(100mg,0.20mmol)溶于甲醇(1.5mL)中,加入碳酸铵(58.6mg,0.60mmol)和乙二酸碘苯(261.9mg,0.81mmol),室温搅拌1小时。停止反应,将反应液旋干,加饱和碳酸氢钠溶液水(2ml)淬灭,水相用乙酸乙酯(5ml×3)萃取,合并有机相,经饱和食盐水(3ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经反相制备色谱(洗脱体系:氨水,水,乙腈)纯化得到标题化合物(10mg,收率:9.4%,白色固体)。
MS(ESI):m/z 539.8[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.28(s,1H),8.62(d,J=4.9Hz,1H),8.42(s,1H),7.82(s,1H),7.72(s,1H),7.16(s,1H),4.36(s,1H),3.39(brs,2H),3.19(d,J=9.6Hz,2H),3.14(s,3H),2.85(brs,2H),2.32(brs,2H),1.98(brs,2H).
实施例19
5-氯-2-((4,4-二氟环己基)氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物19)
第一步:5-氯-2-((4,4-二氟环己基)氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(100mg,0.274mmol)和4,4-二氟环己-1-醇(150mg,1.097mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸铯(268mg,0.823mmol),油浴100℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(200mg,收率:98.1%,白色固体)。
MS(ESI):m/z 481.0[M+H]+
第二步:5-氯-2-((4,4-二氟环己基)氧基)-N-(2-(S-甲基亚胺代磺酰基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物19)
将5-氯-2-((4,4-二氟环己基)氧基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(190mg,0.4mmol)用甲醇(2mL)溶解,加入碳酸铵(114mg,1.19mmol)、二乙酸碘苯(510mg,1.58mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:10)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(81.2mg,收率:39.8%,白色固体)。
MS(ESI):m/z 512.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.14(s,1H),8.63(d,J=5.4Hz,1H),8.40(s,1H),7.92(s,1H),7.78(d,J=5.2Hz,1H),7.66(s,1H),4.96(brs,1H),4.37(s,1H),3.14(s,3H),2.01–1.83(m,8H).
实施例20
5-氯-2-(4-氟-2-甲基苯氧基)-N-(6-(S-甲基亚胺代磺酰基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(化合物20)
第一步:6-(甲硫基)哒嗪-4-胺
将6-氯哒嗪-4-胺(1g,7.7mmol)溶解在甲醇(10mL)中,加入甲硫醇钠(2.8g,38.6mmol),封管100℃搅拌反应16小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:10)分离纯化得到标题化合物(800mg,白色固体,收率:73.4%)。
MS(ESI):m/z 142.0[M+H]+
第二步:5-氯-2-氟-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺
将6-(甲硫基)哒嗪-4-胺(75mg,0.53mmol)溶解在吡啶(2mL)中,加入5-氯-2-氟-4-(三氟甲基)苯甲酸(129mg,0.53mmol),0℃下滴加三氯氧磷(244mg,1.59mmol),继续搅拌反应2小时。停止反应,将反应液缓慢滴加到冰水(10ml)中,用1N盐酸调pH至5~6,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(90mg,收率:46.6%,黄色固体)。
MS(ESI):m/z 365.0[M+H]+
第三步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(80mg,0.22mmol)溶解在DMF(1mL)中,加入4-氟-2-甲基苯酚(41.4mg,0.33mmol),加入碳酸铯(142.7mg,0.44mmol),油浴100℃搅拌反应2小时。停止反应,反应液加水(10ml)淬灭,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(69mg,收率:67.0%,白色固体)。
MS(ESI):m/z 472.0[M+H]+
第四步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(6-(S-甲基亚胺代磺酰基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(化合物20)
将5-氯-2-(4-氟-2-甲基苯氧基)-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(60mg,0.13mmol)溶解在甲醇(1mL)中,加入碳酸铵(36.6mg,0.39mmoL)和二乙酸碘苯(164mg,0.51mmoL),室温搅拌1小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离得产物粗品,粗品经反相制备色谱(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(17.45mg,收率:27.3%,白色固体)。
MS(ESI):m/z 503.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.66(s,1H),9.39(s,1H),8.63(d,J=2.0Hz,1H),8.21(s,1H),8.14(s,1H),7.24–7.17(m,1H),7.11(d,J=5.3Hz,3H),4.86(s,1H),3.31(s,3H),2.16(s,3H).
实施例21
5-氯-N-(6-(S-甲基亚胺代磺酰基)哒嗪-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物21)
第一步:5-氯-N-(6-(甲硫基)哒嗪-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(70mg,0.192mmol)和4-(三氟甲氧基)苯酚(68.3mg,0.384mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸钾(53mg,0.384mmol),油浴70℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(70mg,收率:69.72%,白色固体)。
MS(ESI):m/z 524.0[M+H]+
第二步:5-氯-N-(6-(S-甲基亚胺代磺酰基)哒嗪-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物21)
将5-氯-N-(6-(甲硫基)哒嗪-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(60mg,0.125mmol)用甲醇(2mL)溶解,加入碳酸铵(36.04mg,0.375mmol)、二乙酸碘苯(161mg,0.5mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:9)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(25.42 mg,收率:36.7%,黄色固体)。
MS(ESI):m/z 555.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.65(s,1H),9.39(d,J=2.4Hz,1H),8.59(d,J=2.4Hz,1H),8.19(s,1H),7.55(s,1H),7.41(d,J=8.8Hz,2H),7.23(d,J=9.1Hz,2H),4.87(s,1H),3.29(s,3H).
实施例22
5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(6-(S-甲基亚胺代磺酰基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(化合物22)
第一步:5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(70mg,0.192mmol)和4,4-二氟氮杂环庚烷盐酸盐(164mg,0.959mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸铯(437mg,1.342mmol),油浴100℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(70mg,收率:75.95%,白色固体)。
MS(ESI):m/z 481.1[M+H]+
第二步:5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(6-(S-甲基亚胺代磺酰基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(化合物22)
将5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(6-(甲硫基)哒嗪-4-基)-4-(三氟甲基)苯甲酰胺(70mg,0.134mmol)用甲醇(2mL)溶解,加入碳酸铵(39mg,0.402mmol)、二乙酸碘苯(172mg,0.535mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:10)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(15.88mg,收率:23.2%,白色固体)。
MS(ESI):m/z 512.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.51(s,1H),9.44(s,1H),8.66(s,1H),7.80(s,1H),7.35(s,1H),4.89(s,1H),3.42–3.40(m,2H),3.38–3.34(m,2H),3.32(s,3H),2.27(brs,2H),2.08–2.00(m,2H),1.80(brs,2H).
实施例23
5-氯-2-(4-氟-2-甲基苯氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物23)
第一步:4-氟-3-(甲硫基)苯胺
将3-溴-4-氟苯胺(1g,5.26mmol)溶解在超干1,4-二氧六环(20mL)中,加入甲硫醇钠(569mg,7.89mmol),Pd2(dba)3(485mg,0.53mmol),XantPhos(609mg,1.05mmol),DIEA(2.04g,15.78mmol),氮气置换,油浴110℃搅拌反应16小时。停止反应,反应液过滤,滤饼用DCM(30ml×3)淋洗,滤液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:2)分离纯化得到标题化合物(600mg,无色油状物,收率:72.6%)。
MS(ESI):m/z 158.0[M+H]+
第二步:5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-4-(三氟甲基)苯甲酸(980mg,4.04mmol)溶解在二氯亚砜(1.5mL)中,氮气置换,80℃下搅拌反应2小时,旋干备用。将4-氟-3-(甲硫基)苯胺(635mg,4.04mmol)溶于超干DCM(10mL),0℃下加入吡啶(1.28g,16.16mmol),然后加入旋干备用物的二氯甲烷溶液(2ml),室温搅拌反应2小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=10:1)分离纯化得到标题化合物(870mg,收率:56.4%,白色固体)。
MS(ESI):m/z 382.0[M+H]+
第三步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(300mg,0.785mmol)溶解在DMF(4mL)中,加入4-氟-2-甲基苯酚(198mg,1.571mmol),加入碳酸铯(512mg,1.571mmol),油浴100℃下搅拌反应2小时。停止反应,反应液加水(20ml)淬灭,水相用乙酸乙酯(20 ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=5:1)分离纯化得到标题化合物(290mg,收率:75.7%,黄色固体)。
MS(ESI):m/z 488.1[M+H]+
第四步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物23)
将5-氯-2-(4-氟-2-甲基苯氧基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(290mg,0.59mmol)溶解在甲醇(3mL)中,加入碳酸铵(171.3mg,1.78mmoL)和二乙酸碘苯(766mg,2.36mmoL),室温搅拌反应1小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:10)分离得产物粗品,粗品经反相制备色谱(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(197mg,收率:64.3%,白色固体)。
MS(ESI):m/z 519.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.92(s,1H),8.25(dd,J=6.5,2.7Hz,1H),8.06(s,1H),7.92–7.85(m,1H),7.43(t,J=9.3Hz,1H),7.25–7.19(m,1H),7.13–7.06(m,3H),4.68(s,1H),3.18(s,3H),2.17(s,3H).
化合物23手性异构体拆分:
(S)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺
(R)-5-氯-2-(4-氟-2-甲基苯氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺
化合物23经手性制备分离柱(色谱柱:250*25mm,10μm;流动相:A-Supercritical CO2;B-MEOH(+0.1%7.0mol/l Ammonia in MEOH),30%B比例洗脱,流速:70mL/min,柱温:室温)分离得到单一构型化合物23E1(较短保留时间)和化合物23E2(较长保留时间)。
实施例24
5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物24)
第一步:5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺
将4-氟-2-(甲氧基-d3)苯酚(95mg,0.654mmol)和5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(50mg,0.131mmol)溶解在N,N-二甲基甲酰胺(1mL)中,加入碳酸钾(36.2mg,0.262mmol),密封后油浴100℃反应2小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=10:1)分离纯化得到标题化合物(80mg,收率:98.2%,白色固体)。
MS(ESI):m/z 507.1[M+H]+
第二步:5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物24)
将5-氯-2-(4-氟-2-(甲氧基-d3)苯氧基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(70mg,0.138mmol)溶于甲醇(2mL),加入碳酸铵(40mg,0.415mmol)、二乙酸碘苯(178mg,0.553mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化,而后经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(39.21mg,收率:52.7%,白色固体)。
MS(ESI):m/z 538.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.88(s,1H),8.28(dd,J=6.4,2.6Hz,1H),8.01(s,1H),7.96–7.90(m,1H),7.44(t,J=9.3Hz,1H),7.28(dd,J=8.8,5.9Hz,1H),7.14(dd,J=10.6,2.8Hz,1H),6.98(s,1H),6.85(td,J=8.5,2.9Hz,1H),4.69(s,1H),3.18(s,3H).
实施例25
5-氯-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物25)
第一步:5-氯-N-(4-氟-3-(甲硫基)苯基)-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(60mg,0.157mmol)和2-(甲氧基-d3)-4-(三氟甲氧基)苯酚(66.4mg,0.314mmol)溶解在N,N-二甲基甲酰胺(1.5mL)中,加入碳酸钾(43.4mg,0.314mmol),油浴100℃反应2小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=10:1)分离纯化得到标题化合物(80mg,收率:89.79%,白色固体)。
MS(ESI):m/z 572.8[M+H]+
第二步:5-氯-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物25)
将5-氯-N-(4-氟-3-(甲硫基)苯基)-2-(2-(甲氧基-d3)-4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(60mg,0.105mmol)用甲醇(2mL)溶解,加入碳酸铵(30.3mg,0.315mmol)、二乙酸碘苯(135.15mg,0.42mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化,而后经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(16.02mg,收率:25.4%,白色固体)。
MS(ESI):m/z 604.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.89(s,1H),8.25(dd,J=6.4,2.6Hz,1H),8.03(s,1H),7.91(dd,J=8.3,3.6Hz,1H),7.43(t,J=9.3Hz,1H),7.28(d,J=8.8Hz,1H),7.20(d,J=2.4Hz,1H),7.13(s,1H),7.00(d,J=8.7Hz,1H),4.68(s,1H),3.18(s,3H).
实施例26
5-氯-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物26)
第一步:5-氯-N-(4-氟-3-(甲硫基)苯基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(100mg,0.26mmol)溶解在DMF(2mL)中,加入4-(三氟甲氧基)苯酚(93.3mg,0.52mmol),加入碳酸铯(170mg,0.52mmol),油浴100℃下搅拌2小时。停止反应,反应液加水(10ml)淬灭,水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(5ml×2)洗涤、无水硫酸钠干燥,过滤后滤液旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=10:1)分离纯化得到标题化合物(100mg,收率:71.4%,白色固体)。
MS(ESI):m/z 540.0[M+H]+
第二步:5-氯-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物26)
将5-氯-N-(4-氟-3-(甲硫基)苯基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(87mg,0.16mmol)溶解在甲醇(1mL)中,加入碳酸铵(46.4mg,0.48mmoL)和二乙酸碘苯(206mg,0.64mmoL),室温搅拌0.5小时。停止反应,反应液旋干拌样,经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离得产物粗品,粗品经反相制备色谱(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(28.96mg,收率:31.5%,白色固体)。
MS(ESI):m/z 571.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.86(s,1H),7.86(s,1H),7.71(d,J=23.3Hz,2H),7.48(d,J=8.0Hz,3H),7.34(s,2H),7.02(s,1H),4.75(s,1H),3.13(s,3H).
实施例27
5-氯-2-(4,4-二氟氮杂环庚烷-1-基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物27)
第一步:5-氯-2-(4,4-二氟氮杂环庚烷-1-基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺
将4,4-二氟氮杂环庚烷盐酸盐(80mg,0.58mmol)溶解在DMF(2mL)中,加入5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(80mg,0.20mmol)和碳酸铯(210mg,0.64mmol),100℃油浴中搅拌12小时。停止反应,反应液加水(5mL)淬灭,水相用乙酸乙酯(10mL×3)萃取,合并有机相,经饱和食盐水(5mL×2)洗涤、无水硫酸钠干燥、真空旋干得到得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=3:1)分离纯化得到标题化合物(45mg,收率:43.2%,黄色固体)。
MS(ESI):m/z 497.1[M+H]+
第二步:5-氯-2-(4,4-二氟氮杂环庚烷-1-基)-N-(4-氟-3-(S-甲亚胺代磺酰基)苯基)-4-三氟甲基苯甲酰胺(化合物27)
将5-氯-2-(4,4-二氟氮杂环庚烷-1-基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(45mg,0.09mmol)溶于甲醇(1.5mL)中,加入碳酸氢铵(27mg,0.28mmol)、二乙酸碘苯(118mg, 0.36mmol),室温搅拌反应1小时。停止反应,将反应液旋干,加水(2ml)淬灭,水相用乙酸乙酯(5ml×3)萃取,合并有机相,经饱和食盐水(3ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经反相制备色谱(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(2.85mg,收率:0.59%,白色固体)。
MS(ESI):m/z 539.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.87(s,1H),8.27(dd,J=6.4,2.5Hz,1H),8.01–7.90(m,1H),7.69(s,1H),7.46(t,J=9.3Hz,1H),7.34(s,1H),4.70(s,1H),3.40(brs,4H),3.20(s,3H),2.25(brs,2H),2.07(brs,2H),1.81(brs,2H).
化合物27手性异构体拆分:
(S)-5-氯-2-(4,4-二氟氮杂环庚烷-1-基)-N-(4-氟-3-(S-甲亚胺代磺酰基)苯基)-4-三氟甲基苯甲酰胺
(R)-5-氯-2-(4,4-二氟氮杂环庚烷-1-基)-N-(4-氟-3-(S-甲亚胺代磺酰基)苯基)-4-三氟甲基苯甲酰胺
化合物27经手性制备分离柱(色谱柱:250*25mm,10μm;流动相:A-Supercritical CO2;B-MEOH(+0.1%7.0mol/l Ammonia in MEOH),45%B比例洗脱,流速:70mL/min,柱温:室温)分离得到单一构型化合物27E1(较短保留时间)和化合物27E2(较长保留时间)。
实施例28
5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物28)
第一步:5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺
将(3aR,6aS)-5,5-二氟八氢环戊二烯并[c]吡咯盐酸盐(100mg,0.681mmol)溶解在DMF(1mL)中,加入5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(130mg,0.340mmol)和碳酸铯(322mg,1.021mmol),氮气置换,100℃油浴中搅拌反应16小时。停止反应,反应液加水(5mL)淬灭,水相用乙酸乙酯(10mL×3)萃取,合并有机相,经饱和食盐水(5mL×2)洗涤、无水硫酸钠干燥,过滤后真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=2:1)分离纯化得到标题化合物(100mg,收率:57.8%,黄色固体)。
MS(ESI):m/z 510.1[M+H]+
第二步:5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物28)
将5-氯-2-((3aR,6aS)-5,5-二氟六氢环戊二烯并[c]吡咯-2(1H)-基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(100mg,0.196mmol)溶于甲醇(1.5mL)中,加入碳酸铵(56.6mg,0.589mmol)和乙二酸碘苯(253.1mg,0.785mmol),室温搅拌反应1小时。停止反应,将反应液旋干,加饱和碳酸氢钠溶液水(2ml)淬灭,水相用乙酸乙酯(5ml×3)萃取,合并有机相,经饱和食盐水(3ml×2)洗涤、无水硫酸钠干燥,过滤后滤液真空旋干得到粗品,粗品经反相制备色谱(洗脱体系:氨水,水,乙腈)纯化得到标题化合物(10mg,收率:9.4%,白色固体)。
MS(ESI):m/z 539.8[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.88(s,1H),8.29(d,J=3.8Hz,1H),8.01–7.85(m,1H),7.65(s,1H),7.45(t,J=9.3Hz,1H),7.14(s,1H),4.68(s,1H),3.39(d,J=7.0Hz,2H),3.23–3.19(m,5H),2.84(brs,2H),2.39–2.25(m,2H),1.96(brs,2H).
实施例29
5-氯-2-((4,4-二氟环己基)氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物29)
第一步:5-氯-2-((4,4-二氟环己基)氧基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-氟-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(80mg,0.21mmol)和4,4-二氟环己-1-醇(114mg,0.84mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸铯(205mg,0.63mmol),油浴100℃反应1小时。停止反应,向反应液中加饱和氯化铵溶液(5mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液减压旋干,经柱层析(硅胶,石油醚:乙酸乙酯=10:1)分离纯化得到标题化合物(120mg,收率:92.3%,白色固体)。
MS(ESI):m/z 498.1[M+H]+
第二步:5-氯-2-((4,4-二氟环己基)氧基)-N-(4-氟-3-(S-甲基亚胺代磺酰基)苯基)-4-(三氟甲基)苯甲酰胺(化合物29)
将5-氯-2-((4,4-二氟环己基)氧基)-N-(4-氟-3-(甲硫基)苯基)-4-(三氟甲基)苯甲酰胺(105mg,0.211mmol)用甲醇(2mL)溶解,加入碳酸铵(61mg,0.634mmol)、二乙酸碘苯(272.2mg,0.845mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化,而后经高效液相色谱制备(洗脱体系:甲酸,水,乙腈)纯化得到标题化合物(71.14mg,收率:64.1%,白色固体)。
MS(ESI):m/z 529.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.72(s,1H),8.27(s,1H),7.86(s,2H),7.64(s,1H),7.44(t,J=9.1Hz,1H),4.94(brs,1H),4.68(s,1H),3.18(s,3H),1.88(brs,8H).
实施例30
5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(1-(S-甲基亚胺代磺酰基)环丙基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物30)
第一步:(4-溴吡啶-2-基)甲醇
将4-溴吡啶-2-甲酸甲酯(2g,9.258mmol)溶解在乙醇(50mL)中,加入硼氢化钠(771mg,20.381mmol),氮气置换后室温反应16小时。停止反应,真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=2:1)分离纯化得到标题化合物(1.23g,收率:70.7%,白色固体)。
MS(ESI):m/z 188.0[M+H]+
第二步:4-溴-2-(氯甲基)吡啶
将(4-溴吡啶-2-基)甲醇(9.7g,51.59mmol)溶解在二氯甲烷(60mL)中,0℃下缓慢滴加二氯亚砜(9.21mg,77.414mmol),室温反应3小时。停止反应,向反应液加饱和碳酸氢钠溶液(100mL)淬灭,二氯甲烷(100mL×3)萃取,无水硫酸钠干燥,过滤后滤液真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=9:1)分离纯化得到标题化合物(10g,收率:85.1%,黄色油状物)。
MS(ESI):m/z 206.0[M+H]+
第三步:4-溴-2-((甲硫基)甲基)吡啶
将4-溴-2-(氯甲基)吡啶(10g,48.433mmol)溶解在N,N-二甲基甲酰胺(80mL)中,0℃下缓慢加入甲硫醇钠(4.07mg,58.076mmol),0℃下反应2小时。停止反应,将反应液倒入水(100mL)中,乙酸乙酯(100mL×3)萃取,合并有机相经饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,过滤后滤液真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=5:1)分离纯化得到标题化合物(9.3g,收率:88.1%,黄色油状物)。
MS(ESI):m/z 218.1[M+H]+
第四步:4-溴-2-((甲基亚磺酰基)甲基)吡啶
将4-溴-2-((甲硫基)甲基)吡啶(4g,18.34mmol)溶解在甲醇(50mL)和水(10mL)的混合溶剂中,0℃下缓慢加入高碘酸钠(3.93g,18.34mmol),室温反应5小时。停止反应,真空旋除溶剂,经柱层析(硅胶,二氯甲烷:甲醇=9:1)分离纯化得到标题化合物(4.1g,收率:96.47%,白色固体)。
MS(ESI):m/z 234.0[M+H]+
第五步:N-(((4-溴吡啶-2-基)甲基)(甲基)(氧代)-λ6-硫亚基)-2,2,2-三氟乙酰胺
将4-溴-2-((甲基亚磺酰基)甲基)吡啶(2g,8.584mmol)溶解在二氯甲烷(20mL)中,加入2,2,2-三氟乙酰胺(1.94g,17.17mmol)、氧化镁(1.34g,34.33mmol)、乙酸铑(190g,0.43mmol)和醋酸碘苯(5.53g,17.17mmol),氮气置换后室温反应16小时。停止反应,真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(1.18g,收率:40.2%,黄色油状物)。
MS(ESI):m/z 345.0[M+H]+
第六步:((4-溴吡啶-2-基)甲基)(亚氨基)(甲基)-λ6-亚砜
将N-(((4-溴吡啶-2-基)甲基)(甲基)(氧代)-λ6-硫亚基)-2,2,2-三氟乙酰胺(1.13g,3.285mmol)溶解在甲醇(10mL)中,加入碳酸钾(2.27g,16.424mmol),室温反应1小时。停止反应,真空旋除溶剂,经柱层析(硅胶,二氯甲烷:甲醇=8:1)分离纯化得到标题化合物(660mg,收率:81.08%,黄色油状物)。
MS(ESI):m/z 249.0[M+H]+
第七步:(((4-溴吡啶-2-基)甲基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((4-溴吡啶-2-基)甲基)(亚氨基)(甲基)-λ6-亚砜(660mg,2.66mmol)溶解在二氯甲烷(10mL)中,加入吡啶(632mg,8mmol),降温至0℃后缓慢滴加氯甲酸苄酯(1.09g,6.4mmol),而后室温反应3小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的稀盐酸调至偏酸性,乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(900mg,收率:88.58%,黄色油状物)。
MS(ESI):m/z 383.1[M+H]+
第八步:(1-(4-溴吡啶-2-基)环丙基)(亚氨基)(甲基)-λ6-亚砜
将(((4-溴吡啶-2-基)甲基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(680mg,1.78mmol)溶解在四氢呋喃(12.8mL)中,加入四丁基溴化铵(57.4mg,0.178mmol)和1,2-二溴乙烷9(1.34g,7.12mmol),缓慢滴入氢氧化钠溶液(50%,3.2mL),而后升温至60℃反应16小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的稀盐酸调至偏酸性,乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,二氯甲烷:甲醇=20:1)分离纯化得到标题化合物(400mg,收率:81.96%,黄色固体)。
MS(ESI):m/z 275.0[M+H]+
第九步:((1-(4-溴吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将(1-(4-溴吡啶-2-基)环丙基)(亚氨基)(甲基)-λ6-亚砜(400mg,1.46mmol)溶解在二氯甲烷(6mL)中,加入吡啶(346mg,4.38mmol),降温至0℃后缓慢滴加氯甲酸苄酯(623g,3.65mmol),而后室温反应3小时。停止反应,真空旋除溶剂,加水(10mL)稀释,用1N的稀盐酸调至偏酸性,乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(400mg,收率:67.11%,黄色油状物)。
MS(ESI):m/z 409.0[M+H]+
第十步:((1-(4-((二苯基亚甲基)氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((1-(4-溴吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(330mg,0.81mmol)和二苯甲亚胺(220mg,1.21mmol)溶解在1,4-二氧六环(5mL)中,加入三(二亚苄基丙酮)二钯(18.2mg,0.08mmol)、(9,9-二甲基-9H-呫吨-4,5-二基)双(二苯基膦)(94mg,0.162mmol)和碳酸铯(790mg,2.43mmol),氮气置换后,100℃油浴下反应2小时。停止反应,向反应液中加饱和氯化铵溶液(10mL)淬灭,用乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:2)分离纯化得到标题化合物(400mg,收率:97.08%,黄色油状物)。
MS(ESI):m/z 510.5[M+H]+
第十一步:((1-(4-氨基吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((1-(4-((二苯基亚甲基)氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(420mg,0.825mmol)溶解在四氢呋喃(4mL)中,缓慢滴入1N的稀盐酸(4mL),室温反应1小时。停止反应,真空旋除溶剂,用1N的氢氧化钠溶液调至偏碱性,乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥。过滤后滤液真空旋除溶剂得到标题化合物(270mg,收率:68.4%,白色固体)。
MS(ESI):m/z 346.1[M+H]+
第十二步:((1-(4-(5-氯-2-氟-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((1-(4-氨基吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(205mg,0.594mmol)和5-氯-2-氟-4-(三氟甲基)苯甲酸(144.1mg,0.594mmol)溶解在吡啶(4mL)中,氮气置换后用冰盐浴降温至-10℃,缓慢滴加三氯氧磷(274mg,1.783mmol),室温反应2小时。将反应液倒入冰水(30mL)中,用1N的稀盐酸调至偏酸性,乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,有机相无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:4)分离纯化得到标题化合物(300mg,收率:88.76%,黄色固体)。
MS(ESI):m/z 570.0[M+H]+
第十三步:((1-(4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((1-(4-(5-氯-2-氟-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(75mg,0.132mmol)和4-氟-2-甲基苯酚(20mg,0.158mmol)用N,N-二甲基甲酰胺(2mL)溶解,加入碳酸铯(86mg,0.264mmol),升温至100℃反应2小时。停止反应,向反应液中加饱和氯化铵溶液(10mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(10mL×3)洗涤,收集有机相无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:3)分离纯化得到标题化合物(110mg,收率:93.2%,黄色油状物)。
MS(ESI):m/z 676.1[M+H]+
第十四步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(1-(S-甲基亚胺代磺酰基)环丙基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物30)
将((1-(4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(70mg,0.103mmol)用三氟乙酸(1.5mL)溶解,升温至60℃反应3小时。停止反应,真空旋除溶剂,用1N的氢氧化钠溶液调至偏碱性,乙酸乙酯(2mL×3)萃取,合并有机相用饱和食盐水(5mL×3)洗涤,无水硫酸钠干燥,真空旋除溶剂。经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(32mg,收率:57.14%,白色固体)。
MS(ESI):m/z 542.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.06(s,1H),8.46(d,J=5.6Hz,1H),8.08(s,1H),7.87(d,J=1.4Hz,1H),7.60(dd,J=5.5,1.9Hz,1H),7.24–7.18(m,1H),7.11–7.08(m,3H),3.71(s,1H),2.91(s,3H),2.16(s,3H),1.81–1.73(m,1H),1.48–1.41(m,1H),1.31–1.20(m,2H).
实施例31
5-氯-N-(2-(1-(S-甲基亚胺代磺酰基)环丙基)吡啶-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物31)
第一步:((1-(4-(5-氯-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((1-(4-(5-氯-2-氟-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(75mg,0.132mmol)和4-(三氟甲氧基)苯酚2(47mg,0.264mmol)用N,N-二甲基甲酰胺(2mL)溶解,加入碳酸钾(37mg,0.264mmol),升温至70℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(10mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:3)分离纯化得到标题化合物(80mg,收率:83%,黄色油状物)。
MS(ESI):m/z 728.0[M+H]+
第二步:5-氯-N-(2-(1-(S-甲基亚胺代磺酰基)环丙基)吡啶-4-基)-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰胺(化合物31)
将((1-(4-(5-氯-2-(4-(三氟甲氧基)苯氧基)-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(70mg,0.102mmol)用三氟乙酸(1.5mL)溶解,升温至60℃反应3小时。停止反应,真空旋除溶剂,用1N的氢氧化钠溶液调至偏碱性,乙酸乙酯(2mL×3)萃取,合并有机相用饱和食盐水(5mL×3)洗涤,无水硫酸钠干燥,真空旋除溶剂。经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(39.27mg,收率:68.42%,白色固体)。
MS(ESI):m/z 594.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),8.44(d,J=5.6Hz,1H),8.12(s,1H),7.83(d,J=1.5Hz,1H),7.57(dd,J=5.5,1.8Hz,1H),7.50(s,1H),7.40(d,J=8.5Hz,2H),7.24–7.19(m,2H),3.71(s,1H),2.90(s,3H),1.76(brs,1H),1.44(brs,1H),1.30–1.19(m,2H).
实施例32
5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(1-(S-甲基亚胺代磺酰基)环丙基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物32)
第一步:((1-(4-(5-氯-2-(4,4-二氟氮杂环庚-1-基)-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((1-(4-(5-氯-2-氟-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(75mg,0.132mmol)和4,4-二氟氮杂环庚烷盐酸盐(113mg,0.66mmol)用N,N-二甲基甲酰胺(2mL)溶解,加入碳酸铯(300mg,0.923mmol),升温至100℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(10mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,经柱层析(硅胶,石油醚:乙酸乙酯=1:3)分离纯化得到标题化合物(80mg,收率:88%,黄色油状物)。
MS(ESI):m/z 685.1[M+H]+
第二步:5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-(1-(S-甲基亚胺代磺酰基)环丙基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物32)
将((1-(4-(5-氯-2-(4,4-二氟氮杂环庚-1-基)-4-(三氟甲基)苯甲酰氨基)吡啶-2-基)环丙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(70mg,0.102mmol)用三氟乙酸(1.5mL)溶解,升温至60℃反应3小时。停止反应,真空旋除溶剂,用1N的氢氧化钠溶液调至偏碱性,乙酸乙酯(2mL×3)萃取,合并有机相用饱和食盐水(5mL×3)洗涤,无水硫酸钠干燥,真空旋除溶剂;经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(19.21mg,收率:34.12%,白色固体)。
MS(ESI):m/z 551.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.97(s,1H),8.47(d,J=5.5Hz,1H),7.85(s,1H),7.73–7.67(m,2H),7.36(s,1H),3.69(s,1H),3.42–3.33(m,4H),2.92(s,3H),2.24(brs,2H),2.11–2.00(m,2H),1.79(brs,3H),1.49–1.43(m,1H),1.31–1.21(m,2H).
实施例33
5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(2-(S-甲基亚胺代磺酰基)乙氧基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物33)
第一步:4-溴-2-(2-(甲硫基)乙氧基)吡啶
将2-(甲硫基)乙烷-1-醇(3.14g,34.10mmol)溶解在N,N-二甲基甲酰胺(60mL)中,0℃下加入钠氢(1.70g,42.60mmol),反应0.5小时,氮气下加入4-溴-2-氟吡啶(5.00g,28.40mmol),0℃下继续反应2小时。停止反应,缓慢加水(250mL)淬灭反应,水相用乙酸乙酯(250ml×3)萃取,合并有机相,经饱和食盐水(250ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=97:3)分离纯化得到标题化合物(5.00g,收率:71.0%,无色油状物)。
MS(ESI):m/z 247.9[M+H]+
第二步:(2-((4-溴吡啶-2-基)氧基)乙基)(亚氨基)(甲基)-λ6-亚砜
将4-溴-2-(2-(甲硫基)乙氧基)吡啶(4.5g,18.13mmol)溶解在甲醇(50mL)中,加入碳酸铵(5.2g,54.40mmol),然后冰浴下加入二乙酸碘苯(23.3g,72.53mmol),氮气置换,室温搅拌反应2小时。停止反应,反应液加水(200ml)淬灭,水相用乙酸乙酯(200ml×3)萃取,合并有机相,经饱和食盐水(200ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品反向柱分离(流动相:水:甲醇=85:15)分离纯化得到标题化合物(900mg,收率:17.7%,黄色油状物)。
MS(ESI):m/z 279.0[M+H]+
第三步:((2-((4-溴吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将(2-((4-溴吡啶-2-基)氧基)乙基)(亚氨基)(甲基)-λ6-亚砜(700mg,2.50mmol)溶解在DCM(10mL)中,加入吡啶(297mg,3.76mmol),氮气置换,冰浴下加CbzCl(513mg,3.00mmol),室温搅拌反应2小时。停止反应,反应液加水(50mL)淬灭,水相用乙酸乙酯(50mL×3)萃取,合并有机相,经饱和食盐水(5mL×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(800mg,收率:77.2%,无色油状物)。
MS(ESI):m/z 415.0[M+H]+
第四步:((2-((4-((二苯基亚甲基)氨基)吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6- 硫亚基)氨基甲酸苄基酯
将((2-((4-溴吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(700mg,1.69mmol)溶于二氧六环(10mL)中,加入二苯基亚胺(458mg,2.54mmol),三(二亚苄基丙酮)二钯(154mg,0.16mmol),(9,9-二甲基-9H-呫吨-4,5-二基)双(二苯基膦)(195mg,0.33mmol)和碳酸铯(1654mg,5.08mmol),氮气置换,100℃搅拌反应1.5小时。停止反应,将反应液冷却加水(100mL),水相用乙酸乙酯(100ml×3)萃取,合并有机相,经饱和食盐水(3ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经薄层层析板分离(展开剂体系:乙酸乙酯)纯化得到标题化合物(550mg,收率:63.2%,黄色固体)。
MS(ESI):m/z 514.2[M+H]+
第五步:((2-((4-氨基吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((2-((4-((二苯基亚甲基)氨基)吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(500mg,0.97mmol),溶解在四氢呋喃(2mL)中,加水(1mL)和盐酸(1mL),氮气置换,室温下反应1.5小时,真空旋干得到粗品标题化合物(400mg,白色固体),直接投下一步反应。
MS(ESI):m/z 350.1[M+H]+
第六步:((2-((4-(5-氯-2-氟-4-(三氟甲基)苯甲酰胺)吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((2-((4-氨基吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(450mg,1.16mmol),溶解在吡啶(15mL)中,加5-氯-2-氟-4-(三氟甲基)苯甲酸(283mg,1.16mmol),氮气置换,冰浴下加三氯氧磷(536mg,3.50mmol),室温反应2小时。停止反应,冰浴下反应液加水(100mL),水相用乙酸乙酯(100ml×3)萃取,合并有机相,经饱和食盐水(10ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(450mg,收率:67.0%,黄色固体)。
MS(ESI):m/z 574.1[M+H]+
第七步:((2-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺)吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯
将((2-((4-(5-氯-2-氟-4-(三氟甲基)苯甲酰胺)吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(80mg,0.13mmol)溶解在DMF(2mL)中,加碳酸钾(90mg,2.70mmol),加4-氟-2-甲基苯酚(35mg,0.27mmol),氮气置换,70℃反应12小时。停止反应,反应液加水(15mL),水相用乙酸乙酯(15ml×3)萃取,合并有机相,经饱和食盐水(10ml×2)洗涤、无水硫酸钠干燥、真空旋干得到粗品,粗品经硅胶柱层析(流动相:石油醚:乙酸乙酯=2:1) 分离纯化得到标题化合物(100mg,收率:86.4%,黄色固体)。
MS(ESI):m/z 680.1[M+H]+
第八步:5-氯-2-(4-氟-2-甲基苯氧基)-N-(2-(2-(S-甲基亚胺代磺酰基)乙氧基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物33)
将((2-((4-(5-氯-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺)吡啶-2-基)氧基)乙基)(甲基)(氧代)-λ6-硫亚基)氨基甲酸苄基酯(80mg,0.118mmol)用三氟乙酸(1.5mL)溶解,升温至60℃反应3小时。停止反应,真空旋除溶剂,用1N的氢氧化钠溶液调至偏碱性,乙酸乙酯(2mL×3)萃取,合并有机相用饱和食盐水(5mL×3)洗涤,无水硫酸钠干燥,过滤后滤液真空旋除溶剂,经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(34.87mg,收率:54.22%,白色固体)。
MS(ESI):m/z 546.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.75(s,1H),8.06(s,1H),7.67(d,J=7.5Hz,1H),7.22(d,J=9.0Hz,1H),7.09(d,J=5.8Hz,3H),6.82(d,J=1.9Hz,1H),6.42(dd,J=7.5,2.2Hz,1H),4.21(t,J=6.8Hz,2H),3.88(s,1H),3.43(td,J=6.8,2.7Hz,2H),2.91(s,3H),2.16(s,3H).
实施例34
2-(4-乙酰基-1,4-二氮杂环庚烷-1-基)-5-氯-N-(2-(S-甲基磺酰亚胺基)吡啶-4-基)-4-三氟甲基苯甲酰胺(化合物34)
第一步:2-(4-乙酰基-1,4-二氮杂环庚烷-1-基)-5-氯-N-(2-甲硫基)吡啶-4-基)-4-三氟甲基苯甲酰胺
将5-氯-2-氟-N-(2-甲硫基)吡啶-4-基)-4-三氟甲基苯甲酰胺(120mg,0.33mmol)用N,N-二甲基甲酰胺(2mL)溶解,加入1-(1,4-二氮杂环庚烷-1-基)乙烷-1-酮(140.8mg,0.99mmol)和碳酸铯(322.7mg,0.99mmol),氮气置换,100℃反应16小时。停止反应,向反应液加水,用乙酸乙酯萃取(10mL×3),合并有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤后滤液真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=1:3)分离纯化得到标题化合物(90mg,收率:56.3%,白色固体)。
MS(ESI):m/z 487.0[M+H]+
第二步:2-(4-乙酰基-1,4-二氮杂环庚烷-1-基)-5-氯-N-(2-(S-甲基磺酰亚胺基)吡啶-4-基)-4-三氟甲基苯甲酰胺(化合物34)
将2-(4-乙酰基-1,4-二氮杂环庚烷-1-基)-5-氯-N-(2-甲硫基)吡啶-4-基)-4-三氟甲基苯甲酰胺(90mg,0.18mmol)用甲醇(1mL)溶解,再加入碳酸铵(18mg,0.54mmol)和醋酸碘苯(60.3mg,0.72mmol),室温反应1小时。停止反应,将反应液用真空旋除溶剂,经柱层析(硅胶,二氯甲烷:甲醇=20:1)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(2mg,收率:2.1%,白色固体)。
MS(ESI):m/z 518.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ11.30(s,1H),8.62(d,J=5.4Hz,1H),8.40(d,J=3.2Hz,1H),7.84(d,J=5.5Hz,1H),7.73(d,J=17.0Hz,1H),7.37(d,J=10.9Hz,1H),4.38(s,1H),3.62–3.54(m,2H),3.49–3.34(m,6H),3.15(s,3H),1.89(s,3H),1.74(brs,2H).
实施例35
4-乙酰基-1,4-二氮杂环戊-5-氯-N-(4-氟-3--S-甲磺酰亚胺基苯基)-4-三氟甲基苯甲酰胺(化合物35)
第一步:4-乙酰基-1,4-二氮杂环戊-5-氯-4-氟-3-甲硫基苯甲酰胺
将5-氯-2-氟-N-4-氟-3-甲硫基苯甲酰胺(100mg,0.262mmol)和1-(1,4-二氮杂环戊烯基)乙酮(75mg,0.525mmol)溶解在N,N-二甲基甲酰胺(2mL)中,加入碳酸铯(171mg,0.525mmol),油浴100℃反应16小时。停止反应,向反应液中加入饱和氯化铵溶液(10mL)淬灭,用乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(116mg,收率:88%,黄色油状物)。
MS(ESI):m/z 504.1[M+H]+
第二步:4-乙酰基-1,4-二氮杂环戊-5-氯-N-(4-氟-3--S-甲磺酰亚胺基苯基)-4-三氟甲基苯甲酰胺(化合物35)
将4-乙酰基-1,4-二氮杂环戊-5-氯-4-氟-3-甲硫基苯甲酰胺(96mg,0.19mmol)用甲醇(2mL)溶解,加入碳酸铵(54.78mg,0.57mmol)、二乙酸碘苯(244.8mg,0.76mmol),室温反应1小时。停止反应,真空旋除溶剂,由柱层析(硅胶,乙酸乙酯)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(15.76mg,收率:14.85%,白色固体)。
MS(ESI):m/z 535.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.92(s,1H),8.28(d,J=4.3Hz,1H),8.03–7.91(m,1H),7.67(d,J=16.7Hz,1H),7.45(t,J=9.3Hz,1H),7.35(d,J=9.6Hz,1H),4.70(s,1H),3.62–3.35(m,8H),3.20(s,3H),1.90(s,3H),1.76(brs,2H).
实施例36
5-氯-2-(4-(2,2-二氟乙基)-1,4-二氮杂-1-基)-N-(2-(S-甲基磺酰亚胺基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
第一步:4-(4-氯-2-((2-(甲硫基)吡啶-4-基)氨基甲酰基)-5-(三氟甲基)苯基)-1,4-二氮杂-1-羧酸叔丁酯
将5-氯-2-氟-N-2-甲硫基吡啶-4-基-4-三氟甲基苯甲酰胺(200mg,0.55mmol)和1,4-二氮杂-1-羧酸叔丁酯(220.2mg,1.1mmol)溶解在N,N-二甲基甲酰胺(3mL)中,加入碳酸铯(358.4mg,1.1mmol),油浴100℃反应16小时。停止反应,向反应液中加饱和氯化铵溶液(10mL)淬灭,用乙酸乙酯(30mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液经柱层析(硅胶,石油醚:乙酸乙酯=7:3)分离纯化得到标题化合物(215mg,收率:72%,黄色油)。
MS(ESI):m/z 545.1[M+H]+
第二步:5-氯-2-(1,4-二氮杂-1-基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将4-(4-氯-2-((2-(甲硫基)吡啶-4-基)氨基甲酰基)-5-(三氟甲基)苯基)-1,4-二氮杂-1-羧酸叔丁酯(205mg,0.377mmol)用二氯甲烷(6mL)溶解,加入三氟乙酸(2ml),室温反应1小时。停止反应,真空旋除溶剂,得到标题化合物(337mg,收率:98%,黄色油状物)。
MS(ESI):m/z 445.1[M+H]+
第三步:5-氯-2-(4-(2,2-二氟乙基)-1,4-二氮杂-1-基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
将5-氯-2-(1,4-二氮杂-1-基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(337mg)和2,2-二氟乙基三氟甲磺酸酯(96.34mg,0.45mmol)用四氢呋喃(1.5mL)溶解,加入三乙胺(91.5μl)油浴50℃反应16小时。停止反应,真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=6.5:3.5)分离纯化得到标题化合物(148mg,收率:44.85%,黄色油状物)。
MS(ESI):m/z 509.1[M+H]+
第四步:5-氯-2-(4-(2,2-二氟乙基)-1,4-二氮杂-1-基)-N-(2-(S-甲基磺酰亚胺基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺
5-氯-2-(4-(2,2-二氟乙基)-1,4-二氮杂-1-基)-N-(2-(甲硫基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(114mg,0.212mmol)用甲醇(1mL)溶解,然后加入碳酸铵(11.31mg,0.118mmol)和醋酸碘苯(50.25mg,0.156mmol),室温反应1小时。停止反应,真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=6.5:3.5)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(91mg,收率:67.4%,黄色油状物)。
MS(ESI):m/z 540.1[M+H]+
实施例37
5-氯-N-(2-甲磺酰亚胺基)吡啶-4-基)-2-(4-(2,2,2-三氟乙基)-1,4-二氮杂环戊烷-1-基)-4-三氟甲基苯甲酰胺(化合物37)
第一步:5-氯-N-2-甲硫基吡啶-4-基-2-(2,2,2-三氟乙基)-1,4-二氮杂环戊烷-1-基)-4-三氟甲基苯甲酰胺
将5-氯-2-(1,4-二氮杂环戊烷-1-基)-N-(2-甲硫基)吡啶-4-基)-4-三氟甲基苯甲酰胺(100mg,0.23mmol)和2,2,2-三氟乙基三氟甲磺酸酯(208.89mg,0.9mmol)用四氢呋喃(2.5mL)溶解,加入三乙胺(186.19mg,1.84mmol),油浴50℃反应16小时。停止反应,真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=7:3)分离纯化得到标题化合物(68mg,收率:56.7%,黄色油状物)。
MS(ESI):m/z 527.1[M+H]+
第二步:5-氯-N-(2-甲磺酰亚胺基)吡啶-4-基)-2-(4-(2,2,2-三氟乙基)-1,4-二氮杂环戊烷-1-基)-4-三氟甲基苯甲酰胺(化合物37)
将5-氯-N-2-甲硫基吡啶-4-基-2-(2,2,2-三氟乙基)-1,4-二氮杂环戊烷-1-基)-4-三氟甲基苯甲酰胺(45mg,0.086mmol)用甲醇(1mL)溶解,然后加入碳酸铵(24.7mg,0.257mmol)和醋酸碘苯(110.8mg,0.344mmol),室温反应1小时。停止反应,真空旋除溶剂,经柱层析(硅胶,纯乙酸乙酯)分离纯化,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(57mg,收率:92.3%,黄色油状物)。
MS(ESI):m/z 558.1[M+H]+
实施例38
5-氯-N-(2-((二甲基(氧代)-λ6-亚硫基)氨基)吡啶-4-基)-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺(化合物38)
第一步:(4-氨基吡啶-2-基)亚胺基)二甲基-6-砜
将2-氯-4-硝基吡啶(1000mg,6.31mmol)用无水1,4-二氧六环(20mL)溶解,加入二甲基亚磺酰亚胺(881mg,9.46mmol),碳酸铯(6.2g,18.93mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(730mg,1.26mmol)和三[二亚苄基丙酮]二钯(0)(577mg,0.63mmol)在氮气保护下加热到100℃搅拌反应过夜。停止反应,将反应液旋干,经柱层析(硅胶,石油醚:乙酸乙酯=10:1)纯化得到标题化合物(1.2g,收率:88%,黄色固体)。
MS(ESI):m/z 216.1[M+H]+
第二步:(4-氨基吡啶-2-基)亚氨基)二甲基-λ6-磺胺酮
将二甲基(4-硝基吡啶-2-基)亚氨基)-λ6-磺胺酮(600mg,2.79mmol)用无水甲醇(10mL)溶解,加入钯碳(180mg),氢气置换后室温反应16小时。停止反应,将反应液过滤,滤液真空旋除溶剂,经柱层析(硅胶,石油醚:乙酸乙酯=1:1)分离纯化得到标题化合物(460mg,收率:89%,白色固体)。
MS(ESI):m/z 186.2[M+H]+
第三步:5-氯-N-(2-((二甲基(氧代)-λ6-磺胺亚基)氨基)吡啶-4-基)-2-氟-4-(三氟甲基)苯甲酰胺
将(4-氨基吡啶-2-基)亚氨基)二甲基-λ6-磺胺酮(120mg,0.648mmol)和5-氯-2-氟-4-(三氟甲基)苯甲酸(157.17mg,0.648mmol)用吡啶(2mL)溶解,氮气置换后降温至-10℃加入三氯氧磷(248.64mg,1.62mmol),室温反应2小时。停止反应,向反应液中加入冰水(5mL)淬灭,用1N的盐酸将溶液调至酸性,用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤后滤液真空旋除溶剂,经柱层析(硅胶,二氯甲烷:无水甲醇=9:1)分离纯化得到标题化合物(263.7mg,收率:98%,黄色油状物)。
MS(ESI):m/z 410.0[M+H]+
第四步:5-氯-N-(2-((二甲基(氧代)-λ6-亚硫基)氨基)吡啶-4-基)-2-(4-氟-2-甲基苯氧基)-4-(三氟甲基)苯甲酰胺(化合物38)
将5-氯-N-(2-((二甲基(氧代)-λ6-磺胺亚基)氨基)吡啶-4-基)-2-氟-4-(三氟甲基)苯甲酰胺(132mg,0.324mmol)和4-氟-2-甲基苯酚(81.64mg,0.65mmol)用N,N-二甲基甲酰胺(2mL)溶解,加入碳酸铯(316.7mg,0.972mmol),油浴100℃反应2小时。停止反应,将反应液过滤,而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(38.25mg,收率:22.9%,白色固体)。
MS(ESI):m/z 516.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.71(s,1H),8.03(s,1H),7.99(d,J=5.7Hz,1H),7.21(d,J=9.0Hz,1H),7.10–7.08(m,4H),7.03(d,J=5.7Hz,1H),6.93(s,1H),3.34(s,3H),3.32(s,3H),2.16(s,3H).
实施例39
5-氯-2-(4,4-二氟氮杂环庚-1-基)-N-(2-((二甲基(氧代)-λ6-磺胺亚基)氨基)吡啶-4-基)-4-(三氟甲基)苯甲酰胺(化合物39)
将5-氯-N-(2-((二甲基(氧代)-λ6-磺胺亚基)氨基)吡啶-4-基)-2-氟-4-(三氟甲基)苯甲酰胺(132mg,0.324mmol)和4,4-二氟氮杂环丁烷盐酸盐(322.4mg,1.944mmol)用N,N-二甲基甲酰胺(2mL)溶解,加入碳酸铯(1.056g,3.24mmol),油浴100℃下反应16小时。停止反应,将反应液过滤,滤液而后经高效液相色谱制备(洗脱体系:碳酸氢铵,水,乙腈)纯化得到标题化合物(39.52mg,收率:23.3%,黄色固体)。
MS(ESI):m/z 525.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.66(s,1H),7.99(d,J=5.5Hz,1H),7.64(s,1H),7.33(s,1H),7.03(d,J=8.8Hz,2H),3.41–3.34(m,10H),2.23(brs,2H),2.10–2.01(m,2H),1.80–1.78(m,2H).
生物学测试评价
测试例A、本发明化合物对钠离子通道1.8(NaV1.8)的阻滞活性
实验目的:化合物对CHO细胞稳定表达的人源Nav1.8离子通道抑制率测定
实验方法:
DMSO溶解化合物配制为储备液,用细胞外液(140mM NaCl,3.5mM KCl,1mM MgCl2,2mM CaCl2,10mM D-glucose,10mM HEPES,1.25mM NaH2PO4 2H2O,pH=7.4(NaOH))稀释至特定浓度用于手动膜片钳测试。
用微电极拉制仪将毛细玻璃管拉制成记录电极。在倒置显微镜下操纵微电极操纵仪将记录电极接触到细胞上,给予负压抽吸,形成GΩ封接。形成GΩ封接后进行快速电容补偿,然后继续给予负压,吸破细胞膜,形成全细胞记录模式。然后进行慢速电容的补偿并记录膜电容及串联电阻。不给予漏电补偿。当全细胞记录的Nav1.8电流稳定后开始给药,每个药物浓度作用至5min(或者电流至稳定)后检测下一个浓度,每一个测试化合物检测两个浓度。
全细胞膜片钳记录Nav1.8钠电流的电压刺激方案如下:当形成全细胞封接后细胞电压钳制于-120mV维持30ms。钳制电压除极至0mV维持50ms,然后电压恢复至-50mV(具体电压参考IV测试的半失活电压),维持8s。接着细胞膜电位恢复至-120mV,维持20ms,然后再除极至0mV,维持50ms,最后恢复至钳制电压-120mV,维持30ms。每隔20s重复采集数据。观察药物对钠电流峰值的作用。试验数据由EPC-10放大器(HEKA)进行采集并储存于PatchMaster(HEKA)软件中。
实验结果:

测试例B、大鼠静注或灌胃定量本发明化合物的药代动力学评价
实验目的:该测试例的目的是评价本发明化合物的药代动力学性质
发明人在大鼠体内对本发明的化合物进行了药代动力学评估。其中,动物信息详见表2。
表2:本发明受试动物信息表
实验方法:
将本发明化合物以5%DMSO+5%tween80+90%生理盐水溶液形式,对受试动物进行给药,给药前动物禁食12h,自由饮水。对于静脉注射给药组,给药剂量为1mg/kg;灌胃给药,给药剂量为10mg/kg。动物给药后在以下时间点静脉取血(取血量约0.3mL):0.083、0.25、0.5、1.0、2.0、4.0、8.0和24h,采血管内预先加入EDTA-K2作为抗凝剂,血样在6800g下低温离心6分钟,收集血浆,并于-80℃下保存。
LC-MS/MS测定的样品制备:将20μL血浆样品用400μL含有100ng/mL IS(IS为甲苯磺丁脲)的甲醇进行蛋白质沉淀。将混合物涡旋1分钟,然后以18000g离心7分钟。将400μL上清液转移到96孔板。2μL上清液进行LC-MS/MS分析。分析结果表明,本发明化合物在大鼠内均具有良好的药代动力学性质。
测试例C、小鼠静注或灌胃定量本发明化合物的药代动力学评价
实验目的:该测试例的目的是评价本发明化合物的药代动力学性质
发明人在小鼠体内对本发明的化合物进行了药代动力学评估。其中,动物信息详见表3。
表3:本发明受试动物信息表
实验方法:
将本发明化合物以5%DMSO+5%tween80+90%生理盐水溶媒形式,对受试动物进行给药,给药前动物禁食12h,自由饮水。对于静脉注射给药组,给药剂量为1mg/kg;灌胃给药,给药剂量为10mg/kg。动物给药后在以下时间点静脉取血(取血量约0.3mL):0.083、0.25、0.5、1.0、2.0、4.0、8.0和24h,采血管内预先加入EDTA-K2作为抗凝剂,血样在6800g下低温离心6分钟,收集血浆,并于-80℃下保存。
LC-MS/MS测定的样品制备:将15μL血浆样品用300μL含有100ng/mL IS(IS为甲苯磺丁脲)的甲醇进行蛋白质沉淀。将混合物涡旋1分钟,然后以18000g离心10分钟。将300μL上清液转移到96孔板。2μL上清液进行LC-MS/MS分析。分析结果表明,本发明化合物在小鼠内均具有良好的药代动力学性质。

Claims (39)

  1. 通式(I’)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体:
    其中:
    X为N或CRx
    Y为N或CRy
    Z为N或CRz
    W为N或CRw
    G为N或CRg
    条件是X、Y和Z中至多两者同时为N;
    Rx、Ry和Rz各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1和-L1-R1,其中Rx、Ry和Rz中至少一者为-L1-R1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rw和Rg各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    -L1-为-(CRaRb)m-,在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Ra和Rb与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
    R1为-S(O)qNR’R”、-S(O)(=NR’)R4或-N=S(O)R4R4’;
    A选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经s个R2取代基取代;
    -L2-选自键、-O-、-NR’-、-S-、-C(O)-和-(CRcRd)n-,在化合价允许的情况下,-(CRcRd)n-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Rc和Rd与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
    R’选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R”为-L3-B;
    B选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经t个R5取代基取代;
    -L3-为-(CReRf)p-,在化合价允许的情况下,-(CReRf)p-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Re和Rf与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
    R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R4和R4’各自独立地选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R5各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R6选自选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Ra、Rb、Rc、Rd、Re和Rf独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1和Rs2独立地选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    m为0、1、2、3、4、5或6;
    n为0、1、2、3、4、5或6;
    p为0、1、2、3、4、5或6;
    t为1、2、3、4或5;
    s为1、2、3、4、5、6、7或8;
    q为1或2。
  2. 通式(I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体:
    其中:
    X为N或CRx
    Y为N或CRy
    Z为N或CRz
    W为N或CRw
    G为N或CRg
    条件是X、Y和Z中至多两者同时为N;
    Rx、Ry和Rz各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1和-L1-R1,其中Rx、Ry和Rz中至少一者为-L1-R1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rw和Rg各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    -L1-为-(CRaRb)m-,在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Ra和Rb与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
    R1为-S(O)qNR’R”或-S(O)(=NR’)R4
    A选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经s个R2取代基取代;
    -L2-选自键、-O-、-NR’-、-S-、-C(O)-和-(CRcRd)n-,在化合价允许的情况下,-(CRcRd)n-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Rc和Rd与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
    R’选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R”为-L3-B;
    B选自C3-8环烷基、3-10元杂环基、C6-10芳基和5-10元杂芳基,其任选经t个R5取代基取代;
    -L3-为-(CReRf)p-,在化合价允许的情况下,-(CReRf)p-中的任意一个亚甲基单元任选且独立地被-NR’-、-O-、-S-和-C(O)-替代和/或Re和Rf与它们所连接的碳原子一起形成C3-8环烷基或3-10元杂环基;
    R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R4选自H、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R5各自独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R6选自选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Ra、Rb、Rc、Rd、Re和Rf独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基和C1-6卤代烷 基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1和Rs2独立地选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    m为0、1、2、3、4、5或6;
    n为0、1、2、3、4、5或6;
    p为0、1、2、3、4、5或6;
    t为1、2、3、4或5;
    s为1、2、3、4、5、6、7或8;
    q为1或2。
  3. 权利要求1或2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(II),
  4. 权利要求1-3中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中-L2-为-O-。
  5. 权利要求1-3中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中-L2-为键。
  6. 权利要求1-5中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中A选自苯基、C3-6环烷基、5-6元杂芳基和5-10元杂环基。
  7. 权利要求1-5中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中A选自环戊基、环己基、氮杂环己基、氧杂环己基、硫杂环己基、氮杂环庚基、二氮杂环庚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基、八氢并环戊二烯基、苯基、吡咯基、呋喃基、噻吩基、吡啶基、嘧啶基和哒嗪基。
  8. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中m为0。
  9. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    m为1、2、3、4或5,
    -(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-O-、-S-和-C(O)-替代;和/或Ra和Rb与它们所连接的碳原子一起形成C3-6环烷基和3-5元杂环基;
    Ra选自H、C1-6烷基和C1-6卤代烷基;
    Rb选自H、C1-6烷基和C1-6卤代烷基。
  10. 权利要求2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(III),
  11. 权利要求2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(IV)
    其中:
    X为N或CRx
    Y为N或CRy
    Rx和Ry各自独立地选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R”为-L3-B;
    B为5-7元杂环基,其任选经t个R5取代基取代;
    -L3-为-(CReRf)p-,在化合价允许的情况下,-(CReRf)p-中的任意一个亚甲基单元任选且独立地被-O-、-S-和-C(O)-替代;
    R2a、R2b、R2c、R2d、R2e、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R5各自独立地选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Re和Rf独立地选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1和Rs2独立地选H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    p为0、1、2或3;
    t为1、2或3。
  12. 权利要求11的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N;
    Rx选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R”为-L3-B;
    B为5-6元杂环基,其任选经t个R5取代基取代;
    -L3-为-(CReRf)p-;
    R2a、R2b、R2c、R2d、R2e、R3a、R3b、R3c和R3d独立地选自H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R5各自独立地选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Re和Rf独立地选自H、D、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    p为0、1、2或3;
    t为1、2或3;
    优选地,-L3-B选自
  13. 权利要求11的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N;
    Rx为H或D;
    R’为H;
    R”为-L3-B;
    B为5-6元杂环基,优选为氮杂环戊基或氮杂环己基;
    -L3-为-(CReRf)p-,p为0或1;
    R2a为C1-6烷基优选为甲基;
    R2b为H或D;
    R2c为卤素优选为F;
    R2d为H或D;
    R2e为H或D;
    R3a为H或D;
    R3b为C1-6卤代烷基优选为-CF3
    R3c为卤素优选为了Cl;
    R3d为H或D;
    Re选自H、D和C1-6烷基;
    Rf选自H、D和C1-6烷基;
    优选地,-L3-B选自
  14. 权利要求2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(V)
  15. 权利要求14的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为N或CRx
    Y为N或CRy
    Rx和Ry各自独立地选自H、D、卤素、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    -L2-为键、-O-、-S-或-NH-;
    A选自苯基、C3-8环烷基、5-6元杂芳基和5-10元杂环基,其任选经s个R2取代基取代;
    在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-O-、-S-和-C(O)-替代;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、 -ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Ra和Rb选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    或者Ra和Rb与它们所连接的碳原子一起形成C3-6环烷基和3-5元杂环基;
    Rs1和Rs2独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    m为0、1、2、3或4;
    s为1、2、3、4、5、6、7或8。
  16. 权利要求14的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx和Ry各自独立地选自H、D、卤素和C1-6烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    A选自和5-8元杂环基,其任选经s个R2取代基取代;
    在化合价允许的情况下,-(CRaRb)m-中的任意一个亚甲基单元任选且独立地被-O-替代;
    -L2-为键或-O-;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2各自独立地选自H、D、卤素、C1-6烷基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2a选自H、D、卤素、C1-6烷基、C1-6卤代烷基、-ORs1a和-SRs1a,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H或D;
    R2c为H、D、卤素、-ORs1c或-SRs1c
    R2d为H或D;
    R2e为H或D;
    R3a为H或D;
    R3b为C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3c为卤素;
    R3d为H或D;
    R4选自C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Ra和Rb选自H、D、卤素和C1-6烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    或者Ra和Rb与它们所连接的碳原子一起形成C3-6环烷基;
    Rs1各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1a选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1c选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    m为1、2或3;
    s为1或2。
  17. 权利要求14的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx为H;
    Ry为卤素;
    A选自和氮杂环庚烷基,其经s个R2取代;
    -(CRaRb)m-选自-CH2-、-O-CH2-CH2-、
    -L2-为键或-O-;
    R’为H;
    R2各自独立地选自H和卤素优选为F,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2a为H或C1-6烷基优选为甲基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H;
    R2c为卤素或-ORs1c
    R2d为H;
    R2e为H;
    R3a为H;
    R3b为C1-6卤代烷基优选为-CF3
    R3c为卤素优选为Cl;
    R3d为H;
    R4为C1-6烷基优选为甲基;
    Rs1c为H、C1-6烷基或C1-6卤代烷基,优选为-CF3
    s为1或2。
  18. 权利要求2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(VI):
  19. 权利要求18的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为N或CRx
    Y为N或CRy
    Rx和Ry各自独立地选自H、D、卤素、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基和-ORs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    -L2-为键、-O-、-S-或-NH-;
    A选自苯基、C3-8环烷基、5-10元杂芳基和5-10元杂环基,其任选经s个R2取代基取代;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、ORs1、SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1和Rs2独立地选H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    s为1、2、3、4、5、6、7或8。
  20. 权利要求18的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    A选自环戊基、环己基、氮杂环己基、氧杂环己基、硫杂环己基、氮杂环庚基、二氮杂环庚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基、八氢并环戊二烯基、苯基、吡咯基、呋喃基、噻吩基、 吡啶基、嘧啶基和哒嗪基,优选地,A与其上的取代基选自以下:
    其中,R2a、R2b、R2c和R2d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代。
  21. 权利要求18的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    -L2-为-O-;
    A选自环戊基和环己基,优选为环己基。
  22. 权利要求18的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    -L2-为键;
    A选自氮杂环庚基、二氮杂环庚基、八氢环戊二烯并吡咯基、八氢吡咯并吡咯基和八氢并环戊二烯基,优选地,在A为含氮杂环的情况下通过氮原子与苯基相连。
  23. 权利要求2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其为式(VII)
    其中:
    X为N或CRx
    Y为N或CRy
    Rx和Ry各自独立地选自H、D、卤素、C1-6烷基、C2-6烯基、C2-6炔基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2a、R2b、R2c、R2d、R2e、R3a、R3b、R3c和R3d独立地选自H、D、卤素、CN、C1-6烷基、C2-6烯基、C2-6炔基、C1-6卤代烷基、-ORs1、-SRs1、NRs1Rs2、-C(O)Rs1、-C(O)ORs1、-C(O)NRs1Rs2、-SORs1和-SO2Rs1,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1和Rs2独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
  24. 权利要求23的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为N或CRx
    Y为N或CRy
    Rx和Ry各自独立地选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R’选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2a选自H、D、卤素、-ORs1a、-SRs1a、NRs1aRs2a、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2c选自H、D、卤素、-ORs1c、-SRs1c、NRs1cRs2c、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2d为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2e为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3a为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3b选自H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3c选自H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3d为H、D、卤素、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R4选自H、C1-6烷基和C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1a、Rs2a、Rs1c和Rs2c各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
  25. 权利要求23的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx和Ry独立地选自H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R’为H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2a为-ORs1a、-SRs1a、NRs1aRs2a、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H、D或卤素;
    R2c为H、D、卤素、-ORs1c、-SRs1c或NRs1cRs2c,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2d为H、D或卤素;
    R2e为H、D或卤素;
    R3a为H、D或卤素;
    R3b为H、D、卤素、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3c为H、D或卤素;
    R3d为H、D或卤素;
    R4为H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1a、Rs2a、Rs1c和Rs2c各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    条件是当Y为N时,R2a不为C1-6烷基或C1-6卤代烷基。
  26. 权利要求23的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx和Ry独立地选自H、D或卤素;
    R’为H、C1-6烷基或C1-6卤代烷基;
    R2a为-ORs1a、-SRs1a、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H、D或卤素;
    R2c为H、D、卤素、-ORs1c或-SRs1c,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2d为H或D;
    R2e为H或D;
    R3a为H或D;
    R3b为C1-6卤代烷基;
    R3c为卤素;
    R3d为H或D;
    R4为C1-6烷基或C1-6卤代烷基;
    Rs1a和Rs1c独立地选自C1-6烷基或C1-6卤代烷基;
    条件是当Y为N时,R2a不为C1-6烷基或C1-6卤代烷基。
  27. 权利要求23的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx和Ry独立地选自H、D或卤素;
    R’为H;
    R2a为-ORs1a、-SRs1a或C1-6烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H或D;
    R2c为卤素、-ORs1c或-SRs1c,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2d为H或D;
    R2e为H或D;
    R3a为H或D;
    R3b为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3c为卤素;
    R3d为H或D;
    R4为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1a和Rs1c各自独立地选自H、C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
  28. 权利要求23的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx为H或D;
    Ry为H、D或卤素;
    R’为H;
    R2a为-ORs1a或-SRs1a,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R2b为H或D;
    R2c为卤素、-ORs1c或-SRs1c
    R2d为H或D;
    R2e为H或D;
    R3a为H或D;
    R3b为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    R3c为卤素;
    R3d为H或D;
    R4为C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代;
    Rs1a和Rs1c独立地选自C1-6烷基或C1-6卤代烷基,所述基团任选地被一个或多个氘取代,直至完全氘代。
  29. 权利要求23的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,其中:
    X为CRx
    Y为N或CRy
    Rx为H;
    Ry为卤素优选为F;
    R’为H;
    R2a为-ORs1a,所述基团任选地被一个或多个氘取代,直至完全氘代,优选为-OCD3
    R2b为H;
    R2c为卤素或-ORs1c,优选为F或-OCF3
    R2d为H;
    R2e为H;
    R3a为H;
    R3b为C1-6卤代烷基优选为-CF3
    R3c为卤素,优选为Cl;
    R3d为H;
    R4为C1-6烷基;
    Rs1a为C1-6烷基;
    Rs1c为C1-6卤代烷基。
  30. 权利要求1或2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,所述化合物选自:



  31. 药物组合物,其包含根据权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
  32. 根据权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物在制备用作电压-门控钠通道抑制剂的药物中的用途。
  33. 根据权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物在制备用作钠离子通道1.8(NaV1.8)抑制剂的药物中的用途。
  34. 权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物,其用作电压-门控钠通道抑制剂。
  35. 权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物,其用作钠离子通道1.8(NaV1.8)抑制剂。
  36. 一种在受试者中抑制电压-门控钠通道的方法,包括向所述受试者给药根据权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物。
  37. 一种在受试者中抑制钠离子通道1.8(NaV1.8)的方法,包括向所述受试者给药根据权利要求1-30中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,或包含其的药物组合物。
  38. 根据权利要求32或33的用途或根据权利要求34或35的化合物或药物组合物的用途或根据权利要求36或37的方法,其中所述电压-门控钠通道抑制剂用于治疗选自以下的疾病:急性、慢性、神经性、或炎性疼痛、关节炎、偏头痛、丛集性头痛、三叉神经痛、疱疹性神经痛、一般性神经痛、癫痫或癫痫病症、神经变性疾病、精神障碍如焦虑和抑郁、双相性精神障碍、肌強直、心律失常、运动障碍、神经内分泌障碍、共济失调、多发性硬化、肠易激综合征、失禁、内脏痛、骨关节炎痛、疱瘆后神经痛、糖尿病性神经病、神经根痛、坐骨神经痛、背痛、头痛、颈痛、严重或顽固性疼痛、伤害性疼痛、贯穿性疼痛、手术后疼痛、癌症疼痛、中风、脑缺血、外伤性脑损伤、肌萎缩性侧索硬化、应激或运动诱发的心绞痛、心悸、高血压、偏头痛或异常的胃肠活动。
  39. 根据权利要求32-38中任一项的用途、化合物或药物组合物的用途或方法,其中所述疾病选自神经根痛、坐骨神经痛、背痛、头痛、颈痛、顽固性疼痛、急性疼痛、手术后疼痛、背痛、耳鸣或癌症疼痛。
PCT/CN2023/112584 2022-08-12 2023-08-11 用作电压-门控钠通道抑制剂的化合物 WO2024032774A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210971471.1 2022-08-12
CN202210971471 2022-08-12
CN202211124412.7 2022-09-15
CN202211124412 2022-09-15

Publications (1)

Publication Number Publication Date
WO2024032774A1 true WO2024032774A1 (zh) 2024-02-15

Family

ID=89851022

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/112584 WO2024032774A1 (zh) 2022-08-12 2023-08-11 用作电压-门控钠通道抑制剂的化合物

Country Status (3)

Country Link
CN (1) CN117586180A (zh)
TW (1) TW202406894A (zh)
WO (1) WO2024032774A1 (zh)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101466665A (zh) * 2006-04-11 2009-06-24 沃泰克斯药物股份有限公司 适用作电压-门控钠通道抑制剂的组合物
CN101855210A (zh) * 2007-10-11 2010-10-06 沃泰克斯药物股份有限公司 用作电压-门控钠通道抑制剂的酰胺类
CN105683157A (zh) * 2013-07-19 2016-06-15 沃泰克斯药物股份有限公司 用作钠通道调节剂的磺酰胺类
CN112457294A (zh) * 2021-01-27 2021-03-09 上海济煜医药科技有限公司 一种作为NaV1.8阻滞剂的化合物及其制备方法和用途
WO2021188880A1 (en) * 2020-03-20 2021-09-23 Amathus Therapeutics, Inc. Benzenesulfonamide derivatives as trap1 modulators and uses thereof
CN114031518A (zh) * 2020-12-08 2022-02-11 成都海博为药业有限公司 一种苄胺或苄醇衍生物及其用途
CN114437062A (zh) * 2020-04-30 2022-05-06 成都海博为药业有限公司 一种可作为钠通道调节剂的化合物及其用途
WO2023150201A2 (en) * 2022-02-04 2023-08-10 Latigo Biotherapeutics, Inc. Sodium channel blocking compounds, derivatives thereof, and methods of their use

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101466665A (zh) * 2006-04-11 2009-06-24 沃泰克斯药物股份有限公司 适用作电压-门控钠通道抑制剂的组合物
CN101855210A (zh) * 2007-10-11 2010-10-06 沃泰克斯药物股份有限公司 用作电压-门控钠通道抑制剂的酰胺类
CN105683157A (zh) * 2013-07-19 2016-06-15 沃泰克斯药物股份有限公司 用作钠通道调节剂的磺酰胺类
WO2021188880A1 (en) * 2020-03-20 2021-09-23 Amathus Therapeutics, Inc. Benzenesulfonamide derivatives as trap1 modulators and uses thereof
CN114437062A (zh) * 2020-04-30 2022-05-06 成都海博为药业有限公司 一种可作为钠通道调节剂的化合物及其用途
CN114031518A (zh) * 2020-12-08 2022-02-11 成都海博为药业有限公司 一种苄胺或苄醇衍生物及其用途
CN112457294A (zh) * 2021-01-27 2021-03-09 上海济煜医药科技有限公司 一种作为NaV1.8阻滞剂的化合物及其制备方法和用途
WO2023150201A2 (en) * 2022-02-04 2023-08-10 Latigo Biotherapeutics, Inc. Sodium channel blocking compounds, derivatives thereof, and methods of their use

Also Published As

Publication number Publication date
CN117586180A (zh) 2024-02-23
TW202406894A (zh) 2024-02-16

Similar Documents

Publication Publication Date Title
RU2476430C2 (ru) Производные аминодигидротиазина, замещенные циклической группой
KR102469161B1 (ko) Rho-관련 단백질 키나아제 억제제, Rho-관련 단백질 키나아제 억제제를 함유하는 약학 조성물, 제조 방법 및 약학 조성물의 용도
WO2022135572A1 (zh) 一种五并五元环衍生物及其在医药上的应用
US10167281B2 (en) Substituted thiazole or oxazole P2X7 receptor antagonists
EP2842939B1 (en) Benzamide derivative
JP7207634B2 (ja) P2x3及び/又はp2x2/3受容体アンタゴニスト、それを含む医薬組成物及びその使用
CN109071516B (zh) 新颖的化合物
US8895562B2 (en) Piperazin-1-yl-trifluoromethyl-substituted-pyridines as fast dissociating dopamine 2 receptor antagonists
CN116583502A (zh) 二环-杂环衍生物及其作为食欲素-2受体激动剂的用途
WO2024032774A1 (zh) 用作电压-门控钠通道抑制剂的化合物
CN110997641A (zh) 新的白纹黄单胞菌毒素衍生物、其用途及合成
EP3398941A1 (en) Heterocyclic p2x7 antagonists
WO2021228216A1 (zh) 可用作RORγ调节剂的联芳基类化合物
ES2816204T3 (es) Antagonistas del receptor p2x7 derivados de N-[2-(4-fenoxipiperidin-1-il)-2-(1,3-tiazol-5-il)etil]benzamida y N-[2-(4-benciloxipiperidin-1-il)-2-(1,3-tiazol-5-il)etil]benzamida sustituidas
EP3290417A1 (en) 2-chloro-n-[2-(1,3-thiazol-5-yl)ethyl]-5-(5-fluoropyrimidin-2-yl)-benzamides and their use as p2x7 receptor antagonists
EP4169915A1 (en) Crystalline form of compound
WO2023202687A1 (zh) 一种吡嗪酮衍生物及其在医药上的应用
WO2022223022A1 (zh) 一种并环杂环衍生物及其在医药上的应用
CN111902401A (zh) 受体抑制剂、包含其的药物组合物及其用途
WO2023202642A1 (zh) Rho相关蛋白激酶抑制剂及其制备和应用
WO2023031319A1 (en) 2,4-dihydro-3h-1,2,4-triazol-3-one p2x7 antagonists

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: 23851994

Country of ref document: EP

Kind code of ref document: A1