WO2020019994A1 - 一类具有神经保护作用的化合物及其制备方法和用途 - Google Patents

一类具有神经保护作用的化合物及其制备方法和用途 Download PDF

Info

Publication number
WO2020019994A1
WO2020019994A1 PCT/CN2019/095648 CN2019095648W WO2020019994A1 WO 2020019994 A1 WO2020019994 A1 WO 2020019994A1 CN 2019095648 W CN2019095648 W CN 2019095648W WO 2020019994 A1 WO2020019994 A1 WO 2020019994A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
hydrogen
mmol
group
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2019/095648
Other languages
English (en)
French (fr)
Inventor
梁波
金秋
陈焕明
Original Assignee
上海挚盟医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海挚盟医药科技有限公司 filed Critical 上海挚盟医药科技有限公司
Priority to CN201980048641.5A priority Critical patent/CN112469708B/zh
Publication of WO2020019994A1 publication Critical patent/WO2020019994A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • 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
    • 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
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • 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
    • 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
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • 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
    • A61P25/24Antidepressants
    • 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
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/04Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/061,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles
    • C07D271/071,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention belongs to the field of biomedicine, and in particular, the invention relates to a class of compounds having neuroprotective effects.
  • the compounds are effective in preventing and treating diseases and conditions affected by neuron damage, such as pain, stroke, depression, etc .
  • Stroke is an acute cerebrovascular disease, including ischemic and hemorrhagic stroke.
  • the incidence of ischemic stroke accounts for 60% to 70% of the total number of strokes. Stroke has the characteristics of high morbidity, high mortality, and high disability. So far, clinically effective therapeutic drugs are rare.
  • NMDAR N-methyl-D aspartate receptor
  • PSD-95 post-synaptic dense protein 95
  • nNOS neuronal nitric oxide synthase
  • PSD-95 neuronal nitric oxide synthase
  • the object of the present invention is to provide a new class of compounds with neuroprotective effects.
  • An object of the present invention is to provide a method for preparing the above-mentioned compound.
  • the present invention also provides a method for preventing or treating a neuron damage-related disease, which method comprises administering to a subject in need thereof a compound of the present invention or a pharmaceutical composition containing the compound of the present invention.
  • a first aspect of the present invention provides a compound represented by Formula A or a pharmaceutically acceptable salt thereof:
  • Z is N, C (-OH), C (-OR 9 ) or CH;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 are each independently selected from: hydrogen, halogen, amino (-NH 2 ), R 9 C (O) O-, (R 9 ) (R 10 ) NC (O) S-, -OR 11 OR 10 , hydroxyl, carboxyl (-COOH), nitro, cyano, C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkyl, halo C 1-6 alkoxy, C 3-6 cycloalkyl, halo C 3-6 cycloalkyl, C 1-6 alkylamino, R 9 C (O) NH-;
  • X and Y are each independently and differently selected from: methylene, oxygen, carbonyl, sulfonyl, and imino; wherein the methylene and imino are independently optionally selected from one or more of: halogen, C 1- 6 alkyl, halo C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 ring Substituted by alkyl, C 3-6 cycloalkenyl, C 6-10 aryl, C 3-10 heteroaryl substituents;
  • Each R 9 and R 10 is independently selected from: C 1-6 alkyl, halo C 1-6 alkyl, C 3-6 cycloalkyl, halo C 3-6 cycloalkyl;
  • R 11 is C 1-6 alkylene or C 2-6 alkenylene.
  • Z is N or C (-OH);
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 are each independently selected from hydrogen, halogen, amino (-NH 2 ), hydroxyl, carboxy (-COOH), nitro, cyan Alkyl, C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkyl, halo C 1-6 alkoxy, C 3-6 cycloalkyl, halo C 3-6 Cycloalkyl, C 1-6 alkylamino;
  • X and Y are each independently and differently selected from methylene, oxygen, carbonyl, sulfonyl, and imino; wherein methylene and imino are independently optionally selected from one or more of C 1-6 alkyl, Halo C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, C 6-10 aryl, C 3-10 heteroaryl are substituted.
  • Z is N, C (-OH), C (-OR 9 ) or CH;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 are each independently selected from: hydrogen, halogen, amino, hydroxyl, carboxyl, nitro, cyano, C 1-6 alkyl , C 1-6 alkoxy, halo C 1-6 alkyl, halo C 1-6 alkoxy, C 3-6 cycloalkyl, halo C 3-6 cycloalkyl, C 1-6 Alkylamino, R 9 C (O) O-, R 9 C (O) NH-, (R 9 ) (R 10 ) NC (O) S-, -OR 11 OR 10 ;
  • Each R 9 and R 10 is independently selected from: C 1-6 alkyl, halo C 1-6 alkyl, C 3-6 cycloalkyl, halo C 3-6 cycloalkyl;
  • Z is N, C (-OH), C (-OR 9 ) or CH;
  • R 1 , R 2 , R 3 , R 4 , and R 5 are each independently selected from: hydrogen, halogen, amino, hydroxyl, R 9 C (O) O-, R 9 C (O) NH-, (R 9 ) ( R 10 ) NC (O) S-, -OR 11 OR 10 ; the definitions of R 9 , R 10 , and R 11 are as described above;
  • R 6 , R 7 and R 8 are hydrogen
  • X and Y are each independently and differently selected from: methylene, oxygen, and imino.
  • Z is N, C (-OH), C (-OR 9 ) or CH;
  • R 1 , R 2 , R 3 , R 4 , and R 5 are each independently selected from: hydrogen, halogen, amino, hydroxyl, R 9 C (O) O-, (R 9 ) (R 10 ) NC (O) S- ; R 9 and R 10 are methyl groups;
  • R 6 , R 7 and R 8 are hydrogen
  • X and Y are each independently and differently selected from: methylene, oxygen, and imino.
  • ring A is
  • R 6 , R 7 and R 8 are hydrogen
  • X and Y are each independently and differently selected from: methylene, oxygen, and imino;
  • R 1 is a hydroxyl group
  • R 2 and R 4 are chlorine
  • R 3 and R 5 are hydrogen
  • R 9 is methyl, R 2 and R 4 are chlorine, R 3 and R 5 are hydrogen;
  • R 9 and R 10 are methyl groups, R 2 and R 4 are chlorine, and R 3 and R 5 are hydrogen;
  • R 1 is a hydroxyl group.
  • R 2 and R 4 are chlorine; and R 3 and R 5 are hydrogen.
  • X and Y are each independently and differently selected from methylene, oxygen, and imino.
  • the compound is selected from the group consisting of:
  • a second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising one or more pharmaceutically acceptable carriers or diluents and a compound described in the first aspect of the present invention or a pharmaceutically acceptable salt thereof.
  • the disease or disorder is selected from the group consisting of neuropathic pain, migraine, inflammatory pain, chronic pain, stroke, brain injury, depression, Alzheimer's disease, epilepsy, and emotional disorders. Diseases, neurodegenerative diseases.
  • the present invention provides a method for preparing the compound represented by Formula A or a pharmaceutically acceptable salt thereof, but is not limited to the following methods:
  • W and Q are the two groups that react. These include, but are not limited to, dehydration of hydroxy groups to ethers, dehydration condensation reactions of aldehyde amino groups, coupling reactions of halogens and alkynes, reactions of hydroxy substituted halogens, reactions of amine substituted halogens, dehydration condensation reactions of carboxyl groups and amine groups, esters Condensation reaction of amine group and amine group, condensation reaction of sulfonyl chloride group and amine group, etc.
  • G includes, but is not limited to, COOH, CHO, CN, F, Cl, Br, I, B (OH) 2 , NH 2 , OMs, OTf, etc.
  • ring A is formed by the functional group conversion or coupling reaction of G;
  • the reaction uses a condensation agent such as CDI to perform a condensation reaction to obtain a product.
  • the compound of the invention has good neuroprotective effect and can be used for treating diseases such as pain and stroke.
  • the present invention has been completed on this basis.
  • each chiral carbon atom may optionally be in the R configuration or the S configuration, or a mixture of the R configuration and the S configuration.
  • C 1-6 alkoxy when used alone or as part of another substituent refers to C 1-6 alkyl-oxy-, including, for example, methoxy, ethoxy, propoxy, iso Propoxy, butoxy, isobutoxy, sec-butoxy, tert-butoxy, or similar groups.
  • C 3-6 cycloalkyl when used alone or as part of another substituent refers to a cyclic alkyl group having 3 to 6 carbon atoms, including, for example, cyclopropyl, cyclobutyl, cyclopentyl Group, cyclohexyl, or similar groups.
  • C 3-6 cycloalkenyl refers to a cyclic alkenyl group having 3-6 carbon atoms, which may have one or two alkenyl groups, including, for example, a ring Butenyl, cyclopentenyl, cyclohexenyl, or similar groups.
  • C 2-6 alkenyl refers to a straight or branched chain alkenyl group having 2 to 6 carbon atoms, which may have one or more alkenyl groups, such as Including vinyl, propenyl, butenyl, or similar groups.
  • C 2-6 alkynyl when used alone or as part of another substituent refers to a branched or straight chain alkynyl group having 2-6 carbon atoms, which may have one or more alkynyl groups, Examples include ethynyl, propynyl, butynyl, or similar groups.
  • halogen is fluorine, chlorine, bromine or iodine.
  • halo refers to fluoro, chloro, bromo or iodo.
  • C 6-10 aromatic ring or “C 6-10 aryl group” means an aromatic ring having 6 to 10 carbon atoms, such as a benzene ring, a naphthalene ring, and the like.
  • a 3-8 membered saturated or unsaturated heterocyclic ring containing one or more (e.g., 2, 3, or 4) selected from N, S, and O” means having 3-8 (preferably 5- 6) heterocyclic ring atoms containing one or more (e.g., 2, 3, or 4) heteroatoms selected from N, S, and O, including, for example,
  • CDI refers to N, N'-carbonyldiimidazole.
  • amino has the following structure: -NH 2.
  • amino has the structure: -NH-.
  • C 1-6 alkylamino has the structure: -NH (C 1-6 alkyl).
  • C 1-6 alkylene refers to a group formed by the loss of a hydrogen atom of "C 1-6 alkyl”, and includes, for example, methylene, ethylene, propylene, isopropylidene Propyl, butylene, etc.
  • C 2-6 alkenylene refers to a group formed by "C 2-6 alkenyl” losing one hydrogen atom, and includes, for example, vinylidene, propenylene, isopropenylene, butylene. Alkenyl, etc.
  • the compound of the present invention refers to a compound represented by Formula A or a stereoisomer or optical isomer thereof, or a pharmaceutically acceptable salt thereof.
  • the "pharmaceutically acceptable salt” refers to a salt formed by the compound of the present invention and a pharmaceutically acceptable inorganic acid and organic acid.
  • preferred inorganic acids include (but are not limited to): hydrochloric acid, hydrobromic acid , Phosphoric acid, nitric acid, sulfuric acid, trifluoroacetic acid (TFA);
  • preferred organic acids include (but are not limited to): formic acid, acetic acid, propionic acid, succinic acid, naphthalenedisulfonic acid (1,5), sulfite, Oxalic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, valeric acid, diethylacetic acid, malonic acid, succinic acid, fumaric acid, pimelic acid, adipic acid, maleic acid, malic acid, sulfamic acid , Phenylpropionic acid, gluconic acid, ascorbic acid, niacin, isonicotin
  • stereoisomer or “optical isomer” means that the chiral carbon atom involved in the compound of the present invention may be in the R configuration, S configuration, or a combination thereof.
  • Patent CN104045552A reports compound 04006 as a neuroprotective agent, and as a decoupling agent of nNOS and PSD-95, the compound shows good neuron protection in vitro cell activity experiments.
  • the exposed carboxyl group on the compound 04006 caused excessive molecular polarity and poor lipophilicity. It could not pass the blood-brain barrier (BBB) and was not of value for continued development as a clinical therapeutic drug.
  • BBB blood-brain barrier
  • the present invention enables the compound to effectively pass through the blood-brain barrier (such as the compound 04017 of the present invention), and plays a good neuroprotective role in related animal pharmacodynamic experiments.
  • the method for preparing the compound of formula A according to the present invention is described in more detail below, but these specific methods do not limit the present invention in any way.
  • the compounds of the present invention can also be conveniently prepared by combining various synthetic methods described in the specification or known in the art, and such combinations can be easily performed by those skilled in the art to which the present invention belongs.
  • the preparation process of the compound of the present invention is as follows.
  • the raw materials and reagents used therein can be purchased through commercial channels unless otherwise specified.
  • W is selected from the group consisting of aldehyde, amino, hydroxyl, halogen, C 2-6 alkynyl, halomethylene, carboxyl, ester, and sulfonyl;
  • Q is selected from the group consisting of amino, aldehyde, hydroxyl, halogen, C 2-6 alkynyl, halomethylene, carboxyl, ester, and sulfonyl;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , Z, X, Y and ring A are as described above.
  • ring A is constructed by the functional group conversion or coupling reaction of G;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , Z, X, Y and ring A are as described above.
  • the compound of the present invention Since the compound of the present invention has excellent neuroprotective effects, the compound of the present invention and a pharmaceutical composition containing the compound of the present invention as a main active ingredient can be used to treat, prevent, and alleviate diseases related to neuronal damage. According to the prior art, the compounds of the present invention can be used to treat (but not limited to) the following diseases: inflammatory pain, neuropathic pain, migraine, stroke, neurodegenerative diseases, and the like.
  • the pharmaceutical composition of the present invention contains the compound of the present invention in a safe and effective amount and a pharmacologically acceptable excipient or carrier.
  • a pharmaceutical composition contains from 1 to 2000 mg of a compound / agent of the invention, and more preferably from 5 to 200 mg of a compound / agent of the invention.
  • the "one dose” is a capsule or tablet.
  • the "pharmaceutically acceptable carrier” refers to one or more compatible solid or liquid fillers or gel substances, which are suitable for human use and must have sufficient purity and low enough toxicity. "Compatibility” herein means that each component of the composition can blend with the compound of the present invention and each other without significantly reducing the pharmacological effect of the compound.
  • pharmaceutically acceptable carriers are cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, and solid lubricants (such as stearic acid).
  • Magnesium stearate calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween ), Wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • vegetable oils such as soybean oil, sesame oil, peanut oil, olive oil, etc.
  • polyols such as propylene glycol, glycerin, mannitol, sorbitol, etc.
  • emulsifiers such as Tween
  • Wetting agents such as sodium lauryl sulfate
  • the administration method of the compound or pharmaceutical composition of the present invention is not particularly limited, and representative administration methods include (but are not limited to): oral, rectal, parenteral (intravenous, intramuscular, or subcutaneous), and 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) a filler or compatibilizer, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, such as hydroxymethyl cellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) humectants, For example, glycerol; (d) disintegrating agents, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) Absorption accelerators, such as quaternary amine compounds; (g) wetting agents, such as cetyl alcohol and g
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared using coatings and shell materials, such as casings and other materials known in the art. They may contain opaque agents and the release of the active compound or compounds in such a composition may be released in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be used are polymeric substances and waxes. If necessary, the active compound may also be microencapsulated with one or more of the aforementioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs.
  • liquid dosage forms may include inert diluents conventionally used in the art, such as water or other solvents, solubilizers, and emulsifiers.
  • inert diluents conventionally used in the art, such as water or other solvents, solubilizers, and emulsifiers.
  • ethanol isopropanol
  • ethyl carbonate ethyl acetate
  • propylene glycol 1
  • 3-butanediol dimethylformamide
  • oils especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil, and sesame oil, or mixtures thereof.
  • composition may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweeteners, flavoring agents, and perfumes.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweeteners, flavoring agents, and perfumes.
  • the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these, and the like.
  • suspending agents for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these, and the like.
  • compositions for parenteral injection may include physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous vehicles, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • Dosage forms of the compounds of the invention for topical administration include ointments, powders, patches, sprays and inhalants.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants that may be required if necessary.
  • the compounds of the invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • a safe and effective amount of a compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dose when administered is a pharmaceutically considered effective dose.
  • a mammal such as a human
  • the dosage is usually 1 to 2000 mg, preferably 5 to 500 mg.
  • the specific dosage should also consider factors such as the route of administration, the patient's health, etc., which are all within the skill of a skilled physician.
  • the present invention provides a class of compounds with novel neuroprotective effects.
  • the compounds have better neuroprotective activity, better pharmacokinetic properties, better in vivo pharmacological effects, and better safety.
  • the compounds of the present invention can effectively cross the blood-brain barrier (BBB).
  • BBB blood-brain barrier
  • the compounds of the present invention are expected to be useful in the treatment and prevention of diseases and conditions affected by nerve damage.
  • Step 1 5-((3,5-Dichloro-2-hydroxybenzyl) amino) cyanopyridine (Compound 3)
  • Step 2 5-((3,5-Dichloro-2-hydroxybenzyl) amino) -N-hydroxymethylpyridine (Compound 4)
  • reaction solution was quenched with ice water, then extracted with ethyl acetate (3x 100mL), washed with saturated brine (2x 100mL), dried over anhydrous sodium sulfate, and filtered and concentrated to give compound 3 (2.66g, 82%) as white solid.
  • reaction solution was quenched with ice water, then extracted with ethyl acetate (3 ⁇ 50 mL), washed with saturated brine (2 ⁇ 50 mL), dried over anhydrous sodium sulfate, and then concentrated by filtration.
  • the basal medium of PC12 rat adrenal pheochromaffin cells is 5% (v / v) fetal bovine serum (Pan), 10% (v / v) horse serum (Gibco), and 1% (v / v) penicillin-streptomyces RPMI 1640 (Gibco) medium.
  • PC12 cells were seeded at a density of 2 ⁇ 10 4 cells / well in a 96-well plate pre-coated with polylysine (sigma). 24 hours after the cells were seeded, a basal medium (final compound concentration: 10 ⁇ M) containing the test compound was added and cultured for 1 hour.
  • OGD hypoxia-glucose
  • the OGD medium is a sugar-free 1640 medium with a final concentration of 10 mM sodium dithionite (Sigma).
  • Each group of OGD medium contains a test compound at a final concentration of 10 ⁇ M.
  • wash with basal medium for 5 minutes add basal medium containing the final concentration of the test compound to 10 ⁇ M and continue the culture.
  • the lactate dehydrogenase (LDH) leakage rate was measured using a CytoTox96 Non-Radioactive kit from Promega to assess the level of cell damage. According to the kit instructions, the LDH in the cell culture medium and the total LDH after cell lysis were determined, and the cell damage rate (Cytotoxicity%) was the percentage of LDH in the culture medium to the total LDH. The test results are shown in Table 1.
  • SD Sprague Dawley
  • a model of focal cerebral ischemia reperfusion (MCAO) in rats was prepared by internal carotid artery suture method. Rats were intraperitoneally injected with 7% trichloroacetaldehyde hydrate (6.0 mL / kg), and the depth of anesthesia was appropriate to severely stimulate the animals to respond slightly. The rats under anesthesia were restrained with rubber bands on the limbs (the hind limbs were fixed above the knee joint, the forelimbs were fixed above the wrist joint) and the head. Hair, alcohol disinfects the skin. The neck was cut in the middle, and the subcutaneous tissue was bluntly separated.
  • MCAO focal cerebral ischemia reperfusion
  • the proximal carotid end of the common carotid artery is clamped, all the mouth is made from the distal end of the ligature of the external carotid artery, the plug line is inserted, the bifurcation of the common carotid artery enters the internal carotid artery, and then it is slowly inserted until there is slight resistance (from the bifurcation 20mm), blocking all blood supply to the middle cerebral artery.
  • the external carotid artery was ligated with a suture fixed to the suture, and the skin was sutured and disinfected.
  • the experimental animals were divided into three groups: the compound 04017 group (5mg / kg) and the positive drug edaravone group (6mg / kg). After preparing the ischemic model, the animals were equally and blindly assigned to each group. The animals were administered intravenously once immediately after reperfusion, and the animals in the model group were given an equal volume of normal saline. Symptoms of neurological deficits were evaluated 24 hours after ischemia, and then the animals were sacrificed, brains were taken, stained, and the area of cerebral infarction was measured.
  • the modified Bederson 5-point system was used to evaluate the symptoms of neurological deficits.
  • the single-blind method was used to evaluate the symptoms of neurological deficits in rats after cerebral ischemia. That is, the experimenter marked the animals in groups and scored the neurodeficiency symptoms. The testers did not know the grouping of the animals. The scoring results of various marks were submitted to the designer, and the designer was unblinded to obtain the score of each animal in each experimental group.
  • the degree of cerebral infarction was measured by TTC staining. Animals were sacrificed with CO 2 after evaluation of the symptoms of neurological deficits, the brain was decapitated, the olfactory bulb, cerebellum, and lower brainstem were removed, and the blood on the surface of the brain was washed with physiological saline, and the remaining water on the surface was sucked off. After leaving at -20 ° C for 20 minutes, it was removed. Immediately make a coronal section perpendicular to the line of sight, and cut a slice every 2mm backward. Incubate the brain slices in 1% TTC staining solution (37 ° C for 20min). The normal brain tissue is stained dark red, and the ischemic brain The tissue was pale, and after flushing with physiological saline, the brain slices were quickly arranged in a row from front to back, and the residual water marks on the surface were blotted and photographed.
  • Quantitative data were expressed as mean ⁇ standard error.
  • One-way ANOVA was performed for each pharmacodynamic index using GraphPad Prism (6.01) software. After the variance test was significant, Fisher ’s LSD test was used to test for differences between groups. P ⁇ 0.05 was defined as having a significant difference.
  • SD Sprague Dawley
  • Compounds 40017 and 04006 were administered via tail vein injection at a dose of 2.5 mg / kg, and brain tissues were collected at time points 5 and 30 minutes (1 (Rats / time point), rinsed with physiological saline, blotted with filter paper, weighed, frozen and stored at -20 ° C until use.
  • the hydrophilic-COOH group of compound 04006 is converted into Later, while effectively improving the neuroprotective effect of the compound (comparing the test results of compounds 04006 and 04017 in Table 1), the compound can also effectively pass the blood-brain barrier (Table 3).

Landscapes

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

Abstract

一类具有神经保护作用的化合物及其制备方法和用途。具体地,公开了通式A 所示的化合物或其药学上可接受的盐。还公开了化合物的制备方法及其在神经保护方面的用途。

Description

一类具有神经保护作用的化合物及其制备方法和用途 技术领域
本发明属于生物医药领域,具体而言,本发明涉及一类具有神经保护作用的化合物,该类化合物对预防和治疗受神经元损伤影响的疾病和病症有效果,如疼痛、脑卒中、抑郁等。
背景技术
脑卒中是一种急性脑血管疾病,包括缺血性和出血性卒中,其中缺血性卒中的发病率占脑卒中总数的60%~70%。脑卒中具有发病率高、死亡率高和致残率高的特点,临床上迄今少见疗效确切的治疗药物。
研究表明,选择阻断N-甲基-D天冬氨酸受体(NMDAR)与突触后致密蛋白95(PSD-95)或者神经元型一氧化氮合成酶(nNOS)与PSD-95的相互作用,可以在不影响NMDAR和nNOS生理功能的前提下抑制一氧化氮(NO)的病理性释放,获得没有明显中枢神经系统副作用的安全有效的神经病理性疼痛的治疗药物。选择性阻断nNOS与PSD-95的相互作用,阻止NO的病理性释放,也是预防和治疗缺血性脑卒中等疾病更理想的方式。
目前尚没有针对以上作用机制的药物上市,开发新型有效的针对PSD-95的具有神经保护作用的药物是非常必要的。
发明内容
本发明的目的是提供一类新型的具有神经保护作用的化合物。
本发明的目的是提供上述化合物的制备方法。
本发明的目的还有提供上述化合物在作为神经保护剂的应用,用于治疗脑卒中。
本发明还提供了一种预防或治疗与神经元损伤相关疾病的方法,所述方法包括给需要的对象施用本发明化合物或含有本发明化合物的药物组合物。
本发明第一方面提供了式A所示的化合物或其药学上可接受的盐:
Figure PCTCN2019095648-appb-000001
其中:
Z为N、C(-OH)、C(-OR 9)或CH;
环A为取代或未取代的C 6-10芳环或取代或未取代的3-8元饱和或不饱和的含有一个或多个(例如2、3、或4个)选自N、S和O的杂环;所述取代是指被选自下组的取代基所取代:氧代(=O)、羟基、巯基;
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8分别独立选自:氢、卤素、氨基(-NH 2)、R 9C(O)O-、(R 9)(R 10)NC(O)S-、-OR 11OR 10、羟基、羧基(-COOH)、硝基、氰基、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基、卤代C 1-6烷氧基、C 3-6环烷基、卤代C 3-6环烷基、C 1-6烷基胺基、R 9C(O)NH-;
X和Y各自独立且不相同地选自:亚甲基、氧、羰基、磺酰基、亚氨基;其中亚甲基、亚氨基独立可选地被一个或多个选自:卤素、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烯基、C 6-10芳基、C 3-10杂芳基的取代基所取代;
各R 9、R 10分别独立选自:C 1-6烷基、卤代C 1-6烷基、C 3-6环烷基、卤代C 3-6环烷基;
R 11为C 1-6亚烷基或C 2-6亚烯基。
在另一优选例中,Z为N或C(-OH);
环A为取代或未取代的C 6-10芳环或取代或未取代的3-8元饱和或不饱和的含有一个或多个(例如2、3、或4个)选自N、S和O的杂环;所述取代是指被选自下组的取代基所取代:氧代(=O)、羟基、巯基;
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8分别独立选自氢、卤素、氨基(-NH 2)、羟基、羧基(-COOH)、硝基、氰基、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基、卤代C 1-6烷氧基、C 3-6环烷基、卤代C 3-6环烷基、C 1-6烷基胺基;
X和Y各自独立且不相同地选自亚甲基、氧、羰基、磺酰基、亚氨基;其中亚甲基、亚氨基独立可选地被一个或多个选自C 1-6烷基、卤代C 1-6烷基、C 1-6 烷氧基、卤代C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烯基、C 6-10芳基、C 3-10杂芳基的取代基所取代。
在另一优选例中,
Z为N、C(-OH)、C(-OR 9)或CH;
环A为
Figure PCTCN2019095648-appb-000002
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8分别独立选自:氢、卤素、氨基、羟基、羧基、硝基、氰基、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基、卤代C 1-6烷氧基、C 3-6环烷基、卤代C 3-6环烷基、C 1-6烷基胺基、R 9C(O)O-、R 9C(O)NH-、(R 9)(R 10)NC(O)S-、-OR 11OR 10
X和Y各自独立且不相同地选自:亚甲基、氧、羰基、磺酰基、亚氨基;其中亚甲基、亚氨基独立可选地被一个或多个选自卤素、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烯基、C 6-10芳基、C 3-10杂芳基的取代基所取代;
各R 9、R 10分别独立选自:C 1-6烷基、卤代C 1-6烷基、C 3-6环烷基、卤代C 3-6环烷基;
R 11为C 1-6亚烷基或C 2-6亚烯基。
在另一优选例中,Z为N、C(-OH)、C(-OR 9)或CH;
环A为
Figure PCTCN2019095648-appb-000003
R 1、R 2、R 3、R 4、R 5分别独立选自:氢、卤素、氨基、羟基、R 9C(O)O-、R 9C(O)NH-、(R 9)(R 10)NC(O)S-、-OR 11OR 10;R 9、R 10、R 11的定义如上文所述;
R 6、R 7、R 8为氢;
X和Y各自独立且不相同地选自:亚甲基、氧、亚氨基。
在另一优选例中,Z为N、C(-OH)、C(-OR 9)或CH;
环A为
Figure PCTCN2019095648-appb-000004
R 1、R 2、R 3、R 4、R 5分别独立选自:氢、卤素、氨基、羟基、R 9C(O)O-、(R 9)(R 10)NC(O)S-;R 9、R 10为甲基;
R 6、R 7、R 8为氢;
X和Y各自独立且不相同地选自:亚甲基、氧、亚氨基。
在另一优选例中,环A为
Figure PCTCN2019095648-appb-000005
R 6、R 7、R 8为氢;
X和Y各自独立且不相同地选自:亚甲基、氧、亚氨基;
当R 1为羟基时,R 2、R 4为氯,R 3、R 5为氢;
当R 1为R 9C(O)O-时,R 9为甲基,R 2、R 4为氯,R 3、R 5为氢;
当R 1为(R 9)(R 10)NC(O)S-时,R 9、R 10为甲基,R 2、R 4为氯,R 3、R 5为氢;
当R 1为氨基或氢时,R 2、R 4为氢,R 3、R 5为氯。
在另一优选例中,R 1为羟基。
在另一优选例中,R 2和R 4为氯;以及R 3和R 5为氢。
在另一优选例中,X和Y各自独立且不相同地选自亚甲基、氧、亚氨基。
在另一优选例中,环A为取代或未取代的5-6元饱和或不饱和的含有2、3、或4个选自N、S和O的杂环;所述取代是指被选自下组的取代基所取代:氧代(=O)、羟基、巯基。
在另一优选例中,所述化合物选自下组:
Figure PCTCN2019095648-appb-000006
在另一优选例中,所述化合物选自下组:
Figure PCTCN2019095648-appb-000007
本发明第二方面提供了一种药物组合物,其包含一种或多种可药用的载体或稀释剂和本发明第一方面所述的化合物或其药学上可接受的盐。
本发明第三方面提供了本发明第一方面所述的化合物或其药学上可接受的盐或第二方面所述的药物组合物的用途,用于制备预防或治疗与神经损伤相关的疾病或病症的药物。
在另一优选例中,所述疾病或病症选自下组:神经性疼痛、偏头痛、炎症痛、慢性疼痛、脑卒中、脑损伤、抑郁症、阿尔茨海默病、癫痫、情感障碍性疾病、神经退行性疾病。
本发明提供了式A所示的化合物或其药学上可接受的盐的制备方法,但不仅限于下列方法:
路线一:
Figure PCTCN2019095648-appb-000008
其中W和Q为发生反应的两个基团。其中包括但不限于羟基脱水成醚反应,醛基胺基脱水缩合反应,卤素和炔的耦联反应,羟基取代卤素的反应,胺基取代卤素的反应,羧基和胺基的脱水缩合反应,酯基和胺基的缩合反应,磺酰氯基和胺基的缩合反应等。
路线二:
Figure PCTCN2019095648-appb-000009
其中G包括但不限于COOH、CHO、CN、F、Cl、Br、I、B(OH) 2、NH 2、OMs、OTf等,通过G的官能团转换或者耦联反应构建环A;或G为
Figure PCTCN2019095648-appb-000010
所述反应用CDI等缩合剂进行缩合反应得到产物。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
具体实施方式
本发明人通过广泛而深入的研究,首次发现一类结构新颖的具有神经保护作用的化合物。本发明化合物具有良好的神经保护作用,可用于治疗疼痛、脑卒中等疾病。在此基础上完成了本发明。
术语
除特别说明之处,本文中提到的“或”具有与“和/或”相同的意义(指“或”以及“和”)。
除特别说明之处,本发明的所有化合物之中,各手性碳原子(手性中心)可以任选地为R构型或S构型,或R构型和S构型的混合物。
如本文所用,在单独或作为其他取代基一部分时,术语“烷基”指只含碳原子的直链(即,无支链)或支链饱和烃基,或直链和支链组合的基团。当烷基前具有碳原子数限定(如C 1-6烷基)时,指所述的烷基含有1-6个碳原子,例如包括甲基、乙基、丙基、异丙基、丁基、异丁基、仲丁基、叔丁基、或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“C 1-6烷氧基”指C 1-6烷基-氧-,例如包括甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、异丁氧基、仲丁氧基、叔丁氧基、或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“C 3-6环烷基”指具有3-6个碳原子的环状烷基,例如包括环丙基、环丁基、环戊基、环己基,或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“C 3-6环烯基”指具有3-6个碳原子的环状烯基,可以具有一个或两个烯基,例如包括环丁烯基、环戊烯基、环己烯基,或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“C 2-6烯基”指具有2-6个碳原子的直链或支链烯基,可以具有一个或多个烯基,例如包括乙烯基、 丙烯基、丁烯基,或类似基团。
如本文所用,在单独或作为其他取代基一部分时,术语“C 2-6炔基”指具有2-6个碳原子的支链或直链的炔基,可以具有一个或多个炔基,例如包括乙炔基、丙炔基、丁炔基,或类似基团。
如本文所用,“卤素”为氟、氯、溴或碘。
如本文所用,“卤代”是指氟代、氯代、溴代或碘代。
如本文所用,“C 6-10芳环”或“C 6-10芳基”表示具有6-10个碳原子的芳香性环,例如苯环、萘环等。
如本文所用,“3-8元饱和或不饱和的含有一个或多个(例如2、3、或4个)选自N、S和O的杂环”表示具有3-8个(优选5-6个)环原子的杂环,所述杂环含有一个或多个(例如2、3、或4个)的选自N、S和O的杂原子,例如包括
Figure PCTCN2019095648-appb-000011
如本文所用,术语“CDI”是指N,N’-羰基二咪唑。
如本文所用,术语“氨基”具有如下结构:-NH 2
如本文所用,术语“亚氨基”具有如下结构:-NH-。
如本文所用,术语“C 1-6烷基胺基”具有如下结构:-NH(C 1-6烷基)。
如本文所用,术语“C 1-6亚烷基”是指“C 1-6烷基”失去一个氢原子所形成的基团,例如包括亚甲基、亚乙基、亚丙基、亚异丙基、亚丁基等。
如本文所用,术语“C 2-6亚烯基”是指“C 2-6烯基”失去一个氢原子所形成的基团,例如包括亚乙烯基、亚丙烯基、亚异丙烯基、亚丁烯基等。
活性成分
本发明化合物是指通式A所示的化合物或其立体异构体或光学异构体,或其药学上可接受的盐。
所述的“药学上可接受的盐”指本发明化合物与药学上可接受的无机酸和有机酸所形成的盐,其中,优选的无机酸包括(但并不限于):盐酸、氢溴酸、磷酸、硝酸、硫酸、三氟乙酸(TFA);优选的有机酸包括(但并不限于):甲酸、乙酸、丙酸、丁二酸、萘二磺酸(1,5)、亚细亚酸、草酸、酒石酸、乳酸、水杨酸、苯甲酸、戊酸、二乙基乙酸、丙二酸、琥珀酸、富马酸、庚二酸、己二酸、马来酸、苹果酸、氨基磺酸、苯丙酸、葡糖酸、抗坏血酸、烟酸、异烟 酸、甲磺酸、对甲苯磺酸、柠檬酸,以及氨基酸。
所述的“立体异构体”或“光学异构体”指本发明化合物所涉及手性碳原子可以为R构型,也可以为S构型,或其组合。
专利CN104045552A报道了作为神经保护剂的化合物04006,作为nNOS与PSD-95的解耦联剂,该化合物在体外细胞活性实验中表现出良好的神经元保护作用。然而,研究过程中发现化合物04006分子上裸露的羧基导致分子极性过大造成亲脂性较差,无法通过血脑屏障(BBB),不具备继续开发成为临床治疗药物的价值。本发明通过合理的分子设计,使得化合物可以有效的通过血脑屏障(如本发明化合物04017),在相关的动物药效实验中起到了良好的神经保护作用。
Figure PCTCN2019095648-appb-000012
制备方法
下面更具体地描述本发明式A结构化合物的制备方法,但这些具体方法不对本发明构成任何限制。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便地制得,这样的组合可由本发明所属领域的技术人员容易地进行。
典型地,本发明化合物的制备工艺流程如下,其中所用原料和试剂如无特殊说明,均可通过商业途径购买。
流程一:
Figure PCTCN2019095648-appb-000013
其中,W选自下组:醛基、氨基、羟基、卤素、C 2-6炔基、卤代亚甲基、羧基、酯基、磺酰基;
Q选自下组:氨基、醛基、羟基、卤素、C 2-6炔基、卤代亚甲基、羧基、酯基、磺酰基;
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、Z、X、Y和环A的定义如上文所述。
流程二:
Figure PCTCN2019095648-appb-000014
其中,当G为
Figure PCTCN2019095648-appb-000015
时,所述反应通过缩合剂进行缩合反应得到产物;
当G选自COOH、CHO、CN、卤素、B(OH) 2、NH 2、OMs、OTf时,通过G的官能团转换或者耦联反应构建环A;
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、Z、X、Y和环A的定义如上文所述。
药物组合物和施用方法
由于本发明化合物具有优异神经保护作用,因此本发明化合物以及含有本发明化合物为主要活性成分的药物组合物可用于治疗、预防以及缓解与神经元损伤相关的疾病。根据现有技术,本发明化合物可用于治疗以下疾病(但并不限于):炎症性疼痛、神经性疼痛、偏头痛、脑卒中、神经变性疾病等。
本发明的药物组合物包含安全有效量范围内的本发明化合物及药理上可以接受的赋形剂或载体。
所述的“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有5-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
所述的“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温
Figure PCTCN2019095648-appb-000016
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、 防腐剂、无热原水等。
本发明化合物或药物组合物的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、直肠、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
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、蔗糖和阿拉伯胶;(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、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,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。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。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,例知,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,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等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,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、甲醇铝和琼脂或这些物质的混合物等。
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、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉 末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。
本发明化合物可以单独给药,或者与其他药学上可接受的化合物联合给药。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选5~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
本发明的主要优点在于:
本发明提供了一类结构新颖的具有神经保护作用的化合物,该类化合物具有更好的神经保护活性、更好的药代性质、更好的体内药效以及更优的安全性。
与类似的化合物相比,本发明的化合物可以有效通过血脑屏障(BBB)。
本发明化合物有望用于治疗和预防受神经损伤影响的疾病和病症。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数是重量百分比和重量份数。
以下实施例中所用的实验材料和试剂如无特别说明均可从市售渠道获得。
实施例1化合物04001的制备
Figure PCTCN2019095648-appb-000017
步骤一、6-(1H-四唑-5-基)吡啶-3-胺(化合物2)
在冰浴冷却下,往3-氨基-6-氰基吡啶(1.0g,8.4mmol)的DMF(15mL)溶液中分批加入叠氮化钠(4.4g,67.7mmol),反应混合物加热到140℃反应40小时。冷却到室温,过滤,滤液用饱和碳酸氢钠水溶液调节PH值到6左 右,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩得到黄色固体化合物2(0.30g,收率:22.0%)。
MS(ESI):Calcd.for C 6H 6N 6 162;Found 163[M+H] +.
步骤二、2-(((6-(1H-四唑-5-基)吡啶-3-基)氨基)甲基)-4,6-二氯苯酚(化合物04001)
往化合物2(100mg,0.62mmol)、3,5-二氯-2-羟基苯甲醛(120mg,0.63mmol)的甲醇(10mL)溶液中加入冰醋酸(4滴),混合物在氮气保护下室温搅拌2小时,然后加入氰基硼氢化钠(101mg,1.57mmol),室温搅拌一小时,浓缩除去溶剂,剩余物通过制备色谱得到白色化合物04001(27.66mg,收率:13.3%)。
MS(ESI):Calcd.for C 13H 10Cl 2N 6O 336;Found 337[M+H] +.
HNMR(400MHz,CD 3OD):δ8.12(s,1H),7.94–7.88(d,J=8.4Hz,1H),7.28-7.25(d,J=2.0Hz,1H),7.19-7.16(d,J=2.0Hz,1H),7.09-7.04(dd,J=2.0Hz,8.4Hz,1H),4.45(s,2H).
实施例2化合物04017的制备
Figure PCTCN2019095648-appb-000018
步骤一、5-((3,5-二氯-2-羟基苯甲基)氨基)氰基吡啶(化合物3)
往3-氨基-6-氰基吡啶(0.60g,5.1mmol)、3,5-二氯-2-羟基苯甲醛(1.0g,5.2mmol)的甲醇(10mL)溶液中加入冰醋酸(1mL),混合物在室温下搅拌2小时,然后加入氰基硼氢化钠(1.0g,15.7mmol),继续搅拌一小时,浓缩除去溶剂,硅胶柱分离得到白色化合物3(600mg,收率:39.5%)。
MS(ESI):Calcd.for C 13H 9Cl 2N 3O 293;Found 294[M+H] +.
步骤二、5-((3,5-二氯-2-羟基苯甲基)氨基)-N-羟基甲基吡啶脒(化合物 4)
往化合物3(0.20g,0.68mmol)的乙醇(6mL)和水(3mL)的混合溶液中加入盐酸羟胺(47mg,1.4mmol)和碳酸氢钠(114mg,1.4mmol)。混合物加热到80℃并搅拌2小时。冷却到室温,浓缩除去溶剂,加入水(10mL),室温搅拌30分钟,二氯甲烷萃取,无水硫酸钠干燥,过滤,浓缩得到白色化合物4(166mg,收率:74.9%)。
MS(ESI):Calcd.for C 13H 12Cl 2N 4O 2 326;Found 327[M+H] +.
步骤三、3-(5-((3,5-二氯-2-羟基苯甲基)氨基)吡啶-2-基)-1,2,4-噁二唑-5(4H)-酮(化合物04017)
往化合物4(0.13g,0.40mmol)的二甲基亚砜(6mL)溶液中加入CDI(129mg,0.80mmol)和DBU(122mg,0.80mmol)。混合溶液室温搅拌60分钟,加水(50mL)稀释,用1N HCl调节PH=3,乙酸乙酯萃取,无水硫酸钠干燥,过滤,制备色谱纯化得到白色化合物04017(52mg,收率:36.9%)。
MS(ESI):Calcd.for C 14H 10Cl 2N 4O 3 352;Found 353[M+H] +.
HNMR(400MHz,d 6-DMSO):δ12.70(s,1H),9.86(s,1H),8.12-8.07(d,J=2.0Hz,1H),7.73–7.66(d,J=8.8Hz,1H),7.46-7.42(d,J=2.0Hz,1H),7.24-7.16(m,2H),7.06-7.01(dd,J=2.0Hz,8.8Hz,1H),4.42-4.34(d,J=5.6Hz,2H).
实施例3化合物04159和化合物04059的制备
Figure PCTCN2019095648-appb-000019
步骤一、合成中间体3
将化合物1(5g,26.3mmol,1.0eq)溶于二氯甲烷(100mL)中,然后加入DIEA(6.79g,52.6mmol,2.0eq)和DMAP(3.21g,26.3mmol,1.0 eq)。然后化合物2(6.52g,52.6mmol,2.0eq)加入到反应液中,反应液在室温下搅拌3小时。冷却至室温后,反应液浓缩,固体用1mol/L稀盐酸洗涤2次,然后用乙酸乙酯(3x 100mL)萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤浓缩后得到化合物3(6.0g,97%)为白色固体。
步骤二、合成中间体4
将化合物3(3.0g,12.8mmol,1.0eq)溶于四氢呋喃(100mL)中,冰浴条件下分批加入硼氢化钠(1.94g,51.2mmol,4.0eq),然后慢慢升至室温。混合物在室温下搅拌2小时。反应液用乙酸乙酯(3x 100mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥,过滤浓缩后得到化合物4(3.0g,99%)为白色固体。
步骤三、合成中间体6
将化合物4(3.0g,12.7mmol,1.0eq)溶于N,N-二甲基甲酰胺(50mL)中,冰浴条件下加入氢化钠(763mg,19.1mmol,1.5eq),继续搅拌5分钟,然后化合物5(3.47g,19.1mmol,1.5eq)加入到反应液中。然后反应液升至室温,在室温下搅拌1小时。反应液用水稀释淬灭,然后乙酸乙酯(3x50mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=4/1)纯化,得到化合物6(4.2g,99%)为白色固体。
LCMS:[M+H] +=339.0.
步骤四、合成中间体7
将化合物6(1.5g,4.4mmol,1.0eq)溶于乙醇(10mL)和水(5mL)中,加入盐酸羟胺(617mg,8.8mmol,2.0eq)和碳酸氢钠(745mg,8.8mmol,2.0eq),反应液在80℃下搅拌2小时。反应液浓缩后,固体用乙酸乙酯(100mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥,过滤浓缩后得到化合物7(1.4g,86%)为白色固体。
LCMS:[M+H] +=372.0.
步骤五、合成化合物04159
将化合物7(1.4g,3.7mmol,1.0eq)溶于二甲基亚砜(10mL)中,加入CDI(1.22g,7.5mmol,2.0eq)和DBU(1.15g,7.5mmol,2.0eq),反应液在室温下搅拌1小时。反应液用1N HCl调节pH为3,然后用二氯甲烷(100mL)萃取,饱和食盐水(3x 50mL)洗涤,用无水硫酸钠干燥,过滤浓缩后得到粗产品化合物04159(1.0g,67%)为黄色固体。
LCMS:[M+H] +=398.0.
步骤六、合成化合物04059
将化合物04159(1.0g,2.5mmol,1.0eq)溶于二氯甲烷(5mL)中,加入三氟乙酸(10mL),反应液在室温下搅拌2小时。反应液浓缩后,固体用乙酸乙酯(100mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥后浓缩。残留物通过反相柱层析(0.1%甲酸/乙腈/水)纯化得到化合物04059(27.3mg,3%)为白色固体。
LCMS:[M+H] +=354.0.
1H NMR(400MHz,DMSO)δ13.03(s,1H),10.06(s,1H),8.52(d,J=2.4Hz,1H),7.97(d,J=8.8Hz,1H),7.69(dd,J=8.8,2.4Hz,1H),7.55(d,J=2.4Hz,1H),7.47(d,J=2.4Hz,1H),5.26(s,2H).
实施例4化合物04060的制备
Figure PCTCN2019095648-appb-000020
步骤一、合成中间体2
将化合物1(500mg,4.20mmol,1.0eq)溶于CCl 4(6mL)中,然后依次加入NBS(818mg,4.60mmol,1.1eq)和AIBN(68mg,0.42mmol,0.1eq)。反应液在80度下搅拌3小时。冷却至室温,浓缩后得到的残留物用硅胶柱层析(石油醚/乙酸乙酯=8/1)纯化后得到化合物2(300mg,35%)为白色固体。
LCMS:[M+H] +=197.1。
步骤二、合成中间体4
将化合物2(300mg,1.52mmol,1.0eq)溶于乙腈(5mL)中,然后依次加入化合物3(240mg,1.52mmol,1.0eq),碘化钾(24mg,0.15mmol,0.1eq)和DIEA(0.25mL)。反应液在80度下反应3小时。冷却至室温,浓缩后得到的残留物用硅胶柱层析(石油醚/乙酸乙酯=8/1)纯化后得到化合物4(120mg,26%)为白色固体。
LCMS:[M+H] +=294.0
步骤三、合成中间体5
将化合物4(90mg,0.3mmol,1.0eq)溶于乙醇/水(3mL/1.5mL)的混合溶剂中,然后依次加入盐酸羟胺(40mg,0.6mmol,2.0eq)和碳酸氢钠(50mg,0.6mmol 2.0eq)。反应液在80度下搅拌2小时。反应液浓缩后,加入5毫升水,用乙酸乙酯(3x 10mL)萃取。合并后的有机相用无水硫酸钠干燥,浓缩后得到化合物5(105mg,95%)为白色固体。
LCMS:[M+H] +=327.1.
步骤四、合成化合物04060
将化合物5(120mg,0.37mmol,1.0eq)溶于DMSO(4.0mL),然后加入CDI(178mg,1.10mmol 3.0eq)和DBU(167mg,1.10mmol,3.0eq)。反应液在50℃下搅拌3小时。冷却至室温,加入20mL水,用乙酸乙酯(3x15mL)萃取。合并后的有机相用无水硫酸钠干燥并浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=4/1)纯化后得到化合物04060(40mg,30%)为白色固体。
LCMS:[M+H] +=353.1
1H NMR(400MHz,CDCl 3)δ13.19(s,1H),8.87(d,J=1.6Hz,1H),8.20(dd,J=8.1,2.1Hz,1H),8.04(d,J=8.0Hz,1H),6.71(d,J=2.4Hz,1H),6.67(d,J=2.5Hz,1H),5.69(s,2H),5.02(s,2H).
实施例5化合物04162和化合物04062的制备
Figure PCTCN2019095648-appb-000021
步骤一、合成中间体2
将化合物1(5.0g,17.9mmol,1.0eq)溶于N,N-二甲基甲酰胺(50mL)中,然后加入氰化锌(2.3g,21.6mmol,1.2eq)和四(三苯基膦)钯(1.0g,0.9mmol,0.05eq)。反应液用氮气保护且在80℃下搅拌4小时。冷却至室温后,反应液加水稀释,然后用乙酸乙酯(3x 100mL)萃取,用饱和食盐水洗涤,用无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=4/1)纯化后得到化合物2(2.97g,94%)为白色固体。
LCMS:[M+H] +=178.0.
步骤二、合成中间体3
将化合物2(2.97g,16.8mmol,1.0eq)溶于N,N-二甲基甲酰胺(50mL)中,冰浴条件下分批加入钠氢(60%,1.0g,25.2mmol,1.5eq),继续搅拌5分钟,然后碘甲烷(3.57g,25.2mmol,1.5eq)加入到反应液中。加料完毕后反应液升至室温,混合物在室温下搅拌3小时。反应液用冰水淬灭,然后用乙酸乙酯(3x 100mL)萃取,饱和食盐水(2x 100mL)洗涤,用无水硫酸钠干燥,过滤浓缩后得到化合物3(2.66g,82%)为白色固体。
LCMS:[M+H] +=192.0.
步骤三、合成中间体4
将化合物3(2.46g,12.8mmol,1.0eq)溶于四氢呋喃(50mL)中,冰浴条件下加入硼氢化锂(0.56g,25.6mmol,2.0eq),混合物在65℃下搅拌3小时。冷却至室温后,反应液加水稀释,然后用乙酸乙酯(3x 100mL) 萃取,饱和食盐水洗涤,用无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物4(1.33g,64%)为白色固体。LCMS:[M+H] +=164.0.
步骤四、合成中间体6
将化合物4(1.33g,8.1mmol,1.0eq)溶于N,N-二甲基甲酰胺(20mL)中,冰浴条件下加入氢化钠(60%,653mg,16.3mmol,2.0eq),继续搅拌5分钟,然后化合物5(2g,16.3mmol,2.0eq)加入到反应液中。加料完毕后反应液升至室温并在室温下搅拌1小时。反应液用冰水淬灭,然后用乙酸乙酯(3x 50mL)萃取,饱和食盐水(2x 50mL)洗涤,用无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化得到化合物6(1.25g,74%)为白色固体。
LCMS:[M+H] +=208.1.
步骤五、合成中间体7
将化合物6(600mg,2.9mmol,1.0eq)溶于乙醇(5mL)和水(2.5mL)中,加入盐酸羟胺(400mg,5.8mmol,2.0eq)和碳酸氢钠(487mg,5.8mmol,2.0eq),反应液在80℃下搅拌2小时。反应液浓缩后,固体用乙酸乙酯(50mL)溶解,饱和食盐水洗涤,用无水硫酸钠干燥,过滤浓缩后得到化合物7(664mg,95%)为透明油状液体。
LCMS:[M+H] +=241.0.
步骤六、合成中间体8
将化合物7(664mg,2.7mmol,1.0eq)溶于二甲基亚砜(5mL)中,加入CDI(897mg,5.5mmol,2.0eq)和DBU(843mg,5.5mmol,2.0eq),反应液在室温下搅拌1小时。反应液用1N HCl调节pH为3,然后用乙酸乙酯(100mL)萃取,饱和食盐水(2x 50mL)洗涤,用无水硫酸钠干燥,过滤浓缩后得到化合物8(700mg,97%)为黄色固体。
LCMS:[M+H] +=267.1.
步骤七、合成中间体9
将化合物8(700mg,2.6mmol,1.0eq)溶于二氯甲烷(4mL)中,加入三氟乙酸(2mL),反应液在室温下搅拌1小时。反应液浓缩后,固体用乙酸乙酯(100mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥后浓缩。残留物通过硅胶柱层析(二氯甲烷/甲醇=20/1)纯化得到化合物9(150mg,26%)为白色固体。
LCMS:[M+H] +=223.0.
步骤八、合成中间体10
将化合物9(150mg,0.67mmol,1.0eq)溶于二氯甲烷(10mL)中,加入二氧化锰(587mg,6.7mmol,10.0eq),反应液在40℃下搅拌3小时。将二氧化锰滤去,滤液浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20/1)纯化得到化合物10(70mg,47%)为白色固体。
LCMS:[M+H] +=221.0.
步骤九、合成化合物04162
将化合物10(25mg,0.11mmol,1.0eq)溶于甲醇(3mL)中,加入化合物11(30mg,0.17mmol,1.5eq),乙酸(0.5mL)和氰基硼氢化钠(17.9mg,0.28mmol,2.5eq)。混合物在室温下搅拌2小时。反应液浓缩后,用碳酸氢钠水溶液调节pH为7~8,然后用乙酸乙酯(100mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥后浓缩。残留物通过硅胶柱层析(二氯甲烷/甲醇=20/1)纯化得到化合物04162(39mg,93%)为黄色固体。
LCMS:[M+H] +=382.0.
步骤十、合成化合物04062
将化合物04162(39mg,0.1mmol,1.0eq)溶于二氯甲烷(5mL)中,冰浴条件下缓慢滴加三溴化硼(253mg,1.0mmol,10.0eq)。混合物在室温下搅拌2小时。反应液加水稀释,然后用乙酸乙酯(100mL)萃取,饱和食盐水洗涤,用无水硫酸钠干燥后浓缩。残留物通过硅胶柱层析(二氯甲烷/甲醇=20/1)纯化得到化合物04062(5.7mg,15%)为白色固体。
LCMS:[M+H] +=368.0.
1H NMR(400MHz,MeOD)δ7.64(d,J=7.9Hz,1H),6.96(d,J=8.8 Hz,2H),6.54(d,J=2.4Hz,1H),6.25(d,J=2.4Hz,1H),4.38(s,2H).
实施例6化合物04161和化合物04061的制备
Figure PCTCN2019095648-appb-000022
步骤一、合成中间体2
将化合物1(2.0g,11.2mmol,1.0eq)溶于乙醇(60mL)中,然后加入氢氧化钠(898mg,22.4mmol,2eq)的水(20mL)溶液,升温至90℃并加入盐酸羟胺(1.56g,22.4mmol,2.0eq)。反应液在90℃下搅拌7小时。冷却至室温后将反应液浓缩,然后用乙酸乙酯(200mL)稀释,有机相用水(3x 200mL)洗涤。合并后的有机相用无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物2(1.0g,42%)为黄色固体。
LCMS:[M+H] +=212.2
步骤二、合成中间体3
将化合物2(850mg,4.0mmol,1.0eq)溶于二甲基亚砜(60mL)中,然后加入CDI(1.3g,8.0mmol,2.0eq)和DBU(1.23g,8.0mmol,2.0eq),反应液在室温下搅拌1小时。反应液用水(150mL)稀释,然后用1M的盐酸水溶液调节PH值至3。水相用乙酸乙酯(200mL)萃取,合并后的有机相用无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物3(800mg,85%)为类白色固体。
LCMS:[M+H] +=238.1.
步骤三、合成中间体4
将化合物3(750mg,3.2mmol,1.0eq)溶于乙醇(20mL)中,然后加入二氯化锡(5.7g,32mmol,10eq)。反应液在90℃下搅拌2小时。冷却至室温后,反应液先浓缩,然后用乙酸乙酯(150mL)稀释。有机相用水(3x 150mL)洗涤,无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物4(422mg,63%)为类白色固体。
LCMS:[M+H] +=208.1.
步骤四、合成化合物04161
将化合物4(432mg,1.8mmol,1.0eq)溶于甲醇/乙酸(10/1,66mL)的混合溶剂中,然后加入化合物5(343mg,1.8mmol,1.0eq)。反应液在25℃下搅拌1小时后,加入氰基硼氢化钠(339mg,5.4mmol,3.0eq)。反应液在25℃下再次搅拌1小时。反应液先浓缩,然后用乙酸乙酯(300mL)稀释。有机相用饱和食盐水洗涤,无水硫酸钠干燥后过滤浓缩。残留物通过硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物04161(565mg,83%)为粉色固体。
LCMS:[M+H] +=382.1.
步骤五、合成化合物04061
将化合物04161(100mg,0.26mmol,1.0eq)溶于二氯甲烷(5mL)中,冷却至0℃后加入三溴化硼(2g,8mmol,30eq)。反应液在室温下搅拌2小时。将反应液冷却至0℃后用饱和食盐水稀释,然后用乙酸乙酯(100mL)萃取。有机相用用饱和食盐水洗涤,无水硫酸钠干燥后过滤浓缩。残留物用二氯甲烷打浆,过滤得到的固体干燥后得到化合物04061(48.8mg,51%)。
LCMS:[M+H] +=368.1.
1H NMR(400MHz,DMSO-d 6)δ11.76(s,1H),10.08(s,1H),9.78(s,1H),7.40(d,J=2.8Hz,1H),7.29(d,J=8.8Hz,1H),7.08(d,J=2.4Hz,1H),6.20(dd,J=8.8,2.4Hz,1H),6.08(d,J=2Hz,1H),4.26(s,2H).
实施例7化合物04063的制备
Figure PCTCN2019095648-appb-000023
步骤一、合成化合物04063
将化合物04017(50mg,0.14mmol,1.0eq)溶于DCM(5mL)中,冷却到0℃后加入TEA(28mg,0.28mmol,2.0eq),然后加入乙酰氯(11mg,0.14mmol,1.0eq),反应液在0℃下搅拌2小时后,将反应液浓缩,得到的残留物用pre-HPLC(0.1%甲酸/乙腈/水)纯化后得到化合物04063(6.5mg,10.3%)为白色固体。
LCMS:[M+H] +=395.0
实施例8化合物04066的制备
Figure PCTCN2019095648-appb-000024
步骤一、合成中间体3
将化合物1(2g,10.2mmol,1.0eq)溶于乙腈(30mL)中,然后依次加入化合物2(1.8g,10.2mmol,1.0eq),碘化钾(0.17g,1.02mmol,0.1eq)和DIEA(2.6g,20.4mmol,2.0eq)。反应液在100度下反应2小时。冷却至室温,浓缩后得到的残留物用硅胶柱层析(石油醚/乙酸乙酯=5/1)纯化后得到化合物3(1.08g,44%)为黄色固体。
LCMS:[M+H] +=293.1
步骤二、合成中间体4
将化合物3(500mg,1.7mmol,1.0eq)溶于乙醇/水(15mL/7.5mL)的混合溶剂中,然后依次加入盐酸羟胺(240mg,3.4mmol,2.0eq)和碳酸氢钠(290mg,3.4mmol,2.0eq)。反应液在80度下搅拌2小时。反应液浓缩后,加入10毫升水,用乙酸乙酯(3x 10mL)萃取。合并后的有机相 用无水硫酸钠干燥,浓缩后得到化合物4(516mg,94%)为橙色固体。
LCMS:[M+H] +=326.1
步骤三、合成化合物04066
将化合物4(6.7mg,0.02mmol,1.0eq)溶于DMSO(5.0mL)混合溶剂中,然后加入CDI(10mg,0.06mmol,3.0eq)和DBU(9mg,0.06mmol,3.0eq)。反应液在50℃下搅拌3小时。冷却至室温,加入5mL水,用乙酸乙酯(3x 5mL)萃取。合并后的有机相用无水硫酸钠干燥并浓缩,然后有硅胶制备板纯化得到化合物04066(6mg,95%)为灰色固体。
LCMS:[M+H] +=352.0
1H NMR(400MHz,DMSO):δ13.00(s,1H),7.84(d,J=8.3Hz,2H),7.74(d,J=8.3Hz,2H),6.71(d,J=2.5Hz,1H),6.67(d,J=2.5Hz,1H),5.61(br,2H),4.94(s,2H).
实施例9化合物04065的制备
Figure PCTCN2019095648-appb-000025
步骤一、合成中间体2
将化合物1(3.0g,25.4mmol,1.0eq)溶于CCl 4(30mL)中,然后依次加入NBS(4.9g,27.9mmol,1.1eq)和AIBN(0.4g,2.5mmol,0.1eq)。反应液在80℃下搅拌5小时。冷却至室温,浓缩后得到的残留物用硅胶柱层析(石油醚/乙酸乙酯=8/1)纯化后得到化合物2(2.2g,44%)为白色固体。
LCMS:[M+H] +=197.1
步骤二、合成中间体4
将化合物2(500mg,2.5mmol,1.0eq)溶于乙腈(20mL)中,然后依次加入化合物3(400mg,2.5mmol,1.0eq),碘化钾(15mg,0.1mmol,0.04eq)和DIEA(0.25mL)。反应液在80℃下反应2小时。冷却至室温,浓缩后得到的残留物用硅胶柱层析(石油醚/乙酸乙酯=8/1-1/1)纯化后得到化合物4(450mg,64%)为白色固体。
LCMS:[M+H] +=279.0
步骤三、合成中间体5
将化合物4(200mg,0.7mmol,1.0eq)溶于乙醇/水(8mL/4mL)的混合溶剂中,然后依次加入盐酸羟胺(120mg,1.4mmol,2.0eq)和碳酸氢钠(100mg,1.4mmol 2.0eq)。反应液在80℃下搅拌2小时。反应液浓缩后,加入10毫升水,用乙酸乙酯(3x 15mL)萃取。合并后的有机相用无水硫酸钠干燥,浓缩后得到化合物5(200mg,91%)为白色固体。
LCMS:[M+H] +=312.0
步骤四、合成化合物04065
将化合物5(100mg,0.3mmol,1.0eq)溶于DMSO(3.0mL)中,然后加入CDI(145mg,0.9mmol 3.0eq)和DBU(136mg,0.9mmol,3.0eq)。反应液在50℃下搅拌3小时。冷却至室温,先加入20mL水,然后加入1N HCl调节pH值为3。水相用乙酸乙酯(3x 15mL)萃取。合并后的有机相用无水硫酸钠干燥并浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=10/1)纯化后得到化合物04065(10.1mg,10%)为白色固体。
LCMS:[M+H] +=338.0
1H NMR(400MHz,DMSO)δ13.17(s,1H),8.86(d,J=1.2,1H),8.12-8.10(m,1H),8.06-8.04(m,1H),7.63(s,1H),7.43-7.41(m,1H),7.32-7.30(m,1H),5.39(s,2H).
实施例10化合物04164和化合物04064的制备
Figure PCTCN2019095648-appb-000026
步骤一、合成中间体3
将化合物1(2.0g,16.2mmol,1.0eq)溶于DMF(80mL)中,然后加入DABCO(1.8g,16.2mmol,1.0eq)和化合物2(3.1g,16.2mmol,1.0eq)。反应液在室温下搅拌2小时。反应结束将反应液用乙酸乙酯(300mL)稀释,然后用饱和食盐水洗涤(3x 200mL)后将有机相用Na 2SO 4干燥并浓缩。残留物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物3(3.4g,76%)为浅棕红色固体。
LCMS:[M+H] +=278.0
步骤二、合成中间体4
将化合物3(500mg,1.8mmol,1.0eq)在200℃下搅拌12小时。反应结束冷却至室温后用用硅胶柱层析(石油醚/乙酸乙酯=10/1)纯化后得到化合物4(147mg,29%)为淡黄色固体。
LCMS:[M+H] +=278.0
步骤三、合成化合物04164
将化合物4(100mg,0.37mmol,1.0eq)溶于MeOH/AcOH=10/1(44mL)中,然后加入化合物5(77mg,0.37mmol,1.0eq)。反应液在室温下搅拌1小时。在反应液中加入NaBH 3CN(68mg,1.1mmol,3.0eq)。反应液在室温下再次搅拌1小时。反应结束将溶剂旋干。残留物用乙酸乙酯(200ml)溶解,然后用饱和食盐水洗涤(3x 200mL)后将有机相用Na 2SO 4干燥并浓缩。残留物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化后得到化合物04164(70mg,70%) 为淡黄色固体。
LCMS:[M+H] +=469.04
1H NMR(400MHz,CDCl 3)δ9.33(s,1H),7.78(s,1H),7.76(s,1H),7.51(s,1H),7.37(d,J=2.4Hz,1H),6.32(dd,J=8.8,2.0Hz,1H),6.13(d,J=2.0Hz,1H),4.50(s,2H),3.89(s,3H),3.16(s,3H),3.03(s,3H).
步骤四、合成化合物04064
将化合物04164(40mg,0.085mmol,1.0eq)溶于DCM(4mL)中,然后加入BBr 3(701mg,2.8mmol,33.0eq)。反应液在室温下搅拌1小时。反应结束在冰浴下用水将BBr3淬灭,然后用NaHCO 3水溶液将pH调至弱碱性,接着用乙酸乙酯萃取。将乙酸乙酯相旋干,残留物制备纯化(0.1%碳酸氢钠/乙腈/水)得到化合物04064(1.8mg,6%)为白色固体。
LCMS:[M+H] +=455.0
1H NMR(400MHz,DMSO)δ7.73(d,J=2.4Hz,1H),7.33(d,J=2.4Hz,1H),7.28(d,J=8.6Hz,1H),6.13-6.11(m,1H),6.03(s,1H),4.35(d,J=6.4Hz,2H),3.11(s,3H),2.94(s,3H).
OGD模型神经元保护作用测试
PC12大鼠肾上腺嗜铬细胞的基础培养基为5%(v/v)胎牛血清(Pan),10%(v/v)马血清(Gibco)和1%(v/v)青霉素-链霉素的RPMI 1640(Gibco)培养基。PC12细胞以2×10 4细胞/孔的密度接种于多聚赖氨酸(sigma)预包被的96孔板。细胞接种24小时后,加入含待测化合物的基础培养基(化合物终浓度10μM),培养1小时。对于氧糖剥夺模型,先用无糖RPMI 1640培养基洗5分钟,再用缺氧缺糖(OGD)培养基培养30分钟。OGD培养基是终浓度为10mM连二亚硫酸钠(Sigma)的无糖1640培养基,各组OGD培养基中含终浓度10μM的待测化合物。缺氧缺糖培养30分钟后,用基础培养基洗5分钟,加入含待检测化合物终浓度10μM的基础培养基继续培养。24小时后,使用Promega公司CytoTox96 Non-Radioactive试剂盒检测乳酸脱氢酶(LDH)漏出率以评估细胞损伤水平。根据试剂盒说明,测定在细胞培养基中的LDH和细胞裂解后的总LDH,细胞损伤率(Cytotoxicity%)为培养基中的LDH与总LDH的百分化 比率。测试结果见表1。
Figure PCTCN2019095648-appb-000027
化合物04017对局灶性脑缺血再灌注的保护作用研究
1.材料和方法
1.1实验动物
Sprague Dawley(SD)大鼠,雄性,SPF级,体重250~280克
1.2受试药品
化合物04017
市售依达拉奉注射液,南京先声东元制药有限公司产品,规格10mg/mL
1.3实验方法
1.3.1局灶性脑缺血再灌注模型的制备
采用颈内动脉线栓法制备大鼠局灶性脑缺血再灌模型(Middle cerebral artery occlusion,MCAO)。大鼠腹腔注射7%水合三氯乙醛(6.0mL/kg),麻醉深度以重度刺激动物有轻微反应为宜。将处于麻醉状态的大鼠用皮筋束紧四肢(后肢固定膝关节以上,前肢固定腕关节以上)及头部,动物仰卧位固定于手术台上,以动物剃毛器自头端向胸部方向剃毛,酒精消毒皮肤。颈部正中切开,钝性分离皮下组织。分离颈前三角表面的薄层筋膜,拨起低边-锁骨舌骨肌下缘,可见与此肌肉平行的纵行搏动的动脉,打开动脉壳,暴露右侧颈动脉分叉,分离右侧颈总动脉、颈外动脉、颈内动脉,轻轻剥离迷走神经,结扎并剪断颈外动脉。夹闭颈总动脉近心端,从颈外动脉的结扎线的远端作一切口,插入栓线,经过颈总动脉分叉进入颈内动脉,然后徐徐插入至有轻微阻力为止(自分叉处约20mm),阻断大脑中动脉的所有血供。以丝线在颈外动脉切口 下方稍固定栓线,松开颈总动脉近心端夹闭丝线,将浸有消毒生理盐水的纱布覆盖于创面上,将大鼠放置在保温垫上保温。右侧脑缺血2.0h后,轻轻拔出栓线,恢复血供进行再灌注,用固定栓线的丝线结扎颈外动脉,缝合皮肤,消毒。
1.3.2动物分组与给药
实验动物分为化合物04017组(5mg/kg)、阳性药依达拉奉组(6mg/kg)剂模型组共3组。制备缺血模型后,将动物机率均等单盲分配至各组。动物于再灌注后立即静脉给药1次,模型组动物给与等体积的生理盐水。缺血后24小时评价神经缺陷症状,而后处死动物,取脑,染色,拍照测定脑梗死面积。
1.3.3神经缺陷症状评分及脑梗死面积的测定
采用改良Bederson 5分制法进行神经缺陷症状评价。采用单盲法评价脑缺血后大鼠的神经缺陷症状,即由实验设计者将动物按组标记,对神经缺陷症状进行评分的试验者不知道动物的分组情况,评分结束后,评分者将各种标记的评分结果呈交设计者,由设计者揭盲,获得各实验组每只动物的评分。
Figure PCTCN2019095648-appb-000028
脑梗死程度的测定,采用TTC染色法进行脑梗死程度的测定。动物神经缺陷症状评价完毕后以CO 2处死,断头取脑,去除嗅球、小脑和低位脑干,用生理盐水冲洗大脑表面血迹,吸去表面残留水迹,于-20℃放置20min,取出后立即于视线交叉平面垂直向下作冠状切面,并向后每隔2mm切一片,将脑片置于1%TTC染液中孵育(37℃20min),正常脑组织染成深红色,缺血脑组织则呈苍白色,用生理盐水冲洗后,迅速将脑片从前向后按顺序排成一排,吸干表面残留水迹,拍照。
脑梗死面积的计算:照片用Image J软件处理,计算左脑相应面积以及右脑非梗死灶面积,求出梗死范围百分比。
2.实验结果
2.1统计分析
定量资料表示为均值±标准误,各药效指标采用GraphPad Prism(6.01)软件进行单因素方差分析(one-way ANOVA),方差检验显著后再采用Fisher’s LSD test检验组间差异。将P<0.05定义为具有显著性差异。
2.2化合物04017对大鼠脑缺血再灌急性损伤的影响
各组动物神经缺陷症状的程度及脑梗死面积情况见表2。可以看出,依达拉奉6mg/kg组(F (4,63)=3.819,P=0.0458)和化合物04017 5.0mg/kg组(F (4, 63)=3.819,P=0.0016)均能显著降低大鼠脑梗死面积,化合物04017的药效优于依达拉奉。
Figure PCTCN2019095648-appb-000029
化合物04017与化合物04006透过血脑屏障(BBB)的对比
1.材料和方法
1.1实验动物
Sprague Dawley(SD)大鼠,雄性,SPF级,体重250~280克
1.2受试化合物
化合物04017和化合物04006(参考专利CN104045552A的报道合成)
1.3实验方法
取健康雄性SD大鼠4只,2只鼠/组,分成2组,分别尾静脉注射给予化合物04017和化合物04006,剂量均为2.5mg/kg,分别于时间点5和30min采集脑组织(1只鼠/时间点),经生理盐水冲洗干净后用滤纸吸干,称重,-20℃冻存备用。(化合物配制:以5%DMAC+5%Solutol HS 15+90%Saline配制成浓度为1.0mg/mL的溶液)。
1.4实验结果
用LC-MS/MS的方法检测血浆和脑中的药物浓度,试验结果见表3.
Figure PCTCN2019095648-appb-000030
以上试验表明,化合物04017可以有效的通过血脑屏障,而化合物04006(专利CN104045552A报道的已知化合物)基本不能通过血脑屏障。
综上所述,将化合物04006的亲水性-COOH基团变换为
Figure PCTCN2019095648-appb-000031
后,在有效提高化合物的神经保护作用(对比表1中化合物04006与04017的试验结果)的同时还使得化合物可以有效的通过血脑屏障(表3)。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (10)

  1. 式A所示的化合物或其药学上可接受的盐:
    Figure PCTCN2019095648-appb-100001
    其中:
    Z为N、C(-OH)、C(-OR 9)或CH;
    环A为
    Figure PCTCN2019095648-appb-100002
    R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8分别独立选自:氢、卤素、氨基、羟基、羧基、硝基、氰基、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基、卤代C 1-6烷氧基、C 3-6环烷基、卤代C 3-6环烷基、C 1-6烷基胺基、R 9C(O)O-、R 9C(O)NH-、(R 9)(R 10)NC(O)S-、-OR 11OR 10
    X和Y各自独立且不相同地选自:亚甲基、氧、羰基、磺酰基、亚氨基;其中亚甲基、亚氨基独立可选地被一个或多个选自卤素、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烯基、C 6-10芳基、C 3-10杂芳基的取代基所取代;
    各R 9、R 10分别独立选自:C 1-6烷基、卤代C 1-6烷基、C 3-6环烷基、卤代C 3-6环烷基;
    R 11为C 1-6亚烷基或C 2-6亚烯基。
  2. 如权利要求1所述的化合物或其药学上可接受的盐,其特征在于,Z为N、C(-OH)、C(-OR 9)或CH;
    环A为
    Figure PCTCN2019095648-appb-100003
    R 1、R 2、R 3、R 4、R 5分别独立选自:氢、卤素、氨基、羟基、R 9C(O)O-、R 9C(O)NH-、(R 9)(R 10)NC(O)S-、-OR 11OR 10;R 9、R 10、R 11的定义如权利要求1中所述;
    R 6、R 7、R 8为氢;
    X和Y各自独立且不相同地选自:亚甲基、氧、亚氨基。
  3. 如权利要求1或2所述的化合物或其药学上可接受的盐,其特征在于,
    Z为N、C(-OH)、C(-OR 9)或CH;
    环A为
    Figure PCTCN2019095648-appb-100004
    R 1、R 2、R 3、R 4、R 5分别独立选自:氢、卤素、氨基、羟基、R 9C(O)O-、(R 9)(R 10)NC(O)S-;R 9、R 10为甲基;
    R 6、R 7、R 8为氢;
    X和Y各自独立且不相同地选自:亚甲基、氧、亚氨基。
  4. 如权利要求1-3任一所述的化合物或其药学上可接受的盐,其特征在于,环A为
    Figure PCTCN2019095648-appb-100005
    R 6、R 7、R 8为氢;
    X和Y各自独立且不相同地选自:亚甲基、氧、亚氨基;
    当R 1为羟基时,R 2、R 4为氯,R 3、R 5为氢;
    当R 1为R 9C(O)O-时,R 9为甲基,R 2、R 4为氯,R 3、R 5为氢;
    当R 1为(R 9)(R 10)NC(O)S-时,R 9、R 10为甲基,R 2、R 4为氯,R 3、R 5为氢;
    当R 1为氨基或氢时,R 2、R 4为氢,R 3、R 5为氯。
  5. 如权利要求1所述的化合物或其药学上可接受的盐,其特征在于,所述化合物选自下组:
    Figure PCTCN2019095648-appb-100006
  6. 一种权利要求1所述的化合物或其药学上可接受的盐的制备方法,其特征在于,通过以下反应式合成式A所述的化合物:
    Figure PCTCN2019095648-appb-100007
    其中,W选自下组:醛基、氨基、羟基、卤素、C 2-6炔基、卤代亚甲基、羧基、酯基、磺酰基;
    Q选自下组:氨基、醛基、羟基、卤素、C 2-6炔基、卤代亚甲基、羧基、酯基、磺酰基;
    R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、Z、X、Y和环A的定义如权利要求1所述;
    或通过以下反应式合成式A所述的化合物:
    Figure PCTCN2019095648-appb-100008
    其中,当G为
    Figure PCTCN2019095648-appb-100009
    时,所述反应通过缩合剂进行缩合反应得到产物;
    当G选自COOH、CHO、CN、卤素、B(OH) 2、NH 2、OMs、OTf时,通过G的官能团转换或者耦联反应构建环A;
    R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、Z、X、Y和环A的定义如权利要求1所述。
  7. 一种药物组合物,其特征在于,其包含一种或多种可药用的载体或稀释剂和权利要求1-5中任意一项所述的化合物或其药学上可接受的盐。
  8. 一种权利要求7所述的药物组合物的用途,其特征在于,用于制备用于预防或治疗受神经元损伤影响的疾病或病症的药物。
  9. 如权利要求8所述的用途,其特征在于,所述受神经元损伤影响的疾病或病症选自下组:神经性疼痛、偏头痛、炎症痛、慢性疼痛、脑卒中、脑损伤、抑郁症、阿尔茨海默病、癫痫、情感障碍性疾病、神经退行性疾病。
  10. 一种神经保护剂,其特征在于,包含一种或多种权利要求1所述的化合物或其药学上可接受的盐。
PCT/CN2019/095648 2018-07-25 2019-07-11 一类具有神经保护作用的化合物及其制备方法和用途 WO2020019994A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980048641.5A CN112469708B (zh) 2018-07-25 2019-07-11 一类具有神经保护作用的化合物及其制备方法和用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810827608 2018-07-25
CN201810827608.X 2018-07-25

Publications (1)

Publication Number Publication Date
WO2020019994A1 true WO2020019994A1 (zh) 2020-01-30

Family

ID=69180575

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/095648 WO2020019994A1 (zh) 2018-07-25 2019-07-11 一类具有神经保护作用的化合物及其制备方法和用途

Country Status (2)

Country Link
CN (1) CN112469708B (zh)
WO (1) WO2020019994A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0317165A1 (en) * 1987-11-13 1989-05-24 Riker Laboratories, Inc. DI-t-butylphenols substituted by an alkoxy or benzyloxy group or a benzylthio group
WO1994005153A1 (en) * 1992-09-09 1994-03-17 E.I. Du Pont De Nemours And Company Herbicidal benzene compounds
WO2004080377A2 (en) * 2003-03-11 2004-09-23 Neurosearch A/S Kcnq channel modulating compounds and their pharmaceutical use
WO2012025460A1 (en) * 2010-08-23 2012-03-01 Bayer Cropscience Ag Oxadiazolinone derivatives and use thereof in pest control
CN104045552A (zh) * 2013-03-13 2014-09-17 上海先声药物研究有限公司 作为神经保护剂的药用化合物

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0223147D0 (en) * 2002-10-05 2002-11-13 Man Mat Ltd Matrix assembly

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0317165A1 (en) * 1987-11-13 1989-05-24 Riker Laboratories, Inc. DI-t-butylphenols substituted by an alkoxy or benzyloxy group or a benzylthio group
WO1994005153A1 (en) * 1992-09-09 1994-03-17 E.I. Du Pont De Nemours And Company Herbicidal benzene compounds
WO2004080377A2 (en) * 2003-03-11 2004-09-23 Neurosearch A/S Kcnq channel modulating compounds and their pharmaceutical use
WO2012025460A1 (en) * 2010-08-23 2012-03-01 Bayer Cropscience Ag Oxadiazolinone derivatives and use thereof in pest control
CN104045552A (zh) * 2013-03-13 2014-09-17 上海先声药物研究有限公司 作为神经保护剂的药用化合物

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BOEZIO, A. A. 2: "4-Triazolsulfone: A Novel Isosteric Replacement of Acylsulfonamides in the Context of NaVl. 7 Inhibition", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 28, no. 11, 17 April 2018 (2018-04-17), pages 2103 - 2108, XP085402234, ISSN: 1464-3405 *
DESFORGES, G.: "An Efficient and Expeditious Synthesis of Di- and Trisubstituted Amino-phenyl and -benzyl Derivatives of Tetrazole and [1, 3, 4]Oxadiazol-2-one", JOURNAL OF COMBINATORIAL CHEMISTRY, vol. 10, no. 5, 16 August 2008 (2008-08-16), pages 671 - 680, XP055681799 *
HAFTCHENARY, S: "Potent Targeting of the STAT3 Protein in Brain Cancer Stem Cells: A Promising Route for Treating Glioblastoma", ACS MEDICINAL CHEMISTRY LETTERS, vol. 4, no. 11, 8 September 2013 (2013-09-08), pages 1102 - 1107, XP055681783, ISSN: 1948-5875 *
LEBRETON, L.: "Selective and Potent Monoamine Oxidase Type B Inhibitors: 2-Substituted 5-Aryltetrazole Derivatives", JOURNAL OF MEDICINAL CHEMISTRY, vol. 38, no. 24, 1 November 1995 (1995-11-01), pages 4786 - 4792, XP055681801, ISSN: 0022-2623 *

Also Published As

Publication number Publication date
CN112469708B (zh) 2023-08-01
CN112469708A (zh) 2021-03-09

Similar Documents

Publication Publication Date Title
DE60303381T2 (de) Neue indolylpiperidin-derivate als potente antihistaminische und antiallergische mittel
CN108699078A (zh) 噻吩并嘧啶二酮acc抑制剂的固体形式及其制备方法
WO2022007979A1 (zh) 稠合咪唑类衍生物、其制备方法及其在医药上的应用
BR122022005149B1 (pt) Compostos com atividade anticâncer, composição farmacêutica que os compreende e seus usos
TW202012412A (zh) 一種含有醯胺類衍生物的醫藥組成物及其製備方法和應用
JP2021152056A (ja) 疾患を治療するためのmct4阻害剤
WO2021121210A1 (zh) 并环类衍生物、其制备方法及其在医药上的应用
WO2022105852A1 (zh) 三嗪二酮类衍生物、其制备方法及其在医药上的应用
WO2012020725A1 (ja) Npy y5受容体拮抗作用を有するヘテロ環誘導体
WO2023227052A1 (zh) 双环衍生物parp抑制剂及其用途
WO2020019994A1 (zh) 一类具有神经保护作用的化合物及其制备方法和用途
ES2217533T3 (es) Derivados de indolomorfinano y medicamentos/preventivos para transtornos cerebrales.
EP4174068A1 (en) Novel compound, and pharmaceutical composition for preventing or treating resistant cancer, comprising same
CA3203698A1 (en) 2-pyridone derivative, and preparation method therefor and pharmaceutical application thereof
WO2022116968A1 (zh) 一种新型n-杂环bet溴结构域抑制剂、其制备方法及医药用途
CN106674245B (zh) 吡喃葡萄糖基衍生物的制备及医药上的应用
JP7450980B2 (ja) 神経保護効果を有する化合物およびその調製方法と用途
TWI831325B (zh) 作為atr抑制劑的萘啶衍生物及其製備方法
CN114149423B (zh) 四氢吡啶并嘧啶二酮类衍生物、其制备方法及其在医药上的应用
CN112300161B (zh) 一类用于治疗和/或预防乙型肝炎病毒感染的化合物及其制备方法和应用
CN103923105B (zh) 2-中氮茚甲酰胺类化合物及其制备和用途
WO2022068915A1 (zh) 6-氧代-1,6-二氢哒嗪类衍生物、其制备方法及其在医药上的应用
WO2024012565A1 (zh) 美金刚衍生物、其药物组合物及其用途
WO2024051795A1 (zh) 用作泛素-特异性蛋白酶抑制剂的取代嘌呤酮衍生物
CN114292260A (zh) 取代的哌啶类多靶点化合物及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19840899

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19840899

Country of ref document: EP

Kind code of ref document: A1