WO2014029102A1 - 噻嗪酰胺衍生物及其药物组合物和用途 - Google Patents

噻嗪酰胺衍生物及其药物组合物和用途 Download PDF

Info

Publication number
WO2014029102A1
WO2014029102A1 PCT/CN2012/080539 CN2012080539W WO2014029102A1 WO 2014029102 A1 WO2014029102 A1 WO 2014029102A1 CN 2012080539 W CN2012080539 W CN 2012080539W WO 2014029102 A1 WO2014029102 A1 WO 2014029102A1
Authority
WO
WIPO (PCT)
Prior art keywords
carbon atoms
alkyl group
linear
branched alkyl
compound
Prior art date
Application number
PCT/CN2012/080539
Other languages
English (en)
French (fr)
Inventor
李松
肖军海
韩丹
钟武
王莉莉
郑志兵
谢云德
周辛波
李行舟
王晓奎
姜丹
陈伟
刘洪英
Original Assignee
中国人民解放军军事医学科学院毒物药物研究所
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 中国人民解放军军事医学科学院毒物药物研究所 filed Critical 中国人民解放军军事医学科学院毒物药物研究所
Priority to US14/423,205 priority Critical patent/US9359314B2/en
Priority to PCT/CN2012/080539 priority patent/WO2014029102A1/zh
Priority to KR1020157007305A priority patent/KR101899777B1/ko
Priority to JP2015527749A priority patent/JP6078153B2/ja
Priority to ES12883262.3T priority patent/ES2629019T3/es
Priority to CA2882289A priority patent/CA2882289C/en
Priority to EP12883262.3A priority patent/EP2889293B1/en
Publication of WO2014029102A1 publication Critical patent/WO2014029102A1/zh
Priority to HK15106563.7A priority patent/HK1206019A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/121,4-Thiazines; Hydrogenated 1,4-thiazines not condensed with other rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to a thiazolamide derivative, a pharmaceutical composition containing the same, and a method or use thereof for preventing or treating a neurodegenerative disease.
  • Neurodegenerative diseases are a group of diseases caused by progressive pathological changes in the nervous system, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and muscle atrophy. Lateral sclerosis (ALS) ischemic or hemorrhagic stroke. Due to the complex causes of these diseases, the pathogenesis is not clear, and no effective treatment has been found.
  • ALS Lateral sclerosis
  • the FKBPs protein is named for its binding to the immunosuppressant FK506 and is an important mediator of FK506's immunosuppressive effect. Its physiological function has not yet been fully elucidated.
  • Biological evaluations include the use of in vitro tests (chicken embryo dorsal root nerve growth, PC12 cell differentiation, and oxidative damage of nerve cell lines) and various animal models (rat sciatic nerve injury model, diabetic rat peripheral neurodegenerative disease) Models, animal models of Parkinson's disease, animal models of Alzheimer's disease, etc.), some compounds based on structural design and synthesis of FKBPs have significant nerve growth and protective functions.
  • a typical representative of these compounds is GPI1485 of Guilford Pharmaceuticals Inc., which has completed phase II clinical studies based on the company's use of GPI1485 as a preventive or therapeutic agent for Parkinson's disease and stroke. Phase III clinical trials are also underway, with At the same time, a large number of highly active compounds are also emerging, making FKBPs an important target for the prevention or treatment of neurodegenerative diseases.
  • Chinese Invention Patent Application No. 01142744.2 (Replacement of six-membered nitrogen heterocyclic compounds and their use as neuromodulators) discloses a novel structure of FKBP ligands with neurotropic regeneration, of which compound 4 is the most preferred compound.
  • a novel thiazolamide derivative in a first aspect, the thiazolamide derivative of the present invention,
  • R 2 is a linear or branched alkyl group of 1 to 4 carbon atoms, or a linear or branched alkyl group of 1 to 4 carbon atoms which is substituted by a benzene ring.
  • the invention provides a thiazinamide derivative having the structure of Formula I, or a pharmaceutically acceptable salt, solvate or hydrate thereof, wherein:
  • R 2 is a linear or branched alkyl group of 1 to 4 carbon atoms, or a linear or branched alkyl group of 1 to 4 carbon atoms which is substituted by a benzene ring.
  • the present invention provides a thiazolamide derivative having the structure of Formula I, or a pharmaceutically acceptable salt, solvate or hydrate thereof, wherein: a straight chain or a branch of from 1 to 4 carbon atoms Alkenyl;
  • R 2 is a linear or branched alkyl group of 1 to 4 carbon atoms, or a linear or branched alkyl group of 1 to 4 carbon atoms which is substituted by a benzene ring.
  • the present invention provides a thiazolamide derivative having the structure of Formula I, or a pharmaceutically acceptable salt, solvate or hydrate thereof, wherein: a straight chain or a branch of from 1 to 4 carbon atoms Alkenyl;
  • R 2 is a linear or branched alkyl group of 1 to 4 carbon atoms.
  • the present invention provides a thiazolamide derivative having the structure of Formula I, or a pharmaceutically acceptable salt, solvate or hydrate thereof, wherein: a straight chain or a branch of from 1 to 4 carbon atoms Alkenyl; R 2 is a linear or branched alkyl group of 1 to 4 carbon atoms which is substituted by a benzene ring.
  • the present invention provides a thiazinamide derivative having the structure of Formula I, or a pharmaceutically acceptable salt, solvate or hydrate thereof, wherein: Ri is isobutyl;
  • R 2 is a linear or branched alkyl group of 1 to 4 carbon atoms, or 1 - substituted by a benzene ring
  • a linear or branched alkyl group of 4 carbon atoms A linear or branched alkyl group of 4 carbon atoms.
  • the present invention provides a thiazinamide derivative having the structure of Formula I, or a pharmaceutically acceptable salt, solvate or hydrate thereof, the compound being selected from the group consisting of:
  • the present invention provides a pharmaceutical composition comprising the compound of any one of the above first embodiments, and one or more pharmaceutically acceptable excipients.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising, in addition to the compound of any one of the above first aspects, further suitable pharmaceutically active compounds, and one or more pharmaceuticals Acceptable excipients.
  • the present invention provides the use of a compound according to any one of the preceding aspects in the preparation of a medicament for preventing or treating a neurodegenerative disorder caused by a physiological or physical injury or a progressive lesion. Sexual disease.
  • the neurodegenerative disease is selected from the group consisting of Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, acquired immunodeficiency Related neuropathy, multiple sclerosis of the cerebral palsy, stroke or physical stimulation associated with brain damage and various degenerative diseases affecting the central or peripheral nervous system.
  • the present invention provides the compound according to any one of the above first aspects, which is for use in the prevention or treatment of a neurodegenerative disease caused by a physiological or physical injury or a progressive lesion.
  • the neurodegenerative disease is selected from the group consisting of Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, neuropathy associated with acquired immunodeficiency, and multiple cerebral palsy Sexual sclerosis, stroke or physical stimulation associated with brain damage and various degenerative diseases affecting the central or peripheral nervous system.
  • the present invention provides a method for preventing or treating a neurodegenerative disease caused by a physiological or physical injury or a progressive lesion in a subject, comprising an effective amount of any one of the above first aspects
  • the compound, or the pharmaceutical composition of the above second aspect is administered to the subject.
  • the neurodegenerative disease is selected from the group consisting of Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, neuropathy associated with acquired immunodeficiency, and multiple cerebral palsy Sexual sclerosis, stroke or physical stimulation associated with brain damage and various degenerative diseases affecting the central or peripheral nervous system.
  • the compounds of the invention can be prepared by the methods shown in the following schemes:
  • a pharmaceutical composition of the compound using the same
  • the following terms used have the following meanings when the compound prevents or treats a method of neurodegenerative diseases caused by physiological or physical damage or progressive lesions in a subject. If the terminology used is not specifically defined herein, the term has the meaning as commonly understood by one of ordinary skill in the art.
  • linear or branched alkyl group of 1 - 6 carbon atoms means a straight or branched alkyl group having 1, 2, 3, 4, 5 or 6 carbon atoms, typically methyl, B.
  • Base n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl and hexyl.
  • linear or branched alkyl group of 1 - 4 carbon atoms means a straight or branched alkyl group having 1, 2, 3 or 4 carbon atoms, typically methyl, ethyl. , n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl.
  • salts means a salt of a compound of the invention which is pharmaceutically acceptable and which possesses the desired pharmacological activity of the parent compound.
  • Such salts include: acid addition salts with inorganic acids or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, etc.; organic acids such as acetic acid, propionic acid, Caproic acid, cyclopentanoic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methylsulfonate Acid, ethanesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, gluconic acid, glutamic acid, hydroxynaph
  • solvate means a substance which is usually formed by combining a compound of the present invention with a pharmaceutically acceptable solvent.
  • Pharmaceutically acceptable solvents include water, ethanol, acetic acid and the like.
  • Solvates include stoichiometric amounts of solvates and non-stoichiometric amounts of solvates, preferably hydrates.
  • the compound of the present invention may be crystallized or recrystallized with water or various organic solvents, in which case various solvates may be formed.
  • subject includes mammals or humans, preferably humans.
  • an effective amount means an amount of a compound which, when administered to a subject in need thereof, is sufficient to produce a prophylactic or therapeutic effect on the disease. "Effective amount” can be based on the compound, the disease and its severity, and the treatment The subject's age, weight, etc. change.
  • treatment means to ameliorate or eliminate one or more symptoms of the disease in a subject.
  • preventing means reducing the risk of a subject suffering from the disease, ie, causing at least one of the clinical symptoms of the disease not to occur in a subject who may be exposed to or susceptible to the disease, the subject being tested The person does not experience or exhibit symptoms of the disease.
  • pharmaceutically acceptable excipient means any excipient which is conventionally used in the field of pharmaceutical preparations.
  • the choice of a particular excipient will depend on the mode of administration or the type and state of the disease used to treat a particular patient.
  • it may be used as a pharmaceutically acceptable excipient including conventional diluents, carriers, fillers, binders, wetting agents, disintegrating agents, absorption enhancers, surfactants, adsorption carriers, lubricants, and the like in the pharmaceutical field.
  • a flavoring agent, a preservative, a sweetener, and the like may also be added to the pharmaceutical composition as necessary.
  • suitable pharmaceutical compositions for a particular mode of administration is well within the knowledge of those skilled in the pharmaceutical arts.
  • the pharmaceutical composition of the present invention can be administered by any means and in any form conventionally used in the art.
  • the pharmaceutical composition of the present invention may be administered in a form selected from the group consisting of oral administration, spray inhalation, rectal administration, nasal administration, vaginal administration, topical administration, parenteral administration such as subcutaneous, intravenous, and intramuscular.
  • parenteral administration such as subcutaneous, intravenous, and intramuscular.
  • Internal, intraperitoneal, intrathecal, intraventricular, intrasternal or intracranial injection preferably administered orally, intramuscularly, internally or intravenously.
  • the pharmaceutical compositions of this invention may be administered in unit dosage form.
  • the dosage form can be a liquid dosage form or a solid dosage form.
  • the liquid dosage form can be a solution, a colloid, an emulsion dosage form or a suspension dosage form.
  • the solid dosage form can be a tablet, a powder, a suppository, a granule or a capsule or the like.
  • Other dosage forms include aerosols, implants, patches or liniments.
  • the compound of the invention is administered in an amount of from about 1 to 1000 mg, preferably from 5 to 500 mg, per 24 hours, whether in human or veterinary medicine. If appropriate, the daily dose can be administered in divided doses in multiple dosage units to achieve the desired effect.
  • the compound of the present invention may be contained in the dosage unit in an amount of from 1 to 200 mg, preferably from 1 to 100 mg.
  • the specific administration dose depends on the kind and weight of the subject, the nature and severity of the disease, the type of the preparation, the mode of administration of the drug, and the administration cycle or time interval.
  • the compound of the formula I can penetrate the blood-brain barrier and is superior to the preferred compound 4 of the Chinese Patent Application No. 01142744.2, thereby having a higher bioavailability.
  • the compound of the formula I is a white crystal which has more excellent processability than the preferred compound 4 of the Chinese Patent Application No. 01142744.2, which is a stable high-melting crystalline substance which is solid and has a good flow. Sex. It is therefore suitable for industrial large-scale preparation and processing, particularly pharmaceutical processing processes that require heat or heat generation, such as grinding, heat drying, fluidized bed drying, and high temperature autoclaving.
  • the compound of the present invention is superior to the preferred compound 4 of Chinese Patent Application No. 01142744.2 in the evaluation of the efficacy of anti-mouse incomplete global cerebral ischemia.
  • the compounds of the present invention can promote nerve growth and regeneration in various neuropathy states, including neurological diseases associated with neurodegeneration, as well as by various physical injuries (such as machinery). Neuropathy caused by injury or shock) or disease (such as diabetes or autoimmune deficiency), thereby preventing or treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis or stroke .
  • Example 4 -4-p-methyl-1,4-thiazin-3-carboxylic acid 13.0 g -1,4-thiazine
  • the -3-carboxylic acid hydrochloride was suspended in 120 ml of dichloromethane, cooled to 0 ° C, and slowly added dropwise 30 ml of triethylamine. After stirring for 1 h, 120 ml of dichloromethane dissolved in 13.5 g of p-toluenesulfonyl chloride was slowly added dropwise. The solution was reacted at room temperature for 24 h, and the white precipitate was filtered.
  • Example 11 Evaluation of Neurotrophic Activity of Compounds
  • the neurotrophic activity of the compounds of the present invention can be demonstrated in a variety of in vitro biological models, such as a serum-free culture model of chicken embryo dorsal root ganglia in vitro.
  • the chicken embryo in a sterile environment, exposes its spine and bilateral ganglia under a dissecting microscope.
  • the dorsal root ganglion is removed one by one with a sharp sputum, and inoculated into a culture bottle with rat tail collagen, each bottle is inoculated 5 ⁇ 6 bottles, 2 bottles per dose.
  • Set 37 The dorsal root ganglion is removed one by one with a sharp sputum, and inoculated into a culture bottle with rat tail collagen, each bottle is inoculated 5 ⁇ 6 bottles, 2 bottles per dose.
  • Example 8 ( ⁇ ) +NGF ( 0.15 ng/mL ) 2.25
  • Example 12 In vivo pharmacodynamic evaluation of compounds in stroke
  • Kunming mice were used as experimental subjects, and two administration modes of intravenous administration and intragastric administration were used.
  • the bilateral carotid artery occlusion with low blood pressure (BCAO) was applied.
  • the -LBP model, the protective effect of the compound of the present invention on the incomplete global cerebral ischemia in mice was examined by measuring the neurological function score of the mouse and the MDA content in the brain.
  • the compound of the present invention was formulated into a solution of 30 mg/kg with 0.7% CMC-Na, and thoroughly mixed by ultrasonication to obtain a solution of 1.5 mg/ml.
  • the drug was administered by intragastric administration (i.g.) at 0.2 ml/10 g.
  • mice Twenty-eight mice that had been acclimated to the laboratory environment for one week were divided into groups according to their body weight, and i.g. were given 0.7% CMC-Na or each compound of the present invention once a day for 3 days.
  • the specific groups are as follows:
  • Cerebral ischemia model group 12, i.g 0.7% CMC-Na solution
  • Dosing group 12, respectively, i.g different compound solutions of the present invention, dosage
  • mice Twenty-eight mice that had been acclimated for one week in the laboratory environment were divided into groups according to their body weight, and i.v. were given 10% DMSO or each compound of the present invention once a day for 3 days.
  • the specific groups are as follows:
  • Muscle tension disorder torsional involuntary movement, resulting in persistent, often weird posture
  • seizures seidden loss of consciousness, falling to the ground, head tilting, limb rigidity
  • myoclonus twitching
  • 1 point excitement jump 1-2 times 1 minute jump 3-5 times 2 points jump 5 times or more 3 points stand still and wheezing (if there is partiality, then according to hemiplegia) 2 points
  • Control 4 33.15 ⁇ 2.75 0.00 ⁇ 0.00 Medium 11 40.94 ⁇ 1.754* 3.92 ⁇ 0.25**
  • Example 5 (30 mg/kg) 12 36.09 ⁇ 1.85 3.62 ⁇ 0.31
  • Example 6 (30 mg/kg) 12 30.87 ⁇ 0.95### 3.08 ⁇ 0.40
  • Example 7 (30 mg/kg) 11 34 ⁇ 90 ⁇ 1 ⁇ 65## 2.91 ⁇ 0.28
  • Example 8 (30 mg/kg) 12 33 ⁇ 27 ⁇ 1 ⁇ 91## 2 ⁇ 67 ⁇ 0 ⁇ 31#
  • MDCK-MDR1 cells are monolayers of cells that express the P-gp transporter after transfection of the MDR1 gene in MDCK (canine kidney epithelial cells). This cell is similar to the structure of the blood-brain barrier (BBB) due to its compactness of monolayer cells and high expression of drug efflux proteins, and is currently used as one of the models for evaluating BBB permeability.
  • BBB blood-brain barrier
  • the compound of the present invention must pass through the BBB because its target site is at the center, so MDCK-MDR1 cells should be used to study its membrane permeability, and the ability to penetrate BBB is initially evaluated.
  • Culture medium preparation DMEM was added with 10% FBS, 1% glutamine, 100 U mL-1 penicillin and streptomycin double solution, 1% non-essential amino acid, G418 1.2 mg 'L-l.
  • Digestion solution preparation Weigh chymotrypsin lg, EDTA 80mg, add 400mL of brick buffer, 0.22 ⁇ filter membrane sterilization, -20. C is frozen for backup.
  • the frozen MDCK-MDR1 cells were taken at 37. Quickly thaw in the C 7j bath.
  • the cells after the addition were added to DMEM medium containing 10% FBS at 37. C, 5% C02, relative humidity 90% culture in the incubator, medium change every other day.
  • the digest was mixed with 0.25 % trypsin-EDTA (0.2%), digested at 37 °C, subcultured at a certain ratio, and the cell number used for the experiment was 40 - 60 passages.
  • the cells reached 80% confluence, and after digestion, the cells were suspended in complete medium and inoculated onto Millicell plates at 1 x 106 'mL-l.
  • the culture medium was changed once every 2 days, and the liquid was changed every 1 day after 1 week. After 5 days of incubation, the resistance value reached Pingtai (> 200 ⁇ « ⁇ 2), which can be used for transport experiments.
  • the electrode When measuring the transmembrane resistance, the electrode was immersed in DMEM culture solution for 24 hours, taken out and immersed in 70% alcohol for 15 minutes, placed at room temperature and allowed to dry naturally, and then equilibrated in sterile DMEM medium for 15 min. During the experiment, the two ends of the electrode were sequentially inserted into the upper and lower cells of each well of a 24-well Millicell plate to measure the resistance value. Each hole was measured three times at any point, the resistance value was recorded, and the resistance value of the blank hole was measured, and the transmembrane resistance was calculated according to the following formula. Value (TEER).
  • Rt is the measured resistance value
  • R0 is the blank hole resistance value
  • S is the effective membrane area
  • Rho-123 (Rhodamine 123) was used as the positive control compound, and the compound was diluted to 5 mol.L1 with HBSS. The medium in each well was discarded before the experiment, using 37 V. Wash the HBSS solution twice, then incubate in a 37 °C incubator, add Rho-123 to the upper pool, add HBSS solution to the lower tank, incubate in a constant temperature shaker, and collect the lower pool at 0, 30, 90, 120 min at each time point. Solution, -20. C Store the test. The amount of Rho-123 permeation in the lower cell was measured by a fluorescence spectrophotometer. The emission wavelength was set to 430 nm, and the excitation wavelength was set to 530 nm. The Papp value of Rho-123 in this experiment is consistent with the literature reports.
  • the sample taken out is precisely added to the internal standard solution 50 ⁇ L f ethyl acetate 350 ⁇ , shake and mix, centrifuge 12000rmp, 5min, take the supernatant 300 ⁇ , dry, 50 ⁇ acetonitrile reconstituted, take ⁇ Sample determination.
  • the permeability from the basal surface to the luminal surface is added to the bottom side (BL), and the top side ( ⁇ ) is added to the blank HBSS solution. The following steps are the same as the permeability test of the cavity surface to the basal plane.
  • the apparent permeability coefficient (Papp) of the drug reflects the ability of the drug to pass through the monolayer and the rate and extent of drug absorption. It can be calculated by:
  • AQ is the amount of drug permeated during the At period
  • A is the cell surface area
  • in this model is the area of the support membrane (0.6 cm2)
  • CO is the initial concentration.
  • the units of Papp are usually expressed in centimeters per hour (cm*hl) or centimeters per second (cm*sl).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Psychology (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen- Or Sulfur-Containing Heterocyclic Ring Compounds With Rings Of Six Or More Members (AREA)

Abstract

本发明涉及噻嗪酰胺衍生物及其医药用途,具体涉及式I化合物(式中变量如说明书中所述)、其制药上可接受的盐、溶剂化物或水合物。本发明还涉及该化合物的制备方法、含有该化合物的药物组合物及其用于预防或治疗神经退行性疾病的方法或用途。

Description

噻嗪酰胺衍生物及其药物组合物和用途 技术领域
本发明涉及噻嗪酰胺衍生物、 含有该化合物的药物组合物及 其用于预防或治疗神经退行性疾病的方法或用途。 背景技术
神经退行性疾病是一类由神经系统进行性病变所引起的疾 病, 包括阿尔茨海默氏病(Alzheimer's disease) , 帕金森氏病 (Parkinson's disease), 亨廷顿氏病 (Huntington's disease)及肌姜 缩性侧索硬化症 (ALS) 缺血性或出血性脑卒中等, 由于这类疾病 的发生原因复杂, 致病机制不很明确, 目前尚没有找到有效的治 疗药物。
FKBPs 蛋白由于其与免疫抑制剂 FK506 结合而命名, 是 FK506产生免疫抑制作用的重要介质,其生理功能仍未完全阐明。
Steiner J.P.等人于 1992年发现 FKBPs ( FKBP家族)在脑和外 周的浓度远大于其在免疫组织中的浓度, 这就让人猜想 FKBPs 与神经系统可能存在某种关系。 Dawson 等人的研究结果表明 FK506能够阻断谷氛酸激活 NMDA受体引起的神经兴奋性毒性, 据推测这可能是抑制了 Calcineurin 后增加了一氧化氮合成酶 ( NOS )的磷酸化水平, 抑制了 NOS的催化活性, 避免了神经元 受到 NO的损伤。 另外研究发现与神经元的增长密切相关的蛋白 — GAP43同时也是 Calcineurin的底物, 面神经和坐骨神经损伤 的神经再生总伴有明显的 GAP43 的 mRNA 水平增加, 而同时 FKBPs的 mRNA 7j平也相应提高。 这些研究结果都表明 FKBPs 与神经生长可能存在一定的关系。最终促使人们从 FKBPs的配体 中找到了可促神经生长的有机小分子化合物,而 FKBPs也因此被 称为神经亲免素。
在这种思想的指导下, 1994年, Lyons等人研究发现, 免疫 抑制剂 FK506在体外有显著的促神经生长活性,开创了有机小分 子神经生长促进剂研究的先河。虽然 FKBPs家族配体促神经生长 和保护的机理目前尚未完全弄清楚, 但越来越多的研究表明 FKBPs参与介导了这一过程。 生物学上的评价表明, 包括使用体 外试验(鸡胚背根神经生长、 PC12 细胞分化以及神经细胞株氧 化损伤等)和多种动物模型 (大鼠外周围坐骨神经损伤模型、 糖 尿病鼠外周神经变性病模型、 帕金森氏症动物模型、 早老性痴呆 症动物模型等 ),一些基于 FKBPs结构设计和合成的化合物具有 显著的促神经生长和保护功能。 这些化合物的典型代表是 Guilford Pharmaceuticals Inc.的 GPI1485, 根据该公司将 GPI1485作为治疗帕金森氏症及脑卒中的预防或治疗药已完成了 II期临床研究, III期临床也正在进行之中, 与此同时大量的高活 性化合物也正不断涌现,使 FKBPs成为神经退行性疾病预防或治 疗药物的重要靶标。
中国发明专利申请 No. 01142744.2(取代六元氮杂环类化合物 及其作为神经调节剂的用途)披露了一类全新结构的具有促神经 再生的 FKBP配体, 其中化合物 4是最优化合物。 但研究发现其 血脑屏障通过能力较差, 且由于熔点较低, 常温下呈油状, 不适 合用于制备神经退行性疾病的预防或治疗药物。
因此, 需要发现和开发一类新的血脑屏障通过能力更强的可 用于预防或治疗神经退行性疾病的化合物。 发明内容
本发明的目的是提供新的噻嗪酰胺衍生物、 含有该化合物的 药物组合物及其用于预防或治疗神经退行性疾病的方法或用途。 第一方面, 本发 的噻嗪酰胺衍生物,
Figure imgf000005_0001
或其制药上可接受的盐、 溶剂化物或水合物, 其中:
为氢, 或 1 - 6个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基,或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
在一个实施方案中, 本发明提供具有式 I结构的噻嗪酰胺衍 生物, 或其制药上可接受的盐、 溶剂化物或水合物, 其中:
为氢, 或 1 - 4个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基,或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
在另一个实施方案中, 本发明提供具有式 I结构的噻嗪酰胺 衍生物, 或其制药上可接受的盐、 溶剂化物或水合物, 其中: 为 1 - 4个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基, 或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
在另一个实施方案中, 本发明提供具有式 I结构的噻嗪酰胺 衍生物, 或其制药上可接受的盐、 溶剂化物或水合物, 其中: 为 1 - 4个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基。
在另一个实施方案中, 本发明提供具有式 I结构的噻嗪酰胺 衍生物, 或其制药上可接受的盐、 溶剂化物或水合物, 其中: 为 1 - 4个碳原子的直链或支链烷基; 以及 R2为被苯环取代的 1 - 4个碳原子的直链或支链烷基。
在另一个实施方案中, 本发明提供具有式 I结构的噻嗪酰胺 衍生物, 或其制药上可接受的盐、 溶剂化物或水合物, 其中: Ri为异丁基; 以及
R2为 1 - 4个碳原子的直链或支链烷基,或被苯环取代的 1 -
4个碳原子的直链或支链烷基。
在再一个实施方案中, 本发明提供具有式 I结构的噻嗪酰胺 衍生物, 或其制药上可接受的盐、 溶剂化物或水合物, 该化合物 选自如下化合物:
(3R)-4-[(4-甲基苯磺酰基 )】-1,4-噻嗪 -3-羧酸 -D-亮氨酸,
(3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸乙酯, (3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸丙酯, (3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸异丙酯, 以及
(3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸苯甲酯, 或其制药上可接受的盐、 溶剂化物或水合物。
第二方面, 本发明提供一种药物组合物, 其包含上述第一方 面任意一种实施方案所述的化合物, 以及一种或多种制药上可接 受的赋形剂。
在一个实施方案中, 本发明提供一种药物组合物, 其除包含 上述第一方面任意一种实施方案所述的化合物外, 还包含其它合 适的药物活性化合物, 以及一种或多种制药上可接受的赋形剂。
第三方面, 本发明提供上述第一方面任意一种实施方案所述 的化合物在制备药物中的用途, 其中所述药物用于预防或治疗由 于生理或物理损伤或进行性病变所致的神经退行性疾病。
在一个实施方案中,所述的神经退行性疾病选自阿尔兹海默、 帕金森氏症、 享廷顿氏病、 肌萎缩性侧索硬化、 获得性免疫缺陷 相关的神经病变、 脑脊髄多发性硬化、 中风或物理刺激相关的脑 损伤和各种影响中枢或周围神经系统的退行性疾病。
第四方面, 本发明提供上述第一方面任意一种实施方案所述 的化合物, 该化合物用于预防或治疗由于生理或物理损伤或进行 性病变所致的神经退行性疾病。
在一个实施方案中,所述的神经退行性疾病选自阿尔兹海默、 帕金森氏症、 享廷顿氏病、 肌萎缩性侧索硬化、 获得性免疫缺陷 相关的神经病变、 脑脊髄多发性硬化、 中风或物理刺激相关的脑 损伤和各种影响中枢或周围神经系统的退行性疾病。
第五方面, 本发明提供一种预防或治疗受试者中由于生理或 物理损伤或进行性病变所致的神经退行性疾病的方法, 其包括将 有效量的上述第一方面任意一种实施方案所述的化合物, 或上述 第二方面所述的药物组合物给予该受试者。
在一个实施方案中,所述的神经退行性疾病选自阿尔兹海默、 帕金森氏症、 享廷顿氏病、 肌萎缩性侧索硬化、 获得性免疫缺陷 相关的神经病变、 脑脊髄多发性硬化、 中风或物理刺激相关的脑 损伤和各种影响中枢或周围神经系统的退行性疾病。
本发明的化合物可通过下面的反应路线所示的方法制备:
Figure imgf000007_0001
在描述本发明的化合物、 该化合物的药物组合物、 使用该化 合物预防或治疗受试者中由于生理或物理损伤或进行性病变所致 的神经退行性疾病的方法时, 所用的下列术语具有如下含义。 如 果所用的术语在本文中没有具体定义, 则该术语具有本领域的普 通技术人员通常所理解的含义。
术语 "1 - 6个碳原子的直链或支链烷基" 意指具有 1、 2、 3、 4、 5或 6个碳原子的直链或支链烷基, 典型地为甲基, 乙基, 正 丙基, 异丙基, 正丁基, 异丁基, 叔丁基、 戊基和己基等。
类似地, 术语 "1 - 4 个碳原子的直链或支链烷基" 意指具有 1、 2、 3或 4个碳原子的直链或支链烷基, 典型地为甲基, 乙基, 正丙基, 异丙基、 正丁基, 异丁基和叔丁基等。
术语"制药上可接受的盐 "意指在制药上可接受的并且具有母 体化合物的所需药理学活性的本发明化合物的盐。 这类盐包括: 与无机酸或与有机酸形成的酸加成的盐,所述的无机酸诸如盐酸, 氢溴酸, 硫酸, 硝酸, 磷酸等; 所述的有机酸诸如乙酸, 丙酸, 己酸, 环戊丙酸, 乙醇酸, 丙酮酸, 乳酸, 丙二酸, 琥珀酸, 苹 果酸, 马来酸, 富马酸, 酒石酸, 柠檬酸, 苯甲酸, 肉桂酸, 扁 桃酸, 甲磺酸, 乙磺酸, 苯磺酸, 萘磺酸, 樟脑磺酸, 葡庚糖酸, 葡糖酸, 谷氨酸, 羟基萘甲酸, 水杨酸, 硬脂酸, 粘康酸等; 或 在母体化合物上存在的酸性质子被金属离子, 例如碱金属离子或 碱土金属离子取代时形成的盐; 或与有机碱形成的配位化合物, 所述的有机碱诸如乙醇胺, 二乙醇胺, 三乙醇胺, N-甲基葡糖胺 等。
术语"溶剂化物 "意指通常本发明化合物与制药上可接受的溶 剂结合形成的物质。 制药上可接受的溶剂包括水, 乙醇, 乙酸等。 溶剂化物包括化学计算量的溶剂合物和非化学计算量的溶剂合 物, 优选为水合物。 本发明的化合物可以用水或各种有机溶剂结 晶或重结晶, 在这种情况下, 可能形成各种溶剂化物。 术语"受试者"包括哺乳动物或人, 优选为人。
术语"有效量,,意指在对有需要的受试者给药时足以对所述疾 病产生预防或治疗效果的化合物的用量。 "有效量 "可以根据化合 物, 疾病及其严重性和所治疗的受试者的年龄, 体重等而改变。
术语"治疗 "意指在受试者中改善或消除所述疾病的一个或多 个症状。
术语 "预防 "意指降低受试者患所述疾病的风险, 即在可能接 触或易感该病的受试者中使所述疾病的临床症状中的至少一种不 发生, 所述受试者不经历或表现出该病的症状。
术语"制药上可接受的赋形剂 "意指在药物制剂领域中常规使用 的任何赋形剂。 特定赋形剂的选择将取决于用于治疗特定患者的给 药方式或疾病类型和状态。 例如, 可以作为制药上可接受的赋形剂 包括药学领域常规的稀释剂、 载体、 填充剂、 粘合剂、 湿润剂、 崩 解剂、 吸收促进剂、 表面活性剂、 吸附载体和润滑剂等。 必要时, 还可以在药物组合物中加入香味剂、 防腐剂和甜味剂等。 用于特定 给药模式的合适药物组合物的制备方法完全在药物领域技术人员的 知识范围内。
本发明的药物组合物可以通过本领域中常用的任意方式和任 意形式给药。 例如, 本发明的药物组合物, 可以选自如下的方式 给药: 口服、 喷雾吸入、 直肠给药、 鼻腔给药、 阴道给药、 局部 给药、 非肠道给药如皮下、 静脉、 肌内、 腹膜内、 鞘内、 心室内、 胸骨内或颅内注射, 其中优选口服、 肌注、 内或静脉内注射 给药。
本发明的药物组合物可以制成单位剂量形式给药。 给药剂型 可以是液体剂型或固体剂型。 液体剂型可以是溶液类、 胶体类、 乳剂剂型或混悬剂型等。 固体剂型可以是片剂、 粉剂、 栓剂、 颗 粒剂或胶囊等。 其它剂型包括气雾剂、 埋植剂、 贴剂或擦剂等。 一般而言, 已经证明有利的是无论在人用药物还是在兽用药 物中, 本发明化合物的给药量为每 24小时约 1— 1000 mg, 优选 5— 500mg。 如果合适的话, 该日剂量可以用多个剂量单位分次给 药, 以达到所期望的效果。 本发明化合物在剂量单位中的含量可 以 l—200 mg, 优选为 1-100 mg。 但是, 具体给药的剂量取决于 治疗对象的种类和体重、 疾病的性质和严重程度、 制剂的类型和 药物的给药方式, 以及给药周期或时间间隔等因素。
根据本发明, 式 I化合物可以透过血脑屏障, 且优于中国发 明专利申请 No. 01142744.2中的优选化合物 4, 从而具有更高的 生物利用度。 进一步讲, 式 I化合物为白色晶体, 较中国发明专 利申请 No. 01142744.2中的优选化合物 4具有更优异的可加工性 能, 它们是稳定的高熔点结晶性物质, 其固体松散并具有较好的 流动性。 因此适宜于工业化大规模制备和处理, 特别是需要热或 产生热的药物加工过程, 如研磨、 加热干燥、 硫化床干燥、 及高 温高压消毒等。 更进一步说明, 本发明涉及的化合物在抗小鼠不 完全性全脑缺血药效评价中作用明显优于中国发明专利申请 No. 01142744.2中的优选化合物 4。
基于发明人所完成的实验及其结果, 可以预期本发明化合物 可以促进在各种神经病变状态下的神经生长和再生, 包括与神经 退变有关的神经疾病, 以及由各种物理损伤 (如机械损伤或冲击) 或疾病 (如糖尿病或自身免疫获得缺陷病)引起的神经病变, 从而 实现对神经退行性疾病如早老性痴呆、 帕金森氏病、 肌萎缩侧索 硬化或脑卒中的预防或治疗。 实施例
下面的实施例是用于进一步说明本发明的举例性实施方案, 不应被理解为对本发明的任何限制。 化合物熔点由 RY-1 型熔点仪测定, 温度计未经较正。 ^ NMR由 ARX-400 NMR仪测定。 质镨由 VG-ZabSpec MS 仪测 定。 除非另有说明, 所有反应用的溶剂都经标准化预处理。 实施例 1: 2 -羟乙基半胱氨酸
在 2000ml的圆底烧瓶中加入 109克 (0.9mol)L-半胱氨酸, 用 1000ml蒸馏水溶解, 冰浴冷却至 10。C, 加入 24ml 1M的 NaOH 水溶液中和至 PH~7。在 10。C下移取提前冷却的环氧乙烷 100ml。 加毕, 10。C下恒温反应 1小时, 后升至室温再反应 1.5小时。
用乙醚 400mlx4萃取以除去未反应的环氧乙烷。在低于 60。C 的条件下蒸除体系中的水层, 得黄色固体, 用水: 乙醇 =85ml: 350ml重结晶, 过滤, 用 95%的乙醇充分洗涤, 得目标化合物, 白色鳞片状固体, mp 195~196。C,约 100克,收率 67.5%。 iH-NMR (400MHz, D20) δ: 3.96131(dd, 1
3.80680-3.77293(m , 2Η), 3.1788
14.814Hz ), 3.08224 (dd , 1Η,
Figure imgf000011_0001
2.80103(t, 2Η, = 6·036Ηζ)。 实施例 2: 2 -氯乙基半胱氨酸盐酸盐 在 1000ml圆底烧瓶中加入 44克 2 -羟乙基半胱氨酸, 溶于 600ml浓盐酸中, 加热至外温 90~95。C搅拌反应 7小时。 反应完 毕, 冰箱中冷藏静置过夜, 体系中析出大量针状固体。 抽滤去除 溶剂, 所得固体自然干燥, 得灰白色固体, mpl85~186eC。 约 40 克,收率 >70%。 iH-NMR (400MHz, D20) δ: 4.30477-4.26952 (m, 1Η), 3.81913- 3.78409 (m, 2Η), 3
2=14·984Ηζ), 3.18877(dd , 1Η,
Figure imgf000011_0002
3.04410- 3.00625(m, 2Η)。 实施例 3: -1,4-噻嗪 -3-羧酸盐酸盐
取 20克 2-氯乙基半胱氨酸盐酸盐, 溶于水中, 冰浴下滴加 含 7.2克 NaHC03的水溶液, 加毕, 充分搅拌中和, 用乙酸乙酯 萃取 3次, 合并有机相, Na2S04干燥。
减压蒸除溶剂,加入 400ml无水甲醇的室温下搅拌反应 5天。 减压蒸除溶剂, 用甲醇: 乙醚重结晶, 得近白色固体约 6 克。 mpl60-161 °C!。 iH-NMR (400M Hz, DMSO-d6) δ: 10.0898(brs, 2Η), 4.4214(dd, 1Η,
Figure imgf000012_0001
8.56Hz), 3.7833(s , 3Η), 3.4986-3.4766(m, 1Η), 3.2246-3.0606(m, 3H), 2.9897-2.9593(m, 1H), 2.8763-2.8622(m, 1H); MS (¥AB)m/z: 162.0 (M-35.5), 102.0, 74.0. 实施例 4: -4-对甲^ 酰 -1,4-噻嗪 -3-羧酸 取 13.0g -1,4-噻嗪 -3-羧酸盐酸盐悬浮于 120ml 二氯甲烷 中, 冷却至 0°C, 緩慢滴入 30ml 三乙氨, 搅拌 lh后, 緩慢滴入 120ml 溶有 13.5 g对甲苯磺酰氯的二氯甲烷溶液,室温反应 24h, 过滤除去白色沉淀, 滤液依次用饱和碳酸氢钠溶液、 水洗, 无水 硫酸钠干燥, 除去干燥剂, 蒸馏出二氯甲烷得白色固体, 用乙酸 乙酯和环己烷重结晶,得白色晶体 19.4g,收率 93.5%, mp87-88°C, 比旋光度 [a】D 24 5=-78.1。 (c 2.00, CH2C12)。 ^ NMR (400MHz, CDC13) δ: 7.66938(d, 2H, =7.352Hz), 7.29941(d, 2H, =8.036Hz) , 5.06654(t, 1H, =3.436Hz) , 4.04908-3.99999(m, 1H), 3.63087(s, 3H), 3.45333-3.38197(m, 1H), 3.06102-3.02157(m, 1H), 3.00305-2.95904(m, 1H), 2.82287-2.74967(m, 1H), 2.42975(s, 3H), 2.40451-2.36514(lH 实施例 5: (3R)-4-[(4-甲基^ 酰基)】-l,4-噻嗪- 3-羧酸 -L-亮氨酸乙酯
将 4.2 克 (0.14mol ) (3 )-4-[(4-甲基苯磺酰基)】-1,4-噻嗪-3- 象酸、 3.0g (0.017mmol) L-亮氨酸乙酯盐酸酸盐、 3.2g(0.014mol) DCC 和 1.7g (0.014mol)DMAP 200ml 的 CH2C12 中, 加入 6ml(0.042mol)的 TEA, 室温反应 24小时。 过滤除去固体, 蒸去 溶剂, 以适量的乙酸乙酯溶解残留物, 过滤除去不溶物, 加入乙 酸乙酯稀适, 依次以 10% NaHCO3溶液、 饱和 NaCl溶液洗, 无 水 Na2S04干燥。 除去干燥剂, 蒸去部分乙酸乙酯, 闪色傳柱分 离(洗脱剂: CH2C12: CH3C1=1 : 1), 得油状物 4.0g。 iH-NMR (400MHz, CDC13): δ 7.77337- 7.75377(d , 2H, J=8.4HZ), 7.37582-7.264111(d, 2H, =8.4Hz), 6.79090(d, 1H, =8.644Hz), 4.79698- 4.58466 (m, 2H), 4.31444- 4.08398 (m, 3H), 3.31989- 3.11674 (m, 2H), 2.53154-2.45847 (m, 5H), 2.24620- 2.21545 (m, 1H), 1.69352-1.65150 (m , 7H) , 0.97759- 0.94891(m, 10H); MS(EI)/w/z: 443.4, 397.2, 369.2, 263.1 , 256.1 , 155.0, 139.2, 101.1. 实施例 6: (3R)-4-[(4-甲基^ 酰基)】-l,4-噻嗪- 3-羧酸 -D-亮氨酸乙酯
按照实施例 5, 由 D-亮氨酸乙酯盐酸酸盐制得 (3R)-4-[(4-甲 基苯磺酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸乙酯, 白色晶体, 收率 83.5%, mp93-95°C,比旋光度 [a】D 24 5=-35.4。。 ^ NMR (400MHz, CDC13): δ 7.77337- 7.75377(d, 2Η, J=8.4HZ), 7.37582-7.264111(d, 2H, =8.4Hz), 6.79090(d, 1H, =8.644Hz), 4.79698- 4.58466 (m, 2H), 4.31444-4.08398 (m, 3H) , 3.31989- 3.11674 (m, 2H), 2.53154-2.45847 (m, 5H), 2.24620- 2.21545 (m, 1H), 1.69352-1.65150 (m, 7H), 0.97759- 0.94891(m, 10H); MS(EI)/w/z: 443.4, 397.2, 369.2, 263.1, 256.1, 155.0, 139.2, 101.1. 实施例 7: (3R)-4-[(4-甲基^ 酰基)】-l,4-噻嗪- 3-羧酸 -D-亮氨酸丙酯
按照实施例 5, 由 D-亮氨酸丙酯盐酸酸盐制得 (3R)-4-[(4-甲 基苯磺酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸丙酯, 白色晶体, 收率 87.5%, mp96-98°C,比旋光度 [a】D 245=-38.1。 ^ NMR (400MHz, CDC13): δ 7.76337- 7.74277(d, 2Η, J=8.4HZ), 7.37782-7.35712(d, 2H, =8.4Hz), 6.76390-6.74745(d, 1H, =8.644Hz), 4.79598- 4.70066 (m, 2H), 4.28444- 4.08098 (m, 3H), 3.54489-3.53274 (t, 1H, J=2.6HZ), 3.14892 (d, 1H, =13.676), 2.56954-2.42247 (m, 5H), 2.23720- 2.19945 (m, 1H), 1.70552-1.62350 (m, 5H), 0.95459- 0.93291(m, 9H); MS(EI)/w/z: 457.3, 397.2, 369.3, 256.2, 174.0, 118.1, 101.1. 实施例 8: (3R)-4-[(4-甲基^ 酰基)】-l,4-噻嗪- 3-羧酸 -D-亮氨酸异丙酯
按照实施例 5, 由 D-亮氨酸异丙酯盐酸酸盐制得 (3R)-4-[(4- 甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸异丙酯, 白色晶体, 收 率 91.5%, mp89-91°C, 比旋光度 [a]D 245=-43.9°。 iH-NMR (400MHz, CDC13): δ 7.76237- 7.74077(d , 2H =8·208Ηζ), 7.37382-7.26511(d, 2H, =8.208Hz), 6.75090(d, 1H, =8.944Hz), 5.40112(m.lH), 4.79298- 4.25166 (m, 3H), 3.54989- 3.53674 (t, 1H, J=12.31110) , 3.15292 -3.11800(d , 1H, =13.676HZ) , 2.56054-2.46247 (m, 4H), 2.23220- 2.20345 (m, 1H), 1.62552-1.43450 (m, 4H), 1.26202-1.24745 (m, 6H), 0.94659- 0.93191(m, 6H); MS(EI)/w/z: 457.3, 397.2, 369.2, 256.2, 154.7, 101.1. 实施例 9: (3R)-4-[(4-甲基^ 酰基)】-l,4-噻嗪- 3-羧酸 -D-亮氨酸苯甲酯
按照实施例 5, 由 D-亮氨酸苯甲酯盐酸酸盐制得 (3R)-4-[(4- 甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸苯甲酯, 白色晶体, 收 率 83.5%, mp91-93°C, 比旋光度 [a]D 24 5=-33.1° . ^ NMR (400MHz, CDC13): δ 7.75237- 7.71377(d , 2H, =1·2ΗΖ), 7.36382-7.26322(m, 7H), 6.73090(m, 1H), 5.18600-5.11600(2H, m), 4.78698- 4.75466 (m, 2H), 4.24244- 4.10298 (m, 1H), 3.37689- 3.09274 (m, 2H), 2.52054-2.44547 (m, 5H), 2.06820- 2.04745 (m, 1H), 1.66552-1.25450 (m , 3H) , 0.92259- 0.90691(m , 6H); MS(EI)/w/z: 505.6, 475.1 , 457.6, 434.7, 399.0, 370.8, 336.7, 308.3, 272.4, 232.6, 148.8, 106.5. 实施例 10: (3R)-4-[(4-甲基苯磺酰基 )卜1,4- 噻嗪 -3-羧酸 -D-亮氨酸
按照实施例 5, 由 D-亮氨酸制得 (3R)-4-[(4-甲基苯磺酰 基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸, 白色晶体, 收率 77.5%, mp87-89°C,比旋光度 [a】D 24 5=-86.7。。 ^ NMR (400MHz, CDC13): 6 7.75237- 7.71377(d, 2H, =1.2HZ), 7.36382-7.26322(m, 7H), 6.73090(m, 1H), 5.18600-5.11600(2H, m), 4.78698- 4.75466 (m, 2H), 4.24244- 4.10298 (m, 1H), 3.37689- 3.09274 (m, 2H), 2.52054-2.44547 (m, 5H), 2.06820- 2.04745 (m, 1H), 1.66552-1.25450 (m, 3H), 0.92259- 0.90691(m, 6H); MS(EI)/w/z: 505.6, 475.1 , 457.6, 434.7, 399.0, 370.8, 336.7, 308.3, 272.4, 232.6, 148.8, 106.5. 实施例 11: 化合物的神经营养活性评价 本发明化合物的神经营养活性可在多种体外生物模型上体 现, 如鸡胚背根神经节体外无血清培养模型.取孵育 8 d的鸡胚, 无菌环境中, 在解剖镜下暴露其脊柱及两侧神经节, 用尖镊逐个 摘取背根神经节,接种于铺有鼠尾胶原的培养瓶中,每瓶接种 5~6 个, 每剂量 2瓶. 置 37。C, 5% C02培养箱内贴壁 l h后, 加入含 NGF (0.15ng/mL) 的无血清培养基 DMEM及本发明化合物, 对 照组只加培养基和相同剂量的 NGF. 如上培养箱中培养 48h后, 倒置相差显微镜下观察背根神经节周围突起生长情况, 依突起的 长短及疏密打分: 0: 不长突起; 1: 长稀少突起; 2: 突起较长或 较密; 3: 突起长且密.表 1所示为各实施例化合物在不同剂量下 促鸡胚背根神经节突起生长分值情况, 该分值为 5个神经节平均 分值。 表 1: 化合物促鸡胚背根活性评价结果
组别 平均分值 培养基 +NGF ( 0.15ng/mL ) (对照组 ) 0.33
实施例 5 ( lpM ) +NGF ( 0.15ng/mL ) 0.65
实施例 5 ( ΙΟΟρΜ ) +NGF ( 0.15ng/mL ) 1.55
实施例 6 ( lpM ) +NGF ( 0.15ng/mL ) 1.23
实施例 6 ( ΙΟΟρΜ ) +NGF ( 0.15ng/mL ) 1.88
实施例 7 ( lpM ) +NGF ( 0.15ng/mL ) 1.46
实施例 7 ( ΙΟΟρΜ ) +NGF ( 0.15ng/mL ) 1.95
实施例 8 ( lpM ) +NGF ( 0.15ng/mL ) 1.68
实施例 8 ( ΙΟΟρΜ ) +NGF ( 0.15ng/mL ) 2.25 实施例 12: 化合物在脑卒中的体内药效学评价
1. 实验方案
本实施例以昆明种小鼠为实验对象, 采用静脉给药及灌胃给 药两种给药方式, 应用小鼠默侧颈总动脉结扎合并低血压 ( bilateral carotid artery occlusion with low blood pressure, BCAO-LBP )模型, 通过测定小鼠神经功能学评分及脑内 MDA 含量, 考察本发明化合物预防给药对小鼠不完全性全脑缺血的保 护作用。
2. 实验方法 2.1 药物配制
2.1.1 0.7%CMC-Na的配制: 临用前一天称取 0.7g ( 700mg ) CMC-Na干粉, 加入到 100ml蒸馏水中, 边适度加热边搅拌, 待 全部溶解后, 放置过夜, 使其充分混匀, 密封装。
2.1.2 灌胃给药途径药物的配制: 本发明化合物用 0.7%CMC-Na配制成 30mg/kg, 超声使充分混匀, 得 1.5mg/ml 溶液。 按 0.2ml/10g灌胃 ( i.g )给药。
2.1.3 10%DMSO 的配制: 用微量移液枪吸取 ΙΟΟΟμί 的 DMSO分析纯, 加入 9 ml N.S. 混匀。 现用现配。
2.1.4 静脉给药途径药物的配制: 先用少许 DMSO溶解本发 明化合物, 几分钟后加入 N.S.至所需体积并充分混 ,得 lmg/ml 溶液(DMSO终浓度为 10% ) , 现用现配。 按 0.1ml/10g尾静脉 ( i.v )给药, 则小鼠给药剂量为 10mg/kg。 2.2 分组与给药
2.2.1 灌胃给药观察各本发明化合物的抗脑缺血作用
取已适应实验室环境一周的小鼠 28只,按体重均衡分组,分 别 i.g给予 0.7%CMC-Na或各本发明化合物, 1次 /d, 连续 3d。 具体组别如下:
假手术组: 4只, i.g 0.7%CMC-Na溶液
脑缺血模型组: 12只, i.g 0.7%CMC-Na溶液
给药组: 12 只, 分别 i.g 各不同本发明化合物溶液, 剂量
30mg/kg
2.2.2 静脉给药观察各本发明化合物的抗脑缺血作用
取已适应实验室环境一周的小鼠 28只,按体重均衡分组,分 别 i.v给予 10%DMSO或各本发明化合物, 1次 /d, 连续 3d。 具 体组别如下:
假手术组: 4只, i.v l0%DMSO溶液
模型组: 12只, i.v l0%DMSO溶液
给药组: 12 只, 分别 i.v各不同本发明化合物溶液, 剂量 10mg/kg
2.3 小鼠不完全性全脑缺血及大脑 MDA含量的测定
2.3.1小鼠双侧颈总动脉结扎: 末次给药 lh后, 将小鼠眼眶 放血降压(约为小鼠总血量的 30% )后, 然后将其仰背位固定于 手术板上, 颈部正中开口, 钝性剥离颈总动脉, 每侧备线 2根, 分别结扎, 当第三根线结扎完时开始计时, 然后在两根线中间将 颈总动脉剪断, 缝合切口, 假手术组只剥离颈总动脉不接扎。 手 术结束后迅速松开小鼠,观察并记录 6h小鼠的行为状态(盲法按 下表打分)和死亡时间, 小鼠死亡后迅速取脑, 去除小脑, TBA 法测其大脑全脑的 MDA含量, 6h还没死亡的小鼠处死取脑。 2.3.2 神经功能评分: 评分标准见表 2。 表 2: 神经行为评价表
( 1 )放置小鼠于地面 4分
(同时出现以下几种行为的按最严重的记录, 若小鼠不动, 可轻推其普部刺激其运动)
正常活动 0分 行走路线弯曲, 但不转 « (没有呈现追尾状) 1分 转囤, 呈现追尾状(记录旋转方向, 顺时针或逆时针) 转囤 1-2次 1分 转囤 3-5次 2分 转 B 5次以上 3分 翔滚(记录翔滚方向, 左或右) 翔滚 1-2次 1分 翔滚 3-5次 2分 翔滚 5次以上 3分 偏瘫(记录偏瘫方向, 左或右) 4分
( 2 )异常运动 8分
肌张力陣碍(扭转性的不自主动作, 造成持续的, 往往 怪异的姿势) 、 瘃痫发作(突然意识 丧失、 倒地、 头后仰、 肢体强直)、 肌阵挛(抽搐) 1分 兴奋(跳跃) 跳跃 1-2次 1分 跳跃 3-5次 2分 跳跃 5次以上 3分 静止不动兼或喘息 (若有偏瘫則按偏瘫计) 2分
4分: 手术后立即死亡(十分钟内)
( 3 )反射缺失 1分 耳廓反射(触碰小鼠的耳道时小鼠会摇头) 1分 总计: 13分 2.3.3 小鼠脑 MDA含量测定:
取小鼠大脑,称重,用 N.S制成 15%脑匀浆,取 1.2ml于 37°C 水浴 lh (每 lOmin震荡一次)后取出, 加 20%三氯乙酸 0.6ml, 混匀, 放置 lOmin , 2000r 离心 lOmin , 取上清液 1.2ml, 加 0.67%TBA0.6ml沸水浴 10min, 取出冷却, 于 532nm处测 OD 值。
3. 统计分析
实验数据以 表示,应用 SPSS13.0统计学软件,通过单 因素方差分析 Homogeneity of variance test判断方差是否齐性, 方差齐性采用 LSD检验,方差非齐性采用 Duimett's T3检验, 比 较各组间显著性差异, P<0.05有统计学意义。 结果如表 3。 表 3: 化合物在 BCAO-LBP小鼠上 MDA含量
及 经行为得^^价结果 ( ± SEM)
Figure imgf000020_0001
对照 4 33.15±2.75 0.00±0.00 介质 11 40.94±1.754* 3.92±0.25** 实施例 5 ( 30mg/kg ) 12 36.09±1.85 3.62±0.31 实施例 6 ( 30mg/kg ) 12 30.87±0.95### 3.08±0.40 实施例 7 ( 30mg/kg ) 11 34·90±1·65## 2.91±0.28 实施例 8 ( 30mg/kg ) 12 33·27±1·91## 2·67±0·31#
*ρ<0·05, 与对照组比较; **ρ<0·01, 与对照组比较; #ρ<0·05, 与介质组比较; ##ρ<0.01, 与介质组比较; ###ρ<0.001, 与介质 组比较; 用 ANOVA followed LSD on SPSS 13.0分析。 实施例 12: 化合物血脑屏障通过评价
MDCK-MDR1细胞透膜研究
1. 实验方案
MDCK-MDR1细胞是在 MDCK (犬腎上皮细胞 )中转染了 MDR1基因后, 高表达 P-gp转运体的单层细胞。 该细胞由于其 单层细胞的致密性以及高表达药物外排蛋白, 因此与血脑屏障 ( BBB )结构有相似性,目前可用作评价 BBB通透性的模型之一。 本发明化合物由于其作用靶部位在中枢, 必须透过 BBB, 因此应 用 MDCK-MDR1细胞研究其透膜性, 初步评价透 BBB的能力。
2. 实验方法
2.1 溶液配制
培养液配制: DMEM用时加入 10% FBS, 1 %谷氨酰胺, 100 U mL-1 青霉素和链霉素双抗液, 1% 非必需氛基酸, G418 1.2mg'L-l。
消化液配制: 称取姨蛋白酶 lg, EDTA 80mg, 加 400mL磚 酸盐緩冲液, 0.22μπι滤膜过滤除菌, -20。C冻存备用。
谷氛酰氛储备液配制: 谷氨酰氨 2.92g, 加 lOOmL PBS緩冲 液, 0.22μπι滤膜过滤除菌, lmL分装, -20。C冻存备用。
青链霉素储备液的配制: 青霉素 80万 U,加入 20mL生理盐 水, 链霉素 100万 U, 加入 25mL生理盐水。 将两者 1: 1混匀, 0.22μπι过滤除菌, lmL分装, -20 °C冻存备用。
HBSS液配制: NaCl 8.0g, KC1 0.4g, Na2HP04 H20 0.0475g, KH2P04 0.06g, Hapes 6g, 加入超纯水中使其溶解, 调 pH值至 7.2 - 7.4, 加水至 1L, 0.22μπι滤膜过滤除菌, -20 。C保存备用。 2.2 细胞培养
取冻存的 MDCK-MDR1细胞, 于 37。C 7j浴中快速解冻。 复 苏后的细胞加入含 10 % FBS的 DMEM培养基中,在 37。C, 5% C02, 相对湿度 90% 培养箱中培养, 隔天更换培养基。 生长 1 - 2天细胞融合后, 用 0.25 %胰蛋白酶 -EDTA ( 0.2 % )混合消化 液, 于 37 °C 条件下消化, 按一定比例传代培养, 实验用的细胞 代数为 40 - 60代。
细胞达到 80%融合, 消化后用完全培养基将细胞悬浮, 按 1x106个 'mL-l接种到 Millicell板上。 以后每 2天换培养液 1次, 1 周后每 1天换液。培育 5天, 电阻值达到坪台( > 200Ω·«η2 ), 即可以用于转运实验。
2.3 MDCK-MDR1单层细胞的质控:
2.3.1 跨膜上皮细胞电阻(TEER ) 的测量
测量跨膜电阻时先将电极浸入 DMEM 培养液中平衡 24h, 取出并浸入 70% 酒精中消毒 15min后, 室温放置并使电极自然 干燥, 再放入无菌的 DMEM培养液中平衡 15min。 实验时将电 极两端依次插入 24孔 Millicell培养板各孔的上下池中检测电阻 值, 每孔在任意点测三次, 记录电阻值, 同时测定空白孔的电阻 值, 根据以下公式计算跨膜电阻值 ( TEER ) 。
TEER= ( Rt-R0)xS
其中, Rt 为实测电阻值; R0为空白孔电阻值; S为有效膜面积。
2.3.2阳性化合物质控:
以 Rho-123(罗丹明 123)作为阳性质控化合物,将该化合物用 HBSS稀释到 5 mol.L-l,实验前先吸弃各孔中的培养基,用 37 V 的 HBSS 液洗涤两次, 然后在 37 °C 培养箱孵育, 于上池加入 Rho-123,下池中加入 HBSS液,在恒温摇床中孵育,各时间点 0, 30, 90, 120min, 收集下池的溶液, -20。C 储存备测。 用荧光分 光光度计检测下池中 Rho-123 透过的量。 其中发射波长设为 430nm, 激发波长设为 530nm。 本实验中 Rho-123的 Papp值与 文献报道相符。
2.4 药物转运实验
试验前以 37 。C HBSS液浸泡接种有细胞的 Millicell合适时 间, 轻微冲洗 Millicell, 除去细胞表面的附着物。 腔面到基底面 的通透性:在顶侧( AP )加入含药的 HBSS液 0.35 mL,底侧( BL ) 加入空白 HBSS液 1.2 mL。 置 37 。C, 以 50 r min-1振摇, 并分 别于 0、 30、 90、 120min于下层采样 50 μί, 并补充同体积空白 HBSS液。 每个浓度重复 3个孔, 取出的样品精密加入内标溶液 50μLf 乙酸乙酯 350μ , 震荡混匀, 离心 12000rmp, 5min, 取 上清 300μΙ^, 挥干, 50μ 乙腈复溶, 取 ΙΟμΙ^进样测定。 基底面 到腔面的通透性则将药物加入底侧 (BL ) , 顶侧 (ΑΡ ) 加入空 白的 HBSS液, 以下步骤同腔面到基底面的通透性试验操作。
药物的表观通透系数 (Papp ) 的大小反映了药物透过单层 细胞的能力以及药物吸收的速度、 程度。 它可由下式计算:
尸 = A
αρρ ΛΠ C。
其中, A Q为药物在 A t时间段内透过的量, A为细胞表面积, 在本模型中即为支持膜的面积(0.6 cm2 ), CO为初始浓度。 Papp 的单位常用厘米 /小时(cm*h-l )或厘米 /秒(cm*s-l )表示。 品检测
应用 LC/MS 进行检测。 每个样品浓度应用其标准曲线nM-10000nM )进行定量。
验结果
Figure imgf000024_0001
罗丹明 123 4.89
实施例 6 19.2
实施例 7 49.8
实施例 8 39.5
实施例 9 12.4
实施例 10 34.1
实施例 5 8.38

Claims

Figure imgf000025_0001
具有式 I结构的噻嗪酰胺衍生物,
或其制药上可接受的盐、 溶剂化物或水合物, 其中:
为氢, 或 1-6个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基,或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
2. 权利要求 1所述的化合物, 其中:
为氢, 或 1-4个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基,或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
3. 权利要求 1所述的化合物, 其中:
为 1-4个碳原子的直链或支链烷基; 以及
R2为 1-4个碳原子的直链或支链烷基, 或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
4. 权利要求 1所述的化合物, 其中:
为 1-4个碳原子的直链或支链烷基; 以及
R2为 1 - 4个碳原子的直链或支链烷基。
5. 权利要求 1所述的化合物, 其中:
为 1 - 4个碳原子的直链或支链烷基; 以及
R2为被苯环取代的 1 - 4个碳原子的直链或支链烷基。
6. 权利要求 1所述的化合物, 其中:
Ri为异丁基; 以及
R2为 1 - 4个碳原子的直链或支链烷基,或被苯环取代的 1 - 4个碳原子的直链或支链烷基。
7. 根据权利要求 1的化合物, 该化合物选自如下化合物: (3R)-4-[(4-甲基苯磺酰基 )】-1,4-噻嗪 -3-羧酸 -D-亮氨酸,
(3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸乙酯, (3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸丙酯, (3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸异丙酯, 以及
(3R)-4-[(4-甲基^ 酰基)】-1,4-噻嗪 -3-羧酸 -D-亮氨酸苯甲酯, 或其制药上可接受的盐、 溶剂化物或水合物。
8. 药物组合物, 其包含权利要求 1-7任一项所述的化合物, 以及一种或多种制药上可接受的赋形剂。
9. 权利要求 1-7任一项所述的化合物在制备药物中的用途, 其中所述药物用于预防或治疗由于生理或物理损伤或进行性病变 所致的神经退行性疾病。
10. 权利要求 9所述的用途,其中所述的神经退行性疾病选自 阿尔兹海默、 帕金森氏症、 享廷顿氏病、 肌萎缩性侧索硬化、 获 得性免疫缺陷相关的神经病变、 脑脊髄多发性硬化、 中风或物理 刺激相关的脑损伤和各种影响中枢或周围神经系统的退行性疾 病。
11. 权利要求 1-7任一项所述的化合物, 其用于预防或治疗由 于生理或物理损伤或进行性病变所致的神经退行性疾病。
12. —种预防或治疗受试者中由于生理或物理损伤或进行性病 变所致的神经退行性疾病的方法,其包括将有效量的权利要求 1-7 任一项所述的化合物或权利要求 8所述的药物组合物给予该受试 者。
PCT/CN2012/080539 2012-08-24 2012-08-24 噻嗪酰胺衍生物及其药物组合物和用途 WO2014029102A1 (zh)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US14/423,205 US9359314B2 (en) 2012-08-24 2012-08-24 Thiazine amide derivative and pharmaceutical composition and use thereof
PCT/CN2012/080539 WO2014029102A1 (zh) 2012-08-24 2012-08-24 噻嗪酰胺衍生物及其药物组合物和用途
KR1020157007305A KR101899777B1 (ko) 2012-08-24 2012-08-24 티아진 아미드 유도체 및 그의 약학 조성물 및 용도
JP2015527749A JP6078153B2 (ja) 2012-08-24 2012-08-24 チアンジンアミド誘導体、並びにその医薬組成物及び使用
ES12883262.3T ES2629019T3 (es) 2012-08-24 2012-08-24 Derivado de tiazinamida y composición farmacéutica y uso de los mismos
CA2882289A CA2882289C (en) 2012-08-24 2012-08-24 Thiazine amide derivative and pharmaceutical composition and use thereof
EP12883262.3A EP2889293B1 (en) 2012-08-24 2012-08-24 Thiazine amide derivative and pharmaceutical composition and use thereof
HK15106563.7A HK1206019A1 (zh) 2012-08-24 2015-07-09 噻嗪酰胺衍生物及其藥物組合物和用途

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2012/080539 WO2014029102A1 (zh) 2012-08-24 2012-08-24 噻嗪酰胺衍生物及其药物组合物和用途

Publications (1)

Publication Number Publication Date
WO2014029102A1 true WO2014029102A1 (zh) 2014-02-27

Family

ID=50149362

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/080539 WO2014029102A1 (zh) 2012-08-24 2012-08-24 噻嗪酰胺衍生物及其药物组合物和用途

Country Status (8)

Country Link
US (1) US9359314B2 (zh)
EP (1) EP2889293B1 (zh)
JP (1) JP6078153B2 (zh)
KR (1) KR101899777B1 (zh)
CA (1) CA2882289C (zh)
ES (1) ES2629019T3 (zh)
HK (1) HK1206019A1 (zh)
WO (1) WO2014029102A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017000869A1 (zh) * 2015-06-30 2017-01-05 中国人民解放军军事医学科学院毒物药物研究所 噻嗪酰胺衍生物及其用途

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1422852A (zh) * 2001-12-06 2003-06-11 中国人民解放军军事医学科学院毒物药物研究所 取代六元氮杂环类化合物及其作为神经调节剂的用途
CN102675244A (zh) * 2011-03-16 2012-09-19 中国人民解放军军事医学科学院毒物药物研究所 噻嗪酰胺衍生物及其在制备神经退行性疾病防治药物的用途

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9623000B2 (en) * 2008-07-31 2017-04-18 Dekel Pharmaceuticals Ltd Compositions and methods for treating inflammatory disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1422852A (zh) * 2001-12-06 2003-06-11 中国人民解放军军事医学科学院毒物药物研究所 取代六元氮杂环类化合物及其作为神经调节剂的用途
CN102675244A (zh) * 2011-03-16 2012-09-19 中国人民解放军军事医学科学院毒物药物研究所 噻嗪酰胺衍生物及其在制备神经退行性疾病防治药物的用途

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
E, AIHUA ET AL.: "Drug discovery based on the structure of FKBP 12: Design, synthesis and evaluation of L-1,4-thiazane-3-carboxylic acid derivatives as neuroimmunophilin ligands.", SCIENCE IN CHINA, SERIES B: CHEMISTRY, vol. 50, no. 3, June 2007 (2007-06-01), pages 405 - 417, XP055193465 *
NIE AI HUA ET AL.: "Drug Discovery Based on the Structure of FKBPs: Design, Synthesis and valuation of L-1, 4-Thiazane-3-carboxylic Acid Derivatives as Neuroimmunophilin Ligands.", CHINESE CHEMICAL LETTERS, vol. 16, no. 2, 2005, pages 163 - 166, XP008176548 *
See also references of EP2889293A4 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017000869A1 (zh) * 2015-06-30 2017-01-05 中国人民解放军军事医学科学院毒物药物研究所 噻嗪酰胺衍生物及其用途
CN106316982A (zh) * 2015-06-30 2017-01-11 中国人民解放军军事医学科学院毒物药物研究所 噻嗪酰胺衍生物及其用途
JP2018524340A (ja) * 2015-06-30 2018-08-30 インスティテュート オブ ファーマコロジー アンド トキシコロジー アカデミー オブ ミリタリー メディカル サイエンシズ ピー.エル.エー.チャイナ チアジドアミド誘導体とその利用
US10335417B2 (en) 2015-06-30 2019-07-02 Institute Of Pharmacology And Toxicology Academy Of Military Medical Sciences P.L.A. China Thiazideamide derivative and use thereof
CN106316982B (zh) * 2015-06-30 2020-08-14 中国人民解放军军事医学科学院毒物药物研究所 噻嗪酰胺衍生物及其用途

Also Published As

Publication number Publication date
HK1206019A1 (zh) 2015-12-31
ES2629019T3 (es) 2017-08-07
EP2889293A4 (en) 2015-12-30
JP2015526447A (ja) 2015-09-10
JP6078153B2 (ja) 2017-02-08
KR101899777B1 (ko) 2018-09-20
CA2882289C (en) 2019-09-17
EP2889293A1 (en) 2015-07-01
US9359314B2 (en) 2016-06-07
EP2889293B1 (en) 2017-04-26
CA2882289A1 (en) 2014-02-27
US20150203460A1 (en) 2015-07-23
KR20150046257A (ko) 2015-04-29

Similar Documents

Publication Publication Date Title
JP2017128605A (ja) 抗ウイルス化合物の固体形態
JP5781008B2 (ja) (r)−8−クロロ−1−メチル−2,3,4,5−テトラヒドロ−1h−3−ベンザゼピン塩酸塩の結晶形
JP4316794B2 (ja) イソキノリン誘導体及び医薬
CN102724880B (zh) 1-磷酸鞘氨醇受体调节剂及手性合成方法
WO2017114509A1 (zh) 醛基类化合物及其制法和用途
JP2008031064A (ja) ジアシルピペラジン誘導体
EA032526B1 (ru) Применение ингибитора кинуренин-3-монооксигеназы для лечения заболеваний и состояний, опосредованных активностью кинуренин-3-монооксигеназы
US9359328B2 (en) 2-[[[2-[(hydroxyacetyl)amino]-4-pyridinyl]methyl]thio]-N-[4-(trifluoromethoxy)phenyl]-3-pyridinecarboxamide benzenesulfonate, crystal of same, crystal polymorph thereof, and methods for production thereof
JP2010519328A (ja) H−pgdsの阻害剤としてのニコチンアミド誘導体およびプロスタグランジンd2が媒介する疾患の治療のためのそれらの使用
TW201127835A (en) Phenyloxadiazole derivatives as PGDS inhibitors
JP5096476B2 (ja) アミノピラゾール誘導体、その製造方法、およびそれを含有する虚血性疾患の予防または治療用組成物
KR20230098186A (ko) 치환된 피라졸로피리미딘의 고체 상태 형태 및 이의 용도
TWI462915B (zh) 一種組織胺h3受體拮抗劑的新穎富馬酸鹽
WO2014029102A1 (zh) 噻嗪酰胺衍生物及其药物组合物和用途
JP5888612B2 (ja) 縮合ピリジン化合物塩の結晶
KR20050085038A (ko) 항우울제 및 불안완화제로 사용가능한 수용체 5-ht 및세로토닌의 재흡수를 저해하는피리딘알킬-아미노알킬-1h-인돌 유도체
CN117202923A (zh) 用于调节fgf活性的方法和组合物
JP2003503476A (ja) 環化アミド誘導体
WO2011140682A1 (zh) (2e)-3-苯基-n-[2,2,2-三氯-1-[[(8-喹啉基氨基)硫代甲基]氨基]乙基]-2-丙烯酰胺及其医药用途
WO2017000869A1 (zh) 噻嗪酰胺衍生物及其用途
WO2023246924A1 (zh) 苯并噻唑化合物及其应用
JP2003503479A (ja) N置換化グリシン誘導体
JP2021509398A (ja) 二重作用fkbp12およびfkbp52阻害剤
JP2003503478A (ja) 神経学的疾患の治療のためのβ−アミノ酸誘導体

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: 12883262

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2882289

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015527749

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14423205

Country of ref document: US

REEP Request for entry into the european phase

Ref document number: 2012883262

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012883262

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157007305

Country of ref document: KR

Kind code of ref document: A