WO2012116586A1 - 2-氨基噻唑衍生物及制备方法和应用 - Google Patents

2-氨基噻唑衍生物及制备方法和应用 Download PDF

Info

Publication number
WO2012116586A1
WO2012116586A1 PCT/CN2012/070811 CN2012070811W WO2012116586A1 WO 2012116586 A1 WO2012116586 A1 WO 2012116586A1 CN 2012070811 W CN2012070811 W CN 2012070811W WO 2012116586 A1 WO2012116586 A1 WO 2012116586A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
substituted
aminothiazole
derivative
preparation
Prior art date
Application number
PCT/CN2012/070811
Other languages
English (en)
French (fr)
Inventor
姜凤超
周平
陈建国
王悦
曹宝帅
鄢佳
Original Assignee
华中科技大学
杭州中美华东制药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 华中科技大学, 杭州中美华东制药有限公司 filed Critical 华中科技大学
Priority to EP18020332.5A priority Critical patent/EP3424912B1/en
Priority to EP12752555.8A priority patent/EP2682390A4/en
Priority to CA2828801A priority patent/CA2828801A1/en
Priority to JP2013555731A priority patent/JP2014506897A/ja
Publication of WO2012116586A1 publication Critical patent/WO2012116586A1/zh
Priority to US14/016,126 priority patent/US9771340B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/42Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • 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
    • A61P25/28Drugs 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/44Acylated amino or imino radicals
    • C07D277/46Acylated amino or imino radicals by carboxylic acids, or sulfur or nitrogen analogues thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • C07D417/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to the preparation of a class of 2-aminothiazole compounds of the structure (I) and to the protection of premature dementia (AD) by inhibiting the rapid apoptosis of nerve cells caused by oxidative stress and the like by inhibiting acetylcholinesterase.
  • Therapeutic effect The immunosuppressive effect by inhibiting myeloid differentiation protein (MYD-88), in the treatment of anti-transplant rejection, anti-autoimmune diseases, anti-ischemic reperfusion injury, anti-chronic inflammatory response, anti-endotoxemia, etc. .
  • AD which is a neurodegenerative dysfunction disease based on primary neuronal degeneration
  • AD is not only afflicting more than 7% of the elderly over 60 years old, but also has a tendency to rejuvenate. Therefore, it is necessary to seek treatment for neurodegeneration such as AD.
  • Effective drugs for diseases have become a research hotspot in the world of medicine.
  • AD Alzheimer's disease
  • AD Alzheimer's disease
  • the commonly used methods mainly include the use of High patient cognitive ability, reducing the rapid decline of subcutaneous neuronal system function, such as the use of choline receptor agonists or acetylcholinesterase inhibitors (such as Aricept, Tacrine, donepezil, etc.) to increase acetylcholine in vivo Amounts to enhance cognitive function, using methods that reduce the amount of free radicals (such as VE and selegiline) and avoid intracellular calcium overload (such as calcium antagonist nimodipine) to reduce neuronal and synaptic loss Improve, achieve certain effects, and have a certain improvement effect on early AD.
  • free radicals such as VE and selegiline
  • calcium overload such as calcium antagonist nimodipine
  • the mechanism of action of drugs that inhibit the rapid apoptosis of nerve cells is either to remove the apoptotic signal, prevent the initiation of apoptosis, or to inhibit the initiation of apoptosis. The resulting cascade reaction.
  • PARP Poly(ADP-ribose) poly-merase
  • Inhibition of PARP can reduce organ damage caused by factors such as inflammatory response, and reduce neurotoxic excitotoxicity of NMDA and KA [Kauppinen, TM.; Swanson, RA.
  • the role of poly (ADP-ribose) polymerase - 1 in CNS disease. Neuroscience. 145, 1267-1272 (2007)] has neuroprotective effects, and has received extensive attention in the treatment of diseases such as neurodegenerative diseases, neuroinflammation, and cerebral ischemic injury.
  • AD is a multi-factorial disease, and single-target drugs are often difficult to work with.
  • multi-drugs in addition to the use of multi-drugs in therapy, the advantages of multi-target punctuation drugs have been noted [see Bolognesi ML]. , Minarini An, Tumiatti V, Melchiorre C ; Mini-Reviews in Medicinal Chemistry (2006), 6(1 1), 1269-1274.].
  • Inhibitors have been used to design inhibitors that can act on both AChe and brain MAO (such as ladostigil, rivastigmine, etc.).
  • MyD88 myeloid-differentation protein 88
  • MyD88 itself is a new target for immunosuppressive agents
  • most of the known MyD88 inhibitors are peptidomimetic compounds or small molecular compounds with complex structures, which are difficult to prepare, have high preparation costs, and are difficult to be clinically popularized [see WO 2006/067091 Al].
  • the designer of the present invention has made a series of preliminary research work on immunological inhibitors and the like in the field of computer-assisted drug design and drug synthesis, especially for the etiology and related drug design of diseases such as senile dementia, and proposed for neurodegenerative
  • 2-aminothiazole structure Considering the good stability and oxidation resistance of 2-aminothiazole structure, the basic structure of 2-aminothiazole is introduced into neuroprotective agents, acetylcholinesterase inhibitors and immunosuppressive agents to optimize pharmacological activity and optimize drug pharmacokinetics. The possibility of learning characteristics is obvious.
  • the 2-aminothiazole derivative of the formula (I) proposed by the present invention can obviously act on acetylcholinesterase and PARP-1 associated with Alzheimer's disease, and the predicted activity is more than that of the drug currently used in clinical practice, and has It can simultaneously act on the characteristics of two targets, so it can be used clinically for the treatment of Alzheimer's disease.
  • the aminothiazole derivative of the formula (I) can specifically bind to the key activation site of the MyD88 molecule, and can competitively bind to inhibit the corresponding signal transduction of MyD88, and can be clinically applied to anti-graft rejection and resistance.
  • the present invention proposes a novel class of 2-aminothiazole derivatives.
  • the present invention also provides a process for the simultaneous preparation of the above 2-aminothiazole derivatives and intermediates thereof.
  • the present invention provides a series of medical effects by using 2-aminothiazole derivatives to inhibit acetylcholinesterase and PARP-1, thereby treating various diseases associated with them (such as Alzheimer's disease (AD) and other neurodegenerative diseases). ).
  • the present invention provides an immunosuppressive effect by inhibiting myeloid differentiation protein (MYD-88) by using a 2-aminothiazole derivative, in anti-transplant rejection, anti-autoimmune disease, anti-ischemic reperfusion injury and anti-chronic inflammatory reaction , anti-endotoxemia and other aspects of the therapeutic effect;
  • MYD-88 myeloid differentiation protein
  • the structure of the aminothiazole derivative of formula (I) provided by an embodiment of the present invention is:
  • the synthesis of the 2-aminothiazole derivative of the formula (I) is carried out by first synthesizing the related intermediates (1), (2), (3), such as the hydroxyhalogenation of diethanolamine, and the resulting bis(2-dichloroethyl)
  • the amine hydrochloride and the substituted aromatic amine are cyclized under microwave or conventional conditions, and the substituted aromatic piperazine (intermediate 1) is stored; the substituted methyl aromatic ketone and the substituted thiourea are heated in the presence of iodine molecules to obtain a substitution 2 -Aminothiazole derivative (Intermediate 2), Intermediate 2 is reacted with a halogenated acid halide to give an ⁇ -haloamide derivative (Intermediate 3) of the substituted 2-aminothiazole.
  • the target compound can be obtained by an alkylation reaction or the like from the relevant intermediate.
  • the alkylation reaction of Intermediate 1 and Intermediate 3 the target product (1) containing a heterocyclic ring is obtained.
  • the intermediate 3 is first reacted with benzylamine, and the obtained product is further reacted with the corresponding halogenated hydrocarbon to obtain the objective compound (1) containing a general amino group.
  • the intermediate compound 2 and the substituted aldehyde compound are condensed under base catalysis to obtain a target compound containing a power-containing alkenyl group (D o)
  • X is a target compound of a carbonyl group
  • the present invention has been designed based on the PARP1 receptor, AchE and MYD88 receptors, and thus the compounds designed by the present invention are also useful for diseases caused by related targets including the PARP1 receptor, AchE and MYD88 receptors.
  • the 2-aminothiazole derivative of the formula (I) mentioned in the present invention has a suitable molecular weight, a stable structure, and can penetrate a cell membrane to predict a less toxic side effect.
  • the 2-aminothiazole derivative of the formula (I) mentioned in the present invention is an acetylcholinesterase inhibitor for the treatment of Alzheimer's disease (AD) and other neurodegenerative diseases;
  • the 2-aminothiazole derivative of the formula (I) mentioned in the present invention is used as a neuroprotective agent for the treatment of diseases caused by rapid apoptosis of nerve cells due to oxidative stress and the like.
  • the 2-aminothiazole derivative of the formula (I) mentioned in the present invention is used as a MyD88 inhibitor for reducing the post-transplant rejection reaction and the application and maintenance of transplantation tolerance.
  • the 2-aminothiazole derivative of the formula (I) mentioned in the present invention is used as an immunomodulator to treat various autoimmune diseases, chronic inflammatory diseases and the like.
  • the 2-aminothiazole derivative of the formula (I) mentioned in the present invention is used as an immunomodulator to treat ischemia-reperfusion injury The application of diseases.
  • the invention has the advantage that a 2-aminothiazole derivative of the formula (I) is designed and obtained, and its inhibition on acetylcholinesterase and PARP-1 and the oxidation reaction are fully confirmed in related experiments.
  • the protective effect of rapid apoptosis of nerve cells caused by acute and hypoxic injury may be an effective drug for treating or preventing neurodegenerative diseases such as senile dementia.
  • MyD88 it has been confirmed that it has obvious effects in anti-rejection and induction of immune tolerance after transplantation, treatment of various inflammatory reactions, and prevention of ischemia-reperfusion injury.
  • the 2-aminothiazole structure has good stability and oxidation resistance, and is very beneficial to the pharmacological activity of the derivative and the improvement of the pharmacokinetic properties.
  • the present invention is also advantageous in the synthesis of a class of 2-aminothiazole derivatives of formula (I), such as diethanolamine, iodine molecule Aromatic ketones, thioureas, etc. are inexpensive and readily available, with few steps, and the products are easy to separate and purify.
  • Fig. 3 shows an aminothiazole derivative against the hydrogen peroxide cell protection activity (cell survival improvement rate) of the present invention.
  • Fig. 4 The cytoprotective activity (cell survival rate) of the aminothiazole derivative against the hypoxia injury of cobalt chloride proposed by the present invention
  • Fig. 6 SH-SY5Y cells protected by the aminothiazole derivative proposed by the present invention after the damage of cobalt chloride.
  • Figure 7-10 The up-regulation of the co-stimulatory molecule CD80 induced by LPS and CpG by the aminothiazole derivative proposed by the present invention
  • Figure 11 is a graph showing the reduction of diabetes incidence rate of the aminothiazole derivatives proposed by the present invention
  • Fig. 1 is a bar graph depicting the results of an inhibitory activity test of an aminothiazole derivative (AchE) according to the present invention. Acetylcholinesterase was obtained from SD rats.
  • Figure 2 is a bar graph depicting the results of an inhibitory activity test of an aminothiazole derivative (concentration ⁇ ) for poly ADP-ribose polymerase-1 (PARP-1) according to the present invention.
  • Figure 3 is a graph showing the induction of an aminothiazole derivative according to the present invention for hydrogen peroxide (0 2 ) A histogram of the protective effect of neuronal apoptosis on stress), the cells are human dopaminergic neuroblastoma cells, SH-SY5Y cells.
  • Figure 4 is a bar graph depicting the results of a test for the protective effect of an aminothiazole derivative on the apoptosis of nerve cells induced by cobalt chloride (CoCl 2 ) (human hypoxia injury) according to the present invention, the cells being human dopaminergic neuroblastoma Cells - SH-SY5Y cells.
  • CoCl 2 cobalt chloride
  • Figure 5 is a human dopaminergic neuroblastoma cell induced by cobalt chloride (CoCl 2 ) (hypoxia injury)
  • Fig. 6 is a photograph of human dopaminergic neuroblastoma-SH-SY5Y cells treated with cobalt chloride (CoCl 2 )-induced (anoxic damage) treatment with the aminothiazole derivative proposed by the present invention.
  • LPS, CPG, myocardial tissue homogenate solvent stimulated DC, and the aminothiazole derivative proposed by the present invention down-regulates the expression of CD80.
  • Figure 9 is a dose-dependent down-regulation of CD surface expression of the aminothiazole derivative of the present invention.
  • Figure 10 is a dose-dependent down-regulation of CD80 expression on the surface of macrophages of the aminothiazole derivative proposed in the present invention.
  • the experimental group in Fig. 11 MyD88KO NOD mouse, MyD88KO/+NOD mouse, NOD mouse
  • the aminothiazole derivative drug group proposed by the present invention is a dose-dependent down-regulation of CD surface expression of the aminothiazole derivative of the present invention.
  • Figure 10 is a dose-dependent down-regulation of CD80 expression on the surface of macrophages of the aminothiazole derivative proposed in the present invention.
  • the experimental group in Fig. 11 MyD88KO NOD mouse, MyD88KO/+NOD mouse, NOD mouse
  • the aminothiazole derivative drug group proposed by the present invention The aminothiazole derivative drug group proposed by the present invention.
  • R 2 is a cycloalkyl group; or a substituted aromatic group, a hydrocarbon group of 11, , -CH 2 Ph (benzyl
  • n 0 to 5.
  • R 2 is a cyclic hydrocarbon group comprising a 3 to 7 membered ring; or when R 2 is a hydrocarbon group (including When -CH 3 , -C 2 H 5 , -CH 2 Ph (benzyl), -CH 2 OR, etc.), the substituted aromatic group is a substituted benzene ring (a), benzyl
  • R 4 is a hydrocarbon alkyl group, a cycloalkyl group having a 3 to 7-membered ring, an alkoxy group having a ⁇ hydrocarbyl group, and a ⁇ . a carboxylate group or a halogen group (-Cl, -Br, -CN);
  • substitution position of R 4 may be in the para, ortho or meta position.
  • the amino group-containing group (R 3 ) is piperazine derivative (f), piperidine derivative (g), tetrahydropyrrole derivative (h), pyridine (i) Or an amino derivative (j).
  • R 5 is a substituted benzene ring (a), a substituted pyridine (b) or a substituted pyrrole (c);
  • Ar is -benzyl or 3-pyridylmethyl.
  • the process for the preparation of the 2-aminothiazole derivative of the formula (I) provided by the embodiment of the present invention includes the preparation of an intermediate and a target compound.
  • the powdered thiourea and iodine are mixed uniformly according to a molar ratio of 2:1.
  • the substituted ketone derivative is added and stirred uniformly, and reacted under microwave conditions of suitable power for several minutes. After the reaction is completed, a small amount of diethyl ether is added and filtered. The precipitate was washed with diethyl ether to give the hydroiodide of 2-aminothiazole.
  • the hydroiodide was dissolved in hot water, and a calculated amount of sodium hydrogencarbonate solid was added thereto with stirring to adjust the pH of the solution to neutrality. The resulting precipitate was filtered, washed with water and dried to give a crude 2-aminothiazole.
  • 4 5 -Disubstituted-2-aminothiazole was used in anhydrous THF, and a halogenated acid halide (such as chloroacetyl chloride) was added dropwise in an ice bath, and the molar ratio to 4 5 -disubstituted-2-aminothiazole was 1 :1.2), after the completion of the dropwise addition, the reaction was stirred at room temperature, and the reaction was monitored by TLC until the reaction was completed. The sample was separated by chromatography.
  • a halogenated acid halide such as chloroacetyl chloride
  • Chloroacetyl chloride can also be replaced by acryloyl chloride to form the corresponding 4 5-disubstituted-2-aminothiazole-N-chloropropionamide.
  • Target compound preparation can also be replaced by acryloyl chloride to form the corresponding 4 5-disubstituted-2-aminothiazole-N-chloropropionamide.
  • the aminothiazole derivative (I) for use in therapy can be administered to an individual by any suitable method, either directly (local administration, such as injection, transplantation or topical application to a target tissue site, etc.) and systemic administration (via Injection route and oral administration, etc.).
  • the compound is administered to certain specific sites by parenteral administration, so that A portion of the aqueous or physiologically compatible fluid suspension and aqueous solution are preferably combined.
  • the 2-aminothiazole derivative (I) proposed by the present invention can be used for treating diseases caused by rapid apoptosis of nerve cells caused by certain neurodegenerative diseases such as Alzheimer's disease, oxidative stress, etc. Immune diseases and chronic inflammatory diseases, diseases of ischemia-reperfusion injury, and diseases for reducing post-transplant rejection and induction and maintenance of transplantation immune tolerance.
  • the drug can be provided in one dose or continuously.
  • the new compound can be administered alone or in combination with other agents, for example: compounds for combination therapy.
  • the new drug contains a certain amount of new compounds with therapeutic effects. The amount of the new compound will depend on the patient's condition. The patient's weight, the severity of the condition, the method of administration, and the pharmacist prescribing the prescription should be included in the range of consideration for the appropriate dose.
  • the 2-aminothiazole derivative (I) proposed by the present invention can be administered continuously or intermittently by any route suitable for a drug molecule. Both oral administration and intraperitoneal administration can be used as a suitable route for administration.
  • Example 1 Preparation of 4-phenyl-2-aminothiazole 7.6 g (0.1 mol) of thiourea and 12.7 g (0.05 mol) of iodine were respectively ground and mixed with a mortar, and quickly added 6.0 g (0.05 mol) of acetophenone, stirred and mixed, and then controlled in a microwave reactor. 150W-200W reaction for 2-3min. After completion of the reaction, about 40 ml to 50 ml of diethyl ether was added, and the mixture was stirred rapidly, filtered, and the filter cake was washed with a small amount of diethyl ether to give a pale yellow solid.
  • Example 3 Preparation of 4-phenyl-N-(4-phenyltWazol-2-yl)acetamide 3.5 g (0.02 mol) of 4-phenyl-2-aminothiazole was dissolved in as little tetrahydrofuran as possible, and 2.5 g (0.024 mol) of chloroacetyl chloride was slowly added dropwise under ice bath. The solution was slightly yellow, after the addition was completed. The ice bath was removed, and the reaction was stirred at room temperature for 4 h, and then the mixture was evaporated to ethylamine (ethyl acetate: petroleum ether-1:4, Rf: 0.2).
  • ethylamine ethyl acetate: petroleum ether-1:4, Rf: 0.2
  • Example 10 Preparation of N-2-isonicotinyl-4-phenylthiazole-2-amine (No. TJ-M201050): 4-phenyl-2-aminothiazole (I) lg (5.6 mmol), Isonicotinic acid 1.02 g (8.4 mmol), DMAP 0.68 g (5.6 mmol), DCC 1.73 g (5.6 mmol) were mixed in 15 ml of acetone, and the mixture was shaken in a parallel synthesizer at 25 ° C for 24 h, filtered.
  • Example 11 Preparation of N-2-nicotinoyl-4,5,6,7-tetrahydrobenzo Mthiazole-2-amine (No. TJ-M201057): The reaction process and treatment method were the same as those in Example 10. A pale yellow powdery solid was obtained, yield 34%, m.p.
  • Example 14 Inhibition of acetylcholinesterase by 2-aminothiazole derivatives of TJ-M2010
  • TJ-M201018 (1), TJ-M201005 (2), TJ-M201041 (3), TJ-M201021 (4), TJ- in the aminothiazole derivatives (I) and (II) mentioned in the present invention are selected.
  • Six compounds such as M201064 (5) and TJ-M201057 (6) were tested.
  • a 1% DMSO solution was chosen as the blank control and tacrine was used as the positive control. The results are shown in Figure 1
  • Example 15 Inhibition of PARP-1 by an aminothiazole derivative proposed by the present invention
  • the PA P Universal Color Rendering Kit (R&D Catalog Number 4677-096-K) is based on the principle that the 96-well plate has been fixedly coated with histone-coated and the histone is PARP-1 catalytic reaction. Substrate. A PARP-1 enzyme, a biotin-labeled NAD+, an activator Nicked DNA, and various inhibitors were sequentially added to each well. Activation of PARP-1 by Nicked DNA catalyzes the attachment of PA to histones, resulting in the formation of a PARP-1-PAR-histone complex at the bottom. HRP (horseradish peroxidase) which can develop color is attached to the complex protein.
  • reaction solution is appropriately washed and a substrate TACS which can be catalyzed by HRP is added, and the reading can be carried out at 630 nm using a microplate reader.
  • the reaction can also be stopped with 0.2 N hydrochloric acid and read at 450 nm.
  • the typical PARP-1 inhibitor, 3-AB has an inhibitory effect on PARP-1 in the range of 2 M to 10 mM and is therefore used as a positive control.
  • the aminothiazole derivatives (1), (II) mentioned in the present invention are selected.
  • TJ-M201018 (1), TJ-M201005 (2), TJ-M201041 (3), TJ-M201021 (4), TJ-M201064 (5), TJ-M201057 (6) and TJ-M201061 (7), etc. 7 The compounds were tested. The results showed (see Figure 2) that compounds 1-3 and 7 inhibited PARP-1 by more than 50% at a concentration of 1 M.
  • Example 16 The protective effect of the aminothiazole derivative proposed by the present invention on hydrogen peroxide-induced neuronal apoptosis
  • Human dopaminergic neuroblastoma-SH-SY5Y cells were selected as experimental cells.
  • the SH-SY5Y cells were treated with 3 ⁇ 40 2 at a concentration of ⁇ /L for 12 hours.
  • the cell viability was about 50%, and the model of hydrogen peroxide damage was determined.
  • the aminothiazole derivatives TJ-M201018 (1), TJ-M201005 (2), TJ-M201021 (4) and TJ-M201064 (5) protected by hydrogen peroxide in different concentrations were examined by MTT method. Function, the result is shown in Figure 3.
  • Example 10 Protective effect of the 2-aminothiazole derivative proposed by the present invention on neuronal apoptosis induced by hypoxia injury of cobalt chloride
  • Example 11 The 2-aminothiazole derivative proposed by the present invention is used for treating autoimmune diseases
  • Raw264.7 The number of cells in the 48-well plate is 9*105/well, and the lml medium per well is first pre-incubated with different concentration gradients TJ-M201021 for 2 h, then CPG is added, the final concentration is 40 ug/ml, 37 ° CC02 incubator Incubate overnight (12 h).
  • DC 48-well plate, the number of cells is 1*106/well, 1 ml medium system per well, first pre-incubation with different concentration gradient TJ-M201021 for 2 h, then add LPS, final concentration lug/ml, incubate in 37°CC02 incubator Overnight (12h).
  • aminothiazole derivatives proposed by the present invention exert a concentration-dependent inhibition on the expression of CD80 on the surface of DCs and macrophages, as shown in Fig. 9, Fig. 10.
  • Drug group 1 day before antigen injection, 0-3, 5, 7, 9, 11, 13, 15 days, respectively, intraperitoneal injection of 0.5% CMC in TJ-M2010, 150mg / kg / d; 3. Each group was intraperitoneally injected with mycobacterial antigen and continuously monitored for its concentration;
  • the resulting incidence of type 1 diabetes is shown in Figure 11.
  • the results showed that: MyD88KO heterozygous group gradually increased type I diabetes with time, while MyD88KO homozygous group did not develop type I diabetes.
  • the incidence of type I diabetes in aminothiazole derivative group was similar to that of MyD88KO homozygous group, indicating MyD88 pathway and The occurrence of type 1 diabetes is inextricably linked, and blocking its pathway can reduce the incidence of diabetes. Therefore, the small molecule MyD88 inhibitor TJ-M2010 may become an effective method for prevention and treatment of type I diabetes.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Hospice & Palliative Care (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Description

2-氨基噻唑衍生物及制备方法和应用 技术领域
本发明涉及到结构为 (I) 的一类 2-氨基噻唑化合物的制备及其在通过抑制乙酰胆 碱酯酶, 保护由于氧化应激等因素导致的神经细胞快速凋亡而对早老性痴呆 (AD) 的 治疗作用。通过抑制髓样分化蛋白 (MYD-88 )而产生的免疫抑制作用, 在抗移植排斥、 抗自身免疫疾病、抗缺血再灌注损伤和抗慢性炎症反应、抗内毒素血症等方面的治疗作 用。
Figure imgf000003_0001
背景技术
含 2-氨基噻唑环的化合物在抗菌、消炎、抗过敏等方面有很多应用, 近年来在多巴 胺受体激动剂、血管紧张素受体拮抗剂等类型的药物中也有发现。但在神经退行性疾病 和免疫抑制剂等方面还很少有报道。 虽然报道还很少。
属于原发性神经元变性为基础的神经退行性功能障碍疾病的 AD, 除了使超过 7% 的 60岁以上的老年人蒙受折磨外, 并具有年轻化的趋势, 因此寻找治疗 AD等神经退 行性疾病的有效药物, 已成为世界医学界的研究热点。
导致 AD产生的因素很多:包括由于乙酰胆碱转移酶活性降低以及胆碱摄取能力低 下引起神经元或神经胶质细胞不能提供充分的营养、轴突传递功能受损等导致的皮下神 经元系统功能衰退; 由于体内谷氨酸受体活性过强、 活性氧水平过高 (氧化应激)、 炎 症、病毒感染等导致代谢通路受损、线粒体能量产生减少;由于 β-淀粉样前体蛋白(APP) 经 β-分泌酶(P-secretase)、 γ-分泌酶(γ-secretase)作用产生的 β淀粉样蛋白 (β-amyloid, Αβ) 引起的淀粉样蛋白团块聚集 [6- 9], 由 Tau蛋白的异常磷酸化等导致的神经纤维缠结 以及细胞核或线粒体 DNA突变、 R A等。
目前对 AD的防治是基于对 AD病因的和症状的控制,常用的方法主要包括利用提 高患者的认知能力,减少皮下神经元系统功能的快速衰退, 如利用胆碱受体激动剂或乙 酰胆碱酯酶抑制剂 (如安理申 (Aricept)、 他克林、 多奈哌齐等) 增加体内乙酰胆碱的 量以加强认知功能, 利用降低自由基量 (如 VE和司来吉兰等)和避免胞内钙超载 (如 钙拮抗剂尼莫地平等)药物以减少神经元和突触丢失等方法进行改善,取得一定的效果, 对早期 AD有一定的改善作用。但是迄今为止无论是单纯的乙酰胆碱酯酶抑制剂还是其 它类型的药物的临床效果都不理想。
虽然诱发神经细胞凋亡的调控因素很多,但作用于抑制神经细胞快速凋亡的药物的 作用机制要么是移去细胞凋亡信号、阻止细胞凋亡的启动, 要么是抑制细胞凋亡启动后 所产生的级联反应。 存在于真核细胞中的非组蛋白染色体蛋白聚 ADP-核糖聚合酶 [poly(ADP- ribose) poly-merase, PARP]具有调节许多细胞过程的功能,可修复 DNA和维 持基因组稳定性, 调节转录水平调控有关蛋白的表达, 影响复制和分化 [NgUewa,P. A.; Fuertes, M. A.; Valladares, B.; Alonso, C; Perez, J. M. Poly (ADP-ribose) polymerases: Homology, structural domains and functions. Novel Therapeutical Applications [J]. Prog. Biophys. Mol. Biol, 88, 143-172(2005).] 。 对 PARP的抑制既可以减少由于炎症反应的等 因素对器官造成的损伤, 又可降低 NMDA和 KA的神经兴奋性毒性 [Kauppinen, TM.; Swanson, RA. The role of poly (ADP-ribose) polymerase- 1 in CNS disease. Neuroscience. 145, 1267-1272(2007)] , 具有神经细胞保护作用, 在神经退行性疾病, 神经炎症, 脑缺 血损伤等疾病的治疗中得到广泛关注。
AD属于多因素疾病 (multi-factorial diseases) , 单一靶标药物往往难以奏效的特点, 目前除了在治疗中采用多药联用等方法外, 人们已经注意到多靶标点作用药物的优势 [参见 Bolognesi ML, Minarini An, Tumiatti V, Melchiorre C ; Mini-Reviews in Medicinal Chemistry (2006), 6(1 1), 1269-1274.]。 已经有人利用拼合原理设计出可同时作用于 AChe和脑 MAO的抑制剂 (如拉多替吉 ladostigil、 雷伐斯的明 rivastigmine等) 等。
另外,研究表明各种内源性和外源性的危险因子, 刺激天然免疫系统的各个 TLR, 刺 激信号经关键分子髓样分化蛋白 (myeloid -differentation protein 88 , MyD88)传导, 最后 也激活 NF- B, 产生相应的免疫反应过程 [参见 Maria Loiarro Claudio SetteJ, Grazia Gallo, J BIO CHEM 280, 16 ( 2005 ) 15809-15814]。 因此, MyD88就是天然免疫的关 键分子节点, 阻断了 MyD88就阻断了天然免疫系统的主要反应, 进而产生相应的免疫抑 制效果。 由于 MyD88本身是一个免疫抑制剂新靶点, 已知的 MyD88抑制剂多数都是拟肽 类化合物或结构复杂的小分子化合物, 难以制备, 制备成本较高, 难以在临床上推广应 用 [参见 WO 2006/067091 Al]。
发明内容
本发明设计人在计算机辅助药物设计、药物合成方面, 尤其在对于老年痴呆等疾病 的病因研究和相关药物设计方面,在免疫抑制剂等方面作了一系列前期研究工作, 提出 了针对神经退行性疾病发病的多因素性的多靶点作用药物的想法,构建了多个与神经退 行性疾病密切相关的靶点的药效团模型,设计并合成了多个相应的新型结构的化合物类 型。 考虑到 2-氨基噻唑结构良好的稳定性和抗氧化性, 将 2-氨基噻唑基本结构引入到 神经细胞保护剂、 乙酰胆碱酯酶抑制剂以及免疫抑制剂, 对于提高药理活性, 优化药物 药代动力学特性的可能性是很明显的。
本发明所提出的式 (I) 的 2-氨基噻唑类衍生物, 可以明显的作用于与老年痴呆相 关的乙酰胆碱酯酶和 PARP-1 , 模拟预测的活性超过现用于临床的药物, 并且具有可同 时作用于两个靶点的特性, 因此可在临床上用于老年痴呆的治疗。 同时式 (I) 的氨基 噻唑类衍生物与 MyD88分子的关键活化位点相匹配而能特异性结合, 能够竞争性结合 而抑制 MyD88相应的信号传导, 可在临床上应用于抗移植排斥、 抗自身免疫疾病、 抗 缺血再灌注损伤和抗慢性炎症反应、抗内毒素血症等, 开创了此类小分子物质的应用先 河, 为多种天然免疫相关性疾病找到了一种全新的药物治疗可能。
本发明提出了一类新型的 2-氨基噻唑衍生物。
本发明还提供了同时上述 2-氨基噻唑衍生物及其中间体的制备方法。
本发明提供了利用 2-氨基噻唑衍生物对于乙酰胆碱酯酶和 PARP-1的抑制作用达到 一系列医疗作用, 从而治疗与其相关的多种疾病 (如早老性痴呆 (AD) 及其他神经退 行性疾病) 。
本发明提供了利用 2-氨基噻唑衍生物通过抑制髓样分化蛋白 (MYD-88)而产生的 免疫抑制作用,在抗移植排斥、抗自身免疫疾病、抗缺血再灌注损伤和抗慢性炎症反应、 抗内毒素血症等方面的治疗作用;
本发明的实施方案所提供的式 (I) 的氨基噻唑衍生物的结构为:
Figure imgf000006_0001
(I)
式中 分别为取代芳香基团, R2为 H, 或者 R!R2为 -CH2(CH2)2CH2- 等; R3 为含 c=o H
有氨基的基团; X 为羰基 ( 〜) 或甲烯基 G\ ); n =0〜5。
式 (I) 的 2-氨基噻唑衍生物的合成是先合成相关中间体 (1 )、 (2), (3 ), 如二乙 醇胺的羟基卤代后, 得到的双 (2-二氯乙基)胺盐酸盐与取代芳香胺在微波或常规条件下 环化, 贮备出取代芳香哌嗪 (中间体 1 ); 取代甲基芳香酮和取代硫脲在碘分子存在下 加热反应制得取代 2-氨基噻唑衍生物 (中间体 2), 中间体 2与卤代酰卤反应, 得到取 代 2-氨基噻唑的 ω-卤代酰胺衍生物 (中间体 3 )。 再由相关中间体利用烷基化反应等得 到目标化合物。 如中间体 1和中间体 3通过烷基化反应, 得含有杂环的目标产物 (1)。 中间体 3先与苄胺反应,所得产物再与相应的卤代烃作用, 可得含有一般氨基的目标化 合物(1)。 中间体 2和取代醛基化合物在碱催化下縮合制得权力含甲烯基的目标化合物 ( D o
1、 中间体的合成
Figure imgf000006_0002
2、 目标化合物合成
X为羰基的目标化合物的
方法 1
Figure imgf000006_0003
方法 2
Figure imgf000007_0001
Figure imgf000007_0002
X为甲烯基的目标化合物的合成
Figure imgf000007_0003
另外, 本发明是基于 PARP1受体、 AchE和 MYD88受体为靶标进行设计的, 因此 本发明所设计的化合物对于包括 PARP1受体、 AchE和 MYD88受体等有关的靶点引起 的疾病也有作用。
本发明提到的式 (I) 的 2-氨基噻唑衍生物, 分子量合适, 结构稳定, 可穿透细胞 膜, 预测毒副作用较小。
本发明提到的式 (I) 的 2-氨基噻唑衍生物作为乙酰胆碱酯酶抑制剂, 对于早老性 痴呆 (AD) 及其它神经退行性疾病的治疗作用;
本发明提到的式 (I) 的 2-氨基噻唑衍生物作为神经细胞保护剂, 用于治疗由于氧 化应激等原因导致的神经细胞快速凋亡而引起的疾病。
本发明提到的式(I)的 2-氨基噻唑衍生物作为 MyD88抑制剂, 用于降低移植后排 斥反应及移植免疫耐受诱导和维持方面的应用等。
本发明提到的式 (I) 的 2-氨基噻唑衍生物, 作为免疫调节剂, 治疗多种自身免疫 疾病和慢性炎症性疾病等。
本发明提到的式 (I) 的 2-氨基噻唑衍生物作为免疫调节剂, 治疗缺血再灌注损伤 类疾病上的应用。
本发明的优势在于, 设计并得到了一类结构为式 (I) 的 2-氨基噻唑衍生物, 在相 关的实验中充分证实了其对于乙酰胆碱酯酶和 PARP-1的抑制作用以及对于氧化应激和 缺氧损伤引起的神经细胞快速凋亡的保护作用,有可能作为治疗或预防老年痴呆等神经 退行性疾病的有效药物。 在作为 MyD88特异性抑制剂应用在实验中, 充分证实了该在 用于移植后抗排斥和诱导免疫耐受、治疗各种炎症反应、防治缺血再灌注损伤等方面具 有明显的效果。
2-氨基噻唑结构良好的稳定性和抗氧化性等特性, 非常有利于其衍生物药理活性的 发挥和药物动力学性质的改善。 与原来的乙酰胆碱酯酶抑制剂、 PARP-1抑制剂以及 MyD88抑制剂相比, 本发明的优势还在于式 (I) 的一类 2-氨基噻唑衍生物合成时, 原 料如二乙醇胺, 碘分子, 芳香酮、 硫脲等价廉易得, 步骤少, 产物易于分离和纯化。 附图说明
图 1、 本发明提出的氨基噻唑衍生物对 AchE的抑制作用
图 2、 本发明提出的氨基噻唑衍生物对 PARP-1的抑制作用
图 3、 本发明提出的氨基噻唑衍生物对抗双氧水细胞保护活性 (细胞存活提高率) 图 4 本发明提出的氨基噻唑衍生物对抗氯化钴缺氧损伤的细胞保护活性(细胞存 活提高率)
图 5 氯化钴损伤后的 SH-SY5Y细胞
图 6 氯化钴损伤后加入本发明提出的氨基噻唑衍生物保护后的 SH-SY5Y细胞 图 7-10 本发明提出的氨基噻唑衍生物抑制 LPS、 CpG引起的共刺激分子 CD80 的上调图
图 11 本发明提出的氨基噻唑衍生物减低糖尿病发生率曲线图
附图 1为描述根据本发明所提供的氨基噻唑衍生物 (对于乙酰胆碱酯酶 (AchE) 的抑制活性试验结果的柱状图。 乙酰胆碱酯酶取自于 SD大鼠。
附图 2为描述根据本发明所提供的氨基噻唑衍生物 (浓度为 ΙμΜ) 对于聚 ADP-核 糖聚合酶 -1 (PARP-1 ) 的抑制活性试验结果的柱状图。
附图 3为描述根据本发明所提供的氨基噻唑衍生物对于双氧水( 02)诱发(氧化 应激)的神经细胞凋亡的保护作用试验结果的柱状图, 细胞为人多巴胺能神经母瘤细胞 —— SH-SY5Y细胞。
附图 4为描述根据本发明所提供的氨基噻唑衍生物对于氯化钴 (CoCl2) 诱发 (缺 氧损伤)的神经细胞凋亡的保护作用试验结果的柱状图, 细胞为人多巴胺能神经母瘤细 胞—— SH-SY5Y细胞。
附图 5 为经过氯化钴 (CoCl2 ) 诱发 (缺氧损伤) 的人多巴胺能神经母瘤细胞
—— SH-SY5Y细胞的照片。
附图 6为利用经过氯化钴 (CoCl2) 诱发 (缺氧损伤) 处理, 并用本发明提出的氨 基噻唑衍生物处理后的人多巴胺能神经母瘤细胞一 SH-SY5Y细胞照片。
附图 7 中, 本发明提出的氨基噻唑衍生物剂量相关地减少 T细胞活化;
附图 8中, LPS、 CPG、 心肌组织匀浆溶媒刺激 DC, 本发明提出的氨基噻唑衍生 物下调 CD80的表达。
附图 9为本发明提出的氨基噻唑衍生物剂量相关性下调 DC表面 CD 80表达 附图 10为本发明提出的氨基噻唑衍生物剂量相关性下调巨噬细胞表面 CD80表达。 附图 11中实验分组: MyD88KO NOD小鼠, MyD88KO/+NOD小鼠, NOD小鼠本 发明提出的氨基噻唑衍生物用药组。
1.化合物
本发明不同实施方案中所提及编号为 (I) 的 2-氨基噻唑衍生物, 具有下述所示的
Figure imgf000009_0001
R2为环烷基; 或者 为取代芳香基团, 为11、 的烃基、 -CH2Ph (苄
、c=o 基)或含有 烃基的醚甲基(-CH2OR) ; R3 为含有氨基的基团;X 为羰基( ) 或甲烯基 (-c\ )。 n =0〜5。
本发明的实施方案中, R2为包括含有 3〜7元环的环烃基; 或者当 R2为烃基 (包 -CH3, -C2H5, -CH2Ph (苄基), -CH2OR等)时, 取代芳香基团为取代苯环 (a)、 苄
Figure imgf000010_0001
( a ) ( b ) ( c ) (d) (e) 其巾:
R4为 的烃烷基、 含有 3〜7元环的环烃基、 含 〜 ο烃基的烷氧基、, 含 〜 。烃基的羧酸酯基或卤素基团 (-Cl, -Br, -CN);
R4的取代位置可以在对位、 邻位或间位。
当 (I) 分子中的 X为羰基时, 含有氨基的基团 (R3) 为哌嗪衍生物 (f)、 哌啶衍生 物 (g)、 四氢吡咯衍生物 (h)、 吡啶 (i) 或氨基衍生物 (j)。
Figure imgf000010_0002
R5为取代苯环 (a)、 取代吡啶 (b) 或取代吡咯 (c);
Ar为-苄基或 3-吡啶甲基。
当 X为甲烯基时, 含有氨基的基团 (R5) 为吡啶 (i)、 3-吲哚或 4-咪唑; n=0。 2.化合物的制备方法
本发明的实施方案所提供的式 (I) 的 2-氨基噻唑衍生物的制备方法包括中间体和 目标化合物的制备。
中间体制备
取代苯基哌嗪(1 )的制备 二乙醇胺的羟基卤代后, 得到的双 (2-二氯乙基)胺盐酸 盐与取代芳香胺环化即得, 可利用微波催化法合成, 也可以用常规法合成。
: NHHCI
CIHpCHpC
Figure imgf000010_0003
( 1 ) (微波催化法) 将双 (2-二氯乙基)胺盐酸盐与取代苯胺固体 (摩尔比 1 :1.2)加 入到正丁醇搅拌混合均匀, 195w功率下微波辐射 2 8 min,稍冷却后加入计算量的粉状 无水碳酸钠, 继续微波辐射 15 20 min, 趁热过滤, 滤液静置析晶, 得取代苯基哌嗪。
(2) (常规法) 将双 (2-二氯乙基)胺盐酸盐加入到正丁醇搅拌混合均匀, 搅拌下加 入取代苯胺固体 (与双 (2-二氯乙基)胺盐酸盐的摩尔比 1 : 1.2 ), 加热回流搅拌反应 30-60h, 稍冷却后加入计算量的粉状无水碳酸钠, 继续回流搅拌反应 48-150h, 用 TLC 监测反应至反应完全, 后处理同前。
4,5-二取代 -2-氨基噻唑(2)的制备取代甲基芳香酮和取代硫脲在碘分子存在下加 热反应制得, 可利用微波催化法合成, 也可以用常规法合成。
Figure imgf000011_0001
将研成粉末状的硫脲、 碘按照摩尔比 2:1的比例混合均匀, 加入取代甲酮衍生物搅 拌均匀, 在合适功率的微波条件下反应数分钟, 反应完全后, 加入少量乙醚, 过滤,用 乙醚洗涤沉淀, 得 2-氨基噻唑的氢碘酸盐。将此氢碘酸盐溶于热水中, 搅拌下加入计算 量的碳酸氢钠固体, 调节溶液 pH至中性, 过滤所得沉淀, 用水洗涤, 干燥, 得 2-氨基 噻唑粗品。
4,5-二取代 -2-氨基噻唑 -Ν-ω-卤代酰胺 (3) 的制备 4 5-二取代 2-氨基噻唑与卤代 酰卤反应, 得到取代 2-氨基噻唑的 ω-卤代酰胺衍生物。
Figure imgf000011_0002
将 4 5-二取代 -2-氨基噻唑用于无水 THF中, 冰浴下缓慢滴加卤代酰卤 (如氯乙酰 氯, 与 4 5-二取代 -2-氨基噻唑的摩尔比为 1 :1.2), 滴加完毕后室温搅拌反应, TLC监测 反应至反应完全。 利用层析法分离样品。
氯乙酰氯也可用丙烯酰氯代替, 生成相应的 4 5-二取代 -2-氨基噻唑 -N-氯丙酰胺。 目标化合物制备
方法 1 2- (4-取代哌嗪 -1-基) -N-4, 5-二取代噻唑 -2-乙酰胺的制备 取代 2-氨基 噻唑的 ω-卤代酰胺衍生物 (3 )和取代哌嗪盐酸盐(1 )通过烷基化反应, 得含有杂环的
Figure imgf000012_0001
通法:将等摩尔的取代 2-氨基噻唑的 ω-卤代酰胺衍生物(3 )和取代哌嗪盐酸盐(1 ) 溶于 DMF中, 加入碳酸盐或有机碱及相应的催化剂, 在 50-100°C条件下反应, 用 TLC 监测反应至反应完全。 反应完全后加水处理, 并用有机溶剂提取, 有机层用水洗涤,干 燥, 用色谱法分离。
方法 2 3- (4-取代哌嗪 -1-基) -N-4, 5-二取代噻唑 -2-丙酰胺的制备
制备通法: 将等摩尔的 4,5-二取代 -2-氨基噻唑 -N-丙烯酰胺和取代哌嗪盐酸盐 (1 ) 溶于相关溶剂中, 加入有机碱, 在 50-100°C条件下反应, 用 TLC监测反应至反应完全。 反应完 离。
Figure imgf000012_0002
方法 3 3- (N,N-二取代胺基) -N-4,5-二取代噻唑 -2-酰胺的制备 取代 2-氨基噻唑 的 ω-卤代酰胺衍生物先与 2-胺甲基衍生物反应,所得产物再与相应的卤代烃作用,得含 有一般氨基的目标化合物。 也可用 4,5-二取代 -2-氨基噻唑 -Ν-丙烯酰胺与 2-胺甲基衍生 物进行加成反应, 所得产物再与相关卤代烃发生取代反应, 得相应的 3- (Ν,Ν-二取代胺 基) -Ν-4 5-二取代噻唑 -2-丙酰胺。
Figure imgf000012_0003
将等摩尔的将等摩尔的取代 2-氨基噻唑的 ω-卤代酰胺衍生物 (3 ) 和 2-氨甲基衍生 物溶于相关溶剂中, 加入有机碱, 在 50-100°C条件下反应, 用 TLC监测反应至反应完 全, 减压蒸除溶剂, 再加入 DMF和等摩尔量的卤代烃及相应的碱, 在 50-110°C条件下 搅拌反应, 用 TLC监测反应至反应完全 (约需 4-10h), 反应完全后加水处理, 并用有 机溶剂提取, 有机层用水洗涤, 干燥, 用色谱法分离。
方法 4氨基噻唑衍生物(Π ) 的制备 4,5二取代 -2-氨基噻唑 (2) 与取代甲醛在 有机碱催化下縮合 可利用微波催化法合成, 也可以用常规法合成。
Figure imgf000013_0001
3.治疗方案及方法
用于治疗时的氨基噻唑衍生物 (I) 可以通过任何适当的方法提供给个体, 直接给 药(局部给药, 如注射、 移植或者局部应用于目的组织部位等)和全身系统给药(经注 射途径给药和口服给药等)。 通过静脉注射、 皮下注射、 分子内给药、 眼内给药、 腹腔 给药、 肌肉注射、 口腔含服、、 皮内注射、 经皮肤给药、 气管内给药、 大脑内给药、 颅 内给药、脊柱内给药、心室内给药、鞘膜内给药、脑池内给药、囊内给药和气雾吸入等, 化合物通过非肠胃途径给药应用到某些特定部位,这样就能较好地结合一部分水性的或 者生理学上可以配伍的流质混悬液和水溶液。
本发明所提出的 2-氨基噻唑衍生物 (I) 能用于治疗某些神经退行性疾病, 如早老 性痴呆,氧化应激等原因引起的神经细胞快速凋亡而引起的疾病, 多种自身免疫疾病和 慢性炎症性疾病,缺血再灌注损伤类疾病以及用于降低移植后排斥反应及移植免疫耐受 诱导和维持等方面的疾病。药物能一次性的提供或者连续的提供。新化合物可以单独的 给与或者与其他试剂结合给与, 例如: 化合疗法的化合物。 新药物含有一定量的有治疗 疗效的新化合物。而新化合物的含量将依照病人的病情而定。病人的体重, 病情的严重 程度, 给药方法以及开处方的药师都要列入考虑合适剂量的范围。
本发明所提出的 2-氨基噻唑衍生物 (I) 可以通过适合药物分子的任何途径连续或 者间断给药。 口服给药或腹腔给药都可以做为给药的合适途径。
4.实施例
以下通过不同的实施例对本发明的内容作进一步的描述,但不意味对本发明范围的 限制。
实施例 1: 4-苯基 -2-氨基噻唑的制备 分别将硫脲 7.6g (0.1mol)、 碘 12.7g (0.05mol) 用研钵研细、 混匀, 迅速加入苯乙 酮 6.0g ( 0.05mol), 搅拌混匀后, 在微波反应器中控制 150W-200W反应 2-3min。 反应 完毕后, 加入约 40ml-50ml的乙醚, 快速搅拌, 过滤, 滤饼用少量乙醚洗涤多次, 得微 黄色固体。 将所得固体溶于约 150ml的 80度左右的热水中, 搅拌下加入碳酸氢钠固体 , 调 pH值至 7-8, 抽滤, 并用少量水洗涤多次, 得微黄色固体 5.5g (0.031mol), 粗产 率 62.5%, 无水乙醇重结晶, 静置, 得微黄色的针状结晶
3.4g ( 0.019mol , 产 率 38.6% , 熔 点 153-156°C ) 。 IR ( ΚΒΓ,σ/cm 1 ) 3435(s),3253 (m,v-NH2) ;3113.55 (m,thiazole ring V-HC=); 1599(S,V C=N); 1517(s,Benz ring skelecton) ;1338 (m,vc-N) ;768.8 (thiazole ring skelecton);716(5Benz ring bend.); !H NMR(CDC ,300MHz,5/ppm)7.78(d,J=8Hz,2H,Aryl-H),7.47(t,J=27Hz,2H), 7.28(t,J=15Hz, 1H, Aryl-H), 6.73(s,lH,thiazole-H),5.11(s,2H,-NH2).
实施例 2: 中间体 l-(4-甲氧基苯基)哌嗪的制备
( 1 ) (双(2-二氯乙基)胺烟酸盐的合成, 搅拌下将用氯仿稀释的二乙醇胺的混合 液缓慢滴加到冷却至 0°C左右的氯化亚矾 (与二乙醇胺的摩尔比为 1 :4) 与氯仿的混合 溶液中, 滴加完毕后, 室温条件下继续搅拌反应 2-5 h, 然后逐渐升温至 30-70 °C, 维 持此温反应 2 h, 反应完毕后, 加入无水乙醇, 冷却, 减压过滤, 固体分别用乙醇、 乙 醚洗涤, 干燥得到双 (2-二氯乙基)胺盐酸, 产率 97.8%, mp:205.1〜207.0°C .
(2)将 4.3 g (24 mmol)双 (2-二氯乙基)胺盐酸盐与 2.5 g (20 mmol) 4-甲氧基苯 胺固体加入到 50 mL 正丁醇搅拌混合均匀, 195w功率下微波辐射 6 min, 稍冷却后加 入 1.3g (12 mmol)无水碳酸钠粉末, 继续微波辐射 19 min, 趁热过滤, 少量热的正丁醇 洗涤滤饼, 滤液冷却至室温, 加入一倍体积的无水乙醇混匀后静置。 过滤所得晶体分别 用无水乙醇及乙醚洗涤。乙醇 /乙醚(1 :2)重结晶,真空干燥得白色晶体 3.2 g,产率 76.9%, mp: 203.6〜205.2°C。 UV ( /nm) max=246; HPLC (min) Rt=2.717; ^-ΝΜ^ϋΜΞΟ-βϋ, 300MHz, δ/ppm) 7.03〜6.89(m, 4H, Aryl-H), 4.69(s, 1H, N-H), 3.70 (s, 3H,CH30), 3.32〜 3.32(t, 4H, NCH2), 3.25〜 3.22(t, 4H, NCH2); 13C-NMR(DMSO-6D, 300MHz, δ/ppm) 154.6(C8), 143.6(C5), 119.8(C6, 10), 114.9(C7, 9), 55.6(C11), 47.7(C1, 4), 43.2(C2, 3)
实施例 3: 4-苯基噻唑 -2-氯乙酰胺 (2-chloro-N-(4-phenyltWazol-2-yl) acetamide) 的制备 将 4-苯基 -2-氨基噻唑 3.5g (0.02mol) 溶于尽量少的四氢呋喃中, 冰浴条件下缓慢 滴加氯乙酰氯 2.5g (0.024mol), 溶液呈微黄色, 滴加完毕后撤去冰浴, 室温搅拌反应 4h, TLC监测反应 (乙酸乙酯: 石油醚 -1 :4, Rf: 0.2)。 反应完成后, 产物与硅胶 1 : 15硅胶柱层析,流动相为乙酸乙酯:石油醚 -1 : 4,得到较纯净的黄色颗粒状化合物 1.8g (7.14mmol, 产率 35.7%, 熔点 179-181 °C )。
实施例 4: 2-(4-(4-甲氧基苯基)哌嗪 -1-基) -N-(4-苯基噻唑 -2-基)乙酰胺 (编号为 TJ-M201005) 的制备
将 4-苯基噻唑 -2- (2-氯) -乙酰胺 0.5g (2.1mmol), 1- (4-甲氧基苯基) 哌嗪盐酸 盐 0.48g (2.1mmol)溶于 20mlDMF中, 加入碳酸钾 0.29g (2.1mmol) 以及催化量的碘 化钾, 加热 80°C条件下反应, TLC检测反应进行, 大约反应 5h。 反应完全后, 反应液 中加入饱和食盐水处理, 用二氯甲烷多次萃取, 合并二氯甲烷溶液, 并用饱和食盐水洗 涤, 无水硫酸钠干燥。 浓縮后利用硅胶柱层析法分离, 以乙酸乙酯: 石油醚 (1 : :10)为流 动相,浓縮后得土黄色固体 0.6g ( 1.5mmol产率 70%, 熔点 186-189°C )。 UV (CH3OH) Amax/nm 268.1 , 231.0; !H-NMR (CDCb): . 2.83 (t, J=8.4Hz, 4H), 3.22 (t, J=8.4Hz, 4H), 3.35 (s, 2H), 3.80 (s, 3H), 6.87 (d, J=9.2Hz, 2H), 6.95 (d, J=9.2Hz), 7.18 (s, 1H), 7.27 (s, CDCb), 7.34 (d, J=8.0Hz, 1H), 7.42 (t, J=8.0Hz, 2H), 7.85 (d, J=8.0Hz, 2H); 13C-NMR (CDCb) . 50.85, 53.75, 55.58, 61.10, 107.87, 114.55, 118.66, 126.09, 128.07, 128.76, 134.34, 140.54, 147.35, 150.13, 157.02, 168.38。
实施例 5: 2-(4-(4-甲氧基苯基)哌嗪 -1-基) -N-(4-苯基噻唑 -2-基)丁酰胺 (编号为 TJ-M201018) 的制备
将 4-苯基噻唑 -2- (2-溴) -丁酰胺 0.6g (2.1mmol), 1- (4-甲氧基苯基) 哌嗪盐酸 盐 0.48g (2.1mmol)溶于 20mlDMF中, 加入碳酸钾 0.29g (2.1mmol) 以及催化量的碘 化钾, 反映条件同实施例 4, 得黄色晶体 0.7g ( 1.5mmol产率 73%, 熔点 189-192°C )。 UV (CH3OH) Amax/nm 262.0, 233.4; ¾-NMR (CDCb): 2.68 (m, 2H), 2.83-3.31 (t, J=5.4Hz, 4H), 2.87 (m, 4H), 3.22 (t, J=8.4Hz, 4H), 3.55 (s, 2H), 3.80 (s, 3H), 6.92 (d, J=9.2Hz, 2H), 6.91 (d, J=5.4Hz), 7.15 (s, 1H), 7.34 (d, J=8.0Hz, 1H), 7.42 (t, J=8.0Hz, 2H), 7.85 (d, J=8.0Hz, 2H); 13C-NMR (CDCb), 28.45, 31.26, 50.85, 52.70, 55.59, 61.10, 107.29, 114.55, 118.60, 127. 92, 128.97, 128.76, 134.34, 146.34, 149.35, 155.18, 157.42, 168.38, 206.98。
实施例 6 : 2-(4- (对甲苯基)哌嗪 -1-基) -N-(4-苯基噻唑 -2-基) -乙酰胺 (编号为 TJ-M201002) 的制备
与实施例 5相同,产率 82%,熔点 176-178°C ; UV (CH3OH) Amax/nm 268.1 , 235.1; ¾-NMR (CDCb) . 2.30 (s, 3H), 2.81 (t, J=6.4Hz, 4H), 3.26 (t, J=6.4Hz, 4H), 3.34 (s, 2H) 6.88 (d, J=7.2Hz, 2H), 7.11 (d, J=7.2Hz, 2H) 7.18 (s, 1H) 7.27 (s, CDCb) 6.873-7.868 (m, 5H); 13C-NMR (CDCb) 20.45, 49.85, 53.72, 61.15, 107.86, 116.75 , 126.09, 127.92, 128.60, 128.76, 129.75 , 134.35 , 148.78, 150.14, 157.02, 168.43。
实施例 7: 3-(4-苄基哌嗪 -1-基) -N-(4-苯基噻唑 -2-基)丙酰胺(编号为 TJ-M201015) 的制备
将 0.5g (2.2mmol) 4-苯基噻唑 -2-丙烯酰胺、 0.24g (2.2mmol) 苄基哌嗪溶于 30ml 无水乙醇中, 加入三乙胺 0.22g (2.2mmol), 60°C条件下加热搅拌反应, TLC检测反应 进行 (乙酸乙酯: 石油醚: 三乙胺 -l :4:ld)。 反应完全后减压蒸除大量乙醇, 利用硅胶 柱层析纯化, 流动相 (乙酸乙酯: 石油醚 /三乙胺 -1 :8/1%。), 得到较纯的微黄色粉状固 体化合物 0.5g ( 1.2mmol产率 56.0%, 熔点 197-201 °C )。 UV (CH3OH) Amax/nm 260.0, 225.0; ¾-NMR (CDCb) . 2.62 (t, J=6.0Hz, 2H), 2.70 (s, 8H), 2.79 (t, J=6.0Hz, 2H), 3.63 (s, 2H), 7.15 (s, 1H), 7.27 (s, CDCb), 7.29-7.37 (m, 6H), 7.45 (t, 6.4Hz, 2H), 7.90 (d, J=7.2Hz, 2H); 13C-NMR (CDCb) : . 30.93, 31.17, 52.21 , 52.98, 62.92, 107.28, 125.98, 127.26, 127.82, 128.33, 129.23, 134.61 , 149.82, 157.48, 170.10, 206.97。
实施例 8 : 3-(4-(4-甲氧基苯基)哌嗪 -1-基 -N-(4-苯基噻唑 -2-基)丙酰胺 (编号为 TJ-M201021) 的制备
制备方法同实施例 6, 柱层析流动相 (乙酸乙酯: 石油醚: 三乙胺 -1 : 4: 1%。),产 物为白色片状晶体 0.5g ( 1.2mmol产率 53.9%,熔点 202-204 °C )。 UV (CH3OH) Amax/nm 261.0, 233.2; !H-NMR (CDCb) . 2.68 (m, 4H), 2.83-2.87 (m, 6H), 3.31 (t, J=4.8Hz, 2H), 3.81 (s, 3H), 6.91 (d, J=5.2Hz, 2H), 6.98 (d, J=5.2Hz, 2H), 7.14 (s, 1H), 7.27 (s, CDCb) 7.73 (d, J=7.2Hz, 1H), 7.38 (t, J=7.6Hz, 2H), 7.85 (d, J=5.2Hz, 2H); 13C-NMR (CDCb) . 31.26, 50.80, 52.51, 53.06, 55.59, 107. 28, 114.52, 118.62, 127.81 , 128.62, 134.45, 145.32, 149.86, 154.17, 157.41 , 169.99, 206.98。
实施例 9: 3-(N,N-吡啶甲基-乙基胺) -N-(4-苯基噻唑 -5-甲基 -2-基)丙酰胺(编号为 TJ-M201041) 的合成
将 4-苯基 -5-甲基噻唑 -2-丙烯酰胺 0.5g (2.0mmol)、 2-氨甲基吡啶 0.22g (2.0mmol) 溶于约 30ml无水乙醇, 加入三乙胺 0.21g (2.0mmol), 在 50-100°C条件下搅拌反应, TLC检测反应进行 (乙酸乙酯: 石油醚: 三乙胺 -1 : 3: Id) 反应完全后, 旋蒸除去溶 剂, 加入约 15ml的 DMF及 0.24g (2.2mmol) 的溴乙烷和适量的碳酸钾, 50-100°C条 件下反应 TLC监测反应 (乙酸乙酯: 石油醚: 三乙胺 -4:l :ld)。 反应完毕后, 硅胶柱分 离(流动相为乙酸乙酯:石油醚:三乙胺 -8:1 :1%。),得白色颗粒状化合物 0.41g ( 1.0mmol 产率 50%, 熔点 185-187°C )。 UV ( CH3OH) Amax/nm 269.9, 230.0, ¾-NMR (CDCb) . 1.22 (t, J=7.2Hz, 3H), 2.64 (t, J=6.4Hz, 2H), 2.76 ( q, J=7.2Hz, 2H), 2.89 (t, J=6.4Hz, 2H), 3.25 (s, 3H), 3.78 (s, 2H), 7.27 (s, CDCb), 7.58 (d, J=7.2Hz, 2H), 7.45 (t, J=7.8Hz, 2H), 7.35 (t, J=7.8Hz, 2H), 7.34 (m, 2H), 7.97 (d, J=7.2Hz, 2H), 13C-NMR (CDCb) . 10.36, 31.69, 45.87, 48.11, 58.05, 60.82, 107.13, 125.99, 127.69, 127.77, 128.63, 128.68, 129.48, 134.75, 149.87, 157.30, 170.11 。
实施例 10: N-2-异烟酰基 -4-苯基噻唑 -2-胺(编号为 TJ-M201050) 的制备: 将 4-苯基 -2-氨基噻唑 (I)lg(5.6 mmol) 、 异烟酸 1.02g(8.4mmol)、 DMAP0.68 g (5.6 mmol), DCC1.73 g(5.6 mmol) 混合于 15ml丙酮中, 将混合物在平行合成仪中 25°C条 件下震荡反应 24h, 过滤所得的白色固体, 用丙酮洗涤沉淀(3x3 ml), 合并丙酮溶液并 减压蒸去丙酮, 得到淡黄色固体, 加入蒸馏水洗涤数次至中性, 抽滤得到浅黄色粉末, 用 75%乙醇溶液重结晶, 真空干燥得 0.9g浅黄色针状结晶, 产率 60%, 熔点 213〜216°C 。 UV(MeOH, max/nm): 228.; IR(KBro/cm— 3400(W,VN-H), 3028(W,VAT-H), 1602(m,vc=o) , 1542(s, thiazole ring V-C=N), 704(m, 5Benz ring bend); ^-NMRCCDC ^OOMHz, δ/ρ pm):7.25(dd,lH, Aryl-H), 7.29(dd,2H, Aryl-H), 7.45(d,2H, Aryl-H), 7.58(s,lH, thiazole-H) , 7.67(d,2H,pyridine-H), 8.64(d,2H, pyridine-H), 11.49(s,lH,NH).。
实施例 11: N-2-烟酰基 -4,5,6,7-四氢苯并 M噻唑 -2-胺 (编号为 TJ-M201057) 的制 备: 反应过程和处理方法同实施例 10。得浅黄色粉末状固体,产率 34%,熔点 189〜192°C。 UV(MeOH , max/nm): 212.4.; IR(KBro/cm- 342 l(s, VN-H), 2930(m,pyridine ring v— HC=), 1641(s, vc=o), 1542(s, thiazole ring V-C=N); ^-NMR^CDCb OOMHz, δ/ppm): 1.63(m,4H,CH2): 2.58(m,4H,CH2), 7.38(s,lH, pyridine-H), 8.24(s,lH, pyridine-H), 8.68(s,lH, pyridine-H), 8.77(s,lH, pyridine-H), 9.19(s,lH,-NH -)。
实施例 12 ((1H-吲哚 -3-基)次甲基 )-4-(2,4-二乙氧苯基)噻唑 -2-胺(编号为 TJ-M20106 1) 的制备:
将 4-(2,4-二乙氧基苯基 )-2-氨基噻唑 (c)1.6g (0.006mol)、3-甲醛基吲哚 0.9g (0.006mol) 混合后溶于 20ml无水乙醇, 并滴入 3滴六氢吡啶作催化剂。在微波辐射功率为 65W条 件下搅拌反应 lmin后, 调整微波功率为 130W继续反应 lh, 得到黄色混浊溶液。 趁热 抽滤,滤饼水洗 2〜3次,真空干燥得 2.2g黄色粉末,产率 93.8%, mp 218〜220°C。IR (KBr) /cm"1 3224, 3113, 1523.72, 1486.64. ¾-NMR (DMSO- 6) 1.34 (t,J= 6.6 Hz, 3H), 1.48 (t, J= 6.6 Hz, 3H), 4.01 (q, J= 6.6 Hz, 2H), 4.10 (q, J= 6.6 Hz, 2H), 6.47 (d, J= 2.4 Hz, 1H), 6.48 (dd, J= 7.2 Hz and J= 2.4 Hz, 1H), 7.16-7.21 (m, 2H), 7.58 (d, J= 7.2 Hz, 1H), 7.68 (d, J= 3.6 Hz, 1H), 7.85 (d, J= 7.2 Hz, 1H), 8.15 (d, J = 7.2 Hz, 1H), 8.38 (d, J= 3.6 Hz, 1H), 9.10 (s, 1H), 11.64 (s, 1H). 13C-NMR (DMSO- 6) 171.27, 164.70, 158.31, 156.20, 147.64, 136.52, 134.14, 129.73, 123.84, 122.37, 121.13, 120.62, 115.17, 113.67, 110.96, 104.21, 98.40, 62.74, 62.27, 13.77, 13.69. MS (ESI positive ion) m/z: 392.2 (M + 1).
实施例 13((1H-吲哚 -3-基)次甲基 )-4- (萘 -1-基)噻唑 -2-胺 (编号为 TJ-M201064) 的 合成
将 4- (萘 -1-基) -2-氨基噻唑 0.7g(0.003mol)、 3-甲醛基吲哚 0.4g(0.003mol)和 25ml无 水乙醇混合, 并滴入 3滴六氢吡啶作催化剂。 在微波辐射功率为 65W条件下搅拌反应 lmin后, 调整微波功率为 130W继续反应 1.3h, 得到黄色浊液。 趁热抽滤, 滤饼水洗 2-3次。真空干燥得 l.Og黄色粉末,产率 94.4%,mp 239-241 °C。IR(KBr, σ/cm 1): 3102.94(s, thiazole ring VHC=), 3056.71(W, phenyl ring V=C-H), 1598.24(S, VC=C), 1520.6 l(m, Schiff basevc=N), 1497.64(m, thiazolevc=N), 778.05, 740.33(5 naphthyl ring bend), 740.33(5 phenyl ring bend); -NMI pMSO, 300MHz, δ/ppm): 12.13(s, 1H, NH), 9.21(s, 1H, CH=N), 8.33~8.37(m, 2H, naphthyl-H), 8.27(s, 1H, indyl-H), 7.95〜7.98(m, 2H, naphthyl-H), 7.73, 7.72(d, 1H, indyl-H), 7.64(s, 1H, thiazole-H), 7.54〜7.57(m, 2H, naphthyl-H), 7.54, 7.53(d, 1H, indyl-H), 7.51, 7.50(d, 1H, naphthyl-H), 7.22〜7.27(m, 2H, indyl-H); 13C-NMR(DMSO, 300MHz, δ/ppm): 174.53(C10), 159.57(C9), 152.74(C11), 138.28(C13), 138.16(C8), 134.16(C17), 133.60(C22), 131.39(C1), 129.21(C16), 128.96(C18), 127.96(C21), 127.12(C19), 126.66(C20), 126.52(C15), 126.15(C3), 125.26(C14), 124.18(C5), 122.65(C6), 122.52(C4), 115.63(C7), 114.95(C12), 113.13(C2).
实施例 14: TJ-M2010类 2-氨基噻唑衍生物对乙酰胆碱酯酶的抑制作用
利用 Ellman法测定部分 TJ-M2010氨基噻唑衍生物对乙酰胆碱酯酶的抑制作用。 乙 酰胆碱酯酶取自于 SD大鼠。 SD大鼠 (160-180g) 2只, 低温条件下取大脑海马部, 用 2ml裂解液裂解匀浆, 3000r/min离心 15分钟, 吸取上清液, 分装, -20°C冻存备用。 经 BCA法测定, 匀浆蛋白含量为 11.383mg/mL (吸光度 A=0.428 ), 标准液 10mg/mL (吸 光度 A=0.376)。选取本发明所提到的氨基噻唑衍生物( I )、( II )中的 TJ-M201018 ( 1)、 TJ-M201005 (2)、 TJ-M201041 (3)、 TJ-M201021 (4)、 TJ-M201064 (5)和 TJ-M201057 (6) 等 6个化合物进行试验。 选择 1%的 DMSO酶溶液作为空白对照, 以 tacrine做阳性 对照。 结果见附图 1
实验结果表明, TJ-M2010系列的氨基噻唑衍生物对于 AchE有一定的抑制作用, 其抑制强度接近于 Tacrine。
实施例 15: 本发明提出的氨基噻唑衍生物对 PARP-1的抑制作用
采用的是 PA P通用显色方法试剂盒(R&D Catalog Number 4677-096-K) 原理是: 96孔板的地步已经固定涂布有组蛋白 (histone-coated), 组蛋白为 PARP-1催化反应的 底物。 向每一孔中依次加入 PARP-1酶、 生物素标记了的 NAD+、 激活剂 Nicked DNA、 以及各种抑制剂。 PARP-1被 Nicked DNA激活后会催化 PA 连接到组蛋白上, 从而形 成 PARP-1-PAR-组蛋白复合物固定在底部。 而在复合物蛋白上连接有可以显色的 HRP (辣根过氧化酶)。反应合适的时间后, 吸弃反应液适当清洗并加入可以被 HRP催化显 色的底物 TACS, 可以用酶标仪在 630nm处进行读数。 该反应也可以被 0.2N的盐酸终 止, 并在 450nm处进行读数。
典型的 PARP-1抑制剂 3-AB,在 2 M 到 10 mM 浓度范围内对 PARP-1均有抑制 作用, 因此作为阳性对照。 选取本发明所提到的氨基噻唑衍生物 ( 1 )、 ( II ) 中的 TJ-M201018 ( 1)、 TJ-M201005 (2)、 TJ-M201041 (3)、 TJ-M201021 (4)、 TJ-M201064 ( 5)、 TJ-M201057 (6) 和 TJ-M201061 (7) 等 7个化合物进行试验。 结果表明 (见 附图 2), 化合物 1-3和 7在 1 M的浓度下对 PARP-1的抑制率均大于 50%。
实施例 16: 本发明提出的氨基噻唑衍生物对过氧化氢诱导的神经细胞凋亡的保护 作用
选取人多巴胺能神经母瘤细胞一 SH-SY5Y细胞作为实验细胞, 利用浓度为 ΙΟΟΟμΜ /L 的 ¾02作用 SH-SY5Y细胞 12小时, 细胞存活率能够达到 50%左右, 确定 双氧水损伤模型。 利用 MTT法, 考察本发明提出的氨基噻唑衍生物 TJ-M201018 ( 1)、 TJ-M201005 (2)、 TJ-M201021 (4) 和 TJ-M201064 ( 5) 在不同浓度下对双氧水损伤 细胞的保护作用, 结果见图 3
结果表明, 化合物 4 (TJ-M201021 ) 在 ΙμΜ浓度下对双氧水损伤的 SH-SY5Y细 胞有一定的保护作用, 达到 30%,
实施例 10: 本发明提出的 2-氨基噻唑衍生物对氯化钴缺氧损伤诱导的神经细胞凋 亡的保护作用
选取人多巴胺能神经母瘤细胞一 SH-SY5Y细胞作为实验细胞, 利用浓度为 βΟΟμΜ/L氯化钴作用 24小时, 损伤模型细胞成活率在 55%左右, 确定氯化钴缺氧损伤 模型。利用 ΜΤΤ法,选取本范明所提出的氨基噻唑衍生物 TJ-M201018( 1)、TJ-M201005 (2)、 TJ-M201041 (3)、 TJ-M201021 (4)、 TJ-M201064 (5)、 TJ-M201057 (6)进行 试验, 考察本发明提出的氨基噻唑衍生物的化合物在不同浓度下对氯化钴缺氧损伤细胞 的保护作用, 结果见附图 4
结果表明,本发明提出的氨基噻唑衍生物对氯化钴缺氧损伤的细胞保护具有较好的 保护作用。 在显微镜下观察, 细胞损伤后 (图 5 ) 边缘模糊不清, 细胞数量明显减少; 而加入保护剂 TJ-M201021 (4) 的细胞 (图 6), 细胞数量较多, 细胞边缘明显, 生长 状况良好。
实施例 11: 本发明提出的 2-氨基噻唑衍生物用于治疗自身免疫性疾病
体外实验一细胞流式仪结果: 证明 MyD88抑制剂可阻止 DC成熟, 用于治疗自 身免疫性疾病。
步骤: 1. 应用了本发明提出的氨基噻唑衍生物 TJ-M201021 (见实施例 8)的 BALB/c小 鼠来源骨髓细胞, 破红, 2 xl0 6/ML密度 RPMI1640培养基(加 GM-CSF10ng/ml, IL-4 10ng/ml)
2. 48小时去悬浮细胞, 第六天收悬浮及半贴壁细胞
3. DCs加 TJ-M20102150mM 0培养 1小时,加入坏死心肌上清液, LPS(200ng/ml), Poly I:C ( 20 mg/ml) ,CpG (10 mg/ml)培养 12小时
4. 加流式抗体 FITC标记抗 CD80,CD86,上机检测 TJ-M201021抑制了 RAW264.7 细胞对 TLR刺激物 (LPS、 CpG) 引起的共刺激分子 CD80的上调, 说明 TJ-M201021 能够有效的阻断 TLR信号通路, 进而抑制了细胞的免疫反应, 见图 7, 图 8。
Raw264.7: 48孔板 细胞数为 9*105/孔, 每孔 lml培养基体系, 首先加入不同浓度 梯度 TJ-M201021预孵育 2h, 然后加入 CPG, 终浓度 40ug/ml, 37°CC02培养箱孵育过 夜 (12h)。 加流式抗体 FITC标记抗 CD80,CD86, 上机检测;
DC: 48孔板, 细胞数为 1*106/孔, 每孔 lml培养基体系, 首先加入不同浓度梯度 TJ-M201021预孵育 2h,然后加入 LPS,终浓度 lug/ml, 37°CC02培养箱孵育过夜( 12h)。 加流式抗体 FITC标记抗 CD80,CD86, 检测;
可见本发明提出的氨基噻唑衍生物对 DC和巨噬细胞表面 CD80表达起一浓度相关 性抑制, 见图 9, 图 10。
以上检测结果说明 MyD88抑制剂可降低 CD80表达, 从而阻止 DC细胞的成熟。 DC细胞成熟已被证明为多种自身免疫性疾病如自身免疫性心肌病、 实验自身免疫性葡 萄炎、 I型糖尿病、 多发性硬化症、 红斑狼疮等发病的关键步骤之一。 故 MyD88抑制 剂可用于此类疾病的治疗。
体内试验: 应用 MyD88-/-以及本发明提出的氨基噻唑衍生物对构建 I型糖尿病模 型的影响
实验步骤:
1. 实验分组: MyD88KO NOD小鼠, MyD88KO/+NOD小鼠, NOD小鼠 TJ-M201002
(见实施例 6) 用药;
2. 用药组: 抗原注射前 1天, 第 0-3, 5, 7, 9, 11, 13, 15天分别腹腔注射溶于 0.5%CMC的 TJ-M2010, 150mg/kg/d; 3. 各组均腹腔注射分支杆菌抗原并持续监测其浓度;
4. 清洁级伺养观察 30周, 取尾静脉血, 测非空腹血糖, 连续 2次所测血糖≥22 mmol/L为糖尿病建模标准。
所得 I型糖尿病发生率曲线见图 11。 结果显示: MyD88KO杂合子组随着时间延长 I型糖尿病发生逐渐增加, 而 MyD88KO纯合子组无 I型糖尿病发生, 氨基噻唑衍生物 组 I型糖尿病发生率与 MyD88KO纯合子组相当, 说明 MyD88通路与 I型糖尿病的发 生有必然的联系, 阻断其通路能减少糖尿病的发生, 因此小分子 MyD88抑制剂 TJ-M2010可能成为 I型糖尿病的有效防治方法。
在上文中,结合一些具体的实施方案针对本发明的化合物及其制备方法和应用进行 了描述。但应当理解, 本发明的保护范围不应受到这些具体实施方案的过度限制。 事实 上,对本文所提供的实施方案的修改或调整对于本领域技术人员来说并不偏离本发明的 主旨和范围, 因而包涵在本发明之中。

Claims

权利要求书
1. 具有以下式 (I) 结构 2-
Figure imgf000023_0001
(I)
RiR2为环烷基; 或者 为取代芳香基团, 为8、 〜 !的烃基、 -CH2Ph (苄基) 或含有 〜^!烃基的醚甲基 (-CH2OR) ;
3 为含有氨基的基团:
\
: c=o H
X 为羰基 ( ) 或甲烯基 (^C\)。
n=0
2. 根据权利要求 1所述的中所述的 2-氨基噻唑衍生物, 其特征在于, 所述的环垸 基为含有 3〜7元环的环烃基。
3. 根据权利要求 1所述的中所述的 2-氨基噻唑衍生物, 其特征在于, 所述的取代 芳香 a)、 取代吡啶 (b)、 取代吡咯 (c)、 取代吲哚 (d) 或取代咪唑 (e):
Figure imgf000023_0002
(a) (b) (c) (d) (e) 其巾:
!^为。!〜^的烃烷基、 含有 3〜7元环的环烃基、 含 C CK)烃基的烷氧基、, 含 C! CK)烃基的羧酸酯基或卤素基团 (-CL -Br, -CN);
R4的取代位置可以在对位、 邻位或间位。
4. 根据权利要求 1所述的中所述的 2-氨基噻唑衍生物, 其特征在于, 当 X为羰基 时, 含有氨基的基团 (R3 ) 为哌嗪衍生物 (f)、 哌啶衍生物 (g)、 四氢吡咯衍生物 0 、 吡啶
(1) 或氨基衍生物 ω。
Figure imgf000024_0001
(f) (g) (h)
R5为取代苯环 (a)、 取代吡啶 (b) 或取代吡咯 (c);
Ar为-苄基或 3-吡啶甲基。
5. 根据权利要求 1所述的中所述的 2-氨基噻唑衍生物, 其特征在于, 当 X为甲烯 基时, 含有氨基的基团 (R5) 为吡啶 (i)、 3-吲哚或 4-咪唑; n=0。
6. 根据权利要求 1所述的中所述的 2-氨基噻唑衍生物的制备方法, 其特征在于, 二乙醇胺的羟基用卤素取代后,得到的双 (2-二氯乙基)胺盐酸盐与取代芳香胺在微波或常 规条件下反应, 制得取代芳香哌嗪(中间体 1 ), 取代甲基芳香酮和取代硫脲在碘分子存 在下加热反应制得取代 2-氨基噻唑衍生物(中间体 2), 中间体 2与卤代酰卤反应, 得到 取代 2-氨基噻唑的 ω-卤代酰胺衍生物(中间体 3 ), 中间体 1和中间体 3反应, 可得含有 杂环的目标产物 (1)。 中间体 3先与苄胺反应, 所得产物再与相应的卤代烃作用, 可得 含有一般氨基的目标化合物 (1)。 中间体 2和取代醛基化合物在碱催化下縮合制得权力 要求 5所示的含甲烯基的 (1)。
7. 权利要求 1-5中的 2-氨基噻唑衍生物在制备用于治疗早老性痴呆 (AD) 及其他 神经退行性疾病等方面药物的用途;
8. 权利要求 1-5中的 2-氨基噻唑衍生物在制备作为神经细胞保护剂,用于治疗由于 氧化应激等原因导致的神经细胞凋亡而引起的疾病等方面药物中的用途。
9. 权利要求 1-5中的 2-氨基噻唑衍生物在制备作为 MyD88抑制剂, 用于降低器官 移植后排斥反应等方面药物的用途。
10. 权利要求 1-5中的 2-氨基噻唑衍生物在制备作为免疫调节剂, 用于治疗自身免 疫疾病、 I型糖尿病等疾病和药物的用途。
PCT/CN2012/070811 2011-03-02 2012-02-01 2-氨基噻唑衍生物及制备方法和应用 WO2012116586A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP18020332.5A EP3424912B1 (en) 2011-03-02 2012-02-01 2-aminothiazole derivatives, preparation and use thereof
EP12752555.8A EP2682390A4 (en) 2011-03-02 2012-02-01 2-AMINOTHIAZOL DERIVATIVE, METHOD FOR THE PRODUCTION THEREOF AND USE THEREOF
CA2828801A CA2828801A1 (en) 2011-03-02 2012-02-01 2-aminothiazole derivative, preparation method, and use
JP2013555731A JP2014506897A (ja) 2011-03-02 2012-02-01 2−アミノチアゾール誘導体、その製剤方法および使用
US14/016,126 US9771340B2 (en) 2011-03-02 2013-09-01 2-aminothiazole derivatives and methods of preparing and using the same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201110049687.4 2011-03-02
CN201110049687.4A CN102336720B (zh) 2011-03-02 2011-03-02 2-氨基噻唑衍生物及制备方法和应用

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/016,126 Continuation-In-Part US9771340B2 (en) 2011-03-02 2013-09-01 2-aminothiazole derivatives and methods of preparing and using the same

Publications (1)

Publication Number Publication Date
WO2012116586A1 true WO2012116586A1 (zh) 2012-09-07

Family

ID=45512717

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/070811 WO2012116586A1 (zh) 2011-03-02 2012-02-01 2-氨基噻唑衍生物及制备方法和应用

Country Status (6)

Country Link
US (1) US9771340B2 (zh)
EP (2) EP3424912B1 (zh)
JP (1) JP2014506897A (zh)
CN (1) CN102336720B (zh)
CA (1) CA2828801A1 (zh)
WO (1) WO2012116586A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103224476A (zh) * 2013-05-25 2013-07-31 上高县瑞雅精细化工有限公司 二乙醇胺法制备1-[2-(2-羟基乙氧基)乙基]哌嗪新工艺
WO2015069593A1 (en) * 2013-11-06 2015-05-14 Bristol-Myers Squibb Company Substituted pyridine derivatives useful as gsk-3 inhibitors
US9718804B2 (en) 2013-11-06 2017-08-01 Bristol-Myers Squibb Company GSK-3 inhibitors

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102166214B (zh) * 2011-03-02 2012-12-12 华中科技大学同济医学院附属同济医院 氨基噻唑类MyD88特异性抑制剂在医学上的用途
EP2517898A1 (de) 2011-04-29 2012-10-31 Lanxess Deutschland GmbH Kieselsäurehaltige Kautschukmischungen mit schwefelhaltigen Additiven
RU2621708C2 (ru) 2011-06-22 2017-06-07 ПУРДЬЮ ФАРМА Эл. Пи. Антагонисты trpv1, включающие дигидроксигруппу в качестве заместителя, и их применение
CN103387551B (zh) * 2012-05-11 2015-05-20 中国人民解放军军事医学科学院毒物药物研究所 噻唑类化合物及其用途
CN106668022B (zh) * 2015-11-05 2020-09-15 武汉应内药业有限公司 氨基噻唑类MyD88特异性抑制剂TJM2010-5的应用
US9839267B1 (en) * 2016-12-29 2017-12-12 Shadecraft, Inc. Shading system with artificial intelligence application programming interface
US20190137978A1 (en) * 2017-08-31 2019-05-09 Shadecraft, Inc. Intelligent Umbrella and/or Robotic Shading System Mechanical and Tracking Improvements
CA3090545A1 (en) 2018-02-28 2019-09-06 University Of Southern California Compositions and methods for modulating inflammatory and degenerative disorder
CN109394764B (zh) * 2018-09-03 2021-01-15 温州医科大学 一种n-(噻唑-2-基)-3-(哌嗪-1-基)丙酰胺类化合物在药物制备中的应用
CN113730410A (zh) * 2021-08-05 2021-12-03 武汉应内药业有限公司 一种皮炎膏剂及制备方法
CN115010715B (zh) * 2022-06-24 2023-07-21 河南师范大学 3-(吲哚-2-基)琥珀酰亚胺和吲哚并苯并二氮卓类化合物的合成方法及抗癌活性

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3957766A (en) * 1970-06-19 1976-05-18 Boehringer Mannheim G.M.B.H. Novel nitrofuran compounds and pharmaceutical compositions
WO1993000342A1 (en) * 1991-06-21 1993-01-07 Boehringer Mannheim Italia S.P.A. 2-amino-4-aryl-thiazoles with antiasthmatic and anti-inflammatory activities on the respiratory tract
WO1996037493A1 (de) * 1995-05-26 1996-11-28 Bayer Aktiengesellschaft Pyridyl-thiazole und deren verwendung zum schutz von pflanzen gegen befall durch mikroorganismen
EP1486490A1 (en) * 2002-02-28 2004-12-15 Takeda Chemical Industries, Ltd. Azole compounds
WO2006067091A1 (en) 2004-12-20 2006-06-29 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Myd88 homodimerization inhibitors
WO2007110337A1 (en) * 2006-03-29 2007-10-04 F. Hoffmann-La Roche Ag Pyridine and pyrimidine derivatives as mglur2 antagonists

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2406634A1 (fr) * 1977-10-19 1979-05-18 Fabre Sa Pierre Immunostimulants derives d'amino thiazoles
WO2008101029A2 (en) * 2007-02-13 2008-08-21 Xenon Pharmaceuticals Inc. Use of thiazole, oxazole and imidazole compounds for the treatment of sodium channel-mediated diseases or conditions
WO2010129954A1 (en) * 2009-05-08 2010-11-11 Apogee Biotechnology Corporation Treatment of ischemia-reperfusion injury
WO2011115998A2 (en) * 2010-03-17 2011-09-22 Taivex Therapeutics Inc. Modulators of hec1 activity and methods therefor
CN102166214B (zh) * 2011-03-02 2012-12-12 华中科技大学同济医学院附属同济医院 氨基噻唑类MyD88特异性抑制剂在医学上的用途

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3957766A (en) * 1970-06-19 1976-05-18 Boehringer Mannheim G.M.B.H. Novel nitrofuran compounds and pharmaceutical compositions
WO1993000342A1 (en) * 1991-06-21 1993-01-07 Boehringer Mannheim Italia S.P.A. 2-amino-4-aryl-thiazoles with antiasthmatic and anti-inflammatory activities on the respiratory tract
WO1996037493A1 (de) * 1995-05-26 1996-11-28 Bayer Aktiengesellschaft Pyridyl-thiazole und deren verwendung zum schutz von pflanzen gegen befall durch mikroorganismen
EP1486490A1 (en) * 2002-02-28 2004-12-15 Takeda Chemical Industries, Ltd. Azole compounds
WO2006067091A1 (en) 2004-12-20 2006-06-29 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Myd88 homodimerization inhibitors
WO2007110337A1 (en) * 2006-03-29 2007-10-04 F. Hoffmann-La Roche Ag Pyridine and pyrimidine derivatives as mglur2 antagonists

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BOLOGNESI ML; MINARINI AN; TUMIATTI V; MELCHIORRE C, MINI-REVIEWS IN MEDICINAL CHEMISTRY, vol. 6, no. 11, 2006, pages 1269 - 1274
KAUPPINEN, TM.; SWANSON, RA: "The role of poly (ADP-ribose) polymerase-1 in CNS disease", NEUROSCIENCE, vol. 145, 2007, pages 1267 - 1272
LANDREAU, C. ET AL.: "2-Amino-4-dimethylamino-l-thia-3-azabutadienes as Precursors of Thiazoles", PHOSPHORUS, SULFUR AND SILICON, vol. 177, no. 11, 2002, pages 2651 - 2659, XP008170479 *
MARIA LOIARRO; CLAUDIO SETTE; GRAZIA GALLO, J BIO CHEM, vol. 280, no. 16, 2005, pages 15809 - 15814
NGUEWA, P. A.; FUERTES, M. A.; VALLADARES, B.; ALONSO, C.; PEREZ, J. M.: "Poly (ADP-ribose) polymerases: Homology, structural domains and functions. Novel Therapeutical Applications [J", PROG. BIOPHYS. MOL. BIOL., vol. 88, 2005, pages 143 - 172
PAPADOPOULOU, CHRISTINA ET AL.: "Synthesis and biological evaluation of new thiazolyl/benzothiazolyl-amides, derivatives of 4-phenyl-piperazine", FARMACO, vol. 60, no. 11-12, 2005, pages 969 - 973, XP027697620 *
See also references of EP2682390A4

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103224476A (zh) * 2013-05-25 2013-07-31 上高县瑞雅精细化工有限公司 二乙醇胺法制备1-[2-(2-羟基乙氧基)乙基]哌嗪新工艺
WO2015069593A1 (en) * 2013-11-06 2015-05-14 Bristol-Myers Squibb Company Substituted pyridine derivatives useful as gsk-3 inhibitors
CN105849097A (zh) * 2013-11-06 2016-08-10 百时美施贵宝公司 取代的吡啶衍生物用作gsk-3抑制剂
US9718804B2 (en) 2013-11-06 2017-08-01 Bristol-Myers Squibb Company GSK-3 inhibitors
US9809573B2 (en) 2013-11-06 2017-11-07 Bristol-Myers Squibb Company Substituted pyridine derivatives useful as GSK-3 inhibitors

Also Published As

Publication number Publication date
EP3424912B1 (en) 2022-09-07
EP2682390A1 (en) 2014-01-08
CN102336720B (zh) 2016-01-13
CA2828801A1 (en) 2012-09-07
CN102336720A (zh) 2012-02-01
EP2682390A4 (en) 2014-07-23
EP3424912A1 (en) 2019-01-09
US20140004155A1 (en) 2014-01-02
JP2014506897A (ja) 2014-03-20
US9771340B2 (en) 2017-09-26

Similar Documents

Publication Publication Date Title
WO2012116586A1 (zh) 2-氨基噻唑衍生物及制备方法和应用
AU2007208240B2 (en) Substituted biaryl compounds for inflammation and immune-related uses
TW495498B (en) Amide derivative and a pharmaceutical composition containing the same as a Ca2+ release activated Ca2+ channel inhibitor
KR101088557B1 (ko) 염증성 사이토카인 생산 유리 억제제
EP2555768B1 (en) Ire-1 inhibitors
EP2639229B9 (en) Thiazole Derivative and use thereof as VAP-1 Inhibitor
JP5670266B2 (ja) フラザノベンゾイミダゾール
EP2704701B1 (en) Compounds for inflammation and immune-related uses
AU2007230911A1 (en) Benzimidazolyl-pyridine compounds for inflammation and immune-related uses
JP2020509062A (ja) 2−(置換フェニルヘテロ)芳香族カルボン酸系fto阻害剤、その製造方法およびその使用
JP2004504323A (ja) インドロキナゾリノン類
WO2011001931A1 (ja) 新規オキサジアゾール誘導体
JPH0987282A (ja) チアゾール誘導体
US5599813A (en) Thiazolopyrimidine derivatives
EP0398179B1 (en) Rhodanine derivatives and pharmaceutical compositions
JPS6251272B2 (zh)
JP2013533258A (ja) 細胞増殖を阻害するための化合物
CN108623537B (zh) 含胺基侧链的芳香胺类乙酰胆碱酯酶抑制剂合成与用途
JPH08502517A (ja) 抗炎症薬としてのオキシンドール1−[n−(アルコキシカルボニル)カルボキサミド類および1−(n−カルボキサミド)カルボキサミド類
CN114634500A (zh) 一种ptp1b抑制剂及其合成方法和用途
WO2013063385A1 (en) Compounds for inflammation and immune-related uses
CN108640872B (zh) 一种gsk-3抑制剂及其制备方法与应用
KR20120113886A (ko) 잔틴 옥시다아제 저해제로서 효과적인 신규 화합물 및 그를 함유한 약제학적 조성물
JPH06228095A (ja) カルバゾール誘導体
JP7017797B2 (ja) 新規なドーパミンd3受容体選択的リガンド及びびその調製方法並びに医薬使用

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: 12752555

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2828801

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013555731

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: 2012752555

Country of ref document: EP