WO2015007191A1 - 帕利哌酮氨基酸类衍生物及其应用 - Google Patents

帕利哌酮氨基酸类衍生物及其应用 Download PDF

Info

Publication number
WO2015007191A1
WO2015007191A1 PCT/CN2014/082111 CN2014082111W WO2015007191A1 WO 2015007191 A1 WO2015007191 A1 WO 2015007191A1 CN 2014082111 W CN2014082111 W CN 2014082111W WO 2015007191 A1 WO2015007191 A1 WO 2015007191A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
acceptable salt
group
compound
methyl
Prior art date
Application number
PCT/CN2014/082111
Other languages
English (en)
French (fr)
Inventor
张桂森
陈寅
孙家权
徐祥清
于民权
刘欣
刘笔锋
Original Assignee
江苏恩华药业股份有限公司
武汉珈瑜科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 江苏恩华药业股份有限公司, 武汉珈瑜科技有限公司 filed Critical 江苏恩华药业股份有限公司
Publication of WO2015007191A1 publication Critical patent/WO2015007191A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants

Definitions

  • the invention belongs to the field of medicinal chemistry, and particularly relates to paliperidone amino acid derivatives and applications thereof. Background technique
  • Schizophrenia is the most serious and most harmful of all mental illnesses, with a global incidence of about 1-2%.
  • the lifetime prevalence of schizophrenia patients is 0.7-0.8%, which is not significantly related to gender, ethnicity, or social boundaries, and mortality is 2-3 times higher than that of the general population.
  • the latest research shows that the social burden of mental illness ranks first in Chinese diseases, exceeding the cardiovascular, respiratory and malignant diseases.
  • Typical anti-schizophrenic drugs (such as chlorpromazine and haloperidol) block the dopamine D 2 receptor and have a good effect on positive symptoms of schizophrenia.
  • EPS extrapyramidal
  • prolactin prolactin
  • Atypical anti-schizophrenic drugs represented by clozapine and risperidone, not only have a strong effect on dopamine (D 2 ) receptors, but also have a strong effect on serotonin (5-HT 2A ) receptors. Compared with typical anti-schizophrenic drugs, these drugs have great advantages: good effects on positive symptoms of schizophrenia; side effects such as extrapyramidal reaction and tardive dyskinesia are significantly reduced; some atypical anti-schizophrenia drugs Negative symptoms and cognitive impairment have some improvement.
  • the current clinical application of atypical anti-schizophrenic drugs have varying degrees of QT interval prolongation and high prolactin and other adverse reactions. Therefore, it is very important to find new drugs that can effectively cure schizophrenia and have fewer side effects.
  • GABA Y-aminobutyric acid
  • GABA agonists have hydrophilic groups (such as carboxylic acids and amino groups) that are not easily passed through the blood-brain barrier, their use is limited. However, it has been found that chemically binding to fatty amino acids or peptides allows these compounds to pass through the blood-brain barrier in large amounts. Improve the side effects of the drug and expand the scope of treatment.
  • Paliperidone as atypical antipsychotics, has a high affinity for the 5 D 2 receptors, the affinity of dopamine in the brain and greater than 01 02 receptors and dopamine. Clinical trials have shown that paliperidone is effective in treating both positive and negative symptoms of schizophrenia. However, at the same time, paliperidone has high affinity for histamine receptors and adrenergic receptors, so it is easy to cause weight gain, high prolactin and orthostatic hypotension in the application. During the long-term dosing of splitting, studies have shown that side effects of weight gain are closely related to histamine receptors.
  • the object of the present invention is to provide a novel active paliperidone amino acid derivative based on the prior art.
  • Another object of the present invention is to provide an application of the above-described alipenemone amino acid derivative for the preparation of a medicament for treating neuropsychiatric diseases.
  • the invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof:
  • R ⁇ PR 2 are each independently selected from hydrogen, a substituted or unsubstituted alkyl with; R 3 is hydrogen, or with [ ⁇ connected to form a substituted or unsubstituted C 4 - 6 cycloalkyl group embankment.
  • the said! ⁇ Preferred is hydrogen or methyl.
  • the unsubstituted fluorenyl group is preferably a methyl group, an ethyl group, a propyl group, a butyl group, a pentyl group or an isopentyl group
  • the substituted 5 fluorenyl group is preferably a d- 5 halogenated fluorenyl group or a d- 5 hydroxy fluorenyl group.
  • the unsubstituted C 4 - 6 cycloalkyl is preferably alkyl with from cyclobutyl, cyclopentyl or cyclohexyl group, more preferably cyclopentyl or cyclohexyl.
  • hydrogen, methyl, isobutyl, isopentyl or, in combination with [ ⁇ , constitutes a cyclopentyl group, a cyclohexyl group or a cyclobutyl group, and further preferably is bonded to [ ⁇ , which constitutes a cyclopentyl group or a cyclohexyl group.
  • the compound of the formula (I) or a pharmaceutically acceptable salt thereof according to the invention is preferably selected from any one of the following compounds or a pharmaceutically acceptable salt:
  • Pharmaceutically acceptable salts of the compounds of the invention include, but are not limited to: hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, hydrogen sulfate, phosphate, acid phosphate, acetate , lactate, citrate, tartrate, maleate, fumarate, methanesulfonate, gluconate, saccharide, benzoate, ethanesulfonate, benzenesulfonic acid Salt or p-toluenesulfonate.
  • the pharmaceutically acceptable salt of the compound of the invention is the hydrochloride salt.
  • the invention in another aspect, relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention and/or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier and/or excipient.
  • the present invention relates to a compound of the present invention and/or a pharmaceutically acceptable salt thereof or a compound represented by the formula (I) and/or a pharmaceutically acceptable salt thereof as a main active ingredient of the present invention
  • a compound of the present invention and/or a pharmaceutically acceptable salt thereof or a compound represented by the formula (I) and/or a pharmaceutically acceptable salt thereof as a main active ingredient of the present invention
  • the invention relates to a method of treating a prophylactic or therapeutic neuropsychiatric disorder comprising administering to a subject suffering from a neuropsychiatric disorder a therapeutically effective amount of a compound of the invention and/or a pharmaceutically acceptable salt thereof.
  • mercapto refers to a straight or branched, saturated monovalent hydrocarbon radical having 1, 2, 3, 4 or 5 carbon atoms.
  • fluorenyl groups include methyl, ethyl, and propyl.
  • Base butyl, pentyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1 , 2-dimethylpropyl, neopentyl or 1,1-dimethylpropyl.
  • d- 5 is methyl, ethyl, propyl, butyl, pentyl or isopentyl .
  • the 5- indenyl group in the compound of the formula (I) of the present invention may be optionally substituted by one or more groups selected from a halogen and a hydroxyl group, for example, a 5- halofluorenyl group and a d- 5 hydroxyl group. ⁇ .
  • the halocinyl group is a trifluoromethyl group and the hydroxymethyl group is a hydroxymethyl group.
  • pharmaceutically acceptable salt refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the invention. See, for example, SM Berge et al., "Pharmaceutical Salts,” J. Pharm. Sci 1977, 66, 1-19.
  • the general synthetic method for this class of compounds is to first synthesize (BOC) 20 protection of aminobutyric acid, then connect to the hydroxyl group of paliperidone, and finally remove the BOC protection to obtain the product.
  • BOC BOC 20 protection of aminobutyric acid
  • the compounds of the invention, or pharmaceutically acceptable salts thereof may act systemically and/or locally.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be administered by a suitable method, including, but not limited to, oral, injection, parenteral administration and the like, as needed.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier and/or excipient, may be formulated into a desired administration form, including but not limited to tablets, powders, depending on the route of administration. Capsules, solutions, suspensions, suppositories, patches, granules, ointments, lotions, and the like. This can be done by prior art methods. For example, it can be accomplished by mixing a compound of the present invention or a pharmaceutically acceptable salt thereof with a pharmaceutically suitable excipient.
  • Examples of pharmaceutically suitable excipients that can be used in the present invention include, but are not limited to, solvents, emulsifiers, dispersants, wetting agents, binders, stabilizers, colorants, and odor and/or taste masking agents.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can also be used in combination with other known drugs for treating neuropsychiatric diseases.
  • Drugs known in the art for treating neuropsychiatric disorders include, for example, risperidone, aripiprazole, amisulpride, fluoxetine, alprazolam, midazolam, citalopram, and diazepam. Wait.
  • the present invention is also a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and/or a pharmaceutically acceptable salt thereof, and optionally other known therapeutic neuropsychiatric disorders The drug, as well as pharmaceutically suitable excipients.
  • compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention and/or a pharmaceutically acceptable salt thereof, and a pharmaceutically suitable excipient.
  • the pharmaceutical composition of the present invention may comprise from about 0.01 to 1000 mg, preferably from 1.0 to 300 mg, more preferably from 10 to 150 mg, most preferably 100 mg of the compound of the present invention and/or a pharmaceutically acceptable salt thereof per unit dose.
  • the pharmaceutical composition of the present invention should comprise at least 0.5 wt%, preferably 4 wt% to 70 wt%, more preferably 10 wt% to IJ 50 wt%, most preferably 30 wt%, based on the total weight of the pharmaceutical composition of the present invention.
  • a compound of the invention and/or a pharmaceutically acceptable salt thereof may comprise from about 0.01 to 1000 mg, preferably from 1.0 to 300 mg, more preferably from 10 to 150 mg, most preferably 100 mg of the compound of the present invention and/or a pharmaceutically acceptable salt thereof per unit dose.
  • the pharmaceutical composition of the present invention should comprise at least 0.5 wt%, preferably 4 wt% to 70 wt%, more preferably 10 wt% to IJ 50
  • the dose of the compound of the present invention or a pharmaceutically acceptable salt thereof contained in the pharmaceutical composition of the present invention depends on the type and severity of the disease or condition, and the characteristics of the subject, such as general health, age, sex, body weight and drug Tolerance. Those skilled in the art will be able to determine appropriate dosages of the active compounds of the present invention based on these or other factors.
  • the effective dosage of a central nervous system drug which is generally used is well known to the skilled person, and the total daily dose is usually between about 0.05 mg and 2000 mg.
  • Yet another aspect of the present invention provides a compound of the present invention and/or a pharmaceutically acceptable salt thereof Use in the preparation of a medicament for the prevention or treatment of neuropsychiatric disorders.
  • treating includes overcoming, alleviating, ameliorating, relieving or ameliorating a disease or condition. In some cases, the term “treatment” also includes “prevention.”
  • neuropsychiatric disease refers to a general term for neurological and psychiatric disorders.
  • exemplary neuropsychiatric disorders include, but are not limited to, mental disorders, anxiety disorders, personality disorders, depression.
  • the neuropsychiatric disease in the present invention is schizophrenia.
  • a further aspect of the present invention provides a method for preventing or treating a neuropsychiatric disease using an effective amount of a compound of the present invention and/or a pharmaceutically acceptable salt thereof, which comprises a subject having a neuropsychiatric disorder A therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, is administered.
  • subject as used herein includes mammals, preferably humans.
  • the in vitro receptor binding assay showed that the derivatives of the present invention have affinity for three receptors (D 2 , 5-HT 1A , and 5-HT 2A ) comparable to paliperidone, whereas for H ⁇ P ai Low affinity, less likely to cause side effects of weight gain.
  • paliperidone is linked to an amino acid to form an ester prodrug, which has potential anti-schizophrenia activity similar to paliperidone, and can reduce paliperidone weight gain, prolactin elevation or orthostatic hypotension Such side effects can also improve the symptoms of cognitive impairment.
  • paliperidone amino acid derivatives have low acute toxicity and high therapeutic index compared to paliperidone.
  • the title compound 2 was prepared by the method of Example 1 using 3-aminomethyl-5-methylhexanoic acid in place of 4-aminobutyric acid.
  • the title compound 3 was prepared by the method of Example 1 using 4-amino-2-methylbutyric acid in place of 4-aminobutyric acid.
  • Rats were decapitated, operated on ice, and the cortex was quickly taken. Add 3 ml of homogenate (0.05 M Tris-HCI buffer containing 0.1% ascorbic acid, 10 um eugenin and 4 mM CaCI 2 ), homogenate, and then add 5 ml buffer. The solution (0.05M Tris-HCI buffer containing 0.1% ascorbic acid, 10um eugenin and 4mM CaCI 2 ) was incubated at 37 ° C for 10 min. After incubation, the test tube was adjusted with a balance and centrifuged at 12000 rpm at 4 ° C.
  • homogenate 0.05 M Tris-HCI buffer containing 0.1% ascorbic acid, 10 um eugenin and 4 mM CaCI 2
  • Isotope ligand 3 H-8-OH-DPAT (67.0 Ci/mmol), purchased from PerkinElmer; 5-HT, purchased from RBI; GF/C glass fiber filter paper, purchased from Whatman; Tris imported package 2, 5-Diphenyloxazole (PPO) and 1,4-bis(5-phenyl-2-oxazolyl)benzene (POPOP) were purchased from Shanghai Reagent No. 1 Plant. Beckman LS-6500 multi-function liquid scintillation counter.
  • Radioactive ligands 3 H-8-OH-DPAT ⁇ were added to each reaction tube (two parallel tubes were set for each reaction tube, and each tube was placed on ice when the sample was applied).
  • Inhibition rate (I %) (total binding tube cpm - compound cpm) / (total binding tube cpm - non-specific binding tube cpm) xl00%
  • Rats were decapitated, operated on ice, and the cortex was quickly taken. Add 3 ml of buffer (0.05 M Tris-HCI buffer: 6.05 g Tris dissolved in 1000 ml of double distilled water and adjusted to pH 7.5 with concentrated HCI).
  • buffer 0.05 M Tris-HCI buffer: 6.05 g Tris dissolved in 1000 ml of double distilled water and adjusted to pH 7.5 with concentrated HCI.
  • Isotope ligand [ 3 H]- Ketanserin ( 67.0 Ci/mmol ) available from PerkinElmer; Methysergide, purchased from RBI; GF/C glass fiber filter paper, purchased from Whatman; Tris imported package; PPO, POPOP purchased from Shanghai Reagent No. 1; fat-soluble scintillation fluid. Beckman LS-6500 multi-function liquid scintillation counter.
  • Each reaction tube was separately added with a radioligand 3 H-Ketanserin ⁇ (two parallel tubes were set for each reaction tube, and each tube was placed on ice when the sample was applied).
  • reaction tubes were incubated at 37 ° C for 15 min. After the reaction was completed, the combined ligands were quickly filtered by decompression, washed thoroughly with ice-cold test buffer, and the filter was taken out and placed in a 3 ml scintillation cup, and 2 ml was added. Toluene scintillation solution and mix;
  • Inhibition rate (I %) (total binding tube cpm - compound cpm) / (total binding tube cpm - non-specific binding tube cpm) xl00%
  • Rats were decapitated, operated on ice, and the brain striatum was quickly taken.
  • Add 3 ml of buffer (0.05 M Tris-HCI buffer containing NaCI 120 mM, KCI 5 mM, MgCI 2 lmM, CaCI 2 ImM) in 4 steps 3-4 s homogenate, homogenate 4 times, then add 5ml buffer, adjust the weight of the homogenized test tube with the balance, centrifuge at 12000r, 4 °C for 20 min, discard the supernatant, add 3ml of B solution, mix with a vortex mixer Evenly, add 5 ml of buffer, centrifuge, repeat three times of centrifugation, centrifuge, and discard the supernatant. Store the pellet at -80 °C for later use.
  • Receptor binding experimental materials 0.05 M Tris-HCI buffer containing NaCI 120 mM, KCI 5 mM, MgCI 2 lmM, CaCI 2 ImM
  • Isotope ligand 3 H-Spiperone (67.0 Ci/mmol), purchased from PerkinElmer; Butaclamol, purchased from RBI; GF/C glass fiber filter paper, purchased from Whatman; Tris imported package; PPO, POPOP from Shanghai reagent One plant; fat-soluble scintillation fluid. Beckman LS-6500 multi-function liquid scintillation counter.
  • Each reaction tube was separately charged with a radioactive ligand ⁇ -Spiperone ⁇ (two parallel tubes were set for each reaction tube, and each tube was placed on ice when the sample was applied).
  • Inhibition rate (I %) (total binding tube cpm - compound cpm) / (total binding tube cpm - non-specific binding tube cpm) xl00%
  • Rats were decapitated, operated on ice, and the brain cortex was quickly taken.
  • Add 0.05 M Tris-HCl buffer (pH 7.7) mix with a vortex mixer, centrifuge at 48000 g for 15 min at 4 ° C, discard the supernatant, and take a precipitate.
  • Add 0.05M Tris-HCI buffer (pH 7.7) to wash, repeat three times of centrifugation, centrifuge, and discard the supernatant, and store the pellet at -80 °C for later use.
  • Step 1 Firstly prepare the prepared membrane with a suitable amount of buffer 0.05MTris-HCI (pH 7.7), disperse evenly with a homogenizer, mix 15 tubes into a 100ml container, and add an appropriate amount of homogenate.
  • buffer 0.05MTris-HCI pH 7.7
  • Step 2 Each reaction tube was separately added with a membrane preparation of 100 ⁇ M, and a buffer of 100 ⁇ .
  • Step 3 Total binding tube ( ⁇ ) Add 100 ⁇ _ homogenate, non-specific binding tube ( ⁇ ) to prazosin ⁇ (final concentration 10 - 5 ⁇ ), each test compound specific binding tube (SB) is added 100 ⁇ of test compound (final concentration of 10- 5 M);
  • Step 4 Add radioactive ligands 3 Hp raZ0S in ⁇ to each reaction tube (two parallel tubes are set for each reaction tube, and each tube is placed on ice when loading).
  • Step 5 Incubate each reaction tube at 25 ° C for 60 min. After the reaction is completed, the combined ligands are quickly filtered by decompression, thoroughly washed with ice-cold test buffer, and the filter is taken out and placed in a 3 ml scintillation cup. 2ml of toluene scintillation fluid and mix well;
  • Step 6 Put the scintillation bottle into the liquid scintillation counter
  • Inhibition rate (I %) (total binding tube cpm - compound cpm) / (total binding tube cpm - non-specific binding tube cpm) xl00%
  • Rats were decapitated, operated on ice, quickly taken the rat cerebellum, added buffer (0.05 M Tris-HCI buffer, containing 5 nM EDTA, pH 7.7), mixed with a vortex mixer, at 48000 g, 4 ° C Centrifuge for 10 min, discard the supernatant, take a pellet, and add buffer (0.05 M Tris-HCI buffer containing 5 nM EDTA, pH 7.7) Wash, repeat three times of centrifugation, centrifuge, and discard the supernatant, store the pellet at -80 °C for later use.
  • buffer 0.05 M Tris-HCI buffer, containing 5 nM EDTA, pH 7.7
  • the prepared membrane was firstly mixed with an appropriate amount of buffer (0.05 M Tris-HCI buffer containing 5 nM EDTA, pH 7.7). Disperse evenly with a homogenizer, mix 15 tubes into a 100 ml container, add a suitable amount of buffer to a 50 ml membrane suspension, and set aside.
  • buffer 0.05 M Tris-HCI buffer containing 5 nM EDTA, pH 7.7.
  • Step 2 Each reaction tube was separately added to the membrane preparation 100 ⁇ .
  • the third step The total binding tube ( ⁇ ) was added 100 ⁇ _ buffer, non-specific binding tubes ( ⁇ ) was added ⁇ promethazine (final concentration of 10- 5 M), each test compound specifically binds tube (SB) was added by 100 ⁇ _ of test compound (final concentration of 10- 5 M);
  • Step 4 Each reaction tube is separately added with radioactive ligand 3 H- P y r ilami ne l ( ⁇ L (each reaction tube is provided with 2 parallel tubes, and each tube is placed on ice when the sample is applied).
  • Step 5 Incubate each reaction tube at 30 ° C for 60 min. After the reaction is completed, the combined ligand is rapidly filtered by decompression, thoroughly washed with ice-cold test buffer, and the filter is taken out and placed in a 3 ml scintillation cup. 2ml of toluene scintillation fluid and mix well;
  • Step 6 Put the scintillation bottle into the liquid scintillation counter
  • Inhibition rate (I %) (total binding tube cpm - compound cpm) / (total binding tube cpm - non-specific binding tube cpm) xl00%
  • mice Male and female, weighing (20 ⁇ 2) g, were provided by Nanjing Qinglongshan Animal Breeding Center. Ascorbic acid, Sinopharm Chemical Reagent Co., Ltd.;
  • MK-801 produced by Sigma, USA, preparation method: 0.1% vitamin C is formulated into a solution of lmg/ml;
  • Positive drugs tested haloperidol, clozapine, risperidone, olanzapine, aripiprazole, ziprasidone, quetiapine;
  • mice were randomly divided into a blank group, a model group, a positive control group (risperidone group), and a drug group.
  • the blank group and the model group were intragastrically administered with 10% Tween 0.1 ml/10 g, and the positive control group was given risperidone O.lmg/kg by gavage.
  • the drug group was given the corresponding dose of the drug.
  • the blank group was intraperitoneally injected with 0.1% ascorbic acid 0.1ml/10g
  • the model group, the positive control group (30min) were intraperitoneally injected with MK-801 solution 0.1mg/kg. Thereafter, spontaneous activity of each group of mice was measured within 90 minutes. The results are shown in Table 3.
  • Example 11 Apomorphine-induced mouse climbing experiment
  • Apomorphine supplied by Sigma, dissolves 0.9% NaCI (containing 0.1% vitamin C) before use, and is ready for use;
  • the instrument clamps the climbing cage, the stopwatch.
  • mice male, weighing 18-22 g, were randomly divided into negative control group, model group, quetiapine-positive drug group and compound dose group (see Table below for specific doses), with 10 rats in each group.
  • the negative control group and the model group were given the corresponding solvent double distilled water by gavage, and the positive drug group was given the corresponding positive drug by gavage.
  • the solution is first, add a small amount of acetic acid, and then add double distilled water.
  • Each dose of the compound is intragastrically administered with the corresponding dose of the compound, and the intragastric volume is 0.1 ml/10 g.
  • apomorphine (1 mg/kg) was injected subcutaneously in a volume of 0.1 ml/10 g.
  • Self-made grab bar equipment A stainless steel rod with a diameter of 0.3 cm and a height of 5 cm above the table is placed in the mouse box. experimental method:
  • mice male and female, weighing 20 ⁇ 24g, were randomly divided into negative control group, model group, quetiapine dose group and compound dose group, with 10 groups in each group.
  • the negative control group and the model group were given the corresponding solvent double distilled water by gavage, and the positive drug group was given the corresponding positive drug by intragastric administration (first adding a small amount of acetic acid and then adding double distilled water), and each dose group of the compound was intragastrically administered with the corresponding dose of the compound.
  • the volume of the gavage was 0.1 ml/10 g.
  • Sequential method limit experiment KM mice, male and female, were randomly divided into several groups, 2-5 in each group, respectively, each compound 2000mg/kg group and solvent group, administered by 0.2ml/10g. Animals were observed for death within 3 days. (If the animal survives in 3 or more days within 3 days, if there is no abnormality in the state of life, continue to observe until the end of the experiment after 7 days. If the animal dies within 3 or 3 days within 3 days, the median lethal dose method is used. Determine its LD50.
  • mice were pre-tested by median lethal dose method, male and female, randomly divided into several groups, 4 in each group, respectively, each group of 1500mg/kg, 1000mg/kg, 500mg/kg group and solvent group, according to 0.2ml/10g
  • the rats were administered by gavage, and the animals were observed for death within 1-3 days.
  • mice were given a single dose of LD 5 . More than 2000mg/kg, much higher than paliperidone (800mg/kg), with less acute toxicity.
  • Example 14 study of mouse obesity and prolactin
  • mice 100 male ICR mice were randomly divided into three groups: the negative control group, the compound 4 low, medium and high dose groups, and the paliperidone low, medium and high dose groups.
  • the drug was administered once a day for 28 days and the body weight was monitored daily. After the end of the administration, the eyeballs were taken for blood collection, and the blood was allowed to stand at room temperature for 30 minutes, and then centrifuged at 4 ° C, 3000 r / min for 10 minutes using a low-temperature high-speed centrifuge. The collected serum was stored at -20 ° C and the prolactin content was measured. Serum Prolactin was measured using the mouse PRL EIisa kit and the results showed that Compound 4 significantly reduced prolactin levels and weight gain in mice compared to paliperidone. The results are shown in Figures 1 and 2.
  • Active ingredient (any one of the present compounds 1 to 4) lOOmg
  • the original auxiliary material has been sieved for 80 mesh, and the prescribed amount of active ingredient, microcrystalline cellulose, lactose, povidone K30 is weighed, added to the high-speed mixing machine, mixed at low speed, uniformly mixed, added with appropriate amount of purified water, low-speed stirring, high-speed cutting Granulation, wet granules were dried at 60 ° C for 3 h, and sieved through a 24-mesh sieve. The prescribed amount of sodium carboxymethyl starch, silica and magnesium stearate were added, and the mixture was mixed and compressed by a rotary tableting machine.
  • Example 16 Capsule (230 mg)
  • Active ingredient (any one of the present compounds 1 to 4) lOOmg
  • the original auxiliary material has been sieved for 80 mesh, and the active ingredients, lactose, starch and povidone K30 are weighed into the high-speed mixing machine.
  • the mixture is mixed at low speed, and the appropriate amount of purified water is added.
  • the mixture is stirred at a low speed and cut at a high speed.
  • the wet granules were dried at 60 ° C for 3 h, and sieved through a 24-mesh sieve.
  • the prescribed amount of silica and magnesium stearate were added, and the mixture was mixed, and the capsule filling machine filled the capsules.

Landscapes

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

Abstract

本发明属于药物化学领域,公开了帕利哌酮氨基酸类衍生物及其应用。经实验发现,该类化合物可应用于制备治疗神经精神类疾病药物方面。

Description

帕利哌酮氨基酸类衍生物及其应用 技术领域
本发明属于药物化学领域, 具体涉及帕利哌酮氨基酸类衍生物及其应用。 背景技术
精神分裂症是所有精神疾病中最严重,危害最大的一种疾病,全球发病率约 为 1-2%。精神分裂症患者终生患病率为 0.7-0.8%, 与性别, 种族, 或社会界限没 有明显相关性, 同时死亡率比一般人群高出 2-3倍。 最新研究显示, 精神疾病的 社会负担在中国疾病中排名居首,超过了心脑血管、呼吸系统及恶性肿瘤等疾患。
现有精神分裂药物主要有两大类: 典型抗精神分裂药物和非典型抗精神分裂药 物。 典型抗精神分裂药物 (如氯丙嗪和氟哌啶醇) 阻断多巴胺 D2受体, 对精神分 裂症阳性症状具有良好疗效。 但由于强烈阻断多巴胺受体, 导致了锥体外系反应 ( EPS),迟发性运动障碍以及泌乳素增加等不良反应], 而且对精神分裂症阴性症状 无效。
非典型抗精神分裂药物, 是以氯氮平和利培酮为代表, 不仅对多巴胺 (D2 ) 受体有较强作用, 同时对 5-羟色胺 (5-HT2A) 受体也有较强作用。 与典型抗精神 分裂药物相比这类药物有很大的优势:对精神分裂症阳性症状有良好疗效;锥体 外系反应和迟发性运动障碍等副作用显著降低;部分非典型抗精神分裂药物对阴 性症状和认知障碍有一定改善作用。然而, 目前临床应用的非典型抗精神分裂药 物都有不同程度的 QT间期延长和高泌乳素等不良反应。 因此, 寻找新的既能有 效地治愈精神分裂症而且副作用小的药物是非常重要。
Y-氨基丁酸 (GABA) 是脑内最主要的抑制性神经递质, 由脑内最主要的兴奋 性神经递质谷氨酸在谷氨酸脱羧酶的催化下脱羧而成, 生成的 GABA与突触后膜 的 GABA受体结合, 发挥生物学效应。 1972年, Robert首先提出精神分裂症存在 Y-氨基丁酸系统的缺损。 近年来的研究表明, 精神分裂症患者前额皮质和海马中 GABA能神经传递异常。 研究认为, 精神分裂症的多巴胺能神经元通路上, GABA 的抑制作用减弱, 可造成抑制性神经冲动不足, 使多巴胺功能亢进, 从而引起行 为异常。 而且, 直接用低剂量 GABA激动剂会抑制僵住症的行为, 高剂量会加剧 氟哌啶醇诱导的僵住症。还有研究表明 GABA激动剂能抗惊厥和改善认知的作用。 这些研究结果可以看出 Y-氨基丁酸系统与精神分裂症间接相关。 因为 GABA 激动剂有亲水性基团 (如羧酸和氨基), 不容易通过血脑屏障, 使用受到限制。 然而, 发现通过化学方法与脂肪氨基酸或缩氨酸结合, 能使这些化合物大量通过 血脑屏障。 改善药物的副作用, 扩大治疗范围。 如在二期临床药物 BL-1020 (奋 乃静和 GABA的结合化合物, WO03026563和 J. Med, Chem. 2008, 51, 2858-2862 ) 与原药奋乃静相比, 不仅能降低 EPS的发生率, 而且因为 GABA通过血脑屏障, 起到改善认知障碍的作用。
帕利哌酮作为非典型抗精神分裂药物, 对 5 丁2受体具有高度亲和力, 且大 于对脑中多巴胺 01和多巴胺 02受体的亲和力。 临床试验显示, 帕利哌酮对治疗 精神分裂症的阳性和阴性症状均有效。 但是, 同时帕利哌酮对组织胺受体 ^和 肾上腺素能(^受体同样有高亲和力, 所以在应用中易导致体重增加、 高泌乳素 和体位性低血压的副作用。 同时在治疗精神分裂的长期服药过程中,研究表明体 重增加的副作用与组胺 ^受体密切相关。
因此, 仍然需要有一种以改善治疗活性和减少副作用为特征的精神分裂症药 物。 发明内容
本发明的目的是在现有技术的基础上,提供一种新的具有活性的帕利哌酮氨 基酸类衍生物。
本发明的另一目的是提供一种上述帕利哌酮氨基酸类衍生物在制备治疗神 经精神类疾病药物方面的应用。
本发明的目的可以通过以下技术方案实现:
一方面, 本发明涉及一种通式 (I)的化合物或其药学上可接受的盐:
Figure imgf000005_0001
(I)
其中:
R^PR2分别独立地选自氢、 取代或未取代的 垸基; R3为氢, 或者 与[^ 相连, 构成取代或未取代的 C46环垸基。
其中, 所述的!^优选氢或甲基。
所述的未取代的 垸基优选自甲基、 乙基、 丙基、 丁基、 戊基或异戊基, 取代的 5垸基优选自 d— 5卤代垸基或 d— 5羟垸基。
所述的未取代的 C46环垸基优选自环丁基、 环戊基或环己基, 进一步优选环 戊基或环己基。
所述的!^进一步优选氢, 甲基, 异丁基, 异戊基, 或者 与[^相连, 构成环 戊基、 环己基或环丁基, 更进一步优选 与[^相连, 构成环戊基或环己基。
本发明所述的通式 (I ) 的化合物或其药学上可接受的盐最优选自以下任意 一种化合物或药学上可接受的盐:
( 1) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -4-氨基丁酸酯;
( 2) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -3- (胺甲基) -5-甲基己酸酯;
( 3 ) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H-吡啶并 [l,2-a]9-嘧啶基 -4-氨基 -2-甲基丁酸酯;
(4) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -2- ( 1- (胺甲基) 环己基) -乙酸酯。
本发明化合物的药学上可接受的盐包括但不限于: 盐酸盐、氢溴酸盐、氢碘 酸盐、 硝酸盐、 硫酸盐、 硫酸氢盐、 磷酸盐、 酸式磷酸盐、 乙酸盐、 乳酸盐、 柠 檬酸盐、 酒石酸盐、 马来酸盐、 富马酸盐、 甲磺酸盐、 葡糖酸盐、 糖二酸盐、 苯 甲酸盐、 乙磺酸盐、 苯磺酸盐或对甲苯磺酸盐。 优选地, 本发明化合物的药学上 可接受的盐是盐酸盐。
另一方面,本发明涉及一种药物组合物,包含治疗有效量的本发明的化合物 和 /或其药学上可接受的盐, 以及药物学可接受的载体和 /或赋形剂。
再一方面, 本发明涉及本发明的化合物和 /或其药学上可接受的盐或本发明 所述的以式 (I ) 所示化合物和 /或其药学上可接受的盐为主要有效成分的组合物 在制备用于预防或治疗神经精神类疾病的药物中的应用。
又一方面,本发明涉及治疗预防或治疗神经精神类疾病的方法,其包括对患 有神经精神类疾病的对象施用治疗有效量的本发明的化合物和 /或其药学上可接 受的盐。
除非另有指明, 否则在整个此说明书中以下术语将具有以下含义。
本文使用的术语" 垸基' '是指具有 1、 2、 3、 4或 5个碳原子的直链或支链 的、 饱和的一价烃基。 垸基的实例包括甲基、 乙基、 丙基、 丁基、 戊基、 异 丙基、 异丁基、 仲丁基、 叔丁基、 异戊基、 2-甲基丁基、 1-甲基丁基、 1-乙基丙 基、 1,2-二甲基丙基、新戊基或 1,1-二甲基丙基。优选地, d— 5垸基为甲基、 乙基、 丙基、 丁基、 戊基或异戊基。
进一步地, 本发明通式 (I)的化合物中的 5垸基可选地被一个或多个选自卤 素和羟基的基团相同或不同地取代, 例如 5卤代垸基和 d— 5羟垸基。 优选地, 卤代垸基是三氟甲基, 且 羟垸基是羟甲基。
进一步地, 本发明通式 (I)的化合物中的 C46环垸基为环戊基、 环己基或环丁 基
本文使用的术语"药学上可接受的盐' '指本发明化合物的相对无毒的、无机酸 或有机酸加成盐。例如,参见 S.M. Berge等人, "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19。
本类化合物的通用合成方法是先合成氨基丁酸的 (BOC)20保护, 然后与帕利 哌酮的羟基相连, 最后脱掉 BOC保护得到产物。 本发明的化合物或其药学上可接受的盐可在全身和 /或局部起作用。根据需 要,本发明的化合物或其药学上可接受的盐可通过适合的方法施用,其包括但不 限于口服、 注射、 胃肠外施用等。
根据不用施用途径,可将本发明的化合物或其药学上可接受的盐与药学上可 接受的载体和 /或赋形剂一起配制为所需的施用形式, 包括但不限于片剂、 散剂、 胶囊剂、 溶液、 混悬液、 栓剂、 贴剂、 颗粒剂、 膏剂、 洗液等。 这可以通过现有 技术的方法完成。例如,可以通过将本发明的化合物或其药学上可接受的盐与药 学上适合的辅料混合来完成。可用于本发明的药学上适合的辅料的实例包括但不 限于溶剂、 乳化剂、 分散剂、 润湿剂、 粘结剂、 稳定剂、 着色剂和气味和 /或味 道掩蔽剂。
本发明的化合物或其药学上可接受的盐还可与其它已知的治疗神经精神类 疾病的药物组合使用。 本领域已知的治疗神经精神类疾病的药物包括例如利培 酮、 阿立哌唑、 氨磺必利、 氟西汀、 阿普唑仑、 咪达唑仑、 西酞普兰,、 地西泮 等。 因此, 另一方面, 本发明还涉及一种药物组合物, 其包括治疗有效量的本发 明的化合物和 /或其药学上可接受的盐、 和可选地其它已知的治疗神经精神类疾 病的药物、 以及药学上适合的辅料。
在一个实施方式中,本发明的药物组合物包括治疗有效量的本发明的化合物 和 /或其药学上可接受的盐以及药学上适合的辅料。
本发明的药物组合物每单位剂量可包含约 0.01到 1000 mg, 优选 1.0到 300 mg, 更优选 10到 150 mg, 最优选 lOOmg的本发明的化合物和 /或其药学上可接 受的盐。或者, 基于本发明的药物组合物的总重量, 本发明的药物组合物应包含 至少 0.5wt%, 优选 4 wt%至 70 wt%, 更优选 10wt%至 IJ 50 wt%, 最优选 30 wt%的 本发明的化合物和 /或其药学上可接受的盐。
本发明药物组合物中所含的本发明的化合物或其药学上可接受的盐的剂量 取决于疾病或病症的类型和严重性, 以及对象的特征, 例如一般健康、 年龄、 性 别、体重和药物耐受性。本领域技术人员能够根据这些或其它因素来确定适当的 本发明的活性化合物剂量。通常所用的中枢神经系统药物的有效剂量是技术人员 熟知的, 其每日总剂量通常在约 0.05mg到 2000mg之间。
本发明的又一方面是提供一种本发明的化合物和 /或其药学上可接受的盐在 制备用于预防或治疗神经精神类疾病的药物中的应用。
本文使用的术语 "治疗"包括克服、 缓解、 减轻、 解除或改善疾病或病症。 在某些情况下, 术语 "治疗"也包括 "预防"。
本文使用的术语 "神经精神类疾病"是指神经类疾病与精神类疾病的总称。 示例性的神经精神类疾病包括但不限于精神障碍、 焦虑症、 人格障碍、 抑郁症。 优选地, 本发明中的神经精神类疾病是精神分裂症。
本发明的再一方面是提供一种使用有效量的本发明的化合物和 /或其药学上 可接受的盐来预防或治疗神经精神类疾病的方法,其包括对患有神经精神类疾病 的对象施用治疗有效量的本发明的化合物或其药学上可接受的盐。
本文使用的术语 "对象"包括哺乳动物, 优选人。
有益效果:
体外受体结合试验表明, 本发明所涉及的衍生物对三种受体的亲和力 (D2, 5-HT1A,和 5-HT2A) 与帕利哌酮相当, 而对 H^Pai的亲和力低, 产生体重增加的副 作用可能性较小。
动物试验结果显示, 这类化合物既能明显改善 MK-801诱导的高活动性, 又 能有效改善阿扑吗啡诱导的攀爬症状, 并且在有效剂量下不引起 EPS。 由于这些 体外作用靶点和体内药理模型与多巴胺功能紊乱导致的神经系统疾病,特别是精 神分裂症密切相关, 因此本发明涉及的化合物具有治疗神经精神类疾病的作用, 尤其对精神分裂症有治疗作用。
因此, 将帕利哌酮与氨基酸相连形成酯类前药, 具有潜在的与帕利哌酮类似 的抗精神分裂症活性, 且能降低帕利哌酮体重增加、泌乳素增高或体位性低血压 等副作用, 同时能改善认知障碍的症状。 同时, 帕利哌酮氨基酸衍生物与帕利哌 酮相比, 具有低的急性毒性和高的治疗指数。 附图说明
图 1、 化合物 4, 帕利哌酮在小鼠 (每组 10只) 中连续给药 28天的体重变化情 况, 其中 P < 0.05表示差异有统计学意义。
图 2、 化合物 4, 帕利哌酮在小鼠 (每组 10只) 中连续给药 28天的泌乳素的变 化情况, 其中 P < 0.05表示差异有统计学意义 具体实施方式
下面的实施例只是以说明为目的而不作为本发明的限制
A、 合成方面的实施例
Figure imgf000009_0001
实施例 1、 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -4-氨基丁酸酯盐酸盐
( 1)取 4-氨基丁酸(10.3g, 100 mmol ), 加入到反应瓶中, 加入 50ml水, 以及 100ml丙酮, 加入三乙胺(110 mmol ), 在室温下搅拌至全部溶解, 分批加入 Boc 酸酐 (26.2g, 120 mmol), 在室温下反应 4h。 停止反应, 减压蒸去丙酮, 用乙醚 50mlx2萃取,弃掉有几层,将水层用 10%稀盐酸调 PH至 4-5,用乙酸乙酯 200mlx3 萃取, 合并有几层, 用无水硫酸镁干燥, 过滤, 将滤液旋干得无色油状物 18.5g, 收率 91.1%。 MS (ESI) m/z 204.2 ([M+H]+).
( 2) 取第一步产物 2.0g ( 10 mmol ), 加入 50ml二氯甲垸, 在室温下加入帕利 哌酮 3.4g ( 9 mmol ), 加入 4-二甲氨基吡啶 (DMAP) 0.3g, 分批加入 1,3-二环 己基碳二亚胺 0:2.^ ( 11 010101 ),在室温下搅拌反应 24h,停止搅拌,放置 4h, 过滤, 将滤液旋干, 加入 50ml乙酸乙酯, 静置 lh, 过滤, 滤液用 10%柠檬酸洗 涤 2次, 再用饱和碳酸氢钠洗涤, 有几层用无水硫酸镁干燥, 过滤, 旋干溶剂得 淡黄色油状物 4.9g, 收率 86.3% MS (ESI) m/z 612..3 ([M+H]+).
( 3 )取第二步产物 2g, 加入 30ml乙酸乙酯溶解, 加入 10ml饱和乙酸乙酯盐酸 气, 在室温下搅拌反应 4h, 有白色固体析出, 停止反应, 旋干溶剂, 加入乙醚, 过滤得白色固体 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四 氢 -4H-吡啶并 [l,2-a]9-嘧啶基 -4-氨基丁酸酯盐酸盐 1.8g, 收率 88.6%, 结构式见 表 1,熔点: 166-167 V。 XH NMR (DMSO) δ 11,19 (s, IH), 8.20-8.25 (m, IH), 7.71-7.76 (m, IH), 7.32-7.35 (m, IH), 5.72 (t, J = 8 Hz, 2H), 3.91-3.95 (m, IH), 3.72-3.75 (m, 3H), 3.45-3.49 (m, 4H), 3.14-3.18 (m, 4H), 2.95-3.01 (m, IH), 2.51-2.58 (m, 4H), 2.38-2.41 (m, 2H), 2.31 (s, 3H), 2.23-2.27 (m, 3H), 1.89-1.99 (m, 5H) 0 MS (ESI) m/z 512.2 ([M+H]+). 实施例 2、 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -3- (胺甲基) -5-甲基-己酸酯盐酸盐
用 3-氨甲基 -5-甲基己酸代替 4-氨基丁酸, 按实施例 1的方法制备目标化合物 2。
目标化合物 2结构式见表 1,熔点: 185-187 "C o ^ N MR (DMSO) δ 11.15 (s, IH), 8.19-8.23 (m, IH), 7,69-7,73 (m, IH), 7.30-7.35 (m, IH), 5.68 (t, J = 8 Hz, 2H), 3.95-3.98 (m, IH), 3.71-3.76 (m, 3H), 3.45-3.51 (m, 4H), 3.11-3.18 (m, 4H), 2.92-2.96 (m, IH), 2.51-2.56 (m,4H), 2.51 (s, 2H), 2.38-2.40 (m, 2H), 2.31 (s, 3H), 2.23-2.27 (m, 2H), 1.72-1.99 (m, 7H), 1.02 (d, J = 6.6 Hz, 6H)。 MS (ESI) m/z 568.3 ([M+H]+), 实施例 3、 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -4-氨基 -2-甲基丁酸酯 盐酸盐
用 4-氨基 -2-甲基丁酸代替 4-氨基丁酸, 按实施例 1的方法制备目标化合物 3。
目标化合物 3结构式见表 1,熔点: 188-189 Η NMR (DMSO) δ 11.20 (s, IH), 8.17-8.20 (m, IH), 7.70-7.76 (m, IH), 7.29-7.33 (m, IH), 5.70 (t, J = 8 Hz, 2H), 3.89-3.92 (m, IH), 3.72-3.77 (m, 3H), 3.45-3.51 (m, 4H), 3.14-3.18 (m, 4H), 2.95-2.99 (m, IH), 2.51-2.57 (m, 4H), 2.38-2.41 (m, 2H), 2.99 (s, 3H), 2.23-2.27 (m, 3H), 1.92-1.99 (m, 5H), 1.43 (d, J = 6 Hz, 2H)。 MS (ESI) m/z 526.3 ( [M+H]+), 实施例 4、 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -2- ( 1- (胺甲基) 环己基) -乙酸酯 盐酸盐
用 1- (氨甲基)环己垸乙酸代替 4-氨基丁酸, 按实施例 1的方法制备目标化合物
4。
目标化合物 2结构式见表 4,熔点: 196-198 V ^H N MR (DMSO) δ 11.15 (s, IH), 8.21-8.25 (m, IH), 7.73-7.78 (m, IH), 7.33-7.38 (m, IH), 5.75 (t, J = 8 Hz, 2H), 3.90-3.95 (m, IH), 3.72-3.77 (m, 3H), 3.45-3.51 (m, 4H), 3.14-3.22 (m, 4H), 2.95-2.99 (m, IH), 2.56-2.58 (m, 2H), 2.51 (s, 2H), 2.38-2.41 (m, 2H), 2.31 (s, 3H), 2.23-2.27 (m, 2H), 1.92-1.99 (m, 3H), 1.38-1.52 (m, 10H)。 MS (ESI) m/z 580.3 ([M+H]+), 表 1 实施例制备的优选化合物编号及其结构式
Figure imgf000011_0001
B、 药理方面的实施例
实施例 5
5 "^膜的制备
大鼠断头, 冰上操作, 迅速取脑皮层, 加入 3ml匀浆液 (0.05M 的 Tris-HCI 缓冲液, 含 0.1%的抗坏血酸、 10um优降宁和 4mMCaCI2), 匀浆, 然后加入 5ml 缓冲液 (0.05M 的 Tris-HCI缓冲液, 含 0.1%的抗坏血酸、 10um优降宁和 4mM CaCI2), 于 37°C孵化 10min, 孵化完后试管用天平调整重量, 在 12000r, 4°C离 心 20min, 弃上清液, 加入 3ml B液, 用旋涡混合器混匀, 再加入 5ml 缓冲液, 离心, 重复三次离心, 离心完毕, 弃上清液, 将沉淀于 -80°C储存备用。
受体结合实验材料:
同位素配基 3H-8-OH-DPAT (67.0Ci/mmol), 购自 PerkinElmer公司; 5-HT, 购 自 RBI公司; GF/C玻璃纤维滤纸, 购自 Whatman公司; Tris进口分装 2,5-二苯基 噁唑 (PPO)、 1,4-双 (5-苯基 -2-恶唑基)苯 (POPOP)购自上海试剂一厂。 Beckman LS-6500型多功能液体闪烁计数仪。
实验方法:
(1) 取 5-^/^受体膜, 加入适量的匀浆液, 用匀浆机分散均匀, 呈 50ml 膜的混悬液, 备用。
(2) 各反应管分别加入膜制备物 100μΙ_, 缓冲液 100μί。
(3) 总结合管 (ΤΒ) 加入 100μΙ_缓冲液, 非特异性结合管 (ΝΒ) 加入 5-ΗΤ ΙΟΟμί (终浓度 10— 5Μ), 各受试化合物特异性结合管 (SB) 加入 100μΙ_受试化合 物 (终浓度 10— 5M);
(4) 各反应管分别加入放射性配体 3H-8-OH-DPAT ΙΟμί (各反应管均设 2个 平行管, 加样时各管置于冰上)。
(5) 将各反应管 37°C温孵 10 min, 反应完毕, 结合的配基通过减压快速过 滤, 用冰冷的试验缓冲液充分洗涤, 将滤片取出放到 3ml 闪烁杯中, 加入 2ml 的甲苯闪烁液 (PP02.5g, POPOP 0.05 g溶于 500 mL甲苯) 并混匀;
(6) 将闪烁瓶放入液闪计数仪计数
抑制率 (I %)=(总结合管 cpm—化合物 cpm)/ (总结合管 cpm一非特异结合管 cpm)xl00%
化合物每次实验做两复管, 进行两次单独实验。 实验结果见表 2 实施例 6
5-HT2/J 的制备
大鼠断头, 冰上操作, 迅速取脑皮层, 加入 3ml缓冲液 (0.05M 的 Tris-HCI 缓冲液: 取 6.05gTris溶于 1000ml双蒸水中, 用浓 HCI调 PH为 7.5 )于 4档 3-4 s 匀浆, 匀浆 4次, 然后加入 5ml缓冲液, 于 37°C孵化 10min, 孵化完后试管用天 平调整重量, 在 12000r, 4°C离心 20 min, 弃上清液, 加入 3ml缓冲液, 用旋涡 混合器混匀, 再加入 5ml缓冲液, 离心, (重复三次离心), 离心完毕, 弃上清液, 将沉淀于 -80°C储存备用。
受体结合实验材料:
同位素配基 [3H]- Ketanserin ( 67.0Ci/mmol ) , 购自 PerkinElmer 公司; Methysergide, 购自 RBI公司; GF/C玻璃纤维滤纸, 购自 Whatman公司; Tris进 口分装; PPO、 POPOP购自上海试剂一厂; 脂溶性闪烁液。 Beckman LS-6500型 多功能液体闪烁计数仪。
实验方法:
( 1 )先将制备好的 5-HT2A膜用适量的缓冲液, 用匀浆机分散均匀, 将 15只 试管混入到 100ml的容器中, 加入适量的缓冲液呈 50ml膜的混悬液, 备用。
( 2 ) 各反应管分别加入膜制备物 100μΙ_, 缓冲液 100μί。
( 3 ) 总结合管 (ΤΒ ) 加入 100μΙ_ 缓冲液, 非特异性结合管 (ΝΒ ) 加入 Methysergide 100μΙ_ (终浓度 10— 5Μ ),各受试化合物特异性结合管(SB)加入 100μΙ_ 受试化合物 (终浓度 10— 5M );
(4) 各反应管分别加入放射性配体 3H-Ketanserin ΙΟμί (各反应管均设 2个 平行管, 加样时各管置于冰上)。
( 5 ) 将各反应管 37°C温孵 15 min, 反应完毕, 结合的配基通过减压快速过 滤, 用冰冷的试验缓冲液充分洗涤, 将滤片取出放到 3ml 闪烁杯中, 加入 2ml 的甲苯闪烁液并混匀;
( 6 ) 将闪烁瓶放入液闪计数仪计数
抑制率 (I %)=(总结合管 cpm—化合物 cpm)/ (总结合管 cpm一非特异结合管 cpm)xl00%
化合物每次实验做两复管, 进行两次单独实验。 实验结果见表 2 实施例 7
02膜的制备
大鼠断头, 冰上操作, 迅速取脑纹状体, 加入 3ml缓冲液(0.05M的 Tris-HCI 缓冲液,含 NaCI 120mM、 KCI 5mM、 MgCI2 lmM、 CaCI2 ImM ),于 4档 3-4 s匀浆, 匀浆 4次, 然后加入 5ml缓冲液, 将匀浆完的试管用天平调整重量, 在 12000r, 4°C离心 20 min, 弃上清液, 加入 3mlB液, 用旋涡混合器混匀, 再加入 5ml缓 冲液, 离心, 重复三次离心, 离心完毕, 弃上清液, 将沉淀于 -80°C储存备用。 受体结合实验材料:
同位素配基 3H-Spiperone( 67.0Ci/mmol ),购自 PerkinElmer公司; Butaclamol, 购自 RBI公司; GF/C玻璃纤维滤纸, 购自 Whatman公司; Tris进口分装; PPO、 POPOP购自上海试剂一厂; 脂溶性闪烁液。 Beckman LS-6500型多功能液体闪烁 计数仪。
实验方法:
( 1 ) 先将制备好的 D2膜用适量的缓冲液, 用匀浆机分散均匀, 将 15只试 管混入到 100ml的容器中, 加入适量的缓冲液呈 50ml膜的混悬液, 备用。
( 2 ) 各反应管分别加入膜制备物 100μΙ_, 缓冲液 100μί。
( 3 )总结合管 (ΤΒ)加入 100μΙ_缓冲液, 非特异性结合管 ( ΝΒ )加入 100μΙ_ Butaclamol (终浓度 10— 5M ), 各受试化合物特异性结合管 (SB )加入 100μΙ_受试 化合物 (终浓度 10— 5M );
(4) 各反应管分别加入放射性配体 ^-Spiperone ΙΟμί (各反应管均设 2个 平行管, 加样时各管置于冰上)。
( 5 ) 将各反应管 37°C温孵 20 min, 反应完毕, 结合的配基通过减压快速过 滤, 用冰冷的试验缓冲液充分洗涤, 将滤片取出放到 3ml 闪烁杯中, 加入 2ml 的甲苯闪烁液并混匀;
( 6 ) 将闪烁瓶放入液闪计数仪计数
抑制率 (I %)=(总结合管 cpm—化合物 cpm)/ (总结合管 cpm一非特异结合管 cpm)xl00%
化合物每次实验做两复管, 进行两次单独实验。 实验结果见表 2。 实施例 8
αι去甲肾上腺素受体膜的制备
大鼠断头,冰上操作,迅速取脑皮层,加入 0.05MTris-HCl缓冲液(PH7.7) 用旋涡混合器混匀, 在 48000g, 4°C离心 15 min, 弃上清液, 取沉淀, 再加 入 0.05M的 Tris-HCI缓冲液 (PH7.7) 洗涤, 重复三次离心, 离心完毕, 弃 上清液, 将沉淀于 -80°C储存备用。
实验方法:
第一步: 先将制备好的膜用适量的缓冲液 0.05MTris-HCI ( PH7.7), 用匀浆机 分散均匀, 将 15只试管混入到 100ml的容器中, 加入适量的匀浆液呈
50ml膜的混悬液, 备用。
第二步: 各反应管分别加入膜制备物 100μΙ_, 缓冲液 100μί。
第三步: 总结合管 (ΤΒ ) 加入 100μΙ_匀浆液, 非特异性结合管 (ΝΒ) 加入 prazosin ΙΟΟμί (终浓度 10- 5Μ ), 各受试化合物特异性结合管 (SB ) 加 入 100μΙ_受试化合物 (终浓度 10— 5M );
第四步: 各反应管分别加入放射性配体 3H-praZ0Sin ΙΟμί (各反应管均设 2个 平行管, 加样时各管置于冰上)。
第五步: 将各反应管 25°C温孵 60 min, 反应完毕, 结合的配基通过减压快速 过滤, 用冰冷的试验缓冲液充分洗涤, 将滤片取出放到 3ml闪烁杯中, 加入 2ml的甲苯闪烁液并混匀;
第六步: 将闪烁瓶放入液闪计数仪计数
抑制率 (I %)=(总结合管 cpm—化合物 cpm)/ (总结合管 cpm一非特异结合管 cpm)xl00%
化合物每次实验做两复管, 进行两次单独实验。 实验结果见表 2。 实施例 9
组胺 ^受体膜的制备
大鼠断头, 冰上操作, 迅速取滕鼠小脑, 加入缓冲液 (0.05M 的 Tris-HCI缓 冲液, 含 5nM EDTA, PH7.7 ), 用旋涡混合器混匀, 在 48000g, 4°C离心 10 min, 弃上清液,取沉淀,再加入缓冲液(0.05M的 Tris-HCI缓冲液,含 5nM EDTA, PH7.7) 洗涤, 重复三次离心, 离心完毕, 弃上清液, 将沉淀于 -80°C储存备用。
实验方法:
第一步:先将制备好的膜用适量的缓冲液(0.05M的 Tris-HCI缓冲液,含 5nM EDTA, PH7.7)。, 用匀浆机分散均匀, 将 15只试管混入到 100ml的容 器中, 加入适量的缓冲液呈 50ml膜的混悬液, 备用。
第二步: 各反应管分别加入膜制备物 100μί。
第三步: 总结合管 (ΤΒ ) 加入 100μΙ_缓冲液, 非特异性结合管 (ΝΒ) 加入 ΙΟΟμί promethazine (终浓度 10— 5M ),各受试化合物特异性结合管(SB) 加入 100μΙ_受试化合物 (终浓度 10— 5M );
第四步: 各反应管分别加入放射性配体 3H-Pyrilamine l(^L (各反应管均设 2 个平行管, 加样时各管置于冰上)。
第五步: 将各反应管 30°C温孵 60 min, 反应完毕, 结合的配基通过减压快速 过滤, 用冰冷的试验缓冲液充分洗涤, 将滤片取出放到 3ml闪烁杯中, 加入 2ml的甲苯闪烁液并混匀;
第六步: 将闪烁瓶放入液闪计数仪计数
抑制率 (I %)=(总结合管 cpm—化合物 cpm)/ (总结合管 cpm一非特异结合管 cpm)xl00%
化合物每次实验做两复管, 进行两次单独实验。 实验结果见表 2。
表 2 化合物对各受体的亲和力
Figure imgf000016_0001
体外实验结果表明化合物 4对三种受体(D2, 5-HT1A,和 5-HT2A)与帕利哌酮相 当, 而对 ^和31的亲和力低, 产生体重增加的副作用可能性较小。 实施例 10、 MK-801诱导的高活动性化合物体内抗精神分裂活性 实验动物及试剂
健康昆明种小鼠,雌雄各半,体重 (20±2)g, 由南京青龙山动物养殖中心提供。 抗坏血酸, 国药集团化学试剂有限公司;
MK-801,由美国 Sigma公司生产,配制方法:用 0.1%的维生素 C配成 lmg/ml 的溶液;
受试阳性药物: 氟哌啶醇、 氯氮平、利培酮、 奥氮平、 阿立哌唑、 齐拉西酮、 奎硫平;
吐温 80, 浓度 10%。
实验方法
选择体重合格的小鼠, 随机分为空白组、 模型组、 阳性对照组 (利培酮组)、 药物组。 空白组、 模型组灌胃 10%吐温 0.1ml/10g, 阳性对照组灌胃给利培酮 O.lmg/kg, 药物组分别灌胃给与相应剂量药物。给药后 lh 空白组腹腔注射 0.1% 抗坏血酸 0.1ml/10g, 模型组、 阳性对照组 (30min)、 药物组腹腔注射 MK-801溶 液 0.1mg/kg。 其后测定各组小鼠 90分钟内自发活动。 结果见表 3。 实施例 11、 阿扑吗啡诱导小鼠攀爬实验
实验动物
健康 KM小鼠, 雄性, 体重 18〜22g, 由南京青龙山动物养殖中心提供。
主要试剂
奎硫平;
阿扑吗啡, Sigma公司提供, 临用前 0.9 % NaCI (含 0.1% 维生素 C) 溶解, 现配现用;
维生素 C, F20061113, 国药集团化学试剂有限公司;
氯化钠注射液, H32026305, 徐州市第五制药厂有限公司。
仪器洎制攀爬笼, 秒表。
实验方法:阿扑吗啡诱导小鼠攀爬实验
KM小鼠, 雄性, 体重 18〜22g, 随机分为阴性对照组、 模型组、 奎硫平阳性 药物各剂量组以及化合物各剂量组 (具体给药剂量见下表), 每组 10只。 阴性对 照组和模型组灌胃给予相应溶剂双蒸水, 阳性药物组灌胃给予相应阳性药物(溶 解时先加微量乙酸, 再加双蒸水), 化合物各剂量组灌胃给予相应剂量化合物, 灌胃体积为 0.1ml/10g。 灌胃给药 1小时后皮下注射阿扑吗啡 (lmg/kg), 体积为 0.1ml/10g。 注射阿扑吗啡后, 立即放入攀爬笼中, 适应 5分钟, 观察注射阿扑吗 啡后第 10-11, 20-21, 30-31分钟的行为并进行评分, 评分标准: 四足在地板上得 分为 0; 两前足在网笼上得分为 1; 四只足在网笼上得分为 2。 结果见表 3。 实施例 12、 僵住症实验方法
实验动物
健康昆明种小鼠, 雌雄各半, (22±2)g, 由南京青龙山动物养殖中心提供。 主要试剂:
受试药、 帕利哌酮
仪器:
自制抓棒器材: 小鼠盒内放置直径 0.3 cm,高于工作台 5 cm的不锈钢棒。 实验方法:
KM小鼠, 雌雄各半, 体重 20〜24g, 随机分为阴性对照组、 模型组、 奎硫平 各剂量组以及化合物各剂量组,每组 10只。阴性对照组和模型组灌胃给予相应溶 剂双蒸水, 阳性药物组灌胃给予相应阳性药物(溶解时先加微量乙酸, 再加双蒸 水), 化合物各剂量组灌胃给予相应剂量化合物, 灌胃体积为 0.1ml/10g。 灌胃给 药 30min、 60min 90min时, 将小鼠两只前爪轻柔地放在长 20cm,直径 0.3cm,高于 工作台 5.5cm的小棒上,再将动物后肢轻放于盒底面,记录小鼠两只前爪在棒上保 持姿势的持续时间, 以 30s僵直不动为阳性反应。 如果小鼠前爪一直没有放下 ,60 s时终止观察。 统计每个化合物剂量组阳性反应动物数。 结果见表 3。
表 3.优选化合物体内动物模型试验结果
Figure imgf000019_0001
本实验结果表明:与模型组相比,帕利哌酮和化合物 4既能明显改善 MK-801 诱导的高活动性, 又能有效的改善阿扑吗啡诱导的攀爬症状, 并且在有效剂量下 不引起 EPS, 而且化合物 4的治疗指数明显高于帕利哌酮。 实施例 13、 急性毒性研究
序贯法之限度实验 取 KM小鼠, 雌雄各半, 随机分为若干组, 每组 2-5只, 分别为各化合物 2000mg/kg组和溶剂组, 按 0.2ml/10g灌胃给药。 观察动物 3日 内的死亡情况。 (如果动物在三日内有 3只或 3只以上存活, 生命状态无明显异 常时, 继续观察, 直至 7日后实验结束。如果动物在三日内死亡 3只或 3只以上 时, 采用半数致死量法测定其 LD50。)
半数致死量法预试验 取 KM小鼠, 雌雄各半, 随机分若干组, 每组 4只, 分别为各化合物 1500mg/kg、 1000mg/kg、 500mg/kg组和溶剂组, 按 0.2ml/10g 灌胃给药, 观察动物 1-3日内的死亡情况。
结果:小鼠单次灌服的 LD5。大于 2000mg/kg,远远高于帕利哌酮(800mg/kg), 具有较小的急性毒性。 实施例 14、 小鼠肥胖及泌乳素的研究
雄性 ICR小鼠 100只, 随机分为阴性对照组、 化合物 4低、 中、 高三个剂量 组, 帕利哌酮低、 中、 高三个剂量组。 每天给药一次, 连续给药 28天, 每天监 测体重。给药结束后, 摘眼球取血, 血液室温静置 30min后, 用低温高速离心机 4°C, 3000r/min离心 10分钟。 采集血清储存在 -20°C中, 待测泌乳素含量。血清 泌乳素采用小鼠 PRL EIisa试剂盒测定, 结果显示化合物 4与帕利哌酮相比,可以 显著降低小鼠泌乳素水平和体重增加。 结果见图 1和图 2。
C 组合物实施例
实施例 15、 片剂
活性成分 (本发明化合物 1~4中的任意一种) lOOmg
微晶纤维素 50mg
乳糖 lOOmg
聚维酮 K30 9 mg
羧甲淀粉钠 12 mg
二氧化硅 2.5mg
硬脂酸镁 1.5mg
原辅料过 80目筛备用, 称取处方量活性成分、 微晶纤维素、 乳糖、 聚维酮 K30, 加入到高速混合制剂机中, 低速搅拌混合均匀, 加入适量纯化水, 低速搅 拌, 高速切割制粒, 湿颗粒 60°C干燥 3h, 24目筛整粒, 加入处方量羧甲淀粉钠、 二氧化硅和硬脂酸镁, 总混, 旋转压片机压片。 实施例 16、 胶囊剂 (230mg)
活性成分(本发明化合物 1~4中的任意一种) lOOmg
乳糖 80 mg
淀粉 40 mg
聚维酮 K30 7 mg
二氧化硅 2 mg
硬脂酸镁 1 mg
原辅料过 80 目筛备用, 称取处方量活性成分、 乳糖、 淀粉、 聚维酮 K30, 加入到高速混合制剂机中, 低速搅拌混合均匀, 加入适量纯化水, 低速搅拌, 高 速切割制粒, 湿颗粒 60°C干燥 3h, 24目筛整粒, 加入处方量二氧化硅和硬脂酸 镁, 总混, 胶囊灌装机填充胶囊。

Claims

m ^
1、 通式 (I)所示的化合物或其药学上可接受的盐:
Figure imgf000021_0001
(I)
其中:
R^P R2分别独立地选自氢、 取代或未取代的 垸基; R3为氢, 或者 与[^ 相连, 构成取代或未取代的 C46环垸基。
2、根据权利要求 1所述的化合物或其药学上可接受的盐, 其特征在于, 所述 的未取代的 垸基选自甲基、 乙基、 丙基、 丁基、 戊基或异戊基, 取代的 5 垸基选自 卤代垸基、 d— 5羟垸基。
3、根据权利要求 2所述的化合物或其药学上可接受的盐, 其特征在于: 所述 为氢或甲基。
4、根据权利要求 1所述的化合物或其药学上可接受的盐, 其特征在于: 所述 的未取代的 C46环垸基为环戊基或环己基;所述的取代的 C46环垸基为被一个或多 个选自 C1-3垸基或卤素的基团相同或不同地取代。
5、根据权利要求 1、 2或 4所述的化合物或其药学上可接受的盐, 其特征在 于: 所述 R2为氢, 甲基, 异丁基或异戊基; 或 R3与 R2相连, 构成环戊基、 环己 基或环丁基, 进一步优选 与[^相连, 构成环戊基或环己基。
6、 根据权利要求 1所述的化合物或其药学上可接受的盐, 其特征在于所述 的通式 (I)所示的化合物或其药学上可接受的盐选自以下任意一种化合物或其药 学上可接受的盐: ( 1) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -4-氨基丁酸酯;
( 2) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -3- (胺甲基) -5-甲基己酸酯;
( 3 ) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H-吡啶并 [l,2-a]9-嘧啶基 -4-氨基 -2-甲基丁酸酯;
(4) 3-(2-(4-(6-氟-苯并异恶唑) -3-哌啶基)-乙基) -2-甲基 -4-氧 -6,7,8,9-四氢 -4H- 吡啶并 [l,2-a]9-嘧啶基 -2- ( 1- (胺甲基) 环己基) -乙酸酯。
7、如权利要求 1所述的化合物或其药学上可接受的盐,其特征在于所述的药 学上可接受的盐选自: 盐酸盐、 氢溴酸盐、 氢碘酸盐、 硝酸盐、 硫酸盐或硫酸氢 盐、 磷酸盐或酸式磷酸盐、 乙酸盐、 乳酸盐、 柠檬酸盐、 酒石酸盐、 马来酸盐、 富马酸盐、 甲磺酸盐、 葡糖酸盐、 糖二酸盐、 苯甲酸盐、 乙磺酸盐、 苯磺酸盐或 对甲苯磺酸盐。
8、 一种药物组合物, 其特征在于包括治疗有效量的权利要求 1所述的化合 物和 /或其药学上可接受的盐。
9、权利要求 1所述的化合物或其药学上可接受的盐在制备用于预防或治疗神 经精神类疾病的药物中的应用。
10、如权利要求 9所述的应用,其特征在于所述的神经精神类疾病是精神分 裂症。
11、 权利要求 8所述的组合物在制备用于预防或治疗神经精神类疾病的药物 中的应用。
12、 如权利要求 11所述的应用, 其特征在于所述的神经精神类疾病是精神 分裂症。
PCT/CN2014/082111 2013-07-16 2014-07-14 帕利哌酮氨基酸类衍生物及其应用 WO2015007191A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201310296582.8 2013-07-16
CN201310296582.8A CN104292226B (zh) 2013-07-16 2013-07-16 帕利哌酮氨基酸类衍生物及其应用

Publications (1)

Publication Number Publication Date
WO2015007191A1 true WO2015007191A1 (zh) 2015-01-22

Family

ID=52312212

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2014/082111 WO2015007191A1 (zh) 2013-07-16 2014-07-14 帕利哌酮氨基酸类衍生物及其应用

Country Status (2)

Country Link
CN (1) CN104292226B (zh)
WO (1) WO2015007191A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115260190A (zh) * 2022-08-30 2022-11-01 南开大学 一种帕利哌酮前药、制剂及其制备方法和应用

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106749219A (zh) * 2015-11-20 2017-05-31 江苏恩华药业股份有限公司 一种内酰胺类衍生物及其应用
CN112704678B (zh) * 2020-12-02 2024-02-23 江苏恩华药业股份有限公司 一种丙酰胺衍生物在制备治疗精神分裂症药物中应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011073997A2 (en) * 2009-12-14 2011-06-23 Cadila Healthcare Limited Process for preparing paliperidone and pharmaceutically acceptable salts thereof
WO2012042368A1 (en) * 2010-09-30 2012-04-05 Aurobindo Pharma Limited Process for preparation of paliperidone
CA2837251A1 (en) * 2011-05-30 2012-12-06 Cipla Limited Process for the preparation of paliperidone
CN102993200A (zh) * 2011-09-10 2013-03-27 鲁翠涛 帕潘立酮氨基酸酯及其制备方法
US20140057298A1 (en) * 2012-08-21 2014-02-27 Janssen Pharmaceutica Nv Antibodies to Paliperidone and Use Thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110166156A1 (en) * 2010-01-07 2011-07-07 Alkermes, Inc. Prodrugs for the Treatment of Schizophrenia and Bipolar Disease

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011073997A2 (en) * 2009-12-14 2011-06-23 Cadila Healthcare Limited Process for preparing paliperidone and pharmaceutically acceptable salts thereof
WO2012042368A1 (en) * 2010-09-30 2012-04-05 Aurobindo Pharma Limited Process for preparation of paliperidone
CA2837251A1 (en) * 2011-05-30 2012-12-06 Cipla Limited Process for the preparation of paliperidone
CN102993200A (zh) * 2011-09-10 2013-03-27 鲁翠涛 帕潘立酮氨基酸酯及其制备方法
US20140057298A1 (en) * 2012-08-21 2014-02-27 Janssen Pharmaceutica Nv Antibodies to Paliperidone and Use Thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115260190A (zh) * 2022-08-30 2022-11-01 南开大学 一种帕利哌酮前药、制剂及其制备方法和应用

Also Published As

Publication number Publication date
CN104292226A (zh) 2015-01-21
CN104292226B (zh) 2016-12-28

Similar Documents

Publication Publication Date Title
TWI382984B (zh) 雜環化合物
TWI458726B (zh) 具有抑制一氧化氮合成酶活性的苯並噁嗪、苯並噻嗪及其相關化合物
US8569512B2 (en) P2X3 receptor antagonists for treatment of pain
EP2995617B1 (en) Benzoisothiazole compounds and use in preparation of antipsychotic drugs
CN101792417A (zh) 用于治疗5ht2c受体相关疾病的苯并氮杂卓衍生物
JP6437119B2 (ja) 選択的s1p1レセプターアゴニストを含む薬学的合剤
TW201306842A (zh) 使用pi3k/mtor吡啶並嘧啶酮抑制劑及苯達莫司汀及/或利妥昔單抗治療惡性血液疾病之組合療法
JP6977024B2 (ja) 5−ht2c受容体アゴニストおよび組成物ならびに使用方法
JP6789578B2 (ja) 5−ht2c受容体アゴニストおよび組成物、ならびに使用方法
JP2022070895A (ja) ニューロキニン-1受容体アンタゴニストとしての化合物およびその使用
WO2013120438A1 (zh) 治疗或缓解疼痛的物质
JP2008500295A (ja) 4−ヒドロキシ−4−メチル−ピペリジン−1−カルボン酸(4−メトキシ−7−モルホリン−4−イル−ベンゾチアゾール−2−イル)−アミド
CA2962569A1 (en) N-alkylaryl-5-oxyaryl-octahydro-cyclopenta[c]pyrrole negative allosteric modulators of nr2b
WO2008000142A1 (fr) Agoniste du transporteur de la dopamine et ses utilisations
WO2015007191A1 (zh) 帕利哌酮氨基酸类衍生物及其应用
WO2017045599A1 (zh) 环己烷衍生物或其立体异构体或盐及其制备与应用
WO2011133212A1 (en) Methods, compounds and pharmaceutical compositions for treating anxiety and mood disorders
JP2007262022A (ja) 新規2−チオフェンカルボキサミド誘導体
CN105384676B (zh) 三重再摄取抑制剂及其应用方法
TW201605856A (zh) 5-HTc受體促效劑
AU2006291059A1 (en) Salicylate and gentisate salts of a piperazine compound
RU2322443C2 (ru) Новое применение замещенных аминометилхроманов
WO2013017068A1 (zh) 取代的苯并吡喃酮类衍生物及其应用
CN109476584B (zh) N-(2-(取代-萘-1-基)乙基)取代酰胺类化合物、其制备及其用途
CN100427484C (zh) 2-甲氧基甲基-3-(3,4-二氯苯基)-8-氮杂双环[3.2.1]辛烷酒石酸盐

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: 14826960

Country of ref document: EP

Kind code of ref document: A1

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14826960

Country of ref document: EP

Kind code of ref document: A1