Compounds as potassium channel modulators, their preparation and use

文档序号:298124 发布日期:2021-11-26 浏览:19次 中文

阅读说明:本技术 作为钾通道调节剂的化合物及其制备和应用 (Compounds as potassium channel modulators, their preparation and use ) 是由 梁波 刘刚 陈焕明 于 2021-10-27 设计创作,主要内容包括:本发明涉及作为钾通道调节剂的化合物及其制备和应用。具体地,本发明化合物具有式A所示结构。本发明还公开了所述化合物的制备方法及其作为钾通道调节剂的用途。式A。(The invention relates to potassium asCompounds which are channel modulators, their preparation and use. Specifically, the compound has a structure shown in formula A. The invention also discloses a preparation method of the compound and application of the compound as a potassium channel regulator.)

1. A compound of formula A or a pharmaceutically acceptable salt thereof,

formula A.

2. A process for preparing a compound of claim 1, or a pharmaceutically acceptable salt thereof, comprising the steps of:

1)andreacting to obtain

2)Andand reacting to obtain the compound of the formula A.

3. Use of a compound of claim 1, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for the treatment or prevention of a disease, disorder or condition selected from the group consisting of: epilepsy, convulsion, inflammatory pain, neuropathic pain, migraine, depression, stroke, neurodegenerative disease.

4. The use of claim 3, wherein the disease, disorder or condition is selected from the group consisting of: epilepsy and convulsion.

5. The use of claim 3, wherein the disease, disorder or condition is selected from the group consisting of: inflammatory pain, neuropathic pain, migraine.

6. The use of claim 3, wherein the disease, disorder or condition is depression.

7. The use of claim 3, wherein the disease, disorder, or condition is stroke.

8. The use of claim 3, wherein the disease, disorder or condition is a neurodegenerative disease.

9. A pharmaceutical composition comprising one or more pharmaceutically acceptable carriers and a therapeutically effective amount of one or more compounds of claim 1 or pharmaceutically acceptable salts thereof.

Technical Field

The invention relates to the field of biomedicine, in particular to a compound serving as a potassium channel regulator and preparation and application thereof.

Background

Kv7 potassium channel is a voltage-dependent potassium ion channel and has the characteristics of low threshold activation, slow activation and non-inactivation. The Kv7 potassium channel has five family members (Kv7.1-Kv7.5), and all Kv7 potassium channel members have similar topologies, i.e., a functional channel is composed of four subunits, each of which contains six transmembrane segments (S1-S6). Wherein S4 is a voltage sensing region and has important functions in sensing membrane potential change, controlling conformational change and the like; SS-S6 is the major component of the pore region of the channel and is the major component and region of action of potassium channel openers. KV7.1 potassium channels are a non-neuronal pathway, distributed in peripheral tissues and expressed in the heart to mediate myocardial Iks, mutations of which can lead to Long Q-T syndrome. Kv7.2-Kv7.5 potassium channels are the basis of M current in neurons, are widely distributed in the nervous system, and have various physiological activities. Mutations in the Kv7.2 and Kv7.3 potassium channel genes can lead to a variety of different epileptic phenotypes, such as Benign Familial Neonatal Convulsions (BFNC), which fully explain the role of M current in regulating neuronal excitability. Kv7.4 potassium channels are highly expressed in the outer hair cells of the cochlea and brainstem auditory nucleus, and mutations thereof may lead to hereditary deafness. Kv7.5 potassium channels are highly expressed in skeletal muscle and brain, and mutations thereof may lead to retinopathy. Many diseases such as epilepsy, anxiety, deafness, etc. have a common feature of membrane hyperexcitability, and the Kv7 potassium channel, which is a molecular basis for M current, can be opened by sensing a change in membrane potential to up-regulate inhibitory potassium current, thereby controlling membrane excitability, so that the Kv7 potassium channel has a great significance in pain and mental diseases represented by nerve hyperexcitability.

Retigabine (Retigabine) is a drug for the treatment of epilepsy and is currently approved for the market in the uk, germany, danish. Research has confirmed that retigabine's action is related to voltage-gated potassium ion channels (KCNQs), of which the main mechanism of action is acting on the KCNQ2/3 channel to regulate M-type potassium current.

Retigabine (RTG) was the first Kv7 potassium channel opener marketed in 2011 for the adjuvant treatment of adult partial-onset epilepsy. In addition to having an anti-epileptic effect, RTG can be used for treating anxiety, neuralgia, neurodegenerative diseases, and the like. RTGs are effective in reducing or arresting seizures in a variety of models of epilepsy. RTG showed potent antiepileptic effects on both tonic seizures induced by the Maximal Electrical Shock (MES) model and PTZ-induced clonic seizures. In addition, RTG can prevent epileptic seizure due to N-methyl-D-aspartate (NMDA), penicillin, tetrandrine, Kainic Acid (KA), etc. The ignition model is suitable for screening various antiepileptic drugs, and the effect of RTG on the model is stronger than that of other models. RTG is less selective due to its broad effect on all Kv7 potassium channel members and other channels, making it potentially undesirable. A great deal of literature reports that the RTG has high incidence rate of adverse events related to the central nervous system, and can cause dizziness, fatigue, aphasia, speech disorder, balance disorder and other adverse reactions including kidney and urinary system diseases such as renal calculus and urinary retention, heart related diseases such as cardiac arrest and transient non-persistent ventricular tachycardia, and can also cause retinal discoloration, blue/purple pigmentation of skin, nails and the like.

Disclosure of Invention

The invention aims to provide a compound shown as a formula A, a preparation method thereof and application thereof as a potassium channel regulator.

In a first aspect of the invention, there is provided a compound of formula A or a pharmaceutically acceptable salt thereof,

formula A.

In a second aspect of the present invention, there is provided a process for the preparation of a compound according to the first aspect of the present invention, or a pharmaceutically acceptable salt thereof, comprising the steps of:

1)andreacting to obtain

2)Andand reacting to obtain the compound of the formula A.

In another preferred embodiment, in step 1),andin a molar ratio of 1: 1-1.2, preferably 1: 1.1.

in another preferred embodiment, in step 2),andis 0.8 to 1.1, preferably 0.8 to 1.

In another preferred example, the method comprises the following steps:

in a third aspect of the invention, there is provided a use of a compound according to the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition which is affected by modulation of potassium ion channels.

In a fourth aspect of the invention, there is provided a use of a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition affected by modulation of the potassium ion channel KCNQ 2.

In a fifth aspect of the invention, there is provided a use of a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition affected by modulation of the potassium channel KCNQ 2/3.

In a sixth aspect of the invention, there is provided a use of a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition affected by modulation of the potassium channel KCNQ 3.

In a seventh aspect of the invention, there is provided a use of a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition affected by modulation of the potassium ion channel KCNQ 3/5.

In an eighth aspect of the invention there is provided the use of a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition affected by modulation of the potassium channel KCNQ 4.

In another preferred embodiment, the disease, disorder or condition is a central nervous system disease.

In another preferred embodiment, the central nervous system disease is selected from the group consisting of: epilepsy, convulsions, inflammatory pain, neuropathic pain, migraine, depression, anxiety disorders, stroke, Alzheimer's disease, neurodegenerative diseases, cocaine abuse, nicotine withdrawal, alcohol withdrawal, tinnitus.

In a ninth aspect of the invention, there is provided a pharmaceutical composition comprising one or more pharmaceutically acceptable carriers and a therapeutically effective amount of one or more compounds of the first aspect of the invention or a pharmaceutically acceptable salt thereof.

In a tenth aspect of the invention, there is provided a method of preventing or treating a disease, disorder or condition affected by modulation of potassium ion channels, comprising the steps of: administering to a patient in need thereof a therapeutically effective amount of a compound according to the first aspect of the invention or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to the ninth aspect of the invention.

It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.

Detailed Description

The present inventors have conducted extensive and intensive studies for a long time and unexpectedly prepared a compound represented by formula a, which has excellent potassium channel opening activity, potassium channel maximal activation rate, pharmacokinetics (e.g., cerebral hematocrit performance, etc.), in vivo efficacy and safety, and a novel structure, by structural optimization. On this basis, the inventors have completed the present invention.

Term(s) for

In the present invention, unless otherwise specified, the terms used have the ordinary meanings well known to those skilled in the art.

Compound (I)

The invention provides a compound shown as a formula A or a pharmaceutically acceptable salt thereof.

As used herein, the term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, and the like; organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid and the like; and amino acids such as proline, phenylalanine, aspartic acid, glutamic acid, etc.

Another preferred class of salts are those of the compounds of the invention with bases, for example alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., magnesium or calcium salts), ammonium salts (e.g., lower alkanolammonium salts and other pharmaceutically acceptable amine salts), for example methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butylamine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, triethanolamine salts, and amine salts formed from morpholine, piperazine, lysine, respectively.

Preparation method

The process for the preparation of the compounds of formula A according to the invention is described in more detail below, but these particular processes do not limit the invention in any way. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.

Typically, the process for the preparation of the compounds of the present invention is as follows, wherein the starting materials and reagents used are commercially available without specific reference.

Pharmaceutical compositions and methods of administration

The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 5-1000mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.

"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g., propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (e.g., tween), wetting agents (e.g., sodium lauryl sulfate), colorants, flavors, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.

The medicine composition is injection, capsule, tablet, pill, powder or granule.

The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.

Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.

Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.

In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.

Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.

Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.

The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.

The treatment methods of the present invention can be administered alone or in combination with other therapeutic means or agents.

When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 1 to 2000mg, preferably 5 to 1000 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.

Compared with the prior art, the invention has the following main advantages:

(1) the compound has better pharmacokinetic performance, such as better brain-blood ratio, half-life period, exposure, metabolic stability and the like;

(2) the compound has better potassium ion channel opening activity, better maximum activation rate of the potassium ion channel, better ion channel selectivity, better in-vivo drug effect and better safety;

(3) the compounds are expected to be useful in the treatment and/or prevention of diseases and disorders which are affected by the activity of potassium ion channels.

The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, molecular cloning is generally performed according to conventional conditions such as Sambrook et al: the conditions described in the Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer's recommendations. Unless otherwise indicated, percentages and parts are by weight.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred embodiments and materials described herein are intended to be exemplary only.

The test materials and reagents used in the following examples are commercially available without specific reference.

EXAMPLE 1 preparation of Compound A

Step one, compound 2

Compound 1 (2.0 g, 10.0 mmol, 1.0 eq) was dissolved in ethyl acetate (100 mL), and 2- (1-methylcyclopropyl) acetic acid (cas:71199-15-0, 1.26 g, 11.0 mmol, 1.1 eq), pyridine (7.9 g, 99.96 mmol, 10.0 eq) and T were added3P (50%, 31.8 g, 49.97 mmol, 5.0 eq), heated to 50 ℃ and stirred for 16 hours. After cooling to 25 ℃ water was added to dilute and extracted with ethyl acetate (3 × 100 mL). The combined organic phases were washed with saturated sodium chloride solution and dried over anhydrous sodium sulfate, and the concentrated residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate =10/1) to give compound 2 (2.5 g, 84%) as a white solid.

LCMS: [M+H]+= 296.0

Step two, compound A

Compound 3(224mg, 0.61mmol, 1.2eq) was dissolved in toluene (5 mL), and Compound 2(150 mg, 0.51mmol, 1.0 eq), potassium tert-butoxide (172mg, 1.53mmol, 3.0 eq), Dave-phos (40 mg, 0.10 mmol, 0.2 eq), and Pd were added in that order2(dba)3(47 mg, 0.051 mmol, 0.1 eq), and is stirred at 80 ℃ for 16 hours under the protection of nitrogen. The reaction solution is cooled to 25 ℃, diluted by ethyl acetate (30mL), washed by water and saturated sodium chloride solution in turn,the organic phase was dried over anhydrous sodium sulfate, and the residue obtained after concentration was purified by silica gel column chromatography (petroleum ether/ethyl acetate =2/1) to obtain compound a (52.3 mg, 28%).

LCMS: [M+H] + = 367.2

1H NMR (400 MHz, DMSO-d 6) δ 8.80 (s, 1H), 7.27-7.24 (m, 1H), 7.02 – 6.99 (m, 2H), 6.71 (s, 2H), 4.31 (s, 2H), 3.48 (t, J = 6.0 Hz, 2H), 2.90 (t, J = 6.0 Hz, 2H), 2.17 (s, 2H), 2.10 (d, 6H), 1.14 (s, 3H), 0.55-0.52 (m, 2H), 0.32 – 0.29 (m, 2H).

Example 2 Potassium channel opener agonist Rate test (FDSS/. mu.CELL assay)

1. The experimental method comprises the following steps:

1.1 Experimental procedures

Cell preparation: CHO-KCNQ2 cells cultured at 175 cm2In a culture flask, when the cell density reaches 60-80%, the culture solution is removed, washed once with 7 mL of PBS (Phosphate Buffered Saline), and then digested with 3 mL of 0.25% Trypsin. After digestion was complete, 7 mL of culture medium (90% DMEM/F12 + 10% FBS + 500. mu.g/mL G418) was added for neutralization, centrifuged at 800 rpm for 3 minutes, the supernatant was aspirated, 5mL of culture medium was added for resuspension, and the cells were counted.

Cell plating: based on the cell count results, the density was adjusted to 3 × 104Standing at room temperature for 30 min, and placing in CO at 37 ℃2The incubator is used for culturing overnight for 16-18 hours, and the cell density reaches about 80%.

Incubation with fluorescent dye: the cell culture medium was discarded, 80. mu.L/well of the loading buffer was added, and the cells were incubated for 60 minutes at room temperature in the absence of light.

Compound incubation: the loading buffer was discarded, 80. mu.L/well of the prepared compound solution was added, and the mixture was incubated at room temperature for 20 minutes in the absence of light.

Fluorescence data acquisition: and (3) recording a real-time fluorescent signal by adopting an FDSS/mu CELL instrument, wherein the excitation wavelength is 480 nm, the emission wavelength is 540 nm, recording is carried out for 1 time per second, after the baseline of 10 seconds is recorded, 20 mu L/hole of stimulation buffer solution is added, and the recording is continuously carried out until 180 seconds are finished.

1.2 preparation of solution

Loading buffer solution: 10 mL/plate, the preparation method is as follows:

testing a buffer sample: 100 mL/plate, the formulation was as follows:

stimulation buffer: 5 mL/plate, the preparation method is as follows:

the buffer is derived from a commercially available kit under the name FluxOR porosity ion channel assay.

1.2 preparation of Compounds

Preparing 20 mM DMSO compound mother liquor, taking 10 mu L of 20 mM compound mother liquor to 20 mu L DMSO solution, and diluting 3 times to 8 intermediate concentrations in series; then compounds of intermediate concentration were added to the test buffer, diluted 200 times to the final concentration to be tested, and 80 μ L was added to the assay plate.

The highest concentration tested was 100. mu.M, which was in turn 100, 33.33, 11.11, 3.70, 1.23, 0.41, 0.137, 0.045. mu.M for 8 concentrations. Each concentration was 3 replicates.

The DMSO content in the final assay concentration did not exceed 0.5%, and this concentration of DMSO had no effect on the KCNQ2 potassium channel.

1.3 data analysis

The experimental data were analyzed by Excel 2007, GraphPad Prism 5.0 software, and the ratio of 180 seconds was counted to calculate the agonistic effect. The compound agonistic effect is calculated by the following formula:

1.4 quality control

Environment: temperature of 25 DEG C

Reagent: FluxORTM assay kit (Invitrogen, Cat # F0017)

The experimental data in the report must meet the following criteria: z' Factor > 0.5

2. And (3) measuring results: see table 1 for details.

TABLE 1

References to the above test methods: ZhaobingGao et al Journal of Biological chemistry 2010, 285(36): 28322-28332.

The compound B is a compound disclosed in patents WO2008024398 and WO2011094186, and has a structure. By comparing the maximum activation rates of compound A and compound B, it can be seen that the tert-butyl group of compound B is changed toThen, the maximum activation rate of the compound on a potassium ion channel KCNQ2 is greatly improved (4.5 times).

EXAMPLE 3 study of the ability of Compounds to cross the blood brain Barrier

1) The research aims are as follows: to obtain a test Compound crossing the blood brain Barrier

2) Content of the experiment

Dividing 9 healthy male ICR mice (weight range of 18-22 g) into 3 groups, 3 mice/group, orally administering test compound after fasting overnight, collecting blood by cardiac puncture at time points of 1h, 2h and 4h, collecting at least 0.5 mL of whole blood to EDTA-K2And (4) centrifuging the anticoagulation tube within half an hour to obtain plasma (6000 rpm, 8 minutes and 4 ℃), and freezing and storing at the temperature of minus 20 ℃ for later use. Meanwhile, brain tissue is collected, washed by normal saline, sucked by absorbent paper, weighed and frozen at-20 ℃ for later use.

The experimental results are as follows: according to the obtained blood concentration data, the pharmacokinetic parameters after drug administration are calculated by adopting a non-compartmental model of WinNonlin 7.0 software (Pharsight, USA).

TABLE 2 cerebral blood ratios at various time points following a single oral administration in male ICR mice

The cerebral blood ratio is important to nerve drugs, and the higher the cerebral blood ratio is, the stronger the compound has the ability to penetrate the blood brain barrier. From a comparison of the data in table 2: the cerebral blood ratio of the compound A is obviously better than that of the compound B disclosed in the patents WO2008024398 and WO2011094186 (more than 4 times).

All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

11页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种布南色林工艺杂质B的制备方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!