CN1736485A - Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease - Google Patents

Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease Download PDF

Info

Publication number
CN1736485A
CN1736485A CN 200510027292 CN200510027292A CN1736485A CN 1736485 A CN1736485 A CN 1736485A CN 200510027292 CN200510027292 CN 200510027292 CN 200510027292 A CN200510027292 A CN 200510027292A CN 1736485 A CN1736485 A CN 1736485A
Authority
CN
China
Prior art keywords
receptor agonist
vanillin
vanillin receptor
substituted
capsaicin
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN 200510027292
Other languages
Chinese (zh)
Inventor
陈春麟
毛晨
张劲涛
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Medicilon Inc
Original Assignee
Shanghai Medicilon Inc
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 Shanghai Medicilon Inc filed Critical Shanghai Medicilon Inc
Priority to CN 200510027292 priority Critical patent/CN1736485A/en
Publication of CN1736485A publication Critical patent/CN1736485A/en
Pending legal-status Critical Current

Links

Images

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention relates to the use of vanillic aldehyde receptor activator in preparing products against Alzheimer's disease, wherein the vanillic aldehyde receptor activator comprises vanillic aldehyde and its derivative, capsaicin analogue and derivatives.

Description

Use of vanillin receptor agonist for preparing anti-Alzheimer's disease product
Technical Field
The invention relates to the technical field of medicines, foods, beverages and the like, in particular to application of a vanillin receptor agonist, and more particularly to application of the vanillin receptor agonist in preparation of an anti-Alzheimer disease product.
Background
Development of vanilloid receptor agonists
Transient Receptor Potential (TRP) channels are widely found in both vertebrates and invertebrates, in activated and non-activated cells, and are the major channels for regulating calcium flow. The mammalian transient receptor potential family consists of 6 protein families: the receptor family of classical transient receptor potential (short for TRPCS), the receptor family of transient receptor potential vanillin (short for TRPVS), the family of transient receptor potential melastatin or long transient receptor potential (short for TRPMS), the family of transient receptor potential mucrolipin (short for TRPMLS), the family of transient receptor potential polycysteine (short for TRPAPS) and the family of anktom protein transmembrane protein 1 (ANKTM1 short for TRPA 1). The transient receptor potential family, which is commonly functionally related to G-protein coupled receptors, growth factor receptors and phospholipases, is found in eukaryotes, from yeast to mammalian cells. The family of transient receptor potentials is widely distributed in the brain, with almost all subunits of transient receptor potentials present.
The transient receptor potential, the vanillin receptor (TRPV for short), plays an increasingly important role in the conversion of the sensory and non-sensory functions, and therefore this receptor family is gaining increasing importance, as the different channels in this group exhibit a unique sensitivity to a large group of harmful and harmless environmental stimuli, including heat, pepper, osmotic pressure and mechanical stimuli. Functionally, the large TRPV families of 6 mammals can be subdivided into two groups: TRPV1, TRPV2, TRPV3 through TRPV4 are high temperature sensitive, non-selective calcium ion channels. TRPV5 and TRPV6 are low temperature sensitive, highly selective calcium ion channels.
TRPV1 (also called vanillin Receptor1, VR1) or capsaicin Receptor in transient Receptor potential vanillin Receptor is found in cloning expression of capsaicin and thermosensitive gated channel, is an externally regulated cation selective ion channel, and is inclined to calcium (PCa/PNa10) or magnesium (P)Mg/PNa5) Depending on one of the aspartate molecules in the core group of the protein. TRPV1 was also activated by warmth (. gtoreq.43 ℃) and by moderate acids (. gtoreq.5.9) as a pool of stimuli caused by chemical and mechanical pain. The pathway caused by heat is direct, and the moderate acid (pH ≦ 5.9) lowers the temperature threshold required for activation, enhancing the response to capsaicin. Capsaicin and resiniferatoxin are potent exogenous agonists of the vanillin receptor. Endogenous agonists include cannabinoid receptor agonists (e.g., peanut tetraenol ethanolamine, etc.) and eicosanoid products of several lipoxidases.
The hydropathic analysis showed that VR1 has 6 transmembrane domains, with a short, porous hydrophobic structure between the 5 th and 6 th transmembrane domains that is involved in and regulates the process of capsaicin activation of VR1, with little relation to hydrogen ion and temperature. The amino-terminal hydrophilic region (432 amino acids) contains a proline-rich region (proline-rich region) linked to three hook-like repeats presumably involved in the interaction between receptor proteins; the carboxy terminus (154 amino acids) contains some unrecognizable motifs (motifs) whose function is not known. VR1 has 23% amino acid sequence homology to some members of other transient receptor potential families.
The capsaicin receptor is expressed on nociceptive receptors of central sensory neurons which sense painThe cation channel is closely related to the feeling of nociceptive impulse and the generation of pain sensation. Under experimental conditions, capsaicin receptor is excited, and the body can generate pain response. Animals with a knockout of capsaicin receptor are responsible for some nociceptive stimuli, such as capsaicin and H+(pH < 5.9) and heat (. gtoreq.43 ℃) are extremely insensitive. The VR1 expressed by artificial clone can be activated by vanillin compound, temperature (43 ℃ or higher) and acid (pH is less than or equal to 5.9), so that VR1 is considered as a molecular complex causing pain by chemical and physical stimulation. There are also experimental results that do not support this hypothesis. Nagy et al compared the cellular membrane responses of rat primary sensory cells to capsaicin and noxious thermal stimuli by electrophysiological and measurement of ion flux, demonstrated that the properties of the ion channels activated by capsaicin or thermal stimuli were much the same, but with an important difference that the permeability of the heat-activated channel calcium ion was lower than that of the capsaicin-activated channel. The channels that respond to thermal stimuli or capsaicin are monosensitive, with few ion channels having dual sensitivity to heat and capsaicin. At the whole cell level, each cell can respond to heat or capsaicin. It is concluded that capsaicin is substantially different from the molecule that causes a cellular response to thermal stimuli and may be associated with multiple subtypes of VR 1.
Capsaicin (capsaicin), the main pungent component in capsicum. Thresh first isolated it from paprika in the middle of the 19 th century and named capsaicin. In 1919 Nelson reported that capsaicin has the structure trans-8-methyl-N-vanillyl-6-nonenamide. Capsaicin has a very pronounced cellular specificity and activates the vanillin receptor (i.e., capsaicin receptor) resulting in biological activity. Current research on capsaicin has focused primarily on exploring the mechanisms underlying pain transmission and on studying its potential utility in pain therapy.
Vanillin is a group of compounds that can act by coupling through its receptor to a non-specific cation channel. Capsaicin is a typical vanilloid receptor agonist, and micromoles of capsaicin inhibit VR 1. A group of compounds structurally similar to capsaicin, including 4-hydroxy-3-methoxybenzyl (i.e., vanillyl) groups, called capsaicin family compounds, has been developed, including dihydrocapsaicin, norcapsaicin, homocapsaicin, homodihydrocapsaicin I, nordihydrocapsaicin, cis-capsaicin, nonivamide, olvanil (NE-19550), NE-21610, N-oleylcharamide (NE-28345), Do-APmarceutical (DA-5018), resiniferatoxins, and the like, wherein the most potent capsaicin group is resiniferatoxin.
The structure-activity relationship of capsaicin for producing analgesia comprises the following components: suitable length of the hydrophobic hydrocarbon chain is 8 to 18 carbons; the 3-methoxy group on the aromatic ring plays an important but not essential role; the phenolic hydroxyl group is indispensable, and the para-position is preferable; amide linkage is essential; amino and cyclic through CH2The connection is best. Some vanillin receptor agonists/inhibitors lacking the known vanillyl group are discovered in succession, as anandamide et al.
(II) progress in the study of Alzheimer's disease
1. Overview
Dementia includes Alzheimer's Disease (or Alzheimer's Disease, abbreviated as AD), multi-infarct Dementia (Multimfarct Dementia), Alcoholic Dementia (Alcoholic Dementia), and Normal brain Pressure hydroencephalopathy (Normal Pressure Hychocephalus).
AD is a chronic primary and progressive brain degenerative disease, a progressive neurodegenerative disease occurring in the elderly or in the early stages of the elderly, a common morbid reaction after the onset of the later years, and Alzheimer's disease which belongs to an abnormal aging state. Since AD is well-developed in elderly people over the age of 60, the habit is called Alzheimer's disease or Alzheimer's disease.
AD is characterized by hypomnesia, cognitive impairment, behavioral abnormalities and speech disability, and is characterized by overall impairment of acquired higher functions of the cerebral cortex, significant impairment of cognitive function, degenerative decline of brain function, i.e., rapid deterioration of memory and impairment of cognitive function, with behavioral or personality changes.
The disease course is usually long, about 3-20 years. The nursing cost of AD patients throughout the year is huge, which not only has great influence on the medical guarantee system of the whole society, but also is a great challenge to the small family model of the future society of China. Therefore, the occurrence of the disease brings heavy economic burden and mental burden to both society and families, and also brings great pain to patients. At present, the disease is still one of the more intractable diseases in the world, and active research and development of new treatment methods are matters which are not slow.
2. Pathological manifestations and pathogenesis
The main pathological manifestations of AD are the formation of extracellular neuritic plaques in the forebrain basement, hippocampus and cerebral cortex, the appearance of intracellular neurofibrillary tangles and the reduction in the number of nerve cells and synapses. The spots are mainly composed of amyloid beta (Abeta) aggregates, which are the proteolytic products of Amyloid Precursor Protein (APP). Under normal conditions, the APP hydrolysate is soluble Abeta40Whereas under pathological conditions, APP hydrolysates are long, fibre-forming A β1-42It first exists as an immature, non-fibrous, discrete spot, which eventually undergoes a conformational change and polymerizes into a toxic fibrous mass, i.e., a spot. Neurofibrillary tangles, which are mainly present in the intracellular and abnormal neurites of pyramidal neurons, are a water-insoluble structure consisting mainly of a double-helical fibrillary structure (abbreviated as PHF) with a diameter of 10nm, and the main component is a microtubule-associated protein, tau protein, which stabilizes the interaction of tubulin with normal tau proteins, while tau proteins in PHFs exist in a hyperphosphorylated form.
The etiology of AD is complex, and there are two major hypotheses at present: the tau protein hypothesis and the amyloid protein hypothesis. the main arguments of the tau protein hypothesis are: hyperphosphorylation of tau protein results in destabilization of tubulin, which causes alterations in golgi structure, thereby affecting APP metabolism and producing excess a β. This hypothesis has not been confirmed at the cellular or animal individual level. The main arguments of the amyloid hypothesis are: the aggregation of peripheral amyloid that occurs in extracellular neuropathies leads to the formation of neuritic plaques and the subsequent formation of neurofibrillary tangles. Thus, amyloid is the cause of AD pathological chain formation. Evidence supporting the amyloid hypothesis is extensive.
In AD patients, a key warning for neuronal loss of function and death is the proteolytic process of APP, resulting in increased production of a β, which accumulates in the brain in neurotoxic forms. A beta is a protein structure containing 40-43 amino acids, one part is embedded in a cell membrane, and the other part is exposed outside the cell. The precursor of A beta protein is Amyloid Precursor Protein (APP), which has 695-770 amino acids in total, most of the A beta protein is exposed outside the cell, and a small part of the A beta protein is embedded in the cell membrane and cytoplasm. The 3 enzymes capable of cleaving APP are: alpha-secretase, beta-secretase (BACE) and gamma-secretase. The formation of a β requires that APP be cleaved by β secretase to form the N-terminal a β and by γ secretase to form the C-terminal a β. Alpha secretase is able to cleave APP internally within A β, thereby hindering A β formation. The products cleaved by the α -secretase and γ -secretase are smaller than A β and are called P3 protein. Both A.beta.and P3 are released from the cell membrane, wherein the free A.beta.self-aggregates to form so-called spots.
In the development of drugs targeting specific sites of the neurodegenerative cascade, since the conversion from APP to the neurotoxic form of a β is a crucial event in the pathological development of AD, it is a hot spot to develop drugs that block β -secretase or γ -secretase. A specific gamma secretase inhibitor that reduces a β production has been developed, but its use in humans is not feasible due to its side effects of blocking gamma secretase from cleaving Notch and other protein substrates. BACE-deficient mice are able to reduce A β production without significant abnormal physiological phenomena, demonstrating that BACE inhibitors are able to reduce A β production without significant side effects, but there is the possibility that other physiologically important BACE substrates than APP may be present, inhibiting their formation may cause other serious problems. Despite much effort in inhibiting β -secretase and γ -secretase, direct action on α -secretase has been overlooked. Few currently contemplated drug targets are alpha secretases that break down within the ap sequence to produce a large, soluble extracellular fragment (abbreviated as sAPPa) and a small intracellular fragment (abbreviated as C83) that do not exhibit pathotoxicity and that may also have neuroprotective properties. Since alpha and beta secretases compete for APP, promoting alpha secretase can result in reduced pathological fragments. In theory, both direct and indirect promoters of alpha secretase are beneficial for AD treatment.
3. Epidemiological conditions
With the increasing life span of both developed and developing countries, the number of elderly suffering from dementia is rapidly increasing. The proportion of AD is highest (about 50% to 60%) among all dementias.
According to data reports, the incidence rate of dementia is continuously increased under the influence of human tissue aging and genes along with the increase of age; wherein AD rapidly increases with age, and doubles every 4 to 5 years from the age of 60; the incidence of AD is about 10% in the elderly around 65 years of age, and about 47% in the elderly over 85 years of age, and has become the fourth killer of humans.
In the past, academia thought that china was a low risk country for AD, however, this was a challenge in the second chinese academy of dementia and depression, international academy of senile dementia, held in tunxi, anhui. 5 research reports show that China is also an AD high-risk country, and a large amount of research results reported in the meeting also show that the research of China on AD is rapidly approaching the international advanced level.
At present, the number of AD patients worldwide is estimated to be about 1200 ten thousand, the number of the existing senile dementia patients in China is about 300-400 ten thousand, the current senile dementia patients in China are in a rapid rising trend, and the patients die within about 7-9 years after diagnosis. The data show that the population of China over 60 years old reaches 1.2 hundred million as early as 1998, and the population increases at a rate of 3.2% per year, which is greatly higher than the population growth rate. The elderly people over 75 years old have 2000 ten thousand, the elderly people over 80 years old have 800 ten thousand, and the elderly people increase at a rate of 5.4% per year, and can reach about 4 hundred million in the middle of the next century, so that the elderly people in the world will become the most old people.
The research of Shanghai Mitsubishi in 1988 shows that the incidence rate of dementia of the elderly people over 65 years old in China is 4.6%, wherein the proportion of AD is 63%, the proportion of multi-infarct dementia is 28%, and the dementia caused by brain tissue injury sequelae, chemical drug poisoning and nervous system disorder or nutrition deficiency accounts for about 10%.
Epidemiological survey in Shanghai shows that the prevalence of dementia in China above 55 and 65 years old is 2.57% and 4.6%, respectively. The investigation of 2788 old people over 60 years old in Beijing by the Beijing medical research center for senile diseases shows that 208 old people suffer from dementia, and the prevalence rate reaches 7.5%. Wherein, 139 female patients have the prevalence rate of 9.7 percent, which is obviously higher than 5.8 percent of male patients. Their studies also indicate that elderly, low education level, living in rural areas are a high risk factor for senile dementia. The investigation of 1728 departed retired cadres in the general hospital of the liberation army shows that the family history of dementia, the coercion in the 'Wen leather', the exposure of electromagnetic field and the brain trauma are also risk factors of senile dementia. In the survey of the prevalence of dementia in elderly people over 75 years of age in Guangzhou urban area, the prevalence of AD was 7.49%.
4. Medicine for treating senile dementia
AD is one of the common diseases of the old, is a progressive neurodegenerative disorder, the etiology of the disease is not fully elucidated at present, and is a troublesome problem in treatment.
The research and development of anti-dementia drugs has attracted high attention from the medical world of various countries in the world. In recent years, with the progress of research on neurophysiology, biochemistry, pharmacology and the like of the elderly, development and research of related drugs have been advanced. 1270 products were put on the market in 2001, 90% of the products were discovered in the 80 s and successfully researched and developed in the 90 s, and the number of new products developed by the medicines exceeds that of any other therapeutic medicines.
The recent 10 years of clinical application show that acetylcholinesterase (AChE) inhibitor has certain therapeutic effect on AD, and the main medicines of tacrine, rivastigmine, donepezil and galantamine are representative varieties in the field, thereby promoting the development process of the dementia treatment drug market.
(1) Brain metabolism agonists
Researches show that senile dementia patients have sugar metabolism and metabolic system disorders such as nucleic acid, protein, lipid and the like, and the cerebral blood flow and oxygen consumption of the senile dementia patients are obviously lower than those of normal people of the same age. Therefore, the brain metabolism stimulant and the brain circulation improving agent, in particular the brain metabolism stimulant with the function of cerebral vessel dilation, become the optional medicines for treating the disease. The medicine includes piracetam, Naofukang, pyritinol, Naofumei, hydrogenated ergot alkali, camptotheca, Coxidect, vincamine, long-acting vincamine, vinpocetine, nimodipine, nimotine, cinnarizine, cyclamen, cloxacarb, pentoxifylline, patritane, ergocristatum, etc. The medicines have different improving effects on some symptoms of the senile dementia, such as hypomnesis, reduced environmental adaptation capability and the like.
(2) Cholinomimetic
It is known that the content of acetylcholine in brain is closely related to memory, and the quantity of acetylcholine in brain of the elderly or dementia patients is reduced, and the choline-supplementing medicines can improve the memory and thinking ability. But choline or lecithin directly administered did not increase acetylcholine. Some people try to use choline or lecithin and a cholinesterase inhibitor physostigmine which can pass through a blood brain barrier to be used for patients with the disease together, so that the memory of the patients can be improved, and the effect of singly applying the lecithin is not obvious. In addition, the physostigmine is singly used for intravenous injection to the patient, the performance of memory determination is improved by about 20 percent compared with that of a placebo, and the physostigmine is found to be effective to the behavior disorder. It is thought that cholinesterase inhibitors may delay the metabolic breakdown of acetylcholine, thereby prolonging the excitation of postsynaptic receptors. In addition, the domestically developed haberein (huperzine A) is a new alkaloid separated from huperzine serrate belonging to huperzine by Chinese scholars, and pharmacological experiments prove that the product has strong cholinomic activity and is a high-efficiency reversible cholinesterase inhibitor.
(3) Acetylcholinesterase Inhibitors (Acetylcholinesterase Inhibitors)
Acetylcholinesterase inhibitors include physostigmine (trade name: Synapton, manufactured by Forest laboratories), tetrahydroaminoacridine (trade name: Cognex, manufactured by Parke-Davis), donepezil (trade name: Aricept, manufactured by Perey/Wei Co.), metrifonate (manufactured by Bayer), and rivastigmine (trade name: Exelon, manufactured by Nowa). These compounds concentrate acetylcholine in nerve tendons by inhibiting acetylcholinesterase and can last longer.
Tetrahydroaminoacridine is the first cholinesterase inhibitor to pass the FDA and clinically treat alzheimer's disease, but the regulations still need to be improved. The product has lower bioavailability than rivastigmine and donepezil, and has greater side effects. Donepezil (Donopozil) is a second generation cholinesterase inhibitor which is also the second approved by the FDA in the united states for the treatment of alzheimer's disease (senile dementia), and is approved by the FDA in 1997 for clinical use, and has effects of promoting cognitive ability of patients with mild and moderate senile dementia, improving mental state of patients and maintaining brain functional activity. Compared with the first generation cholinesterase inhibitor, the product has multiple advantages: (1) the action time is long, and the medicine is taken orally once a day; (2) the drug effect is strong and the curative effect is high; (3) high safety, high selectivity and less untoward reaction of the medicine. At present, the product is sold in China and is also produced by manufacturers at home.
The first acetylcholinesterase inhibitor, Reminyl (galantamine), marketed in Europe was announced by Sanochemia Pharmazeutika AG (Vienna, Austria). The article is approved for marketing in Sweden and is submitted to the European Union in anticipation of admission to other markets in Europe.
NeuroSearch corporation of wheat recently developed clinical studies using NS 2330. NS2330 increased dopamine and norepinephrine activity, while also activating its inhibitory mechanism to stimulate acetylcholine release in the cerebral cortex (including the sensing site). Thus the function of all three neurotransmitters will be affected by alzheimer's disease. The company expects NS2330 to have a better therapeutic effect, and the phase I clinical work for this drug is now completed, and the FDA also agrees to continue the phase II clinical work for this company.
Other compounds to be investigated include xanomeline (Nound and Nodek/Li Co.), besteridine (Herilast Co.) and talsaclidine (Boringer Invitrogen/Framex Puqia Co.).
(ii) tacrine
Tacrine has the chemical name of Tetrahydroaminoacridine (THA) and the trade name of Cognex, is a non-competitive reversible cholinesterase inhibitor of the central nervous system developed by the company Huan-Lanbert in the United states and first marketed, is first marketed in the United states after obtaining FDA approval in 1993, is a new drug for improving AD cognition and an elderly intelligence-benefiting drug, is marketed in the United kingdom, France, Canada and other countries in the same year, is exclusively favorable for becoming due in 1996, and is mainly a product of the United states 'Pade-Davis' in the market at present.
Tacrine has obvious curative effect especially on female AD, can obtain ideal effect when being used together with lecithin, and can obviously improve the memory of patients, so the tacrine is considered to be one of only a few medicines for treating and preventing Alzheimer's disease.
Tacrine has the disadvantages of having great influence on liver function and transaminase index, restricting market development, and the sales volume of tacrine is far less increased than that of other AD medicines. Through further research, the addition screening is carried out on the basis of tacrine mother nucleus, and the Winnakline is developed by German Herstrusel company. The medicine is a tacrine 1-hydroxyl derivative, the action mechanism of the medicine is correspondingly improved, but some adverse reactions still exist, and the medicine is replaced by a new generation of acetylcholinesterase inhibitor along with the development of medicine.
(ii) donepezil
Donepezil is a highly selective and reversible drug for treating AD, is a second generation central acetylcholinesterase inhibitor, is a drug developed by japan toilet pharmaceutical companies, and is approved by the FDA for clinical use on 11/25/1996, with the trade name aricept. The world market was first introduced in the united states in early 1997, co-developed by wei-material/fei, introduced in china in aricept 10 in 1999, and now formed a sales network in over 50 countries of the world.
Donepezil is the second approved medicine for treating senile dementia, and has the advantages of small standard treating dosage, low toxic side effect and high tolerance. Experts generally consider that the drug is in the leading position in AD treatment drugs, and accounts for 60% of the market share in the four main varieties, and the advantage is expected to continue until new drugs which can exert important influence on the treatment of diseases are available.
Reported in the Pharma Business journal: of the 500 drugs sold worldwide in 2000, the sales of the japanese toilet company is $ 4.31 billion and the sales of the fevery company is $ 1.19 billion, which are increased by 27.3% and 30.8% respectively over the last year. Donepezil ranked 101 th worldwide with a total sales of $ 6.83 billion in 2001 and more than 1100 billion yen worldwide in 2002 has been superior in the AD market.
Donepezil raw material medicines and tablet products thereof were developed in 2001 by the chinese medicine research and development center and the chongqing mulberry field pharmaceutical industry, and were approved by the national drug administration for production of four types of new medicines.
③ rivastigmine
Rivastigmine is an amino acid-based brain-selective cholinesterase inhibitor, belonging to the second generation of this class of drugs, developed by norwalk pharmaceutical uk, switzerland under the trade name esnerat, first marketed in switzerland the next year in the uk in 12 months in 1997. FDA approval was obtained at 21 days 4/2000 and marketed in the united states at the end of 6 months in the same year, thereby expanding the market for alzheimer's disease treatment drugs.
The research result shows that: although the half-life period of the medicine is relatively short, the inhibition effect on cholinesterase can reach 10 hours, the medicine does not undergo liver and P450 metabolism, has better tolerance on mild and moderate Alzheimer's disease, has the effect of inhibiting butyrylcholinesterase in brain, and obtains higher evaluation in prospective and random multi-center double-blind research in 45 countries such as Europe, America and the like.
As rivastigmine gains more and more share in the pharmaceutical market, sales increased 83% over the last year in $ 1.195 billion in 2000, and increased 101% in 2001 worldwide in rivastigmine, reaching $ 2.4 billion.
Galanthamine
Galantamine belongs to the second generation acetylcholinesterase inhibitor, and its medicinal components are the same as alkaloid extracted from Narcissus tazetta bulb in European mountain area, and the plant medicine has been used clinically for over 30 years in some countries and regions, and can be used for treating reversal of neuromuscular blockade, myasthenia gravis, infantile cerebral palsy, etc.
Galantamine is a chemically synthesized drug developed by combining Hilei and Qiangsheng company, and the preparation is tablets, capsules, oral liquid and the like, and is clinically used for improving the overall functions of AD patients. The medicine has double action mechanisms, can better stimulate and inhibit acetylcholinesterase, can regulate nicotine receptor sites in brain, can obviously improve cognitive function of light and moderate Alzheimer's disease patients, and delays the process of brain cell hypofunction.
Galantamine was first marketed in the uk, ireland after approval by the european union in 7 months of 2000, was approved in 2001 by the FDA for the treatment of alzheimer's disease in the united states, and is now marketed in 25 countries. Hilei corporation is responsible for sales in the United kingdom, Ireland, and Qiangsheng corporation is responsible for sales in the United states and other European countries. The sale of galantamine in 2001 was reported abroad to be $ 1.36 million.
The Shanghai Shenxing pharmaceutical factory in 1998 has produced galanthamine raw material medicines, the national drug administration approves the sixth pharmaceutical factory in Suzhou in 1999 to produce four new medicines of galanthamine hydrobromide capsules, the Shanghai Shenxing yield in 2001 is increased by 3 times, and the annual yield reaches 30 kg. After the medicine enters the market, the clinical popularization is greatly advanced, and the horn of the head is exposed in the anti-dementia medicine in key hospitals in main cities of China in 2000.
In recent years, acetylcholinesterase inhibitors have become first-line treatment drugs for AD, the status of effectively relieving cognitive dysfunction is confirmed, the sales volume of products occupies the greatest share of anti-dementia drugs, researches show that huperzine A, physostigmine and melittin have certain treatment effect on Alzheimer's disease, huperzine A compounds developed by Shanghai drugs of Chinese academy of sciences have applied for international patents, the action mechanism of the huperzine A compounds is deeply researched, and Beijing tetracyclic, Ningbo Hua drugs, Shanghai Han Yin pharmaceutical industry and Shanghai medical university Douglas pharmaceutical factories obtain new drug certificates and production literature numbers.
(4) Potassium channel blockers
Studies have shown that non-selective potassium channel blockers cause increased release of neurotransmitters, including Ach, by delaying depolarization and prolonging calcium influx to presynaptic nerve terminals. If such drugs are intended for the treatment of senile dementia, the drugs should have the advantage of increasing the release of acetylcholine without decreasing the composition of the membrane phosphatidylcholine, which may be a side effect of the presence of AchE inhibitors. Phosphatidylcholine is a reservoir of choline used by neurons to synthesize Ach. Loss of membrane phosphatidylcholine limits acetylcholine synthesis, resulting in decreased choline output. Clinical studies have shown that non-selective potassium channel blockers have only a modest effect in improving the patient's cognitive ability, and the lack of such effects may be due to low central nervous system penetration, poor selectivity or insufficient activity of these drugs, and are to be perfected by further studies.
(5) Glutamate receptor modulators
In the brains of elderly patients with dementia, there is a disorganization and degeneration of nerve fibers in the cortical and cortical pyramidal cells. These pyramidal cells are reported to have glutamate as an excitatory transmitter. These neurons are damaged and when function is lost it can lead to senile dementia; however, if the functional activity is too strong, excitotoxicity will occur, leading to neuronal death, and causing various neurodegenerative diseases. Therefore, modulation of synaptic activity of degenerated glutamate neurons is promising for the treatment of Alzheimer's disease. Direct activation of postsynaptic receptors has been shown to favor glutamate delivery, and partial agonists have the advantage of acting as agonists when endogenous glutamate is below normal levels and as antagonists when glutamate is released in excess, and thus, partial agonists may provide neuroprotective effects to excitotoxic conditions. Drugs such as memantine (memantine) have been reported to reduce the neurotoxic effects of glutamate when it is released in pathological amounts; when the release of glutamic acid is low, memantine can improve the transfer of glutamic acid required in the memory process, and clinical researches show that the memantine has better tolerance for senile dementia patients and produces mild and statistically significant improvement in psychopathology and behavior determination.
(6) 5-hydroxytryptamine 3 receptor antagonists
The 5-hydroxytryptamine 3 (5-HT 3) receptor antagonist is used for stopping vomit, and the prior research speculates that the antagonist should have other central nervous system effects according to the distribution state of 5-HT3 receptors in the brain. Such as ondansetron, a 5-HT3 antagonist
The pinosylvin can improve the recognition capability of animals with normal and insufficient cholinergic nerve functions, such as mice, marmosets and the like; ondansetron improved memory equivalent to 6 years of memory loss in clinical trials in age-related impaired memory (AAMI) patients over the age of 50. Therefore, ondansetron and other 5-HT3 receptor antagonists (tropisetron, granisetron, etc. may contribute to the foeing of patients with senile dementia.
(7) Research on natural medicine
Many pharmaceutical companies today are also looking to natural drugs in an attempt to develop effective therapeutic drugs therefrom. Progress has also been made in this regard.
Apolipoprotein E4(ApoE4)
As the incidence of Alzheimer's disease is known to be closely related to the genetic factors, the apolipoprotein E4ApoE4 is particularly worthy of attention and research in the direction of treating and preventing Alzheimer's disease. In the drug development, ApoE4 has been subjected to human drug clinical trials, and is likely to be a drug with relatively definite therapeutic effect.
② Beta-amylin (Beta-amyloid)
Beta-amylin is an insoluble polypeptide. The material can effectively prevent the destruction of diethyl bromoacetamide caused in the violent oxidation process. In the theory of the research of Alzheimer's disease, beta-amylin is utilized to limit the generation of free radicals and eliminate the free radicals so as to prevent the damage caused by the free radicals. This class of compounds includes idebenone (martial arts, Osaka japan), which is both a scavenger of free radicals and a stimulator of certain nerve growth factors. Early studies indicate that idebenone can effectively treat moderate dementia. Although the company Gelanwekang (UK) and the company US household products have been marketed in a few countries, it has been withdrawn from the market from Japan and its phase III clinical trial has been stopped.
(8) Progress in the study of other types of drugs
In the last half year of 2000, some studies show that estrogen replacement therapy can significantly delay the onset of Alzheimer's disease in women and can reduce the severity of Alzheimer's disease. Individual clinical studies have also found that estrogen treatment can improve perception. The ability of estrogens to act as antioxidants and anti-inflammatories can reduce the incidence of disease, drive acetylcholine production, and promote neuronal cell growth and survival.
In 2000, Toyama Chemical company, Japan, expected to begin phase II clinical studies of T-588 in the United kingdom and enable earlier trials in the United states. Under the cooperation of New York university, the company found that T-588 can protect brain nerve cells.
FK-960 from Nippon Tengze pharmaceutical company was performed in Japan and USA. The medicine can improve perception, and has new action mechanism. In recent research reports, chewing may prevent memory deterioration in elderly people, but the mechanism of action is still not clear. Researchers investigated the signs of human aging by studying genetic changes in mice. Experiments have shown that mice with teeth pulled out to prevent chewing have a memory that is inferior to that of the normal control group. In addition, researchers have studied brain activity while chewing and found that jaw activity can enhance brain hippocampal signaling.
Researchers from the kentucky university have found that reducing consumption can protect the brain from age-induced disorders similar to alzheimer's disease. Recently published research papers have shown that researchers find that damage to their brains can be reduced by feeding mice with reduced amounts of food. Studies on caloric intake and neurodegenerative diseases in humans have not yet been initiated, but there are several related studies showing that china and japan have a reduced incidence of alzheimer's disease due to the intake of less calories than in the united states and canada.
Neurobiotechnology companies in canada announced a more specific product, memantine, in the first half of 2000. The product is a noncompetitive N-methyl-D-aspartic acid (NMDA) blocker, and is used for treating Alzheimer's disease by performing phase III clinical test in the United states.
5. Market development prospect of senile dementia
The world seven drug market in 2000 showed that the AD drug market has nearly doubled from more than 4 billion dollars in the middle of the 90 s, with the global AD market value being nearly $ 12 billion in 2001; foreign analysts predict that AD patients will increase to about 20% in the next decade, and the growth rate of the AD drug market will increase to a greater extent.
The market sales of AD therapeutic drugs have been steadily increasing due to the increasing number of patients. In the 90 s, these drugs have become popular and have reached $ 50 billion worldwide in 1995. The sales of the medicines exceed the market shares of the first three-ranked medicines for treating cardiovascular diseases, gastrointestinal diseases and anti-infective medicines at the beginning of the 21 st century, and the development of the medicines is good.
At present, the diseases are more and more highly valued by China and society, and because the diseases are similar to rich and expensive diseases such as diabetes and the like and need to be taken for a long time, the medicine market of the diseases is gradually expanded along with the aging population, and the market prospect is good. Based on the above, in order to know the market condition of the AD treatment medicines in China in time, the information center of the southern medicine and economic research institute of the food and drug administration of China selects hundreds of sampling hospitals, 60 medical experts and 120 consumers of six main medication cities (Beijing, Shanghai, Guangzhou, Nanjing, Hangzhou and Chengdu) in China, and carries out comprehensive market research activities on the medication market of senile dementia (1999 and 2000) in China and main competitive varieties thereof.
(1) Analysis of hospital medication amount of Chinese medicine for treating senile dementia in 1999-2000
From the composition of the medicine amount for treating senile dementia in six cities (Beijing, Shanghai, Guangzhou, Nanjing, Hangzhou and Chengdu) sampling hospitals from 1999 to 2000, the total sales amount of the six cities sampling hospitals from 2000 is increased by 28.49 percent compared with 1999. In the main varieties of the medicine for treating senile dementia, the varieties with larger usage amount percentage are ranked as piracetam, ginkgo leaf preparation, hydrogenated ergot alkali, amitrazine/raubasine, aniracetam, nicergoline, citicoline, huperzine A, donepezil, pyritinol, vinpocetine, galantamine, clofazid and idebenone in sequence. The market share (monetary percentage) of the first four varieties is large.
In a specific variety, the market share (sum percentage) of piracetam is increased by 8 percent in 2000 compared with 1999, and still stays at the top, and the sum increase rate reaches 55.59%. The ginkgo leaf preparation and the hydrogenated ergoline keep relatively stable market share, and the use amount is increased to a certain extent. The varieties with faster use amount increase comprise huperzine A and galanthamine, wherein the galanthamine has the highest increase amplitude. Overall market atrophy of amitrazine/raubasine, aniracetam, nicergoline, citicoline, vinpocetine and pyritinol occurred, while idebenone essentially quits market competition. Donepezil is not sold in 1999 due to its newer variety, but has a market share (percentage of used money) of 0.45% in 2000, showing a certain market potential.
(2) Analysis of hospital medication amount trend of Chinese medicine for treating senile dementia in 1999-2000
From the trend of the dosage amount of senile dementia in six cities from 1999 to 2000, the overall dosage level in 2000 is obviously higher than that in 1999, and an increasing trend appears. Wherein the 1999 peak of drug use and the 2000 peak of drug use both occur in quarters (the 2000 peak of drug use is the third quarter), indicating that the use of the high priced species in quarters two is relatively high in 2000. The medication trend of Chinese cities can be reflected in the medication situation of six cities.
(3) Analysis of medication condition of urban hospital mainly using medicine
From the view of the ordering of the drug use amount of each variety in each city, the varieties with better sale performance mainly comprise ginkgo leaf preparations, piracetam, hydrogenated ergot alkali and amitrazine/raubasine. The ginkgo leaf preparation is ranked the first in Shanghai, Guangzhou, Hangzhou and Chengdu areas in 2000, ranked the second in Beijing area and ranked the third in Nanjing area, has quite good performance, is increased compared with 1999, and has popular market reactions of the brands of Dana kang, gingko Tianbao, Huoxuening capsules, Bailuda capsules, Tianbaoning and Yinchuo; secondly, Piracetam is the better one, the Piracetam occupies the first place in Beijing and Nanjing areas in 2000, the Piracetam also occupies the first three names in other cities, and the Piracetam is weaker only in Hangzhou areas (rank 5); the overall situation of the hydrogenated ergoline is ranked third, but the ranking in 2000 is improved compared with the ranking in 1999. The ranking of the ergoline in each city remains substantially unchanged and increases in individual cities such as Shanghai and Chengdu areas, while in Nanjing areas the ranking decreases by one place.
(4) Market evaluation in other aspects
With the popularization of the clinical popularization of drugs for treating AD, AD has been more and more appreciated by society and families, from the view of information feedback of investigated doctors, 68% of doctors consider that the society has begun to concern and appreciate AD, meanwhile, 8.16% of doctors consider that the market supply is seriously insufficient, and 63.2% of doctors consider that the market supply is insufficient. Therefore, the market supply of AD drugs is insufficient, the market still has space, and nearly eight doctors have good market prospects.
Similarly, with the rapid development of OTC market in recent years, a considerable number of varieties (oral dosage forms) are sold in retail drug stores, and due to the characteristic of long-term administration, more and more patients choose to directly take drug treatment from the drug stores. The monitoring results of the medicines from the retail research department of the southern medical and economic research institute of the State drug administration in China show that the market share (percentage of sales) of the ginkgo biloba leaf preparation accounts for about 4 of the market, the Chinese patent medicines are favored by the retail market, the market share of the ginkgo biloba leaf preparation (amitrazine/raubasine) accounts for about 3 of the ginkgo biloba leaf preparation, and the rest varieties account for the rest of the market share. As reflected by the investigated consumer profile, 67.80% of the consumers obtained the required drug from the hospital pharmacy; and 27.97% of consumers go directly to the pharmacy for purchase; 4.24% of patients were obtained from the unit clinic; still 0.85% of patients were purchased at the wholesaler. This means that hospitals are currently the main sales channels for elderly people to take drugs, but it can also be seen that retail channels also account for a non-negligible proportion.
Treatment of AD accounts for the seventh of the world's drug market, costing $ 61 billion by the end of 2005. The existing anti-AD medicines mainly relieve symptoms and do not inhibit the development of diseases. Therefore, it is necessary to develop an effective treatment to prevent the underlying pathogenic processes, and it is considered that significant social and economic benefits can be achieved in developing products, particularly drugs, for the prevention, diagnosis, detection, protection and treatment of AD.
However, no report has been found so far about the use of a vanillin receptor agonist as an anti-alzheimer product through a literature search or the like.
Disclosure of Invention
The technical problem to be solved by the invention is to disclose a new application of the vanillin receptor stimulant so as to overcome the defects in the prior art.
That is, the invention aims to specify the specific application of the vanillin receptor agonist in the aspect of the Alzheimer disease, and further the vanillin receptor agonist is used for preparing an anti-Alzheimer disease product; the anti-Alzheimer disease product is a product for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases thereof, and comprises one or more of medicaments, reagents, foods and the like, preferably medicaments.
Technical idea
The independent development of innovative drugs is a current urgent task in China, the Chinese medicine industry has a long development history, rich experience is accumulated in the aspects of preventing and treating diseases and the like, and the search of effective active ingredients is an effective way and is the advantage of the development of the Chinese innovative drugs.
The current anti-AD drugs mainly have the effect of relieving symptoms and do not inhibit the development of diseases. Therefore, it is necessary to develop an effective prophylactic, diagnostic, therapeutic and research approach to prevent the underlying pathogenic processes.
Two characteristics of AD are spots and tangles, the amyloid-forming spots being the cause of the AD pathological cascade. The spots are Abeta obtained by transforming Amyloid Precursor Protein (APP)1-42Polymerized to form. During conversion of APP to a β, there are several kinases such as GSK3 β, CDK5, and PKC ∈ in play. Activated PKC epsilon increases alpha secretase, leads to increased APP secretion, and decreases A beta1-42And (4) generating.
It has recently been discovered that non-selective cation channels, such as the transient receptor potential vanilloid receptor (abbreviated as TRPV1), play an important role in calcium regulation. Calcium ion is one of the Protein Kinase C (PKC) activating factors in neuron cell, PKC promotes alpha secretase activity and reduces A beta1-42And (4) generating. Therefore, it is an object of the present invention to modulate calcium ion by novel TRPV1 agonists which are autonomously developed, thereby modulating PKC activity, yet further leading to potential a β1-42The generation is reduced, and the effect of treating AD is finally achieved.
Memory impairment or loss associated with AD involves the modulation of such molecular events by PKC agonism. PKC is also involved in APP transformation, and previous studies have shown that PKC agonists-phorbol esters (phorbol esters), benzene lactams (BL for short) and Bryostatin (Bryostatin) greatly increase the secretion of the alpha secretase product sAPPa in fibroblasts of AD patients, reducing A β. These data show that PKC and its activation are a potential and important approach to improve AD pathophysiology and cognitive impairment, thus providing more promising drug development targets. However, these new PKC agonists are not currently available for the development of anti-AD drugs due to safety concerns.
As is well known, calcium ion (abbreviated as Ca)2+) Is one of the PKC activating factors in neuronal cells. TRPV1 channel at Ca2+Plays an important role in regulation. Therefore, the present invention hypothesizes that by modulating TRPV1 channel, PKC activity can be modulated, alpha secretase activity can be promoted, APP progression can be affected, and secretion of sAPPa with neuronal protection can be increasedReducing the production of toxic A β in neurons. The experimental data of the present invention provide strong evidence for this hypothesis. In contrast to PKC, the TRPV1 channel is selectively expressed in neurons. Thus, more specific modulation of TRPV1 activity in neuronal cells may specifically modulate PKC activity.
Ca2+Plays a fundamental role in learning and memory, and is also involved in the survival and death of nerve cells. One aspect of AD pathology is the loss of the ability of nerve cells to regulate calcium balance, ultimately leading to loss of nerve cell function and death. Pathological gene studies on AD patients and on APP and PS1 variants support a chaotic calcium regulation effect in AD disease. Regulation of intracellular calcium concentration plays an important role in controlling various calcium-dependent cellular responses such as gene expression, cell differentiation, and the like. Regulation of intracellular calcium concentration is complex, with inflow and outflow through a variety of pathways. Calcium influx into the cytoplasm is usually due to the release of calcium from internal storage organelles (mainly sarcoplasmic/endoplasmic reticulum SERs) via endoplasmic reticulum calcium release channels (either the triose phosphate receptor or the ryanodine receptor) or via calcium channels on the cell membrane. A new group of calcium channels, the large family of Transient Receptor Potential (TRP) channels, has recently been discovered. This finding reveals molecular entities and signaling mechanisms that control calcium flux in many cells.
According to literature search, the inventor finds out through experiments that the vanilloid receptor agonist has various significant pharmacological activities. The experiment and research result proves and proves that the vanillin receptor stimulant has obvious activity in the aspects of preventing, diagnosing, detecting, protecting and treating AD and the like.
(di) vanilloid receptor agonists
The vanillin receptor stimulant comprises one or more of vanillin and derivatives thereof, capsaicin analogues and derivatives thereof and the like.
Wherein, the vanillin and the derivatives thereof comprise one or more of capsaicin, capsaicin family compounds and derivatives thereof. The vanillin is a group of compounds which can generate effects by coupling a vanillin receptor serving as a receptor to a nonspecific cation channel.
The chemical structural general formula of the vanillin and the derivatives thereof is as follows:
Figure A20051002729200191
wherein n is: 0.1 or 2, etc., preferably 1;
R1comprises the following steps: hydroxy (OH), alkyl, alkoxy, acyloxy, aminoalkoxy, hydrogen (H), amino (NH)2) Or halogen, etc., preferably a hydroxyl group;
R2comprises the following steps: alkoxy, hydrogen, hydroxyl, amino, alkyl, aliphatic or aromatic amine, amine alkoxy, or acyloxy, preferably alkoxy, more preferably methoxy;
R3comprises the following steps: an alkyl or substituted alkyl group having 5 to 23 carbon atoms, an alkenyl or substituted alkenyl group, a diterpene alkenyl group, a phenyl or substituted phenyl group, an adamantyl or substituted adamantyl group, or a piperazinyl or substituted piperazinyl group having 5 to 23 carbon atoms; preferably one of an alkyl group or a substituted alkyl group having 7 to 18 carbon atoms, or an alkenyl group or a substituted alkenyl group having 7 to 18 carbon atoms, etc.; further preferably an alkyl group having 7 to 18 carbon atoms or a substituted alkyl group.
X is: NHC (O), C (O) NH, C (O) O, NHC (O) O, NHC (O) NH, NHC (S) NH, or NH (O) S (O); preferably NHC (O);
the alkyl group herein includes one of a straight-chain alkyl group, a branched-chain alkyl group, a cyclic alkyl group, etc., having 5 to 23 carbon atoms, preferably includes one of an isobutyl group, a tert-butyl group, a sec-butyl group, a pentyl group, a tert-pentyl group, a hexyl group, a cyclopentyl group, a cyclohexyl group, etc., and particularly preferably includes one of a straight-chain alkyl group, a branched-chain alkyl group, a cyclic alkyl group, etc., having 8 to 12 carbon atoms;
the alkyl group in the alkoxy group includes one of a straight-chain alkyl group, a branched-chain alkyl group, a cyclic alkyl group, etc., having 1 to 6 carbon atoms, and includes one of a methyl group, an ethyl group, a propyl group, an isopropyl group, a butyl group, an isobutyl group, a tert-butyl group, a sec-butyl group, a pentyl group, a tert-pentyl group, a hexyl group, a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group, etc., preferably includes one of an alkyl group, etc., having 1 to 3 carbon atoms, and particularly preferably includes one of a methyl group, an ethyl group, etc.;
the aliphatic amine, i.e., a substituted aliphatic primary, secondary or tertiary amine, includes methylamine, ethylamine, propylamine, isopropylamine, four isomers of butylamine, dimethylamine, diethylamine, diethanolamine, dipropylamine, diisopropylamine, di-N-butylamine, tetrahydropyrrole, piperidine, morpholine, 3, 5-dimethylmorpholine, piperazine, N-methylpiperazine, N-ethylpiperazine, N-isopropylpiperazine, 2-methylpiperazine, 3-methylpiperazine, 2, 6-dimethylpiperazine, trimethylamine, triethylamine or tripropylamine, and the like, and preferably includes one of ammonia, methylamine, ethylamine, propylamine, diethylamine, diethanolamine, triethylamine, morpholine, or N-methylpiperazine, and particularly preferably includes one of ammonia, methylamine, triethylamine, morpholine, and the like;
the aromatic amine, i.e., the substituted primary, secondary or tertiary aromatic amine, herein includes one of aniline, substituted aniline, N-methylaniline, N-ethylaniline, pyridine, substituted pyridine, quinoline, substituted quinoline, isoquinoline, substituted isoquinoline or quinine, etc., preferably substituted quinoline, and among the substituted quinolines preferably (5-ethyl-1-aza-bicyclo [2.2.2] -oct-2-yl) -quinolin-4-yl-methanol.
For example, R2Is H, OH, OCH3、OCH2CH3、OCH2CH2CH3、OCH2CH2CH2CH3、OC6H5、CH2NH2、CH2CH2NH2、CH2CH2CH2NH2、OC(O)CH3、OC(O)CH2CH3、OC(O)CH2CH2CH3、OC(O)CH2CH2CH2CH3Or OC (O) C6H5One of the like; preferably OCH3、OCH2CH3、OCH2CH2CH3Or OCH2CH2CH2CH3One of the like; further preferably OCH3
For example, X is one of NHC (O), NHC (O) O, NHC (O) NH, NHC (S) NH, C (O) O or NH (O) S (O), etc.; preferably one of NHC (O) or C (O) O, etc.; further preferred is NHC (O).
Wherein, the capsaicin analogues and the derivatives thereof are a group of compounds which do not contain vanillyl and have similar structure-activity relationship with capsaicin.
The structural formula of part of capsaicin analogues and derivatives thereof is as follows:
Figure A20051002729200211
Figure A20051002729200212
N-(2-Hydroxyethyl)-5,8,11,14-eicosatetraenamide
for short: anandamide
Phorbol 12-phenylacetate
For short: PPHV
The structural-activity relationship is similar to that of capsaicin, and the structural-activity relationship comprises one or more of the following structural-activity relationships:
the appropriate length of the hydrophobic hydrocarbon chain is 8-18 carbons; the aromatic ring 3-methoxy plays an important but not essential role; the phenolic hydroxyl group is indispensable, and the most suitable position is the para position; the amide chain is indispensable; amino and cyclic through CH2Ligation is suitable (J toxicol. Sci.1991 Feb;16 Suppl 1:3-20);
② the compound has three hypothetical binding sites: a-vanillyl, B-amido, C-fatty chain; among them, the fatty end of the compound shows great flexibility and overall hydrophobicity to accommodate the C site.
In the C region, a rigid ring structure can be introduced, including one or more of amantadine-2-amine, amantadine-2-ol, (5-ethyl-1-aza-bicyclo [2.2.2] -oct-2-yl) -quinolin-4-yl-methanol, 1-phenyl-piperazine, isoindole-1, 3-dione, or 2-methyl-2-propanol, etc.
Determination of the magnitude of the compound's anti-AD activity is required by testing its ability to penetrate the blood brain barrier.
The key rules for the penetration of molecules across the blood-brain barrier are: (ii) a small to medium volume; fat soluble; ③ no electric charge.
These rigid ring structures enable the C domain to be more compact and less bulky, mostly lipid soluble (i.e., hydrophobic), with the exception of the piperazine substituent, and without charge, and contain some structures that successfully penetrate the BBB, which all indicate that these compounds are preferred suitable structures for anti-AD drugs.
Pharmacological Activity of (tri) vanilloid receptor agonists
Two characteristics of AD are spots and tangles, the amyloid-forming spots being the cause of the AD pathological cascade. The spots are Abeta obtained by transforming Amyloid Precursor Protein (APP)1-42Polymerized to form. During conversion of APP to a β, there are several kinases such as GSK3 β, CDK5, and PKC ∈ in play. Activated PKC epsilon increases alpha secretase, leads to increased APP secretion, and decreases A beta1-42And (4) generating.
It has recently been discovered that non-selective cation channels, such as the transient receptor potential vanilloid receptor (abbreviated as TRPVI), play an important role in calcium regulation. Calcium ion is one of the Protein Kinase C (PKC) activators in neuronal cells, and PKC promotes alpha secretionEnzymatic activity, reduction of A beta1-42And (4) generating. Therefore, it is an object of the present invention to modulate calcium ion by novel TRPV1 agonists which are autonomously developed, thereby modulating PKC activity, yet further leading to potential a β1-42The generation is reduced, and the effect of treating AD is finally achieved.
Memory impairment or loss associated with AD involves the modulation of such molecular events by PKC agonism. PKC is also involved in APP transformation, and previous studies have shown that PKC agonists-phorbol esters (phorbol esters), benzene lactams (BL for short) and Bryostatin (Bryostatin) greatly increase the secretion of the alpha secretase product sAPPa in fibroblasts of AD patients, reducing A β. These data show that PKC and its activation are a potential and important approach to improve AD pathophysiology and cognitive impairment, thus providing more promising drug development targets. However, the safety of the new PKC agonists for humans remains to be verified.
As is well known, calcium ion (abbreviated as Ca)2+) Is one of the PKC activating factors in neuronal cells. TRPV1 channel at Ca2+Plays an important role in regulation. Therefore, the present invention hypothesizes that by modulating the TRPV1 channel, PKC activity can be modulated, α secretase activity can be promoted, APP progression can be affected, sAPPa secretion with neuronal protection can be increased, and the production of neuronal toxic a β can be reduced. The experimental data of the present invention have confirmed this. In contrast to PKC, the TRPV1 channel is selectively expressed in neurons. Thus, more specific modulation of TRPV1 activity in neuronal cells may specifically modulate PKC activity.
Ca2+Plays a fundamental role in learning and memory, and is also involved in the survival and death of nerve cells. One aspect of AD pathology is the loss of the ability of nerve cells to regulate calcium balance, ultimately leading to loss of nerve cell function and death. Pathological gene studies on AD patients and on APP and PS1 variants support a chaotic calcium regulation effect in AD disease. Regulation of intracellular calcium concentration plays an important role in controlling various calcium-dependent cellular responses such as gene expression, cell differentiation, and the like. The regulation of intracellular calcium concentration is very complicated and there are various inflows and outflowsA channel. Calcium influx into the cytoplasm is usually due to the release of calcium from internal storage organelles (mainly sarcoplasmic/endoplasmic reticulum SERs) via endoplasmic reticulum calcium release channels (either the triose phosphate receptor or the ryanodine receptor) or via calcium channels on the cell membrane. A new group of calcium channels, the large family of Transient Receptor Potential (TRP) channels, has recently been discovered. This finding reveals molecular entities and signaling mechanisms that control calcium flux in many cells.
The invention carries out various tests on the activity of the vanillin receptor stimulant in the aspects of prevention, diagnosis, protection, treatment, research and the like of the Alzheimer disease.
Use of (tetra) vanilloid receptor agonists
1. Overview
The invention aims to provide a product for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases thereof, wherein the product comprises one or more of medicines, reagents, foods, beverages and the like, preferably medicines.
Pharmacological activity screening proves that the vanillin receptor stimulant has the activity of preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases. The completed acute toxicity experiment proves that the maximum tolerance of the mouse gavage administration to the active site exceeds 2.0g/kg, which is equivalent to 440 times of the clinical recommended dosage, and the vanillin receptor stimulant is safe and reliable.
In conclusion, the inventor conducts theoretical exploration on the vanillin receptor agonist, and finds that the vanillin receptor agonist has remarkable activities of preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases through a large number of experimental researches, particularly long-term pharmacological tests. Therefore, the vanillin receptor stimulant and the composition thereof can be used for preparing anti-Alzheimer disease products, and preferably medicaments prepared by taking the vanillin receptor stimulant as a raw material are selected.
2. Methods of use and requirements for vanilloid receptor agonists and compositions thereof
The vanillin receptor agonist of the invention can be used alone or in combination with other active components, and is used for preparing products for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases thereof, including medicaments, reagents, foods, beverages and the like, in particular medicaments.
In particular uses, the vanillin receptor agonists of the invention can be used alone, but can also be used with many other chemicals. Whether or not these chemical substances have biological activity or function for treating diseases, including auxiliary functions such as synergistic amplification, antagonism or alleviation of side effects of vanillin receptor agonists, etc., these chemical substances include one or more of pharmaceutically acceptable carriers, foods, natural products, chemically synthesized drugs or human drugs, etc.; preferably comprises one or more of pharmaceutically acceptable carriers or food, etc.; further preferably a pharmaceutically acceptable carrier.
As used herein, "pharmaceutically acceptable carrier" includes any and all physiologically acceptable solvents, dispersion media, integuments, antibacterial and antifungal agents, isotonic or absorption delaying agents, and the like. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, or ethanol, and the like, and combinations thereof. In many cases, it will be desirable to include isotonic agents, for example, one or more of sugars, polyalcohols such as mannitol, sorbitol, sodium chloride and the like in the composition. The pharmaceutically acceptable carrier may also contain minor amounts of auxiliary substances, such as one or more of wetting or emulsifying agents, preservatives or buffers, and the like, which enhance the efficacy or longevity of the vanillin receptor agonist.
In a specific classification, the pharmaceutically acceptable carrier refers to a conventional pharmaceutical carrier in the pharmaceutical field, and includes excipients, such as one or more of starch, water and the like; one or more of a lubricant, such as glycerin or magnesium stearate, and the like; disintegrants, such as microcrystalline cellulose and the like; fillers, such as one or more of starch or lactose; a binder such as one or more of pregelatinized starch, dextrin, cellulose derivatives, alginate, gelatin, or polyvinylpyrrolidone, etc.; osmotic pressure regulators, such as one or more of glucose, sucrose, sorbitol, or mannitol; diluents such as water and the like; disintegrating agents, such as one or more of agar, calcium carbonate or sodium bicarbonate; absorption accelerators such as quaternary ammonium compounds and the like; surfactants such as cetyl alcohol and the like; an adsorption carrier, such as one or more of kaolin, bentonite, etc.; lubricants, such as one or more of talc, calcium stearate, magnesium stearate, or polyethylene glycol; in addition, other adjuvants such as one or more of flavoring agents or sweeteners may also be added to the composition.
For example, an injectable formulation can be prepared by dissolving, suspending or emulsifying the active ingredient vanillin receptor agonist in a suitable aqueous solvent (e.g., one or more of distilled water, physiological saline or a solution of green) or oily solvent (e.g., one or more of vegetable oil such as olive oil, sesame oil, cottonseed oil, corn oil or propylene glycol) which may contain a dispersing agent (e.g., one or more of polysorbate 80, polyoxyethylene hardened castor oil 60, polyethylene glycol, benzyl alcohol, chlorobutanol or phenol, etc.), an osmotic pressure regulator (e.g., one or more of sodium chloride, glycerol, D9-mannose, D-sorbitol or glucose, etc.). In this case, additives such as solubilizing agents (e.g., one or more of sodium salicylate, sodium acetate, or the like), stabilizers (e.g., human serum albumin, or the like), analgesics (e.g., benzyl alcohol, or the like), and the like may be added, if necessary.
The vanillin receptor agonists described herein can also be used in combination in the form of a composition, in particular a composition for the treatment of animals, especially mammals including humans or other animals, with other chemicals such as drugs, or a similar composition. The mammal includes one or more of human, mouse, rat, sheep, monkey, cow, pig, horse, rabbit, dog, chimpanzee, baboon, marmoset, macaque or rhesus monkey, preferably one or more of human, mouse, rat, monkey, pig, rabbit or dog, and more preferably one or more of human, rat or monkey. For example, a vanillin receptor agonist of the invention can be added to a pharmaceutical composition suitable for administration to a subject. Typically, the pharmaceutical composition comprises a vanillin receptor agonist of the invention and a pharmaceutically acceptable carrier.
The composition of the vanillin receptor agonist, particularly the pharmaceutical composition, can be in various forms including one or more of dosage forms such as liquid, semi-solid, and solid; wherein the pharmaceutical composition comprises a therapeutically effective amount of a vanillin receptor agonist as an active ingredient, and one or more pharmaceutically acceptable carriers.
The pharmaceutical compositions of the vanillin receptor agonist can be formulated into various dosage forms using conventional manufacturing methods well known in the art, such as by mixing the active ingredient with one or more carriers and then formulating into the desired dosage form. The dosage form comprises one or more of tablets, capsules, granules, suspensions, emulsions, solutions, syrups or injections, and the like, and one or more administration routes of oral administration or injection (including one or more of intravenous injection, intravenous drip, intramuscular injection or subcutaneous injection, and the like), mucosal dialysis and the like are adopted for treating or scientifically researching the AD and related diseases thereof.
The pharmaceutical composition preferably contains 0.5-99% by weight of the active ingredient of the vanillin receptor agonist, further preferably contains 1-95% by weight of the active ingredient of the vanillin receptor agonist, and most preferably contains 5-90% by weight of the active ingredient of the vanillin receptor agonist.
Pharmaceutical compositions of vanillin receptor agonists generally must be sterile and stable under the conditions of manufacture and storage. The compositions may be formulated as solutions, microemulsions, dispersions, liposomes or other ordered structures suitable for high drug concentrations. Sterile injectable solutions are prepared by incorporating the vanillin receptor agonist in the required amount in an appropriate solvent with one or a combination of the ingredients enumerated above, as required, followed by sterile filtration. Generally, dispersions are prepared by adding the vanillin receptor agonist to a sterile vehicle containing a basic dispersion medium and the required other ingredients described above. In the case of sterile powders for the preparation of sterile injectable solutions, the recommended methods of preparation are vacuum drying and freeze drying. Proper fluidity of the solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. Prolonged absorption of the injectable compositions can be achieved by including in the composition an agent which delays absorption, for example, monostearate salts or gelatin.
When the dose of the vanillin receptor agonist is 5-20 mg/kg-d, the dose or the dosage is generally determined according to the age and the weight of a patient or a user and the physical condition or the condition of symptoms of the patient. That is, the following dosage ranges are safe and reliable, and the dose of the vanillin receptor agonist is 1-3600 mg/d (calculated by 70kg body weight); 1-2000 mg, preferably 50-1000 mg, is orally taken; injecting 2-400 mg, preferably 50-200 mg; 0.5-5% of ointment (deposition), preferably 1-2%.
The vanillin receptor agonists and pharmaceutical compositions thereof of the invention may comprise a "therapeutically effective amount" or a "prophylactically effective amount" of the vanillin receptor agonists of the invention. A "therapeutically effective amount" is an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic effect. A therapeutically effective amount of a vanillin receptor agonist can vary depending on factors such as the condition, age, sex, and weight of the individual and the ability of the vanillin receptor agonist to elicit a desired response in the individual. A therapeutically effective amount also refers to an amount of the vanillin receptor agonist that has a beneficial therapeutic effect in excess of any toxic or deleterious effects thereof. A "prophylactically effective amount" is an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic effect. Because prophylactic doses are used in subjects before or at an early stage of the disease, the prophylactically effective amount is generally less than the therapeutically effective amount. A typical, non-limiting range of therapeutically or prophylactically effective amounts of the vanillin receptor agonists of the invention is 5-20 mg/kg, more preferably 5-10 mg/kg. It is to be noted that the dosage value will vary depending on the type and severity of the disease to be alleviated, that is, when administered to a patient, the dosage or amount of the vanillin receptor agonist described herein will generally be determined depending on the age and weight of the patient or user and the physical condition or condition of the patient's symptoms. In addition, it is to be understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the dosage ranges set forth herein are exemplary only and do not limit the scope or practice of the claimed compositions.
That is, the dose or amount per and/or daily of the vanillin receptor agonist of the invention is required to vary according to the subject to be treated, the route of administration, the disease and condition to be treated, and the like. For example, the vanillin receptor agonist is administered intravenously to a mammal, particularly an adult (e.g., a human with a body weight of 60kg), in a single dose of about 5 to 10mg, preferably about 10mg, preferably 1 to 3 times daily. Dosage units may be adjusted to provide the optimum desired response (e.g., therapeutic or prophylactic response). For example, a bolus dose may be given, several aliquots may be given over a period of time or the dose may be proportionally reduced or increased depending on the exigencies of the therapeutic situation. It is particularly advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units of unit dosage suitable for the mammalian subject to be treated; each unit containing a predetermined quantity of an active vanillin receptor agonist calculated to produce the desired therapeutic effect in association with the desired pharmaceutical carrier. The specification for the dosage unit forms of the invention is determined by and directly depends on the following (a) unique characteristics of the vanillin receptor agonist and the particular therapeutic or prophylactic effect to be achieved, and (b) the inherent limitations in the art of admixing such a vanillin receptor agonist for the treatment of an individual sensitive to.
3. Pharmaceutical dosage forms and routes of administration of vanilloid receptor agonists and compositions thereof
The vanillin receptor agonist and the composition thereof are prepared into products for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases, wherein the products prepared according to the requirements of the technical fields of beverages and foods can be used for preventing, protecting and treating the Alzheimer disease and the related diseases; the product prepared according to the requirements of the medical technical field can be used for treating or protecting the health of patients, can be directly used for preparing medicaments for treating or protecting the health independently, and can also be mixed or combined with a plurality of chemical substances to be directly or indirectly used for preparing the medicaments for treating or protecting the health. The chemistry described herein is the same as described above in this section.
In the invention, the required materials comprise the raw materials of the invention, the chemical substances used in coordination with the raw materials and the like, and food-grade or medicinal-grade materials are adopted according to actual conditions and requirements.
The vanillin receptor agonists and compositions thereof of the invention can be administered by a variety of methods known in the art, although the recommended route/mode of administration in many therapeutic applications is by spray or oral administration. However, the skilled artisan will appreciate that the route/mode of administration will vary depending on the desired result. In certain implementations, the active compound can be formulated, for example, as an empty release formulation, including one or more of a graft delivery system, a transdermal patch delivery system, or a microencapsulated delivery system, along with a carrier that protects the compound from rapid release. In addition, biodegradable, biocompatible polymers may also be used, such as one or more of ethyl vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, or polylactic acid, among others. Many methods of preparing such formulations are patented or generally known to those skilled in the art (see, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, eds., Marcel Dekker, Inc., New York, 1978).
The vanillin receptor agonists and compositions thereof of the invention are administered to a patient in need of such treatment, typically by one or more of oral, nasal inhalation, rectal or parenteral administration, and the like.
For oral administration, it may be formulated into one or more conventional solid preparations such as tablets, powders, granules or capsules. In practice, the vanillin receptor agonist of the invention may be administered orally, for example, with an inert diluent or an assimilable edible carrier. The vanillin receptor agonist (and other components, if desired) can also be encapsulated in a hard or soft shell gelatin capsule, compressed into a tablet, or added directly to the diet of the subject. For oral therapeutic administration, the vanillin receptor agonist may be added with excipients and used in the form of one or more of an edible tablet, buccal tablet, lozenge, capsule, suspension, syrup, or wafer, and the like.
In order to administer the present vanillin receptor agonist in addition to parenteral administration, it may be desirable to coat the vanillin receptor agonist with a material that prevents inactivation thereof or to administer it together with the vanillin receptor agonist. Supplementary active compounds may also be added to the composition. In particular implementations, the vanillin receptor agonist of the invention is co-formulated and/or co-administered with one or more other therapeutic agents that may be used to treat a disease. Such a combination may advantageously utilize lower doses of the administered therapeutic agent, thus avoiding possible toxicity or complications associated with various monotherapies.
Making into liquid preparation such as aqueous solution, oil suspension or other liquid preparation, such as syrup or elixir; for parenteral administration, it may be formulated into one or more of solution for injection, aqueous solution, oily suspension, etc.
Among the above-mentioned forms of use, preferred forms are one or more of tablets, coated tablets, capsules, suppositories, injections and the like, more preferred are one or more of tablets, capsules, injections and the like, and particularly preferred are injections.
In conclusion, the vanillin receptor agonist and the composition thereof can be used for products, preferably medicines and foods, and further preferably medicines, for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases thereof.
(IV) technical specialties
The invention expands the new medical application of the vanillin receptor stimulant and provides a new medicine source for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases thereof. The vanillin receptor agonist has the advantages of safety, low toxicity, strong pharmacological action, wide raw material source, low price and simple preparation process, and can be used for preparing products for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases thereof.
The invention researches the vanillin receptor stimulant with pertinence, the vanillin receptor stimulant has strong pharmacological action, the raw material source is rich, the preparation process is simple, and the yield is high; the multifunctional electric heating cooker is safe to use, multipurpose, capable of playing a role to the maximum extent, and wide in application range, so that the multifunctional electric heating cooker is easy to popularize and apply, and can generate great social benefits and economic benefits in a short time.
The active form of the vanillin receptor stimulant in the gastrointestinal tract biotransformation directly uses the vanillin receptor stimulant, so that the bioavailability of the medicine is high, and the dosage of the medicine can be accurately controlled. The vanillin receptor agonist has stable quality, and the prepared preparation has stable quality.
In addition, the vanillin receptor agonist has stable chemical property, and has obvious effects of preventing, diagnosing, detecting, protecting, treating and researching the Alzheimer disease and related diseases, so the vanillin receptor agonist is more suitable for industrial production of products for preventing, diagnosing, detecting, protecting, treating and researching the Alzheimer disease and related diseases.
In a word, the invention actively adapts to the working requirements and the requirements of humanized services in the field of modern medical treatment and scientific research, and is a safe raw material for preventing, diagnosing, detecting, protecting, treating and researching Alzheimer disease and related diseases.
Drawings
FIG. 1 shows: capsaicin activity was measured in SY5Y/VR1/SWAPP cells using the FLIPR assay;
the english headings in the upper part of the figure are: dose-response curves for capsaicin for SY5Y-hVR1 and CHO/hVR1/r VR1 cell lines;
the ordinate in the figure is: relative fluorescence units;
the abscissa in the figure is: capsaicin log concentration;
FIG. 2 shows: detection of capsaicin vs. Abeta by ELISA1-42The impact of the generation;
the english headings in the upper part of the figure are: activation of the VR1 ion channel results in A β1-42A reduction in generation;
the ordinate in the figure is: abeta (beta)1-42Concentration;
the abscissa in the figure is: capsaicin concentration;
FIG. 3 shows: quantitative Western Blot (Western Blot) method PKC epsilon kinase activity was determined.
The english headings in the upper part of the figure are: activation of the VR1 channel results in enhanced PKC activity;
the ordinate in the figure is: PKC activity;
the abscissa in the figure is: capsaicin concentration.
Detailed Description
The invention researches the new pharmacological action and the new application of the existing vanillin receptor stimulant, provides a raw material for preparing products for preventing, diagnosing, protecting and treating the Alzheimer disease and the like, and is convenient for safe use in the fields of medical industry and related industries such as food, beverage and the like.
(one) specific examples of existing vanillin and derivatives thereof
The capsaicin is a typical vanilloid receptor agonist, and micromoles of capsaicin can inhibit VR1, and the structural formula is as follows:
Figure A20051002729200281
the described capsaicine compound and its derivative are a group of compounds whose structure-activity relationship is similar to that of capsaicine, i.e. capsaicine group compound containing vanillyl group (4-hydroxy-3-methoxybenzyl-vanillyl group) and its derivative, including dihydrocapsaicin (dihydrocapsaicin), norhydrocapsaicin (nordihydrocapsaicin), homocapsaicin (homocapsaicin), homodihydrocapsaicin (homodihydrocapsaicin), homodihydrocapsaicin I, norhydrocapsaicin, nordihydrocapsaicin (nordihydrocapsaicin), cis-capsaicin (cis-capsaicin), nonivamide, olvanillyl (NE-19550), NE-21610, N-olenyl-vanillylamide (NE-28501), Dong-APharmaceutical (APharmaceutical-DA 8) and other resins. Among the most potent compounds of the capsaicin family are the resiniferatoxins and other compounds.
The vanillyl-containing vanilloid receptor agonist has the following structural formula:
Figure A20051002729200282
Capsaicin
Dihydrocapsaicin
Homocapsaicin
Figure A20051002729200293
Homodihydrocapsaicin
Figure A20051002729200294
Norcapsaicin
Figure A20051002729200295
Nordihydrocapsaicin
Nonivamide
Figure A20051002729200302
Resiniferatoxin
(all Z)-N-(4-Hydroxyphenyl)-5,8,11,14-eicosatetraenamide
AM 404
Figure A20051002729200311
N-[(4-Hydroxy-3-methoxyphenyl)methyl]-5Z,8Z,11Z,14Z-eicosatetraeamideArvanil
Figure A20051002729200312
DA-5018
N-Oleoyldopamine
OLDA
Figure A20051002729200322
NE-19550N-vanillyloleamide
the vanillyl-free vanilloid receptor agonist has the following structural formula:
Figure A20051002729200323
N-(2-Hydroxyethyl)-5,8,11,14-eicosatetraenamide
Anandamide
Phorbol 12-phenylacetate
PPAHV
synthesis and design of (di) capsaicin derivatives
1. Synthesis method of partial compound
Method for synthesis of vanilloid receptor agonist a:
the carboxylic acid was dissolved in 2-10 times of oxalyl chloride, and two drops of DMF (N, N-dimethylformamide) were added and stirred for 3 hours or until bubbling ceased. Excess oxalyl chloride can be distilled off under reduced pressure, but usually without further purification. Vanillylamine hydrochloride is dissolved in DMF (or THF, tetrahydrofuran), 2 times of 5N NaOH is added, the mixture is stirred for 30 minutes, then cooled to 0 ℃, 1-1.2 times of ether of acyl chloride or chloroform solution is added dropwise, and the mixture is stirred for 3-24 hours at room temperature. The reaction solution was poured into water at about 10 times the volume of DMF (when THF was used as the solvent, it was evaporated and ether was added). After separation of layers, the aqueous layer was extracted with ether or chloroform. The combined extracts were washed sequentially with 1N HCl, saturated NaHCO3Water and brine, then MgSO4Drying and evaporating to dryness. The crude product was purified by silica gel column chromatography.
Synthesis of vanillin receptor agonists method B:
4-acetoxyl-3-methoxyphenylacetic acid and 3-12 times of SOCl2Is refluxed until bubbling ceases. Excess SOCl2Is evaporated to dryness. Adding benzene and evaporating to remove trace amount of SOCl2. The resulting acid chloride was dissolved in about 20ml/10mmol of benzene and cooled to 0 deg.C and added dropwise to a 2.0 fold solution of the amine in ether or amine in chloroform (about half the volume of benzene used). The reaction was stirred at room temperature for more than 3 h. The precipitate was filtered and washed with ether. The filtrate was washed with water and brine, and MgSO4And (5) drying. The solvent is then vacuum stripped to dryness to afford the acylated amino compound. The amino compound was dissolved in 25ml/10mmol of MeOH, and 1.2-2.0 times of 5N NaOH was added. The mixture was stirred at room temperature for 3h and the solvent was evaporated. The residue was dissolved in 1N NaOH, extracted with ether, cooled, acidified with HCl and extracted three times with ether. Mixed extract H2Wash with O and brine over MgSO4And (5) drying. The solvent was removed in vacuo to give a crude product.
Method C for synthesis of vanilloid receptor agonist:
1 equivalent of amine and 1-1.2 equivalents of ethyl vanillin were mixed and heated to 170 ℃ to volatilize the EtOH produced. After 3h, the remaining EtOH was distilled off with a weak vacuum. The reaction solution is cooled, diluted with ether, and sequentially treated with water, 1NHCl and NaHCO3Washed with brine and then MgSO4And (5) drying. The solvent was evaporated to obtain a crude product.
4-acetoxy-3-methoxyphenylacetic acid. Homovanillic acid (30g, 0.17mol), acetic anhydride (75ml, 0.79mol) and 2 drops of concentrated H2SO4The mixture was stirred at room temperature overnight, and the reaction solution was poured into 1.8L H2And stirring for 3 hours in the O solution. The precipitate was filtered, the filtrate was concentrated to a volume of about 400 ml, all the precipitate was collected by filtration, recrystallized in hot water, filtered and dried to yield 29.4g (79%) of product, mp 138-139 ℃ (lit.*mp 139~140℃)。
2. The synthetic 12 vanillin receptor agonists of the invention
The synthesis of twelve compounds is illustrated below.
Figure A20051002729200351
Synthesis of compounds 1 and 2:
dissolving 3-methoxy-4-hydroxybenzoic acid or 3-methoxy-4-hydroxyphenylacetic acid in dimethyl adamantyl-2-amine, adding HATU, heating to 180 ℃, reacting for 3 hours, and detecting by TLC to complete the reaction. The reaction residue was washed with 1N hydrochloric acid, a saturated sodium bicarbonate solution, and water in this order. The crude product was purified by column chromatography.
Synthesis of compounds 3, 4:
mixing 3-methoxy-4-hydroxybenzoic acid or 3-methoxy-4-hydroxyphenylacetic acid and adamantyl-2-ol, slowly adding concentrated sulfuric acid, heating to 120 ℃, reacting for 2 hours, and detecting by TLC to complete the reaction. The reaction residue was washed with 1N hydrochloric acid, a saturated sodium bicarbonate solution, and water in this order. The crude product was purified by column chromatography.
Synthesis of compounds 5, 6:
mixing 3-methoxy-4-hydroxybenzoic acid or 3-methoxy-4-hydroxyphenylacetic acid and (5-ethyl-1-aza-bicyclo [2.2.2] -oct-2-yl) -quinolin-4-yl-methanol, slowly adding concentrated sulfuric acid, heating to 120 ℃, reacting for 2 hours, and detecting by TLC that the reaction is complete. The reaction residue was washed with saturated sodium bicarbonate solution and water in this order. Purifying the crude product by column chromatography
Synthesis of compound 7:
3-methoxy-4-hydroxybenzoic acid and Boc protected 2-bromoethylamine were reacted in triethylamine at room temperature for 3 hours, after which the solvent was removed by rotary evaporation. Then, dimethyladamantyl-2-amine and HATU were added thereto, and the reaction was stirred at 120 ℃ overnight. Diluting with water, extracting with ethyl acetate, drying, and spin-drying to obtain crude product. Dissolving the crude product in dioxane, adding hydrochloric acid, and stirring for reaction. After the reaction, water was added to dilute the reaction mixture, and the mixture was extracted with ether, and the organic phase was washed with a sodium hydrogencarbonate solution, a saturated saline solution and dried over anhydrous sodium sulfate. The crude product obtained by spin drying was recrystallized from 95% ethanol.
Synthesis of compound 8:
3-methoxy-4-hydroxyphenylacetic acid and Boc protected 2-bromoethylamine were reacted in triethylamine at room temperature for 3 hours, after which the solvent was removed by rotary evaporation. Then, dimethyladamantyl-2-amine and HATU were added thereto, and the reaction was stirred at 120 ℃ overnight. Diluting with water, extracting with ethyl acetate, drying, and spin-drying to obtain crude product. Dissolving the crude product in dioxane, adding hydrochloric acid, and stirring for reaction. After the reaction, water was added to dilute the reaction mixture, and the mixture was extracted with ether, and the organic phase was washed with a sodium hydrogencarbonate solution, a saturated saline solution and dried over anhydrous sodium sulfate. The crude product obtained by spin drying was recrystallized from 95% ethanol.
Synthesis of compound 9:
dissolving 3-methoxy-4-hydroxybenzylamine and adamantyl-2-carboxylic acid in acetonitrile, adding DCC (dicyclohexylcarbodiimide), and stirring for reaction for 1 hour. The extract was washed with dilute hydrochloric acid, saturated sodium bicarbonate solution and water, and dried over anhydrous sodium sulfate. And (5) purifying by column chromatography to obtain the compound 9.
Synthesis of compound 10:
reacting 3-methoxy-4-hydroxybenzylamine with phthalic anhydride under acetic acid, and stirring for 1 hour. The extract was washed with dilute hydrochloric acid, saturated sodium bicarbonate solution and water, and dried over anhydrous sodium sulfate. Purifying by column chromatography to obtain compound 10.
Synthesis of compound 11:
dissolving 3-methoxy-4-hydroxybenzylamine and 1-methyl-piperazinoic acid in acetonitrile, adding DCC (dicyclohexylcarbodiimide), and stirring for reaction for 1 hour. The extract was washed with dilute hydrochloric acid, saturated sodium bicarbonate solution and water, and dried over anhydrous sodium sulfate. And (5) purifying by column chromatography to obtain the compound 11.
Synthesis of compound 12:
dissolving 3-methoxy-4-hydroxybenzylamine and 1-phenyl-piperazinoic acid in acetonitrile, adding DCC (dicyclohexylcarbodiimide), and stirring for reaction for 1 hour. The extract was washed with dilute hydrochloric acid, saturated sodium bicarbonate solution and water, and dried over anhydrous sodium sulfate. And (5) purifying by column chromatography to obtain the compound 12.
(III) evaluation of A beta caused by potential activation of VR1 ion channel by TRPV1 agonist using in vitro assay1-42Production amount reducing effect
A group of existing TRPV1 agonists have been evaluated in the present invention, including OLDA, Anadamide, NE-28345, DA-5018, NE-19550, Olvanil, resiniferatoxin, Arvanil, AM404, PPAHV, NADA; these reagents are either purchased from commercial suppliers or obtained from trusted parties. The present invention tested novel compounds synthesized.
The present invention hypothesizes that these compounds may produce similar reductions in A β through activation of TRPV11-42The function of (1). These compounds were evaluated in preliminary studies by in vitro experiments to establish structure-activity relationships and lay the foundation for subsequent studies. In vitro experiments the cell line from human neuroblasts, SH-SY5Y, was stably transfected with human APP695swe and human TRPV 1. The cell line can produce a large amount of A beta and A beta1-42High levels of TRPV 1-regulated Ca2+Active window, expressing high levels of PKC epsilon. Experiments have shown that vanilloid receptor agonists reduce a β production.
1. Biological experiments
Establishment of cell lines: it is SH-SY5Y cells (SY5Y/VR1/SWAPP) obtained from human neuroblasts stably transfected with TRPV1 and APP65 swe.
(1) FLIPR assay for potency of TRPV1(VR1) agonists at VR1
The efficacy (EC50 value) of existing VR1 agonists and designed capsaicin derivatives is planned to be assessed using the fluorescence imaging microplate assay method (FLIPR).
FLIPR assays were performed on 96-well PLIRP plates (unplated clean plate Corning # 3603). 6 ten thousand SY5Y/VR1/SWAPP cells were spotted in advance on individual wells of a FLIPR plate 24 hours prior to FLIPR analysis. Staining loading buffer (50. mu.l SY5Y medium/2.5. mu.M probenecid, 5. mu.M Fluo-3, 0.1% berlonic) was added to the wells of the FLIPR plate and left at 37 ℃ for 1 hour and 15 minutes. Each well was washed twice with 75. mu.L of wash buffer (HBSS, containing 20mM HEPES, 2.5mM probenecid). Each compound was diluted into eleven different concentration series of assay buffers (Wash buffer + 0.25% BSA) ranging from 100. mu.M to 0.001. mu.M. The plates were loaded into a FLIPR analyzer and 70 μ l of different concentrations of compound were added. The FLIPR signal is captured and output in a statistical format. Data EC50 values were calculated using the Prism Graphpad analysis method.
(2) The effect of TRPV1 agonists on Α β production was analyzed using ELISA:
detection of A beta and A beta Using Sandwich ELISA1-42The total amount of (a). Briefly, the following samples were taken in sampling buffer (0.6% BSA, 8mM Na)2HPO4*7H2O,1.5mM NaH2PO4*H2O, 145mM NaCl, 0.05% thimerosal, 0.05% Triton X-405) diluted cell samples and A β1-42Standards (0-1000 pg/ml) (Bachem, Torrence, CA) were loaded individually onto immulon-4 microplates (CMS, Chicago, IL) and total A β protein was coated with 15 μ g/ml 266 capture antibody (for binding human A β 13-28 residues), A β 1-425 μ g/ml of 21F12 antibody (for binding to human A β 33-42 residues) was applied and incubated overnight at 4 ℃. The diluted samples were loaded onto 1.2 μ M Durapore 96 well titer plates (Millipore) and vacuum filtered using a multi-functional vacuum multi-layer filtration system. The supernatant was collected on a 96-well polypropylene microplate, and 100. mu.l of the sample was transferred to a 96-well immulon-4 microplate and coated with a capture antibody. The plates were incubated overnight at 4 ℃. The next day, plates (containing cell or tissue samples) were washed with PBS containing 0.05% Tween-20, pH 7.4, and then incubated for one hour at room temperature with 3D6 biotinylated indicator antibody (Harlan, Madison, Wis.) specific for A β 1-5 (1: 2000 diluted in 0.25% casein buffer). The above procedure was repeated to wash the plates, then Horse Radish Peroxidase (HRP) (0.25% casein buffer dilution 1: 1000, incubated for one hour at room temperature, after a final series of washes, TMB substrate (Pierce, Rockford, IL) was added at room temperature for 15 minutes, 2N sulfuric acid was added to stop the enzymatic reaction, and the reaction products were quantified by the difference in absorbance at 450nm and 650 nm.
(3) Quantitative western blot method PKC epsilon kinase activity:
to determine the activity of PKC epsilon kinase in cells treated with compounds, SY5Y/VR1/SWAPP cells were transiently transfected with RD α (a unique indoor cultured Pan-PKC substrate expressing strain). PKC epsilon kinase activity was determined by quantitative western blot method using indoor culture of anti-phosphor-RD alpha antibodies. The resulting dots were analyzed by densitometry. Densitometry data were normalized by densitometric analysis of β -actin from parallel loaded dots.
SY5Y/VR1/SWAPP cells transfected with RD α were treated with different concentrations of the designed compound. Study cells were harvested using a spatula and rinsed once with 1 × PBS. Resuspend the cell pellet in cytolytic buffer (10mM K)2HPO4,pH 7.2/1mM EDTA/5mM EGTA/10mM MgCl250mM beta-Glycerol phosphate/1 mM Na3VO4A solution was obtained in 2mM DTT/1% Triton X-100/1. mu.M Microcystin/Complex protease inhibitor (Roche)) in ice for 30 minutes. All lysates were centrifuged at 14000rpm for 30 minutes at 4 ℃ in a microcentrifuge, and the supernatants were separated. The supernatant was assayed for all protein concentrations using a BCA protein analyzer (PIERCE) according to the instrument instructions. Mu.g of all proteins from each sample were removed and separated on a 10% NuPAGEBI-Tris gel and transferred to a 0.2 μm nitrocellulose membrane (Novex, San Diego, Calif.) using a Hoefer Transblotter (Semiphor, San Francisco, Calif.). The nitrocellulose membrane was washed twice with TST (10mM Tris-HCl, 150mM NaCl, 0.1% Tween-20, pH 7.5) for 5 minutes each, blocked with 5% BSA-TST for 1 hour at room temperature on a shaking plate (Lab-Line), and then incubated with the original antibody overnight at 4 ℃ respectively. The next day, the film was washed 3 times with TST for 5 minutes each. HRP-conjugated secondary antibody was diluted 1: 2000 with 5% milk-TST and incubated with the membrane at room temperature for 1 hour with shaking. Finally, the membrane was washed with PBS for 5 minutes after washing with the method described above. The proteins on the membranes were detected separately by the chemiluminescence reality method (Amersham Life Science).
2. Pharmacological experiments
7-week old female Balb/c mice were used. The room in which the mice were housed was kept at 22 + -1 deg.C with a relative humidity of 60 + -10% for 12 hours with 12 hours light and 12 hours without light. Mice were euthanized with ketamine and xylazine.
(1) Pharmacokinetic studies in mice. 0.1ml of vanillin solution (in 5% DMSO in PBS) was administered intravenously to mice at a dose of 10 mg/kg. Mice can tolerate these DMSO when injected intravenously rapidly. Three mice were used for pharmacokinetic studies at each time point. Blood samples (. about.0.5 ml) were drawn from the orbital venous plexus using retro- orbital venipuncture 0, 2, 5, 10, 15, 30, 45 minutes and 1, 2, 4, 6, 8, 24 hours after intravenous injection. After 30 min, 4 and 8 hours, 3 mice were sacrificed at each cervical dislocation and their brains were removed. After the blood sample was taken, heparin was added at 7000rpm for 5 minutes, and plasma was separated after refrigerated centrifugation. Brain was homogenized by adding 3 volumes of water. Plasma and brain homogenate samples obtained from mice are extracted by a liquid-liquid extraction method, then the plasma vanillin concentration is analyzed and determined by an HPLC (high performance liquid chromatography) or LC/MS/MS (liquid chromatography/mass spectrometry) method, and a standard solution containing 0-2000 ng/ml of test compound is prepared by diluting 0.1ml of plasma samples containing vanillin with different concentrations.
(2) And (4) carrying out pharmacokinetic analysis. Pharmacokinetic models and parameter calculation methods were established using pharmacokinetic software from WinNonlin Standard Version 2.0(Pharsight, inc., Mountain View, CA). And selecting a suitable model on the basis of the lowest weighted square residual error, the lowest Schwartz Standard (SC), the lowest Akaike's information standard (AIC), the lowest Ses suitable for the parameters and the discreteness of the residual error. Calculating t by linear regression analysis of the final phase of the plasma-concentration-time curve1/2(elimination half-life). The AUC is divided by the dose to give the systemic Clearance (CL). The ratio of drug concentrations in brain and plasma will also be calculated.
(3) And (4) metabolic research. Metabolism of vanillin in liver microsomes. Mouse, rat, dog, monkey and human liver microsomes will be purchased from Vitro Technologies or Gentese (Woburn, MA). All culture fluids contained the following components at final concentrations (200 μ l): 1 XPBS, microsomes (1mg/ml), glucose 6-phosphate (10mM), glucose 6-phosphate dehydrogenase (2 units/ml), MgCl2(5 mM). The mixture was pre-incubated at 37 ℃ for 5 minutes. Metabolic processes were initiated by the addition of VR1 agonists at various concentrations. After 10 minutes of incubation, the reaction was stopped by adding 800. mu.l of acetone and thoroughly mixed using a vortex apparatus. HPLC extraction was performed as followsAnd separating the mixture. The contribution of cytochrome P450 to vanillin metabolism was determined by adding SKF-525A to the microsomes. To determine the non-enzymatic metabolism: microsomes will not be cultured in culture medium, eliminating the source of enzymes; removing NADPH generation system from the culture solution, and eliminating enzyme cofactor; the microparticles were inactivated by addition for 5 minutes at 90 ℃. The above culture method was also used to study the effect of liver cytosol. The reduction of vanillin precursors can be detected by HPLC or LC/MS/MS. The structure of the metabolite was determined by LC/MS/MS.
3. Experiment of drug
The powder injection preparation of the invention generally adopts a conventional freeze-drying method, takes water as a solvent, and comprises the following steps: adding excipient into vanillin receptor agonist, dissolving in water, adding active carbon, filtering, sterilizing, bottling, half-plugging, freeze drying, plugging, and capping. The excipient is selected from one or more of mannitol, hydrolyzed gelatin, glucose, lactose, dextran, etc. Each bottle contains 10-100 mg of vanillin receptor agonist.
The powder injection preparation of the invention can also adopt a spray drying method, takes water as a solvent, and comprises the following steps: dissolving vanillin receptor agonist with or without excipient (the above excipient), adding active carbon, filtering for sterilization, spray drying, packaging under sterile condition, and capping. Each bottle contains 10-100 mg of vanillin receptor agonist.
When the small injection is prepared, water for injection is used as a solvent for preparation, and a proper amount of auxiliary materials can be added, wherein the auxiliary materials are selected from one or more of ethanol, propylene glycol, glycerol, polyethylene glycol, benzyl benzoate and dimethylacetamide. Each of the herbal medicine composition contains 10-100 mg of vanillin receptor agonist.
The glucose infusion or sodium chloride infusion is prepared by taking water for injection as a solvent and adding a proper amount of glucose or sodium chloride, and also can be prepared by adding a proper amount of auxiliary materials, wherein the auxiliary materials are selected from one or more of ethanol, propylene glycol, glycerol, polyethylene glycol, benzyl benzoate and dimethylacetamide. Each bottle contains 10-100 mg of vanillin receptor agonist.
The invention can be prepared into tablets, capsules, granules, oral liquid and other oral preparations, and the auxiliary materials can be lactose, starch, dextrin, stearate and the like, and are prepared according to the conventional technology.
In the present invention, the above-described embodiments and the following examples are provided to better illustrate the present invention and are not intended to limit the scope of the present invention.
The present invention will be described in detail by examples.
Example 1 FLIPR assay method for determining the potency of capsaicin for VR1
First, the efficacy of capsaicin for VR1 was assessed using fluorescence imaging microplate assay (FLIPR) in established SHSY5Y cells stably transfected with TRPV1 and APP695swe (SY5Y/VR 1/swap) from a human neuroblastoma cell line (EC50 values, fig. 1). Capsaicin showed dose-dependent activity in SY5Y-hVR1 and CHO/hVR1/rVR1 cell lines.
As shown in FIG. 1 (capsaicin activity measured by FLIPR assay in SY5Y/VR1/SWAPP cells), blue, green, and red curves represent the response of SY5Y-hVR1, CHO/hVR1, and CHO/rVR1 cells, respectively, to different concentrations of capsaicin. CHO/hVR1 and CHO/rVR1 cell lines were used as controls. The table shows the EC50 values for capsaicin in the three cell lines calculated from the data measured in the dose-dependent experiments described above. FLIPR analysis was performed on a 96-well PLIRP plate (uncoated clean bottom plate Coming # 3603). 6 ten thousand SY5Y/VR11/SWAPP cells were spotted in advance on individual wells of a FLIPR plate 24 hours prior to FLIPR analysis. Staining loading buffer (50. mu. lSY5Y medium/2.5. mu.M probenecid, 5. mu.M Fluo-3, 0.1% Berlonic) was added to the wells of the FLIPR plate and left at 37 ℃ for 1 hour and 15 minutes. Each well was washed twice with 75. mu.L of wash buffer (HBSS, containing 20mM HEPES, 2.5mM probenecid). Each compound was diluted into eleven different concentration series of assay buffers (Wash buffer + 0.25% BSA) ranging from 100. mu.M to 0.001. mu.M. The plates were loaded into a FLIPR analyzer and 75. mu.l of different concentrations of compound were added. The FLIPR signal is captured and output in a statistical format. Data EC50 values were calculated using the Prism Graphpad analysis method.
Example 2 detection of capsaicin for A β by ELISA1-42Influence of generation
The following sandwich ELISA (enzyme linked immuno-labeling assay) showed that capsaicin-induced activation of the VR1 channel leads to A β activation1-42Production reduction (FIG. 2: detection of capsaicin for A.beta.by ELISA1-42The resulting effect). SY5Y/VR1/SWAPP cells were transfected with RD for 48hr, washed twice with PBS, and various concentrations of capsaicin were added to the medium after fresh medium was added (as shown in FIG. 2). After two hours, collecting the culture medium, and detecting Abeta by using an ELISA method; cells were collected for later PKC epsilon kinase activity assay. As shown in FIG. 2, capsaicin at a concentration of 0.1. mu.M or higher can greatly reduce A.beta.1-42And (4) generating.
Quantification of Abeta and Abeta Using Sandwich ELISA1-42. Briefly, the sampling buffer (0.6% BSA, 8mM Na) was used2HPO4*7H2O,1.5mM NaH2PO4*H2O, 145mM NaCl, 0.05% thimerosal, 0.05% Triton X-405) diluted cell samples and A β1-42Standards (0-1000 pg/ml) (Bachem, Torrence, CA) were loaded individually into immulon-4 microplates (CMS, Chicago, IL) and total A β protein was coated with 15 μ g/ml 266 capture antibody (for binding human A β 13-28 residues), A β 1-425 μ g/ml of 21F12 antibody (for binding to human A β 33-42 residues) was applied and incubated overnight at 4 ℃. The diluted samples were loaded onto 1.2 μ M Durapore 96 well titer plates (Millipore) and vacuum filtered using a multi-functional vacuum multi-layer filtration system. The supernatant was collected on a 96-well polypropylene microplate and 100. mu.l of the sample was transferred to a 96-well microplate coated with the capture antibody immulon-4. The plates were incubated overnight at 4 ℃. The next day, plates (containing cell or tissue samples) were washed with PBS containing 0.05% Tween-20, pH 7.4, followed by a biotin-labeled indicator antibody (Harlan, Madison, Wis.) specific for A.beta.1-5 (1: 2000 diluted in 0.25% casein buffer) at room temperature with 3D 6. The incubation was carried out for one hour,the above procedure was repeated to wash the plates, and horse radish peroxidase (Amersham Life Sciences, Arlington heights, IL) diluted 1: 1000 with 0.25% casein buffer dilution was added and incubated at room temperature for one hour. After a final series of washes, TMB substrate (Pierce, Rockford, IL) was added at room temperature and incubated for 15 minutes at room temperature. The enzymatic reaction was stopped by addition of 2N sulfuric acid. The reaction product was quantified by the difference in absorbance at 450nm and 650 nm.
Example 3 Western blot quantitative determination of PKC epsilon kinase Activity
Subsequent quantitative western blot assays showed that activation of the VR1 channel resulted in an increase in PKC epsilon kinase activity (see fig. 3: quantitative western blot assays for PKC epsilon kinase activity). Abeta in cell lysate1-42The significant reduction of PKC epsilon kinase activity was consistent with that measured by the phosphorescence-RD α quantitative Western blot method (PKC epsilon is the predominant PKC isomer expressed in SY5Y cell line). As shown in fig. 3, capsaicin above 0.1M significantly increased PKC kinase activity.
SY5Y/VR1/SWAPP cells were transiently transfected with RD α (a unique Pan-PKC substrate expressing strain). PKC epsilon kinase activity was determined by quantitative western blot method using anti-phosphor-RD alpha antibodies. The resulting dots were analyzed by densitometry. Densitometry data was normalized by densitometric analysis of β -actin from parallel loaded blots.
EXAMPLE 4 preparation of Vanillin receptor agonist powder
Taking 30g of vanillin receptor stimulant, adding 30g of dextran, adding 500ml of water for injection, and stirring to dissolve the vanillin receptor stimulant; adding water for injection to 2000ml, adding 3.0g of active carbon for injection, and fully stirring for 30 minutes; decarbonizing and filtering; filtering with 0.22 μm microporous membrane; filling into sterile penicillin bottles with 2ml per bottle, and half-rolling a stopper; freeze drying, and then plugging and capping.
EXAMPLE 5 preparation of Vanillin receptor agonist powder
Taking 60g of vanillin receptor stimulant, adding 500ml of water for injection, and stirring to dissolve the vanillin receptor stimulant; adding 1000ml of water for injection, adding 1g of active carbon for injection, and fully stirring for 30 minutes; decarbonizing and filtering; filtering with 0.22 μm microporous membrane; freeze drying to obtain sterile powder, and subpackaging into 1000 bottles.
EXAMPLE 6 preparation of Vanillin receptor agonist powder
Taking 40g of vanillin receptor stimulant, adding 50g of lactose and 100ml of water for injection, and stirring to dissolve the vanillin receptor stimulant; adding water for injection to 1000ml, adding 1.5g of active carbon for injection, and fully stirring for 30 minutes; decarbonizing and filtering; filtering with 0.22 μm microporous membrane; spray drying to obtain sterile powder, and subpackaging into 1000 bottles.
EXAMPLE 7 preparation of Vanillin receptor agonist powder
(1) Prescription
Semi-finished product of vanillin receptor agonist for injection 150g
Mannitol 400g
Adding 10000ml of water for injection
Made into 10000 bottles
(2) Preparation process
Weighing vanillin receptor agonist according to the prescription, adding into proper amount of water for injection, and stirring to dissolve; adding mannitol in a prescription amount, stirring to completely dissolve, and adding water for injection to the full amount; adding 0.1% of liquid amount of needle activated carbon, and fully stirring for 30 minutes; decarbonizing and filtering; filtering with 0.22 μm microporous membrane; filling and half-rolling stopper; freeze drying, and capping. 9735 bottles are prepared, and the yield is 97.35%.
EXAMPLE 8 preparation of a Vanillin receptor agonist ampoules
Taking 5g of vanillin receptor stimulant, adding 100ml of water for injection, and stirring to dissolve the vanillin receptor stimulant; adding water for injection to 1000ml, and filtering with 0.22 μm microporous membrane; subpackaging and encapsulating, 10ml per bottle, and sterilizing.
EXAMPLE 9 preparation of a Vanillin receptor agonist ampoules
Taking 10g of vanillin receptor stimulant, adding 30g of propylene glycol, adding 200ml of water for injection, and stirring to dissolve; adding water for injection to 1000ml, adding 1.5g of active carbon for injection, and fully stirring for 30 minutes; decarbonizing and filtering; filtering with 0.22 μm microporous membrane; subpackaging and encapsulating, 5ml per bottle, and sterilizing.
Example 10 preparation of glucose infusion solution containing vanilloid receptor agonist
Taking 2g of vanillin receptor stimulant, adding 10g of polyethylene glycol, adding 500g of glucose, adding 2000ml of water for injection, and stirring to dissolve the water for injection; adding water for injection to 5000 ml; filtering with 0.22 μm microporous membrane; packaging, bottling, and sterilizing, wherein each bottle contains 100 ml.
Example 11 preparation of glucose infusion solution containing vanilloid receptor agonist
Taking 2g of vanillin receptor stimulant, adding 250g of glucose, adding 1000ml of water for injection, and stirring to dissolve the vanillin receptor stimulant; adding water for injection to 5000 ml; filtering with 0.22 μm microporous membrane; subpackaging and encapsulating, 250ml per bottle, and sterilizing.
EXAMPLE 12 preparation of sodium chloride infusion solution as vanilloid receptor agonist
Taking 1g of vanillin receptor stimulant, adding 90g of sodium chloride, adding 1000ml of water for injection, and stirring to dissolve; adding water for injection to 10000 ml; filtering with 0.22 μm microporous membrane; subpackaging and encapsulating, 250ml per bottle, and sterilizing.
Example 13 preparation of vanilloid receptor agonist tablets
(1) Prescription
Vanillin receptor agonist 1000.0g
Microcrystalline cellulose 1170.0g
Pregelatinized starch 690.0g
Lactose 125.0g
Proper amount of 5% PVP absolute ethyl alcohol
Magnesium stearate 15.0g
Making into 10000 tablets
(2) Preparation process
The main medicine and the auxiliary materials are respectively weighed according to the prescription, the main medicine and the auxiliary materials are uniformly mixed according to an equivalent progressive method, the processes of soft material preparation, granulation, drying, granule straightening and the like are carried out according to the prescription process, a single-punch tablet machine and a 10.5mm shallow concave punch die are used for tabletting after the tablet weight is calculated, the hardness of a bare chip is controlled to be 5-7 kg, tablets 9698 are prepared, and the yield is 96.98%. Coating by adopting a rolling spraying method, wherein the coating process comprises the following steps:
preparing a coating solution: gastric soluble film coating material: 85G61235 from Shanghai Kalekang coating technology Co., Ltd
The coating process comprises the following steps: putting a bare chip (with the hardness of 5 kg-7 kg) to be coated into a coating pan, starting a stirring device and an air blast heating device, opening a spray gun to aim at 1/3 of a tablet bed to spray coating liquid for coating when the temperature of the bare chip rises to 40 ℃, controlling the temperature of the tablet bed to be 38-42 ℃, the gas pressure to be 6kg, the flow rate of the coating liquid to be 50mL/min, and the weight of a coating film to be 3% of that of the coating tablet.
Example 14 preparation of vanilloid receptor agonist tablets
Taking 100g of vanillin receptor agonist, 80g of microcrystalline cellulose, 15g of lactose and 60g of pregelatinized starch, sieving, uniformly mixing, preparing a soft material by using a proper amount of 10% PVP alcohol solution, granulating, drying, adding 3g of magnesium stearate, granulating, tabletting and preparing into 1000 tablets.
Example 15 preparation of vanilloid receptor agonist capsules
(1) Prescription
Vanillin receptor agonist 1000.0g
Microcrystalline cellulose 1000g
Sodium carboxymethyl starch 140g
Proper amount of absolute ethyl alcohol
Talcum powder 80g
Making into 10000 capsule
(2) The preparation process comprises the steps of respectively taking the vanillin receptor agonist of the raw material medicine and other auxiliary materials in the prescription, respectively sieving the raw material medicine and the other auxiliary materials in the prescription through a 100-mesh sieve, drying at 60 ℃, weighing the vanillin receptor agonist of the prescription, uniformly mixing the vanillin receptor agonist, the microcrystalline cellulose and the sodium carboxymethyl starch in an equivalent progressive method, preparing a soft material by using a proper amount of absolute ethyl alcohol, granulating through a 30-mesh sieve, drying at 50-60 ℃ for 2 hours, granulating through a 30-mesh sieve, adding the talcum powder and the sodium carboxymethyl starch in the prescription, and uniformly mixing.
Example 16 preparation of vanilloid receptor agonist capsules
Taking 100g of vanillin receptor agonist, 100g of microcrystalline cellulose, 14g of sodium carboxymethyl starch and 8g of talcum powder, respectively sieving the materials with a 100-mesh sieve, drying the materials at 60 ℃, taking the vanillin receptor agonist, the microcrystalline cellulose and the sodium carboxymethyl starch in equal quantity, gradually and uniformly mixing the vanillin receptor agonist, the microcrystalline cellulose and the sodium carboxymethyl starch, preparing a soft material by using a proper amount of absolute ethyl alcohol, granulating the mixture with a 30-mesh sieve, drying the mixture for 2 hours at 50-60 ℃, granulating the dried mixture with a 30-mesh sieve, adding the talcum powder and the sodium carboxymethyl starch in the formula, and uniformly mixing the talcum powder and the sodium.

Claims (35)

1. Application of vanillin receptor stimulant in preparation of anti-Alzheimer disease products.
2. Application of a vanillin receptor agonist composition in preparing an anti-Alzheimer disease product.
3. The use of the vanillin receptor agonist according to claim 1 or 2, wherein the anti-alzheimer product is a product for preventing, diagnosing, detecting, protecting, treating and studying alzheimer's disease and related diseases.
4. The use of the vanillin receptor agonist of claim 3, wherein the anti-Alzheimer's disease product comprises one or more of a drug, an agent, a food, or a beverage.
5. The use of the vanillin receptor agonist according to claim 1 or 2, wherein the vanillin receptor agonist is one or more selected from the group consisting of vanillin and derivatives thereof, capsaicin analogs and derivatives thereof.
6. The use of the vanillin receptor agonist of claim 5, wherein the vanillin and derivatives thereof include one or more of capsaicin, a compound of the capsaicin family, and derivatives thereof.
7. The use of the vanillin receptor agonist according to claim 6, wherein the vanillin and derivatives thereof have the following general chemical structure:
wherein,
R1is one of hydroxyl, alkyl, alkoxy, acyloxy, amine alkoxy, hydrogen, amino or halogen;
R2is one of alkoxy, hydrogen, hydroxyl, amino, alkyl, aliphatic amine or aromatic amine, amine alkoxy or acyloxy.
R3Is one of alkyl or substituted alkyl, alkenyl or substituted alkenyl, diterpene alkenyl, phenyl or substituted phenyl, adamantyl or substituted adamantyl with 5-23 carbon atoms, or piperazinyl or substituted piperazinyl with 5-23 carbon atoms;
n is one of 0, 1 or 2;
x is one of NHC (O), C (O) NH, C (O) O, NHC (O) NH, NHC (S) NH, or NH (O) S (O).
8. The use of the vanillin receptor agonist according to claim 7, wherein the alkyl group is one of a linear, branched, or cyclic alkyl group having 5 to 23 carbon atoms.
9. The use of the vanillin receptor agonist of claim 8, wherein the alkyl group is one of isobutyl, tert-butyl, sec-butyl, pentyl, tert-pentyl, hexyl, cyclopentyl, or cyclohexyl.
10. The use of the vanillin receptor agonist according to claim 8, wherein the alkyl group is one of a linear, branched, or cyclic alkyl group having 8 to 12 carbon atoms.
11. The use of the vanillin receptor agonist according to claim 7, wherein the alkyl group of the alkoxy group is one of a linear, branched or cyclic alkyl group having 1 to 6 carbon atoms.
12. The use of the vanillin receptor agonist of claim 11, wherein the alkyl of the alkoxy group is one of methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, sec-butyl, pentyl, tert-pentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
13. The use of the vanillin receptor agonist according to claim 11, wherein the alkyl group of the alkoxy group is one including an alkyl group having 1 to 3 carbon atoms.
14. The use of the vanillin receptor agonist according to claim 13, wherein the alkyl group of the alkoxy group is one of a methyl group and an ethyl group.
15. The use of the vanillin receptor agonist of claim 7, wherein the fatty amine is one of a substituted primary, secondary or tertiary aliphatic amine.
16. The use of the vanillin receptor agonist according to claim 15, wherein the substituted aliphatic primary, secondary or tertiary amine is one of methylamine, ethylamine, propylamine, isopropylamine, the four butylamine isomers, dimethylamine, diethylamine, diethanolamine, dipropylamine, diisopropylamine, di-N-butylamine, tetrahydropyrrole, piperidine, morpholine, 3, 5-dimethylmorpholine, piperazine, N-methylpiperazine, N-ethylpiperazine, N-isopropylpiperazine, 2-methylpiperazine, 3-methylpiperazine, 2, 6-dimethylpiperazine, trimethylamine, triethylamine or tripropylamine.
17. The use of the vanillin receptor agonist of claim 16, wherein the substituted aliphatic primary, secondary or tertiary amine is one of ammonia, methylamine, ethylamine, propylamine, diethylamine, diethanolamine, triethylamine, morpholine or N-methylpiperazine.
18. The use of the vanillin receptor agonist of claim 17, wherein the substituted aliphatic primary, secondary or tertiary amine is one comprising ammonia, methylamine, triethylamine or morpholine.
19. The use of the vanillin receptor agonist of claim 7, wherein the aromatic amine is one comprising a substituted aromatic primary, secondary or tertiary amine.
20. The use of the vanillin receptor agonist of claim 19, wherein the substituted aromatic primary, secondary or tertiary amine is one of aniline, substituted aniline, N-methylaniline, N-ethylaniline, pyridine, substituted pyridine, quinoline, substituted quinoline, isoquinoline, substituted isoquinoline, or quinine.
21. The use of the vanillin receptor agonist according to claim 20, wherein the substituted aromatic primary, secondary or tertiary amine is a substituted quinoline.
22. The use of a vanillin receptor agonist according to claim 21, wherein the substituted quinoline is (5-ethyl-1-aza-bicyclo [2.2.2] -oct-2-yl) -quinolin-4-yl-methanol.
23. The use of a vanillin receptor agonist according to claim 7, wherein n is 1.
24. The use of the vanillin receptor agonist according to claim 7, wherein R is1Is a hydroxyl group.
25. The use of the vanillin receptor agonist according to claim 7, wherein R is2Is an alkoxy group.
26. The use of the vanillin receptor agonist of claim 25, wherein R is2Is methoxy.
27. The use of the vanillin receptor agonist according to claim 7, wherein R is2Is composed of H, OH and OCH3、OCH2CH3、OCH2CH2CH3、OCH2CH2CH2CH3、OC6H5、CH2NH2、CH2CH2NH2、CH2CH2CH2NH2、OC(O)CH3、OC(O)CH2CH3、OC(O)CH2CH2CH3、OC(O)CH2CH2CH2CH3Or OC (O) C6H5One kind of (1).
28. The use of a vanillin receptor agonist according to claim 27, wherein R is2Is composed of OCH3、OCH2CH3、OCH2CH2CH3Or OCH2CH2CH2CH3One kind of (1).
29. The use of the vanillin receptor agonist according to claim 7, wherein R is3The alkyl group or substituted alkyl group has 7 to 18 carbon atoms, or one of an alkenyl group or substituted alkenyl group has 7 to 18 carbon atoms.
30. The use of the vanillin receptor agonist according to claim 7, wherein X is one of NHC (O), C (O) NH, C (O) O, NHC (O) O, NHC (O) NH, NHC (S) NH, or NH (O) S (O).
31. The use of the vanillin receptor agonist according to claim 7, wherein X is NHC (O).
32. The use of the vanillin receptor agonist of claim 5, wherein the capsaicin analog or derivative is a group of compounds that do not include an oxalyl group but have a structure-activity relationship similar to capsaicin, such structure-activity relationship being similar to capsaicin and including one or more of the following structure-activity relationships:
the appropriate length of the hydrophobic hydrocarbon chain is 8-18 carbons; the aromatic ring 3-methoxy plays an important but not essential role; the phenolic hydroxyl group is indispensable, and the most suitable position is the para position; the amide chain is indispensable; amino and cyclic through CH2The attachment is suitable;
② the compound has three hypothetical binding sites: a-vanillyl, B-amido, C-fatty chain; among them, the fatty end of the compound shows great flexibility and overall hydrophobicity to accommodate the C site;
a ring structure capable of introducing rigidity in the C region, comprising one or more of amantadine-2-amine, amantadine-2-ol, (5-ethyl-1-aza-bicyclo [2.2.2] -oct-2-yl) -quinolin-4-yl-methanol, 1-phenyl-piperazine, isoindole-1, 3-dione, or 2-methyl-2-propanol.
33. The use of the vanillin receptor agonist as defined in claim 32, wherein the capsaicin analog or derivative thereof has a chemical formula as follows:
34. the use of the vanillin receptor agonist of claim 6, wherein the capsaicin family compound and the derivative thereof are vanillyl-containing capsaicin family compounds and derivatives thereof.
35. The use of the vanillin receptor agonist of claim 34, wherein the capsaicin family compound and the derivatives thereof include one or more of dihydrocapsaicin, norhydrocapsaicin, homocapsaicin, homodihydrocapsaicin, homomodalipsin I, norhydrocapsaicin, nordihydrocapsaicin, cis-capsaicin, nonivamide, olvanil, NE-21610, N-oleyl-homovanillamide, Dong-a Pharmaceutical, or resiniferatoxin.
CN 200510027292 2005-06-29 2005-06-29 Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease Pending CN1736485A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN 200510027292 CN1736485A (en) 2005-06-29 2005-06-29 Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN 200510027292 CN1736485A (en) 2005-06-29 2005-06-29 Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease

Publications (1)

Publication Number Publication Date
CN1736485A true CN1736485A (en) 2006-02-22

Family

ID=36079557

Family Applications (1)

Application Number Title Priority Date Filing Date
CN 200510027292 Pending CN1736485A (en) 2005-06-29 2005-06-29 Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease

Country Status (1)

Country Link
CN (1) CN1736485A (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008174456A (en) * 2007-01-16 2008-07-31 Idemitsu Kosan Co Ltd Adamanthylcarbonyl group-containing aromatic diol, its derivative and methods for producing these
US8048908B2 (en) 2006-07-13 2011-11-01 High Point Pharmaceuticals, Llc 11β-hydroxysteroid dehydrogenase type 1 active compounds
US8053431B2 (en) 2005-11-01 2011-11-08 High Point Pharmaceuticals, Llc Pharmaceutical use of substituted amides
US8053447B2 (en) 2006-04-07 2011-11-08 High Point Pharmaceuticals, Llc 11β-hydroxysteroid dehydrogenase type 1 active compounds
US8138342B2 (en) 2004-10-12 2012-03-20 High Point Pharmacueticals, LLC 11β-hydroxysteroid dehydrogenase type 1 active spiro compounds
US8153798B2 (en) 2007-03-09 2012-04-10 High Point Pharmaceuticals, Llc Indole- and benzimidazole amides as hydroxysteroid dehydrogenase inhibitors
US8334305B2 (en) 2007-02-23 2012-12-18 High Point Pharmaceuticals, Llc N-adamantyl benzamides as inhibitors of 11-β-hydroxysteroid dehydrogenase
US8383683B2 (en) 2007-04-24 2013-02-26 High Point Pharmaceuticals, Llc Pharmaceutical use of substituted amides
US8383820B2 (en) 2007-02-23 2013-02-26 High Point Pharmaceuticals, Llc N-adamantyl benzamides as inhibitors of 11-β-hydroxysteroid dehydrogenase
CN103006631A (en) * 2012-12-28 2013-04-03 武汉华士特工业生物技术开发有限公司 Application of N-benzyl fatty acyl amide compound to preparation of neuroprotective drugs
US8927549B2 (en) 2008-11-21 2015-01-06 High Point Pharmaceuticals, Llc Adamantyl benzamide derivatives
US10159268B2 (en) 2013-02-08 2018-12-25 General Mills, Inc. Reduced sodium food products
TWI650121B (en) * 2016-01-18 2019-02-11 國立臺灣師範大學 Use of a compound for the preparation of a pharmaceutical composition for a Tau protein disease

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8138342B2 (en) 2004-10-12 2012-03-20 High Point Pharmacueticals, LLC 11β-hydroxysteroid dehydrogenase type 1 active spiro compounds
US8053431B2 (en) 2005-11-01 2011-11-08 High Point Pharmaceuticals, Llc Pharmaceutical use of substituted amides
US8053447B2 (en) 2006-04-07 2011-11-08 High Point Pharmaceuticals, Llc 11β-hydroxysteroid dehydrogenase type 1 active compounds
US8048908B2 (en) 2006-07-13 2011-11-01 High Point Pharmaceuticals, Llc 11β-hydroxysteroid dehydrogenase type 1 active compounds
JP2008174456A (en) * 2007-01-16 2008-07-31 Idemitsu Kosan Co Ltd Adamanthylcarbonyl group-containing aromatic diol, its derivative and methods for producing these
US8334305B2 (en) 2007-02-23 2012-12-18 High Point Pharmaceuticals, Llc N-adamantyl benzamides as inhibitors of 11-β-hydroxysteroid dehydrogenase
US8383820B2 (en) 2007-02-23 2013-02-26 High Point Pharmaceuticals, Llc N-adamantyl benzamides as inhibitors of 11-β-hydroxysteroid dehydrogenase
US8809540B2 (en) 2007-02-23 2014-08-19 High Point Pharmaceuticals, Llc N-adamantyl benzamides as inhibitors of 11-beta-hydroxysteroid dehydrogenase
US8153798B2 (en) 2007-03-09 2012-04-10 High Point Pharmaceuticals, Llc Indole- and benzimidazole amides as hydroxysteroid dehydrogenase inhibitors
US8383683B2 (en) 2007-04-24 2013-02-26 High Point Pharmaceuticals, Llc Pharmaceutical use of substituted amides
US8927549B2 (en) 2008-11-21 2015-01-06 High Point Pharmaceuticals, Llc Adamantyl benzamide derivatives
CN103006631A (en) * 2012-12-28 2013-04-03 武汉华士特工业生物技术开发有限公司 Application of N-benzyl fatty acyl amide compound to preparation of neuroprotective drugs
US10159268B2 (en) 2013-02-08 2018-12-25 General Mills, Inc. Reduced sodium food products
US11540539B2 (en) 2013-02-08 2023-01-03 General Mills, Inc. Reduced sodium food products
TWI650121B (en) * 2016-01-18 2019-02-11 國立臺灣師範大學 Use of a compound for the preparation of a pharmaceutical composition for a Tau protein disease

Similar Documents

Publication Publication Date Title
CN1736485A (en) Use of vanillin receptor agonist in preparation of product for resisting Alzheimer disease
CN1279040C (en) Tryasolyl tropane derivatives for therapy
CN1186332C (en) New thryoid receptor ligands and process II
CN1078889C (en) Non-peptide tachykinin receptor antagonists
JP2020503298A (en) Monocyclic OGA inhibitor compounds
CN1090188C (en) Benzonaphthyrindines as bronchial therapeutics
CN1913778A (en) Aminocyclopentyl pyridopyrazinone modulators of chemokine receptor activity
CN1450998A (en) Inhibitors of copper-containing amine oxidases
CN1533390A (en) Drugs containing triazaspiro [5,5] undecane derivatives as the active ingredient
CN1520290A (en) Carbocyclic hydrorazino inhibitors of copper-containing amine oxidases
CN1302207A (en) Combined therapy for bipolar disorders
CN1129942A (en) Aza spiro-compounds acting on the cholinergic system with muscarinic agonist activity
CN1329589A (en) N-aralkylaminotetralins as ligands for neuroptide YY5 receptor
CN1864675A (en) Use of naringenin and derivative in preparation of product for resisting cardiovascular and cerebrovascular disease
CN1270585A (en) Substituted 1,2,3,4-tetrahydronaphthalene derivatives
JP2012509879A (en) Adamantylbenzamide compounds
CN1281432A (en) Alpha-aryl-N-alkylnitrones and pharmaceutical compositions containing same
CN1918145A (en) Amino heterocyclic modulators of chemokine receptor activity
CN1036064C (en) Benzene derivatives having NGF production-promoting activity
CN1500085A (en) Lipid lowering biphenylcarboxamides
CN1968931A (en) Pyridine analogs as C5A antagonists
CN1054601A (en) Azabicyclo and azepine cyclic oximes and amine cholinergic agents thing and methods of treatment thereof
CN1921841A (en) Amines and amides for the treatment of diseases
CN1146562C (en) Thtrahydrothiopy 2,3-diazanaphthylaone derivatives as PDE inhibitors
CN1826115A (en) Substituted piperidine compounds and methods of their use

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
C02 Deemed withdrawal of patent application after publication (patent law 2001)
WD01 Invention patent application deemed withdrawn after publication