CN1893957A - Nitric oxide donating derivatives for the treatment of cardiovascular disorders - Google Patents

Nitric oxide donating derivatives for the treatment of cardiovascular disorders Download PDF

Info

Publication number
CN1893957A
CN1893957A CN 200480036099 CN200480036099A CN1893957A CN 1893957 A CN1893957 A CN 1893957A CN 200480036099 CN200480036099 CN 200480036099 CN 200480036099 A CN200480036099 A CN 200480036099A CN 1893957 A CN1893957 A CN 1893957A
Authority
CN
China
Prior art keywords
nitrooxy
chemical compound
ocor
patient
hydroxyl
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 200480036099
Other languages
Chinese (zh)
Inventor
N·黄
J·塔克
D·R·麦卡弗里
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.)
Resverlogix Corp
Original Assignee
Resverlogix Corp
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 Resverlogix Corp filed Critical Resverlogix Corp
Publication of CN1893957A publication Critical patent/CN1893957A/en
Pending legal-status Critical Current

Links

Abstract

Compounds for inducing the expression of ApoA1, comprising a donatable nitric oxide moiety and a free-radical scavenging moiety. Preferably, nitric oxide derivatives of stilbenes, polyphenols, flavonoids and isoflavonoids and methods of their use are provided for treating patients suffering from diseases including hypercholesterolemia, vascular oxidative stress and endothelial dysfunction.

Description

The nitric oxide donating derivatives of treatment cardiovascular disease
Invention field
The present invention relates to synthetic and use is applicable to the field of the flavone compound (flavonoids) that is added to food, medicine, health product (nutraceuticals) and derivant thereof and with this type of material treatment the method that needs individuality is arranged.
Background of invention
Cardiovascular disease is the generic term that is used to illustrate a class heart and angiopathy, comprises hypertension, coronary heart disease, cerebrovascular disease, peripheral vascular disease, heart failure, rheumatic heart disease, congenital heart disease and cardiomyopathy.The main cause of cardiovascular disease is that the lipid precipitation on atherosclerosis, the arterial wall is accumulated.High blood cholesterol levels is with that atherosclerotic risk takes place is closely related, therefore drops into huge medical research in the therapy of blood cholesterol levels falls in exploitation.
Atherosclerosis is relevant with endothelial function disturbance, endothelial function disturbance is the impaired disease of vascular system internal layer normal function wherein, except being multiple other cardiovascular disease, outside the major risk factors of for example angina pectoris, myocardial infarction and cerebrovascular disease, it still is atherosclerotic pathogenesis.The feature of endothelial function disturbance comprises that oxidisability blood vessel stress improves and vasoconstriction, and high blood cholesterol levels, and they all impel mutual acceleration generation cardiovascular disease.For the most effective prevention disease takes place, need to improve the therapeutic strategy of anti-multiple cardiovascular disease cause of disease risk factor.
Cholesterol metabolism
Because it is insoluble, cholesterol is lipid and the transhipment of proteinic complex by being called lipoprotein in blood.It is believed that low density lipoprotein, LDL (LDL) is responsible for cholesterol is transported to other tissue of health from liver, thereby becomes " the bad cholesterol " that is commonly called as.The LDL granule is transformed by intermediate density lipoprotein (IDL) (IDL), and intermediate density lipoprotein (IDL) itself generates by deglycerizin three acid esters from very low density lipoprotein (VLDL) (VLDL).VLDL is synthetic by triglyceride and several apolipoprotein in liver, then thus their direct secretion in blood flow.
It is believed that, high density lipoprotein (HDL) is the main carrier molecule that cholesterol is transported to liver from extrahepatic tissue, catabolism takes place in it in liver, eliminate in the process that is called reverse cholesterol transport (RCT) then, thereby HDL obtains the nickname of " good cholesterol ".In the elimination process that liver takes place, cholesterol is converted into bile acid, excretes then.
Treat the method for hyperlipemia at present
The medicine of the cholesterol reducing of approval provides the treatment benefit by a plurality of differences of attacking on the normal cholesterol metabolic pathway at present.The bile acid binding resin, for example colestyramine is adsorbed onto bile acid, excretes, and causing cholesterol to be converted into bile acid increases, and the result reduces blood cholesterol levels.Resin only reduces maximum 20% serum cholesterol, causes gastrointestinal side-effect, and can not with other medicines administration simultaneously because resin can in conjunction with, cause this type of other medicines to be drained.
It is synthetic that nicotinic acid suppresses lipoprotein, reduces the needed VLDL granule of preparation LDL and generate.When the administration of needs high concentration when increasing the HDL level, serious adverse, for example flushing take place.
It is believed that, fibrate (Fibrates), for example chlorine Bei Te and fenofibrate activate the transcription factor of peroxisome proliferation-activated receptors (PPAR) family that belongs to nuclear hormone receptor.These transcription factor raise and relate to the gene that HDL generates, and downward modulation relates to the gene that LDL generates.Fibrate is used for the treatment of hyperlipemia, because they partly reduce S-TG by reducing VLDL.But because the effectiveness that the xenogenesis character of its lipid reactant in the patient and shortage observe in patients with coronary heart disease, they ratify to be the medicine of hypercholesterolemia as yet in the U.S..The use of fibrate is also relevant with serious adverse, for example human primary gastrointestinal cancers, gallbladder disease and Gao Fei coronary heart disease death incidence rate.
Statins (Statins) is called HMG CoA reductase inhibitor again and reduces VLDL, LDL and IDL cholesterol by the synthetic rate-limiting enzyme of blocking-up liver cholesterol.Statins only increases the HDL level on a small quantity, and many livers are relevant with these medicines of use with the renal dysfunction side effect.
Ezetimibe is the medicine of first approval in the new treating cardiovascular disease of a class, and it works by cholesterol picked-up in the inhibition intestinal.Ezetimibe reduces LDL, but does not increase the HDL level basically, does not relate to synthetic cholesterol in the health, does not also relate to the cholesterol that circulates in blood flow or be present in atheromatous plaque.Also find to influence other chemical compound of cholesterol absorption, comprise bile acid bonding agent colestyramine and plant sterol.
Although developed these Therapeutic Method, increase blood HDL horizontal aspect progress not almost, the curative effect of all medicines of approval is subjected to the restriction of side effect and effectiveness at present.Therefore, need to improve Therapeutic Method improving HDL safely, thereby increase the speed of reverse cholesterol transport, to reduce the level of blood cholesterol levels.
Endothelial function disturbance and atherosclerosis
Endothelial function is impaired to occur in early stage that atherosclerosis takes place, in fact occur in detect lipid precipitation before.The characteristic symptoms of endothelial function disturbance is vasoconstriction, and causes hypertension, and it is other cardiovascular disease, and for example apoplexy and myocardial infarction knows risk factor.The endothelial function reduction of discovering the atherosclerotic is relevant with the bioavailability minimizing of nitrogen oxide (NO), and nitrogen oxide is the signaling molecule of induction of vascular diastole.
The bioavailability of NO reduces other mechanism that works that also activates in incidence of atherosclerosis.For example, knowing the NO inhibition is the platelet aggregation of the lipid plaque progression steps necessary of feature with the atherosclerosis.NO also is the important endogenous medium that suppresses the leukocyte adhesion, and the leukocyte adhesion is the key step of progression of atherosclerosis, and may be the result that the reaction of hyperlipidemia patient blood vessel oxidative stress increases.The adhesion leukocyte also increases the oxidant stress reaction by discharging a large amount of levels of reactive oxygen species.
The reaction of blood vessel oxidative stress increases and hypercholesterolemia is accredited as the reason that causes the NO bioavailability to reduce respectively.Another inducer that the oxidation increase also causes atherosclerotic lesions to form, the i.e. lipid peroxidation of free radical mediated.In a word, positive feedback loop will occur, wherein these three kinds of principal elements: each during hypercholesterolemia, the reaction of blood vessel oxidative stress and NO bioavailability reduce increases the degree and the pathology seriousness of other factors.
The nitrogen oxide therapy that is used for atherosclerosis-endothelial function disturbance
It is clinical that use provides the treatment pattern of the known compound of NO to be used for, attempts to break atherosclerosis-endothelial function disturbance circulation, but do not succeed.The exogenous NO that has confirmed confession NO drug release not only generates NO, and is generated as the nitrous acid anion excessively of active oxidizer, and it further increases the oxidative stress reaction.The peroxynitrite that produces by the NO donor and by the oxidative stress reaction increase cause reduce reaction subsequently to NO, may be the basis that in organic nitrates long-term treatment patient, produces well tolerable property record.It is believed that also need by the transition of mercaptan intermediate before they discharge NO for the NO medicine, the bioavailability of mercaptan significantly reduces under this type of oxidative stress reaction condition.
Antioxidant/nitrogen oxide conjoint therapy
Worsen attempting to alleviate, unite and offer patient's antioxidant and NO donor for the drug induced blood vessel oxidative stress reaction of NO.Some studies have shown that antioxidant and the associating of NO donor significantly increase the endothelium-dependent relaxation vasodilation of hypercholesterolemiapatients patients.
But, other people query that these results are subjected to not finding with this Therapeutic Method that the endothelium-dependent relaxation vasodilation improves, may be owing to due to the interior dosage of born of the same parents that is difficult to reach enough, increase the relevant fact with any delay progression of atherosclerosis or movable atherosclerotic's life-span far away with the treatment of NO donor.In addition, NO donor and NO donor/antioxidant conjoint therapy is not all directly at atherosclerosis-endothelial function disturbance circulation hypercholesterolemia aspect.
Neither very ideal method for NO and antioxidant and treatment based on the existing therapy associating of atherosclerotic hypercholesterolemia, because the medicine of approval can not effectively utilize the HDL increase cholesterol is transported out health effectively at present.
But people may suppose that preferred anti-oxidants and/or the combination of NO donor can guarantee that antioxidant is present in same position in the identical time with the NO donor in health, so that make antioxidant offset the potential oxidisability side effect of NO most effectively.The problem that satisfies this demand with several different pharmaceutical combinations of different rates of release and bioavailability is probably because in a single day NO discharges from donor molecule, the short and aggravation of its half-life in cellular environment.
Stilbene (stilbenes), polyhydric phenols and flavone compound antioxidant
Can breathe the levels of reactive oxygen species (ROS) that produces by normal cell is the main cause of oxidative damage in the health.One of method of the most effective anti-ROS is by providing and the bonded anti-oxidant compounds of ROS " removing " active group, thus prevent they with cell in key protein and DNA is unsuitable combines.The very effective anti-oxidant compounds that can eliminate ROS contains at least a phenol ring structure usually.The phenol ring is an active structure, and ROS and phenol ring can form covalent bond, thereby it eliminates the strong oxidation activity of ROS.Stilbene, polyhydric phenols and flavone compound all contain at least two phenol ring structures, thereby make them become potential effective anti-oxidants.
1 dose of the short ApoA of resveratrol (resveratrol), other stilbene and polyhydric phenols and flavone compound
Except that their antioxidant activity, stilbene, polyhydric phenols and flavone compound also have the activity that is used for the treatment of hypercholesterolemia.For example, the natural polyhydric phenols that the someone proposes to find in certain plants, promptly a kind of know 1, the 2-stilbene, resveratrol (3,4 ', the trans stilbene of 5-trihydroxy) is the basis that is called the epidemiologic observation of " French contradiction (French Paradox) ".This contradiction is meant does not consider comparable high fat diet, and the probability of observing French population trouble cardiovascular disease is 1/3 of a north American population.Compare with the red wine of north American population consumption, French contradiction is relevant with a large amount of red wines of French population consumption.Resveratrol is very abundant in the Radix seu Herba Tetrastigmatis Hypoglauci skin, therefore finds that its amount in red wine is remarkable, and exists hardly in Chinese liquor or other alcoholic beverage.
The mechanism of resveratrol minimizing cardiovascular disease generation probability is still arguement and wants when many exercise questions the hypothesis of several competitions is arranged.Confirmed that resveratrol is an effective anti-oxidants, it shows and causes low LDL granule levels of peroxide and suppress atherosclerosis subsequently forming.Resveratrol also is inferred as the inhibitor of leukocyte adhesion and platelet aggregation.In addition and since described it regulate the ability of P21 and p53 activity level, studying the probability of resveratrol as potential anticancer therapy medicine.
Resveratrol has been accredited as anti-inflammatory drug, and the mechanism of proposition comprises that inhibition cyclo-oxygenase-1 enzyme is (referring to United States Patent (USP) 6,541,045; J Nat Prod.1993 such as Jayatilake October; 56 (10): 1805-10; United States Patent (USP) 6,414,037) and Profilin kinases (U.S. Patent application 0030171429).Therefore, resveratrol may have as analgesics, antipyretic curative use treatment of arthritis, asthma, psoriasis, gastroenteropathy, oculopathy, pneumonia disease, cancer or the treatment potentiality with angiopathy, central nervous system disorder and antibacterial, fungus and the relevant inflammation of viral infection.
In recent years, resveratrol is described as activating the chemical compound of sirtuin, prompting causes reducing p53 by directly interacting the growth life-span with SirT1.Also known resveratrol antagonism aromatic hydrocarbon receptor, excited estrogen receptor and being described as by activating ERK 1/2 approach and active by increasing the active mediation of transcription factor egr-1.
More in recent years, we confirm that resveratrol has the ability that ApoA 1 is transcribed that increases, and infers by the mediation of the nucleotide sequence S site in the promoter region of ApoA-1 gene (PCT/CA 03/01220).
The sequence A GCCCCCGC that will find in the S site is described as " Egr-1 response element " consensus sequence.This primitive is included in the leap-196 of people APO AI promoter, and (Kilbourne etc. 1995, JBC, 270 (12): 7004-10) to-174 nucleotide.Be not subjected to any concrete theory constraint, this AGCCCCCGC element of propose finding to be included in the S site is that resveratrol passes through it and mediates its active sequence, but this does not get rid of the element of other potential demand.
It is believed that, when being connected with allogeneic promoter, comprising the nucleotide sequence in S site or any approximately 8 adjacent bases of AGCCCCCGC element and play the enhancer element effect with the expression of adjusting reporter gene.
The shortcoming that stilbene, flavone compound and the treatment of other polyhydric phenols are used
Unfortunately, for example resveratrol and other polyhydric phenols and flavone compound have problem to use stilbene as medicine.
Can not consume resveratrol resource the abundantest and that can utilize for consumer every day in a large number, i.e. red wine is because the illeffects of excessive alcohol consumption has many records.In other words, when not having ethanol, the effect of resveratrol may be better or safer.
Can prepare many stilbene, polyhydric phenols and flavone compounds with potential useful quality, they can not naturally synthesize, and describe as yet or preparation for test.Before people's clinical research test, must be at this compounds of prepared in laboratory, test is to prove useful therapeutic activity in measuring in suitable external and body.
The stilbene of known many biogenetic derivations, polyhydric phenols and flavone compound are because they pass through phytosynthesis usually.Tested the potential beneficial characteristics of many these chemical compounds, for example they known in antioxidation in vitro ability, deduction their anticancer effectiveness and they are to the apparent beneficial effect of cardiovascular disease.Though this compounds is done some research, and the result is very indeterminate, and still contradiction sometimes.For example, the discovery of people's clinical research also must the proof clear and definite evidence useful to main clinical endpoint, for example cardiovascular event that atheromatous plaque is big or small or minimizing is for example had a heart attack.In the situation of in some zooscopy, finding, with research and the people's result of study onrelevant of for example rabbit or rat.For example, give naringenin (as an example of the many flavone compound components that give to find in mandarin orange (Citrus) juice) increase HDL though in people's research, observe, but LDL or triglyceride there is not influence, but find when giving the rabbit naringenin, reduce LDL, but HDL is not had influence.
In addition, the flavone compound that does not have clinical research to describe to be used for the treatment of people's cardiovascular disease so far naringenin or the stilbene suitable dose of resveratrol or other polyhydric phenols for example for example.
Chemical compound with blocking group can experience longer serum half-life, increase and render a service, reduce toxicity and improve therapeutic effect
As medicine have need individual chemical compound usually before drainage in health metabolism be multiple metabolite.This metabolite usually toxicity, render a service with reside in serum in time aspect different with parent compound.For chemical compound lot, metabolite is effective not as parent compound, and toxicity may be bigger.
In the treatment chemical compound that the metabolism external source gives, multiple different variation may take place, for example add various chemical parts or remove crucial group.Occurring in an intravital metabolic response is to remove hydroxyl.From having flavone compound, 1,2-stilbene and other polyphenol compound, comprise in the chemical compound of mother nucleus structure of nitric oxide donating derivatives of The compounds of this invention and remove hydroxyl, can significantly reduce the ability that just influencing of these chemical compounds, because aforementioned hydroxyl is the major part of the active site of this quasi-molecule to cholesterol metabolism.Therefore, if thereby utilize some protection hydroxyl to increase the mechanism that chemical compound resides in the time in the health to reduce metabolic rate, help improving the beneficial effect that gives The compounds of this invention.
A kind of protection mechanism that is usually used in the laboratory chemical reaction is to use blocking group, and it is connected with the unstable chemical group of macromolecular easy modification, so that prevent unstable group degeneration or lose.Blocking group can be removed later on to recover initial molecule, do not changed any covalent bond in the whole molecule.Can use similar protection form to the predetermined chemical compound that gives the patient, the reaction of bioactive molecule takes place to reformulate in the wherein known health probably.The speed that the may command blocking group discharges from molecule is to influence the rate of release of medicine.The purpose of this invention is to provide half-life prolongation in vivo and/or postpone excretory active compound.
There are the needs that improve the cardiovascular disease therapy
In view of the above, clearly, need the exploitation can be safely and reduce blood cholesterol levels effectively, reduce endothelial function disturbance simultaneously and reduce the improvement therapy and the chemical compound of the reaction of blood vessel oxidative stress.
Summary of the invention
One aspect of the present invention provides all kinds of noval chemical compounds, and these noval chemical compounds have supplies with nitrogen oxide and the ability of co (co-localized) with the antioxidant molecule release of removing free radical simultaneously; And with their the treatment method.These noval chemical compounds have the ability of inducing ApoAl to express simultaneously, and thereby increase blood HDL level and reduce blood cholesterol levels.In addition, these chemical compounds have other useful activity, comprise the activity level that suppresses HMG-CoA reductase, increase PPAR activity, inhibition ACAT, increase ABCA-1 activity and reduce blood LDL and triglyceride.The associating multiple action of these chemical compounds can be used for reducing endothelial function disturbance, reduces the reaction of blood vessel oxidative stress and reduces hyperlipemia, thereby treatment cardiovascular disease, for example atherosclerosis, hypertension, coronary artery disease, cerebrovascular disease etc.
According to various aspects of the present invention and principle, provide following chemical compound.
The stilbene chemical compound that contains following structure:
Figure A20048003609900171
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate (glucoronidate) [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900181
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2And wherein X can be single, double or triple bond.
The flavone compound that contains following structure:
Figure A20048003609900182
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10, R 13And R 14Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10Or R 13Or R 14In at least one is nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be O, CR 13Or NR 13
Y can be the ketone that CO[still keeps 6 atom ring structures], CR 14Or NR 14With
Z can be singly-bound or two key.
The isoflavonoid (isoflavonoid) that contains following structure:
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10, R 13And R 14Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10Or R 13Or R 14In at least one is nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be O, CR 13Or NR 13
Y can be the ketone that CO[still keeps 6 atom ring structures], CR 14Or NR 14With
Z can be singly-bound or two key.
Chalcone (chalcone) chemical compound that contains following structure:
Figure A20048003609900201
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10And R 13Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10Or R 13In at least one is nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900202
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be singly-bound or two key;
Y can be singly-bound or two key; With
Z can be CO[ketone], CR 13Or NR 13
If condition is X and Y not all is that two keys and Z are CO, then Y is not two keys.
The polyphenol compound that contains following structure:
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900212
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2With
X can be C, S, (CO), SO, AKA ketone, (SO 2) N, (CO) C, (CO) N, (CO) O, C-N[singly-bound], the two keys of C=N[], C-O, N-O, N-N[singly-bound] or the two keys of N=N[].
In addition, also provide treatment the patient and method cardiovascular, cholesterol or lipid diseases associated, this method comprises any aforesaid compound that needs the patient treatment effective dose for the treatment of.Another kind of preferably be used to induce patient's ApoA1 to express, provide the Therapeutic Method of antioxidant activity to comprise simultaneously and give described patient any aforesaid compound.The method that other the present invention preferably is used to reduce total serum cholesterol in patients also comprises and gives described patient any aforesaid compound.
In addition, the method of treatment or prevention patient's presenile dementia, diabetes, obesity, ischemic damage and reperfusion damage, congestive heart failure and relevant disease also is provided, and this method comprises any aforesaid compound of the patient treatment effective dose that needs treatment.Low serum hdl level increases relevant (2003 J.Gerontol ABiol.Sci.Med.Sci.58 (9) such as Suryadevara: M859-61) with the presenile dementia risk.Find The compounds of this invention adjusting PPAR gamma activity; Relevant (2004 Diabetes such as Ferre, the 53 [1:S43-50 of PPAR γ dysfunction with diabetes, obesity, ischemic damage and reperfusion damage, congestive heart failure and diseases related; Yue 2003 Drugs Today (Bare) 39 (12): 949-60).
The present invention and optimal mode describe in detail
According to principle of the present invention and each side, provide detailed description about the method, chemical compound and the description latent effect mechanism that supply NO analogue treatment cardiovascular disease of using stilbene, polyhydric phenols and flavone compound and derivant thereof.Understand the potential mechanism of action and can cause increasing exploitation derivant and the analog with the further raising of treatment benefit more, they also within the scope of the invention.
The morbidity of many factor affecting cardiovascular disease is clearly arranged.Lack three main aspects that disease takes place in treatment simultaneously in the modern medicines, i.e. blood vessel oxidative stress reaction increases, the bioavailability of nitrogen oxide reduces and the method for hypercholesterolemia also is conspicuous.Therefore, the present invention new have antioxidant activity, nitrogen oxide and discharges active and induce the molecule of reverse cholesterol transport ability by providing single, and the method at all three factors is described in detail in detail simultaneously.When discharging according to nitrogen oxide, oxidation resistance and nitrogen oxide are supplied with the simultaneous fact, make that The compounds of this invention and Therapeutic Method are more effective, therefore especially preferred they.The invention provides with following material have any kind favourable that be connected and still keep the invention provides active for the NO part: form the present invention and expect 1 of chemical compound parent nucleus, the almost any part of 2-stilbene, other polyhydric phenols and flavone compound, or keep antioxidant properties and induce ApoA 1 transcriptional capability or have any derivant that increases reverse cholesterol transport or reduce active this compounds of serum cholesterol, or even antioxidant is arranged and reduce any chemical compound of serum cholesterol ability.
Chemical compound provided by the invention comprises having when giving the patient, can discharge the analog of resveratrol, other stilbene, other polyhydric phenols and the flavone compound of the coupling part of nitrogen oxide.This compounds include but not limited to resveratrol, other 1, the analog of 2-stilbene, other polyhydric phenols and flavone compound, wherein nitric oxide donating partly belongs to organic nitrates, alkoxyl nitrate, diazeniumdiolate, sulfo-nitrooxy type chemical constitutions such as (thionitroxy).
Implementing the present invention does not need understanding to change The compounds of this invention cutter reason really by it.Mechanism disclosed herein is nonrestrictive, only is used for describing better the present invention.Though bound by theory not it is believed that, because the activity of forming by at least one aromatic ring structure and must parent nucleus, and have the molecular structure of at least one hydroxyl on the aromatic ring, resveratrol produces aforementioned effect.The resveratrol of natural generation itself especially is made up of two aromatic rings, and two hydroxyls are positioned at 3 and 5 an of ring, and a hydroxyl is positioned at 4 of another ring ' position, and two aromatic rings connect by two carbon atoms, between two carbon atoms two keys are arranged.It is believed that, other chemical compound of this big class, described class is to contain at least one aromatic ring structure, and has those chemical compounds of a hydroxyl on the ring at least, has the result identical with generation with those identical abilities of the resveratrol of enumerating.
Therefore, the stilbene that contains two aromatic rings that connect by two carbon atoms; Other polyhydric phenols for example contains those polyhydric phenols of two or more preferred two aromatic rings that connect by 1,2 or 3 atom, and described atom independently is selected from nitrogen, carbon, oxygen and sulfur, they can or cannot by side group for example the oxygen of ketone independently replace; And flavone compound, such as but not limited to naturally occurring flavone compound, such as but not limited to naringenin, Quercetin, piceatannol, butein, fisetin, isoliquiritigenin and hesperetin (hesperitin), be all chemical compounds with those character that are similar to described resveratrol.Found that, when being used for prevention or treatment disease, obstacle or disease, especially but not limited to regulating relevant those diseases, obstacle or disease with cholesterol, cardiovascular disease, hypertension, oxidative damage, unusual lipidemia, ApoA 1 or apoB, or changing or the metabolic others of cholesterol regulating, for example suppress HMG CoA reductase, increase PPAR activity, inhibition ACAT, increase ABCA-1 activity, rising HDL or reduce LDL or triglyceride, can think that any of these chemical compound and resveratrol function are interchangeable.For example at United States Patent (USP) 5,877, illustrated in 208,6,455,577,5,763,414,5,792,461,6,165,984 and 6,133,241 not contain the activity that has potential reduction serum cholesterol with the flavone compound that nitric oxide donating partly is connected.
Similarly, transcribe from the AGCCCCCGC element regulation from the S site when being used for, maybe ought be used to suppress leukocyte adhesion or platelet aggregation, or when suppressing COX-1, can think that such any stilbene, other polyhydric phenols and flavone compound and resveratrol function are interchangeable.This does not mean all chemical compounds on each top of these functions or ability, or the activity level aspect of toxicity in vivo or effectiveness or bioavailability is identical.In the process of simple experiment, do not need over-drastic experiment by the easy operation of those skilled in the art, these chemical compounds proof wherein some chemical compound provides and is equivalent to ability or the function that other chemical compound improves, therefore than other chemical compound preferably as medicine.
Also known phenolic hydroxyl group, for example those phenolic hydroxyl groups of finding in the improved alkali cpd of the present invention are easy to impel excretory glucuronic acid esterification (glucoronidation) and sulfuric acid esterification.By blocking-up phenolic hydroxyl group and other chemical group, for example nitrate (is called organic nitrates or ONO again 2) base, alkoxyl nitrooxy or oppositely the ester nitrooxy react the protection that stops these reactions further prolong molecule in health half-life and postpone to drain.
As an example, resveratrol contained three infers that important and therapeutic activity hydroxyl can be by (being called nitrooxy or ONO again with nitrate 2, be called " nitre oxygen base " once in a while, but it not should with NO 2Obscure), the alkoxyl nitrooxy or oppositely the ester nitrooxy replace and protect, and in vivo, pass them in time and reformulated the resveratrol reactive compound by the hydroxyl displacement.Because within a certain period of time, the nitric oxide donating group is at every turn by the displacement of hydroxyl, and the resveratrol molecule that contains one or two nitric oxide donating group still has the part activity, and therefore resveratrol in vivo prolongs active effective half-life.This strategy also allows to use with respect to the resveratrol parent compound of the hydroxylating form resveratrol of the nitrate form of low dosage more, and the resveratrol of nitrate form is less to the side effect that the patient causes then.Obviously, this method is also effective to other stilbene, other polyhydric phenols and the flavone compound of the present invention's expectation, also comprises one or more hydroxyls that can form molecular activity position major part because expect them.
The invention provides above-mentioned 1, synthetic, the compositions of the NItroxyderivatives of chemical compound and processing method beyond 2-stilbene, polyhydric phenols and the flavone compound, wherein the described chemical compound by its synthesizing nitryl oxygen radical derivative contains aromatic ring or hetero-aromatic ring, one or more hydroxyl and known adjusting serum cholesterol level.An example that contains this compounds of aromatic ring or hetero-aromatic ring, one or more hydroxyl and known adjusting serum cholesterol level comprises HMG CoA reductase inhibitor, is called statins again.The invention provides the NItroxyderivatives of commercially available statins, commercially available statins comprises atorvastatin, lovastatin, pravastatin, simvastatin, fluvastatin, cerivastatin and rosuvastatin.Two kinds of other chemical compounds that contain aromatic ring or hetero-aromatic ring, one or more hydroxyl and known adjusting serum cholesterol level in this description scope are ezetimibe and nicotinic acid.Therefore, the present invention also provides the NItroxyderivatives of ezetimibe and nicotinic acid.
The nitric oxide donating derivatives of synthetic stilbene, polyhydric phenols, flavone compound, statins and ezetimibe
Use known method, for example wherein the nitrooxy Hakimelahi method that replaces hydroxyl on the parent molecule can (be called nitrooxy, nitrate, ONO again with organic nitrates 2, be called " nitre oxygen base " once in a while, but it not should with NO 2Obscure) base is added to (1984.Helv.Chim.Acta.67:906-915 such as Hakimelahi) on the chemical compound.
Method with explanation in the United States Patent (USP) 5,861,426 for example can be added to the alkoxyl nitrooxy on the chemical compound.Can pass through the whole bag of tricks, comprise the synthetic Diazeniumdolates of method of explanation in the United States Patent (USP) 4,954,526,5,039,705,5,155,137,5,405,919 and 6,232,336 for example, all these patents are attached to herein by reference.
The nitric oxide donating part preferably can be passed through covalent bond or ionic bond and 1, for example resveratrol, polyhydric phenols or flavone compound are for example described in naringenin or the present invention and other chemical compound that provides for the 2-stilbene, and for example statins member or derivatives thereof or analog connect.Preferred nitric oxide donating part or each several part connect by one or more covalent bonds.The nitric oxide donating part can advantageously be connected with any part of molecule.In one embodiment, nitric oxide donating has partly been replaced one or more hydroxyls.In a preferred embodiment, be that the organic nitrates base replaces hydroxyl.In another preferred embodiment, be that the organic nitrates base that is connected with ester or reverse ester replaces hydroxyl.In another preferred embodiment, each nitric oxide donating partly has been provided by for example resveratrol, polyhydric phenols or flavone compound for example all hydroxyl or derivatives thereofs of any member of statins or all hydroxyls of analog of naringenin or of the present invention and other chemical compound of providing for example of stilbene.
For all chemical compounds of the present invention, also expect and provide by fluorion, chloride ion, bromide ion, CF 3Group, CCl 3Group, CBr 3, 1-18 carbon atom alkyl chain of the optional optional side chain that replaces or the optional optional side chain that replaces the hydroxyl of 1-18 carbon atom alkoxy chain replacement, so modify parent compound and be known increase medicine stability and do not change the common practice of the mechanism of action, those skilled in the art can easily finish.
For all The compounds of this invention, also expect and provide the acetyl derivatives of chemical compound, so modify parent compound and be the beneficial effect of known raising medicine and do not change the common practice of the mechanism of action, those skilled in the art can easily finish.Acetyl derivatives comprises ester, reverse ester, has the ester that connects nitric oxide donating part (including but not limited to nitrooxy) and is connected the partly reverse ester of (including but not limited to nitrooxy) of nitric oxide donating.
For all The compounds of this invention, also expect and provide the Phosphation derivant of chemical compound, so modify parent compound and be the beneficial effect of known raising medicine and do not change the common practice of the mechanism of action, those skilled in the art can easily finish.
Expect also that herein the present invention expects the glucuronic acid esterification derivant of chemical compound, because the glucuronic acid esterification is the spontaneous generation process as stilbene, other polyhydric phenols and a flavone compound metabolism part in the body.In case offer the patient, most The compounds of this invention can be modified in vivo, therefore can be present in the body with the form of glucuronic acid esterification.Therefore, before administration, chemical compound function or the treatment effectiveness that research records in the body is not got rid of in puting together of glucuronic acid and The compounds of this invention.Therefore, think that the function of the The compounds of this invention that is connected with other sugar moieties can be compared with parent compound, therefore the invention provides them.End user's hepatomicrosome (2002.Drug Metab.Disp.30 (5) such as Otake: 576-581), can realize the glucuronic acid esterification that makes any stilbene, polyhydric phenols or flavone compound derivant that the present invention expects in the Otake method for example.
Similarly, also expect the Sulfation derivant of desired compounds of the present invention herein, because Sulfation is the spontaneous generation process as stilbene, other polyhydric phenols and a flavone compound metabolism part in the body.In case offer the patient, some The compounds of this invention can be modified in vivo, therefore can be present in the body with the form of Sulfation.Therefore, Sulfation is not got rid of chemical compound function or the treatment effectiveness that research records in the body.Therefore, think and to compare with parent compound, therefore the invention provides them through the function of the The compounds of this invention of sulfuric acid esterification.Ion-air of available Varin (ion-air) extracting process 1987.Anal.Biochem.161:176-180 such as () Varin for example, that realizes the present invention's expectation makes any stilbene, polyhydric phenols or flavone compound derivant Sulfation.
The present invention also provides the salt of the described chemical compound herein that comprises those salt that are preferred for pharmaceutical preparation.
Desired compounds of the present invention
For clearly, furnishing an explanation property of chemical compound chemical constitution provided by the invention, but this can not be limited in following compounds with the scope of the invention.When using term " nitrooxy ", it represents itrate group-ONO 2When using term " hydroxyl ", it represents group-OH.When using term " oppositely ester ", it represents group
Wherein the O-key is connected with the parent compound of flavone compound, stilbene or polyhydric phenols structure,
And R is C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, optional for side chain with can have one or more by S, N or the metathetical C atom of O.
When using term " oppositely ester nitrooxy ", it represents group
Figure A20048003609900282
Wherein the O-key is connected with the parent compound of flavone compound, stilbene or polyhydric phenols structure,
And R is C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, optional for side chain with can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The invention provides chemical compound with stilbene universal architecture:
Figure A20048003609900283
It can further be divided into following structure:
Figure A20048003609900291
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900301
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides following universal architecture chemical compound:
Figure A20048003609900311
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900312
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, optionally be side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
And X and Y can be C, N, O independently separately, and condition is that then another is not C if any of X or Y is C.
The present invention also provides following universal architecture chemical compound:
Figure A20048003609900321
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900322
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides the chemical compound with polyhydric phenols universal architecture:
Figure A20048003609900331
It can be further divided into following structure:
Wherein
X is C or S,
And R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900333
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides the chemical compound with flavone compound universal architecture:
Figure A20048003609900341
It can be further divided into following structure:
Figure A20048003609900351
The present invention also provides the chemical compound with isoflavonoid universal architecture:
Figure A20048003609900371
It can be further divided into following structure:
Figure A20048003609900372
Figure A20048003609900381
Figure A20048003609900391
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10, R 11, R 12, R 15And R 16Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 13, R 14, OR 13, OR 14, OCOR 13, OCOR 14, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 12Or R 15Or R 16In at least one is nitrooxy, R 14, OR 14Or OCOR 14With
Wherein OCOR represents
Figure A20048003609900392
And R is R 13Or R 14,
R wherein 13Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 14Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be O, CR 15Or NR 15
Y can be the ketone that CO[still keeps 6 atom ring structures], CR 16Or NR 16With
Z can be singly-bound or two key.
The present invention also provides the chemical compound with chalcone (chalcone) universal architecture:
Figure A20048003609900401
Its some structure is represented by following structure:
Figure A20048003609900402
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10And R 11Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 13, R 12, OR 13, OR 12, OCOR 13, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 11In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900421
And R is R 12Or R 13,
R wherein 13Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2With
Wherein
X can be singly-bound or two key;
Y can be singly-bound or two key; With
Z can be CO[ketone], CR 11Or NR 11
Other of the cholesterol reducing chemical compound that provides among the present invention comprises for the NO derivant:
The present invention also provides following general formula compound
Figure A20048003609900422
Wherein
R 1, R 2, R 3, R 4Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 4In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900431
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides chemical compound
Figure A20048003609900432
Wherein
R 1Be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900441
And R is R 12,
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides chemical compound
Figure A20048003609900442
Wherein
R 1Be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 12,
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides following general formula compound
Wherein
R 1, R 2, R 3Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 3In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides following general formula compound
Figure A20048003609900461
Wherein
R 1, R 2, R 3Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 3In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides following general formula compound
Figure A20048003609900471
Wherein
R 1, R 2, R 3Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 3In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900472
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides following general formula compound
Figure A20048003609900481
Wherein
R 1, R 2, R 3Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 3In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900482
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides following general formula compound
Figure A20048003609900491
Wherein
R 1, R 2Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 2In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900492
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The present invention also provides chemical compound
Wherein
R 1Be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A20048003609900502
And R is R 12,
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
The method for the NO derivant of synthetic 1,2-stilbene, polyhydric phenols and flavone compound
To those skilled in the art, there be for example for example naringenin or other antioxidant, serum cholesterol-lowering or to activate the reverse cholesterol transport or the nitric oxide donating analog of high density lipoprotein increasing chemical compound or the method for derivant be conspicuous of resveratrol, polyhydric phenols or flavone compound of many synthetic stilbene.Although known method is arranged, this compounds was never described in the past or was synthesized.Preferred this compounds is and the bonded stilbene of nitric oxide donating part for example naringenin or other antioxidant, serum cholesterol-lowering or activate the analog or the derivant of the chemical compound of reverse cholesterol transport or high density lipoprotein increasing of resveratrol, polyhydric phenols or flavone compound for example.Most preferably this compounds is to use one or more ONO 2The l of group (being called itrate group, organic nitrates base or nitrooxy again) alternative precursor chemical compound hydroxyl, 2-stilbene be for example naringenin or other antioxidant, serum cholesterol-lowering or activate the analog or the derivant of the chemical compound of reverse cholesterol transport or high density lipoprotein increasing of resveratrol, polyhydric phenols or flavone compound for example.
An example of chemical compound provided by the invention is the resveratrol that the organic nitrates base with 3 hydroxyls that replace being present in natural resveratrol replaces.This chemical compound called after 3,4 ', trans stilbene of 5-trinitro-oxygen base or resveratrol trisnitrate, or be 1 with the IUPAC nomenclature, 3-two-nitrooxy-5-[2-(4-nitrooxy-phenyl)-vinyl]-benzene.Another example of this chemical compound provided by the invention is the naringenin that the organic nitrates base with 3 hydroxyls that replace being present in natural naringenin replaces.This chemical compound called after naringenin trisnitrate, or be 5 with the IUPAC nomenclature, 7-two-nitrooxy-2-(4-nitrooxy-phenyl)-4-Chromanone.Another example of chemical compound provided by the invention is the reverse ester nitrooxy analog of naringenin; its three hydroxyls are substituted; be 5-nitrooxy-valeric acid 4-[5,7-two-(5-nitrooxy-valeryl oxygen base)-4-oxo-benzodihydropyran-2-yl]-phenylester.Although be not subjected to the restriction of those chemical compounds of example description herein, partly provide more example in the embodiment of the invention.
The trans-resveratrol raw material that is used to react can be from Bio-Stat Limited (Stockport, U.K.) or Sigma Chemical Co. (St.Louis, MO, USA) purchase obtains, method (1995) Am.J.Enol.Vitic.46 (2) with Goldberg etc.: 159-165 separates from wine.Perhaps, can be by Toppo at United States Patent (USP) 6,048, the method for explanation in 903, or by the Wittig reaction of Waterhouse by Moreno-Manas and Pleixats method improvement is a raw material by the phenol of suitable replacement, synthesizing trans-resveratrol.
As the naringenin of synthetic reaction composition is naturally occurring chemical compound, obtains from source, market widely easily, and perhaps available well-known process is for example separated the citrus juice from natural origin, need not over-drastic experiment.
Administration
For treating above-mentioned disease, can use chemical compound separately, but the pharmaceutically acceptable dosage form that contains acceptable carrier or excipient more preferably is provided.Although in any concrete condition, optimal form of medication depends on the degree of the disease for the treatment of and seriousness and the character of using particular compound, but these preparations comprise those preparations that are applicable to oral, rectum, part, mouthful cheek and parenteral (for example subcutaneous, intramuscular, Intradermal or intravenous) administration.
The preparation that is applicable to oral administration can provide by discontinuous unit, for example capsule, cachet, lozenge or tablet, each self-contained predetermined quantitative compound powder or granule; Be water or on-aqueous liquid solution or suspension; Or be oil-in-water or water in oil emulsion.By explanation, can prepare this type of preparation by any proper drug method, these methods comprise the step that makes reactive compound and carrier or excipient (they can form one or more annexing ingredients) combination.According to preparation in other component compatibility and principle that must be harmless to the receiver, carrier must be acceptable.Carrier can be solid or liquid or both, preferably is mixed with unit dose formulations with chemical compound, tablet for example, and it can contain 0.05%-95% (weight) reactive compound.Also can use other pharmacological active substance that comprises other chemical compound.Can be by any medicament technology preparation of knowing basically by mixing the preparation of the present invention that each component is formed.
For solid composite, conventional non-toxic solid carrier comprises for example pharmaceutical grade mannitol, lactose, starch, magnesium stearate, saccharin sodium, Pulvis Talci, cellulose, glucose, sucrose, magnesium carbonate etc.Can for example pass through described reactive compound and the medicinal auxiliary agent dissolving of choosing wantonly, dispersion etc. herein at excipient, for example in water, saline, D/W, glycerol, the ethanol etc., thereby form solution or suspension, the preparation pharmacology goes up spendable fluid composition.Generally speaking, preferably by with reactive compound with liquid or solid carrier fine powder or both are all even mixes fully, then as need make the product shaping, prepare appropriate formulation.For example, can be by powder or granule compression or molded preparation tablet with chemical compound and optional and one or more helper components.Can be by on suitable machine, compression optional with binding agent, lubricant, inert diluent and/or the blended free-flowing form of surfactant/dispersant for example powder or graininess chemical compound, preparation compressed tablet.Can be by on suitable machine, with compound powder inert liquid diluent moistening, by molded preparation molded tablet.
The preparation that is applicable to a mouthful cheek (Sublingual) administration comprises that containing flavoring substrate is generally the lozenge of sucrose and arabic gum (atacia) or tragacanth and chemical compound and contains chemical compound and the inert base pastille (pastilles) of gelatin and glycerol or sucrose and arabic gum for example.
The preparation of the present invention that is applicable to parenteral comprises the sterile aqueous preparation of chemical compound, and they and receiver's blood waits substantially and oozes.These preparations give through intravenous, also can give by subcutaneous, intramuscular or intradermal injection.Can mix with water by making chemical compound, with the solution sterilization that obtains and make it and blood etc. oozes, prepare this type of preparation expediently.Composition for injection of the present invention contains the 0.1-5%w/w reactive compound usually.
The preparation that is applicable to rectally is provided with unit dose suppository.Can by with the conventional solid carrier of chemical compound and one or more for example cocoa butter mix, make then to obtain mixture shaping, prepare these preparations.
Be applicable to that the preparation that local skin is used preferably adopts ointment, cream, lotion, paste, gel, spray, aerosol or oily form.Spendable carrier and excipient comprise vaseline, lanoline (lanoline), Polyethylene Glycol, pure and mild wherein two or more combination.Usually the concentration of the reactive compound that exists is counted 0.1-15%w/w with compositions, for example 0.5-2%.
The amount that gives reactive compound depends on the mode of patient to be treated, patient's body weight, administration and prescriber's judgement naturally.In the methods of the invention, dosage regimen is usually directed to every day or gives the envelope capsule chemical compound of intelligible 1 μ g-1000mg dosage per half a day.The help of envelope capsule enters site of action, allows to give simultaneously each active component, produces synergism in theory.According to the standard dosage regimen, the doctor determines optimal dose easily, can easily improve administration to reach such dosage.
Embodiment
Propose following examples and understand the present invention, but should not be construed as described herein and concrete qualification claimed invention with help.This type of version of the present invention in those skilled in the art are familiar with scope comprises that now known or exploitation thereafter is equal to the replacement of chemical compound, comprises that preparation changes or the less change of experimental design, thinks in the scope of the invention that is combined in herein.
For all embodiment that provide herein, unless otherwise indicated, any chemical compound that provides among the present invention is provided for term " all cpds " or " chemical compound ".
Embodiment 1: preparation 1,3-two-nitrooxy-5-[2-(4-nitrooxy-phenyl)-vinyl]-benzene
Under 25 ℃, to 1mmol 5-[(E)-2-(4-hydroxyl-phenyl)-vinyl]-benzene-1,3-diphenol (synonym: resveratrol; 3,4 ', the trans stilbene of 5-trihydroxy) the 5ml anhydrous THF solution in add 3mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether), solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products (1,3-two-nitrooxy-5-[(E)-2-(4-nitrooxy-phenyl)-vinyl]-benzene) and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 2: preparation piceatannol tetranitrate
Under 25 ℃, to 1mmol (E)-4-(2-(3, the 5-dihydroxy phenyl) vinyl)-1,2-Benzodiazepines (synonym: add 4mmolSOCl (NO in 5ml anhydrous THF solution piceatannol) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products (piceatannol tetranitrate) and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 3: preparation butein tetranitrate
Under 25 ℃, to 1mmol 3,4,2 ', 4 '-tetrahydroxy chalcone (synonym: add 4mmol SOCl (NO in 5ml anhydrous THF solution butein) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) makes the solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products butein tetranitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 4: preparation isoliquiritigenin trisnitrate
Under 25 ℃, to 1mmol 4,2 ', 4 '-trihydroxy chalcone (synonym: add 3mmol SOCl (NO in 5ml anhydrous THF solution isoliquiritigenin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products isoliquiritigenin tetranitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 5: preparation fisetin tetranitrate
Under 25 ℃, to 1mmol 3,7,3 ', 4 '-kaempferol (synonym: add 4mmol SOCl (NO in 5ml anhydrous THF solution fisetin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products fisetin tetranitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 6: preparation Quercetin five nitrates
Under 25 ℃, to 1mmol 3,5,7,3 ', 4 '-pentahydroxyflavone (synonym: add 5mmol SOCl (NO in 5ml anhydrous THF solution Quercetin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products Quercetin five nitrates and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 7: preparation N-(3,5-two-nitrooxy-phenyl)-N '-(4-nitrooxy-phenyl)-hydrazine
Under 25 ℃, to 1mmol 5-[N '-(4-hydroxyl-phenyl)-diazanyl]-benzene-1, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products N-(3,5-two-nitrooxy-phenyl)-N '-(4-nitrooxy-phenyl)-hydrazine and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 8: preparation 1,3-two-nitrooxy-5-(4-nitrooxy-phenyl disulfide group)-benzene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(4-hydroxyl-phenyl disulfide group)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1, (wherein any hydroxyl is independent of ONO for 3-two-nitrooxy-5-(4-nitrooxy-phenyl disulfide group)-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 9: preparation 1,3-two-nitrooxy-5-(4-nitrooxy-phenyl peroxy)-benzene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(4-hydroxyl-phenyl peroxy)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1, (wherein any hydroxyl is independent of ONO for 3-two-nitrooxy-5-(4-nitrooxy-phenyl peroxy)-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 10: preparation 1,3-two-nitrooxy-5-(4-nitrooxy-phenyl sulfenyl methyl)-benzene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(4-hydroxyl-phenyl sulfenyl methyl)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1, (wherein any hydroxyl is independent of ONO for 3-two-nitrooxy-5-(4-nitrooxy-phenyl sulfenyl methyl)-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 11: and preparation N-(3,5-two-nitrooxy-phenyl-O-(4-nitrooxy-phenyl)-hydroxylamine
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(4-hydroxyl-benzene oxygen amino)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(3, (wherein any hydroxyl is independent of ONO for 5-two-nitrooxy-phenyl-O-(4-nitrooxy-phenyl)-hydroxylamine and part nitric acid esterification products to make complete nitric acid esterification products N- 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 12: preparation benzyl-(4-nitrooxy-phenyl)-amine
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 4-benzylamino-phenol, add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products benzyl-(4-nitrooxy-phenyl)-amine pass through the silica gel column chromatography purification and separate.
Embodiment 13: preparation 2-(salicylidene amino) phenol dinitrate
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 2-(salicylidene amino) phenol, add 2mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein arbitrary hydroxyl is independent of ONO to make complete nitric acid esterification products 2-(salicylidene amino) phenol dinitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 14: preparation (2,4-two-nitrooxy-phenyl)-(2-nitrooxy-phenyl)-diazene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol (synonym: 4-((2-hydroxy phenyl) azo group)-1,3-Benzodiazepines) to 1mmol 4-(2-hydroxyl-phenylazo)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2,4-two-nitrooxy-phenyl)-(wherein any hydroxyl is independent of ONO for (2-nitrooxy-phenyl)-diazene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 15: preparation two-(2,2 '-nitrooxy-phenyl)-diazene
Under 25 ℃, to 1mmol two-(2,2 '-hydroxyl-phenyl)-diazene (synonym: add 2mmolSOCl (NO in 5ml anhydrous THF solution 1-hydroxyl-2-(2-hydroxy phenyl azo group) benzene) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein arbitrary hydroxyl is independent of ONO to make complete nitric acid esterification products two-(2,2 '-nitrooxy-phenyl)-diazene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 16: preparation N-(3-nitrooxy-phenyl)-benzsulfamide
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol N-(3-hydroxyl-phenyl)-benzsulfamide (synonym: N-(3-hydroxy phenyl) benzsulfamide), add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products N-(3-nitrooxy-phenyl)-benzsulfamide pass through the silica gel column chromatography purification and separate.
Embodiment 17: preparation N-(4-nitrooxy-phenyl)-benzsulfamide
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol N-(4-hydroxyl-phenyl)-benzsulfamide (synonym: N-(4-hydroxy phenyl) benzsulfamide), add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products N-(4-nitrooxy-phenyl)-benzsulfamide pass through the silica gel column chromatography purification and separate.
Embodiment 18: preparation 3,3 ', 4,5 '-tetranitro oxygen base bibenzyl
Under 25 ℃, to 1mmol 3,3 ', 4,5 '-add 4mmol SOCl (NO in the 5ml anhydrous THF solution of tetrahydroxy bibenzyl 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 3,3 ', 4,5 '-(wherein any hydroxyl is independent of ONO for tetranitro oxygen base bibenzyl and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 19: preparation 1-benzyloxy-2-nitrooxy-benzene
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 2-benzyloxy-phenol, add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products 1-benzyloxy-2-nitrooxy-benzene pass through the silica gel column chromatography purification and separate.
Embodiment 20: preparation benzoic acid 3-nitrooxy-phenylester
Under 25 ℃, to 1mmol benzoic acid 3-hydroxyl-phenylester (synonym: add 1mmol SOCl (NO in 5ml anhydrous THF solution resorcinol monobenzoate) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products benzoic acid 3-nitrooxy-phenylester pass through the silica gel column chromatography purification and separate.
Embodiment 21: preparation 2-nitrooxy-phenol benzoate
Under 25 ℃, to 1mmol 2-hydroxy-benzoic acid phenyl ester (synonym: add 1mmol SOCl (NO in 5ml anhydrous THF solution phenyl salicytate) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products 2-nitrooxy-phenol benzoate pass through the silica gel column chromatography purification and separate.
Embodiment 22: preparation 2-nitrooxy-N-(4-nitrooxy-phenyl)-Benzoylamide
Under 25 ℃, to 1mmol 2-hydroxy-n-(4-hydroxyl-phenyl)-Benzoylamide (synonym: add 2mmol SOCl (NO in 5ml anhydrous THF solution osalmide (osalmid)) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein arbitrary hydroxyl is independent of ONO to make complete nitric acid esterification products 2-nitrooxy-N-(4-nitrooxy-phenyl)-Benzoylamide and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 23: preparation 2-nitrooxy-N-(3-nitrooxy-phenyl)-Benzoylamide
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 2-hydroxy-n-(3-hydroxyl-phenyl)-Benzoylamide, add 2mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein arbitrary hydroxyl is independent of ONO to make complete nitric acid esterification products 2-nitrooxy-N-(3-nitrooxy-phenyl)-Benzoylamide and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 24: preparation 3,4,5-three-nitrooxy-N-phenyl-Benzoylamide
Under 25 ℃, to 1mmol 3,4,5-trihydroxy-N-((Z)-1-methylene-but-2-ene base)-Benzoylamide (synonym: add 3mmolSOCl (NO in 5ml anhydrous THF solution gallanilide) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 3,4, (wherein any hydroxyl is independent of ONO for 5-three-nitrooxy-N-phenyl-Benzoylamide and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 25: preparation 1-(2,4-two-nitrooxy-phenyl)-2-phenyl-ethyl ketone
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 1-(2,4-hydroxyl-phenyl)-2-phenyl-ethyl ketone (synonym: benzyl 2,4-dihydroxy phenyl ketone), add 2mmolSOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein arbitrary hydroxyl is independent of ONO to make complete nitric acid esterification products 1-(2,4-two-nitrooxy-phenyl)-2-phenyl-ethyl ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 26: preparation 1,2-two-nitrooxy-3-phenoxy group-benzene
Under 25 ℃,, add 2mmol SOCl (NO in the 5ml anhydrous THF solution of 2-diphenol to 1mmol 3-phenoxy group-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1, (wherein arbitrary hydroxyl is independent of ONO for 2-two-nitrooxy-3-phenoxy group-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 27: preparation 1,2-two-nitrooxy-3-(2-nitrooxy-phenoxy group)-benzene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 2-diphenol to 1mmol 3-(2-hydroxyl-phenoxy group)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1, (wherein any hydroxyl is independent of ONO for 2-two-nitrooxy-3-(2-nitrooxy-phenoxy group)-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 28: preparation 1-nitrooxy-2-phenoxy group-benzene
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 2-phenoxy group-phenol, add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products 1-nitrooxy-2-phenoxy group-benzene pass through the silica gel column chromatography purification and separate.
Embodiment 29: preparation 5, the two resorcinol tetranitrates of 5-sulfinyl
Under 25 ℃,, add 4mmol SOCl (NO in the 5ml anhydrous THF solution of the two resorcinol of 5-sulfinyl to 1mmol 5 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 5, (wherein any hydroxyl is independent of ONO for two resorcinol tetranitrates of 5-sulfinyl and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 30: preparation 4,4 '-thiobis 1,3-Benzodiazepines tetranitrate
Under 25 ℃,, add 4mmol SOCl (NO in the 5ml anhydrous THF solution of 3-Benzodiazepines to 1mmol 4,4 '-thiobis 1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 4,4 '-thiobis 1, (wherein any hydroxyl is independent of ONO for 3-Benzodiazepines tetranitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 31: preparation 2,2 '-thiobis phenol dinitrate
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 2,2 '-thiobis phenol, add 2mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2,2 '-(wherein arbitrary hydroxyl is independent of ONO for thiobis phenol dinitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 32: preparation 1-benzyl-2,4-two-nitrooxy-benzene
Under 25 ℃,, add 2mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol (synonym: 3-phenyl methyl-1,3-Benzodiazepines) to 1mmol 4-benzyl-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1-benzyl-2, (wherein arbitrary hydroxyl is independent of ONO for 4-two-nitrooxy-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 33: preparation 2-benzyl-1,4-two-nitrooxy-benzene
Under 25 ℃,, add 2mmolSOCl (NO in the 5ml anhydrous THF solution of 4-diphenol (synonym: 4-phenyl methyl-1,4-Benzodiazepines) to 1mmol 2-benzyl-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2-benzyl-1, (wherein arbitrary hydroxyl is independent of ONO for 4-two-nitrooxy-benzene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 34: preparation (2,3,4-three-nitrooxy-phenyl)-(3,4,5-three-nitrooxy-phenyl)-ketone
Under 25 ℃, to 1mmol (2,3,4-trihydroxy-phenyl)-(3,4,5-trihydroxy-phenyl)-ketone (synonym: add 6mmolSOCl (NO in 5ml anhydrous THF solution exifone) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products (2,3,4-three-nitrooxy-phenyl)-(3,4,5-three-nitrooxy-phenyl)-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 35: preparation (2-nitrooxy-phenyl)-aniline
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 2-phenyl amino-phenol, add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products (2-nitrooxy-phenyl)-aniline pass through the silica gel column chromatography purification and separate.
Embodiment 36: preparation 2-(3,5-two-nitrooxy-phenyl)-6-nitrooxy-4H-.alpha.-5:6-benzopyran
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(6-hydroxyl-4H-.alpha.-5:6-benzopyran-2-yl)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products 2-(3,5-two-nitrooxy-phenyl)-6-nitrooxy-4H-.alpha.-5:6-benzopyran and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 37: preparation 2-(3,5-two-2 nitrooxies-phenyl)-6-nitrooxy-1,4-dihydro-naphthalene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(6-hydroxyl-1,4-dihydro-naphthalene-2-yl)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2-(3,5-two-nitrooxy-phenyl)-6-nitrooxy-1, (wherein any hydroxyl is independent of ONO for 4-dihydro-naphthalene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 38: preparation 2-(3,5-two-nitrooxy-phenyl)-6-nitrooxy-1,2,3,4-tetrahydrochysene-naphthalene
Under 25 ℃,, add 3mmol SOCl (NO in the 5ml anhydrous THF solution of 3-diphenol to 1mmol 5-(6-hydroxyl-1,2,3,4-tetrahydrochysene-naphthalene-2-yl)-benzene-1 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2-(3,5-two-nitrooxy-phenyl)-6-nitrooxy-1,2,3, (wherein any hydroxyl is independent of ONO for 4-tetrahydrochysene-naphthalene and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 39: preparation 5,7-two-nitrooxy-2-(4-nitrooxy-phenyl)-4-Chromanone
Under 25 ℃, to 1mmol 5,7-dihydroxy-2-(4-hydroxyl-phenyl)-4-Chromanone (synonym: add 3mmolSOCl (NO in 5ml anhydrous THF solution naringenin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 5, (wherein any hydroxyl is independent of ONO for 7-two-nitrooxy-2-(4-nitrooxy-phenyl)-4-Chromanone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 40: preparation 5,7-two-nitrooxy-2-(4-nitrooxy-phenyl)-.alpha.-5:6-benzopyran-4-ketone
Under 25 ℃, to 1mmol 5,7-dihydroxy-2-(4-hydroxyl-phenyl)-.alpha.-5:6-benzopyran-4-ketone (synonym: add 3mmol SOCl (NO in the 5ml anhydrous THF solution celery flavin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 5, (wherein any hydroxyl is independent of ONO for 7-two-nitrooxy-2-(4-nitrooxy-phenyl)-.alpha.-5:6-benzopyran-4-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 41: preparation 5,7-two-nitrooxy-3-(4-nitrooxy-phenyl)-.alpha.-5:6-benzopyran-4-ketone
Under 25 ℃, to 1mmol 5,7-dihydroxy-3-(4-hydroxyl-phenyl)-.alpha.-5:6-benzopyran-4-ketone (synonym: add 3mmolSOCl (NO in 5ml anhydrous THF solution genistein) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 5, (wherein any hydroxyl is independent of ONO for 7-two-nitrooxy-3-(4-nitrooxy-phenyl)-.alpha.-5:6-benzopyran-4-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 42: preparation 2-(3,4-two-nitrooxy-phenyl)-3,4,5,7-four-nitrooxy-benzodihydropyran
Under 25 ℃, to 1mmol 2-(3,4-dihydroxy-phenyl)-benzodihydropyran-3,4,5,7-tetrol (synonym: add 6mmolSOCl (NO in 5ml anhydrous THF solution leucocianidol) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2-(3,4-two-nitrooxy-phenyl)-3,4,5, (wherein any hydroxyl is independent of ONO for 7-four-nitrooxy-benzodihydropyran and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 43: preparation 6-hydroxyl-7-nitrooxy-3-(4-nitrooxy-phenyl)-4-Chromanone
Under 25 ℃, to 1mmol 6,7-dihydroxy-3-(4-hydroxyl-phenyl)-4-Chromanone (synonym: 6,7,4 '-the trihydroxy isoflavanone) the 5ml anhydrous THF solution in add 3mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products 6-hydroxyl-7-nitrooxy-3-(4-nitrooxy-phenyl)-4-Chromanone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 44: preparation Quracol B tetranitrate
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol Quracol B, add 4mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products Quracol B tetranitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 45: preparation 1-(4-hydroxyl-2,6-two-nitrooxy-phenyl)-3-(4-nitrooxy-phenyl)-third-1-ketone
Under 25 ℃, to 1mmol 3-(4-hydroxyl-phenyl)-1-(2,4,6-trihydroxy-phenyl)-third-1-ketone (synonym: add 4mmol SOCl (NO in 5ml anhydrous THF solution phloretin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein any hydroxyl is independent of ONO to make complete nitric acid esterification products 1-(4-hydroxyl-2,6-two-nitrooxy-phenyl)-3-(4-nitrooxy-phenyl)-third-1-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 46: preparation 1-nitrooxy-4-((Z)-3-phenyl-pi-allyl)-benzene
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 4-((Z)-3-phenyl-pi-allyl)-phenol (synonym: (E)-4-(3-phenyl-2-acrylic)-phenol), add 1mmolSOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 1-nitrooxy-4-((Z)-3-phenyl-pi-allyl)-benzene pass through the silica gel column chromatography purification and separate.
Embodiment 47: preparation 1-nitrooxy-4-((E)-3-phenyl-acrylic)-benzene
Under 25 ℃, in the 5ml anhydrous THF solution of 1mmol 4-((E)-3-phenyl-acrylic)-phenol, add 1mmol SOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products 1-nitrooxy-4-((E)-3-phenyl-acrylic)-benzene pass through the silica gel column chromatography purification and separate.
Embodiment 48: preparation 5,6,7-three-nitrooxy-2-phenyl-.alpha.-5:6-benzopyran-4-ketone
Under 25 ℃, to 1mmol 5,6,7-trihydroxy-2-phenyl-.alpha.-5:6-benzopyran-4-ketone (synonym: add 3mmol SOCl (NO in 5ml anhydrous THF solution baicalein) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 5,6, (wherein any hydroxyl is independent of ONO for 7-three-nitrooxy-2-phenyl-.alpha.-5:6-benzopyran-4-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 49: preparation rutin tetranitrate
Under 25 ℃, to 1mmol 2-(3,4-dihydroxy-phenyl)-5,7-dihydroxy-3-[(2S, 3R, 5S, 6R)-3,4,5-trihydroxy-6-((2R, 3R, 4R, 5R, 6S)-3,4,5-trihydroxy-6-methyl-tetrahydrochysene-pyrans-2-base oxygen ylmethyl)-tetrahydrochysene-pyrans-2-base oxygen base]-.alpha.-5:6-benzopyran-4-ketone (synonym: add 4mmol SOCl (NO in 5ml anhydrous THF solution rutin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2-(3,4-two-nitrooxy-phenyl)-5,7-two-nitrooxy-3-[(2S, 3R, 5S, 6R)-3,4,5-trihydroxy-6-((2R, 3R, 4R, 5R, 6S)-3,4,5-trihydroxy-6-methyl-tetrahydrochysene-pyrans-2-base oxygen ylmethyl)-tetrahydrochysene-pyrans-2-base oxygen base]-(wherein any hydroxyl independence is by ONO for .alpha.-5:6-benzopyran-4-ketone (rutin tetranitrate) and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 50: preparation 5-hydroxyl-2-(4-hydroxy phenyl)-7-(2-O-α-L-Fructus rhamni (Rhamnus davurica Pall.) pyrans glycosyl-β-D-glucopyranosyl oxygen base)-4-benzodihydropyrone (chromanon) dinitrate
Under 25 ℃, to 1mmol 5-hydroxyl-2-(4-hydroxy phenyl)-7-(2-O-α-L-Fructus rhamni (Rhamnus davurica Pall.) pyrans glycosyl-β-D-glucopyranosyl oxygen base)-4-benzodihydropyrone (synonym: add 2mmol SOCl (NO in 5ml anhydrous THF solution naringin) 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.(wherein arbitrary hydroxyl is independent of ONO to make complete nitric acid esterification products 5-hydroxyl-2-(4-hydroxy phenyl)-7-(2-O-α-L-Fructus rhamni (Rhamnus davurica Pall.) pyrans glycosyl-β-D-glucopyranosyl oxygen base)-4-benzodihydropyrone dinitrate and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 51: preparation (E)-(3S, 5R)-7-[3-(4-fluoro-phenyl)-1-isopropyl-1H-indole-2-yl]-1,3,5-three-nitrooxy-heptan-6-alkene-1-ketone
Under 25 ℃, to 1mmol (E)-(3S, 5R)-7-[3-(4-fluoro-phenyl)-1-isopropyl-1H-indole-2-yl]-3,5-dihydroxy-heptan-6-olefin(e) acid (synonym: fluvastatin; Novartis) add 3mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products (E)-(3S, SR)-7-[3-(4-fluoro-phenyl)-1-isopropyl-1H-indole-2-yl]-1,3,5-three-nitrooxy-heptan-(wherein any hydroxyl is independent of ONO for 6-alkene-1-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 52: preparation N-phenyl-[5-(4-fluoro-phenyl)-2-isopropyl-4-phenyl-1-((3R, 5R)-3,5,7-three-nitrooxy-7-oxo-heptyl)-1H-pyrroles-1-yl]-the 3-Methanamide
Under 25 ℃, to 1mmol (3R, 5R)-7-[2-(4-fluoro-phenyl)-5-isopropyl-3-phenyl-4-phenyl amino formoxyl-pyrroles-1-yl]-3,5-dihydroxy-enanthic acid (synonym: atorvastatin; Parke-Davis) add 3mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products N-phenyl-[5-(4-fluoro-phenyl)-2-isopropyl-4-phenyl-1-((3R, 5R)-3,5,7-three-nitrooxy-7-oxo-heptyl)-1H-pyrroles-1-yl]-(wherein any hydroxyl is independent of ONO for 3-Methanamide and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 53: preparation (E)-(3R, 5S)-7-[4-(4-fluoro-phenyl)-2,6-diisopropyl-5-methoxy-pyridin-3-yl]-1,3,5-three-nitrooxy-heptan-6-alkene-1-ketone
Under 25 ℃, to 1mmol (E)-(3R, 5S)-7-[4-(4-fluoro-phenyl)-2,6-diisopropyl-5-methoxy-pyridin-3-yl]-3,5-dihydroxy-heptan-6-olefin(e) acid (synonym: cerivastatin; Bayer) add 3mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products (E)-(3R, 5S)-7-[4-(4-fluoro-phenyl)-2,6-diisopropyl-5-methoxy-pyridin-3-yl]-1,3,5-three-nitrooxy-heptan-(wherein any hydroxyl is independent of ONO for 6-alkene-1-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 54: preparation (S)-2-methyl-butanoic acid (1S, 3S, 7S, 8S, 8aR)-7-methyl-3-nitro oxygen base-8-((4R, 6R)-3,5,7-three-nitrooxy-7-oxo-heptyl)-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester
Under 25 ℃, to 1mmol (2R, 4R)-3,5-dihydroxy-7-[(1S, 2S, 6S, 8S, 8aR)-6-hydroxy-2-methyl-8-((S)-2-methyl-bytyry oxygen base)-1,2,6,7,8,8a-six hydrogen-naphthalene-1-yl]-enanthic acid (synonym: pravastatin; Bristol-Myers Squibb) adds 4mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products (S)-2-methyl-butanoic acid (1S, 3S, 7S, 8S, 8aR)-7-methyl-3-nitro oxygen base-8-((4R, 6R)-3,5,7-three-nitrooxy-7-oxo-heptyl)-1,2,3,7,8, (wherein any hydroxyl is independent of ONO for 8a-six hydrogen-naphthalene-1-base ester and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 55: preparation 2,2-dimethyl-butanoic acid (1S, 3R, 7S, 8S, 8aR)-3,7-dimethyl-8-[2-((2R, 4R)-4-nitrooxy-6-oxo-tetrahydrochysene-pyrans-2-yl)-ethyl]-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester
Under 25 ℃, to 1mmol 2,2-dimethyl-butanoic acid (1S, 3R, 7S, 8S, 8aR)-8-[2-((2R, 4R)-4-hydroxyl-6-oxo-tetrahydrochysene-pyrans-2-yl)-ethyl]-3,7-dimethyl-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester (synonym: simvastatin; Merck) add 1mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products 2,2-dimethyl-butanoic acid (1S, 3R, 7S, 8S, 8aR)-3,7-dimethyl-8-[2-((2R, 4R)-4-nitrooxy-6-oxo-tetrahydrochysene-pyrans-2-yl)-ethyl]-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester passes through the silica gel column chromatography purification and separates.
Embodiment 56: preparation (S)-2-methyl-butanoic acid (1S, 3R, 7S, 8S, 8aR)-3,7-dimethyl-8-[2-((2R, 4R)-4-nitrooxy-6-oxo-tetrahydrochysene-pyrans-2-yl)-ethyl]-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester
Under 25 ℃, to 1mmol (S)-2-methyl-butanoic acid (1S, 3R, 7S, 8S, 8aR)-8-[2-((2R, 4R)-4-hydroxyl-6-oxo-tetrahydrochysene-pyrans-2-yl)-ethyl]-3,7-dimethyl-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester (synonym: lovastatin; Merck) add 1mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products (S)-2-methyl-butanoic acid (1S, 3R, 7S, 8S, 8aR)-3,7-dimethyl-8-[2-((2R, 4R)-4-nitrooxy-6-oxo-tetrahydrochysene-pyrans-2-yl)-ethyl]-1,2,3,7,8,8a-six hydrogen-naphthalene-1-base ester passes through the silica gel column chromatography purification and separates.
Embodiment 57: preparation N-[4-(4-fluoro-phenyl)-6-isopropyl-5-((E)-(3R, SR)-3,5,7-three-nitrooxy-7-oxo-heptan-1-thiazolinyl)-pyrimidine-2-base]-N-methyl-Methanesulfomide
Under 25 ℃, to 1mmol (E)-(3R, SR)-7-[4-(4-fluoro-phenyl)-6-isopropyl-2-(mesyl-methyl-amino)-pyrimidine-5-yl]-3,5-dihydroxy-heptan-6-olefin(e) acid (synonym: rosuvastatin; Astra-Zeneca) add 1mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products N-[4-(4-fluoro-phenyl)-6-isopropyl-5-((E)-(3R, SR)-3,5,7-three-nitrooxy-7-oxo-heptan-1-thiazolinyl)-pyrimidine-2-base]-N-methyl-Methanesulfomide is by the silica gel column chromatography purification and separate.
Embodiment 58: preparation nitrooxy-pyridin-3-yl-ketone
Under 25 ℃, in the 5ml of 1mmol nicotinic acid anhydrous THF solution, add 1mmolSOCl (NO 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make nitric acid esterification products nitrooxy-pyridin-3-yl-ketone pass through the silica gel column chromatography purification and separate.
Embodiment 59: preparation (S)-1-(4-fluoro-phenyl)-3-[(S)-3-(4-fluoro-phenyl)-3-nitrooxy-propyl group]-4-(4-nitrooxy-phenyl)-nitrogen heterocyclic din-2-ketone
Under 25 ℃, to 1mmol (S)-1-(4-fluoro-phenyl)-3-[(S)-3-(4-fluoro-phenyl)-3-hydroxyl-propyl group]-4-(4-hydroxyl-phenyl)-nitrogen heterocyclic din-2-ketone (synonym: ezetimibe; Merck) add 2mmol SOCl (NO in the 5ml anhydrous THF solution 3) or SO (NO 3) 2Behind the 1h, add Et 2O (ether) is with solution with water washing, drying, evaporation.Make complete nitric acid esterification products (S)-1-(4-fluoro-phenyl)-3-[(S)-3-(4-fluoro-phenyl)-3-nitrooxy-propyl group]-(wherein any hydroxyl is independent of ONO for 4-(4-nitrooxy-phenyl)-nitrogen heterocyclic din-2-ketone and part nitric acid esterification products 2Group displacement) passes through the silica gel column chromatography purification and separate.
Embodiment 60: the method that makes the esterification of The compounds of this invention glucuronic acid
This embodiment describes the method for preparing glucuronic acid esterification chemical compound of the present invention.In this specific embodiment, the resveratrol of dinitric acid esterification form (press 3,4 of embodiment 1 preparation '-nitrooxy-5-hydroxyl resveratrol) (50-1000 μ M) and 10 μ l people intestinal, 25 μ l colons or 10 μ l hepatomicrosomes (being respectively 200,400,200 μ g protein), the 20 μ l UDP-glucuronyl transferases (400 μ g albumen) of recombinating are being contained 10mM MgCl 2The final volume of 50mM Tris HCl buffer (pH 7.8) solution be 500 μ l solution, at 37 ℃ of following precincubation 5min.Add 1mM 5 '-diphosphonic acid glucuronic acid initiation reaction.With reactant mixture at 37 ℃ of following incubation 60min.With sample in cooled on ice, with the 1cc C of suitable (oasis) hydrophile-lipophile balance value 18(Waters Corp, Milford MA) carry out Solid-Phase Extraction to extraction column.With post 1-ml washed with methanol, use the 1-ml water balance.After loading the 0.5ml sample, with post 5% washed with methanol, with 2ml 100% methanol-eluted fractions.Under 40 ℃, nitrogen,, sample is dissolved in 250 μ l mobile phases again, is used for HPLC and analyzes the meoh eluate drying.
Embodiment 61: the method that makes the The compounds of this invention Sulfation
This embodiment describes the method for preparing Sulfation chemical compound of the present invention.In this specific embodiment, with aforementioned ion pair extraction method (1987.Anal.Biochem.161:176-180 such as Varin), by the resveratrol of sulfotransferase with dinitro acidify form, promptly press 3,4 of embodiment 1 preparation '-nitrooxy-5-hydroxyl resveratrol Sulfation.The type reaction mixture of cumulative volume 100 μ l contains 0.1-200 μ M 3,4 '-nitrooxy-5-hydroxyl resveratrol, 1 μ M[ 35S] PAPS and 2.5 μ l mixing human liver cell colloidal sols (50 μ g albumen), 2.5 μ l people jejunum cytosoles (30 μ g), Caco-2 cytosol (225 μ g) or 0.25 μ l reorganization sulfotransferase and 33mM Tris-HCl buffer (pH 7.4) and 8mM dithiothreitol, DTT and 0.0625% bovine serum albumin.Sample at 37 ℃ of following incubation 30min, is added 10 μ l, 2.5% acetic acid, 20 μ l, 0.1 μ M 4-butyl ammonium hydrogen sulfate and 500 μ l ethyl acetate cessation reactions.Through mixing and centrifugal after, add biodegradable counting scintillator (Amersham Biosciences, Piscataway, NJ) after, make 400 μ l acetic acid ethyl acetate extracts experience liquid scintillation counting.
Embodiment 62: measure ACAT and suppress
Can pass through known method, for example at United States Patent (USP) 6,165,984 explanations with the method for the following stated, measure activity as the The compounds of this invention of ACAT inhibitor.
At first, by the sacrificed by decapitation rat, hepatectomize.Each liver is got 1g, puts into 5ml homogenize medium (0.1M KH 2PO 4, pH 7.4,0.1mM EDTA and 10mM beta-mercaptoethanol) and middle homogenize.Under 4 ℃, with the centrifugal 10min of homogenate, under 4 ℃, with the centrifugal 15min of the supernatant that obtains, obtain supernatant with 15,000 .g with 3,000 .g.Under 4 ℃, supernatant is put into ultracentrifugation pipe (Beckman), with centrifugal 1 hour of 100,000 .g, obtain the microsome precipitation, then it is suspended in the 3ml homogenize medium, under 4 ℃, with centrifugal 1 hour of 100,000 .g.The precipitation that so obtains is suspended in the 1ml homogenize medium.Measure the protein concentration in the suspension obtain by the LowryShi method, transfer to 4-8mg/ml then.The suspension that obtains is stored in cold closet (Biofreezer, Forma ScientificInc.).
The acetone soln of 6.67 μ l 1mg/ml cholesterol is mixed with the acetone soln of 6 μ l 10%Triton WR-1339 (Sigma Co.), remove acetone in the mixture with nitrogen by evaporation then.Distilled water is added in the mixture that obtains, and the amount of adding transfers to 30mg/ml with the concentration of cholesterol.
In the cholesterol aqueous solution that 10 μ l obtain, add 10 μ l 1M KH 2PO 4(pH 7.4), 5 μ l 0.6mM bovine serum albumin (BSA), at (10 μ l microsome solution that obtain in the step 1) and 55 μ l distilled water (amounting to 90 μ l).In water-bath, under 37 ℃, with mixture precincubation 30min.
With 10 μ l (1- 14C) oleoyl-CoA solution (0.05 μ Ci, final concentration: 10 μ M) add in the mixture of precincubation, under 37 ℃, with the mixture that obtains water-bath incubation 30min.In mixture, add 500 μ l isopropyl alcohols: heptane mixture (4: 1 (v/v)), 300 μ l heptane and 200 μ l 0.1M KH 2PO 4(pH 7.4) by rotating violent blend mixture, at room temperature leave standstill 2min then.
The 200 μ l supernatant that obtain are put into scintillation vial, to wherein adding 4ml scintillation solution (Lumac).Radioactivity with the liquid flashing counter measuring mixture.Calculate ACAT activity (pmole/min/mg albumen) by the synthetic oleic acid cholesterol of the every min. of every mg albumen pmol number.The ACAT activity that observes the rat group that gives The compounds of this invention is less than matched group.
Embodiment 63: measure the HMG-CoA reductase and suppress
Available known method, for example at United States Patent (USP) 5,877, the method for explanation is measured the effectiveness that The compounds of this invention suppresses the HMG-CoA reductase in 208.This method is summarized as follows.
By the sacrificed by decapitation rat, hepatectomize, put into ice-cooled homogenize medium (50mMKH immediately 2PO 4(pH 7.0), 0.2M sucrose, 2mM dithiothreitol, DTT (DTT)) in.With Waring blender (in Potter-Elvehjem type glass homogenizer with engine-driven Teflon mallet bump 3 times) with liver homogenize 15 seconds in homogenize medium (2ml medium/g liver).With the centrifugal 10min of homogenate, the supernatant that obtains with 100,000 centrifugal 75min of .g, is obtained the microsome precipitation with 15,000 .g, then with its resuspending in the homogenize medium that contains 50mM EDTA, with 100,000 centrifugal 60min of .g.Contain MC supernatant as the enzyme source.
By the method (Biochemica et Biophysica Acta, 370,369-377 (1974)) of following Shapiro etc., with radiolabeled 14C HMG-CoA measures the activity of HMG-CoA reductase.
Under 37 ℃, will be in that (what obtain in the step 1) contains enzyme activation 30min in the microsome supernatant.Join and include 20 μ l HMG-CoA reductases mensuration buffer (0.25M KH 2PO 4(pH 7.0), 8.75mM EDTA, 25mM DTT, 0.45M KCl and 0.25mg/mlBSA), 5 μ l 50mM NADPH, 5 μ l are radiolabeled 14C HMG-CoA (0.05 μ Ci/ pipe, final concentration 120 μ M) and 10 μ l activate in the reaction tube of microsomal enzymes (0.03-0.04mg), with mixture at 37 ℃ of following incubation 30min.By 10 μ l 6M HCl are joined cessation reaction in the mixture, at 37 ℃ of following incubation 15min, product is lactonized fully in mixture.By removing precipitation with 10,000 centrifugal 1min of .g, with supernatant point silica gel 60G TLC plate (Altech, Inc., Newark, U.S.A.) on, use benzene then: acetone (1: 1, v/v) launch.(cover slips) scrapes off suitable zone with disposable cover plate, and (Wallacoy Finland) measures radioactivity with 1450 Microbeta liquid scintillation counters.Calculate enzymatic activity (pmol/min/mg albumen) by the synthetic mevalonic acid pmol of the every min. of every mg albumen number.Control rats shows high relatively HMG-CoA reductase activity, and it is low to observe the HMG-CoA specific activity matched group of the rat that gives The compounds of this invention.
Embodiment 64: measure The compounds of this invention and activate PPAR
By several known methods, for example at United States Patent (USP) 6,369,098 following those methods that illustrated are measured the ability that The compounds of this invention is regulated PPAR γ and PPAR alpha active.
Based on suppressing the method that the chemical compound of PPAR γ and PPAR alpha active is regulated in the screening of NF-kB activation
The test The compounds of this invention suppresses the ability of NF-kB activity.Known person endotheliocyte and vascular smooth muscle cell (VSMC) are expressed PPAR γ and PPAR α BiochemPharmacol. such as (, 60:1245-1250 (2000)) Neve BP.Perhaps, be that for example Jurkat T cell line can be used for these experiments from the isolating people's peripheral T lymphocyte of normal healthy people donor or with PPAR γ and/or PPAR alpha expression carrier mammalian cells transfected.In order to one or more stimulate one of cell type of these selections down: phytohemagglutinin/phorbol-12-myristate-13-acetas (PHA/PMA), TNF-α, other factor of interferon-or activation NF-κ B.Can be by measuring the activation of NF-κ B with aforementioned Proc Natl Acad Sci USA such as (, 94:746-50 (1997)) Rossi A similar electrophoretic mobility shift assay.Same cell with 5 μ mol The compounds of this invention precincubation 2 hours, is added the agent of NF-kB activation then, when these chemical compounds do not exist, observe NF-κ B activation and be suppressed.
Based on suppressing the method that the chemical compound of PPAR γ and PPAR alpha active is regulated in NFAT activation screening
Activate in the factor of NFAT one or more to stimulate from the isolating people's peripheral T lymphocyte of normal healthy people donor or PPAR γ and/or PPAR alpha expression carrier mammalian cells transfected be Jurkat T cell line for example with PHA/PMA, TNF-α, interferon-or other.Can by with J Biol Chem. such as aforementioned Yang; The similar electrophoretic mobility shift assay of 275:4541-4 (2000) is measured the transcriptional activation of NFAT.With same cell and 5 μ mol The compounds of this invention precincubation 2 hours, add the NFAT activator then, the NFAT that observes when no described chemical compound exists activates and is suppressed.
Produce the method that the chemical compound of PPAR γ and PPAR alpha active is regulated in screening based on suppressing IL-2
With in the factor of PHA/PMA, TNF-α, interferon-or other activation-inducing IL-2 gene expression one or more stimulate isolating human T lymphocyte or with PPAR α and/or PPAR γ expression vector mammalian cells transfected system as Jurkat T cell line.Press J Biol Chem. such as Yang, 275:4541-4 (2000) is described, with the concentration of IL-2 in Endogen test kit (Wolbum) the measurement cell conditioned medium liquid, measures the IL-2 that produces.With same cell and 5 μ mol The compounds of this invention precincubation 12 hours, add IL-2 then and produce activator, the IL-2 that observes when no described chemical compound exists produces to activate and is suppressed.
Embodiment 65: measure the method that The compounds of this invention activates the ABCA-1 ability
With United States Patent (USP) 6,548, the known method of 548 explanations, this test confirms the effectiveness of The compounds of this invention to ABCA-1 gene expression.Briefly, (Wis.) construction recombination plasmid is to measure reporter gene expression under the control of ABCA-1 promoter for Promega, Madison with pGL3 luciferase reporter gene carrier system.
With plasmid pGL3-Basic (Promega, Madison, Wis.; Catalog number #E1751) work contains the control plasmid of no promoter luciferase genes.By 5 ' flanking region (hAPR1 5 ' promoter with the ABCA-1 gene, nucleotide 1080-1643 corresponding to SEQ ID No:3) genomic fragment is cloned into the SacI position generation plasmid GL-6a of GL3-Basic plasmid, and preparation contains the reporter gene member of ABCA-1 promoter and luciferase genes.Next step is with SpeI and Acc65I digested plasmid GL-6a.Representative is connected on the residue carrier/ABCA-I promoter fragment that is produced by this digestion corresponding to the BsiWI-SpeI fragment by the excision of λ sub-clone of the ABCA-1 genome sequence of the nucleotide 1-1534 of SEQ ID No:3.The plasmid pAPR1 that obtains transcribes under the control 1.75kb people ABCA-1 promoter sequence, coding luciferase reporter gene.
Contrast or transfection are kept among the DMEM that contains 10% hyclone to the pAPR1 plasmid wisas of RAW 264.7 cell fusion culture medium.Each Kong Zhongyong pGL3-Basic, pGL3-promoter or pAPR1 DNA in 12 hole wares (1 μ g), luciferase plasmid DNA (1 μ g) and 12 μ l Geneporter reagent (Gene Therapy Systems, San Diego, Calif.; Catalog number #T201007) transfection is 5 hours.In addition, add 0.1 μ g pCMV β plasmid DNA, (Clontech, Palo Alto, Calif., catalog number #6177-1) contrast as transfection efficiency.After 5 hours, in acetylation LDL (100 μ g/ml) existence or not, with the DMEM/BSA replacement culture medium of serum-free, incubation 24 hours.
After the transfection, with the dissolving 1 time in the molten born of the same parents' agent of 70 μ l cells (Promega, Madison, Wis., catalog number #E3971) of the cell in each hole, experience 1 freeze-thaw circulation, by with 12,000g made molten born of the same parents' thing clarify in centrifugal 5 minutes.After centrifugal, with 100 μ l luciferase assay reagent (Promega, Madison, Wis.; Catalog number #E1501) joins in the molten born of the same parents' thing of 10 μ l.Measure the luciferase activity of each molten born of the same parents' thing with light unit with luminometer.In addition, by manufacturer's (Tropix Inc., Bedford, Mass. catalog number #BL100G) explanation, measure the betagalactosidase activity of each molten born of the same parents' thing with the chemical luminescent detecting reagent that provides in the Galacto-light test kit.Divided by the beta galactosidase value that records, measure the normalized luciferase activity of each molten born of the same parents' thing by the luciferase activity value, report with relative light unit.
In this was measured, The compounds of this invention confirmed that ABCA-1 gene expression increases.
Embodiment 66: measure human apolipoprotein A1 protein expression
The effect of The compounds of this invention to ApoA 1 protein level of the endogenous APO AI gene expression in by CaCO2 cell, people's enterocyte system or Hep G2 cell, human liver cell being measured in this research.The compounds of this invention is dissolved in the appropriate solvent, offers CaCO2 or Hep G2 cell in containing the serum cell culture medium then, send back in the incubator for tissue culture, cultivated 12,24,36 or 48 hours down at 37 ℃.With the culture medium flushing cell that does not contain serum,, dissolve subsequently cell fixation, with commercially available human apolipoprotein A1 antibody (mouse anti human ApoA 1 antibody for example, Intracel Resources LLC, Frederick, MD, USA) existence of detection ApoA 1.Observe the cell ApoA 1 protein expression abundance of handling with The compounds of this invention and only use difference between the expression abundance of solvent processing cell.By with about 0.1pmol to each chemical compound of the variable concentrations that is up to about 100mmol by 2 times of concentration amplitude repeated experiments, measure and be used to detect its apolipoprotein and express the optium concentration of regulating active each chemical compound.Detecting with the bonded antibody number of times increase of cell confirmation chemical compound induces ApoA 1 to express increase.
Embodiment 67: measure the ApoA-1 promoter and induce
With CaCO2 or Hep G2 cellular exposure in the The compounds of this invention of valid density.Use standard technique, with reporter gene member, pAI.474-Luc and pRSV-B tilactase transfectional cell together, pRSV-B tilactase monitoring transfection efficiency.PAI.474-Luc is the member that makes up with conventional molecular biotechnology, contains-474 to-7 the rat APO AI promoter nucleotide that is fused to reporter gene, and this member is the luciferase (Luc) (U.S. Patent application 10/222,013) of Lampyridea.The compounds of this invention is dissolved in appropriate solvent (for example DMSO), adds culture medium then, kept 16 hours.When processing finished, harvesting was measured luciferase activity with standard available luciferase assay scheme.Also measure and be exposed to the proteic content of its APO AI in the culture medium of cell after 36 hours with the western blot analysis method.The luciferase activity increase shows that The compounds of this invention has ApoA 1 induced expression activity in molten born of the same parents' thing of cell or the exhausted culture medium.
Embodiment 68: measure the AGCCCCCGC sequential element and induce
With CaCO2 or Hep G2 cellular exposure in the The compounds of this invention of valid density.Use earlier standard technique, with contain 9 nucleotide 5 as enhancer element '-the reporter gene member transfectional cell (Kilbourne etc. of one or more templates of AGCCCCCGC-3 ', JBC, 270 (12): 7004-7010,1995), can be connected with promoter (for example thymidine kinase (TK) promoter), can with the pRSV-B tilactase together with reporter gene (luciferase for example, CAT or ApoA 1 gene) connect, pRSV-B tilactase monitoring transfection efficiency (as U.S. Patent application 10/222,013 explanation like that).Then The compounds of this invention is dissolved in appropriate solvent (for example DMSO), adds culture medium then, kept 16 hours.When processing finished, harvesting was measured reporter gene activity with the standard available assay method.Reporter gene activity increase or reduce show The compounds of this invention have adjusting from contain 9 nucleotide sequences 5 '-ability of the promoter transcription of AGCCCCCGC-3 ', it is believed that it comprises the egr-1 response element.Therefore, The compounds of this invention can be used for treating active relevant disease, disease or obstacle with egr-1.
Embodiment 69: measure the vasodilatory activity of chemical compound with ring test
Determine chemical compound usefulness provided by the invention with the isolating vascular ring of standard.Under 37 ℃, the New Zealand white rabbit thoracic aortic ring is suspended in pH 7.4 buffer, add the 10g load in advance for every ring.After the balance 2 hours, these ring norepinephrine preshrinking.Add the organ bath measurement by the The compounds of this invention that concentration is increased progressively continuously and induce loose gram number, set up the dose effect curve of each chemical compound.Make positive control with sodium nitroprusside and nitroglycerin.
Also, measure the inhibition that epinephrine causes contraction, in separating rat aorta, measure hemangiectasis activity by in the tissue for preparing in (J.Pharmacol.Exp.Therap.252,915,1990) described methods such as pressing Reynolds.
By adding the loose increase that The compounds of this invention causes, prove the hemangiectasis activity and the treatment of these chemical compounds or prevent many and hypertension diseases associated, for example effectiveness of cardiovascular disease.
Embodiment 70: measuring N O supplies with
Be the effectiveness of proof The compounds of this invention, The compounds of this invention is dissolved in the phosphate buffer of appropriate solvent and pH 7.4, incubation in 37 ℃ of water-baths as the nitrogen oxide releasing agent.By using the inert nitrogen rinse solution, then newly-generated NO is purged the theory of evolution luminescence detector, every 4-7 minute signal integration, periodic measurement NO rate of release with generation.With respect to negative control, NO discharges increase, potential suitable negative control is for example hydroxylating but not the same chemical compound of nitric acid esterified form, confirms that NO discharges active and treatment or prevention obstacle, disease or the disease relevant with hypertension, for example effectiveness of cardiovascular disease.
Embodiment 71: measure the antioxidant effectiveness
By with disclosed colour measurement method (FOX algoscopy based on dyestuff, referring to Zadeh, " Methods in Enzymology ", 300,58 (1999)) measurement proves the The compounds of this invention antioxidant properties by the degree of copper catalysis autoxidation low density lipoprotein, LDL hydrogen peroxide.Make positive control with only containing the sample that LDL and copper sulfate do not contain test substances, with the equal mixture contrast that contains test substances.
The phosphate-buffered aqueous solution pH-7.4 of human ldl (Sigma Chemical Company L2139) is mixed with copper sulfate.With the The compounds of this invention of effective dose under 25 ℃ or 37 ℃ under normal pressure incubation, finish oxidation, between zero-time point and 3-20 hour incubation period,, measure hydrogen peroxide with the FOX algoscopy from the mixture sampling.Read sample with the microtitration card reader.The hydrogen peroxide that the FOX algoscopy is measured reduces the effectiveness of the antioxidant activity that shows The compounds of this invention and their treatments or prevention obstacle, disease or the disease relevant with oxidation, or benefited from the use antioxidant.From the example of the benefited this disease of antioxidant therapy is cardiovascular disease.
Embodiment 72: measure antioxidant activity by the LDL oxidimetry:
Can use Esterbauer method (Esterbauer, H., Striegl, G., Puhl, H., Rotheneder, M., " Continuous monitoring of in vitro oxidation of humanlow density lipoprotein " (external continuous monitoring oxidation human ldl), FreeRadic.Res.Commun, 1989; 6 (1): 67-75), some improvement is as follows:
Chemical compound and suitable solubilizing agent are dissolved in phosphate buffer (PBS, 0.15M NaCl-0.05M sodium phosphate buffer-pH 7.4).Write down definite concentration (about 30-60 μ g/mL extract to be measured).In 100 these solution of μ L, add 900 μ L oxidation buffer liquid (by people LDL (120 μ L5mg/mL solution, d=1.019-1.063g/mL wherein, available from PerImmune, Rockville, Md.) and the 8mL PBS formulations prepared from solutions of copper sulfate (20 μ L 10mM aqueous solution)).Also prepare blank sample with 100 μ L PBS and the preparation of 900 μ L oxidation buffer liquid.Then each solution is transferred to the 1cm quartz colorimetric utensil, this cuvette is put into thermostat (37 ℃).With the HP-8452A diode array spectrophotometer at 234nm (OD 234) measuring light density, measured once in per 5 minutes.With the lag time of the maximum value calculation oxidation of first derivant of optical density curve.Measure the standard substance that contain ascorbic acid during each mensuration.
Embodiment 73: measure and contrast HDL, LDL, VLDL and triglyceride level
In morning every day, give the male Sprague-Dawley rat or the female fat Zucker rat chemical compound of food raising or give solvent separately, continuous 7 days by per os feed 1.5% carboxymethyl cellulose/0.2% tween 20 (administration solvent).In whole research, weigh animal every day, lets alone freely to eat rodent food and drinking-water.An eye frame blood sample is gathered in fasting after 6 hours before the beginning administration, also blood sample collection after the 7th administration.After the 7th administration, put to death animal evening, measure serum total cholesterol and triglyceride, lipoprotein cholesterol curve, VLDL and add and mix LDL cholesterol (being called the apo B or the non--HDL cholesterol that contain lipoprotein cholesterol again), HDL cholesterol, and HDL cholesterol and VLDL add the ratio of LDL cholesterol.
Embodiment 74: measure and contrast the reaction that people HDL, LDL, VLDL and triglyceride level give chemical compound
Every day administration of human experimenter The compounds of this invention.Monitor other diet picked-up, be consistent between the individuality.Before giving chemical compound, weekly at blood sample collection on the 0th, continue the 3-6 month.Measure serum total cholesterol and triglyceride, lipoprotein cholesterol curve, VLDL and add mixing LDL cholesterol (being called the apo B or the non--HDL cholesterol that contain lipoprotein cholesterol again), HDL cholesterol, HDL 2And HDL 3Cholesterol components, and HDL cholesterol and VLDL add the ratio of LDL cholesterol, with the commercially available cholesterol test method of standard, for example (AL), it can repeatability measure these parameters from a small amount of human blood sample for Atherotech Inc, Birmingham in the VAP test.Perhaps, can pass through Kulkarni method 1997.J.LipidRes.38:2353-64 such as () Kulkarni or measure the HDL in the blood by Gidez method 1982.J.Lipid Res.23:1206-23 such as () Gidez 2And HDL 3The total HDL of increase that in this blood test, records, increase HDL 2, the The compounds of this invention that reduces total LDL, reduce VLDL, reduce triglyceride or increase HDL/ T-CHOL or HDL/LDL ratio is used for the treatment of and cholesterol or lipid diseases associated.
Embodiment 75: measure the atherosclerotic lesions size with Dan Baijutang-combination-defective LDL
Available nucleic acid member produces mice expressing protein polysaccharide-combination-defective LDL.Continuous 17 weeks give the food that transgenic mice contains 1.2% cholesterol, 0.5% cholate and 20% fat.Put to death mice then, aortic perfusion is fixed, wherein that whole aorta is smooth fixing with en face methods analyst, with Sudan IV dyeing, measure image analysis system (image-1/AT) analyze quantitative atherosclerotic degree with form.
Embodiment 76: measure live animal hypertension and descend
By containing the heparinized saline conduit pressure transducer is connected with right carotid.The record mean arterial pressure and the rhythm of the heart.Use pentobarbital,, adopt littler injected dose anesthetized rat in addition as needs with predose 35mg/kg body weight.Chemical compound is dissolved in pharmaceutical carrier (for example 5% glucose USP of Abbott), injects to rat by right femur venous duct.Spendable positive control comprises sodium nitroprusside and NaNO 2, and available NaNO 3Make negative control.The result shows that chemical compound provided by the invention is effective depressor, its significantly blood pressure lowering.Should reach the blood pressure drops peak value at short notice, for example inject about 1 minute after, and after this blood pressure should begin to rise very soon, should recover fully by blood pressure in about 10-15 minute.
Embodiment 77: the minimizing degree of measuring hypercholesterolemia rabbit arterial damage back restenosis
Can use U.S. Patent number 5,595, in 974 and Goodman at United States Patent (USP) 6,022, the 901 Tomaru methods that further describe are estimated the effectiveness that The compounds of this invention prevent hypercholesterolemia rabbit restenosis.
Embodiment 78: be used to prevent people's restenosis
Can implement (1997) such as Tardif described in 901 by Goodman at United States Patent (USP) 6,022, New England J.Med.337 (6): the method for 365-67, difference are to substitute the trans-resveratrol check with our chemical compound.
Embodiment 79: measure platelet anticoagulant collection activity
Can be by (Science 220,517,1983) described methods such as Bertele, in-vitro evaluation is to the platelet anticoagulant collection activity of stimulated by thrombin human blood platelets.
Embodiment 80: measure ADP is induced the agglutinative influence of rabbit platelet
Platelet aggregation test: the syringe with silicon-coating is gathered the Sanguis Leporis seu oryctolagi sample by the rabbit heart puncture.This blood sample is mixed with the ratio of 3.8% sodium citrate with 9: 1, by 1,000rpm rotation 6 minutes.1ml is rich among the 2ml Xiao Chi that hematoblastic blood plasma is transferred to silicon-coating, mixes, write down transmittance (Ti) with spectrophotometer.Add 0.02ml ADP (10 μ M), stir, per minute once writes down the transmittance that contains platelet blood plasma, obtains maximum transmission degree (Tm) in 10 minutes.Blood sample with 3000rpm rotation 45 minutes, is write down transmittance.
Embodiment 81: measure the inductive thrombocytopenic influence of collagen in the body
With pentobarbital sodium (65mg/kg, Vet Labs, Limited, Inc., Lenexa, KA) anesthesia male rat (Charles River, CRL:CD (SD), 400-450g).Prepare two otch, expose two jugular veins.(Harvard Apparatus, South Natick Mass.) with the 5cc syringe, press 0.39ml/min speed with 19g butterfly (butterfly), and test compounds or solvent are infused into left jugular vein, continue 3min with infusion pump.Behind infusion chemical compound/solvent 2min, (Helena Laboratories, Beaumont TX) are expelled to right jugular vein with collagen (60 μ g/kg) with the 1ml syringe.Open body cavity, expose caval vein, blood sample collection.Behind the injection collagen 1min, stop the infusion chemical compound, add the 3cc syringe of 150 μ M indomethacins from caval vein blood sample collection (in 30 seconds) with containing 0.3mg 4.5%EDTA/Tris (0.1M) (pH 7.35).By being rich in hematoblastic blood plasma (PRP) with 126 centrifugal blood sample 10min preparations of .g.On the Coulter register, in 20ml Isoton.RTM.III, 5 μ l PRP are counted.Compare by the platelet count that will in handling animal, add up and the platelet count that does not give the collagen animal and the platelet count that gives the animal of solvent and collagen, calculate collagen-induced agglutinative inhibition percentage rate.Suppress to estimate usefulness according to collagen-induced thrombocytopenia.
Embodiment 82: antipsoriatic effectiveness in the measuring body
The infection site of suffering from psoriatic people patient contains the topical preparation of The compounds of this invention.The control formulation that does not contain The compounds of this invention is applied on this patient's the equal position.After the administration the 3rd day and the 7th day, by analyze with the position of using control formulation relatively, the inflammation at administered compound position is improved and proliferative cell reduces, the effectiveness of mensuration chemical compound.
Embodiment 83: measure protein kinase and suppress
In suitable buffer, The compounds of this invention and radiolabeled ATP, suitable protein kinase and suitable substrate are mixed.Behind the incubation,, quantitatively add ATP difference, its measurement and the downtrod protein kinase amount of comparing of substrate with scintillation counter by some cessation reaction on filter paper.
Embodiment 84: the inhibition of measuring neutrophil activation
(Tudan.1999.Biochem.Pharmacol.58:1869-80 tests The compounds of this invention with the Tudan scheme.This test confirms that test compounds suppresses for example ability of the neutrophil activation that causes of fMLP of crystallization and chemical inhibitor.
Embodiment 85: the inhibition of the inflammation that measurement TPA causes
By improved Marks method 1976.Cancer Res.36:2636 such as () Marks test The compounds of this invention, with the anti-effectiveness of proof chemical compound with 12-O-tetradecanoyl phorbol-inflammation that 13-acetas (TPA) brings out.Compound administration on mouse ear, is used TPA subsequently.With the perforation of mouse ear biopsy method, weigh after 4 hours, measure edema, with the biopsy method perforation contrast of another ear that does not use chemical compound.
Embodiment 86: measure CO X-1 suppresses
By Van der Ouderaa method (Van der Ouderaa.1982.Methods Enzymol.86:60) test The compounds of this invention.By arachidonic acid being added in the mixture of 0.1M sodium phosphate (pH 7.4) solution, 1.0mM phenol, 0.01mM hemin and the COX-1 enzyme that contain test compounds initiation reaction.
Embodiment 87: the inhibition of the inflammation that the measurement carrageenin brings out
By Slowing method (1994.J.WrhnophEMxol.43:9 such as Slowing), on the Wistar rat, test The compounds of this invention.With the Freund adjuvant percutaneous injection in the animal tail.After 7 days, give test compounds, after 1 hour, the normal saline suspension of carrageenin is given in the left back pawl.By water volume tracing measuring claw volume, with the contrast contrast.
Embodiment 88: measure the cancer chemoprevention activity
By Mondal method 1976.Cancer Res.36:2254-2260 such as () Mondal, handle C3H/10T1/2 with The compounds of this invention and clone 8 cells (ATCC).Cell in the culture medium was handled 24 hours with 3-MECA, incubation 5 days in fresh culture medium subsequently, washing.Subsequently TPA is added and contain or do not contain in the culture medium of test compounds.After realizing merging for 7 weeks, fix, show the focus (foci) that II and III type transform,, suppress the effectiveness that two phases transformed with the proof test compounds with its scoring with Giemsa dyeing with methanol.
Embodiment 89: before one or more hydroxyls are by one or more nitrooxy displacements, and the method for synthesizing the fluorine derivative of the The compounds of this invention that comprises stilbene, polyhydric phenols and flavone compound
Because may need to be used as with the raising chemical compound effectiveness of medicine with one or more hydroxyls of fluorine displacement The compounds of this invention, method (Cramerand Coffman according to Cramer and Coffman, 1961 J Org.Chem.26:4164), provide a description the embodiment that how replaces the hydroxyl that is connected with aromatic ring at this with fluorine.This method is highly suitable for any hydroxyl with any chemical compound of fluorine displacement the present invention, need not those skilled in the art and carries out over-drastic experiment.Because described in this embodiment some harshness of fluorination conditions, so can be by nonfluorinated prepares the yield that chemical compound improves some The compounds of this invention by the structure body.When chemical compound with fluoro for hydroxyl, and when replacing other hydroxyl (for example in embodiment 1-59), should finish the fluorine additive reaction earlier with one or more nitrooxies, carry out the nitrooxy additive reaction then.
Below the fluorine derivative of synthesizing resveratrol is described in reaction.
In the stainless steel-lined autoclave of 400mL capacity, add 250mmol 5-[(E)-2-(4-hydroxyl-phenyl)-vinyl]-benzene-1, and the 3-diphenol (synonym: resveratrol), evacuation.Add 500mmol tetrafluoro sulfur oxide, the vibrations reactant mixture heated 9 hours down at 150 ℃.To be mainly the gaseous product of sulfuryl fluoride-49 ℃ to-44 ℃ distillations down.With residue with 5% sodium hydrate aqueous solution and water washing.When distillation, find that this liquid contains following complete and partially fluorinated mixture of products: 3-fluoro-5-[(E)-2-(4-hydroxyl-phenyl)-vinyl]-phenol, 5-[(E)-2-(4-fluoro-phenyl)-vinyl]-benzene-1,3-diphenol, 4-[(E)-2-(3,5-two fluoro-phenyl)-vinyl]-phenol and 1,3-two fluoro-5-[(E)-2-(4-fluoro-phenyl)-vinyl-benzene.By the silica gel column chromatography purification with separate various products.
After separating the fluorine derivative of resveratrol, can be by the described further modified compound of embodiment 1-59 to contain nitrooxy.This method need not undo experimentation and fluorine is added on any The compounds of this invention.
Embodiment 90: synthetic containing by containing-(CO) method of two aromatic ring polyhydric phenols connecting of the linking group of NH-
The polyphenol compound of the present invention's expectation comprises that containing two passes through the interconnective aromatic compound of linking group, and wherein said linking group contains following group:
By this reaction, with the easily synthetic following general formula polyphenol compound of the raw material that is easy to obtain
Wherein X is NH,
And R 1-10Independently be selected from H or OH separately.
These chemical compounds of the present invention are as midbody compound, NItroxyderivatives described in available their synthetic subsequently embodiment 1-59 and the NItroxyderivatives that can further modify, fluorine phosphate-based to comprise, ester group and other modification group.Synthetic intermediate examples of compounds N-(3,5-dihydroxy-phenyl)-4-hydroxyl-Benzoylamide by the following method, available subsequently it prepare its NItroxyderivatives.
In dry DMF (15ml) solution of 4-hydroxy-benzoic acid (6mmol), add EDCI (9mmol), HOBt (9mmol) and triethylamine (12mmol).After at room temperature stirring 24 hours, be added dropwise to 5-amino-benzene-1, the 3-diphenol continues reaction 48 hours under room temperature, argon.Add entry (300ml) then, mixture is stirred 5min.Then product is extracted with ethyl acetate (5*50ml).The organic extract liquid that merges is washed with saline (40ml),, filter, remove and desolvate through dried over sodium sulfate.Finish purified product N-(3,5-dihydroxy-phenyl)-4-hydroxyl-Benzoylamide by silica gel column chromatography.
Perhaps, with 5-amino methyl-benzene-1, the 3-diphenol replaces 5-amino-benzene-1, and the 3-diphenol reacts, and causes synthetic N-(3,5-dihydroxy-benzyl)-4-hydroxyl-Benzoylamide.Should syntheticly prove that the wherein X of synthetic this embodiment was NHCH 2The method of general formula compound.Similarly, as proof, Pyrogentisinic Acid's alkyl modification can cause the identical modification to the linking group of products therefrom.
By for example fluoridize, bromination, chlorination or acetylation aryl or by the heteroaromatic aryl, or pass through C 1-18Alkyl, or by bicyclic aryl etc. replaced with the R group that the amino reagent that provides is connected should syntheticly be described and (promptly replaced 5-amino-benzene-1, the benzene 1 of 3-diphenol, 3-diphenol group), can generate the product of suitable modification, this will be apparent to those skilled in the art.
Best, can be used as the midbody compound that is used for the NO of confession synthetic of the present invention, NItroxyderivatives by the synthetic product of this method.
Embodiment 91: synthetic containing by containing-method of two aromatic ring polyhydric phenols that the linking group of C-NH-connects
The polyphenol compound of the present invention's expectation comprises the chemical compound that contains by interconnective two aromatic rings of linking group, and wherein said linking group contains the carbon atom singly-bound that is connected with nitrogen-atoms.By this reaction, by the easily synthetic following general formula polyphenol compound of the raw material reagent that is easy to obtain.
Wherein X is CH 2, Y is NH, or X is NH, Y is CH 2,
And R 1-10Independently be selected from H or OH separately.
These chemical compounds of the present invention are as midbody compound, NItroxyderivatives described in available their synthetic subsequently embodiment 1-59 and the NItroxyderivatives that can further modify, fluorine phosphate-based to comprise, ester group and other modification group.Synthetic intermediate examples of compounds 5-(4-hydroxyl-benzylamino)-benzene 1 by the following method, the 3-diphenol, available it prepare its NItroxyderivatives.
With 5-amino-benzene-1,3-diphenol (1.5mmol) adds in benzene (40ml) solution of 4-hydroxyl-benzaldehyde (1.5mmol), under argon, with the Dean-Stark trap mixture heated is refluxed 24 hours.Then reactant mixture is concentrated to remove benzene fully, residue is dissolved in methanol (15ml).Stir simultaneously, in 30min, divide three parts to add sodium cyanoborohydride (3mmol), at room temperature, with reactant mixture restir 1 hour.In reactant mixture, add the saturated NaCl solution (100ml) that contains 37%HCl then.Reactant mixture is extracted with ethyl acetate (3*50ml).The organic layer that merges, concentrates through dried over sodium sulfate with saline (10ml) washing, obtains crude product 5-(4-hydroxyl-benzylamino)-benzene 1, and the 3-diphenol is further purified it by silica gel column chromatography.
Perhaps, replace 4-hydroxyl-benzaldehyde to react, cause synthetic 5-[2-(4-hydroxyl-phenyl)-ethylamino with 4-hydroxyl-phenyl-acetaldehyde]-benzene-1, the 3-diphenol.Should syntheticly prove that the wherein X of synthetic this embodiment was (CH 2) 2, Y is the method for the general formula compound of NH.Similarly, as proof, Pyrogentisinic Acid's alkyl modification can cause the identical modification to the linking group of products therefrom.
Best, can be used as the midbody compound that is used for the NO of confession synthetic of the present invention, NItroxyderivatives by the synthetic product of this method.
Embodiment 92: the synthetic method that contains by-two aromatic ring polyhydric phenols that the CO-linking group connects
The polyphenol compound of the present invention's expectation comprises and containing by interconnective two aromatic compounds of linking group that wherein said linking group contains the carbon atom singly-bound that is connected with oxygen atom.By this reaction, by the easily synthetic following general formula polyphenol compound of the raw material reagent that is easy to obtain,
Wherein X is CH 2, Y is an oxygen,
And R 1-10Independently be selected from H or OH separately.
These chemical compounds of the present invention are as midbody compound, NItroxyderivatives described in available their synthetic subsequently embodiment 1-59 and the NItroxyderivatives that can further modify, fluorine phosphate-based to comprise, ester group and other modification group.Synthetic intermediate examples of compounds 5-(4-hydroxyl-phenoxymethyl)-benzene 1 by the following method, the 3-diphenol, available it prepare its NItroxyderivatives.
At argon with under stirring, with the anhydrous N of tertiary butyl chloride for dimethylsilane solid (25mmol) adding 4-hydroxyl-benzaldehyde (17mmol) and imidazoles (42.5mmol), dinethylformamide (100ml) solution.After 4 hours, in reactant mixture impouring water, use extracted with diethyl ether.With organic extract liquid water and salt water washing, drying is concentrated into coloured grease.Filtering by silicagel pad, is eluant with 20% ethyl acetate-hexane, obtains silyl ether 4-(tert-butyl group-dimethyl-silicon alkoxyl)-benzaldehyde.With 4-(tert-butyl group-dimethyl-silicon alkoxyl)-benzaldehyde (14mmol) and-dichloromethane (100ml) vlil of chlorine benzylhydroperoxide (20mmol) 2 hours, at room temperature place then and spend the night.Then reactant mixture is extracted in the ether, subsequently with organic layer sodium hydrate aqueous solution (1M), water and salt water washing, drying, reduction vaporization obtains solid.Its preadsorption to silica gel, is filtered by the silica gel plug experience then fast.With the formic acid esters that obtains, promptly methanol (70ml) solution of formic acid 4-(tert-butyl group-dimethyl-silicon alkoxyl)-phenylester adds in the potassium carbonate (10mmol).After 20 minutes, add 1-bromomethyl-3,5-two-(tert-butyl group-dimethyl-silicon alkoxyl)-benzene (14mmol is basically by the silylation guard method preparation identical with above formic acid 4-(tert-butyl group-dimethyl-silicon alkoxyl)-phenylester).After 6 hours, the volume of reactant mixture reduces, and adds entry, with solution aqueous hydrochloric acid solution (1M) acidify.It is used extracted with diethyl ether, make ether extraction liquid pass through Pearson method (1967J Org Chem 32:2358 such as Pearson) and carry out post processing.Make eluent with 2-80% ethyl acetate-hexane, the gradient elution dry chromatography obtains 1,3-two-(tert-butyl group-dimethyl-silicon alkoxyl)-5-[4-(tert-butyl group-dimethyl-silicon alkoxyl)-phenoxymethyl]-benzene.Make 1,3-two-(tert-butyl group-dimethyl-silicon alkoxyl)-5-[4-(tert-butyl group-dimethyl-silicon alkoxyl)-phenoxymethyl]-tetrahydrofuran solution of benzene handles with four-n-butyl ammonium fluoride trihydrate.3.5 after hour, add entry and ether.Water layer with aqueous hydrochloric acid solution (1M) acidify, is extracted with ether again.By the Pearson method organic extract liquid is carried out post processing then.Filter by silica gel plug (20% ethyl acetate-hexane), obtain grease.Grind with hexane, in acetone-the dry ice bath, cool off this grease crystallization simultaneously.By dichloromethane-hexane recrystallization, obtain product 5-(4-hydroxyl-phenoxymethyl)-benzene 1, the 3-diphenol is further purified it by silica gel column chromatography.
Best, can be used as the midbody compound that is used for the NO of confession synthetic of the present invention, NItroxyderivatives by the synthetic product of this method.
Embodiment 93: synthetic containing by containing-method of two aromatic ring polyhydric phenols that the linking group of C=N-connects
The polyphenol compound of the present invention's expectation comprises and containing by interconnective two aromatic compounds of linking group that wherein said linking group contains the two keys of the carbon atom that is connected with nitrogen-atoms.By this reaction, by the easily synthetic following general formula polyphenol compound of the raw material reagent that is easy to obtain,
Figure A20048003609900901
Wherein X is CH, and Y is N, or X is N, and Y is CH,
And R 1-10Independently be selected from H or OH separately.
These chemical compounds of the present invention are as midbody compound, NItroxyderivatives described in available their synthetic subsequently embodiment 1-59 and the NItroxyderivatives that can further modify, fluorine phosphate-based to comprise, ester group and other modification group.Synthetic intermediate examples of compounds 5-{[(E by the following method)-4-hydroxyl-phenylimino]-methyl-benzene 1, the 3-diphenol, available it prepare its NItroxyderivatives.
In Dean and Stark device, with 3, toluene (5ml) vlil of 5-dihydroxy-benzaldehyde (1mmol) and 4-amino-phenol (1mmol) 16 hours.After the solvent vacuum removed, with product 5-{[(E)-4-hydroxyl-phenylimino]-methyl }-benzene 1, the 3-diphenol is further purified by recrystallizing methanol with by silica gel column chromatography.
Best, can be used as the midbody compound that is used for the NO of confession synthetic of the present invention, NItroxyderivatives by the synthetic product of this method.
Embodiment 94: synthetic containing by containing-universal method of the stilbene (with the dihydro stilbene) of two aromatic rings that the linking group of C=C-connects
The stilbene chemical compound of the present invention's expectation comprises and containing by interconnective two aromatic compounds of linking group that wherein said linking group contains the two keys of the carbon atom that is connected with another carbon atom.By this reaction, by the easily synthetic following general formula stilbene chemical compound of the raw material reagent that is easy to obtain,
Figure A20048003609900911
Wherein X is CH, and Y is CH,
And R 1-10Independently be selected from H or OH separately.
These chemical compounds of the present invention are as midbody compound, NItroxyderivatives described in available their synthetic subsequently embodiment 1-59 and the NItroxyderivatives that can further modify, fluorine phosphate-based to comprise, ester group and other modification group.By the following method synthetic intermediate examples of compounds resveratrol (synonym: 5-[(E)-2-(4-hydroxyl-phenyl)-vinyl]-benzene 1, the 3-diphenol), available it prepare its NItroxyderivatives.
In sealed tube, with 3, the mixture of 5-dihydroxy-benzyl-bromine (10mmol) and NSC 6513 (30mmol) heated 8 hours in 180 ℃ of oil baths.After the mixture cooling, excessive NSC 6513 vacuum is removed.Residue obtains product (3,5-dihydroxy-benzyl)-dimethyl phosphonate behind quick short column chromatography purification.At room temperature, to the well-beaten 2ml CH that also contains fresh powdery KOH (2mmol), hexaoxacyclooctadecane-6-6 (0.1mmol) of (3,5-dihydroxy-benzyl)-dimethyl phosphonate 2Cl 2Add aromatic aldehyde 4-hydroxyl-benzaldehyde (1mmol) in the suspension of solution.After mixture stirred 6 hours, with mixture 15ml CH 2Cl 2Dilution, water (10ml) and saline (2*10ml) washing.Organic layer is through dried over mgso, vacuum concentration.Residue is dissolved in 2mlCH 2Cl 2
In this solution, add GirardShi reagent T (0.5mmol) and AcOH (5mmol), at room temperature, the mixture that obtains was stirred 2 hours.The filtering insoluble matter with the filtrate vacuum concentration, is dissolved in 15ml EtOAc with residue.Solution with saline (3*10ml) washing, through dried over mgso, is removed the solvent vacuum, obtained the E and the Z isomer mixture of resveratrol.Add the iodine of catalytic amount in heptane (5ml) solution of this mixture, reflux is 12 hours then.Reactant mixture with the dilution of 20ml ether, is washed with saturated aqueous solution of sodium bisulfite (10ml) and saline (2*10ml).Organic layer is through dried over mgso, and vacuum concentration obtains the E-resveratrol that needs.
Best, with the synthetic any stilbene chemical compound of this method, it is the midbody compound for NO, NItroxyderivatives of synthetic the present invention's expectation.
The dihydro stilbene is the derivant of corresponding stilbene, and difference is to have singly-bound between two carbon atoms of linking group, preferably can be synthetic by the stilbene parent compound.Universal method is as follows.
Under 40psi, in the presence of 10% palladium on carbon (60mg), make ethanol (120ml) solution hydrogenation 18-24 hour of stilbene (1mmol).Catalyst is removed by filter by Celite pad,, obtain dihydro stilbene derivant the solvent evaporation in the filtrate.
The most handy this method is synthesized any dihydro stilbene chemical compound, and it is the midbody compound for NO, NItroxyderivatives of synthetic the present invention's expectation.
Embodiment 95: the method for synthetic phosphate ester-derivant of the present invention
Best, can make phosphate-based replacement hydroxyl replace some The compounds of this invention, because phosphate-based metabolism and the half-life that changes chemical compound in the serum.As the embodiment of the phosphate derivative of described synthesizing resveratrol herein, but be not limited to this embodiment, be preferably in other hydroxyl and taken place after fluorine, ester and nitrooxy (being nitrate or the itrate group) displacement.
Press earlier derivant (for example 3-[(E)-2-(4-hydroxyl-phenyl)-vinyl of the single nitrooxy replacement of synthetic described in the embodiment 1, separation and purified resveratrol]-5-nitrooxy-phenol).With derivant (4g) and N that single nitrooxy replaces, anhydrous acetonitrile (30ml) solution of N-(dimethylamino) pyridine (0.2g) is cooled to-10 ℃, adds carbon tetrachloride (5 equivalent) and DIEA (2 equivalent).Under-10 ℃ and argon, mixture is stirred 30min, add phosphate dibenzyl ester (1 equivalent), with solution stirring 12 hours, impouring 0.5M potassium dihydrogen phosphate then.With the mixture ethyl acetate extraction, the solvent vacuum of organic facies is removed, obtain coloured grease.Make its experience rapid column chromatography (4: 1 hexane/ethyl acetate), phosphate product is reclaimed, be coloured grease.
Under 0 ℃, in anhydrous methylene chloride (15ml) solution of this phosphate product, add bromo trimethyl silane (2 equivalent), mixture was stirred 2 hours.Add entry (10ml), with solution stirring 1 hour, with the ethyl acetate washing, with the water lyophilization to white solid.In this solid ethanol (30ml) solution, add Feldalat NM (0.6g), suspension was stirred 12 hours.The solvent vacuum is removed, the coloured grease that obtains is water-soluble.Solution is washed with ethyl acetate, and lyophilization then obtains the colorless solid that high yield, high-purity contain the Verakanol derivative mixture with phosphate-based, hydroxyl and nitrooxy.By silica gel column chromatography, the derivant of needs is separated and purification.
Best, available this synthetic method makes phosphate ester replace hydroxyl to replace any chemical compound of the present invention.
Embodiment 96: the method for synthetic acetyl derivative of the present invention
Best, can make acetyl group replace hydroxyl to replace some chemical compound of the present invention, because some acetyl group can change the lipotropy degree, thereby change the metabolic rate and the half-life of chemical compound in the serum.For example, with one or more hydroxyl displacements of resveratrol, the acetic ester derivative of formation resveratrol reduces metabolic rate and the prolong half-life in the serum.Describe herein and be preferably in that to add one or more nitrates (be ONO 2Or itrate group) group and add phosphate-based before (like this if desired, but fluoridizing the back) acetic ester derivative of synthesizing resveratrol.
Resveratrol (0.5mmol) is dissolved in anhydrous methylene chloride (5ml).Add excessive anhydrous pyridine, add the 1mmol acetic anhydride subsequently.The solution that obtains was at room temperature stirred 5 hours.Reactant mixture is concentrated, be dissolved in dichloromethane (20ml) again.With organic layer with hydrogen chloride solution (0.1M, 10ml), sodium bicarbonate (saturated, 10ml) and the salt water washing.Organic layer is through dried over mgso, filters, and concentrates, and obtains the mixture of the resveratrol acetic ester derivative of high yield and purity, wherein 1,2 or all 3 hydroxyls replaced by acetate groups.Various products pass through the silica gel column chromatography purification and separate.
Also available acetyl halide (for example chloroacetic chloride) or activation acetas (for example N-hydroxy-succinamide ester) synthesis of acetic acid ester derivant.Use same procedure, replace acetic anhydride with other Acibenzolar or carboxylic acid halides, similarly other ester of synthetic The compounds of this invention.The example that can replace this type of ester of any hydroxyl on any chemical compound of the present invention expectation is described by following formula:
Wherein R can be C 1-18, aryl, heteroaryl and the optional derivant that replaces thereof.
Best, can be combined to step by carrying out nitrate described in the embodiment 1-59, the synthesis of acetic acid ester derivant separates and purification by silica gel column chromatography subsequently.
Embodiment 97: synthesize the methoxyl group of the The compounds of this invention that is used as midbody compound and the method for ethyoxyl derivant, by these midbody compound synthesizing nitryl oxygen radical derivatives, they are all chemical compounds of the present invention's expectation.
Preferably some chemical compound of the present invention can have the methoxyl group (OCH that offers the R group 3) or ethyoxyl (OCH 2CH 3), because known methoxyl group and ethyoxyl are lipophilic, thereby can change the drug disposition half-life and do not reduce its activity.The many methoxyl groups and the ethyoxyl derivant of known aromatic hydrocarbons (for example benzene, phenol etc.), and on market, obtain easily, or synthetic by the method for knowing easily.Therefore obtain being used to prepare the structure body of stilbene class and other polyhydric phenols easily, and can for example use among the embodiment 90-94.Therefore, can synthesize the polyhydric phenols and the stilbene of the application's definition, comprise methoxyl group (OCH so that the R group can be chosen wantonly independently 3) or ethyoxyl (OCH 2CH 3).
Embodiment 98: the method that proves the The compounds of this invention antifungal activity
With United States Patent (USP) 6,165, the method proof antifungal compound of the present invention of 998 explanations.Briefly, with about 10 6Individual Candida albicans (C.albicans) or saccharomyces cerevisiae (S.cerevisiae) cellular exposure continue 45 minutes in 25 μ g/ml antifungal compound of the present invention, do not stay detectable colony-forming units.
In addition, antifungal compound of the present invention is effective in murine systemic candidiasis model.Mean survival time of antifungal compound extended treatment mice of the present invention and time-to-live intermediate value.IP gives chemical compound, produces and the similar survival mode of oral positive control chemical compound fluconazol.Antifungal compound of the present invention and fluconazol all reduce the recovered bacterium colony of treatment animal kidney.When general candidiasis (Candida) mice infected that per os is set up, antifungal compound of the present invention is also effective.By measuring the time-to-live, curing percentage rate and kidney burden (burden), it is similar to fluconazol to prove that per os gives the effectiveness of chemical compound.Antifungal compound of the present invention is also effective to the systemic candidiasis that Candida albicans (C.albicans) bacterial strain of anti-fluconazol causes.
Embodiment 99: the method that proves the The compounds of this invention active anticancer
Press United States Patent (USP) 5,145, the method for 839 explanations is treated with following animal cancer model with The compounds of this invention, proves the active anticancer of The compounds of this invention.
By 10 every group, will have the O BALB C mice of lymphoma YC8 (ascitic type) and have Swiss mice (20-22g, the Charles River breeding) random packet of Ou Lixishi (Ehrlich) ascites cells.Accept respectively for every group:
Group I contrast, tumor cell and NaCl isosmotic solution (0.2ml/ mice, 2 times/day, i.p. approach);
Group II: the mice with tumor cell is accepted The compounds of this invention 0.2ml/ mice, and 2 times/day, by the i.p administration;
Group III: the mice with tumor cell is accepted The compounds of this invention: the 0.2ml/ mice, and 2 times/day, i.p. approach and i.p give chemotherapeutics;
Group IV: the mice with tumor cell is accepted The compounds of this invention: 0.2ml/ mice, 2 times/day, i.m administration.
The ascites tumour cell that will have the mice of these cells adds in the aseptic culture medium, keeps 15-20 days.With 0.1ml ascites suspension and 10ml buffer (pH 7.2): (NaCl 7.2g/l; Na 2HPO 44.3g/l and KH 2PO 40.4g/l) mix.Measure cell number (by the Malassez cell), with the cell suspending liquid dilution, so that make cell number near 40.000-50.000/ml.By the i.p. approach this suspension of 0.1ml is expelled in group I, II and the III mice and by the i.m. approach immediately and is expelled in the group IV mice.
Injection tumor cell after 48 hours: group II mice accept (i.p) 37 ℃ of heating, on microporous filter membrane filtering The compounds of this invention, treated in continuous 5 days; The continuous 5 days mixture process with one of The compounds of this invention and antibiotic (i.p) group III mices; Continuous 5 days group I mice (contrast) acceptance (i.p) isosmotic solution; Group IV mice was accepted (i.m) The compounds of this invention in continuous 15 days.After stopping treatment, observe mice 1 or 2 months.Only consider the surviving animals that physical qualification is good.Therefore as this test confirmation, The compounds of this invention useful as anti-cancer agents thing.
Embodiment 100: the method that proves the The compounds of this invention anti-diabetic activity
With United States Patent (USP) 6,410, the method proof The compounds of this invention hypoglycemic activity of 596 explanations.This test confirms The compounds of this invention C57BL/ks diabetes (db/db) mice, reduces the activity of plasma glucose levels in promptly art-recognized noninsulindependent diabetes (NIDDM) model.
Embodiment 101: the method that proves the The compounds of this invention antiviral activity
Interior evaluating agathis australis bisflavone (Robustaflavone) in Mus influenza model
Carry out experiment in the body, effective with the proof The compounds of this invention influenza infection that anti-experiment is brought out in no specificity cause of disease BALB/c mouse.United States Patent (USP) 6,399 press in these experiments basically, and the method for embodiment 11 explanations is carried out with The compounds of this invention replacement agathis australis bisflavone in 654.
Embodiment 102: preparation 5-nitrooxy-valeric acid 4-[5,7-two-(5-nitrooxy-valeryl oxygen base)-4-oxo-benzodihydropyran-2-yl]-phenylester
Figure A20048003609900961
Synthetic 5-nitrooxy-valeric acid
Under 40 ℃, with 5-bromo-valeric acid (180mg, 1mmol), (255mg, 1.5mmol) mixture in acetonitrile stirs silver nitrate.By thin layer chromatography (TLC) monitoring reaction.After the end, add dichloromethane, mixture is washed with water,, filter, concentrate through anhydrous sodium sulfate drying.Crude product is through column chromatography purification.
Figure A20048003609900971
Synthetic 5-nitrooxy-valeric acid (2,5-dioxo-pyrrolidine-1-yl) ester
At room temperature and N 2Down, with 5-nitrooxy-valeric acid (163mg, 1mmol), N-hydroxy-succinamide (173mg, 1.5mmol), (1.5mmol) mixture in dichloromethane stirs N-(3-dimethylaminopropyl)-N '-ethyl carbodiimide for EDC, 288mg.By the TLC monitoring reaction.After the end, add dichloromethane, mixture is washed with water.Organic layer filters through anhydrous sodium sulfate drying, concentrates.Residue is through column chromatography purification.
Figure A20048003609900972
The reverse ester nitrooxy analog of synthetic naringenin
At room temperature or 40 ℃, N 2Down, with naringenin (758mg, 1mmol), (1.3g, 5mmol) and N, (646mg, 5mmol) mixture in acetonitrile stirs the N-diisopropylethylamine 5-nitrooxy-valeric acid (2,5-dioxo-pyrrolidine-1-yl) ester.By the TLC monitoring reaction.After the end, add dichloromethane, with mixture hydrochloric acid solution (0.1N), saturated sodium bicarbonate aqueous solution and water washing.Organic layer filters through dried over sodium sulfate, concentrates.Crude product is through column chromatography purification.
Perhaps, prepare this product with other activating carboxy acid's analog (acyl chlorides, anhydride etc.).
The synthetic reverse ester NItroxyderivatives of this method of all chemical compounds that can be by being used for the present invention expectation, for example, the any starting compound that provides the method for synthetic naringenin to can be used for embodiment 1-59, it causes generating the reverse ester NItroxyderivatives of described starting compound but not the NItroxyderivatives of described starting compound then subsequently.
Method 1: the method for synthetic " oppositely ester nitrooxy " chemical compound of suggestion
Method 2: the method for synthetic " oppositely ester nitrooxy " chemical compound of suggestion
Figure A20048003609900982
Embodiment 103: preparation 5-nitrooxy-valeric acid 4-[5,7-two-(5-nitrooxy-valeryl oxygen base)-4-oxo-benzodihydropyran-2-yl]-alternative approach of phenylester
Figure A20048003609900991
Synthetic 5-bromo-valeric acid (2,5-dioxo-pyrrolidine-1-yl) ester
At room temperature, N 2Down, with 5-bromo-valeric acid (180mg, 1mmol), N-hydroxy-succinamide (173mg, 1.5mmol), (1.5mmol) mixture in dichloromethane stirs N-(3-dimethylaminopropyl)-N '-ethyl carbodiimide for EDC, 288mg.By the TLC monitoring reaction.After the end, add dichloromethane, mixture is washed with water.Organic layer filters through anhydrous sodium sulfate drying, concentrates.Make residue pass through column chromatography purification.
Figure A20048003609900992
With three-(5-bromo-valerate) of the synthetic naringenin of activation 5-bromo-valeric acid analog
At room temperature or 40 ℃, N 2Down, with naringenin (272mg, 1mmol), (1.39g, 5mmol) and N, (646mg, 5mmol) mixture in acetonitrile stirs the N-diisopropylethylamine 5-bromo-valeric acid (2,5-dioxo-pyrrolidine-1-yl) ester.By the TLC monitoring reaction.After the end, add dichloromethane, with mixture hydrochloric acid solution (0.1N), saturated sodium bicarbonate aqueous solution and water washing.Organic layer filters through dried over sodium sulfate, concentrates.Make crude product through column chromatography purification.
Perhaps, with other activating carboxy acid's analog (acyl chlorides, anhydride etc.) preparation product.
Figure A20048003609900993
The reverse ester nitrooxy analog of synthetic naringenin
Under 40 ℃, with three of naringenin-(5-bromo-valerate) (758mg, 1mmol) and silver nitrate (850mg 5mmol) stirs in acetonitrile.By the TLC monitoring reaction.After the end, add dichloromethane, mixture is washed with water,, filter, concentrate through anhydrous sodium sulfate drying.Make crude product pass through column chromatography purification.
The synthetic reverse ester NItroxyderivatives of this method of all chemical compounds that can be by being used for the present invention expectation, for example, the any starting compound that provides the method for synthetic naringenin to can be used for embodiment 1-59, it causes generating the reverse ester NItroxyderivatives of described starting compound but not the NItroxyderivatives of described starting compound subsequently then.

Claims (24)

1. induce the ApoA1 expression to cause blood HDL level to raise and the chemical compound of blood cholesterol levels reduction for one kind, but described chemical compound comprise the antioxidant molecule of nitric oxide donating part and removing free radical.
2. the chemical compound of claim 1, described chemical compound comprises the flavone compound that contains following structure:
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10, R 13And R 14Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10Or R 13Or R 14In at least one is nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be O, CR 13Or NR 13
Y can be the ketone that CO[still keeps 6 atom ring structures], CR 14Or NR 14With
Z can be singly-bound or two key.
3. Pharmaceutical composition, described compositions comprises the flavone compound of claim 2 and the pharmaceutically acceptable carrier of applied in any combination.
4. method for the treatment of patient's cardiovascular, cholesterol or lipid relevant disease, described method comprise the flavone compound of the claim 2 of the patient treatment effective dose that needs treatment.
5. induce the method that ApoA1 expresses provides antioxidant activity simultaneously for one kind in the patient, described method comprises the flavone compound that gives described patient's claim 2.
6. method that reduces total serum cholesterol in patients, described method comprises the flavone compound that gives described patient's claim 2.
7. the chemical compound of claim 1, described chemical compound comprises the isoflavonoid that contains following structure:
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10, R 13And R 14Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10Or R 13Or R 14In at least one is nitrooxy, R 12, OR 12Or OCOR 12And
Wherein OCOR represents
Figure A2004800360990004C1
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more metathetical by S, N or O CAtom and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be O, CR 13Or NR 13
Y can be the ketone that CO[still keeps 6 atom ring structures], CR 14Or NR 14With
Z can be singly-bound or two key.
8. Pharmaceutical composition, described compositions comprises the isoflavonoid of claim 7 and the pharmaceutically acceptable carrier of applied in any combination.
9. method for the treatment of patient's cardiovascular, cholesterol or lipid relevant disease, described method comprise the isoflavonoid of the claim 7 of the patient treatment effective dose that needs treatment.
10. induce the method that ApoA1 expresses provides antioxidant activity simultaneously for one kind in the patient, described method comprises the isoflavonoid that gives described patient's claim 7.
11. a method that reduces total serum cholesterol in patients, described method comprises the isoflavonoid that gives described patient's claim 7.
12. the chemical compound of claim 1, described chemical compound comprise the stilbene chemical compound that contains following structure:
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
Figure A2004800360990005C2
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2With
Wherein X can be singly-bound, two key or triple bond.
13. a Pharmaceutical composition, described compositions comprise the stilbene chemical compound of claim 12 and the pharmaceutically acceptable carrier of applied in any combination.
14. a method for the treatment of patient's cardiovascular, cholesterol or lipid relevant disease, described method comprise the stilbene chemical compound of the claim 12 of the patient treatment effective dose that needs treatment.
15. induce the method that ApoA1 expresses provides antioxidant activity simultaneously for one kind in the patient, described method comprises the stilbene chemical compound that gives described patient's claim 12.
16. a method that reduces total serum cholesterol in patients, described method comprise the stilbene chemical compound that gives described patient's claim 12.
17. the chemical compound of claim 1, described chemical compound comprise the chalcone chemical compound that contains following structure:
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9, R 10, R 13And R 14Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10Or R 13Or R 14In at least one is nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2
X can be singly-bound or two key;
Y can be singly-bound or two key;
Z can be CO[ketone], CR 13Or NR 13
18. a method for the treatment of patient's cardiovascular, cholesterol or lipid relevant disease, described method comprise the chemical compound of the claim 17 of the patient treatment effective dose that needs treatment.
19. induce the method that ApoA1 expresses provides antioxidant activity simultaneously for one kind in the patient, described method comprises the chemical compound that gives described patient's claim 17.
20. a method that reduces total serum cholesterol in patients, described method comprises the chemical compound that gives described patient's claim 17.
21. the chemical compound of claim 1, described chemical compound comprises the polyphenol compound that contains following structure:
Figure A2004800360990007C1
Wherein
R 1, R 2, R 3, R 4, R 5, R 6, R 7, R 8, R 9And R 10Can be hydrogen, hydroxyl [OH], hydroxyalkyl, aminoalkyl, bromine (Br), iodine (I), nitrooxy [ONO independently separately 2], methoxyl group [OCH 3], ethyoxyl [OCH 2CH 3], fluorine [F], chlorine [Cl], CF 3, CCl 3, phosphate-based, R 11, R 12, OR 11, OR 12, OCOR 11, OCOR 12, O-sulfuric ester [sulfuric ester conjugate] or O-glucuronate [glucuronic acid (AKA glucuronic acid) conjugate], condition is R 1-R 10In at least one be nitrooxy, R 12, OR 12Or OCOR 12With
Wherein OCOR represents
And R is R 11Or R 12,
R wherein 11Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted and to choose wantonly and be side chain, and can have one or more by S, N or the metathetical C atom of O and
R wherein 12Be C 1-18, aryl, heteroaryl or derivatives thereof, wherein said derivant is optional to be substituted, and optionally is side chain, can have one or morely by S, N or the metathetical C atom of O, and contains one or more ONO 2With
X can be C, S, (CO), SO, AKA ketone, (SO 2) N, (CO) C, (CO) N, (CO) O, C-N[singly-bound], the two keys of C=N[], C-O, N-O, N-N[singly-bound] or the two keys of N=N[].
22. a method for the treatment of patient's cardiovascular, cholesterol or lipid relevant disease, described method comprise the chemical compound of the claim 21 of the patient treatment effective dose that needs treatment.
23. induce the method that ApoA1 expresses provides antioxidant activity simultaneously for one kind in the patient, described method comprises the chemical compound that gives described patient's claim 21.
24. a method that reduces total serum cholesterol in patients, described method comprises the chemical compound that gives described patient's claim 21.
CN 200480036099 2003-10-07 2004-10-07 Nitric oxide donating derivatives for the treatment of cardiovascular disorders Pending CN1893957A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US50915603P 2003-10-07 2003-10-07
US60/509,156 2003-10-07
US10/762,796 2004-01-22
US10/807,800 2004-03-24

Publications (1)

Publication Number Publication Date
CN1893957A true CN1893957A (en) 2007-01-10

Family

ID=37598186

Family Applications (1)

Application Number Title Priority Date Filing Date
CN 200480036099 Pending CN1893957A (en) 2003-10-07 2004-10-07 Nitric oxide donating derivatives for the treatment of cardiovascular disorders

Country Status (1)

Country Link
CN (1) CN1893957A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108069954A (en) * 2017-03-03 2018-05-25 上海华汇拓医药科技有限公司 The quinazolinones of the donor containing NO
CN108349911A (en) * 2015-09-07 2018-07-31 浙江华海药业股份有限公司 Releasable nitric oxide production prodrugs
CN110183312A (en) * 2019-07-25 2019-08-30 江西中医药大学 A kind of isopentene group talan and its purposes in preparation treatment diseases associated with inflammation drug
CN113831352A (en) * 2020-12-16 2021-12-24 顺德职业技术学院 Novel flavane compound of dianella root and preparation method and application thereof

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108349911A (en) * 2015-09-07 2018-07-31 浙江华海药业股份有限公司 Releasable nitric oxide production prodrugs
CN108069954A (en) * 2017-03-03 2018-05-25 上海华汇拓医药科技有限公司 The quinazolinones of the donor containing NO
CN108069954B (en) * 2017-03-03 2018-11-23 上海华汇拓医药科技有限公司 The quinazolinones of the donor containing NO
CN110183312A (en) * 2019-07-25 2019-08-30 江西中医药大学 A kind of isopentene group talan and its purposes in preparation treatment diseases associated with inflammation drug
CN113831352A (en) * 2020-12-16 2021-12-24 顺德职业技术学院 Novel flavane compound of dianella root and preparation method and application thereof

Similar Documents

Publication Publication Date Title
CN1925862A (en) Treatment of diseases associated with the EGR-1 enhancer element
CN1040433C (en) Azole compounds, their production and use
CN1289467C (en) Dibenzylamine compound and medicinal use thereof
CN100344616C (en) Compounds for the treatment of metabolic disorders
CN1279902C (en) Apoptosis inducing adamantyl derivatives and their usage as anti-cancer agents
CN1154486C (en) Method of treating malignant tumours with thyroxine analogues having no significant hormonal activity
CN1168631A (en) Methods for treating resistant tumors
CN1950333A (en) Substituted methyl aryl or heteroaryl amide compounds
CN1751038A (en) Substituted aryl and heteroaryl derivatives as modulators of glucose metabolism and the prophylaxis and treatment of disorders thereof
CN1494425A (en) Carboxamide derivatives as therapeutic agents
CN1909902A (en) Triazole, oxadiazole and thiadiazole derivatives as PPAR modulators for the treatment of diabetes
CN1950334A (en) Ortho substituted aryl or heteroaryl amide compounds
CN1646504A (en) Substituted benzoxazoles and analogues as estrogenic agents
CN1934091A (en) Imidazole compounds for the treatment of neurodegenerative disorders
CN87103953A (en) Pharmaceutically active 1, 5-diaryl-3-substituted pyrazoles and synthesis method thereof
CN1111247A (en) Protein kinase C inhibitors
CN1738788A (en) Substituted phenyl naphthalenes as estrogenic agents
CN101031540A (en) Hexafluoroisopropanol substituted ether derivatives
CN1143366A (en) 4-amino derivatives of mycophenolic acid
CN1768040A (en) Isoquinoline-5-sulfonic acid amides as inhibitors of AKT (protein kinase B)
CN1117732C (en) Novel dihydronaphthalene compound and process and producing the same
CN1090275A (en) The prodrug of imidazole carboxylic acids as angiotensin II receptor antagonists
CN101044126A (en) Prodrug substituted benzoxazoles as estrogenic agents
CN1826111A (en) Imidazole derivatives for treatment of allergic and hyperproliferative disorders
CN1869027A (en) Synthesis and antiprotozoal activity of dicationic 3,5-diphenylisoxazoles

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