CN101213281B - New dha derivatives and their use as medicaments - Google Patents

New dha derivatives and their use as medicaments Download PDF

Info

Publication number
CN101213281B
CN101213281B CN200680024064.9A CN200680024064A CN101213281B CN 101213281 B CN101213281 B CN 101213281B CN 200680024064 A CN200680024064 A CN 200680024064A CN 101213281 B CN101213281 B CN 101213281B
Authority
CN
China
Prior art keywords
compound
group
dha
prb
formula
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.)
Expired - Fee Related
Application number
CN200680024064.9A
Other languages
Chinese (zh)
Other versions
CN101213281A (en
Inventor
莫滕·布赖恩
安妮·K·霍尔梅德
简·科佩基
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.)
Pronova Biopharma Norge AS
Original Assignee
Pronova Biocare AS
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 Pronova Biocare AS filed Critical Pronova Biocare AS
Priority claimed from PCT/IB2006/001155 external-priority patent/WO2006117664A1/en
Publication of CN101213281A publication Critical patent/CN101213281A/en
Application granted granted Critical
Publication of CN101213281B publication Critical patent/CN101213281B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Landscapes

  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

Compounds of formula (I); wherein - R1 and R2 are the same or different and may be selected from the group consisting of a hydrogen atom, a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, an alkylsulfmyl group, an alkylsulfonyl group, an amino group, and an alkylamino group; and - X represents a carboxylic acid group, a carboxylate group, or a carboxamide group; or any pharmaceutically acceptable salt, solvate, complex or pro-drug thereof, with the provisos that the compound of formula (I) is not (all-Z)-4,7,10,13,16,19- docosahexaenoic acid (DHA), alpha-methyl DHA, alpha-methyl DHA methyl ester, alpha-methyl DHA ethyl ester or alpha-hydroxy DHA ethyl ester, are disclosed. A fatty acid composition and a pharmaceutical composition comprising such compounds are also disclosed. The use of such compounds as medicaments, in particular for the treatment of diabetes type 2, is also disclosed.

Description

New DHA derivative and as the purposes of medicine
Technical field
The present invention relates to the compound of general formula (I):
And they are as the purposes of medicine, particularly treat diabetes B and the purposes in early stage thereof, also relate to the pharmaceutical composition that contains formula (I) compound, and the fatty acid composition that contains formula (I) compound.
Background of invention
Diabetes B sickness rate in the worldwide increases the successful execution of prevention and therapeutic strategy has been caused huge publilc health and the challenge of medical aspect.The overweight disease that is closely related with diabetes B and the concurrent treatment that hinders diabetes of obesity, and increased the possibility of hypertension, blood lipid dysbolism and atherosclerosis relative disease.
The pathologic, physiologic illness in the early stage that diabetes B forms is relevant to the effect reduction of peripheral tissues with Regular Insulin, is referred to as insulin resistant.These tissues are mainly muscle, fat and liver.Muscle tissue is the Main Tissues relevant with insulin resistant in diabetes B.Syndromes take insulin resistant, hypertension, blood lipid dysbolism and whole body proinflammatory state as feature is called metabolic syndrome.The sickness rate of metabolic syndrome is 22-39% (Meighs 2003) in developed country's Grown living.
Recently the most promising approach that alleviates and stops metabolic syndrome is the combination with the mode of life interference of the increase of weight reducing, the minimizing that consumes saturated fatty, physical exertion and suitable pharmacotherapy.Stop the health diet of the excessive absorption of energy to comprise that list or polyunsaturated fatty acid are to the replacement of saturated fatty.The long-chain omega-fatty acid that particularly comes from the fattiness fish, that is timnodonic acid (EPA) and docosahexenoic acid (DHA) to be proved to be in the prevention diabetes B be useful.
EPA and DHA have effect to the multiple physiological process that affects normal health and chronic disease, such as the adjusting of blood lipid level, cardiovascular and immunologic function, insulin action and neurodevelopment and sight function.Existing strong evidence shows that they play in prevention and treatment coronary heart disease, blood lipid dysbolism, diabetes B, insulin resistant and hypertension that useful (Simonopoulos 1999; Geleijnse 2002; Storlien 1998).
Nearest studies show that omega-fatty acid as the important amboceptor of genetic expression, its expression that is controlled at the gene that lipid and glucose metabolism and adipocyte relate in forming by nuclear receptor such as peroxisome proliferation-activated receptors (PPARs) work (Jump 2002).PPARs is the nuclear Fatty Acid Receptors, and it plays an important role in the relevant metabolic disease of obesity such as hyperlipidaemia, insulin resistant and coronary heart disease.
Three kinds of subtype alphas, γ and δ have different expression patterns, evolve to be the sensation composition of different lipoprotein and the homeostasis of regulating lipid according to the needs of particular organization.It is the molecular target of the special class of lipopenicillinase shellfish (fibrates) that PPAR α strengthens lipid acid katabolism and its in liver.PPAR γ is vital to Adipocyte Differentiation on the other hand, and it regulates the activity of the thiazolidinediones (glitazone) of Regular Insulin-sensitivity, and its mechanism does not make fully that (Chih-Hao 2003 clear; Yki-J
Figure 2006800240649_0
Rvinen 2004).
Recently, there has been the medicine of the part that is used as PPAR γ acceptor to be introduced in (Yki-J in the treatment of diabetes B
Figure 2006800240649_1
Rvinen 2004).The compound that these compounds are called thiazolidinediones or glitazone is the medicine that reverses insulin resistant, and wherein insulin resistant is the pathophysiological basis of the formation of metabolic syndrome and diabetes B.These compounds, wherein rosiglitazone and pioglitazone have been used as medicine, reduce the concentration of fasting and GLPP concentration (it shows as the test of pathology glucose tolerance), plasma insulin and free fatty acids.In this regard, glitazone is as insulin sensitisers.
Yet these improve the increase (Adams 1997) that usually is accompanied by weight increase and subcutaneous lipids-liver mass.The use of thiazolidinediones not only is associated with weight to be increased, and the sub-fraction patient has fluid retention and Plasma volume expansion, thereby causes the periphery oedema.Increasing with the increase of Incidence of Heart Failure of body weight and oedema is associated, and namely why a reason of warning is provided in the descriptive information of rosiglitazone (being provided by Avandia) and pioglitazone (being provided by Takeda) in food and drug administration for this.These untoward reactions have limited the especially application in patients with coronary heart disease is arranged of glitazone.Significantly, it need to have the positive reaction to insulin resistant, the potentiality that have the active of weight reducing and do not have the tendency of fluid retention simultaneously concerning new drug.
Polyunsaturated fatty acid (PUFAs) is not only the structure of its lipid acid to the effect of PPARs and to the affinity of acceptor.The factor that is of value to the composition of cell Nei Fei-esterified fatty acid (NEFA) degree also is vital.This type of NEFA storehouse is subject to entering the impact of the amount of the concentration of Exogenous Fatty Acid of cell and inherent synthetic lipid acid, and removing of they can be finished (Pawar 2003) by being attached to lipid and their oxidative pathway.
Although omega-fatty acid is weak PPARs agonist, but when relatively coming word with pharmacological agonist such as thiazolidinediones, these lipid acid are verified to have improvement effect (Storlien 1987) in glucose absorption and insulin sensitivity.Have been reported when increasing the ratio of polyunsaturated and saturated lipid acid in diet, adipocyte is more responsive and transmit more glucose (Field1990) to Regular Insulin.Generally speaking, these data show the lipid acid of 20-and 22-carbon, are called EPA and DHA can play prevention in the formation of insulin resistant effect.
Because limited stability and lack biologic specificity in the body, PUFAs is not widely used as therapeutical agent.In order to change or increase their metabolic effects, the n-3 polyunsaturated fatty acid of chemical modifying carries out in several research groups.
For example, the effect for reducing fat of EPA by the α of EPA-or β-position introduce methyl or ethyl strengthened (Vaagenes 1999).When this compound when EPA EE does not have effect also can reduce blood plasma free fatty acid.
In the works of recently L.Larsen publication (Larsen 2005), the author discloses than EPA/DHA, and the Alpha-Methyl derivative of EPA and DHA has increased the activity of nuclear receptors PPAR's α, and therefore increases the expression of L-FABP.The EPA that has ethyl at alpha-position is equal to Alpha-Methyl EPA to the intensity of activation of PPAR α.The author thinks that the katabolism that postpones these Alpha-Methyls EPA can help their effects to increase owing to cause the minimizing of the β-oxidation of peroxysome oxygenizement in plastosome.
Than EPA, Alpha-Methyl EPA (Willumsen 1998) in external (Larsen 1998) and body has been indicated as stronger anticoagulant.
Publication number is that the Japanese patent abstract of 05-00974 discloses the DHA that is replaced by the OH-base at alpha-position, yet it only is as intermediate.Test to the possible curative effect of medication of this compound is not disclosed.
Laxdale Limited discloses the purposes (US6689812) of α substitutive derivative in treatment psychosis or nervus centralis obstacle of EPA.
Figure S2006800240649D00031
(A) Alpha-Methyl EPA
Yet these improved lipid acid neither ones have demonstrated gratifying pharmaceutical activity, and neither one enters pharmaceutical market.
Summary of the invention
The purpose of this invention is to provide the new DHA-derivative with therapeutic activity.
Based on the present invention, there are in the claims a lot of aspects, some of them are;
1. compounds, that is, and the polyunsaturated fatty acid derivative of some alpha-substitution.
2. the compound of the novelty that is used as medicine and in treatment, uses.
3. the fatty acid composition or the pharmaceutical composition that contain compounds.
4. the fatty acid composition that contains compounds that is used as medicine and in treatment, uses.
5. compounds is for the preparation of the purposes in the medicine that prevents and/or treats the diabetes in the human or animal.
6. compounds is for the preparation of the purposes in the medicine that treats and/or prevents obesity or overweight disease.
7. the purposes of compounds in the medicine that increases for the preparation of control weight loss and/or prevention body weight.
8. compounds is for the preparation of the purposes in the medicine that treats and/or prevents the relevant disease of amyloidosis.
9. compounds is for the preparation of the purposes in the medicine for the treatment of or the multiple risk factors of preventing cardiovascular disease.
10. compounds is for the preparation of the purposes in the medicine of the prevention apoplexy relevant with the atherosclerosis of several arteries or transient ischemic attack.
11. special for treating diabetes, the method for preferred diabetes B.
12. method of controlling weight loss, the increase of prevention body weight and/or treating and/or preventing obesity or overweight disease.
13. method that treats and/or prevents the relevant disease of amyloidosis.
14. the method for a treatment or the multiple risk factors of preventing cardiovascular disease.
15. the apoplexy that a prevention is relevant with the atherosclerosis of several arteries or the method for transient ischemic attack.
16. the method for preparation novel fatty acid analog according to the present invention.
The present invention relates to formula (I) compound:
Figure S2006800240649D00051
Wherein
-R 1And R 2Be identical or different, and can be selected from following group: hydrogen atom, hydroxyl, alkyl, halogen atom, alkoxyl group, acyloxy, acyl group, alkenyl, alkynyl, aryl, alkylthio, alkoxy carbonyl, alkyl sulphinyl, alkyl sulphonyl, amino and alkylamino; And
-X represents carboxylic acid group (carboxylic acid group), carboxylic acid ester groups (carboxylate group) or carboxamide groups (carboxamide group),
Or its any pharmacologically acceptable salt, solvate, mixture or prodrug,
Condition is:
■ formula (I) compound be not (complete-Z)-DHA (DHA), Alpha-Methyl DHA, Alpha-Methyl DHA methyl esters, Alpha-Methyl DHA-EE or Alpha-hydroxy DHA-EE.
Described condition is corresponding to following situation:
■ works as R 1Hydrogen atom, R then 2It or not hydrogen atom;
■ works as R 2Hydrogen atom, R then 1It or not hydrogen atom;
■ works as R 1Methyl, R then 2Be not hydrogen atom, and X not carboxylic acid group, methoxycarbonyl (metyhlcarboxylate) or ethoxycarbonyl;
■ works as R 2Methyl, R then 1Be not hydrogen atom, and X not the carboxylic acid group, methoxycarbonyl or ethoxycarbonyl;
■ works as R 1Hydroxyl, R then 2Be not hydrogen atom, and X not ethoxycarbonyl; And
■ works as R 2Hydroxyl, R then 1Be not hydrogen atom, and X not ethoxycarbonyl.
In the compound of the present invention, described alkyl can be selected from following group: methyl, ethyl, n-propyl, sec.-propyl, normal-butyl, sec-butyl, n-hexyl and benzyl; Described halogen atom can be selected from following group: fluorine, chlorine, bromine and iodine; Described alkoxyl group can be selected from following group: methoxyl group, oxyethyl group, propoxy-, isopropoxy, sec-butoxy, phenoxy group, benzyloxy, OCH 2CF 3And OCH 2CH 2OCH 3Described acyloxy can be selected from acetoxyl group, propionyloxy and butyryl acyloxy; Described alkenyl can be selected from following group: allyl group, crotyl and 3-hexenyl; Described alkynyl can be selected from following group: propargyl, 2-butyne base and 3-hexin base; Described aryl is phenyl; Described alkylthio can be selected from following group: methylthio group, ethylmercapto group, isopropyl sulfenyl and thiophenyl; Described alkoxy carbonyl can be selected from following group: methoxycarbonyl, ethoxy carbonyl, propoxycarbonyl and butoxy carbonyl; Described alkyl sulphinyl can be selected from following group: methylsulfinyl, ethyl sulfinyl and sec.-propyl sulfinyl; Described alkyl sulphonyl can be selected from following group: methyl sulphonyl, ethylsulfonyl and sec.-propyl alkylsulfonyl; Described alkylamino can be selected from following group: methylamino, dimethylamino, ethylamino and diethylamino; Described ester group can be selected from following group: ethoxycarbonyl, methoxycarbonyl, n-propyl ester group, sec.-propyl ester group, normal-butyl ester group, sec-butyl ester group and n-hexyl ester group; Described carboxamide groups can be selected from following group: uncle's carboxamide groups (primarycarboxamide), N-methyl carboxamide groups (N-methyl carboxamide), N, N-dimethyl carboxamide groups, N-ethyl carboxamide groups and N, N-diethyl carboxamide groups.
In one embodiment of the invention, R 1And R 2Be selected from following group: hydrogen atom, hydroxyl, alkyl, halogen atom, alkoxyl group, alkylthio, alkyl sulphinyl, alkyl sulphonyl, amino and alkylamino.
In another embodiment of the invention, R 1And R 2Be selected from following group: hydrogen atom, hydroxyl, C 1-C 7Alkyl, halogen atom, C 1-C 7Alkoxyl group, C 1-C 7Alkylthio, C 1-C 7Alkyl sulphinyl, C 1-C 7Alkyl sulphonyl, amino and C 1-C 7Alkylamino.In addition, described C 1-C 7Alkyl can be methyl, ethyl or benzyl; Described halogen atom can be fluorine or iodine; Described C 1-C 7Alkoxyl group can be methoxy or ethoxy; Described C 1-C 7Alkylthio can be methylthio group, ethylmercapto group or thiophenyl; Described C 1-C 7Alkyl sulphinyl can be the ethyl sulfinyl; Described C 1-C 7Alkyl sulphonyl can be ethylsulfonyl; Described C 1-C 7Alkylamino can be ethylamino or diethylamino; And X can represent ethoxycarbonyl or carboxamide groups.
In another embodiment of the invention, R 1And R 2Be selected from following group: hydrogen atom, C 2-C 7Alkyl, halogen atom, C 1-C 7Alkoxyl group, C 1-C 7Alkylthio, C 1-C 7Alkyl sulphinyl, C 1-C 7Alkyl sulphonyl, amino and C 1-C 7Alkylamino; And X represents carboxylic acid ester groups.In addition, described C 2-C 7Alkyl can be ethyl or benzyl; Described halogen atom can be fluorine or iodine; Described C 1-C 7Alkoxyl group can be methoxy or ethoxy; Described C 1-C 7Alkylthio can be methylthio group, ethylmercapto group or thiophenyl; Described C 1-C 7Alkyl sulphinyl can be the ethyl sulfinyl; Described C 1-C 7Alkyl sulphonyl can be ethylsulfonyl; Described C 1-C 7Alkylamino can be ethylamino or diethylamino; And X represents ethoxycarbonyl.
In the compound of the formula according to the present invention (I), R 1And R 2Can be identical or different.When they not simultaneously, formula (I) compound can exist with the form of steric isomer.Should be appreciated that all optically active isomers and the mixture that comprises racemic modification of the formula of the present invention includes (I) compound.
Therefore, work as R 1With R 2When not identical, the present invention includes formula (I) compound, it is the form of racemic modification or enantiomeric pure, (S) or (R) enantiomer.Therefore, work as R 1With R 2When not identical, the present invention includes formula (I) compound, it is the form of racemic modification or enantiomeric pure, (S) or (R) enantiomer.
Within the scope of the present invention, the enantiomer of formula (I) compound as defined above.In addition, the enantiomer of DHA derivative of the present invention can be the form of carboxylic acid, or its pharmacologically acceptable salt, any ester, acid anhydrides or acid amides (primary, secondary and tertiary).Acid derivative can with phosphatide or three-, two-or monoglyceride.
In the embodiment according to formula of the present invention (I) compound, R 1And R 2In one represent C 2-C 7Alkyl is ethyl or benzyl for example, and another represents hydrogen atom.Preferably, alkyl is ethyl.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2In a representation alkoxy for example oxyethyl group or methoxyl group, and another represents hydrogen atom.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2In one represent halogen atom for example fluorine or iodine, and another represents hydrogen atom.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2In one represent alkylthio for example ethylmercapto group, methylthio group or thiophenyl, and another represents hydrogen atom.Preferably, alkylthio is ethylmercapto group.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2In one represent for example ethylsulfonyl of alkyl sulphonyl, and another represents hydrogen atom.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2In one represent aminoly, and another represents hydrogen atom.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2In one represent alkyl-amino for example ethyl-amino or diethyl-amino, and another represents hydrogen atom.
In another embodiment according to formula of the present invention (I) compound, R 1And R 2Be identical, and represent C 1-C 7-alkyl, preferable methyl or ethyl.
In the preferred embodiment of formula (I) compound, X is for example ethoxycarbonyl of carboxylic acid ester groups.
Can be with phosphatide according to compound of the present invention, three-, two-or the form of monoglyceride (monoglyceride) or free acid exist.
The good pharmaceutical active that has shockingly demonstrated according to the DHA-derivative of alpha-substitution of the present invention.Especially, derivative of fatty acid according to the present invention has huge potentiality in treating and/or preventing diabetes and the use in its early stage.
Another aspect of the present invention relates to formula (I) compound as medicine.
The present invention also relates to the method for preparation formula (I) compound.For example, formula (I) compound can from (complete-Z)-DHA (DHA) preparation.DHA can for example obtain from vegetables, microorganism and/or animal source such as deep sea fish oil.Another important advantage of formula (I) compound be its fatty acid analog can from (complete-Z)-directly preparation of DHA (DHA).
In a preferred embodiment of the invention, the fatty acid analog of formula (I) prepares from DHA, and wherein said DHA is from least a vegetables, microorganism and animal source, or its combination obtains.Therefore the present invention includes the derivative for preparing in the oil that from microbial source, contains DHA.Suitably, described DHA for example produces in the fish oil from marine animal oil.
Another aspect of the present invention relates to and contains formula (I) compound as the pharmaceutical composition of active ingredient.This pharmaceutical composition can further comprise pharmaceutically acceptable carrier.Suitably, pharmaceutical composition according to the present invention is mixed with the oral administration form, as with capsule or wafer (sachet) form.Suitable per daily dose according to formula of the present invention (I) compound is 10mg-10g, especially the described compound of 100mg-1g.
In addition, the present invention relates to contain the fatty acid composition of formula (I) compound.At least 60% or at least 90 % by weight of this fatty acid composition are comprised of described compound.This fatty acid composition can further comprise (entirely-Z)-5,8,11,14,17-timnodonic acid (EPA), (complete-Z)-4,7,10,13,16,19-docosahexenoic acid (DHA), (complete-Z)-6,9,12,15,18-, 21 carbon 5 alkene acids (HPA) and/or (complete-Z)-7,10,13,16,19-clupanodonic acid (DPA).Lipid acid can exist with the form of derivative.Can further comprise pharmaceutically acceptable antioxidant such as tocopherol according to fatty acid composition of the present invention.Above-described fatty acid composition as medicine also within the scope of the invention.
On the other hand, the present invention relates to formula (I) compound loses weight and/or prevents purposes in the medicine that body weight increases in preparation control; It treats and/or prevents purposes in the medicine of obesity and overweight disease (overweight) in preparation; It prevents and/or treats diabetes in the animal in preparation, particularly the purposes in the diabetes B medicine; It treats and/or prevents purposes in the medicine of the relevant disease of amyloidosis in preparation; It is at the multiple risk factors of preparation treatment or preventing cardiovascular disease, is preferred for treating the purposes in the medicine that blood fat raises; Its purposes in the medicine for preparing the prevention apoplexy relevant with the atherosclerosis of several arteries or transient ischemic attack.
In addition, the present invention relates to control the method for weight loss and/or the increase of prevention body weight; Treat and/or prevent the method for obesity or overweight disease; Prevent and/or treat the particularly method of diabetes B of diabetes; Treat and/or prevent the method for the relevant disease of amyloidosis; The method of the multiple risk factors for the treatment of or preventing cardiovascular disease; Prevent the apoplexy relevant with the atherosclerosis of several arteries or the method for transient ischemic attack, wherein the formula of administration human or animal medicine effective quantity (I) compound.Compatibly, formula (I) compound is by the oral administration human or animal.
Brief Description Of Drawings:
Fig. 1 is the synoptic diagram of free fatty acids storehouse theory.
Fig. 2 represents in order to illustrate that model that the present invention uses and method are to the synoptic diagram of the effect of metabolic syndrome and diabetes B.
Fig. 3 has described the free-fat acid concentration of the different compounds of the present invention in animal liver tissue, and wherein these compounds give this animal with 1.5% concentration of total lipid content.
Fig. 4 describes the IC of the DHA in the animal liver tissue, and wherein different compound of the present invention gives this animal with 1.5% concentration of total lipid content.
Fig. 5 describes different compound of the present invention to the binding affinity of PPAR γ acceptor.
Fig. 6 describes different compound of the present invention to the binding affinity of nuclear receptors PPAR's α.
Fig. 7 describes different compound of the present invention to the binding affinity of nuclear receptor RXR α.
Fig. 8 describes the release of luciferase from the transfectional cell of the different compound treatment of process the present invention.
Fig. 9 represents the research and design of the 4th group of experiment.
Figure 10 represents through the body weight change in the diet interference phase within 2 weeks after 8 all HF diet.
Figure 11 represents the result that uciferase activity (giving birth to PPAR γ-activity namely) obtains.
Figure 12 represents interior living luciferase in the different compounds of the present invention and the comparison among the DHA.
Figure 13 is illustrated in before the animalizing compound with insulin resistant and typical blood sugar is afterwards eliminated curve, and the compound that wherein gives animal is to have the compound that weakens the insulin resistant effect.
Figure 14,15 and 16 represents that DHA derivative of the present invention is to the different-effect of metabolic syndrome and insulin resistant.
Detailed Description Of The Invention
In nearest work of the present invention, prepared new DHA-derivative, it has demonstrated good pharmaceutical activity.
Lipid acid enters in cell or toughness (trough) G-albumen coupling transportation subsystem such as the fatty acid transport protein passively.They by in conjunction with albumen (fatty acid binding protein, FABP) well temporary transient combination in cell, for metabolism and genetic expression its at guiding lipid acid (the Pawar ﹠amp that plays an important role to different intracellular region chambers; Jump 2003) (Fig. 1 liver cell).
Fatty acid ester changes into triglyceride level, and polar lipid and cholesteryl ester and their β-oxidation form (plastosome and peroxysome) need lipid acid to be converted into acyl-CoA monothioester (thioesters).Other approach, synthetic such as list-oxidised form and eicosanoid that microsome NADPH-relies on, utilize the lipid acid of non--esterification as substrate.All these reactions all might affect the cell levels of free fatty acids (non--esterification) and therefore affect the value volume and range of product that can be used as the lipid acid of nuclear receptor ligands.Because therefore known PPARs, expects that the composition in free fatty acids storehouse is that important determinative also is rational in conjunction with the lipid acid of non--esterification in control PPAR activity.
The composition in free fatty acids storehouse is subject to entering the impact of the speed of the Exogenous Fatty Acid concentration of cell and they removing by approach listed above.Because the lipid acid of short chain and moderate-length chain is added to these approach effectively, only has long chain polyunsaturated fatty acids can be coordinated to nuclear receptor in the reality.In addition, fatty acid structure also is important determinative.Even in a series of list and polyunsaturated fatty acid that demonstrates PPAR α receptor affinity, in the experiment of using rat hepatocytes, EPA and DHA have shown the highest binding ability (Pawar ﹠amp; Jump 2003).
Seek available lipid acid material standed for and be used for coming albumen is carried out genetic modification by the interaction with nuclear receptor such as PPARs, most important be exactly separately lipid acid of conclusive evidence will enrichment in the free fatty acids storehouse.
The DHA that enters cell is converted into rapidly fatty acid acyl-CoA monothioester and is bonded in the phosphatide, because these, the DHA level is relatively low in the cell.These DHA-CoA also are the substrates of main β-oxidation in peroxysome, and this has caused the DHA reverse to turn to EPA, referring to Fig. 1.Owing to be bonded to fast neutral fat, therefore in the free fatty acids storehouse, can't continue the long time at oxidative pathway DHA.Thus, DHA may be for limited to the effect of genetic expression.
The object of the invention is to obtain the accumulation of derivative of fatty acid in the free fatty acids storehouse, rather than is bonded in the phosphatide.The inventor has found to introduce the minimizing that at least a substituting group will cause rate of oxidation to slow down and be bonded to neutral fat surprisingly in the alpha-position of DHA.Because the DHA derivative will particularly gather in liver, muscle and adipocyte and cause that the karyomerite receptor active arrives than in the larger degree of DHA organizing, this will increase the impact on genetic expression.
Can cause derivative that the affinity of lipid acid bind receptor is changed according to different substituents of the present invention.Thereby also might change the biological activity of DHA derivative that affinity to fatty acid binding protein changes the alpha-substitution of these formulas (I).In a word, than DHA, these changes have caused the result for the treatment of according to DHA derivative increase of the present invention.
EPA (complete-Z)-EPA in the past once by α-and β-position alkylation be used for suppressing mitochondrial β-oxidation.DHA is not oxidized in plastosome, but is incorporated in in the phosphatide.Although some DHA are reversed and change into EPA in peroxysome.Thus, the substituting group in the alpha-position of EPA and DHA can affect different pathways metabolisms.Have been reported in early days that Alpha-Methyl EPA and Beta-methyl EPA are incorporated in to phosphatide and tri-glyceride α-ethyl EPA does not then have (Larsen 1998).In this research, the substrate that this derivative conduct relates in the eicosanoid cascade and/or enzymeinhibition agent are tested.For these enzymes, because most substrate is the lipid acid that discharges, therefore expect that these derivatives can be incorporated in in the phosphatide from phosphatide.On the contrary, as previously mentioned, the derivative debond that we need but is accumulated in the NEFA storehouse to lipid.
In the whole specification sheets, abbreviation " PRB-x ", wherein X is integer, will use when describing according to specific compound of the present invention.The below has listed each structural formula and the popular name of these compounds.
Figure S2006800240649D00111
PRB-1 Alpha-Methyl docosahexenoic acid ethyl ester
PRB-2 α-ethyl docosahexenoic acid ethyl ester
Figure S2006800240649D00113
PRB-3 α-oxyethyl group docosahexenoic acid ethyl ester
Figure S2006800240649D00121
PRB-4 α-fluorine docosahexenoic acid ethyl ester
Figure S2006800240649D00122
PRB-5 α, alpha-alpha-dimethyl docosahexenoic acid ethyl ester
Figure S2006800240649D00123
PRB-6 alpha-methylmercapto docosahexenoic acid ethyl ester
Figure S2006800240649D00124
PRB-7 α-ethylmercapto group docosahexenoic acid ethyl ester
Figure S2006800240649D00125
PRB-8 α, α-diethyl docosahexenoic acid ethyl ester
Figure S2006800240649D00131
PRB-9 α-benzyl docosahexenoic acid ethyl ester
Figure S2006800240649D00132
PRB-10 α-ethyl sulfinyl docosahexenoic acid ethyl ester
Figure S2006800240649D00133
PRB-11 α-thiophenyl docosahexenoic acid ethyl ester
Figure S2006800240649D00134
PRB-12 Alpha-hydroxy docosahexenoic acid ethyl ester
Figure S2006800240649D00135
PRB-13 Alpha-Methyl docosahexenoic acid acid amides
Figure S2006800240649D00141
PRB-14 α-methoxyl group docosahexenoic acid ethyl ester
Figure S2006800240649D00142
PRB-15 alpha-iodine docosahexenoic acid ethyl ester
Figure S2006800240649D00143
PRB-17 alpha-amino group docosahexenoic acid ethyl ester
Figure S2006800240649D00144
PRB-18 (4R, 5S)-3-two dodecahexaenes acyl group-4-methyl-5-phenyl- Azoles alkane-2-ketone
Figure S2006800240649D00145
PRB-19 (4R, 5S)-3-[(S)-α-ethyl two dodecahexaene acyl groups]-4-methyl-5-phenyl-
Figure 2006800240649_3
Azoles alkane-2-ketone
Figure S2006800240649D00151
PRB-20 (S)-(+)-α-ethyl docosahexenoic acid ethyl ester
Figure S2006800240649D00152
PRB-21 (4S, 5R)-3-two dodecahexaenes acyl group-4-methyl-5-phenyl-
Figure 2006800240649_4
Azoles alkane-2-ketone
Figure S2006800240649D00153
PRB-22 (4S, 5R)-3-[(R)-α-ethyl two dodecahexaene acyl groups]-4-methyl-5-phenyl-
Figure 2006800240649_5
Azoles alkane-2-ketone
Figure S2006800240649D00154
PRB-23 (R)-(-)-α-ethyl docosahexenoic acid ethyl ester
Figure S2006800240649D00161
PRB-24 2-(1,3-dioxo-1,3-dihydro-isoindole-2-yl)-docosahexenoic acid ethyl ester
Figure S2006800240649D00162
PRB-25 α-ethyl-amino docosahexenoic acid ethyl ester
Figure S2006800240649D00163
PRB-26 α-diethyl-amino docosahexenoic acid ethyl ester
PRB-1 is corresponding to formula (I) compound, wherein R 1Or R 2Be methyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-2 is corresponding to formula (I) compound, wherein R 1Or R 2Be ethyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-3 is corresponding to formula (I) compound, wherein R 1Or R 2Be oxyethyl group, and another is hydrogen, and X is ethoxycarbonyl.
PRB-4 is corresponding to formula (I) compound, wherein R 1Or R 2Be fluorine, and another is hydrogen, and X is ethoxycarbonyl.
PRB-5 is corresponding to formula (I) compound, wherein R 1And R 2Be methyl, and X is ethoxycarbonyl.
PRB-6 is corresponding to formula (I) compound, wherein R 1Or R 2Be methylthio group, and X is ethoxycarbonyl.
PRB-7 is corresponding to formula (I) compound, wherein R 1Or R 2Be ethylmercapto group, and another is hydrogen, and X is ethoxycarbonyl.
PRB-8 is corresponding to formula (I) compound, wherein R 1And R 2Be ethyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-9 is corresponding to formula (I) compound, wherein R 1Or R 2Be benzyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-10 is corresponding to formula (I) compound, wherein R 1Or R 2Be the ethyl sulfinyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-11 is corresponding to formula (I) compound, wherein R 1Or R 2Be thiophenyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-12 is corresponding to formula (I) compound, wherein R 1Or R 2Be hydroxyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-13 is corresponding to formula (I) compound, wherein R 1Or R 2Be methyl, and another is hydrogen, and X is uncle's carboxamide groups.
PRB-14 is corresponding to formula (I) compound, wherein R 1Or R 2Be methoxyl group, and another is hydrogen, and X is ethoxycarbonyl.
PRB-15 is corresponding to formula (I) compound, wherein R 1Or R 2Be iodine, and another is hydrogen, and X is ethoxycarbonyl.
PRB-17 is corresponding to formula (I) compound, wherein R 1Or R 2Be amino, and another is hydrogen, and X is ethoxycarbonyl.
PRB-20 is corresponding to (S) steric isomer of formula (I) compound, wherein R 1Or R 2Be ethyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-23 is corresponding to (R) steric isomer of formula (I) compound, wherein R 1Or R 2Be ethyl, and another is hydrogen, and X is ethoxycarbonyl.
PRB-24 is corresponding to formula (I) compound, wherein R 1Or R 2Be N phlhalimide base (phtalimide), and another is hydrogen, and X is ethoxycarbonyl.
PRB-25 is corresponding to formula (I) compound, wherein R 1Or R 2Be ethyl-amino, and another is hydrogen, and X is uncle's carboxamide groups.
PRB-26 is corresponding to formula (I) compound, wherein R 1Or R 2Be diethyl-amino, and another is hydrogen, and X is ethoxycarbonyl.
Most preferred compound is PRB-2 according to the present invention.Other preferred compound of the present invention is PRB-5, PRB-7 and PRB-8.
Should be appreciated that any possible pharmaceutically useful salt, solvate, mixture or the prodrug of the formula of the present invention includes (I) compound.
" prodrug " for can have the entity that maybe can not have pharmacological activity, but it can be by administration (for example clothes or administered parenterally) thereby is subject in vivo afterwards the medicine of the present invention that bioactivation forms pharmacological activity.
When X was carboxylic acid, the present invention also comprised the salt of this carboxylic acid.The salt of suitable pharmaceutically acceptable carboxylic acid comprises metal-salt such as aluminium salt, an alkali metal salt such as lithium, sodium or sylvite, alkaline earth salt such as calcium or magnesium salts, and the ammonium salt of ammonium salt or replacement.
" treatment significant quantity " refers to the amount that effectively reaches its expection therapeutic interest agent.What yet individual patient was required can change, and the optimum range of the significant quantity of each nitric oxide adduct really fixes on this area and knows.Be used for sanatory common dosage regimen with compound of the present invention and/or composition and select according to many factors, comprise patient's type, age, sex, diet and medical condition.
" medicine " refers to the compound according to formula (I), and it is any form for example form/composite preparation or product, diet product, foodstuff raw material or the food fill-in of medicinal product that is suitable for medical purpose.
In the context of specification sheets of the present invention, term " treatment " comprises " prevention " unless opposite special instruction is arranged, and has also therefore made up term " treatment " and " treatment ground ".
Treatment comprises uses people or the inhuman useful any treatment of animal.Particularly preferably mammiferous treatment.The treatment of people and beasts all is included in the scope of the present invention.Medical needle is to being already present illness or preventative treatment.It can be in adult, teenager, baby, fetus or aforesaid any part (for example organ, tissue, cell or nucleic acid molecule)." chronicity treatment " refers to treatment and continues several weeks or several years.
" treatment and medicine effective quantity " refers to and can cause required pharmacology and/or the amount of result for the treatment of.
Can for example be included in foodstuff raw material, food fill-in, nutritional supplement or the diet product according to compound of the present invention.
The DHA derivative of alpha-substitution and EPA (or about this DHA) can combine and be bonded on the tri-glyceride by esterification process, and this process is reacted between α-derivative, EPA and glycerine by Novozym 435 (a kind of can from Candidaantarctica commercially available machine made lipase) catalysis.
Formula (I) compound has activity as medicine, particularly as the initiator of receptor active.Therefore, the present invention also relates to as defined above formula (I) compound, its pharmacologically acceptable salt, solvate, mixture or prodrug, it is used as medicine and/or uses in treatment.Preferably, can use novel compound of the present invention or its its pharmacologically acceptable salt, solvate, mixture or prodrug:
-be used for preventing and/or treating the diabetes among the human or animal;
-be used for controlling weight loss and/or prevent body weight to increase;
-be used for preventing and/or treating obesity or overweight disease among the human or animal;
-be used for treating and/or preventing the relevant disease of amyloidosis;
-be used for treating or the multiple risk factors of preventing cardiovascular disease;
-be used for preventing apoplexy or the transient ischemic attack relevant with the atherosclerosis of several arteries.
-be used for treating TBC or HIV.
The main diabetes of two classes are arranged: a class is type 1 diabetes, is known as insulin-dependent diabetes (IDDM), and another kind of is diabetes B, is known as non-insulin-dependent diabetes mellitus (NIDDM) (NIDDM).It is relevant that diabetes B and obesity/overweight disease and lacking taken exercise, and usually occurs gradually in the adult, and be that reduction by insulin sensitivity causes, so be called peripheral insulin resistance.This has caused the increase of compensation in Regular Insulin generates.This stage before being completed into diabetes B is referred to as metabolic syndrome, and it often causes the Atherosclerosis of artery to turn to feature with hyperinsulinemia, insulin resistant, obesity, glucose intolerance, hypertension, blood lipid dysbolism, high coagulopathy (hypercoagulopathia), blood lipid dysbolism and inflammation.After the Regular Insulin generation stopped, diabetes B formed.
In preferred embodiments, can be used for treating diabetes B according to the compound of formula (I).Compound according to formula (I) also can be used to treat the diabetes that other is selected from following type: metabolic syndrome, secondary diabetes, such as pancreas, the outer/inner secretion of pancreas or medicine-diabetes of inducing or the diabetes of other exception form, such as lipoatrophy type diabetes, myatonia (myatonic) diabetes or because the disease that the insulin receptor disorder causes.The present invention also comprises the treatment of diabetes B.Compatibly, can active nuclei acceptor, preferably PPAR (peroxisome proliferation-activated receptors) α and/or γ such as the defined formula in front (I) compound.
Formula (I) compound also can be used to treat and/or prevent obesity.Obesity is associated with the insulin resistant increase usually, and the obese person is emitting the very high danger that forms diabetes B, and it is again the Major Risk Factors that forms cardiovascular disorder.Obesity is a kind of chronic disease that is tormenting a part of crowd who is increasing in Western society, and it is not only relevant with social characteristic, and is also with the reduction in life-span and a lot of problems, relevant such as diabetes, insulin resistant and hypertension.Therefore the present invention has realized that for a long time to the needs of medicine, this medicine can reduce the amount of TBW or fatty tissue, the amount of preferred obese person's fatty tissue, for they standard body weight and do not have obvious harmful side effect.
Compound according to formula (I) also can be used for preventing and/or treating the relevant disease of amyloidosis.Amyloidosis associated conditions or the disease relevant with amyloid deposition, the result who preferably forms as protofibril or thrombocyte, comprise Alzheimer or dementia, parkinsonism, amyotrophic lateral sclerosis, spongiform encephalopathy, such as amyloid deposition in creutzfeldt-Jacob disease, cystic fibrosis, primary or Secondary cases renal amyloidosis, IgA nephropathy and artery, cardiac muscle and the middle sex organization.These diseases can for that distribute, heredity or even with infect relevant such as TBC or HIV, more early stage even mode of inheritance can appear at, but they appear at old age usually.All the aggregation with specific albumen or these albumen is relevant for every kind of disease, and these albumen are considered to the direct sources with the pathologic condition of this disease-related connection.The treatment of the disease of amyloidosis-relevant can for or (acutely) or long-term (chronically) of short-term.
Formula (I) compound also can be used for treating because the disease that amyloid assemble to reduce, prevention can cause being called protein mis folding that protofibril or thrombocyte form, treatment because the disease of the minimizing of the generation of precursor protein such as A beta-protein (amyloid beta protein), and prevents and/or treats owing to suppress or reduce the disease of proteinogen fiber, aggregate or hematoblastic formation.Prevent fibriilar gathering or formation to be also included among the present invention by administration formula (I) compound as defined above.In one embodiment, as defined above compounds, its pharmacologically acceptable salt, solvate, mixture or prodrug is used for treating TBC (tuberculosis) or HIV (HIV (human immunodeficiency virus)).
In addition, formula (I) compound can be had by administration the atherosclerosis patient of apoplexy or transient ischemic attack symptom for example of the artery of supply brain, to reduce further outbreak that may be fatal.
Formula (I) compound also can be used to treat the hyperlipidemia among the people.
In addition, formula as defined above (I) compound also is of great value treating and/or preventing the known multiple risk factors of cardiovascular disorder, and these type of Hazard Factor such as hypertension, hypertriglyceridemia and high proconvertin phospholipid complex are active.Preferably, formula (I) compound is the hyperlipidemia for the treatment of among the people.
Formula (I) compound and pharmacologically acceptable salt, solvate, prodrug or mixture can use separately, but generally with the form administration of pharmaceutical composition, its Chinese style (I) compound (activeconstituents) is used in combination with pharmaceutically acceptable auxiliary agent, diluent or carrier.
Therefore the present invention also provides and contains formula of the present invention (I) compound of treat significant quantity and the pharmaceutical composition of pharmaceutically acceptable carrier, auxiliary agent or vehicle (comprising that it makes up).
This treats the composition of the pharmaceutically active agents of significant quantity for containing or comprising (comprises or consists).It preferably contains pharmaceutically acceptable carrier, thinner or vehicle (comprising its combination).Acceptable carrier or thinner as therepic use are known at pharmaceutical field.The selection of pharmaceutical carrier, vehicle or thinner can as required route of administration and standard drug be put into practice to carry out.This pharmaceutical composition can comprise as-or interpolation-carrier, vehicle or thinner, any suitable binding agent, lubricant, suspension agent, Drug coating, solubilizing agent.
Pharmaceutical composition in the scope of the invention can comprise the additive below one or more: sanitas, solubilizing agent, stablizer, wetting agent, emulsifying agent, sweeting agent, tinting material, seasonings, reodorant, (itself can provide the compounds of this invention salt with pharmaceutical acceptable salt), buffered soln, Drug coating, antioxidant, suspension agent, auxiliary agent, vehicle and thinner.
Preferably be mixed with oral administration human or animal's preparation according to pharmaceutical composition of the present invention.This pharmaceutical composition can be mixed with also that activeconstituents can effectively be absorbed and utilize through any other administration, the preparation of intravenous administration, subcutaneous administration, intramuscular administration, intranasal administration, rectal administration, intravaginal administration or topical for example.
Special embodiment of the present invention, this pharmaceutical composition is made capsule form, and it also can be for producing the microcapsule of powder or face powder.This capsule can be for fragrance.It all is capsule with fragrance that this embodiment also comprises capsule and the fatty acid composition of the present invention that wraps into.Capsule with fragrance has more magnetism concerning the user.For therepic use above-mentioned, yes for the dosage of administration along with the compound that uses, administering mode, the treatment that needs and the illness of pointing out change.
This pharmaceutical composition can be mixed with per daily dose is provided is 10mg to 10g.Preferably, this pharmaceutical composition is mixed with provides the described composition of per daily dose between 50mg and 5g.Most preferably, this pharmaceutical composition is mixed with provides the described composition of per daily dose between 100mg and 1g.The meaning of per daily dose is per 24 hours dosage.
Certainly, the dosage of administration can change along with employed compound, administering mode, required treatment and the illness of pointing out.Generally speaking, the attending doctor will determine the optimal actual dose of single patient.Concrete dosage level and dose frequency can change concerning any specific patient, it will depend on many factors, comprise the mode of the metabolic stability of activity, this compound activity of employed specific compound and time length, age, body weight, general health situation, sex, diet, administration and time, the speed of drainage, the combination of medicine, severity and the single treatment of carrying out of particular disorder.Medicament of the present invention and/or pharmaceutical composition can be according to 1-10 scheme administrations every day, as once a day or twice.For oral and administered parenterally people patient, the dosage that the dosage level of medicament can be single dose or separates.
The present invention relates to the fatty acid composition that contains formula (I) compound on the other hand.This fatty acid composition that contains formula (I) compound has increased the natural biological effect of DHA, and it is the result that genetic expression is regulated, and will accumulate in the free fatty acids storehouse according to derivative of the present invention.
This fatty acid composition can comprise formula (I) compound of 60-100 % by weight scope, and all weight percents are based on that total weight of fatty acid composition calculates.In the preferred embodiments of the invention, at least 80 % by weight of this fatty acid composition are formula (I) compound.More preferably, at least 90 of this fatty acid composition % by weight are formula (I) compound.Most preferably, formula (I) compound has consisted of the fatty acid composition that surpasses 95 % by weight.
This fatty acid composition can further comprise at least a lipid acid (entirely-Z)-5,8,11,14,17-timnodonic acid (EPA), (complete-Z)-4,7,10,13,16,19-docosahexenoic acid (DHA), (complete-Z)-6,9,12,15,18-, 21 carbon 5 alkene acids (HPA) and (complete-Z)-7,10,13,16,19-clupanodonic acid (DPAn-3), (complete-Z)-8,11,14,17-eicosatetraenoic acid (ETAn-3) or its combination.In addition, this fatty acid composition can comprise (complete-Z)-4,7,10,13,16-clupanodonic acid (DPAn-6) and/or (complete-Z)-Arachidonic Acid (ARA) or derivatives thereof.This fatty acid composition also can comprise at least these lipid acid or its combination of derivative form.This derivative compatibly replaces in the mode identical with the DHA derivative of as defined above formula (I).
Can comprise (complete-Z ω-3)-6,9,12,15 according to fatty acid composition of the present invention, 18-21 carbon 5 alkene acids (HPA) or derivatives thereof, its amount is at least 1% weight or the weight of 1-4%.
In addition, can comprise the omega-fatty acid or derivatives thereof rather than have EPA and the DHA of 20,21 or 22 carbon atoms according to fatty acid composition of the present invention, its amount position at least 1.5 % by weight or at least 3% weight.
In the special embodiment of the present invention, this fatty acid composition is pharmaceutical composition, nutritive compositions or dietary composition.
This fatty acid composition can further comprise the pharmaceutically acceptable antioxidant of significant quantity.Preferably, this antioxidant is the mixture of tocopherol or tocopherols.In preferred embodiments, this fatty acid composition further comprises its amount and is every gram fatty acid composition at the most tocopherol of 4mg or the mixture of tocopherols.Preferably, this fatty acid composition comprises the tocopherol of every gram composition 0.2-0.4mg based on the gross weight of composition.
Another aspect of the present invention provides fatty acid composition or its any pharmacologically acceptable salt, solvate, prodrug or the mixture of formula (I) compound that contains as defined above, mixture its as medicine and/or be used in the treatment.This type of fatty acid composition can be used to prevent and/or treat such as the listed identical illness out of top formula (I) compound.
When this fatty acid composition was used as medicine, it was with the amount administration for the treatment of or pharmaceutical activity.
In preferred embodiments, this fatty acid composition oral administration human or animal.
The present invention also provides the compound of formula (I) as defined above, or the purposes of its pharmacologically acceptable salt, solvate, prodrug or mixture, its purposes in the medicine of preparation control weight loss and/or the increase of prevention body weight; It treats and/or prevents purposes in the medicine of obesity or overweight disease in preparation; It prevents and/or treats purposes in the medicine of diabetes among the human or animal in preparation; It treats and/or prevents purposes in the medicine of the relevant disease of amyloidosis in preparation; Its purposes in the medicine of the multiple risk factors for preparing treatment and preventing cardiovascular disease such as hypertension, hypertriglyceridemia and high proconvertin phospholipid complex activity; Its purposes in preparation treatment TBC or HIV medicine; Its purposes in the medicine for preparing the prevention apoplexy relevant with the atherosclerosis of several arteries or transient ischemic attack; Its preparation reduce in the mammalian triglyceride level and/or in the people patients serum purposes in the medicine of rising HDL cholesterol levels; It treats and/or prevents the purposes in the medicine of the multiple metabolic syndrome that is called " metabolic syndrome " in preparation.These all embodiments also comprise the purposes of the fatty acid composition of the formula for the preparation of medicine (I) compound that contains as defined above.The present invention relates to the purposes of Alpha-hydroxy-DHA in preparation medicine as listed above in addition.
The present invention also relates to control the method for weight loss and the increase of prevention body weight, wherein contain this fatty acid composition administration human or animal of at least formula (I) compound as defined above.
In addition, the present invention relates to treat and/or prevent the method for obesity or overweight disease, wherein contain this fatty acid composition administration human or animal of at least formula (I) compound as defined above.
In the preferred embodiments of the invention, the present invention relates to prevent and/or treat the method for diabetes, wherein contain this fatty acid composition administration human or animal of at least formula (I) compound as defined above.Preferably, diabetes are diabetes Bs.
Other side of the present invention relates to;
-treat and/or prevent the method for the relevant disease of amyloidosis;
-treat and/or prevent the method for the multiple risk factors of cardiovascular disorder;
The apoplexy that-prevention is relevant with the atherosclerosis of several arteries or the method for transient ischemic attack;
The composition administration human or animal of wherein containing this lipid acid of at least formula (I) compound as defined above.
The derivative of fatty acid of formula (I) can prepare most effectively from DHA.Impure DHA (namely be not 100% DHA) such as starting raw material, then final fatty acid composition will contain the mixture of the DHA derivative that limits just like the front, and a certain amount of lipid acid rather than DHA, wherein these lipid acid replace in the mode identical with the fatty acid analog of the novelty of formula (I).The present invention also comprises this type of embodiment.
In another embodiment of the invention, formula (I) compound from (complete-Z)-DHA (DHA) preparation, wherein said DHA obtains from plant, microorganism and/or animal source or their combination.Preferably, described DHA derives from marine animal oil, such as fish oil.
Lipid acid in the composition also can obtain from plant, microorganism and/or animal source or their combination.Therefore, the present invention also comprises the fatty acid composition for preparing from microbial oil.
The invention provides the method for the fatty acid analog of preparation new formula (I) as defined above.
DHA produces from biogenic such as ocean, microorganism or vegetation fat.All possible starting material are the mixture of fatty acid triglycercide form, and wherein DHA has only consisted of the part of lipid acid.Usually the concentration of DHA is 40% in microorganism fat, is 10-25% in marine animal fat.The vegetation fat that contains DHA is developed, and the fat that contains high DHA concentration is desirably in the future and occurs.
The first step is generally triglyceride level and is converted into free lipid acid or monoesters.Preferred ester is methyl esters or ethyl ester, but also can be other ester.In this way, the lipid acid that is combined on the triglyceride level is out separated from one another with three, three ground, thereby so that is separated into possibility.Separate DHA and can use several method from other lipid acid, wherein the most general method is to utilize the plain precipitator method separation of fatty acids of urea according to volatility with the short-path distillation separation of fatty acids and according to degree of saturation not.The with good grounds degree of unsaturation of other the method for having reported uses the effect of Silver Nitrate mixture to separate, separate and use by the enzymatic esterification coupling of lipid acid selected fat short-path distillation the method for the counter-current extraction of supercritical co.
The most important challenge that the production of pure DHA is relevant is exactly to separate in its other C20-22 highly unsaturated fatty acids that exists from all can be originated.These lipid acid have the character similar to DHA, so that there is not a kind of method above-mentioned that the separation of abundant degree is provided.To certain micro-organisms height DHA fat, it has low-down C20-22 content of highly unsaturated fatty acids, and independent short-path distillation or other method coupling of mentioning can provide the purity greater than 90%.
Great majority contain the C20-22 highly unsaturated fatty acids that fatty DHA also contains a great deal of, for example EPA (20:5n-3), n-3DPA (22:5n-3), HPA (21:5n-3) and other unsaturated fatty acids.From then on DHA is separated only methods availalbe in the class lipid acid is the high performance liquid chromatography of preparation type, and its stationary phase is the silica gel of silica gel or Silver Nitrate dipping, and moving phase is selected from organic solvent or supercritical co.Use this method can obtain surpassing the DHA of 97% purity.Yet, should be noted that production cost along with concentration sharply increases, the cost of the DHA such as 97% exceeds more than 5 times than the cost of 90% DHA.
The DHA that has purity and be 90,95 eller 97% contains other a small amount of lipid acid.For example, purity is that 97% DHA contains n-3DPA (22:5n-3), longer chain fatty acid is also arranged, for example EPA (20:5n-3), HPA (21:5n-3) and other lipid acid.Yet this other lipid acid will react in the mode that is similar to DHA, thereby the derivative of alpha-substitution is provided.
Because DHA and n-6DPA (22:5n-6 generally exists with low-down concentration) known onlyly can provide the lipid acid of gamma lactone by come cyclisation with first pair of key, so organic synthesis can provide purification process.Lactonization after the purifying and hydrolysis may turn back to the method for DHA and also can use, but this path is higher than HPLC cost.
In one embodiment, R wherein 1(or R 2) prepare according to the methods below (scheme 1) for the formula of hydrogen (I) compound.Through suitable improvement, these methods also can be for the preparation of R wherein 1And R 2All be for example C 1-C 7The compound of the general formula of alkyl, benzyl, halogen, benzyl, alkenyl or alkynyl (I) representative.
R wherein 1Be hydrogen and R 2Be C 1-C 7The compound of the general formula of alkyl, benzyl, halogen, benzyl, alkenyl or alkynyl (I) representative reacts preparation by DHA ester and strong non-nucleophilicity alkali such as lithium diisopropylamine or hexamethyl dimethyl silanyl potassium amide/sodium at solvent such as tetrahydrofuran (THF), ether under-60 to-78 ℃ temperature, obtain the enol form (step 1) of ester.
(scheme 1)
Figure DEST_PATH_G200680024064920080311D000011
The enol form of this ester and electrophilic reagent such as alkylogen such as iodoethane, benzyl chloride, carboxylic acid halides such as Acetyl Chloride 98Min., benzoyl bromide, the reactions such as acid anhydrides such as diacetyl oxide or close electric halide reagent such as N-fluorobenzene sulphonamide (NFSI) obtain mono-substituted derivative (step 1).The ester that obtains is 15-40 ℃ temperature, and in solvent such as ethanol or methyl alcohol, the aqueous solution by adding alkali such as lithium hydroxide or sodium hydroxide further hydrolysis obtains carboxylic acid derivative.
When processing DHA EE with highly basic, claisen condensation occurs in DHA EE.This condensation product has interesting biological activity.Therefore, in one embodiment of the invention have also disclosed condensation product above-mentioned (intermediate) and this product is used for treating and/or preventing disease according to the present invention purposes.
In another embodiment, the compound of general formula (I) representative is by following method synthetic (scheme 2).
(scheme 2)
Figure DEST_PATH_G200680024064920080311D000021
R wherein 1Be hydrogen and R 2In solvent such as tetrahydrofuran (THF), ether, reaction obtains the enol form (step 4) of ester to the compound of the general formula of representation hydroxy, alkoxyl group, acyloxy (I) representative under-60 to-78 ℃ temperature by DHA ester and strong non-nucleophilic base such as lithium diisopropylamine or hexamethyl dimethyl silanyl potassium amide/sodium.The enol form of this ester and oxygen source such as third ring (dioxirane), 2-(phenyl sulfonyl)-3-phenyl oxygen azepine the third pyridine (oxaziridine) of dimethyl dioxa, molecular oxygen; from different additive such as trimethyl phosphite or different catalyzer such as the reaction of Ni (II) mixture, obtain Alpha-hydroxy DHA ester (step 5).The reaction in solvent such as THF or DMF of secondary alcohol and alkali such as sodium hydride produces alkoxide (alkoxide), and it reacts (step 6) with different electrophilic reagent such as alkyl iodide such as methyl iodide, iodoethane, bromobenzyl or carboxylic acid halides such as Acetyl Chloride 98Min., benzoyl bromide.Under 15-40 ℃ temperature, ester obtains carboxylic acid derivative (step 7) by the aqueous hydrolysis that adds alkali such as lithium hydroxide or sodium hydroxide in solvent such as ethanol or methyl alcohol.
Introduce in other functional group at alpha-position of the present invention, this hydroxyl-DHA ester is useful intermediate.From the reactions such as different nucleophilic reagent such as ammonia, amine, mercaptan before, this hydroxy functional group can be by being converted into halogenide or tosylate activates.Can utilize equally the Mitsunobu reaction that hydroxyl is converted into other functional group (Mitsunobu, O, Synthesis, 1981,1).
The compound of general formula as defined above (I) representative also can synthesize by the combination of previously described different methods.The method of mentioning above the present invention includes.
The present invention further provides the method for preparing pharmaceutical composition of the present invention, it comprises at least as top defined formula (I) compound or its pharmacologically acceptable salt, solvate, mixture or prodrug, mixes with pharmaceutically acceptable auxiliary agent, diluent or carrier.
The compound of enantiomeric pure can prepare by the resolution of racemate such as formula defined above (I) compound.The fractionation of formula (I) compound can utilize known method for splitting to carry out, and such as the auxiliary agent reaction of through type (I) compound and enantiomeric pure, obtains the mixture of diastereomer, and it can separate by chromatography.After this two kinds of enantiomers of formula (I) compound obtain from the diastereomer that has separated such as hydrolysis again by the method for routine.
May be by carrying out such as the asymmetric introducing of top defined substituting group at the alpha-position of DHA with stoichiometric chirality assistant agent.Proved the use chirality
Figure 2006800240649_6
Azoles alkane-2-ketone is special effective means.From chirality N-acyl group
Figure 2006800240649_7
The enolate that azoles alkane is derived can be with multiple electrophilic reagent with high three-dimensional control mode quencher (Schaad 1996 for Ager, Prakash).
Embodiment
The present invention will describe in further detail by the following examples, and it is not construed as limiting the invention.Structure is by mass spectrum (MS) conclusive evidence in an embodiment.Should be noted that derivative of fatty acid can prepare with the starting material of the DHA that contains low levels and moderate content (i.e. the DHA of about 40-60 % by weight).
Synthetic schemes
The preparation of Alpha-Methyl DHA EE (PRB-1)
At N 2Under 0 ℃, with butyllithium (228ml, 0.37mol, 1.6M hexane solution) be added drop-wise to the diisopropylamine (59.5ml of stirring, 0.42mol) anhydrous THF (800ml) solution in, the solution that obtains is 0 ℃ of lower stirring 30 minutes, is cooled to-78 ℃ and restir 30 minutes, then in 2 hours, drip anhydrous THF (500ml) solution of DHA EE (100g, 0.28mol).Then blackish green solution add MeI (28ml, 0.45mol)-78 ℃ of lower stirrings 30 minutes.Solution was risen to-20 ℃ in 1.5 hours, in so fall back (1.51), and with heptane extraction (2 * 800ml).The organic phase that merges is washed with 1MHCl (11), dry (Na 2SO 4), filter and vaporising under vacuum.Product uses the anhydrous flash chromatography method of silica gel purifying, with heptane/EtOAc (99: 1) wash-out, obtains 50g (48%) title compound, is light yellow oil;
1H-NMR(200MHz,CDCl 3)δ1.02(t,J?7.5Hz,3H),1.20(d,J?6.8Hz,3H),1.29(t,J?7.1Hz,3H),2.0-2.6(m,5H),2.8-3.0(m,10H),4.17(t,J?7.1Hz,2H),5.3-5.5(m,12H);
MS (electron spray(ES)): 393[M+Na].
The preparation of α-ethyl DHA EE (PRB-2)
At N 2Under 0 ℃, with butyllithium (440ml, 0.67mol, 1.6M hexane solution) be added drop-wise to the diisopropylamine (111ml of stirring, 0.78mol) anhydrous THF (750ml) solution in, the solution that obtains was stirred 45 minutes under-78 ℃, then drip anhydrous THF (1.6l) solution of DHA EE (200g, 0.56mol).Dropwising of 4 hours lactones.Blackish green solution-78 ℃ of lower stirrings 30 minutes, is then added MeI (65ml, 0.81mol).Solution is warming up to-40 ℃, and then adds EtI (5ml, 0.06mol), rise at last-15 ℃ (from 3 hours-78 ℃ of times spent), then mixture is poured into water and with hexane extraction (2x).The organic phase that merges 1M HCl and water washing, dry (Na 2SO 4), filter and vaporising under vacuum.Product uses flash chromatography on silica gel method purifying, uses heptane/EtOAc (99: 1 then 50: 1) wash-out, obtains 42.2g (20%) title compound, is yellow oil;
1H-NMR(200MHz,CDCl 3)δ0.8-1.0(m,6H),1.2-1.4(m,4H),1.5-1.7(m,2H),2.12(m,2H),2.3-2.5(m,2H),2.8-3.0(m,10H),4.18(t,J?7.1Hz,2H),5.3-5.6(m,12H);
MS (electron spray(ES)): 407[M+Na].
The preparation of α-oxyethyl group DHA-EE (PRB-3)
Figure S2006800240649D00291
At N 2Under-78 ℃, NaH (84.1mg to 60%, 2.1mmol) the suspension of THF (5mL) in drip Alpha-hydroxy-DHA-EE (PRB-12) (372mg, 1.00mmol) the solution of THF (5mL), the mixture that obtains is-78 ℃ of lower stirrings 20 minutes, then drip iodoethane (0.24mL, 3.01mmol).Reaction mixture is warming up to ambient temperature overnight gradually.Add saturated NH 4The Cl aqueous solution (15mL) is also used extracted with diethyl ether mixture (25mL * 2), and organic phase is washed with salt solution (25mL), dry (Na 2SO 4), to filter, evaporation and use flash chromatography on silica gel method purifying use heptane/EtOAc (95: 5) wash-out under the vacuum, obtain 68mg (17%) product, are yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.94(t,J=7.5Hz,3H),1.16-1.29(m,6H),2.05(quint,J=7.2Hz,2H),2.50(m,2H),2.76-2.84(m,10H),3.33-3.48(m,1H),3.53-3.71(m,1H),3.83(dd,J=6.8Hz,J=6.2Hz,1H),4.18(q,J=7.1Hz,2H),5.31-5.45(m,12H);
13C NMR (50MHz, CDCl 3) δ 14.2,15.1,20.5,25.5,25.6,25.7,31.0,60.8,66.0,78.7,124.1,127.0,127.8,127.9,128.0 (2 signals), 128.2 (2 signals), 128.5,130.7,132.0,172.5 (3 signal hidings)
MS (electron spray(ES)): 423[M+Na] +
The preparation of α-fluorine DHA EE (PRB-4)
At N 2Under-78 ℃, in 15 minutes, anhydrous THF (10ml) solution of LDA (2.1ml, 4.2mol, the THF/ heptane of 2M/ethylbenzene solution) is added drop-wise in anhydrous THF (30ml) solution of DHA EE (1g, 2.8mmol).Then add NFSi (1.06g, 3.4mmol).Solution is warming up to room temperature and stirred 70 hours.Mixture is poured into water and extracts with hexane (2x).The organic phase that merges 1M HCl and water washing, dry (Na 2SO 4), filter and vaporising under vacuum; MS (electron spray(ES)): 397[M+Na].
α, the preparation of alpha-alpha-dimethyl DHA EE (PRB-5)
At N 2Under 0 ℃, butyllithium (100ml, 0.17mol, the hexane solution of 1.6M) is added drop-wise in anhydrous THF (100ml) solution of diisopropylamine (28ml, 0.20mol) of stirring.The solution that obtains 0 ℃ of lower stirring 30 minutes, is cooled to-78 ℃ and drip anhydrous THF (200ml) solution of DHA EE (50g, 0.14mol).The cyan solution that obtains was stirred 30 minutes under-78 ℃, then add MeI (17ml, 0.28mol).This solution is warming up to-10 ℃, then is poured into water and extracts with hexane (2x).The organic phase that merges is washed with 1M HCl, dry (Na 2SO 4), filter and vaporising under vacuum.
Repeat above-mentioned steps, but use Alpha-Methyl DHA EE crude product, rather than DHA EE.Product uses the anhydrous flash chromatography method of silica gel purifying, uses heptane/EtOAc (99: 1 then 98: 2) wash-out, obtains 31.6g (59%) title compound, is light yellow oil;
1H-NMR(200MHz,CDCl 3)δ1.01(t,J?7.5Hz,3H),1.21(s,6H),1.28(t,J7.1Hz,3H),2.08(m,2H),2.34(d,J?6.8Hz,2H),2.8-3.0(m,10H),4.15(q,J?7.5Hz,2H),5.3-5.6(m,12H);
13C-NMR (50MHz, CDCl 3) δ 14.7,21.0,25.3,26.0,26.1,38.3,42.8,60.7,125.8,127.4,128.3,128.5,128.6,128.7,129.0,130.7,132.4,177.9; MS (electron spray(ES)): 385[M+H].
The preparation of alpha-methylmercapto DHA (PRB-6)
At N 2Under 0 ℃, alpha-iodine DHA EE (0.5g, 1.04mmol) is dissolved among the 20mL THF.Add MeSNa (80mg, 1.14mmol), reaction mixture stirs several minutes, then dilutes with heptane.
Organic phase water (2x) washing, dry (Na 2SO 4) and vaporising under vacuum.Required product uses flash chromatography method (heptane/EtOAc (30: 1)) to separate, and obtains alpha-methylmercapto DHA EE, is light yellow oil.Alpha-methylmercapto DHA EE is dissolved among 10mL EtOH and the 10mL THF.Add the LiOH (0.39g, 9.2mmol) that is dissolved in the 5mL water to this solution.Reaction mixture at room temperature stirs and spends the night, then water and heptane dilution.Organic moiety is with 1M LiOH (2x) extraction, and with the water that merges with 5M HCl acidifying after with extracted with diethyl ether (2x).The organic phase that merges salt solution and water washing, dry (Na 2SO 4) and vaporising under vacuum, obtain 183mg (47%) title compound, be light yellow oil;
1H-NMR(200MHz,CDCl 3)δ.0.98(t,J?6.6Hz,3H),1.95-2.65(m,7H),2.72-3.05(m,10H),3.12-3.43(m,1H),5.20-5.70(m,12H),10.65(br?s,1H);
13H-NMR(50MHz,CDCl 3)δ14.7,21.0,25.9,26.0,26.2,28.8,125.4,127.4,128.1,128.3,128.4,128.7,128.9,129.0,131.6,132.4,177.0。
The preparation of α-ethylmercapto group DHA EE (PRB-7)
At N 2Under 0 ℃, alpha-iodine DHA EE (11g, 23mmol) is dissolved among the 100mL THF.Add EtSNa (2.1g, 25mmol) in this solution, then 0 ℃ of lower stirring 1 hour.Then reaction dilutes with heptane with 1M HCl quencher.Organic phase water (2x) washing, dry (Na 2SO 4) and vaporising under vacuum.Required product uses the flash chromatography method, and (heptane/EtOAc (30: 1) separates, and obtains 7.3g (76%) title compound, is light yellow oil;
1H-NMR(200MHz,CDCl 3)δ1.1-1.3(m,9H),2.05(m,2H),2.3-2.7(m,4H),2.7-2.9(m,10H),3.25(m,1H),4.17(q,J?7.1Hz,2H),5.3-5.5(m,12H);
MS (electron spray(ES)): 439[M+Na].
α, the preparation of α-diethyl DHA EE (PRB-8)
At N 2Under 0 ℃, butyllithium (38.6ml, 0.62mol, the hexane solution of 1.6M) is added drop-wise in anhydrous THF (200ml) solution of diisopropylamine (9.1ml, 0.65mol) of stirring.The solution that obtains is 0 ℃ of lower stirring 30 minutes, is cooled to-78 ℃ and drip anhydrous THF (100ml) solution of DHA EE (20.0g, 0.56mol).The blackish green solution that obtains was stirred 30 minutes under-78 ℃, then add EtI (6.8ml, 0.84mol).This solution is warming up to-10 ℃, then is poured into water and extracts with hexane (2x).The organic phase that merges is washed with 1M HCl, dry (Na 2SO 4), filter and vaporising under vacuum.
Repeat above-mentioned steps, but use α-ethyl DHAEE crude product, rather than DHAEE.After adding EtI, reaction mixture is warming up to envrionment temperature and stirs spend the night.Product uses the anhydrous flash chromatography method of silica gel purifying, uses heptane/EtOAc (99: 1 then 98: 2) wash-out, obtains 10.0g (43%) title compound, is light yellow oil;
1H-NMR(200MHz,CDCl 3)δ0.83(t,J?7.4Hz,6H),0.94(t,J?5.8Hz,3H),1.28(t,J?7.1Hz,3H),1.63(q,J?7.4Hz,4H),2.10(m,2H),2.34(d,J?6.9Hz,2H),2.8-3.0(m,10H),4.15(q,J?7.5Hz,2H),5.3-5.6(m,12H);
13C-NMR(50MHz,CDCl 3)δ8.9,14.7,21.0,23.1,25.9,26.0,26.2,27.4,31.2,50.1,60.6,125.5,127.4,128.3,128.6,128.9,130.5,132.4,177.1;
MS (electron spray(ES)): 413.3[M+H], 435.3[M+Na].
The preparation of α-benzyl DHA EE (PRB-9)
Remain under 0 ℃, anhydrous THF (20mL) solution to the diisopropylamine (0.91mL, 6.46mmol) that stirs under inert atmosphere drips n-BuLi (hexane solution of 1.6M, 3.86mL, 6.18mmol).Mixture is 0 ℃ of lower stirring 30 minutes, is down to-78 ℃ and stirred 5 minutes under this temperature.Drip anhydrous THF (10mL) solution of DHA EE (2.0g, 5.62mmol), and this mixture was stirred 20 minutes under-78 ℃, then add bromobenzyl (0.80mL, 6.74mmol).The solution that obtains was warming up to 0 ℃ in 3 hours, and is distributed in water (100mL) and the heptane (100mL).Water layer washs with 1M HCl and dry (Na with heptane (50mL) extraction, the organic layer of merging 2SO 4).And under reduced pressure concentrated and with flash chromatography method purifying (heptane: EtOAc 99: 1), obtain 1.05g (42%) title compound, be colorless oil;
1H-NMR(200MHz,CDCl 3):δ0.99(t,3H),1.18(t,3H),2.08-2.16(m,2H),2.35-2.42(m,2H),2.74-2.98(m,13H),4.09(q,4H),5.38-5.50(m,10H),7.19-7.36(m,5H);
13C-NMR(50MHz,CDCl 3):δ14.61,14.71,20.99,25.98,26.07,30.07,38.32,48.02,60.88,126.75,126.83,127.46,128.31,128.45,128.53,128.58,128.86,128.77,129.01,129.35,130.55,132.46,138.89,175.39。
MS (electron spray(ES)): 447.3[M+H], 469.3[M+Na].
The preparation of α-ethyl sulfinyl DHA EE (PRB-10)
Remain under-20 ℃, under inert atmosphere to the 15mL CHCl of α-ethylmercapto group DHA EE (0.5g, 1.3mmol) 3Solution adds the 10mL CHCl of MCPBA (0.22g, 1.3mmol) 3Solution.Reaction mixture was stirred 2 hours under this temperature, filter and use saturated NaHCO 3Solution washing.Water CHCl 3Extract 2 times, and with the organic phase water and the salt water washing that merge, use Na 2SO 4Drying is filtered and is concentrated.Product separates from residue through flash chromatography method (using 8: 2 wash-outs of hexane: EtOAc), obtains 0.35g (70%) title compound.
1H?NMR(200MHz,CDCl 3):δ0.99(t,3H),1.27-1.45(m,6H),2.09(m,2H),2.79-2.94(m,14H),3.55(m,1H),4.25(q,2H),5.37-5.59(m,12H)。
13C?NMR(50MHz,CDCl 3):δ7.97,14.58,14.68,20.95,23.68,25.17,25.93,26.04,44.20,45.15,62.30,64.08,123.91,124.47,127.41,127.86,128.26,128.40,128.44,128.72,128.72,128.96,129.12,132.42,132.47,174.55。
MS (electron spray(ES)): 455.3[M+Na].
The preparation of α-thiophenyl DHA-EE (PRB-11)
Drip acetone (110mL) solution of thiophenol sodium (sodium phenyl sulfide) (1.039g, 7.86mmol) to acetone (20mL) solution of alpha-iodine-DHA-EE (PRB-15) (3.40g, 7.05mmol).The mixture that obtains was stirred one and a half hours at ambient temperature, evaporation and use flash chromatography on silica gel method purifying under the vacuum, use heptane/EtOAc 200: 1-95: 5 wash-outs, obtain 2.35g (72%) product, be yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.97(t,J=7.5Hz,3H),1.18(t,J=7.1Hz,3H),2.09(quint,J=7.1Hz,2H),2.54-2.66(m,2H),2.83-2.86(m,10H),3.67(dd,J=6.8Hz,J=8.3Hz,1H),4.12(q,J=7.1Hz,2H),5.24-5.49(m,12H),7.28-7.33(m,3H),7.46-7.50(m,2H)。
13C NMR (50MHz, CDCl 3) δ 14.0,14.2,20.5,25.5,25.6,25.7,29.4,50.6,61.1,125.1,127.0,127.7,127.9,128.0,128.3,128.42,128.45,128.9,131.2,132.0,133.0,133.2,174.1 (5 signal hidings).
MS (electron spray(ES)): 465[M+H] +, 487[M+Na] +
HRMS (EI) calculated value: C 30H 40O 2S:464.2749, measured value: 464.2741.
The preparation of Alpha-hydroxy-DHA-EE (PRB-12)
Figure S2006800240649D00332
At N 2Under atmosphere and-78 ℃, drip hexane (87.5mL, the 140mmol) solution of 1.6M BuLi to anhydrous THF (40mL) solution of diisopropylamine (19.76mL, 140mmol).The mixture that obtains was stirred 15 minutes under-78 ℃, then drip THF (80mL) solution of DHA-EE (24.99g, 70.1mmol).The blackish green reaction mixture that obtains was stirred 1 hour under-78 ℃, then drip triethyl-phosphite (12.2mL 70.1mmol), use O 2Spend the night by the reaction mixture bubbling, keep simultaneously reaction mixture-78 ℃ lower 5 hours, then slowly be warming up to room temperature.Add saturated NaHCO 3(100mL) aqueous solution, mixture extracts with ether (200mL * 2).With the dry (Na of organic phase 2SO 4), filter and vaporising under vacuum, use flash chromatography on silica gel method purifying, use heptane/EtOAc99: 1-95: 5 wash-outs, obtain 4.52g (17%) product, be yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.92(t,J=7.5Hz,3H),1.24(t,J=7.1Hz,3H),2.02(quint,J=7.1Hz,2H),2.44-2.54(m,2H),2.74-2.87(m,10H),4.13-4.24(m,3H),5.25-5.94(m,12H)。
13C NMR (50MHz, CDCl 3) δ 14.0,14.1,20.4,25.4,25.5,25.6,32.0,61.5,69.9,123.3,126.9,127.7,127.9,128.08,128.1,128.2,128.4,131.3,131.8,174.4 (4 signal hidings).
MS (electron spray(ES)): 395[M+Na] +
HRMS (ES) calculated value: C 24H 36O 3Na:395.2556, measured value: 395.2543.
The preparation of Alpha-Methyl-DHA acid amides (PRB-13)
Figure S2006800240649D00341
To Alpha-Methyl-DHA (PRB-1 FA) (3.13g, 9.1mmol) and oxalyl chloride (8.0mL, 94.5mmol) toluene (90mL) solution in add DMF (0.1mL), the mixture that obtains was stirred 15.5 hours under envrionment temperature and nitrogen atmosphere.Then with the mixture vaporising under vacuum, resistates is dissolved in THF (100mL), is cooled to 0 ℃ and drip NH 3The aqueous solution (20mL).Remove ice bath and mixture was stirred 4 hours at ambient temperature, add entry (50mL), water is with ether (2 * 100mL) extractions.The saturated NH of organic phase 4The Cl aqueous solution (50mL) washing, dry (Na 2SO 4), filter and vaporising under vacuum, use flash chromatography on silica gel method purifying, use CH 2Cl 2/ 2M NH 397.5: 2.5 wash-outs of MeOH solution, obtain 2.51g (80%) product, be yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.91(t,J=7.5Hz,3H),1.10(d,J=9.8Hz,3H),1.94-2.11(m,3H),2.19-2.35(m,2H),2.76-2.77(m,10H),5.18-5.45(m,12H),6.03(s,1H),6.72(s,1H)。
13C NMR (50MHz, CDCl 3) δ 14.6,17.6,20.8,25.8,25.9,32.0,41.0,127.3,128.1,128.4,128.6,128.8,130.1,132.2,179.6 (8 signal hidings).
MS (electron spray(ES)): 342[M+H] +, 364[M+Na] +
HRMS (EI) calculated value: C 23H 35NO:341.2719, measured value: 341.2707.
The preparation of α-methoxyl group-DHA-EE (PRB-14)
Figure S2006800240649D00351
At N 2Under atmosphere and-78 ℃, to 60%NaH (61.1mg, 1.53mmol) THF (5mL) solution drip Alpha-hydroxy-DHA-EE (PRB-12) (373mg, 1.00mmol) THF (5mL) solution, the mixture that obtains is-78 ℃ of lower stirrings 20 minutes, then drip methyl iodide (0.13mL, 2.09mmol).In 5 hours reaction mixture is warming up to room temperature gradually.Add saturated NH 4The Cl aqueous solution (15mL) and mixture use salt solution (25mL) to wash with ether (25mL * 2) extraction, organic phase, dry (Na 2SO 4) filter, evaporation and use flash chromatography on silica gel method purifying under the vacuum, use heptane/EtOAc99: 1-4: 1 wash-out, obtain 136mg (35%) product, be yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.92(t,J=7.5Hz,3H),1.24(t,J=7.1Hz,3H),2.03(quint,J=7.3Hz,2H),2.48(t,J=5.7Hz,2H),2.73-2.82(m,10H),3.34(s,3H),3.74(t,J=6.2Hz,1H),4.17(q,J=7.1Hz,2H),5.24-5.43(m,12H)。
13C NMR (50MHz, CDCl 3) δ 14.1,20.4,25.4,25.5,25.7,30.6,57.9,60.9,80.8,123.7,126.9,127.71,127.73,127.92,127.94,128.07,128.1,128.2,128.4,130.7,131.8,171.9 (3 signal hidings).
MS (electron spray(ES)): 409[M+Na] +
HRMS (ES) calculated value: C 25H 38O 3Na:409.2713, measured value: 409.2711.
The preparation of alpha-iodine DHA EE (PRB-15)
At N 2Under-20 ℃, diisopropylamine (20mL, 140mmol) is dissolved among the 150mL THF, in this mixture, drip n-BuLi (88mL, 140mmol, 1.6M), then solution is cooled to-78 ℃.THF (250mL) solution of DHA EE (50g, 140mmol) is added drop-wise in the top solution, and reaction mixture at room temperature stirred 30 minutes.At-78 ℃ and N 2Lower, the mixture that obtains is added drop-wise to I 2In the THF of (42.8g, 169mmol) (400mL) solution.Reaction is diluted with 1M HCl quencher and with heptane.Organic phase is with 10% Na 2S 2O 3(2x) washing, dry (Na 2SO 4), filter and vaporising under vacuum.Required product uses flash chromatography method (heptane/EtOAc (100: 1)) to separate, and obtains 11.0 g (16%) title compound, is light yellow oil; MS (electron spray(ES)): 505[M+Na].
The preparation of alpha-iodine-DHA-EE (PRB-15)
Under-78 ℃ and nitrogen atmosphere, drip hexane (158mL, the 253mmol) solution of 1.6M BuLi in anhydrous THF (150mL) solution of diisopropylamine (42mL, 298mmol).The mixture that obtains was stirred 35 minutes under-78 ℃, then drip THF (300mL) solution of DHA-EE (75.05g, 210mmol).The blackish green reaction mixture that obtains was stirred 30 minutes under-78 ℃, then drip I 2The THF of (91.06g, 359mmol) (200mL) solution.Reaction mixture is-78 ℃ of lower stirrings 20 minutes, and then water (200mL) quencher is reacted, and extracts with heptane (300mL).Organic phase is washed dry (Na with 1M HCl (150mL) and water (200mL) 2SO 4), filter and vaporising under vacuum.The crude product that obtains uses heptane/EtOAc (100: 1) wash-out with flash chromatography on silica gel method purifying, obtains 26.14g (26%) product, is yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.94(t,J=7.5Hz,3H),1.24(t,J=7.1Hz,3H),2.04(quint,J=7.1Hz,2H),2.69-2.84(m,12H),4.17(q,J=7.1Hz,2H),4.22(t,J=7.9Hz,1H),5.24-5.49(m,12H)。
13C NMR (50MHz, CDCl 3) δ 13.7,14.2,25.5,26.0 (2 signals), 25.8,34.0,61.7,126.1,127.0,127.4,127.8,127.9,128.0,128.2,128.5,128.5,131.6,131.9,170.9 (4 signal hidings).
MS (electron spray(ES)): 505[M+Na] +
The preparation of alpha-amino group-DHA-EE (PRB-17)
Figure S2006800240649D00362
To α-phthalimide-based-DHA-EE (313.5mg, 0.62mmol) EtOH (5mL) solution in add hydrazine hydrate (46 μ l, 0.95mmol), the mixture that obtains was refluxed 15.5 hours under nitrogen atmosphere, follow vaporising under vacuum, and use flash chromatography on silica gel method purifying, use CH 2Cl 2: 7M NH 3Methanol solution (99: 1-95: 1) wash-out, obtain 149mg (64%) product, be yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.91(t,J=7.5Hz,3H),1.22(t,J=7.1Hz,3H),1.72(bs,2H),2.02(quint.,J=7.2Hz,2H),2.39-2.46(m,2H),2.73-2.82(m,10H),3.47(bs,1H),4.13(q,2H),5.23-5.56(m,12H)。
13C NMR (50MHz, CDCl 3) δ 14.1,20.4,25.4,25.5,25.6,54.1,60.8,124.4,126.9,127.7 (2 signals), 127.9,128.2,128.3,128.4,131.4,131.9,189.3 (6 signal hidings).
MS (electron spray(ES)): 372[M+H] +
(S)-(+)-preparation of α-ethyl DHA EE (PRB-20)
Intermediate PRB-18's is synthetic:
Figure S2006800240649D00371
Remain under 0 ℃ and the inert atmosphere, DHA (3.00g, 18.3mmol) is dissolved in anhydrous CH 2Cl 2(120mL), add DMAP (2.45g, 20.1mmol) and DCC (3.96g, 19.2mmol).Mixture 0 ℃ of lower stirring 20 minutes, is added (4R, 5S)-(+)-4-methyl-5-phenyl-2-
Figure 2006800240649_8
Oxazolidone (3.24g, 18.3mmol) also stirred 20 hours at ambient temperature.Filter and use flash chromatography method purifying (heptane: EtOAc 6: 1), obtain 3.00g (34%) intermediate PRB-18, be colorless oil.
1H-NMR(200MHz,CDCl 3):δ0.93-1.05(t+d,6H),2.11(m,2H),2.51(m,2H),2.80-3.00(m,10H),3.05(m,2H),4.77(m,1H),5.34-5.68(m,12H),5.70(d,1H),7.28.7.32(m,2H),7.37-7.47(m,3H)。
Intermediate PRB-19's is synthetic:
Figure S2006800240649D00381
Remain under-78 ℃ and the inert atmosphere, with anhydrous THF (10mL) solution of PRB-18 (1.80g, 3.70mmol) be added drop-wise to LiHMDS (the THF solution of 1M, 4.00mL, 4, in anhydrous THF (15mL) solution 00mmol).Mixture-78 ℃ of lower stirrings 30 minutes, is added EtI (0.89mL, 11.1mmol) and slowly be warming up to 0 ℃ in 1 hour.Then this mixture was stirred 18 hours under 0 ℃, and be distributed in saturated NH 4In Cl (50mL) solution and the ether (50mL).Water layer extracts with ether (50mL), and the organic layer that merges is washed with 0.1M HCl (50mL) and salt solution (50mL).Dry (Na 2SO 4) and use flash chromatography method purifying (heptane: EtOAc 95: 5), obtain 0.52g (27%) intermediate PRB-19, be colorless oil.
1H-NMR(200MHz,CDCl 3):δ0.88-1.01(m,9H),1.64-1.78(m,2H),2.08(m,2H),2.31(m,1H),2.48(m,1H),2.87(m,10H),3.87(m,1H),4.75(m,1H),5.32(m,12H),5.63(d,J?7.1Hz,1H),7.32(m,2H),7.42(m,3H)。
13C-NMR(50MHz,CDCl 3):δ7.26,11.75,14.67,14.98,20.95,25.57,25.93,26.04,29.93,44.59,55.31,79.10,125.21,126.01,127.17,127.42,128.27,128.50,128.55,128.67,128.95,129.09,130.35,132.42,133.80,153.18,176.25。
MS (electron spray(ES)): 538.2[M+Na].
Under 0 ℃ and inert atmosphere, PRB-19 (0.25g, 0.485mmol) is dissolved among the anhydrous EtOH (5mL).Add NaOEt (the EtOH solution of 1M, 0.54mL, 0.54mmol) and also this mixture was stirred 30 minutes under 0 ℃, then be distributed in water and the heptane.Water layer extracts with heptane, and the organic layer of merging is also dry with 0.1M HCl washing.Use flash chromatography method purifying, obtain 0.025g (13%) title compound PRB-20, be colorless oil.
1H-NMR(200MHz,CDCl 3)δ0.8-1.0(m,6H),1.2-1.4(m,4H),1.5-1.7(m,2H),2.12(m,2H),2.3-2.5(m,2H),2.8-3.0(m,10H),4.18(t,2H),5.3-5.6(m,12H)。
MS (electron spray(ES)): 407[M+Na].
D+ 1.7 ° (c=1.5, ethanol).
(R)-(-)-preparation of α-ethyl DHA EE (PRB-23)
Intermediate PRB-21's is synthetic:
Figure S2006800240649D00391
Remain under 0 ℃ and the inert atmosphere, DHA (1.00g, 3.05mmol) is dissolved in anhydrous CH 2Cl 2(20mL), add DMAP (0.41g, 3.35mmol) and DCC (0.66g, 3.20mmol).Mixture 0 ℃ of lower stirring 20 minutes, is added (4S, 5R)-(-)-4-methyl-5-phenyl-2-
Figure 2006800240649_9
Oxazolidone (0.54g, 3.05mmol) also stirred 20 hours at ambient temperature.Filter and use flash chromatography method purifying (heptane: EtOAc 6: 1), obtain 1.08g (73%) intermediate PRB-21, be colorless oil.
1H-NMR(200MHz,CDCl 3):δ0.93-1.05(t+d,6H),2.11(m,2H),2.51(m,2H),2.80-3.00(m,10H),3.05(m,2H),4.77(m,1H),5.34-5.68(m,12H),5.70(d,1H),7.28.7.32(m,2H),7.37-7.47(m,3H)。
Intermediate PRB-22's is synthetic:
Figure S2006800240649D00401
Under-78 ℃ and inert atmosphere, with PRB-21 (3.25g, 6.67mmol) anhydrous THF (15mL) solution be added drop-wise to LiHMDS (the THF solution of 1M, 7.34mL, 7, in anhydrous THF (35mL) solution 34mmol), mixture is-78 ℃ of lower stirrings 30 minutes, add EtI (1.6mL, 20.0mmol) and in 1 hour, slowly be warming up to 0 ℃.Then this mixture was stirred 18 hours under 0 ℃, and be distributed in NH 4In Cl (50mL) and the ether (50mL).Water layer washs with 0.1M HCl (50mL) and salt solution (50mL) with ether (50mL) extraction, the organic layer of merging, dry (Na 2SO 4), and use flash chromatography method purifying (heptane: EtOAc 95: 5), and obtain 1.50g (44%) intermediate PRB-22, be colorless oil.
1H-NMR(200MHz,CDCl 3):δ0.88-1.01(m,9H),1.64-1.78(m,2H),2.08(m,2H),2.31(m,1H),2.48(m,1H),2.87(m,10H),3.87(m,1H),4.75(m,1H),5.32(m,12H),5.63(d,J?7.1Hz,1H),7.32(m,2H),7.42(m,3H)。
13C-NMR(50MHz,CDCl 3):δ7.26,11.75,14.67,14.98,20.95,25.57,25.93,26.04,29.93,44.59,55.31,79.10,125.21,126.01,127.17,127.42,128.27,128.50,128.55,128.67,128.95,129.09,130.35,132.42,133.80,153.18,176.25。
MS (electron spray(ES)): 538.2[M+Na].
Figure S2006800240649D00402
Inert atmosphere and 0 ℃ PRB-22 (0.25g, 0.485mmol) is dissolved among the anhydrous EtOH (5mL).Add NaOEt (the EtOH solution of 1M, 0.54mL, 0.54mmol), mixture 0 ℃ of lower stirring 30 minutes, then is distributed in water and the heptane.Water layer extracts with heptane, and the organic layer of merging is also dry with 0.1M HCl washing.Use flash chromatography method purifying, obtain 0.025g (13%) title compound PRB-23, be colorless oil.
1H-NMR(200MHz,CDCl 3)δ0.8-1.0(m,6H),1.2-1.4(m,4H),1.5-1.7(m,2H),2.12(m,2H),2.3-2.5(m,2H),2.8-3.0(m,10H),4.18(t,2H),5.3-5.6(m,?12H);
MS (electron spray(ES)): 407[M+Na].
D-1.3 ° (c=1.00, ethanol).
The preparation of α-phthalimide-based (phtalimide)-DHA-EE (PRB-24)
Figure S2006800240649D00411
At N 2Under the atmosphere, with Alpha-hydroxy-DHA-EE (PRB-12) (373.5mg, 1.00mmol), phthalic imidine (178mg, 1.21mmol) and triphenylphosphine (313.9mg, 1.20mmol) THF (10mL) solution of mixture is cooled to 0 ℃, then drips azo-2-carboxylic acid's diisopropyl ester (0.24mL, 1.24mmol).Remove ice bath, and reaction mixture was stirred 18 hours at ambient temperature, then vaporising under vacuum, and use flash chromatography on silica gel method purifying, use heptane/EtOAc (99: 1-95: 1) wash-out, obtain 323mg (64%) product, be yellow liquid.
1H?NMR(200MHz,CDCl 3)δ0.95(t,J=7.5Hz,3H),1.22(t,J=7.1Hz,3H),2.05(m,2H),2.72-2.84(m,11H),3.02-3.22(1H),4.20(q,J=7.1Hz,2H),4.87(dd,J=11Hz,J=4.9Hz,1H),5.17-5.40(m,12H),7.68-7.75(m,2H),7.79-7.85(m,2H)。
13C NMR (50MHz, CDCl 3) δ 14.0,14.1,20.4,25.4,25.4,25.5,27.0,51.8,61.7,123.8,124.3,126.9,127.5,127.7,127.9,127.9,128.1,128.1,128.3,128.4,131.6,131.8,131.8,134.0,167.3,168.7 (2 signal hidings).
MS (electron spray(ES)): 502[M+H] +, 524[M+Na] +
The preparation of α-ethylamino-DHA-EE (PRB-25) and α-diethylamino-DHA-EE (PRB-26)
Figure S2006800240649D00412
To alpha-amino group-DHA-EE (PRB-17) (746.5mg, 2.01mmol), LiOHH 2O (171.6mg, 4.09mmol) and 4
Figure 2006800240649_10
DMF (4mL) solution of molecular sieve (599mg) mixture adds monobromethane (3.0ml, 40.2mmol), and the mixture that obtains was stirred 71 hours at ambient temperature.Mixture dilutes with ether (100mL) and filters.Organic phase is washed with 1M NaOH (20mL) and salt solution (20mL), dry (Na 2SO 4), filter and vaporising under vacuum, use flash chromatography on silica gel method purifying, use heptane: EtOAc (95: 5)-CH 2Cl 2: 2M NH 3MeOH solution (99: 1) wash-out, obtain the PRB-26 of 458mg (53%), be yellow liquid, and the PRB-25 of 152mg (19%), be yellow liquid.
PRB-26:
1H?NMR(200MHz,CDCl 3)δ0.89(t,J=7.5Hz,3H),1.03(t,3H),1.24(t,J=7.1Hz,6H),2.05(quint,J=7.1Hz,2H),2.52(m,4H),2.76-2.85(m,12H),3.35(t,1H),4.13(q,J=7.1Hz,2H),5.28-5.44(m,12H)。
13C NMR (75MHz, CDCl 3) δ 14.1,14.3,14.4,20.5,22.6,25.5,25.6,25,7,31.9,44.4,60.1,62,9,127.0,127.8,128.05,128.13,128.17,128.22,128.5,132.0,173.3 (5 signal hidings).
Embodiment
The model that uses among the present invention and method have been shown among Fig. 2 have been used for proving synoptic diagram on the impact of metabolic syndrome and diabetes B.Carried out 5 groups and tested to illustrate that the DHA derivative is on the reduction of insulin resistant and/or the impact that alleviates metabolic syndrome.Embodiment shown in the present invention is not limited to and embodiment.
Embodiment 1: the endocellular liberation lipid acid in the liver cell (non--esterified fatty acid) is analyzed (among Fig. 2 the 1st group)
Background
In first group of experiment (Fig. 2), analyze the free unesterified fatty acid that comes from the hepatic tissue that gives PRB-1,2,5 and 7 animals.These animals are by shifting in the 5th group of experiment (the pharmacokinetics impact of DHA derivative in the animal model of metabolic syndrome).Given these animal DHA (diet of 15% lipid content) or DHA-derivative (in their diet, 1.5% lipid content) 8 weeks, and supposition is in DHA with cell inner stablity level and the stable state of DHA-derivative.Because the speed of metabolism is very fast in liver, therefore choose hepatic tissue.
Method
The liver sample is homogenized in cold PBS damping fluid, immediately with Yoshinox BHT (the butylated hydroxy tolune that contains 0.2mM, BHT) chloroform: methyl alcohol (2: 1) solution extraction, use cis-10-heptadecenoic acid as interior mark.Analyze in order to carry out RP-HPLC MS/MS, organic phase is dry under nitrogen, use the acetonitrile that contains 0.1% acetic acid and 10 μ M BHT again to dissolve.Total protein content uses the Bio-Rad method to measure after homogenizing.
Reversed-phase column uses Agilent (the Supelco Ascentis C of 1100 systems 18Post, 25cm * 4.6mm, internal diameter 5 μ m) separation DHA and its PRB derivative in 22 minutes.Moving phase is the acetonitrile-H that contains 0.1% acetic acid of constant gradient 2O (87+13, v/v).The post furnace temperature is set in 35 ℃.The post eluate determines in the negative electrode electron spray ionisation and quantizes that it uses the many reaction detection pattern in three utmost point tandem quadrupole mass spectrometers (ABI Qtrap-4000).Mother and sons' ion pair is respectively 327.3/327.3 (DHA), 341.3/341.3 (PRB-1), 355.3/355.3 (PRB-2 and PRB-5), 387.3/387.3 (PRB-7), 267.2/267.2 (I.S.FA 17:1) under the unit resolution rate.The signal collection residence time all is 100msec, except the 200msec that is set as of FA 17:1.The accurate conclusive evidence of isomer PRB compound is undertaken by the combination of retention time and feature matter/lotus ratio.Behind Internal standard correction methods, quantize with the quadratic regression typical curve.
The result
Total amount with albumen in the μ g/g liver cell has provided the concentration of different DHA-derivatives and the concentration of DHA.Fig. 3 has described the concentration of the different PRBs that come from animal, gives these animal PRB-1,2,5 and 7 with 1.5% total lipid content in high fat diet.
For PRB-2, obtained the IC of the highest PRBs.Although do not reach the degree of PRB-2, PRB-5 also is enrichment in the cell.This discovery is unexpected.
Fig. 4 has described the IC that comes from the DHA in the animal liver tissue that gives different PRBs.Than other 3 DHA-derivatives, give that DHA has reached obvious higher level in the animal of PRB-7.Give to have minimum DHA concentration in the animal of PRB-2.As if PRB-7 transform to a certain extent and got back to DHA.
PRB-2 has reached the highest IC.This means that PRB-2 is more available as the part of nuclear receptor, is a kind of pattern that can be transformed into therapeutic action in processing blood sugar and blood fat.
Embodiment 2: area of computer aided affinity test (among Fig. 2 the 2nd group)
Background
Nuclear receptor sorts, and for PPARs and other associated receptor of using in the Gene Handling of glucose and fat, its aminoacid sequence is known.Therefore X-ray crystal structure and the NMR wave spectrum of PPAR are available, can test to assess binding kinetics with the computerized affinity that lipid acid is bonded to acceptor.Binding geometry is commonly referred to binding pattern or posture, has both comprised that position with respect to the part of acceptor also comprises the conformational state of part and acceptor, therefore can analyze effective part docking.
Part is defined by two different parameters the affinity of acceptor: part (DHA derivative) is docked to the combining site of acceptor and specific amino acids and the carbonyl in the lipid acid front portion or the electrostatic bonding between the side chain (Krumrine) of acceptor.
Known such as the front, PPAR α acceptor mixes more than PPAR γ, means that PPAR α can accept more lipid acid as part than PPAR γ.Yet, because the patient with metabolic syndrome or diabetes B is generally fat or overweight and has ill blood fat, be mainly high triglyceride level and low height-density cholesterol (HDL-chol), so the activation of PPAR α acceptor is very important.The ideal medicament for the treatment of metabolic syndrome or diabetes B should play part to these acceptors, preferably PPAR γ acceptor is had the highest affinity.
Method
Calculate the grade (Ranking) of different DHA-derivatives according to their binding affinity, and provide with minimum binding affinity (LBA) and average binding affinity (ABE).
Use computer docking calculation (computerized docking method) to test altogether 15 DHA derivatives (PRB-1 is to PRB-15).Some derivatives such as PRB-1, PRB-2, PRB-7, PRB-9, PRB-10, PRB-11, PRB-12, PRB-13, PRB-14 and PRB-15 represent with r and s enantiomer, in the case to both testing.PPAR γ part rosiglitazone and pioglitazone all are r and s configuration, in order more all to test.These compounds are the medicine of the treatment diabetes of registration.
The result
The result is presented in the table 1, the minimum combination energy (LBE) of the single conformation (confirmation) of expression parameter testing compound, pose (posed) conformation of the average binding energy (ABE) of the pose of correction (posed) conformation and correction is to the mark (fbound) of 20 the lowest energy conformation of ICM-preservation.With the affinity of same setting mensuration to RXR α.This RXR α acceptor and PPAR acceptor interaction have formed heterodimer by conjugated fatty acid.
Fig. 5 has described the binding affinity of PPAR γ acceptor, this receptor mainly participate in blood sugar process in the transcribing of albumen of institute's combination.The r of PRB-2 and s stereoisomer form all have good affinity to PPAR γ acceptor significantly.The PRB-5 value is somewhat on the low side, and PRB-8 has the highest ABE value.These discoveries are very unexpected and can change more effectively the transcribing of gene of the activation of PPAR γ separately into that wherein this gene determines the processing to blood sugar.
Fig. 6 has described the binding affinity to nuclear receptors PPAR's α, and this nuclear receptors PPAR's α is the major cause of metabolism of fat, blood fat, fatty tissue physiology and weight control.Several DHA-derivatives have high binding affinity, but PRB8's is the highest.This equally also is very unexpected.
Fig. 7 has described the binding affinity to nuclear receptor RXR α.The physiology result who is bonded to RXR α acceptor does not firmly set up.Existing known RXR is bonded to and therefore forms heterodimer on the PPAR acceptor, and it has begun to limit transcribing of gene subsequently.
Table 1
Figure 2006800240649A00800011
ND=is without docking, and c=is two keys of alltrans form.The r=R enantiomer, the s=S enantiomer.The ROSI=rosiglitazone, the PIO=pioglitazone
With parent compound DHA relatively, several PRBs even PPAR α and PPAR γ acceptor had higher LBE and ABE value also have higher value for PPAR γ acceptor rosiglitazone and pioglitazone (being r and s configuration).This interesting phenomenon shows that these several PRBs can be used as the rival likely of established resisting-Rezulin rosiglitazone pioglitazone.
The ethyl derivative of the α position of identical lipid acid for r and s configuration, does not improve affinity, particularly to PPAR γ acceptor.As previously mentioned, PPAR α acceptor be mix therefore can be in conjunction with wide serial lipid acid.
In a word, the DHA-derivative of many tests has demonstrated PPAR α and the interesting affinity of PPAR γ acceptor, and its binding affinity is better than rosiglitazone and pioglitazone.
Embodiment 3: affinity in transfectional cell test (among Fig. 2 the 3rd group)
Background
The release of luciferase is relevant with transcribing of gene.Part to the zygotic induction of nuclear receptor such as PPAR γ transcribing of gene separately, thereby discharge luciferase.Therefore this technology provides the part affinity of a kind of measurement to acceptor and the method for the activation of responsive genes.
Method
As described by Graham and van der Eb (Graham) the COS-1 cell carried out temporary transient transfection in the 6-orifice plate.For total length PPAR transfection research, the pSV-beta-galactosidase enzymes of reporter gene construction (construct), 2.5 μ g of 5 μ g is accepted as the pSG5-PPAR γ 2 of inherence contrast, 0.4 μ g in each hole.Collecting cell after 72 hours is measured uciferase activity according to scheme (Promega).Uciferase activity is according to betagalactosidase activity normalization method.Use the pTK Renilla luciferase of the pSG5-PPAR γ of fat transfection amine (Life Technologies Inc.), 0.2 μ g of fat transfection amine Plus reagent, the 4 μ l of 16 μ l and 100ng as the contrast of transfection efficiency (afficiency), with adipocyte in the upper transfection of the D11 of differentiation (differentiation).After the transfection 3 hours, cell cultivated in containing the substratum of serum and incubation 48 hours in the same substratum that contains suitable reagent.(Dual-Luciferase is tested, Promega) to measure uciferase activity according to the method for supplier's recommendation.All transfections are carried out 3 times.
Lipid acid (BRL or DHA) and PRBs (stock solution) in DMSO solubilising to the ultimate density of 0.1M.Then, in DMSO fatty solubilising to 10mM and be stored in 1.5ml with in the test tube (homopolymer, plastic test tube) of nitrogen wash ,-20 ℃ of lower storages.After the transfection 5 hours, the PRBs of 10 μ M or lipid acid and DMSO (contrast) are added to substratum.Before by the cracking of reporter gene lysis buffer, the cell of transfection was kept 24 hours.The combination of the LBD of PRBs or lipid acid and PPAR has activated GAL4 and has been combined with UAS, and it also stimulates the tk promotor to order about luciferase expression.Use luminometer (TD-20/20 luminometer; Turner Designs, Sunnycvale, CA) measure uciferase activity, and according to the protein content normalization.
The result
Fig. 8 has described from the release of the luciferase of the transfectional cell of processing with different PRBs.The result shows that PRB-1,2,6,7 and 14 has higher luciferase releasing value than PRB-3,5,9,10,11,12 and 16.
Embodiment 4: affinity in having fatty susceptible (prone) animal of metabolic syndrome test (among Fig. 2 the 4th)
Background
Come from the luciferase that discharges in the adipocyte in these animals with the animal model of the metabolic syndrome of the mouse of the fatty susceptible of C57BL/6J system by measurement and test PRB-2,5 and 8 affinity, with 97%DHA and antidiabetic compound rosiglitazone the affinity of PPAR γ is compared.Give these animals (n=8 in every group) high fat diet (fat has consisted of 60% total calorie, with using identical diet in the 5th group of experiment) 8 weeks.After this give other 2 week of these animal PRBs, its dosage is the lipid content of 1.5% diet.The rosiglitazone group is fed with the amount of 100mg/KG diet.Control group continues to give only high fat diet or full diet (standard chow).Fig. 9 shows research and design.
Method
Behind the sacrifice of animal, fatty tissue (epididymis and subcutaneous) is cleared out from other tissue, and be cut into the piece of mm in size.With fatty tissue with 0.9% NaCl drip washing, and in the water-bath of shaking under 37 ℃, contained in the Krebs-Ringer solution of collagenase of the glucose of adenosine, 2nM of Hepes, FAF bovine serum albumin, 200nM and 260U/mL digestion 1.5 hours at 5mL.Behind collagenase digesting, adipocyte is separated from fragment of tissue by filtering.Then with the Krebs-Ringer solution washing of cell with the glucose that contains the adenosine of Hepes, FAF bovine serum albumin, 200nM and 2nM, and under 37 ℃, remain in the water-bath of shaking maximum 30 minutes until electroporation.
Nascent (primary) adipocyte that separates is measured specific PPAR γ response factors (PPRE) activity by electroporation transfection.In the case, under the PPRE control of acyl-CoA oxydase (acyl-CoA-oxidase) gene, we sneak into plasmid-encoded Photinus pyralis LUC cDNA.With the cell plasmid co-transfection that contains Renilla luciferase cDNA, it is by the active promotor control of composition.The Photinus pyralis LUC activity that PPRE induces is come normalization method according to the Renilla luciferase, therefore proofreaies and correct difference possible on the cell quantity of transfection.We use Dual-Luciferase
Figure 2006800240649_11
Reporter gene detection system (Promega, USA) is measured the luciferase signal.
In order to copy, to gather enough epididymises (epidydimal) fatty tissue and come the separating out fat cell.Each group is put to death in 2 days of separating, and concerning each diet group, has obtained 4 independently transfections.
The result
Than giving full diet (chow diet) control group mice (4.5%w/w), (33.7% fat, Mouse Weight w/w) increases gradually to give the HF diet during front 8 weeks.Rear 2 weeks giving the experimental diet high fat diet and giving high fat diet and continue to increase in conjunction with the animal weight of rosiglitazone, almost identical with the speed of front.In the PRB-8 that is rich in the HF diet and PRB-5 situation, weight increases and reduces.Yet, under PRB-2 and DHA (5%w/w) diet situation, stopped the weight increase fully even also caused weight reducing (Figure 10).Recorded at random (4x) food intake amount.There is not difference at HF with between interfering.
In higher fatty acid and situation in conjunction with rosiglitazone, the intrinsic activity of PPAR γ is than other all diet group height nearly 2 times (Figure 11).In addition, these adipocytes become more responsive to the stimulation (5,12-doubly stimulates) of extra external use 5uM rosiglitazone than HF diet self (1,5-doubly stimulates).This type of rosiglitazone-susceptibility impact has also been observed in PRB-2 and PRB-5 diet group (2,6-doubly stimulates).
From then on the data that obtain of research have clearly illustrated that the activity of nuclear PPAR acceptor, and the group that particularly gives PRB-2 in the impact on weight is the most significant.Even giving the animal of PRB-5 and PRB-8 does not increase in weight as giving high-fat diet group.What make us interest is that to give the animal of rosiglitazone identical with the animal animal that only gives high fat diet in the degree that weight increases.This has clearly illustrated and has only given PPAR γ part such as glitazone, namely can reduce insulin resistant, but the danger that has weight to increase.Yet when consider the PPAR gamma activity of testing such as glitazone the uciferase activity test in this experiment, rosiglitazone is all higher than the value of any PRBs.In PRB group, PRB-2 and PRB-5 are only than the value high (Figure 12) of PRB-8 and DHA.
Embodiment 5: the pharmacokinetics effect of the DHA derivative in the animal model of metabolic syndrome (among Fig. 2 the 5th group)
Background
Prove the common typical laboratory of metabolic syndrome and the impact of pathological anatomy feature with the animal model of the metabolic syndrome of the mouse of the fatty susceptible of C57BL/6J system.When nursing contained the high fat diet of 60% fat, it is fat that this animal becomes, and develops into hyperinsulinism serum level, the test of pathology glucose tolerance, high serum triglyceride and non--esterified fatty acid and fatty liver.
Embodiment 5a: diet intervention is the effect of DHA derivative in the mouse of fatty susceptible during 4 months
Method
All experiments are male C57BL/6 mouse, perhaps subbreed C57BL/N (supplier: CharlesRiver, Germany, n=160, experiment A-C, referring to following), or subbreed C57BL/6J (supplier: theJackson laboratory, Bar Harbor, ME, USA, n=32, experiment D) on carry out.In order to select total quantity of the animal of use large (n difference=170 and 36).In the situation of back, institute of physiology breeding several generations animal (<20).In when beginning treatment, animal is that 1 4-age in week and their weight range are at 23.6-27.1g.Week before research beginning, with animal according to their weight sorting and be assigned in the group of identical mean body weight (n=8).This method allows to select the animal that shows respectively minimum and maximum body weight of about 5-10%.The animal that gets rid of from this research in this stage is shifted its execution by neck.By supplier Charles River these mouse are carried out the thoroughness physical examination, before the research beginning, carry out Serological testing with ANLAB (Prague, Czech Republic).In addition, in Animal House, carry out conventional physical examination, use sentry box mouse and Serological testing (ANLAB) with 3 months intervals.In all tests, animal is away from specific pathogenic agent.
Diet
Animal is fed 3 kinds of experimental diet:
(i) full diet (Chow diet) (ssniff R/M-H from S SNIFF Spezialdieten Gmbh, Soest, Germany; See also Http:// ssniff.de), have albumen, fat and carbohydrate, form respectively 33,9 and 58% energy
(ii) height-fat diet that in the laboratory, prepares (high fat diet) (cHF diet), it has albumen, fat and carbohydrate, form respectively 15,59 and 26% energy, and the fatty acid composition of superperformance (has the most of lipid that derives from Semen Maydis oil; See Ruzickova 2004)
(iii) cHF diet, wherein 0.15,0.5 and 1.5% fat (specific Semen Maydis oil component) is by various PRB-compounds, and namely PRB1, PRB2, PRB5, PRB7 and PRB8 substitute, or are substituted by DHA.These all compounds are the ethyl ester form, are provided in sealed vessel by Pronova Biocare a.s..The chemical constitution of PRB-compound is unknown to the laboratory (Institute of Physiology, Academy of Sciences Prague, Czech Republic) of testing.
Behind the arrival, this PRB-compound is housed in the refrigerator with original container.Only before the preparation experiment diet, just open this container.Diet is kept in the plastics bag that is full of nitrogen, and-70 ℃ of lower storages, with the amount in one week of enough nutrition purposes, especially for feeding animals of aliquot.With 2 days intervals or give fresh ratio every day.
Summary
Research is based on 4 independent experiments.In each experiment, (or DHA, respectively), it is mixed to the cHF diet with 3 different concentration (lipid content 0.15,0.5 and 1.5%) to have tested different PRB-compounds.In each experiment, comprise the mouse of the usual cHF diet of a small group-nursing in contrast.Divide 4 groups to raise in cages and feed full diet until 3 monthly ages mouse, given animal (n=8-13) is with different test diet randomly.After giving 2 months of this new diet (5 monthly ages), animal is by overnight fasting and carry out in the morning intraperitoneal glucose tolerance test (GTT).Give experimental diet after 4 months, putting to death animal in the time of 7 monthly ages, carrying out end-point analysis.
The research parameter
Parameter in the research is: body weight increases (gram), tests (mMol * area under curve (AUC) 180min), plasma insulin (ng/ml), serum triglyceride (TAGs from the intraperitoneal glucose tolerance, mmol/l) and non--esterified fatty acid (NEFA, mmol/l).
Figure 13 is illustrated in before the compound that the animal with insulin resistant has the effect that reduces insulin resistant and afterwards, typical blood sugar is eliminated curve.The minimizing of area means because the minimizing blood sugar of insulin resistant has been eliminated more effectively below curve.
The result
The result is presented in the lower tabulation 2,3 and 4.( *=than HF diet marked difference (P<0.05))
Table 2 has demonstrated than giving full diet (STD), combination high fat diet (cHF) or 97%DHA, gives the impact in the PRB test compounds animal of 1.5% concentration.Than the animal that gives high fat diet (cHF), body weight is increased in the animal that gives PRB-2 and reduces significantly.The food decrease to some degree of in this group, taking in.In identical group, even in giving the animal of PRB-1, observed the most significant reduction of AUC in the glucose tolerance test.Than the cHF control group, plasma insulin reduces significantly in the PRB-2 group, even the animal of PRB-1 and PRB-5 treatment has also demonstrated some impacts on this parameter.The PRB-2 group shows the maximum reduction amount of triglyceride level (TAGs) and non--esterified fatty acid (NEFA).
Table 3 shows than giving full diet (STD), combination high fat diet (cHF) or 97%DHA, gives the impact of the PRB test compounds of animal low concentration (0.5%).Body weight increases decrease to some degree in the animal that gives PRB-2 and PRB-5.Yet the AUC of glucose tolerance test and plasma insulin only reduce in the PRB-2 group significantly.
Table 4 shows the result who gives minimum PRB concentration (0.15%).Herein, difference is very little.The weight increase has to a certain degree reduction in PRB-1 and PRB-2 group, and AUC only reduces in the PRB-2 group significantly.Plasma insulin reduces in PRB-1,2 and 7.
Figure 2006800240649A00800021
[0468]?
Figure 2006800240649A00800031
[0470]?
Figure 2006800240649A00800041
[0472]In a word, in 4 months, PBR-1,2,5 and 7 shows clear and unquestionable test PRB impact in having the fatty susceptible animal of insulin resistant and metabolic syndrome, DHA-derivative PBR-2 particularly, the reduction of the AUC in the symptom of insulin resistant and metabolic syndrome such as weight reducing, the test of intraperitoneal glucose tolerance, lower Regular Insulin/blood plasma level and triglyceride level and the non--esterified fatty acid of reduction.In 1.5% and 0.5% dosage group, observe this impact.Make some difference even also be obvious in the group of lowest dose level (0.15%).
It is slower that the test of PRB-8 compound begins, and therefore only provided the data that three groups of dosage (1.5%, 0.5% and 0.15%) were interfered in 2 months.Give with 1.5% group in, than the 29.6+0.9 of control group, body weight (BW) is 28.0 ± 0.7 grams, AUC is 1031 ± 104, than 1074 ± 91.These differences are little but its trend is interesting.For than low dosage 0.5% and 0.15%, interfering with control group does not have difference.The trend that shows its weight reducing and AUC from the PRB-8 related data of pharmacological agent in 2 months.
Embodiment 5b:DHA derivative is on the metabolic syndrome proved conclusively and the impact of insulin resistant
Method
In other experiment, in the animal of same breed, test PRB-2, PRB-5 and PRB-7.In this experiment, originally give animal high fat diet (identical with the experiment 5a of front) 8 weeks, to form insulin resistant and metabolic syndrome, then give PRBs.The dosage of beginning is for to replace with PRBs with 15% of lipid content, but these animals are held and can't stand this dosage.Through after the time in other 2 weeks, give PRB-2,5% and 1.5% the PRB-5 of animal 1.5% and 1.5% and 0.5% PRB-7.
The result
It is very good to give in the animal of PRB-2 weight reducing.Even give also to have shown some weight reducings in the animal of PRB-5, except in 5% higher dosage.Than the control animal that gives compound high fat diet, use that triglyceride level reduces in the animal of all test derivatives.Use PRB-2 and PRB-5 non--minimizing of esterified fatty acid is the most obvious, yet be with different dosage (referring to Figure 14).
Blood cholesterol reduces in the animal that gives PRB-2 and PRB-5.Because these animals are in the early stage-diabetes stage, because the generation of hyperinsulinism has normal glucose, so its blood sugar and unaffected.Yet important is, plasma insulin has reduced significantly in the PRB-2 group, much lower with DHA-derivative PRB-5 concentration best in second group.Even PRB-7 has demonstrated some impacts (referring to Figure 15) on insulin concentration.
Than baseline value, PRB-2 has demonstrated the reduction of the significant AUC blood sugar on the statistical significance on all time points of curve.After this means the treatment through 1.5% PRB-2, blood sugar has been removed effectively.PBR-5 has also shown some effects with PBR-7 but has not reached identical degree (referring to Figure 16).
These effects be very beyond thought and with metabolic syndrome and diabetes B in positive impact very relevant.These patient's great majority all are overweight or fat, and medicine is very definite to the effect of weight reducing.The medicine of now the most frequently used treatment diabetes B, thiazolidinediones, it is effective PPAR γ part thereby reduces insulin resistant, usually causes weight to increase, it is the height side effect (Yki-J in this class patient
Figure 2006800240649_12
Rvinen 2004).
The reduction of serum triglyceride is another very important impact, and it shows in experiment.Patient with metabolic syndrome and diabetes B has high triglyceride level usually.The reducing effect of the triglyceride level of DHA-derivative is a positive discovery, and PRB-2 has also demonstrated the effective influence of use lowest dose level.Very definite effect at plasma insulin and glucose-tolerant property testing is very likely and fully beyond thought.Considering the effect of using DHA-derivative, particularly PRB-2 to obtain, is very likely, and it consists of the good basis of the exploitation of antidiabetic medicine.
Embodiment 5c: the test of the DHA derivative on liver fat
Method
To come from and use the tissue samples of the animal of DHA derivative to carry out histologic analysis in the experiment.Behind paraffin section (paraffination), the tissue samples that comes from liver, fatty tissue, skeletal muscle, pancreas and kidney dyes with Yihong-phenodin (eosin-hematoxylin).
The result
Except the tissue from liver, the tissue of detection does not have the discovery of pathology.The control animal that gives high fat diet has formed fatty liver (steatosis).Fat granule in the liver is easy to distinguish very different from normal liver cell.Animal through RB-1,5 and 7 treatments has the fatty liver of low degree.Yet the animal liver cell of the PRB-2 treatment through 1.5% is fully normal, the steatosis that has not a particle of.
This is a very important discovery, and very relevant with treating the patient with insulin resistant, obesity and diabetes B.Fatty degeneration of liver often finds that in these patients it is usually excessive relevant with triglyceride level with lipid acid, the biological marker that exists in its formation for insulin resistant and metabolic syndrome.The DHA-derivative has reduced fatty degeneration of liver, and PRB-2 is showing the compounds effective of this effect.
Discuss and conclusion
The present invention has clearly determined the active nuclei acceptor that a class is new, therefore the compound of PPAR γ and PPAR α particularly provides a series of result for the treatment of in the disease treatment relevant with other atherosclerosis of insulin resistant, metabolic syndrome, diabetes B, cardiovascular disorder.
Member in this class is for having the DHA derivative of different types of side chain in the α position of molecule.Tested the DHA derivative of a large amount of alpha-substitution and compared in contrast with pure DHA and EPA.Proved several in the test compounds have potential anti--interesting biology effect that diabetes medicament is relevant.
Interesting and can not expect in advance, in test group, α-ethyl DHA-EE (PRB-2) is noticeable more effective, therefore this test is used for illustrating the effect relevant with insulin resistant, and the disease that causes of main thus pathologic, physiologic symptom such as metabolic syndrome, diabetes B, disease that cardiovascular disorder is relevant with other atherosclerosis.α-ethyl DHA-EE is enriched in the hepatic tissue of animal, and this animal is given different DHA test derivatives (1 district), has shown that this compound is not used to synthetic glycerine three esters, eicosanoid (eikosanoids) or other Metabolic Intermediate.This means that indirectly α-ethyl DHA can be used for being attached to nuclear receptor such as PPARs.
In using the affinity of computer docking technique test to PPAR γ and PPAR α, a large amount of DHA-derivatives has demonstrated two acceptors especially affinity of PPAR γ, and PPAR γ may be the most important nuclear receptor that participates in the gene activation of metabolism of blood glucose.Particularly α-ethyl DHA (PRB-2) and α-diethyl DHA (PBR-8) has outstanding affinity to these nuclear receptors.Than α-diethyl DHA, α-ethyl DHA has two kinds of steric isomers, r and s configuration.Use two kinds of steric isomers of docking technique to have approximately identical affinity for PPAR γ and PPAR α, this means that r or s configuration all do not have benefit than racemic modification.In fact this racemic modification may be more profitable than every kind of steric isomer.
When testing affinity in the transfectional cell that is carrying nuclear receptor and DNA response factor subsequently, several PRBs have demonstrated good affinity, and this affinity is measured with the release of luciferase.α-ethyl DHA (PRB-2) and PRB-6,7 and 14 have demonstrated best effect.
5 DHA derivatives the C57BL/6 mouse model in test on a large scale, develop into insulin resistant and metabolic syndrome when giving this mouse high fat diet.α-ethyl DHA (PRB-2) is through testing in 3 independent experiments, and PRB-1,5 and 7 tests in 2, and α-diethyl DHA (PRB-8) tests in 1 experiment.All derivatives have shown the far reaching biological action.Yet α-ethyl DHA (PRB-2) has shown the most promising consistent effect to following aspect: AUC reduction, plasma insulin and serum triglyceride and non--esterified fatty acid in the reduction of body weight, the test of intraperitoneal glucose tolerance reduce.Dosage 1.5 and 0.5% obtains result for the treatment of.Minimum proof load 0.15% does not obtain the result of conviction.Also having shown the normalizing of fatty liver with the α of 1.5% dosage-ethyl DHA (PRB-2), is that important pathology are found in the humans and animals with insulin resistant and metabolic syndrome.
As if compare with pure DHA, α-ethyl DHA (PRB-2) has 10-30 effectiveness doubly.These all discoveries and all uncertain and be all beyond one's expectations than the effectiveness of parent molecule DHA.
Because α-ethyl DHA (PRB-2) it seems by simultaneously in conjunction with nuclear receptors PPAR's α and PPAR γ effect, therefore this compound is not only to glucose and lipid metabolism, especially in the patient with insulin resistant, metabolic syndrome and diabetes B, have interesting result for the treatment of, and have weight reducing and general antiphlogistic effects.Directly or by the front interfere risk factor, α-ethyl DHA (PRB-2) has forming cardiovascular disorder such as myocardium infarct and and the prophylactic effect of cerebral apoplexy and the effect of prevention cardiovascular death.
Pharmaceuticals as PPAR γ part have gone on the market, even but these compounds have glucose metabolism are had positive effect, but they by as the triglyceride level that increases, the increase of weight and the side effect of oedema hinder.The DHA derivative of the alpha-substitution that proposes among the application has PPAR γ and the PPAR α effect of combination, its may be correlated with and the patient with insulin resistant, metabolic syndrome and diabetes B had benefit.In addition, these compound actions are to blood fat, inflammatory symptoms, atherosclerosis, and also have important effect in the cardiovascular disorder thus.
Embodiment shown in the present invention is not limited in and embodiment.
Reference:
Simonopoulos?AP.Essential?fatty?acids?in?health?and?chronic?disease.Am?JClin?Nutr?1999;70(Suppl):560S-569S
Geleijnse?JM,Giltay?EJ,Grobbee?DE,et?al.Blood?pressure?response?to?fishoil?supplementation:metaregression?analysis?of?randomized?trials.J?Hypertension2002;20:1493-1499
Storlien?LH,Hulbert?AJ,and?Else?PL.Polyunsaturated?fatty?acids,membrane?function?and?metabolic?syndrome?such?as?diabetes?and?obesity.CurrOpin?Clin?Nutr?Metab?Care?1998;1:559-563
Jump?DB.The?biochemistry?of?n-3?polyunsaturated?fatty?acids.J?Biol?Chem2002;277:8755-8758
Pawar?A?and?Jump?D.Unsaturated?fatty?acid?regulation?of?peroxisomesproliferator-activated?receptor?alfa?activity?in?rat?primary?hepatocytes.J?BiolChem?2003;278:35931-35939
Meigs?JB,Wilson?PWF,Nathan?DM,et?al.Prevalence?and?characteristics?ofthe?metabolic?syndrome?in?the?San?Antonio?Heart?and?Framingham?offspringstudies.Diabetes?2003;52:2160-2167
Storlien?LH,KraegenWE,Chisholm?DJ,et?al.Fish?oil?prevents?indulinresistance?induced?by?high?fat?feeding?in?rats.Science?1987;237:885-888
Field?CJ,Ryan?EA,Thomson?ABR,et?al.Diet?fat?composition?altersmembrane?phospholipid?composition,insulin?binding?and?glucose?metabolism?inadipocytes?from?control?and?diabetic?animals.J?Biol?Chemistry?1990;265:11143-11150
Yki-J
Figure 2006800240649_13
rvinen,H.Thiazolidinediones.NEJM?2004;351:1106-1118
Adams?M,Montague?CT,Prins?JB,et?al.Activators?of?peroxisomesproliferator-activated?receptor?gamma?have?depot-specific?effects?on?humanpreadipocyte?differentiation.J?Clin?Invest?1997;100:3149-3153
Ruzickovaj,Rossmeisl?M,Prazak?T,et?al.Omega-3?PUFA?of?marine?originlimit?diet-induced?obesity?in?mice?by?reducing?cellularity?of?adipose?tissue,Lipids2004;39:1177-1185
Vaagenes?H,Madsen?L,Dyr
Figure 2006800240649_14
y?E,et?al.The?hypolipidaemic?effect?of?EPA?ispotentiated?by?2-and?3-methylation.Biochim?Pharmacol?1999;58:1133-1143
Larsen?L,Granslund?L,Holmeide?AK,et?al.Sulfur-substituted?andα-methylated?fattyacids?as?peroxisome?proliferator-activated?receptor?activators.?Lipids?2005;40:49-57
Larsen?L,H rvik?K,S
Figure 2006800240649_16
rensen?HIN,et?al.Polyunsaturated?thia-andoxa-fatty?acids:incorporation?into?cell-lipids?and?their?effects?on?arachidonicacid-and?eikosanoids?synthesis.Biochim?et?Biophys?Acta?1997;1348:346-354
Larsen,et.al?Biochemical?Pharmacology?1998;55,405
Chih-Hao?L,Olson?P,and?Evans?RM.Lipid?metabolism,metabolic?diseases,and?peroxisome.Endocrinology?2003;144:2201-2207
Willumsen?N,Waagenes?H,Holmsen?H,et?al.On?the?effect?of?2-deuterium-and?2-methyl-eicosapentaenoic?acid?derivatives?on?triglycerides,peroxisomalbeta-oxidation?and?platelet?aggregation?in?rats.Biochim?Biophys?Acta?1998;1369:193-203
Mitsunobu?O,Synthesis?1981;1
Ager?DJ,Prakash?I,and?Schaad?DR.1,2-amino?alcohols?and?theirheterocyclic?derivatives?as?chiralauxiliaries?in?asymmetric?synthesis?Chem?Rev1996;96:835-876

Claims (36)

1. formula (I) compound:
Figure FSB00000926953400011
Wherein
X represents hydroxy-acid group, carboxylate group, carboxylic acid anhydride group or carboxylacyl amine group, or its any pharmacologically acceptable salt, solvate, mixture or prodrug.
2. according to claim 1 compound, its Chinese style (I) compound is tri-glyceride or phosphatide.
3. according to claim 2 compound, its Chinese style (I) compound is formula (II) tri-glyceride:
Figure FSB00000926953400012
4. according to claim 1 compound, it is take X as COO -Z +The form of salt, Z wherein +Be Li +, Na +, K +, NH 4 +Or the NH that replaces 4 +
5. according to claim 1 compound, it is with the form of following formula salt:
Z wherein 2+Be Mg 2+Or Ca 2+
6. according to claim 1 compound, wherein said carboxylate group is selected from: ethoxycarbonyl, methoxycarbonyl, n-propyl ester group, sec.-propyl ester group, normal-butyl ester group, sec-butyl ester group and n-hexyl ester group.
7. according to claim 1 compound, wherein said carboxylacyl amine group is selected from: uncle's carboxamide groups, N-methyl carboxamide groups, N, N-dimethyl carboxamide groups, N-ethyl carboxamide groups and N, N-diethyl carboxamide groups.
8. according to claim 6 compound, wherein said carboxylate group is selected from: methoxycarbonyl or ethoxycarbonyl.
9. according to claim 8 compound, its be following formula (complete-Z)-2-ethyl DHA ethyl ester:
Figure FSB00000926953400021
10. according to claim 1 compound, wherein said compound exists as S enantiomorph isomer.
11. compound according to claim 1, wherein said compound exists as R enantiomorph isomer.
12. compound according to claim 1, wherein said compound exists as the mixture of R and S enantiomorph isomer.
13. compound according to claim 12, wherein said mixture are racemic modification.
14. compound according to claim 1, its Chinese style (I) compound is Diglyceride.
15. composition, it comprises at least one compound according to claim 1.
16. composition according to claim 15 also comprises the compound of following formula:
Figure FSB00000926953400022
17. each compound or its any pharmacologically acceptable salt, solvate, mixture or prodrug are for the preparation of the purposes that treats and/or prevents in the medicine that is selected from following disease or illness among the claim 1-14: peripheral insulin resistance; Diabetes; Obesity or overweight disease.
18. purposes according to claim 17, wherein said diabetes are type ii diabetes.
19. each compound or its any pharmacologically acceptable salt, solvate, mixture or prodrug are for the preparation of the purposes in the medicine that treats and/or prevents blood lipid dysbolism among the claim 1-14.
20. purposes according to claim 19, wherein said blood lipid dysbolism are hyperlipidaemia.
21. purposes according to claim 19, wherein said blood lipid dysbolism comprise that triglyceride level and/or non--HDL cholesterol raise.
22. purposes according to claim 21, wherein said non--the HDL cholesterol is LDL-C and/or VLDL cholesterol.
23. each compound or its any pharmacologically acceptable salt, solvate, mixture or prodrug are for the preparation of the purposes in the medicine that reduces insulin resistant, blood sugar and/or S-TG level among the claim 1-14.
24. each compound or its any pharmacologically acceptable salt, solvate, mixture or prodrug are for the preparation of the purposes that reduces in the medicine that body weight and/or prevention body weight increase among the claim 1-14.
25. each compound or its any pharmacologically acceptable salt, solvate, mixture or prodrug are for the preparation of the purposes in the medicine of activation peroxisome proliferator-activated receptor alpha and/or γ among the claim 1-14.
26. pharmaceutical composition, it comprises the compound of formula (I)
Wherein X represents hydroxy-acid group, carboxylate group, carboxylic acid anhydride group or carboxylacyl amine group,
Or its any pharmacologically acceptable salt, solvate, mixture or prodrug.
27. pharmaceutical composition according to claim 26, it is formulated as oral administration.
28. pharmaceutical composition according to claim 26, it is formulated as capsule or wafer.
29. pharmaceutical composition according to claim 26, it is formulated as vein, subcutaneous or intramuscular administration.
30. pharmaceutical composition according to claim 26, it is formulated as the per daily dose that 10mg to 10g is provided.
31. pharmaceutical composition according to claim 30, it is formulated as the per daily dose that 100mg to 1g is provided.
32. lipid composition, it comprises the compound of formula (I)
Figure FSB00000926953400032
Wherein X represents hydroxy-acid group, carboxylate group, carboxylic acid anhydride group or carboxylacyl amine group,
Or its any pharmacologically acceptable salt, solvate, mixture or prodrug.
33. lipid composition according to claim 32, the compound of its Chinese style (I) exists with the concentration that accounts at least TL composition 60% weight.
34. lipid composition according to claim 33, the compound of its Chinese style (I) exists with the concentration that accounts at least TL composition 90% weight.
35. lipid composition according to claim 32, it also comprises pharmaceutically acceptable antioxidant.
36. lipid composition according to claim 35, wherein said pharmaceutically acceptable antioxidant is tocopherol.
CN200680024064.9A 2005-05-04 2006-05-04 New dha derivatives and their use as medicaments Expired - Fee Related CN101213281B (en)

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US67735005P 2005-05-04 2005-05-04
US67735105P 2005-05-04 2005-05-04
SE0501045 2005-05-04
SE0501044-2 2005-05-04
US60/677,350 2005-05-04
SE05010459 2005-05-04
SE0501044 2005-05-04
US60/677,351 2005-05-04
SE05010442 2005-05-04
SE0501045-9 2005-05-04
PCT/IB2006/001155 WO2006117664A1 (en) 2005-05-04 2006-05-04 New dha derivatives and their use as medicaments

Related Child Applications (2)

Application Number Title Priority Date Filing Date
CN201210494826.9A Division CN103058867B (en) 2005-05-04 2006-05-04 Novel DHA derivatives for and uses thereof as medicament
CN2010105396933A Division CN102050720B (en) 2005-05-04 2006-05-04 Novel DHA derivative and its usage as medicine

Publications (2)

Publication Number Publication Date
CN101213281A CN101213281A (en) 2008-07-02
CN101213281B true CN101213281B (en) 2013-03-13

Family

ID=39612495

Family Applications (1)

Application Number Title Priority Date Filing Date
CN200680024064.9A Expired - Fee Related CN101213281B (en) 2005-05-04 2006-05-04 New dha derivatives and their use as medicaments

Country Status (1)

Country Link
CN (1) CN101213281B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9394228B2 (en) 2010-11-05 2016-07-19 Pronova Biopharma Norge As Methods of treatment using lipid compounds

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2345241B1 (en) * 2009-03-16 2011-09-08 Lipopharma Therapeutics USE OF 2-HYDROXIDERIVATES OF POLYINSATURATED FATTY ACIDS AS MEDICINES.
JP5872453B2 (en) * 2009-04-17 2016-03-01 ナタク ファルマ エセ.エレ. Composition with low phytanic acid content and rich in omega-3 fatty acids
US8735436B2 (en) * 2009-05-08 2014-05-27 Pronova Biopharma Norge As Polyunsaturated fatty acids for the treatment of diseases related to cardiovascular, metabolic and inflammatory disease areas
EP2961384B1 (en) 2013-02-28 2019-08-28 Basf As A composition comprising a lipid compound, a triglyceride, and a surfactant, and methods of using the same
CN107530306A (en) 2015-04-28 2018-01-02 普罗诺瓦生物医药挪威公司 Purposes of the sulfur-bearing aliphatic acid of structure enhancing in preventing and/or treating nonalcoholic fatty liver disease
US11925614B2 (en) 2017-12-06 2024-03-12 Basf As Fatty acid derivatives for treating non-alcoholic steatohepatitis
CN114689704B (en) * 2020-12-26 2023-05-09 四川汇宇制药股份有限公司 Method for detecting 1,3-dihydroxyacetone and related impurities
CN115433144B (en) * 2022-10-09 2023-09-19 燃点(南京)生物医药科技有限公司 Fatty acid ester compound and preparation method and application thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656667A (en) * 1988-08-11 1997-08-12 Norsk Hydro As Fatty acid composition
CN1204638A (en) * 1997-07-02 1999-01-13 湖南天海林实业股份有限公司 Preparation of docosahexenoic acid and esterification technology
EP1310249A1 (en) * 2001-11-12 2003-05-14 Quatex N.V. Use of polyunsatured fatty acids for the primary prevention of major cardiovascular events
US20040162348A1 (en) * 1999-01-27 2004-08-19 Laxdale Limited Highly purified ethyl EPA and other EPA derivatives for psychiatric and neurological disorders
WO2004078166A2 (en) * 2003-03-05 2004-09-16 Solvay Pharmaceuticals Gmbh Use of omega-3-fatty acids in the treatment of diabetic patients
EP1544281A1 (en) * 2002-08-07 2005-06-22 Kao Corporation Fat composition

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656667A (en) * 1988-08-11 1997-08-12 Norsk Hydro As Fatty acid composition
CN1204638A (en) * 1997-07-02 1999-01-13 湖南天海林实业股份有限公司 Preparation of docosahexenoic acid and esterification technology
US20040162348A1 (en) * 1999-01-27 2004-08-19 Laxdale Limited Highly purified ethyl EPA and other EPA derivatives for psychiatric and neurological disorders
EP1310249A1 (en) * 2001-11-12 2003-05-14 Quatex N.V. Use of polyunsatured fatty acids for the primary prevention of major cardiovascular events
EP1544281A1 (en) * 2002-08-07 2005-06-22 Kao Corporation Fat composition
WO2004078166A2 (en) * 2003-03-05 2004-09-16 Solvay Pharmaceuticals Gmbh Use of omega-3-fatty acids in the treatment of diabetic patients

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Larsen L. et al,.Sulfur-Substituted and a-Methylated Fatty Acids as Peroxisome Proliferator-Activated Receptor Activators.Lipids.2005,40(1),49-57. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9394228B2 (en) 2010-11-05 2016-07-19 Pronova Biopharma Norge As Methods of treatment using lipid compounds

Also Published As

Publication number Publication date
CN101213281A (en) 2008-07-02

Similar Documents

Publication Publication Date Title
CN102050720B (en) Novel DHA derivative and its usage as medicine
CN101213281B (en) New dha derivatives and their use as medicaments
CN102159200B (en) Novel sulphur containing lipids for use as food supplement or as medicament
KR101783817B1 (en) Polyunsaturated fatty acids for the treatment of diseases related to cardiovascular, metabolic and inflammatory disease areas
JP4213205B2 (en) 1,3-propanediol derivatives as biologically active compounds
CN101535238A (en) Alpha-substituted omega-3 lipids that are activators or modulators of the peroxisome proliferators-activated receptor (PPAR)
CN101054344B (en) Lipoxin A4 analogs
CA2816949C (en) Methods of treatment using lipid compounds
WO1998018751A1 (en) Presentation of bioactives
KR101544584B1 (en) -3 omega-3 lipid compounds
JP2011502113A (en) Novel DHA derivatives and their use as pharmaceuticals
CN100391930C (en) 1, 3-propane diol derivatives as bioactive compounds
WO2011149766A2 (en) Lipid-tailored pharmaceutical agents
JP5746730B2 (en) New compounds
USRE40546E1 (en) 1,3-Propane diol esters and ethers and methods for their use in drug delivery
WO2003004484A1 (en) Novel aliphatic compounds, synthesis method and method of using the same

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1115409

Country of ref document: HK

C14 Grant of patent or utility model
GR01 Patent grant
REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1115409

Country of ref document: HK

CF01 Termination of patent right due to non-payment of annual fee
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20130313

Termination date: 20200504