MXPA97005771A - Employment of xantina derivatives for the modulation of the apopto - Google Patents

Employment of xantina derivatives for the modulation of the apopto

Info

Publication number
MXPA97005771A
MXPA97005771A MXPA/A/1997/005771A MX9705771A MXPA97005771A MX PA97005771 A MXPA97005771 A MX PA97005771A MX 9705771 A MX9705771 A MX 9705771A MX PA97005771 A MXPA97005771 A MX PA97005771A
Authority
MX
Mexico
Prior art keywords
alkyl
formula
radical
compound
hydroxy
Prior art date
Application number
MXPA/A/1997/005771A
Other languages
Spanish (es)
Other versions
MXPA02005771A (en
Inventor
Mullner Stefan
Dax Claudia
Original Assignee
Hoechst Aktiengesellschaft
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
Priority claimed from US08/461,698 external-priority patent/US5733529A/en
Priority claimed from US08/462,599 external-priority patent/US5665374A/en
Priority claimed from US08/462,930 external-priority patent/US5645841A/en
Priority claimed from US08/462,203 external-priority patent/US5651959A/en
Priority claimed from US08/464,403 external-priority patent/US5711936A/en
Application filed by Hoechst Aktiengesellschaft filed Critical Hoechst Aktiengesellschaft
Priority claimed from PCT/US1996/008714 external-priority patent/WO1996039116A1/en
Publication of MXPA97005771A publication Critical patent/MXPA97005771A/en
Publication of MXPA02005771A publication Critical patent/MXPA02005771A/en

Links

Abstract

The present invention relates to: Compound of the formula I (See Formula) wherein one of the radicals R1 and R3 represents a radical of the formula II - (CH2) nA-CH3 (II), wherein A represents a covalent bond, -C (O) -ó-C (R4) (OH) -, are suitable for the preparation of drugs for the modulation of apoptosis. A combination preparation containing a compound of the formula I and a compound of the formula IV and / or V (See Formula) is suitable for the preparation of medicaments for the modulation of apoptos

Description

Use of xanthine derivatives for the modulation of apoptosis In the case of apoptosis, it is, in contrast to necrosis, a death of the cells that runs in a genetically controlled (programmed) manner, which is an essential component of the life of multicellular organisms.
In contrast to this process of normal and necessary apoptosis for life, numerous forms of diseases or their symptoms are the expression of an abnormal apotosis, ie a) without limits or b) oppressed [a]: infarction, stroke or neurodegeneration, b) hypertrophic diseases]. Accordingly, the healing processes of the diseases may be possible by oppression or activation of apoptosis (for example transverse paralysis, immune response, etc.). The apoptosis proceeds, after the induction of definite deadly signals, for example by stimulation of certain receptors (for example Fas receptor) through a complex cascade induced in a secondary way of entangled biochemical events, at the end of which is the dissolution of the intact cell in packed units in the membrane that can only be removed from the body without or with little damage to the surrounding cells (as opposed to necrosis). With this, in some cases the transitions between necrosis and apoptosis are fluid; thus, there are cases in which necrosis leads to apoptosis (or vice versa) (for example infarction, stroke, etc.). Cofilin, an actin-binding protein of 19 Da size, plays a key role in the immune reaction as a co-stimulatory factor in T cells. Cofilin is present in a phosphorylated form in cytosol and is transported to the nucleus of the cell after dephosphorylation. In this case, it evidently serves as a trailing molecule for the actin protein that does not possess any nuclear recognition sequence and is known as an inhibitor of DNase I. By this mechanism, the degree of phosphorylation of cytosolic cofilin can have a regulatory influence and modulator on cell apoptosis. It has now been found that certain xanthine derivatives are suitable for inhibiting dephosphorylation of cofilin and, thereby, have a modulating influence on apoptosis. Therefore, the invention relates to the use of at least one xanthine derivative of the formula I and / or a possibly stereoisomeric form of the xanthine derivative of the formula I, wherein R2 represents alkyl (Cx-C4), one of the radicals R1 or R3 represents a radical of the formula II - (CH2) n-R-CH3 (II) where R represents a) a single covalent bond, and where n means the integer zero, 1, 2, 3, 4, 5, 6 or 7, b) a radical -CO-, and where n means the number integer 1, 2, 3, 4, 5 or 6, or c) a radical -C (R4) (OH) -, and where n means the integer 1, 2, 3, 4, 5 or 6 and R * represents a) a hydrogen atom or b) alkyl (Cx-C3), and the other radical R3 or R1 represents a) a hydrogen atom, b) alkyl (C ^ C,), c) (C4-C8) cycloalkyl-alkyl or d) alkyl having 2 to 6 carbon atoms, wherein the carbon chain is interrupted by an oxygen atom, for the preparation of drugs for the modulation of apoptosis. Preferably, xanthine derivatives of the formula I are used, wherein R 2 represents alkyl (C 1 C and one of the radicals R 1 or R 3 represents a radical of the formula II, wherein R represents a) a radical -CO- or b) a radical -C (R) (0H) -, and n means the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or alkyl (C ^ C,) and the other radical R3 or R1 represents alkyl (C) ^ C-) or (C4-C8) cycloalkyl-alkyl. Particularly preferably, xanthine derivatives of the formula I are used, wherein R 2 represents alkyl (C x C 2), R 1 represents the radical of formula II, wherein R represents a) -CO- or b radical) a radical -C (R4) (0H) -, and n means the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or alkyl (C ^ Cj) and R3 represents alkyl (C ^ C,) or (C4-C8) cycloalkyl-alkyl. Particularly preferred are xanthine derivatives of the formula I, wherein R 2 represents alkyl (C x -C 2), R 1 represents a radical of the formula II, wherein R represents a) a radical -CO- or b) a radical - C (R4) (0H) -, and n means the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or (C1-C2) alkyl and R3 represents (C2-C5) alkyl or (C4-C6) cycloalkyl ) -I rent.
Very particularly preferably, 1- (5-hydroxy-5-methylhexyl) -3-methyl-7-propylxanthine is used. The alkyl radicals of the formula I are straight-chain or branched. The term "(C4-C8) cycloalkyl-alkyl" defines alkyl radicals which are substituted by (C3-C6) cycloalkyl, the sum of all C atoms being less than or equal to 8. They include, for example, cyclopropyl-methyl-bis-pentyl, cyclobutyl-methyl-bis-butyl, cyclopentyl-methyl-bis-propyl, as well as cyclohexyl-methyl and -ethyl ester. The radical "(O)" represents an oxygen atom. By "modulation of apoptosis" is meant the inhibition or induction of apoptosis. The xanthine derivatives of the formula I are prepared according to known procedures (US 3,737,433; US 4,108,995; US 4,833,146). One way of proceeding, for example, is to rent a 3-alkylxanthine of formula II, wherein R2 represents an alkyl group with 1 to 4 C atoms, A represents a hydrogen atom, (C 4 -C 7) -alkyl, (C 2 -C 6) alkoxy-alkyl or the radical of the formula II and B represents a hydrogen atom, (C 4 -C 8) -alkyl-alkyl, (C2-C2) alkoxy-alkyl, the radical of the formula II, the benzyl or diphenyl radical, but wherein at least one of these radicals A and B means a hydrogen atom, directly or in the presence of a condensing agent of basic character or in the form of one of its salts in position 1 and / or 7, in a step or stepwise manner, with corresponding alkylating agents of the general formula III X-Q (III) wherein X means a halogen atom or an ester grouping of sulfonic acid or phosphoric acid ester and Q means (C4-C8) cycloalkyl-alkyl, (C2-C6) alkoxy-alkyl or a radical of the formula II, subsequent reductive separation of radical B, when it represents a benzyl or diphenylmethyl group, or optionally hydrolytic removal of an alkoxymethyl radical from the position of radical B and / or reduction of the keto group in the alcohol function when A or B means a radical oxoalkyl, at a reaction temperature between 0 ° C and the boiling point of the reaction medium in each case used. The starting substances of the reactions are known or can be easily prepared according to methods known from the literature. The invention also relates to medicaments for the modulation of apoptosis which contain at least an effective amount of a xanthine derivative of the formula I together with pharmaceutically suitable and physiologically compatible support substances, diluents and / or other active substances and adjuvants. . The invention also relates to processes for the preparation of a medicament for the modulation of apoptosis, which are characterized in that a suitable form of administration is brought to at least one xanthine derivative of the formula I with a physiologically acceptable support and other substances active ingredients, additives or coadjuvants. The medicaments according to the invention are administered parenterally, orally, rectally, or optionally also topically. Suitable forms of galenic, solid or liquid preparations are, for example, granules, powders, dragees, tablets, (micro) capsules, suppositories, syrups, juices, suspensions, emulsions, drops or injectable solutions, as well as preparations with delayed release of the substance. active, in the preparation of which are found suitable auxiliary agents, such as support substances, disintegrating agents, binders, coating, expansion, glidants or lubricants, flavoring substances, sweeteners or dissolution inducers. As frequently used adjuvants there may be mentioned, for example, magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, milk albumin, gelatin, starch, cellulose and its derivatives, animal and vegetable oils, polyethylene glycols and solvents, such as for example sterile water and monovalent or polyvalent alcohols, for example glycerol.
By virtue of the pharmacological properties of the xanthine derivatives of the formula I, these compounds can be used for the directed modulation of apoptosis. Therefore, diseases with excessive apoptosis can be treated, such as infarction, myoma, muscular atrophy, muscular dystrophy, cachexia, systemic inflammation response syndrome (SIRS), respiratory distress syndrome in adults (ARDS), cerebral malaria, chronic pulmonary inflammation, pulmonary sarcosidosis, reperfusion injury, scarring, intestinal inflammation, acquired immunodeficiency syndrome (AIDS), cancer, diseases with increased protein loss, stroke, neurodegeneration, chronic renal failure, burn injuries or diseases hypertrophic Preferably, the pharmaceutical preparations are produced and administered in dosage units, each unit containing as active component a certain dose of at least one xanthine derivative of the formula I. In the case of solid dosage units, such as tablets, capsules, dragees or suppositories, this dose amounts to approximately 1000 mg, but preferably approximately 100-600 mg, and in the case of solutions for injection in the form of ampoules up to 300 mg, preferably 20-200 mg. For the treatment of a patient (70 kg), an intravenous infusion treatment of 100-2000 mg per day is indicated in the early stages. In the subsequent rehabilitation phase, an oral administration of 3 times 400 mg per day is indicated, in particular 1- (5-hydroxy-5-methylhexyl) -3-methyl-7-propylxanthine. In certain circumstances, however, higher or lower doses are also suitable. The administration of the dose can be effected either by single administration in the form of a single dosage unit, or of several smaller dosage units, as well as by multiple administration of subdivided doses at certain intervals. Lastly, the xanthine derivatives of the formula I and / or optionally their corresponding salts can be formulated in the preparation of the aforementioned pharmaceutical preparation forms also together with other suitable active substances, for example active substances that take up the free oxygen radicals , for example 1, 5-hydro-4H-pi-razolo (3,4-d) pyrimidin-4-one, superoxide dismutase, dimethyl sulfoxide or mannitol, heparin, ascorbic acid or deferoxamine. In addition, a combination preparation containing a xanthine derivative of the formula I and a compound of the formula IV or V shows a superadditive inhibitory effect on the dephosphorylation of cofilin and, thereby, on the activation of cofilin leading to a modulation of apoptosis. By virtue of the magnitude of this effect, the application of this combination preparation can be prolonged to sectors that until now were not accessible, for example in an immunosuppressive therapy through the individual components. Therefore, the invention further relates to a combination preparation, which contains 1) at least one xanthine derivative of the formula I as defined above, 2) a compound of the formula IV and / or V, and / or a possibly stereoisomeric form of the compound of the formula IV or V and / or a physiologically compatible salt of the compound of the formula V, wherein represents a) (C 1 -C 4) alkyl, b) (C 3 -C 5) cycloalkyl, Or (C2-C6) alkenyl or d) alkynyl (C2-C6), represents a) -CF ,, c) -S-CF3, d) -OH, e) -N0a, f) halogen, g) benzyl, ) phenyl, i) -0-phenyl, k) -CN or 1) -O-phenyl, substituted one or more times with 1) alkyl (Cx-C4), 2) halogen, 3) -0-CF3 or 4) -0-CH3, R7 represents a) alkyl (-C, b) halogen, or c) a hydrogen atom, and X represents a) a group -CH or b) a nitrogen atom, and 3) a pharmaceutical support, for modulation of apoptosis. The use of a compound of the formula IV and / or V and / or of an optionally stereoisomeric form of the compound of the formula IV or V and / or a salt of the compound of the formula V is preferred., wherein R 5 represents a) methyl b) cyclopropyl or c) alkynyl (C 1 -C 5), R 6 represents -CF 3 or -CN, R 7 represents a hydrogen or methyl atom, and X represents a group -CH-, in combination with xanthine derivatives of the formula I, wherein R 2 represents alkyl (Ci-Cj), R 1 represents a radical of the formula II, wherein R represents a) -CO- radical or a radical -C (R 4) (0H) -, and n means the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or alkyl (C.-C,) and R3 represents (C2-C3) alkyl or (C4-C6) cycloalkyl- I rent. The use of N- (4-trifluoromethylphenyl) -2-cyan-3-hydroxy-crotonic acid amide, 2-cyano-3-cyclopropyl-3-hydroxy- (4-cyanophenyl) -amide is particularly preferred. N- (4-trifluoromethylphenyl) -2-cyan-3-hydroxy-hept-2-en-6-in-carboxylic acid acrylic or amide in combination with 1- (5-hydroxy-5-methylhexyl) -3- methyl-7-propylxanthine. The preparation of the compound of formula IV or V is carried out according to known processes, such as described in EP 484 223; EP 529 500; US 4 061 767; EP 538 783 or EP 551 230. Starting substances for chemical reactions are known or can be easily prepared according to methods known from the literature. By the term alkyl, alkenyl or alkynyl radicals are understood, whose carbon chain can be linear or branched. In addition, the alkenyl or alkynyl radicals may also contain several double bonds or several triple bonds. Cyclic alkyl radicals are for example 3 to 5 membered monocycles, such as cyclopropyl, cyclobutyl or cyclopentyl. The term "superadditive" is understood to mean effects that are greater than the sum of the individual effects. The combination preparation according to the invention is suitable, for example, for the treatment of transplants, autoimmune diseases, infarction, stroke, inflammations, neurodegeneration, myoma, muscular atrophy, muscular dystrophy, cachexia, response syndrome to systemic inflammation ( SIRS), adult respiratory distress syndrome (ARDS), cerebral malaria, chronic lung inflammation, pulmonary sarcosidosis, reperfusion injury, scarring, intestinal inflammation, burn injuries, acquired immunodeficiency syndrome (AIDS), cancer, diseases with a increased protein loss, neurodegeneration, chronic renal failure or hypertrophic diseases. The combination preparation according to the invention can also comprise compositions or combinations packages, in which the components are arranged side by side and, therefore, can be applied simultaneously, separately or staggered in time to one and the same. same human or animal body.
The invention also relates to processes for the production of a combination preparation for the modulation of apoptosis, characterized in that at least one xanthine derivative of the formula I and a compound of the formula IV or V are brought into a form of suitable administration with a physiologically acceptable support and other active substances, additives or suitable adjuvants. The combination preparation according to the invention can be present as a dosage unit in the form of medicinal forms such as capsules (including micro-capsules which, in general, do not contain any pharmaceutical support), tablets (including dragees and pills) or suppositories, where, in the case of use of capsules, the material of the capsule assumes the function of the support and the content may be present for example in the form of powder, gel, solution, emulsion or dispersion. However, it is particularly advantageous and simple to prepare oral (peroral) formulations with the two active components 1) and 2) containing the calculated amounts of the active substances together with each desired pharmaceutical support. A corresponding formulation (suppositories) can also be used for rectal therapy. Also, transdermal application in the form of ointments or creams, parenteral injection (intraperitoneal, subcutaneous, intramuscular) or oral application of solutions containing the combinations according to the invention is possible. Ointments, pastes, creams and powders may contain, together with the active substances, the usual support substances, for example animal and vegetable fats, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicone, silicic acid, hydroxide, aluminum, talcum, zinc oxide, lactose, bentite, calcium silicate and polyamide powder, or mixtures of these substances. Tablets, pills or granules can be prepared according to processes such as the pressing, dipping or fluidized-bed process or boiling and contain customary support agents and other adjuvants, such as gelatin, agarose, starch (for example starch). of potato, corn or wheat), cellulose, such as ethylcellulose, silicon dioxide, magnesium carbonate, different sugars, such as lactose and / or calcium phosphates. The dragee solution usually consists of sugar and / or starch syrup and most often contains gelatin, synthetic cellulose esters, gum arabic, polyvinylpyrrolidone, pigments, surfactants, plasticizers and similar additives in a manner corresponding to the known state of the art. The technique. For production of the preparation forms, any conventional flow regulator, lubricant or glidant, such as magnesium stearate and separating agents, can be used. Preferably, the preparations have the formula of shell / core tablets or multilayer tablets, the active component 2 being in the shell or in the core or in a layer, while the active component 1 is in the core, in the shell. envelope or in another layer. The components of active substances can also be present in a delayed form or be adsorbed to a delayed release material or be included in a delayed release material (for example based on cellulose or polystyrene resin, for example hydroxyethylcellulose). A delayed release of the active substances can also be achieved by providing the corresponding layer or the compartment with customary insoluble coatings in the gastric juices. The dosage to be used naturally depends on different factors, such as the living being to be treated (ie man or animal), age, weight, general state of health, degree of severity of the symptoms, of the disease to be treated, of eventual diseases concomitant (in case they arise) the type of accompanying treatment with other medications, or the periodicity of the treatment. The dosages are usually administered several times a day and, preferably, once to three times a day. The amounts used of the individual active substance are in this case oriented to the recommended daily dose of the respective individual active substance and should, in general, be in the combination preparation in 10% to 100% of the recommended daily dose, preferably of 20% to 80%, in particular 50%. The appropriate therapy with the combinations according to the invention therefore consists, for example, in the administration of one, two or three individual dosages of the preparation consisting of the sodium salt of N-4- (trifluoromethylphenyl) -2-cyan acid amide. -3-hydroxy-crotonic in an amount of 2 mg to 250 mg, preferably 5 mg to 150 mg, in particular 10 mg to 50 mg, particularly preferably 10 mg to 20 mg and 1- (5-hydroxy-5 -methylhexyl) -3-methyl-7-propyl-xanthine in an amount of 100 to 600 mg, in particular 150 to 300 mg, preferably 20 to 200 mg.
Example 1 Preparation of 1- (5-hydroxy-5-methylhexyl) -3-methy-7-propylxan-ina To a suspension of 61.3 g (0.2 mol) of 3-methyl-1- (5-oxohexyl) -7-propylxanthine in 2 l of anhydrous ether are added dropwise, under vigorous stirring at room temperature, 22.4 g (0.3 mol) of methylmagnesium chloride in the form of a 20% solution in tetrahydrofuran, the internal temperature rising to about 30 ° C. It is then heated for 2 hours under stirring and reflux to decompose the formed alkanolate with saturated aqueous ammonium chloride solution, w separates the organic phase and is washed twice with 500 ml of water each time. The combined water phases are extracted again thoroughly with dichloromethane.
The extract is combined in dichloromethane with the ether phase, dried over sodium sulfate, filtered and concentrated by evaporation under reduced pressure, obtaining 59.0 g of crude product (91.5% of theory) w is purified by recrystallization in diisopropyl ether. Yield 49.8 g (77.2% of theory); Melting point: 81-82 ° C C16H26N403 (MW = 322.4) Analysis: Calculated: C 59.61% H 8.13% N 17.38% Found: C 59.72% H 8.09% N 17 , 44% Example 2 Pharmacological Assay 2.1 Cell Culture The murine macrophage cell line RAW 264.7 was purchased from ATCC (Rockville, MD) and cultured in DMEM (Sigma, St. Louis, MO) with 4.5 g glucose / 1, 110 mg of sodium pyruvate / 1, 10% FCS inactivated by heat (Gibco, Grand Island, NY) and penicillin / streptomycin (50 U / 50 mg / ml). The macrophages were passed every 2-3 days, and one day before the start of the experiment, they were separated in the order of 2-106 cells in tissue culture flasks (75 cm2, Falcon, Becton Dickinson GmbH, Heidelberg, Germany) . The cells were supplied with fresh medium and the preparations were added at the corresponding concentrations. 1- (5-hydroxy-5-methylhexyl) -3-methyl-7-propylxanthine (compound 1) was dissolved in the concentration of 20 mM in cell medium. From this, 100 μl (100 μM) and 50 μl (50 μM) were pipetted into 20 ml of medium. Sodium salt of N-4- (trifluoromethylphenyl) -2-cyan-3-hydroxy-crotonic acid amide (compound 2) was dissolved in a concentration of 12 mM in cell medium. Of this, 100 μl (final concentration 60 μM), 33 μl (final concentration 20 μM) and 16.7 μl (final concentration 10 μM) were pipetted into each 20 ml of medium. Stimulation with lipopolysaccharides (LPS; E. coli, serotype 0127.-B 8 Next, St. Louis, MO) was carried out at a concentration of 10 ng / ml for 1 hour after the previous incubation with the preparation. Aliquots of a standard solution of lipopolysaccharides (LPS 1 mg / ml in 10% dimethylsulfoxide (DMSO)) were diluted in the medium to a concentration of 1 μg / ml and stored at -20 ° C. The cells were incubated for 24 hours (h) at 37 ° C in 10% C02. 2. 2 Preparation of the samples All the chemicals used were analytically pure or with an electrophoresis quality, and were purchased from Millipore Co. (Bedford, MA) or Sigma (St. Louis, MO), if other sources are not expressly indicated. acquisition. The 2-D electrophoresis (2 -DE) was carried out with the Investigator System® (Millipore), and the samples were treated according to the manufacturer's prescription with minor modifications. Adherent murine macrophages were washed on ice three times every 60 seconds with 10 ml of ice-cold PBS. The cells were then lysed in 1 ml of boiling lysis buffer, consisting of 0.3 g of SDS, 3.088 g of DTT, 0.444 g of TrisHCl and 0.266 g of Tris-base, in 100 ml. The used cell was scraped off and cured in boiling water in a 2 ml sample container for 10 minutes (min). The polynucleotides were separated by the addition of Benzonase® (Merck, Darmstadt, Germany) for 30 min at 37 ° C. At this point of the sample preparation an aliquot was removed, and the protein content was determined according to the Popov method. For 2-DE, the proteins in the sample were precipitated by dropwise addition to ice-cold acetone (80% v / v). The sample was cooled for 20 min on ice and then centrifuged at 240 g for 10 min.
The dried pellet was collected in one part of lysis buffer and four parts of a sample buffer with a protein content of 5 mg / ml. The sample buffer consists of 59.7 g of urea, 4.0 ml of NP-40, 1.54 g of. DTT, 5.5 ml of support ampholytes (pH 3-10, optimized by 2 -DE) in 100 ml. The undissolved material was separated from the electrophoresis by centrifugation of the samples at 16,000 x g. 2. 3 Gel electrophoresis 2 -DE High dissolution two-dimensional gel electrophoresis was carried out according to the O'Farrell method with modifications, as already described by Garrels. For this, the Millipore 2-D Investigator electrophoresis system (Millipore Co., Bedford, MA) was used. The isoelectric focus was carried out in glass capillaries (1 mm in diameter) with a thread of 0.08 mm thickness, which prevents a dilation and a break of the rod. The IEF gel consists of a polyacrylamide matrix of 4.1% T, 2.4% C, which was prepared from a standard solution of 30.8% T, 2.6% C, urea 9 , 5 M, 2.0% (v / v) of NP-40, 10 mM Chaps and 2% (v / v) of support ampholytes (pH 3-10, optimized by 2-DE). 0.01 M H3P04 was used as the anode buffer, and 0.1 M NaOH was used as the cathode buffer. Before the previous approach for the formation of the pH gradient, 15 μl of a coating buffer of the samples was applied, consisting of 0.5 M urea, 0.2% (v / v) of NP-40, 0.1% (v / v) of ampholytes of support and DTT 50 M. The maximum voltage of 1500 volts was reached in the space 90 minutes at a maximum current of 110 μA / gel. After the previous focus 20 μl of the sample (100 μg of protein) and another 15 μl of coating buffer were applied. The isoelectric focus of the proteins was carried out in the space of 18,000 Vh. After the electrophoresis was complete, the rodlets were chilled on ice and equilibrated in a buffer consisting of 0.3 M Tris-base, 0.075 M Tris HC1, 6% SDS, 50 mM DTT and 0.01% blue of bromophenol. The rod-shaped gels were transferred directly onto the vertical gel surface of the second dimension or stored at -20 ° C until use. The second dimension was carried out in an SDS gradient gel (10-17%) without a harvester gel. The gradient was prepared by mixing two gel solutions. A: 100 ml of acrylamide (30.5% of T, 1.64% of C), 73 ml of Tris (1.5 M, pH 8.8), 123 ml of H20, 3 ml of SDS (up to 10 ml). %), 150 μl of TEMED and 750 μl of ammonium peroxodisulfate (10%). B: 170 ml of acrylamide, 73 ml of Tris, 66.78 g of glycerol, 3 ml of SDS, 150 ml of TEMED, 750 ml of ammonium peroxodisulfate. The electrophoresis was carried out overnight at a constant temperature in an elution buffer, consisting of 25 mM Tris-base, 192 mM glycine and 0.1% SDS, until the bromophenol blue front was approximately 1 cm apart. cm from the end of the gel. After the electrophoresis was complete, the proteins were stained in gel with silver reagent according to Heukeshoven and Dernick. The analysis of the 2-D gels and the preparation of synthetic formers was carried out with the Biolmage system (Biolmage Systems Co.). The protein model obtained was explored with a Kodak Magaplus model 1.4 camera and the data was processed by a HAM station. 2. 4 Results The results of the unstimulated controls were established at 100%. The addition of LPS (10 ng / ml) led to a 50% dephosphorylation of cofilin. The simultaneous application of LPS (10 ng / ml) and of compound 1 (100 μM) leads to a dephosphorylation of 10% cofilin. Therefore, the inhibition of dephosphorylation is 80% compared to macrophages treated only with LPS. Table 1 shows the results. In contrast to a final concentration at 100 μM, a final concentration of 50 μM of compound 1 is ineffective. Compound 2 also lacks effect up to a final concentration of 20 μM and is only active at 60 μM. If compound 1 and compound 2 are combined in a range of concentrations in which each individual compound is ineffective, a superadditive effect results surprisingly.
Comp. represents Compound

Claims (11)

1. - Use of a xanthine derivative of the formula I and / or a possibly stereoisomeric form of the compound of the formula I, wherein R2 represents alkyl (Cx-C4), one of the radicals R1 or R3 represents a radical of the formula II - (CH2) n-R-CH3 (II) where R represents a) a single covalent bond, and where n means the integer zero, 1, 2, 3, 4, 5, 6 or 7, b) a radical -CO-, and where n means the number integer 1, 2, 3, 4, 5 or 6, or c) a radical -C (R4) (OH) -, and where n means the integer 1, 2, 3, 4, 5 or 6 and R4 represents ) one hydrogen atom or b) alkyl (C? -C3), and the other radical R3 or R1 represents a) a hydrogen atom, b) alkyl (C ^ -C-,), c) (C4-C8) cycloalkyl) -alkyl or d) alkyl having 2 to 6 carbon atoms, wherein the carbon chain is interrupted by an oxygen atom, for the preparation of drugs for the modulation of apoptosis.
2. Use according to claim 1, characterized in that a compound of the formula I is used, wherein R 2 represents alkyl (C 1 C and one of the radicals R 1 or R 3 represents a radical of the formula II, wherein R represents ) a radical -CO- or b) a radical -C (R4) (0H) -, and n means the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or alkyl (Cx-C3) and the other radical R3 or R1 represents alkyl (C ^ C?) Or (C4-C8) cycloalkyl-alkyl.
3. Use according to claim 1 or 2, characterized in that a xanthine derivative of the formula I is used, wherein R2 represents alkyl (Cx-C2), R1 represents the radical of the formula II, wherein R represents a) a radical -CO- or b) a radical -C (R4) (0H) -, and n means the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or alkyl (C ^ C.) and R3 represents alkyl (C ^ C?) Or (C4-C8) cycloalkyl-alkyl.
4. Use according to one or more of claims 1 to 3, characterized in that a xanthine derivative of the formula I is used, wherein R2 represents alkyl (L-C2), R1 represents a radical of the formula II, wherein R a) represents a radical -CO- or b) a radical -C (R4) (0H) -, and n signifies the integer 3, 4, 5 or 6 and R4 represents a hydrogen atom or alkyl (C ^, ) and R3 represents (C2-C5) alkyl or (C4-C6) cycloalkyl-alkyl.
5. Use according to one or more of claims 1 to 4, characterized in that 1- (5-hydroxy-5-methylhexyl) -3-methyl-7-propylxanthine is used.
6.- Combination preparation, which contains 1) at least one xanthine derivative of the formula I according to claims 1 to 5, 2) a compound of the formula IV and / or V, and / or a possibly stereoisomeric form of the compound of the formula IV or V and / or a physiologically compatible salt of the compound of the formula V, wherein represents a) (C 1 -C, C) alkyl (C 3 -C 3) alkyl, c ) (C2-C6) alkenyl or d) (C2-C6) alkynyl, R6 represents a) -CF3, b) -0-CF3, O -S-CF3, d) -OH, e) -N02, f) halogen , g) benzyl, h) phenyl, i) -O-phenyl, k) -CN or 1) -O-phenyl, substituted once or several times with 1) alkyl (C ^ C, 2) halogen, 3) -0 -CF3 or 4) -0-CH3, R7 represents a) (Cx-C4) alkyl, b) halogen, or c) a hydrogen atom, and X represents a) a group -CH or b) a nitrogen atom, and ) a pharmaceutical support.
7.- Combination preparation according to claim 6, characterized in that a compound of the formula IV and / or V is used, wherein R5 represents a) methyl b) cyclopropyl or c) alkynyl (C3-C3), Rβ represents -CF3 or -CN, R7 represents a hydrogen or methyl atom, and X represents a group -CH- in combination with xanthine derivatives of the formula I, wherein R2 represents alkyl (Cx-C2), R1 represents a radical of the formula II , wherein R represents a) a radical -CO- or b) a radical C (R4) (0H) -, and n stands for the integer 3, 4, 5 or 6 and R4 represents a hydrogen or alkylene atom (C ^ C ^ and R3 represents (C2-C5) alkyl or (C4-C6) cycloalkyl-alkyl.
8. - Combination preparation according to claim 6 or 7, characterized in that N- (4-trifluoromethylphenyl) -2-cyan-3-hydroxy-crotonic acid, 2-cyano-3-cyclopropyl-3-hydroxy (4-cyanophenyl) -amide is used as compound V N- (4-trifluoromethylphenyl) -2-cyano-3-hydroxy-hept-2-en-6-in-carboxylic acid acrylic acid amide and 1- (5-hydroxy-5-hydroxy) -methylhexyl) -3-methyl-7-propylxanthine.
9.- Combination preparation according to one of claims 6 to 8, characterized in that N- (4-trifluoromethyl-phenyl) -2-cyano-3-hydroxy-chromic acid amide and 1- (5-idroxy-methylhexyl) are used. -3 -me il-7-propylxan ina.
10. Use of a compound of formula I according to one or more of claims 1 to 5 and a compound of formula IV and / or V according to the definition in one or several of claims 6 to 9 for the preparation of a medication for the modulation of apoptosis.
11. Employment according to claim 10, for the treatment of transplants, autoimmune diseases, infarction, stroke, inflammations, neurodegeneration, myoma, muscular atrophy, muscular dystrophy, cachexia, response to systemic inflammation syndrome (S, pain syndrome respiratory disease in adults (ARDS), cerebral malaria, chronic pulmonary inflammation, pulmonary sarcosidosis, re-perfusion injuries, scarring, intestinal inflammations, burn injuries, acquired immunodeficiency syndrome (AIDS), cancer, diseases with increased protein loss , chronic renal failure or hypertrophic diseases.
MXPA02005771A 1995-06-05 1996-06-04 Oral care ultramulsion based products. MXPA02005771A (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US08/461,698 US5733529A (en) 1995-06-05 1995-06-05 Ultramulsion based antigingivitis toothpaste compositions
US08/462,599 US5665374A (en) 1995-06-05 1995-06-05 Ultramulsion containing interdental delivery devices
US08/462,930 US5645841A (en) 1995-06-05 1995-06-05 Ultramulsion based oral care rinse compositions
US08/462,203 US5651959A (en) 1995-06-05 1995-06-05 Ultramulsion based oral care compositions
US08/464,403 US5711936A (en) 1995-06-05 1995-06-05 Ultramulsion based ingestible compositions
PCT/US1996/008714 WO1996039116A1 (en) 1995-06-05 1996-06-04 Oral care ultramulsion based products
DE19630837.2 1996-07-31
DE19640556.4 1996-10-01

Publications (2)

Publication Number Publication Date
MXPA97005771A true MXPA97005771A (en) 1998-11-12
MXPA02005771A MXPA02005771A (en) 2005-02-22

Family

ID=27541677

Family Applications (2)

Application Number Title Priority Date Filing Date
MXPA02005771A MXPA02005771A (en) 1995-06-05 1996-06-04 Oral care ultramulsion based products.
MX9709735A MX9709735A (en) 1995-06-05 1997-12-05 Oral care ultramulsion based products.

Family Applications After (1)

Application Number Title Priority Date Filing Date
MX9709735A MX9709735A (en) 1995-06-05 1997-12-05 Oral care ultramulsion based products.

Country Status (8)

Country Link
EP (1) EP0831762A4 (en)
JP (1) JPH11506769A (en)
CN (1) CN1190342A (en)
AU (1) AU712266B2 (en)
BR (1) BR9608343A (en)
CA (1) CA2222407A1 (en)
MX (2) MXPA02005771A (en)
WO (1) WO1996039116A1 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0955836B1 (en) * 1996-07-25 2003-10-29 Whitehill Oral Technologies Inc. Toothbrush with improved cleaning and abrasion efficiency
DE19721590A1 (en) * 1997-05-23 1998-11-26 Henkel Ecolab Gmbh & Co Ohg Non-drying, viscous surface treatment agent
US6190706B1 (en) * 1997-12-23 2001-02-20 Wm. Wrigley Jr. Company Chewing gum and gum bases having reduced adhesion to environmental surfaces and method for making
US6596298B2 (en) 1998-09-25 2003-07-22 Warner-Lambert Company Fast dissolving orally comsumable films
BR0101911A (en) * 2001-04-30 2002-12-24 Johnson & Johnson Ind Com Composition for oral hygiene
CN102038963B (en) * 2003-06-30 2013-09-11 日本制药株式会社 Method for preparing a stabilised menthol emulsion
WO2009067605A2 (en) * 2007-11-21 2009-05-28 The Procter & Gamble Company Preparations, methods and kits useful for treatment of cough
TWI392514B (en) 2010-01-29 2013-04-11 Colgate Palmolive Co Fluoride free and anionic surfactant free dentifrice having a high micro efficacy
CN107252396A (en) * 2010-11-12 2017-10-17 高露洁-棕榄公司 Oral care product and its use and preparation method
US9877929B2 (en) 2011-10-13 2018-01-30 Premier Dental Products Company Topical vitamin D and ubiquinol oral supplement compositions
WO2014059157A1 (en) * 2012-10-12 2014-04-17 Premier Dental Products Company Topical vitamin d oral supplement compositions
AU2013329156B2 (en) * 2012-10-12 2018-07-12 Premier Dental Products Company Topical vitamin D and UBIQUINOL oral supplement compositions
US9724542B2 (en) * 2012-10-12 2017-08-08 Premier Dental Products Company Remineralizing and desensitizing compositions, treatments and methods of manufacture
WO2015157683A1 (en) * 2014-04-11 2015-10-15 Premier Dental Products Company Enamel protectant and repair toothpaste
US9877930B2 (en) * 2012-10-12 2018-01-30 Premier Dental Products Company Topical ubiquinol oral supplement compositions with amorphous calcium phosphate
CN103646763A (en) * 2013-11-28 2014-03-19 宝电电子(张家港)有限公司 Transformer
JP2015224233A (en) * 2014-05-29 2015-12-14 サンメディカル株式会社 Composition for tooth cleaning treatment
EP3151861B1 (en) 2014-06-09 2022-04-06 Klox Technologies Inc. Silicone-based biophotonic compositions and uses thereof
WO2020097051A1 (en) * 2018-11-05 2020-05-14 Inspired Material Solutions, LLC Dimethicone-based oral varnish

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057306A (en) * 1983-11-06 1991-10-15 Hill Ira D Method of manufacturing oral hygiene gels
US5057307A (en) * 1986-11-06 1991-10-15 Hill Ira D Method of relieving gum discomfort
US5098711A (en) * 1988-11-14 1992-03-24 Ira Hill Method of treating the oral cavity with dental floss containing chemotherapeutic agents
US5380530A (en) * 1992-12-29 1995-01-10 Whitehill Oral Technologies Oral care composition coated gum

Similar Documents

Publication Publication Date Title
MXPA97005771A (en) Employment of xantina derivatives for the modulation of the apopto
WO2019040706A1 (en) Compositions and methods for treatment of vitiligo
KR101290745B1 (en) Pharmaceutical Composition for Prevention or Treatment of Inflammatory or Allergy Diseases Comprising Ramalin
WO2004103959A2 (en) Heterocyclic compounds and uses thereof
IL127717A (en) Anticonvulsant derivatives useful in treating amyotrophic lateral sclerosis (als)
CZ212394A3 (en) Enantiomeric hydroxy-xanthine derivatives
EP0552373A1 (en) Apoptosis regulator
HU217132B (en) Use of riluzole for preparing pharmaceutical compositions treating aids-related neural disorders
US20180303780A1 (en) Derivatives used in the treatment of muscle atroph
JPH0710875A (en) Selective phosphodiesterase iv inhibitor
ATE254463T1 (en) USE OF VALACICLOVIR FOR THE MANUFACTURE OF A MEDICINAL PRODUCT FOR THE TREATMENT OF GENITAL HERPES BY ONCE DAILY ADMINISTRATION
JPH07173063A (en) Remedy for malaria containing pharmaceutically active compound
CA2212205C (en) Use of xanthine derivatives for the modulation of apoptosis
US6011051A (en) Use of isoxazole and crotonamide derivatives for the modulation of apoptosis
CA2555862A1 (en) Polyamine analogs that activate antizyme frameshifting
US5981536A (en) Use of xanthine derivatives for the modulation of apoptosis
US5962480A (en) Drug for ameliorating brain diseases
KR20050047036A (en) The use of benzisoselenazolone compounds against ischemic myocardial damage
GB2074858A (en) Compositions containing 3-deazaaden-osine
JP2001500526A (en) Use of 2-methyl-thiazolidine-2,4-dicarboxylic acid as a mucolytic agent
EP1598079B1 (en) Remedies for allergic contact dermatitis
WO2021100847A1 (en) Carbalysophosphatidic acid
EP0998924A1 (en) Antitumor agents
JPH0717857A (en) Circulatory disease therapeutic agent
KR960014874B1 (en) Uricosuric composition