Unnamed: 0
int64 0
350k
| ApplicationNumber
int64 9.75M
96.1M
| ArtUnit
int64 1.6k
3.99k
| Abstract
stringlengths 7
8.37k
| Claims
stringlengths 3
292k
| abstract-claims
stringlengths 68
293k
| TechCenter
int64 1.6k
3.9k
|
---|---|---|---|---|---|---|
600 | 14,505,965 | 1,615 | A tablet that includes a solid solution of soluble fiber and one or more sugar alcohols, the solid solution having a glass transition temperature of less than 40° C., and one or more additives dispersed in the solid solution such that at least one additive is released from the tablet when the tablet is chewed or dissolved within an oral cavity. | 1. A dissolvable-chewable tablet comprising:
a solid solution of soluble fiber and one or more sugar alcohols, the solid solution having a glass transition temperature of less than 40° C.; and one or more additives dispersed in the solid solution such that the at least one additive is released from the tablet when the tablet is chewed or dissolved within an oral cavity. 2. The tablet of claim 1, wherein the solid solution comprises at least 20 weight percent of soluble fiber. 3. A dissolvable-chewable tablet comprising:
a solid solution comprising:
at least 20 weight percent soluble fiber, and
at least 20 weight percent of one or more sugar alcohols; and
one or more additives dispersed in the solid solution such that at least one additive is released from the tablet when the tablet is chewed or dissolved within an oral cavity. 4. The tablet of claim 3, wherein the solid solution has a glass transition temperature of less than 40° C. 5. The tablet of claim 1, wherein the solid solution has a glass transition temperature of between −50° C. and 40° C. 6. The tablet of claim 5, wherein the solid solution has a glass transition temperature of between −20° C. and 10° C. 7. The tablet of claim 1, wherein the tablet further comprises cellulosic fibers dispersed in the solid solution. 8. The tablet of claim 7, wherein the tablet comprises at least one additive absorbed into the cellulosic fibers. 9. The tablet of claim 1, further comprising oil dispersed in the solid solution. 10. The tablet of claim 9, wherein the tablet comprises 2-15 weight percent of oil. 11. The tablet of claim 9, wherein the oil is selected from the group consisting of partially hydrogenated oil, oil, palm kernel oil, coconut oil, corn oil, cotton seed oil, olive oil, peanut oil, canola oil, sesame oil, soybean oil, rapeseed oil, safflower oil, sunflower oil, mustard oil, almond oil, beech nut oil, cashew oil, hazelnut oil, macadamia oil, pecan oil, pine nut oil, pistachio oil, walnut oil, grapefruit seed oil, lemon oil, orange oil, pumpkin oil, watermelon seed oil, citrus oils, oils from melons and gourd seeds, flaxseed oil, cocoa butter. 12. The tablet of claim 1, further comprising a plasticizer dispersed in the solid solution. 13. The tablet of claim 12, wherein the plasticizer comprises propylene glycol, glycerin, vegetable oil, triglycerides, or a combination thereof. 14. The tablet of claim 1, wherein the soluble fiber is a dietary fiber. 15. The tablet of claim 1, wherein the soluble fiber comprises maltodextrin. 16. The tablet of claim 15, wherein the tablet comprises at least 20 weight percent maltodextrin. 17. The tablet of claim 1, wherein the soluble fiber comprises a digestion resistant soluble fiber. 18. The tablet of claim 17, wherein the soluble fiber is a digestion resistant maltodextrin. 19. The tablet of claim 1, wherein the one or more sugar alcohols comprises is selected from the group consisting of mannitol, sorbitol, xylitol, erythritol, isomalt, lactitol, maltitol, maltitol syrup, and hydrogenated starch hydrolysates [HSH]. 20. The tablet of claim 1, wherein the one or more sugar alcohols comprise sorbitol. 21. The tablet of claim 1, wherein the one or more sugar alcohols comprises mannitol. 22. The tablet of claim 1, further comprising an antioxidant. 23. The tablet of claim 1, the tablet having a water content of between 0.5 weight percent and 7 weight percent. 24. The tablet of claim 1, wherein the tablet is disk shaped. 25. The tablet of claim 1, wherein the tablet is part of a sheet structure configured for subdivision into individual tablets. 26. The tablet of claim 1, wherein the one or more additives includes at least one additive selected from the group consisting of minerals, vitamins, dietary supplements, nutraceuticals, energizing agents, soothing agents, amino acids, chemesthic agents, antioxidants, botanicals, teeth whitening agents, therapeutic agents, and combinations thereof. 27. The tablet of claim 1, wherein the one or more additives includes at least one therapeutic agent selected from the group consisting of Gerd, Buprenorphin, Nitroglycerin, Diclofenac, Fentanyl, Carbamazepine, Galantamine, Acyclovir, Polyamidoamine Nanoparticles, Chlorpheniramine, Testosterone, Estradiol, Progesterone, Calcitonin, Fluorouracil, Naltrexone, Odansetron, Decitabine, Selegiline, Lamotrigine, Prochlorperazine, and combinations thereof. 28. The tablet of claim 1, further comprising a flavorant dispersed in the body such that the flavorant is released when the tablet is held or chewed within a mouth of a consumer, wherein the flavorant is selected from the group consisting of licorice, wintergreen, cherry and berry type flavorants, Dramboui, bourbon, scotch, whiskey, spearmint, peppermint, lavender, cinnamon, cardamon, apium graveolents, clove, cascarilla, nutmeg, sandalwood, bergamot, geranium, honey essence, rose oil, vanilla, lemon oil, orange oil, Japanese mint, cassia, caraway, cognac, jasmin, chamomile, menthol, ylang ylang, sage, fennel, piment, ginger, anise, chai, coriander, coffee, mint oils from a species of the genus Mentha, and combinations thereof. 29. The tablet of claim 1, wherein the one or more additives comprises a color additive. 30. The tablet of claim 1, wherein the solid solution comprises between 30 weight percent and 50 weight percent of soluble fiber and between 30 weight percent and 50 weight percent of the one or more sugar alcohols. 31. The tablet of claim 1, wherein the tablet is chewable. 32. A method of forming a dissolvable-chewable tablet:
forming a molten mixture of soluble fiber, one or more sugar alcohols, one or more additives, and water while maintaining a mixture temperature of less than 150° C., the molten mixture including at least 20 weight percent of soluble fiber, at least 20 weight percent of one or more sugar alcohols, and less than 15 weight percent water; and portioning the molten mixture into a plurality of dissolvable-chewable tablets. 33. The method of claim 32, wherein the molten mixture is formed in an extruder, wherein the extruder comprises multiple stages, further comprising mixing the molten mixture with a plasticizer prior to portioning the molten mixture into a plurality of tablets. 34. The method of claim 32, further comprising cutting the molten mixture into individual tablets sized to be at least partially received in an oral cavity of a consumer. 35. The method of claim 33, wherein the plasticizer is partially hydrogenated vegetable oil, propylene glycol, or a combination thereof. | A tablet that includes a solid solution of soluble fiber and one or more sugar alcohols, the solid solution having a glass transition temperature of less than 40° C., and one or more additives dispersed in the solid solution such that at least one additive is released from the tablet when the tablet is chewed or dissolved within an oral cavity.1. A dissolvable-chewable tablet comprising:
a solid solution of soluble fiber and one or more sugar alcohols, the solid solution having a glass transition temperature of less than 40° C.; and one or more additives dispersed in the solid solution such that the at least one additive is released from the tablet when the tablet is chewed or dissolved within an oral cavity. 2. The tablet of claim 1, wherein the solid solution comprises at least 20 weight percent of soluble fiber. 3. A dissolvable-chewable tablet comprising:
a solid solution comprising:
at least 20 weight percent soluble fiber, and
at least 20 weight percent of one or more sugar alcohols; and
one or more additives dispersed in the solid solution such that at least one additive is released from the tablet when the tablet is chewed or dissolved within an oral cavity. 4. The tablet of claim 3, wherein the solid solution has a glass transition temperature of less than 40° C. 5. The tablet of claim 1, wherein the solid solution has a glass transition temperature of between −50° C. and 40° C. 6. The tablet of claim 5, wherein the solid solution has a glass transition temperature of between −20° C. and 10° C. 7. The tablet of claim 1, wherein the tablet further comprises cellulosic fibers dispersed in the solid solution. 8. The tablet of claim 7, wherein the tablet comprises at least one additive absorbed into the cellulosic fibers. 9. The tablet of claim 1, further comprising oil dispersed in the solid solution. 10. The tablet of claim 9, wherein the tablet comprises 2-15 weight percent of oil. 11. The tablet of claim 9, wherein the oil is selected from the group consisting of partially hydrogenated oil, oil, palm kernel oil, coconut oil, corn oil, cotton seed oil, olive oil, peanut oil, canola oil, sesame oil, soybean oil, rapeseed oil, safflower oil, sunflower oil, mustard oil, almond oil, beech nut oil, cashew oil, hazelnut oil, macadamia oil, pecan oil, pine nut oil, pistachio oil, walnut oil, grapefruit seed oil, lemon oil, orange oil, pumpkin oil, watermelon seed oil, citrus oils, oils from melons and gourd seeds, flaxseed oil, cocoa butter. 12. The tablet of claim 1, further comprising a plasticizer dispersed in the solid solution. 13. The tablet of claim 12, wherein the plasticizer comprises propylene glycol, glycerin, vegetable oil, triglycerides, or a combination thereof. 14. The tablet of claim 1, wherein the soluble fiber is a dietary fiber. 15. The tablet of claim 1, wherein the soluble fiber comprises maltodextrin. 16. The tablet of claim 15, wherein the tablet comprises at least 20 weight percent maltodextrin. 17. The tablet of claim 1, wherein the soluble fiber comprises a digestion resistant soluble fiber. 18. The tablet of claim 17, wherein the soluble fiber is a digestion resistant maltodextrin. 19. The tablet of claim 1, wherein the one or more sugar alcohols comprises is selected from the group consisting of mannitol, sorbitol, xylitol, erythritol, isomalt, lactitol, maltitol, maltitol syrup, and hydrogenated starch hydrolysates [HSH]. 20. The tablet of claim 1, wherein the one or more sugar alcohols comprise sorbitol. 21. The tablet of claim 1, wherein the one or more sugar alcohols comprises mannitol. 22. The tablet of claim 1, further comprising an antioxidant. 23. The tablet of claim 1, the tablet having a water content of between 0.5 weight percent and 7 weight percent. 24. The tablet of claim 1, wherein the tablet is disk shaped. 25. The tablet of claim 1, wherein the tablet is part of a sheet structure configured for subdivision into individual tablets. 26. The tablet of claim 1, wherein the one or more additives includes at least one additive selected from the group consisting of minerals, vitamins, dietary supplements, nutraceuticals, energizing agents, soothing agents, amino acids, chemesthic agents, antioxidants, botanicals, teeth whitening agents, therapeutic agents, and combinations thereof. 27. The tablet of claim 1, wherein the one or more additives includes at least one therapeutic agent selected from the group consisting of Gerd, Buprenorphin, Nitroglycerin, Diclofenac, Fentanyl, Carbamazepine, Galantamine, Acyclovir, Polyamidoamine Nanoparticles, Chlorpheniramine, Testosterone, Estradiol, Progesterone, Calcitonin, Fluorouracil, Naltrexone, Odansetron, Decitabine, Selegiline, Lamotrigine, Prochlorperazine, and combinations thereof. 28. The tablet of claim 1, further comprising a flavorant dispersed in the body such that the flavorant is released when the tablet is held or chewed within a mouth of a consumer, wherein the flavorant is selected from the group consisting of licorice, wintergreen, cherry and berry type flavorants, Dramboui, bourbon, scotch, whiskey, spearmint, peppermint, lavender, cinnamon, cardamon, apium graveolents, clove, cascarilla, nutmeg, sandalwood, bergamot, geranium, honey essence, rose oil, vanilla, lemon oil, orange oil, Japanese mint, cassia, caraway, cognac, jasmin, chamomile, menthol, ylang ylang, sage, fennel, piment, ginger, anise, chai, coriander, coffee, mint oils from a species of the genus Mentha, and combinations thereof. 29. The tablet of claim 1, wherein the one or more additives comprises a color additive. 30. The tablet of claim 1, wherein the solid solution comprises between 30 weight percent and 50 weight percent of soluble fiber and between 30 weight percent and 50 weight percent of the one or more sugar alcohols. 31. The tablet of claim 1, wherein the tablet is chewable. 32. A method of forming a dissolvable-chewable tablet:
forming a molten mixture of soluble fiber, one or more sugar alcohols, one or more additives, and water while maintaining a mixture temperature of less than 150° C., the molten mixture including at least 20 weight percent of soluble fiber, at least 20 weight percent of one or more sugar alcohols, and less than 15 weight percent water; and portioning the molten mixture into a plurality of dissolvable-chewable tablets. 33. The method of claim 32, wherein the molten mixture is formed in an extruder, wherein the extruder comprises multiple stages, further comprising mixing the molten mixture with a plasticizer prior to portioning the molten mixture into a plurality of tablets. 34. The method of claim 32, further comprising cutting the molten mixture into individual tablets sized to be at least partially received in an oral cavity of a consumer. 35. The method of claim 33, wherein the plasticizer is partially hydrogenated vegetable oil, propylene glycol, or a combination thereof. | 1,600 |
601 | 15,119,883 | 1,613 | The present invention relates to an aqueous co-formulation of metalaxyl, which contains:
i. metalaxyl, in particular metalaxyl-M; ii. at least one organic pesticide compound PC1, which has a solubility in water of at most 1 g/l at 20° C. and a melting point in the range from 40 to 100° C.; and iii. an aqueous phase containing water and at least one surfactant;
wherein the at least one organic pesticide compound PC1 is present in the form of particles suspended in the aqueous phase and where metalaxyl is essentially present dissolved in the aqueous phase and where the surfactant comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups.
The invention also relates to a process for preparing such formulations, which comprises the following steps:
a) providing a suspension of metalaxyl in an aqueous phase containing water, at least one surfactant, which comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups, b) treating the suspension until metalaxyl is essentially dissolved, to obtain an aqueous composition of metalaxyl, wherein metalaxyl is present in dissolved form; c) mixing the aqueous composition of metalaxyl obtained in step b) with an aqueous suspension of the further organic pesticide compound PC1. | 1-17. (canceled) 18. An aqueous co-formulation of metalaxyl comprising:
i. metalaxyl; ii. at least one organic pesticide compound PC1, which has a solubility in water of at most 1 g/l at 20° C. and a melting point in the range from 40 to 100° C.; and iii. an aqueous phase containing water and at least one surfactant; wherein the at least one organic pesticide compound PC1 is present in the form of particles suspended in the aqueous phase and where at least 95% of the metalaxyl present in the aqueous formulation is present dissolved in the aqueous phase and where the surfactant comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups. 19. The formulation of claim 18, wherein the oligomer or polymer having a plurality of arylsulfonyl groups is selected from the group consisting of the salts of naphthalinsulfonic acid formaldehyde condensates, phenolsulfonic acid formaldehyde condensates, naphthalinsulfonic acid urea formaldehyde condensates and phenolsulfonic acid formaldehyde urea condensates, and mixtures thereof. 20. The formulation of claim 18, wherein the surfactant further comprises an oligomeric or polymeric surfactant having at least one poly(C2-C4-alkylene oxide) group. 21. The formulation of claim 20, wherein the oligomeric or polymeric surfactant having at least one poly(C2-C4-alkylene oxide) group is selected from the group consisting of ethylenoxide-co-propylenoxide block copolymers, graft or comb polymers having a plurality of poly(C2-C4-alkylene oxide) side chains attached to a polymeric backbone of polymerized ethylenically unsaturated monomers, graft or comb polymers containing a poly-C2-C4-alkylene oxide backbone and polymeric side chains of polymerized ethylenically unsaturated monomers, and salts of the sulfates or phosphates of ethoxylated di- or tristyrylphenol. 22. The formulation of claim 21, wherein the oligomeric or polymeric surfactant having at least one poly(C2-C4-alkylene oxide) group comprises an ethylenoxide-co-propylenoxide block copolymer. 23. The formulation of claim 18, wherein the organic pesticide compound PC1 is selected from the group consisting of pyraclostrobin, imazalil, dodemorph acetate, pyrimethanil, difenoconazole, ipconazole, trifloxystrobin, fenoxanil, carboxin, metrafenone and acetamiprid, and mixtures thereof. 24. The formulation of claim 23, wherein the organic pesticide compound PC1 is pyraclostrobin. 25. The formulation of claim 18, further comprising:
i. 0.2 to 5% by weight, based on the total weight of the formulation, of metalaxyl; ii. 0.2 to 15% by weight, based on the total weight of the formulation, of the at least one organic pesticide compound PC1; iii. 0.2 to 10% by weight, based on the total weight of the formulation, of at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups, the total amount of surfactant being 0.5 to 20% by weight, based on the total weight of the formulation; and iv. at least 50% by weight, based on the total weight of the formulation, of water. 26. The formulation of claim 18, further comprising an organic pesticide compound PC2, having a melting point above 100° C. and having a water-solubility of at most 5 g/l at 20° C. 27. The formulation of claim 26, wherein the organic pesticide compound PC2 is selected from the group consisting of triticonazole, fluxapyroxad, boscalid, metconazole, dimethomorph, prochloraz, thiophanate-methyl, iprodione, epoxiconazole, fenpropimorph, chlorothalonil, fludioxonil, prothioconazole, tebuconazole, propiconazole, thiram, metiram, dithianon, mancozeb, dimoxystrobine, ametoctradin, fipronil, rynaxypyr, thiametoxam, clothianidin, thiacloprid, imidacloprid and dinotefuran, and mixtures thereof. 28. The formulation of claim 26, wherein the organic pesticide compound PC2 is selected from the group consisting of 2-[2-chloro-4-(4-chlorophenoxy)phenyl]-1-(1,2,4-triazol-1-yl)pent-3-yn-2-ol, 2-[4-(4-chlorophenoxy)-2-(trifluoromethyl)phenyl]-1-(1,2,4-triazol-1-yl)propan-2-ol and 2-[4-(4-chlorophenoxy)-2-(trifluoromethyl)phenyl]-3-methyl-1-(1,2,4-triazol-1-yl)butan-2-ol, and mixtures thereof. 29. The formulation of claim 26, wherein the concentration of the organic pesticide compound PC2 is from 0.1 to 25% by weight, in particular based on the weight of the formulation, 30. The formulation of claim 18, further comprising at least one aliphatic alcohol having at least one OH group. 31. The formulation of claim 18, further comprising at least one thickener. 32. The formulation of claim 31, wherein the at least one thickener is a polysaccharide based thickener. 33. A method for producing the aqueous co-formulation of metalaxyl as defined in claim 18, wherein the process comprises the following steps:
a) providing a suspension of metalaxyl in an aqueous phase containing water, at least one surfactant, which comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups; b) treating the suspension until metalaxyl is essentially dissolved, to obtain an aqueous composition of metalaxyl, wherein metalaxyl is present in dissolved form; and c) mixing the aqueous composition of metalaxyl obtained in step b) with an aqueous suspension of the further organic pesticide compound PC1. 34. The method of claim 33, wherein step b) comprises mixing a suspension of metalaxyl with an aqueous solution of the surfactant. | The present invention relates to an aqueous co-formulation of metalaxyl, which contains:
i. metalaxyl, in particular metalaxyl-M; ii. at least one organic pesticide compound PC1, which has a solubility in water of at most 1 g/l at 20° C. and a melting point in the range from 40 to 100° C.; and iii. an aqueous phase containing water and at least one surfactant;
wherein the at least one organic pesticide compound PC1 is present in the form of particles suspended in the aqueous phase and where metalaxyl is essentially present dissolved in the aqueous phase and where the surfactant comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups.
The invention also relates to a process for preparing such formulations, which comprises the following steps:
a) providing a suspension of metalaxyl in an aqueous phase containing water, at least one surfactant, which comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups, b) treating the suspension until metalaxyl is essentially dissolved, to obtain an aqueous composition of metalaxyl, wherein metalaxyl is present in dissolved form; c) mixing the aqueous composition of metalaxyl obtained in step b) with an aqueous suspension of the further organic pesticide compound PC1.1-17. (canceled) 18. An aqueous co-formulation of metalaxyl comprising:
i. metalaxyl; ii. at least one organic pesticide compound PC1, which has a solubility in water of at most 1 g/l at 20° C. and a melting point in the range from 40 to 100° C.; and iii. an aqueous phase containing water and at least one surfactant; wherein the at least one organic pesticide compound PC1 is present in the form of particles suspended in the aqueous phase and where at least 95% of the metalaxyl present in the aqueous formulation is present dissolved in the aqueous phase and where the surfactant comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups. 19. The formulation of claim 18, wherein the oligomer or polymer having a plurality of arylsulfonyl groups is selected from the group consisting of the salts of naphthalinsulfonic acid formaldehyde condensates, phenolsulfonic acid formaldehyde condensates, naphthalinsulfonic acid urea formaldehyde condensates and phenolsulfonic acid formaldehyde urea condensates, and mixtures thereof. 20. The formulation of claim 18, wherein the surfactant further comprises an oligomeric or polymeric surfactant having at least one poly(C2-C4-alkylene oxide) group. 21. The formulation of claim 20, wherein the oligomeric or polymeric surfactant having at least one poly(C2-C4-alkylene oxide) group is selected from the group consisting of ethylenoxide-co-propylenoxide block copolymers, graft or comb polymers having a plurality of poly(C2-C4-alkylene oxide) side chains attached to a polymeric backbone of polymerized ethylenically unsaturated monomers, graft or comb polymers containing a poly-C2-C4-alkylene oxide backbone and polymeric side chains of polymerized ethylenically unsaturated monomers, and salts of the sulfates or phosphates of ethoxylated di- or tristyrylphenol. 22. The formulation of claim 21, wherein the oligomeric or polymeric surfactant having at least one poly(C2-C4-alkylene oxide) group comprises an ethylenoxide-co-propylenoxide block copolymer. 23. The formulation of claim 18, wherein the organic pesticide compound PC1 is selected from the group consisting of pyraclostrobin, imazalil, dodemorph acetate, pyrimethanil, difenoconazole, ipconazole, trifloxystrobin, fenoxanil, carboxin, metrafenone and acetamiprid, and mixtures thereof. 24. The formulation of claim 23, wherein the organic pesticide compound PC1 is pyraclostrobin. 25. The formulation of claim 18, further comprising:
i. 0.2 to 5% by weight, based on the total weight of the formulation, of metalaxyl; ii. 0.2 to 15% by weight, based on the total weight of the formulation, of the at least one organic pesticide compound PC1; iii. 0.2 to 10% by weight, based on the total weight of the formulation, of at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups, the total amount of surfactant being 0.5 to 20% by weight, based on the total weight of the formulation; and iv. at least 50% by weight, based on the total weight of the formulation, of water. 26. The formulation of claim 18, further comprising an organic pesticide compound PC2, having a melting point above 100° C. and having a water-solubility of at most 5 g/l at 20° C. 27. The formulation of claim 26, wherein the organic pesticide compound PC2 is selected from the group consisting of triticonazole, fluxapyroxad, boscalid, metconazole, dimethomorph, prochloraz, thiophanate-methyl, iprodione, epoxiconazole, fenpropimorph, chlorothalonil, fludioxonil, prothioconazole, tebuconazole, propiconazole, thiram, metiram, dithianon, mancozeb, dimoxystrobine, ametoctradin, fipronil, rynaxypyr, thiametoxam, clothianidin, thiacloprid, imidacloprid and dinotefuran, and mixtures thereof. 28. The formulation of claim 26, wherein the organic pesticide compound PC2 is selected from the group consisting of 2-[2-chloro-4-(4-chlorophenoxy)phenyl]-1-(1,2,4-triazol-1-yl)pent-3-yn-2-ol, 2-[4-(4-chlorophenoxy)-2-(trifluoromethyl)phenyl]-1-(1,2,4-triazol-1-yl)propan-2-ol and 2-[4-(4-chlorophenoxy)-2-(trifluoromethyl)phenyl]-3-methyl-1-(1,2,4-triazol-1-yl)butan-2-ol, and mixtures thereof. 29. The formulation of claim 26, wherein the concentration of the organic pesticide compound PC2 is from 0.1 to 25% by weight, in particular based on the weight of the formulation, 30. The formulation of claim 18, further comprising at least one aliphatic alcohol having at least one OH group. 31. The formulation of claim 18, further comprising at least one thickener. 32. The formulation of claim 31, wherein the at least one thickener is a polysaccharide based thickener. 33. A method for producing the aqueous co-formulation of metalaxyl as defined in claim 18, wherein the process comprises the following steps:
a) providing a suspension of metalaxyl in an aqueous phase containing water, at least one surfactant, which comprises at least one salt of an oligomer or polymer having a plurality of arylsulfonyl groups; b) treating the suspension until metalaxyl is essentially dissolved, to obtain an aqueous composition of metalaxyl, wherein metalaxyl is present in dissolved form; and c) mixing the aqueous composition of metalaxyl obtained in step b) with an aqueous suspension of the further organic pesticide compound PC1. 34. The method of claim 33, wherein step b) comprises mixing a suspension of metalaxyl with an aqueous solution of the surfactant. | 1,600 |
602 | 15,169,498 | 1,634 | The present invention relates to methods for identifying variations that occur in the human genome and relating these variations to the genetic basis of disease and drug response. In particular, the present invention relates to identifying individual SNPs, determining SNP haplotype blocks and patterns, and, further, using the SNP haplotype blocks and patterns to dissect the genetic bases of disease and drug response. The methods of the present invention are useful in whole genome analysis. | 1-26. (canceled) 27. A system for genomic analysis to determine pharmacogenomic-related genetic loci without a priori knowledge of the location of said loci, the system comprising:
one or more databases that store information obtained from genotyping genomic DNA of individuals in a case population which comprises individuals who exhibit a response to a drug to determine frequency of one or more informative genetic variations in the case population and information obtained from genotyping genomic DNA of individuals in a control population which comprises individuals who do not exhibit the response to the drug to determine frequency of one or more informative genetic variations in the control population, wherein the one or more informative genetic variations distinguish one haplotype pattern from other haplotype patterns of a haplotype block; a server comprising:
one or more processors; and
a computer memory comprising processor-executable instructions that, when executed by the one or more processors, cause the one or more processors to perform the following:
access the one or more databases;
compare frequencies of the one or more informative genetic variations between the case population and the control population;
select the one or more informative genetic variations which have a frequency in the case population which is significantly different to the control population as pharmacogenomic-related genetic loci; and
provide a report to a client device in communication with the server across a communications network, wherein the report includes a description of the one or more informative genetic variations which have a frequency in the case population which is significantly different to the frequency in the control population as pharmacogenomic-related genetic loci. 28. The system of claim 27, wherein the one or more processors are configured to receive a request for the report from the client device. 29. The system of claim 27, wherein the one or more informative genetic variations are single-nucleotide polymorphisms. 30. A computer-implemented method of genomic analysis, the method implemented by a server in communication with a client device across a communications network, the server comprising one or more processors, the method comprising:
accessing, by the one or more processors, one or more databases that store information obtained from genotyping genomic DNA of individuals in a case population which comprises individuals who exhibit a response to a drug to determine frequency of one or more informative genetic variations in the case population and information obtained from genotyping genomic DNA of individuals in a control population which comprises individuals who do not exhibit the response to the drug to determine frequency of one or more informative genetic variations in the control population; wherein the one or more informative genetic variations distinguish one haplotype pattern from other haplotype patterns in a haplotype block; comparing, by the one or more processors, the determined frequency of the one or more informative genetic variations in the case population with the determined frequency of the one or more informative genetic variations in the control population; determining, by the one or more processors, and based on the comparison, differences in the frequency of the informative genetic variations between the case population and the control population; selecting, by the one or more processors, and based on the determined frequency differences, the one or more informative genetic variations having a frequency in the case population that is significantly different than a frequency in the control population; adding, by the one or more processors, the selected one or more informative genetic variations as pharmacogenomic-related genetic loci; and providing to the client device, by the one or more processors, a report including a description of the one or more informative genetic variations having a frequency in the case population that is significantly different than a frequency in the control population and an identification of the selected one or more informative genetic variations as pharmacogenomic-related genetic loci. 31. The computer-implemented method of claim 30, wherein the server is in communication with the database across the communications network. 32. The computer-implemented method of claim 30, wherein the server comprises the database. 33. The computer-implemented method of claim 30, wherein the server is configured to receive a request for the report from the client device. 34. The computer-implemented method of claim 30, wherein the one or more informative genetic variations are single-nucleotide polymorphisms. | The present invention relates to methods for identifying variations that occur in the human genome and relating these variations to the genetic basis of disease and drug response. In particular, the present invention relates to identifying individual SNPs, determining SNP haplotype blocks and patterns, and, further, using the SNP haplotype blocks and patterns to dissect the genetic bases of disease and drug response. The methods of the present invention are useful in whole genome analysis.1-26. (canceled) 27. A system for genomic analysis to determine pharmacogenomic-related genetic loci without a priori knowledge of the location of said loci, the system comprising:
one or more databases that store information obtained from genotyping genomic DNA of individuals in a case population which comprises individuals who exhibit a response to a drug to determine frequency of one or more informative genetic variations in the case population and information obtained from genotyping genomic DNA of individuals in a control population which comprises individuals who do not exhibit the response to the drug to determine frequency of one or more informative genetic variations in the control population, wherein the one or more informative genetic variations distinguish one haplotype pattern from other haplotype patterns of a haplotype block; a server comprising:
one or more processors; and
a computer memory comprising processor-executable instructions that, when executed by the one or more processors, cause the one or more processors to perform the following:
access the one or more databases;
compare frequencies of the one or more informative genetic variations between the case population and the control population;
select the one or more informative genetic variations which have a frequency in the case population which is significantly different to the control population as pharmacogenomic-related genetic loci; and
provide a report to a client device in communication with the server across a communications network, wherein the report includes a description of the one or more informative genetic variations which have a frequency in the case population which is significantly different to the frequency in the control population as pharmacogenomic-related genetic loci. 28. The system of claim 27, wherein the one or more processors are configured to receive a request for the report from the client device. 29. The system of claim 27, wherein the one or more informative genetic variations are single-nucleotide polymorphisms. 30. A computer-implemented method of genomic analysis, the method implemented by a server in communication with a client device across a communications network, the server comprising one or more processors, the method comprising:
accessing, by the one or more processors, one or more databases that store information obtained from genotyping genomic DNA of individuals in a case population which comprises individuals who exhibit a response to a drug to determine frequency of one or more informative genetic variations in the case population and information obtained from genotyping genomic DNA of individuals in a control population which comprises individuals who do not exhibit the response to the drug to determine frequency of one or more informative genetic variations in the control population; wherein the one or more informative genetic variations distinguish one haplotype pattern from other haplotype patterns in a haplotype block; comparing, by the one or more processors, the determined frequency of the one or more informative genetic variations in the case population with the determined frequency of the one or more informative genetic variations in the control population; determining, by the one or more processors, and based on the comparison, differences in the frequency of the informative genetic variations between the case population and the control population; selecting, by the one or more processors, and based on the determined frequency differences, the one or more informative genetic variations having a frequency in the case population that is significantly different than a frequency in the control population; adding, by the one or more processors, the selected one or more informative genetic variations as pharmacogenomic-related genetic loci; and providing to the client device, by the one or more processors, a report including a description of the one or more informative genetic variations having a frequency in the case population that is significantly different than a frequency in the control population and an identification of the selected one or more informative genetic variations as pharmacogenomic-related genetic loci. 31. The computer-implemented method of claim 30, wherein the server is in communication with the database across the communications network. 32. The computer-implemented method of claim 30, wherein the server comprises the database. 33. The computer-implemented method of claim 30, wherein the server is configured to receive a request for the report from the client device. 34. The computer-implemented method of claim 30, wherein the one or more informative genetic variations are single-nucleotide polymorphisms. | 1,600 |
603 | 15,104,698 | 1,617 | The invention relates to a pharmaceutical matrix tablet which can be administered orally once or twice per day with gastro-retentive controlled release of Baclofen. | 1. A matrix-type pharmaceutical tablet administrable by oral route once to twice per day with controlled release of baclofen by gastric retention, comprising granules of baclofen and one or more diluents within a matrix comprising at least a gelling agent, a gas-generating agent and a superdisintegrant. 2. The tablet according to claim 1, comprising baclofen granules and mannitol within a matrix comprising at least a gelling agent, a gas-generating agent and a superdisintegrant. 3. The tablet according to claim 1, comprising from 1 to 30% by weight of superdisintegrant relative to the total weight of the tablet. 4. The tablet according to claim 1, comprising from 5 to 50% by weight of gelling agent relative to the total weight of the tablet. 5. The tablet according to claim 1, comprising from 2 to 25% by weight of gas generator relative to the total weight of the tablet. 6. The tablet according to claim 5, comprising 5% by weight of gas generator relative to the total weight of the tablet. 7. The tablet according to claim 1, not comprising sodium alginate. 8. The tablet according to claim 1, wherein the gelling agent and the gas generator are in a weight ratio of gelling agent to gas generator of 1:10 to 10:1. 9. The tablet according to claim 1, comprising:
granules comprising baclofen and mannitol within a matrix comprising a superdisintegrant which is croscarmellose or crospovidone in proportions of between 3% and 8% by weight of the total weight of the tablet, a gas—generating agent which is sodium bicarbonate in proportions of between 4% and 8% by weight of the total weight of the tablet and a gelling agent which is either xanthan gum, Polyox™ or Methocel™ in proportions of between 10% and 30% by weight of the total weight of the tablet. 10. A method of treatment of alcohol dependence or of maintaining alcohol abstinence, comprising the administration once to twice per day of a tablet according to claim 1 to a patient in need thereof. 11. The tablet according to claim 2, comprising from 1 to 30% by weight of superdisintegrant relative to the total weight of the tablet. | The invention relates to a pharmaceutical matrix tablet which can be administered orally once or twice per day with gastro-retentive controlled release of Baclofen.1. A matrix-type pharmaceutical tablet administrable by oral route once to twice per day with controlled release of baclofen by gastric retention, comprising granules of baclofen and one or more diluents within a matrix comprising at least a gelling agent, a gas-generating agent and a superdisintegrant. 2. The tablet according to claim 1, comprising baclofen granules and mannitol within a matrix comprising at least a gelling agent, a gas-generating agent and a superdisintegrant. 3. The tablet according to claim 1, comprising from 1 to 30% by weight of superdisintegrant relative to the total weight of the tablet. 4. The tablet according to claim 1, comprising from 5 to 50% by weight of gelling agent relative to the total weight of the tablet. 5. The tablet according to claim 1, comprising from 2 to 25% by weight of gas generator relative to the total weight of the tablet. 6. The tablet according to claim 5, comprising 5% by weight of gas generator relative to the total weight of the tablet. 7. The tablet according to claim 1, not comprising sodium alginate. 8. The tablet according to claim 1, wherein the gelling agent and the gas generator are in a weight ratio of gelling agent to gas generator of 1:10 to 10:1. 9. The tablet according to claim 1, comprising:
granules comprising baclofen and mannitol within a matrix comprising a superdisintegrant which is croscarmellose or crospovidone in proportions of between 3% and 8% by weight of the total weight of the tablet, a gas—generating agent which is sodium bicarbonate in proportions of between 4% and 8% by weight of the total weight of the tablet and a gelling agent which is either xanthan gum, Polyox™ or Methocel™ in proportions of between 10% and 30% by weight of the total weight of the tablet. 10. A method of treatment of alcohol dependence or of maintaining alcohol abstinence, comprising the administration once to twice per day of a tablet according to claim 1 to a patient in need thereof. 11. The tablet according to claim 2, comprising from 1 to 30% by weight of superdisintegrant relative to the total weight of the tablet. | 1,600 |
604 | 13,000,241 | 1,611 | The present invention relates to a novel directly compressible matrix for the production of tablets which disintegrate rapidly in the presence of moisture, in particular in the mouth. | 1. Co-mixture for the production of rapidly disintegrating tablets in a direct tableting process, consisting of 90-98 parts by weight of a sprayed mannitol and 10-2 parts by weight of a crosslinked sodium carboxymethylcellulose and characterised in that the co-mixture has a BET surface area of greater than 1.5 m2/g. 2. Co-mixture for the production of rapidly disintegrating tablets in a direct tableting process according to Claim 1, consisting of 95 parts by weight of a sprayed mannitol and 5 parts by weight of a crosslinked sodium carboxymethylcellulose and characterised in that the co-mixture has a BET surface area of greater than 1.5 m2/g. 3. Co-mixture according to Claim 1, characterised in that the co-mixture has a BET surface area in the range from 1.9 to 4.0 m2/g. 4. Co-mixture according to claim 1, characterised in that the co-mixture has a BET surface area in the range from 1.9 to 2.6 m2/g. 5. Co-mixture according to claim 1, characterised by a bulk density of 0.45 to 0.60 g/ml, a tapped density of 0.60 to 0.75 g/ml, an angle of repose of 30 to 38°. 6. Co-mixture according to claim 1, characterised by an average particle diameter (laser) in the range between 60 and 200 μm. 7. Co-mixture according to claim 1, characterised by an average particle diameter (laser) in the range between 64 and 114 μm. 8. Co-mixture according to claim 1 having a water content<1% by weight. 9. Co-mixture according to claim 1, characterised in that it can be compressed to give tablets, and, after compression at a pressing force of 20 kN, tablets having hardnesses>250N, a friability≦0.14% and a disintegration time≦70 seconds are obtained. 10. Active compound- and/or aroma-containing tablet formulations prepared using a co-mixture according to one or more of claim 1 as excipient material. 11. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise vitamins, mineral substances, trace elements, functional food constituents. 12. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise plant constituents and plant extracts. 13. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise synthetic and natural dyes, natural and nature-identical aromas and other flavouring substances, such as, for example, sweeteners from the group aspartame, sachcharin, acesulfame K, neohesperidin DC, sucralose, thaumatin and stevioside, or fruit aromas, peppermint aromas, menthol, fruit acids from the group citric acid and tartaric acid or flavouring plant extracts. 14. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise substances having a pharmacological action, such as, for example, antacids, antiinfectives, also for local action in the mouth and throat area, analgesics, including opioids, antiallergics, antiemetics, antidiarrhoeal agents. | The present invention relates to a novel directly compressible matrix for the production of tablets which disintegrate rapidly in the presence of moisture, in particular in the mouth.1. Co-mixture for the production of rapidly disintegrating tablets in a direct tableting process, consisting of 90-98 parts by weight of a sprayed mannitol and 10-2 parts by weight of a crosslinked sodium carboxymethylcellulose and characterised in that the co-mixture has a BET surface area of greater than 1.5 m2/g. 2. Co-mixture for the production of rapidly disintegrating tablets in a direct tableting process according to Claim 1, consisting of 95 parts by weight of a sprayed mannitol and 5 parts by weight of a crosslinked sodium carboxymethylcellulose and characterised in that the co-mixture has a BET surface area of greater than 1.5 m2/g. 3. Co-mixture according to Claim 1, characterised in that the co-mixture has a BET surface area in the range from 1.9 to 4.0 m2/g. 4. Co-mixture according to claim 1, characterised in that the co-mixture has a BET surface area in the range from 1.9 to 2.6 m2/g. 5. Co-mixture according to claim 1, characterised by a bulk density of 0.45 to 0.60 g/ml, a tapped density of 0.60 to 0.75 g/ml, an angle of repose of 30 to 38°. 6. Co-mixture according to claim 1, characterised by an average particle diameter (laser) in the range between 60 and 200 μm. 7. Co-mixture according to claim 1, characterised by an average particle diameter (laser) in the range between 64 and 114 μm. 8. Co-mixture according to claim 1 having a water content<1% by weight. 9. Co-mixture according to claim 1, characterised in that it can be compressed to give tablets, and, after compression at a pressing force of 20 kN, tablets having hardnesses>250N, a friability≦0.14% and a disintegration time≦70 seconds are obtained. 10. Active compound- and/or aroma-containing tablet formulations prepared using a co-mixture according to one or more of claim 1 as excipient material. 11. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise vitamins, mineral substances, trace elements, functional food constituents. 12. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise plant constituents and plant extracts. 13. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise synthetic and natural dyes, natural and nature-identical aromas and other flavouring substances, such as, for example, sweeteners from the group aspartame, sachcharin, acesulfame K, neohesperidin DC, sucralose, thaumatin and stevioside, or fruit aromas, peppermint aromas, menthol, fruit acids from the group citric acid and tartaric acid or flavouring plant extracts. 14. Use of an excipient material according to claim 1 for the preparation of tablet formulations which comprise substances having a pharmacological action, such as, for example, antacids, antiinfectives, also for local action in the mouth and throat area, analgesics, including opioids, antiallergics, antiemetics, antidiarrhoeal agents. | 1,600 |
605 | 14,291,401 | 1,611 | Disclosed is a non-aqueous hair care composition comprising: a base oil being a volatile isoparaffin; a silicone elastomer; a solubilizer for the silicone elastomer which is a non-volatile silicone having a viscosity from about 25 to about 100,000 mm 2 ·s −1 . The composition of the present invention has improved stability such as reduced phase separation at lower temperature, and also has a balanced dry performance. | 1. A non-aqueous hair care composition comprising by weight:
(a) from about 50% to about 99.9% of a base oil being a volatile isoparaffin; (b) from about 0.025% to about 15% of a silicone elastomer; and (c) from about 1% to about 40% of a solubilizer for the silicone elastomer which is a non-volatile silicone having a viscosity from about 25 to about 100,000 mm2·s−1. 2. The composition of claim 1 wherein the non-volatile silicones having a viscosity from about 50 to about 50,000 mm2·s−1. 3. The composition of claim 1 wherein the non-volatile silicones having a viscosity from about 100 to about 30,000 mm2·s−1. 4. The composition of claim 1 wherein the non-volatile silicones having a viscosity from about 7,000 to about 20,000 mm2·s−1. 5. The composition of claim 1 wherein the non-volatile silicones is a non-volatile non-amino modified silicone. 6. The composition of claim 1 wherein the non-volatile silicones is polydimethylsiloxane. 7. The composition of claim 1 wherein the weight ratio of the solubilizer to the silicone elastomer is from about 1:1 to about 30:1. 8. The composition of claim 1 wherein the weight ratio of the solubilizer to the silicone elastomer is from about 2:1 to about 20:1. 9. The composition of claim 1 wherein the weight ratio of the solubilizer to the silicone elastomer is from about 3:1 to about 10:1. 10. The composition of claim 1 wherein the composition is substantially free of surfactants. 11. The composition of claim 1 wherein the composition is for leave-on use. | Disclosed is a non-aqueous hair care composition comprising: a base oil being a volatile isoparaffin; a silicone elastomer; a solubilizer for the silicone elastomer which is a non-volatile silicone having a viscosity from about 25 to about 100,000 mm 2 ·s −1 . The composition of the present invention has improved stability such as reduced phase separation at lower temperature, and also has a balanced dry performance.1. A non-aqueous hair care composition comprising by weight:
(a) from about 50% to about 99.9% of a base oil being a volatile isoparaffin; (b) from about 0.025% to about 15% of a silicone elastomer; and (c) from about 1% to about 40% of a solubilizer for the silicone elastomer which is a non-volatile silicone having a viscosity from about 25 to about 100,000 mm2·s−1. 2. The composition of claim 1 wherein the non-volatile silicones having a viscosity from about 50 to about 50,000 mm2·s−1. 3. The composition of claim 1 wherein the non-volatile silicones having a viscosity from about 100 to about 30,000 mm2·s−1. 4. The composition of claim 1 wherein the non-volatile silicones having a viscosity from about 7,000 to about 20,000 mm2·s−1. 5. The composition of claim 1 wherein the non-volatile silicones is a non-volatile non-amino modified silicone. 6. The composition of claim 1 wherein the non-volatile silicones is polydimethylsiloxane. 7. The composition of claim 1 wherein the weight ratio of the solubilizer to the silicone elastomer is from about 1:1 to about 30:1. 8. The composition of claim 1 wherein the weight ratio of the solubilizer to the silicone elastomer is from about 2:1 to about 20:1. 9. The composition of claim 1 wherein the weight ratio of the solubilizer to the silicone elastomer is from about 3:1 to about 10:1. 10. The composition of claim 1 wherein the composition is substantially free of surfactants. 11. The composition of claim 1 wherein the composition is for leave-on use. | 1,600 |
606 | 15,627,415 | 1,653 | The biological functionality of living microbial spores is modified using phenotypic engineering to endow the resulting modified spores with novel functionality that extends the usefulness of the spores for a variety of practical applications including, for example, sterility testing, the release of active compounds, and cell-based biosensing systems. An embodiment entails engineering Bacillus spores to acquire synthetic new functions that enable the modified spores to sense and rapidly transduce specific germination signals in their surroundings. The newly acquired functions allow the spores to perform, for example, as self-reporters of cellular viability, self-indicating components of cell-based biosensors, and in other analytical systems. Also disclosed are methods for testing adequate sterility of a system by using engineered spores. | 1. A method of using engineered spores to test adequacy of a sterilization process for a system, comprising:
a) introducing the engineered spores into the system; b) sterilizing the system, wherein the system with the engineered spores is subjected to the sterilization process; c) exposing the engineered spores to a germinant for a predetermined germination period; d) measuring fluorescence, wherein the fluorescence of the engineered spores is measured to obtain a fluorescence measurement; and e) determining sterilization adequacy, such that:
if the fluorescence measurement is above a predetermined zero-baseline value, the sterilization process is determined to be inadequate, and
if the fluorescence measurement is equal to or less than the predetermined zero-baseline level, the sterilization process is determined to be adequate;
wherein the engineered spores each comprise: a first spore; and an at least partially hydrophobic compound, which is incorporated into the first spore; wherein the at least partially hydrophobic compound is fluorogenic, such that the at least partially hydrophobic compound is configured to become fluorescent by hydrolysis; wherein the sole fluorogenic compound in the engineered spores is the at least partially hydrophobic compound that is incorporated into the first spore in each of the engineered spores; wherein the engineered spore is configured to be capable of germination; and wherein the engineered spore is non-fluorescent; wherein the engineered spore does not comprise a germinant; such that the engineered spore is configured to become fluorescent upon germination. 2. The method of using engineered spores of claim 1, wherein the sterilization process is dry heat sterilization, such that sterilizing the system comprises exposing the system with the engineered spores to dry heat in a temperature range of 140-160 degrees Celsius. 3. The method of using engineered spores of claim 1, wherein the sterilization process is steam heat sterilization, such that sterilizing the system comprises exposing the system with the engineered spores to steam heat. 4. The method of using engineered spores of claim 1, wherein the first spore is selected from the group consisting of bacteria, fungi, plants, and yeast. 5. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is an entirely hydrophobic compound. 6. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is an amphiphilic compound. 7. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is dipropionylfluorescein. 8. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is diacetyl fluorescein. 9. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is dibutyryl fluorescein. 10. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is SYTO 9. 11. The method of using engineered spores of claim 1, wherein the first spore is a spore of Geobacillus stearothermophilus. 12. The method of using engineered spores of claim 1, wherein the first spore is a spore of Bacillus cereus. 13. The method of using engineered spores of claim 1, wherein the first spore is a spore of Bacillus atrophaeus. 14. The method of using engineered spores of claim 1, wherein the first spore is a spore of Bacillus megaterium. 15. A sterilization testing method for testing the adequacy of a sterilization process by using an engineered spore suspension, comprising:
a) sterilizing the engineered spore suspension, wherein the engineered spore suspension is subjected to the sterilization process; b) incorporating a germinant into the engineered spore suspension; c) waiting for a predetermined germination period; d) measuring fluorescence, wherein the fluorescence of the engineered spore suspension is measured to obtain a fluorescence measurement; and e) determining sterilization adequacy, such that:
if the fluorescence measurement is above a predetermined zero-baseline value, the sterilization process is determined to be inadequate, and
if the fluorescence measurement is equal to or less than the predetermined zero-baseline level, the sterilization process is determined to be adequate;
wherein the engineered spore suspension comprises: a sterile liquid solution; and a plurality of engineered spores, wherein each engineered spore comprises:
a first spore; and
an at least partially hydrophobic compound, which is incorporated into the first spore;
wherein the plurality of engineered spores is suspended in the sterile liquid solution; wherein the at least partially hydrophobic compound is fluorogenic, such that the at least partially hydrophobic compound is configured to become fluorescent by hydrolysis; wherein the sole fluorogenic compound in the engineered spore suspension is the at least partially hydrophobic compound that is incorporated into the first spore of each engineered spore; wherein the engineered spores are configured to be capable of germination; and wherein the engineered spores are non-fluorescent; wherein the engineered spore suspension does not comprise a germinant; such that the engineered spores are configured to become fluorescent upon germination. 16. The sterilization testing method of claim 15, wherein the sterilization process is dry heat sterilization, such that sterilizing the engineered spore suspension comprises exposing the engineered spore suspension to dry heat in a temperature range of 140-160 degrees Celsius. 17. The sterilization testing method of claim 15, wherein the sterilization process is steam heat sterilization, such that sterilizing the engineered spore suspension comprises exposing the engineered spore suspension to steam heat. 18. The sterilization testing method of claim 15, wherein the first spore is selected from the group consisting of bacteria, fungi, plants, and yeast. 19. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is dipropionylfluorescein. 20. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is diacetyl fluorescein. 21. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is dibutyryl fluorescein. 22. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is SYTO 9. 23. The sterilization testing method of claim 15, wherein the first spore is a spore of Geobacillus stearothermophilus. 24. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus cereus. 25. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus atrophaeus. 26. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus megaterium. 27. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus globigii. 28. The sterilization testing method of claim 15, wherein the sterile liquid solution is sterile distilled water. 29. A method of using engineered spores to test adequate sterility of a system, comprising sterilizing the system together with the engineered spores, subsequently incubating the engineered spores with a germinant, and finally measuring fluorescence of the engineered spores;
wherein the engineered spores each comprise:
a first spore; and
an at least partially hydrophobic compound, which is incorporated into the first spore;
wherein the at least partially hydrophobic compound is fluorogenic, such that the at least partially hydrophobic compound is configured to become fluorescent by hydrolysis; wherein the sole fluorogenic compound in the engineered spores is the at least partially hydrophobic compound that is incorporated into the first spore in each of the engineered spores; wherein the engineered spore is configured to be capable of germination; and wherein the engineered spore is non-fluorescent; wherein the engineered spore does not comprise a germinant; such that the engineered spore is configured to become fluorescent upon germination. | The biological functionality of living microbial spores is modified using phenotypic engineering to endow the resulting modified spores with novel functionality that extends the usefulness of the spores for a variety of practical applications including, for example, sterility testing, the release of active compounds, and cell-based biosensing systems. An embodiment entails engineering Bacillus spores to acquire synthetic new functions that enable the modified spores to sense and rapidly transduce specific germination signals in their surroundings. The newly acquired functions allow the spores to perform, for example, as self-reporters of cellular viability, self-indicating components of cell-based biosensors, and in other analytical systems. Also disclosed are methods for testing adequate sterility of a system by using engineered spores.1. A method of using engineered spores to test adequacy of a sterilization process for a system, comprising:
a) introducing the engineered spores into the system; b) sterilizing the system, wherein the system with the engineered spores is subjected to the sterilization process; c) exposing the engineered spores to a germinant for a predetermined germination period; d) measuring fluorescence, wherein the fluorescence of the engineered spores is measured to obtain a fluorescence measurement; and e) determining sterilization adequacy, such that:
if the fluorescence measurement is above a predetermined zero-baseline value, the sterilization process is determined to be inadequate, and
if the fluorescence measurement is equal to or less than the predetermined zero-baseline level, the sterilization process is determined to be adequate;
wherein the engineered spores each comprise: a first spore; and an at least partially hydrophobic compound, which is incorporated into the first spore; wherein the at least partially hydrophobic compound is fluorogenic, such that the at least partially hydrophobic compound is configured to become fluorescent by hydrolysis; wherein the sole fluorogenic compound in the engineered spores is the at least partially hydrophobic compound that is incorporated into the first spore in each of the engineered spores; wherein the engineered spore is configured to be capable of germination; and wherein the engineered spore is non-fluorescent; wherein the engineered spore does not comprise a germinant; such that the engineered spore is configured to become fluorescent upon germination. 2. The method of using engineered spores of claim 1, wherein the sterilization process is dry heat sterilization, such that sterilizing the system comprises exposing the system with the engineered spores to dry heat in a temperature range of 140-160 degrees Celsius. 3. The method of using engineered spores of claim 1, wherein the sterilization process is steam heat sterilization, such that sterilizing the system comprises exposing the system with the engineered spores to steam heat. 4. The method of using engineered spores of claim 1, wherein the first spore is selected from the group consisting of bacteria, fungi, plants, and yeast. 5. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is an entirely hydrophobic compound. 6. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is an amphiphilic compound. 7. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is dipropionylfluorescein. 8. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is diacetyl fluorescein. 9. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is dibutyryl fluorescein. 10. The method of using engineered spores of claim 1, wherein the at least partially hydrophobic compound is SYTO 9. 11. The method of using engineered spores of claim 1, wherein the first spore is a spore of Geobacillus stearothermophilus. 12. The method of using engineered spores of claim 1, wherein the first spore is a spore of Bacillus cereus. 13. The method of using engineered spores of claim 1, wherein the first spore is a spore of Bacillus atrophaeus. 14. The method of using engineered spores of claim 1, wherein the first spore is a spore of Bacillus megaterium. 15. A sterilization testing method for testing the adequacy of a sterilization process by using an engineered spore suspension, comprising:
a) sterilizing the engineered spore suspension, wherein the engineered spore suspension is subjected to the sterilization process; b) incorporating a germinant into the engineered spore suspension; c) waiting for a predetermined germination period; d) measuring fluorescence, wherein the fluorescence of the engineered spore suspension is measured to obtain a fluorescence measurement; and e) determining sterilization adequacy, such that:
if the fluorescence measurement is above a predetermined zero-baseline value, the sterilization process is determined to be inadequate, and
if the fluorescence measurement is equal to or less than the predetermined zero-baseline level, the sterilization process is determined to be adequate;
wherein the engineered spore suspension comprises: a sterile liquid solution; and a plurality of engineered spores, wherein each engineered spore comprises:
a first spore; and
an at least partially hydrophobic compound, which is incorporated into the first spore;
wherein the plurality of engineered spores is suspended in the sterile liquid solution; wherein the at least partially hydrophobic compound is fluorogenic, such that the at least partially hydrophobic compound is configured to become fluorescent by hydrolysis; wherein the sole fluorogenic compound in the engineered spore suspension is the at least partially hydrophobic compound that is incorporated into the first spore of each engineered spore; wherein the engineered spores are configured to be capable of germination; and wherein the engineered spores are non-fluorescent; wherein the engineered spore suspension does not comprise a germinant; such that the engineered spores are configured to become fluorescent upon germination. 16. The sterilization testing method of claim 15, wherein the sterilization process is dry heat sterilization, such that sterilizing the engineered spore suspension comprises exposing the engineered spore suspension to dry heat in a temperature range of 140-160 degrees Celsius. 17. The sterilization testing method of claim 15, wherein the sterilization process is steam heat sterilization, such that sterilizing the engineered spore suspension comprises exposing the engineered spore suspension to steam heat. 18. The sterilization testing method of claim 15, wherein the first spore is selected from the group consisting of bacteria, fungi, plants, and yeast. 19. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is dipropionylfluorescein. 20. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is diacetyl fluorescein. 21. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is dibutyryl fluorescein. 22. The sterilization testing method of claim 15, wherein the at least partially hydrophobic compound is SYTO 9. 23. The sterilization testing method of claim 15, wherein the first spore is a spore of Geobacillus stearothermophilus. 24. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus cereus. 25. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus atrophaeus. 26. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus megaterium. 27. The sterilization testing method of claim 15, wherein the first spore is a spore of Bacillus globigii. 28. The sterilization testing method of claim 15, wherein the sterile liquid solution is sterile distilled water. 29. A method of using engineered spores to test adequate sterility of a system, comprising sterilizing the system together with the engineered spores, subsequently incubating the engineered spores with a germinant, and finally measuring fluorescence of the engineered spores;
wherein the engineered spores each comprise:
a first spore; and
an at least partially hydrophobic compound, which is incorporated into the first spore;
wherein the at least partially hydrophobic compound is fluorogenic, such that the at least partially hydrophobic compound is configured to become fluorescent by hydrolysis; wherein the sole fluorogenic compound in the engineered spores is the at least partially hydrophobic compound that is incorporated into the first spore in each of the engineered spores; wherein the engineered spore is configured to be capable of germination; and wherein the engineered spore is non-fluorescent; wherein the engineered spore does not comprise a germinant; such that the engineered spore is configured to become fluorescent upon germination. | 1,600 |
607 | 15,754,598 | 1,648 | A process for assaying viral vector manufactured by large-scale viral vector manufacturing processes to assure the resulting vector has acceptable purity and potency. The process entails three different types of assays, each one of which is optionally useful on a stand-alone basis, and which together provide the first system able to assure the quality of viral vector produced by large-scale vector manufacturing processes. | 1. An assay to determine the infectivity of a recombinant viral vector, the assay comprising:
a. obtaining a recombinant viral vector; and b. obtaining cells that support replication of said viral vector and then dividing said cells into a first cell culture and a second cell culture; and then c. contacting each of said first cell culture and said second cell culture with said recombinant viral vector, whereby the vector particles per cell contacted to said first cell culture is different from the vector particles per cell contacted to said second cell culture; and then d. determining the percentage of cells in each cell culture which have been infected with the viral vector; and then e. calculating a linear response curve to measure relative infectivity using the Slope Ratio method. 2. The assay of claim 1, where said determining the percentage of infected cells in the cell cultures involves flow cytometry. 3. The assay of claim 1, where said determining the percentage of infected cells in the cell cultures involves fluorescence activated cell sorting. 4. The assay of claim 1, where said determining the percentage of infected cells in the cell cultures involves utilizing an antibody against a viral vector antigen. 5. The assay of claim 1, where the number of particles per cell in said first cell culture and the number of particles per cell in said second cell culture are selected to provide a linear response curve for viral vector infectivity. 6. The assay of claim 1,
step b further comprising dividing said cells into a negative control cell culture. 7. The assay of claim 1,
step b further comprising dividing said cells into a reference standard control cell culture, and step c further comprising contacting the reference standard control cell culture with a reference standard virus which is different from the recombinant viral vector. 8. The assay of claim 1,
step b further comprising dividing said cells into a positive control cell culture, and step c further comprising contacting the positive control cell culture with a different manufacturing lot of the recombinant viral vector. 9. The assay of claim 1, wherein the cell cultures are seeded to produce, at time of contacting the cell cultures with the recombinant viral vector, a cell density of about 80%. 10. A recombinant viral vector manufactured by the process comprising:
a. from a batch of recombinant viral vector, removing a sample; and then b. assaying the sample using the assay of claim 1; and then c. if the infectivity of the sample is not acceptable, then discarding said batch of recombinant viral vector. 11. The recombinant vector of claim 10, the process further comprising:
d. if the infectivity of the sample is acceptable, then packaging and releasing said batch of recombinant viral vector. 12. A process for manufacturing a viral vector having a transgene, comprising: obtaining a sample, and then assaying said sample by a. measuring viral vector infectious titer by calculating a linear response curve to measure relative infectivity using the Slope Ratio method; and using at least two assays selected from the group consisting of: b. measuring transgene expression, c. measuring the activity of the polypeptide expressed by said transgene, and d. measuring viral particle titer. 13. The process of claim 12, wherein said assaying said sample comprises a. measuring viral vector infectious titer by calculating a linear response curve to measure relative infectivity using the Slope Ratio method, b. measuring transgene expression and c. measuring the activity of the polypeptide expressed by said transgene. 14. The process of claim 12, wherein said assaying said sample comprises d. measuring viral particle titer. 15. The process of claim 12, wherein said assaying said sample comprises: a. measuring viral vector infectious titer, b. measuring transgene expression, c. measuring the activity of the polypeptide expressed by said transgene, and d. measuring viral particle titer. 16. A viral vector produced by the manufacturing process of claim 12. 17. A viral vector produced by the manufacturing process of claim 13. 18. A viral vector produced by the manufacturing process of claim 14. 19. A viral vector produced by the manufacturing process of claim 15. 20.-34. (canceled) | A process for assaying viral vector manufactured by large-scale viral vector manufacturing processes to assure the resulting vector has acceptable purity and potency. The process entails three different types of assays, each one of which is optionally useful on a stand-alone basis, and which together provide the first system able to assure the quality of viral vector produced by large-scale vector manufacturing processes.1. An assay to determine the infectivity of a recombinant viral vector, the assay comprising:
a. obtaining a recombinant viral vector; and b. obtaining cells that support replication of said viral vector and then dividing said cells into a first cell culture and a second cell culture; and then c. contacting each of said first cell culture and said second cell culture with said recombinant viral vector, whereby the vector particles per cell contacted to said first cell culture is different from the vector particles per cell contacted to said second cell culture; and then d. determining the percentage of cells in each cell culture which have been infected with the viral vector; and then e. calculating a linear response curve to measure relative infectivity using the Slope Ratio method. 2. The assay of claim 1, where said determining the percentage of infected cells in the cell cultures involves flow cytometry. 3. The assay of claim 1, where said determining the percentage of infected cells in the cell cultures involves fluorescence activated cell sorting. 4. The assay of claim 1, where said determining the percentage of infected cells in the cell cultures involves utilizing an antibody against a viral vector antigen. 5. The assay of claim 1, where the number of particles per cell in said first cell culture and the number of particles per cell in said second cell culture are selected to provide a linear response curve for viral vector infectivity. 6. The assay of claim 1,
step b further comprising dividing said cells into a negative control cell culture. 7. The assay of claim 1,
step b further comprising dividing said cells into a reference standard control cell culture, and step c further comprising contacting the reference standard control cell culture with a reference standard virus which is different from the recombinant viral vector. 8. The assay of claim 1,
step b further comprising dividing said cells into a positive control cell culture, and step c further comprising contacting the positive control cell culture with a different manufacturing lot of the recombinant viral vector. 9. The assay of claim 1, wherein the cell cultures are seeded to produce, at time of contacting the cell cultures with the recombinant viral vector, a cell density of about 80%. 10. A recombinant viral vector manufactured by the process comprising:
a. from a batch of recombinant viral vector, removing a sample; and then b. assaying the sample using the assay of claim 1; and then c. if the infectivity of the sample is not acceptable, then discarding said batch of recombinant viral vector. 11. The recombinant vector of claim 10, the process further comprising:
d. if the infectivity of the sample is acceptable, then packaging and releasing said batch of recombinant viral vector. 12. A process for manufacturing a viral vector having a transgene, comprising: obtaining a sample, and then assaying said sample by a. measuring viral vector infectious titer by calculating a linear response curve to measure relative infectivity using the Slope Ratio method; and using at least two assays selected from the group consisting of: b. measuring transgene expression, c. measuring the activity of the polypeptide expressed by said transgene, and d. measuring viral particle titer. 13. The process of claim 12, wherein said assaying said sample comprises a. measuring viral vector infectious titer by calculating a linear response curve to measure relative infectivity using the Slope Ratio method, b. measuring transgene expression and c. measuring the activity of the polypeptide expressed by said transgene. 14. The process of claim 12, wherein said assaying said sample comprises d. measuring viral particle titer. 15. The process of claim 12, wherein said assaying said sample comprises: a. measuring viral vector infectious titer, b. measuring transgene expression, c. measuring the activity of the polypeptide expressed by said transgene, and d. measuring viral particle titer. 16. A viral vector produced by the manufacturing process of claim 12. 17. A viral vector produced by the manufacturing process of claim 13. 18. A viral vector produced by the manufacturing process of claim 14. 19. A viral vector produced by the manufacturing process of claim 15. 20.-34. (canceled) | 1,600 |
608 | 14,379,613 | 1,613 | Compositions comprising a mixture of at least two types of particles wherein a) the first type of particles comprise dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof; and b) the second type of particles comprise at least one pharmaceutically acceptable organic acid, use of said compositions in the reduction of the risk of stroke and systemic embolism in patients with non-valvular atrial fibrillation and/or in the prevention of venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery and processes for the preparation of said compositions. | 1-20. (canceled) 21. A composition comprising a mixture of at least two types of particles wherein a) the first type of particles comprise dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof; and b) the second type of particles comprise at least one pharmaceutically acceptable organic acid. 22. The composition according to claim 21, wherein at least one type of particles is coated with a protective coating layer. 23. The composition according to claim 21, further comprising at least one pharmaceutically acceptable excipient. 24. The composition according to claim 21, wherein the first type of particles is free from acids. 25. The composition according to claim 21, wherein the second type of particles is free from dabigatran etexilate. 26. The composition according to claim 21, comprising from 0.01 wt % to 90 wt % of dabigatran etexilate (expressed as dabigatran etexilate mesylate). 27. The composition according to claim 21, wherein at least 90% by weight of the organic acid present in the composition is contained in the second type of particles and the rest (if any) of the organic acid forms part of the pharmaceutically acceptable excipients. 28. The composition according to claim 21, wherein the first type of particles is coated with a protective coating layer. 29. The composition according to claim 21, wherein the second type of particles is coated with a protective coating layer. 30. The composition according to claim 21, comprising from 2 wt % to 95 wt % of the at least one pharmaceutically acceptable organic acid. 31. The composition according to claim 21, wherein at least 90% by weight of the organic acid present in the composition is contained in the second type of particles and the rest (if any) of the organic acid forms part of the excipients. 32. A unit dosage form prepared from the composition according to claim 21, which comprises from 50 mg to 200 mg of dabigatran etexilate mesylate. 33. A method for reducing the risk of stroke and systemic embolism in patients with non-valvular atrial fibrillation and/or preventing venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery, comprising administering to the patient in need thereof a composition according to claim 21. 34. A method for reducing the risk of stroke and systemic embolism in patients with non-valvular atrial fibrillation and/or preventing venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery, comprising administering to the patient in need thereof a unit dose according to claim 32. 35. A process for the preparation of the composition according to claim 23, comprising the step of mixing the first type of particles and the second type of particles with the at least one at least one pharmaceutically acceptable excipient. 36. The process of claim 35, wherein the first type of particles are prepared by granulation. 37. The process according to claim 36, comprising the steps of:
i) blending dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof and at least one pharmaceutically acceptable excipient; ii) granulating the blend of step (i) with a binder solution to form granules of dabigatran etexilate; iii) blending at least one organic acid and at least one pharmaceutically acceptable excipient; iv) granulating the blend of step (iii) with a binder solution to form organic acid granules; v) coating the organic acid granules with a protective coating layer; vi) blending the granules of step (ii) with the coated granules of step (v) to form a mixture of at least two types of granules; vii) optionally blending the mixture of at least two types of granules of step (vi) with at least one pharmaceutically acceptable excipient; viii) adding a lubricant to the blend of step (vii); ix) filling the lubricated mixture of step (viii) into suitable hard capsules. 38. The process according to claim 36, comprising the steps of:
i) blending dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof and at least one pharmaceutically acceptable excipient; ii) granulating the blend of step (i) with a binder solution to form granules of dabigatran etexilate; iii) blending at least one organic acid and at least one pharmaceutically acceptable excipient; iv) granulating the blend of step (iii) with a binder solution to form organic acid granules; v) coating the granules of dabigatran etexilate with a protective coating layer; vi) blending the granules of step (iv) with the coated granule of step (v) to form a mixture of at least two types of granules; vii) optionally blending the mixture of at least two types of granules of step (vi) with at least one pharmaceutically acceptable excipient; viii) adding a lubricant to the blend of step (vii); ix) filling the lubricated mixture of step (viii) into suitable hard capsules. 39. The process according to claim 36, comprising the steps of:
blending dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof and at least one pharmaceutically acceptable excipient; ii) granulating the blend of step (i) with a binder solution to form granules of the dabigatran etexilate; iii) blending at least one organic acid and at least one pharmaceutically acceptable excipient; iv) extruding and a heron zing the blend of step (iii) to form organic acid pellets; v) coating the organic acid pellets of step (iv) with a protective coating layer; vi) blending the granules of step (ii) with the coated pellets of step (v) to form a mixture of at least two types of particles; vii) optionally blending the mixture of at least two types of granules of step (vi) with at least one pharmaceutically acceptable excipient; viii) adding a lubricant to the blend of step (vii); ix) filling the lubricated mixture of step (viii) into suitable hard capsules. | Compositions comprising a mixture of at least two types of particles wherein a) the first type of particles comprise dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof; and b) the second type of particles comprise at least one pharmaceutically acceptable organic acid, use of said compositions in the reduction of the risk of stroke and systemic embolism in patients with non-valvular atrial fibrillation and/or in the prevention of venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery and processes for the preparation of said compositions.1-20. (canceled) 21. A composition comprising a mixture of at least two types of particles wherein a) the first type of particles comprise dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof; and b) the second type of particles comprise at least one pharmaceutically acceptable organic acid. 22. The composition according to claim 21, wherein at least one type of particles is coated with a protective coating layer. 23. The composition according to claim 21, further comprising at least one pharmaceutically acceptable excipient. 24. The composition according to claim 21, wherein the first type of particles is free from acids. 25. The composition according to claim 21, wherein the second type of particles is free from dabigatran etexilate. 26. The composition according to claim 21, comprising from 0.01 wt % to 90 wt % of dabigatran etexilate (expressed as dabigatran etexilate mesylate). 27. The composition according to claim 21, wherein at least 90% by weight of the organic acid present in the composition is contained in the second type of particles and the rest (if any) of the organic acid forms part of the pharmaceutically acceptable excipients. 28. The composition according to claim 21, wherein the first type of particles is coated with a protective coating layer. 29. The composition according to claim 21, wherein the second type of particles is coated with a protective coating layer. 30. The composition according to claim 21, comprising from 2 wt % to 95 wt % of the at least one pharmaceutically acceptable organic acid. 31. The composition according to claim 21, wherein at least 90% by weight of the organic acid present in the composition is contained in the second type of particles and the rest (if any) of the organic acid forms part of the excipients. 32. A unit dosage form prepared from the composition according to claim 21, which comprises from 50 mg to 200 mg of dabigatran etexilate mesylate. 33. A method for reducing the risk of stroke and systemic embolism in patients with non-valvular atrial fibrillation and/or preventing venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery, comprising administering to the patient in need thereof a composition according to claim 21. 34. A method for reducing the risk of stroke and systemic embolism in patients with non-valvular atrial fibrillation and/or preventing venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery, comprising administering to the patient in need thereof a unit dose according to claim 32. 35. A process for the preparation of the composition according to claim 23, comprising the step of mixing the first type of particles and the second type of particles with the at least one at least one pharmaceutically acceptable excipient. 36. The process of claim 35, wherein the first type of particles are prepared by granulation. 37. The process according to claim 36, comprising the steps of:
i) blending dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof and at least one pharmaceutically acceptable excipient; ii) granulating the blend of step (i) with a binder solution to form granules of dabigatran etexilate; iii) blending at least one organic acid and at least one pharmaceutically acceptable excipient; iv) granulating the blend of step (iii) with a binder solution to form organic acid granules; v) coating the organic acid granules with a protective coating layer; vi) blending the granules of step (ii) with the coated granules of step (v) to form a mixture of at least two types of granules; vii) optionally blending the mixture of at least two types of granules of step (vi) with at least one pharmaceutically acceptable excipient; viii) adding a lubricant to the blend of step (vii); ix) filling the lubricated mixture of step (viii) into suitable hard capsules. 38. The process according to claim 36, comprising the steps of:
i) blending dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof and at least one pharmaceutically acceptable excipient; ii) granulating the blend of step (i) with a binder solution to form granules of dabigatran etexilate; iii) blending at least one organic acid and at least one pharmaceutically acceptable excipient; iv) granulating the blend of step (iii) with a binder solution to form organic acid granules; v) coating the granules of dabigatran etexilate with a protective coating layer; vi) blending the granules of step (iv) with the coated granule of step (v) to form a mixture of at least two types of granules; vii) optionally blending the mixture of at least two types of granules of step (vi) with at least one pharmaceutically acceptable excipient; viii) adding a lubricant to the blend of step (vii); ix) filling the lubricated mixture of step (viii) into suitable hard capsules. 39. The process according to claim 36, comprising the steps of:
blending dabigatran etexilate in the form of the free base or in the form of pharmaceutically acceptable salts, polymorphs, solvates or hydrates thereof and at least one pharmaceutically acceptable excipient; ii) granulating the blend of step (i) with a binder solution to form granules of the dabigatran etexilate; iii) blending at least one organic acid and at least one pharmaceutically acceptable excipient; iv) extruding and a heron zing the blend of step (iii) to form organic acid pellets; v) coating the organic acid pellets of step (iv) with a protective coating layer; vi) blending the granules of step (ii) with the coated pellets of step (v) to form a mixture of at least two types of particles; vii) optionally blending the mixture of at least two types of granules of step (vi) with at least one pharmaceutically acceptable excipient; viii) adding a lubricant to the blend of step (vii); ix) filling the lubricated mixture of step (viii) into suitable hard capsules. | 1,600 |
609 | 15,835,403 | 1,633 | The invention relates to the use of RNA and an aqueous injection buffer containing a sodium salt, a calcium salt and optionally a potassium salt and optionally lactate, in the preparation of a RNA injection solution for increasing RNA transfer and/or RNA translation into/in a host organism. The invention relates further to a RNA injection solution and to a method for increasing the RNA transfer and/or RNA translation of RNA in vivo and in vitro. | 1-21. (canceled) 22. A method for increasing the expression of a mRNA in an organism comprising:
a) obtaining an aqueous mRNA injection solution comprising a calcium salt and a purified mRNA encoding a selected antigen, wherein said mRNA is capped and comprises a poly-A tail; and b) injecting the aqueous mRNA injection solution into an organism intradermally, thereby expressing the mRNA in the organism. 23. The method of claim 22, wherein the aqueous mRNA injection solution comprises salts with the anions selected from the group consisting of chlorides, iodides, bromides, hydroxides, carbonates, hydrogen carbonates or sulfates 24. The method of claim 22, wherein the aqueous mRNA injection solution contains at least 50 mM sodium chloride (NaCl), at least 0.01 mM calcium chloride (CaCl2) and optionally at least 3 mM potassium chloride (KCl). 25. The method of claim 24, wherein the aqueous mRNA injection solution contains from 50 mM to 800 mM sodium chloride (NaCl), from 0.01 mM to 100 mM calcium chloride (CaCl2), and optionally from 3 mM to 500 mM potassium chloride (KCl). 26. The method of claim 22, wherein the aqueous mRNA injection solution further comprises lactate. 27. The method of claim 26, wherein the aqueous mRNA injection solution contains at least 50 mM sodium chloride (NaCl), at least 0.01 mM calcium chloride (CaCl2), at least 15 mM lactate, and optionally at least 3 mM potassium chloride (KCl). 28. The method of claim 26, wherein the aqueous mRNA injection solution contains from 15 mM to 500 mM lactate. 29. The method of claim 22, wherein the mRNA is complexed with a polycationic polypeptide. 30. The method of claim 29, wherein the polycationic polypeptide is poly-L-lysine, poly-L-arginine, or protamine. 31. The method of claim 30, wherein the poly cationic polypeptide is protamine. 32. The method of claim 22, wherein the mRNA is a modified mRNA comprising at least one modification selected from the group consisting of: a) having an increased G/C content in the coding region of the mRNA relative to the wild-type (WT) mRNA, b) having at least one analog of a naturally occurring ribonucleotide, c) free of one or more destabilizing sequences in the WT sequence, d) having an increased A/U content in the ribosome binding site of the mRNA relative to the WT sequence, e) having one or more codon substitutions, f) having a heterologous poly(A) tail, g) having an IRES sequence, h)having a 3′ or 5′ stabilization sequence, and i) having a sequence encoding a signal peptide. 33. The method of claim 22, wherein the mRNA does not comprise a chemical modification. 34. The method of claim 22, wherein the mRNA encodes a tumor antigen. 35. The method of claim 34, wherein the tumor antigen is selected from the group consisting of MAGE, NY-ESO-1, PSA, and mucin. 36. The method of claim 22, wherein the aqueous mRNA injection solution is a Ringer's Lactate solution. 37. The method of claim 22, wherein the aqueous mRNA injection solution does not contain mono-, di- or polysaccharides. 38. The method of claim 22, wherein the aqueous mRNA injection solution comprises from 0.5 mM to 100 mM calcium salt. 39. The method of claim 38, wherein the aqueous mRNA injection solution comprises from 1.5 mM to 40 mM calcium salt. 40. The method of claim 22, wherein the organism suffers from a disease or disorder selected from cancer, allergies, autoimmune diseases, viral infection, and bacterial infection. 41. A method for eliciting an immune response in an organism comprising injecting an aqueous mRNA injection solution into an organism intradermally, thereby expressing the mRNA within the organism, wherein the aqueous mRNA injection solution comprises a calcium salt and a purified mRNA encoding a selected antigen. | The invention relates to the use of RNA and an aqueous injection buffer containing a sodium salt, a calcium salt and optionally a potassium salt and optionally lactate, in the preparation of a RNA injection solution for increasing RNA transfer and/or RNA translation into/in a host organism. The invention relates further to a RNA injection solution and to a method for increasing the RNA transfer and/or RNA translation of RNA in vivo and in vitro.1-21. (canceled) 22. A method for increasing the expression of a mRNA in an organism comprising:
a) obtaining an aqueous mRNA injection solution comprising a calcium salt and a purified mRNA encoding a selected antigen, wherein said mRNA is capped and comprises a poly-A tail; and b) injecting the aqueous mRNA injection solution into an organism intradermally, thereby expressing the mRNA in the organism. 23. The method of claim 22, wherein the aqueous mRNA injection solution comprises salts with the anions selected from the group consisting of chlorides, iodides, bromides, hydroxides, carbonates, hydrogen carbonates or sulfates 24. The method of claim 22, wherein the aqueous mRNA injection solution contains at least 50 mM sodium chloride (NaCl), at least 0.01 mM calcium chloride (CaCl2) and optionally at least 3 mM potassium chloride (KCl). 25. The method of claim 24, wherein the aqueous mRNA injection solution contains from 50 mM to 800 mM sodium chloride (NaCl), from 0.01 mM to 100 mM calcium chloride (CaCl2), and optionally from 3 mM to 500 mM potassium chloride (KCl). 26. The method of claim 22, wherein the aqueous mRNA injection solution further comprises lactate. 27. The method of claim 26, wherein the aqueous mRNA injection solution contains at least 50 mM sodium chloride (NaCl), at least 0.01 mM calcium chloride (CaCl2), at least 15 mM lactate, and optionally at least 3 mM potassium chloride (KCl). 28. The method of claim 26, wherein the aqueous mRNA injection solution contains from 15 mM to 500 mM lactate. 29. The method of claim 22, wherein the mRNA is complexed with a polycationic polypeptide. 30. The method of claim 29, wherein the polycationic polypeptide is poly-L-lysine, poly-L-arginine, or protamine. 31. The method of claim 30, wherein the poly cationic polypeptide is protamine. 32. The method of claim 22, wherein the mRNA is a modified mRNA comprising at least one modification selected from the group consisting of: a) having an increased G/C content in the coding region of the mRNA relative to the wild-type (WT) mRNA, b) having at least one analog of a naturally occurring ribonucleotide, c) free of one or more destabilizing sequences in the WT sequence, d) having an increased A/U content in the ribosome binding site of the mRNA relative to the WT sequence, e) having one or more codon substitutions, f) having a heterologous poly(A) tail, g) having an IRES sequence, h)having a 3′ or 5′ stabilization sequence, and i) having a sequence encoding a signal peptide. 33. The method of claim 22, wherein the mRNA does not comprise a chemical modification. 34. The method of claim 22, wherein the mRNA encodes a tumor antigen. 35. The method of claim 34, wherein the tumor antigen is selected from the group consisting of MAGE, NY-ESO-1, PSA, and mucin. 36. The method of claim 22, wherein the aqueous mRNA injection solution is a Ringer's Lactate solution. 37. The method of claim 22, wherein the aqueous mRNA injection solution does not contain mono-, di- or polysaccharides. 38. The method of claim 22, wherein the aqueous mRNA injection solution comprises from 0.5 mM to 100 mM calcium salt. 39. The method of claim 38, wherein the aqueous mRNA injection solution comprises from 1.5 mM to 40 mM calcium salt. 40. The method of claim 22, wherein the organism suffers from a disease or disorder selected from cancer, allergies, autoimmune diseases, viral infection, and bacterial infection. 41. A method for eliciting an immune response in an organism comprising injecting an aqueous mRNA injection solution into an organism intradermally, thereby expressing the mRNA within the organism, wherein the aqueous mRNA injection solution comprises a calcium salt and a purified mRNA encoding a selected antigen. | 1,600 |
610 | 12,920,524 | 1,639 | The present invention concerns a stable pharmaceutical solution, a process of the preparation thereof and therapeutic uses thereof. | 1. A stable solution of a pharmaceutical compound being an 2-oxo-1-pyrrolidine derivative of formula (I),
wherein,
R1 is C1-10 alkyl or C2-6 alkenyl;
R2 is C1-10 alkyl or C2-6 alkenyl;
X is —CONR4R5, —COOH, —COORS or —CN,
R3 is C1-10 alkyl;
R4 is hydrogen or C1-10 alkyl;
R5 is hydrogen or C1-10 alkyl,
characterized in that it has a pH value of between 4.5 and 6.5. 2. The solution according to claim 1, characterized in that the pH values are between 5.0 and 6.0. 3. The solution according to claim 1, characterized in that
R1 is n-propyl or 2,2-difluororovinyl; R2 is ethyl; X is —CONH2. 4. The solution according to claim 1, characterized in that it is an injectable solution, the amount of the pharmaceutical compound being in the range of 0.01 mg per ml to 200 mg per ml. 5. The solution according to claim 1, characterized in that it is an oral solution, the amount of the pharmaceutical compound being in the range of 0.01 mg per ml to 100 mg per ml. 6. The solution according to claim 1, characterized in that it is an injectable solution having a pH value of 5.5±0.2. 7. The solution according to claim 1, characterized in that it is an oral solution having a pH value of 5.5±0.2. | The present invention concerns a stable pharmaceutical solution, a process of the preparation thereof and therapeutic uses thereof.1. A stable solution of a pharmaceutical compound being an 2-oxo-1-pyrrolidine derivative of formula (I),
wherein,
R1 is C1-10 alkyl or C2-6 alkenyl;
R2 is C1-10 alkyl or C2-6 alkenyl;
X is —CONR4R5, —COOH, —COORS or —CN,
R3 is C1-10 alkyl;
R4 is hydrogen or C1-10 alkyl;
R5 is hydrogen or C1-10 alkyl,
characterized in that it has a pH value of between 4.5 and 6.5. 2. The solution according to claim 1, characterized in that the pH values are between 5.0 and 6.0. 3. The solution according to claim 1, characterized in that
R1 is n-propyl or 2,2-difluororovinyl; R2 is ethyl; X is —CONH2. 4. The solution according to claim 1, characterized in that it is an injectable solution, the amount of the pharmaceutical compound being in the range of 0.01 mg per ml to 200 mg per ml. 5. The solution according to claim 1, characterized in that it is an oral solution, the amount of the pharmaceutical compound being in the range of 0.01 mg per ml to 100 mg per ml. 6. The solution according to claim 1, characterized in that it is an injectable solution having a pH value of 5.5±0.2. 7. The solution according to claim 1, characterized in that it is an oral solution having a pH value of 5.5±0.2. | 1,600 |
611 | 14,383,665 | 1,616 | The present invention relates to a liquid concentrate formulation comprising a pyripyropene pesticide of the formula I as defined below and an alkoxylated aliphatic alcohol as an adjuvant. Moreover, the invention relates to the use of the formulations for the treatment of plants and seed and to corresponding methods. | 1-29. (canceled) 30. A liquid concentrate formulation, which comprises
a) 0.5 to 30 wt %, based on the total weight of the formulation, of a compound of formula I;
b) 20 to 80 wt %, based on the total weight of the formulation, of an aromatic hydrocarbon solvent or aromatic hydrocarbon solvent mixture,
c) 10 to 50 wt %, based on the total weight of the formulation, of at least one alkoxylated aliphatic alcohol of formula (A)
Ra—O—(CmH2mO)x—(C6H2nO)y—(CpH2pO)z—Rb (A)
in which
Ra represents C8-C36-alkyl, C8-C36-alkenyl or a mixture thereof;
Rb represents H or C1-C12-alkyl;
m, n, p represent, independently of one another, an integer from 2 to 16;
x, y, z represent, independently of one another, a number from 0 to 50; and
x+y+z corresponds to a value from 2 to 50,
further comprising
d) at least one anionic surfactant, and
e) at least one non-ionic surfactant S which is selected from the group consisting of C2-C3-polyalkoxylates of C10-C22-hydroxy fatty acid triglycerides and C2-C3-polyalkoxylates of C10-C22-fatty acid mono- or diglycerides. 31. The formulation according to claim 30, wherein the non-ionic surfactant S is selected from the group consisting of polyethoxylates of C12-C22-hydroxyfatty acid triglycerides and polyethoxylates of C12-C22-fatty acid mono- or diglycerides. 32. The formulation according to claim 30, wherein the non-ionic surfactant S is selected from polyethoxylates of C12-C22-hydroxyfatty acid triglycerides. 33. The formulation according to claim 32, wherein the non-ionic surfactant S is castor oil ethoxylate. 34. The formulation according to claim 30 where the average degree of alkoxylation in the non-ionic surfactant S is from 20 to 70. 35. The formulation according to claim 30, containing the non-ionic surfactant S in an amount from 1 to 20 wt %, based on the total weight of the formulation. 36. The formulation according to claim 30, wherein the anionic surfactant is selected from the group consisting of C1-C22-alkylarylsulfonates, sulfates of ethoxylated C4-C22-alkylphenols, phosphates of ethoxylated C4-C22-alkylphenols, phosphates of ethoxylated polyarylphenols and sulfates of ethoxylated polyarylphenols. 37. The formulation according to claim 35, wherein the anionic surfactant is selected from the group consisting of C8-C20-alkylbenzene sulfonates. 38. The formulation according to claim 30, containing the anionic surfactant in an amount from 0.5 to 20 wt %, based on the total weight of the formulation. 39. The formulation according to claim 30, wherein Ra in formula (A) represents a linear C14-C36-alkyl, C14-C36-alkenyl, or a mixture thereof. 40. The formulation according to claim 30, wherein the variables m, n, p in formula (A) represent, independently of one another, an integer from 2 to 5, in particular an integer 2 or 3. 41. The formulation according to claim 30, wherein the sum x+y+z of the variables x, y and z in formula (A) corresponds to a value from 5 to 50, in particular 10 to 30. 42. The formulation according to claim 30, further comprising a ketone having from 6 to 10 carbon atoms. 43. The formulation according to claim 42, wherein the ketone is selected from the group consisting of acetophenone, cycloheptanone, cyclohexanone, 2-hexanone and 2-heptanone. 44. The formulation according to claim 42, containing the ketone in an amount from 5 to 30 wt %, based on the total weight of the formulation. 45. The formulation according to claim 30, further comprising at least one non-ionic block copolymer P comprising at least one polyethylene oxide moiety PEO and at least one polyether moiety PAO consisting of repeating units derived from C3-C6-alkylene oxides and/or styrene oxide, where the block copolymer P does not have alkyl or alkenyl groups with more than 6 carbon atoms. 46. The formulation according to claim 45, wherein the non-ionic block copolymer P is present in an amount from 0.5 to 10 wt %, based on the total weight of the formulation. 47. The formulation according to claim 30, wherein the non-ionic block copolymer P has a number average molecular weight MN ranging from 500 to 10,000 Dalton, in particular 750 to 6,000 Dalton. 48. The formulation according to claim 30, further comprising at least one pH adjuster. 49. The formulation according to claim 48, wherein the pH adjuster is an organic amine containing at least one tertiary amino group. 50. The formulation according to claim 30, which comprises
a) 1 to 10 wt %, based on the total weight of the formulation, of the compound of formula I, b) 30 to 70 wt %, based on the total weight of the formulation, of an aromatic hydrocarbon solvent or aromatic hydrocarbon solvent mixture, c) 10 to 40 wt %, based on the total weight of the formulation, of at the least one alkoxylated aliphatic alcohol of the formula (A), d) 1 to 10 wt %, based on the total weight of the formulation, of the at least one anionic surfactant, e) 1 to 10 wt %, based on the total weight of the formulation, of the at least one non-ionic surfactant S, f) optionally 5 to 30 wt %, based on the total weight of the formulation, of the at least one a ketone having from 6 to 10 carbon atoms, and g) optionally 0.5 to 10 wt %, based on the total weight of the formulation, at least one non-ionic block copolymer P comprising at least one polyethylene oxide moiety PEO and at least one polyether moiety PAO consisting of repeating units derived from C3-C6-alkylene oxides and/or styrene oxide, where the block copolymer P does not have alkyl or alkenyl groups with more than 6 carbon atoms, where the total amount of components a), b), c), d), e) and, if present, components f) and g) add up to at least 90 wt % of the total amount of the formulation. 51. The formulation according to claim 30, which is a non-aqueous formulation. 52. An aqueous ready-to-use preparation obtained by diluting the formulation according to claim 30 with water. 53. A method for protecting plants from attack or infestation by invertebrate pests comprising contacting the plant, or the soil or water in which the plant is growing, with a formulation according to claim 30 in pesticidally effective amounts. 54. A method for controlling invertebrate pests comprising contacting an invertebrate pest or their food supply, habitat, breeding grounds or their locus with a formulation according to claim 30 in pesticidally effective amounts. 55. A method for protection of plant propagation material against invertebrate pests comprising contacting the plant propagation material with a formulation according to claim 30 in pesticidally effective amounts. 56. Seed treated with the formulation according to claim 30. | The present invention relates to a liquid concentrate formulation comprising a pyripyropene pesticide of the formula I as defined below and an alkoxylated aliphatic alcohol as an adjuvant. Moreover, the invention relates to the use of the formulations for the treatment of plants and seed and to corresponding methods.1-29. (canceled) 30. A liquid concentrate formulation, which comprises
a) 0.5 to 30 wt %, based on the total weight of the formulation, of a compound of formula I;
b) 20 to 80 wt %, based on the total weight of the formulation, of an aromatic hydrocarbon solvent or aromatic hydrocarbon solvent mixture,
c) 10 to 50 wt %, based on the total weight of the formulation, of at least one alkoxylated aliphatic alcohol of formula (A)
Ra—O—(CmH2mO)x—(C6H2nO)y—(CpH2pO)z—Rb (A)
in which
Ra represents C8-C36-alkyl, C8-C36-alkenyl or a mixture thereof;
Rb represents H or C1-C12-alkyl;
m, n, p represent, independently of one another, an integer from 2 to 16;
x, y, z represent, independently of one another, a number from 0 to 50; and
x+y+z corresponds to a value from 2 to 50,
further comprising
d) at least one anionic surfactant, and
e) at least one non-ionic surfactant S which is selected from the group consisting of C2-C3-polyalkoxylates of C10-C22-hydroxy fatty acid triglycerides and C2-C3-polyalkoxylates of C10-C22-fatty acid mono- or diglycerides. 31. The formulation according to claim 30, wherein the non-ionic surfactant S is selected from the group consisting of polyethoxylates of C12-C22-hydroxyfatty acid triglycerides and polyethoxylates of C12-C22-fatty acid mono- or diglycerides. 32. The formulation according to claim 30, wherein the non-ionic surfactant S is selected from polyethoxylates of C12-C22-hydroxyfatty acid triglycerides. 33. The formulation according to claim 32, wherein the non-ionic surfactant S is castor oil ethoxylate. 34. The formulation according to claim 30 where the average degree of alkoxylation in the non-ionic surfactant S is from 20 to 70. 35. The formulation according to claim 30, containing the non-ionic surfactant S in an amount from 1 to 20 wt %, based on the total weight of the formulation. 36. The formulation according to claim 30, wherein the anionic surfactant is selected from the group consisting of C1-C22-alkylarylsulfonates, sulfates of ethoxylated C4-C22-alkylphenols, phosphates of ethoxylated C4-C22-alkylphenols, phosphates of ethoxylated polyarylphenols and sulfates of ethoxylated polyarylphenols. 37. The formulation according to claim 35, wherein the anionic surfactant is selected from the group consisting of C8-C20-alkylbenzene sulfonates. 38. The formulation according to claim 30, containing the anionic surfactant in an amount from 0.5 to 20 wt %, based on the total weight of the formulation. 39. The formulation according to claim 30, wherein Ra in formula (A) represents a linear C14-C36-alkyl, C14-C36-alkenyl, or a mixture thereof. 40. The formulation according to claim 30, wherein the variables m, n, p in formula (A) represent, independently of one another, an integer from 2 to 5, in particular an integer 2 or 3. 41. The formulation according to claim 30, wherein the sum x+y+z of the variables x, y and z in formula (A) corresponds to a value from 5 to 50, in particular 10 to 30. 42. The formulation according to claim 30, further comprising a ketone having from 6 to 10 carbon atoms. 43. The formulation according to claim 42, wherein the ketone is selected from the group consisting of acetophenone, cycloheptanone, cyclohexanone, 2-hexanone and 2-heptanone. 44. The formulation according to claim 42, containing the ketone in an amount from 5 to 30 wt %, based on the total weight of the formulation. 45. The formulation according to claim 30, further comprising at least one non-ionic block copolymer P comprising at least one polyethylene oxide moiety PEO and at least one polyether moiety PAO consisting of repeating units derived from C3-C6-alkylene oxides and/or styrene oxide, where the block copolymer P does not have alkyl or alkenyl groups with more than 6 carbon atoms. 46. The formulation according to claim 45, wherein the non-ionic block copolymer P is present in an amount from 0.5 to 10 wt %, based on the total weight of the formulation. 47. The formulation according to claim 30, wherein the non-ionic block copolymer P has a number average molecular weight MN ranging from 500 to 10,000 Dalton, in particular 750 to 6,000 Dalton. 48. The formulation according to claim 30, further comprising at least one pH adjuster. 49. The formulation according to claim 48, wherein the pH adjuster is an organic amine containing at least one tertiary amino group. 50. The formulation according to claim 30, which comprises
a) 1 to 10 wt %, based on the total weight of the formulation, of the compound of formula I, b) 30 to 70 wt %, based on the total weight of the formulation, of an aromatic hydrocarbon solvent or aromatic hydrocarbon solvent mixture, c) 10 to 40 wt %, based on the total weight of the formulation, of at the least one alkoxylated aliphatic alcohol of the formula (A), d) 1 to 10 wt %, based on the total weight of the formulation, of the at least one anionic surfactant, e) 1 to 10 wt %, based on the total weight of the formulation, of the at least one non-ionic surfactant S, f) optionally 5 to 30 wt %, based on the total weight of the formulation, of the at least one a ketone having from 6 to 10 carbon atoms, and g) optionally 0.5 to 10 wt %, based on the total weight of the formulation, at least one non-ionic block copolymer P comprising at least one polyethylene oxide moiety PEO and at least one polyether moiety PAO consisting of repeating units derived from C3-C6-alkylene oxides and/or styrene oxide, where the block copolymer P does not have alkyl or alkenyl groups with more than 6 carbon atoms, where the total amount of components a), b), c), d), e) and, if present, components f) and g) add up to at least 90 wt % of the total amount of the formulation. 51. The formulation according to claim 30, which is a non-aqueous formulation. 52. An aqueous ready-to-use preparation obtained by diluting the formulation according to claim 30 with water. 53. A method for protecting plants from attack or infestation by invertebrate pests comprising contacting the plant, or the soil or water in which the plant is growing, with a formulation according to claim 30 in pesticidally effective amounts. 54. A method for controlling invertebrate pests comprising contacting an invertebrate pest or their food supply, habitat, breeding grounds or their locus with a formulation according to claim 30 in pesticidally effective amounts. 55. A method for protection of plant propagation material against invertebrate pests comprising contacting the plant propagation material with a formulation according to claim 30 in pesticidally effective amounts. 56. Seed treated with the formulation according to claim 30. | 1,600 |
612 | 14,372,088 | 1,641 | A method and composition for extracting an analyte from a test sample such as grain, so as to determine whether the test sample is contaminated with a toxin. The method is particularly useful for detecting the presence in a batch of grain of a mycotoxin, such as for example aflatoxin, ochratoxin, T2, zearalanone, vomitoxin (deoxynivalenol a/k/a DON), patulin and fumonisin. Extraction is performed with use of a composition that includes a proteinaceous material, such as albumin, as an extraction agent. | 1. A method for extracting one or more analytes from a dry test sample comprising the steps of:
a) mixing the sample with a composition comprising a proteinaceous material to form an admixture; b) providing conditions for the admixture to separate into a settled layer and a liquid layer, and c) collecting at least a portion of said liquid layer, wherein said at least a portion of said liquid layer is an extract containing said analyte. 2. The method of claim 1, wherein, in said mixing step, said composition comprising a proteinaceous material is dry and is mixed with the sample to form a dry admixture, and wherein said method further comprises adding a solvent to the dry admixture to form a wet admixture. 3. The method of claim 1, wherein the proteinaceous material is an amino acid. 4. The method of claim 1, wherein the proteinaceous material is an albumin. 5. The method of claim 4, wherein said albumin is bovine serum albumin. 6. The method of claim 4, wherein said albumin is porcine albumin. 7. The method claim 1, wherein the proteinaceous material is Primatone RL. 8. The method of claim 1, wherein the proteinaceous material is a collagen. 9. The method of claim 1, wherein the proteinaceous material is a peptone. 10. The method of claim 9, wherein the proteinaceous material is gelatin peptone. 11. The method of claim 9, wherein the proteinaceous material is soy peptone. 12. The method of claim 1, wherein at least one of the analytes is a mycotoxin. 13. The method of claim 12, wherein the at least one of the analytes is fumonisin. 14. The method of claim 12, wherein the at least one of the analytes is ochratoxin. 15. The method of claim 12, wherein the at least one of the analytes is zearlanone. 16. The method of claim 12, wherein the at least one of the analytes is aflatoxin. 17. The method of claim 1, wherein the test sample comprises a grain. 18. The method of claim 17, wherein the grain comprises corn. 19. The method of claim 17, wherein the grain comprises maize. 20. The method of claim 17, wherein the grain comprises distiller's grain. 21. The method of claim 17, wherein the grain comprises a rice. 22. The method of claim 1, wherein the composition comprising the proteinaceous material is in the form of a tablet. 23. The method of claim 1, wherein the composition comprising the proteinaceous material is in the form of a hydratable solid. 24. The method of claim 1, wherein the composition comprising the proteinaceous material is in the form of a powder. 25. The method of claim 1, wherein the composition comprising the proteinaceous material further comprises a preservative. 26. The method of claim 1, wherein the composition comprising the proteinaceous material further comprises KATHON®. 27. The method of claim 1, wherein the composition comprising the proteinaceous material further comprises a salt. 28. A method for detecting whether a mycotoxin analyte is present in a dry test sample, comprising:
a) preparing an extract containing said mycotoxin analyte according to the method of any of the preceding claims; b) contacting the extract with a labeled receptor to form a mobile phase, the labeled receptor characterized by an ability to bind to the analyte to provide, in the mobile phase, a labeled receptor-analyte complex and further characterized by its ability to provide a detectable signal when the labeled receptor is captured on a solid support; c) contacting the mobile phase with a first test area on a solid support, the first test area comprising a first test area capture agent immobilized on the solid support, said first test area capture agent configured to capture labeled receptor unbound by the analyte from the sample and not to capture the labeled receptor-analyte complex; and d) measuring the intensity of the detectable signal at the first test area, wherein the intensity of the detectable signal is related to the concentration of the analyte in the sample. 29. The method of claim 28, wherein said solid support comprises nitrocellulose. 30. A composition comprising a proteinaceous material for use in the method of claim 1. 31. A composition comprising a proteinaceous material for use in the method of claim 28. | A method and composition for extracting an analyte from a test sample such as grain, so as to determine whether the test sample is contaminated with a toxin. The method is particularly useful for detecting the presence in a batch of grain of a mycotoxin, such as for example aflatoxin, ochratoxin, T2, zearalanone, vomitoxin (deoxynivalenol a/k/a DON), patulin and fumonisin. Extraction is performed with use of a composition that includes a proteinaceous material, such as albumin, as an extraction agent.1. A method for extracting one or more analytes from a dry test sample comprising the steps of:
a) mixing the sample with a composition comprising a proteinaceous material to form an admixture; b) providing conditions for the admixture to separate into a settled layer and a liquid layer, and c) collecting at least a portion of said liquid layer, wherein said at least a portion of said liquid layer is an extract containing said analyte. 2. The method of claim 1, wherein, in said mixing step, said composition comprising a proteinaceous material is dry and is mixed with the sample to form a dry admixture, and wherein said method further comprises adding a solvent to the dry admixture to form a wet admixture. 3. The method of claim 1, wherein the proteinaceous material is an amino acid. 4. The method of claim 1, wherein the proteinaceous material is an albumin. 5. The method of claim 4, wherein said albumin is bovine serum albumin. 6. The method of claim 4, wherein said albumin is porcine albumin. 7. The method claim 1, wherein the proteinaceous material is Primatone RL. 8. The method of claim 1, wherein the proteinaceous material is a collagen. 9. The method of claim 1, wherein the proteinaceous material is a peptone. 10. The method of claim 9, wherein the proteinaceous material is gelatin peptone. 11. The method of claim 9, wherein the proteinaceous material is soy peptone. 12. The method of claim 1, wherein at least one of the analytes is a mycotoxin. 13. The method of claim 12, wherein the at least one of the analytes is fumonisin. 14. The method of claim 12, wherein the at least one of the analytes is ochratoxin. 15. The method of claim 12, wherein the at least one of the analytes is zearlanone. 16. The method of claim 12, wherein the at least one of the analytes is aflatoxin. 17. The method of claim 1, wherein the test sample comprises a grain. 18. The method of claim 17, wherein the grain comprises corn. 19. The method of claim 17, wherein the grain comprises maize. 20. The method of claim 17, wherein the grain comprises distiller's grain. 21. The method of claim 17, wherein the grain comprises a rice. 22. The method of claim 1, wherein the composition comprising the proteinaceous material is in the form of a tablet. 23. The method of claim 1, wherein the composition comprising the proteinaceous material is in the form of a hydratable solid. 24. The method of claim 1, wherein the composition comprising the proteinaceous material is in the form of a powder. 25. The method of claim 1, wherein the composition comprising the proteinaceous material further comprises a preservative. 26. The method of claim 1, wherein the composition comprising the proteinaceous material further comprises KATHON®. 27. The method of claim 1, wherein the composition comprising the proteinaceous material further comprises a salt. 28. A method for detecting whether a mycotoxin analyte is present in a dry test sample, comprising:
a) preparing an extract containing said mycotoxin analyte according to the method of any of the preceding claims; b) contacting the extract with a labeled receptor to form a mobile phase, the labeled receptor characterized by an ability to bind to the analyte to provide, in the mobile phase, a labeled receptor-analyte complex and further characterized by its ability to provide a detectable signal when the labeled receptor is captured on a solid support; c) contacting the mobile phase with a first test area on a solid support, the first test area comprising a first test area capture agent immobilized on the solid support, said first test area capture agent configured to capture labeled receptor unbound by the analyte from the sample and not to capture the labeled receptor-analyte complex; and d) measuring the intensity of the detectable signal at the first test area, wherein the intensity of the detectable signal is related to the concentration of the analyte in the sample. 29. The method of claim 28, wherein said solid support comprises nitrocellulose. 30. A composition comprising a proteinaceous material for use in the method of claim 1. 31. A composition comprising a proteinaceous material for use in the method of claim 28. | 1,600 |
613 | 15,986,592 | 1,617 | A hygiene product, a hygiene product pod, and a method of using the hygiene product pod, the hygiene product pod including a water soluble envelope and the hygiene product sealed in the envelope. The hygiene product includes a carrier comprising butylene glycol in an amount ranging from about 40 wt % to about 70 wt %, based on the total weigh of hygiene product, and an active agent including at least one surfactant. | 1-9. canceled 10. A single-use hygiene product pod comprising:
a water-soluble envelope; and a hygiene product sealed in the envelope, the hygiene product comprising: a carrier comprising butylene glycol in an amount ranging from about 25 wt % to about 70 wt %, based on the total weight of the hygiene product; and an active agent comprising at least one surfactant. 11. The pod of claim 10, wherein:
the butylene glycol comprises 1,3-butanediol; and the hygiene product comprises from about 40 wt % to about 65 wt % 1,3-butanediol, based on the total weight of the hygiene product. 12. The pod of claim 10, wherein the envelope has a pH ranging from about 4.0 to about 9.0, at a temperature of about 25° C. 13. The pod of claim 10, wherein the envelope comprises a polyvinyl alcohol (PVOH) film or a woven or non-woven polyvinyl alcohol (PVOH) fiber. 14. The pod of claim 10, wherein the envelope has a thickness ranging from about 0.5 mm to about 5.0 mm. 15. The pod of claim 10, wherein the hygiene product has a free water content of about 20 wt % or less, based on the total weight of the hygiene product. 16. The pod of claim 10, wherein the hygiene product further comprises a thickener configured to reduce diffusion of the carrier through the envelope. 17. The pod of claim 16, wherein the thickener comprises a lamellar gel network comprising a nonionic surfactant in an amount ranging from about 12 wt % to about 18 wt %, based on the total weight of the hygiene product. 18. The pod of claim 16, wherein the thickener comprises carboxymethyl cellulose in an amount ranging from about 0.15 wt % to about 0.25 wt %, based on the total weight of the hygiene product. 19. A single-use hygiene product pod comprising:
a water-soluble envelope; and a hygiene product sealed in the envelope, the hygiene product comprising:
a carrier comprising butylene glycol in an amount ranging from about 40 wt % to about 70 wt %, based on the total weight of hygiene product; and
an active agent comprising at least one surfactant. 20. The hygiene product pod of claim 19, wherein:
the butylene glycol comprises 1,3-butanediol; the hygiene product further comprises carboxymethyl cellulose in an amount ranging from about 0.15 wt % to about 0.25 wt %, based on the total weight of the hygiene product; the envelope comprises a polyvinyl alcohol (PVOH) film or a woven or non-woven polyvinyl alcohol (PVOH) fiber; and the active agent comprises at least two compounds selected from:
sodium cocoyl isethionate;
cocamidopropyl betaine;
capryloyl/caproyl methyl glucamide and lauroyl/myristoyl methyl glucamide;
cocamidopropyl PG-dimonium chloride phosphate;
linoleamidopropyl PG-dimonium chloride phosphate; or lauramine oxide. 21-25. canceled | A hygiene product, a hygiene product pod, and a method of using the hygiene product pod, the hygiene product pod including a water soluble envelope and the hygiene product sealed in the envelope. The hygiene product includes a carrier comprising butylene glycol in an amount ranging from about 40 wt % to about 70 wt %, based on the total weigh of hygiene product, and an active agent including at least one surfactant.1-9. canceled 10. A single-use hygiene product pod comprising:
a water-soluble envelope; and a hygiene product sealed in the envelope, the hygiene product comprising: a carrier comprising butylene glycol in an amount ranging from about 25 wt % to about 70 wt %, based on the total weight of the hygiene product; and an active agent comprising at least one surfactant. 11. The pod of claim 10, wherein:
the butylene glycol comprises 1,3-butanediol; and the hygiene product comprises from about 40 wt % to about 65 wt % 1,3-butanediol, based on the total weight of the hygiene product. 12. The pod of claim 10, wherein the envelope has a pH ranging from about 4.0 to about 9.0, at a temperature of about 25° C. 13. The pod of claim 10, wherein the envelope comprises a polyvinyl alcohol (PVOH) film or a woven or non-woven polyvinyl alcohol (PVOH) fiber. 14. The pod of claim 10, wherein the envelope has a thickness ranging from about 0.5 mm to about 5.0 mm. 15. The pod of claim 10, wherein the hygiene product has a free water content of about 20 wt % or less, based on the total weight of the hygiene product. 16. The pod of claim 10, wherein the hygiene product further comprises a thickener configured to reduce diffusion of the carrier through the envelope. 17. The pod of claim 16, wherein the thickener comprises a lamellar gel network comprising a nonionic surfactant in an amount ranging from about 12 wt % to about 18 wt %, based on the total weight of the hygiene product. 18. The pod of claim 16, wherein the thickener comprises carboxymethyl cellulose in an amount ranging from about 0.15 wt % to about 0.25 wt %, based on the total weight of the hygiene product. 19. A single-use hygiene product pod comprising:
a water-soluble envelope; and a hygiene product sealed in the envelope, the hygiene product comprising:
a carrier comprising butylene glycol in an amount ranging from about 40 wt % to about 70 wt %, based on the total weight of hygiene product; and
an active agent comprising at least one surfactant. 20. The hygiene product pod of claim 19, wherein:
the butylene glycol comprises 1,3-butanediol; the hygiene product further comprises carboxymethyl cellulose in an amount ranging from about 0.15 wt % to about 0.25 wt %, based on the total weight of the hygiene product; the envelope comprises a polyvinyl alcohol (PVOH) film or a woven or non-woven polyvinyl alcohol (PVOH) fiber; and the active agent comprises at least two compounds selected from:
sodium cocoyl isethionate;
cocamidopropyl betaine;
capryloyl/caproyl methyl glucamide and lauroyl/myristoyl methyl glucamide;
cocamidopropyl PG-dimonium chloride phosphate;
linoleamidopropyl PG-dimonium chloride phosphate; or lauramine oxide. 21-25. canceled | 1,600 |
614 | 15,106,778 | 1,639 | A composition for therapy of a peripheral neuropathy disorder in a subject in need thereof. The composition comprises an effective amount of an agent selected from a group consisting of pirenzepine, oxybutynin, muscarinic toxin 7, a muscarinic receptor antagonist, and combinations thereof, and a pharmacologically acceptable carrier and/or an excipient. The composition is useful for therapy of peripheral neuropathies exemplified by peripheral neuropathies induced by systemic diseases, peripheral neuropathies induced by metabolic diseases, chemotherapy-induced peripheral neuropathies, compression-induced peripheral neuropathies, peripheral neuropathies induced by exposure to dichloroacetate, immune-mediated peripheral neuropathies, peripheral neuropathies induced by infections, and genetically acquired peripheral neuropathies. | 1. A composition for therapy of a peripheral neuropathy disorder in a subject in need thereof, the composition comprising:
an effective amount of an agent selected from a group consisting of pirenzepine, oxybutynin, muscarinic toxin 7, a muscarinic receptor antagonist, and combinations thereof; and a pharmacologically acceptable carrier and/or an excipient. 2. The composition according to claim 1, wherein the composition is injectable. 3. The composition according to claim 1, wherein the composition is administrable by a topical application. 4. The composition according to claim 3, wherein the composition is one of a lotion, a cream, a gel, and a viscous fluid. 5. The composition according to claim 3, additionally comprising one or more of a penetration enhancer, an emollient, an emulsifying agent, a water miscible solvent, an alcohol, and mixtures thereof. 6. The composition according to claim 1, wherein the composition is administrable with a transdermal patch. 7. The composition according to claim 1, wherein the composition is administrable by an oral dosage. 8. The composition according to claim 1, wherein the peripheral neuropathy disorder is a peripheral neuropathy induced by a systemic disease, a peripheral neuropathy induced by a metabolic disease, a chemotherapy-induced peripheral neuropathy, a compression-induced peripheral neuropathy, a peripheral neuropathy induced by an exposure to dichloroacetate, an immune-mediated peripheral neuropathy, a peripheral neuropathy induced by an infection, and a genetically acquired peripheral neuropathy. 9. Use of the composition of claim 1 for therapy of peripheral neuropathy disorder in a subject in need thereof. 10. The use according to claim 9, wherein the peripheral neuropathy is one of a peripheral neuropathy induced by a systemic disease, a peripheral neuropathy induced by a metabolic disease, a chemotherapy-induced peripheral neuropathy, a compression-induced peripheral neuropathy, a peripheral neuropathy induced by an exposure to dichloroacetate, an immune-mediated peripheral neuropathy, a peripheral neuropathy induced by an infection, and a genetically acquired peripheral neuropathy. 11. A method of treating a peripheral neuropathy disorder in a subject in need thereof, the method comprising administering at least one dosage of the composition of claim 1. 12. The method according to claim 10, wherein administration is by an injection. 13. The method according to claim 11, wherein administration is by a subcutaneous injection. | A composition for therapy of a peripheral neuropathy disorder in a subject in need thereof. The composition comprises an effective amount of an agent selected from a group consisting of pirenzepine, oxybutynin, muscarinic toxin 7, a muscarinic receptor antagonist, and combinations thereof, and a pharmacologically acceptable carrier and/or an excipient. The composition is useful for therapy of peripheral neuropathies exemplified by peripheral neuropathies induced by systemic diseases, peripheral neuropathies induced by metabolic diseases, chemotherapy-induced peripheral neuropathies, compression-induced peripheral neuropathies, peripheral neuropathies induced by exposure to dichloroacetate, immune-mediated peripheral neuropathies, peripheral neuropathies induced by infections, and genetically acquired peripheral neuropathies.1. A composition for therapy of a peripheral neuropathy disorder in a subject in need thereof, the composition comprising:
an effective amount of an agent selected from a group consisting of pirenzepine, oxybutynin, muscarinic toxin 7, a muscarinic receptor antagonist, and combinations thereof; and a pharmacologically acceptable carrier and/or an excipient. 2. The composition according to claim 1, wherein the composition is injectable. 3. The composition according to claim 1, wherein the composition is administrable by a topical application. 4. The composition according to claim 3, wherein the composition is one of a lotion, a cream, a gel, and a viscous fluid. 5. The composition according to claim 3, additionally comprising one or more of a penetration enhancer, an emollient, an emulsifying agent, a water miscible solvent, an alcohol, and mixtures thereof. 6. The composition according to claim 1, wherein the composition is administrable with a transdermal patch. 7. The composition according to claim 1, wherein the composition is administrable by an oral dosage. 8. The composition according to claim 1, wherein the peripheral neuropathy disorder is a peripheral neuropathy induced by a systemic disease, a peripheral neuropathy induced by a metabolic disease, a chemotherapy-induced peripheral neuropathy, a compression-induced peripheral neuropathy, a peripheral neuropathy induced by an exposure to dichloroacetate, an immune-mediated peripheral neuropathy, a peripheral neuropathy induced by an infection, and a genetically acquired peripheral neuropathy. 9. Use of the composition of claim 1 for therapy of peripheral neuropathy disorder in a subject in need thereof. 10. The use according to claim 9, wherein the peripheral neuropathy is one of a peripheral neuropathy induced by a systemic disease, a peripheral neuropathy induced by a metabolic disease, a chemotherapy-induced peripheral neuropathy, a compression-induced peripheral neuropathy, a peripheral neuropathy induced by an exposure to dichloroacetate, an immune-mediated peripheral neuropathy, a peripheral neuropathy induced by an infection, and a genetically acquired peripheral neuropathy. 11. A method of treating a peripheral neuropathy disorder in a subject in need thereof, the method comprising administering at least one dosage of the composition of claim 1. 12. The method according to claim 10, wherein administration is by an injection. 13. The method according to claim 11, wherein administration is by a subcutaneous injection. | 1,600 |
615 | 14,796,539 | 1,618 | A method of tracking immune cells to detect immune response. The method including steps of identifying a patient having a disease associated with an organ; administering biocompatible magnetic nanoparticles into the blood stream of the patient; and obtaining a magnetic resonance image of the organ. The presence of hyperintense or hypointense spots in the magnetic resonance image indicates immune response in the patient. | 1. A method of tracking immune cells, the method comprising:
identifying a patient having a disease associated with an organ; providing an aqueous suspension containing biocompatible magnetic nanoparticles, the aqueous suspension being free of particles having a size greater than 1000 nm, the biocompatible magnetic nanoparticles each containing a superparamagnetic core that is covered by one or more biocompatible polymers, each of which has a polyethylene glycol group, a silane group, and a linker linking, via a covalent bond, the polyethylene glycol group and the silane group; administering the aqueous suspension into the blood stream of the patient; and after the administration step, obtaining a magnetic resonance image of the organ, wherein the presence of hyperintense or hypointense spots in the magnetic resonance image indicates immune response in the patient. 2. The method of claim 1, wherein the magnetic resonance image is a T2 or T2* weighted magnetic resonance image. 3. The method of claim 1, wherein the disease is cancer or rejection of a transplanted organ. 4. The method of claim 3, wherein the transplanted organ is heart or kidney. 5. The method of claim 3, wherein the cancer is lymphoma. 6. The method of claim 1, wherein the organ is heart, kidney, or lymph node. 7. The method of claim 1, wherein the superparamagnetic core contains an iron oxide, a cobalt oxide, a nickel oxide, or a combination thereof; the polyethylene glycol group has 5-1000 oxyethylene units; the silane group contains a C1-10 alkylene group; and the linker is O, S, Si, C1-C6 alkylene, a carbonyl moiety containing two carbonyl groups and 2-20 carbon atoms, or a group having one of the following formula:
in which each of m, n, p, q, and t, independently, is 1-6; W is O, S, or NRb; each of L1, L3, L5, L7, and L9, independently, is a bond, O, S, or Nab; each of L2, L4, L6, L8, and L10, independently, is a bond, O, S, or NRd; and V is ORe, SRf, or NRgRh, each of Ra, Rb, Rc, Rd, Re, Rf, Rg, and Rh, independently, being H, OH, a C1-C10 oxyaliphatic radical, a C1-C10 monovalent aliphatic radical, a C1-C10 monovalent heteroaliphatic radical, a monovalent aryl radical, or a monovalent heteroaryl radical. 8. The method of claim 7, wherein the biocompatible magnetic nanoparticles each have a particle size of 10-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 9. The method of claim 8, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 10. The method of claim 1, wherein the superparamagnetic core is a superparamagnetic iron oxide nanoparticle; the polyethylene glycol group has 10 to 200 oxyethylene units; the silane group contains C3-C10 alkylene; and the linker is a carbonyl moiety of the following formula: 11. The method of claim 10, wherein the biocompatible magnetic nanoparticles each have a particle size of 1-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 12. The method of claim 11, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 13. The method of claim 1, wherein the biocompatible magnetic nanoparticles each have a particle size of 1-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 14. The method of claim 13, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 15. The method of claim 1, wherein the superparamagnetic core is covered by one or more biocompatible polymers having the following formula:
in which
R is H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C10 cycloalkyl, C1-C10 heterocycloalkyl, aryl, heteroaryl, a C1-C10 carbonyl group, or a C1-C10 amine group;
L is a linker;
m is 1 to 10; and
n is 5 to 1000. 16. The method of claim 15, wherein the linker is O, S, Si, C1-C6 alkylene, a carbonyl moiety containing two carbonyl groups and 2-20 carbon atoms, or a group having one of the following formula:
in which each of m, n, p, q, and t, independently, is 1-6; W is O, S, or NRb; each of L1, L3, L5, L7, and L9, independently, is a bond, O, S, or NRc; each of L2, L4, L6, L8, and L10, independently, is a bond, O, S, or NRd; and V is ORe, SRf, or NRgRh, each of Ra, Rb, Rc, Rd, Re, Rf, Rg, and Rh, independently, being H, OH, a C1-C10 oxyaliphatic radical, a C1-C10 monovalent aliphatic radical, a C1-C10 monovalent heteroaliphatic radical, a monovalent aryl radical, or a monovalent heteroaryl radical. 17. The method of claim 1, wherein the superparamagnetic core is covered by one or more biocompatible polymers having the following formula:
in which
R1 is H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C10 cycloalkyl, C1-C10 heterocycloalkyl, aryl, heteroaryl, a C1-C10 carbonyl group, or a C1-C10 amine group;
R2 is H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C10 cycloalkyl, C1-C10 heterocycloalkyl, aryl, or heteroaryl;
m is 1 to 10; and
n is 5 to 1000. 18. The method of claim 17, wherein the biocompatible magnetic nanoparticles each have a particle size of 10-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 19. The method of claim 18, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 20. The method of claim 19, wherein the disease is rejection of a transplanted heart or kidney; the superparamagnetic core is a superparamagnetic iron oxide nanoparticle; the polyethylene glycol group has 10 to 200 oxyethylene units; the silane group contains C3-C10 alkylene; the linker is a carbonyl moiety of the following formula:
and the magnetic resonance image is a T2 or T2* weighted magnetic resonance image. 21. The method of claim 17, wherein R1 is H; R2 is H, C1-C6 alkyl, a C1-C10 carbonyl group, or a C1-C10 amine group; m is 3 to 10; and n is 10 to 200. 22. The method of claim 21, wherein the biocompatible magnetic nanoparticles each have a particle size of 10-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 23. The method of claim 22, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. | A method of tracking immune cells to detect immune response. The method including steps of identifying a patient having a disease associated with an organ; administering biocompatible magnetic nanoparticles into the blood stream of the patient; and obtaining a magnetic resonance image of the organ. The presence of hyperintense or hypointense spots in the magnetic resonance image indicates immune response in the patient.1. A method of tracking immune cells, the method comprising:
identifying a patient having a disease associated with an organ; providing an aqueous suspension containing biocompatible magnetic nanoparticles, the aqueous suspension being free of particles having a size greater than 1000 nm, the biocompatible magnetic nanoparticles each containing a superparamagnetic core that is covered by one or more biocompatible polymers, each of which has a polyethylene glycol group, a silane group, and a linker linking, via a covalent bond, the polyethylene glycol group and the silane group; administering the aqueous suspension into the blood stream of the patient; and after the administration step, obtaining a magnetic resonance image of the organ, wherein the presence of hyperintense or hypointense spots in the magnetic resonance image indicates immune response in the patient. 2. The method of claim 1, wherein the magnetic resonance image is a T2 or T2* weighted magnetic resonance image. 3. The method of claim 1, wherein the disease is cancer or rejection of a transplanted organ. 4. The method of claim 3, wherein the transplanted organ is heart or kidney. 5. The method of claim 3, wherein the cancer is lymphoma. 6. The method of claim 1, wherein the organ is heart, kidney, or lymph node. 7. The method of claim 1, wherein the superparamagnetic core contains an iron oxide, a cobalt oxide, a nickel oxide, or a combination thereof; the polyethylene glycol group has 5-1000 oxyethylene units; the silane group contains a C1-10 alkylene group; and the linker is O, S, Si, C1-C6 alkylene, a carbonyl moiety containing two carbonyl groups and 2-20 carbon atoms, or a group having one of the following formula:
in which each of m, n, p, q, and t, independently, is 1-6; W is O, S, or NRb; each of L1, L3, L5, L7, and L9, independently, is a bond, O, S, or Nab; each of L2, L4, L6, L8, and L10, independently, is a bond, O, S, or NRd; and V is ORe, SRf, or NRgRh, each of Ra, Rb, Rc, Rd, Re, Rf, Rg, and Rh, independently, being H, OH, a C1-C10 oxyaliphatic radical, a C1-C10 monovalent aliphatic radical, a C1-C10 monovalent heteroaliphatic radical, a monovalent aryl radical, or a monovalent heteroaryl radical. 8. The method of claim 7, wherein the biocompatible magnetic nanoparticles each have a particle size of 10-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 9. The method of claim 8, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 10. The method of claim 1, wherein the superparamagnetic core is a superparamagnetic iron oxide nanoparticle; the polyethylene glycol group has 10 to 200 oxyethylene units; the silane group contains C3-C10 alkylene; and the linker is a carbonyl moiety of the following formula: 11. The method of claim 10, wherein the biocompatible magnetic nanoparticles each have a particle size of 1-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 12. The method of claim 11, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 13. The method of claim 1, wherein the biocompatible magnetic nanoparticles each have a particle size of 1-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 14. The method of claim 13, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 15. The method of claim 1, wherein the superparamagnetic core is covered by one or more biocompatible polymers having the following formula:
in which
R is H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C10 cycloalkyl, C1-C10 heterocycloalkyl, aryl, heteroaryl, a C1-C10 carbonyl group, or a C1-C10 amine group;
L is a linker;
m is 1 to 10; and
n is 5 to 1000. 16. The method of claim 15, wherein the linker is O, S, Si, C1-C6 alkylene, a carbonyl moiety containing two carbonyl groups and 2-20 carbon atoms, or a group having one of the following formula:
in which each of m, n, p, q, and t, independently, is 1-6; W is O, S, or NRb; each of L1, L3, L5, L7, and L9, independently, is a bond, O, S, or NRc; each of L2, L4, L6, L8, and L10, independently, is a bond, O, S, or NRd; and V is ORe, SRf, or NRgRh, each of Ra, Rb, Rc, Rd, Re, Rf, Rg, and Rh, independently, being H, OH, a C1-C10 oxyaliphatic radical, a C1-C10 monovalent aliphatic radical, a C1-C10 monovalent heteroaliphatic radical, a monovalent aryl radical, or a monovalent heteroaryl radical. 17. The method of claim 1, wherein the superparamagnetic core is covered by one or more biocompatible polymers having the following formula:
in which
R1 is H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C10 cycloalkyl, C1-C10 heterocycloalkyl, aryl, heteroaryl, a C1-C10 carbonyl group, or a C1-C10 amine group;
R2 is H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C10 cycloalkyl, C1-C10 heterocycloalkyl, aryl, or heteroaryl;
m is 1 to 10; and
n is 5 to 1000. 18. The method of claim 17, wherein the biocompatible magnetic nanoparticles each have a particle size of 10-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 19. The method of claim 18, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. 20. The method of claim 19, wherein the disease is rejection of a transplanted heart or kidney; the superparamagnetic core is a superparamagnetic iron oxide nanoparticle; the polyethylene glycol group has 10 to 200 oxyethylene units; the silane group contains C3-C10 alkylene; the linker is a carbonyl moiety of the following formula:
and the magnetic resonance image is a T2 or T2* weighted magnetic resonance image. 21. The method of claim 17, wherein R1 is H; R2 is H, C1-C6 alkyl, a C1-C10 carbonyl group, or a C1-C10 amine group; m is 3 to 10; and n is 10 to 200. 22. The method of claim 21, wherein the biocompatible magnetic nanoparticles each have a particle size of 10-1000 nm and a transverse magnetic relaxivity rate of 50 to 400. 23. The method of claim 22, wherein the biocompatible magnetic nanoparticles each have a particle size of 15-200 nm and a transverse magnetic relaxivity rate of 120 to 400. | 1,600 |
616 | 14,806,586 | 1,632 | Chemotherapeutic agents, methods of producing the chemotherapeutic agents, and methods of treating cancerous cells using the chemotherapeutic agents are provided herein. In an embodiment, a chemotherapeutic agent includes a chimeric gene comprising an anti-apoptotic Bcl-2 family promoter region and a pro-apoptotic Bcl-2 family coding region. A method of producing the chemotherapeutic agent includes isolating an anti-apoptotic Bcl-2 family gene and isolating a pro-apoptotic Bcl-2 family gene. An anti-apoptotic Bcl-2 family promoter region is cleaved from the anti-apoptotic Bcl-2 family gene and a pro-apoptotic Bcl-2 family coding region is cleaved from the pro-apoptotic Bcl-2 family gene. The anti-apoptotic Bcl-2 family promoter region and the pro-apoptotic Bcl-2 family coding region are ligated to form the chimeric gene. A method of treating cancerous cells includes introducing the chimeric gene into the nucleus of the cancerous cells. | 1. A chemotherapeutic agent comprising:
a chimeric gene comprising an anti-apoptotic Bcl-2 family promoter region and a pro-apoptotic Bcl-2 family coding region. 2. The chemotherapeutic agent of claim 1, wherein the anti-apoptotic Bcl-2 family promoter region is chosen from Bcl-2 or Bcl-xL. 3. The chemotherapeutic agent of claim 1, wherein the pro-apoptotic Bcl-2 family coding region is chosen from Bax or Bak. 4. The chemotherapeutic agent of claim 1, wherein the anti-apoptotic Bcl-2 family promoter region is chosen from Bcl-2 or Bcl-xL and wherein the pro-apoptotic Bcl-2 family coding region is chosen from Bax or Bak. 5. The chemotherapeutic agent of claim 1, wherein the anti-apoptotic Bcl-2 family promoter region and the pro-apoptotic Bcl-2 family coding region are linked together through a linking region comprising complementary bases from an anti-apoptotic Bcl-2 family promoter region segment and a pro-apoptotic Bcl-2 family coding region segment. 6. The chemotherapeutic agent of claim 1, wherein the chimeric gene is free from additional promoter regions other than Bcl-2 family promoter regions. 7. The chemotherapeutic agent of claim 1, wherein the chimeric gene further comprises a marker region. 8. The chemotherapeutic agent of claim 1, further comprising a delivery vector wherein the chimeric gene is contained within or bound to the delivery vector. 9. The chemotherapeutic agent of claim 8, wherein the delivery vector targets specific cell types. 10. The chemotherapeutic agent of claim 9, wherein the delivery vector is chosen from oncoretroviruses, lentiviruses, or adenoviruses. 11. A method of treating cancerous cells, wherein the method comprises:
introducing a chimeric gene comprising an anti-apoptotic Bcl-2 family promoter region and a pro-apoptotic Bcl-2 family coding region into the nucleus of the cancerous cells. 12. The method of claim 11, wherein introducing the chimeric gene into the nucleus of the cancerous cells comprises delivering the chimeric gene into the nucleus of the cancerous cells using a delivery vector. 13. The method of claim 11, wherein introducing the chimeric gene wherein introducing the chimeric gene into the nucleus of the cancerous cells comprises introducing the chimeric gene into the nucleus of the cancerous cells through an ex vivo introduction technique. 14. The method of claim 11, wherein introducing the chimeric gene into the nucleus of the cancerous cells comprises introducing the chimeric gene into the nucleus of the cancerous cells through an in vivo introduction technique within a mammal. 15. The method of claim 11, further comprises identifying a defect in cell cycle regulation of candidate cancerous cells to be treated. 16. The method of claim 15, wherein identifying the defect comprises identifying over-production of Bcl-2 in the candidate cancerous cells to be treated. 17. The method of claim 15, wherein identifying the defect comprises identifying over-production of a protein that targets the anti-apoptotic Bcl-2 family promoter region in the candidate cancerous cells to be treated. 18. The method of claim 17, wherein identifying over-production of the protein that targets the anti-apoptotic Bcl-2 family promoter region comprises identifying over-production of the protein chosen from STAT, Myc, Lsf, E2f, and/or NFkB in the candidate cancerous cells to be treated. 19. A method of producing a chemotherapeutic agent, wherein the method comprises:
isolating an anti-apoptotic Bcl-2 family gene; isolating a pro-apoptotic Bcl-2 family gene; cleaving an anti-apoptotic Bcl-2 family promoter region from the anti-apoptotic Bcl-2 family gene and a pro-apoptotic Bcl-2 family coding region from the pro-apoptotic Bcl-2 family gene; ligating the anti-apoptotic Bcl-2 family promoter region and the pro-apoptotic Bcl-2 family coding region to form a chimeric gene. 20. The method of claim 19, further comprising introducing the chimeric gene into a delivery vector. | Chemotherapeutic agents, methods of producing the chemotherapeutic agents, and methods of treating cancerous cells using the chemotherapeutic agents are provided herein. In an embodiment, a chemotherapeutic agent includes a chimeric gene comprising an anti-apoptotic Bcl-2 family promoter region and a pro-apoptotic Bcl-2 family coding region. A method of producing the chemotherapeutic agent includes isolating an anti-apoptotic Bcl-2 family gene and isolating a pro-apoptotic Bcl-2 family gene. An anti-apoptotic Bcl-2 family promoter region is cleaved from the anti-apoptotic Bcl-2 family gene and a pro-apoptotic Bcl-2 family coding region is cleaved from the pro-apoptotic Bcl-2 family gene. The anti-apoptotic Bcl-2 family promoter region and the pro-apoptotic Bcl-2 family coding region are ligated to form the chimeric gene. A method of treating cancerous cells includes introducing the chimeric gene into the nucleus of the cancerous cells.1. A chemotherapeutic agent comprising:
a chimeric gene comprising an anti-apoptotic Bcl-2 family promoter region and a pro-apoptotic Bcl-2 family coding region. 2. The chemotherapeutic agent of claim 1, wherein the anti-apoptotic Bcl-2 family promoter region is chosen from Bcl-2 or Bcl-xL. 3. The chemotherapeutic agent of claim 1, wherein the pro-apoptotic Bcl-2 family coding region is chosen from Bax or Bak. 4. The chemotherapeutic agent of claim 1, wherein the anti-apoptotic Bcl-2 family promoter region is chosen from Bcl-2 or Bcl-xL and wherein the pro-apoptotic Bcl-2 family coding region is chosen from Bax or Bak. 5. The chemotherapeutic agent of claim 1, wherein the anti-apoptotic Bcl-2 family promoter region and the pro-apoptotic Bcl-2 family coding region are linked together through a linking region comprising complementary bases from an anti-apoptotic Bcl-2 family promoter region segment and a pro-apoptotic Bcl-2 family coding region segment. 6. The chemotherapeutic agent of claim 1, wherein the chimeric gene is free from additional promoter regions other than Bcl-2 family promoter regions. 7. The chemotherapeutic agent of claim 1, wherein the chimeric gene further comprises a marker region. 8. The chemotherapeutic agent of claim 1, further comprising a delivery vector wherein the chimeric gene is contained within or bound to the delivery vector. 9. The chemotherapeutic agent of claim 8, wherein the delivery vector targets specific cell types. 10. The chemotherapeutic agent of claim 9, wherein the delivery vector is chosen from oncoretroviruses, lentiviruses, or adenoviruses. 11. A method of treating cancerous cells, wherein the method comprises:
introducing a chimeric gene comprising an anti-apoptotic Bcl-2 family promoter region and a pro-apoptotic Bcl-2 family coding region into the nucleus of the cancerous cells. 12. The method of claim 11, wherein introducing the chimeric gene into the nucleus of the cancerous cells comprises delivering the chimeric gene into the nucleus of the cancerous cells using a delivery vector. 13. The method of claim 11, wherein introducing the chimeric gene wherein introducing the chimeric gene into the nucleus of the cancerous cells comprises introducing the chimeric gene into the nucleus of the cancerous cells through an ex vivo introduction technique. 14. The method of claim 11, wherein introducing the chimeric gene into the nucleus of the cancerous cells comprises introducing the chimeric gene into the nucleus of the cancerous cells through an in vivo introduction technique within a mammal. 15. The method of claim 11, further comprises identifying a defect in cell cycle regulation of candidate cancerous cells to be treated. 16. The method of claim 15, wherein identifying the defect comprises identifying over-production of Bcl-2 in the candidate cancerous cells to be treated. 17. The method of claim 15, wherein identifying the defect comprises identifying over-production of a protein that targets the anti-apoptotic Bcl-2 family promoter region in the candidate cancerous cells to be treated. 18. The method of claim 17, wherein identifying over-production of the protein that targets the anti-apoptotic Bcl-2 family promoter region comprises identifying over-production of the protein chosen from STAT, Myc, Lsf, E2f, and/or NFkB in the candidate cancerous cells to be treated. 19. A method of producing a chemotherapeutic agent, wherein the method comprises:
isolating an anti-apoptotic Bcl-2 family gene; isolating a pro-apoptotic Bcl-2 family gene; cleaving an anti-apoptotic Bcl-2 family promoter region from the anti-apoptotic Bcl-2 family gene and a pro-apoptotic Bcl-2 family coding region from the pro-apoptotic Bcl-2 family gene; ligating the anti-apoptotic Bcl-2 family promoter region and the pro-apoptotic Bcl-2 family coding region to form a chimeric gene. 20. The method of claim 19, further comprising introducing the chimeric gene into a delivery vector. | 1,600 |
617 | 15,534,450 | 1,648 | The present disclosure provides a rabies composition comprising IPRV and PIKA adjuvant, and the pharmaceutical use thereof. The present disclosure also discloses a method for prophylaxis or therapeutic treatment of rabies virus infection, the method comprises a step of administering the rabies vaccine composition to a host. The rabies composition is more stable and safe, and is able to induce earlier and higher titers of neutralizing antibody. | 1. A rabies vaccine composition, comprising:
a) inactivated purified rabies virus (IPRV), b) PIKA adjuvant, and c) human serum albumin (HSA);
wherein
the amount of IPRV is from 0.2 IU to 4.0 IU per unit dose,
the amount of PIKA adjuvant is from 250 μg to 5000 μg per unit dose, and
the amount of HSA is from 0.1% to 0.9% by concentration. 2. The rabies vaccine composition according to claim 1, wherein the amount of said IPRV is selected from the group consisting of 0.2 IU, 0.5 IU, 1.0 IU, 1.5 IU, 2.0 IU, 2.5 IU, 3.0 IU, 3.5 IU, 4.0 IU per unit dose, and the range between any two of the following amounts: 0.2 IU, 0.5 IU, 1.0 IU, 1.5 IU, 2.0 IU, 2.5 IU, 3.0 IU, 3.5 IU, and 4.0 IU per unit dose 3. The rabies vaccine composition according to claim 2, wherein the amount of said IPRV is from 0.5 IU to 3.0 IU per unit dose; preferably from 1.0 IU to 2.5 IU per unit dose. 4. The rabies vaccine composition according to claim 1, wherein the amount of said PIKA adjuvant is selected from the group consisting of 250 μg, 500 μg, 1000 μg, 1500 μg, 2000 μg, 3000 μg, 4000 μg, 5000 μg per unit dose, and the range between any two of the following amounts: 250 μg, 500 μg, 1000 μg, 1500 μg, 2000 μg, 3000 μg, 4000 μg, and 5000 μg per unit dose. 5. The rabies vaccine composition according to claim 4, wherein the amount of said PIKA adjuvant is between from 500 μg to 4000 μg per unit dose, or from 1000 μg to 3000 μg, or from 1500 μg to 2500 μg, optionally about 2000 μg per unit dose. 6. The rabies vaccine composition according to claim 1, wherein the amount of said HSA is selected from the group consisting of 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6% by concentration, and the range between any two of the following amounts: 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, and 0.6% by concentration. 7. The rabies vaccine composition according to claim 1, wherein the ratio of IPRV to PIKA adjuvant is selected from the group consisting of: 1 IU/100 μg, 1 IU/125 μg, 1 IU/200 μg, 1 IU/250 μg, 1 IU/300 μg, 1 IU/350 μg, 1 IU/400 μg, 1 IU/450 μg, 1 IU/500 μg, 1 IU/550 μg, 1 IU/600 μg, 1 IU/700 μg, 1 IU/800 μg, 1 IU/1000 μg, 1 IU/1500 μg, 1 IU/2000 μg, and the ratio between any two of the following ratios: 1 IU/100 μg, 1 IU/125 μg, 1 IU/200 μg, 1 IU/250 μg, 1 IU/300 μg, 1 IU/350 μg, 1 IU/400 μg, 1 IU/4501 μg, 1 IU/500 μg, 1 IU/550 μg, 1 IU/600 μg, 1 IU/700 μg, 1 IU/800 μg, 1 IU/1000 μg, 1 IU/1500 μg, and 1 IU/2000 μg; optionally the ratio of IPRV to PIKA adjuvant is 1 IU/500 μg. 8. The rabies vaccine composition according to any of claims 1-7, wherein the amount of IPRV is 2.0 IU per unit dose, and the amount of PIKA adjuvant is 1000 μg per unit dose. 9. The rabies vaccine composition according to any of claims 1-8, wherein the amount of IPRV is 2.0 IU per unit dose, the amount of PIKA adjuvant is 2000 μg per unit dose, and the amount of HSA is 0.3% by concentration. 10. The rabies vaccine composition according to any of claims 1-7, wherein the amount of IPRV is 1.5 IU per unit dose, the amount of PIKA adjuvant is 2000 μg per unit dose, and the amount of HSA is 0.3% by concentration. 11. The rabies vaccine composition according to any of claims 1-7, wherein the amount of IPRV is 4.0 IU per unit dose, and the amount of PIKA adjuvant is 1000 μg per unit dose, and the amount of HSA is 0.5% by concentration. 12. The rabies vaccine composition according to claim 1, said composition further comprises pharmaceutical acceptable buffer; and the pH of said buffer is between pH 7.0 to pH 8.0. 13. The rabies vaccine composition according to claim 1, wherein the said composition does not comprise gelatin. 14. The rabies vaccine composition according to claim 1, wherein the said composition further comprises maltose, wherein the amount of said maltose is between 1.0% and 6.0% by concentration. 15. The rabies vaccine composition according to any one of claims 1-14, wherein said IPRV is selected from the group consisting of CTN strain, PM strain, aG strain and PV strain; optionally CTN-1 strain, Pitman-Moore L503 strain or PV-2061 strain. 16. The rabies vaccine composition according to claim 1, wherein the said IPRV is prepared in Vero cell or primary hamster kidney cell. 17. The rabies vaccine composition according to any one of claims 1-16, wherein said composition is in a liquid form, optionally in solution or in suspension. 18. The rabies vaccine composition according to any one of claims 1-17, wherein the concentration of said IPRV is from 0.05 IU/ml to 40.0 IU/ml. 19. The rabies vaccine composition according any one of claims 1-18, wherein said unit dose is prepared into a volume which is selected from the group consisting of 0.1 ml, 0.15 ml, 0.2 ml, 0.5 ml, 1.0 ml, 1.5 ml, 2.0 ml, and any volume between any two of the following volumes: 0.1 ml, 0.15 ml, 0.2 ml, 0.5 ml, 1.0 ml, 1.5 ml, and 2.0 ml. 20. A rabies vaccine composition, which is the solid form of the rabies vaccine composition according to any one of claims 1-19, optionally said solid form is lyophilized form. 21. A pharmaceutical kit, comprising or consisting of:
a) the rabies vaccine composition of any of claims 1-20, b) a vial, c) an instruction for use, and d) optionally water for injection,
preferably, said instruction for use suggests the following administration regimen:
1) 2-2-1 regimen for post exposure treatment: 2 unit dose administered on days 0, 2 unit dose administered on days 3 and 1 unit dose administered on days 7 post exposure; and/or
2) 2-1 regimen for prophylaxis: 2 unit dose administered on days 0, and 1 unit dose administered on days 7. 22. Use of rabies vaccine composition according to any one of claims 1-20 in manufacture of a medicament for the treatment or prophylaxis of rabies virus infection. 23. A method for treatment or prevention of rabies caused by rabies virus infection, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host has been exposed to rabies virus. 24. A method for inducing an immune response to rabies virus infection, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host has been exposed to rabies virus. 25. The method according to claim 23 or 24, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host for 5 times within 7 days post exposure of rabies virus. 26. The method according to claim 25, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host based on the following regimen:
a first administration on 0 day post exposure,
a second administration on 0 day post exposure,
a third administration on 2 or 3 day post exposure,
a fourth administration on 2 or 3 day post exposure, and
a fifth administration on 6 or 7 day post exposure, respectively. 27. The method according to claim 23 or 24, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host based on the following regimen:
a 1st and a 2nd administration on 0 day post exposure,
a 3rd and a 4th administration on 2 or 3 day post exposure, and
a 5th and/or a 6th administration on 7 day post exposure, respectively. 28. A method for prophylaxis of rabies caused by rabies virus infection, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host is not exposed to rabies virus. 29. A method of inducing an immune response to a rabies virus, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host is not exposed to rabies virus. 30. The method according to claim 28 or 29, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host for 3 times within 7 days. 31. The method according to claim 30, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host based on the following regimen:
a first administration on day 0,
a second administration on day 0, and
a third administration on day 7, respectively. 32. The method according to any one of claims 23-29, wherein said administration is by intramuscular injection, intradermal delivery, transdermal delivery, subcutaneous injection, parenteral injection, intraperitoneal injection, intravenous injection, inhalation, rectal delivery, nasal delivery, oral delivery, or topical delivery. 33. The method according to any one of claims 23-29, wherein said host is human or non-human animal. 34. The rabies vaccine composition according to any one of claims 1-20 for use in a method of medical treatment, wherein the method involves administering the rabies vaccine composition five or six times within seven days. 35. The rabies vaccine composition for use according to claim 34, wherein the method of treatment involves administering the rabies vaccine composition based on the following regimen:
a 1st and a 2nd administration on 0 day post exposure, a 3rd and a 4th administration on 2 or 3 day post exposure, and a 5th and/or a 6th administration on 7 day post exposure, respectively. 36. The rabies vaccine composition according to any one of claims 1-20 for use in a method of prevention of rabies, wherein the method involves administering the rabies vaccine composition three times within seven days. 37. The rabies vaccine composition for use according to claim 36, wherein the method of prevention involves administering the rabies vaccine composition base on the following regimen:
a first administration on day 0, a second administration on day 0, and a third administration on day 7, respectively. | The present disclosure provides a rabies composition comprising IPRV and PIKA adjuvant, and the pharmaceutical use thereof. The present disclosure also discloses a method for prophylaxis or therapeutic treatment of rabies virus infection, the method comprises a step of administering the rabies vaccine composition to a host. The rabies composition is more stable and safe, and is able to induce earlier and higher titers of neutralizing antibody.1. A rabies vaccine composition, comprising:
a) inactivated purified rabies virus (IPRV), b) PIKA adjuvant, and c) human serum albumin (HSA);
wherein
the amount of IPRV is from 0.2 IU to 4.0 IU per unit dose,
the amount of PIKA adjuvant is from 250 μg to 5000 μg per unit dose, and
the amount of HSA is from 0.1% to 0.9% by concentration. 2. The rabies vaccine composition according to claim 1, wherein the amount of said IPRV is selected from the group consisting of 0.2 IU, 0.5 IU, 1.0 IU, 1.5 IU, 2.0 IU, 2.5 IU, 3.0 IU, 3.5 IU, 4.0 IU per unit dose, and the range between any two of the following amounts: 0.2 IU, 0.5 IU, 1.0 IU, 1.5 IU, 2.0 IU, 2.5 IU, 3.0 IU, 3.5 IU, and 4.0 IU per unit dose 3. The rabies vaccine composition according to claim 2, wherein the amount of said IPRV is from 0.5 IU to 3.0 IU per unit dose; preferably from 1.0 IU to 2.5 IU per unit dose. 4. The rabies vaccine composition according to claim 1, wherein the amount of said PIKA adjuvant is selected from the group consisting of 250 μg, 500 μg, 1000 μg, 1500 μg, 2000 μg, 3000 μg, 4000 μg, 5000 μg per unit dose, and the range between any two of the following amounts: 250 μg, 500 μg, 1000 μg, 1500 μg, 2000 μg, 3000 μg, 4000 μg, and 5000 μg per unit dose. 5. The rabies vaccine composition according to claim 4, wherein the amount of said PIKA adjuvant is between from 500 μg to 4000 μg per unit dose, or from 1000 μg to 3000 μg, or from 1500 μg to 2500 μg, optionally about 2000 μg per unit dose. 6. The rabies vaccine composition according to claim 1, wherein the amount of said HSA is selected from the group consisting of 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6% by concentration, and the range between any two of the following amounts: 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, and 0.6% by concentration. 7. The rabies vaccine composition according to claim 1, wherein the ratio of IPRV to PIKA adjuvant is selected from the group consisting of: 1 IU/100 μg, 1 IU/125 μg, 1 IU/200 μg, 1 IU/250 μg, 1 IU/300 μg, 1 IU/350 μg, 1 IU/400 μg, 1 IU/450 μg, 1 IU/500 μg, 1 IU/550 μg, 1 IU/600 μg, 1 IU/700 μg, 1 IU/800 μg, 1 IU/1000 μg, 1 IU/1500 μg, 1 IU/2000 μg, and the ratio between any two of the following ratios: 1 IU/100 μg, 1 IU/125 μg, 1 IU/200 μg, 1 IU/250 μg, 1 IU/300 μg, 1 IU/350 μg, 1 IU/400 μg, 1 IU/4501 μg, 1 IU/500 μg, 1 IU/550 μg, 1 IU/600 μg, 1 IU/700 μg, 1 IU/800 μg, 1 IU/1000 μg, 1 IU/1500 μg, and 1 IU/2000 μg; optionally the ratio of IPRV to PIKA adjuvant is 1 IU/500 μg. 8. The rabies vaccine composition according to any of claims 1-7, wherein the amount of IPRV is 2.0 IU per unit dose, and the amount of PIKA adjuvant is 1000 μg per unit dose. 9. The rabies vaccine composition according to any of claims 1-8, wherein the amount of IPRV is 2.0 IU per unit dose, the amount of PIKA adjuvant is 2000 μg per unit dose, and the amount of HSA is 0.3% by concentration. 10. The rabies vaccine composition according to any of claims 1-7, wherein the amount of IPRV is 1.5 IU per unit dose, the amount of PIKA adjuvant is 2000 μg per unit dose, and the amount of HSA is 0.3% by concentration. 11. The rabies vaccine composition according to any of claims 1-7, wherein the amount of IPRV is 4.0 IU per unit dose, and the amount of PIKA adjuvant is 1000 μg per unit dose, and the amount of HSA is 0.5% by concentration. 12. The rabies vaccine composition according to claim 1, said composition further comprises pharmaceutical acceptable buffer; and the pH of said buffer is between pH 7.0 to pH 8.0. 13. The rabies vaccine composition according to claim 1, wherein the said composition does not comprise gelatin. 14. The rabies vaccine composition according to claim 1, wherein the said composition further comprises maltose, wherein the amount of said maltose is between 1.0% and 6.0% by concentration. 15. The rabies vaccine composition according to any one of claims 1-14, wherein said IPRV is selected from the group consisting of CTN strain, PM strain, aG strain and PV strain; optionally CTN-1 strain, Pitman-Moore L503 strain or PV-2061 strain. 16. The rabies vaccine composition according to claim 1, wherein the said IPRV is prepared in Vero cell or primary hamster kidney cell. 17. The rabies vaccine composition according to any one of claims 1-16, wherein said composition is in a liquid form, optionally in solution or in suspension. 18. The rabies vaccine composition according to any one of claims 1-17, wherein the concentration of said IPRV is from 0.05 IU/ml to 40.0 IU/ml. 19. The rabies vaccine composition according any one of claims 1-18, wherein said unit dose is prepared into a volume which is selected from the group consisting of 0.1 ml, 0.15 ml, 0.2 ml, 0.5 ml, 1.0 ml, 1.5 ml, 2.0 ml, and any volume between any two of the following volumes: 0.1 ml, 0.15 ml, 0.2 ml, 0.5 ml, 1.0 ml, 1.5 ml, and 2.0 ml. 20. A rabies vaccine composition, which is the solid form of the rabies vaccine composition according to any one of claims 1-19, optionally said solid form is lyophilized form. 21. A pharmaceutical kit, comprising or consisting of:
a) the rabies vaccine composition of any of claims 1-20, b) a vial, c) an instruction for use, and d) optionally water for injection,
preferably, said instruction for use suggests the following administration regimen:
1) 2-2-1 regimen for post exposure treatment: 2 unit dose administered on days 0, 2 unit dose administered on days 3 and 1 unit dose administered on days 7 post exposure; and/or
2) 2-1 regimen for prophylaxis: 2 unit dose administered on days 0, and 1 unit dose administered on days 7. 22. Use of rabies vaccine composition according to any one of claims 1-20 in manufacture of a medicament for the treatment or prophylaxis of rabies virus infection. 23. A method for treatment or prevention of rabies caused by rabies virus infection, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host has been exposed to rabies virus. 24. A method for inducing an immune response to rabies virus infection, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host has been exposed to rabies virus. 25. The method according to claim 23 or 24, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host for 5 times within 7 days post exposure of rabies virus. 26. The method according to claim 25, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host based on the following regimen:
a first administration on 0 day post exposure,
a second administration on 0 day post exposure,
a third administration on 2 or 3 day post exposure,
a fourth administration on 2 or 3 day post exposure, and
a fifth administration on 6 or 7 day post exposure, respectively. 27. The method according to claim 23 or 24, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host based on the following regimen:
a 1st and a 2nd administration on 0 day post exposure,
a 3rd and a 4th administration on 2 or 3 day post exposure, and
a 5th and/or a 6th administration on 7 day post exposure, respectively. 28. A method for prophylaxis of rabies caused by rabies virus infection, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host is not exposed to rabies virus. 29. A method of inducing an immune response to a rabies virus, the method comprises a step of administering the rabies vaccine composition of any of claims 1 to 20 to a host, wherein said host is not exposed to rabies virus. 30. The method according to claim 28 or 29, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host for 3 times within 7 days. 31. The method according to claim 30, wherein the rabies vaccine composition of any of claims 1 to 20 is administered to a host based on the following regimen:
a first administration on day 0,
a second administration on day 0, and
a third administration on day 7, respectively. 32. The method according to any one of claims 23-29, wherein said administration is by intramuscular injection, intradermal delivery, transdermal delivery, subcutaneous injection, parenteral injection, intraperitoneal injection, intravenous injection, inhalation, rectal delivery, nasal delivery, oral delivery, or topical delivery. 33. The method according to any one of claims 23-29, wherein said host is human or non-human animal. 34. The rabies vaccine composition according to any one of claims 1-20 for use in a method of medical treatment, wherein the method involves administering the rabies vaccine composition five or six times within seven days. 35. The rabies vaccine composition for use according to claim 34, wherein the method of treatment involves administering the rabies vaccine composition based on the following regimen:
a 1st and a 2nd administration on 0 day post exposure, a 3rd and a 4th administration on 2 or 3 day post exposure, and a 5th and/or a 6th administration on 7 day post exposure, respectively. 36. The rabies vaccine composition according to any one of claims 1-20 for use in a method of prevention of rabies, wherein the method involves administering the rabies vaccine composition three times within seven days. 37. The rabies vaccine composition for use according to claim 36, wherein the method of prevention involves administering the rabies vaccine composition base on the following regimen:
a first administration on day 0, a second administration on day 0, and a third administration on day 7, respectively. | 1,600 |
618 | 15,621,584 | 1,648 | Antibodies and method of making antibodies, either monoclonal or polyclonal wherein said antibodies have dual or multi-specific binding capacity to more than one type of antigenic epitope. The antibodies have simultaneous or independent recognition subsites to each of the epitopes. Antigenic epitopes include lipids, peptides, proteins, amino acid sequences, sugars and carbohydrates. Monoclonal antibodies and a method of making monoclonal antibodies of the invention include monoclonal antibodies that are broadly neutralizing to HIV-1 or other envelop viruses wherein the monoclonal antibody has subsites that simultaneously recognize protein and lipid epitopes from the virus. | 1-54. (canceled) 55. A modified liposome comprising: a) one or more lipids found in a host cell plasma membrane lipid bilayer in a region of the lipid raft or one or more types of lipids normally found in HIV-1; b) an adjuvant, c) a protein or peptide epitope from HIV-1 virus and d) nef or nef with env antigens, wherein said modified liposome produces monoclonal antibodies having specificity for both the lipid epitopes and the protein or peptide epitope. 56. The modified liposome of claim 55, wherein said lipid epitopes comprise one or more of phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, GalCer, SGalCer, CTH, GM1, and GM3. 57. The modified liposome of claim 55, wherein said protein or peptide epitopes are from HIV-1 and comprise one or more of gp160, gp 140, gp120 and gp41. 58. The modified liposome of claim 55, wherein the proteins or peptide epitope are attached to the surface of the liposomes, intercalated into a liposomal membrane bilayer, or encapsulated in an aqueous space inside of the liposome. 59. The modified liposome of claim 55, wherein the adjuvant is Lipid A. 60. A modified liposome comprising:
a. lipid epitopes to HIV-1, wherein said lipid epitopes comprise one or more of phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, GalCer, SGalCer, CTH, GM1 and GM3; b. an adjuvant; c. a protein or peptide epitopes from HIV-1 virus, wherein said protein or peptide epitopes are from HIV-1 comprise one or more of gp160, gp 140, gp120 and gp41; and d. nef or nef with env antigens, wherein said modified liposome has a property that it is capable of producing monoclonal antibodies having specificity for both the lipid and the HIV protein or peptide. 61. The modified liposome of claim 60, wherein the adjuvant is Lipid A. 62. A method of making liposomes that that have a property that they can produce monoclonal antibodies having specificity for both HIV lipid and HIV protein or peptide, said method comprising the steps of:
a. obtaining liposomes having one or more lipids found in a host cell plasma membrane lipid bilayer in a region of a lipid raft or one or more types of lipids normally found in HIV-1; b. modifying said liposomes by incorporating: (1) an adjuvant, (2) a protein or peptide epitope from HIV-1 virus, and (3) nef or nef with env antigens. 63. The method of claim 62, wherein said lipids comprise one or more of phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, GalCer, SGalCer, CTH, GM1 and GM3. 64. The method of claim 62, wherein said protein or peptide epitopes are from HIV-1 and comprise one or more of gp160, gp 140, gp120 and gp41. 65. The method of claim 62, wherein said adjuvant is Lipid A. 66. A vaccine comprising the modified liposome of claim 55. 67. A vaccine comprising the modified liposome of claim 60. 68. An isolated dual- or multi-specific monoclonal antibody that has an antigen binding site that is dual- or multi-specific in binding both lipid and lipid associated HIV-1 protein antigen, wherein said monoclonal antibody was made by the following process:
a) obtaining liposomes having lipid epitopes to HIV-1 and modifying said organized lipid structure by incorporating (1) an adjuvant and (2) a protein or peptide epitope from HIV-1 virus; b) immunizing a mammal with said liposomes; c) producing said antibodies, wherein said antibodies have simultaneous recognition subsites to said lipid epitopes in said liposome and to said protein or peptide epitope to HIV-1 virus; d) identifying said antibodies with simultaneous recognition subsites to said lipid epitopes in said liposomes and to said protein or peptide epitope of said HIV-1 virus; e) isolating only said antibodies with simultaneous recognition subsites to said lipid epitopes in said liposome and to said protein or peptide epitope of said HIV-1; and f) cloning said antibodies to obtain monoclonal dual-multi specific antibodies having dual or multi-specific antigen binding sites binding more than one antigenic epitope selected from the lipid epitope and the HIV-1 epitope. 69. A method of making monoclonal antibodies that have an antigen binding site that is dual- or multi-specific in binding more than one type of antigenic epitope comprising:
a) obtaining an organized lipid structure having a first lipid epitope and modifying said organized lipid structure by incorporating (1) an adjuvant and (2) a protein or peptide epitope; b) immunizing a mammal with said organized lipid structure; c) producing said antibodies, wherein said antibodies have simultaneous recognition subsites to said lipid epitopes in said organized lipid structure and to said protein or peptide epitope; d) identifying said antibodies with simultaneous recognition subsites to said lipid epitopes in said organized lipid structure and to said protein or peptide epitope; e) isolating only said antibodies with simultaneous recognition subsites to said lipid epitopes in said organized lipid structure and to said protein or peptide epitope; and f) cloning said antibodies to obtain monoclonal dual-multi specific antibodies having dual or multi-specific antigen binding sites binding more than one antigenic epitope selected from the lipid epitope and the protein or peptide epitope. 70. The method of claim 69, wherein said lipid and protein or peptide epitopes are from the same entity. 71. The method of claim 70 wherein said same entity is selected from the group consisting of viruses, bacteria, hormones, fungi, cancer cells and protozoa. 72. The method of claim 69, wherein said adjuvant is Lipid A. 73. The method of claim 69, wherein said organized lipid structure is a lipid or liposome. 74. The method of claim 69, wherein said lipid epitope comprise one or more of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, phosphatidyl glycerol, GalCer, SGalCer, CTH, GM1, GM3 and cholesterol. 75. A method of making antibodies that have an antigen binding site that is dual- or multi-specific in binding more than one type of antigenic epitope comprising:
a) obtaining liposomes having a first antigenic epitope; b) modifying said liposomes by including a second antigenic epitope in said organized lipid structure; c) immunizing a mammal with said liposomes; d) producing said antibodies, wherein said antibodies have simultaneous recognition subsites to said first antigenic epitopes in said liposome and to said second antigenic epitopes; e) identifying said antibodies with simultaneous recognition subsites to said first antigenic epitopes in said liposome and to said second antigenic epitope; f) isolating only said antibodies with simultaneous recognition subsites to said first epitopes in said liposome and to said second antigenic epitope; g). cloning said antibodies to obtain monoclonal dual-multi specific antibodies having dual or multi-specific antigen binding sites binding more than one antigenic epitope selected from said first and second antigenic epitopes. 76. The method of claim 75, wherein said first antigenic epitope and second antigenic epitope is selected from the group consisting of protein, peptide, polypeptide, amino acid sequence, lipid, sugar and carbohydrate. 77. The method of claim 75, wherein said lipid epitope comprise one or more of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, phosphatidyl glycerol, GalCer, SGalCer, CTH, GM1, GM3 and cholesterol. 78. A method of making a monoclonal antibody to anionic phospholipids comprising: incorporating said anionic phospholipids and lipid A into a liposome; inserting said liposome into a mammal wherein said mammal produces monoclonal antibodies to said phospholipids. 79. The method of claim 78, wherein the anionic phospholipids is phosphatidylinositol phosphate (PIP). 80. The method of claim 78, wherein said anionic phospholipids is cardiolipin. 81. A method of making a monoclonal antibody to phosphatidylinositol phosphate (PIP) comprising:
incorporating PIP and Lipid A into a liposome; inserting said liposome into a mammal, wherein said mammal produces monoclonal antibodies to said PIP. 82. A method of binding PIP antigen and or cardiolipin (CL) comprising:
administering anti-PIP antibody of claim 81 to a medium containing PIP antigen or CL antigen. 83. A method of inhibition of infection of HIV-1 in primary cultures of peripheral blood mononuclear cells by HIV-1 comprising administering the anti PIP antibody of claim 81. | Antibodies and method of making antibodies, either monoclonal or polyclonal wherein said antibodies have dual or multi-specific binding capacity to more than one type of antigenic epitope. The antibodies have simultaneous or independent recognition subsites to each of the epitopes. Antigenic epitopes include lipids, peptides, proteins, amino acid sequences, sugars and carbohydrates. Monoclonal antibodies and a method of making monoclonal antibodies of the invention include monoclonal antibodies that are broadly neutralizing to HIV-1 or other envelop viruses wherein the monoclonal antibody has subsites that simultaneously recognize protein and lipid epitopes from the virus.1-54. (canceled) 55. A modified liposome comprising: a) one or more lipids found in a host cell plasma membrane lipid bilayer in a region of the lipid raft or one or more types of lipids normally found in HIV-1; b) an adjuvant, c) a protein or peptide epitope from HIV-1 virus and d) nef or nef with env antigens, wherein said modified liposome produces monoclonal antibodies having specificity for both the lipid epitopes and the protein or peptide epitope. 56. The modified liposome of claim 55, wherein said lipid epitopes comprise one or more of phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, GalCer, SGalCer, CTH, GM1, and GM3. 57. The modified liposome of claim 55, wherein said protein or peptide epitopes are from HIV-1 and comprise one or more of gp160, gp 140, gp120 and gp41. 58. The modified liposome of claim 55, wherein the proteins or peptide epitope are attached to the surface of the liposomes, intercalated into a liposomal membrane bilayer, or encapsulated in an aqueous space inside of the liposome. 59. The modified liposome of claim 55, wherein the adjuvant is Lipid A. 60. A modified liposome comprising:
a. lipid epitopes to HIV-1, wherein said lipid epitopes comprise one or more of phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, GalCer, SGalCer, CTH, GM1 and GM3; b. an adjuvant; c. a protein or peptide epitopes from HIV-1 virus, wherein said protein or peptide epitopes are from HIV-1 comprise one or more of gp160, gp 140, gp120 and gp41; and d. nef or nef with env antigens, wherein said modified liposome has a property that it is capable of producing monoclonal antibodies having specificity for both the lipid and the HIV protein or peptide. 61. The modified liposome of claim 60, wherein the adjuvant is Lipid A. 62. A method of making liposomes that that have a property that they can produce monoclonal antibodies having specificity for both HIV lipid and HIV protein or peptide, said method comprising the steps of:
a. obtaining liposomes having one or more lipids found in a host cell plasma membrane lipid bilayer in a region of a lipid raft or one or more types of lipids normally found in HIV-1; b. modifying said liposomes by incorporating: (1) an adjuvant, (2) a protein or peptide epitope from HIV-1 virus, and (3) nef or nef with env antigens. 63. The method of claim 62, wherein said lipids comprise one or more of phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, GalCer, SGalCer, CTH, GM1 and GM3. 64. The method of claim 62, wherein said protein or peptide epitopes are from HIV-1 and comprise one or more of gp160, gp 140, gp120 and gp41. 65. The method of claim 62, wherein said adjuvant is Lipid A. 66. A vaccine comprising the modified liposome of claim 55. 67. A vaccine comprising the modified liposome of claim 60. 68. An isolated dual- or multi-specific monoclonal antibody that has an antigen binding site that is dual- or multi-specific in binding both lipid and lipid associated HIV-1 protein antigen, wherein said monoclonal antibody was made by the following process:
a) obtaining liposomes having lipid epitopes to HIV-1 and modifying said organized lipid structure by incorporating (1) an adjuvant and (2) a protein or peptide epitope from HIV-1 virus; b) immunizing a mammal with said liposomes; c) producing said antibodies, wherein said antibodies have simultaneous recognition subsites to said lipid epitopes in said liposome and to said protein or peptide epitope to HIV-1 virus; d) identifying said antibodies with simultaneous recognition subsites to said lipid epitopes in said liposomes and to said protein or peptide epitope of said HIV-1 virus; e) isolating only said antibodies with simultaneous recognition subsites to said lipid epitopes in said liposome and to said protein or peptide epitope of said HIV-1; and f) cloning said antibodies to obtain monoclonal dual-multi specific antibodies having dual or multi-specific antigen binding sites binding more than one antigenic epitope selected from the lipid epitope and the HIV-1 epitope. 69. A method of making monoclonal antibodies that have an antigen binding site that is dual- or multi-specific in binding more than one type of antigenic epitope comprising:
a) obtaining an organized lipid structure having a first lipid epitope and modifying said organized lipid structure by incorporating (1) an adjuvant and (2) a protein or peptide epitope; b) immunizing a mammal with said organized lipid structure; c) producing said antibodies, wherein said antibodies have simultaneous recognition subsites to said lipid epitopes in said organized lipid structure and to said protein or peptide epitope; d) identifying said antibodies with simultaneous recognition subsites to said lipid epitopes in said organized lipid structure and to said protein or peptide epitope; e) isolating only said antibodies with simultaneous recognition subsites to said lipid epitopes in said organized lipid structure and to said protein or peptide epitope; and f) cloning said antibodies to obtain monoclonal dual-multi specific antibodies having dual or multi-specific antigen binding sites binding more than one antigenic epitope selected from the lipid epitope and the protein or peptide epitope. 70. The method of claim 69, wherein said lipid and protein or peptide epitopes are from the same entity. 71. The method of claim 70 wherein said same entity is selected from the group consisting of viruses, bacteria, hormones, fungi, cancer cells and protozoa. 72. The method of claim 69, wherein said adjuvant is Lipid A. 73. The method of claim 69, wherein said organized lipid structure is a lipid or liposome. 74. The method of claim 69, wherein said lipid epitope comprise one or more of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, phosphatidyl glycerol, GalCer, SGalCer, CTH, GM1, GM3 and cholesterol. 75. A method of making antibodies that have an antigen binding site that is dual- or multi-specific in binding more than one type of antigenic epitope comprising:
a) obtaining liposomes having a first antigenic epitope; b) modifying said liposomes by including a second antigenic epitope in said organized lipid structure; c) immunizing a mammal with said liposomes; d) producing said antibodies, wherein said antibodies have simultaneous recognition subsites to said first antigenic epitopes in said liposome and to said second antigenic epitopes; e) identifying said antibodies with simultaneous recognition subsites to said first antigenic epitopes in said liposome and to said second antigenic epitope; f) isolating only said antibodies with simultaneous recognition subsites to said first epitopes in said liposome and to said second antigenic epitope; g). cloning said antibodies to obtain monoclonal dual-multi specific antibodies having dual or multi-specific antigen binding sites binding more than one antigenic epitope selected from said first and second antigenic epitopes. 76. The method of claim 75, wherein said first antigenic epitope and second antigenic epitope is selected from the group consisting of protein, peptide, polypeptide, amino acid sequence, lipid, sugar and carbohydrate. 77. The method of claim 75, wherein said lipid epitope comprise one or more of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, phosphatidylserine, phosphatidylinositol-4-phosphate, phosphatidylinositol, phosphatidyl glycerol, GalCer, SGalCer, CTH, GM1, GM3 and cholesterol. 78. A method of making a monoclonal antibody to anionic phospholipids comprising: incorporating said anionic phospholipids and lipid A into a liposome; inserting said liposome into a mammal wherein said mammal produces monoclonal antibodies to said phospholipids. 79. The method of claim 78, wherein the anionic phospholipids is phosphatidylinositol phosphate (PIP). 80. The method of claim 78, wherein said anionic phospholipids is cardiolipin. 81. A method of making a monoclonal antibody to phosphatidylinositol phosphate (PIP) comprising:
incorporating PIP and Lipid A into a liposome; inserting said liposome into a mammal, wherein said mammal produces monoclonal antibodies to said PIP. 82. A method of binding PIP antigen and or cardiolipin (CL) comprising:
administering anti-PIP antibody of claim 81 to a medium containing PIP antigen or CL antigen. 83. A method of inhibition of infection of HIV-1 in primary cultures of peripheral blood mononuclear cells by HIV-1 comprising administering the anti PIP antibody of claim 81. | 1,600 |
619 | 15,221,379 | 1,631 | The invention provides methods and systems of determining biopolymer profiles and correlations between structural units (residues) of a biopolymer based on sampling of the conformational space available to the molecule. The correlations between these structural units can further be used to find networks within a biopolymer such as the coupled residue networks in a protein. The invention also provides for designing and engineering biopolymers including polypeptides, nucleic acids and carbohydrates using the information derived from the conformation clustering and subsequent methods described herein. | 1. A method of engineering a variant of a biopolymer, wherein the biopolymer comprises a plurality of component structural units, the method comprising:
a) performing a simulation to calculate at least one trajectory for a plurality of geometric metrics using a three-dimensional atomic model of the biopolymer, wherein
the plurality of geometric metrics comprises a intra-component structural unit plane metric or an intra-component structural unit dihedral angle metric or an intra-component structural unit distance metric for each individual component structural unit in the plurality of component structural units;
b) determining at least one structural unit conformation for each corresponding component structural unit in the plurality of component structural units, wherein
the at least one structural unit conformation of a corresponding component structural unit in the plurality of component structural units is obtained by clustering first conformation information for the corresponding component structural unit in the at least one trajectory, wherein the first conformation information comprises a subset of geometric metrics or the plurality of geometric metrics for the corresponding component structural unit;
c) determining a conformational frequency of each the at least one structural unit conformation of each component structural unit in the plurality of component structural units; d) determining a variance in the first conformation information of each the at least one structural unit conformation of each component structural unit in the plurality of component structural units; and e) assigning a mobility index to each respective component structural unit in the plurality of component structural units based on the corresponding conformational frequency and the variance in the first conformation information of each structural unit conformation in the at least one structural unit conformation of the respective component structural unit; and f) making the variant of the biopolymer, wherein the variant of the biopolymer comprises the sequence of the biopolymer in which a first component structural unit of the biopolymer has been substituted in the variant of the biopolymer for a different component structural unit at least partly on the basis of the mobility index for the component structural unit. 2. The method of claim 1, wherein each component structural unit in the plurality of component structural units is a residue. 3-4. (canceled) 5. The method of claim 2 wherein a structural unit conformation in the at least one structural unit conformation of a second component structural unit in the plurality of component structural units is an off rotamer conformation of the second component structural unit. 6. (canceled) 7. The method of claim 2 wherein the plurality of geometric metrics comprises a residue metric selected from the group consisting of a dihedral angle, a position vector and a plane angle metric or a distance metric. 8. The method of claim 7 wherein the plane angle metric is calculated using a first and a second plane of a component structural unit, the first plane defined by a first set of atoms comprising a backbone atom of the component structural unit and the second plane defined by a second set of atoms comprising an atom selected from the group consisting of a backbone atom and a terminal atom of the component structural unit. 9. The method of claim 8 wherein the first set of atoms is selected from the group consisting of (Cα, CO, N) and (Cα, CO, O). 10. The method of claim 1 wherein the clustering first conformation information for the corresponding component structural unit comprises using a multidimensional clustering method. 11-14. (canceled) 15. The method of claim 1 wherein the biopolymer is selected from the group consisting of a structural protein, an antibody, an enzyme and a signaling protein. 16-37. (canceled) 38. The method of claim 1 wherein the simulation is performed using a Monte Carlo sampling technique or an experimental method. 39. The method of claim 38 wherein the experimental method is performed using data from NMR spectroscopy or X-ray crystallography. 40. The method of claim 1, wherein the first component structural unit of the biopolymer has been substituted in the variant of the biopolymer for the different component structural unit at least partly on the basis of (i) the mobility index for the component structural unit and (ii) a solvent accessibility of the component structural unit in the three-dimensional atomic model of the biopolymer. 41. The method of claim 1, wherein the simulation is a molecular dynamics simulation using the three-dimensional atomic model of the biopolymer | The invention provides methods and systems of determining biopolymer profiles and correlations between structural units (residues) of a biopolymer based on sampling of the conformational space available to the molecule. The correlations between these structural units can further be used to find networks within a biopolymer such as the coupled residue networks in a protein. The invention also provides for designing and engineering biopolymers including polypeptides, nucleic acids and carbohydrates using the information derived from the conformation clustering and subsequent methods described herein.1. A method of engineering a variant of a biopolymer, wherein the biopolymer comprises a plurality of component structural units, the method comprising:
a) performing a simulation to calculate at least one trajectory for a plurality of geometric metrics using a three-dimensional atomic model of the biopolymer, wherein
the plurality of geometric metrics comprises a intra-component structural unit plane metric or an intra-component structural unit dihedral angle metric or an intra-component structural unit distance metric for each individual component structural unit in the plurality of component structural units;
b) determining at least one structural unit conformation for each corresponding component structural unit in the plurality of component structural units, wherein
the at least one structural unit conformation of a corresponding component structural unit in the plurality of component structural units is obtained by clustering first conformation information for the corresponding component structural unit in the at least one trajectory, wherein the first conformation information comprises a subset of geometric metrics or the plurality of geometric metrics for the corresponding component structural unit;
c) determining a conformational frequency of each the at least one structural unit conformation of each component structural unit in the plurality of component structural units; d) determining a variance in the first conformation information of each the at least one structural unit conformation of each component structural unit in the plurality of component structural units; and e) assigning a mobility index to each respective component structural unit in the plurality of component structural units based on the corresponding conformational frequency and the variance in the first conformation information of each structural unit conformation in the at least one structural unit conformation of the respective component structural unit; and f) making the variant of the biopolymer, wherein the variant of the biopolymer comprises the sequence of the biopolymer in which a first component structural unit of the biopolymer has been substituted in the variant of the biopolymer for a different component structural unit at least partly on the basis of the mobility index for the component structural unit. 2. The method of claim 1, wherein each component structural unit in the plurality of component structural units is a residue. 3-4. (canceled) 5. The method of claim 2 wherein a structural unit conformation in the at least one structural unit conformation of a second component structural unit in the plurality of component structural units is an off rotamer conformation of the second component structural unit. 6. (canceled) 7. The method of claim 2 wherein the plurality of geometric metrics comprises a residue metric selected from the group consisting of a dihedral angle, a position vector and a plane angle metric or a distance metric. 8. The method of claim 7 wherein the plane angle metric is calculated using a first and a second plane of a component structural unit, the first plane defined by a first set of atoms comprising a backbone atom of the component structural unit and the second plane defined by a second set of atoms comprising an atom selected from the group consisting of a backbone atom and a terminal atom of the component structural unit. 9. The method of claim 8 wherein the first set of atoms is selected from the group consisting of (Cα, CO, N) and (Cα, CO, O). 10. The method of claim 1 wherein the clustering first conformation information for the corresponding component structural unit comprises using a multidimensional clustering method. 11-14. (canceled) 15. The method of claim 1 wherein the biopolymer is selected from the group consisting of a structural protein, an antibody, an enzyme and a signaling protein. 16-37. (canceled) 38. The method of claim 1 wherein the simulation is performed using a Monte Carlo sampling technique or an experimental method. 39. The method of claim 38 wherein the experimental method is performed using data from NMR spectroscopy or X-ray crystallography. 40. The method of claim 1, wherein the first component structural unit of the biopolymer has been substituted in the variant of the biopolymer for the different component structural unit at least partly on the basis of (i) the mobility index for the component structural unit and (ii) a solvent accessibility of the component structural unit in the three-dimensional atomic model of the biopolymer. 41. The method of claim 1, wherein the simulation is a molecular dynamics simulation using the three-dimensional atomic model of the biopolymer | 1,600 |
620 | 15,287,909 | 1,636 | The prior art largely suggests that immune competence in a cancer patient would destroy an antigenic viral vector before it transfects host cells with a transgene-rendering therapy futile. We surprisingly found the opposite is true: cancer patients with competent immune systems obtain the most therapeutic benefit from antigenic viral vector, apparently because the antigenic viral vector, apart from transfecting cells with a transgene, induces a humoral immune response, which in turn attacks cells bearing cancer antigen. Our new therapy works particularly well in combination with cytotoxic chemotherapeutic drugs. | 1. In a method of treating cancer in a human, the improvement comprising:
a. Determining the level of immunity against a viral vector, b. Confirming said human has a measurable level of immunity against said vector, and c. Administering to said human said viral vector. 2. The method of claim 1, wherein said administering to said human said viral vector comprises administering to a cavity created by a resection of said tumor. 3. The method of claim 1, wherein said viral vector comprises adenovirus. 4. The method of claim 1, wherein said viral vector comprises a transgene. 5. The method of claim 4, wherein said transgene comprises nucleic acid sequence coding for thymidine kinase enzyme. 6. The method of claim 1, wherein said cancer comprises glioma. 7. The method of claim 1, further comprising administering to said human patient a cytotoxic chemotherapeutic. 8. The method of claim 6, further comprising administering to said human patient temozolomide. 9. In a method of treating cancer in a human, the improvement comprising:
a. Inducing an immune response against a viral vector, and then b. Administering to said human said viral vector. 10. The method of claim 9, wherein said administering to said human said viral vector comprises administering to a cavity created by a resection of said tumor. 11. The method of claim 9, wherein said viral vector comprises adenovirus. 12. The method of claim 9, wherein said viral vector comprises a transgene. 13. The method of claim 12, wherein said transgene comprises nucleic acid sequence coding for thymidine kinase enzyme. 14. The method of claim 9, wherein said cancer comprises glioma. 15. The method of claim 9, further comprising administering to said human patient a cytotoxic chemotherapeutic. 14. The method of claim 13, further comprising administering to said human patient temozolomide. | The prior art largely suggests that immune competence in a cancer patient would destroy an antigenic viral vector before it transfects host cells with a transgene-rendering therapy futile. We surprisingly found the opposite is true: cancer patients with competent immune systems obtain the most therapeutic benefit from antigenic viral vector, apparently because the antigenic viral vector, apart from transfecting cells with a transgene, induces a humoral immune response, which in turn attacks cells bearing cancer antigen. Our new therapy works particularly well in combination with cytotoxic chemotherapeutic drugs.1. In a method of treating cancer in a human, the improvement comprising:
a. Determining the level of immunity against a viral vector, b. Confirming said human has a measurable level of immunity against said vector, and c. Administering to said human said viral vector. 2. The method of claim 1, wherein said administering to said human said viral vector comprises administering to a cavity created by a resection of said tumor. 3. The method of claim 1, wherein said viral vector comprises adenovirus. 4. The method of claim 1, wherein said viral vector comprises a transgene. 5. The method of claim 4, wherein said transgene comprises nucleic acid sequence coding for thymidine kinase enzyme. 6. The method of claim 1, wherein said cancer comprises glioma. 7. The method of claim 1, further comprising administering to said human patient a cytotoxic chemotherapeutic. 8. The method of claim 6, further comprising administering to said human patient temozolomide. 9. In a method of treating cancer in a human, the improvement comprising:
a. Inducing an immune response against a viral vector, and then b. Administering to said human said viral vector. 10. The method of claim 9, wherein said administering to said human said viral vector comprises administering to a cavity created by a resection of said tumor. 11. The method of claim 9, wherein said viral vector comprises adenovirus. 12. The method of claim 9, wherein said viral vector comprises a transgene. 13. The method of claim 12, wherein said transgene comprises nucleic acid sequence coding for thymidine kinase enzyme. 14. The method of claim 9, wherein said cancer comprises glioma. 15. The method of claim 9, further comprising administering to said human patient a cytotoxic chemotherapeutic. 14. The method of claim 13, further comprising administering to said human patient temozolomide. | 1,600 |
621 | 14,415,434 | 1,617 | The present disclosure relates generally to crosslinked cation-binding polymers comprising monomers containing carboxylic acid groups and pKa decreasing groups, wherein the polymer contains i) calcium cations that are counterions to about 5% to about 75% of the carboxylic acid groups in the polymer; or ii) calcium cations and magnesium cations that are counterions to about 5% to about 75% of the carboxylic acid groups in the polymer, wherein the magnesium cations are counterions to no more than about 35% of the carboxylate groups in the polymer. The present disclosure also relates to methods of preparation of the poly mers, and compositions, formulations, and dosage forms containing the polymers, and methods of using the polymers, compositions, formulations, and/or dosage forms to treat various diseases or disorders. | 1. A crosslinked cation-binding polymer comprising:
a. monomers that comprise carboxylate groups and pKa decreasing groups; and b. calcium cations; wherein the calcium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 2. The composition of claim 1, wherein the polymer is crosslinked with about 4.0 mol % to about 20.0 mol % of one or more crosslinkers. 3. The composition of claim 2, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 4. The composition of claim 1, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 5. The composition of claim 4, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 6. The crosslinked cation-binding polymer of claim 1, wherein the pKa-decreasing group is an electron-withdrawing substituent. 7. The crosslinked cation-binding polymer of claim 1, wherein the electron-withdrawing substituent is located adjacent to the carboxylic acid group of the monomer. 8. The crosslinked cation-binding polymer of claim 1, wherein the electron-withdrawing substituent is located in the alpha or beta position of the carboxylic acid group of the monomer. 9. The crosslinked cation-binding polymer of claim 1, wherein the electron-withdrawing substituent is a hydroxyl group, an ethereal group, an ester group or a halide atom. 10. The crosslinked cation-binding polymer of claim 9, wherein the halide atom is fluorine (F). 11. The polymer of claim 1, wherein the calcium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of of the carboxylate groups in said polymer. 12. The polymer of claim 1, wherein counterions that are not calcium or sodium are hydrogen. 13. The polymer of any of claims 1-12, wherein the counterions to said carboxylate groups consist of said calcium counterions and hydrogen counterions. 14. A crosslinked cation-binding polymer comprising:
a. monomers that comprise carboxylate groups and pkA decreasing groups; and b. calcium cations and magnesium cations, wherein the calcium cations and magnesium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, wherein the magnesium cations are counterions to no more than about 35% of the carboxylate groups in the polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 15. The composition of claim 14, wherein the polymer is crosslinked with about 5.0 mol % to about 20.0 mol % of one or more crosslinkers. 16. The composition of claim 15, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 17. The composition of claim 14, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 18. The composition of claim 17, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 19. The crosslinked cation-binding polymer of claim 14, wherein the pKa-decreasing group is an electron-withdrawing substituent. 20. The crosslinked cation-binding polymer of claim 14, wherein the electron-withdrawing substituent is located adjacent to the carboxylic acid group of the monomer. 21. The crosslinked cation-binding polymer of claim 14, wherein the electron-withdrawing substituent is located in the alpha or beta position of the carboxylic acid group of the monomer. 22. The crosslinked cation-binding polymer of claim 14, wherein the electron-withdrawing substituent is a hydroxyl group, an ethereal group, an ester group or a halide atom. 23. The crosslinked cation-binding polymer of claim 22, wherein the halide atom is fluorine (F). 24. The polymer of claim 14, wherein the calcium cations and magnesium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of the carboxylate groups in the polymer. 25. The polymer of claim 14, wherein counterions that are not calcium, magnesium or sodium are hydrogen. 26. The polymer of any of claims 14-25, wherein the counterions to said carboxylate groups consist of said calcium counterions, magnesium counterions and hydrogen counterions. 27. The polymer of any one of claims 1-26, wherein the monomer is acrylic acid, an acrylic acid derivative, or a salt thereof. 28. The polymer of any of claims 1-27, wherein the polymer is crosslinked with a crosslinker selected from diethelyeneglycol diacrylate (diacryl glycerol), triallylamine, tetraallyloxyethane, allylmethacrylate, 1,1,1-trimethylolpropane triacrylate (TMPTA), derivatives of TMPTA, divinyl benzene, 1,7-ocatadiene and divinyl glycol. 29. The polymer of claim 28, wherein the crosslinked polymer is derived from acrylic acid monomers and TMPTA. 30. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 212 microns to about 500 microns. 31. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 75 microns to about 150 microns. 32. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 100 microns. 33. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 75 microns or less. 34. A composition comprising the polymer of any of claims 1-33. 35. The composition of claim 34 further comprising an additional base. 36. The composition of claim 35, wherein the additional base is a pharmaceutically acceptable base, a salt thereof, or a combination thereof. 37. The composition of claim 35, wherein the additional base is selected from an alkali metal hydroxide, an alkali metal acetate, an alkali metal carbonate, an alkali metal bicarbonate, an alkali metal oxide, an alkaline earth metal hydroxide, an alkaline earth metal acetate, an alkaline earth metal carbonate, an alkaline earth metal bicarbonate, an alkaline earth metal oxide, an organic base, choline, lysine, arginine, histidine, an acetate, a butyrate, a propionate, a lactate, a succinate, a citrate, an isocitrate, a fumarate, a malate, a malonate, an oxaloacetate, a pyruvate, a phosphate, a carbonate, a bicarbonate, a benzoate, an oxide an oxalate, a hydroxide, an amine, a hydrogen citrate, calcium bicarbonate, calcium carbonate, calcium oxide, calcium hydroxide, magnesium oxide, magnesium hydroxide, magnesium carbonate, magnesium bicarbonate, aluminum carbonate, aluminum hydroxide, sodium bicarbonate, potassium citrate, and combinations thereof. 38. The composition of any of claims 34-37, wherein the polymer is a polyacrylate polymer. 39. The composition of any of claims 35-37, wherein the additional base is calcium carbonate. 40. A composition comprising a crosslinked polyacrylate polymer comprising acrylic acid repeat units that comprise carboxylic acid groups, pKa decreasing groups, and calcium cations, wherein said calcium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 41. The composition of claim 40, wherein the polymer is crosslinked with about 5.0 mol % to about 20.0 mol % of one or more crosslinkers. 42. The composition of claim 41, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 43. The composition of claim 40, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 44. The composition of claim 43, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 45. The composition of claim 40, wherein the calcium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of the carboxylate groups in the polymer. 46. The composition of any of claims 40-44, wherein the counterions to said carboxylate groups consist of said calcium counterions and hydrogen counterions. 47. A composition comprising a crosslinked polyacrylate polymer comprising acrylic acid repeat units that comprise carboxylic acid groups, pKa decreasing groups, and calcium cations and magnesium cations, wherein said calcium and magnesium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, wherein the magnesium cations are counterions to no more than about 35% of the carboxylate groups in the polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 48. The composition of claim 47, wherein the polymer is crosslinked with about 5.0 mol % to about 20.0 mol % of one or more crosslinkers. 49. The composition of claim 48, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 50. The composition of claim 47, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 51. The composition of claim 50, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 52. The composition of claim 47, wherein the calcium cations and magnesium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of the carboxylate groups in the polymer. 53. The composition of any of claims 47-51, wherein the counterions to said carboxylate groups consist of said calcium counterions, magnesium counterions and hydrogen counterions. 54. The composition of any one of claims 40-53, wherein remaining counterions to said carboxylate groups consist of hydrogen cations. 55. The composition of any one of claims 40-54 further comprising calcium carbonate. 56. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 212 microns to about 500 microns. 57. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 75 microns to about 150 microns. 58. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 100 microns. 59. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 75 microns or less. 60. A dosage form comprising the polymer of any of claims 1-33 or the composition of any of claims 34-59. 61. The dosage form of claim 60 further comprising an additional base. 62. The dosage form of claim 61, wherein the additional base is selected from an alkali metal hydroxide, an alkali metal acetate, an alkali metal carbonate, an alkali metal bicarbonate, an alkali metal oxide, an alkaline earth metal hydroxide, an alkaline earth metal acetate, an alkaline earth metal carbonate, an alkaline earth metal bicarbonate, an alkaline earth metal oxide, an organic base, choline, lysine, arginine, histidine, an acetate, a butyrate, a propionate, a lactate, a succinate, a citrate, an isocitrate, a fumarate, a malate, a malonate, an oxaloacetate, a pyruvate, a phosphate, a carbonate, a bicarbonate, a benzoate, an oxide, an oxalate, a hydroxide, an amine, a hydrogen citrate, calcium bicarbonate, calcium carbonate, calcium oxide, calcium hydroxide, magnesium oxide, magnesium hydroxide, magnesium carbonate, magnesium bicarbonate, aluminum carbonate, aluminum hydroxide, sodium bicarbonate, potassium citrate, and combinations thereof. 63. The dosage form of any of claims 60-62, further comprising one or more pharmaceutically acceptable excipients. 64. The dosage form of any of claims 60-63, wherein the dosage form is a tablet, a chewable tablet, a capsule, a suspension, an oral suspension, a powder, a gel block, a gel pack, a confection, a chocolate bar, a flavored bar, a pudding, or a sachet. 65. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 1 g to about 30 g of the polymer. 66. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 4 g to about 15 g of the polymer. 67. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 8 g to about 15 g of the polymer. 68. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 8 g of the polymer. 69. The dosage form of any of claims 60-64, wherein the dosage form is a capsule comprising about 0.1 g to about 1 g of the polymer. 70. The dosage form of any of claims 60-64, wherein the dosage form is a capsule containing about 0.25 g to about 0.75 g of the polymer. 71. The dosage form of any of claims 60-64, wherein the dosage form is a capsule comprising about 0.5 g of the polymer. 72. The dosage form of any of claims 60-64, wherein the dosage form is a tablet comprising about 0.1 g to about 1.0 g of the polymer. 73. The dosage form of any of claims 60-64, wherein the dosage form is a tablet comprising about 0.3 g to about 0.8 g of the polymer. 74. The dosage form of any of claims 60-64, wherein the dosage form is a sachet, flavored bar, gel block, gel pack, pudding, or powder comprising about 1 g to about 30 g of the polymer. 75. The dosage form of any of claims 60-64, wherein the dosage form is a sachet, flavored bar, gel block, gel pack, pudding, or powder comprising about 4 g to about 20 g of the polymer. 76. The dosage form of any of claims 60-64, wherein the dosage form is a sachet, flavored bar, gel block, gel pack, pudding, or powder comprising about 4 g to about 8 g of the polymer. 77. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 0.04 g of the polymer per mL of suspension to about 1 g of the polymer per mL of suspension. 78. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 0.1 g of the polymer per mL of suspension to about 0.8 g of the polymer per mL of suspension. 79. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 0.3 g of the polymer per mL of suspension. 80. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 1 g to about 30 g of the polymer. 81. The dosage form of any of claims 77-80, wherein the suspension is an oral suspension. 82. The dosage form of any of claims 60-81, further comprising one or more additional agent. 83. The dosage form of claim 82, wherein the one or more additional agent is known to increase serum potassium. 84. The dosage form of claim 82, wherein the one or more additional agent is selected from a tertiary amine, spironolactone, fluoxetine, pyridinium and its derivatives, metoprolol, quinine, loperamide, chlorpheniramine, chlorpromazine, ephedrine, amitryptyline, imipramine, loxapine, cinnarizine, amiodarone, nortriptyline, a mineralocorticosteroid, propofol, digitalis, fluoride, succinylcholine, eplerenone, an alpha-adrenergic agonist, a RAAS inhibitor, an ACE inhibitor, an angiotensin II receptor blocker, a beta blocker, an aldosterone antagonist, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril, candesartan, eprosartan, irbesartan, losartan, valsartan, telmisartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, nadolol, propranolol, sotalol, timolol, canrenone, aliskiren, aldosterone synthesis inhibitors, VAP antagonists, amiloride, triamterine, a potassium supplement, heparin, a low molecular weight heparin, a non-steroidal anti-inflammatory drug, ketoconazole, trimethoprim, pentamide, a potassium sparing diuretic, amiloride, triamterene, and combinations thereof. 85. A method of treating hyperkalemia in a subject, the method comprising administering to said subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 86. A method of treating hyperkalemia in a subject, the method comprising:
a. identifying the subject as having, or as having a risk of developing, hyperkalemia; and b. administering to said subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 87. The method of claim 85 or 86, wherein the hyperkalemia is acute hyperkalemia or chronic hyperkalemia. 88. The method of claim 85 or 86, wherein the hyperkalemia is induced by a drug. 89. The method of claim 88, wherein the drug is a RAAS blocker, a beta blocker, trimethoprim, a renin inhibitor, an aldosterone synthase inhibitor, a non-steroidal anti-inflammatory drug, or heparin. 90. The method of claim 85 or 86, further comprising, after administering the polymer or composition, determining a potassium level in said subject, wherein the potassium level is within a normal potassium level range for the subject. 91. The method of any of claims 85-90, further comprising co-administering to said subject one or more of: mannitol, sorbitol, calcium acetate, sevelamer carbonate, sevelamer hydrochloride, a tertiary amine, spironolactone, fluoxetine, pyridinium and its derivatives, metoprolol, quinine, loperamide, chlorpheniramine, chlorpromazine, ephedrine, amitryptyline, imipramine, loxapine, cinnarizine, amiodarone, nortriptyline, a mineralocorticosteroid, propofol, digitalis, fluoride, succinylcholine, eplerenone, an alpha-adrenergic agonist, a RAAS inhibitor, an ACE inhibitor, an angiotensin II receptor blocker, a beta blocker, an aldosterone antagonist, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril, candesartan, eprosartan, irbesartan, losartan, valsartan, telmisartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, nadolol, propranolol, sotalol, timolol, canrenone, aliskiren, aldosterone synthesis inhibitors, VAP antagonists, amiloride, triamterine, a potassium supplement, heparin, a low molecular weight heparin, a non-steroidal anti-inflammatory drug, ketoconazole, trimethoprim, pentamide, a potassium sparing diuretic, amiloride, triamterene, and combinations thereof. 92. The method of any of claims 85-91, further comprising:
a. before administering the polymer or composition, determining a baseline level of potassium in the subject; and b. after administering the polymer or composition, determining a second level of potassium in the subject, wherein the second level of potassium is substantially less than said baseline level of potassium. 93. The method of any of claims 85-92, wherein an acid/base status associated with the subject does not significantly change within about 1 day of administration of the polymer or composition. 94. A method of treating heart failure in a subject, the method comprising administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 95. A method of treating heart failure in a subject, the method comprising:
a. identifying a subject as having heart failure; and b. administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 96. The method of claim 94 or 95 further comprising:
a. before administering said polymer or composition, determining one or more of: a baseline level of one or more ions in the subject, a baseline total body weight associated with the subject, a baseline total body water level associated with the subject, a baseline total extracellular water level associated with the subject, and a baseline total intracellular water level associated with the subject; and
b. after administering said polymer or composition, determining one or more of: a second level of one or more ions in the subject, a second total body weight associated with the subject, a second total body water level associated with the subject, a second total extracellular water level associated with the subject, and a second total intracellular water level associated with the subject,
wherein the second level is substantially lower than the baseline level. 97. The method of claim 96, wherein the one or more ions are selected from sodium, potassium, calcium, lithium, and magnesium. 98. The method of any of claims 94-97, wherein an acid/base status associated with said subject does not significantly change within about 1 day of administration of the polymer or composition. 99. The method of any of claims 94-98, wherein a blood pressure level associated with the subject after administration of the polymer or composition is substantially lower than a baseline blood pressure level associated with the subject before administration of the polymer or composition. 100. The method of claim 99, wherein the blood pressure level is one or more of: a systolic blood pressure level, a diastolic blood pressure level, and a mean arterial pressure level. 101. The method of any of claims 94-100, wherein a symptom of fluid overload associated with said subject, determined after administration of the polymer or composition, is reduced compared to a baseline level determined before administration of the composition. 102. The method of claim 101, wherein the symptom is one or more of: difficulty breathing when lying down, difficulty breathing with normal physical activity, ascites, fatigue, shortness of breath, increased body weight, peripheral edema, and pulmonary edema. 103. The method of any of claims 94-102, wherein the subject is on concomitant diuretic therapy. 104. The method of claim 103, wherein the diuretic therapy is reduced or discontinued after administration of the composition. 105. The method of any of claims 94-104, further comprising co-administering to the subject an agent known to increase serum potassium levels. 106. The method of claim 105, wherein the agent is one or more of: a tertiary amine, spironolactone, fluoxetine, pyridinium and its derivatives, metoprolol, quinine, loperamide, chlorpheniramine, chlorpromazine, ephedrine, amitryptyline, imipramine, loxapine, cinnarizine, amiodarone, nortriptyline, a mineralocorticosteroid, propofol, digitalis, fluoride, succinylcholine, eplerenone, an alpha-adrenergic agonist, a RAAS inhibitor, an ACE inhibitor, an angiotensin II receptor blocker, a beta blocker, an aldosterone antagonist, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril, candesartan, eprosartan, irbesartan, losartan, valsartan, telmisartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, nadolol, propranolol, sotalol, timolol, canrenone, aliskiren, aldosterone synthesis inhibitors, VAP antagonists, amiloride, triamterine, a potassium supplement, heparin, a low molecular weight heparin, a non-steroidal anti-inflammatory drug, ketoconazole, trimethoprim, pentamide, a potassium sparing diuretic, amiloride, triamterene, and combinations thereof. 107. The method of claim 106, wherein a dose of the agent is increased after administration of the polymer or composition. 108. The method of any of claims 94-107, wherein the subject is co-administered a blood pressure medication. 109. The method of claim 108, wherein a dose of the blood pressure medication is reduced after administration of the polymer or composition. 110. A method of treating chronic kidney disease in a subject, the method comprising administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 111. A method of treating chronic kidney disease in a subject, the method comprising:
a. identifying the subject as having a chronic kidney disease; and b. administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 112. The method of claim 110 or 111, wherein a symptom of fluid overload is reduced after administration of the polymer or composition. 113. The method of claim 112, wherein the symptom is one or more of: difficulty breathing at rest, difficulty breathing during normal physical activity, edema, pulmonary edema, hypertension, peripheral edema, leg edema, ascites, and/or increased body weight. 114. The method of claim any of claims 110-113, wherein a blood pressure level associated with the subject after administration of the polymer or composition is substantially lower than a baseline blood pressure level associated with the subject before administration of the composition. 115. The method of claim 114, wherein the blood pressure level is one or more of:
a systolic blood pressure level, a diastolic blood pressure level, and a mean arterial pressure level. 116. The method of any of claims 110-115, wherein a co-morbidity of chronic kidney disease is reduced or alleviated after administration of the composition. 117. The method of claim 116, wherein the co-morbidity is one or more of: fluid overload, edema, pulmonary edema, hypertension, hyperkalemia, excess total body sodium, and uremia. 118. The method of any of claims 110-117, further comprising:
a. before administering the polymer or composition, determining one or more of: a baseline level of one or more ions in the subject, a baseline total body weight associated with the subject, a baseline total body water level associated with the subject, a baseline total extracellular water level associated with the subject, and a baseline total intracellular water level associated with the subject; and b. after administering the polymer or composition, determining one or more of: a second level of said one or more ions in the subject, a second total body weight associated with the subject, a second total body water level associated with the subject, a second total extracellular water level associated with the subject, and a second total intracellular water level associated with the subject wherein the second level is substantially less than the baseline level. 119. The method of claim 118, wherein the one or more ions are selected from sodium, potassium, calcium, lithium, magnesium, and ammonium. 120. The method of any of claims 110-119, wherein an acid/base status associated with said subject does not significantly change within about 1 day of administration of the polymer or composition. 121. A method of treating a disease or disorder in a subject, the method comprising administering to the subject the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 122. A method of treating a disease or disorder in a subject, the method comprising:
a. identifying a disease or a disorder in the subject, or identifying a risk that the subject will develop a disease or disorder; and b. administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 123. The method of claim 121 or 122, wherein the disease or disorder is one or more of: heart failure, a renal insufficiency disease, end stage renal disease, liver cirrhosis, chronic renal insufficiency, chronic kidney disease, fluid overload, fluid maldistribution, edema, pulmonary edema, peripheral edema, lymphedema, nephrotic edema, idiopathic edema, ascites, cirrhotic ascites, interdialytic weight gain, high blood pressure, hyperkalemia, hypernatremia, abnormally high total body sodium, hypercalcemia, tumor lysis syndrome, head trauma, an adrenal disease, hyporeninemic hypoaldosteronism, hypertension, salt-sensitive hypertension, refractory hypertension, renal tubular disease, rhabdomyolysis, crush injuries, renal failure, acute tubular necrosis, insulin insufficiency, hyperkalemic periodic paralysis, hemolysis, malignant hyperthermia, pulmonary edema secondary to cardiogenic pathophysiology, pulmonary edema with non-cardiogenic origin, drowning, acute glomerulonephritis, allergic pulmonary edema, high altitude sickness, Adult Respiratory Distress Syndrome, traumatic edema, cardiogenic edema, acute hemorrhagic edema, heatstroke edema, facial edema, eyelid edema, angioedema, cerebral edema, scleral edema, nephritis, nephrosis, nephrotic syndrome, glomerulonephritis, and/or renal vein thrombosis. 124. The method of any of claims 121-123, wherein said polymer or composition is administered from 1 time every 3 days to about 4 times per day. 125. The method of any of claims 121-123, wherein said polymer or composition is administered from 1 to 4 times per day. 126. The method of any of claims 121-123, wherein said polymer or composition is administered from 1 to 2 times per day. | The present disclosure relates generally to crosslinked cation-binding polymers comprising monomers containing carboxylic acid groups and pKa decreasing groups, wherein the polymer contains i) calcium cations that are counterions to about 5% to about 75% of the carboxylic acid groups in the polymer; or ii) calcium cations and magnesium cations that are counterions to about 5% to about 75% of the carboxylic acid groups in the polymer, wherein the magnesium cations are counterions to no more than about 35% of the carboxylate groups in the polymer. The present disclosure also relates to methods of preparation of the poly mers, and compositions, formulations, and dosage forms containing the polymers, and methods of using the polymers, compositions, formulations, and/or dosage forms to treat various diseases or disorders.1. A crosslinked cation-binding polymer comprising:
a. monomers that comprise carboxylate groups and pKa decreasing groups; and b. calcium cations; wherein the calcium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 2. The composition of claim 1, wherein the polymer is crosslinked with about 4.0 mol % to about 20.0 mol % of one or more crosslinkers. 3. The composition of claim 2, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 4. The composition of claim 1, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 5. The composition of claim 4, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 6. The crosslinked cation-binding polymer of claim 1, wherein the pKa-decreasing group is an electron-withdrawing substituent. 7. The crosslinked cation-binding polymer of claim 1, wherein the electron-withdrawing substituent is located adjacent to the carboxylic acid group of the monomer. 8. The crosslinked cation-binding polymer of claim 1, wherein the electron-withdrawing substituent is located in the alpha or beta position of the carboxylic acid group of the monomer. 9. The crosslinked cation-binding polymer of claim 1, wherein the electron-withdrawing substituent is a hydroxyl group, an ethereal group, an ester group or a halide atom. 10. The crosslinked cation-binding polymer of claim 9, wherein the halide atom is fluorine (F). 11. The polymer of claim 1, wherein the calcium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of of the carboxylate groups in said polymer. 12. The polymer of claim 1, wherein counterions that are not calcium or sodium are hydrogen. 13. The polymer of any of claims 1-12, wherein the counterions to said carboxylate groups consist of said calcium counterions and hydrogen counterions. 14. A crosslinked cation-binding polymer comprising:
a. monomers that comprise carboxylate groups and pkA decreasing groups; and b. calcium cations and magnesium cations, wherein the calcium cations and magnesium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, wherein the magnesium cations are counterions to no more than about 35% of the carboxylate groups in the polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 15. The composition of claim 14, wherein the polymer is crosslinked with about 5.0 mol % to about 20.0 mol % of one or more crosslinkers. 16. The composition of claim 15, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 17. The composition of claim 14, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 18. The composition of claim 17, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 19. The crosslinked cation-binding polymer of claim 14, wherein the pKa-decreasing group is an electron-withdrawing substituent. 20. The crosslinked cation-binding polymer of claim 14, wherein the electron-withdrawing substituent is located adjacent to the carboxylic acid group of the monomer. 21. The crosslinked cation-binding polymer of claim 14, wherein the electron-withdrawing substituent is located in the alpha or beta position of the carboxylic acid group of the monomer. 22. The crosslinked cation-binding polymer of claim 14, wherein the electron-withdrawing substituent is a hydroxyl group, an ethereal group, an ester group or a halide atom. 23. The crosslinked cation-binding polymer of claim 22, wherein the halide atom is fluorine (F). 24. The polymer of claim 14, wherein the calcium cations and magnesium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of the carboxylate groups in the polymer. 25. The polymer of claim 14, wherein counterions that are not calcium, magnesium or sodium are hydrogen. 26. The polymer of any of claims 14-25, wherein the counterions to said carboxylate groups consist of said calcium counterions, magnesium counterions and hydrogen counterions. 27. The polymer of any one of claims 1-26, wherein the monomer is acrylic acid, an acrylic acid derivative, or a salt thereof. 28. The polymer of any of claims 1-27, wherein the polymer is crosslinked with a crosslinker selected from diethelyeneglycol diacrylate (diacryl glycerol), triallylamine, tetraallyloxyethane, allylmethacrylate, 1,1,1-trimethylolpropane triacrylate (TMPTA), derivatives of TMPTA, divinyl benzene, 1,7-ocatadiene and divinyl glycol. 29. The polymer of claim 28, wherein the crosslinked polymer is derived from acrylic acid monomers and TMPTA. 30. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 212 microns to about 500 microns. 31. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 75 microns to about 150 microns. 32. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 100 microns. 33. The polymer of any one of claims 1-29, wherein no less than about 70% of the polymer has a particle size of about 75 microns or less. 34. A composition comprising the polymer of any of claims 1-33. 35. The composition of claim 34 further comprising an additional base. 36. The composition of claim 35, wherein the additional base is a pharmaceutically acceptable base, a salt thereof, or a combination thereof. 37. The composition of claim 35, wherein the additional base is selected from an alkali metal hydroxide, an alkali metal acetate, an alkali metal carbonate, an alkali metal bicarbonate, an alkali metal oxide, an alkaline earth metal hydroxide, an alkaline earth metal acetate, an alkaline earth metal carbonate, an alkaline earth metal bicarbonate, an alkaline earth metal oxide, an organic base, choline, lysine, arginine, histidine, an acetate, a butyrate, a propionate, a lactate, a succinate, a citrate, an isocitrate, a fumarate, a malate, a malonate, an oxaloacetate, a pyruvate, a phosphate, a carbonate, a bicarbonate, a benzoate, an oxide an oxalate, a hydroxide, an amine, a hydrogen citrate, calcium bicarbonate, calcium carbonate, calcium oxide, calcium hydroxide, magnesium oxide, magnesium hydroxide, magnesium carbonate, magnesium bicarbonate, aluminum carbonate, aluminum hydroxide, sodium bicarbonate, potassium citrate, and combinations thereof. 38. The composition of any of claims 34-37, wherein the polymer is a polyacrylate polymer. 39. The composition of any of claims 35-37, wherein the additional base is calcium carbonate. 40. A composition comprising a crosslinked polyacrylate polymer comprising acrylic acid repeat units that comprise carboxylic acid groups, pKa decreasing groups, and calcium cations, wherein said calcium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 41. The composition of claim 40, wherein the polymer is crosslinked with about 5.0 mol % to about 20.0 mol % of one or more crosslinkers. 42. The composition of claim 41, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 43. The composition of claim 40, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 44. The composition of claim 43, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 45. The composition of claim 40, wherein the calcium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of the carboxylate groups in the polymer. 46. The composition of any of claims 40-44, wherein the counterions to said carboxylate groups consist of said calcium counterions and hydrogen counterions. 47. A composition comprising a crosslinked polyacrylate polymer comprising acrylic acid repeat units that comprise carboxylic acid groups, pKa decreasing groups, and calcium cations and magnesium cations, wherein said calcium and magnesium cations are counterions to about 5% to about 75% of the carboxylate groups in said polymer, wherein the magnesium cations are counterions to no more than about 35% of the carboxylate groups in the polymer, and wherein the polymer optionally comprises no more than about 5% sodium cations as counterions to the carboxylate groups in said polymer. 48. The composition of claim 47, wherein the polymer is crosslinked with about 5.0 mol % to about 20.0 mol % of one or more crosslinkers. 49. The composition of claim 48, wherein the polymer is crosslinked with about 4.0 mol % to about 10.0 mol %, 4.0 mol % to about 15.0 mol %, 8.0 mol % to about 10.0 mol %, 8.0 mol % to about 15.0 mol %, 8.0 mol % to about 20.0 mol %, or 12.0 mol % to about 20.0 mol % of one or more crosslinkers. 50. The composition of claim 47, wherein the polymer is crosslinked with about 0.025 mol % to about 3.0 mol % of one or more crosslinkers. 51. The composition of claim 50, wherein the polymer is crosslinked with about 0.025 mol % to about 0.3 mol %, about 0.025 mol % to about 0.17 mol %, about 0.025 mol % to about 0.34 mol %, or about 0.08 mol % to about 0.2 mol % of one or more crosslinkers. 52. The composition of claim 47, wherein the calcium cations and magnesium cations are counterions to about 5% to about 10%, about 5% to about 15%, about 5% to about 20%, about 5% to about 25%, about 5% to about 30%, about 5% to about 35%, about 5% to about 40%, about 5% to about 45%, about 5% to about 50%, about 5% to about 55%, about 5% to about 60%, about 5% to about 65%, about 5% to about 70%, about 5% to about 75%, about 10% to about 15%, about 10% to about 20%, about 10% to about 25%, about 10% to about 30%, about 10% to about 35%, about 10% to about 40%, about 10% to about 45%, about 10% to about 50%, about 10% to about 55%, about 10% to about 60%, about 10% to about 65%, about 10% to about 70%, about 10% to about 75%, about 15% to about 20%, about 15% to about 25%, about 15% to about 30%, about 15% to about 35%, about 15% to about 40%, about 15% to about 45%, about 15% to about 50%, about 15% to about 55%, about 15% to about 60%, about 15% to about 65%, about 15% to about 70%, about 15% to about 75%, about 20% to about 25%, about 20% to about 30%, about 20% to about 35%, about 20% to about 40%, about 20% to about 45%, about 20% to about 50%, about 20% to about 55%, about 20% to about 60%, about 20% to about 65%, about 20% to about 70%, about 20% to about 75%, about 25% to about 30%, about 25% to about 35%, about 25% to about 40%, about 25% to about 45%, about 25% to about 50%, about 25% to about 55%, about 25% to about 60%, about 25% to about 65%, about 25% to about 70%, about 25% to about 75%, about 30% to about 35%, about 30% to about 40%, about 30% to about 45%, about 30% to about 50%, about 30% to about 55%, about 30% to about 60%, about 30% to about 65%, about 30% to about 70%, about 30% to about 75%, about 35% to about 40%, about 35% to about 45%, about 35% to about 50%, about 35% to about 55%, about 35% to about 60%, about 35% to about 65%, about 35% to about 70%, about 35% to about 75%, about 40% to about 45%, about 40% to about 50%, about 40% to about 55%, about 40% to about 60%, about 40% to about 65%, about 40% to about 70%, about 40% to about 75%, about 45% to about 50%, about 45% to about 55%, about 45% to about 60%, about 45% to about 65%, about 45% to about 70%, about 45% to about 75%, about 50% to about 55%, about 50% to about 60%, about 50% to about 65%, about 50% to about 70%, about 50% to about 75%, about 55% to about 60%, about 55% to about 65%, about 55% to about 70%, about 55% to about 75%, about 60% to about 65%, about 60% to about 70%, about 60% to about 75%, about 65% to about 70%, about 65% to about 75% or about 70% to about 75% of the carboxylate groups in the polymer. 53. The composition of any of claims 47-51, wherein the counterions to said carboxylate groups consist of said calcium counterions, magnesium counterions and hydrogen counterions. 54. The composition of any one of claims 40-53, wherein remaining counterions to said carboxylate groups consist of hydrogen cations. 55. The composition of any one of claims 40-54 further comprising calcium carbonate. 56. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 212 microns to about 500 microns. 57. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 75 microns to about 150 microns. 58. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 100 microns. 59. The composition of any one of claims 40-55, wherein no less than about 70% of the polymer has a particle size of about 75 microns or less. 60. A dosage form comprising the polymer of any of claims 1-33 or the composition of any of claims 34-59. 61. The dosage form of claim 60 further comprising an additional base. 62. The dosage form of claim 61, wherein the additional base is selected from an alkali metal hydroxide, an alkali metal acetate, an alkali metal carbonate, an alkali metal bicarbonate, an alkali metal oxide, an alkaline earth metal hydroxide, an alkaline earth metal acetate, an alkaline earth metal carbonate, an alkaline earth metal bicarbonate, an alkaline earth metal oxide, an organic base, choline, lysine, arginine, histidine, an acetate, a butyrate, a propionate, a lactate, a succinate, a citrate, an isocitrate, a fumarate, a malate, a malonate, an oxaloacetate, a pyruvate, a phosphate, a carbonate, a bicarbonate, a benzoate, an oxide, an oxalate, a hydroxide, an amine, a hydrogen citrate, calcium bicarbonate, calcium carbonate, calcium oxide, calcium hydroxide, magnesium oxide, magnesium hydroxide, magnesium carbonate, magnesium bicarbonate, aluminum carbonate, aluminum hydroxide, sodium bicarbonate, potassium citrate, and combinations thereof. 63. The dosage form of any of claims 60-62, further comprising one or more pharmaceutically acceptable excipients. 64. The dosage form of any of claims 60-63, wherein the dosage form is a tablet, a chewable tablet, a capsule, a suspension, an oral suspension, a powder, a gel block, a gel pack, a confection, a chocolate bar, a flavored bar, a pudding, or a sachet. 65. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 1 g to about 30 g of the polymer. 66. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 4 g to about 15 g of the polymer. 67. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 8 g to about 15 g of the polymer. 68. The dosage form of any of claims 60-64, wherein the dosage form is a sachet comprising about 8 g of the polymer. 69. The dosage form of any of claims 60-64, wherein the dosage form is a capsule comprising about 0.1 g to about 1 g of the polymer. 70. The dosage form of any of claims 60-64, wherein the dosage form is a capsule containing about 0.25 g to about 0.75 g of the polymer. 71. The dosage form of any of claims 60-64, wherein the dosage form is a capsule comprising about 0.5 g of the polymer. 72. The dosage form of any of claims 60-64, wherein the dosage form is a tablet comprising about 0.1 g to about 1.0 g of the polymer. 73. The dosage form of any of claims 60-64, wherein the dosage form is a tablet comprising about 0.3 g to about 0.8 g of the polymer. 74. The dosage form of any of claims 60-64, wherein the dosage form is a sachet, flavored bar, gel block, gel pack, pudding, or powder comprising about 1 g to about 30 g of the polymer. 75. The dosage form of any of claims 60-64, wherein the dosage form is a sachet, flavored bar, gel block, gel pack, pudding, or powder comprising about 4 g to about 20 g of the polymer. 76. The dosage form of any of claims 60-64, wherein the dosage form is a sachet, flavored bar, gel block, gel pack, pudding, or powder comprising about 4 g to about 8 g of the polymer. 77. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 0.04 g of the polymer per mL of suspension to about 1 g of the polymer per mL of suspension. 78. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 0.1 g of the polymer per mL of suspension to about 0.8 g of the polymer per mL of suspension. 79. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 0.3 g of the polymer per mL of suspension. 80. The dosage form of any of claims 60-64, wherein the dosage form is a suspension comprising about 1 g to about 30 g of the polymer. 81. The dosage form of any of claims 77-80, wherein the suspension is an oral suspension. 82. The dosage form of any of claims 60-81, further comprising one or more additional agent. 83. The dosage form of claim 82, wherein the one or more additional agent is known to increase serum potassium. 84. The dosage form of claim 82, wherein the one or more additional agent is selected from a tertiary amine, spironolactone, fluoxetine, pyridinium and its derivatives, metoprolol, quinine, loperamide, chlorpheniramine, chlorpromazine, ephedrine, amitryptyline, imipramine, loxapine, cinnarizine, amiodarone, nortriptyline, a mineralocorticosteroid, propofol, digitalis, fluoride, succinylcholine, eplerenone, an alpha-adrenergic agonist, a RAAS inhibitor, an ACE inhibitor, an angiotensin II receptor blocker, a beta blocker, an aldosterone antagonist, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril, candesartan, eprosartan, irbesartan, losartan, valsartan, telmisartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, nadolol, propranolol, sotalol, timolol, canrenone, aliskiren, aldosterone synthesis inhibitors, VAP antagonists, amiloride, triamterine, a potassium supplement, heparin, a low molecular weight heparin, a non-steroidal anti-inflammatory drug, ketoconazole, trimethoprim, pentamide, a potassium sparing diuretic, amiloride, triamterene, and combinations thereof. 85. A method of treating hyperkalemia in a subject, the method comprising administering to said subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 86. A method of treating hyperkalemia in a subject, the method comprising:
a. identifying the subject as having, or as having a risk of developing, hyperkalemia; and b. administering to said subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 87. The method of claim 85 or 86, wherein the hyperkalemia is acute hyperkalemia or chronic hyperkalemia. 88. The method of claim 85 or 86, wherein the hyperkalemia is induced by a drug. 89. The method of claim 88, wherein the drug is a RAAS blocker, a beta blocker, trimethoprim, a renin inhibitor, an aldosterone synthase inhibitor, a non-steroidal anti-inflammatory drug, or heparin. 90. The method of claim 85 or 86, further comprising, after administering the polymer or composition, determining a potassium level in said subject, wherein the potassium level is within a normal potassium level range for the subject. 91. The method of any of claims 85-90, further comprising co-administering to said subject one or more of: mannitol, sorbitol, calcium acetate, sevelamer carbonate, sevelamer hydrochloride, a tertiary amine, spironolactone, fluoxetine, pyridinium and its derivatives, metoprolol, quinine, loperamide, chlorpheniramine, chlorpromazine, ephedrine, amitryptyline, imipramine, loxapine, cinnarizine, amiodarone, nortriptyline, a mineralocorticosteroid, propofol, digitalis, fluoride, succinylcholine, eplerenone, an alpha-adrenergic agonist, a RAAS inhibitor, an ACE inhibitor, an angiotensin II receptor blocker, a beta blocker, an aldosterone antagonist, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril, candesartan, eprosartan, irbesartan, losartan, valsartan, telmisartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, nadolol, propranolol, sotalol, timolol, canrenone, aliskiren, aldosterone synthesis inhibitors, VAP antagonists, amiloride, triamterine, a potassium supplement, heparin, a low molecular weight heparin, a non-steroidal anti-inflammatory drug, ketoconazole, trimethoprim, pentamide, a potassium sparing diuretic, amiloride, triamterene, and combinations thereof. 92. The method of any of claims 85-91, further comprising:
a. before administering the polymer or composition, determining a baseline level of potassium in the subject; and b. after administering the polymer or composition, determining a second level of potassium in the subject, wherein the second level of potassium is substantially less than said baseline level of potassium. 93. The method of any of claims 85-92, wherein an acid/base status associated with the subject does not significantly change within about 1 day of administration of the polymer or composition. 94. A method of treating heart failure in a subject, the method comprising administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 95. A method of treating heart failure in a subject, the method comprising:
a. identifying a subject as having heart failure; and b. administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 96. The method of claim 94 or 95 further comprising:
a. before administering said polymer or composition, determining one or more of: a baseline level of one or more ions in the subject, a baseline total body weight associated with the subject, a baseline total body water level associated with the subject, a baseline total extracellular water level associated with the subject, and a baseline total intracellular water level associated with the subject; and
b. after administering said polymer or composition, determining one or more of: a second level of one or more ions in the subject, a second total body weight associated with the subject, a second total body water level associated with the subject, a second total extracellular water level associated with the subject, and a second total intracellular water level associated with the subject,
wherein the second level is substantially lower than the baseline level. 97. The method of claim 96, wherein the one or more ions are selected from sodium, potassium, calcium, lithium, and magnesium. 98. The method of any of claims 94-97, wherein an acid/base status associated with said subject does not significantly change within about 1 day of administration of the polymer or composition. 99. The method of any of claims 94-98, wherein a blood pressure level associated with the subject after administration of the polymer or composition is substantially lower than a baseline blood pressure level associated with the subject before administration of the polymer or composition. 100. The method of claim 99, wherein the blood pressure level is one or more of: a systolic blood pressure level, a diastolic blood pressure level, and a mean arterial pressure level. 101. The method of any of claims 94-100, wherein a symptom of fluid overload associated with said subject, determined after administration of the polymer or composition, is reduced compared to a baseline level determined before administration of the composition. 102. The method of claim 101, wherein the symptom is one or more of: difficulty breathing when lying down, difficulty breathing with normal physical activity, ascites, fatigue, shortness of breath, increased body weight, peripheral edema, and pulmonary edema. 103. The method of any of claims 94-102, wherein the subject is on concomitant diuretic therapy. 104. The method of claim 103, wherein the diuretic therapy is reduced or discontinued after administration of the composition. 105. The method of any of claims 94-104, further comprising co-administering to the subject an agent known to increase serum potassium levels. 106. The method of claim 105, wherein the agent is one or more of: a tertiary amine, spironolactone, fluoxetine, pyridinium and its derivatives, metoprolol, quinine, loperamide, chlorpheniramine, chlorpromazine, ephedrine, amitryptyline, imipramine, loxapine, cinnarizine, amiodarone, nortriptyline, a mineralocorticosteroid, propofol, digitalis, fluoride, succinylcholine, eplerenone, an alpha-adrenergic agonist, a RAAS inhibitor, an ACE inhibitor, an angiotensin II receptor blocker, a beta blocker, an aldosterone antagonist, benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril, candesartan, eprosartan, irbesartan, losartan, valsartan, telmisartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, nadolol, propranolol, sotalol, timolol, canrenone, aliskiren, aldosterone synthesis inhibitors, VAP antagonists, amiloride, triamterine, a potassium supplement, heparin, a low molecular weight heparin, a non-steroidal anti-inflammatory drug, ketoconazole, trimethoprim, pentamide, a potassium sparing diuretic, amiloride, triamterene, and combinations thereof. 107. The method of claim 106, wherein a dose of the agent is increased after administration of the polymer or composition. 108. The method of any of claims 94-107, wherein the subject is co-administered a blood pressure medication. 109. The method of claim 108, wherein a dose of the blood pressure medication is reduced after administration of the polymer or composition. 110. A method of treating chronic kidney disease in a subject, the method comprising administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 111. A method of treating chronic kidney disease in a subject, the method comprising:
a. identifying the subject as having a chronic kidney disease; and b. administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 112. The method of claim 110 or 111, wherein a symptom of fluid overload is reduced after administration of the polymer or composition. 113. The method of claim 112, wherein the symptom is one or more of: difficulty breathing at rest, difficulty breathing during normal physical activity, edema, pulmonary edema, hypertension, peripheral edema, leg edema, ascites, and/or increased body weight. 114. The method of claim any of claims 110-113, wherein a blood pressure level associated with the subject after administration of the polymer or composition is substantially lower than a baseline blood pressure level associated with the subject before administration of the composition. 115. The method of claim 114, wherein the blood pressure level is one or more of:
a systolic blood pressure level, a diastolic blood pressure level, and a mean arterial pressure level. 116. The method of any of claims 110-115, wherein a co-morbidity of chronic kidney disease is reduced or alleviated after administration of the composition. 117. The method of claim 116, wherein the co-morbidity is one or more of: fluid overload, edema, pulmonary edema, hypertension, hyperkalemia, excess total body sodium, and uremia. 118. The method of any of claims 110-117, further comprising:
a. before administering the polymer or composition, determining one or more of: a baseline level of one or more ions in the subject, a baseline total body weight associated with the subject, a baseline total body water level associated with the subject, a baseline total extracellular water level associated with the subject, and a baseline total intracellular water level associated with the subject; and b. after administering the polymer or composition, determining one or more of: a second level of said one or more ions in the subject, a second total body weight associated with the subject, a second total body water level associated with the subject, a second total extracellular water level associated with the subject, and a second total intracellular water level associated with the subject wherein the second level is substantially less than the baseline level. 119. The method of claim 118, wherein the one or more ions are selected from sodium, potassium, calcium, lithium, magnesium, and ammonium. 120. The method of any of claims 110-119, wherein an acid/base status associated with said subject does not significantly change within about 1 day of administration of the polymer or composition. 121. A method of treating a disease or disorder in a subject, the method comprising administering to the subject the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 122. A method of treating a disease or disorder in a subject, the method comprising:
a. identifying a disease or a disorder in the subject, or identifying a risk that the subject will develop a disease or disorder; and b. administering to the subject an effective amount of the polymer of any of claims 1-33, the composition of any of claims 34-59, or the dosage form of any of claims 60-84. 123. The method of claim 121 or 122, wherein the disease or disorder is one or more of: heart failure, a renal insufficiency disease, end stage renal disease, liver cirrhosis, chronic renal insufficiency, chronic kidney disease, fluid overload, fluid maldistribution, edema, pulmonary edema, peripheral edema, lymphedema, nephrotic edema, idiopathic edema, ascites, cirrhotic ascites, interdialytic weight gain, high blood pressure, hyperkalemia, hypernatremia, abnormally high total body sodium, hypercalcemia, tumor lysis syndrome, head trauma, an adrenal disease, hyporeninemic hypoaldosteronism, hypertension, salt-sensitive hypertension, refractory hypertension, renal tubular disease, rhabdomyolysis, crush injuries, renal failure, acute tubular necrosis, insulin insufficiency, hyperkalemic periodic paralysis, hemolysis, malignant hyperthermia, pulmonary edema secondary to cardiogenic pathophysiology, pulmonary edema with non-cardiogenic origin, drowning, acute glomerulonephritis, allergic pulmonary edema, high altitude sickness, Adult Respiratory Distress Syndrome, traumatic edema, cardiogenic edema, acute hemorrhagic edema, heatstroke edema, facial edema, eyelid edema, angioedema, cerebral edema, scleral edema, nephritis, nephrosis, nephrotic syndrome, glomerulonephritis, and/or renal vein thrombosis. 124. The method of any of claims 121-123, wherein said polymer or composition is administered from 1 time every 3 days to about 4 times per day. 125. The method of any of claims 121-123, wherein said polymer or composition is administered from 1 to 4 times per day. 126. The method of any of claims 121-123, wherein said polymer or composition is administered from 1 to 2 times per day. | 1,600 |
622 | 14,706,747 | 1,633 | Disclosed herein are methods and compositions for treating or preventing Huntington's Disease. | 1. A method of modifying a neuron in a subject with Huntington's Disease (HD), the method comprising administering a repressor of a mutant Htt (mHtt) allele to the subject such that the neuron is modified. 2. The method of claim 1, wherein the neuron is an HD neuron. 3. The method of claim 1, wherein the modification comprises reducing aggregation of Htt in the neuron; increasing energy metabolism in the neuron; reducing susceptibility to apoptosis in the neuron and combinations thereof. 4. The method of claim 3, wherein intracellular ATP levels are increased. 5. A method of preventing or reducing the formation of Htt aggregates in an HD neuron, the method comprising modifying the HD neuron according to the method of claim 2. 6. A method of increasing cellular activity in an HD neuron, the method comprising modifying the HD neuron according to the method of claim 3. 7. A method of reducing apoptosis in an HD neuron, the method comprising modifying the HD neuron according to the method of claim 3. 8. A method for reducing motor deficits in an HD subject in need thereof, the method comprising modifying an HD neuron in the subject according to the method of claim 2. 9. The method of claim 8, wherein the motor deficit comprises clasping. 10. The method of claim 1, further comprising detecting mHtt levels in the subject. 11. The method of claim 10, wherein the mHtt levels are detected in cerebrospinal fluid (CSF). 12. The method of claim 1, wherein the repressor comprises a zinc finger protein transcription factor (ZF)-TF), a TAL-effector domain transcription factor (TALE-TF), a CRISPR/Cas-transcription factor, a zinc finger nuclease (ZFN), TAL-effector domain nuclease (TALEN) or a CRISPR/Cas nuclease system. 13. The method of claim 1, wherein the repressor is delivered to the neuron as a protein or as polynucleotide. 14. The method of claim 13, wherein the polynucleotide comprises mRNA or an expression construct selected from the group consisting of a plasmid and a viral vector. 15. The method of claim 1, wherein the repressor reduces mHtt expression by at least 85%. 16. A kit comprising an Htt repressor and reagents for detecting and/or quantifying mHtt protein levels. | Disclosed herein are methods and compositions for treating or preventing Huntington's Disease.1. A method of modifying a neuron in a subject with Huntington's Disease (HD), the method comprising administering a repressor of a mutant Htt (mHtt) allele to the subject such that the neuron is modified. 2. The method of claim 1, wherein the neuron is an HD neuron. 3. The method of claim 1, wherein the modification comprises reducing aggregation of Htt in the neuron; increasing energy metabolism in the neuron; reducing susceptibility to apoptosis in the neuron and combinations thereof. 4. The method of claim 3, wherein intracellular ATP levels are increased. 5. A method of preventing or reducing the formation of Htt aggregates in an HD neuron, the method comprising modifying the HD neuron according to the method of claim 2. 6. A method of increasing cellular activity in an HD neuron, the method comprising modifying the HD neuron according to the method of claim 3. 7. A method of reducing apoptosis in an HD neuron, the method comprising modifying the HD neuron according to the method of claim 3. 8. A method for reducing motor deficits in an HD subject in need thereof, the method comprising modifying an HD neuron in the subject according to the method of claim 2. 9. The method of claim 8, wherein the motor deficit comprises clasping. 10. The method of claim 1, further comprising detecting mHtt levels in the subject. 11. The method of claim 10, wherein the mHtt levels are detected in cerebrospinal fluid (CSF). 12. The method of claim 1, wherein the repressor comprises a zinc finger protein transcription factor (ZF)-TF), a TAL-effector domain transcription factor (TALE-TF), a CRISPR/Cas-transcription factor, a zinc finger nuclease (ZFN), TAL-effector domain nuclease (TALEN) or a CRISPR/Cas nuclease system. 13. The method of claim 1, wherein the repressor is delivered to the neuron as a protein or as polynucleotide. 14. The method of claim 13, wherein the polynucleotide comprises mRNA or an expression construct selected from the group consisting of a plasmid and a viral vector. 15. The method of claim 1, wherein the repressor reduces mHtt expression by at least 85%. 16. A kit comprising an Htt repressor and reagents for detecting and/or quantifying mHtt protein levels. | 1,600 |
623 | 14,640,318 | 1,642 | The present invention is directed to methods and compositions for modulating proteins involved in the immune response. The present invention further provides methods and compositions for treatment of autoimmune disease and cancer by modulating the expression and activity of such proteins. | 1. A method for the treatment of cancer, the method comprising administering to a subject in need thereof a combination of a pharmaceutically effective amount of (i) at least one immune checkpoint inhibitor and (ii) an inhibitor of Toso activity. 2. The method of claim 1, wherein the at least one immune checkpoint inhibitor is a member selected from the group consisting of: a PD-1 inhibitor, a CTLA-4 inhibitor, a LAG3 inhibitor, and a TIM3 inhibitor. 3. The method of claim 1, wherein the inhibitor of Toso activity is a soluble Toso protein. 4. The method of claim 1, wherein the inhibitor of Toso activity is an antibody to Toso. 5. The method of claim 1, wherein the combination comprises a PD-1 inhibitor, a CTLA-4 inhibitor, and a soluble Toso protein. 6. The method of claim 5, wherein the PD-1 inhibitor is an anti-PD-1 antibody and the CTLA-4 inhibitor is an anti-CTLA-4 antibody. 7. A method for increasing a level of infiltrating lymphocytes in a tumor, the method comprising treating the tumor with a soluble Toso protein, wherein the level of infiltrating lymphocytes that results from the treating is higher than the level without the treating. 8. The method of claim 7, wherein the infiltrating lymphocytes comprise CD3+ cells, CD8+ cells, or a combination of CD3+ cells and CD8+ cells. 9. The method of claim 7, wherein the method further comprises treating the tumor with one or more of a member selected from a PD-1 inhibitor, a CTLA-4 inhibitor, and a PDL-1 inhibitor. 10. A method of dampening the immune response, said method comprising decreasing expression and/or activity of Toso. 11. A method according to claim 10, said method further comprising increasing expression and/or activity of IgM. 12. A method according to claim 10, said method further comprising decreasing expression and/or activity of PAMPs and/or DAMPs. 13. A method of treating autoimmune disease, said method comprising one or more of the following steps:
(a) decreasing the expression and/or activity of Toso; (b) increasing the expression and/or activity of IgM; and (c) decreasing the expression and/or activity of PAMPs and/or DAMPs. 14. A method of inducing the immune response, said method comprising increasing expression and/or activity of Toso. 15. A method according to claim 14, said method further comprising decreasing expression and/or activity of IgM. 16. A method according to claim 14, said method further comprising increasing expression and/or activity of PAMPs and/or DAMPs. 17. A method according to claim 14, said method further comprising modulating activity of a scramblase protein. 18. A method of treating cancer, said method comprising one or more of the following steps:
(a) increasing the expression and/or activity of Toso; (b) decreasing the expression and/or activity of IgM; (c) increasing the expression and/or activity of PAMPs and/or DAMPs; (d) modulating the activity of a scramblase protein. | The present invention is directed to methods and compositions for modulating proteins involved in the immune response. The present invention further provides methods and compositions for treatment of autoimmune disease and cancer by modulating the expression and activity of such proteins.1. A method for the treatment of cancer, the method comprising administering to a subject in need thereof a combination of a pharmaceutically effective amount of (i) at least one immune checkpoint inhibitor and (ii) an inhibitor of Toso activity. 2. The method of claim 1, wherein the at least one immune checkpoint inhibitor is a member selected from the group consisting of: a PD-1 inhibitor, a CTLA-4 inhibitor, a LAG3 inhibitor, and a TIM3 inhibitor. 3. The method of claim 1, wherein the inhibitor of Toso activity is a soluble Toso protein. 4. The method of claim 1, wherein the inhibitor of Toso activity is an antibody to Toso. 5. The method of claim 1, wherein the combination comprises a PD-1 inhibitor, a CTLA-4 inhibitor, and a soluble Toso protein. 6. The method of claim 5, wherein the PD-1 inhibitor is an anti-PD-1 antibody and the CTLA-4 inhibitor is an anti-CTLA-4 antibody. 7. A method for increasing a level of infiltrating lymphocytes in a tumor, the method comprising treating the tumor with a soluble Toso protein, wherein the level of infiltrating lymphocytes that results from the treating is higher than the level without the treating. 8. The method of claim 7, wherein the infiltrating lymphocytes comprise CD3+ cells, CD8+ cells, or a combination of CD3+ cells and CD8+ cells. 9. The method of claim 7, wherein the method further comprises treating the tumor with one or more of a member selected from a PD-1 inhibitor, a CTLA-4 inhibitor, and a PDL-1 inhibitor. 10. A method of dampening the immune response, said method comprising decreasing expression and/or activity of Toso. 11. A method according to claim 10, said method further comprising increasing expression and/or activity of IgM. 12. A method according to claim 10, said method further comprising decreasing expression and/or activity of PAMPs and/or DAMPs. 13. A method of treating autoimmune disease, said method comprising one or more of the following steps:
(a) decreasing the expression and/or activity of Toso; (b) increasing the expression and/or activity of IgM; and (c) decreasing the expression and/or activity of PAMPs and/or DAMPs. 14. A method of inducing the immune response, said method comprising increasing expression and/or activity of Toso. 15. A method according to claim 14, said method further comprising decreasing expression and/or activity of IgM. 16. A method according to claim 14, said method further comprising increasing expression and/or activity of PAMPs and/or DAMPs. 17. A method according to claim 14, said method further comprising modulating activity of a scramblase protein. 18. A method of treating cancer, said method comprising one or more of the following steps:
(a) increasing the expression and/or activity of Toso; (b) decreasing the expression and/or activity of IgM; (c) increasing the expression and/or activity of PAMPs and/or DAMPs; (d) modulating the activity of a scramblase protein. | 1,600 |
624 | 15,102,957 | 1,615 | Novel dry powder compositions comprising and methods relating thereto are provided. The dry powder compositions comprise PDE5 inhibitors, such as vardenafil, or pharmaceutically acceptable salts or esters thereof. The dry powder compositions may optionally include an carrier/excipient. The concentration of active agent may be at least about 2% by weight. Methods of aerosolizing the dry powder compositions and using them to treat various diseases are also disclosed. | 1. A powder pharmaceutical composition comprising a) at least about 2% by weight of a PDE5 inhibitor or a pharmaceutically acceptable salt or ester thereof relative to the total weight of the overall pharmaceutical composition, and b) at least one pharmaceutically acceptable carrier. 2. The powder pharmaceutical composition of claim 1, wherein the PDE5 inhibitor is at least one of vardenafil, sildenafil, tadalafil, avanafil, benzamidenafil, lodenafil, mirodenafil, udenafil, or zaprinast, or a pharmaceutically acceptable salt or ester thereof. 3. The powder pharmaceutical composition of claim 1, wherein the composition comprises at least about 2% to about 20% by weight of the PDE5 inhibitor. 4. The powder pharmaceutical composition of claim 1, wherein the composition comprises at least about 2% to about 20% by weight of vardenafil or a pharmaceutically acceptable salt or ester thereof. 5. The powder pharmaceutical composition of claim 1, wherein the at least one pharmaceutically acceptable carrier comprises lactose, mannitol, trehalose, or starch. 6. The powder pharmaceutical composition of claim 5, wherein the at least one pharmaceutically acceptable carrier comprises at least one of a mono-, di- or polysaccharide, or their derivatives, calcium stearate, magnesium stearate, leucine or its derivatives, lecithin, human serum albumin, polylysine, polyarginine, or other force control agents, or combinations thereof. 7. The powder pharmaceutical composition of claim 1, wherein the PDE5 inhibitor or a pharmaceutically acceptable salt or ester is micronized. 8. The powder pharmaceutical composition of claim 1, wherein the composition is packaged to have a nominal load of about 3 mg to 30 mg. 9. The powder pharmaceutical composition of claim 1, wherein the composition is packaged to have a nominal dose of at least about 0.25 mg. 10. The powder pharmaceutical composition of claim 1, wherein the composition is packaged to have a delivered dose of at least about 0.075 mg. 11. A method of aerosolizing a powder pharmaceutical composition comprising a) at least 2% by weight of a PDE5 inhibitor, or a pharmaceutically acceptable salt or ester thereof, relative to the total weight of the overall pharmaceutical composition, and b) at least one pharmaceutically acceptable carrier, the method comprising:
providing an inhaler comprising a dispersion chamber having an inlet and an outlet, the dispersion chamber containing an actuator that is movable reciprocatable along a longitudinal axis of the dispersion chamber; and inducing air flow through the outlet channel to cause air and the powder pharmaceutical composition to enter into the dispersion chamber from the inlet, and to cause the actuator to oscillate within the dispersion chamber to assist in dispersing the powder pharmaceutical composition from the outlet for delivery to a subject through the outlet. 12. The method of claim 11, wherein the PDE5 inhibitor is at least one of vardenafil, sildenafil, tadalafil, avanafil, benzamidenafil, lodenafil, mirodenafil, udenafil, or zaprinast, or a pharmaceutically acceptable salt or ester thereof. 13. The method of claim 11, wherein the composition comprises at least about 2% to about 20% by weight of the PDE5 inhibitor. 14. The method of claim 11, wherein the composition comprises at least about 2% to about 20% by weight of vardenafil or a pharmaceutically acceptable salt or ester thereof. 15. The method of claim 11, wherein the at least one pharmaceutically acceptable carrier comprises lactose, mannitol, trehalose, or starch. 16. The method of claim 11, wherein the at least one pharmaceutically acceptable carrier comprises at least one of a mono-, di- or poly-saccharide, or their derivatives, calcium stearate, magnesium stearate, leucine or its derivatives, lecithin, human serum albumin, polylysine, polyarginine, or other force control agents, or combinations thereof. 17. The method of claim 11, wherein the composition has a mass median aerodynamic diameter of between 0.5 μm and 5 μm upon aerosolization. 18. The method of claim 11, wherein the composition has a fine particle fraction of at least about 20% upon aerosolization. 19. The method of claim 11, wherein the composition has an emitted dose of at least about 40% upon aerosolization. 20. The method of claim 11, wherein the powdered medicament is stored within a storage compartment, and wherein the powder pharmaceutical composition is transferred from the storage compartment, through the inlet and into the dispersion chamber. 21. The method of claim 11, wherein the inlet is in fluid communication with an initial chamber, and wherein the powder pharmaceutical composition is received into the initial chamber prior to passing through the inlet and into the dispersion chamber. 22. A method of treating a disease in a subject in need thereof, the method comprising administering to the subject via a pulmonary route an effective amount of a powder pharmaceutical composition comprising a) at least about 2% of a PDE5 inhibitor, or a pharmaceutically acceptable salt or ester thereof, by weight relative to the total weight of the overall pharmaceutical composition dose, and b) at least one pharmaceutically acceptable carrier. 23. The method of claim 22, wherein the disease is a lung disease or a heart disease. 24. The method of claim 23, wherein the lung disease is pulmonary hypertension or cystic fibrosis. 25. The method of claim 23, wherein the heart disease is congestive heart failure. 26. The method of claim 22, wherein the powder pharmaceutical composition is administered as an aerosol. 27. The method of claim 22, wherein the powder pharmaceutical composition is administered using a dry powder inhaler or a metered dose inhaler. 28. The method of claim 22, wherein the powder pharmaceutical composition is administered by
providing an inhaler comprising a dispersion chamber having an inlet and an outlet, the dispersion chamber containing an actuator that is movable reciprocatable along a longitudinal axis of the dispersion chamber; and inducing air flow through the outlet channel to cause air and the powder pharmaceutical composition to enter into the dispersion chamber from the inlet, and to cause the actuator to oscillate within the dispersion chamber to assist in dispersing the powder pharmaceutical composition from the outlet for delivery to a subject through the outlet. 29. The method of claim 22, wherein the PDE5 inhibitor is at least one of vardenafil, sildenafil, tadalafil, avanafil, benzamidenafil, lodenafil, mirodenafil, udenafil, or zaprinast, or a pharmaceutically acceptable salt or ester thereof. 30. The method of claim 22, wherein the composition comprises at least about 2% to about 20% by weight of the PDE5 inhibitor. 31. The method of claim 22, wherein the composition comprises at least about 2% to about 20% by weight of vardenafil or a pharmaceutically acceptable salt or ester thereof. 32. The method of claim 22, wherein the composition further comprises at least one pharmaceutically acceptable salt, and wherein the at least one pharmaceutically acceptable carrier comprises lactose, mannitol, trehalose, or starch. 33. The method of claim 22, wherein the composition further comprises at least one pharmaceutically acceptable salt, and wherein the at least one pharmaceutically acceptable carrier comprises at least one of a mono-, di- or poly-saccharide, or their derivatives, calcium stearate, magnesium stearate, leucine or its derivatives, lecithin, human serum albumin, polylysine, polyarginine, or other force control agents, or combinations thereof. 34. The method of claim 22, wherein a delivered dose of about 0.25 mg to about 20 mg is delivered to the subject upon aerosolization. | Novel dry powder compositions comprising and methods relating thereto are provided. The dry powder compositions comprise PDE5 inhibitors, such as vardenafil, or pharmaceutically acceptable salts or esters thereof. The dry powder compositions may optionally include an carrier/excipient. The concentration of active agent may be at least about 2% by weight. Methods of aerosolizing the dry powder compositions and using them to treat various diseases are also disclosed.1. A powder pharmaceutical composition comprising a) at least about 2% by weight of a PDE5 inhibitor or a pharmaceutically acceptable salt or ester thereof relative to the total weight of the overall pharmaceutical composition, and b) at least one pharmaceutically acceptable carrier. 2. The powder pharmaceutical composition of claim 1, wherein the PDE5 inhibitor is at least one of vardenafil, sildenafil, tadalafil, avanafil, benzamidenafil, lodenafil, mirodenafil, udenafil, or zaprinast, or a pharmaceutically acceptable salt or ester thereof. 3. The powder pharmaceutical composition of claim 1, wherein the composition comprises at least about 2% to about 20% by weight of the PDE5 inhibitor. 4. The powder pharmaceutical composition of claim 1, wherein the composition comprises at least about 2% to about 20% by weight of vardenafil or a pharmaceutically acceptable salt or ester thereof. 5. The powder pharmaceutical composition of claim 1, wherein the at least one pharmaceutically acceptable carrier comprises lactose, mannitol, trehalose, or starch. 6. The powder pharmaceutical composition of claim 5, wherein the at least one pharmaceutically acceptable carrier comprises at least one of a mono-, di- or polysaccharide, or their derivatives, calcium stearate, magnesium stearate, leucine or its derivatives, lecithin, human serum albumin, polylysine, polyarginine, or other force control agents, or combinations thereof. 7. The powder pharmaceutical composition of claim 1, wherein the PDE5 inhibitor or a pharmaceutically acceptable salt or ester is micronized. 8. The powder pharmaceutical composition of claim 1, wherein the composition is packaged to have a nominal load of about 3 mg to 30 mg. 9. The powder pharmaceutical composition of claim 1, wherein the composition is packaged to have a nominal dose of at least about 0.25 mg. 10. The powder pharmaceutical composition of claim 1, wherein the composition is packaged to have a delivered dose of at least about 0.075 mg. 11. A method of aerosolizing a powder pharmaceutical composition comprising a) at least 2% by weight of a PDE5 inhibitor, or a pharmaceutically acceptable salt or ester thereof, relative to the total weight of the overall pharmaceutical composition, and b) at least one pharmaceutically acceptable carrier, the method comprising:
providing an inhaler comprising a dispersion chamber having an inlet and an outlet, the dispersion chamber containing an actuator that is movable reciprocatable along a longitudinal axis of the dispersion chamber; and inducing air flow through the outlet channel to cause air and the powder pharmaceutical composition to enter into the dispersion chamber from the inlet, and to cause the actuator to oscillate within the dispersion chamber to assist in dispersing the powder pharmaceutical composition from the outlet for delivery to a subject through the outlet. 12. The method of claim 11, wherein the PDE5 inhibitor is at least one of vardenafil, sildenafil, tadalafil, avanafil, benzamidenafil, lodenafil, mirodenafil, udenafil, or zaprinast, or a pharmaceutically acceptable salt or ester thereof. 13. The method of claim 11, wherein the composition comprises at least about 2% to about 20% by weight of the PDE5 inhibitor. 14. The method of claim 11, wherein the composition comprises at least about 2% to about 20% by weight of vardenafil or a pharmaceutically acceptable salt or ester thereof. 15. The method of claim 11, wherein the at least one pharmaceutically acceptable carrier comprises lactose, mannitol, trehalose, or starch. 16. The method of claim 11, wherein the at least one pharmaceutically acceptable carrier comprises at least one of a mono-, di- or poly-saccharide, or their derivatives, calcium stearate, magnesium stearate, leucine or its derivatives, lecithin, human serum albumin, polylysine, polyarginine, or other force control agents, or combinations thereof. 17. The method of claim 11, wherein the composition has a mass median aerodynamic diameter of between 0.5 μm and 5 μm upon aerosolization. 18. The method of claim 11, wherein the composition has a fine particle fraction of at least about 20% upon aerosolization. 19. The method of claim 11, wherein the composition has an emitted dose of at least about 40% upon aerosolization. 20. The method of claim 11, wherein the powdered medicament is stored within a storage compartment, and wherein the powder pharmaceutical composition is transferred from the storage compartment, through the inlet and into the dispersion chamber. 21. The method of claim 11, wherein the inlet is in fluid communication with an initial chamber, and wherein the powder pharmaceutical composition is received into the initial chamber prior to passing through the inlet and into the dispersion chamber. 22. A method of treating a disease in a subject in need thereof, the method comprising administering to the subject via a pulmonary route an effective amount of a powder pharmaceutical composition comprising a) at least about 2% of a PDE5 inhibitor, or a pharmaceutically acceptable salt or ester thereof, by weight relative to the total weight of the overall pharmaceutical composition dose, and b) at least one pharmaceutically acceptable carrier. 23. The method of claim 22, wherein the disease is a lung disease or a heart disease. 24. The method of claim 23, wherein the lung disease is pulmonary hypertension or cystic fibrosis. 25. The method of claim 23, wherein the heart disease is congestive heart failure. 26. The method of claim 22, wherein the powder pharmaceutical composition is administered as an aerosol. 27. The method of claim 22, wherein the powder pharmaceutical composition is administered using a dry powder inhaler or a metered dose inhaler. 28. The method of claim 22, wherein the powder pharmaceutical composition is administered by
providing an inhaler comprising a dispersion chamber having an inlet and an outlet, the dispersion chamber containing an actuator that is movable reciprocatable along a longitudinal axis of the dispersion chamber; and inducing air flow through the outlet channel to cause air and the powder pharmaceutical composition to enter into the dispersion chamber from the inlet, and to cause the actuator to oscillate within the dispersion chamber to assist in dispersing the powder pharmaceutical composition from the outlet for delivery to a subject through the outlet. 29. The method of claim 22, wherein the PDE5 inhibitor is at least one of vardenafil, sildenafil, tadalafil, avanafil, benzamidenafil, lodenafil, mirodenafil, udenafil, or zaprinast, or a pharmaceutically acceptable salt or ester thereof. 30. The method of claim 22, wherein the composition comprises at least about 2% to about 20% by weight of the PDE5 inhibitor. 31. The method of claim 22, wherein the composition comprises at least about 2% to about 20% by weight of vardenafil or a pharmaceutically acceptable salt or ester thereof. 32. The method of claim 22, wherein the composition further comprises at least one pharmaceutically acceptable salt, and wherein the at least one pharmaceutically acceptable carrier comprises lactose, mannitol, trehalose, or starch. 33. The method of claim 22, wherein the composition further comprises at least one pharmaceutically acceptable salt, and wherein the at least one pharmaceutically acceptable carrier comprises at least one of a mono-, di- or poly-saccharide, or their derivatives, calcium stearate, magnesium stearate, leucine or its derivatives, lecithin, human serum albumin, polylysine, polyarginine, or other force control agents, or combinations thereof. 34. The method of claim 22, wherein a delivered dose of about 0.25 mg to about 20 mg is delivered to the subject upon aerosolization. | 1,600 |
625 | 15,791,677 | 1,628 | wherein X, R1 and R2 are defined herein. Also provided are pharmaceutical compositions, kits and articles of manufacture comprising such compounds, methods and intermediates useful for making the compounds, and methods of using the compounds and compositions. | 1. A compound comprising:
a) the basic skeleton of an organic compound having two to eight five and/or six-membered rings provided that the basic skeleton is not the basic skeleton of argentatin, betulinic acid, lanostane, oleanic acid, boswellic acid, glycyrrhetinic acid, ursolic acid, or tricyclic-bis-enone; b) a structural unit of the formula:
wherein:
the carbon atoms labeled 1, 2 and 3 are part of a five or six-membered ring;
X is cyano or —C(O)Ra, wherein Ra is:
hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), alkoxy(C≤12), alkenyl-oxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkylamino(C≤12), heteroarylamino(C≤12), heteroaralkyl-amino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkyl-silyloxy(C≤12), or substituted versions of any of these groups; and
R1 and R2 are each independently:
hydrogen; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤8), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkoxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), hetero-aralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkyl-amino(C≤12), arylamino(C≤12), aralkylamino(C≤12), hetero-arylamino(C≤12), heteroaralkylamino(C≤12), amido(C≤12), or a substituted version of any of these groups; or
R1 and R2 are taken together and are alkanediyl(C≤12), alkenediyl(C≤12), alkanediyl(C≤12) or alkenediyl(C≤12); and
c) from 0 to 8 chemical groups attached to a carbon atom of the basic skeleton other than carbon atoms 1, 2, 3 or 4, wherein each chemical group is independently:
hydroxy, halo, oxo, amino, hydroxyamino, nitro, imino, cyano, azido, mercapto, or thio; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkylidene(C≤12), alkoxy(C≤12), alkenyloxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyl-oxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkyl-amino(C≤12), heteroarylamino(C≤12), heteroaralkylamino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkylimino(C≤12), alkenyl-imino(C≤12), alkynylimino(C≤12), arylimino(C≤12), aralkylimino(C≤12), heteroarylimino(C≤12), heteroaralkylimino(C≤12), acylimino(C≤12), alkylthio(C≤12), alkenylthio(C≤12), alkynylthio(C≤12), arylthio(C≤12), aralkylthio(C≤12), heteroarylthio(C≤12), heteroaralkylthio(C≤12), acyl-thio(C≤12), thioacyl(C≤12), alkylsulfonyl(C≤12), alkenylsulfonyl(C≤8), alkynylsulfonyl(C≤12), arylsulfonyl(C≤12), aralkylsulfonyl(C≤12), heteroarylsulfonyl(C≤12), heteroaralkylsulfonyl(C≤12), alkyl-ammonium(C≤12), alkylsulfonium(C≤12), alkylsilyl(C≤12), alkylsilyl-oxy(C≤12), or a substituted version of any of these groups;
or pharmaceutically acceptable salts, esters, hydrates, solvates, tautomers, acetals, ketals, prodrugs, or optical isomers thereof. 2. The compound of claim 1, wherein R1 and R2 are each independently hydrogen, alkyl(C≤8) or substituted alkyl(C≤8). 3-6. (canceled) 7. A compound comprising:
a) the basic skeleton of a natural product having two to eight rings, provided that the natural product is not argentatin, betulinic acid, lanostane, oleanic acid, or ursolic acid; b) a structural unit of the formula:
wherein:
the carbon atoms labeled 1, 2 and 3 are part of a six-membered ring;
X is cyano, fluoroalkyl(C≤8), substituted, fluoroalkyl(C≤8), or —C(O)Ra, wherein Ra is:
hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), alkoxy(C≤12), alkenyl-oxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkylamino(C≤12), heteroarylamino(C≤12), heteroaralkyl-amino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkyl-silyloxy(C≤12), or substituted versions of any of these groups; and
R1 and R2 are each independently:
hydrogen; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkoxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), hetero-aralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkyl-amino(C≤12), arylamino(C≤12), aralkylamino(C≤12), hetero-arylamino(C≤12), heteroaralkylamino(C≤12), amido(C≤12), or a substituted version of any of these groups; or
R1 and R2 are taken together and are alkanediyl(C≤12), alkenediyl(C≤12), alkanediyl(C≤12) or alkenediyl(C≤12); and
c) from 0 to 8 chemical groups attached to a carbon atom of the basic skeleton other than carbon atoms 1, 2, 3 or 4, wherein each chemical group is independently:
hydroxy, halo, oxo, amino, hydroxyamino, nitro, imino, cyano, azido, mercapto, or thio; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkylidene(C≤12), alkoxy(C≤12), alkenyloxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyl-oxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkyl-amino(C≤12), heteroarylamino(C≤12), heteroaralkylamino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkylimino(C≤12), alkenyl-imino(C≤12), alkynylimino(C≤12), arylimino(C≤12), aralkylimino(C≤12), heteroarylimino(C≤12), heteroaralkylimino(C≤12), acylimino(C≤12), alkylthio(C≤12), alkenylthio(C≤12), alkynylthio(C≤12), arylthio(C≤12), aralkylthio(C≤12), heteroarylthio(C≤12), heteroaralkylthio(C≤12), acyl-thio(C≤12), thioacyl(C≤8), alkylsulfonyl(C≤12), alkenylsulfonyl(C≤12), alkynylsulfonyl(C≤12), arylsulfonyl(C≤12), aralkylsulfonyl(C≤12), heteroarylsulfonyl(C≤12), heteroaralkylsulfonyl(C≤12), alkyl-ammonium(C≤12), alkylsulfonium(C≤12), alkylsilyl(C≤12), alkylsilyl-oxy(C≤12), or a substituted version of any of these groups;
or pharmaceutically acceptable salts, esters, hydrates, solvates, tautomers, acetals, ketals, prodrugs, or optical isomers thereof. 8. The compound of claim 7, wherein R1 and R2 are each independently hydrogen, alkyl(C≤8) or substituted alkyl(C≤8). 9-15. (canceled) 16. The compound of claim 7, wherein X is —CN. 17-18. (canceled) 19. The compound of claim 16, wherein the structural unit is further defined as: 20-173. (canceled) 174. A method of making a compound of claim 1, comprising:
(a) obtaining an organic compound having two to eight five and/or six-membered rings, provided that the organic compounds is not argentatin, betulinic acid, lanostane, oleanic acid, boswellic acid, glycyrrhetinic acid, ursolic acid, or tricyclic-bis-enone; (b) reacting the organic compound in a series of one of more steps to obtain a compound according to claim 1. 175. The method of claim 174, wherein the compound's induction of Nrf2 activity is at least ten times higher than the organic compound's induction of Nrf2 activity. 176. The method of claim 174, wherein the compound's inhibition of NF-κB activity is at least ten times higher than the organic compound's inhibition of NF-κB activity. 177. The method of claim 174, wherein the compound's inhibition of IFNγ induced NO activity is at least ten times higher than the organic compound's inhibition of IFNγ induced NO activity. | wherein X, R1 and R2 are defined herein. Also provided are pharmaceutical compositions, kits and articles of manufacture comprising such compounds, methods and intermediates useful for making the compounds, and methods of using the compounds and compositions.1. A compound comprising:
a) the basic skeleton of an organic compound having two to eight five and/or six-membered rings provided that the basic skeleton is not the basic skeleton of argentatin, betulinic acid, lanostane, oleanic acid, boswellic acid, glycyrrhetinic acid, ursolic acid, or tricyclic-bis-enone; b) a structural unit of the formula:
wherein:
the carbon atoms labeled 1, 2 and 3 are part of a five or six-membered ring;
X is cyano or —C(O)Ra, wherein Ra is:
hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), alkoxy(C≤12), alkenyl-oxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkylamino(C≤12), heteroarylamino(C≤12), heteroaralkyl-amino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkyl-silyloxy(C≤12), or substituted versions of any of these groups; and
R1 and R2 are each independently:
hydrogen; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤8), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkoxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), hetero-aralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkyl-amino(C≤12), arylamino(C≤12), aralkylamino(C≤12), hetero-arylamino(C≤12), heteroaralkylamino(C≤12), amido(C≤12), or a substituted version of any of these groups; or
R1 and R2 are taken together and are alkanediyl(C≤12), alkenediyl(C≤12), alkanediyl(C≤12) or alkenediyl(C≤12); and
c) from 0 to 8 chemical groups attached to a carbon atom of the basic skeleton other than carbon atoms 1, 2, 3 or 4, wherein each chemical group is independently:
hydroxy, halo, oxo, amino, hydroxyamino, nitro, imino, cyano, azido, mercapto, or thio; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkylidene(C≤12), alkoxy(C≤12), alkenyloxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyl-oxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkyl-amino(C≤12), heteroarylamino(C≤12), heteroaralkylamino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkylimino(C≤12), alkenyl-imino(C≤12), alkynylimino(C≤12), arylimino(C≤12), aralkylimino(C≤12), heteroarylimino(C≤12), heteroaralkylimino(C≤12), acylimino(C≤12), alkylthio(C≤12), alkenylthio(C≤12), alkynylthio(C≤12), arylthio(C≤12), aralkylthio(C≤12), heteroarylthio(C≤12), heteroaralkylthio(C≤12), acyl-thio(C≤12), thioacyl(C≤12), alkylsulfonyl(C≤12), alkenylsulfonyl(C≤8), alkynylsulfonyl(C≤12), arylsulfonyl(C≤12), aralkylsulfonyl(C≤12), heteroarylsulfonyl(C≤12), heteroaralkylsulfonyl(C≤12), alkyl-ammonium(C≤12), alkylsulfonium(C≤12), alkylsilyl(C≤12), alkylsilyl-oxy(C≤12), or a substituted version of any of these groups;
or pharmaceutically acceptable salts, esters, hydrates, solvates, tautomers, acetals, ketals, prodrugs, or optical isomers thereof. 2. The compound of claim 1, wherein R1 and R2 are each independently hydrogen, alkyl(C≤8) or substituted alkyl(C≤8). 3-6. (canceled) 7. A compound comprising:
a) the basic skeleton of a natural product having two to eight rings, provided that the natural product is not argentatin, betulinic acid, lanostane, oleanic acid, or ursolic acid; b) a structural unit of the formula:
wherein:
the carbon atoms labeled 1, 2 and 3 are part of a six-membered ring;
X is cyano, fluoroalkyl(C≤8), substituted, fluoroalkyl(C≤8), or —C(O)Ra, wherein Ra is:
hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), alkoxy(C≤12), alkenyl-oxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkylamino(C≤12), heteroarylamino(C≤12), heteroaralkyl-amino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkyl-silyloxy(C≤12), or substituted versions of any of these groups; and
R1 and R2 are each independently:
hydrogen; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkoxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), hetero-aralkoxy(C≤12), acyloxy(C≤12), alkylamino(C≤12), dialkyl-amino(C≤12), arylamino(C≤12), aralkylamino(C≤12), hetero-arylamino(C≤12), heteroaralkylamino(C≤12), amido(C≤12), or a substituted version of any of these groups; or
R1 and R2 are taken together and are alkanediyl(C≤12), alkenediyl(C≤12), alkanediyl(C≤12) or alkenediyl(C≤12); and
c) from 0 to 8 chemical groups attached to a carbon atom of the basic skeleton other than carbon atoms 1, 2, 3 or 4, wherein each chemical group is independently:
hydroxy, halo, oxo, amino, hydroxyamino, nitro, imino, cyano, azido, mercapto, or thio; or
alkyl(C≤12), alkenyl(C≤12), alkynyl(C≤12), aryl(C≤12), aralkyl(C≤12), heteroaryl(C≤12), heteroaralkyl(C≤12), acyl(C≤12), alkylidene(C≤12), alkoxy(C≤12), alkenyloxy(C≤12), alkynyloxy(C≤12), aryloxy(C≤12), aralkoxy(C≤12), heteroaryloxy(C≤12), heteroaralkoxy(C≤12), acyl-oxy(C≤12), alkylamino(C≤12), dialkylamino(C≤12), alkoxyamino(C≤12), alkenylamino(C≤12), alkynylamino(C≤12), arylamino(C≤12), aralkyl-amino(C≤12), heteroarylamino(C≤12), heteroaralkylamino(C≤12), alkylsulfonylamino(C≤12), amido(C≤12), alkylimino(C≤12), alkenyl-imino(C≤12), alkynylimino(C≤12), arylimino(C≤12), aralkylimino(C≤12), heteroarylimino(C≤12), heteroaralkylimino(C≤12), acylimino(C≤12), alkylthio(C≤12), alkenylthio(C≤12), alkynylthio(C≤12), arylthio(C≤12), aralkylthio(C≤12), heteroarylthio(C≤12), heteroaralkylthio(C≤12), acyl-thio(C≤12), thioacyl(C≤8), alkylsulfonyl(C≤12), alkenylsulfonyl(C≤12), alkynylsulfonyl(C≤12), arylsulfonyl(C≤12), aralkylsulfonyl(C≤12), heteroarylsulfonyl(C≤12), heteroaralkylsulfonyl(C≤12), alkyl-ammonium(C≤12), alkylsulfonium(C≤12), alkylsilyl(C≤12), alkylsilyl-oxy(C≤12), or a substituted version of any of these groups;
or pharmaceutically acceptable salts, esters, hydrates, solvates, tautomers, acetals, ketals, prodrugs, or optical isomers thereof. 8. The compound of claim 7, wherein R1 and R2 are each independently hydrogen, alkyl(C≤8) or substituted alkyl(C≤8). 9-15. (canceled) 16. The compound of claim 7, wherein X is —CN. 17-18. (canceled) 19. The compound of claim 16, wherein the structural unit is further defined as: 20-173. (canceled) 174. A method of making a compound of claim 1, comprising:
(a) obtaining an organic compound having two to eight five and/or six-membered rings, provided that the organic compounds is not argentatin, betulinic acid, lanostane, oleanic acid, boswellic acid, glycyrrhetinic acid, ursolic acid, or tricyclic-bis-enone; (b) reacting the organic compound in a series of one of more steps to obtain a compound according to claim 1. 175. The method of claim 174, wherein the compound's induction of Nrf2 activity is at least ten times higher than the organic compound's induction of Nrf2 activity. 176. The method of claim 174, wherein the compound's inhibition of NF-κB activity is at least ten times higher than the organic compound's inhibition of NF-κB activity. 177. The method of claim 174, wherein the compound's inhibition of IFNγ induced NO activity is at least ten times higher than the organic compound's inhibition of IFNγ induced NO activity. | 1,600 |
626 | 13,115,815 | 1,629 | A flavored oral care composition and method of making same for preventing and/or treating bacterial, fungal or inflammatory diseases and conditions of the oral cavity may be in the form of a rinse, spray, or the like, and free of glycerin and free of castor oil and castor oil derivatives, incorporates a chlorine dioxide source, a phosphate buffer, a flavoring system containing a polyoxyethylene sorbitan ester (emulsifier/suspender) and flavoring agent, a sweetener and water. With the addition of a fluoride ion source, the composition may be used as an anticaries agent. | 1. A flavored oral care single phase composition free of glycerin, castor oil and castor oil derivatives comprising:
(a) a source of chlorine dioxide; (b) a phosphate buffer; (c) a source of flavoring containing a polyoxyethylene sorbitan ester and a flavoring agent; (d) a sweetener; and (e) water. 2. The composition as set forth in claim 1 wherein the concentration of said polyoxyethylene sorbitan ester in the final composition is in the range of about 0.001% to about 40.000% (w/w). 3. The composition as set forth in claim 1 wherein the concentration of said flavoring agent in the final composition is in the range of about 0.005% to about 10.000% (w/w). 4. The composition as set forth in claim 3 wherein the concentration of said polyoxyethylene sorbitan ester in the final composition is in the range of about 0.01% to about 40.0% (w/w). 5. The composition as set forth in claim 1 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.000% (w/w). 6. The composition as set forth in claim 1 wherein the amount of said water in the final composition is the difference between the amount of the total composition and the amount of the sum of all other components of said composition. 7. The composition as set forth in claim 2 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.0% (w/w). 8. The composition as set forth in claim 3 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.0% (w/w). 9. The composition as set forth in claim 4 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.0% (w/w). 10. The composition as set forth in claim 1 wherein said phosphate buffer maintains said composition at a pH in the range of about 6.5 to about 8.0. 11. The composition as set forth in claim 10 wherein the pH of said composition is maintained in the range of 7.3 to 7.5. 12. The composition as set forth in claim 1 including a fluoride ion source. 13. The composition as set forth in claim 12 wherein said fluoride ion source is selected from the group consisting of stannous fluoride, sodium fluoride and sodium monofluorophosphate. 14. The composition as set forth in claim 12 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.025% to about 0.080% (w/w). 15. The composition as set forth in claim 13 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.025% to about 0.080% (w/w). 16. The composition as set forth in claim 12 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.080% to about 0.150% (w/w). 17. The composition as set forth in claim 13 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.080% to about 0.150% (w/w). 18. A method for making a single phase oral rinse containing a chlorine dioxide source, said method comprising the steps of:
(a) mixing a flavoring agent with an emulsifier; (b) further mixing water, a sweetener, a phosphate buffer and stabilized chlorine dioxide; (c) combining the mixtures obtained from exercise of said mixing and said further mixing; (d) adjusting the pH of the mixture obtained by exercise of said step of combining; and (e) wherein said step of adjusting is carried out by the step of adding a weak organic acid to obtain a pH in the range of about 6.5 to about 8.0. 19. The method as set forth in claim 18 wherein the step of adding is carried out to obtain a pH of 7.2. 20. The method as set forth in claim 18 wherein said step of adjusting is carried out by selecting the weak organic acid from the group consisting of acetic acid, fumaric acid, and citric acid. 21. A method for making a single phase flavored oral spray containing a chlorine dioxide source, said method comprising the steps of:
(a) mixing a flavoring agent and an emulsifier in a first container; (b) further mixing water, a phosphate buffer, a sweetener, and a stabilized chlorine dioxide liquid in a second container; (c) combining the mixtures in the first and second containers; and (d) transferring the combined mixtures into a spray dispenser. 22. The method as set forth in claim 21 including the step of selecting the phosphate buffer from a group consisting of sodium phosphate monobasic and sodium phosphate dibasic. 23. The method as set forth in claim 21 including the step of adjusting the pH of the combined mixtures to a pH in the range of about 6.8 to about 7.0. 24. The method as set forth in claim 23 wherein said step of adjusting is carried out by adding a quantity of the phosphate buffer. | A flavored oral care composition and method of making same for preventing and/or treating bacterial, fungal or inflammatory diseases and conditions of the oral cavity may be in the form of a rinse, spray, or the like, and free of glycerin and free of castor oil and castor oil derivatives, incorporates a chlorine dioxide source, a phosphate buffer, a flavoring system containing a polyoxyethylene sorbitan ester (emulsifier/suspender) and flavoring agent, a sweetener and water. With the addition of a fluoride ion source, the composition may be used as an anticaries agent.1. A flavored oral care single phase composition free of glycerin, castor oil and castor oil derivatives comprising:
(a) a source of chlorine dioxide; (b) a phosphate buffer; (c) a source of flavoring containing a polyoxyethylene sorbitan ester and a flavoring agent; (d) a sweetener; and (e) water. 2. The composition as set forth in claim 1 wherein the concentration of said polyoxyethylene sorbitan ester in the final composition is in the range of about 0.001% to about 40.000% (w/w). 3. The composition as set forth in claim 1 wherein the concentration of said flavoring agent in the final composition is in the range of about 0.005% to about 10.000% (w/w). 4. The composition as set forth in claim 3 wherein the concentration of said polyoxyethylene sorbitan ester in the final composition is in the range of about 0.01% to about 40.0% (w/w). 5. The composition as set forth in claim 1 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.000% (w/w). 6. The composition as set forth in claim 1 wherein the amount of said water in the final composition is the difference between the amount of the total composition and the amount of the sum of all other components of said composition. 7. The composition as set forth in claim 2 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.0% (w/w). 8. The composition as set forth in claim 3 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.0% (w/w). 9. The composition as set forth in claim 4 wherein the concentration of said sweetener in the final composition is in the range of about 0.005% to about 2.0% (w/w). 10. The composition as set forth in claim 1 wherein said phosphate buffer maintains said composition at a pH in the range of about 6.5 to about 8.0. 11. The composition as set forth in claim 10 wherein the pH of said composition is maintained in the range of 7.3 to 7.5. 12. The composition as set forth in claim 1 including a fluoride ion source. 13. The composition as set forth in claim 12 wherein said fluoride ion source is selected from the group consisting of stannous fluoride, sodium fluoride and sodium monofluorophosphate. 14. The composition as set forth in claim 12 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.025% to about 0.080% (w/w). 15. The composition as set forth in claim 13 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.025% to about 0.080% (w/w). 16. The composition as set forth in claim 12 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.080% to about 0.150% (w/w). 17. The composition as set forth in claim 13 wherein the concentration of the fluoride ion source in the final composition is in the range of about 0.080% to about 0.150% (w/w). 18. A method for making a single phase oral rinse containing a chlorine dioxide source, said method comprising the steps of:
(a) mixing a flavoring agent with an emulsifier; (b) further mixing water, a sweetener, a phosphate buffer and stabilized chlorine dioxide; (c) combining the mixtures obtained from exercise of said mixing and said further mixing; (d) adjusting the pH of the mixture obtained by exercise of said step of combining; and (e) wherein said step of adjusting is carried out by the step of adding a weak organic acid to obtain a pH in the range of about 6.5 to about 8.0. 19. The method as set forth in claim 18 wherein the step of adding is carried out to obtain a pH of 7.2. 20. The method as set forth in claim 18 wherein said step of adjusting is carried out by selecting the weak organic acid from the group consisting of acetic acid, fumaric acid, and citric acid. 21. A method for making a single phase flavored oral spray containing a chlorine dioxide source, said method comprising the steps of:
(a) mixing a flavoring agent and an emulsifier in a first container; (b) further mixing water, a phosphate buffer, a sweetener, and a stabilized chlorine dioxide liquid in a second container; (c) combining the mixtures in the first and second containers; and (d) transferring the combined mixtures into a spray dispenser. 22. The method as set forth in claim 21 including the step of selecting the phosphate buffer from a group consisting of sodium phosphate monobasic and sodium phosphate dibasic. 23. The method as set forth in claim 21 including the step of adjusting the pH of the combined mixtures to a pH in the range of about 6.8 to about 7.0. 24. The method as set forth in claim 23 wherein said step of adjusting is carried out by adding a quantity of the phosphate buffer. | 1,600 |
627 | 15,349,727 | 1,617 | Treatment of vaginal mycoses, bacterial vaginoses, and other forms of the vaginitis (inflammation of the vagina) by clinoptilolite having a particle size of between 0.2 and 10 μm. Clinoptilolite, when used externally, is effective in the treatment of these vaginal disorders in mammals and humans, and also for restoring a healthy vaginal microbiota. The clinoptilolite may be used with one or more of the following adjuvants: pharmaceutically acceptable carrier materials, viable microorganisms and/or extracts thereof, nutrients for the healthy vaginal microbiota (e.g. lactose, etc.), and/or substances which favorably influence the vaginal environment for the healthy vaginal microbiota (e.g. estradiol, organic acids, etc.). The composition used may be applied locally, preferably in one of the following administration forms: foam, suppository, vaginal tablet, ovule, gel, aerosol, powder, rinse, douche, cream/ointment, or suspension. | 1-4. (canceled) 5. A method of treating vaginitis in a mammal, comprising applying a therapeutically effective amount of a pharmaceutically acceptable composition of clinoptilolite having a particle size of between 0.2 and 10 μm. 6. The method of claim 5, wherein applying the composition of clinoptilolite includes externally applying the composition of clinoptilolite. 7. The method of claim 5, wherein the composition of clinoptilolite is substantially free of heavy metals. 8. The method of claim 5, wherein the method is used to treat one or more of bacterial vaginosis, vulvovaginal candidiasis, and trichomoniasis. 9. The method of claim 5, wherein the method includes treating vaginitis in a human. 10. The method of claim 5, wherein the composition of clinoptilolite applied to the mammal includes one or more pharmaceutically acceptable adjuvants. 11. The method of claim 10, wherein the composition of clinoptilolite applied to the mammal includes one or more of a pharmaceutically acceptable carrier material, a viable microorganisms, a microorganism extract, a nutrient for healthy vaginal microbiota, and a substances which favorably influences the vaginal environment for healthy vaginal microbiota. 12. The method of claim 11, wherein the composition of clinoptilolite applied to the mammal includes one or more of lactose, estradiol, and an organic acid. 13. The method of claim 5, wherein applying the composition of clinoptilolite includes applying a foam, a suppository, a vaginal tablet, an ovule, a gel, an aerosol, a powder, a rinse, a douche, a cream, an ointment, or a suspension. 14. A method of restoring a healthy vaginal microbiota in a mammal, comprising applying a therapeutically effective amount of a pharmaceutically acceptable composition of clinoptilolite having a particle size of between 0.2 and 10 μm. 15. The method of claim 14, wherein applying the composition of clinoptilolite includes externally applying the composition of clinoptilolite. 16. The method of claim 14, wherein the composition of clinoptilolite is substantially free of heavy metals. 17. The method of claim 14, wherein the method includes applying the composition of clinoptilolite to a human. 18. The method of claim 14, wherein the composition of clinoptilolite applied to the mammal includes one or more adjuvants. 19. The method of claim 14, wherein the composition of clinoptilolite applied to the mammal includes one or more of a pharmaceutically acceptable carrier material, a viable microorganisms, a microorganism extract, a nutrient for healthy vaginal microbiota, and a substances which favorably influences the vaginal environment for healthy vaginal microbiota. 20. The method of claim 14, wherein applying the composition of clinoptilolite includes applying a foam, a suppository, a vaginal tablet, an ovule, a gel, an aerosol, a powder, a rinse, a douche, a cream, an ointment, or a suspension. 21. A pharmaceutically acceptable composition of clinoptilolite comprising particles sized between 0.2 and 10 μm for use in the treatment of vaginitis in mammals, and for restoring a healthy vaginal microbiota. 22. The pharmaceutically acceptable composition of claim 21, wherein the composition is substantially free of heavy metals. 23. The pharmaceutically acceptable composition of claim 21, further comprising one or more pharmaceutically acceptable carriers or pharmaceutically acceptable adjuvants. 24. The pharmaceutically acceptable composition of claim 21, consisting essentially of clinoptilolite particles between 0.2 and 10 μm in size. | Treatment of vaginal mycoses, bacterial vaginoses, and other forms of the vaginitis (inflammation of the vagina) by clinoptilolite having a particle size of between 0.2 and 10 μm. Clinoptilolite, when used externally, is effective in the treatment of these vaginal disorders in mammals and humans, and also for restoring a healthy vaginal microbiota. The clinoptilolite may be used with one or more of the following adjuvants: pharmaceutically acceptable carrier materials, viable microorganisms and/or extracts thereof, nutrients for the healthy vaginal microbiota (e.g. lactose, etc.), and/or substances which favorably influence the vaginal environment for the healthy vaginal microbiota (e.g. estradiol, organic acids, etc.). The composition used may be applied locally, preferably in one of the following administration forms: foam, suppository, vaginal tablet, ovule, gel, aerosol, powder, rinse, douche, cream/ointment, or suspension.1-4. (canceled) 5. A method of treating vaginitis in a mammal, comprising applying a therapeutically effective amount of a pharmaceutically acceptable composition of clinoptilolite having a particle size of between 0.2 and 10 μm. 6. The method of claim 5, wherein applying the composition of clinoptilolite includes externally applying the composition of clinoptilolite. 7. The method of claim 5, wherein the composition of clinoptilolite is substantially free of heavy metals. 8. The method of claim 5, wherein the method is used to treat one or more of bacterial vaginosis, vulvovaginal candidiasis, and trichomoniasis. 9. The method of claim 5, wherein the method includes treating vaginitis in a human. 10. The method of claim 5, wherein the composition of clinoptilolite applied to the mammal includes one or more pharmaceutically acceptable adjuvants. 11. The method of claim 10, wherein the composition of clinoptilolite applied to the mammal includes one or more of a pharmaceutically acceptable carrier material, a viable microorganisms, a microorganism extract, a nutrient for healthy vaginal microbiota, and a substances which favorably influences the vaginal environment for healthy vaginal microbiota. 12. The method of claim 11, wherein the composition of clinoptilolite applied to the mammal includes one or more of lactose, estradiol, and an organic acid. 13. The method of claim 5, wherein applying the composition of clinoptilolite includes applying a foam, a suppository, a vaginal tablet, an ovule, a gel, an aerosol, a powder, a rinse, a douche, a cream, an ointment, or a suspension. 14. A method of restoring a healthy vaginal microbiota in a mammal, comprising applying a therapeutically effective amount of a pharmaceutically acceptable composition of clinoptilolite having a particle size of between 0.2 and 10 μm. 15. The method of claim 14, wherein applying the composition of clinoptilolite includes externally applying the composition of clinoptilolite. 16. The method of claim 14, wherein the composition of clinoptilolite is substantially free of heavy metals. 17. The method of claim 14, wherein the method includes applying the composition of clinoptilolite to a human. 18. The method of claim 14, wherein the composition of clinoptilolite applied to the mammal includes one or more adjuvants. 19. The method of claim 14, wherein the composition of clinoptilolite applied to the mammal includes one or more of a pharmaceutically acceptable carrier material, a viable microorganisms, a microorganism extract, a nutrient for healthy vaginal microbiota, and a substances which favorably influences the vaginal environment for healthy vaginal microbiota. 20. The method of claim 14, wherein applying the composition of clinoptilolite includes applying a foam, a suppository, a vaginal tablet, an ovule, a gel, an aerosol, a powder, a rinse, a douche, a cream, an ointment, or a suspension. 21. A pharmaceutically acceptable composition of clinoptilolite comprising particles sized between 0.2 and 10 μm for use in the treatment of vaginitis in mammals, and for restoring a healthy vaginal microbiota. 22. The pharmaceutically acceptable composition of claim 21, wherein the composition is substantially free of heavy metals. 23. The pharmaceutically acceptable composition of claim 21, further comprising one or more pharmaceutically acceptable carriers or pharmaceutically acceptable adjuvants. 24. The pharmaceutically acceptable composition of claim 21, consisting essentially of clinoptilolite particles between 0.2 and 10 μm in size. | 1,600 |
628 | 14,105,662 | 1,653 | The present invention relates to a device and method for the detection of mastitis or other disease from a body fluid of a mammal for example from cow's milk. The device and method relates to a wedge microfluidic chamber for using a minimal amount of fluid and being able to use the device to observe leukocytes in a mono-layer for the purpose of disease detection, cell counts or the like. | 1. A method for performing a leukocyte differential assay on a milk sample comprising:
a) adding a milk sample comprising live cells to a microfluidic chamber assembly comprising: i. a wedge base; and ii. a wedge top having a first top edge and an opposite second edge positioned on the base such that the second top edge is in an elevated configuration, wherein the wedge top is secured to the base in the elevated position, and at least one of the top or base is optically transparent and wherein there is a leukocyte observation colorant positioned within the chamber; b) observing an image morphology of live cell leukocytes in the chamber; c) enumerating and calculating the leukocytes in the milk containing chamber into sub-populations based on the observation of the image morphology of live cell leukocytes in the chamber; and d) deriving a differential count of the leukocyte sub-populations; wherein enumerating and calculating the leukocytes in the milk comprises determining a health status based on a percentage of lymphocytes, neutrophils and macrophages in the sample. 2. A method for performing a leukocyte differential assay on a milk sample comprising:
a) adding a milk sample comprising live cells to a microfluidic chamber assembly comprising: i. a wedge base; ii. a wedge top having a first top edge and an opposite second edge positioned on the base such that the second top edge is in an elevated configuration; and iii. a cut out well positioned to add the milk sample comprising live cells to a top portion of the wedge at the second top edge; wherein the wedge top is secured to the base in the elevated position, wherein at least one of the top or base is optically transparent and wherein there is a leukocyte observation colorant positioned within the chamber; b) observing an image morphology of live cell leukocytes in the chamber; c) enumerating and calculating the leukocytes in the milk containing chamber into sub-populations based on the observation of the image morphology of live cell leukocytes in the chamber; and d) deriving a differential count of the leukocyte sub-populations. 3. A method according to claim 2, further comprising determining the presence of mastitis in the milk responsive to the differential count of leukocyte sub-populations. 4. A method according to claim 2, wherein the assay is performed in an automatic reader wherein a differential is performed by the reader on a milk sampler placed in the microfluidic chamber and the chamber placed in the reader. 5. A method according to claim 2, wherein the step of enumerating and calculating the leukocytes in the milk sample is performed on fluorescing live cells in a fluid milk sample and the leukocyte observation colorant is a fluorescing colorant. 6. A method according to claim 5, wherein the leukocyte observation colorant is a mixture of fluorescing colorants. 7. A method according to claim 11, wherein the microfluidic chamber assembly further comprises a wedge forming device that secures the first top edge and the opposite second edge to the wedge base so that the second top edge is in an elevated configuration. 8. A method according to claim 7, wherein the wedge forming device comprises a sheet having an adhesive and configured to mount the top to the base. 9. A method according to claim 8, wherein when the sample is received in the well, the sample flows through a void between the top and the base and into the chamber via capillary action. 10. A method according to claim 8, wherein at least one of the top or base is optically transparent. 11. A method according to claim 8, wherein the wedge forming device further comprises a liquid entry well for liquid addition to the chamber. 12. A method according to claim 2, wherein each base comprises one or more chambers thereon. 13. A method according to claim 2, wherein observing an image morphology of fluorescing live cells comprises generating a computer-enhanced image using recognition software. 14. A method according to claim 2, wherein adding a milk sample comprising live cells to a microfluidic chamber assembly comprises a self-preparing wet smear. 15. A method according to claim 2, wherein enumerating and calculating the leukocytes in the milk comprises determining a health status based on a percentage of lymphocytes, neutrophils and macrophages in the sample. 16. A method according to claim 15, wherein the health status is a stage of disease. 17. A method according to claim 16, wherein the stage of disease comprises a stage of mastitis. 18. A method according to claim 16, wherein the stage of disease comprises mastitis in peripartum and/or early lactation. 19. A method according to claim 2, wherein the milk sample forms a mono-layer of cells in the chamber. 20. A method for performing a leukocyte differential assay on a milk sample to determine a stage and/or status of mastitis, the method comprising:
a) adding a milk sample comprising live cells as a self-preparing wet smear to a microfluidic chamber assembly comprising: i. a wedge base; ii. a wedge top having a first top edge and an opposite second edge positioned on the base such that the second top edge is in an elevated configuration; iii. a cut out well positioned to add the milk sample comprising live cells to a top portion of the wedge at the first top edge; wherein the wedge top is secured to the base in the elevated position, wherein at least one of the top or base is optically transparent and wherein there is a fluorescing leukocyte observation colorant positioned within the chamber; and iv. a wedge forming device that secures the first top edge and the opposite second edge to the wedge base so that the second top edge is in an elevated configuration; b) generating a computer-enhanced image using recognition software to identify an image morphology of fluorescing live cell leukocytes in the chamber; c) enumerating and calculating the leukocytes in the milk containing chamber into sub-populations based on the identification of the image morphology of fluorescing live cell leukocytes in the chamber; d) deriving a differential count of the leukocyte sub-populations; e) determining a health status comprising a stage of mastitis based on a percentage of lymphocytes, neutrophils and macrophages in the sample. | The present invention relates to a device and method for the detection of mastitis or other disease from a body fluid of a mammal for example from cow's milk. The device and method relates to a wedge microfluidic chamber for using a minimal amount of fluid and being able to use the device to observe leukocytes in a mono-layer for the purpose of disease detection, cell counts or the like.1. A method for performing a leukocyte differential assay on a milk sample comprising:
a) adding a milk sample comprising live cells to a microfluidic chamber assembly comprising: i. a wedge base; and ii. a wedge top having a first top edge and an opposite second edge positioned on the base such that the second top edge is in an elevated configuration, wherein the wedge top is secured to the base in the elevated position, and at least one of the top or base is optically transparent and wherein there is a leukocyte observation colorant positioned within the chamber; b) observing an image morphology of live cell leukocytes in the chamber; c) enumerating and calculating the leukocytes in the milk containing chamber into sub-populations based on the observation of the image morphology of live cell leukocytes in the chamber; and d) deriving a differential count of the leukocyte sub-populations; wherein enumerating and calculating the leukocytes in the milk comprises determining a health status based on a percentage of lymphocytes, neutrophils and macrophages in the sample. 2. A method for performing a leukocyte differential assay on a milk sample comprising:
a) adding a milk sample comprising live cells to a microfluidic chamber assembly comprising: i. a wedge base; ii. a wedge top having a first top edge and an opposite second edge positioned on the base such that the second top edge is in an elevated configuration; and iii. a cut out well positioned to add the milk sample comprising live cells to a top portion of the wedge at the second top edge; wherein the wedge top is secured to the base in the elevated position, wherein at least one of the top or base is optically transparent and wherein there is a leukocyte observation colorant positioned within the chamber; b) observing an image morphology of live cell leukocytes in the chamber; c) enumerating and calculating the leukocytes in the milk containing chamber into sub-populations based on the observation of the image morphology of live cell leukocytes in the chamber; and d) deriving a differential count of the leukocyte sub-populations. 3. A method according to claim 2, further comprising determining the presence of mastitis in the milk responsive to the differential count of leukocyte sub-populations. 4. A method according to claim 2, wherein the assay is performed in an automatic reader wherein a differential is performed by the reader on a milk sampler placed in the microfluidic chamber and the chamber placed in the reader. 5. A method according to claim 2, wherein the step of enumerating and calculating the leukocytes in the milk sample is performed on fluorescing live cells in a fluid milk sample and the leukocyte observation colorant is a fluorescing colorant. 6. A method according to claim 5, wherein the leukocyte observation colorant is a mixture of fluorescing colorants. 7. A method according to claim 11, wherein the microfluidic chamber assembly further comprises a wedge forming device that secures the first top edge and the opposite second edge to the wedge base so that the second top edge is in an elevated configuration. 8. A method according to claim 7, wherein the wedge forming device comprises a sheet having an adhesive and configured to mount the top to the base. 9. A method according to claim 8, wherein when the sample is received in the well, the sample flows through a void between the top and the base and into the chamber via capillary action. 10. A method according to claim 8, wherein at least one of the top or base is optically transparent. 11. A method according to claim 8, wherein the wedge forming device further comprises a liquid entry well for liquid addition to the chamber. 12. A method according to claim 2, wherein each base comprises one or more chambers thereon. 13. A method according to claim 2, wherein observing an image morphology of fluorescing live cells comprises generating a computer-enhanced image using recognition software. 14. A method according to claim 2, wherein adding a milk sample comprising live cells to a microfluidic chamber assembly comprises a self-preparing wet smear. 15. A method according to claim 2, wherein enumerating and calculating the leukocytes in the milk comprises determining a health status based on a percentage of lymphocytes, neutrophils and macrophages in the sample. 16. A method according to claim 15, wherein the health status is a stage of disease. 17. A method according to claim 16, wherein the stage of disease comprises a stage of mastitis. 18. A method according to claim 16, wherein the stage of disease comprises mastitis in peripartum and/or early lactation. 19. A method according to claim 2, wherein the milk sample forms a mono-layer of cells in the chamber. 20. A method for performing a leukocyte differential assay on a milk sample to determine a stage and/or status of mastitis, the method comprising:
a) adding a milk sample comprising live cells as a self-preparing wet smear to a microfluidic chamber assembly comprising: i. a wedge base; ii. a wedge top having a first top edge and an opposite second edge positioned on the base such that the second top edge is in an elevated configuration; iii. a cut out well positioned to add the milk sample comprising live cells to a top portion of the wedge at the first top edge; wherein the wedge top is secured to the base in the elevated position, wherein at least one of the top or base is optically transparent and wherein there is a fluorescing leukocyte observation colorant positioned within the chamber; and iv. a wedge forming device that secures the first top edge and the opposite second edge to the wedge base so that the second top edge is in an elevated configuration; b) generating a computer-enhanced image using recognition software to identify an image morphology of fluorescing live cell leukocytes in the chamber; c) enumerating and calculating the leukocytes in the milk containing chamber into sub-populations based on the identification of the image morphology of fluorescing live cell leukocytes in the chamber; d) deriving a differential count of the leukocyte sub-populations; e) determining a health status comprising a stage of mastitis based on a percentage of lymphocytes, neutrophils and macrophages in the sample. | 1,600 |
629 | 15,130,728 | 1,641 | A method for predicting risk of pre-eclampsia in a pregnant individual includes measuring one or more biochemical markers including an RBP4 biochemical marker in a blood sample obtained from the pregnant individual to determine one or more biomarker levels including an RBP4 biomarker level, identifying, for each of the one or more measured biochemical markers, a difference between the measured biomarker level and a corresponding predetermined control level, and, responsive to the identifying, determining a prediction corresponding to a relative risk of the pregnant individual having or developing pre-eclampsia. | 1. A method for predicting risk of pre-eclampsia in a pregnant individual, the method comprising:
measuring two or more biochemical markers including a retinol binding protein (RBP4) biochemical marker and a placental growth factor (PlGF) biochemical marker in a blood sample obtained from the pregnant individual to determine two or more biomarker levels including an RBP4 biomarker level and a PlGF biochemical marker; identifying, by a processor of a computing device, for each of the two or more measured biochemical markers, a difference between the measured biomarker level and a corresponding predetermined control level; and responsive to the identifying, determining, by the processor, a prediction corresponding to a relative risk of the pregnant individual developing pre-eclampsia. 2. The method of claim 1, wherein measuring the two or more biochemical markers comprises measuring one or more of a P-Selectin biochemical marker, a pappalysin 1 (PAPP-A) biochemical marker, an alpha-fetal protein (AFP) biochemical marker, and a soluble tumor necrosis factor receptor 1 (sTNFR1) biochemical marker. 3. (canceled) 4. The method of claim 1, wherein the prediction corresponds to a relative risk of the pregnant individual developing at least one of severe pre-eclampsia (PeG) and severe early-onset pre-eclampsia (PeG34). 5. (canceled) 6. The method of claim 1, wherein the prediction is based in part upon at least one maternal history factor of the pregnant individual. 7. The method of claim 6, wherein the at least one maternal history factor comprises one of a gestational age, a weight, a BMI, a family history status, an ethnicity, and a smoking status. 8. The method of claim 1, wherein the prediction is positive based at least in part upon identifying the RBP4 biomarker level reflects a statistically significant increase in comparison to a respective control level. 9-13. (canceled) 14. The method of claim 1, wherein the blood sample comprises one of a plasma sample and a serum sample. 15. The method of claim 1, wherein measuring the one or more biochemical markers comprises performing a quantitative immunoassay. 16. The method of claim 1, wherein measuring the one or more biochemical markers comprises determining a concentration of each respective biochemical marker. 17. (canceled) 18. A system for predicting risk of pre-eclampsia in a pregnant individual comprising:
an in vitro diagnostics kit comprising testing instruments for testing a blood sample obtained from the pregnant individual for two or more biochemical markers including an RBP4 biochemical marker and a PlGF biochemical marker, the kit comprising one or more reagents for detecting the amount of RBP4 and one or more reagents for detecting the amount of PlGF; and a non-transitory computer-readable medium having instructions stored thereon, wherein the instructions, when executed by a processor, cause the processor to:
retrieve two or more measured biomarker levels from the blood sample, wherein each biomarker level of the two or more biomarker levels corresponds to a biochemical marker tested for using the in vitro diagnostics kit, and wherein the retrieved one or more biomarker levels comprises an RBP4 biomarker level and a PlGF biochemical marker level; and
calculate a risk assessment score corresponding to a relative risk of the pregnant individual developing pre-eclampsia, wherein the risk assessment score comprises consideration of levels of the RBP4 biomarker level and the PlGF biomarker level. 19. The system of claim 18, wherein measuring the two or more biochemical markers comprises measuring one or more of a P-Selectin biochemical marker, a PAPP-A biochemical marker, an AFP biochemical marker, and a sTNFR1 biochemical marker. 20. The system of claim 18, wherein the risk assessment score is based at least in part upon a comparison of the RBP4 biochemical marker level and the PlGF biochemical marker level with respective predetermined control levels. 21. The system of claim 18, wherein the instructions cause the processor to, prior to calculating the risk assessment score, access at least one maternal history factor of the pregnant individual. 22. The system of claim 21, wherein accessing the at least one maternal history factor of the pregnant individual comprises causing presentation of a graphical user interface at a display device, wherein the graphical user interface comprises one or more input fields for submitting maternal history factor information regarding the pregnant individual. 23. (canceled) 24. The system of claim 18, wherein the instructions cause the processor to, after calculating the risk assessment score, cause presentation of the risk assessment score at a display device. 25. The system of claim 24, wherein causing presentation of the risk assessment score comprises:
causing presentation of a numeric value corresponding to a proportional risk of the pregnant individual developing pre-eclampsia; a ranking on a scale of a relative risk of the pregnant individual developing pre-eclampsia; a percentage likelihood of the pregnant individual developing pre-eclampsia; or graphical illustration expressing a relative risk of the pregnant individual having or developing pre-eclampsia. 26. The system of claim 18, wherein the testing instruments comprise an assay buffer. 27. The system of claim 26, wherein the testing instruments comprise one or more of a coated plate, a tracer, and calibrators. 28-37. (canceled) 38. A system for predicting risk of pre-eclampsia in a pregnant individual comprising:
an in vitro diagnostics kit comprising testing instruments for testing a blood sample obtained from the pregnant individual for two or more biochemical markers, wherein
a first biomarker of the two or more biochemical markers comprises RBP4, and
a second biomarker of the two or more biochemical markers comprises PlGF; and
a non-transitory computer-readable medium having instructions stored thereon, wherein the instructions, when executed by a processor, cause the processor to:
retrieve two or more biomarker levels, wherein each biomarker level of the two or more biomarker levels corresponds to a biochemical marker tested for using the in vitro diagnostics kit, and wherein the retrieved two or more biomarker levels comprises an RBP4 biomarker level, and a PlGF biomarker level;
identify, for each of the two or more measured biochemical markers, a difference between the measured biomarker level and a corresponding predetermined control level; and
responsive to the identifying, determine a prediction corresponding to a relative risk of the pregnant individual developing pre-eclampsia. 39. The system of claim 38, wherein a second biomarker of the one or more biochemical markers is one of a, a P-Selectin biochemical marker, a PAPP-A biochemical marker, an AFP biochemical marker, and a sTNFR1 biochemical marker. 40-41. (canceled) 42. The system of claim 18, wherein the blood sample is obtained from the pregnant individual at 9 to 37 weeks gestation. | A method for predicting risk of pre-eclampsia in a pregnant individual includes measuring one or more biochemical markers including an RBP4 biochemical marker in a blood sample obtained from the pregnant individual to determine one or more biomarker levels including an RBP4 biomarker level, identifying, for each of the one or more measured biochemical markers, a difference between the measured biomarker level and a corresponding predetermined control level, and, responsive to the identifying, determining a prediction corresponding to a relative risk of the pregnant individual having or developing pre-eclampsia.1. A method for predicting risk of pre-eclampsia in a pregnant individual, the method comprising:
measuring two or more biochemical markers including a retinol binding protein (RBP4) biochemical marker and a placental growth factor (PlGF) biochemical marker in a blood sample obtained from the pregnant individual to determine two or more biomarker levels including an RBP4 biomarker level and a PlGF biochemical marker; identifying, by a processor of a computing device, for each of the two or more measured biochemical markers, a difference between the measured biomarker level and a corresponding predetermined control level; and responsive to the identifying, determining, by the processor, a prediction corresponding to a relative risk of the pregnant individual developing pre-eclampsia. 2. The method of claim 1, wherein measuring the two or more biochemical markers comprises measuring one or more of a P-Selectin biochemical marker, a pappalysin 1 (PAPP-A) biochemical marker, an alpha-fetal protein (AFP) biochemical marker, and a soluble tumor necrosis factor receptor 1 (sTNFR1) biochemical marker. 3. (canceled) 4. The method of claim 1, wherein the prediction corresponds to a relative risk of the pregnant individual developing at least one of severe pre-eclampsia (PeG) and severe early-onset pre-eclampsia (PeG34). 5. (canceled) 6. The method of claim 1, wherein the prediction is based in part upon at least one maternal history factor of the pregnant individual. 7. The method of claim 6, wherein the at least one maternal history factor comprises one of a gestational age, a weight, a BMI, a family history status, an ethnicity, and a smoking status. 8. The method of claim 1, wherein the prediction is positive based at least in part upon identifying the RBP4 biomarker level reflects a statistically significant increase in comparison to a respective control level. 9-13. (canceled) 14. The method of claim 1, wherein the blood sample comprises one of a plasma sample and a serum sample. 15. The method of claim 1, wherein measuring the one or more biochemical markers comprises performing a quantitative immunoassay. 16. The method of claim 1, wherein measuring the one or more biochemical markers comprises determining a concentration of each respective biochemical marker. 17. (canceled) 18. A system for predicting risk of pre-eclampsia in a pregnant individual comprising:
an in vitro diagnostics kit comprising testing instruments for testing a blood sample obtained from the pregnant individual for two or more biochemical markers including an RBP4 biochemical marker and a PlGF biochemical marker, the kit comprising one or more reagents for detecting the amount of RBP4 and one or more reagents for detecting the amount of PlGF; and a non-transitory computer-readable medium having instructions stored thereon, wherein the instructions, when executed by a processor, cause the processor to:
retrieve two or more measured biomarker levels from the blood sample, wherein each biomarker level of the two or more biomarker levels corresponds to a biochemical marker tested for using the in vitro diagnostics kit, and wherein the retrieved one or more biomarker levels comprises an RBP4 biomarker level and a PlGF biochemical marker level; and
calculate a risk assessment score corresponding to a relative risk of the pregnant individual developing pre-eclampsia, wherein the risk assessment score comprises consideration of levels of the RBP4 biomarker level and the PlGF biomarker level. 19. The system of claim 18, wherein measuring the two or more biochemical markers comprises measuring one or more of a P-Selectin biochemical marker, a PAPP-A biochemical marker, an AFP biochemical marker, and a sTNFR1 biochemical marker. 20. The system of claim 18, wherein the risk assessment score is based at least in part upon a comparison of the RBP4 biochemical marker level and the PlGF biochemical marker level with respective predetermined control levels. 21. The system of claim 18, wherein the instructions cause the processor to, prior to calculating the risk assessment score, access at least one maternal history factor of the pregnant individual. 22. The system of claim 21, wherein accessing the at least one maternal history factor of the pregnant individual comprises causing presentation of a graphical user interface at a display device, wherein the graphical user interface comprises one or more input fields for submitting maternal history factor information regarding the pregnant individual. 23. (canceled) 24. The system of claim 18, wherein the instructions cause the processor to, after calculating the risk assessment score, cause presentation of the risk assessment score at a display device. 25. The system of claim 24, wherein causing presentation of the risk assessment score comprises:
causing presentation of a numeric value corresponding to a proportional risk of the pregnant individual developing pre-eclampsia; a ranking on a scale of a relative risk of the pregnant individual developing pre-eclampsia; a percentage likelihood of the pregnant individual developing pre-eclampsia; or graphical illustration expressing a relative risk of the pregnant individual having or developing pre-eclampsia. 26. The system of claim 18, wherein the testing instruments comprise an assay buffer. 27. The system of claim 26, wherein the testing instruments comprise one or more of a coated plate, a tracer, and calibrators. 28-37. (canceled) 38. A system for predicting risk of pre-eclampsia in a pregnant individual comprising:
an in vitro diagnostics kit comprising testing instruments for testing a blood sample obtained from the pregnant individual for two or more biochemical markers, wherein
a first biomarker of the two or more biochemical markers comprises RBP4, and
a second biomarker of the two or more biochemical markers comprises PlGF; and
a non-transitory computer-readable medium having instructions stored thereon, wherein the instructions, when executed by a processor, cause the processor to:
retrieve two or more biomarker levels, wherein each biomarker level of the two or more biomarker levels corresponds to a biochemical marker tested for using the in vitro diagnostics kit, and wherein the retrieved two or more biomarker levels comprises an RBP4 biomarker level, and a PlGF biomarker level;
identify, for each of the two or more measured biochemical markers, a difference between the measured biomarker level and a corresponding predetermined control level; and
responsive to the identifying, determine a prediction corresponding to a relative risk of the pregnant individual developing pre-eclampsia. 39. The system of claim 38, wherein a second biomarker of the one or more biochemical markers is one of a, a P-Selectin biochemical marker, a PAPP-A biochemical marker, an AFP biochemical marker, and a sTNFR1 biochemical marker. 40-41. (canceled) 42. The system of claim 18, wherein the blood sample is obtained from the pregnant individual at 9 to 37 weeks gestation. | 1,600 |
630 | 14,888,673 | 1,612 | Advantageously, para-chloroaniline (PCA) is minimal in antimicrobial articles prepared according to the method of the invention. A method of forming an antimicrobial article according to the invention comprises steps of: providing a polymerizable composition; incorporating an antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent into the polymerizable composition; and, polymerizing the polymerizable composition to form chlorhexidine-containing polymer of the antimicrobial article, wherein processing temperature during the method is less than about 80° C. | 1. A method of forming an antimicrobial article, the method comprising steps of:
providing a polymerizable composition; incorporating an antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent into the polymerizable composition; and, polymerizing the polymerizable composition to form chlorhexidine-containing polymer of the antimicrobial article,
wherein processing temperature during the method is less than about 80° C. 2. The method of claim 1, wherein the processing temperature during the method is less than about 40° C. 3. The method of claim 1, wherein the processing temperature during the method is about room temperature. 4. The method of claim 1, wherein the processing temperature during the method is less than that temperature at which chlorhexidine decomposes to para-chloroaniline. 5. The method of claim 1, wherein the antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent in the antimicrobial article consists essentially of chlorhexidine free base. 6. The method of claim 1, wherein the antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent comprises chlorhexidine salt. 7. The method of claim 1, wherein method comprises continuously forming the chlorhexidine-containing polymer on a web. 8. The method of claim 1, wherein the polymerizable composition is essentially free of solvents. 9. The method of claim 1, wherein the chlorhexidine-containing polymer is essentially free of unreacted solvent. 10. The method of claim 1, wherein polymerization of the polymerizable composition is initiated using at least one radiation source selected from ultraviolet radiation and electron beam radiation. 11. The method of claim 1, wherein polymerization of the polymerizable composition is initiated without use of an external source of thermal radiation. 12. The method of claim 1, wherein the chlorhexidine-containing polymer is a polymer film. 13. The method of claim 1, wherein chlorhexidine-containing polymer is an adhesive. 14. The method of claim 1, wherein chlorhexidine-containing polymer is a backing for an adhesive-coated antimicrobial article. 15. The method of claim 1, wherein 100% reactive chemistry is used to form the chlorhexidine-containing polymer. 16. The method of claim 1, wherein the chlorhexidine-containing polymer comprises at least one base polymer selected from polycarbonates, polyvinyl fluorides, poly(meth)acrylates, polyurethanes, and modified polymers thereof. 17. The method of claim 1, wherein the chlorhexidine-containing polymer is polyurethane-based. 18. The method of claim 1, wherein the chlorhexidine-containing polymer is (meth)acrylate-based. 19. The antimicrobial article prepared according to the method of claim 1. 20. An antimicrobial article comprising at least one chlorhexidine-containing polymer, wherein less than about 0.0001 mg/mL detectable para-chloroaniline is present in the chlorhexidine-containing polymer. 21. The antimicrobial article of claim 20, wherein the article comprises about 10 weight % chlorhexidine free base based on total weight of the chlorhexidine-containing polymer. 22. The antimicrobial article of claim 20, wherein essentially no detectable para-chloroaniline is present in the chlorhexidine-containing polymer. | Advantageously, para-chloroaniline (PCA) is minimal in antimicrobial articles prepared according to the method of the invention. A method of forming an antimicrobial article according to the invention comprises steps of: providing a polymerizable composition; incorporating an antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent into the polymerizable composition; and, polymerizing the polymerizable composition to form chlorhexidine-containing polymer of the antimicrobial article, wherein processing temperature during the method is less than about 80° C.1. A method of forming an antimicrobial article, the method comprising steps of:
providing a polymerizable composition; incorporating an antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent into the polymerizable composition; and, polymerizing the polymerizable composition to form chlorhexidine-containing polymer of the antimicrobial article,
wherein processing temperature during the method is less than about 80° C. 2. The method of claim 1, wherein the processing temperature during the method is less than about 40° C. 3. The method of claim 1, wherein the processing temperature during the method is about room temperature. 4. The method of claim 1, wherein the processing temperature during the method is less than that temperature at which chlorhexidine decomposes to para-chloroaniline. 5. The method of claim 1, wherein the antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent in the antimicrobial article consists essentially of chlorhexidine free base. 6. The method of claim 1, wherein the antimicrobially effective amount of at least one chlorhexidine-containing antimicrobial agent comprises chlorhexidine salt. 7. The method of claim 1, wherein method comprises continuously forming the chlorhexidine-containing polymer on a web. 8. The method of claim 1, wherein the polymerizable composition is essentially free of solvents. 9. The method of claim 1, wherein the chlorhexidine-containing polymer is essentially free of unreacted solvent. 10. The method of claim 1, wherein polymerization of the polymerizable composition is initiated using at least one radiation source selected from ultraviolet radiation and electron beam radiation. 11. The method of claim 1, wherein polymerization of the polymerizable composition is initiated without use of an external source of thermal radiation. 12. The method of claim 1, wherein the chlorhexidine-containing polymer is a polymer film. 13. The method of claim 1, wherein chlorhexidine-containing polymer is an adhesive. 14. The method of claim 1, wherein chlorhexidine-containing polymer is a backing for an adhesive-coated antimicrobial article. 15. The method of claim 1, wherein 100% reactive chemistry is used to form the chlorhexidine-containing polymer. 16. The method of claim 1, wherein the chlorhexidine-containing polymer comprises at least one base polymer selected from polycarbonates, polyvinyl fluorides, poly(meth)acrylates, polyurethanes, and modified polymers thereof. 17. The method of claim 1, wherein the chlorhexidine-containing polymer is polyurethane-based. 18. The method of claim 1, wherein the chlorhexidine-containing polymer is (meth)acrylate-based. 19. The antimicrobial article prepared according to the method of claim 1. 20. An antimicrobial article comprising at least one chlorhexidine-containing polymer, wherein less than about 0.0001 mg/mL detectable para-chloroaniline is present in the chlorhexidine-containing polymer. 21. The antimicrobial article of claim 20, wherein the article comprises about 10 weight % chlorhexidine free base based on total weight of the chlorhexidine-containing polymer. 22. The antimicrobial article of claim 20, wherein essentially no detectable para-chloroaniline is present in the chlorhexidine-containing polymer. | 1,600 |
631 | 13,612,925 | 1,662 | Provided are novel compositions for use in herbicide activity. Specifically, methods and compositions that modulate 5-enolpyruvylshikimate-3-phosphate synthase in plant species. The present invention also provides for combinations of compositions and methods that enhance weed control. | 1. A method of plant control comprising: treating a plant with a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an EPSPS gene sequence or fragment thereof, or to a portion of an RNA transcript of said EPSPS gene sequence or fragment thereof, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, whereby said plant growth or development or reproductive ability is reduced or said plant is more sensitive to an EPSPS inhibitor herbicide relative to a plant not treated with said composition. 2. The method as claimed in claim 1, wherein said transfer agent is an organosilicone surfactant composition or compound contained therein. 3. The method as claimed in claim 1, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 4. The method as claimed in claim 3, wherein said polynucleotide fragment is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 5. The method as claimed in claim 1, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Lolium multiflorum, Lolium rigidium, Ambrosia artemisiifolia, Ambrosia trifida, Euphorbia heterophylla, Kochia scoparia, Abutilon theophrasti, Sorghum halepense, Chenopodium album, Commelina diffusa, Conyza candensis, Digitaria sanguinalis. 6. The method as claimed in claim 1, wherein said composition further comprises said EPSPS inhibitor herbicide and external application to a plant with said composition. 7. The method as claimed in claim 6, wherein said composition further comprises one or more herbicides different from said EPSPS inhibitor herbicide. 8. The method as claimed in claim 7, wherein said composition further comprises an auxin-like herbicide. 9. The method as claimed in claim 8, wherein said auxin-like herbicide is dicamba or 2,4-D. 10. The method as claimed in claim 3, wherein said composition comprises any combination of two or more of said polynucleotide fragments and external application to a plant with said composition. 11. A composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an EPSPS gene sequence or fragment thereof, or to an RNA transcript of said EPSPS gene sequence, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 12. The composition of claim 11, wherein said transfer agent is an organosilicone composition. 13. The composition of claim 11, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 14. The composition of claim 11, wherein said polynucleotide is selected from the group consisting of SEQ ID NO:121-3222. 15. The composition of claim 11, wherein said polynucleotide is selected from the group consisting of SEQ ID NO:3223-3542. 16. The composition of claim 11, further comprising an EPSPS inhibitor herbicide. 17. The composition of claim 16, wherein said EPSPS inhibitor molecule is glyphosate. 18. The composition of claim 17, further comprising a co-herbicide. 19. The composition of claim 18, wherein said co-herbicide is an auxin-like herbicide. 20. The method as claimed in claim 8, wherein said auxin-like herbicide is dicamba or 2,4-D. 21. A method of reducing expression of an EPSPS gene in a plant comprising: external application to a plant of a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an EPSPS gene sequence, or to the RNA transcript of said EPSPS gene sequence, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, whereby said expression of said EPSPS gene is reduced relative to a plant in which the composition was not applied. 22. The method as claimed in claim 21, wherein said transfer agent is an organosilicone compound. 23. The method as claimed in claim 21, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 24. The method as claimed in 21, wherein said polynucleotide molecule is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 25. A microbial expression cassette comprising a polynucleotide fragment of 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 26. A method of making a polynucleotide comprising a) transforming the microbial expression cassette of claim 25 into a microbe; b) growing said microbe; c) harvesting a polynucleotide from said microbe, wherein said polynucleotide is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 27. A method of identifying polynucleotides useful in modulating EPSPS gene expression when externally treating a plant comprising: a) providing a plurality of polynucleotides that comprise a region essentially identical or essentially complementary to a polynucleotide fragment of 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120; b) externally treating said plant with one or more of said polynucleotides and a transfer agent; c) analyzing said plant or extract for modulation of EPSPS gene expression, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 28. The method as claimed in 27, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Lolium multiflorum, Lolium rigidium, Ambrosia artemisiifolia, Ambrosia trifida, Euphorbia heterophylla, Kochia scoparia, Abutilon theophrasti, Sorghum halepense, Chenopodium album, Commelina diffusa, Conyza candensis, Digitaria sanguinalis. 29. The method as claimed in 27, wherein said EPSPS gene expression is reduced relative to a plant not treated with said polynucleotide fragment and a transfer agent. 30. The method as claimed in 27, wherein said transfer agent is an organosilicone compound. 31. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a co-herbicide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an EPSPS gene sequence or fragment thereof, or to a portion of an RNA transcript of said EPSPS gene sequence or fragment thereof, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 32. The agricultural chemical composition of claim 31, wherein said co-herbicide is selected from the group consisting of amide herbicides, arsenical herbicides, benzothiazole herbicides, benzoylcyclohexanedione herbicides, benzofuranyl alkylsulfonate herbicides, carbamate herbicides, cyclohexene oxime herbicides, cyclopropylisoxazole herbicides, dicarboximide herbicides, dinitroaniline herbicides, dinitrophenol herbicides, diphenyl ether herbicides, dithiocarbamate herbicides, halogenated aliphatic herbicides, imidazolinone herbicides, inorganic herbicides, nitrile herbicides, organophosphorus herbicides, oxadiazolone herbicides, oxazole herbicides, phenoxy herbicides, phenylenediamine herbicides, pyrazole herbicides, pyridazine herbicides, pyridazinone herbicides, pyridine herbicides, pyrimidinediamine herbicides, pyrimidinyloxybenzylamine herbicides, quaternary ammonium herbicides, thiocarbamate herbicides, thiocarbonate herbicides, thiourea herbicides, triazine herbicides, triazinone herbicides, triazole herbicides, triazolone herbicides, triazolopyrimidine herbicides, uracil herbicides, and urea herbicides. 33. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a pesticide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an EPSPS gene sequence, or to a portion of an RNA transcript of said EPSPS gene sequence, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 34. The agricultural chemical composition of claim 33, wherein said pesticide is selected from the group consisting of insecticides, fungicides, nematocides, bactericides, acaricides, growth regulators, chemosterilants, semiochemicals, repellents, attractants, pheromones, feeding stimulants, and biopesticides. 35. A polynucleotide molecule applied to the surface of a plant that enhances said plant sensitivity to a glyphosate containing herbicide composition, wherein said polynucleotide comprises a homologous or complementary polynucleotide having at least 85 percent idendity to a polynucleotide selected from the group consisting of SEQ ID NO: 3781-3789. | Provided are novel compositions for use in herbicide activity. Specifically, methods and compositions that modulate 5-enolpyruvylshikimate-3-phosphate synthase in plant species. The present invention also provides for combinations of compositions and methods that enhance weed control.1. A method of plant control comprising: treating a plant with a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an EPSPS gene sequence or fragment thereof, or to a portion of an RNA transcript of said EPSPS gene sequence or fragment thereof, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, whereby said plant growth or development or reproductive ability is reduced or said plant is more sensitive to an EPSPS inhibitor herbicide relative to a plant not treated with said composition. 2. The method as claimed in claim 1, wherein said transfer agent is an organosilicone surfactant composition or compound contained therein. 3. The method as claimed in claim 1, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 4. The method as claimed in claim 3, wherein said polynucleotide fragment is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 5. The method as claimed in claim 1, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Lolium multiflorum, Lolium rigidium, Ambrosia artemisiifolia, Ambrosia trifida, Euphorbia heterophylla, Kochia scoparia, Abutilon theophrasti, Sorghum halepense, Chenopodium album, Commelina diffusa, Conyza candensis, Digitaria sanguinalis. 6. The method as claimed in claim 1, wherein said composition further comprises said EPSPS inhibitor herbicide and external application to a plant with said composition. 7. The method as claimed in claim 6, wherein said composition further comprises one or more herbicides different from said EPSPS inhibitor herbicide. 8. The method as claimed in claim 7, wherein said composition further comprises an auxin-like herbicide. 9. The method as claimed in claim 8, wherein said auxin-like herbicide is dicamba or 2,4-D. 10. The method as claimed in claim 3, wherein said composition comprises any combination of two or more of said polynucleotide fragments and external application to a plant with said composition. 11. A composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an EPSPS gene sequence or fragment thereof, or to an RNA transcript of said EPSPS gene sequence, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 12. The composition of claim 11, wherein said transfer agent is an organosilicone composition. 13. The composition of claim 11, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 14. The composition of claim 11, wherein said polynucleotide is selected from the group consisting of SEQ ID NO:121-3222. 15. The composition of claim 11, wherein said polynucleotide is selected from the group consisting of SEQ ID NO:3223-3542. 16. The composition of claim 11, further comprising an EPSPS inhibitor herbicide. 17. The composition of claim 16, wherein said EPSPS inhibitor molecule is glyphosate. 18. The composition of claim 17, further comprising a co-herbicide. 19. The composition of claim 18, wherein said co-herbicide is an auxin-like herbicide. 20. The method as claimed in claim 8, wherein said auxin-like herbicide is dicamba or 2,4-D. 21. A method of reducing expression of an EPSPS gene in a plant comprising: external application to a plant of a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an EPSPS gene sequence, or to the RNA transcript of said EPSPS gene sequence, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, whereby said expression of said EPSPS gene is reduced relative to a plant in which the composition was not applied. 22. The method as claimed in claim 21, wherein said transfer agent is an organosilicone compound. 23. The method as claimed in claim 21, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 24. The method as claimed in 21, wherein said polynucleotide molecule is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 25. A microbial expression cassette comprising a polynucleotide fragment of 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 26. A method of making a polynucleotide comprising a) transforming the microbial expression cassette of claim 25 into a microbe; b) growing said microbe; c) harvesting a polynucleotide from said microbe, wherein said polynucleotide is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120. 27. A method of identifying polynucleotides useful in modulating EPSPS gene expression when externally treating a plant comprising: a) providing a plurality of polynucleotides that comprise a region essentially identical or essentially complementary to a polynucleotide fragment of 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an EPSPS gene sequence selected from the group consisting of SEQ ID NO:1-120; b) externally treating said plant with one or more of said polynucleotides and a transfer agent; c) analyzing said plant or extract for modulation of EPSPS gene expression, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 28. The method as claimed in 27, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Lolium multiflorum, Lolium rigidium, Ambrosia artemisiifolia, Ambrosia trifida, Euphorbia heterophylla, Kochia scoparia, Abutilon theophrasti, Sorghum halepense, Chenopodium album, Commelina diffusa, Conyza candensis, Digitaria sanguinalis. 29. The method as claimed in 27, wherein said EPSPS gene expression is reduced relative to a plant not treated with said polynucleotide fragment and a transfer agent. 30. The method as claimed in 27, wherein said transfer agent is an organosilicone compound. 31. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a co-herbicide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an EPSPS gene sequence or fragment thereof, or to a portion of an RNA transcript of said EPSPS gene sequence or fragment thereof, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 32. The agricultural chemical composition of claim 31, wherein said co-herbicide is selected from the group consisting of amide herbicides, arsenical herbicides, benzothiazole herbicides, benzoylcyclohexanedione herbicides, benzofuranyl alkylsulfonate herbicides, carbamate herbicides, cyclohexene oxime herbicides, cyclopropylisoxazole herbicides, dicarboximide herbicides, dinitroaniline herbicides, dinitrophenol herbicides, diphenyl ether herbicides, dithiocarbamate herbicides, halogenated aliphatic herbicides, imidazolinone herbicides, inorganic herbicides, nitrile herbicides, organophosphorus herbicides, oxadiazolone herbicides, oxazole herbicides, phenoxy herbicides, phenylenediamine herbicides, pyrazole herbicides, pyridazine herbicides, pyridazinone herbicides, pyridine herbicides, pyrimidinediamine herbicides, pyrimidinyloxybenzylamine herbicides, quaternary ammonium herbicides, thiocarbamate herbicides, thiocarbonate herbicides, thiourea herbicides, triazine herbicides, triazinone herbicides, triazole herbicides, triazolone herbicides, triazolopyrimidine herbicides, uracil herbicides, and urea herbicides. 33. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a pesticide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an EPSPS gene sequence, or to a portion of an RNA transcript of said EPSPS gene sequence, wherein said EPSPS gene sequence is selected from the group consisting of SEQ ID NO:1-120 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 34. The agricultural chemical composition of claim 33, wherein said pesticide is selected from the group consisting of insecticides, fungicides, nematocides, bactericides, acaricides, growth regulators, chemosterilants, semiochemicals, repellents, attractants, pheromones, feeding stimulants, and biopesticides. 35. A polynucleotide molecule applied to the surface of a plant that enhances said plant sensitivity to a glyphosate containing herbicide composition, wherein said polynucleotide comprises a homologous or complementary polynucleotide having at least 85 percent idendity to a polynucleotide selected from the group consisting of SEQ ID NO: 3781-3789. | 1,600 |
632 | 14,765,828 | 1,618 | A polymeric film includes a polymer matrix having at least one of a discrete RSNO adduct or a polymeric RSNO adduct associated therewith, by: covalent attachment to the polymer matrix; dispersion within the polymer matrix; or both, with the at least one of the discrete RSNO adduct or the polymeric RSNO adduct capable of releasing nitric oxide (NO). The polymer matrix is a polyurethane polymer matrix, a silicone rubber polymer matrix, or a copolymer matrix of polyurethane and silicone rubber. The polymeric film is to exhibit stability under dry conditions at 37° C. and prolonged and controllable NO release rates, when exposed to moisture or light capable of photolyzing an RSNO bond, for a predetermined amount of time from the at least one of the discrete RSNO adduct or the polymeric RSNO adduct. | 1. A polymeric film, comprising:
a polymer matrix having at least one of a discrete RSNO adduct or a polymeric RSNO adduct associated therewith, by: covalent attachment to the polymer matrix; dispersion within the polymer matrix; or both, with the at least one of the discrete RSNO adduct or the polymeric RSNO adduct capable of releasing nitric oxide (NO), the polymer matrix being a polyurethane polymer matrix, a silicone rubber polymer matrix, or a copolymer matrix of polyurethane and silicone rubber; the polymeric film to exhibit stability under dry conditions at 37° C. and prolonged and controllable NO release rates, when exposed to moisture or light capable of photolyzing an RSNO bond, for a predetermined amount of time from the at least one of the discrete RSNO adduct or the polymeric RSNO adduct. 2. The polymeric film as defined in claim 1 wherein the polymer matrix comprises a hydrophobic polymer or a polymer with both hydrophobic and hydrophilic domains. 3. The polymeric film as defined in claim 1 wherein the polymer matrix is a siloxane-based polyurethane elastomer, and wherein the discrete RSNO adduct is S-nitroso-N-acetylpenicillamine (SNAP) or the polymeric RSNO adduct is a polymer possessing an appended SNAP species. 4. The polymeric film as defined in claim 3 wherein the discrete RSNO adduct is a SNAP derivative in which a carboxyl group of SNAP, an amine group of SNAP, or both are modified with an alkyl group. 5. The polymeric film as defined in claim 3 wherein the discrete RSNO adduct is a SNAP derivative in which i) an alkyl group including from 4 carbon atoms to 10 carbon atoms is attached to an amine nitrogen of SNAP, ii) an ester linkage is present between an alkyl group and a carboxyl group of SNAP, or both i and ii. 6. The polymeric film as defined in claim 1 wherein the discrete RSNO adduct or the polymeric RSNO adduct is dispersed within the polymer matrix, and wherein the discrete RSNO adduct or the polymeric RSNO adduct is present in an amount of about 5 wt %. 7. The polymeric film as defined in claim 1 wherein the discrete RSNO adduct or the polymeric RSNO adduct is dispersed within the polymer matrix, and wherein the discrete RSNO adduct or the polymeric RSNO adduct is present in an amount of about 10 wt %. 8. A polymeric composition, comprising:
a base polymer layer; a top polymer layer disposed on the base polymer layer; and at least one active layer intermediate to the base polymer layer and the top polymer layer, and the at least one active intermediate layer including the polymeric film as defined in claim 3. 9. The polymeric composition as defined in claim 8 wherein:
the base polymer layer is selected from a siloxane-based polyurethane elastomer, poly(vinyl chloride), crosslinked polyurethane, crosslinked silicone rubber, and polytetrafluoroethylene; and
the top polymer layer is a siloxane-based polyurethane elastomer, poly(vinyl chloride), crosslinked polyurethane, crosslinked silicone rubber, and polytetrafluoroethylene. 10. The polymeric composition as defined in claim 8 wherein the discrete RSNO adduct or the polymeric RSNO adduct is dispersed within the polymer matrix, and wherein the discrete RSNO adduct or the polymeric RSNO adduct is present in an amount ranging about 5 wt % to about 10 wt %. 11. A method for making an NO-releasing polymeric composition, comprising the steps of:
selecting a polymer matrix to at least one of increase, prolong, and control NO release rates from at least one of a discrete RSNO adduct and a polymeric RSNO adduct, the selected polymer matrix acting to stabilize the at least one of the discrete RSNO adduct and the polymeric RSNO adduct; and dispersing the at least one of the discrete RSNO adduct and the polymeric RSNO adduct within the polymer matrix, the at least one of the discrete RSNO adduct and the polymeric RSNO adduct capable of releasing nitric oxide (NO). 12. The method as defined in claim 11 wherein the polymer matrix is a siloxane-based polyurethane elastomer, and wherein the discrete RSNO adduct is S-nitroso-N-acetylpenicillamine (SNAP) or a polymer possessing an appended SNAP species. 13. The method as defined in claim 11 wherein the dispersing includes:
dissolving the selected polymer matrix in a solvent to form a solution;
adding the at least one of the discrete RSNO adduct and the polymeric RSNO adduct to the solution; and
stirring the solution for a predetermined time. 14. The method as defined in claim 13, further comprising:
casting the solution on a base polymer layer; and drying the cast solution to form a layer of the NO-releasing polymeric composition on the base polymer layer. 15. The method as defined in claim 14, further comprising coating an other polymer on the layer of the NO-releasing polymeric composition to form a top polymer layer, wherein the other polymer does not include the at least one of the discrete RSNO adduct and the polymeric RSNO adduct therein. | A polymeric film includes a polymer matrix having at least one of a discrete RSNO adduct or a polymeric RSNO adduct associated therewith, by: covalent attachment to the polymer matrix; dispersion within the polymer matrix; or both, with the at least one of the discrete RSNO adduct or the polymeric RSNO adduct capable of releasing nitric oxide (NO). The polymer matrix is a polyurethane polymer matrix, a silicone rubber polymer matrix, or a copolymer matrix of polyurethane and silicone rubber. The polymeric film is to exhibit stability under dry conditions at 37° C. and prolonged and controllable NO release rates, when exposed to moisture or light capable of photolyzing an RSNO bond, for a predetermined amount of time from the at least one of the discrete RSNO adduct or the polymeric RSNO adduct.1. A polymeric film, comprising:
a polymer matrix having at least one of a discrete RSNO adduct or a polymeric RSNO adduct associated therewith, by: covalent attachment to the polymer matrix; dispersion within the polymer matrix; or both, with the at least one of the discrete RSNO adduct or the polymeric RSNO adduct capable of releasing nitric oxide (NO), the polymer matrix being a polyurethane polymer matrix, a silicone rubber polymer matrix, or a copolymer matrix of polyurethane and silicone rubber; the polymeric film to exhibit stability under dry conditions at 37° C. and prolonged and controllable NO release rates, when exposed to moisture or light capable of photolyzing an RSNO bond, for a predetermined amount of time from the at least one of the discrete RSNO adduct or the polymeric RSNO adduct. 2. The polymeric film as defined in claim 1 wherein the polymer matrix comprises a hydrophobic polymer or a polymer with both hydrophobic and hydrophilic domains. 3. The polymeric film as defined in claim 1 wherein the polymer matrix is a siloxane-based polyurethane elastomer, and wherein the discrete RSNO adduct is S-nitroso-N-acetylpenicillamine (SNAP) or the polymeric RSNO adduct is a polymer possessing an appended SNAP species. 4. The polymeric film as defined in claim 3 wherein the discrete RSNO adduct is a SNAP derivative in which a carboxyl group of SNAP, an amine group of SNAP, or both are modified with an alkyl group. 5. The polymeric film as defined in claim 3 wherein the discrete RSNO adduct is a SNAP derivative in which i) an alkyl group including from 4 carbon atoms to 10 carbon atoms is attached to an amine nitrogen of SNAP, ii) an ester linkage is present between an alkyl group and a carboxyl group of SNAP, or both i and ii. 6. The polymeric film as defined in claim 1 wherein the discrete RSNO adduct or the polymeric RSNO adduct is dispersed within the polymer matrix, and wherein the discrete RSNO adduct or the polymeric RSNO adduct is present in an amount of about 5 wt %. 7. The polymeric film as defined in claim 1 wherein the discrete RSNO adduct or the polymeric RSNO adduct is dispersed within the polymer matrix, and wherein the discrete RSNO adduct or the polymeric RSNO adduct is present in an amount of about 10 wt %. 8. A polymeric composition, comprising:
a base polymer layer; a top polymer layer disposed on the base polymer layer; and at least one active layer intermediate to the base polymer layer and the top polymer layer, and the at least one active intermediate layer including the polymeric film as defined in claim 3. 9. The polymeric composition as defined in claim 8 wherein:
the base polymer layer is selected from a siloxane-based polyurethane elastomer, poly(vinyl chloride), crosslinked polyurethane, crosslinked silicone rubber, and polytetrafluoroethylene; and
the top polymer layer is a siloxane-based polyurethane elastomer, poly(vinyl chloride), crosslinked polyurethane, crosslinked silicone rubber, and polytetrafluoroethylene. 10. The polymeric composition as defined in claim 8 wherein the discrete RSNO adduct or the polymeric RSNO adduct is dispersed within the polymer matrix, and wherein the discrete RSNO adduct or the polymeric RSNO adduct is present in an amount ranging about 5 wt % to about 10 wt %. 11. A method for making an NO-releasing polymeric composition, comprising the steps of:
selecting a polymer matrix to at least one of increase, prolong, and control NO release rates from at least one of a discrete RSNO adduct and a polymeric RSNO adduct, the selected polymer matrix acting to stabilize the at least one of the discrete RSNO adduct and the polymeric RSNO adduct; and dispersing the at least one of the discrete RSNO adduct and the polymeric RSNO adduct within the polymer matrix, the at least one of the discrete RSNO adduct and the polymeric RSNO adduct capable of releasing nitric oxide (NO). 12. The method as defined in claim 11 wherein the polymer matrix is a siloxane-based polyurethane elastomer, and wherein the discrete RSNO adduct is S-nitroso-N-acetylpenicillamine (SNAP) or a polymer possessing an appended SNAP species. 13. The method as defined in claim 11 wherein the dispersing includes:
dissolving the selected polymer matrix in a solvent to form a solution;
adding the at least one of the discrete RSNO adduct and the polymeric RSNO adduct to the solution; and
stirring the solution for a predetermined time. 14. The method as defined in claim 13, further comprising:
casting the solution on a base polymer layer; and drying the cast solution to form a layer of the NO-releasing polymeric composition on the base polymer layer. 15. The method as defined in claim 14, further comprising coating an other polymer on the layer of the NO-releasing polymeric composition to form a top polymer layer, wherein the other polymer does not include the at least one of the discrete RSNO adduct and the polymeric RSNO adduct therein. | 1,600 |
633 | 10,995,203 | 1,611 | A combination comprising creatine and/or creatinine and/or a derivative thereof and one or more retinoids. This Abstract is not intended to define the invention disclosed in the specification, nor intended to limit the scope of the invention in any way. | 1. An active substance combination comprising
(b) at least one of creatine, creatinine, a creatine derivative and a creatinine derivative, and (b) at least one retinoid. 2. The active substance combination of claim 1, wherein a weight ratio of creatinine to creatine is from about 10:1 to about 1:10. 3. The active substance combination of claim 2, wherein the weight ratio is from about 4:1 to about 3:7. 4. The active substance combination of claim 2, wherein the weight ratio is from about 2:1 to about 1:2. 5. The active substance combination of claim 1, wherein the at least one retinoid comprises at least one of retinol and retinyl palmitate. 6. The active substance combination of claim 1, wherein the at least one retinoid comprises retinyl palmitate. 7. The active substance combination of claim 1, wherein the at least one retinoid comprises retinol. 8. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 1. 9. The preparation of claim 8, wherein the preparation comprises from about 0.0005% to about 50% by weight of the active substance combination. 10. The preparation of claim 9, wherein the preparation comprises from about 0.01% to about 20% by weight of the active substance combination. 11. The preparation of claim 10, wherein the preparation comprises from about 0.02% to about 10% by weight of the active substance combination. 12. The preparation of claim 11, wherein the preparation comprises from about 0.02% to about 5% by weight of the active substance combination. 13. The preparation of claim 12, wherein the preparation comprises from about 0.5% to about 3% by weight of the active substance combination. 14. The preparation of claim 8, wherein the preparation comprises from about 0.001% to about 10% by weight of at least one of creatine and a creatine derivative. 15. The preparation of claim 14, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatine and a creatine derivative. 16. The preparation of claim 8, wherein the preparation comprises from about 0.001% to about 10% by weight of at least one of creatinine and a creatinine derivative. 17. The preparation of claim 15, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatinine and a creatinine derivative. 18. The preparation of claim 8, wherein the preparation comprises from about 0.001% to about 10% by weight of the at least one retinoid. 19. The preparation of claim 12, wherein the preparation comprises from about 0.01% to about 1% by weight of the at least one retinoid. 20. The preparation of claim 19, wherein the at least one retinoid comprises at least one of retinol and retinyl palmitate. 21. The preparation of claim 8, wherein the preparation further comprises from about 0.001% to about 30% by weight of glycerin. 22. The preparation of claim 9, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 23. The preparation of claim 10, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 24. The preparation of claim 11, wherein the preparation further comprises from about 1% to about 7% by weight of glycerin. 25. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 2. 26. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 3. 27. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 4. 28. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 5. 29. A cosmetic or dermatological preparation which comprises from about 0.01% to about 20% by weight of an active substance combination comprising
(a) at least one of creatine, creatinine, a creatine derivative and a creatinine derivative, and (b) at least one retinoid. 30. The preparation of claim 29, wherein a weight ratio of creatinine to creatine in the active substance combination is from about 4:1 to about 3:7. 31. The preparation of claim 30, wherein the weight ratio is from about 2:1 to about 1:2. 32. The preparation of claim 29, wherein the preparation comprises from about 0.02% to about 10% by weight of the active substance combination. 33. The preparation of claim 30, wherein the preparation comprises from about 0.5% to about 3% by weight of the active substance combination. 34. The preparation of claim 32, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatine and a creatine derivative. 35. The preparation of claim 32, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatinine and a creatinine derivative. 36. The preparation of claim 31, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 37. The preparation of claim 30, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 38. The preparation of claim 32, wherein the preparation further comprises from about 1% to about 7% by weight of glycerin. 39. The preparation of claim 32, wherein the preparation comprises from about 0.001% to about 10% by weight of the at least one retinoid. 40. The preparation of claim 30, wherein the preparation comprises from about 0.01% to about 1% by weight of the at least one retinoid. 41. The preparation of claim 40, wherein the at least one retinoid comprises at least one of retinol and retinyl palmitate. 42. An O/W cream which comprises the active substance combination of claim 1. 43. An O/W emulsion which comprises the active substance combination of claim 1. 44. A W/O emulsion which comprises the active substance combination of claim 1. 45. A gel cream which comprises the active substance combination of claim 1. 46. A hydrodispersion which comprises the active substance combination of claim 1. 47. A method for the prophylaxis or treatment of UV or ozone-induced skin damage, wherein the method comprises applying to at least a part of the skin the preparation of claim 8. 48. A method for the prophylaxis or treatment of inflammatory and degenerative skin, wherein the method comprises applying to at least a part of the skin the preparation of claim 8. | A combination comprising creatine and/or creatinine and/or a derivative thereof and one or more retinoids. This Abstract is not intended to define the invention disclosed in the specification, nor intended to limit the scope of the invention in any way.1. An active substance combination comprising
(b) at least one of creatine, creatinine, a creatine derivative and a creatinine derivative, and (b) at least one retinoid. 2. The active substance combination of claim 1, wherein a weight ratio of creatinine to creatine is from about 10:1 to about 1:10. 3. The active substance combination of claim 2, wherein the weight ratio is from about 4:1 to about 3:7. 4. The active substance combination of claim 2, wherein the weight ratio is from about 2:1 to about 1:2. 5. The active substance combination of claim 1, wherein the at least one retinoid comprises at least one of retinol and retinyl palmitate. 6. The active substance combination of claim 1, wherein the at least one retinoid comprises retinyl palmitate. 7. The active substance combination of claim 1, wherein the at least one retinoid comprises retinol. 8. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 1. 9. The preparation of claim 8, wherein the preparation comprises from about 0.0005% to about 50% by weight of the active substance combination. 10. The preparation of claim 9, wherein the preparation comprises from about 0.01% to about 20% by weight of the active substance combination. 11. The preparation of claim 10, wherein the preparation comprises from about 0.02% to about 10% by weight of the active substance combination. 12. The preparation of claim 11, wherein the preparation comprises from about 0.02% to about 5% by weight of the active substance combination. 13. The preparation of claim 12, wherein the preparation comprises from about 0.5% to about 3% by weight of the active substance combination. 14. The preparation of claim 8, wherein the preparation comprises from about 0.001% to about 10% by weight of at least one of creatine and a creatine derivative. 15. The preparation of claim 14, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatine and a creatine derivative. 16. The preparation of claim 8, wherein the preparation comprises from about 0.001% to about 10% by weight of at least one of creatinine and a creatinine derivative. 17. The preparation of claim 15, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatinine and a creatinine derivative. 18. The preparation of claim 8, wherein the preparation comprises from about 0.001% to about 10% by weight of the at least one retinoid. 19. The preparation of claim 12, wherein the preparation comprises from about 0.01% to about 1% by weight of the at least one retinoid. 20. The preparation of claim 19, wherein the at least one retinoid comprises at least one of retinol and retinyl palmitate. 21. The preparation of claim 8, wherein the preparation further comprises from about 0.001% to about 30% by weight of glycerin. 22. The preparation of claim 9, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 23. The preparation of claim 10, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 24. The preparation of claim 11, wherein the preparation further comprises from about 1% to about 7% by weight of glycerin. 25. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 2. 26. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 3. 27. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 4. 28. A cosmetic or dermatological preparation which comprises an effective amount of the active substance combination of claim 5. 29. A cosmetic or dermatological preparation which comprises from about 0.01% to about 20% by weight of an active substance combination comprising
(a) at least one of creatine, creatinine, a creatine derivative and a creatinine derivative, and (b) at least one retinoid. 30. The preparation of claim 29, wherein a weight ratio of creatinine to creatine in the active substance combination is from about 4:1 to about 3:7. 31. The preparation of claim 30, wherein the weight ratio is from about 2:1 to about 1:2. 32. The preparation of claim 29, wherein the preparation comprises from about 0.02% to about 10% by weight of the active substance combination. 33. The preparation of claim 30, wherein the preparation comprises from about 0.5% to about 3% by weight of the active substance combination. 34. The preparation of claim 32, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatine and a creatine derivative. 35. The preparation of claim 32, wherein the preparation comprises from about 0.01% to about 1% by weight of at least one of creatinine and a creatinine derivative. 36. The preparation of claim 31, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 37. The preparation of claim 30, wherein the preparation further comprises from about 0.01% to about 15% by weight of glycerin. 38. The preparation of claim 32, wherein the preparation further comprises from about 1% to about 7% by weight of glycerin. 39. The preparation of claim 32, wherein the preparation comprises from about 0.001% to about 10% by weight of the at least one retinoid. 40. The preparation of claim 30, wherein the preparation comprises from about 0.01% to about 1% by weight of the at least one retinoid. 41. The preparation of claim 40, wherein the at least one retinoid comprises at least one of retinol and retinyl palmitate. 42. An O/W cream which comprises the active substance combination of claim 1. 43. An O/W emulsion which comprises the active substance combination of claim 1. 44. A W/O emulsion which comprises the active substance combination of claim 1. 45. A gel cream which comprises the active substance combination of claim 1. 46. A hydrodispersion which comprises the active substance combination of claim 1. 47. A method for the prophylaxis or treatment of UV or ozone-induced skin damage, wherein the method comprises applying to at least a part of the skin the preparation of claim 8. 48. A method for the prophylaxis or treatment of inflammatory and degenerative skin, wherein the method comprises applying to at least a part of the skin the preparation of claim 8. | 1,600 |
634 | 15,591,301 | 1,653 | A method of producing jellyfish collagen extract by combining hard water, frozen jellyfish, protease enzymes, and sodium bisulfate to form a mixture. Heating the mixture for a period of time to permit the mixture to react. The filter, concentrate, and dry the mixture. | 1. A method of producing jellyfish collagen extract, comprising the steps of:
obtaining a quantity of frozen jellyfish; adding the frozen jellyfish to a tank containing hard water; adding protease enzymes and sodium bisulfate to the tank to form a mixture; heating the tank for a period of time that permits a reaction in the mixture; filtering the mixture to remove sediment concentrate from the mixture to form a solid; and drying the solid to form a jellyfish collagen extract. 2. The method of claim 1 wherein the tank is heated to 60° C. 3. The method of claim 1 wherein a period between two and twenty hours permits the mixture to react. 4. The method of claim 1 wherein the mixture is concentrated through ultrafiltration. 5. The method of claim 1 wherein the mixture is concentrated through evaporation. 6. The method of claim 1 wherein the solid is spray dried. 7. The method of claim 1 wherein the jellyfish collagen extract includes type I, II, IV, and V collagen. 8. The method of claim 1 wherein the jellyfish collagen extract includes Aequorin, Calmodulin, and Coelenterazine. 9. The method of claim 1 wherein the jellyfish collagen extract includes mucopolysaccharides. 10. The method of claim 9 wherein the mucopolysaccharides include chondroitin and hyaluronic acid. 11. The method of claim 1 wherein the jellyfish collagen extract includes calcium binding proteins. 12. The method of claim 1 wherein the jellyfish collagen extract having 10.59% wt of chondroitin sulfate, 0.19% wt of glucosamine; 3.21% wt of hyaluronic acid, 6.58% wt hydroxyproline, and 29 to 40% wt of mucopolysaccharides. 13. The method of claim 1 wherein the jellyfish collagen extract having 2103 pmol/μg of coelenterazine, 2011 mg/kg of aequorin, and 406 mg/kg of calmodulin. 14. The method of claim 1 wherein the jellyfish collagen extract having a collagen acid soluble protein breakdown of 0.1% 3-hydroxyproline, 4.5% 4-hydroxyproline, 7.8% aspartic acid, 3.1% threonine, 4.6% serine, 9.7% glutamic acid, 7.5% proline, 30.2% glycine, 7.8% alanine, 0.6% half-cystine, 3.1% valine, 1.3% methionine, 2.1% isoleucine, 3.2% leucine, 0.9% tyrosine, 1.2% phenylalanine, 0.1% tryptophan, 3.2% hydoxylysine, and 3.1% lysine. 15. A method of producing jellyfish collagen extract, comprising the steps of:
obtaining a quantity of frozen jellyfish; adding hard water to a tank, wherein calcium has been removed from the hard water prior to being added to the tank; adding the frozen jellyfish to the tank; adding protease enzymes and sodium bisulfate to the tank to form a mixture; heating the tank for a period of time that permits a reaction in the mixture; filtering the mixture to remove sediment concentrate from the mixture to form a solid; and drying the solid to form a jellyfish collagen extract. 16. The method of claim 15 wherein the jellyfish collagen extract includes calcium binding proteins. 17. The method of claim 15 wherein the jellyfish collagen extract having 10.59% wt of chondroitin sulfate, 0.19% wt of glucosamine; 3.21% wt of hyaluronic acid, 6.58% wt hydroxyproline, and 29 to 40% wt of mucopolysaccharides. 18. The method of claim 15 wherein the jellyfish collagen extract having 2103 pmol/μg of coelenterazine, 2011 mg/kg of aequorin, and 406 mg/kg of calmodulin. 19. The method of claim 15 wherein the jellyfish collagen extract having a collagen acid soluble protein breakdown of 0.1% 3-hydroxyproline, 4.5% 4-hydroxyproline, 7.8% aspartic acid, 3.1% threonine, 4.6% serine, 9.7% glutamic acid, 7.5% proline, 30.2% glycine, 7.8% alanine, 0.6% half-cystine, 3.1% valine, 1.3% methionine, 2.1% isoleucine, 3.2% leucine, 0.9% t tyrosine, 1.2% phenylalanine, 0.1% tryptophan, 3.2% hydoxylysine, and 3.1% lysine. 20. A method of producing jellyfish collagen extract, comprising the steps of:
obtaining a quantity of frozen jellyfish; adding the frozen jellyfish to a tank containing water; adding protease enzymes and sodium bisulfate to the tank to form a mixture; heating the tank for a period of time that permits a reaction in the mixture; filtering the mixture to remove sediment concentrate from the mixture to form a solid; and drying the solid to form a jellyfish collagen extract. | A method of producing jellyfish collagen extract by combining hard water, frozen jellyfish, protease enzymes, and sodium bisulfate to form a mixture. Heating the mixture for a period of time to permit the mixture to react. The filter, concentrate, and dry the mixture.1. A method of producing jellyfish collagen extract, comprising the steps of:
obtaining a quantity of frozen jellyfish; adding the frozen jellyfish to a tank containing hard water; adding protease enzymes and sodium bisulfate to the tank to form a mixture; heating the tank for a period of time that permits a reaction in the mixture; filtering the mixture to remove sediment concentrate from the mixture to form a solid; and drying the solid to form a jellyfish collagen extract. 2. The method of claim 1 wherein the tank is heated to 60° C. 3. The method of claim 1 wherein a period between two and twenty hours permits the mixture to react. 4. The method of claim 1 wherein the mixture is concentrated through ultrafiltration. 5. The method of claim 1 wherein the mixture is concentrated through evaporation. 6. The method of claim 1 wherein the solid is spray dried. 7. The method of claim 1 wherein the jellyfish collagen extract includes type I, II, IV, and V collagen. 8. The method of claim 1 wherein the jellyfish collagen extract includes Aequorin, Calmodulin, and Coelenterazine. 9. The method of claim 1 wherein the jellyfish collagen extract includes mucopolysaccharides. 10. The method of claim 9 wherein the mucopolysaccharides include chondroitin and hyaluronic acid. 11. The method of claim 1 wherein the jellyfish collagen extract includes calcium binding proteins. 12. The method of claim 1 wherein the jellyfish collagen extract having 10.59% wt of chondroitin sulfate, 0.19% wt of glucosamine; 3.21% wt of hyaluronic acid, 6.58% wt hydroxyproline, and 29 to 40% wt of mucopolysaccharides. 13. The method of claim 1 wherein the jellyfish collagen extract having 2103 pmol/μg of coelenterazine, 2011 mg/kg of aequorin, and 406 mg/kg of calmodulin. 14. The method of claim 1 wherein the jellyfish collagen extract having a collagen acid soluble protein breakdown of 0.1% 3-hydroxyproline, 4.5% 4-hydroxyproline, 7.8% aspartic acid, 3.1% threonine, 4.6% serine, 9.7% glutamic acid, 7.5% proline, 30.2% glycine, 7.8% alanine, 0.6% half-cystine, 3.1% valine, 1.3% methionine, 2.1% isoleucine, 3.2% leucine, 0.9% tyrosine, 1.2% phenylalanine, 0.1% tryptophan, 3.2% hydoxylysine, and 3.1% lysine. 15. A method of producing jellyfish collagen extract, comprising the steps of:
obtaining a quantity of frozen jellyfish; adding hard water to a tank, wherein calcium has been removed from the hard water prior to being added to the tank; adding the frozen jellyfish to the tank; adding protease enzymes and sodium bisulfate to the tank to form a mixture; heating the tank for a period of time that permits a reaction in the mixture; filtering the mixture to remove sediment concentrate from the mixture to form a solid; and drying the solid to form a jellyfish collagen extract. 16. The method of claim 15 wherein the jellyfish collagen extract includes calcium binding proteins. 17. The method of claim 15 wherein the jellyfish collagen extract having 10.59% wt of chondroitin sulfate, 0.19% wt of glucosamine; 3.21% wt of hyaluronic acid, 6.58% wt hydroxyproline, and 29 to 40% wt of mucopolysaccharides. 18. The method of claim 15 wherein the jellyfish collagen extract having 2103 pmol/μg of coelenterazine, 2011 mg/kg of aequorin, and 406 mg/kg of calmodulin. 19. The method of claim 15 wherein the jellyfish collagen extract having a collagen acid soluble protein breakdown of 0.1% 3-hydroxyproline, 4.5% 4-hydroxyproline, 7.8% aspartic acid, 3.1% threonine, 4.6% serine, 9.7% glutamic acid, 7.5% proline, 30.2% glycine, 7.8% alanine, 0.6% half-cystine, 3.1% valine, 1.3% methionine, 2.1% isoleucine, 3.2% leucine, 0.9% t tyrosine, 1.2% phenylalanine, 0.1% tryptophan, 3.2% hydoxylysine, and 3.1% lysine. 20. A method of producing jellyfish collagen extract, comprising the steps of:
obtaining a quantity of frozen jellyfish; adding the frozen jellyfish to a tank containing water; adding protease enzymes and sodium bisulfate to the tank to form a mixture; heating the tank for a period of time that permits a reaction in the mixture; filtering the mixture to remove sediment concentrate from the mixture to form a solid; and drying the solid to form a jellyfish collagen extract. | 1,600 |
635 | 13,852,654 | 1,615 | Disclosed is a hair conditioning composition comprising: a mono-alkyl amine cationic surfactant; a di-alkyl quaternized ammonium salt cationic surfactant; a high melting point fatty compound; a deposition polymer having specific monomers; a silicone compound; and an aqueous carrier. The composition of the present invention provides improved deposition of cationic surfactant, fatty compounds, and/or silicone compounds, especially silicone compounds on damaged hair. | 1. A hair conditioning composition comprising by weight:
(a) from about 0.1% to about 8% of a mono-alkyl amine cationic surfactant which is a primary, secondary, and tertiary amines having one long alkyl or alkenyl group of from about 12 to about 30 carbon atoms; (b) from about 0.05% to about 6% of a di-alkyl quaternized ammonium salt cationic surfactant having two long alkyl groups of from about 12 to about 30 carbon atoms; (c) from about 1% to about 15% of a high melting point fatty compound; (d) from about 0.05% to about 6% of a deposition polymer which is a copolymer comprising: a vinyl monomer (A) with a carboxyl group in the structure; and a vinyl monomer (B) expressed by the following formula (1):
CH2═C(R1)—CO—X-(Q-O)r—R2 (1)
wherein: R1 represents a hydrogen atom or a methyl group; R2 represents a hydrogen atom or an alkyl group with from 1 to 5 carbon atoms, which may have a substitution group; Q represents an alkylene group with from 2 to 4 carbon atoms which may also have a substitution group; r represents an integer from 2 to 15; and X represents an oxygen atom or an NH group; and, in the following structure -(Q-O)r—R2, the number of atoms bonded in a straight chain is 70 or less;
and wherein the vinyl monomer (A) is contained at a level of from about 10 mass % to about 50 mass %, and the vinyl monomer (B) is contained at level of from about 50 mass % to about 90 mass %; and
(e) from about 0.05% to about 15% of a silicone compound; and
(f) an aqueous carrier. 2. The composition of claim 1 wherein, in the formula (1), r represents from about 3 to about 12. 3. The composition of claim 1 wherein, in the formula (1), X represents an oxygen atom. 4. The composition of claim 1 wherein the vinyl monomer (A) is expressed by the following formula (2) or the following formula (3):
CH2═C(R3)—CO—(O—(CH2)m—CO)—OH (2)
wherein R3 represents a hydrogen atom or a methyl group, m represents an integer of 1 through 4, and n represents an integer of 0 through 4;
CH2═C(R4)—COO—(CH2)p—OOC—(CH2)q—COOH (3)
wherein R4 represents a hydrogen atom or a methyl group, p and q independently represent an integer of 2 through 6. 5. The composition of claim 1 wherein the deposition polymer has a weighted average molecular weight of from about 3,000 to about 2,000,000. 6. The composition of claim 1 wherein the deposition polymer is anionic. 7. The hair conditioning composition of claim 1 wherein the amine is a tertiary amidoamine having an alkyl group of from about 16 to about 22 carbon atoms. 8. The hair conditioning composition of claim 1 wherein the mono-alkyl amine cationic surfactant is used in combination with an acid selected from the group consisting of l-glutamic acid, lactic acid, hydrochloric acid, malic acid, succinic acid, acetic acid, fumaric acid, tartaric acid, citric acid, l-glutamic hydrochloride, maleic acid, and mixtures thereof, at a level such that the mole ratio of the amine to the acid is from about 1:0.3 to about 1:2. 9. The composition of claim 1 wherein the silicone compound is selected from polyalkyl siloxanes, polyaryl siloxanes, polyalkylaryl siloxanes, polyether siloxane copolymers, amino substituted silicones, quaternized silicones including quaternized silicones comprising terminal ester groups, having a viscosity of lower than 100,000 mPa·s and a D block length of greater than 200 D units, and mixtures thereof. | Disclosed is a hair conditioning composition comprising: a mono-alkyl amine cationic surfactant; a di-alkyl quaternized ammonium salt cationic surfactant; a high melting point fatty compound; a deposition polymer having specific monomers; a silicone compound; and an aqueous carrier. The composition of the present invention provides improved deposition of cationic surfactant, fatty compounds, and/or silicone compounds, especially silicone compounds on damaged hair.1. A hair conditioning composition comprising by weight:
(a) from about 0.1% to about 8% of a mono-alkyl amine cationic surfactant which is a primary, secondary, and tertiary amines having one long alkyl or alkenyl group of from about 12 to about 30 carbon atoms; (b) from about 0.05% to about 6% of a di-alkyl quaternized ammonium salt cationic surfactant having two long alkyl groups of from about 12 to about 30 carbon atoms; (c) from about 1% to about 15% of a high melting point fatty compound; (d) from about 0.05% to about 6% of a deposition polymer which is a copolymer comprising: a vinyl monomer (A) with a carboxyl group in the structure; and a vinyl monomer (B) expressed by the following formula (1):
CH2═C(R1)—CO—X-(Q-O)r—R2 (1)
wherein: R1 represents a hydrogen atom or a methyl group; R2 represents a hydrogen atom or an alkyl group with from 1 to 5 carbon atoms, which may have a substitution group; Q represents an alkylene group with from 2 to 4 carbon atoms which may also have a substitution group; r represents an integer from 2 to 15; and X represents an oxygen atom or an NH group; and, in the following structure -(Q-O)r—R2, the number of atoms bonded in a straight chain is 70 or less;
and wherein the vinyl monomer (A) is contained at a level of from about 10 mass % to about 50 mass %, and the vinyl monomer (B) is contained at level of from about 50 mass % to about 90 mass %; and
(e) from about 0.05% to about 15% of a silicone compound; and
(f) an aqueous carrier. 2. The composition of claim 1 wherein, in the formula (1), r represents from about 3 to about 12. 3. The composition of claim 1 wherein, in the formula (1), X represents an oxygen atom. 4. The composition of claim 1 wherein the vinyl monomer (A) is expressed by the following formula (2) or the following formula (3):
CH2═C(R3)—CO—(O—(CH2)m—CO)—OH (2)
wherein R3 represents a hydrogen atom or a methyl group, m represents an integer of 1 through 4, and n represents an integer of 0 through 4;
CH2═C(R4)—COO—(CH2)p—OOC—(CH2)q—COOH (3)
wherein R4 represents a hydrogen atom or a methyl group, p and q independently represent an integer of 2 through 6. 5. The composition of claim 1 wherein the deposition polymer has a weighted average molecular weight of from about 3,000 to about 2,000,000. 6. The composition of claim 1 wherein the deposition polymer is anionic. 7. The hair conditioning composition of claim 1 wherein the amine is a tertiary amidoamine having an alkyl group of from about 16 to about 22 carbon atoms. 8. The hair conditioning composition of claim 1 wherein the mono-alkyl amine cationic surfactant is used in combination with an acid selected from the group consisting of l-glutamic acid, lactic acid, hydrochloric acid, malic acid, succinic acid, acetic acid, fumaric acid, tartaric acid, citric acid, l-glutamic hydrochloride, maleic acid, and mixtures thereof, at a level such that the mole ratio of the amine to the acid is from about 1:0.3 to about 1:2. 9. The composition of claim 1 wherein the silicone compound is selected from polyalkyl siloxanes, polyaryl siloxanes, polyalkylaryl siloxanes, polyether siloxane copolymers, amino substituted silicones, quaternized silicones including quaternized silicones comprising terminal ester groups, having a viscosity of lower than 100,000 mPa·s and a D block length of greater than 200 D units, and mixtures thereof. | 1,600 |
636 | 13,612,929 | 1,662 | The present invention provides novel compositions for use to enhance weed control. Specifically, the present invention provides for methods and compositions that modulate acetolactate synthase in weed species. The present invention also provides for combinations of compositions and methods that enhance weed control. | 1. A method of plant control comprising: treating a plant with a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an ALS gene sequence or fragment thereof, or to an RNA transcript of said ALS gene sequence or fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, whereby said plant growth or development or reproductive ability is reduced or said plant is more sensitive to an ALS inhibitor herbicide relative to a plant not treated with said composition. 2. The method as claimed in claim 1, wherein said transfer agent is an organosilicone surfactant composition or compound contained therein. 3. The method as claimed in claim 1, wherein said polynucleotide fragment is 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 4. The method as claimed in claim 3, wherein said polynucleotide fragment is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 5. The method as claimed in claim 1, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus chlorostachys, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Ambrosia trifida, Kochia scoparia, Abutilon theophrasti, Chenopodium album, Commelina diffusa, Conyza candensis Digitaria sanguinalis Euphorbia heterophylla, and Lolium multiflorum. 6. The method as claimed in claim 1, wherein said composition further comprises said ALS inhibitor herbicide and external application to a plant with said composition. 7. The method as claimed in claim 6, wherein said composition further comprises one or more herbicides different from said ALS inhibitor herbicide. 8. The method as claimed in claim 3, wherein said composition comprises any combination of two or more of said polynucleotide fragments and external application to a plant with said composition. 9. A composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an ALS gene sequence or fragment thereof, or to an RNA transcript of said ALS gene sequence or fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an ALS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 10. The composition of claim 9, wherein said transfer agent is an organosilicone composition. 11. The composition of claim 9, wherein said polynucleotide fragment is 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 12. The composition of claim 9, wherein said polynucleotide is selected from the group consisting of SEQ ID NO: 46-1363 and 1789-4166. 13. The composition of claim 9, wherein said polynucleotide is selected from the group consisting of SEQ ID NO: 1364-1691 and 4167-4201. 14. The composition of claim 9, further comprising an ALS inhibitor herbicide. 15. The composition of claim 14, wherein said ALS inhibitor molecule is selected from the group consisting of amidosulfuron, azimsulfuron, bensulfuron-methyl, chlorimuron-ethyl, chlorsulfuron, cinosulfuron, cyclosulfamuron, ethametsulfuron-methyl, ethoxysulfuron, flazasulfuron, flupyrsulfuron-methyl-Na, foramsulfuron, halosulfuron-methyl, imazosulfuron, iodosulfuron, metsulfuron-methyl, nicosulfuron, oxasulfuron, primisulfuron-methyl, prosulfuron, pyrazosulfuron-ethyl, rimsulfuron, sulfometuron-methyl, sulfosulfuron, thifensulfuron-methyl, triasulfuron, tribenuron-methyl, trifloxysulfuron, triflusulfuron-methyl, tritosulfuron, imazapic, imazamethabenz-methyl, imazamox, imazapyr, imazaquin, imazethapyr, cloransulam-methyl, diclosulam, florasulam, flumetsulam, metosulam, bispyribac-Na, pyribenzoxim, pyriftalid, pyrithiobac-Na, pyriminobac-methyl, flucarbazone-Na, and procarbazone-Na. 16. The composition of claim 14, further comprising a co-herbicide. 17. A method of reducing expression of an ALS gene in a plant comprising: external application to a plant of a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an ALS gene sequence or fragment thereof, or to the RNA transcript of said ALS gene sequence or fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, whereby said expression of said ALS gene is reduced relative to a plant in which the composition was not applied. 18. The method as claimed in claim 17, wherein said transfer agent is an organosilicone compound. 19. The method as claimed in claim 17, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 20. The method as claimed in 17, wherein said polynucleotide molecule is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 21. A microbial expression cassette comprising a polynucleotide fragment of 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 22. A method of making a polynucleotide comprising a) transforming the microbial expression cassette of claim 21 into a microbe; b) growing said microbe; c) harvesting a polynucleotide from said microbe, wherein said polynucleotide is 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 23. A method of identifying polynucleotides useful in modulating ALS gene expression when externally treating a plant comprising: a) providing a plurality of polynucleotides that comprise a region essentially identical or essentially complementary to a polynucleotide fragment of 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788; b) externally treating said plant with one or more of said polynucleotides and a transfer agent; c) analyzing said plant or extract for modulation of ALS gene expression, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 24. The method as claimed in 23, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus chlorostachys, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Ambrosia trifida, Kochia scoparia, Abutilon theophrasti, Chenopodium album, Commelina diffusa, Conyza candensis Digitaria sanguinalis Euphorbia heterophylla, and Lolium multiflorum. 25. The method as claimed in 23, wherein said ALS gene expression is reduced relative to a plant not treated with said polynucleotide fragment and a transfer agent. 26. The method as claimed in 23, wherein said transfer agent is an organosilicone compound. 27. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a co-herbicide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an ALS gene sequence, or to a portion of an RNA transcript of said ALS gene sequence, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO: 1-45 and 1692-1788 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an ALS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 28. The agricultural chemical composition of claim 27, wherein said co-herbicide is selected from the group consisting of amide herbicides, arsenical herbicides, benzothiazole herbicides, benzoylcyclohexanedione herbicides, benzofuranyl alkylsulfonate herbicides, carbamate herbicides, cyclohexene oxime herbicides, cyclopropylisoxazole herbicides, dicarboximide herbicides, dinitroaniline herbicides, dinitrophenol herbicides, diphenyl ether herbicides, dithiocarbamate herbicides, halogenated aliphatic herbicides, imidazolinone herbicides, inorganic herbicides, nitrile herbicides, organophosphorus herbicides, oxadiazolone herbicides, oxazole herbicides, phenoxy herbicides, phenylenediamine herbicides, pyrazole herbicides, pyridazine herbicides, pyridazinone herbicides, pyridine herbicides, pyrimidinediamine herbicides, pyrimidinyloxybenzylamine herbicides, quaternary ammonium herbicides, thiocarbamate herbicides, thiocarbonate herbicides, thiourea herbicides, triazine herbicides, triazinone herbicides, triazole herbicides, triazolone herbicides, triazolopyrimidine herbicides, uracil herbicides, and urea herbicides. 29. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a pesticide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an ALS gene sequence or a fragment thereof, or to a portion of an RNA transcript of said ALS gene sequence or a fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an ALS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 30. The agricultural chemical composition of claim 29, wherein said pesticide is selected from the group consisting of insecticides, fungicides, nematocides, bactericides, acaricides, growth regulators, chemosterilants, semiochemicals, repellents, attractants, pheromones, feeding stimulants, and biopesticides. 31. A polynucleotide molecule applied to the surface of a plant that enhances said plant sensitivity to a glyphosate containing herbicide composition, wherein said polynucleotide comprises a homologous or complementary polynucleotide having at least 85 percent identity to a polynucleotide selected from the group consisting of SEQ ID NO:4202 and 4218-4247. | The present invention provides novel compositions for use to enhance weed control. Specifically, the present invention provides for methods and compositions that modulate acetolactate synthase in weed species. The present invention also provides for combinations of compositions and methods that enhance weed control.1. A method of plant control comprising: treating a plant with a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an ALS gene sequence or fragment thereof, or to an RNA transcript of said ALS gene sequence or fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, whereby said plant growth or development or reproductive ability is reduced or said plant is more sensitive to an ALS inhibitor herbicide relative to a plant not treated with said composition. 2. The method as claimed in claim 1, wherein said transfer agent is an organosilicone surfactant composition or compound contained therein. 3. The method as claimed in claim 1, wherein said polynucleotide fragment is 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 4. The method as claimed in claim 3, wherein said polynucleotide fragment is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 5. The method as claimed in claim 1, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus chlorostachys, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Ambrosia trifida, Kochia scoparia, Abutilon theophrasti, Chenopodium album, Commelina diffusa, Conyza candensis Digitaria sanguinalis Euphorbia heterophylla, and Lolium multiflorum. 6. The method as claimed in claim 1, wherein said composition further comprises said ALS inhibitor herbicide and external application to a plant with said composition. 7. The method as claimed in claim 6, wherein said composition further comprises one or more herbicides different from said ALS inhibitor herbicide. 8. The method as claimed in claim 3, wherein said composition comprises any combination of two or more of said polynucleotide fragments and external application to a plant with said composition. 9. A composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an ALS gene sequence or fragment thereof, or to an RNA transcript of said ALS gene sequence or fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an ALS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 10. The composition of claim 9, wherein said transfer agent is an organosilicone composition. 11. The composition of claim 9, wherein said polynucleotide fragment is 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 12. The composition of claim 9, wherein said polynucleotide is selected from the group consisting of SEQ ID NO: 46-1363 and 1789-4166. 13. The composition of claim 9, wherein said polynucleotide is selected from the group consisting of SEQ ID NO: 1364-1691 and 4167-4201. 14. The composition of claim 9, further comprising an ALS inhibitor herbicide. 15. The composition of claim 14, wherein said ALS inhibitor molecule is selected from the group consisting of amidosulfuron, azimsulfuron, bensulfuron-methyl, chlorimuron-ethyl, chlorsulfuron, cinosulfuron, cyclosulfamuron, ethametsulfuron-methyl, ethoxysulfuron, flazasulfuron, flupyrsulfuron-methyl-Na, foramsulfuron, halosulfuron-methyl, imazosulfuron, iodosulfuron, metsulfuron-methyl, nicosulfuron, oxasulfuron, primisulfuron-methyl, prosulfuron, pyrazosulfuron-ethyl, rimsulfuron, sulfometuron-methyl, sulfosulfuron, thifensulfuron-methyl, triasulfuron, tribenuron-methyl, trifloxysulfuron, triflusulfuron-methyl, tritosulfuron, imazapic, imazamethabenz-methyl, imazamox, imazapyr, imazaquin, imazethapyr, cloransulam-methyl, diclosulam, florasulam, flumetsulam, metosulam, bispyribac-Na, pyribenzoxim, pyriftalid, pyrithiobac-Na, pyriminobac-methyl, flucarbazone-Na, and procarbazone-Na. 16. The composition of claim 14, further comprising a co-herbicide. 17. A method of reducing expression of an ALS gene in a plant comprising: external application to a plant of a composition comprising a polynucleotide and a transfer agent, wherein said polynucleotide is essentially identical or essentially complementary to an ALS gene sequence or fragment thereof, or to the RNA transcript of said ALS gene sequence or fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, whereby said expression of said ALS gene is reduced relative to a plant in which the composition was not applied. 18. The method as claimed in claim 17, wherein said transfer agent is an organosilicone compound. 19. The method as claimed in claim 17, wherein said polynucleotide fragment is 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 20. The method as claimed in 17, wherein said polynucleotide molecule is selected from the group consisting of sense or anti-sense ssDNA or ssRNA, dsRNA, or dsDNA, or dsDNA/RNA hybrids. 21. A microbial expression cassette comprising a polynucleotide fragment of 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 22. A method of making a polynucleotide comprising a) transforming the microbial expression cassette of claim 21 into a microbe; b) growing said microbe; c) harvesting a polynucleotide from said microbe, wherein said polynucleotide is 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788. 23. A method of identifying polynucleotides useful in modulating ALS gene expression when externally treating a plant comprising: a) providing a plurality of polynucleotides that comprise a region essentially identical or essentially complementary to a polynucleotide fragment of 18 contiguous, 19 contiguous nucleotides, 20 contiguous nucleotides or at least 21 contiguous nucleotides in length and at least 85 percent identical to an ALS gene sequence selected from the group consisting of SEQ ID NO:1-45 and 1692-1788; b) externally treating said plant with one or more of said polynucleotides and a transfer agent; c) analyzing said plant or extract for modulation of ALS gene expression, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an EPSPS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 24. The method as claimed in 23, wherein said plant is selected from the group consisting of Amaranthus palmeri, Amaranthus rudis, Amaranthus chlorostachys, Amaranthus graecizans, Amaranthus hybridus, Amaranthus lividus, Amaranthus spinosus, Amaranthus thunbergii, Amaranthus viridis, Ambrosia trifida, Kochia scoparia, Abutilon theophrasti, Chenopodium album, Commelina diffusa, Conyza candensis Digitaria sanguinalis Euphorbia heterophylla, and Lolium multiflorum. 25. The method as claimed in 23, wherein said ALS gene expression is reduced relative to a plant not treated with said polynucleotide fragment and a transfer agent. 26. The method as claimed in 23, wherein said transfer agent is an organosilicone compound. 27. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a co-herbicide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an ALS gene sequence, or to a portion of an RNA transcript of said ALS gene sequence, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO: 1-45 and 1692-1788 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an ALS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 28. The agricultural chemical composition of claim 27, wherein said co-herbicide is selected from the group consisting of amide herbicides, arsenical herbicides, benzothiazole herbicides, benzoylcyclohexanedione herbicides, benzofuranyl alkylsulfonate herbicides, carbamate herbicides, cyclohexene oxime herbicides, cyclopropylisoxazole herbicides, dicarboximide herbicides, dinitroaniline herbicides, dinitrophenol herbicides, diphenyl ether herbicides, dithiocarbamate herbicides, halogenated aliphatic herbicides, imidazolinone herbicides, inorganic herbicides, nitrile herbicides, organophosphorus herbicides, oxadiazolone herbicides, oxazole herbicides, phenoxy herbicides, phenylenediamine herbicides, pyrazole herbicides, pyridazine herbicides, pyridazinone herbicides, pyridine herbicides, pyrimidinediamine herbicides, pyrimidinyloxybenzylamine herbicides, quaternary ammonium herbicides, thiocarbamate herbicides, thiocarbonate herbicides, thiourea herbicides, triazine herbicides, triazinone herbicides, triazole herbicides, triazolone herbicides, triazolopyrimidine herbicides, uracil herbicides, and urea herbicides. 29. An agricultural chemical composition comprising an admixture of a polynucleotide and a glyphosate herbicide and a pesticide, wherein said polynucleotide is essentially identical or essentially complementary to a portion of an ALS gene sequence or a fragment thereof, or to a portion of an RNA transcript of said ALS gene sequence or a fragment thereof, wherein said ALS gene sequence is selected from the group consisting of SEQ ID NO:1-45 and 1692-1788 or a polynucleotide fragment thereof, and whereby a plant treated with said composition has its growth or development or reproductive ability regulated, suppressed or delayed or said plant is more sensitive to an ALS inhibitor herbicide as a result of said polynucleotide containing composition relative to a plant not treated with said composition. 30. The agricultural chemical composition of claim 29, wherein said pesticide is selected from the group consisting of insecticides, fungicides, nematocides, bactericides, acaricides, growth regulators, chemosterilants, semiochemicals, repellents, attractants, pheromones, feeding stimulants, and biopesticides. 31. A polynucleotide molecule applied to the surface of a plant that enhances said plant sensitivity to a glyphosate containing herbicide composition, wherein said polynucleotide comprises a homologous or complementary polynucleotide having at least 85 percent identity to a polynucleotide selected from the group consisting of SEQ ID NO:4202 and 4218-4247. | 1,600 |
637 | 14,971,343 | 1,619 | A cationic compatible metal oxide and oral care compositions containing the metal oxide are disclosed. Preferably, the oral care compositions contain a cationic compatible metal oxide and a cationic ingredient, such as for example, a cationic antibacterial agent such as cetyl pyridinium chloride (“CPC”). The cationic compatible metal oxide, e.g. silica, comprise metal oxide particles having a subtantially negative surface charge and a vitamin deposited onto or reacted with the surface of the metal oxide particles in an amount sufficient to provide a substantially positive surface charge on the metal oxide particle. Processes for making and using the vitamin modified metal oxide in oral care compositions are also disclosed. | 1-11. (canceled) 13. An oral care composition comprising
(1) an antibacterial effective amount of at least one cationic antibacterial agent; and (2) a cationic compatible metal oxide comprising a metal oxide particle having a negative surface charge and a vitamin molecule immobilized on the surface of the metal oxide particle in an amount sufficient to provide a substantially positive charge on the surface of the metal oxide particle, said vitamin molecule being present in an amount sufficient to repel or minimize any reaction of the metal oxide with said at least one cationic antibacterial agent. 14. The oral care composition of claim 13, wherein the cationic antibacterial agent is cetylpridinium chloride (CPC). 15. The oral care composition of claim 13, wherein the metal oxide of the metal oxide particle is selected from the group consisting of silica, titania, zirconia, silica alumina and mixtures thereof. 16. The oral care compositon of claim 13, wherein the amount of vitamin molecule immobilized on the surface of the metal oxide particle ranges from about 0.01 wt % to about 10.0 wt % of the metal oxide. 17. The oral care composition of claim 16, wherein the vitamin molecule is immobilized directly onto the surface of the metal oxide particle. 18. The oral care composition of claim 13, wherein the vitamin molecule is selected from the group consisting of vitamin B, vitamin M, vitamin U, their salts and derivatives, and mixtures thereof. 19. The oral care composition of claim 18, wherein the vitamin molecule is a vitamin B. 20. The oral care composition of claim 19, wherein the vitamin B is selected from the group consisting of vitamin B1, vitamin B2, vitamin B6, salts thereof, and mixtures thereof. 21. The oral care composition of claim 20, wherein the metal oxide particle is a silica particle. 22. The oral care composition of claim 17, wherein the vitamin molecule is impregnated onto the surface of the metal oxide particle. 23. The oral care composition of claim 17, wherein the vitamin molecule is adsorbed directly onto the surface of the metal oxide particle. 24. The oral care composition of claim 17, wherein the vitamin molecule is chemically reacted with the surface of the metal oxide particle. 25. The oral care composition of claim 13, wherein the composition comprises a dentrifice, a chewing gum, a mouthwash, or a mixture thereof. 26. The oral care composition of claim 25, wherein the composition is a dentrifice selected from the group consisting of toothpastes, tooth powders, denture creams and mixtures thereof. 27. The oral care composition of claim 13, further comprising at least one material selected from the group consisting of thickening agents, whiteners, abrasive agents, flavorants, humectants, detergents, surfactants, fluoride supplying compounds, desensitizing agents and mixtures thereof. 28. The oral care composition of claim 13, further compring at least one sweetening agent. 29. The oral care composition of claim 13, further comprising at least one additional cationic antibacterial agent. 30. A method of reducing the presence of microorganisms in an oral cavity of a warm-blooded animal, said method comprising administering to the oral cavity of the warm-blooded animal an effective amount of the oral care composition of claim 13. 31-35. (canceled) 36. The oral care composition of claim 21, wherein the silica particle has a % cetyl pridinium chloride (CPC) computability of at least 10%, wherein % CPC compatibility indicates a molar percentage of CPC in a test solution resulting from (i) contacting 3 grams of vitamin treated silica with 27 grams of a 0.3 wt % CPC solution for 7 days at 140° F. to form a heated solution, (ii) cooling the heated solution to form a cooled solution, (iii) centrifuging the cooled solution at 10,000 rpm for 10 minutes to form two phases comprising vitamin treated silica and a separated solution, (iv) extracting a 1.0 gram sample of fluid from the separated solution, and diluting the 1.0 gram sample of fluid with deionized water at a ratio of 20 parts deionized water to 1 part sample of fluid. 37. The oral care composition of claim 13, wherein the metal oxide particle is a silica particle, and the amount of vitamin molecule immobilized on the surface of the silica particle ranges from about 0.5 to about 1.0 wt % of the silica particle. | A cationic compatible metal oxide and oral care compositions containing the metal oxide are disclosed. Preferably, the oral care compositions contain a cationic compatible metal oxide and a cationic ingredient, such as for example, a cationic antibacterial agent such as cetyl pyridinium chloride (“CPC”). The cationic compatible metal oxide, e.g. silica, comprise metal oxide particles having a subtantially negative surface charge and a vitamin deposited onto or reacted with the surface of the metal oxide particles in an amount sufficient to provide a substantially positive surface charge on the metal oxide particle. Processes for making and using the vitamin modified metal oxide in oral care compositions are also disclosed.1-11. (canceled) 13. An oral care composition comprising
(1) an antibacterial effective amount of at least one cationic antibacterial agent; and (2) a cationic compatible metal oxide comprising a metal oxide particle having a negative surface charge and a vitamin molecule immobilized on the surface of the metal oxide particle in an amount sufficient to provide a substantially positive charge on the surface of the metal oxide particle, said vitamin molecule being present in an amount sufficient to repel or minimize any reaction of the metal oxide with said at least one cationic antibacterial agent. 14. The oral care composition of claim 13, wherein the cationic antibacterial agent is cetylpridinium chloride (CPC). 15. The oral care composition of claim 13, wherein the metal oxide of the metal oxide particle is selected from the group consisting of silica, titania, zirconia, silica alumina and mixtures thereof. 16. The oral care compositon of claim 13, wherein the amount of vitamin molecule immobilized on the surface of the metal oxide particle ranges from about 0.01 wt % to about 10.0 wt % of the metal oxide. 17. The oral care composition of claim 16, wherein the vitamin molecule is immobilized directly onto the surface of the metal oxide particle. 18. The oral care composition of claim 13, wherein the vitamin molecule is selected from the group consisting of vitamin B, vitamin M, vitamin U, their salts and derivatives, and mixtures thereof. 19. The oral care composition of claim 18, wherein the vitamin molecule is a vitamin B. 20. The oral care composition of claim 19, wherein the vitamin B is selected from the group consisting of vitamin B1, vitamin B2, vitamin B6, salts thereof, and mixtures thereof. 21. The oral care composition of claim 20, wherein the metal oxide particle is a silica particle. 22. The oral care composition of claim 17, wherein the vitamin molecule is impregnated onto the surface of the metal oxide particle. 23. The oral care composition of claim 17, wherein the vitamin molecule is adsorbed directly onto the surface of the metal oxide particle. 24. The oral care composition of claim 17, wherein the vitamin molecule is chemically reacted with the surface of the metal oxide particle. 25. The oral care composition of claim 13, wherein the composition comprises a dentrifice, a chewing gum, a mouthwash, or a mixture thereof. 26. The oral care composition of claim 25, wherein the composition is a dentrifice selected from the group consisting of toothpastes, tooth powders, denture creams and mixtures thereof. 27. The oral care composition of claim 13, further comprising at least one material selected from the group consisting of thickening agents, whiteners, abrasive agents, flavorants, humectants, detergents, surfactants, fluoride supplying compounds, desensitizing agents and mixtures thereof. 28. The oral care composition of claim 13, further compring at least one sweetening agent. 29. The oral care composition of claim 13, further comprising at least one additional cationic antibacterial agent. 30. A method of reducing the presence of microorganisms in an oral cavity of a warm-blooded animal, said method comprising administering to the oral cavity of the warm-blooded animal an effective amount of the oral care composition of claim 13. 31-35. (canceled) 36. The oral care composition of claim 21, wherein the silica particle has a % cetyl pridinium chloride (CPC) computability of at least 10%, wherein % CPC compatibility indicates a molar percentage of CPC in a test solution resulting from (i) contacting 3 grams of vitamin treated silica with 27 grams of a 0.3 wt % CPC solution for 7 days at 140° F. to form a heated solution, (ii) cooling the heated solution to form a cooled solution, (iii) centrifuging the cooled solution at 10,000 rpm for 10 minutes to form two phases comprising vitamin treated silica and a separated solution, (iv) extracting a 1.0 gram sample of fluid from the separated solution, and diluting the 1.0 gram sample of fluid with deionized water at a ratio of 20 parts deionized water to 1 part sample of fluid. 37. The oral care composition of claim 13, wherein the metal oxide particle is a silica particle, and the amount of vitamin molecule immobilized on the surface of the silica particle ranges from about 0.5 to about 1.0 wt % of the silica particle. | 1,600 |
638 | 14,898,606 | 1,617 | Present invention relates to an aqueous oxidizing composition comprising at least one hydrophobic dye which provides improved mixing of such composition with an alkalizing composition prior to use and/or application such as onto hair. | 1. An aqueous emulsion composition comprising:
one or more oxidizing agent, one or both of: one or more fatty alcohol and one or more oil, one or more surfactant, and one or more dyestuff having a log P value at 25° C. of higher than or equal to 2, wherein the composition is free of solid peroxides or persulphates, or free of both solid peroxides and persulphates, and has a pH below 7. 2. The composition according to claim 1, wherein the one or more oxidizing agent is hydrogen peroxide and present at a concentration of 1 to 20% by weight calculated to the total composition. 3. The composition according to claim 1, wherein the one or more fatty alcohol is selected from compounds according to general structure
R30—OH
wherein R30 is a saturated or unsaturated, straight or branched alkyl chain having 12 to 30 C atoms which may also be substituted with one or more OH groups. 4. The composition according to claim 3, wherein the one or more fatty alcohol is selected from arachidyl alcohol, behenyl alcohol, brassica alcohol, C9-11 alcohols, C10-16 alcohols, C12-13 alcohols, C12-15 alcohols, C12-16 alcohols, C14-15 alcohols, C14-22 alcohols, C20-22 alcohols, caprylyl alcohol, cetearyl alcohol, cetyl alcohol, coconut alcohol, decyl alcohol, hydrogenated brassica alcohol, hydrogenated jojoba alcohol, hydrogenated rapeseed alcohol, hydrogenated tallow alcohol, hydroxystearyl alcohol, jojoba alcohol, lauryl alcohol, myristyl alcohol, oleyl alcohol, olive alcohol, palm alcohol, palm kernel alcohol, stearyl alcohol, tallow alcohol, tridecyl alcohol and mixtures thereof. 5. The composition according to claim 1, wherein the one or more fatty alcohol is present at a concentration of 0.5 to 30%, by weight calculated to the total composition. 6. The composition according to claim 1, wherein the one or more oil is selected from synthetic oils and natural oils, and
wherein synthetic oils are selected from silicones, fatty acid fatty alcohol esters according to the general structure
R1C(O)R2
wherein R1 is a straight or branched, saturated or unsaturated alkyl with 11 to 21 C atoms and R2 is a straight or branched, saturated or unsaturated alkyl with 1 to 22 C atoms, and fatty alcohol ethers according to general structure
R3OR4
wherein R3 and R4 are same or different, straight or branched, saturated or unsaturated alkyl with 8 to 22 C atoms, and natural oils are selected from mineral oils and plant derived oils. 7. The composition according to claim 1, wherein the one or more oil is present at a concentration in the range of 1 to 20% by weight calculated to the total composition. 8. The composition according to claim 1, wherein the one or more fatty alcohol and the one or more oil are present at a total concentration in the range of 1 to 35% by weight calculated to the total composition. 9. The composition according to claim 1, wherein the one or more surfactant selected from anionic, cationic and nonionic surfactants,
wherein the anionic surfactants are selected from alkyl sulphates, alkyl ether sulfates, carboxylic acids or alkali salts thereof, fatty acid salts, alkyl/alkenyl succinates, and anionic amino acid surfactants, and the nonionic surfactants are selected from fatty alcohol polyglycol ethers according to general structure
R30(OCH2CH2)nOH
wherein R30 is same as above and n is a number between 1 and 50, fatty acid alkanolamides, amineoxides, and C8-C18-alkyl polyglucosides of the general structure
R6—O—(R7O)nO—Zx
wherein R6 is an alkyl group with 8 to 18 carbon atoms, R7 is an ethylene or propylene group, Z is a saccharide group with 5 to 6 carbon atoms, n is a number from 0 to 10 and x is a number between 1 and 5, and the cationic surfactants are mono alkyl quaternary ammonium salts of the following general structure
where R8 is a saturated or unsaturated, branched or non-branched alkyl chain with 8-22 C atoms, or
R12CONH(CH2)n
where R12 is saturated or unsaturated, branched or non-branched alkyl chain with 7-21 C atoms and n has typical value of 0-4, or
R13COO(CH2)n
where R13 is saturated or unsaturated, branched or non-branched alkyl chain with 7-21 C atoms and n has typical value of 0-4,
and R9, R10 and R11 are independent from each other H or lower alkyl chain with 1 to 4 carbon atoms, and X is an anion. 10. The composition according to claim 1, wherein the one or more dyestuff is free of transition metal ions, and free of anionic or cationic solubilizing groups or both anionic and cationic solubilizing groups. 11. (canceled) 12. A process for lightening hair color comprising:
(a) mixing a first composition according to claim 1 with second composition comprising one or more alkalizing agent, (b) applying a mixture obtained from step a) onto hair, (c) leaving the mixture obtained from step a) on hair for 5 to 45 min, and (d) rinsing the mixture obtained from step a) off from hair. 13. The process according to claim 12, wherein the second composition further comprises one or more oxidative dye precursor. 14. The process according to claim 12, wherein the first composition has a viscosity in the range of 500 to 50000 mPa·s, and the second composition has a viscosity in the range of 500 to 50000 mPa·s, measured at 20° C. with a Brookfield viscosimeter with a suitable spindle. 15. A kit comprising two or more compositions, wherein one of the two or more compositions is a composition according to claim 1. 16. The composition according to claim 4, wherein the one or more fatty alcohol is a mixture of cetyl and stearyl alcohols. 17. The composition of claim 5, wherein the one or more fatty alcohol is present at a concentration of 1 to 25% by weight calculated to the total composition. 18. The composition of claim 6, wherein the one or more oil is a nonvolatile silicone. 19. The composition of claim 7, wherein the one or more oil is present at a concentration in the range of 1 to 15% by weight calculated to the total composition. 20. The composition of claim 8, wherein the one or more fatty alcohol and the one or more oil are present at a total concentration in the range of 1 to 30% calculated to the total composition. 21. The composition of claim 9, wherein the one or more surfactant is present at a total concentration in the range of 0.05% to 15% by weight calculated to total composition. | Present invention relates to an aqueous oxidizing composition comprising at least one hydrophobic dye which provides improved mixing of such composition with an alkalizing composition prior to use and/or application such as onto hair.1. An aqueous emulsion composition comprising:
one or more oxidizing agent, one or both of: one or more fatty alcohol and one or more oil, one or more surfactant, and one or more dyestuff having a log P value at 25° C. of higher than or equal to 2, wherein the composition is free of solid peroxides or persulphates, or free of both solid peroxides and persulphates, and has a pH below 7. 2. The composition according to claim 1, wherein the one or more oxidizing agent is hydrogen peroxide and present at a concentration of 1 to 20% by weight calculated to the total composition. 3. The composition according to claim 1, wherein the one or more fatty alcohol is selected from compounds according to general structure
R30—OH
wherein R30 is a saturated or unsaturated, straight or branched alkyl chain having 12 to 30 C atoms which may also be substituted with one or more OH groups. 4. The composition according to claim 3, wherein the one or more fatty alcohol is selected from arachidyl alcohol, behenyl alcohol, brassica alcohol, C9-11 alcohols, C10-16 alcohols, C12-13 alcohols, C12-15 alcohols, C12-16 alcohols, C14-15 alcohols, C14-22 alcohols, C20-22 alcohols, caprylyl alcohol, cetearyl alcohol, cetyl alcohol, coconut alcohol, decyl alcohol, hydrogenated brassica alcohol, hydrogenated jojoba alcohol, hydrogenated rapeseed alcohol, hydrogenated tallow alcohol, hydroxystearyl alcohol, jojoba alcohol, lauryl alcohol, myristyl alcohol, oleyl alcohol, olive alcohol, palm alcohol, palm kernel alcohol, stearyl alcohol, tallow alcohol, tridecyl alcohol and mixtures thereof. 5. The composition according to claim 1, wherein the one or more fatty alcohol is present at a concentration of 0.5 to 30%, by weight calculated to the total composition. 6. The composition according to claim 1, wherein the one or more oil is selected from synthetic oils and natural oils, and
wherein synthetic oils are selected from silicones, fatty acid fatty alcohol esters according to the general structure
R1C(O)R2
wherein R1 is a straight or branched, saturated or unsaturated alkyl with 11 to 21 C atoms and R2 is a straight or branched, saturated or unsaturated alkyl with 1 to 22 C atoms, and fatty alcohol ethers according to general structure
R3OR4
wherein R3 and R4 are same or different, straight or branched, saturated or unsaturated alkyl with 8 to 22 C atoms, and natural oils are selected from mineral oils and plant derived oils. 7. The composition according to claim 1, wherein the one or more oil is present at a concentration in the range of 1 to 20% by weight calculated to the total composition. 8. The composition according to claim 1, wherein the one or more fatty alcohol and the one or more oil are present at a total concentration in the range of 1 to 35% by weight calculated to the total composition. 9. The composition according to claim 1, wherein the one or more surfactant selected from anionic, cationic and nonionic surfactants,
wherein the anionic surfactants are selected from alkyl sulphates, alkyl ether sulfates, carboxylic acids or alkali salts thereof, fatty acid salts, alkyl/alkenyl succinates, and anionic amino acid surfactants, and the nonionic surfactants are selected from fatty alcohol polyglycol ethers according to general structure
R30(OCH2CH2)nOH
wherein R30 is same as above and n is a number between 1 and 50, fatty acid alkanolamides, amineoxides, and C8-C18-alkyl polyglucosides of the general structure
R6—O—(R7O)nO—Zx
wherein R6 is an alkyl group with 8 to 18 carbon atoms, R7 is an ethylene or propylene group, Z is a saccharide group with 5 to 6 carbon atoms, n is a number from 0 to 10 and x is a number between 1 and 5, and the cationic surfactants are mono alkyl quaternary ammonium salts of the following general structure
where R8 is a saturated or unsaturated, branched or non-branched alkyl chain with 8-22 C atoms, or
R12CONH(CH2)n
where R12 is saturated or unsaturated, branched or non-branched alkyl chain with 7-21 C atoms and n has typical value of 0-4, or
R13COO(CH2)n
where R13 is saturated or unsaturated, branched or non-branched alkyl chain with 7-21 C atoms and n has typical value of 0-4,
and R9, R10 and R11 are independent from each other H or lower alkyl chain with 1 to 4 carbon atoms, and X is an anion. 10. The composition according to claim 1, wherein the one or more dyestuff is free of transition metal ions, and free of anionic or cationic solubilizing groups or both anionic and cationic solubilizing groups. 11. (canceled) 12. A process for lightening hair color comprising:
(a) mixing a first composition according to claim 1 with second composition comprising one or more alkalizing agent, (b) applying a mixture obtained from step a) onto hair, (c) leaving the mixture obtained from step a) on hair for 5 to 45 min, and (d) rinsing the mixture obtained from step a) off from hair. 13. The process according to claim 12, wherein the second composition further comprises one or more oxidative dye precursor. 14. The process according to claim 12, wherein the first composition has a viscosity in the range of 500 to 50000 mPa·s, and the second composition has a viscosity in the range of 500 to 50000 mPa·s, measured at 20° C. with a Brookfield viscosimeter with a suitable spindle. 15. A kit comprising two or more compositions, wherein one of the two or more compositions is a composition according to claim 1. 16. The composition according to claim 4, wherein the one or more fatty alcohol is a mixture of cetyl and stearyl alcohols. 17. The composition of claim 5, wherein the one or more fatty alcohol is present at a concentration of 1 to 25% by weight calculated to the total composition. 18. The composition of claim 6, wherein the one or more oil is a nonvolatile silicone. 19. The composition of claim 7, wherein the one or more oil is present at a concentration in the range of 1 to 15% by weight calculated to the total composition. 20. The composition of claim 8, wherein the one or more fatty alcohol and the one or more oil are present at a total concentration in the range of 1 to 30% calculated to the total composition. 21. The composition of claim 9, wherein the one or more surfactant is present at a total concentration in the range of 0.05% to 15% by weight calculated to total composition. | 1,600 |
639 | 15,403,940 | 1,642 | A method of treating a malignancy in a human subject by analyzing pseudo-projection images of cells obtained from a sputum specimen obtained from a subject employs a biological specimen classifier that identifies cells from the sputum specimen as normal or abnormal. If abnormal cells are detected, then the abnormal cells are further classified as dysplastic or cancerous. If the cells are classified as dysplastic, then an immunomodulating agent is administered to the subject over a predetermined time period designed to achieve a therapeutic dosage. | 1. A method of treating a malignancy in a human subject comprising:
analyzing 3D images of cells based on pseudo-projections obtained from a sputum specimen obtained from a subject; operating a biological specimen classifier to identify cells from the sputum specimen as normal or abnormal; further classifying identified abnormal cells as pre-cancerous or cancerous; further classifying pre-cancerous cells as pre-cancerous cells of glandular origin, mild-moderate dysplasia, moderate to severe dysplasia or severe dysplasia; when the cells are classified as cancerous, then performing a biopsy to verify suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion;
when the cells are classified as pre-cancerous of glandular origin, then conducting immunotherapy by administering an immunomodulating agent to a human subject over a predetermined time period to assist the immune system of the human subject in eradicating cancerous cells; when cells are classified as mild, moderate or severe dysplasia, then administering an immunomodulating agent to the subject over a predetermined time period; and when both pre-cancerous and cancer cells are discovered, then verifying the cancer status of any suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion, and
administering an immunomodulating agent to the subject over a predetermined time period. 2. The method of claim 1 wherein the immunomodulating agent comprises a drug selected from the group consisting of a chimeric immunoreceptor, a prostacyclin analog, iloprost, a chimeric antigen receptor (CAR) for T-cells, Vorinostat, HDAC inhibitors, cholecalciferol, calcitriol and combinations thereof. 3. The method of claim 1 further comprising:
repeating operation of the biological specimen classifier to classify cells as normal or abnormal;
when abnormal cells are detected, then further classifying the abnormal cells cancerous, precancerous cells of glandular origin, as mild to moderate dysplasia, moderate to severe dysplasia, severe dysplasia or cancerous; and
comparing the classification results of the repeated operation with classification results from the previous operation to determine the effectiveness of the immunomodulating agent. 4. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 12-24 months later if the cell data indicates a normal and metaplasia condition. 5. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 6 months later if the cell data indicates mild to moderate dysplasia. 6. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates severe dysplasia. 7. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates pre-cancerous conditions of glandular origin. 8. The method of claim 1 wherein the classification of the severe dysplasia is verified, followed by immunotherapy and/or chemotherapy. 9. The method of claim 1 further comprising scheduling follow-up diagnostic testing such as CT scans and bronchoscopy within 6 months later severe dysplasia indicates CIS and/or cancer. 10. The method of claim 1 further comprising administering a cancer chemoprevention pharmaceutical to the subject over a predetermined time period if the cell is dysplastic. 11. A method of immunotherapy for treating a malignancy in a human subject comprising:
analyzing 3D images of cells based on pseudo-projections obtained from a sputum specimen obtained from a subject; operating a biological specimen classifier to identify cells from the sputum specimen as normal or abnormal; further classifying identified abnormal cells as pre-cancerous or cancerous; further classifying pre-cancerous cells as pre-cancerous cells of glandular origin, mild-moderate dysplasia, moderate to severe dysplasia or severe dysplasia; when the cells are classified as cancerous, then performing a biopsy to verify suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion;
when the cells are classified as pre-cancerous of glandular origin, then administering an immunomodulation agent to the subject over a predetermined time period; when cells are classified as mild, moderate or severe dysplasia, then administering iloprost to the subject over a predetermined time period; and when both pre-cancerous and cancer cells are discovered, then verifying the cancer status of any suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion, and
administering immunotherapy and/or chemotherapy to the subject over a predetermined time period. 12. The method of claim 11 wherein the act of administering immunotherapy further includes administering an immunomodulation agent that comprises a drug selected from the group consisting of a chimeric immunoreceptor, a prostacyclin analog, a chimeric antigen receptor (CAR) for T-cells, Vorinostat, HDAC inhibitors, cholecalciferol, calcitriol and combinations thereof. 13. The method of claim 11 further comprising:
obtaining a second sputum specimen from the subject;
repeating operation of the biological specimen classifier to classify cells as normal or abnormal;
when abnormal cells are detected, then further classifying the abnormal cells cancerous, precancerous cells of glandular origin, as mild to moderate dysplasia, moderate to severe dysplasia, severe dysplasia or cancerous; and
comparing the classification results of the repeated operation with classification results from the previous operation to determine the effectiveness of the iloprost. 14. A method for automated detection and monitoring of dysplasia by analyzing 3D images of cells obtained from a sputum sample comprising:
providing 3D imaging data for a cell contained in the sputum sample; providing a sample adequacy determination for the sputum sample; if the sample adequacy determination meets predetermined criteria, then identifying the cell data as normal and metaplasia conditions, pre-cancerous cells of glandular origin, mild to moderate dysplasia or severe dysplasia or CIS and/or cancer. 15. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 12-24 months later if the cell data indicates a normal and metaplasia condition. 16. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 6 months later if the cell data indicates mild to moderate dysplasia. 17. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates severe dysplasia. 18. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates pre-cancerous conditions of glandular origin. 19. The method of claim 14 further comprising administering a cancer chemoprevention pharmaceutical to the subject over a predetermined time period if the cell is dysplastic. 20. The method of claim 19 wherein the immunomodulation agent comprises a drug selected from the group consisting of a chimeric immunoreceptor, a prostacyclin analog, iloprost, a chimeric antigen receptor (CAR) for T-cells, Vorinostat, HDAC inhibitors, cholecalciferol, calcitriol and combinations thereof. | A method of treating a malignancy in a human subject by analyzing pseudo-projection images of cells obtained from a sputum specimen obtained from a subject employs a biological specimen classifier that identifies cells from the sputum specimen as normal or abnormal. If abnormal cells are detected, then the abnormal cells are further classified as dysplastic or cancerous. If the cells are classified as dysplastic, then an immunomodulating agent is administered to the subject over a predetermined time period designed to achieve a therapeutic dosage.1. A method of treating a malignancy in a human subject comprising:
analyzing 3D images of cells based on pseudo-projections obtained from a sputum specimen obtained from a subject; operating a biological specimen classifier to identify cells from the sputum specimen as normal or abnormal; further classifying identified abnormal cells as pre-cancerous or cancerous; further classifying pre-cancerous cells as pre-cancerous cells of glandular origin, mild-moderate dysplasia, moderate to severe dysplasia or severe dysplasia; when the cells are classified as cancerous, then performing a biopsy to verify suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion;
when the cells are classified as pre-cancerous of glandular origin, then conducting immunotherapy by administering an immunomodulating agent to a human subject over a predetermined time period to assist the immune system of the human subject in eradicating cancerous cells; when cells are classified as mild, moderate or severe dysplasia, then administering an immunomodulating agent to the subject over a predetermined time period; and when both pre-cancerous and cancer cells are discovered, then verifying the cancer status of any suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion, and
administering an immunomodulating agent to the subject over a predetermined time period. 2. The method of claim 1 wherein the immunomodulating agent comprises a drug selected from the group consisting of a chimeric immunoreceptor, a prostacyclin analog, iloprost, a chimeric antigen receptor (CAR) for T-cells, Vorinostat, HDAC inhibitors, cholecalciferol, calcitriol and combinations thereof. 3. The method of claim 1 further comprising:
repeating operation of the biological specimen classifier to classify cells as normal or abnormal;
when abnormal cells are detected, then further classifying the abnormal cells cancerous, precancerous cells of glandular origin, as mild to moderate dysplasia, moderate to severe dysplasia, severe dysplasia or cancerous; and
comparing the classification results of the repeated operation with classification results from the previous operation to determine the effectiveness of the immunomodulating agent. 4. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 12-24 months later if the cell data indicates a normal and metaplasia condition. 5. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 6 months later if the cell data indicates mild to moderate dysplasia. 6. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates severe dysplasia. 7. The method of claim 1 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates pre-cancerous conditions of glandular origin. 8. The method of claim 1 wherein the classification of the severe dysplasia is verified, followed by immunotherapy and/or chemotherapy. 9. The method of claim 1 further comprising scheduling follow-up diagnostic testing such as CT scans and bronchoscopy within 6 months later severe dysplasia indicates CIS and/or cancer. 10. The method of claim 1 further comprising administering a cancer chemoprevention pharmaceutical to the subject over a predetermined time period if the cell is dysplastic. 11. A method of immunotherapy for treating a malignancy in a human subject comprising:
analyzing 3D images of cells based on pseudo-projections obtained from a sputum specimen obtained from a subject; operating a biological specimen classifier to identify cells from the sputum specimen as normal or abnormal; further classifying identified abnormal cells as pre-cancerous or cancerous; further classifying pre-cancerous cells as pre-cancerous cells of glandular origin, mild-moderate dysplasia, moderate to severe dysplasia or severe dysplasia; when the cells are classified as cancerous, then performing a biopsy to verify suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion;
when the cells are classified as pre-cancerous of glandular origin, then administering an immunomodulation agent to the subject over a predetermined time period; when cells are classified as mild, moderate or severe dysplasia, then administering iloprost to the subject over a predetermined time period; and when both pre-cancerous and cancer cells are discovered, then verifying the cancer status of any suspicious lesions;
when cancer is found, then administering surgical procedures to remove the cancer lesion, and
administering immunotherapy and/or chemotherapy to the subject over a predetermined time period. 12. The method of claim 11 wherein the act of administering immunotherapy further includes administering an immunomodulation agent that comprises a drug selected from the group consisting of a chimeric immunoreceptor, a prostacyclin analog, a chimeric antigen receptor (CAR) for T-cells, Vorinostat, HDAC inhibitors, cholecalciferol, calcitriol and combinations thereof. 13. The method of claim 11 further comprising:
obtaining a second sputum specimen from the subject;
repeating operation of the biological specimen classifier to classify cells as normal or abnormal;
when abnormal cells are detected, then further classifying the abnormal cells cancerous, precancerous cells of glandular origin, as mild to moderate dysplasia, moderate to severe dysplasia, severe dysplasia or cancerous; and
comparing the classification results of the repeated operation with classification results from the previous operation to determine the effectiveness of the iloprost. 14. A method for automated detection and monitoring of dysplasia by analyzing 3D images of cells obtained from a sputum sample comprising:
providing 3D imaging data for a cell contained in the sputum sample; providing a sample adequacy determination for the sputum sample; if the sample adequacy determination meets predetermined criteria, then identifying the cell data as normal and metaplasia conditions, pre-cancerous cells of glandular origin, mild to moderate dysplasia or severe dysplasia or CIS and/or cancer. 15. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 12-24 months later if the cell data indicates a normal and metaplasia condition. 16. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 6 months later if the cell data indicates mild to moderate dysplasia. 17. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates severe dysplasia. 18. The method of claim 14 further comprising scheduling a subsequent optical tomography sputum test within 3 months later if the cell data indicates pre-cancerous conditions of glandular origin. 19. The method of claim 14 further comprising administering a cancer chemoprevention pharmaceutical to the subject over a predetermined time period if the cell is dysplastic. 20. The method of claim 19 wherein the immunomodulation agent comprises a drug selected from the group consisting of a chimeric immunoreceptor, a prostacyclin analog, iloprost, a chimeric antigen receptor (CAR) for T-cells, Vorinostat, HDAC inhibitors, cholecalciferol, calcitriol and combinations thereof. | 1,600 |
640 | 13,210,331 | 1,637 | Disclosed is a method for amplifying and detecting polynucleotides which can provide sensitive, specific detection of multiple targets from a clinical specimen within a relatively short time. | 1. A method comprising
amplifying one or more target nucleic acids using high-concentration target-specific primers in a first amplification reaction, thereby producing at least one nucleic acid amplicon containing at least one common primer binding site; rescuing the at least one nucleic acid amplicon; and amplifying the at least one nucleic acid amplicon in a second amplification reaction using at least one common primer which binds to the at least one common primer binding site. 2. A method as in claim 1 wherein the high-concentration target-specific primers are nested primers. 3. A method as in claim 1 wherein the one or more target nucleic acids are chosen from among the group consisting of viral, bacterial, and fungal nucleic acids. 4. A method as in claim 1 wherein the one or more target nucleic acids are obtained from a human clinical sample. 5. A method as in claim 1 wherein the one or more target nucleic acids are obtained from a clinical sample from an animal. 6. A method as in claim 1 wherein the at least one common primer is chosen from among the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and combinations thereof. 7. A method as in claim 1 wherein the step of rescuing the at least one nucleic acid amplicon further comprises taking a small sampling from a completed amplification in a first reaction system to provide amplicons for a second amplification in a second reaction system. | Disclosed is a method for amplifying and detecting polynucleotides which can provide sensitive, specific detection of multiple targets from a clinical specimen within a relatively short time.1. A method comprising
amplifying one or more target nucleic acids using high-concentration target-specific primers in a first amplification reaction, thereby producing at least one nucleic acid amplicon containing at least one common primer binding site; rescuing the at least one nucleic acid amplicon; and amplifying the at least one nucleic acid amplicon in a second amplification reaction using at least one common primer which binds to the at least one common primer binding site. 2. A method as in claim 1 wherein the high-concentration target-specific primers are nested primers. 3. A method as in claim 1 wherein the one or more target nucleic acids are chosen from among the group consisting of viral, bacterial, and fungal nucleic acids. 4. A method as in claim 1 wherein the one or more target nucleic acids are obtained from a human clinical sample. 5. A method as in claim 1 wherein the one or more target nucleic acids are obtained from a clinical sample from an animal. 6. A method as in claim 1 wherein the at least one common primer is chosen from among the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and combinations thereof. 7. A method as in claim 1 wherein the step of rescuing the at least one nucleic acid amplicon further comprises taking a small sampling from a completed amplification in a first reaction system to provide amplicons for a second amplification in a second reaction system. | 1,600 |
641 | 15,381,638 | 1,651 | The present disclosure provides robust, high-throughput, and clinically applicable methods for simultaneously separating and quantifying the biologically active forms of Vitamin B1 (TPP) and Vitamin B6 (PLP) from human whole blood. | 1. A method of quantifying both native Thiamine Pyrophosphate (TPP) and native Pyridoxal 5-Phosphate (PLP) from a single sample of human whole blood using mass spectrometry, comprising:
eluting TPP and PLP at time of less than 20 seconds apart from one another from a liquid chromatography column; generating a mass spectrometry signal during elution of the TPP and PLP; and quantifying the TPP and PLP using one or more calibration standards. 2. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises augmenting human whole blood with TPP and/or PLP, and optionally diluting the human whole blood with phosphate buffered saline to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 3. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises augmenting TPP and PLP depleted whole blood with TPP and PLP to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 4. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises the use of surrogate matrix calibrators prepared from phosphate buffered saline solution optionally containing human serum albumin or bovine serum albumin and optionally augmented with TPP and PLP to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 5. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises the use of surrogate analyte calibrators prepared in human whole blood by the addition of a different stable labeled isotope to that of the internal standard for TPP and PLP to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 6. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises the use placing one or more calibration standards in the same sample together with the TPP and PLP eluted from the column, to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 7. The method of claim 1, wherein the one or more calibrators are provided in the form of a kit. 8. A method of separating both native Thiamine Pyrophosphate (TPP) and native Pyridoxal 5-Phosphate (PLP) from whole blood comprising eluting TPP and PLP at time of less than 20 seconds apart from one another from a liquid chromatography column. 9. The method of claim 1, comprising eluting TPP and PLP at time of less than 17 seconds apart from one another from the liquid chromatography column 10. The method of claim 1, comprising eluting TPP and PLP at time of less than 15 seconds apart from one another from the liquid chromatography column. 11. The method of claim 1, comprising eluting TPP and PLP at time of less than 13 seconds apart from one another from the liquid chromatography column 12. The method of claim 1, wherein both the TPP and PLP have a retention time on the liquid chromatography column of 35 seconds or greater. 13. The method of claim 1, wherein both the TPP and PLP have a retention time on the liquid chromatography column of 40 seconds or greater. 14. The method of claim 1, wherein both the TPP and PLP have a retention time on the liquid chromatography column of 45 seconds or greater. 15. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 3.5 minutes. 16. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 3.0 minutes. 17. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 2.5 minutes. 18. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 2.0 minutes. 19. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column. 20. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column having a 3.5 μm particle size. 21. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column having a 3.5 μm particle size, a greater than 15% carbon load, and a pore size of greater than or equal to 100 Å. 22. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column having a 3.5 μm particle size, a 15% to 20% carbon load, and a pore size of 100 Å. 23. The method of claim 1, wherein the TPP and PLP are eluted from the liquid chromatography column using a mobile phase comprising water (H2O), methanol (MeOH) and formic acid. 24. The method of claim 1, wherein the TPP and PLP are eluted from the liquid chromatography column using a mobile phase comprising H2O together with 0.1% formic acid (v/v) and MeOH together with 0.1% formic acid (v/v). 25. The method of claim 1, wherein the TPP and PLP are eluted from the liquid chromatography column using H2O together with 0.1% formic acid (v/v) and MeOH together with 0.1% formic acid (v/v), subject to a gradient comprising the following conditions:
% Total of H2O
% Total of MeOH
Time
Flow Rate
together with 0.1%
together with 0.1%
(mm)
(mL/min)
formic acid (v/v)
formic acid (v/v)
Curve
0
0.60
97
3
Initial
0.60
0.60
70
30
6
1.20
0.80
3
97
11
1.70
0.80
97
3
11
1.90
0.60
97
3
11. 26. The method of claim 1, further comprising the step of protein precipitating the TPP and PLP from whole blood prior to elution on the liquid chromatography column 27. The method of claim 1, wherein prior to eluting TPP and PLP, the TPP and PLP are mixed with trichloroacetic acid. | The present disclosure provides robust, high-throughput, and clinically applicable methods for simultaneously separating and quantifying the biologically active forms of Vitamin B1 (TPP) and Vitamin B6 (PLP) from human whole blood.1. A method of quantifying both native Thiamine Pyrophosphate (TPP) and native Pyridoxal 5-Phosphate (PLP) from a single sample of human whole blood using mass spectrometry, comprising:
eluting TPP and PLP at time of less than 20 seconds apart from one another from a liquid chromatography column; generating a mass spectrometry signal during elution of the TPP and PLP; and quantifying the TPP and PLP using one or more calibration standards. 2. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises augmenting human whole blood with TPP and/or PLP, and optionally diluting the human whole blood with phosphate buffered saline to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 3. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises augmenting TPP and PLP depleted whole blood with TPP and PLP to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 4. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises the use of surrogate matrix calibrators prepared from phosphate buffered saline solution optionally containing human serum albumin or bovine serum albumin and optionally augmented with TPP and PLP to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 5. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises the use of surrogate analyte calibrators prepared in human whole blood by the addition of a different stable labeled isotope to that of the internal standard for TPP and PLP to generate the one or more calibration standards over a measuring range to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 6. The method of claim 1, wherein quantifying the TPP and PLP using one or more calibration standards comprises the use placing one or more calibration standards in the same sample together with the TPP and PLP eluted from the column, to which the amount of the TPP and PLP eluted from the column can be determined by mathematical expression. 7. The method of claim 1, wherein the one or more calibrators are provided in the form of a kit. 8. A method of separating both native Thiamine Pyrophosphate (TPP) and native Pyridoxal 5-Phosphate (PLP) from whole blood comprising eluting TPP and PLP at time of less than 20 seconds apart from one another from a liquid chromatography column. 9. The method of claim 1, comprising eluting TPP and PLP at time of less than 17 seconds apart from one another from the liquid chromatography column 10. The method of claim 1, comprising eluting TPP and PLP at time of less than 15 seconds apart from one another from the liquid chromatography column. 11. The method of claim 1, comprising eluting TPP and PLP at time of less than 13 seconds apart from one another from the liquid chromatography column 12. The method of claim 1, wherein both the TPP and PLP have a retention time on the liquid chromatography column of 35 seconds or greater. 13. The method of claim 1, wherein both the TPP and PLP have a retention time on the liquid chromatography column of 40 seconds or greater. 14. The method of claim 1, wherein both the TPP and PLP have a retention time on the liquid chromatography column of 45 seconds or greater. 15. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 3.5 minutes. 16. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 3.0 minutes. 17. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 2.5 minutes. 18. The method of claim 1, wherein the total elution time of TPP and PLP does not exceed 2.0 minutes. 19. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column. 20. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column having a 3.5 μm particle size. 21. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column having a 3.5 μm particle size, a greater than 15% carbon load, and a pore size of greater than or equal to 100 Å. 22. The method of claim 1, wherein the liquid chromatography column is a reverse phase C18 HPLC column having a 3.5 μm particle size, a 15% to 20% carbon load, and a pore size of 100 Å. 23. The method of claim 1, wherein the TPP and PLP are eluted from the liquid chromatography column using a mobile phase comprising water (H2O), methanol (MeOH) and formic acid. 24. The method of claim 1, wherein the TPP and PLP are eluted from the liquid chromatography column using a mobile phase comprising H2O together with 0.1% formic acid (v/v) and MeOH together with 0.1% formic acid (v/v). 25. The method of claim 1, wherein the TPP and PLP are eluted from the liquid chromatography column using H2O together with 0.1% formic acid (v/v) and MeOH together with 0.1% formic acid (v/v), subject to a gradient comprising the following conditions:
% Total of H2O
% Total of MeOH
Time
Flow Rate
together with 0.1%
together with 0.1%
(mm)
(mL/min)
formic acid (v/v)
formic acid (v/v)
Curve
0
0.60
97
3
Initial
0.60
0.60
70
30
6
1.20
0.80
3
97
11
1.70
0.80
97
3
11
1.90
0.60
97
3
11. 26. The method of claim 1, further comprising the step of protein precipitating the TPP and PLP from whole blood prior to elution on the liquid chromatography column 27. The method of claim 1, wherein prior to eluting TPP and PLP, the TPP and PLP are mixed with trichloroacetic acid. | 1,600 |
642 | 14,769,563 | 1,653 | The present invention relates to a method for isolating stem cells comprising preparing a cell suspension from uterine cervix tissue, to the stem cells isolated by said method, and to the conditioned medium obtained from the culture of said stem cells. The invention also encompasses the use of said stem cells or conditioned medium for treating or preventing cancer, precancerous lesions, inflammatory diseases, autoimmune diseases, chronic pathologies or infectious diseases, diseases associated to tissue loss, or for use in diagnostic, prognostic or treatment of fertility disorders, as well as for cosmetic treatment. | 1. A method for isolating stem cells comprising:
(a) Preparing a cell suspension from uterine cervix tissue, (b) Recovering the cells from said cell suspension, (c) Incubating said cells in a suitable cell culture medium and under conditions which allow cells to proliferate, and (d) Selecting the stem cells. 2. Method according to claim 1, wherein the step (a) comprises enzymatically disaggregating the cervical mucus. 3. Method according to claim 1 or 2, wherein the isolated stem cells:
(a) express the cell markers CD29, CD44, CD73, CD90, CD105, vimentin, cytokeratin (CKAE1AE3), Klf4, Oct4 and Sox-2, and
(b) do not express at least one cell marker selected from the group consisting of desmin, actin HHF35, β-catenin, p63, E-cadherin, CD117, CD133, HLA-DR, TRA1-81, CD45, CD34 and CD31. 4. Method according to claim 3, wherein the stem cells further show
(a) a proliferating rate from 0.4 to 2.1 doublings per 24 hours in growth medium, (b) a fibroblast-like morphology, (c) a stable karyotype for at least 10 cell passages, (d) capacity to grow in monolayer and to adhere to a substrate, (e) capacity to be differentiated into endodermal, ectodermal or mesodermal cell lineage, (f) a non tumorigenic capacity and/or (g) capacity to form spheres. 5. Method according to any one of claims 1 to 4, wherein the uterine cervix tissue is a mammalian uterine cervix tissue, preferably, a human uterine cervix tissue. 6. Method according to any one of claims 1 to 5, wherein the uterine cervix tissue is a non-cancerous tissue. 7. Use of isolated uterine cervix tissue according to any one of claims 1 to 6, for obtaining uterine cervix stem cells. 8. Use according to claim 1 wherein the uterine cervix tissue is non-cancerous. 9. An isolated uterine cervix stem cell, wherein said cell:
(a) expresses the cell markers CD29, CD44, CD73, CD90, CD105, vimentin, cytokeratin (CKAE1AE3), Klf4, Oct4 and Sox-2, and (b) does not express at least one cell marker selected from the group consisting of desmin, actin HHF35, β-catenin, p63, E-cadherin, CD117, CD133, HLA-DR, TRA1-81, CD45, CD34 and CD31. 10. Isolated stem cell according to claim 9, wherein the cell further shows
(a) a proliferating rate from 0.4 to 2.1 doublings per 24 hours in growth medium, (b) a fibroblast-like morphology, (c) a stable karyotype for at least 10, preferably, 20 cell passages, (d) capacity to grow in monolayer and to adhere to a substrate, (e) capacity to be differentiated into an adipogenic, osteogenic, neural or myocytic cell linage, (f) a non tumorigenic capacity and/or (g) capacity to form spheres. 11. Isolated stem cell according to claim 9 or 10, wherein the cell is from a mammal, preferably, from a human, more preferably, from a human in a non-menstrual phase. 12. A cell population comprising an isolated stem cell according to any one of claims 9 to 11. 13. A conditioned medium obtained by a method comprising:
(a) Incubating an isolated stem cell according to any one of claims 9 to 11 or a cell population according to claim 12, and (b) Removing the cells from the culture medium. 14. A pharmaceutical composition comprising an isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, or a conditioned medium according to claim 13, and an acceptable pharmaceutically carrier and/or an adjuvant. 15. An isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, a conditioned medium according to claim 13 or a pharmaceutical composition according to claim 14 for use as a medicament. 16. An isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, a conditioned medium according to claim 13 or a pharmaceutical composition according to claim 14, for use in the treatment or prevention of cancer, precancerous lesions, inflammatory diseases, autoimmune diseases, chronic pathologies or infectious diseases, diseases associated to tissue loss, or for use in diagnostic, prognostic or treatment of fertility disorders or for use in a cosmetic treatment. 17. An isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, a conditioned medium according to claim 13 or a pharmaceutical composition according to claim 14, for inhibiting or decreasing the proliferation and/or metastasis of tumor cells, the monocytic differentiation, the peripheral blood mononuclear cells proliferation or the pathogenic microorganism growth and/or replication, or for enhancing or inducing the apoptosis of tumor cells, or for enhancing tissue regeneration and selection of germ cells. 18. A kit comprising an isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, or a conditioned medium according to claim 13, or the pharmaceutical composition according to claim 14. 19. Use of the kit according to claim 18 for the treatment or prevention of cancer, precancerous lesions, inflammatory diseases, autoimmune diseases, chronic pathologies or infectious diseases, diseases associated to tissue loss, or for use in diagnostic, prognostic or treatment of fertility disorders or for use in a cosmetic treatment. 20. Use of the kit according to claim 18 for inhibiting or decreasing the proliferation and/or metastasis of tumor cells, the monocytic differentiation or peripheral blood mononuclear cells proliferation, or for enhancing or inducing the apoptosis of tumor cells, or for enhancing tissue regeneration, or for the selection of germ cells. | The present invention relates to a method for isolating stem cells comprising preparing a cell suspension from uterine cervix tissue, to the stem cells isolated by said method, and to the conditioned medium obtained from the culture of said stem cells. The invention also encompasses the use of said stem cells or conditioned medium for treating or preventing cancer, precancerous lesions, inflammatory diseases, autoimmune diseases, chronic pathologies or infectious diseases, diseases associated to tissue loss, or for use in diagnostic, prognostic or treatment of fertility disorders, as well as for cosmetic treatment.1. A method for isolating stem cells comprising:
(a) Preparing a cell suspension from uterine cervix tissue, (b) Recovering the cells from said cell suspension, (c) Incubating said cells in a suitable cell culture medium and under conditions which allow cells to proliferate, and (d) Selecting the stem cells. 2. Method according to claim 1, wherein the step (a) comprises enzymatically disaggregating the cervical mucus. 3. Method according to claim 1 or 2, wherein the isolated stem cells:
(a) express the cell markers CD29, CD44, CD73, CD90, CD105, vimentin, cytokeratin (CKAE1AE3), Klf4, Oct4 and Sox-2, and
(b) do not express at least one cell marker selected from the group consisting of desmin, actin HHF35, β-catenin, p63, E-cadherin, CD117, CD133, HLA-DR, TRA1-81, CD45, CD34 and CD31. 4. Method according to claim 3, wherein the stem cells further show
(a) a proliferating rate from 0.4 to 2.1 doublings per 24 hours in growth medium, (b) a fibroblast-like morphology, (c) a stable karyotype for at least 10 cell passages, (d) capacity to grow in monolayer and to adhere to a substrate, (e) capacity to be differentiated into endodermal, ectodermal or mesodermal cell lineage, (f) a non tumorigenic capacity and/or (g) capacity to form spheres. 5. Method according to any one of claims 1 to 4, wherein the uterine cervix tissue is a mammalian uterine cervix tissue, preferably, a human uterine cervix tissue. 6. Method according to any one of claims 1 to 5, wherein the uterine cervix tissue is a non-cancerous tissue. 7. Use of isolated uterine cervix tissue according to any one of claims 1 to 6, for obtaining uterine cervix stem cells. 8. Use according to claim 1 wherein the uterine cervix tissue is non-cancerous. 9. An isolated uterine cervix stem cell, wherein said cell:
(a) expresses the cell markers CD29, CD44, CD73, CD90, CD105, vimentin, cytokeratin (CKAE1AE3), Klf4, Oct4 and Sox-2, and (b) does not express at least one cell marker selected from the group consisting of desmin, actin HHF35, β-catenin, p63, E-cadherin, CD117, CD133, HLA-DR, TRA1-81, CD45, CD34 and CD31. 10. Isolated stem cell according to claim 9, wherein the cell further shows
(a) a proliferating rate from 0.4 to 2.1 doublings per 24 hours in growth medium, (b) a fibroblast-like morphology, (c) a stable karyotype for at least 10, preferably, 20 cell passages, (d) capacity to grow in monolayer and to adhere to a substrate, (e) capacity to be differentiated into an adipogenic, osteogenic, neural or myocytic cell linage, (f) a non tumorigenic capacity and/or (g) capacity to form spheres. 11. Isolated stem cell according to claim 9 or 10, wherein the cell is from a mammal, preferably, from a human, more preferably, from a human in a non-menstrual phase. 12. A cell population comprising an isolated stem cell according to any one of claims 9 to 11. 13. A conditioned medium obtained by a method comprising:
(a) Incubating an isolated stem cell according to any one of claims 9 to 11 or a cell population according to claim 12, and (b) Removing the cells from the culture medium. 14. A pharmaceutical composition comprising an isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, or a conditioned medium according to claim 13, and an acceptable pharmaceutically carrier and/or an adjuvant. 15. An isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, a conditioned medium according to claim 13 or a pharmaceutical composition according to claim 14 for use as a medicament. 16. An isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, a conditioned medium according to claim 13 or a pharmaceutical composition according to claim 14, for use in the treatment or prevention of cancer, precancerous lesions, inflammatory diseases, autoimmune diseases, chronic pathologies or infectious diseases, diseases associated to tissue loss, or for use in diagnostic, prognostic or treatment of fertility disorders or for use in a cosmetic treatment. 17. An isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, a conditioned medium according to claim 13 or a pharmaceutical composition according to claim 14, for inhibiting or decreasing the proliferation and/or metastasis of tumor cells, the monocytic differentiation, the peripheral blood mononuclear cells proliferation or the pathogenic microorganism growth and/or replication, or for enhancing or inducing the apoptosis of tumor cells, or for enhancing tissue regeneration and selection of germ cells. 18. A kit comprising an isolated stem cell according to any one of claims 9 to 11, a cell population according to claim 12, or a conditioned medium according to claim 13, or the pharmaceutical composition according to claim 14. 19. Use of the kit according to claim 18 for the treatment or prevention of cancer, precancerous lesions, inflammatory diseases, autoimmune diseases, chronic pathologies or infectious diseases, diseases associated to tissue loss, or for use in diagnostic, prognostic or treatment of fertility disorders or for use in a cosmetic treatment. 20. Use of the kit according to claim 18 for inhibiting or decreasing the proliferation and/or metastasis of tumor cells, the monocytic differentiation or peripheral blood mononuclear cells proliferation, or for enhancing or inducing the apoptosis of tumor cells, or for enhancing tissue regeneration, or for the selection of germ cells. | 1,600 |
643 | 14,537,463 | 1,644 | A method of comparative ligand mapping to identify peptide ligands presented by MHC positive cells that distinguish an infected/transfected cell from an uninfected/non-transfected cell is disclosed. | 1. A method for identifying at least one individual, endogenously loaded peptide ligand for an individual class I MHC molecule that distinguishes a transfected cell from a non-transfected cell, comprising the steps of:
culturing a non-transfected cell line and a transfected cell line, each containing a construct that encodes an individual soluble class I MHC molecule, and the non-transfected cell line being able to naturally process proteins into peptide ligands capable of being loaded into antigen binding grooves of class I MHC molecules, wherein the cell lines are cultured under conditions which allow for expression of the individual soluble class I MHC molecules from the construct, such conditions also allowing for endogenous loading of a peptide ligand in the antigen binding groove of each individual soluble class I MHC molecule prior to secretion of the individual soluble class I MHC molecules from the cell, and wherein the transfected cell line is produced by transfecting a non-transfected cell line with at least one of a gene from a microorganism and a tumor gene; isolating the secreted individual soluble class I MHC molecules having the endogenously loaded peptide ligands bound thereto from the non-transfected cell line and the transfected cell line; separating the endogenously loaded peptide ligands from the individual soluble class I MHC molecules from the non-transfected cell line and separating the endogenously loaded peptide ligands from the individual soluble class I MHC molecules from the transfected cell line; isolating the endogenously loaded peptide ligands from the non-transfected cell line and the endogenously loaded peptide ligands from the transfected cell line; comparing the endogenously loaded peptide ligands isolated from the transfected cell line to the endogenously loaded peptide ligands isolated from the non-transfected cell line; identifying at least one individual, endogenously loaded peptide ligand wherein:
(a) the peptide ligand is presented by the individual soluble class I MHC molecule on the transfected cell line that is not presented by the individual soluble class I MHC molecule on the non-transfected cell line; or
(b) the peptide ligand is presented by the individual soluble class I MHC molecule on the non-transfected cell line that is not presented by the individual soluble class I MHC molecule on the transfected cell line; and
identifying a source protein from which the at least one individual, endogenously loaded peptide ligand presented by the individual soluble class I MHC molecule on the transfected cell line and not presented by the individual soluble class I MHC molecule on the non-transfected cell line is obtained. 2. The method of claim 1, wherein the identified peptide ligand is obtained from a protein encoded by at least one of the gene from a microorganism and a tumor gene with which the cell line was transfected to form the transfected cell line. 3. The method of claim 1, wherein the identified peptide ligand is obtained from a protein encoded by the non-transfected cell line. 4. The method of claim 1, wherein the construct further encodes a tag which is attached to the individual soluble class I MHC molecule and aids in isolating the individual soluble class I MHC molecule. 5. The method of claim 1, wherein the non-transfected cell line is class I MHC negative. 6. The method of claim 1, wherein the non-transfected cell line expresses endogenous class I MHC molecules. 7. The method of claim 1, wherein the transfected cell is further defined as a tumorigenic cell, and the non-transfected cell is further defined as a non-tumorigenic cell. 8. The method of claim 1, further comprising the step of transfecting a portion of the non-transfected cell line with at least one gene from a microorganism and/or a tumor gene, thereby providing the transfected cell line. 9. The method of claim 1, wherein the cell lines containing the construct that encodes the individual soluble class I MHC molecule are produced by a method comprising the steps of:
obtaining genomic DNA or cDNA encoding at least one class I MHC molecule; identifying an allele encoding an individual class I MHC molecule in the genomic DNA or cDNA; PCR amplifying the allele encoding the individual class I MHC molecule in a locus specific manner such that a PCR product produced therefrom encodes a truncated, soluble form of the individual class I MHC molecule; cloning the PCR product into an expression vector, thereby forming a construct that encodes the individual soluble class I MHC molecule; and
transfecting the construct into an uninfected cell line. 10. A method for identifying at least one peptide ligand for a class I MHC molecule, wherein the at least one peptide ligand distinguishes an infected cell from an uninfected cell, the method comprising the steps of:
separately culturing an uninfected cell line and an infected cell line, each containing a construct that encodes a soluble class I MHC molecule, under conditions that allow for expression and secretion of recombinant soluble class I MHC molecules that have endogenously produced and loaded peptide ligands bound thereto; isolating the peptide ligands from the soluble class I MHC molecules secreted from the infected and uninfected cell lines; comparing the peptide ligands isolated from the infected cell line with the peptide ligands isolated from the uninfected cell line to identify at least one peptide ligand that distinguishes infected cells from uninfected cells; and identifying a source protein from which the at least one peptide ligand is obtained. 11. The method of claim 10, wherein the infected cell line is infected with a microorganism, and wherein the at least one peptide ligand identified as distinguishing infected cells from uninfected cells is presented by the class I MHC molecules on the infected cell line but not on the uninfected cell line. 12. The method of claim 11, wherein the at least one peptide ligand is obtained from a protein encoded by the microorganism. 13. The method of claim 10, wherein the uninfected and infected cell lines do not express endogenous class I MHC molecules. 14. The method of claim 10, wherein the uninfected cell line expresses endogenous class I MHC molecules. 15. The method of claim 10, wherein the at least one peptide ligand is obtained from a protein encoded by the uninfected cell line. 16. The method of claim 10, wherein the at least one peptide ligand is presented by the class I MHC molecules on the uninfected cell line but not on the infected cell line. 17. The method of claim 10, further comprising the step of infecting a portion of the uninfected cell line with a microorganism, thereby providing the infected cell line. 18. The method of claim 10, wherein the construct further encodes a tag which is attached to the individual soluble class I MHC molecule and aids in isolating the individual soluble class I MHC molecule. 19. The method of claim 10, wherein the cell lines containing the construct that encodes the individual soluble class I MHC molecule are produced by a method comprising the steps of:
obtaining genomic DNA or cDNA encoding at least one class I MHC molecule; identifying an allele encoding an individual class I MHC molecule in the genomic DNA or cDNA; PCR amplifying the allele encoding the individual class I MHC molecule in a locus specific manner such that a PCR product produced therefrom encodes a truncated, soluble form of the individual class I MHC molecule; cloning the PCR product into an expression vector, thereby forming a construct that encodes the individual soluble class I MHC molecule; and
transfecting the construct into an uninfected cell line. | A method of comparative ligand mapping to identify peptide ligands presented by MHC positive cells that distinguish an infected/transfected cell from an uninfected/non-transfected cell is disclosed.1. A method for identifying at least one individual, endogenously loaded peptide ligand for an individual class I MHC molecule that distinguishes a transfected cell from a non-transfected cell, comprising the steps of:
culturing a non-transfected cell line and a transfected cell line, each containing a construct that encodes an individual soluble class I MHC molecule, and the non-transfected cell line being able to naturally process proteins into peptide ligands capable of being loaded into antigen binding grooves of class I MHC molecules, wherein the cell lines are cultured under conditions which allow for expression of the individual soluble class I MHC molecules from the construct, such conditions also allowing for endogenous loading of a peptide ligand in the antigen binding groove of each individual soluble class I MHC molecule prior to secretion of the individual soluble class I MHC molecules from the cell, and wherein the transfected cell line is produced by transfecting a non-transfected cell line with at least one of a gene from a microorganism and a tumor gene; isolating the secreted individual soluble class I MHC molecules having the endogenously loaded peptide ligands bound thereto from the non-transfected cell line and the transfected cell line; separating the endogenously loaded peptide ligands from the individual soluble class I MHC molecules from the non-transfected cell line and separating the endogenously loaded peptide ligands from the individual soluble class I MHC molecules from the transfected cell line; isolating the endogenously loaded peptide ligands from the non-transfected cell line and the endogenously loaded peptide ligands from the transfected cell line; comparing the endogenously loaded peptide ligands isolated from the transfected cell line to the endogenously loaded peptide ligands isolated from the non-transfected cell line; identifying at least one individual, endogenously loaded peptide ligand wherein:
(a) the peptide ligand is presented by the individual soluble class I MHC molecule on the transfected cell line that is not presented by the individual soluble class I MHC molecule on the non-transfected cell line; or
(b) the peptide ligand is presented by the individual soluble class I MHC molecule on the non-transfected cell line that is not presented by the individual soluble class I MHC molecule on the transfected cell line; and
identifying a source protein from which the at least one individual, endogenously loaded peptide ligand presented by the individual soluble class I MHC molecule on the transfected cell line and not presented by the individual soluble class I MHC molecule on the non-transfected cell line is obtained. 2. The method of claim 1, wherein the identified peptide ligand is obtained from a protein encoded by at least one of the gene from a microorganism and a tumor gene with which the cell line was transfected to form the transfected cell line. 3. The method of claim 1, wherein the identified peptide ligand is obtained from a protein encoded by the non-transfected cell line. 4. The method of claim 1, wherein the construct further encodes a tag which is attached to the individual soluble class I MHC molecule and aids in isolating the individual soluble class I MHC molecule. 5. The method of claim 1, wherein the non-transfected cell line is class I MHC negative. 6. The method of claim 1, wherein the non-transfected cell line expresses endogenous class I MHC molecules. 7. The method of claim 1, wherein the transfected cell is further defined as a tumorigenic cell, and the non-transfected cell is further defined as a non-tumorigenic cell. 8. The method of claim 1, further comprising the step of transfecting a portion of the non-transfected cell line with at least one gene from a microorganism and/or a tumor gene, thereby providing the transfected cell line. 9. The method of claim 1, wherein the cell lines containing the construct that encodes the individual soluble class I MHC molecule are produced by a method comprising the steps of:
obtaining genomic DNA or cDNA encoding at least one class I MHC molecule; identifying an allele encoding an individual class I MHC molecule in the genomic DNA or cDNA; PCR amplifying the allele encoding the individual class I MHC molecule in a locus specific manner such that a PCR product produced therefrom encodes a truncated, soluble form of the individual class I MHC molecule; cloning the PCR product into an expression vector, thereby forming a construct that encodes the individual soluble class I MHC molecule; and
transfecting the construct into an uninfected cell line. 10. A method for identifying at least one peptide ligand for a class I MHC molecule, wherein the at least one peptide ligand distinguishes an infected cell from an uninfected cell, the method comprising the steps of:
separately culturing an uninfected cell line and an infected cell line, each containing a construct that encodes a soluble class I MHC molecule, under conditions that allow for expression and secretion of recombinant soluble class I MHC molecules that have endogenously produced and loaded peptide ligands bound thereto; isolating the peptide ligands from the soluble class I MHC molecules secreted from the infected and uninfected cell lines; comparing the peptide ligands isolated from the infected cell line with the peptide ligands isolated from the uninfected cell line to identify at least one peptide ligand that distinguishes infected cells from uninfected cells; and identifying a source protein from which the at least one peptide ligand is obtained. 11. The method of claim 10, wherein the infected cell line is infected with a microorganism, and wherein the at least one peptide ligand identified as distinguishing infected cells from uninfected cells is presented by the class I MHC molecules on the infected cell line but not on the uninfected cell line. 12. The method of claim 11, wherein the at least one peptide ligand is obtained from a protein encoded by the microorganism. 13. The method of claim 10, wherein the uninfected and infected cell lines do not express endogenous class I MHC molecules. 14. The method of claim 10, wherein the uninfected cell line expresses endogenous class I MHC molecules. 15. The method of claim 10, wherein the at least one peptide ligand is obtained from a protein encoded by the uninfected cell line. 16. The method of claim 10, wherein the at least one peptide ligand is presented by the class I MHC molecules on the uninfected cell line but not on the infected cell line. 17. The method of claim 10, further comprising the step of infecting a portion of the uninfected cell line with a microorganism, thereby providing the infected cell line. 18. The method of claim 10, wherein the construct further encodes a tag which is attached to the individual soluble class I MHC molecule and aids in isolating the individual soluble class I MHC molecule. 19. The method of claim 10, wherein the cell lines containing the construct that encodes the individual soluble class I MHC molecule are produced by a method comprising the steps of:
obtaining genomic DNA or cDNA encoding at least one class I MHC molecule; identifying an allele encoding an individual class I MHC molecule in the genomic DNA or cDNA; PCR amplifying the allele encoding the individual class I MHC molecule in a locus specific manner such that a PCR product produced therefrom encodes a truncated, soluble form of the individual class I MHC molecule; cloning the PCR product into an expression vector, thereby forming a construct that encodes the individual soluble class I MHC molecule; and
transfecting the construct into an uninfected cell line. | 1,600 |
644 | 15,295,170 | 1,634 | The present disclosure describes the thermodynamic design and concentrations necessary to design probe compositions with desired optimal specificity that enable enrichment, detection, quantitation, purification, imaging, and amplification of rare-allele-bearing species of nucleic acids (prevalence <1%) in a large stoichiometric excess of a dominant-allele-bearing species (wildtype). Being an enzyme-free and homogeneous nucleic acid enrichment composition, this technology is broadly compatible with nearly all nucleic acid-based biotechnology, including plate reader and fluorimeter readout of nucleic acids, microarrays, PCR and other enzymatic amplification reactions, fluorescence barcoding, nanoparticle-based purification and quantitation, and in situ hybridization imaging technologies. | 1. A nucleic acid detection composition comprising:
a target nucleic acid probe comprising a first target probe oligonucleotide and a second target probe oligonucleotide, wherein the first target probe oligonucleotide comprises a target probe complement region that is complementary to a target nucleic acid sequence, wherein the second target probe oligonucleotide comprises a target probe protector region that is complementary to a first target probe complement subsequence of the target probe complement region thereby providing a target double-stranded probe portion and a target single-stranded probe portion, wherein the target double-stranded probe portion comprises the first target probe complement subsequence and the target probe protector region, and the target single-stranded probe portion comprises a second target probe complement subsequence of the target probe complement region; a target auxiliary oligonucleotide, wherein the target auxiliary oligonucleotide is the second target probe oligonucleotide separate from the target nucleic acid probe; a variant nucleic acid probe comprising a first variant probe oligonucleotide and a second variant probe oligonucleotide, wherein the first variant probe oligonucleotide comprises a variant probe complement region that is complementary to a variant nucleic acid sequence, wherein the second variant probe oligonucleotide comprises a variant probe protector region that is complementary to a first variant probe complement subsequence of the variant probe complement region thereby providing a variant double-stranded probe portion and a variant single-stranded probe portion, wherein the variant double-stranded probe portion comprises the first variant probe complement subsequence and the variant probe protector region, and the variant single-stranded probe portion comprises a second variant probe complement subsequence of the variant probe complement region, wherein the target probe complement region and the variant probe complement region share at least two conserved sequences separated by a non-conserved sequence; and a variant auxiliary oligonucleotide, wherein the variant auxiliary oligonucleotide is the second variant oligonucleotide separate from the variant nucleic acid probe. 2. The composition of claim 1 wherein the non-conserved sequence is a single nucleotide. 3. The composition of claim 1 wherein the target probe complement region and the variant probe complement region share two conserved sequences separated by a non-conserved sequence. 4. The composition of claim 3 wherein the non-conserved sequence is a single nucleotide. 5. The composition of claim 1 wherein the first target probe oligonucleotide comprises a detectable label or a capture moiety conjugated thereto. 6. The composition of claim 5 wherein the second target probe oligonucleotide comprises a quencher sufficient to prevent detection of the detectable label or capture of the capture moiety. 7. The composition of claim 1 further comprising a third target probe oligonucleotide hybridized to a target probe non-complement region of the first target probe oligonucleotide, wherein the third target probe oligonucleotide comprises a detectable label or a capture moiety conjugated thereto, and a fourth target probe oligonucleotide hybridized to a target probe non-protector region of the second target probe oligonucleotide, wherein the fourth target probe oligonucleotide comprises a quencher sufficient to prevent detection of the detectable label or capture of the capture moiety. 8. The composition of claim 1 wherein the concentration of the variant nucleic acid probe relative to the concentration of the target nucleic acid probe is from greater than 1:1 to about less than 100000:1. 9. The composition of claim 1 wherein the concentration of the target auxiliary oligonucleotide relative to the concentration of the target nucleic acid probe is from greater than 1:1000 to less than 100000:1. 10. The composition of claim 1 wherein the concentration of the variant auxiliary oligonucleotide relative to the concentration of the variant nucleic acid probe is from greater than 1:1000 to less than 100000:1. 11. The composition of claim 1 wherein the target nucleic acid probe has a target reaction standard free energy (ΔG°rxn1), wherein the variant nucleic acid probe has a variant reaction standard free energy (ΔG°rxn2), and wherein ΔG°rxn1 is greater than the sum of ΔG°rxn2+1 kcal/mol. 12. The composition of claim 1 wherein the target nucleic acid probe has a concentration-adjusted target reaction standard free energy defined as ΔG°rxn1+Rτ ln([Pt]/[PtCt]), wherein the variant nucleic acid probe has a variant reaction standard free energy defined as ΔG°rxn2+Rτ ln([Pv]/[PvCv]), where R is the ideal gas constant, is the temperature in Kelvin, Pt is the initial concentration of the target auxiliary oligonucleotide, PtCt is the initial concentration of the target nucleic acid probe, Pv is the initial concentration of the variant auxiliary oligonucleotide, PvCv is the initial concentration of the variant nucleic acid probe, wherein the concentration-adjusted reaction standard free energy of the target nucleic acid probe is greater than the sum of the concentration-adjusted reaction standard free energy of the variant nucleic acid probe +1 kcal/mol. 13. The composition of claim 1 wherein the target nucleic acid probe has a target reaction standard free energy (ΔG°rxn1), wherein the variant nucleic acid probe has a variant reaction standard free energy (ΔG°rxn2), and wherein ΔG°rxn1 is greater than ΔG°rxn2, and ΔG°rxn2 is greater than −7 kcal/mol. 14. The composition of claim 1 wherein the target nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first target probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first target probe oligonucleotide-specific subsequence is complementary to a target probe non-complement region of the first target probe oligonucleotide, wherein the fourth oligonucleotide comprises a second target probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second target probe oligonucleotide-specific subsequence is complementary to a target probe non-protector region of the second target probe oligonucleotide, wherein the target probe non-protector region of the second target probe oligonucleotide does not overlap with the target probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the target auxiliary oligonucleotide further comprises the fourth oligonucleotide. 15. The composition of claim 14 wherein the fourth oligonucleotide-specific subsequence of the third oligonucleotide comprises a detectable label or a capture moiety conjugated thereto, and wherein the third oligonucleotide-specific subsequence of the fourth oligonucleotide comprises a quencher sufficient to prevent detection of the detectable label or capture of the capture moiety. 16. The composition of claim 1 wherein the variant nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first variant probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-complement region of the first variant probe oligonucleotide, wherein the fourth oligonucleotide comprises a second variant probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-protector region of the second variant probe oligonucleotide, wherein the variant probe non-protector region of the second variant probe oligonucleotide does not overlap with the variant probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the variant auxiliary oligonucleotide further comprises the fourth oligonucleotide. 17. A nucleic acid detection composition comprising:
a target nucleic acid probe comprising a first target probe oligonucleotide and a second target probe oligonucleotide, wherein the first target probe oligonucleotide comprises a target probe complement region that is complementary to a target nucleic acid sequence, wherein the second target probe oligonucleotide comprises a target probe protector region that is complementary to a first target probe complement subsequence of the target probe complement region thereby providing a target double-stranded probe portion and a target single-stranded probe portion, wherein the target double-stranded probe portion comprises the first target probe complement subsequence and the target probe protector region, and the target single-stranded probe portion comprises a second target probe complement subsequence of the target probe complement region; a target auxiliary oligonucleotide, wherein the target auxiliary oligonucleotide is the second target probe oligonucleotide separate from the target nucleic acid probe; and a variant nucleic acid probe comprising a first variant probe oligonucleotide, wherein the first variant probe oligonucleotide comprises a variant probe complement region that is complementary to a variant nucleic acid sequence, wherein the target probe complement region and the variant probe complement region share at least two conserved sequences separated by a non-conserved sequence. 18. The composition of claim 17 wherein the target nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first target probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first target probe oligonucleotide-specific subsequence is complementary to a target probe non-complement region of the first target probe oligonucleotide, wherein the fourth oligonucleotide comprises a second target probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second target probe oligonucleotide-specific subsequence is complementary to a target probe non-protector region of the second target probe oligonucleotide, wherein the target probe non-protector region of the second target probe oligonucleotide does not overlap with the target probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the target auxiliary oligonucleotide further comprises the fourth oligonucleotide. 19. A nucleic acid detection composition comprising:
a target nucleic acid probe comprising a first target probe oligonucleotide, wherein the first target probe oligonucleotide comprises a target probe complement region that is complementary to a target nucleic acid sequence; a variant nucleic acid probe comprising a first variant probe oligonucleotide and a second variant probe oligonucleotide, wherein the first variant probe oligonucleotide comprises a variant probe complement region that is complementary to a variant nucleic acid sequence, wherein the second variant probe oligonucleotide comprises a variant probe protector region that is complementary to a first variant probe complement subsequence of the variant probe complement region thereby providing a variant double-stranded probe portion and a variant single-stranded probe portion, wherein the variant double-stranded probe portion comprises the first variant probe complement subsequence and the variant probe protector region, and the variant single-stranded probe portion comprises a second variant probe complement subsequence of the variant probe complement region, wherein the target probe complement region and the variant probe complement region share at least two conserved sequences separated by a non-conserved sequence; and a variant auxiliary oligonucleotide, wherein the variant auxiliary oligonucleotide is the second variant oligonucleotide separate from the variant nucleic acid probe. 20. The composition of claim 19 wherein the variant nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first variant probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-complement region of the first variant probe oligonucleotide, wherein the fourth oligonucleotide comprises a second variant probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-protector region of the second variant probe oligonucleotide, wherein the variant probe non-protector region of the second variant probe oligonucleotide does not overlap with the variant probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the variant auxiliary oligonucleotide further comprises the fourth oligonucleotide. | The present disclosure describes the thermodynamic design and concentrations necessary to design probe compositions with desired optimal specificity that enable enrichment, detection, quantitation, purification, imaging, and amplification of rare-allele-bearing species of nucleic acids (prevalence <1%) in a large stoichiometric excess of a dominant-allele-bearing species (wildtype). Being an enzyme-free and homogeneous nucleic acid enrichment composition, this technology is broadly compatible with nearly all nucleic acid-based biotechnology, including plate reader and fluorimeter readout of nucleic acids, microarrays, PCR and other enzymatic amplification reactions, fluorescence barcoding, nanoparticle-based purification and quantitation, and in situ hybridization imaging technologies.1. A nucleic acid detection composition comprising:
a target nucleic acid probe comprising a first target probe oligonucleotide and a second target probe oligonucleotide, wherein the first target probe oligonucleotide comprises a target probe complement region that is complementary to a target nucleic acid sequence, wherein the second target probe oligonucleotide comprises a target probe protector region that is complementary to a first target probe complement subsequence of the target probe complement region thereby providing a target double-stranded probe portion and a target single-stranded probe portion, wherein the target double-stranded probe portion comprises the first target probe complement subsequence and the target probe protector region, and the target single-stranded probe portion comprises a second target probe complement subsequence of the target probe complement region; a target auxiliary oligonucleotide, wherein the target auxiliary oligonucleotide is the second target probe oligonucleotide separate from the target nucleic acid probe; a variant nucleic acid probe comprising a first variant probe oligonucleotide and a second variant probe oligonucleotide, wherein the first variant probe oligonucleotide comprises a variant probe complement region that is complementary to a variant nucleic acid sequence, wherein the second variant probe oligonucleotide comprises a variant probe protector region that is complementary to a first variant probe complement subsequence of the variant probe complement region thereby providing a variant double-stranded probe portion and a variant single-stranded probe portion, wherein the variant double-stranded probe portion comprises the first variant probe complement subsequence and the variant probe protector region, and the variant single-stranded probe portion comprises a second variant probe complement subsequence of the variant probe complement region, wherein the target probe complement region and the variant probe complement region share at least two conserved sequences separated by a non-conserved sequence; and a variant auxiliary oligonucleotide, wherein the variant auxiliary oligonucleotide is the second variant oligonucleotide separate from the variant nucleic acid probe. 2. The composition of claim 1 wherein the non-conserved sequence is a single nucleotide. 3. The composition of claim 1 wherein the target probe complement region and the variant probe complement region share two conserved sequences separated by a non-conserved sequence. 4. The composition of claim 3 wherein the non-conserved sequence is a single nucleotide. 5. The composition of claim 1 wherein the first target probe oligonucleotide comprises a detectable label or a capture moiety conjugated thereto. 6. The composition of claim 5 wherein the second target probe oligonucleotide comprises a quencher sufficient to prevent detection of the detectable label or capture of the capture moiety. 7. The composition of claim 1 further comprising a third target probe oligonucleotide hybridized to a target probe non-complement region of the first target probe oligonucleotide, wherein the third target probe oligonucleotide comprises a detectable label or a capture moiety conjugated thereto, and a fourth target probe oligonucleotide hybridized to a target probe non-protector region of the second target probe oligonucleotide, wherein the fourth target probe oligonucleotide comprises a quencher sufficient to prevent detection of the detectable label or capture of the capture moiety. 8. The composition of claim 1 wherein the concentration of the variant nucleic acid probe relative to the concentration of the target nucleic acid probe is from greater than 1:1 to about less than 100000:1. 9. The composition of claim 1 wherein the concentration of the target auxiliary oligonucleotide relative to the concentration of the target nucleic acid probe is from greater than 1:1000 to less than 100000:1. 10. The composition of claim 1 wherein the concentration of the variant auxiliary oligonucleotide relative to the concentration of the variant nucleic acid probe is from greater than 1:1000 to less than 100000:1. 11. The composition of claim 1 wherein the target nucleic acid probe has a target reaction standard free energy (ΔG°rxn1), wherein the variant nucleic acid probe has a variant reaction standard free energy (ΔG°rxn2), and wherein ΔG°rxn1 is greater than the sum of ΔG°rxn2+1 kcal/mol. 12. The composition of claim 1 wherein the target nucleic acid probe has a concentration-adjusted target reaction standard free energy defined as ΔG°rxn1+Rτ ln([Pt]/[PtCt]), wherein the variant nucleic acid probe has a variant reaction standard free energy defined as ΔG°rxn2+Rτ ln([Pv]/[PvCv]), where R is the ideal gas constant, is the temperature in Kelvin, Pt is the initial concentration of the target auxiliary oligonucleotide, PtCt is the initial concentration of the target nucleic acid probe, Pv is the initial concentration of the variant auxiliary oligonucleotide, PvCv is the initial concentration of the variant nucleic acid probe, wherein the concentration-adjusted reaction standard free energy of the target nucleic acid probe is greater than the sum of the concentration-adjusted reaction standard free energy of the variant nucleic acid probe +1 kcal/mol. 13. The composition of claim 1 wherein the target nucleic acid probe has a target reaction standard free energy (ΔG°rxn1), wherein the variant nucleic acid probe has a variant reaction standard free energy (ΔG°rxn2), and wherein ΔG°rxn1 is greater than ΔG°rxn2, and ΔG°rxn2 is greater than −7 kcal/mol. 14. The composition of claim 1 wherein the target nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first target probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first target probe oligonucleotide-specific subsequence is complementary to a target probe non-complement region of the first target probe oligonucleotide, wherein the fourth oligonucleotide comprises a second target probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second target probe oligonucleotide-specific subsequence is complementary to a target probe non-protector region of the second target probe oligonucleotide, wherein the target probe non-protector region of the second target probe oligonucleotide does not overlap with the target probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the target auxiliary oligonucleotide further comprises the fourth oligonucleotide. 15. The composition of claim 14 wherein the fourth oligonucleotide-specific subsequence of the third oligonucleotide comprises a detectable label or a capture moiety conjugated thereto, and wherein the third oligonucleotide-specific subsequence of the fourth oligonucleotide comprises a quencher sufficient to prevent detection of the detectable label or capture of the capture moiety. 16. The composition of claim 1 wherein the variant nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first variant probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-complement region of the first variant probe oligonucleotide, wherein the fourth oligonucleotide comprises a second variant probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-protector region of the second variant probe oligonucleotide, wherein the variant probe non-protector region of the second variant probe oligonucleotide does not overlap with the variant probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the variant auxiliary oligonucleotide further comprises the fourth oligonucleotide. 17. A nucleic acid detection composition comprising:
a target nucleic acid probe comprising a first target probe oligonucleotide and a second target probe oligonucleotide, wherein the first target probe oligonucleotide comprises a target probe complement region that is complementary to a target nucleic acid sequence, wherein the second target probe oligonucleotide comprises a target probe protector region that is complementary to a first target probe complement subsequence of the target probe complement region thereby providing a target double-stranded probe portion and a target single-stranded probe portion, wherein the target double-stranded probe portion comprises the first target probe complement subsequence and the target probe protector region, and the target single-stranded probe portion comprises a second target probe complement subsequence of the target probe complement region; a target auxiliary oligonucleotide, wherein the target auxiliary oligonucleotide is the second target probe oligonucleotide separate from the target nucleic acid probe; and a variant nucleic acid probe comprising a first variant probe oligonucleotide, wherein the first variant probe oligonucleotide comprises a variant probe complement region that is complementary to a variant nucleic acid sequence, wherein the target probe complement region and the variant probe complement region share at least two conserved sequences separated by a non-conserved sequence. 18. The composition of claim 17 wherein the target nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first target probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first target probe oligonucleotide-specific subsequence is complementary to a target probe non-complement region of the first target probe oligonucleotide, wherein the fourth oligonucleotide comprises a second target probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second target probe oligonucleotide-specific subsequence is complementary to a target probe non-protector region of the second target probe oligonucleotide, wherein the target probe non-protector region of the second target probe oligonucleotide does not overlap with the target probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the target auxiliary oligonucleotide further comprises the fourth oligonucleotide. 19. A nucleic acid detection composition comprising:
a target nucleic acid probe comprising a first target probe oligonucleotide, wherein the first target probe oligonucleotide comprises a target probe complement region that is complementary to a target nucleic acid sequence; a variant nucleic acid probe comprising a first variant probe oligonucleotide and a second variant probe oligonucleotide, wherein the first variant probe oligonucleotide comprises a variant probe complement region that is complementary to a variant nucleic acid sequence, wherein the second variant probe oligonucleotide comprises a variant probe protector region that is complementary to a first variant probe complement subsequence of the variant probe complement region thereby providing a variant double-stranded probe portion and a variant single-stranded probe portion, wherein the variant double-stranded probe portion comprises the first variant probe complement subsequence and the variant probe protector region, and the variant single-stranded probe portion comprises a second variant probe complement subsequence of the variant probe complement region, wherein the target probe complement region and the variant probe complement region share at least two conserved sequences separated by a non-conserved sequence; and a variant auxiliary oligonucleotide, wherein the variant auxiliary oligonucleotide is the second variant oligonucleotide separate from the variant nucleic acid probe. 20. The composition of claim 19 wherein the variant nucleic acid probe further comprises a third oligonucleotide and a fourth oligonucleotide, wherein the third oligonucleotide comprises a first variant probe oligonucleotide-specific subsequence and a fourth oligonucleotide specific subsequence, wherein the first variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-complement region of the first variant probe oligonucleotide, wherein the fourth oligonucleotide comprises a second variant probe oligonucleotide-specific subsequence and a third oligonucleotide-specific subsequence, wherein the second variant probe oligonucleotide-specific subsequence is complementary to a variant probe non-protector region of the second variant probe oligonucleotide, wherein the variant probe non-protector region of the second variant probe oligonucleotide does not overlap with the variant probe protector region, wherein the fourth oligonucleotide-specific subsequence is complementary to the third oligonucleotide-specific subsequence, and wherein the variant auxiliary oligonucleotide further comprises the fourth oligonucleotide. | 1,600 |
645 | 15,702,462 | 1,613 | Magnetically driven biocompatible microrobots comprising a porous body having a magnetic layer and a biocompatible layer configured to carry and deliver cells to desired sites are described. Embodiments of microrobots are configured with enhanced cell-loading ability, such as by including a plurality of burr members disposed upon the porous body for configuring the microrobot for enhanced cell-loading. The magnetic layer of embodiments may be provided on some portion or all of a surface of the microrobot for configuring the microrobot to be controlled with an external magnetic field. The biocompatible layer of embodiments may be provided on some portion or all of a surface of the microrobot, possibly coating some or all of the aforementioned magnetic layer, for configuring the microrobot for improved biostability and biocompatibility. | 1. A microrobot configured to be magnetically driven and biocompatible, the microrobot comprising:
a porous body having a three-dimensional structures; a plurality of burr members disposed on the porous body, wherein burr members of the plurality of burr members extend orthogonally from an outer surface of the porous body and are configured for carrying cells to desired sites by the microrobot between adjacent burr members of the plurality of burr members; a magnetic layer coating at least a portion of the porous body or at least a portion of the burr members; and a biocompatible layer coating at least a portion of the burr members for cell adhesion between adjacent burr members of the plurality of burr members. 2. The microrobot of claim 1, wherein the porous body comprises:
a photocurable polymer. 3. The microrobot of claim 1, wherein the three-dimensional structure comprises a structure selected from the group consisting of:
a cylinder; a hexahedron; an ellipsoid; a polyhedron; a circular cone; and a sphere. 4. The microrobot of claim 1, wherein pores of the porous body are sized in correspondence with a type of cells to be carried by the microrobot. 5. The microrobot of claim 1, wherein the porous body is configured to mimic an extracellular matrix in which nutrients are supplied for tissue vascularization to yield functional tissues. 6. The microrobot of claim 1, wherein the magnetic layer comprises a metal selected from the group consisting of:
nickel (Ni); iron (Fe); cobalt (Co); neodymium (Nd); and combinations thereof. 7. The microrobot of claim 1, wherein the biocompatible layer fully covers the magnetic layer. 8. The microrobot of claim 1, wherein the biocompatible layer comprises a metal selected from the group consisting of:
titanium (Ti); medical stainless steel; alumina (Al203): gold (Au); and combinations thereof. 9. The microrobot of claim 1, wherein burr members of the plurality of burr members include ball terminal structures at a distal end thereof configured for minimizing potential for damage to tissues by the microrobot. 10. The microrobot of claim 1, wherein pores of the porous body are sized to facilitate supply of nutrients for tissue vascularization, and wherein adjacent burr members of the plurality of burr members have an interspacing corresponding with a type of cells to be carried by the microrobot. 11. The microrobot of claim 1, wherein each burr member of the plurality of burr members comprises:
a protuberance extending from a surface of the porous body, wherein the protuberance is formed in a shape selected from the group consisting of a cylinder, a hexahedron, an ellipsoid, and a polyhedron. 12. The microrobot of claim 1, wherein the plurality of burr members are configured for culturing cells between adjacent burr members of the plurality of burr members. 13. A system for delivery of cells to a desired biological site, the system comprising:
a plurality of microrobots configured to be magnetically driven and biocompatible, each microrobot of the plurality of microrobots including a porous body having a three-dimensional structure with a plurality of burr members extending orthogonally from an outer surface of the porous body, a magnetic layer coating at least a portion of the porous body and configured to enable the microrobot to be positioned at a target site using a magnetic field, and a biocompatible layer coating at least a portion of the burr members for cell adhesion between adjacent burr members of the plurality of burr members and configured for microrobot biostability and biocompatibility, wherein the plurality of burr members are configured for carrying cells to desired sites by the microrobot between adjacent burr members of the plurality of members. 14. The system of claim 13, wherein the porous body of microrobots of the plurality of micro comprises:
a photocurable polymer formed in a structure selected from the group consisting of a cylinder, a hexahedron, an ellipsoid, a polyhedron, a circular cone, and a sphere. 15. The system of claim 13, wherein burr members of the plurality of burr members include ball terminal structures at a distal end thereof configured for minimizing potential for damage to tissues by the plurality of microrobots. 16. The system of claim 13, wherein each burr member of the plurality of burr members comprises:
a protuberance extending from a surface of the porous body, wherein the protuberance is formed in a shape selected from the group consisting of a cylinder, a hexahedron, an ellipsoid, and a polyhedron. 17. The system of claim 13, wherein the plurality of burr members are configured for culturing cells between adjacent burr members of the plurality of burr members. 18. The system of claim 13, wherein pores of the porous body are sized to facilitate supply of nutrients for tissue vascularization, and wherein adjacent burr members of the plurality of burr members of a microrobot of the plurality of microrobots have an interspacing corresponding with a type of cells to be carried by the microrobot. 19. The system of claim 13, wherein pores of the porous body of the microrobot of the plurality of microrobots are sized in correspondence with the type of cells to be carried by the microrobot, wherein the interspacing of the adjacent burr members is defined by a respective pore sized in correspondence with the type of cells to be carried by the microrobot. 20. The system of claim 13, wherein the magnetic layer comprises a metal selected from the group consisting of nickel (Ni), iron (Fe), cobalt (Co), neodymium (Nd), and combinations thereof, and wherein the biocompatible layer comprises a metal selected from the group consisting of titanium (Ti), medical stainless steel, alumina (Al203), gold (Au), and combinations thereof. 21. A method for fabricating a microrobot configured to be magnetically driven and biocompatible, the method comprising:
providing a porous body having a three-dimensional structure with a plurality of burr members, wherein the plurality of burr members are configured for carrying cells to desired sites by the microrobot between adjacent burr members of the plurality of members; coating at least a portion of the porous body with a magnetic layer, wherein the magnetic layer is configured to enable the microrobot to be positioned at a target site using a magnetic field; and coating at least a portion of the burr members with a biocompatible layer for cell adhesion between adjacent burr members of the plurality of burr members, wherein the biocompatible layer is configured for microrobot biostability and biocompatibility. 22. The method of claim 21, wherein the providing the porous body comprises:
using a lithographic process with a photocurable polymer to form the porous body. 23. The method of claim 21, wherein the coating the at least a portion of the porous body with a magnetic layer with the magnetic layer comprises disposing a metal on the at least a portion of the porous body selected from the group consisting of nickel (Ni), iron (Fe), cobalt (Co), neodymium (Nd), and combinations thereof. 24. The method of claim 21, wherein the coating the at least a portion of the burr members with the biocompatible layer comprises disposing a metal on the at least a portion of the burr members selected from the group consisting of titanium (Ti), medical stainless steel, alumina (Al203), gold (Au), and combinations thereof. 25. The method of claim 21, wherein the plurality of burr members are configured for culturing cells between adjacent burr members of the plurality of burr members. 26. The method of claim 25, wherein the providing the plurality of burr members comprises:
using a lithographic process with a photocurable polymer to form both the porous body and the plurality of burr members. 27. The method of claim 25, further comprising:
cultivating cells on the microrobot between adjacent burr members of the plurality of burr members. | Magnetically driven biocompatible microrobots comprising a porous body having a magnetic layer and a biocompatible layer configured to carry and deliver cells to desired sites are described. Embodiments of microrobots are configured with enhanced cell-loading ability, such as by including a plurality of burr members disposed upon the porous body for configuring the microrobot for enhanced cell-loading. The magnetic layer of embodiments may be provided on some portion or all of a surface of the microrobot for configuring the microrobot to be controlled with an external magnetic field. The biocompatible layer of embodiments may be provided on some portion or all of a surface of the microrobot, possibly coating some or all of the aforementioned magnetic layer, for configuring the microrobot for improved biostability and biocompatibility.1. A microrobot configured to be magnetically driven and biocompatible, the microrobot comprising:
a porous body having a three-dimensional structures; a plurality of burr members disposed on the porous body, wherein burr members of the plurality of burr members extend orthogonally from an outer surface of the porous body and are configured for carrying cells to desired sites by the microrobot between adjacent burr members of the plurality of burr members; a magnetic layer coating at least a portion of the porous body or at least a portion of the burr members; and a biocompatible layer coating at least a portion of the burr members for cell adhesion between adjacent burr members of the plurality of burr members. 2. The microrobot of claim 1, wherein the porous body comprises:
a photocurable polymer. 3. The microrobot of claim 1, wherein the three-dimensional structure comprises a structure selected from the group consisting of:
a cylinder; a hexahedron; an ellipsoid; a polyhedron; a circular cone; and a sphere. 4. The microrobot of claim 1, wherein pores of the porous body are sized in correspondence with a type of cells to be carried by the microrobot. 5. The microrobot of claim 1, wherein the porous body is configured to mimic an extracellular matrix in which nutrients are supplied for tissue vascularization to yield functional tissues. 6. The microrobot of claim 1, wherein the magnetic layer comprises a metal selected from the group consisting of:
nickel (Ni); iron (Fe); cobalt (Co); neodymium (Nd); and combinations thereof. 7. The microrobot of claim 1, wherein the biocompatible layer fully covers the magnetic layer. 8. The microrobot of claim 1, wherein the biocompatible layer comprises a metal selected from the group consisting of:
titanium (Ti); medical stainless steel; alumina (Al203): gold (Au); and combinations thereof. 9. The microrobot of claim 1, wherein burr members of the plurality of burr members include ball terminal structures at a distal end thereof configured for minimizing potential for damage to tissues by the microrobot. 10. The microrobot of claim 1, wherein pores of the porous body are sized to facilitate supply of nutrients for tissue vascularization, and wherein adjacent burr members of the plurality of burr members have an interspacing corresponding with a type of cells to be carried by the microrobot. 11. The microrobot of claim 1, wherein each burr member of the plurality of burr members comprises:
a protuberance extending from a surface of the porous body, wherein the protuberance is formed in a shape selected from the group consisting of a cylinder, a hexahedron, an ellipsoid, and a polyhedron. 12. The microrobot of claim 1, wherein the plurality of burr members are configured for culturing cells between adjacent burr members of the plurality of burr members. 13. A system for delivery of cells to a desired biological site, the system comprising:
a plurality of microrobots configured to be magnetically driven and biocompatible, each microrobot of the plurality of microrobots including a porous body having a three-dimensional structure with a plurality of burr members extending orthogonally from an outer surface of the porous body, a magnetic layer coating at least a portion of the porous body and configured to enable the microrobot to be positioned at a target site using a magnetic field, and a biocompatible layer coating at least a portion of the burr members for cell adhesion between adjacent burr members of the plurality of burr members and configured for microrobot biostability and biocompatibility, wherein the plurality of burr members are configured for carrying cells to desired sites by the microrobot between adjacent burr members of the plurality of members. 14. The system of claim 13, wherein the porous body of microrobots of the plurality of micro comprises:
a photocurable polymer formed in a structure selected from the group consisting of a cylinder, a hexahedron, an ellipsoid, a polyhedron, a circular cone, and a sphere. 15. The system of claim 13, wherein burr members of the plurality of burr members include ball terminal structures at a distal end thereof configured for minimizing potential for damage to tissues by the plurality of microrobots. 16. The system of claim 13, wherein each burr member of the plurality of burr members comprises:
a protuberance extending from a surface of the porous body, wherein the protuberance is formed in a shape selected from the group consisting of a cylinder, a hexahedron, an ellipsoid, and a polyhedron. 17. The system of claim 13, wherein the plurality of burr members are configured for culturing cells between adjacent burr members of the plurality of burr members. 18. The system of claim 13, wherein pores of the porous body are sized to facilitate supply of nutrients for tissue vascularization, and wherein adjacent burr members of the plurality of burr members of a microrobot of the plurality of microrobots have an interspacing corresponding with a type of cells to be carried by the microrobot. 19. The system of claim 13, wherein pores of the porous body of the microrobot of the plurality of microrobots are sized in correspondence with the type of cells to be carried by the microrobot, wherein the interspacing of the adjacent burr members is defined by a respective pore sized in correspondence with the type of cells to be carried by the microrobot. 20. The system of claim 13, wherein the magnetic layer comprises a metal selected from the group consisting of nickel (Ni), iron (Fe), cobalt (Co), neodymium (Nd), and combinations thereof, and wherein the biocompatible layer comprises a metal selected from the group consisting of titanium (Ti), medical stainless steel, alumina (Al203), gold (Au), and combinations thereof. 21. A method for fabricating a microrobot configured to be magnetically driven and biocompatible, the method comprising:
providing a porous body having a three-dimensional structure with a plurality of burr members, wherein the plurality of burr members are configured for carrying cells to desired sites by the microrobot between adjacent burr members of the plurality of members; coating at least a portion of the porous body with a magnetic layer, wherein the magnetic layer is configured to enable the microrobot to be positioned at a target site using a magnetic field; and coating at least a portion of the burr members with a biocompatible layer for cell adhesion between adjacent burr members of the plurality of burr members, wherein the biocompatible layer is configured for microrobot biostability and biocompatibility. 22. The method of claim 21, wherein the providing the porous body comprises:
using a lithographic process with a photocurable polymer to form the porous body. 23. The method of claim 21, wherein the coating the at least a portion of the porous body with a magnetic layer with the magnetic layer comprises disposing a metal on the at least a portion of the porous body selected from the group consisting of nickel (Ni), iron (Fe), cobalt (Co), neodymium (Nd), and combinations thereof. 24. The method of claim 21, wherein the coating the at least a portion of the burr members with the biocompatible layer comprises disposing a metal on the at least a portion of the burr members selected from the group consisting of titanium (Ti), medical stainless steel, alumina (Al203), gold (Au), and combinations thereof. 25. The method of claim 21, wherein the plurality of burr members are configured for culturing cells between adjacent burr members of the plurality of burr members. 26. The method of claim 25, wherein the providing the plurality of burr members comprises:
using a lithographic process with a photocurable polymer to form both the porous body and the plurality of burr members. 27. The method of claim 25, further comprising:
cultivating cells on the microrobot between adjacent burr members of the plurality of burr members. | 1,600 |
646 | 16,278,008 | 1,656 | The present invention provides a microorganism-derived soluble coenzyme-binding glucose dehydrogenase which catalyzes a reaction for oxidizing glucose in the presence of an electron acceptor, has an activity to maltose as low as 5% or less, and is inhibited by 1,10-phenanthroline. The invention also provides a method for producing the coenzyme-binding glucose dehydrogenase, and a method and a reagent for measuring employing the coenzyme-binding glucose dehydrogenase. According to the invention, the coenzyme-binding glucose dehydrogenase can be applied to an industrial field, and a use becomes possible also in a material production or analysis including a method for measuring or eliminating glucose in a sample using the coenzyme-binding glucose dehydrogenase as well as a method for producing an organic compound. It became also possible to provide a glucose sensor capable of accurately measuring a blood sugar level. Therefore, it became possible to provide an enzyme having a high utility, such as an ability of being used for modifying a material in the fields of pharmaceuticals, clinical studies and food products. | 1-22. (canceled) 23. A method for producing a biosensor for measuring glucose in a sample liquid comprising:
(i) obtaining a soluble flavin compound-binding glucose dehydrogenase secreted from an Aspergillus fungal body, which has enzymatic activity to glucose comprising catalyzing a reaction for oxidizing glucose in the presence of an electron acceptor, and (ii) forming a biosensor comprising an electrode system and an enzymatic reaction layer on an electrode of the electrode system, the enzymatic reaction layer comprising the soluble flavin compound-binding glucose dehydrogenase and an electron acceptor, wherein: (a) enzymatic activity to maltose is 5% or less relative to the enzymatic activity to glucose, and (b) enzymatic activity to D-fructose is not more than enzymatic activity to D-mannose. 24. The method for producing a biosensor of claim 23, wherein the enzymatic activity to maltose is 3% or less relative to the enzymatic activity to glucose. | The present invention provides a microorganism-derived soluble coenzyme-binding glucose dehydrogenase which catalyzes a reaction for oxidizing glucose in the presence of an electron acceptor, has an activity to maltose as low as 5% or less, and is inhibited by 1,10-phenanthroline. The invention also provides a method for producing the coenzyme-binding glucose dehydrogenase, and a method and a reagent for measuring employing the coenzyme-binding glucose dehydrogenase. According to the invention, the coenzyme-binding glucose dehydrogenase can be applied to an industrial field, and a use becomes possible also in a material production or analysis including a method for measuring or eliminating glucose in a sample using the coenzyme-binding glucose dehydrogenase as well as a method for producing an organic compound. It became also possible to provide a glucose sensor capable of accurately measuring a blood sugar level. Therefore, it became possible to provide an enzyme having a high utility, such as an ability of being used for modifying a material in the fields of pharmaceuticals, clinical studies and food products.1-22. (canceled) 23. A method for producing a biosensor for measuring glucose in a sample liquid comprising:
(i) obtaining a soluble flavin compound-binding glucose dehydrogenase secreted from an Aspergillus fungal body, which has enzymatic activity to glucose comprising catalyzing a reaction for oxidizing glucose in the presence of an electron acceptor, and (ii) forming a biosensor comprising an electrode system and an enzymatic reaction layer on an electrode of the electrode system, the enzymatic reaction layer comprising the soluble flavin compound-binding glucose dehydrogenase and an electron acceptor, wherein: (a) enzymatic activity to maltose is 5% or less relative to the enzymatic activity to glucose, and (b) enzymatic activity to D-fructose is not more than enzymatic activity to D-mannose. 24. The method for producing a biosensor of claim 23, wherein the enzymatic activity to maltose is 3% or less relative to the enzymatic activity to glucose. | 1,600 |
647 | 15,122,451 | 1,616 | The present invention relates to an emulsifiable or self-emulsifying liquid composition for plant growth control which contains cyanamide. | 1.-12. (canceled) 13. An emulsifiable or self-emulsifying liquid composition for regulating plant growth, comprising:
a) 5 to 50 wt. % cyanamide; b) 10 to 95 wt. % of a mixture of at least one oil and at least one organic solvent that is miscible with water; and, c) 0.1 to 10 wt. % of at least one emulsifier, wherein the cyanamide is present in the composition in dissolved form. 14. The composition of claim 13, wherein the organic solvent is selected from the group consisting of water-soluble alcohols, ketones, nitriles and amides, and mixtures thereof. 15. The composition of claim 13, wherein the organic solvent is selected from the group consisting of ethanol, isopropanol, n-propanol, acetone, methyl ethyl ketone, acetonitrile, propionitrile, formamide, dimethylformamide and N-methyl pyrrilidone, and mixtures thereof. 16. The composition of claim 13, wherein the composition contains less than 5 wt. % water. 17. The composition of claim 13, wherein the composition contains less than 3 wt. % water. 18. The composition of claim 13, wherein the oil is selected from the group consisting of mineral oil, paraffin oil, white oil, saturated linear or branched aliphatic hydrocarbons, esters of saturated or unsaturated fatty acids having monovalent, divalent or trivalent C1 to C5 alcohols and ethers of fatty alcohols having C1 to C5 alcohols, and mixtures thereof 19. The composition of claim 13, wherein the mixture contains the organic solvent and the oil in a ratio range of organic solvent to oil of from 4:1 to 1:4. 20. The composition of claim 13, wherein the mixture contains the organic solvent and the oil in a ratio range of organic solvent to oil of from 2:1 to 1:2. 21. The composition of claim 13, wherein the emulsifier is an ionic, a non-ionic or a zwitterionic emulsifier. 22. The composition of claim 13, wherein the emulsifier is selected from the group consisting of oil-in-water emulsifiers (O/W emulsifier), water-in-oil emulsifiers (W/O emulsifier), oil-in-water-in-oil emulsifiers (O/W/O emulsifier) and water-in-oil-in-water emulsifiers (W/O/W emulsifier). 23. The composition of claim 13, further comprising at least one additional additive. 24. The composition of claim 23 wherein the additional additive is selected from the group consisting of viscosity modifiers, stabilisers, dyes and bitter principles. 25. The composition of claim 13, wherein the composition is a self-emulsifying composition. 26. The composition of claim 13, wherein the composition has a viscosity of less than 1 Pa*s. 27. The composition of claim 25, wherein the composition has a viscosity of less than 1 Pa*s. 28. An emulsion for regulating plant growth comprising water and the composition of claim 13. | The present invention relates to an emulsifiable or self-emulsifying liquid composition for plant growth control which contains cyanamide.1.-12. (canceled) 13. An emulsifiable or self-emulsifying liquid composition for regulating plant growth, comprising:
a) 5 to 50 wt. % cyanamide; b) 10 to 95 wt. % of a mixture of at least one oil and at least one organic solvent that is miscible with water; and, c) 0.1 to 10 wt. % of at least one emulsifier, wherein the cyanamide is present in the composition in dissolved form. 14. The composition of claim 13, wherein the organic solvent is selected from the group consisting of water-soluble alcohols, ketones, nitriles and amides, and mixtures thereof. 15. The composition of claim 13, wherein the organic solvent is selected from the group consisting of ethanol, isopropanol, n-propanol, acetone, methyl ethyl ketone, acetonitrile, propionitrile, formamide, dimethylformamide and N-methyl pyrrilidone, and mixtures thereof. 16. The composition of claim 13, wherein the composition contains less than 5 wt. % water. 17. The composition of claim 13, wherein the composition contains less than 3 wt. % water. 18. The composition of claim 13, wherein the oil is selected from the group consisting of mineral oil, paraffin oil, white oil, saturated linear or branched aliphatic hydrocarbons, esters of saturated or unsaturated fatty acids having monovalent, divalent or trivalent C1 to C5 alcohols and ethers of fatty alcohols having C1 to C5 alcohols, and mixtures thereof 19. The composition of claim 13, wherein the mixture contains the organic solvent and the oil in a ratio range of organic solvent to oil of from 4:1 to 1:4. 20. The composition of claim 13, wherein the mixture contains the organic solvent and the oil in a ratio range of organic solvent to oil of from 2:1 to 1:2. 21. The composition of claim 13, wherein the emulsifier is an ionic, a non-ionic or a zwitterionic emulsifier. 22. The composition of claim 13, wherein the emulsifier is selected from the group consisting of oil-in-water emulsifiers (O/W emulsifier), water-in-oil emulsifiers (W/O emulsifier), oil-in-water-in-oil emulsifiers (O/W/O emulsifier) and water-in-oil-in-water emulsifiers (W/O/W emulsifier). 23. The composition of claim 13, further comprising at least one additional additive. 24. The composition of claim 23 wherein the additional additive is selected from the group consisting of viscosity modifiers, stabilisers, dyes and bitter principles. 25. The composition of claim 13, wherein the composition is a self-emulsifying composition. 26. The composition of claim 13, wherein the composition has a viscosity of less than 1 Pa*s. 27. The composition of claim 25, wherein the composition has a viscosity of less than 1 Pa*s. 28. An emulsion for regulating plant growth comprising water and the composition of claim 13. | 1,600 |
648 | 12,840,420 | 1,653 | The invention is generally directed to reducing inflammation by means of cells that secrete factors that reduce leukocyte extravasation. Specifically, the invention is directed to methods using cells that secrete factors that downregulate the expression of cellular adhesion molecules in leukocytes. Downregulating expression of cellular adhesion molecules reduces leukocyte adhesion to endothelial cells such that extravasation is reduced. The end result is a reduction of inflammation. The cells are non-embryonic non-germ cells that have pluripotent characteristics. These may include expression of pluripotential markers and broad differentiation potential. | 1. A method of obtaining a cell that has a desired potency for one or more of the following: (1) reduce leukocyte extravasation, (2) reduce leukocyte adhesion to vascular endothelium or to isolated endothelial cells, (3) reduce Fut-7 expression, (4) reduce expression of CD15s on a leukocyte, the method comprising assessing cells for and selecting cells that have a desired potency for one or more of (1)-(4) above, the cells being non-embryonic, non-germ cells that express one or more of oct4, telomerase, rex-1, or rox-1 and/or can differentiate into cell types of at least two of endodermal, ectodermal, and mesodermal germ layers. 2. A method to treat inflammation in a subject, said method comprising administering the selected cells of claim 1 to the subject in a therapeutically effective amount and for a time sufficient to achieve a therapeutic result. 3. A method to construct a cell bank, said method comprising expanding and storing the selected cells of claim 1 for future administration to a subject. 4. A method for drug discovery, said method comprising exposing the selected cells to an agent to assess the effect of the agent on the ability of the cells to reduce any of events (1)-(4). 5. A composition comprising cells selected for the desired potency to achieve one or more of the following: 1) reduce leukocyte extravasation, (2) reduce leukocyte adhesion to vascular endothelium or to isolated endothelial cells, (3) reduce Fut-7 expression, (4) reduce expression of CD15s on a leukocyte;
the cells being non-embryonic, non-germ cells that express one or more of oct4, telomerase, rex-1, or rox-1 and/or can differentiate into cell types of at least two of endodermal, ectodermal, and mesodermal germ layers. 6. The method of claim 1 wherein adhesion is of E-selectin and/or P-selectin to CD15s. 7. The method of claim 1 wherein the leukocyte is a lymphocyte. 8. The method of claim 7 wherein the lymphocyte is a CD4+ of CD8+ lymphocyte. 9. The method of claim 6 wherein the leukocyte is a lymphocyte. 10. The method of claim 9 wherein the lymphocyte is a CD4+ of CD8+ lymphocyte. 11. The method of claim 1 wherein the leukocyte is a neutrophil. 12. The method of claim 6 wherein the leukocyte is a neutrophil. | The invention is generally directed to reducing inflammation by means of cells that secrete factors that reduce leukocyte extravasation. Specifically, the invention is directed to methods using cells that secrete factors that downregulate the expression of cellular adhesion molecules in leukocytes. Downregulating expression of cellular adhesion molecules reduces leukocyte adhesion to endothelial cells such that extravasation is reduced. The end result is a reduction of inflammation. The cells are non-embryonic non-germ cells that have pluripotent characteristics. These may include expression of pluripotential markers and broad differentiation potential.1. A method of obtaining a cell that has a desired potency for one or more of the following: (1) reduce leukocyte extravasation, (2) reduce leukocyte adhesion to vascular endothelium or to isolated endothelial cells, (3) reduce Fut-7 expression, (4) reduce expression of CD15s on a leukocyte, the method comprising assessing cells for and selecting cells that have a desired potency for one or more of (1)-(4) above, the cells being non-embryonic, non-germ cells that express one or more of oct4, telomerase, rex-1, or rox-1 and/or can differentiate into cell types of at least two of endodermal, ectodermal, and mesodermal germ layers. 2. A method to treat inflammation in a subject, said method comprising administering the selected cells of claim 1 to the subject in a therapeutically effective amount and for a time sufficient to achieve a therapeutic result. 3. A method to construct a cell bank, said method comprising expanding and storing the selected cells of claim 1 for future administration to a subject. 4. A method for drug discovery, said method comprising exposing the selected cells to an agent to assess the effect of the agent on the ability of the cells to reduce any of events (1)-(4). 5. A composition comprising cells selected for the desired potency to achieve one or more of the following: 1) reduce leukocyte extravasation, (2) reduce leukocyte adhesion to vascular endothelium or to isolated endothelial cells, (3) reduce Fut-7 expression, (4) reduce expression of CD15s on a leukocyte;
the cells being non-embryonic, non-germ cells that express one or more of oct4, telomerase, rex-1, or rox-1 and/or can differentiate into cell types of at least two of endodermal, ectodermal, and mesodermal germ layers. 6. The method of claim 1 wherein adhesion is of E-selectin and/or P-selectin to CD15s. 7. The method of claim 1 wherein the leukocyte is a lymphocyte. 8. The method of claim 7 wherein the lymphocyte is a CD4+ of CD8+ lymphocyte. 9. The method of claim 6 wherein the leukocyte is a lymphocyte. 10. The method of claim 9 wherein the lymphocyte is a CD4+ of CD8+ lymphocyte. 11. The method of claim 1 wherein the leukocyte is a neutrophil. 12. The method of claim 6 wherein the leukocyte is a neutrophil. | 1,600 |
649 | 13,464,452 | 1,631 | Computational methods are used to create cardiovascular simulations having desired hemodynamic features. Cardiovascular modeling methods produce descriptions of blood flow and pressure in the heart and vascular networks. Numerical methods optimize and solve nonlinear equations to find parameter values that result in desired hemodynamic characteristics including related flow and pressure at various locations in the cardiovascular system, movements of soft tissues, and changes for different physiological states. The modeling methods employ simplified models to approximate the behavior of more complex models with the goal of to reducing computational expense. The user describes the desired features of the final cardiovascular simulation and provides minimal input, and the system automates the search for the final patient-specific cardiovascular model. | 1.-19. (canceled) 20. A system for determining cardiovascular information for a patient, the system comprising:
at least one computer system configured to:
receive patient-specific data regarding a geometry of an anatomical structure of the patient;
create, based on the patient-specific data, a three-dimensional model representing at least a portion of the anatomical structure of the patient, the three-dimensional model including portions representing at least one inlet and a plurality of outlets for blood flow;
create at least one boundary condition model representing blood flow through at least one of the at least one inlet or the plurality of outlets, the three-dimensional model and the at least one boundary condition model forming a first model;
determine a blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the first model; and
create, based on the patient-specific data and on the determined blood flow characteristic, a second model having a lower level of detail than the first model, the second model representing at least part of the portion of the anatomical structure of the patient represented by the three-dimensional model. 21. The system of claim 20, wherein the second model reproduces, within a tolerance, the blood flow characteristic determined using the first model. 22. The system of claim 20, wherein the at least one computer system is further configured to determine the blood flow characteristic within the anatomical structure of the patient using the second model. 23. The system of claim 22, wherein the at least one computer system is further configured to:
compare the blood flow characteristics determined using the first model with the blood flow characteristics determined using the second model; and determine whether to adjust the second model based on the comparison. 24. The system of claim 23, wherein the at least one computer system is further configured to determine whether to adjust the second model based on whether differences between the blood flow characteristics determined using the first model and the blood flow characteristics determined using the second model are outside a tolerance. 25. The system of claim 24, wherein the at least one computer system is further configured to adjust the second model by increasing the level of detail of the second model when the differences are outside the tolerance. 26. The system of claim 23, wherein the at least one computer system is further configured to adjust the second model by adjusting the level of detail of the second model. 27. The system of claim 20, wherein the at least one computer system is further configured to determine whether to adjust the second model to include a lower level of detail while reproducing, within a tolerance, the blood flow characteristic determined using the first model. 28. The system of claim 20, wherein:
the first model includes a three-dimensional mesh created based on the three-dimensional model; and the second model has less than three dimensions. 29. The system of claim 20, wherein:
the first model includes a three-dimensional mesh created based on the three-dimensional model; and the second model includes a mesh that is less refined than the three-dimensional mesh of the first model. 30. The system of claim 20, wherein the at least one computer system is further configured to determine the blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the second model, the first model being associated with a more complete polynomial basis for solving the equations governing blood flow than the second model. 31. The system of claim 20, wherein:
the first model represents flow in the at least one blood vessel using an elastance-based model of the patient's cardiac chamber; and the second model includes a prescribed flow in the at least one blood vessel in the anatomical structure of the patient. 32. The system of claim 20, wherein the at least one computer system is further configured to:
determine the blood flow characteristic by solving a system of differential equations with numerical methods for temporal integration using the first model; and determine the blood flow characteristic by solving a system of differential equations with Fourier analysis using the second model. 33. The system of claim 20, wherein the second model includes a model assuming vascular walls that are more rigid than the corresponding vascular walls of the first model. 34. The system of claim 20, wherein:
the first model includes a model assuming rigid vascular walls; and the second model includes a model assuming vascular walls that are less rigid than the corresponding vascular walls of the first model. 35. The system of claim 20, wherein:
the anatomical structure includes at least a portion of a plurality of coronary arteries emanating from the aorta; and the portion of the anatomical structure represented by the three-dimensional model includes at least a portion of the plurality of coronary arteries, the three-dimensional model representing at least one fluid flow inlet and at least one fluid flow outlet. 36. The system of claim 20, wherein the at least one computer system is further configured to adjust the second model to predict the results of a treatment option. 37. The system of claim 20, wherein the at least one computer system is further configured to use the second model to predict the results of a treatment option. 38. The system of claim 20, wherein the patient-specific data includes imaging data. 39. The system of claim 20, wherein the blood flow characteristic includes at least one of blood flow or pressure within the anatomical structure of the patient. 40. A method for determining cardiovascular information for a patient using at least one computer system, the method comprising:
inputting into the at least one computer system patient-specific data regarding a geometry of an anatomical structure of the patient; creating, using the at least one computer system and based on the patient-specific data, a three-dimensional model representing at least a portion of the anatomical structure of the patient, the three-dimensional model including portions representing at least one inlet and a plurality of outlets for blood flow; creating, using the at least one computer system, at least one boundary condition model representing blood flow through at least one of the at least one inlet or the plurality of outlets, the three-dimensional model and the at least one boundary condition model forming a first model; determining, using the at least one computer system, a blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the first model; and creating, using the at least one computer system and based on the patient-specific data and on the determined blood flow characteristic, a second model having a lower level of detail than the first model, the second model representing at least part of the portion of the anatomical structure of the patient represented by the three-dimensional model. 41. The method of claim 40, wherein the second model reproduces, within a tolerance, the blood flow characteristic determined using the first model. 42. The method of claim 40, further including adjusting, using the at least one computer system, the second model to predict the results of a treatment option. 43. The method of claim 40, further including determining, using the at least one computer system, the blood flow characteristic within the anatomical structure of the patient using the second model. 44. A non-transitory computer readable medium for use on at least one computer system containing computer-executable programming instructions for performing a method for determining patient-specific cardiovascular information, the method comprising:
receiving patient-specific data regarding a geometry of an anatomical structure of the patient; creating, based on the patient-specific data, a three-dimensional model representing at least a portion of the anatomical structure of the patient, the three-dimensional model including portions representing at least one inlet and a plurality of outlets for blood flow; creating at least one boundary condition model representing blood flow through at least one of the at least one inlet or the plurality of outlets, the three-dimensional model and the at least one boundary condition model forming a first model; determining a blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the first model; and creating, based on the patient-specific data and on the determined blood flow characteristic, a second model having a lower level of detail than the first model, the second model representing at least part of the portion of the anatomical structure of the patient represented by the three-dimensional model. 45. The non-transitory computer readable medium of claim 45, wherein the second model reproduces, within a tolerance, the blood flow characteristic determined using the first model. 46. The non-transitory computer readable medium of claim 45, the method further including adjusting the second model to predict the results of a treatment option. 47. The non-transitory computer readable medium of claim 45, the method further including determining, using the at least one computer system, the blood flow characteristic within the anatomical structure of the patient using the second model. 48. The non-transitory computer readable medium of claim 47, the method further including:
comparing the blood flow characteristics determined using the first model with the blood flow characteristics determined using the second model; and determining whether to adjust the second model based on the comparison. 49. The non-transitory computer readable medium of claim 48, the method further including:
determining whether to adjust the second model based on whether differences between the blood flow characteristics determined using the first model and the blood flow characteristics determined using the second model are outside a tolerance. | Computational methods are used to create cardiovascular simulations having desired hemodynamic features. Cardiovascular modeling methods produce descriptions of blood flow and pressure in the heart and vascular networks. Numerical methods optimize and solve nonlinear equations to find parameter values that result in desired hemodynamic characteristics including related flow and pressure at various locations in the cardiovascular system, movements of soft tissues, and changes for different physiological states. The modeling methods employ simplified models to approximate the behavior of more complex models with the goal of to reducing computational expense. The user describes the desired features of the final cardiovascular simulation and provides minimal input, and the system automates the search for the final patient-specific cardiovascular model.1.-19. (canceled) 20. A system for determining cardiovascular information for a patient, the system comprising:
at least one computer system configured to:
receive patient-specific data regarding a geometry of an anatomical structure of the patient;
create, based on the patient-specific data, a three-dimensional model representing at least a portion of the anatomical structure of the patient, the three-dimensional model including portions representing at least one inlet and a plurality of outlets for blood flow;
create at least one boundary condition model representing blood flow through at least one of the at least one inlet or the plurality of outlets, the three-dimensional model and the at least one boundary condition model forming a first model;
determine a blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the first model; and
create, based on the patient-specific data and on the determined blood flow characteristic, a second model having a lower level of detail than the first model, the second model representing at least part of the portion of the anatomical structure of the patient represented by the three-dimensional model. 21. The system of claim 20, wherein the second model reproduces, within a tolerance, the blood flow characteristic determined using the first model. 22. The system of claim 20, wherein the at least one computer system is further configured to determine the blood flow characteristic within the anatomical structure of the patient using the second model. 23. The system of claim 22, wherein the at least one computer system is further configured to:
compare the blood flow characteristics determined using the first model with the blood flow characteristics determined using the second model; and determine whether to adjust the second model based on the comparison. 24. The system of claim 23, wherein the at least one computer system is further configured to determine whether to adjust the second model based on whether differences between the blood flow characteristics determined using the first model and the blood flow characteristics determined using the second model are outside a tolerance. 25. The system of claim 24, wherein the at least one computer system is further configured to adjust the second model by increasing the level of detail of the second model when the differences are outside the tolerance. 26. The system of claim 23, wherein the at least one computer system is further configured to adjust the second model by adjusting the level of detail of the second model. 27. The system of claim 20, wherein the at least one computer system is further configured to determine whether to adjust the second model to include a lower level of detail while reproducing, within a tolerance, the blood flow characteristic determined using the first model. 28. The system of claim 20, wherein:
the first model includes a three-dimensional mesh created based on the three-dimensional model; and the second model has less than three dimensions. 29. The system of claim 20, wherein:
the first model includes a three-dimensional mesh created based on the three-dimensional model; and the second model includes a mesh that is less refined than the three-dimensional mesh of the first model. 30. The system of claim 20, wherein the at least one computer system is further configured to determine the blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the second model, the first model being associated with a more complete polynomial basis for solving the equations governing blood flow than the second model. 31. The system of claim 20, wherein:
the first model represents flow in the at least one blood vessel using an elastance-based model of the patient's cardiac chamber; and the second model includes a prescribed flow in the at least one blood vessel in the anatomical structure of the patient. 32. The system of claim 20, wherein the at least one computer system is further configured to:
determine the blood flow characteristic by solving a system of differential equations with numerical methods for temporal integration using the first model; and determine the blood flow characteristic by solving a system of differential equations with Fourier analysis using the second model. 33. The system of claim 20, wherein the second model includes a model assuming vascular walls that are more rigid than the corresponding vascular walls of the first model. 34. The system of claim 20, wherein:
the first model includes a model assuming rigid vascular walls; and the second model includes a model assuming vascular walls that are less rigid than the corresponding vascular walls of the first model. 35. The system of claim 20, wherein:
the anatomical structure includes at least a portion of a plurality of coronary arteries emanating from the aorta; and the portion of the anatomical structure represented by the three-dimensional model includes at least a portion of the plurality of coronary arteries, the three-dimensional model representing at least one fluid flow inlet and at least one fluid flow outlet. 36. The system of claim 20, wherein the at least one computer system is further configured to adjust the second model to predict the results of a treatment option. 37. The system of claim 20, wherein the at least one computer system is further configured to use the second model to predict the results of a treatment option. 38. The system of claim 20, wherein the patient-specific data includes imaging data. 39. The system of claim 20, wherein the blood flow characteristic includes at least one of blood flow or pressure within the anatomical structure of the patient. 40. A method for determining cardiovascular information for a patient using at least one computer system, the method comprising:
inputting into the at least one computer system patient-specific data regarding a geometry of an anatomical structure of the patient; creating, using the at least one computer system and based on the patient-specific data, a three-dimensional model representing at least a portion of the anatomical structure of the patient, the three-dimensional model including portions representing at least one inlet and a plurality of outlets for blood flow; creating, using the at least one computer system, at least one boundary condition model representing blood flow through at least one of the at least one inlet or the plurality of outlets, the three-dimensional model and the at least one boundary condition model forming a first model; determining, using the at least one computer system, a blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the first model; and creating, using the at least one computer system and based on the patient-specific data and on the determined blood flow characteristic, a second model having a lower level of detail than the first model, the second model representing at least part of the portion of the anatomical structure of the patient represented by the three-dimensional model. 41. The method of claim 40, wherein the second model reproduces, within a tolerance, the blood flow characteristic determined using the first model. 42. The method of claim 40, further including adjusting, using the at least one computer system, the second model to predict the results of a treatment option. 43. The method of claim 40, further including determining, using the at least one computer system, the blood flow characteristic within the anatomical structure of the patient using the second model. 44. A non-transitory computer readable medium for use on at least one computer system containing computer-executable programming instructions for performing a method for determining patient-specific cardiovascular information, the method comprising:
receiving patient-specific data regarding a geometry of an anatomical structure of the patient; creating, based on the patient-specific data, a three-dimensional model representing at least a portion of the anatomical structure of the patient, the three-dimensional model including portions representing at least one inlet and a plurality of outlets for blood flow; creating at least one boundary condition model representing blood flow through at least one of the at least one inlet or the plurality of outlets, the three-dimensional model and the at least one boundary condition model forming a first model; determining a blood flow characteristic within the anatomical structure of the patient by solving equations governing blood flow using the first model; and creating, based on the patient-specific data and on the determined blood flow characteristic, a second model having a lower level of detail than the first model, the second model representing at least part of the portion of the anatomical structure of the patient represented by the three-dimensional model. 45. The non-transitory computer readable medium of claim 45, wherein the second model reproduces, within a tolerance, the blood flow characteristic determined using the first model. 46. The non-transitory computer readable medium of claim 45, the method further including adjusting the second model to predict the results of a treatment option. 47. The non-transitory computer readable medium of claim 45, the method further including determining, using the at least one computer system, the blood flow characteristic within the anatomical structure of the patient using the second model. 48. The non-transitory computer readable medium of claim 47, the method further including:
comparing the blood flow characteristics determined using the first model with the blood flow characteristics determined using the second model; and determining whether to adjust the second model based on the comparison. 49. The non-transitory computer readable medium of claim 48, the method further including:
determining whether to adjust the second model based on whether differences between the blood flow characteristics determined using the first model and the blood flow characteristics determined using the second model are outside a tolerance. | 1,600 |
650 | 15,753,938 | 1,632 | This disclosure provides, among other things, a transgenic animal and a method of using the same to make antibodies that have a common light chain. In certain embodiments, the transgenic animal may comprising a genome comprising a common light chain transgene, wherein the common light chain transgene comprises a non-immunoglobulin light-chain promoter and a common light-chain coding sequence. In certain embodiments, the common light chain is constitutively expressed. | 1. A transgenic animal comprising a genome comprising a common light chain transgene, wherein the common light chain transgene comprises a non-immunoglobulin light-chain promoter and a common light-chain coding sequence. 2. The transgenic animal of claim 1, wherein the common light chain is constitutively expressed. 3. The transgenic animal of claim 1, wherein the common light chain transgene is a recombinase-inducible common light chain transgene comprising:
i. the non-immunglobulin light-chain promoter; ii. a common light-chain coding sequence and iii. an intervening sequence that prevents the common light-chain from being expressed, unless the intervening sequence is excised by a recombinase. 4. The transgenic animal of claim 3, wherein the intervening sequence of the recombinase-inducible common light-chain transgene is excised when the recombinase is expressed. 5. The transgenic animal of claim 3, wherein the intervening sequence of the recombinase -inducible common light-chain transgene is flanked by recombination sites. 6. The transgenic animal of claim 3, wherein the intervening sequence comprises a selectable marker. 7. The transgenic animal of claim 6, wherein the selectable marker cassette contains a polyadenylation site that prevents downstream transcription. 8. The transgenic animal of claim 1, further comprising a recombinase transgene, wherein said recombinase transgene comprises a recombinase coding sequence operably linked to an immunoglobulin light-chain promoter. 9. The transgenic animal of claim 8, wherein the immunoglobulin light-chain promoter is an endogenous immunoglobulin light-chain promoter. 10. The transgenic animal of claim 8, wherein the recombinase transgene is a self-inactivating recombinase transgene. 11. The transgenic animal of claim 10, wherein the self-inactivating recombinase transgene comprises a recombinase coding sequence flanked by recombination sites. 12. The transgenic animal of claim 1, wherein the recombinase is Cre and the recombination sites are lox sites. 13. The transgenic animal of claim 8, wherein the recombinase transgene and the common light-chain transgene are genetically linked. 14. The transgenic animal of claim 13, wherein recombination between the recombinase -transgene and the common light-chain transgene occurs at a frequency of at most 2.5%. 15. The transgenic animal of claim 13, wherein the transgenic animal is heterozygous for the genetically linked transgenes. 16. The transgenic animal of claim 13, wherein the transgenic animal is homozygous for the genetically linked transgenes. 17. The transgenic animal of claim 1, wherein the common light-chain coding sequence encodes a common immunoglobulin light-chain comprising a pre-rearranged variable region. 18. The transgenic animal of claim 1, wherein the common light-chain coding sequence encodes a human variable region operably linked to an immunoglobulin light-chain constant region. 19. The transgenic animal of claim 1, wherein the transgenic animal is a transgenic bird. 20. The transgenic animal of claim 19, wherein the transgenic bird is a transgenic chicken. 21. The transgenic animal of claim 1, wherein the a common light chain transgene replaces at least part of an endogenous light chain immunoglobulin locus, including any pseudogenes present therein, of the genome of the animal. 22. A method of producing a hybrid animal comprising:
mating a transgenic animal that produces a common light-chain with a second transgenic animal comprising a genome comprising a synthetic heavy-chain knock-in; and collecting the progeny of the mating. 23. The method of claim 22, wherein the synthetic heavy-chain knock-in is a human heavy-chain knock-in. 24. A method for producing an antibody with a common light-chain, wherein said antibody is specific for an antigen, said method comprising:
a) immunizing a transgenic animal of claim 1 with said antigen; b) making hybridomas using cells of said immunized transgenic animal; c) screening said hybridomas to identify an antigen-specific hybridoma; and d) isolating an antigen-specific antibody from said antigen-specific hybridoma. 25. The method of claim 24, wherein the synthetic heavy-chain is a synthetic human heavy-chain. 26. The method of claim 24, wherein the transgenic animal is a transgenic chicken. | This disclosure provides, among other things, a transgenic animal and a method of using the same to make antibodies that have a common light chain. In certain embodiments, the transgenic animal may comprising a genome comprising a common light chain transgene, wherein the common light chain transgene comprises a non-immunoglobulin light-chain promoter and a common light-chain coding sequence. In certain embodiments, the common light chain is constitutively expressed.1. A transgenic animal comprising a genome comprising a common light chain transgene, wherein the common light chain transgene comprises a non-immunoglobulin light-chain promoter and a common light-chain coding sequence. 2. The transgenic animal of claim 1, wherein the common light chain is constitutively expressed. 3. The transgenic animal of claim 1, wherein the common light chain transgene is a recombinase-inducible common light chain transgene comprising:
i. the non-immunglobulin light-chain promoter; ii. a common light-chain coding sequence and iii. an intervening sequence that prevents the common light-chain from being expressed, unless the intervening sequence is excised by a recombinase. 4. The transgenic animal of claim 3, wherein the intervening sequence of the recombinase-inducible common light-chain transgene is excised when the recombinase is expressed. 5. The transgenic animal of claim 3, wherein the intervening sequence of the recombinase -inducible common light-chain transgene is flanked by recombination sites. 6. The transgenic animal of claim 3, wherein the intervening sequence comprises a selectable marker. 7. The transgenic animal of claim 6, wherein the selectable marker cassette contains a polyadenylation site that prevents downstream transcription. 8. The transgenic animal of claim 1, further comprising a recombinase transgene, wherein said recombinase transgene comprises a recombinase coding sequence operably linked to an immunoglobulin light-chain promoter. 9. The transgenic animal of claim 8, wherein the immunoglobulin light-chain promoter is an endogenous immunoglobulin light-chain promoter. 10. The transgenic animal of claim 8, wherein the recombinase transgene is a self-inactivating recombinase transgene. 11. The transgenic animal of claim 10, wherein the self-inactivating recombinase transgene comprises a recombinase coding sequence flanked by recombination sites. 12. The transgenic animal of claim 1, wherein the recombinase is Cre and the recombination sites are lox sites. 13. The transgenic animal of claim 8, wherein the recombinase transgene and the common light-chain transgene are genetically linked. 14. The transgenic animal of claim 13, wherein recombination between the recombinase -transgene and the common light-chain transgene occurs at a frequency of at most 2.5%. 15. The transgenic animal of claim 13, wherein the transgenic animal is heterozygous for the genetically linked transgenes. 16. The transgenic animal of claim 13, wherein the transgenic animal is homozygous for the genetically linked transgenes. 17. The transgenic animal of claim 1, wherein the common light-chain coding sequence encodes a common immunoglobulin light-chain comprising a pre-rearranged variable region. 18. The transgenic animal of claim 1, wherein the common light-chain coding sequence encodes a human variable region operably linked to an immunoglobulin light-chain constant region. 19. The transgenic animal of claim 1, wherein the transgenic animal is a transgenic bird. 20. The transgenic animal of claim 19, wherein the transgenic bird is a transgenic chicken. 21. The transgenic animal of claim 1, wherein the a common light chain transgene replaces at least part of an endogenous light chain immunoglobulin locus, including any pseudogenes present therein, of the genome of the animal. 22. A method of producing a hybrid animal comprising:
mating a transgenic animal that produces a common light-chain with a second transgenic animal comprising a genome comprising a synthetic heavy-chain knock-in; and collecting the progeny of the mating. 23. The method of claim 22, wherein the synthetic heavy-chain knock-in is a human heavy-chain knock-in. 24. A method for producing an antibody with a common light-chain, wherein said antibody is specific for an antigen, said method comprising:
a) immunizing a transgenic animal of claim 1 with said antigen; b) making hybridomas using cells of said immunized transgenic animal; c) screening said hybridomas to identify an antigen-specific hybridoma; and d) isolating an antigen-specific antibody from said antigen-specific hybridoma. 25. The method of claim 24, wherein the synthetic heavy-chain is a synthetic human heavy-chain. 26. The method of claim 24, wherein the transgenic animal is a transgenic chicken. | 1,600 |
651 | 12,910,508 | 1,615 | The present invention relates to methods of protecting patients and sensitizing cancer cells in combination with chemotherapy and/or radiation therapy. More specifically, the invention provides nutritional methods and formulations that are capable of reducing cancer growth without causing chronic weight loss in patients, protecting normal cells, tissues and organs from chemotherapy and/radiation therapy, and sensitizing cancer cells against low, normal and high-dose chemotherapy. Some of the methods also impede cancer growth even without chemotherapy. | 1. A method of alleviating cancer growth or a symptom of cancer, the method comprising:
a) identifying a patient with cancer; b) providing the patient with a first diet for a first predetermined period of time, the first diet providing the patient with at most 50% of the patient's normal caloric intake wherein at least 50% of the kilocalories are derived from fat; and c) providing the patient with a second diet for a second predetermined period of time, the second diet providing the patient with at most 500 kcal/day. 2. The method of claim 1 wherein the first predetermined period of time is about a day and the second predetermined period of time is about 3 days. 3. The method of claim 1 wherein the second diet provides the patient with at most 200 kcal/day. 4. The method of claim 1 wherein the first diet provides the patient with from 700 to 1200 kcal/day. 5. The method of claim 1 further comprising:
providing the patient with a third diet for a third predetermined period of time, the third diet comprising food items having a caloric content greater than 50% of the patient's normal caloric intake and a replenishing composition, the replenishing composition comprising essential amino acids. 6. The method of claim 5 wherein the third predetermined period of time is at least 5 days. 7. The method of claim 5 wherein the replenishing composition further comprises a component selected from the group consisting of vegetable extract, omega-3/6 essential fatty acids, protein and/or essential and non-essential amino acids, vitamins, minerals, and combinations thereof. 8. The method of claim 1, wherein the serum concentration of IGF-I in the patient is reduced by 25-90%. 9. The method of claim 1, wherein the blood glucose concentration in the patient is reduced by 25-75%. 10. A method of sensitizing cancer to chemotherapy drugs, the method comprising:
a) identifying a patient with cancer; and b) providing the patient with a first diet for a first predetermined period of time, the first diet providing the patient with at most 50% of the patient's normal caloric intake wherein at least 50% of the kilocalories are derived from fat; c) providing the patient with a second diet for a second predetermined period of time, the second diet providing the patient with at most 500 kcal/day; and d) administering to the patient a chemotherapy agent during or after the patient consumes the second diet for at least 48 hours. 11. The method of claim 10, wherein the chemotherapy agent is a DNA alkylating agent, oxidant, or topoisomerase inhibitor. 12. The method of claim 10, wherein the chemotherapy agent is methyl methanesulfonate, cyclophosphamide, etoposide, doxorubicin, or menadionecisplatin, carboplatin and other platinum based drugs, gemcitabine, docetaxel, 5-FU, or topoisomerase inhibitors 13. The method of claim 10, wherein the cancer is skin, colon, breast, esophageal, prostate, lung, uterus, ovary, and prostate cancer or glioma, melanoma, neuroblastoma, pheochromocytoma. 14. The method of claim 10 wherein the chemotherapy agent is provided for a longer period of time than is standard practice for the chemotherapy agent when the patient is not provided the first diet and the second diet. 15. The method of claim 10 wherein the first and second diet are administered during the chronic administration of the chemotherapy agent. 16. The method of claim 10 wherein the chemotherapy agent is provided for a shorter period of time than is standard practice for the chemotherapy agent when the patient is not provided the first diet and the second diet. 17. The method of claim 10 wherein the chemotherapy agent is administered in an amount that is at least 10% greater than the amount normally provided. 18. The method of claim 10 wherein the chemotherapy agent is administered in an amount that is at least 10% lower than the amount normally provided. 19. The method of claim 10 wherein the patient has exhibited a symptom of chemotherapeutic-related toxicity prior to step b). 20. The method of claim 10 wherein the patient has been diagnosed as terminal prior to step b). 21. The method of claim 10 wherein the first predetermined period of time is about a day and the second predetermined period of time is about 3 days. 22. The method of claim 10 wherein the second diet is at most 200 kcal/day. 23. The method of claim 10 further comprising:
providing the patient with a third diet for a third predetermined period of time, the third diet which supplements the patient's normal diet, comprises a replenishing composition, the replenishing composition comprising essential amino acids and other amino acids. 24. The method of claim 23 wherein the third predetermined period of time is at least 5 days. 25. The method of claim 23 wherein the replenishing composition further comprises a component selected from the group consisting of vegetable extract, omega-3/6 essential fatty acids, non-essential amino acids, minerals, and combinations thereof. 26. A method of sensitizing cancer to radiation therapy, the method comprising:
a) identifying a patient with cancer; and b) providing the patient with a first diet for a first predetermined period of time, the first diet providing the patient with at most 50% of the patient's normal caloric intake wherein at least 50% of the kilocalories are derived from fat; c) providing the patient with a second diet for a second predetermined period of time, the second diet providing the patient with at most 500 kcal/day; and d) administering to the patient radiation therapy during or after the patient consumes the second diet for at least 48 hours. 27. A therapeutic meal package for providing meals to a cancer patient that retards cancer growth and enhances the efficacy of chemotherapy drugs, the therapeutic meal package comprising:
first meal component portioned into meals that provide the cancer patient at most 50% of the patient's normal caloric intake with at least 50% of the kilocalories derived from fat, the first meal component being in a sufficient amount to provide meal for a first predetermined period of time; a second meal component portioned into meals that provide the cancer patient at most 500 kcal/day, the second meal component being in a sufficient amount to provide meals for a second predetermined period of time; a replenishing composition comprising essential amino acids and other non-essential amino acids, essential fatty acids, minerals, vitamins and/or vegetable extracts for a third predetermined period of time; and instructions for administering the first meal component and the second meal component to the cancer patient. 28. The therapeutic meal package of claim 27 wherein the first meal component, the second meal component, and the third meal component each independently include a component selected from the group consisting of vegetable extracts, minerals, omega-3/6 essential fatty acids, and combinations thereof. | The present invention relates to methods of protecting patients and sensitizing cancer cells in combination with chemotherapy and/or radiation therapy. More specifically, the invention provides nutritional methods and formulations that are capable of reducing cancer growth without causing chronic weight loss in patients, protecting normal cells, tissues and organs from chemotherapy and/radiation therapy, and sensitizing cancer cells against low, normal and high-dose chemotherapy. Some of the methods also impede cancer growth even without chemotherapy.1. A method of alleviating cancer growth or a symptom of cancer, the method comprising:
a) identifying a patient with cancer; b) providing the patient with a first diet for a first predetermined period of time, the first diet providing the patient with at most 50% of the patient's normal caloric intake wherein at least 50% of the kilocalories are derived from fat; and c) providing the patient with a second diet for a second predetermined period of time, the second diet providing the patient with at most 500 kcal/day. 2. The method of claim 1 wherein the first predetermined period of time is about a day and the second predetermined period of time is about 3 days. 3. The method of claim 1 wherein the second diet provides the patient with at most 200 kcal/day. 4. The method of claim 1 wherein the first diet provides the patient with from 700 to 1200 kcal/day. 5. The method of claim 1 further comprising:
providing the patient with a third diet for a third predetermined period of time, the third diet comprising food items having a caloric content greater than 50% of the patient's normal caloric intake and a replenishing composition, the replenishing composition comprising essential amino acids. 6. The method of claim 5 wherein the third predetermined period of time is at least 5 days. 7. The method of claim 5 wherein the replenishing composition further comprises a component selected from the group consisting of vegetable extract, omega-3/6 essential fatty acids, protein and/or essential and non-essential amino acids, vitamins, minerals, and combinations thereof. 8. The method of claim 1, wherein the serum concentration of IGF-I in the patient is reduced by 25-90%. 9. The method of claim 1, wherein the blood glucose concentration in the patient is reduced by 25-75%. 10. A method of sensitizing cancer to chemotherapy drugs, the method comprising:
a) identifying a patient with cancer; and b) providing the patient with a first diet for a first predetermined period of time, the first diet providing the patient with at most 50% of the patient's normal caloric intake wherein at least 50% of the kilocalories are derived from fat; c) providing the patient with a second diet for a second predetermined period of time, the second diet providing the patient with at most 500 kcal/day; and d) administering to the patient a chemotherapy agent during or after the patient consumes the second diet for at least 48 hours. 11. The method of claim 10, wherein the chemotherapy agent is a DNA alkylating agent, oxidant, or topoisomerase inhibitor. 12. The method of claim 10, wherein the chemotherapy agent is methyl methanesulfonate, cyclophosphamide, etoposide, doxorubicin, or menadionecisplatin, carboplatin and other platinum based drugs, gemcitabine, docetaxel, 5-FU, or topoisomerase inhibitors 13. The method of claim 10, wherein the cancer is skin, colon, breast, esophageal, prostate, lung, uterus, ovary, and prostate cancer or glioma, melanoma, neuroblastoma, pheochromocytoma. 14. The method of claim 10 wherein the chemotherapy agent is provided for a longer period of time than is standard practice for the chemotherapy agent when the patient is not provided the first diet and the second diet. 15. The method of claim 10 wherein the first and second diet are administered during the chronic administration of the chemotherapy agent. 16. The method of claim 10 wherein the chemotherapy agent is provided for a shorter period of time than is standard practice for the chemotherapy agent when the patient is not provided the first diet and the second diet. 17. The method of claim 10 wherein the chemotherapy agent is administered in an amount that is at least 10% greater than the amount normally provided. 18. The method of claim 10 wherein the chemotherapy agent is administered in an amount that is at least 10% lower than the amount normally provided. 19. The method of claim 10 wherein the patient has exhibited a symptom of chemotherapeutic-related toxicity prior to step b). 20. The method of claim 10 wherein the patient has been diagnosed as terminal prior to step b). 21. The method of claim 10 wherein the first predetermined period of time is about a day and the second predetermined period of time is about 3 days. 22. The method of claim 10 wherein the second diet is at most 200 kcal/day. 23. The method of claim 10 further comprising:
providing the patient with a third diet for a third predetermined period of time, the third diet which supplements the patient's normal diet, comprises a replenishing composition, the replenishing composition comprising essential amino acids and other amino acids. 24. The method of claim 23 wherein the third predetermined period of time is at least 5 days. 25. The method of claim 23 wherein the replenishing composition further comprises a component selected from the group consisting of vegetable extract, omega-3/6 essential fatty acids, non-essential amino acids, minerals, and combinations thereof. 26. A method of sensitizing cancer to radiation therapy, the method comprising:
a) identifying a patient with cancer; and b) providing the patient with a first diet for a first predetermined period of time, the first diet providing the patient with at most 50% of the patient's normal caloric intake wherein at least 50% of the kilocalories are derived from fat; c) providing the patient with a second diet for a second predetermined period of time, the second diet providing the patient with at most 500 kcal/day; and d) administering to the patient radiation therapy during or after the patient consumes the second diet for at least 48 hours. 27. A therapeutic meal package for providing meals to a cancer patient that retards cancer growth and enhances the efficacy of chemotherapy drugs, the therapeutic meal package comprising:
first meal component portioned into meals that provide the cancer patient at most 50% of the patient's normal caloric intake with at least 50% of the kilocalories derived from fat, the first meal component being in a sufficient amount to provide meal for a first predetermined period of time; a second meal component portioned into meals that provide the cancer patient at most 500 kcal/day, the second meal component being in a sufficient amount to provide meals for a second predetermined period of time; a replenishing composition comprising essential amino acids and other non-essential amino acids, essential fatty acids, minerals, vitamins and/or vegetable extracts for a third predetermined period of time; and instructions for administering the first meal component and the second meal component to the cancer patient. 28. The therapeutic meal package of claim 27 wherein the first meal component, the second meal component, and the third meal component each independently include a component selected from the group consisting of vegetable extracts, minerals, omega-3/6 essential fatty acids, and combinations thereof. | 1,600 |
652 | 14,529,359 | 1,619 | A liquid dispensing razor including a reservoir containing a shave care composition. The shave care composition comprises water; one or more lipophilic skin conditioning agents; one or more thickening agents including electrolyte sensitive polymers; one or more emulsifying agents; and one or more lubricants. The shave care composition is delivered to the skin by the razor to provide skin comfort and moisturization benefits during the shave. The combination of the chemistry in the razor results in step change in skin condition in consumer and technical skin moisturization benefits. | 1. A liquid dispensing razor including a reservoir containing a shave care composition, the shave care composition comprising:
water; one or more lipophilic skin conditioning agents; one or more thickening agents including electrolyte sensitive polymers; one or more emulsifying agents; and one or more lubricants. 2. The liquid dispensing razor of claim 1, wherein the composition comprises from about 1% to about 50% by weight of the one or more lipophilic skin conditioning agents. 3. The liquid dispensing razor of claim 1, wherein composition comprises from about 0.1% to about 5% by weight of the one or more thickening agents. 4. The liquid dispensing razor of claim 1, wherein composition comprises from about 0.1% to about 20% by weight of the one or more emulsifying agents. 5. The liquid dispensing razor of claim 1, wherein composition comprises from about 0.1% to about 8% by weight of the one or more lubricants. 6. The liquid dispensing razor of claim 1, further comprising salicylic acid. 7. The liquid dispensing razor of claim 6, wherein composition comprises from about 0.001% to about 5% by weight of the salicylic acid. 8. The liquid dispensing razor of claim 1 wherein the electrolyte sensitive polymer is selected from the group consisting of Polyacrylamide, Hydroxyethyl Acrylate/Sodium Acryloyldimethyltaurate Copolymer, Sodium Acrylate/Sodium Acryloyldimethyl Taurate Copolymer, Ammonium Polyacrylate, Sodium Acrylate/Acryloyldimethyltaurate/Dimethylacrylamide Crosspolymer, Hydroxyethyl Acrylate/Sodium Acryloyldimethyltaurate Copolymer, Carboxylic Acid Polymers, Carbopol 980 and ETD 2050 or Ammonium Acryloyldimethyltaurate/VP Copolymer, Sodium Acryloyldimethyltaurate/VP Copolymer, Ammonium Acryloyldimethyltaurate/Beheneth-25 Methacrylate Crosspolymer. 9. The liquid dispensing razor of claim 1 wherein the electrolyte sensitive polymer is Polyacrylamide. 10. The liquid dispensing razor of claim 1 wherein the lipophilic skin conditioning agent is comprised of pre-blended lipid materials. 11. The liquid dispensing razor of claim 10 where the pre-blend is composed of Petrolatum with another lipid oil chosen from mineral oil or a fatty acid ester. 12. The liquid dispensing razor according to claim 1 wherein the shave care composition has a Power-Law Index of less than about 0.5 and a Power-Law Consistency of less than about 50 Pa·s and greater than about 20 Pa·s. 13. The liquid dispensing razor according to claim 12 wherein the Power-Law Consistency is less than about 45 Pa·s and greater than about 25 Pa·s. 14. A liquid dispensing razor including a reservoir containing a shave care composition, the shave care composition comprising:
water; from about 1% to about 50% by weight one or more lipophilic skin conditioning agents; from about 0.1% to about 5% by weight one or more thickening agents; from about 0.1% to about 20% by weight one or more emulsifying agents; and from about 0.1% to about 8% by weight one or more lubricants. 15. The liquid dispensing razor of claim 14 wherein the composition comprises from about 5% to about 50% by weight of the one or more lipophilic skin conditioning agents. 16. The liquid dispensing razor of claim 14, wherein composition comprises from about 0.25% to about 3% by weight of the one or more thickening agents. 17. The liquid dispensing razor of claim 14, wherein composition comprises from about 1.0% to about 12% by weight of the one or more emulsifying agents. 18. The liquid dispensing razor of claim 14, wherein composition comprises from about 0.2% to about 3% by weight of the one or more lubricants. 19. The liquid dispensing razor according to claim 14 wherein the shave care composition has a Power-Law Index less than about 0.5 and a Power-Law Consistency of less than about 50 Pa·s and greater than about 20 Pa·s. 20. The liquid dispensing razor according to claim 19 wherein the Power-Law Consistency is less than about 45 Pa·s and greater than about 25 Pa·s. | A liquid dispensing razor including a reservoir containing a shave care composition. The shave care composition comprises water; one or more lipophilic skin conditioning agents; one or more thickening agents including electrolyte sensitive polymers; one or more emulsifying agents; and one or more lubricants. The shave care composition is delivered to the skin by the razor to provide skin comfort and moisturization benefits during the shave. The combination of the chemistry in the razor results in step change in skin condition in consumer and technical skin moisturization benefits.1. A liquid dispensing razor including a reservoir containing a shave care composition, the shave care composition comprising:
water; one or more lipophilic skin conditioning agents; one or more thickening agents including electrolyte sensitive polymers; one or more emulsifying agents; and one or more lubricants. 2. The liquid dispensing razor of claim 1, wherein the composition comprises from about 1% to about 50% by weight of the one or more lipophilic skin conditioning agents. 3. The liquid dispensing razor of claim 1, wherein composition comprises from about 0.1% to about 5% by weight of the one or more thickening agents. 4. The liquid dispensing razor of claim 1, wherein composition comprises from about 0.1% to about 20% by weight of the one or more emulsifying agents. 5. The liquid dispensing razor of claim 1, wherein composition comprises from about 0.1% to about 8% by weight of the one or more lubricants. 6. The liquid dispensing razor of claim 1, further comprising salicylic acid. 7. The liquid dispensing razor of claim 6, wherein composition comprises from about 0.001% to about 5% by weight of the salicylic acid. 8. The liquid dispensing razor of claim 1 wherein the electrolyte sensitive polymer is selected from the group consisting of Polyacrylamide, Hydroxyethyl Acrylate/Sodium Acryloyldimethyltaurate Copolymer, Sodium Acrylate/Sodium Acryloyldimethyl Taurate Copolymer, Ammonium Polyacrylate, Sodium Acrylate/Acryloyldimethyltaurate/Dimethylacrylamide Crosspolymer, Hydroxyethyl Acrylate/Sodium Acryloyldimethyltaurate Copolymer, Carboxylic Acid Polymers, Carbopol 980 and ETD 2050 or Ammonium Acryloyldimethyltaurate/VP Copolymer, Sodium Acryloyldimethyltaurate/VP Copolymer, Ammonium Acryloyldimethyltaurate/Beheneth-25 Methacrylate Crosspolymer. 9. The liquid dispensing razor of claim 1 wherein the electrolyte sensitive polymer is Polyacrylamide. 10. The liquid dispensing razor of claim 1 wherein the lipophilic skin conditioning agent is comprised of pre-blended lipid materials. 11. The liquid dispensing razor of claim 10 where the pre-blend is composed of Petrolatum with another lipid oil chosen from mineral oil or a fatty acid ester. 12. The liquid dispensing razor according to claim 1 wherein the shave care composition has a Power-Law Index of less than about 0.5 and a Power-Law Consistency of less than about 50 Pa·s and greater than about 20 Pa·s. 13. The liquid dispensing razor according to claim 12 wherein the Power-Law Consistency is less than about 45 Pa·s and greater than about 25 Pa·s. 14. A liquid dispensing razor including a reservoir containing a shave care composition, the shave care composition comprising:
water; from about 1% to about 50% by weight one or more lipophilic skin conditioning agents; from about 0.1% to about 5% by weight one or more thickening agents; from about 0.1% to about 20% by weight one or more emulsifying agents; and from about 0.1% to about 8% by weight one or more lubricants. 15. The liquid dispensing razor of claim 14 wherein the composition comprises from about 5% to about 50% by weight of the one or more lipophilic skin conditioning agents. 16. The liquid dispensing razor of claim 14, wherein composition comprises from about 0.25% to about 3% by weight of the one or more thickening agents. 17. The liquid dispensing razor of claim 14, wherein composition comprises from about 1.0% to about 12% by weight of the one or more emulsifying agents. 18. The liquid dispensing razor of claim 14, wherein composition comprises from about 0.2% to about 3% by weight of the one or more lubricants. 19. The liquid dispensing razor according to claim 14 wherein the shave care composition has a Power-Law Index less than about 0.5 and a Power-Law Consistency of less than about 50 Pa·s and greater than about 20 Pa·s. 20. The liquid dispensing razor according to claim 19 wherein the Power-Law Consistency is less than about 45 Pa·s and greater than about 25 Pa·s. | 1,600 |
653 | 14,786,456 | 1,626 | The present invention relates to a process for the preparation of a hydrogel suitable as carrier in a hydrogel-linked prodrug, to hydrogels obtainable from said process, the use of such hydrogel as a carrier in a hydrogel-linked prodrug and to hydrogel-linked prodrugs comprising a covalently conjugated hydrogel of the present invention. The hydrogel prodrug carrier has a reduced drug loading on the outside of the hydrogel carrier. This is achieved by reducing the number of functional groups of the hydrogel, in particular those at its surface. | 1. A process for the preparation of a hydrogel suitable as carrier in a hydrogel-linked prodrug comprising the steps of
(a) providing a hydrogel having groups Ax0, wherein groups Ax0 represent the same or different, preferably same, functional groups; (b) optionally covalently conjugating a spacer reagent of formula (I)
Ax1-SP2-Ax2 (I),
wherein
SP2 is C1-50 alkyl, C2-50 alkenyl or C2-50 alkynyl, which C1-50 alkyl, C2-50 alkenyl and C2-50 alkynyl is optionally interrupted by one or more group(s) selected from the group consisting of —NH—, —N(C1-4alkyl)-, —O—, —S, —C(O)—, —C(O)NH, —C(O)N(C1-4alkyl)-, —O—C(O)—, —S(O)—, —S(O)2—, 4- to 7-membered heterocyclyl, phenyl and naphthyl;
Ax1 is a functional group for reaction with Ax0 of the hydrogel; and
Ax2 is a functional group;
to Ax0 of the hydrogel from step (a); and (c) reacting the hydrogel of step (a) or step (b) with a reagent of formula (II)
Ax3-Z (II),
wherein
Ax3 is a functional group; and
Z is an inert moiety having a molecular weight ranging from 10 Da to 1000 kDa;
such that at most 99 mol-% of Ax or Ax2 react with Ax3. 2. The process of claim 1, wherein Ax0 is selected from the group consisting of maleimide, amine (—NH2 or —NH—), hydroxyl (—OH), thiol, carboxyl (—COOH) and activated carboxyl (—COY1, wherein Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4;
XH is Cl, Br, I, or F). 3. The process of claim 1 or 2, wherein the hydrogel of step (a) is obtainable by a process comprising the steps of:
(a-i) providing a mixture comprising
(a-ia) at least one backbone reagent, wherein the at least one backbone reagent has a molecular weight ranging from 1 to 100 kDa, and comprises at least three functional groups Ax0, wherein each Ax0 is a maleimide, amine (—NH2 or —NH—), hydroxyl (—OH), carboxyl (—COOH) or activated carboxyl (—COY1, wherein Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4
XH is Cl, Br, I, or F);
(a-ib) at least one crosslinker reagent, wherein the at least one crosslinker reagent has a molecular weight ranging from 0.2 to 40 kDa and comprises at least two functional end groups selected from the group consisting of activated ester groups, activated carbamate groups, activated carbonate groups, activated thiocarbonate groups, amine groups and thiol groups;
in a weight ratio of the at least one backbone reagent to the at least one crosslinker reagent ranging from 1:99 to 99:1 and wherein the molar ratio of Ax0 to functional end groups is >1;
(a-ii) polymerizing the mixture of step (a-i) to a hydrogel; and
(a-iii) optionally working-up the hydrogel of step (a-ii). 4. The process of any one of claim 1 to 3, wherein Ax0 is an amine and Ax1 is ClSO2—, R1(C═O)—, I—, Br—, Cl—, SCN—, CN—, O═C═N—, Y1—(C═O)—, Y1—(C═O)—NH—, or Y1—(C═O)—O—,
wherein
R1 is H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-8cycloalkyl, 4- to 7-membered heterocyclyl, 8- to 11-membered heterobicyclyl, phenyl, naphthyl, indenyl, indanyl, or tetralinyl; and
Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4,
XH is Cl, Br, I, or F. 5. The process of any one of claims 1 to 4, wherein Ax2 is selected from the group consisting of -maleimide, —SH, —NH2, —SeH, —N3, —C≡CH, —CR1═CR1aR1b, —OH, —(CH═X)—R, —(C═O)—S—R1, —(C═O)—H, —NH—NH2, —O—NH2, —Ar—X0, —Ar—Sn(R1)(R1a)(R1b), —Ar—B(OH)(OH), Br, I, Y1—(C═O)—, Y1—(C═O)—NH—, Y1—(C═O)—O—,
with optional protecting groups;
wherein
dashed lines indicate attachment to SP2;
X is O, S, or NH,
X0 is —OH, —NR1R1a, —SH, or —SeH,
XH is Cl, Br, I or F;
Ar is phenyl, naphthyl, indenyl, indanyl, or tetralinyl;
R1, R1a, R1b are independently of each other H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-8 cycloalkyl, 4- to 7-membered heterocyclyl, 8- to 11-membered heterobicyclyl, phenyl, naphthyl, indenyl, indanyl, or tetralinyl; and
Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4,
XH is Cl, Br, I, or F. 6. The process of any one of claims 1 to 5, wherein Ax3 is selected from the group consisting of —SH, —NH2, —SeH, -maleimide, —C≡CH, —N3, —CR1═CR1aR1b, —(C═X)—R1, —OH, —(C═O)—S—R1, —NH—NH2, —O—NH2, —Ar—Sn(R1)(R1a)(R1b), —Ar—B(OH)(OH), —Ar—X0,
wherein
dashed lines indicate attachment to Z;
X is O, S, or NH,
X0 is —OH, —NR1R1a, —SH, or —SeH;
R1, R1a, R1b are independently of each other H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-8cycloalkyl, 4- to 7-membered heterocyclyl, 8- to 11-membered heterobicyclyl, phenyl, naphthyl, indenyl, indanyl, or tetralinyl; and
Ar is phenyl, naphthyl, indenyl, indanyl, or tetralinyl.
Y1 is an activated carboxylic acid, activated carbonate or activated carbamate, preferably Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4,
XH is Cl, Br, I, or F. 7. The process of any one of claims 1 to 6, wherein Z is an inert polymer having a molecular weight ranging from 0.5 kDa to 1000 kDa. 8. The process of any one of claims 1 to 7, wherein Z is an inert polymer selected from the group consisting of 2-methacryloyl-oxyethyl phosphoyl cholins, poly(acrylic acids), poly(acrylates), poly(acrylamides), poly(alkyloxy) polymers, poly(amides), poly(amidoamines), poly(amino acids), poly(anhydrides), poly(aspartamides), poly(butyric acids), poly(glycolic acids), polybutylene terephthalates, poly(caprolactones), poly(carbonates), poly(cyanoacrylates), poly(dimethylacrylamides), poly(esters), poly(ethylenes), poly(ethyleneglycols), poly(ethylene oxides), poly(ethyl phosphates), poly(ethyloxazolines), poly(glycolic acids), poly(hydroxyethyl acrylates), poly(hydroxyethyl-oxazolines), poly(hydroxymethacrylates), poly(hydroxypropylmethacrylamides), poly(hydroxypropyl methacrylates), poly(hydroxypropyloxazolines), poly(iminocarbonates), poly(lactic acids), poly(lactic-co-glycolic acids), poly(methacrylamides), poly(methacrylates), poly(methyloxazolines), poly(organophosphazenes), poly(ortho esters), poly(oxazolines), poly(propylene glycols), poly(siloxanes), poly(urethanes), poly(vinyl alcohols), poly(vinyl amines), poly(vinylmethylethers), poly(vinylpyrrolidones), silicones, celluloses, carbomethyl celluloses, hydroxypropyl methylcelluloses, chitins, chitosans, dextrans, dextrins, gelatins, hyaluronic acids and derivatives, functionalized hyaluronic acids, mannans, pectins, rhamnogalacturonans, starches, hydroxyalkyl starches, hydroxyethyl starches and other carbohydrate-based polymers, xylans, and copolymers thereof. 9. The process of any one of claims 1 to 8, wherein Z is an inert linear or branched PEG-based polymer comprising at least 70% PEG. 10. The process of any one of claims 1 to 9, wherein the reagent of formula (II) is used in an amount of at most 0.99 chemical equivalents relative to Ax0 or Ax2. 11. The process of any one of claims 1 to 9, wherein in step (c) the reaction rate is monitored and the reaction is interrupted when at most 0.99 chemical equivalents relative to Ax0 or Ax2 have reacted. 12. The process of any one of claims 1 to 9, wherein no more than 20 mol-% of Ax0 or Ax2 react with Ax3. 13. A hydrogel obtainable from the process of any one of claims 1 to 12. 14. Use of the hydrogel of claim 13 as a carrier in a hydrogel-linked prodrug. 15. A hydrogel-linked prodrug comprising a covalently conjugated hydrogel of claim 13. | The present invention relates to a process for the preparation of a hydrogel suitable as carrier in a hydrogel-linked prodrug, to hydrogels obtainable from said process, the use of such hydrogel as a carrier in a hydrogel-linked prodrug and to hydrogel-linked prodrugs comprising a covalently conjugated hydrogel of the present invention. The hydrogel prodrug carrier has a reduced drug loading on the outside of the hydrogel carrier. This is achieved by reducing the number of functional groups of the hydrogel, in particular those at its surface.1. A process for the preparation of a hydrogel suitable as carrier in a hydrogel-linked prodrug comprising the steps of
(a) providing a hydrogel having groups Ax0, wherein groups Ax0 represent the same or different, preferably same, functional groups; (b) optionally covalently conjugating a spacer reagent of formula (I)
Ax1-SP2-Ax2 (I),
wherein
SP2 is C1-50 alkyl, C2-50 alkenyl or C2-50 alkynyl, which C1-50 alkyl, C2-50 alkenyl and C2-50 alkynyl is optionally interrupted by one or more group(s) selected from the group consisting of —NH—, —N(C1-4alkyl)-, —O—, —S, —C(O)—, —C(O)NH, —C(O)N(C1-4alkyl)-, —O—C(O)—, —S(O)—, —S(O)2—, 4- to 7-membered heterocyclyl, phenyl and naphthyl;
Ax1 is a functional group for reaction with Ax0 of the hydrogel; and
Ax2 is a functional group;
to Ax0 of the hydrogel from step (a); and (c) reacting the hydrogel of step (a) or step (b) with a reagent of formula (II)
Ax3-Z (II),
wherein
Ax3 is a functional group; and
Z is an inert moiety having a molecular weight ranging from 10 Da to 1000 kDa;
such that at most 99 mol-% of Ax or Ax2 react with Ax3. 2. The process of claim 1, wherein Ax0 is selected from the group consisting of maleimide, amine (—NH2 or —NH—), hydroxyl (—OH), thiol, carboxyl (—COOH) and activated carboxyl (—COY1, wherein Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4;
XH is Cl, Br, I, or F). 3. The process of claim 1 or 2, wherein the hydrogel of step (a) is obtainable by a process comprising the steps of:
(a-i) providing a mixture comprising
(a-ia) at least one backbone reagent, wherein the at least one backbone reagent has a molecular weight ranging from 1 to 100 kDa, and comprises at least three functional groups Ax0, wherein each Ax0 is a maleimide, amine (—NH2 or —NH—), hydroxyl (—OH), carboxyl (—COOH) or activated carboxyl (—COY1, wherein Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4
XH is Cl, Br, I, or F);
(a-ib) at least one crosslinker reagent, wherein the at least one crosslinker reagent has a molecular weight ranging from 0.2 to 40 kDa and comprises at least two functional end groups selected from the group consisting of activated ester groups, activated carbamate groups, activated carbonate groups, activated thiocarbonate groups, amine groups and thiol groups;
in a weight ratio of the at least one backbone reagent to the at least one crosslinker reagent ranging from 1:99 to 99:1 and wherein the molar ratio of Ax0 to functional end groups is >1;
(a-ii) polymerizing the mixture of step (a-i) to a hydrogel; and
(a-iii) optionally working-up the hydrogel of step (a-ii). 4. The process of any one of claim 1 to 3, wherein Ax0 is an amine and Ax1 is ClSO2—, R1(C═O)—, I—, Br—, Cl—, SCN—, CN—, O═C═N—, Y1—(C═O)—, Y1—(C═O)—NH—, or Y1—(C═O)—O—,
wherein
R1 is H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-8cycloalkyl, 4- to 7-membered heterocyclyl, 8- to 11-membered heterobicyclyl, phenyl, naphthyl, indenyl, indanyl, or tetralinyl; and
Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4,
XH is Cl, Br, I, or F. 5. The process of any one of claims 1 to 4, wherein Ax2 is selected from the group consisting of -maleimide, —SH, —NH2, —SeH, —N3, —C≡CH, —CR1═CR1aR1b, —OH, —(CH═X)—R, —(C═O)—S—R1, —(C═O)—H, —NH—NH2, —O—NH2, —Ar—X0, —Ar—Sn(R1)(R1a)(R1b), —Ar—B(OH)(OH), Br, I, Y1—(C═O)—, Y1—(C═O)—NH—, Y1—(C═O)—O—,
with optional protecting groups;
wherein
dashed lines indicate attachment to SP2;
X is O, S, or NH,
X0 is —OH, —NR1R1a, —SH, or —SeH,
XH is Cl, Br, I or F;
Ar is phenyl, naphthyl, indenyl, indanyl, or tetralinyl;
R1, R1a, R1b are independently of each other H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-8 cycloalkyl, 4- to 7-membered heterocyclyl, 8- to 11-membered heterobicyclyl, phenyl, naphthyl, indenyl, indanyl, or tetralinyl; and
Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4,
XH is Cl, Br, I, or F. 6. The process of any one of claims 1 to 5, wherein Ax3 is selected from the group consisting of —SH, —NH2, —SeH, -maleimide, —C≡CH, —N3, —CR1═CR1aR1b, —(C═X)—R1, —OH, —(C═O)—S—R1, —NH—NH2, —O—NH2, —Ar—Sn(R1)(R1a)(R1b), —Ar—B(OH)(OH), —Ar—X0,
wherein
dashed lines indicate attachment to Z;
X is O, S, or NH,
X0 is —OH, —NR1R1a, —SH, or —SeH;
R1, R1a, R1b are independently of each other H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-8cycloalkyl, 4- to 7-membered heterocyclyl, 8- to 11-membered heterobicyclyl, phenyl, naphthyl, indenyl, indanyl, or tetralinyl; and
Ar is phenyl, naphthyl, indenyl, indanyl, or tetralinyl.
Y1 is an activated carboxylic acid, activated carbonate or activated carbamate, preferably Y1 is selected from formulas (f-i) to (f-vi):
wherein
the dashed lines indicate attachment to the rest of the molecule,
b is 1, 2, 3 or 4,
XH is Cl, Br, I, or F. 7. The process of any one of claims 1 to 6, wherein Z is an inert polymer having a molecular weight ranging from 0.5 kDa to 1000 kDa. 8. The process of any one of claims 1 to 7, wherein Z is an inert polymer selected from the group consisting of 2-methacryloyl-oxyethyl phosphoyl cholins, poly(acrylic acids), poly(acrylates), poly(acrylamides), poly(alkyloxy) polymers, poly(amides), poly(amidoamines), poly(amino acids), poly(anhydrides), poly(aspartamides), poly(butyric acids), poly(glycolic acids), polybutylene terephthalates, poly(caprolactones), poly(carbonates), poly(cyanoacrylates), poly(dimethylacrylamides), poly(esters), poly(ethylenes), poly(ethyleneglycols), poly(ethylene oxides), poly(ethyl phosphates), poly(ethyloxazolines), poly(glycolic acids), poly(hydroxyethyl acrylates), poly(hydroxyethyl-oxazolines), poly(hydroxymethacrylates), poly(hydroxypropylmethacrylamides), poly(hydroxypropyl methacrylates), poly(hydroxypropyloxazolines), poly(iminocarbonates), poly(lactic acids), poly(lactic-co-glycolic acids), poly(methacrylamides), poly(methacrylates), poly(methyloxazolines), poly(organophosphazenes), poly(ortho esters), poly(oxazolines), poly(propylene glycols), poly(siloxanes), poly(urethanes), poly(vinyl alcohols), poly(vinyl amines), poly(vinylmethylethers), poly(vinylpyrrolidones), silicones, celluloses, carbomethyl celluloses, hydroxypropyl methylcelluloses, chitins, chitosans, dextrans, dextrins, gelatins, hyaluronic acids and derivatives, functionalized hyaluronic acids, mannans, pectins, rhamnogalacturonans, starches, hydroxyalkyl starches, hydroxyethyl starches and other carbohydrate-based polymers, xylans, and copolymers thereof. 9. The process of any one of claims 1 to 8, wherein Z is an inert linear or branched PEG-based polymer comprising at least 70% PEG. 10. The process of any one of claims 1 to 9, wherein the reagent of formula (II) is used in an amount of at most 0.99 chemical equivalents relative to Ax0 or Ax2. 11. The process of any one of claims 1 to 9, wherein in step (c) the reaction rate is monitored and the reaction is interrupted when at most 0.99 chemical equivalents relative to Ax0 or Ax2 have reacted. 12. The process of any one of claims 1 to 9, wherein no more than 20 mol-% of Ax0 or Ax2 react with Ax3. 13. A hydrogel obtainable from the process of any one of claims 1 to 12. 14. Use of the hydrogel of claim 13 as a carrier in a hydrogel-linked prodrug. 15. A hydrogel-linked prodrug comprising a covalently conjugated hydrogel of claim 13. | 1,600 |
654 | 10,708,724 | 1,662 | The present invention relates to a novel culture system for generating transformed corn plants from mature seeds. In particular, the invention relates to the use of plant hormones during germination to affect the culture response. Transgenic corn plants can then be easily produced. | 1. A method of obtaining transformable callus tissue comprising:
germinating a mature seed in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; and culturing the nodal section on a callus induction media to produce embryogenic callus suitable for transformation. 2. The method of claim 1 in which the auxin is picloram and the cytokinin is BAP. 3. The method of claim 4 in which the picloram concentration is between about 0.5 mg/L and about 20 mg/L. 4. The method of claim 4 in which the BAP concentration is between about 0.1 mg/L and about 10 mg/L. 5. The method of claim 1 in which the tissue culture media is solid. 6. The method of claim 1 in which the nodal section is obtained from the seedling between 3 and 30 days after germination. 7. The method of claim 6 in which the nodal section is obtained from the seedling between 7 and 10 days after germination. 8. The method of claim 1 further comprising the steps of:
transforming the callus with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; and
regenerating a transformed plant from the transformed callus containing the nucleic acid sequence. 9. A method of obtaining transformable callus tissue comprising:
germinating a zygotic embryo in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; and culturing the nodal section on a callus induction media to produce embryogenic callus suitable for transformation. 10. The method of claim 9 in which the auxin is picloram and the cytokinin is BAP. 11. The method of claim 10 in which the picloram concentration is between about 0.5 mg/L and about 20 mg/L and the BAP concentration is between about 0.1 mg/L and about 10 mg/L. 12. The method of claim 9 further comprising the steps of:
transforming the callus with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; and
regenerating a transformed plant from the transformed callus to obtain a plant containing the nucleic acid sequence. 13. A method of transforming monocotyledonous plants comprising:
germinating a zygotic embryo from a monocotyledonous plant in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section in a callus induction media to form an embryogenic callus culture; transforming the embryogenic callus culture with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; selecting transformed callus cells; and regenerating a transformed monocotyledonous plant from the transformed callus to obtain a plant containing the nucleic acid sequence. 14. The method of claim 13 in which the monocotyledonous plant is corn. 15. A method of transforming monocotyledonous plants comprising:
germinating a zygotic embryo from a monocotyledonous plant in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section in media to form a multiple bud culture; converting the multiple bud culture to an embryogenic callus culture in callus induction media; transforming the embryogenic callus culture with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; selecting transformed callus cells; and regenerating a transformed monocotyledonous plant from the transformed callus cells to obtain a plant containing the nucleic acid sequence. 16. A method of obtaining transformable callus tissue from a plant comprising:
priming a mature seed; germinating a mature seed in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section on callus induction media to produce embryogenic callus. 17. A method of transforming monocotyledonous plants comprising:
priming a mature seed; germinating the mature seed in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section on callus induction media to form an embryogenic callus culture; transforming the embryogenic callus culture with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; selecting transformed callus cells; and regenerating a transformed plant from the transformed callus to obtain a plant containing the nucleic acid sequence. | The present invention relates to a novel culture system for generating transformed corn plants from mature seeds. In particular, the invention relates to the use of plant hormones during germination to affect the culture response. Transgenic corn plants can then be easily produced.1. A method of obtaining transformable callus tissue comprising:
germinating a mature seed in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; and culturing the nodal section on a callus induction media to produce embryogenic callus suitable for transformation. 2. The method of claim 1 in which the auxin is picloram and the cytokinin is BAP. 3. The method of claim 4 in which the picloram concentration is between about 0.5 mg/L and about 20 mg/L. 4. The method of claim 4 in which the BAP concentration is between about 0.1 mg/L and about 10 mg/L. 5. The method of claim 1 in which the tissue culture media is solid. 6. The method of claim 1 in which the nodal section is obtained from the seedling between 3 and 30 days after germination. 7. The method of claim 6 in which the nodal section is obtained from the seedling between 7 and 10 days after germination. 8. The method of claim 1 further comprising the steps of:
transforming the callus with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; and
regenerating a transformed plant from the transformed callus containing the nucleic acid sequence. 9. A method of obtaining transformable callus tissue comprising:
germinating a zygotic embryo in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; and culturing the nodal section on a callus induction media to produce embryogenic callus suitable for transformation. 10. The method of claim 9 in which the auxin is picloram and the cytokinin is BAP. 11. The method of claim 10 in which the picloram concentration is between about 0.5 mg/L and about 20 mg/L and the BAP concentration is between about 0.1 mg/L and about 10 mg/L. 12. The method of claim 9 further comprising the steps of:
transforming the callus with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; and
regenerating a transformed plant from the transformed callus to obtain a plant containing the nucleic acid sequence. 13. A method of transforming monocotyledonous plants comprising:
germinating a zygotic embryo from a monocotyledonous plant in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section in a callus induction media to form an embryogenic callus culture; transforming the embryogenic callus culture with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; selecting transformed callus cells; and regenerating a transformed monocotyledonous plant from the transformed callus to obtain a plant containing the nucleic acid sequence. 14. The method of claim 13 in which the monocotyledonous plant is corn. 15. A method of transforming monocotyledonous plants comprising:
germinating a zygotic embryo from a monocotyledonous plant in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section in media to form a multiple bud culture; converting the multiple bud culture to an embryogenic callus culture in callus induction media; transforming the embryogenic callus culture with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; selecting transformed callus cells; and regenerating a transformed monocotyledonous plant from the transformed callus cells to obtain a plant containing the nucleic acid sequence. 16. A method of obtaining transformable callus tissue from a plant comprising:
priming a mature seed; germinating a mature seed in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section on callus induction media to produce embryogenic callus. 17. A method of transforming monocotyledonous plants comprising:
priming a mature seed; germinating the mature seed in tissue culture media containing an effective amount of an auxin and an effective amount of a cytokinin to produce a growing seedling containing a nodal section; isolating the nodal section from the seedling; culturing the nodal section on callus induction media to form an embryogenic callus culture; transforming the embryogenic callus culture with a nucleic acid sequence conferring a selected genetic trait to the transformed callus; selecting transformed callus cells; and regenerating a transformed plant from the transformed callus to obtain a plant containing the nucleic acid sequence. | 1,600 |
655 | 14,366,952 | 1,649 | The invention relates to methods for selectively quantifying A-beta aggregates, comprising the immobilization of anti-A-beta antibodies on a substrate, application of the sample to be tested onto the substrate, addition of probes labeled for detection, which mark these by specific binding to A-beta aggregates and detection of the marked aggregates. | 1.-27. (canceled) 28. A method for selectively quantifying and/or characterizing A-beta aggregates in a sample, which method comprises:
(a) applying a sample to be tested onto a substrate, (b) adding probes labeled for detection, which probes label A-beta aggregates by specifically binding to the aggregates, and (c) detecting labeled A-beta aggregates,
it being possible for (b) to be performed prior to (a). 29. The method of claim 28, wherein prior to (a) scavenger molecules are immobilized on the substrate. 30. The method of claim 28, wherein the sample is pretreated. 31. The method of claim 28, wherein glass substrate is employed. 32. The method of claim 28, wherein the substrate comprises a hydrophilic coating. 33. The method of claim 28, wherein the substrate is coated with dextran. 34. The method of claim 28, wherein scavenger molecules are covalently bound to the substrate or to a coating thereof. 35. The method of claim 34, wherein the scavenger molecules are labeled with a fluorescent dye. 36. The method of claim 34, wherein the scavenger molecules are anti-A-beta antibodies. 37. The method of claim 36, wherein the anti-A-beta antibodies specifically bind an epitope of the A-beta aggregate. 38. The method of claim 28, wherein A-beta peptide-specific probes are used. 39. The method of claim 28, wherein the probes are fluorescent dye-labeled anti-A-beta antibodies. 40. The method of claim 28, wherein two or more different probes are used. 41. The method of claim 28, wherein two or more probes with differently labeled fluorescent dyes are used. 42. The method of claim 28, wherein at least one probe is an anti-A-beta antibody which specifically binds to an N-terminal epitope of the A-beta peptide. 43. The method of claim 28, wherein (c) comprises spatial resolution fluorescence microscopy. 44. The method of claim 28, wherein (c) comprises one or more of confocal fluorescence microscopy, fluorescence correlation spectroscopy (FCS), optionally in combination with cross-correlation and single particle immunosolvent laser scanning assay, laser scanning microscopy (LSM), Wetfeld microscopy, TIRF microscopy, and the corresponding super resolution modifications STED, SIM, STORM and dSTORM. 45. The method of claim 44, wherein in (c) sufficient data points for enabling detection of a single aggregate against a background signal are collected. 46. The method of claim 45, wherein a number of read-out values corresponds to a number of spatially resolved events present. 47. The method of claim 28, wherein the sample comprises one or more of spinal fluid (CSF, cerebrospinal fluid), blood, urine. 48. The method of claim 28, wherein an internal or external standard is used for quantifying A-beta aggregates. 49. The method of claim 48, wherein a standard for quantifying A-beta aggregates comprises non-aggregating polymers constructed from polypeptide sequences which with respect to their sequence are identical in a sub-segment or exhibit homology of at least 50% across a sub-segment with endogenous proteins that cause a protein aggregation disease or an amyloid degeneration or protein misfolding disease. 50. A kit for selectively quantifying A-beta aggregates, wherein the kit comprises one or more of:
a glass substrate coated with a hydrophobic substance; a standard; scavenger molecules; a probe; a substrate comprising scavenger molecules; solutions; and a buffer. 51. A method for determining the effectiveness of an active substance and/or therapy for treating AD, wherein the method comprises performing the method of claim 28, comparing active substances and/or therapies with one another in terms of their effect on A-beta aggregate formation, and selecting active substances and/or therapies which exhibit lower A-beta aggregate formation compared with a control. 52. A method for deciding on the acceptance of an individual into a clinical study or test, wherein the method comprises quantifying and/or characterizing A-beta aggregates according to the method of claim 28 and comparing a measured value with a threshold value. 53. A probe, wherein the probe is at least one of A-beta aggregate-specific and A-beta oligomer-specific. 54. A method of using the probe of claim 53, wherein the method comprises specifically binding the probe to a defined A-beta aggregate or A-beta oligomer. | The invention relates to methods for selectively quantifying A-beta aggregates, comprising the immobilization of anti-A-beta antibodies on a substrate, application of the sample to be tested onto the substrate, addition of probes labeled for detection, which mark these by specific binding to A-beta aggregates and detection of the marked aggregates.1.-27. (canceled) 28. A method for selectively quantifying and/or characterizing A-beta aggregates in a sample, which method comprises:
(a) applying a sample to be tested onto a substrate, (b) adding probes labeled for detection, which probes label A-beta aggregates by specifically binding to the aggregates, and (c) detecting labeled A-beta aggregates,
it being possible for (b) to be performed prior to (a). 29. The method of claim 28, wherein prior to (a) scavenger molecules are immobilized on the substrate. 30. The method of claim 28, wherein the sample is pretreated. 31. The method of claim 28, wherein glass substrate is employed. 32. The method of claim 28, wherein the substrate comprises a hydrophilic coating. 33. The method of claim 28, wherein the substrate is coated with dextran. 34. The method of claim 28, wherein scavenger molecules are covalently bound to the substrate or to a coating thereof. 35. The method of claim 34, wherein the scavenger molecules are labeled with a fluorescent dye. 36. The method of claim 34, wherein the scavenger molecules are anti-A-beta antibodies. 37. The method of claim 36, wherein the anti-A-beta antibodies specifically bind an epitope of the A-beta aggregate. 38. The method of claim 28, wherein A-beta peptide-specific probes are used. 39. The method of claim 28, wherein the probes are fluorescent dye-labeled anti-A-beta antibodies. 40. The method of claim 28, wherein two or more different probes are used. 41. The method of claim 28, wherein two or more probes with differently labeled fluorescent dyes are used. 42. The method of claim 28, wherein at least one probe is an anti-A-beta antibody which specifically binds to an N-terminal epitope of the A-beta peptide. 43. The method of claim 28, wherein (c) comprises spatial resolution fluorescence microscopy. 44. The method of claim 28, wherein (c) comprises one or more of confocal fluorescence microscopy, fluorescence correlation spectroscopy (FCS), optionally in combination with cross-correlation and single particle immunosolvent laser scanning assay, laser scanning microscopy (LSM), Wetfeld microscopy, TIRF microscopy, and the corresponding super resolution modifications STED, SIM, STORM and dSTORM. 45. The method of claim 44, wherein in (c) sufficient data points for enabling detection of a single aggregate against a background signal are collected. 46. The method of claim 45, wherein a number of read-out values corresponds to a number of spatially resolved events present. 47. The method of claim 28, wherein the sample comprises one or more of spinal fluid (CSF, cerebrospinal fluid), blood, urine. 48. The method of claim 28, wherein an internal or external standard is used for quantifying A-beta aggregates. 49. The method of claim 48, wherein a standard for quantifying A-beta aggregates comprises non-aggregating polymers constructed from polypeptide sequences which with respect to their sequence are identical in a sub-segment or exhibit homology of at least 50% across a sub-segment with endogenous proteins that cause a protein aggregation disease or an amyloid degeneration or protein misfolding disease. 50. A kit for selectively quantifying A-beta aggregates, wherein the kit comprises one or more of:
a glass substrate coated with a hydrophobic substance; a standard; scavenger molecules; a probe; a substrate comprising scavenger molecules; solutions; and a buffer. 51. A method for determining the effectiveness of an active substance and/or therapy for treating AD, wherein the method comprises performing the method of claim 28, comparing active substances and/or therapies with one another in terms of their effect on A-beta aggregate formation, and selecting active substances and/or therapies which exhibit lower A-beta aggregate formation compared with a control. 52. A method for deciding on the acceptance of an individual into a clinical study or test, wherein the method comprises quantifying and/or characterizing A-beta aggregates according to the method of claim 28 and comparing a measured value with a threshold value. 53. A probe, wherein the probe is at least one of A-beta aggregate-specific and A-beta oligomer-specific. 54. A method of using the probe of claim 53, wherein the method comprises specifically binding the probe to a defined A-beta aggregate or A-beta oligomer. | 1,600 |
656 | 14,175,154 | 1,643 | Administering an amino acid-containing composition, containing the following amino acids (1)-(9), wherein
the molar content ratio of each amino acid to the total content of the amino acids (1)-(9) falls within the following numerical ranges: (1) leucine 35-66%; (2) isoleucine 5.0-15%; (3) valine 5.0-15%; (4) threonine 7.0-14%; (5) lysine 8.0-16%; (6) methionine 2.0-10%; (7) histidine 0.1-3.5%; (8) phenylalanine 2.5-8.0%; and (9) tryptophan 0.1-2.0%,
is effective for enhancing recovery from muscle fatigue. | 1. An amino acid-containing composition for enhancing recovery from muscle fatigue, comprising the following amino acids (1)-(9), wherein
the molar content ratio of each amino acid to the total content of said amino acids (1)-(9) falls within the following numerical ranges: (1) leucine 35-66%; (2) isoleucine 5.0-15%; (3) valine 5.0-15%; (4) threonine 7.0-14%; (5) lysine 8.0-16%; (6) methionine 2.0-10%; (7) histidine 0.1-3.5%; (8) phenylalanine 2.5-8.0%; and (9) tryptophan 0.1-2.0%. 2. The amino acid-containing composition according to claim 1, wherein the muscle fatigue is muscle fatigue caused by muscle damage. 3. The amino acid-containing composition according to claim 2, wherein the muscle damage is muscle damage after exercise. 4. The amino acid-containing composition according to claim 3, wherein the muscle damage after exercise is muscle damage after eccentric contraction exercise. 5. The amino acid-containing composition according to claim 1, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscular pain. 6. The amino acid-containing composition according to claim 1, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle weakness. 7. The amino acid-containing composition according to claim 1, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle damage. 8. A method of enhancing recovery from muscle fatigue, comprising administering to a subject in need thereof an effective amount of an amino acid-containing composition comprising the following amino acids (1)-(9), wherein
the molar content ratio of each amino acid to the total content of said amino acids (1)-(9) falls within the following numerical ranges: (1) leucine 35-66%; (2) isoleucine 5.0-15%; (3) valine 5.0-15%; (4) threonine 7.0-14%; (5) lysine 8.0-16%; (6) methionine 2.0-10%; (7) histidine 0.1-3.5%; (8) phenylalanine 2.5-8.0%; and (9) tryptophan 0.1-2.0%. 9. The method according to claim 8, wherein the muscle fatigue is muscle fatigue caused by muscle damage. 10. The method according to claim 9, wherein the muscle damage is muscle damage after exercise. 11. The method according to claim 10, wherein the muscle damage after exercise is muscle damage after eccentric contraction exercise. 12. The method according to claim 8, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscular pain. 13. The method according to claim 8, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle weakness. 14. The method according to claim 8, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle damage. | Administering an amino acid-containing composition, containing the following amino acids (1)-(9), wherein
the molar content ratio of each amino acid to the total content of the amino acids (1)-(9) falls within the following numerical ranges: (1) leucine 35-66%; (2) isoleucine 5.0-15%; (3) valine 5.0-15%; (4) threonine 7.0-14%; (5) lysine 8.0-16%; (6) methionine 2.0-10%; (7) histidine 0.1-3.5%; (8) phenylalanine 2.5-8.0%; and (9) tryptophan 0.1-2.0%,
is effective for enhancing recovery from muscle fatigue.1. An amino acid-containing composition for enhancing recovery from muscle fatigue, comprising the following amino acids (1)-(9), wherein
the molar content ratio of each amino acid to the total content of said amino acids (1)-(9) falls within the following numerical ranges: (1) leucine 35-66%; (2) isoleucine 5.0-15%; (3) valine 5.0-15%; (4) threonine 7.0-14%; (5) lysine 8.0-16%; (6) methionine 2.0-10%; (7) histidine 0.1-3.5%; (8) phenylalanine 2.5-8.0%; and (9) tryptophan 0.1-2.0%. 2. The amino acid-containing composition according to claim 1, wherein the muscle fatigue is muscle fatigue caused by muscle damage. 3. The amino acid-containing composition according to claim 2, wherein the muscle damage is muscle damage after exercise. 4. The amino acid-containing composition according to claim 3, wherein the muscle damage after exercise is muscle damage after eccentric contraction exercise. 5. The amino acid-containing composition according to claim 1, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscular pain. 6. The amino acid-containing composition according to claim 1, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle weakness. 7. The amino acid-containing composition according to claim 1, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle damage. 8. A method of enhancing recovery from muscle fatigue, comprising administering to a subject in need thereof an effective amount of an amino acid-containing composition comprising the following amino acids (1)-(9), wherein
the molar content ratio of each amino acid to the total content of said amino acids (1)-(9) falls within the following numerical ranges: (1) leucine 35-66%; (2) isoleucine 5.0-15%; (3) valine 5.0-15%; (4) threonine 7.0-14%; (5) lysine 8.0-16%; (6) methionine 2.0-10%; (7) histidine 0.1-3.5%; (8) phenylalanine 2.5-8.0%; and (9) tryptophan 0.1-2.0%. 9. The method according to claim 8, wherein the muscle fatigue is muscle fatigue caused by muscle damage. 10. The method according to claim 9, wherein the muscle damage is muscle damage after exercise. 11. The method according to claim 10, wherein the muscle damage after exercise is muscle damage after eccentric contraction exercise. 12. The method according to claim 8, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscular pain. 13. The method according to claim 8, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle weakness. 14. The method according to claim 8, wherein the enhanced recovery from muscle fatigue is prophylaxis and/or improvement of muscle damage. | 1,600 |
657 | 14,821,072 | 1,613 | Methods of treatment using cadotril compositions are disclosed. | 1. A method for treating a subject experiencing diarrhea, comprising administering to the subject a composition comprising racecadotril, at least one surfactant, and a lipid, wherein said racecadotril achieves a maximum plasma concentration (Cmax) greater than about 300 ng/ml in said subject. 2. The method of claim 1, wherein the subject has been diagnosed with inflammatory bowel disease. 3. The method of claim 1, wherein the subject has been diagnosed with irritable bowel syndrome. 4. The method of claim 1, wherein a maximum plamsa concentratin (Cmax) is achieved at about 1.5 to about 2.5 hours after ingestion. 5. The method of claim 1, wherein the racecadotril is maintained at a level above about 300 ng/ml for at least about 3.5 to about 5 hours after ingestion. 6. The method of claim 1, wherein the composition comprises an average droplet size selected from the group consisting of about 200 nm to about 15 nm, about 70 nm to about 20 nm, about 40 nm to about 20 nm, about 25 nm. 7. The method of claim 1, wherein the AUC v. reference ratio is selected from the group consisting of about 1.8 to about 3.5, about 2 to about 3.3, and about 3.3. 8. The method of claim 1, wherein the composition comprises about 8.0 wt. % to about 10.0 wt. % racecadotril, about 88 wt. % to about 91 wt. % surfactant, and about 1 wt. % to about 2 wt. % lipid, wherein each wt. % is based upon 100 ml of the composition. 9. The method of claim 1, wherein the composition comprises about 3.0 wt. % to about 7.0 wt. % racecadotril, about 40 wt. % to about 53 wt. % of surfactant in total, about 40 wt. % to about 53 wt. % lipid, wherein each wt. % is based upon 100 ml of the composition. | Methods of treatment using cadotril compositions are disclosed.1. A method for treating a subject experiencing diarrhea, comprising administering to the subject a composition comprising racecadotril, at least one surfactant, and a lipid, wherein said racecadotril achieves a maximum plasma concentration (Cmax) greater than about 300 ng/ml in said subject. 2. The method of claim 1, wherein the subject has been diagnosed with inflammatory bowel disease. 3. The method of claim 1, wherein the subject has been diagnosed with irritable bowel syndrome. 4. The method of claim 1, wherein a maximum plamsa concentratin (Cmax) is achieved at about 1.5 to about 2.5 hours after ingestion. 5. The method of claim 1, wherein the racecadotril is maintained at a level above about 300 ng/ml for at least about 3.5 to about 5 hours after ingestion. 6. The method of claim 1, wherein the composition comprises an average droplet size selected from the group consisting of about 200 nm to about 15 nm, about 70 nm to about 20 nm, about 40 nm to about 20 nm, about 25 nm. 7. The method of claim 1, wherein the AUC v. reference ratio is selected from the group consisting of about 1.8 to about 3.5, about 2 to about 3.3, and about 3.3. 8. The method of claim 1, wherein the composition comprises about 8.0 wt. % to about 10.0 wt. % racecadotril, about 88 wt. % to about 91 wt. % surfactant, and about 1 wt. % to about 2 wt. % lipid, wherein each wt. % is based upon 100 ml of the composition. 9. The method of claim 1, wherein the composition comprises about 3.0 wt. % to about 7.0 wt. % racecadotril, about 40 wt. % to about 53 wt. % of surfactant in total, about 40 wt. % to about 53 wt. % lipid, wherein each wt. % is based upon 100 ml of the composition. | 1,600 |
658 | 15,343,570 | 1,628 | The present invention provides methods of treating cancer using pyrimidine and pyridine compounds which are inhibitors of Bruton's tyrosine kinase (BTK). | 1. A method of treating or preventing cancer, comprising administering to a subject a therapeutically effective amount of a compound of formula I
wherein
X denotes CH or N,
R1 denotes NH2, CONH2 or H,
R2 denotes Hal, Ar1 or Het1,
R3 denotes NR5[C(R5)2]nHet2, NR5[C(R5)2]nCyc, Het2, O[C(R5)2]nAr2, NR5[C(R5)2]nAr2, O[C(R5)2]nHet2, NR5(CH2)pNR5R6, O(CH2)pNR5R6 or NR5(CH2)pCR7R8NR5R6,
R4 denotes H, CH3 or NH2,
R5 denotes H or alkyl having 1, 2, 3 or 4 C atoms,
R6 N(R5)2CH2CH═CHCONH, Het3CH2CH═CHCONH, CH2═CHCONH(CH2), Het4(CH2)nCOHet3-diyl-CH2CH═CHCONH, HC≡CCO, CH3C≡CCO, CH2═CH—CO, CH2═C(CH3)CONH, CH3CH═CHCONH(CH2), N≡CCR7R8CONH(CH2)n, Het4NH(CH2)pCOHet3-diyl-CH2CH═CHCONH, Het4(CH2)pCONH(CH2CH2O)p(CH2)pCOHet3-diyl-CH2CH═CHCONH, CH2═CHSO2, ACH═CHCO, CH3CH═CHCO, Het4(CH2)pCONH(CH2)pHet3-diyl-CH2CH═CHCONH, Ar3CH═CHSO2, CH2═CHSO2NH or N(R5)CH2CH═CHCO,
R7, R8 denote together alkylene having 2, 3, 4, or 5 C atoms,
Ar1 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, (CH2)nNH2, CONHAr3, (CH2)nNHCOA, O(CH2)nAr3, OCyc, A, COHet3, OA and/or OHet3 (CH2),
Ar2 denotes phenyl, naphthyl or pyridyl each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, OAr3, (CH2)nNH2, (CH2)nNHCOA and/or Het3,
Ar3 denotes phenyl, which is unsubstituted or mono-, di- or trisubstituted by OH, OA, Hal, CN and/or A,
Het1 denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3,
Het2 denotes a mono- or bicyclic saturated heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, Hal, COOH, OA, COA, COHet3, CycCO, SO2 and/or ═O,
Het3 denotes a monocyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
Het4 denotes a bi- or tricyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O,
Cyc denotes cyclic alkyl having 3, 4, 5 or 6 C atoms, which is unsubstituted, monosubstituted or disubstituted by R6 and/or OH and which may comprise a double bond,
A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F and/or Cl and/or in which one or two non-adjacent CH2 and/or CH-groups may be replaced by O, NH and/or by N,
Hal denotes F, Cl, Br or I,
n denotes 0, 1, 2, 3 or 4,
p denotes 1, 2, 3, 4, 5 or 6,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 2. The method according to claim 1 wherein
Het1 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, benzimidazolyl, benzotriazolyl, indolyl, benzo-1,3-dioxolyl, indazolyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, imidazolidinyl, azetidinyl, azepanyl, benzo-2,1,3-thiadiazolyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl, dihydropyridyl or di hydrobenzodioxinyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 3. The method according to claim 1 wherein
Het1 denotes pyrazolyl, pyridyl, pyrimidinyl, dihydropyridyl or dihydrobenzodioxinyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 4. The method according to claim 1 wherein
Het2 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, 2,7-diazaspiro[3.5]nonyl, 2,8-diazaspiro[4.5]decyl, 2,7-diazaspiro[4.4]nonyl, 3-azabicylo[3.1.0]hexyl, 2-azaspiro[3.3]heptyl, 6-azaspiro[3.4]octyl, 7-azaspiro[3.5]nonyl, 5-azaspiro[3.5]nonyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, tetrahydroimidazolyl, tetrahydropyrazolyl, tetrahydropyridyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, OA, COA, COHet3, CycCO, SO2 and/or ═O, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 5. The method according to claim 1 wherein
Het3 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl or dihydropyridyl, each of which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 6. The method according to claim 1 wherein
Het3 denotes piperidinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, dihydropyrrolyl, dihydropyrazolyl or dihydropyridyl, each of which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 7. The method according to claim 1 wherein
Het4 denotes hexahydrothieno[3,4-d]imidazolyl, benzo[c][1,2,5]oxadiazolyl or 5H-dipyrrolo[1,2-c:2′,1′-f][1,3,2]diazaborinin-4-ium-uidyl, each of which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 8. The method according to claim 1 wherein
X denotes CH or N,
R1 denotes NH2, CON H2 or H,
R2 denotes Hal, Ar1 or Het1,
R3 denotes NR5[C(R5)2]nHet2, NR5[C(R5)2]nCyc, Het2, O[C(R5)2]nAr2, NR5[C(R5)2]nAr2, O[C(R5)2]nHet2, NR5(CH2)pNR5R6, O(CH2)pNR5R6NR5(CH2)pCR7R8NR5R6,
R4 denotes H,
R5 denotes H or alkyl having 1, 2, 3 or 4 C atoms,
R6 N(R5)2CH2CH═CHCONH, Het3CH2CH═CHCONH, CH2═CHCONH(CH2), Het4(CH2)nCOHet3-diyl-CH2CH═CHCONH, HC≡CCO, CH3C≡CCO, CH2═CH—CO, CH2═C(CH3)CONH, CH3CH═CHCONH(CH2)n, N≡CCR7R8CONH(CH2)n, Het4NH(CH2)pCOHet3-diyl-CH2CH═CHCONH, Het4(CH2)pCONH(CH2CH2O)p(CH2)pCOHet3-diyl-CH2CH═CHCONH, CH2═CHSO2, ACH═CHCO, CH3CH═CHCO, Het4(CH2)pCONH(CH2)pHet3-diyl-CH2CH═CHCONH, Ar3CH═CHSO2, CH2═CHSO2NH or N(R5)CH2CH═CHCO,
R7, R8 denote together alkylene having 2, 3, 4, or 5 C atoms,
Ar1 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, (CH2)nNH2, CONHAr3, (CH2)nNHCOA, O(CH2)nAr3, OCyc, A, COHet3, OA and/or OHet3 (CH2),
Ar2 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, OAr3, (CH2)nNH2, (CH2)nNHCOA and/or Het3,
Ar3 denotes phenyl, which is unsubstituted or mono-, di- or trisubstituted by OH, OA, Hal, CN and/or A,
Het1 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, benzimidazolyl, benzotriazolyl, indolyl, benzo-1,3-dioxolyl, indazolyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, imidazolidinyl, azetidinyl, azepanyl, benzo-2,1,3-thiadiazolyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl, dihydropyridyl or di hydrobenzodioxinyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3,
Het2 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, 2,7-diazaspiro[3.5]nonyl, 2,8-diazaspiro[4.5]decyl, 2,7-diazaspiro[4.4]nonyl, 3-azabicylo[3.1.0]hexyl, 2-azaspiro[3.3]heptyl, 6-azaspiro[3.4]octyl, 7-azaspiro[3.5]nonyl, 5-azaspiro[3.5]nonyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, tetrahydroimidazolyl, tetrahydropyrazolyl, tetrahydropyridyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, OA, COA, COHet3, CycCO, SO2 and/or ═O,
Het3 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl or dihydropyridyl, each of which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
Het4 denotes hexahydrothieno[3,4-d]imidazolyl, benzo[c][1,2,5]oxadiazolyl or 5H-dipyrrolo[1,2-c:2′,1′-f][1,3,2]diazaborinin-4-ium-uidyl, each of which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O,
Cyc denotes cyclic alkyl having 3, 4, 5 or 6 C atoms, which is unsubstituted or monosubstituted by R6 and which may comprise a double bond,
A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F and/or Cl and/or in which one or two non-adjacent CH2 and/or CH-groups may be replaced by O, NH and/or by N,
Hal denotes F, Cl, Br or I,
n denotes 0, 1, 2, 3 or 4,
p denotes 1, 2, 3, 4, 5 or 6,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 9. The method according to claim 1, wherein the compound is formula (II):
wherein:
X is H or CH3 or NH2,
Y is H, Hal or is absent,
B is N or CH,
E is NH2 or H,
W is NR, O or a cyclic amine,
Z is, independently, CH2, CH3, CH2—CH2, CH—CH2, H, NH or is absent,
“linker” is (CH2)n, wherein: n is 1, 2 or 3 or an optionally substituted group selected from a phenyl ring, an aryl ring, heteroaryl ring, branched or unbranched alkyl group, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen, a 4-7 membered saturated or partially unsaturated heterocycle having 1-3 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms attached to a hetero saturated ring. Linkers may also be cycloalkanes optionally substituted by heteroatoms (independently selected from nitrogen, or oxygen), cycloalkanes optionally substituted with —NH or OH, fused or bridged rings or optionally substituted spirocyclic rings that optionally contain heteroatoms,
A is a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, and/or O atoms and 5, 6, 7, 8, 9, or 10 skeleton C atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, OH or OR,
Hal is F, Cl, Br or I,
R is independently hydrogen, oxygen or an optionally substituted group selected from C1-6 linear or cyclic aliphatic, benzyl, phenyl, a phenyl group optionally substituted with 1, 2 or 3 O atoms, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen or a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, O atoms and 5, 6, 7, or 8 C skeleton atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, A, OH, NH2, nitrile, and/or CH(Hal)3 or is an unbranched or branched linear alkyl having 1, 2, 3, 4, 5, 6, 7 or 8 C atoms, in which one or two CH2 groups may be replaced by an O atom and/or by an —NH—, —CO—, —NHCOO—, —NHCONH—, —CONH—, —NHCO— or —CH═CH-group, and in which 1-3H atoms may be replaced by Hal,
Rq is selected from —R, --A, halogen, —OR, —O(CH2)rOR, —R(NH), —NO2, —C(O)R, —CO2R, —C(O)N(R)2, —NRC(O)R, —NRC(O)NR2, —NRSO2R, or —N(R)2,
r is 1-4,
n is 0-4, and
Q is an electrophilic group. 10. The method according to claim 1, wherein the compound is selected from Table 2, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 11. The method according to claim 10, wherein the compound is selected from: N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl)pyrimidine-4,6-diamine (A250); and 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 12. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype. 13. A pharmaceutical composition comprising at least one compound of formula I or formula II:
wherein
X denotes CH or N,
R1 denotes NH2, CON H2 or H,
R2 denotes Hal, Ar1 or Het1,
R3 denotes NR5[C(R5)2]nHet2, NR5[C(R5)2]nCyc, Het2, O[C(R5)2]nAr2, NR5[C(R5)2]nAr2, O[C(R5)2]nHet2, NR5(CH2)pNR5R6, O(CH2)pNR5R6 or NR5(CH2)pCR7R8NR5R6,
R4 denotes H, CH3 or NH2,
R5 denotes H or alkyl having 1, 2, 3 or 4 C atoms,
R6 N(R5)2CH2CH═CHCONH, Het3CH2CH═CHCONH, CH2═CHCONH(CH2), Het4(CH2)nCOHet3-diyl-CH2CH═CHCONH, HC≡CCO, CH3C≡CCO, CH2═CH—CO, CH2═C(CH3)CONH, CH3CH═CHCONH(CH2)n, N≡CCR7R8CONH(CH2)n, Het4NH(CH2)pCOHet3-diyl-CH2CH═CHCONH, Het4(CH2)pCONH(CH2CH2O)p(CH2)pCOHet3-diyl-CH2CH═CHCONH, CH2═CHSO2, ACH═CHCO, CH3CH═CHCO, Het4(CH2)pCONH(CH2)pHet3-diyl-CH2CH═CHCONH, Ar3CH═CHSO2, CH2═CHSO2NH or N(R5)CH2CH═CHCO,
R7, R8 denote together alkylene having 2, 3, 4, or 5 C atoms,
Ar1 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, (CH2)nNH2, CONHAr3, (CH2)nNHCOA, O(CH2)nAr3, OCyc, A, COHet3, OA and/or OHet3 (CH2),
Ar2 denotes phenyl, naphthyl or pyridyl each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, OAr3, (CH2)nNH2, (CH2)nNHCOA and/or Het3,
Ar3 denotes phenyl, which is unsubstituted or mono-, di- or trisubstituted by OH, OA, Hal, CN and/or A,
Het1 denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3,
Het2 denotes a mono- or bicyclic saturated heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, Hal, COOH, OA, COA, COHet3, CycCO, SO2 and/or ═O,
Het3 denotes a monocyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
Het4 denotes a bi- or tricyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O,
Cyc denotes cyclic alkyl having 3, 4, 5 or 6 C atoms, which is unsubstituted, monosubstituted or disubstituted by R6 and/or OH and which may comprise a double bond,
A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F and/or Cl and/or in which one or two non-adjacent CH2 and/or CH-groups may be replaced by O, NH and/or by N,
Hal denotes F, Cl, Br or I,
n denotes 0, 1, 2, 3 or 4,
p denotes 1, 2, 3, 4, 5 or 6;
wherein:
X is H or CH3 or NH2,
Y is H, Hal or is absent,
B is N or CH,
E is NH2 or H,
W is NR, O or a cyclic amine,
Z is, independently, CH2, CH3, CH2—CH2, CH—CH2, H, NH or is absent,
“linker” is (CH2)n, wherein: n is 1, 2 or 3 or an optionally substituted group selected from a phenyl ring, an aryl ring, heteroaryl ring, branched or unbranched alkyl group, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen, a 4-7 membered saturated or partially unsaturated heterocycle having 1-3 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms attached to a hetero saturated ring. Linkers may also be cycloalkanes optionally substituted by heteroatoms (independently selected from nitrogen, or oxygen), cycloalkanes optionally substituted with —NH or OH, fused or bridged rings or optionally substituted spirocyclic rings that optionally contain heteroatoms,
A is a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, and/or O atoms and 5, 6, 7, 8, 9, or 10 skeleton C atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, OH or OR,
Hal is F, Cl, Br or I,
R is independently hydrogen, oxygen or an optionally substituted group selected from C1-6 linear or cyclic aliphatic, benzyl, phenyl, a phenyl group optionally substituted with 1, 2 or 3 O atoms, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen or a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, O atoms and 5, 6, 7, or 8 C skeleton atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, A, OH, NH2, nitrile, and/or CH(Hal)3 or is an unbranched or branched linear alkyl having 1, 2, 3, 4, 5, 6, 7 or 8 C atoms, in which one or two CH2 groups may be replaced by an O atom and/or by an —NH—, —CO—, —NHCOO—, —NHCONH—, —CONH—, —NHCO— or —CH═CH-group, and in which 1-3H atoms may be replaced by Hal,
Rq is selected from —R, --A, halogen, —OR, —O(CH2)rOR, —R(NH), —NO2, —C(O)R, —CO2R, —C(O)N(R)2, —NRC(O)R, —NRC(O)NR2, —NRSO2R, or —N(R)2,
r is 1-4,
n is 0-4, and
Q is an electrophilic group;
and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and optionally excipients and/or adjuvants. 14. The pharmaceutical composition according to claim 13 and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active ingredient. 15. The pharmaceutical composition according to claim 13, wherein the compound is selected from Table 2. 16. The pharmaceutical composition according to claim 13, wherein the compound is selected from: N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl) pyrimidine-4,6-diamine (A250); and 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225). 17. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype, and the compound is selected from N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl)pyrimidine-4,6-diamine (A250); and 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 18. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype, and the compound is N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl)pyrimidine-4,6-diamine (A250); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 19. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype, and the compound is 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. | The present invention provides methods of treating cancer using pyrimidine and pyridine compounds which are inhibitors of Bruton's tyrosine kinase (BTK).1. A method of treating or preventing cancer, comprising administering to a subject a therapeutically effective amount of a compound of formula I
wherein
X denotes CH or N,
R1 denotes NH2, CONH2 or H,
R2 denotes Hal, Ar1 or Het1,
R3 denotes NR5[C(R5)2]nHet2, NR5[C(R5)2]nCyc, Het2, O[C(R5)2]nAr2, NR5[C(R5)2]nAr2, O[C(R5)2]nHet2, NR5(CH2)pNR5R6, O(CH2)pNR5R6 or NR5(CH2)pCR7R8NR5R6,
R4 denotes H, CH3 or NH2,
R5 denotes H or alkyl having 1, 2, 3 or 4 C atoms,
R6 N(R5)2CH2CH═CHCONH, Het3CH2CH═CHCONH, CH2═CHCONH(CH2), Het4(CH2)nCOHet3-diyl-CH2CH═CHCONH, HC≡CCO, CH3C≡CCO, CH2═CH—CO, CH2═C(CH3)CONH, CH3CH═CHCONH(CH2), N≡CCR7R8CONH(CH2)n, Het4NH(CH2)pCOHet3-diyl-CH2CH═CHCONH, Het4(CH2)pCONH(CH2CH2O)p(CH2)pCOHet3-diyl-CH2CH═CHCONH, CH2═CHSO2, ACH═CHCO, CH3CH═CHCO, Het4(CH2)pCONH(CH2)pHet3-diyl-CH2CH═CHCONH, Ar3CH═CHSO2, CH2═CHSO2NH or N(R5)CH2CH═CHCO,
R7, R8 denote together alkylene having 2, 3, 4, or 5 C atoms,
Ar1 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, (CH2)nNH2, CONHAr3, (CH2)nNHCOA, O(CH2)nAr3, OCyc, A, COHet3, OA and/or OHet3 (CH2),
Ar2 denotes phenyl, naphthyl or pyridyl each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, OAr3, (CH2)nNH2, (CH2)nNHCOA and/or Het3,
Ar3 denotes phenyl, which is unsubstituted or mono-, di- or trisubstituted by OH, OA, Hal, CN and/or A,
Het1 denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3,
Het2 denotes a mono- or bicyclic saturated heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, Hal, COOH, OA, COA, COHet3, CycCO, SO2 and/or ═O,
Het3 denotes a monocyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
Het4 denotes a bi- or tricyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O,
Cyc denotes cyclic alkyl having 3, 4, 5 or 6 C atoms, which is unsubstituted, monosubstituted or disubstituted by R6 and/or OH and which may comprise a double bond,
A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F and/or Cl and/or in which one or two non-adjacent CH2 and/or CH-groups may be replaced by O, NH and/or by N,
Hal denotes F, Cl, Br or I,
n denotes 0, 1, 2, 3 or 4,
p denotes 1, 2, 3, 4, 5 or 6,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 2. The method according to claim 1 wherein
Het1 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, benzimidazolyl, benzotriazolyl, indolyl, benzo-1,3-dioxolyl, indazolyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, imidazolidinyl, azetidinyl, azepanyl, benzo-2,1,3-thiadiazolyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl, dihydropyridyl or di hydrobenzodioxinyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 3. The method according to claim 1 wherein
Het1 denotes pyrazolyl, pyridyl, pyrimidinyl, dihydropyridyl or dihydrobenzodioxinyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 4. The method according to claim 1 wherein
Het2 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, 2,7-diazaspiro[3.5]nonyl, 2,8-diazaspiro[4.5]decyl, 2,7-diazaspiro[4.4]nonyl, 3-azabicylo[3.1.0]hexyl, 2-azaspiro[3.3]heptyl, 6-azaspiro[3.4]octyl, 7-azaspiro[3.5]nonyl, 5-azaspiro[3.5]nonyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, tetrahydroimidazolyl, tetrahydropyrazolyl, tetrahydropyridyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, OA, COA, COHet3, CycCO, SO2 and/or ═O, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 5. The method according to claim 1 wherein
Het3 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl or dihydropyridyl, each of which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 6. The method according to claim 1 wherein
Het3 denotes piperidinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, dihydropyrrolyl, dihydropyrazolyl or dihydropyridyl, each of which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 7. The method according to claim 1 wherein
Het4 denotes hexahydrothieno[3,4-d]imidazolyl, benzo[c][1,2,5]oxadiazolyl or 5H-dipyrrolo[1,2-c:2′,1′-f][1,3,2]diazaborinin-4-ium-uidyl, each of which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 8. The method according to claim 1 wherein
X denotes CH or N,
R1 denotes NH2, CON H2 or H,
R2 denotes Hal, Ar1 or Het1,
R3 denotes NR5[C(R5)2]nHet2, NR5[C(R5)2]nCyc, Het2, O[C(R5)2]nAr2, NR5[C(R5)2]nAr2, O[C(R5)2]nHet2, NR5(CH2)pNR5R6, O(CH2)pNR5R6NR5(CH2)pCR7R8NR5R6,
R4 denotes H,
R5 denotes H or alkyl having 1, 2, 3 or 4 C atoms,
R6 N(R5)2CH2CH═CHCONH, Het3CH2CH═CHCONH, CH2═CHCONH(CH2), Het4(CH2)nCOHet3-diyl-CH2CH═CHCONH, HC≡CCO, CH3C≡CCO, CH2═CH—CO, CH2═C(CH3)CONH, CH3CH═CHCONH(CH2)n, N≡CCR7R8CONH(CH2)n, Het4NH(CH2)pCOHet3-diyl-CH2CH═CHCONH, Het4(CH2)pCONH(CH2CH2O)p(CH2)pCOHet3-diyl-CH2CH═CHCONH, CH2═CHSO2, ACH═CHCO, CH3CH═CHCO, Het4(CH2)pCONH(CH2)pHet3-diyl-CH2CH═CHCONH, Ar3CH═CHSO2, CH2═CHSO2NH or N(R5)CH2CH═CHCO,
R7, R8 denote together alkylene having 2, 3, 4, or 5 C atoms,
Ar1 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, (CH2)nNH2, CONHAr3, (CH2)nNHCOA, O(CH2)nAr3, OCyc, A, COHet3, OA and/or OHet3 (CH2),
Ar2 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, OAr3, (CH2)nNH2, (CH2)nNHCOA and/or Het3,
Ar3 denotes phenyl, which is unsubstituted or mono-, di- or trisubstituted by OH, OA, Hal, CN and/or A,
Het1 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, benzimidazolyl, benzotriazolyl, indolyl, benzo-1,3-dioxolyl, indazolyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, imidazolidinyl, azetidinyl, azepanyl, benzo-2,1,3-thiadiazolyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl, dihydropyridyl or di hydrobenzodioxinyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3,
Het2 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, azabicyclo[3.2.1]octyl, azabicyclo[2.2.2]octyl, 2,7-diazaspiro[3.5]nonyl, 2,8-diazaspiro[4.5]decyl, 2,7-diazaspiro[4.4]nonyl, 3-azabicylo[3.1.0]hexyl, 2-azaspiro[3.3]heptyl, 6-azaspiro[3.4]octyl, 7-azaspiro[3.5]nonyl, 5-azaspiro[3.5]nonyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, tetrahydroimidazolyl, tetrahydropyrazolyl, tetrahydropyridyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, OA, COA, COHet3, CycCO, SO2 and/or ═O,
Het3 denotes piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, imidazolidinyl, azetidinyl, azepanyl, tetrahydrofuryl, dioxolanyl, tetrahydrothienyl, dihydropyrrolyl, tetrahydroimidazolyl, dihydropyrazolyl, tetrahydropyrazolyl, tetrahydropyridyl or dihydropyridyl, each of which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
Het4 denotes hexahydrothieno[3,4-d]imidazolyl, benzo[c][1,2,5]oxadiazolyl or 5H-dipyrrolo[1,2-c:2′,1′-f][1,3,2]diazaborinin-4-ium-uidyl, each of which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O,
Cyc denotes cyclic alkyl having 3, 4, 5 or 6 C atoms, which is unsubstituted or monosubstituted by R6 and which may comprise a double bond,
A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F and/or Cl and/or in which one or two non-adjacent CH2 and/or CH-groups may be replaced by O, NH and/or by N,
Hal denotes F, Cl, Br or I,
n denotes 0, 1, 2, 3 or 4,
p denotes 1, 2, 3, 4, 5 or 6,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 9. The method according to claim 1, wherein the compound is formula (II):
wherein:
X is H or CH3 or NH2,
Y is H, Hal or is absent,
B is N or CH,
E is NH2 or H,
W is NR, O or a cyclic amine,
Z is, independently, CH2, CH3, CH2—CH2, CH—CH2, H, NH or is absent,
“linker” is (CH2)n, wherein: n is 1, 2 or 3 or an optionally substituted group selected from a phenyl ring, an aryl ring, heteroaryl ring, branched or unbranched alkyl group, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen, a 4-7 membered saturated or partially unsaturated heterocycle having 1-3 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms attached to a hetero saturated ring. Linkers may also be cycloalkanes optionally substituted by heteroatoms (independently selected from nitrogen, or oxygen), cycloalkanes optionally substituted with —NH or OH, fused or bridged rings or optionally substituted spirocyclic rings that optionally contain heteroatoms,
A is a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, and/or O atoms and 5, 6, 7, 8, 9, or 10 skeleton C atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, OH or OR,
Hal is F, Cl, Br or I,
R is independently hydrogen, oxygen or an optionally substituted group selected from C1-6 linear or cyclic aliphatic, benzyl, phenyl, a phenyl group optionally substituted with 1, 2 or 3 O atoms, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen or a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, O atoms and 5, 6, 7, or 8 C skeleton atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, A, OH, NH2, nitrile, and/or CH(Hal)3 or is an unbranched or branched linear alkyl having 1, 2, 3, 4, 5, 6, 7 or 8 C atoms, in which one or two CH2 groups may be replaced by an O atom and/or by an —NH—, —CO—, —NHCOO—, —NHCONH—, —CONH—, —NHCO— or —CH═CH-group, and in which 1-3H atoms may be replaced by Hal,
Rq is selected from —R, --A, halogen, —OR, —O(CH2)rOR, —R(NH), —NO2, —C(O)R, —CO2R, —C(O)N(R)2, —NRC(O)R, —NRC(O)NR2, —NRSO2R, or —N(R)2,
r is 1-4,
n is 0-4, and
Q is an electrophilic group. 10. The method according to claim 1, wherein the compound is selected from Table 2, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 11. The method according to claim 10, wherein the compound is selected from: N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl)pyrimidine-4,6-diamine (A250); and 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 12. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype. 13. A pharmaceutical composition comprising at least one compound of formula I or formula II:
wherein
X denotes CH or N,
R1 denotes NH2, CON H2 or H,
R2 denotes Hal, Ar1 or Het1,
R3 denotes NR5[C(R5)2]nHet2, NR5[C(R5)2]nCyc, Het2, O[C(R5)2]nAr2, NR5[C(R5)2]nAr2, O[C(R5)2]nHet2, NR5(CH2)pNR5R6, O(CH2)pNR5R6 or NR5(CH2)pCR7R8NR5R6,
R4 denotes H, CH3 or NH2,
R5 denotes H or alkyl having 1, 2, 3 or 4 C atoms,
R6 N(R5)2CH2CH═CHCONH, Het3CH2CH═CHCONH, CH2═CHCONH(CH2), Het4(CH2)nCOHet3-diyl-CH2CH═CHCONH, HC≡CCO, CH3C≡CCO, CH2═CH—CO, CH2═C(CH3)CONH, CH3CH═CHCONH(CH2)n, N≡CCR7R8CONH(CH2)n, Het4NH(CH2)pCOHet3-diyl-CH2CH═CHCONH, Het4(CH2)pCONH(CH2CH2O)p(CH2)pCOHet3-diyl-CH2CH═CHCONH, CH2═CHSO2, ACH═CHCO, CH3CH═CHCO, Het4(CH2)pCONH(CH2)pHet3-diyl-CH2CH═CHCONH, Ar3CH═CHSO2, CH2═CHSO2NH or N(R5)CH2CH═CHCO,
R7, R8 denote together alkylene having 2, 3, 4, or 5 C atoms,
Ar1 denotes phenyl or naphthyl, each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, (CH2)nNH2, CONHAr3, (CH2)nNHCOA, O(CH2)nAr3, OCyc, A, COHet3, OA and/or OHet3 (CH2),
Ar2 denotes phenyl, naphthyl or pyridyl each of which is unsubstituted or mono-, di- or trisubstituted by R6, Hal, OAr3, (CH2)nNH2, (CH2)nNHCOA and/or Het3,
Ar3 denotes phenyl, which is unsubstituted or mono-, di- or trisubstituted by OH, OA, Hal, CN and/or A,
Het1 denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, O(CH2)nAr3 and/or (CH2)nAr3,
Het2 denotes a mono- or bicyclic saturated heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by R6, Het3, CycSO2, OH, Hal, COOH, OA, COA, COHet3, CycCO, SO2 and/or ═O,
Het3 denotes a monocyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di- or trisubstituted by Hal, A and/or ═O,
Het4 denotes a bi- or tricyclic unsaturated, saturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms, which may be unsubstituted or mono-, di-, tri- or tetrasubstituted by A, NO2, Hal and/or ═O,
Cyc denotes cyclic alkyl having 3, 4, 5 or 6 C atoms, which is unsubstituted, monosubstituted or disubstituted by R6 and/or OH and which may comprise a double bond,
A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F and/or Cl and/or in which one or two non-adjacent CH2 and/or CH-groups may be replaced by O, NH and/or by N,
Hal denotes F, Cl, Br or I,
n denotes 0, 1, 2, 3 or 4,
p denotes 1, 2, 3, 4, 5 or 6;
wherein:
X is H or CH3 or NH2,
Y is H, Hal or is absent,
B is N or CH,
E is NH2 or H,
W is NR, O or a cyclic amine,
Z is, independently, CH2, CH3, CH2—CH2, CH—CH2, H, NH or is absent,
“linker” is (CH2)n, wherein: n is 1, 2 or 3 or an optionally substituted group selected from a phenyl ring, an aryl ring, heteroaryl ring, branched or unbranched alkyl group, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen, a 4-7 membered saturated or partially unsaturated heterocycle having 1-3 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, or oxygen, or a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms attached to a hetero saturated ring. Linkers may also be cycloalkanes optionally substituted by heteroatoms (independently selected from nitrogen, or oxygen), cycloalkanes optionally substituted with —NH or OH, fused or bridged rings or optionally substituted spirocyclic rings that optionally contain heteroatoms,
A is a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, and/or O atoms and 5, 6, 7, 8, 9, or 10 skeleton C atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, OH or OR,
Hal is F, Cl, Br or I,
R is independently hydrogen, oxygen or an optionally substituted group selected from C1-6 linear or cyclic aliphatic, benzyl, phenyl, a phenyl group optionally substituted with 1, 2 or 3 O atoms, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, or oxygen or a mono- or bicyclic aromatic homo- or heterocycle having 0, 1, 2, 3 or 4 N, O atoms and 5, 6, 7, or 8 C skeleton atoms, which may be unsubstituted or, independently of one another, mono-, di- or trisubstituted by Hal, A, OH, NH2, nitrile, and/or CH(Hal)3 or is an unbranched or branched linear alkyl having 1, 2, 3, 4, 5, 6, 7 or 8 C atoms, in which one or two CH2 groups may be replaced by an O atom and/or by an —NH—, —CO—, —NHCOO—, —NHCONH—, —CONH—, —NHCO— or —CH═CH-group, and in which 1-3H atoms may be replaced by Hal,
Rq is selected from —R, --A, halogen, —OR, —O(CH2)rOR, —R(NH), —NO2, —C(O)R, —CO2R, —C(O)N(R)2, —NRC(O)R, —NRC(O)NR2, —NRSO2R, or —N(R)2,
r is 1-4,
n is 0-4, and
Q is an electrophilic group;
and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and optionally excipients and/or adjuvants. 14. The pharmaceutical composition according to claim 13 and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active ingredient. 15. The pharmaceutical composition according to claim 13, wherein the compound is selected from Table 2. 16. The pharmaceutical composition according to claim 13, wherein the compound is selected from: N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl) pyrimidine-4,6-diamine (A250); and 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225). 17. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype, and the compound is selected from N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl)pyrimidine-4,6-diamine (A250); and 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 18. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype, and the compound is N-[(1-acryloylpiperidin-4-yl)methyl]-5-(4-phenoxyphenyl)pyrimidine-4,6-diamine (A250); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. 19. The method according to claim 1, wherein the cancer is selected from Non-Hodgkin's Lymphoma mantle cell lymphoma and Non-Hodgkins's Lymphoma diffuse large b-cell lymphoma, including abc subtype, and the compound is 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)-4-fluoropiperidin-1-yl)prop-2-en-1-one (A225); and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. | 1,600 |
659 | 15,475,114 | 1,616 | The method for preparing metal nanoparticles includes preparing an extract of myrrh and mixing the extract with an aqueous solution including a metal salt. The mixture changes in color from light yellow to dark brown upon formation of nanoparticles. The extract of myrrh can be a water extract prepared by, for example, soaking a quantity of myrrh in water, filtering the soaked myrrh to obtain a filtered product, and then centrifuging the filtered product. The metal salt can be, for example, silver nitrate (AgNO 3 ). The metal nanoparticles can be spherical, spheroidal, elongated spherical, rod, and/or faceted. The metal nanoparticles can be used to treat Leishmaniasis (lesions) caused by Leishmania major. | 1. A method of preparing metal nanoparticles from myrrh comprising:
preparing an extract of myrrh gum; providing an aqueous solution including a metal salt; and stirring the myrrh gum extract with the aqueous solution of the metal salt solution for about 15 to 20 minutes at a temperature of about 50° C. to about 100° C. to produce the metal nanoparticles. 2. The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the extract and the aqueous solution are mixed at a temperature of about 60° C. 3 The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the metal salt is silver nitrate (AgNO3) and the metal nanoparticles are silver nanoparticles. 4. The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the metal nanoparticles have a mean diameter in the range of from about 5 nm to about 50 nm. 5. The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the metal nanoparticles are spherical, spheroidal, elongated spherical, rod-shaped, and/or faceted. 6. A method of treating epidermal lesions caused by Leishmaniasis comprising:
mixing an extract of myrrh with an aqueous solution of silver nitrate (AgNO3) to form silver nanoparticles in aqueous myrrh suspension, the silver nanoparticles having a mean size of about 5 nm to about 50 nm; and topically applying an effective amount of the silver nanoparticles in aqueous myrrh suspension to a patient in need thereof. 7. The method of treating epidermal lesions caused by leishmaniasis according to claim 6, wherein the leishmaniasis is caused by Leishmania major. 8. The method of treating epidermal lesions caused by leishmaniasis according to claim 6, wherein the lesions are subcutaneous epidermal lesions. 9. The method of treating epidermal lesions caused by leishmaniasis according to claim 6, wherein the metal nanoparticles are silver nanoparticles. 10. The method of treating epidermal lesions caused by leishmaniasis according to claim 9, wherein a concentration of the silver nanoparticles is between about 10−3 to 10−5 moles/L. 11. The method of treating epidermal lesions caused by leishmaniasis according to claim 10, wherein a concentration of the silver nanoparticles is between about 10−3 to 10−4 moles/L. | The method for preparing metal nanoparticles includes preparing an extract of myrrh and mixing the extract with an aqueous solution including a metal salt. The mixture changes in color from light yellow to dark brown upon formation of nanoparticles. The extract of myrrh can be a water extract prepared by, for example, soaking a quantity of myrrh in water, filtering the soaked myrrh to obtain a filtered product, and then centrifuging the filtered product. The metal salt can be, for example, silver nitrate (AgNO 3 ). The metal nanoparticles can be spherical, spheroidal, elongated spherical, rod, and/or faceted. The metal nanoparticles can be used to treat Leishmaniasis (lesions) caused by Leishmania major.1. A method of preparing metal nanoparticles from myrrh comprising:
preparing an extract of myrrh gum; providing an aqueous solution including a metal salt; and stirring the myrrh gum extract with the aqueous solution of the metal salt solution for about 15 to 20 minutes at a temperature of about 50° C. to about 100° C. to produce the metal nanoparticles. 2. The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the extract and the aqueous solution are mixed at a temperature of about 60° C. 3 The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the metal salt is silver nitrate (AgNO3) and the metal nanoparticles are silver nanoparticles. 4. The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the metal nanoparticles have a mean diameter in the range of from about 5 nm to about 50 nm. 5. The method of preparing metal nanoparticles from myrrh according to claim 1, wherein the metal nanoparticles are spherical, spheroidal, elongated spherical, rod-shaped, and/or faceted. 6. A method of treating epidermal lesions caused by Leishmaniasis comprising:
mixing an extract of myrrh with an aqueous solution of silver nitrate (AgNO3) to form silver nanoparticles in aqueous myrrh suspension, the silver nanoparticles having a mean size of about 5 nm to about 50 nm; and topically applying an effective amount of the silver nanoparticles in aqueous myrrh suspension to a patient in need thereof. 7. The method of treating epidermal lesions caused by leishmaniasis according to claim 6, wherein the leishmaniasis is caused by Leishmania major. 8. The method of treating epidermal lesions caused by leishmaniasis according to claim 6, wherein the lesions are subcutaneous epidermal lesions. 9. The method of treating epidermal lesions caused by leishmaniasis according to claim 6, wherein the metal nanoparticles are silver nanoparticles. 10. The method of treating epidermal lesions caused by leishmaniasis according to claim 9, wherein a concentration of the silver nanoparticles is between about 10−3 to 10−5 moles/L. 11. The method of treating epidermal lesions caused by leishmaniasis according to claim 10, wherein a concentration of the silver nanoparticles is between about 10−3 to 10−4 moles/L. | 1,600 |
660 | 12,937,818 | 1,618 | Pearlescent pigments including a largely transparent platelet-shaped substrate having a density ρ S and at least one optically active coating having a density ρ M , the substrate having an average size d 50 of 3 to 8 μm and an average height h S of 40 to 110 nm. The disclosure further relates to a method for producing the pearlescent pigments, and also to the use thereof. | 1. Pearlescent pigments comprising a largely transparent platelet-shaped substrate having a density ρS and at least one optically active coating having a density ρM,
wherein the substrate has an average size d50 of 3 to 8 μm and an average height hS of 40 to 110 nm. 2. The pearlescent pigments of claim 1,
wherein the average height hS is 40 nm to less than 100 nm. 3. The pearlescent pigments of claim 1,
wherein the standard deviation in the height hS is 25% to 80%. 4. The pearlescent pigments of claim 1,
wherein the pearlescent pigments have a size distribution with a d90 value of 7.0 to 11.0 μm. 5. The pearlescent pigments of claim 1,
wherein the optically active coating is a high-index coating. 6. The pearlescent pigments of claim 1,
wherein the optically active layer comprises at least one of a metal oxide layer, a metal hydroxide layer and a metal oxide hydrate layer. 7. The pearlescent pigments of claim 6,
wherein the metal oxide layer comprises at least one metal oxide selected from the group consisting of titanium oxide, iron oxide, cerium oxide, chromium oxide, tin oxide, zirconium oxide, cobalt oxide, and mixtures thereof. 8. The pearlescent pigments of claim 6,
wherein the layer thickness of the metal oxide layer is between 10 and 300 nm. 9. The pearlescent pigments of claim 1,
wherein the average substrate height hS is determined by the following formula:
h
S
=
4
d
M
3
3
r
S
2
+
π
d
M
2
r
S
+
2
·
d
M
ρ
S
ρ
M
·
(
100
c
M
-
1
)
-
(
d
M
r
S
)
2
-
2
·
d
M
r
S
where dM is the average layer thickness of the optically active layer, with dM being 40 to 180 nm,
rS=d50/2 is the average radius of the substrate,
ρS is the density of the substrate and ρM is the density of the optically active coating and cM is the weight fraction in percent of the optically active coating, based on the total weight of substrate and optically active coating. 10. The pearlescent pigments of claim 7,
wherein the metal oxide layer consists of TiO2 and the substrate consists of mica, and the relationship between the TiO2 content in % by weight, based on the total weight of TiO2 and mica, and the average layer thickness of the TiO2 coating is selected from the group consisting of a TiO2 content of 47.5-62% by weight with an average TiO2 layer thickness of above 20 to 30 nm; a TiO2 content of 58-74% by weight with an average TiO2 layer thickness of above 35 to 45 nm; a TiO2 content of 63-78% by weight with an average TiO2 layer thickness of above 45 to 55 nm; a TiO2 content of 67-82% by weight with an average TiO2 layer thickness of above 55 to 65 nm; a TiO2 content of 70-85% by weight with an average TiO2 layer thickness of above 65 to 75 nm; a TiO2 content of 73.5-86.5% by weight with an average TiO2 layer thickness of above 75 to 85 nm; a TiO2 content of 75-88% by weight with an average TiO2 layer thickness of above 85 to 95 nm; a TiO2 content of 76.5-89% by weight with an average TiO2 layer thickness of above 95 to 105 nm; a TiO2 content of 78.5-90% by weight with an average TiO2 layer thickness of above 105 to 115 nm; a TiO2 content of 80-91% by weight with an average TiO2 layer thickness of above 115 to 125 nm; a TiO2 content of 81.5-92% by weight with an average TiO2 layer thickness of above 125 to 135 nm; a TiO2 content of 83-92.5% by weight with an average TiO2 layer thickness of above 135 to 145 nm; a TiO2 content of 84-93% by weight with an average TiO2 layer thickness of above 145 to 155 nm; a TiO2 content of 85-93% by weight with an average TiO2 layer thickness of above 155 to 165 nm; a TiO2 content of 86-93.5% by weight with an average TiO2 layer thickness of above 165 to 175 nm; a TiO2 content of 87-94% by weight with an average TiO2 layer thickness of above 175 to 185 nm; a TiO2 content of 87.5-94% by weight with an average TiO2 layer thickness of above 185 to 195 nm; a TiO2 content of 88-94.5% by weight with an average TiO2 layer thickness of above 195 to 205 nm; and a TiO2 content of 89-95% by weight with an average TiO2 layer thickness of above 205 to 215 nm. 11. The pearlescent pigments of claim 7,
wherein the metal oxide layer consists of TiO2 (largely in the rutile modification), and the substrate consists of mica, and the relationship between the TiO2 content in % by weight, based on the total weight of TiO2 and mica, and the average layer thickness of the TiO2 coating is selected from the group consisting of a TiO2 content of 50.5-62% by weight with an average TiO2 layer thickness of above 20 to 30 nm; a TiO2 content of 61-74% by weight with an average TiO2 layer thickness of above 35 to 45 nm; a TiO2 content of 65.5-78% by weight with an average TiO2 layer thickness of above 45 to 55 nm; a TiO2 content of 69.5-82% by weight with an average TiO2 layer thickness of above 55 to 65 nm; a TiO2 content of 72.5-85% by weight with an average TiO2 layer thickness of above 65 to 75 nm; a TiO2 content of 75-86.5% by weight with an average TiO2 layer thickness of above 75 to 85 nm; a TiO2 content of 77.5-88% by weight with an average TiO2 layer thickness of above 85 to 95 nm; a TiO2 content of 79-89% by weight with an average TiO2 layer thickness of above 95 to 105 nm; a TiO2 content of 80.5-90% by weight with an average TiO2 layer thickness of above 105 to 115 nm; a TiO2 content of 82-91% by weight with an average TiO2 layer thickness of above 115 to 125 nm; a TiO2 content of 83-92% by weight with an average TiO2 layer thickness of above 125 to 135 nm; a TiO2 content of 84.5-92.5% by weight with an average TiO2 layer thickness of above 135 to 145 nm; a TiO2 content of 85.5-93% by weight with an average TiO2 layer thickness of above 145 to 155 nm; a TiO2 content of 86.5-93% by weight with an average TiO2 layer thickness of above 155 to 165 nm; a TiO2 content of 87-93.5% by weight with an average TiO2 layer thickness of above 165 to 175 nm; a TiO2 content of 88-94% by weight with an average TiO2 layer thickness of above 175 to 185 nm; a TiO2 content of 88.5-94% by weight with an average TiO2 layer thickness of above 185 to 195 nm; a TiO2 content of 89-94.5% by weight with an average TiO2 layer thickness of above 195 to 205 nm; and a TiO2 content of 89.5-95% by weight with an average TiO2 layer thickness of above 205 to 215 nm. 12. The pearlescent pigments of claim 1,
wherein the transparent platelet-shaped substrate is selected from the group consisting of mica, synthetic mica, glass flakes, SiO2 platelets, Al2O3 platelets, and mixtures thereof. 13. The pearlescent pigments of claim 12,
wherein the largely transparent platelet-shaped substrate consists of mica, glass flakes or mixtures thereof. 14. The pearlescent pigments of claim 1,
wherein the pearlescent pigments have at least one further layer having a refractive index <2.0. 15. The pearlescent pigments of claim 1,
wherein the pearlescent pigments have at least one further protective layer on the optically active layer. 16. The pearlescent pigments of claim 15,
wherein the at least one further protective layer comprises at least one metal oxide layer whose metal oxides are selected from the group consisting of SiO2, Al2O3, cerium oxide, mixtures thereof and combinations thereof. 17. A method for producing the pearlescent pigments of claim 1,
wherein the method comprises the following steps: a) classifying the largely transparent substrate, to give a substrate having an average height hS of 40 to 110 nm; and b) coating the classified substrate with an optically active layer, to form a pearlescent pigment having an average size d50 of 3 to 8 μm. 18. A method for producing a material selected from the group consisting of paints, printing inks, cosmetics, plastics, glass, enamel and ceramic wherein the method comprises adding to said material the pearlescent pigments of claim 1. 19. The method of claim 18,
wherein the cosmetics are selected from the group consisting of concealer sticks, body powder, face powder, compact and loose powder, face makeup, powder cream, cream makeup, emulsion makeup, wax makeup, foundation, mousse makeup, blusher, eye makeup, lipcare sticks, lipsticks, lip gloss, lip liners, hair-styling compositions, hair-setting products, hair mousse, hair gel, hair wax, hair mascara, permanent and semipermanent hair colors, temporary hair colors, skincare compositions, and nail varnish compositions. 20. A method for producing soft-focus pigment comprising forming a pearlescent pigment by a method according to claim 17. 21. A coating composition comprising one of the pearlescent pigments of claim 1. 22. A method for determining the average layer thickness hS of a platelet-shaped substrate of an effect pigment comprising a platelet-shaped substrate having a density ρS and at least one optically active coating having a density ρM,
in accordance with the following formula:
h
S
=
4
d
M
3
3
r
S
2
+
π
d
M
2
r
S
+
2
·
d
M
ρ
S
ρ
M
·
(
100
c
M
-
1
)
-
(
d
M
r
S
)
2
-
2
·
d
M
r
S
where dM is the ascertained average layer thickness of at least one of a high-index layer and of a semitransparent metal coating,
rS is the ascertained average radius of the substrate,
ρS is the ascertained density of the substrate and ρM is the ascertained density of the optically active coating and cM is the ascertained weight fraction in percent of the optically active coating, based on the total weight of substrate and optically active coating. 23. The pearlescent pigments of claim 5, wherein the high-index coating has a refractive index nM>2.0. 24. The pearlescent pigments of claim 10, wherein the TiO2 is in the rutile modification. 25. The pearlescent pigments of claim 15, wherein the at least one further protective layer is a high index layer. 26. The method of claim 17, wherein the optically active layer has a high index. 27. The coating composition of claim 21, wherein the composition is a cosmetic product. | Pearlescent pigments including a largely transparent platelet-shaped substrate having a density ρ S and at least one optically active coating having a density ρ M , the substrate having an average size d 50 of 3 to 8 μm and an average height h S of 40 to 110 nm. The disclosure further relates to a method for producing the pearlescent pigments, and also to the use thereof.1. Pearlescent pigments comprising a largely transparent platelet-shaped substrate having a density ρS and at least one optically active coating having a density ρM,
wherein the substrate has an average size d50 of 3 to 8 μm and an average height hS of 40 to 110 nm. 2. The pearlescent pigments of claim 1,
wherein the average height hS is 40 nm to less than 100 nm. 3. The pearlescent pigments of claim 1,
wherein the standard deviation in the height hS is 25% to 80%. 4. The pearlescent pigments of claim 1,
wherein the pearlescent pigments have a size distribution with a d90 value of 7.0 to 11.0 μm. 5. The pearlescent pigments of claim 1,
wherein the optically active coating is a high-index coating. 6. The pearlescent pigments of claim 1,
wherein the optically active layer comprises at least one of a metal oxide layer, a metal hydroxide layer and a metal oxide hydrate layer. 7. The pearlescent pigments of claim 6,
wherein the metal oxide layer comprises at least one metal oxide selected from the group consisting of titanium oxide, iron oxide, cerium oxide, chromium oxide, tin oxide, zirconium oxide, cobalt oxide, and mixtures thereof. 8. The pearlescent pigments of claim 6,
wherein the layer thickness of the metal oxide layer is between 10 and 300 nm. 9. The pearlescent pigments of claim 1,
wherein the average substrate height hS is determined by the following formula:
h
S
=
4
d
M
3
3
r
S
2
+
π
d
M
2
r
S
+
2
·
d
M
ρ
S
ρ
M
·
(
100
c
M
-
1
)
-
(
d
M
r
S
)
2
-
2
·
d
M
r
S
where dM is the average layer thickness of the optically active layer, with dM being 40 to 180 nm,
rS=d50/2 is the average radius of the substrate,
ρS is the density of the substrate and ρM is the density of the optically active coating and cM is the weight fraction in percent of the optically active coating, based on the total weight of substrate and optically active coating. 10. The pearlescent pigments of claim 7,
wherein the metal oxide layer consists of TiO2 and the substrate consists of mica, and the relationship between the TiO2 content in % by weight, based on the total weight of TiO2 and mica, and the average layer thickness of the TiO2 coating is selected from the group consisting of a TiO2 content of 47.5-62% by weight with an average TiO2 layer thickness of above 20 to 30 nm; a TiO2 content of 58-74% by weight with an average TiO2 layer thickness of above 35 to 45 nm; a TiO2 content of 63-78% by weight with an average TiO2 layer thickness of above 45 to 55 nm; a TiO2 content of 67-82% by weight with an average TiO2 layer thickness of above 55 to 65 nm; a TiO2 content of 70-85% by weight with an average TiO2 layer thickness of above 65 to 75 nm; a TiO2 content of 73.5-86.5% by weight with an average TiO2 layer thickness of above 75 to 85 nm; a TiO2 content of 75-88% by weight with an average TiO2 layer thickness of above 85 to 95 nm; a TiO2 content of 76.5-89% by weight with an average TiO2 layer thickness of above 95 to 105 nm; a TiO2 content of 78.5-90% by weight with an average TiO2 layer thickness of above 105 to 115 nm; a TiO2 content of 80-91% by weight with an average TiO2 layer thickness of above 115 to 125 nm; a TiO2 content of 81.5-92% by weight with an average TiO2 layer thickness of above 125 to 135 nm; a TiO2 content of 83-92.5% by weight with an average TiO2 layer thickness of above 135 to 145 nm; a TiO2 content of 84-93% by weight with an average TiO2 layer thickness of above 145 to 155 nm; a TiO2 content of 85-93% by weight with an average TiO2 layer thickness of above 155 to 165 nm; a TiO2 content of 86-93.5% by weight with an average TiO2 layer thickness of above 165 to 175 nm; a TiO2 content of 87-94% by weight with an average TiO2 layer thickness of above 175 to 185 nm; a TiO2 content of 87.5-94% by weight with an average TiO2 layer thickness of above 185 to 195 nm; a TiO2 content of 88-94.5% by weight with an average TiO2 layer thickness of above 195 to 205 nm; and a TiO2 content of 89-95% by weight with an average TiO2 layer thickness of above 205 to 215 nm. 11. The pearlescent pigments of claim 7,
wherein the metal oxide layer consists of TiO2 (largely in the rutile modification), and the substrate consists of mica, and the relationship between the TiO2 content in % by weight, based on the total weight of TiO2 and mica, and the average layer thickness of the TiO2 coating is selected from the group consisting of a TiO2 content of 50.5-62% by weight with an average TiO2 layer thickness of above 20 to 30 nm; a TiO2 content of 61-74% by weight with an average TiO2 layer thickness of above 35 to 45 nm; a TiO2 content of 65.5-78% by weight with an average TiO2 layer thickness of above 45 to 55 nm; a TiO2 content of 69.5-82% by weight with an average TiO2 layer thickness of above 55 to 65 nm; a TiO2 content of 72.5-85% by weight with an average TiO2 layer thickness of above 65 to 75 nm; a TiO2 content of 75-86.5% by weight with an average TiO2 layer thickness of above 75 to 85 nm; a TiO2 content of 77.5-88% by weight with an average TiO2 layer thickness of above 85 to 95 nm; a TiO2 content of 79-89% by weight with an average TiO2 layer thickness of above 95 to 105 nm; a TiO2 content of 80.5-90% by weight with an average TiO2 layer thickness of above 105 to 115 nm; a TiO2 content of 82-91% by weight with an average TiO2 layer thickness of above 115 to 125 nm; a TiO2 content of 83-92% by weight with an average TiO2 layer thickness of above 125 to 135 nm; a TiO2 content of 84.5-92.5% by weight with an average TiO2 layer thickness of above 135 to 145 nm; a TiO2 content of 85.5-93% by weight with an average TiO2 layer thickness of above 145 to 155 nm; a TiO2 content of 86.5-93% by weight with an average TiO2 layer thickness of above 155 to 165 nm; a TiO2 content of 87-93.5% by weight with an average TiO2 layer thickness of above 165 to 175 nm; a TiO2 content of 88-94% by weight with an average TiO2 layer thickness of above 175 to 185 nm; a TiO2 content of 88.5-94% by weight with an average TiO2 layer thickness of above 185 to 195 nm; a TiO2 content of 89-94.5% by weight with an average TiO2 layer thickness of above 195 to 205 nm; and a TiO2 content of 89.5-95% by weight with an average TiO2 layer thickness of above 205 to 215 nm. 12. The pearlescent pigments of claim 1,
wherein the transparent platelet-shaped substrate is selected from the group consisting of mica, synthetic mica, glass flakes, SiO2 platelets, Al2O3 platelets, and mixtures thereof. 13. The pearlescent pigments of claim 12,
wherein the largely transparent platelet-shaped substrate consists of mica, glass flakes or mixtures thereof. 14. The pearlescent pigments of claim 1,
wherein the pearlescent pigments have at least one further layer having a refractive index <2.0. 15. The pearlescent pigments of claim 1,
wherein the pearlescent pigments have at least one further protective layer on the optically active layer. 16. The pearlescent pigments of claim 15,
wherein the at least one further protective layer comprises at least one metal oxide layer whose metal oxides are selected from the group consisting of SiO2, Al2O3, cerium oxide, mixtures thereof and combinations thereof. 17. A method for producing the pearlescent pigments of claim 1,
wherein the method comprises the following steps: a) classifying the largely transparent substrate, to give a substrate having an average height hS of 40 to 110 nm; and b) coating the classified substrate with an optically active layer, to form a pearlescent pigment having an average size d50 of 3 to 8 μm. 18. A method for producing a material selected from the group consisting of paints, printing inks, cosmetics, plastics, glass, enamel and ceramic wherein the method comprises adding to said material the pearlescent pigments of claim 1. 19. The method of claim 18,
wherein the cosmetics are selected from the group consisting of concealer sticks, body powder, face powder, compact and loose powder, face makeup, powder cream, cream makeup, emulsion makeup, wax makeup, foundation, mousse makeup, blusher, eye makeup, lipcare sticks, lipsticks, lip gloss, lip liners, hair-styling compositions, hair-setting products, hair mousse, hair gel, hair wax, hair mascara, permanent and semipermanent hair colors, temporary hair colors, skincare compositions, and nail varnish compositions. 20. A method for producing soft-focus pigment comprising forming a pearlescent pigment by a method according to claim 17. 21. A coating composition comprising one of the pearlescent pigments of claim 1. 22. A method for determining the average layer thickness hS of a platelet-shaped substrate of an effect pigment comprising a platelet-shaped substrate having a density ρS and at least one optically active coating having a density ρM,
in accordance with the following formula:
h
S
=
4
d
M
3
3
r
S
2
+
π
d
M
2
r
S
+
2
·
d
M
ρ
S
ρ
M
·
(
100
c
M
-
1
)
-
(
d
M
r
S
)
2
-
2
·
d
M
r
S
where dM is the ascertained average layer thickness of at least one of a high-index layer and of a semitransparent metal coating,
rS is the ascertained average radius of the substrate,
ρS is the ascertained density of the substrate and ρM is the ascertained density of the optically active coating and cM is the ascertained weight fraction in percent of the optically active coating, based on the total weight of substrate and optically active coating. 23. The pearlescent pigments of claim 5, wherein the high-index coating has a refractive index nM>2.0. 24. The pearlescent pigments of claim 10, wherein the TiO2 is in the rutile modification. 25. The pearlescent pigments of claim 15, wherein the at least one further protective layer is a high index layer. 26. The method of claim 17, wherein the optically active layer has a high index. 27. The coating composition of claim 21, wherein the composition is a cosmetic product. | 1,600 |
661 | 13,805,932 | 1,658 | Nutritional compositions that mimic whole foods and methods of using the nutritional compositions are provided. The nutritional compositions may include an increased number and variety of fruits and vegetables, an increased variety of macronutrient sources and an increased amount of other components that are found in whole foods. Methods of administering such nutritional compositions to patients in need of same are also provided. The methods may include administering tube feed formulations to a patient at typical meal times such as, for example, breakfast, lunch and dinner. | 1. A method of administering tube feeding formulations comprising:
administering a first tube feed formulation having a whole food to a patient at a first time of a day corresponding to a typical breakfast time; administering a second tube feed formulation having a whole food to the patient at a second time of the day corresponding to a typical lunch time; and administering a third tube feed formulation having a whole food to the patient at a third time of the day corresponding to a typical dinner time. 2. The method according to claim 1, wherein the first, second and third tube feed formulations each include at least one source of protein, at least one source of fat, at least one source of carbohydrate, and at least one of a processed fruit and a processed vegetable. 3. The method according to claim 2, wherein the at least one of a processed fruit and a processed vegetable of each of the first, second and third tube feed formulations is different. 4. The method according to claim 1, wherein the source of protein of each of the first, second and third tube feed formulations is different. 5. The method according to claim 1, comprising administering a fourth tube feed formulation having a whole food to the patient at a fourth time of the day corresponding to a typical snack time. 6. The method according to claim 1 comprising several daily administrations corresponding to typical snack times. 7. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a daily basis. 8. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a weekly basis. 9. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a monthly basis. 10. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a daily, weekly, or monthly basis, and combinations thereof. 11. The method according to claim 1, wherein the tube feed formulations and a new tube feed formulation include at least one source of protein, at least one source of fat, at least one source of carbohydrate, and at least one of a processed fruit and a processed vegetable. 12. The method according to claim 11, wherein the at least one source of protein of each of the tube feed formulations and the new tube feed formulation is different. 13. The method according to claim 11, wherein the at least one source of carbohydrate of each of the tube feed formulations and the new tube feed formulation is different. 14. The method according to claim 11, wherein the at least one source of fat of each of the tube feed formulations and the new tube feed formulation is different. 15. The method according to claim 11, wherein the at least one of a processed fruit and a processed vegetable of each tube feed formulation and the new tube feed formulation is different. 16. The method according to claim 1, the tube feed formulation comprising a nutrient source that is typically consumed by individuals in a specific region of the world. 17. The method according to claim 16, wherein the nutrient source is selected from the group consisting of a fruit, a vegetable, an herb, a spice, a flavoring, and combinations thereof. 18. A method of administering tube feeding formulations, the method comprising:
administering a first tube feed formulation, a second tube feed formulation, and a third tube feed formulation to a patient at evenly spaced time intervals throughout a 24-hour time period, the evenly spaced time intervals corresponding to typical meal times, wherein the first, second and third tube feed formulations include a whole food component. 19. The method according to claim 18, wherein the first, second and third tube feed formulations each include at least one source of protein, at least one source of fat, at least one source of carbohydrate, and at least one of a processed fruit and a processed vegetable. 20. The method according to claim 18 comprising administering to the patient a fourth tube feed formulation having a whole food at a fourth time of the day corresponding to a typical snack time. | Nutritional compositions that mimic whole foods and methods of using the nutritional compositions are provided. The nutritional compositions may include an increased number and variety of fruits and vegetables, an increased variety of macronutrient sources and an increased amount of other components that are found in whole foods. Methods of administering such nutritional compositions to patients in need of same are also provided. The methods may include administering tube feed formulations to a patient at typical meal times such as, for example, breakfast, lunch and dinner.1. A method of administering tube feeding formulations comprising:
administering a first tube feed formulation having a whole food to a patient at a first time of a day corresponding to a typical breakfast time; administering a second tube feed formulation having a whole food to the patient at a second time of the day corresponding to a typical lunch time; and administering a third tube feed formulation having a whole food to the patient at a third time of the day corresponding to a typical dinner time. 2. The method according to claim 1, wherein the first, second and third tube feed formulations each include at least one source of protein, at least one source of fat, at least one source of carbohydrate, and at least one of a processed fruit and a processed vegetable. 3. The method according to claim 2, wherein the at least one of a processed fruit and a processed vegetable of each of the first, second and third tube feed formulations is different. 4. The method according to claim 1, wherein the source of protein of each of the first, second and third tube feed formulations is different. 5. The method according to claim 1, comprising administering a fourth tube feed formulation having a whole food to the patient at a fourth time of the day corresponding to a typical snack time. 6. The method according to claim 1 comprising several daily administrations corresponding to typical snack times. 7. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a daily basis. 8. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a weekly basis. 9. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a monthly basis. 10. The method according to claim 1, wherein the tube feed formulation is changed to a new tube feed formulation on a daily, weekly, or monthly basis, and combinations thereof. 11. The method according to claim 1, wherein the tube feed formulations and a new tube feed formulation include at least one source of protein, at least one source of fat, at least one source of carbohydrate, and at least one of a processed fruit and a processed vegetable. 12. The method according to claim 11, wherein the at least one source of protein of each of the tube feed formulations and the new tube feed formulation is different. 13. The method according to claim 11, wherein the at least one source of carbohydrate of each of the tube feed formulations and the new tube feed formulation is different. 14. The method according to claim 11, wherein the at least one source of fat of each of the tube feed formulations and the new tube feed formulation is different. 15. The method according to claim 11, wherein the at least one of a processed fruit and a processed vegetable of each tube feed formulation and the new tube feed formulation is different. 16. The method according to claim 1, the tube feed formulation comprising a nutrient source that is typically consumed by individuals in a specific region of the world. 17. The method according to claim 16, wherein the nutrient source is selected from the group consisting of a fruit, a vegetable, an herb, a spice, a flavoring, and combinations thereof. 18. A method of administering tube feeding formulations, the method comprising:
administering a first tube feed formulation, a second tube feed formulation, and a third tube feed formulation to a patient at evenly spaced time intervals throughout a 24-hour time period, the evenly spaced time intervals corresponding to typical meal times, wherein the first, second and third tube feed formulations include a whole food component. 19. The method according to claim 18, wherein the first, second and third tube feed formulations each include at least one source of protein, at least one source of fat, at least one source of carbohydrate, and at least one of a processed fruit and a processed vegetable. 20. The method according to claim 18 comprising administering to the patient a fourth tube feed formulation having a whole food at a fourth time of the day corresponding to a typical snack time. | 1,600 |
662 | 14,220,273 | 1,647 | The present invention relates to injecting a high specificity cytokine antagonist into a diseased intervertebral disc. | 1. A method of inhibiting degeneration of an extracellular matrix of a nucleus pulposus of an intervertebral disc having a degenerating nucleus pulposus and an annulus fibrosus in a patient in need thereof, comprising transdiscally administering into the degenerating nucleus pulposus an effective amount of a formulation comprising:
a) a high specificity cytokine antagonist that inhibits tumor necrosis factor-alpha (TNF-α), and b) one or more additional therapeutic agents selected from the group consisting of a growth factor and viable mesenchymal stem cells. 2. The method of claim 1, wherein the formulation comprises viable mesenchymal stem cells. 3. The method of claim 2, wherein the mesenchymal stem cells are autologous. 4. The method of claim 1, wherein the mesenchymal stem cells are provided in a concentrated form. 5. The method of claim 1, wherein the mesenchymal stem cells are uncultured. 6. The method of claim 1, wherein the formulation comprises a growth factor. 7. The method of claim 6, wherein the growth factor is transforming growth factor beta (TGF-β) or a bone morphogenetic protein (BMP). 8. The method of claim 1, wherein the high specificity cytokine antagonist is a monoclonal antibody. 9. The method of claim 8, wherein the monoclonal antibody is infliximab. 10. The method of claim 1, wherein the high specificity cytokine antagonist is etanercept. 11. The method of claim 1, wherein the high specificity cytokine antagonist is present in the formulation in a concentration of at least 100 mg/ml. 12. The method of claim 1, wherein the high specificity cytokine antagonist is present in the formulation in a dose of no more than 0.5 mg. 13. The method of claim 1, wherein the formulation is suitable for administration into an intervertebral disc at a volume of between 0.5 ml and about 3.0 ml. 14. The method of claim 9, wherein the infliximab is present at a concentration of between about 30 mg/ml and about 60 mg/ml. 15. The method of claim 1, wherein the formulation is delivered in a sustained release device. 16. The method of claim 1, wherein the high specificity cytokine antagonist inhibits the production of TNF-α, binds a membrane-bound TNF-α, binds a solubilized TNF-α or binds a receptor of TNF-α. 17. The method of claim 3, wherein the autologous mesenchymal stem cells are uncultured mesenchymal stem cells from about 7-10 ml of the patient's bone marrow. 18. A method of treating degenerative disc disease in an intervertebral disc having a nucleus pulposus and annulus fibrosus in a patient, comprising transdiscally administering into the degenerating nucleus pulposus an effective amount of a formulation comprising:
a) a high specificity cytokine antagonist that inhibits tumor necrosis factor-alpha (TNF-α), and b) one or more additional therapeutic agents selected from the group consisting of a growth factor and viable mesenchymal stem cells. 19. The method of claim 18, wherein the formulation is administered into the nucleus pulposus of the intervertebral disc. 20. The method of claim 18, wherein the formulation is administered into the annulus fibrosus of the intervertebral disc. | The present invention relates to injecting a high specificity cytokine antagonist into a diseased intervertebral disc.1. A method of inhibiting degeneration of an extracellular matrix of a nucleus pulposus of an intervertebral disc having a degenerating nucleus pulposus and an annulus fibrosus in a patient in need thereof, comprising transdiscally administering into the degenerating nucleus pulposus an effective amount of a formulation comprising:
a) a high specificity cytokine antagonist that inhibits tumor necrosis factor-alpha (TNF-α), and b) one or more additional therapeutic agents selected from the group consisting of a growth factor and viable mesenchymal stem cells. 2. The method of claim 1, wherein the formulation comprises viable mesenchymal stem cells. 3. The method of claim 2, wherein the mesenchymal stem cells are autologous. 4. The method of claim 1, wherein the mesenchymal stem cells are provided in a concentrated form. 5. The method of claim 1, wherein the mesenchymal stem cells are uncultured. 6. The method of claim 1, wherein the formulation comprises a growth factor. 7. The method of claim 6, wherein the growth factor is transforming growth factor beta (TGF-β) or a bone morphogenetic protein (BMP). 8. The method of claim 1, wherein the high specificity cytokine antagonist is a monoclonal antibody. 9. The method of claim 8, wherein the monoclonal antibody is infliximab. 10. The method of claim 1, wherein the high specificity cytokine antagonist is etanercept. 11. The method of claim 1, wherein the high specificity cytokine antagonist is present in the formulation in a concentration of at least 100 mg/ml. 12. The method of claim 1, wherein the high specificity cytokine antagonist is present in the formulation in a dose of no more than 0.5 mg. 13. The method of claim 1, wherein the formulation is suitable for administration into an intervertebral disc at a volume of between 0.5 ml and about 3.0 ml. 14. The method of claim 9, wherein the infliximab is present at a concentration of between about 30 mg/ml and about 60 mg/ml. 15. The method of claim 1, wherein the formulation is delivered in a sustained release device. 16. The method of claim 1, wherein the high specificity cytokine antagonist inhibits the production of TNF-α, binds a membrane-bound TNF-α, binds a solubilized TNF-α or binds a receptor of TNF-α. 17. The method of claim 3, wherein the autologous mesenchymal stem cells are uncultured mesenchymal stem cells from about 7-10 ml of the patient's bone marrow. 18. A method of treating degenerative disc disease in an intervertebral disc having a nucleus pulposus and annulus fibrosus in a patient, comprising transdiscally administering into the degenerating nucleus pulposus an effective amount of a formulation comprising:
a) a high specificity cytokine antagonist that inhibits tumor necrosis factor-alpha (TNF-α), and b) one or more additional therapeutic agents selected from the group consisting of a growth factor and viable mesenchymal stem cells. 19. The method of claim 18, wherein the formulation is administered into the nucleus pulposus of the intervertebral disc. 20. The method of claim 18, wherein the formulation is administered into the annulus fibrosus of the intervertebral disc. | 1,600 |
663 | 15,056,666 | 1,653 | The present invention is directed to a diagnostic composition for use in the viscoelastic analysis of a test liquid, and to a container ( 1 ) comprising same. The composition comprises at least an activator of coagulation, and at least one further constituent selected from CaCl 2 and from one or more inhibitors and/or coagulation components, wherein the composition is present in essentially dry form of all constituents and in an amount sufficient for performing one single viscoelastic analysis of a specified blood or plasma sample and wherein the constituents are not present in a substance mixture, but in a spatially separated form. The present invention is further directed to a method of performing a viscoelastic analysis on a test liquid, and to the use of the diagnostic composition in such a method. | 1. A diagnostic composition for use in the viscoelastic analysis of a test liquid, comprising at least two of the following constituents:
a) one activator of coagulation; and b) a calcium salt in an amount sufficient to ensure recalcification of the test liquid; or c) one inhibitors or other coagulation components or factors; characterized in that the constituents are present in an essentially dry, and wherein the constituents are not present in a substance mixture, but in a spatially separated form. 2. The composition according to claim 1, where the spatial separation of the constituents is realized by incorporating each constituent into a separate carrier material. 3. The composition according to claim 2, where the carrier material comprises at least a carbohydrate. 4. The composition according to claim 3, where the carrier material shows no significant influence on the clotting behaviour, the clot formation behaviour, or the clot lysis behaviour. 5. The diagnostic composition of claim 1, wherein the activator of coagulation is an intrinsic or extrinsic activator. 6. The diagnostic composition of claim 5, wherein the extrinsic activator of coagulation is the Tissue Factor (TF). 7. The diagnostic composition of claim 6, wherein the Tissue Factor is lipidated TF or rTF. 8. The diagnostic composition of claim 5, wherein the intrinsic activator of coagulation is celite, ellagic acid, sulfatit, kaolin, silica, or RNA. 9. The diagnostic composition of claim 1, wherein the inhibitor is a platelet inhibitor, fibrinolysis inhibitor, or heparin inhibitor. 10. The diagnostic composition of claim 9, wherein platelet inhibitor is a cyto-skeletton inhibitor or a GPIIb/IIIa antagonist. 11. The diagnostic composition of claim 9, wherein the fibrinolysis inhibitor is aprotinine, tranexamic acid, or eaca. 12. The diagnostic composition of claim 9, wherein the heparin inhibitor is selected from heparinase, protamine or protamine-related peptides. 13. The diagnostic composition of claim 1, wherein the coagulation factor is a coagulation factor or activated coagulation factor. 14. (canceled) 15. The diagnostic composition of claim 1, wherein the dry form is a lyophilized form. 16. The diagnostic composition of claim 1, which further comprises a stabilizer. 17. The diagnostic composition of claim 16, wherein the stabilizer is albumin or gelatine. 18. A container, comprising the diagnostic composition of claim 1, having an inner portion shaped in a manner that it can be attached to a device for performing viscoelastic measurements. 19.-22. (canceled) 23. A method of performing a viscoelastic analysis on a test liquid, comprising the steps of:
a) adding the test liquid into a container comprising the diagnostic composition of claim 1, thereby dissolving the diagnostic composition contained therein in the test liquid to form a mixture; b) optionally transferring the mixture of said test liquid and said diagnostic composition into an apparatus suitable for performing a viscolelastic analysis; or putting the container into an apparatus suitable for performing a viscolelastic analysis; and c) performing the viscoelastic analysis of said mixture. 24. The method of claim 23, wherein the test liquid is a blood sample. 25. The method of claim 23, wherein the blood sample is whole blood or blood plasma. 26. The method of claim 23, wherein step c) takes about 1-60 sec., preferably about 2-10 sec., more preferably about 5 sec. 27. The method of claim 23, wherein the mixture is transferred in step b) by manually or automatically pipetting the mixture from the container and by transferring it thereby to the apparatus. 28. The method of claim 23, wherein the mixture is transferred to a measuring cup of the apparatus. 29. The method of claim 23, wherein the apparatus is a thromboelastometer or a thrombelastograph. 30. The method of claim 23, wherein the analysis comprises the determination of the clotting time, the clot formation time, the firmness of the clot over time and/or fibrinolysis. 31.-32. (canceled) 33. The diagnostic composition according to claim 1, wherein the calcium salt is CaCl2. 34. The diagnostic composition of claim 33, wherein CaCl2 is present in an amount of about 1-100 μmol/ml of the test liquid. 35. The composition of claim 3, where the carrier material comprises saccharose or cellulose. 36. The diagnostic composition of claim 1, wherein the coagulation factor is FXa or FVa or activated protein C or FVIIa. 37. The method of claim 23, wherein the test liquid is a mammalian blood sample. 38. The method of claim 23, wherein the test liquid is a human blood sample. | The present invention is directed to a diagnostic composition for use in the viscoelastic analysis of a test liquid, and to a container ( 1 ) comprising same. The composition comprises at least an activator of coagulation, and at least one further constituent selected from CaCl 2 and from one or more inhibitors and/or coagulation components, wherein the composition is present in essentially dry form of all constituents and in an amount sufficient for performing one single viscoelastic analysis of a specified blood or plasma sample and wherein the constituents are not present in a substance mixture, but in a spatially separated form. The present invention is further directed to a method of performing a viscoelastic analysis on a test liquid, and to the use of the diagnostic composition in such a method.1. A diagnostic composition for use in the viscoelastic analysis of a test liquid, comprising at least two of the following constituents:
a) one activator of coagulation; and b) a calcium salt in an amount sufficient to ensure recalcification of the test liquid; or c) one inhibitors or other coagulation components or factors; characterized in that the constituents are present in an essentially dry, and wherein the constituents are not present in a substance mixture, but in a spatially separated form. 2. The composition according to claim 1, where the spatial separation of the constituents is realized by incorporating each constituent into a separate carrier material. 3. The composition according to claim 2, where the carrier material comprises at least a carbohydrate. 4. The composition according to claim 3, where the carrier material shows no significant influence on the clotting behaviour, the clot formation behaviour, or the clot lysis behaviour. 5. The diagnostic composition of claim 1, wherein the activator of coagulation is an intrinsic or extrinsic activator. 6. The diagnostic composition of claim 5, wherein the extrinsic activator of coagulation is the Tissue Factor (TF). 7. The diagnostic composition of claim 6, wherein the Tissue Factor is lipidated TF or rTF. 8. The diagnostic composition of claim 5, wherein the intrinsic activator of coagulation is celite, ellagic acid, sulfatit, kaolin, silica, or RNA. 9. The diagnostic composition of claim 1, wherein the inhibitor is a platelet inhibitor, fibrinolysis inhibitor, or heparin inhibitor. 10. The diagnostic composition of claim 9, wherein platelet inhibitor is a cyto-skeletton inhibitor or a GPIIb/IIIa antagonist. 11. The diagnostic composition of claim 9, wherein the fibrinolysis inhibitor is aprotinine, tranexamic acid, or eaca. 12. The diagnostic composition of claim 9, wherein the heparin inhibitor is selected from heparinase, protamine or protamine-related peptides. 13. The diagnostic composition of claim 1, wherein the coagulation factor is a coagulation factor or activated coagulation factor. 14. (canceled) 15. The diagnostic composition of claim 1, wherein the dry form is a lyophilized form. 16. The diagnostic composition of claim 1, which further comprises a stabilizer. 17. The diagnostic composition of claim 16, wherein the stabilizer is albumin or gelatine. 18. A container, comprising the diagnostic composition of claim 1, having an inner portion shaped in a manner that it can be attached to a device for performing viscoelastic measurements. 19.-22. (canceled) 23. A method of performing a viscoelastic analysis on a test liquid, comprising the steps of:
a) adding the test liquid into a container comprising the diagnostic composition of claim 1, thereby dissolving the diagnostic composition contained therein in the test liquid to form a mixture; b) optionally transferring the mixture of said test liquid and said diagnostic composition into an apparatus suitable for performing a viscolelastic analysis; or putting the container into an apparatus suitable for performing a viscolelastic analysis; and c) performing the viscoelastic analysis of said mixture. 24. The method of claim 23, wherein the test liquid is a blood sample. 25. The method of claim 23, wherein the blood sample is whole blood or blood plasma. 26. The method of claim 23, wherein step c) takes about 1-60 sec., preferably about 2-10 sec., more preferably about 5 sec. 27. The method of claim 23, wherein the mixture is transferred in step b) by manually or automatically pipetting the mixture from the container and by transferring it thereby to the apparatus. 28. The method of claim 23, wherein the mixture is transferred to a measuring cup of the apparatus. 29. The method of claim 23, wherein the apparatus is a thromboelastometer or a thrombelastograph. 30. The method of claim 23, wherein the analysis comprises the determination of the clotting time, the clot formation time, the firmness of the clot over time and/or fibrinolysis. 31.-32. (canceled) 33. The diagnostic composition according to claim 1, wherein the calcium salt is CaCl2. 34. The diagnostic composition of claim 33, wherein CaCl2 is present in an amount of about 1-100 μmol/ml of the test liquid. 35. The composition of claim 3, where the carrier material comprises saccharose or cellulose. 36. The diagnostic composition of claim 1, wherein the coagulation factor is FXa or FVa or activated protein C or FVIIa. 37. The method of claim 23, wherein the test liquid is a mammalian blood sample. 38. The method of claim 23, wherein the test liquid is a human blood sample. | 1,600 |
664 | 15,953,141 | 1,636 | The present invention relates to an artificially manipulated neovascularization-associated factor for regulating neovascularization and a use thereof. More particularly, the present invention relates to a system for artificially regulating neovascularization, which includes an artificially manipulated neovascularization-associated factor for regulating neovascularization and/or a composition for artificially manipulating the neovascularization-associated factor. In a specific aspect, a neovascularization regulatory system including a neovascularization-associated factor such as artificially manipulated VEGFA, HIF1A, ANGPT2, EPAS1, or ANGPTL4 and/or an expression product thereof is provided. | 1. A composition for gene manipulation, comprising:
a guide nucleic acid capable of targeting at least one of the target sequence selected from SEQ ID NOs: 1 to 1522 in nucleic acid sequences of one or more genes selected from the group consisting of a VEGFA gene, an HIF1A gene, an ANGPT2 gene, an EPAS1 gene and an ANGPTL4 gene, or a nucleic acid sequence encoding the same; and an editor protein or a nucleic acid sequence encoding the same. 2. The composition for gene manipulation of claim 1, wherein the editor protein includes one or more proteins selected from the group consisting of a Streptococcus pyogenes-derived Cas9 protein, a Campylobacter jejuni-derived Cas9 protein, a Streptococcus thermophilus-derived Cas9 protein, a Streptocuccus aureus-derived Cas9 protein, a Neisseria meningitidis-derived Cas9 protein, and a Cpf1 protein. 3. The composition for gene manipulation of claim 2, wherein the editor protein is a Streptococcus pyogenes-derived Cas9 protein or Campylobacter jejuni-derived Cas9 protein. 4. The composition for gene manipulation of claim 1, wherein the gene manipulation includes one or more modifications of nucleic acids which is
at least one of a deletion or insertion of one or more nucleotides, a substitution with one or more nucleotides different from a wild-type gene, and an insertion of one or more foreign nucleotide, in a proto-spacer-adjacent motif (PAM) sequence in a nucleic acid sequence constituting the neovascularization-associated factor or in a continuous 1 bp to 50 bp the base sequence region adjacent to the 5′ end and/or 3′ end thereof, or a chemical modification of one or more nucleotides in a nucleic acid sequence constituting the neovascularization-associated factor. 5. The composition for gene manipulation of claim 4, wherein the PAM sequence includes one or more of the following sequences (described in the 5′ to 3′ direction):
NGG (N is A, T, C or G);
NNNNRYAC (each N is independently A, T, C or G, R is A or G, and Y is C or T);
NNAGAAW (each N is independently A, T, C or G, and W is A or T);
NNNNGATT (each N is independently A, T, C or G);
NNGRR(T) (each N is independently A, T, C or G, R is A or G, and Y is C or T); and
TTN (N is A, T, C or G). 6. The composition for gene manipulation of claim 1, wherein the composition for gene manipulation is formed in a viral vector system. 7. The composition for gene manipulation of claim 6, wherein the viral vector includes one or more selected from a retrovirus, a lentivirus, an adenovirus, adeno-associated virus (AAV), vaccinia virus, a poxvirus and a herpes simplex virus. 8. The composition for gene manipulation of claim 1, wherein the composition is formed in a ribonucleoprotein which is a complex of a guide nucleic acid and an editor protein, and
wherein the guide nucleic acid is guide RNA. 9. The composition for gene manipulation of claim 1, wherein the composition is used for treating an angiovascular disorder. 10. The composition for treating of claim 9, wherein the angiovascular disorder is ischemic retinopathy or retinopathy of prematurity. 11. A guide nucleic acid, which is capable of targeting at least one of group consisting of target sequences of SEQ ID NOs: 1 to 1522 for artificially manipulating one or more genes selected from the group consisting of a VEGFA gene, an HIF1A gene, an ANGPT2 gene, an EPAS1 gene and an ANGPTL4 gene,
wherein the target sequences of SEQ ID NOs: 1 to 1522 are in the nucleic acid sequences of the genes, and wherein the guide nucleic acid is complexed with an editor protein for artificially manipulating the genes. 12. The guide nucleic acid of claim 11, which includes one or more guide nucleic acids selected from the group consisting of:
guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 3, 4, 7, 9, 10 and 11 in the nucleic acid sequence of the VEGFA gene; guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 14, 18, 19, 20, 26, 29 and 31 in the nucleic acid sequence of the HIF1A gene; guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 33, 34, 37, 38, 39 and 43 in the nucleic acid sequence of the ANGPT2 gene; guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 47, 48, 49, 50, 53, 54 and 55 in the nucleic acid sequence of the EPAS1 gene; and guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 64, 66, 67, 73, 76 and 79 in the nucleic acid sequence of the ANGPTL4 gene. 13. The guide nucleic acid of claim 11, wherein the guide nucleic acid is a nucleotide of 18 to 23 bp. 14. A method for treating an angiovascular disorder, comprising:
introducing (administering) a composition to a subject, wherein the composition comprising: a guide nucleic acid capable of targeting at least one of the target sequence selected from SEQ ID NOs: 1 to 1522 in nucleic acid sequences of one or more genes selected from the group consisting of a VEGFA gene, an HIF1A gene, an ANGPT2 gene, an EPAS1 gene and an ANGPTL4 gene, or a nucleic acid sequence encoding the same; and an editor protein or a nucleic acid sequence encoding the same. 15. The method of claim 14, wherein the angiovascular disorder is ischemic retinopathy or retinopathy of prematurity. 16. The method of claim 14, wherein the editor protein includes one or more proteins selected from the group consisting of a Streptococcus pyogenes-derived Cas9 protein, a Campylobacter jejuni-derived Cas9 protein, a Streptococcus thermophilus-derived Cas9 protein, a Streptocuccus aureus-derived Cas9 protein, a Neisseria meningitidis-derived Cas9 protein, and a Cpf1 protein. 17. The method of claim 14, wherein the introducing is conducted through eyeball. 18. The method of claim 14, wherein the composition is formed in a viral vector system. 19. The method of claim 18, wherein the viral vector includes one or more selected from a retrovirus, a lentivirus, an adenovirus, adeno-associated virus (AAV), vaccinia virus, a poxvirus and a herpes simplex virus. | The present invention relates to an artificially manipulated neovascularization-associated factor for regulating neovascularization and a use thereof. More particularly, the present invention relates to a system for artificially regulating neovascularization, which includes an artificially manipulated neovascularization-associated factor for regulating neovascularization and/or a composition for artificially manipulating the neovascularization-associated factor. In a specific aspect, a neovascularization regulatory system including a neovascularization-associated factor such as artificially manipulated VEGFA, HIF1A, ANGPT2, EPAS1, or ANGPTL4 and/or an expression product thereof is provided.1. A composition for gene manipulation, comprising:
a guide nucleic acid capable of targeting at least one of the target sequence selected from SEQ ID NOs: 1 to 1522 in nucleic acid sequences of one or more genes selected from the group consisting of a VEGFA gene, an HIF1A gene, an ANGPT2 gene, an EPAS1 gene and an ANGPTL4 gene, or a nucleic acid sequence encoding the same; and an editor protein or a nucleic acid sequence encoding the same. 2. The composition for gene manipulation of claim 1, wherein the editor protein includes one or more proteins selected from the group consisting of a Streptococcus pyogenes-derived Cas9 protein, a Campylobacter jejuni-derived Cas9 protein, a Streptococcus thermophilus-derived Cas9 protein, a Streptocuccus aureus-derived Cas9 protein, a Neisseria meningitidis-derived Cas9 protein, and a Cpf1 protein. 3. The composition for gene manipulation of claim 2, wherein the editor protein is a Streptococcus pyogenes-derived Cas9 protein or Campylobacter jejuni-derived Cas9 protein. 4. The composition for gene manipulation of claim 1, wherein the gene manipulation includes one or more modifications of nucleic acids which is
at least one of a deletion or insertion of one or more nucleotides, a substitution with one or more nucleotides different from a wild-type gene, and an insertion of one or more foreign nucleotide, in a proto-spacer-adjacent motif (PAM) sequence in a nucleic acid sequence constituting the neovascularization-associated factor or in a continuous 1 bp to 50 bp the base sequence region adjacent to the 5′ end and/or 3′ end thereof, or a chemical modification of one or more nucleotides in a nucleic acid sequence constituting the neovascularization-associated factor. 5. The composition for gene manipulation of claim 4, wherein the PAM sequence includes one or more of the following sequences (described in the 5′ to 3′ direction):
NGG (N is A, T, C or G);
NNNNRYAC (each N is independently A, T, C or G, R is A or G, and Y is C or T);
NNAGAAW (each N is independently A, T, C or G, and W is A or T);
NNNNGATT (each N is independently A, T, C or G);
NNGRR(T) (each N is independently A, T, C or G, R is A or G, and Y is C or T); and
TTN (N is A, T, C or G). 6. The composition for gene manipulation of claim 1, wherein the composition for gene manipulation is formed in a viral vector system. 7. The composition for gene manipulation of claim 6, wherein the viral vector includes one or more selected from a retrovirus, a lentivirus, an adenovirus, adeno-associated virus (AAV), vaccinia virus, a poxvirus and a herpes simplex virus. 8. The composition for gene manipulation of claim 1, wherein the composition is formed in a ribonucleoprotein which is a complex of a guide nucleic acid and an editor protein, and
wherein the guide nucleic acid is guide RNA. 9. The composition for gene manipulation of claim 1, wherein the composition is used for treating an angiovascular disorder. 10. The composition for treating of claim 9, wherein the angiovascular disorder is ischemic retinopathy or retinopathy of prematurity. 11. A guide nucleic acid, which is capable of targeting at least one of group consisting of target sequences of SEQ ID NOs: 1 to 1522 for artificially manipulating one or more genes selected from the group consisting of a VEGFA gene, an HIF1A gene, an ANGPT2 gene, an EPAS1 gene and an ANGPTL4 gene,
wherein the target sequences of SEQ ID NOs: 1 to 1522 are in the nucleic acid sequences of the genes, and wherein the guide nucleic acid is complexed with an editor protein for artificially manipulating the genes. 12. The guide nucleic acid of claim 11, which includes one or more guide nucleic acids selected from the group consisting of:
guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 3, 4, 7, 9, 10 and 11 in the nucleic acid sequence of the VEGFA gene; guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 14, 18, 19, 20, 26, 29 and 31 in the nucleic acid sequence of the HIF1A gene; guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 33, 34, 37, 38, 39 and 43 in the nucleic acid sequence of the ANGPT2 gene; guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 47, 48, 49, 50, 53, 54 and 55 in the nucleic acid sequence of the EPAS1 gene; and guide nucleic acids capable of targeting at least one of the target sequence selected from SEQ ID NOs: 64, 66, 67, 73, 76 and 79 in the nucleic acid sequence of the ANGPTL4 gene. 13. The guide nucleic acid of claim 11, wherein the guide nucleic acid is a nucleotide of 18 to 23 bp. 14. A method for treating an angiovascular disorder, comprising:
introducing (administering) a composition to a subject, wherein the composition comprising: a guide nucleic acid capable of targeting at least one of the target sequence selected from SEQ ID NOs: 1 to 1522 in nucleic acid sequences of one or more genes selected from the group consisting of a VEGFA gene, an HIF1A gene, an ANGPT2 gene, an EPAS1 gene and an ANGPTL4 gene, or a nucleic acid sequence encoding the same; and an editor protein or a nucleic acid sequence encoding the same. 15. The method of claim 14, wherein the angiovascular disorder is ischemic retinopathy or retinopathy of prematurity. 16. The method of claim 14, wherein the editor protein includes one or more proteins selected from the group consisting of a Streptococcus pyogenes-derived Cas9 protein, a Campylobacter jejuni-derived Cas9 protein, a Streptococcus thermophilus-derived Cas9 protein, a Streptocuccus aureus-derived Cas9 protein, a Neisseria meningitidis-derived Cas9 protein, and a Cpf1 protein. 17. The method of claim 14, wherein the introducing is conducted through eyeball. 18. The method of claim 14, wherein the composition is formed in a viral vector system. 19. The method of claim 18, wherein the viral vector includes one or more selected from a retrovirus, a lentivirus, an adenovirus, adeno-associated virus (AAV), vaccinia virus, a poxvirus and a herpes simplex virus. | 1,600 |
665 | 16,519,434 | 1,651 | Tributyrin and/or tributyrin derivatives are used to selectively increase the levels of Bifidobacteria in the gut. In preferred methods, the tributyrin and/or tributyrin derivatives are orally administered alone or in combination with a food item or other nutritionally acceptable carrier, and may further include Bifidobacteria. Tributyrin-enhanced Bifidobacterium strains are also contemplated that include Bifidobacteria previously cultured in tributyrin and/or tributyrin derivatives. | 1. A method of increasing levels of Faecalibacterium prausnitzii in the gut of a human, the method comprising:
administering a tributyrin-containing composition to the human at a dosage of between 50 to 1,000 mg tributyrin or a tributyrin derivative per day; and wherein the tributyrin-containing composition is effective to selectively increase the levels of Faecalibacterium prausnitzii in the gut. 2. The method of claim 1, wherein the human suffers from irritable bowel disease and/or Crohn's disease. 3. The method of claim 1, wherein the tributyrin-containing composition further comprises at least one odor-masking component. 4. The method of claim 1, wherein the dosage also increases levels of Bifidobacteria in the gut of a human. 5. The method of claim 1, wherein the tributyrin derivative is a butyrate mono-ester, a butyrate di-ester, beta hydroxybutyrate, monobutyrin, dibutyrin, triacetin, tripropionate, glyceryl monoacetate, glyceryl diacetate, or acetoacetate. 6. (canceled) 7. The method of claim 1, further comprising administering at least one probiotic microorganism selected from the group consisting of Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus brevis, Lactobacillus gasseri, Lactobacillus rhamnosus, Bifidobacterium lactis, Bifidobacterium breve, and Bifidobacterium longum. 8. The method of claim 1, wherein the tributyrin-containing composition further comprises at least one additive selected from the group consisting of superoxide dismutase (SOD), compositions comprising activators of SOD, foods or extracts thereof comprising bioavailable SOD, copper I (CuI), selenium (Se), fulvic acid, compositions comprising fulvic acid, Co-enzyme Q10 (ubiquinone), pyrroloquinoline quinone (PQQ), an arabinoxylan (AX), an arabinoxylan oligosaccharide (AXOS), a xylooligosaccharide (XOS), a fructooligosaccharide (FOS), a galactooligosaccharide (GOS), inulin, and pectin. 9. A method of increasing a probiotic benefit of a food item comprising Faecalibacterium prausnitzii, comprising:
combining the food item with a tributyrin-containing composition that comprises tributyrin or a tributyrin derivative; wherein the tributyrin or a tributyrin derivative are present in the food item in an amount that selectively increase the levels of Faecalibacterium prausnitzii in the gut upon ingestion of the food item. 10. The method of claim 9, wherein the tributyrin-containing composition further comprises at least one odor-masking component. 11. The method of claim 9, wherein the tributyrin derivative is a butyrate mono-ester, a butyrate di-ester, beta hydroxybutyrate, monobutyrin, dibutyrin, triacetin, tripropionate, glyceryl monoacetate, glyceryl diacetate, or acetoacetate. 12. The method of claim 9, wherein the food item is a dairy product, a baked good, or a fermented food product. 13. The method of claim 9, wherein the amount of tributyrin or the tributyrin derivative in the tributyrin-containing composition is between 50-1,000 mg. 14. A nutritional supplement comprising:
a nutritionally acceptable carrier in combination with Faecalibacterium prausnitzii and further comprising tributyrin or a tributyrin derivative; wherein the carrier, the Faecalibacterium prausnitzii, and the tributyrin or a tributyrin derivative are formulated in a single oral dosage form. 15. The nutritional supplement of claim 14, wherein the supplement further comprises a Bifidobacterium strain selected from the group consisting of Bifidobacterium lactis, Bifidobacterium bifidum, Bifidobacterium infantis, Bifidobacterium breve, and Bifidobacterium longum. 16. The nutritional supplement of claim 14, wherein the Faecalibacterium prausnitzii is freeze dried. 17. The nutritional supplement of claim 14, wherein the tributyrin derivative is beta hydroxybutyrate, monobutyrin, dibutyrin, triacetin, tripropionate, glyceryl monoacetate, glyceryl diacetate, acetoacetate, a butyrate mono-ester, or a butyrate di-ester. 18. The nutritional supplement of claim 14, wherein the single oral dosage form is in a liquid form or a solid form. 19. The nutritional supplement of claim 14, wherein the single oral dosage form contains the Faecalibacterium prausnitzii in an amount of between 106 and 1012 CFU/g. 20. The nutritional supplement of claim 14, wherein the amount of tributyrin or the tributyrin derivative in the dietary supplement is between 50-1,000 mg. | Tributyrin and/or tributyrin derivatives are used to selectively increase the levels of Bifidobacteria in the gut. In preferred methods, the tributyrin and/or tributyrin derivatives are orally administered alone or in combination with a food item or other nutritionally acceptable carrier, and may further include Bifidobacteria. Tributyrin-enhanced Bifidobacterium strains are also contemplated that include Bifidobacteria previously cultured in tributyrin and/or tributyrin derivatives.1. A method of increasing levels of Faecalibacterium prausnitzii in the gut of a human, the method comprising:
administering a tributyrin-containing composition to the human at a dosage of between 50 to 1,000 mg tributyrin or a tributyrin derivative per day; and wherein the tributyrin-containing composition is effective to selectively increase the levels of Faecalibacterium prausnitzii in the gut. 2. The method of claim 1, wherein the human suffers from irritable bowel disease and/or Crohn's disease. 3. The method of claim 1, wherein the tributyrin-containing composition further comprises at least one odor-masking component. 4. The method of claim 1, wherein the dosage also increases levels of Bifidobacteria in the gut of a human. 5. The method of claim 1, wherein the tributyrin derivative is a butyrate mono-ester, a butyrate di-ester, beta hydroxybutyrate, monobutyrin, dibutyrin, triacetin, tripropionate, glyceryl monoacetate, glyceryl diacetate, or acetoacetate. 6. (canceled) 7. The method of claim 1, further comprising administering at least one probiotic microorganism selected from the group consisting of Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus brevis, Lactobacillus gasseri, Lactobacillus rhamnosus, Bifidobacterium lactis, Bifidobacterium breve, and Bifidobacterium longum. 8. The method of claim 1, wherein the tributyrin-containing composition further comprises at least one additive selected from the group consisting of superoxide dismutase (SOD), compositions comprising activators of SOD, foods or extracts thereof comprising bioavailable SOD, copper I (CuI), selenium (Se), fulvic acid, compositions comprising fulvic acid, Co-enzyme Q10 (ubiquinone), pyrroloquinoline quinone (PQQ), an arabinoxylan (AX), an arabinoxylan oligosaccharide (AXOS), a xylooligosaccharide (XOS), a fructooligosaccharide (FOS), a galactooligosaccharide (GOS), inulin, and pectin. 9. A method of increasing a probiotic benefit of a food item comprising Faecalibacterium prausnitzii, comprising:
combining the food item with a tributyrin-containing composition that comprises tributyrin or a tributyrin derivative; wherein the tributyrin or a tributyrin derivative are present in the food item in an amount that selectively increase the levels of Faecalibacterium prausnitzii in the gut upon ingestion of the food item. 10. The method of claim 9, wherein the tributyrin-containing composition further comprises at least one odor-masking component. 11. The method of claim 9, wherein the tributyrin derivative is a butyrate mono-ester, a butyrate di-ester, beta hydroxybutyrate, monobutyrin, dibutyrin, triacetin, tripropionate, glyceryl monoacetate, glyceryl diacetate, or acetoacetate. 12. The method of claim 9, wherein the food item is a dairy product, a baked good, or a fermented food product. 13. The method of claim 9, wherein the amount of tributyrin or the tributyrin derivative in the tributyrin-containing composition is between 50-1,000 mg. 14. A nutritional supplement comprising:
a nutritionally acceptable carrier in combination with Faecalibacterium prausnitzii and further comprising tributyrin or a tributyrin derivative; wherein the carrier, the Faecalibacterium prausnitzii, and the tributyrin or a tributyrin derivative are formulated in a single oral dosage form. 15. The nutritional supplement of claim 14, wherein the supplement further comprises a Bifidobacterium strain selected from the group consisting of Bifidobacterium lactis, Bifidobacterium bifidum, Bifidobacterium infantis, Bifidobacterium breve, and Bifidobacterium longum. 16. The nutritional supplement of claim 14, wherein the Faecalibacterium prausnitzii is freeze dried. 17. The nutritional supplement of claim 14, wherein the tributyrin derivative is beta hydroxybutyrate, monobutyrin, dibutyrin, triacetin, tripropionate, glyceryl monoacetate, glyceryl diacetate, acetoacetate, a butyrate mono-ester, or a butyrate di-ester. 18. The nutritional supplement of claim 14, wherein the single oral dosage form is in a liquid form or a solid form. 19. The nutritional supplement of claim 14, wherein the single oral dosage form contains the Faecalibacterium prausnitzii in an amount of between 106 and 1012 CFU/g. 20. The nutritional supplement of claim 14, wherein the amount of tributyrin or the tributyrin derivative in the dietary supplement is between 50-1,000 mg. | 1,600 |
666 | 14,895,076 | 1,641 | The invention relates to a method for establishing if a heart failure patient is susceptible to be hospitalized and/or rehospitalized, wherein said method comprises measuring the concentration of IGFBP 2 in a sample obtained from said heart failure patient. | 1. A method for establishing if a heart failure patient is susceptible to be hospitalized and/or rehospitalized, wherein said method comprises
measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient, and selecting a patient for hospitalization or rehospitalization based on the concentration of IGFBP2 measured in said measuring step. 2. The method according to claim 1 wherein said method further comprises the steps of:
(i) measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient,
(ii) comparing the concentration of IGFBP2 measured in step (i) to a threshold value derived from the concentration of IGFBP2 in samples from patients who are at particular stages of heart failure and/or to a threshold value derived from the concentration of IGFBP2 in samples from healthy patients. 3. The method according to claim 1 wherein the heart failure is an asymptomatic heart failure, a chronic heart failure or an acute heart failure. 4. The method according to claim 1, wherein said sample is selected in the group consisting of blood, plasma or plasma sample. 5. The method according to claim 1, wherein the concentration of IGFBP2 is measured by quantifying the level of IGFBP2 protein in the sample. 6. The method according to claim 5, wherein the quantification of the level of IGFBP2 protein is performed by using a set of antibodies directed against IGFBP2. 7. The method according to claim 5, wherein the quantification of the level of IGFBP2 protein is performed by ELISA. 8. The method according to claim 5, wherein the quantification of the level of IGFBP2 protein is performed by capillary electrophoresis-mass spectroscopy technique (CE-MS). 9. A method for establishing if a heart failure patient is susceptible to be further hospitalized, comprising
measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient, and selecting a patient for further hospitalization based on the concentration of IGFBP2 measured in said measuring step. 10. The method according to claim 9 wherein said method further comprises the steps of:
(i) measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient,
(ii) comparing the concentration of IGFBP2 measured in step (i) to a threshold value derived from the concentration of IGFBP2 in samples from patients who are at particular stages of heart failure and/or to a threshold value derived from the concentration of IGFBP2 in samples from healthy patients. | The invention relates to a method for establishing if a heart failure patient is susceptible to be hospitalized and/or rehospitalized, wherein said method comprises measuring the concentration of IGFBP 2 in a sample obtained from said heart failure patient.1. A method for establishing if a heart failure patient is susceptible to be hospitalized and/or rehospitalized, wherein said method comprises
measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient, and selecting a patient for hospitalization or rehospitalization based on the concentration of IGFBP2 measured in said measuring step. 2. The method according to claim 1 wherein said method further comprises the steps of:
(i) measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient,
(ii) comparing the concentration of IGFBP2 measured in step (i) to a threshold value derived from the concentration of IGFBP2 in samples from patients who are at particular stages of heart failure and/or to a threshold value derived from the concentration of IGFBP2 in samples from healthy patients. 3. The method according to claim 1 wherein the heart failure is an asymptomatic heart failure, a chronic heart failure or an acute heart failure. 4. The method according to claim 1, wherein said sample is selected in the group consisting of blood, plasma or plasma sample. 5. The method according to claim 1, wherein the concentration of IGFBP2 is measured by quantifying the level of IGFBP2 protein in the sample. 6. The method according to claim 5, wherein the quantification of the level of IGFBP2 protein is performed by using a set of antibodies directed against IGFBP2. 7. The method according to claim 5, wherein the quantification of the level of IGFBP2 protein is performed by ELISA. 8. The method according to claim 5, wherein the quantification of the level of IGFBP2 protein is performed by capillary electrophoresis-mass spectroscopy technique (CE-MS). 9. A method for establishing if a heart failure patient is susceptible to be further hospitalized, comprising
measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient, and selecting a patient for further hospitalization based on the concentration of IGFBP2 measured in said measuring step. 10. The method according to claim 9 wherein said method further comprises the steps of:
(i) measuring the concentration of IGFBP2 in a sample obtained from said heart failure patient,
(ii) comparing the concentration of IGFBP2 measured in step (i) to a threshold value derived from the concentration of IGFBP2 in samples from patients who are at particular stages of heart failure and/or to a threshold value derived from the concentration of IGFBP2 in samples from healthy patients. | 1,600 |
667 | 14,322,827 | 1,656 | The disclosure relates to the field of glyco-engineering, more specifically, to eukaryotic cells wherein both an endoglucosaminidase and a glycoprotein are present. These cells can be used to deglycosylate or partly deglycosylate the (exogenous) glycoprotein, in particular, without the need for adding an extra enzyme. Methods are also provided for the application of these cells in protein production. According to one specific aspect, the eukaryotic cells are glyco-engineered yeast cells in which, additionally, at least one exogenous enzyme needed for complex glycosylation is present, e.g., allowing easier separation of differentially glycosylated glycoproteins. | 1. A eukaryotic cell comprising:
a first exogenous polynucleotide encoding an endoglucosaminidase enzyme; and a second exogenous polynucleotide encoding a glycoprotein. 2. The eukaryotic cell of claim 1, which does not express an endogenous endoglucosaminidase enzyme. 3. The eukaryotic cell of claim 1, which is selected from the group consisting of a yeast cell, a plant cell, a mammalian cell, an insect cell, an Hek293 cell, and a Pichia cell. 4. The eukaryotic cell of claim 1, which is a glyco engineered yeast cell and further comprises:
at least a third exogenous polynucleotide encoding at least one enzyme needed for complex glycosylation, said enzyme selected from the group consisting of mannosidases and glycosyltransferases, other than mannosyltransferases and phosphomannosyltransferases. 5. The eukaryotic cell of claim 4, wherein the at least one enzyme needed for complex glycosylation is selected from the group consisting of N acetylglucosaminyl transferase I, N acetylglucosaminyl transferase II, mannosidase II, galactosyltransferase, and sialyltransferase. 6. The eukaryotic cell of claim 5, which is a Pichia cell. 7. The eukaryotic cell of claim 4, which is deficient in the functional expression of at least one enzyme involved in the production of high mannose structures. 8. The eukaryotic cell of claim 1, wherein the endoglucosaminidase is a mannosyl glycoprotein endo beta N acetylglucosaminidase (E.C. 3.2.1.96). 9. The eukaryotic cell of claim 1, wherein the glycoprotein is secreted by the cell. 10. The eukaryotic cell of claim 9, wherein the endoglucosaminidase is also secreted by the cell. 11. The eukaryotic cell of claim 1, wherein the endoglucosaminidase is operably linked to an endoplasmic reticulum or Golgi localization signal. 12. The eukaryotic cell of claim 11, wherein the endoplasmic reticulum or Golgi localization signal is from a protein selected from the group consisting of Kre2p, Ste13p, GM2 synthase, α2,6, glycosyltransferase, and α2,6, sialyltransferase. 13. The eukaryotic cell of claim 1, which is deficient in an enzymatic activity needed for complex glycosylation, wherein the enzyme is selected from the group consisting of ER mannosidase I, glucosidase I, glucosidase II, N acetylglucosaminyl transferase I, N acetylglucosaminyl transferase II, mannosidase II, and wherein the cell is not capable of complex glycosylation of glycoproteins. 14. A plant comprising the eukaryotic cell of claim 1. 15. A method for producing single GlcNAc modified proteins in a eukaryotic cell, the method comprising:
providing an eukaryotic cell comprising a first exogenous polynucleotide encoding an endoglucosaminidase enzyme and a second exogenous polynucleotide encoding a glycoprotein, in conditions suitable for expressing the endoglucosaminidase enzyme and the glycoprotein; and recovering the glycoprotein after it has been intracellularly or extracellularly contacted with the endoglucosaminidase. 16. The method according to claim 15, wherein the intracellular contact with the endoglucosaminidase occurs in the Golgi or endoplasmic reticulum. 17. The method according to claim 15, wherein the pH of the medium wherein the extracellular contact takes place is adjusted for optimal enzymatic endoglucosaminidase activity. 18. The method according to claim 15, wherein the eukaryotic cell is incapable of complex glycosylation of glycoproteins. 19. The method according to claim 15, further comprising:
having the glycoprotein processed by a glycosyltransferase after the glycoprotein has been intracellularly or extracellularly processed with the endoglucosaminidase. 20. A method for producing proteins in a glyco engineered yeast cell and depleting proteins with high mannose type glycosylation and/or hybrid type glycosylation, the method comprising:
providing a glyco engineered yeast cell comprising a first exogenous polynucleotide encoding an endoglucosaminidase enzyme, a second exogenous polynucleotide encoding a glycoprotein, and a third exogenous polynucleotide encoding at least one enzyme needed for complex glycosylation, the enzyme being selected from the group consisting of mannosidases and glycosyltransferases other than mannosyltransferases and phosphomannosyltransferases, in conditions suitable for expressing the endoglucosaminidase enzyme, the glycoprotein and the enzyme needed for complex glycosylation; and recovering the glycoprotein after it has been intracellularly contacted with the enzyme needed for complex glycosylation and intracellularly or extracellularly contacted with the endoglucosaminidase. 21. The method according to claim 20, wherein the intracellular contact with the endoglucosaminidase occurs in the Golgi or endoplasmic reticulum, after contact with the at least one enzyme needed for complex glycosylation. 22. The method according to claim 20, wherein the intracellular contact with the endoglucosaminidase occurs in the Golgi or endoplasmic reticulum, before contact with the at least one enzyme needed for complex glycosylation. 23. The method according to claim 20, wherein the pH of the medium wherein the extracellular contact takes place is adjusted for optimal enzymatic endoglucosaminidase activity. 24. A eukaryotic cell comprising:
a first exogenous nucleic acid molecule encoding an endoglucosaminidase enzyme; and a second exogenous nucleic acid molecule encoding a glycoprotein, wherein the endoglucosaminidase enzyme deglycosylates the glycoprotein upon co-expression of the first and second exogenous nucleic acid molecule. 25. A eukaryotic cell comprising:
a first exogenous nucleic acid molecule encoding Endo H; and a second exogenous nucleic acid molecule encoding a glycoprotein. 26. A eukaryotic cell comprising:
a first exogenous nucleic acid molecule encoding Endo M; and a second exogenous nucleic acid molecule encoding a glycoprotein. | The disclosure relates to the field of glyco-engineering, more specifically, to eukaryotic cells wherein both an endoglucosaminidase and a glycoprotein are present. These cells can be used to deglycosylate or partly deglycosylate the (exogenous) glycoprotein, in particular, without the need for adding an extra enzyme. Methods are also provided for the application of these cells in protein production. According to one specific aspect, the eukaryotic cells are glyco-engineered yeast cells in which, additionally, at least one exogenous enzyme needed for complex glycosylation is present, e.g., allowing easier separation of differentially glycosylated glycoproteins.1. A eukaryotic cell comprising:
a first exogenous polynucleotide encoding an endoglucosaminidase enzyme; and a second exogenous polynucleotide encoding a glycoprotein. 2. The eukaryotic cell of claim 1, which does not express an endogenous endoglucosaminidase enzyme. 3. The eukaryotic cell of claim 1, which is selected from the group consisting of a yeast cell, a plant cell, a mammalian cell, an insect cell, an Hek293 cell, and a Pichia cell. 4. The eukaryotic cell of claim 1, which is a glyco engineered yeast cell and further comprises:
at least a third exogenous polynucleotide encoding at least one enzyme needed for complex glycosylation, said enzyme selected from the group consisting of mannosidases and glycosyltransferases, other than mannosyltransferases and phosphomannosyltransferases. 5. The eukaryotic cell of claim 4, wherein the at least one enzyme needed for complex glycosylation is selected from the group consisting of N acetylglucosaminyl transferase I, N acetylglucosaminyl transferase II, mannosidase II, galactosyltransferase, and sialyltransferase. 6. The eukaryotic cell of claim 5, which is a Pichia cell. 7. The eukaryotic cell of claim 4, which is deficient in the functional expression of at least one enzyme involved in the production of high mannose structures. 8. The eukaryotic cell of claim 1, wherein the endoglucosaminidase is a mannosyl glycoprotein endo beta N acetylglucosaminidase (E.C. 3.2.1.96). 9. The eukaryotic cell of claim 1, wherein the glycoprotein is secreted by the cell. 10. The eukaryotic cell of claim 9, wherein the endoglucosaminidase is also secreted by the cell. 11. The eukaryotic cell of claim 1, wherein the endoglucosaminidase is operably linked to an endoplasmic reticulum or Golgi localization signal. 12. The eukaryotic cell of claim 11, wherein the endoplasmic reticulum or Golgi localization signal is from a protein selected from the group consisting of Kre2p, Ste13p, GM2 synthase, α2,6, glycosyltransferase, and α2,6, sialyltransferase. 13. The eukaryotic cell of claim 1, which is deficient in an enzymatic activity needed for complex glycosylation, wherein the enzyme is selected from the group consisting of ER mannosidase I, glucosidase I, glucosidase II, N acetylglucosaminyl transferase I, N acetylglucosaminyl transferase II, mannosidase II, and wherein the cell is not capable of complex glycosylation of glycoproteins. 14. A plant comprising the eukaryotic cell of claim 1. 15. A method for producing single GlcNAc modified proteins in a eukaryotic cell, the method comprising:
providing an eukaryotic cell comprising a first exogenous polynucleotide encoding an endoglucosaminidase enzyme and a second exogenous polynucleotide encoding a glycoprotein, in conditions suitable for expressing the endoglucosaminidase enzyme and the glycoprotein; and recovering the glycoprotein after it has been intracellularly or extracellularly contacted with the endoglucosaminidase. 16. The method according to claim 15, wherein the intracellular contact with the endoglucosaminidase occurs in the Golgi or endoplasmic reticulum. 17. The method according to claim 15, wherein the pH of the medium wherein the extracellular contact takes place is adjusted for optimal enzymatic endoglucosaminidase activity. 18. The method according to claim 15, wherein the eukaryotic cell is incapable of complex glycosylation of glycoproteins. 19. The method according to claim 15, further comprising:
having the glycoprotein processed by a glycosyltransferase after the glycoprotein has been intracellularly or extracellularly processed with the endoglucosaminidase. 20. A method for producing proteins in a glyco engineered yeast cell and depleting proteins with high mannose type glycosylation and/or hybrid type glycosylation, the method comprising:
providing a glyco engineered yeast cell comprising a first exogenous polynucleotide encoding an endoglucosaminidase enzyme, a second exogenous polynucleotide encoding a glycoprotein, and a third exogenous polynucleotide encoding at least one enzyme needed for complex glycosylation, the enzyme being selected from the group consisting of mannosidases and glycosyltransferases other than mannosyltransferases and phosphomannosyltransferases, in conditions suitable for expressing the endoglucosaminidase enzyme, the glycoprotein and the enzyme needed for complex glycosylation; and recovering the glycoprotein after it has been intracellularly contacted with the enzyme needed for complex glycosylation and intracellularly or extracellularly contacted with the endoglucosaminidase. 21. The method according to claim 20, wherein the intracellular contact with the endoglucosaminidase occurs in the Golgi or endoplasmic reticulum, after contact with the at least one enzyme needed for complex glycosylation. 22. The method according to claim 20, wherein the intracellular contact with the endoglucosaminidase occurs in the Golgi or endoplasmic reticulum, before contact with the at least one enzyme needed for complex glycosylation. 23. The method according to claim 20, wherein the pH of the medium wherein the extracellular contact takes place is adjusted for optimal enzymatic endoglucosaminidase activity. 24. A eukaryotic cell comprising:
a first exogenous nucleic acid molecule encoding an endoglucosaminidase enzyme; and a second exogenous nucleic acid molecule encoding a glycoprotein, wherein the endoglucosaminidase enzyme deglycosylates the glycoprotein upon co-expression of the first and second exogenous nucleic acid molecule. 25. A eukaryotic cell comprising:
a first exogenous nucleic acid molecule encoding Endo H; and a second exogenous nucleic acid molecule encoding a glycoprotein. 26. A eukaryotic cell comprising:
a first exogenous nucleic acid molecule encoding Endo M; and a second exogenous nucleic acid molecule encoding a glycoprotein. | 1,600 |
668 | 15,024,265 | 1,618 | Disclosed is an ultrasound (US) mediated delivery of therapeutic agents, such as the delivery of a drug, gene, nanoparticle or radioisotope, using a bi-phasic microparticle system including gas microbubbles, emulsion microdroplets and clusters thereof. Also, disclosed are a cluster composition and a pharmaceutical composition, and their use for delivery of therapeutic agents and as a contrast agent for ultrasound imaging. Moreover, disclosed are a method for delivering such therapeutic agents and the use of said compositions. | 1. A cluster composition which comprises a suspension of clusters in an aqueous biocompatible medium, wherein said clusters have a diameter in the range of 1 to 10 μm, and a circularity of <0.9 and comprises:
(i) a first component which comprises a gas microbubble and a first stabilizer to stabilize said gas microbubble; and
(ii) a second component which comprises a microdroplet comprising an oil phase and a second stabilizer to stabilize said microdroplet, wherein the oil phase comprises a diffusible component capable of diffusing into said gas microbubble so as to at least transiently increase the size thereof, wherein said second component optionally further comprises a first therapeutic agent;
wherein the microbubble and the microdroplet of said first and second components have opposite surface charges and form said clusters via attractive electrostatic interactions. 2. The cluster composition of claim 1, wherein a gas of the gas microbubble of the first component comprises sulphur hexafluoride, a C3-6 perfluorocarbon, nitrogen, air or mixtures thereof. 3. The cluster composition of claim 1, wherein the first stabilizer and the second stabilizer each have a net electrostatic charge that is opposite to that of the other. 4. The cluster composition of claim 3, wherein the first stabilizer and the second stabilizer each independently comprises a phospholipid, a protein, a polymer, a polyethyleneglycol, a fatty acid, a positively charged surfactant, a negatively charged surfactant or a mixture thereof. 5. The cluster composition of claim 1, wherein the oil phase of the microdroplet of the second component comprises a partly halogenated hydrocarbon, a fully halogenated hydrocarbon or a mixture thereof. 6. (canceled) 7. A pharmaceutical composition comprising:
(i) the cluster composition of claim 1; (ii) an optional second therapeutic agent, provided either in mixture with (i), or as a separate composition to (i); wherein said pharmaceutical composition comprises at least one therapeutic agent. 8. The pharmaceutical composition of claim 7, wherein the first therapeutic agent of the cluster composition (i) is absent, and the second therapeutic agent is present and is provided as a separate composition to (i). 9. The pharmaceutical composition of claim 7, wherein the first therapeutic agent of the cluster composition (i) is present, and the second therapeutic agent is present and is provided as a separate composition to (i). 10. The pharmaceutical composition of claim 7, wherein the first and second therapeutic agents are independently selected from the group of: consisting of a drug molecule, a nanoparticle and a nanoparticle delivery system, a gene, and a radioisotope. 11. An ultrasound contrast agent comprising the cluster composition of claim 1. 12. A method of delivering at least one therapeutic agent to a mammalian subject, comprising:
(i) administering the pharmaceutical composition of claim 7 to the mammalian subject; (ii) optionally imaging the microbubbles of said pharmaceutical composition by using ultrasound imaging to identify a region of interest for a treatment within said mammalian subject; (iii) activating a phase shift of the diffusible component of the second component of the cluster composition from the step (i) by ultrasound irradiation of a region of interest within said mammalian subject, such that:
(a) the microbubbles of said clusters are enlarged by said diffusible component of the step (iii) to give enlarged bubbles which are localised at said region of interest due to temporary blocking of a microcirculation at said region of interest by said enlarged bubbles; and
(b) said activating step (iii) facilitates extravasation of the therapeutic agent(s) administered in the step (i).
(iv) optionally, facilitating further extravasation of the therapeutic agent(s) administered in the step (i) by further ultrasound irradiation. 13. The method of claim 12, wherein the ultrasound irradiation applied in the step (iv) has a frequency in the range of 0.05 to 2 MHz. 14. The method of claim 12, wherein the ultrasound irradiation applied in the step (iii) has a mechanical index below 0.7. 15. The method of claim 12, which further comprises a quantification of an amount of the therapeutic agent(s) released by an analysis of an acoustic signature produced by the enlarged bubbles of the step (iii)(a). 16. A method of treatment of the mammalian subject comprising the method of claim 12. 17. A method of treatment of a mammalian subject comprising:
administering the cluster composition of claim 1, and applying a High Intensity Focused Ultrasound (HIFU) to a region of interest in the mammalian subject. 18. (canceled) 19. A method of an ultrasound imaging, comprising:
imaging a mammalian subject previously administered with the ultrasound contrast agent of claim 11. | Disclosed is an ultrasound (US) mediated delivery of therapeutic agents, such as the delivery of a drug, gene, nanoparticle or radioisotope, using a bi-phasic microparticle system including gas microbubbles, emulsion microdroplets and clusters thereof. Also, disclosed are a cluster composition and a pharmaceutical composition, and their use for delivery of therapeutic agents and as a contrast agent for ultrasound imaging. Moreover, disclosed are a method for delivering such therapeutic agents and the use of said compositions.1. A cluster composition which comprises a suspension of clusters in an aqueous biocompatible medium, wherein said clusters have a diameter in the range of 1 to 10 μm, and a circularity of <0.9 and comprises:
(i) a first component which comprises a gas microbubble and a first stabilizer to stabilize said gas microbubble; and
(ii) a second component which comprises a microdroplet comprising an oil phase and a second stabilizer to stabilize said microdroplet, wherein the oil phase comprises a diffusible component capable of diffusing into said gas microbubble so as to at least transiently increase the size thereof, wherein said second component optionally further comprises a first therapeutic agent;
wherein the microbubble and the microdroplet of said first and second components have opposite surface charges and form said clusters via attractive electrostatic interactions. 2. The cluster composition of claim 1, wherein a gas of the gas microbubble of the first component comprises sulphur hexafluoride, a C3-6 perfluorocarbon, nitrogen, air or mixtures thereof. 3. The cluster composition of claim 1, wherein the first stabilizer and the second stabilizer each have a net electrostatic charge that is opposite to that of the other. 4. The cluster composition of claim 3, wherein the first stabilizer and the second stabilizer each independently comprises a phospholipid, a protein, a polymer, a polyethyleneglycol, a fatty acid, a positively charged surfactant, a negatively charged surfactant or a mixture thereof. 5. The cluster composition of claim 1, wherein the oil phase of the microdroplet of the second component comprises a partly halogenated hydrocarbon, a fully halogenated hydrocarbon or a mixture thereof. 6. (canceled) 7. A pharmaceutical composition comprising:
(i) the cluster composition of claim 1; (ii) an optional second therapeutic agent, provided either in mixture with (i), or as a separate composition to (i); wherein said pharmaceutical composition comprises at least one therapeutic agent. 8. The pharmaceutical composition of claim 7, wherein the first therapeutic agent of the cluster composition (i) is absent, and the second therapeutic agent is present and is provided as a separate composition to (i). 9. The pharmaceutical composition of claim 7, wherein the first therapeutic agent of the cluster composition (i) is present, and the second therapeutic agent is present and is provided as a separate composition to (i). 10. The pharmaceutical composition of claim 7, wherein the first and second therapeutic agents are independently selected from the group of: consisting of a drug molecule, a nanoparticle and a nanoparticle delivery system, a gene, and a radioisotope. 11. An ultrasound contrast agent comprising the cluster composition of claim 1. 12. A method of delivering at least one therapeutic agent to a mammalian subject, comprising:
(i) administering the pharmaceutical composition of claim 7 to the mammalian subject; (ii) optionally imaging the microbubbles of said pharmaceutical composition by using ultrasound imaging to identify a region of interest for a treatment within said mammalian subject; (iii) activating a phase shift of the diffusible component of the second component of the cluster composition from the step (i) by ultrasound irradiation of a region of interest within said mammalian subject, such that:
(a) the microbubbles of said clusters are enlarged by said diffusible component of the step (iii) to give enlarged bubbles which are localised at said region of interest due to temporary blocking of a microcirculation at said region of interest by said enlarged bubbles; and
(b) said activating step (iii) facilitates extravasation of the therapeutic agent(s) administered in the step (i).
(iv) optionally, facilitating further extravasation of the therapeutic agent(s) administered in the step (i) by further ultrasound irradiation. 13. The method of claim 12, wherein the ultrasound irradiation applied in the step (iv) has a frequency in the range of 0.05 to 2 MHz. 14. The method of claim 12, wherein the ultrasound irradiation applied in the step (iii) has a mechanical index below 0.7. 15. The method of claim 12, which further comprises a quantification of an amount of the therapeutic agent(s) released by an analysis of an acoustic signature produced by the enlarged bubbles of the step (iii)(a). 16. A method of treatment of the mammalian subject comprising the method of claim 12. 17. A method of treatment of a mammalian subject comprising:
administering the cluster composition of claim 1, and applying a High Intensity Focused Ultrasound (HIFU) to a region of interest in the mammalian subject. 18. (canceled) 19. A method of an ultrasound imaging, comprising:
imaging a mammalian subject previously administered with the ultrasound contrast agent of claim 11. | 1,600 |
669 | 15,423,887 | 1,612 | The present invention concerns an orodispersible self-supporting film free from hydrocolloids comprising:
a) a film-forming substance consisting of a maltodextrin in an amount comprised between 40 and 80% by weight; b) one or more plasticizer in a total amount comprised between 15 and 55% by weight; e) a surfactant System in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, said orodispersible self-supporting film free from hydrocolloids further containing a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 1 and 20% by weight where the percentages are calculated on the total weight of said film. | 1. An orodispersible self-supporting film free from hydrocolloids comprising:
a) a film-forming substance consisting of a maltodextrin in an amount between 40 and 80% by weight; b) one or more plasticizer in a total amount comprised between 15 and 55% by weight; e) one or more surfactant system in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, said orodispersible self-supporting film free from hydrocolloids further containing a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 1 and 20% by weight, where the percentages are calculated on the total weight of said film. 2. The orodispersible self-supporting film according to claim 1 wherein the homopolymer or colpolymer of vinyl acetate is polyvinyl acetate or polyvinylpyrrolidone-vinyl acetate. 3. The orodispersible self-supporting film according to claim 2 wherein polyvinyl acetate or polyvinylpyrrolidone-vinyl acetate is present in a quantity comprised between 2 and 10% by weight. 4. The orodispersible self-supporting film according to claim 1 wherein the active ingredient is selected from an inhibitor of type V phosphodiesterase or a hormone. 5. The orodispersible self-supporting film according to claim 4 wherein the active ingredient is an inhibitor of type V phosphodiesterase selected from, sildenafil citrate and tadalafil. 6. The orodispersible self-supporting film according to claim 4 wherein the active ingredient is a hormone selected from progesterone and testosterone. 7. The orodispersible self-supporting film according to claim 1, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 2 and 10% by weight based on the film total weight. 8. The orodispersible self-supporting film according to claim 1, wherein the maltodextrin has a dextrose content, expressed in equivalents, of less than 50. 9. The orodispersible self-supporting film according to claim 8, wherein the dextrose content is between 5 and 40. 10. The orodispersible self-supporting film according to claim 1, wherein the plasticizer is selected from the class consisting of water, polyalcohols, citric acid esters, sebacic acid esters or mixtures thereof. 11. The orodispersible self-supporting film according to claim 10, wherein the plasticizer is selected from the class consisting of water, propylene glycol, glycerine, sorbitol, maltitol and mixtures thereof. 12. The orodispersible self-supporting film according to claim 1, wherein the surfactant system consists of one or more surfactants. 13. The orodispersible self-supporting film according to claim 12, wherein said surfactants are selected from the group consisting of sorbitan derivatives, sorbitol derivatives, esters of sucrose, fatty acid esters and mixtures thereof. 13. A process for the preparation of self-supporting film according to claim 1, comprising the following steps:
i) dispersing the maltodextrin, the plasticizer, the surfactant system, the homopolymer or copolymer of vinyl acetate and the active ingredient for therapeutic or food use in a polar solvent; ii) laminating the mixture obtained in the previous step on a release liner; iii) drying in the oven at a temperature preferably comprised between 50 and 140° C.; iv) removing the release liner from the film obtained in the preceding step; v) cutting the film with the desired dimensions and packaging them. | The present invention concerns an orodispersible self-supporting film free from hydrocolloids comprising:
a) a film-forming substance consisting of a maltodextrin in an amount comprised between 40 and 80% by weight; b) one or more plasticizer in a total amount comprised between 15 and 55% by weight; e) a surfactant System in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, said orodispersible self-supporting film free from hydrocolloids further containing a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 1 and 20% by weight where the percentages are calculated on the total weight of said film.1. An orodispersible self-supporting film free from hydrocolloids comprising:
a) a film-forming substance consisting of a maltodextrin in an amount between 40 and 80% by weight; b) one or more plasticizer in a total amount comprised between 15 and 55% by weight; e) one or more surfactant system in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, said orodispersible self-supporting film free from hydrocolloids further containing a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 1 and 20% by weight, where the percentages are calculated on the total weight of said film. 2. The orodispersible self-supporting film according to claim 1 wherein the homopolymer or colpolymer of vinyl acetate is polyvinyl acetate or polyvinylpyrrolidone-vinyl acetate. 3. The orodispersible self-supporting film according to claim 2 wherein polyvinyl acetate or polyvinylpyrrolidone-vinyl acetate is present in a quantity comprised between 2 and 10% by weight. 4. The orodispersible self-supporting film according to claim 1 wherein the active ingredient is selected from an inhibitor of type V phosphodiesterase or a hormone. 5. The orodispersible self-supporting film according to claim 4 wherein the active ingredient is an inhibitor of type V phosphodiesterase selected from, sildenafil citrate and tadalafil. 6. The orodispersible self-supporting film according to claim 4 wherein the active ingredient is a hormone selected from progesterone and testosterone. 7. The orodispersible self-supporting film according to claim 1, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 2 and 10% by weight based on the film total weight. 8. The orodispersible self-supporting film according to claim 1, wherein the maltodextrin has a dextrose content, expressed in equivalents, of less than 50. 9. The orodispersible self-supporting film according to claim 8, wherein the dextrose content is between 5 and 40. 10. The orodispersible self-supporting film according to claim 1, wherein the plasticizer is selected from the class consisting of water, polyalcohols, citric acid esters, sebacic acid esters or mixtures thereof. 11. The orodispersible self-supporting film according to claim 10, wherein the plasticizer is selected from the class consisting of water, propylene glycol, glycerine, sorbitol, maltitol and mixtures thereof. 12. The orodispersible self-supporting film according to claim 1, wherein the surfactant system consists of one or more surfactants. 13. The orodispersible self-supporting film according to claim 12, wherein said surfactants are selected from the group consisting of sorbitan derivatives, sorbitol derivatives, esters of sucrose, fatty acid esters and mixtures thereof. 13. A process for the preparation of self-supporting film according to claim 1, comprising the following steps:
i) dispersing the maltodextrin, the plasticizer, the surfactant system, the homopolymer or copolymer of vinyl acetate and the active ingredient for therapeutic or food use in a polar solvent; ii) laminating the mixture obtained in the previous step on a release liner; iii) drying in the oven at a temperature preferably comprised between 50 and 140° C.; iv) removing the release liner from the film obtained in the preceding step; v) cutting the film with the desired dimensions and packaging them. | 1,600 |
670 | 15,217,333 | 1,651 | The present invention relates generally to methods and compositions useful for application to skin and hair comprising saccharide isomerate, Alteromonas ferment extract, and a dermatologically acceptable vehicle, wherein the composition is capable of moisturizing and/or improving the appearance and/or condition of skin and/or hair. | 1. A method of treating a subject in need thereof comprising applying a topical composition comprising saccharide isomerate, Alteromonas ferment extract, and a dermatologically acceptable vehicle to skin, wherein at least one of filaggrin production is increased, skin moisture is increased, occludin production is increased, TNFα production is inhibited, an oxidant is reduced, hyaluronic acid production is increased, hyaluronidase is inhibited, or elastase is inhibited, wherein the saccharide isomerate comprises an exopolysaccharide synthesized by Vibrio alginolyticus, and wherein the Alteromonas ferment extract comprises an exopolysaccharide from Kopara. 2. The method of claim 1, wherein at least one of filaggrin production is increased, skin moisture is increased, occludin production is increased, TNFα production is inhibited, or an oxidant is reduced, and wherein at least one of hyaluronic acid production is increased, hyaluronidase is inhibited, or elastase is inhibited. 3. The method of claim 1, wherein the saccharide isomerate increases production of filaggrin, increases skin moisture, increases production of occluding, inhibits TNFα production, and/or reduces oxidants, and/or wherein the Alteromonas ferment extract increases production of hyaluronic acid, inhibits hyaluronidase, and/or inhibits elastase. 4. The method of claim 1, wherein the composition comprises 0.001% to 2% w/w of saccharide isomerate and 0.0001% to 0.2% w/w of Alteromonas ferment extract. 5. The method of claim 1, wherein the composition is formulated as at least one of a moisturizer, a mask, a freshener, or a cleanser. 6. The method of claim 1, wherein the dermatologically acceptable vehicle comprises water. The method of claim 6, wherein the composition comprises 45% to 95% w/w of water. 8. The method of claim 1, wherein the composition further comprises glycerin, phenoxyethanol, and triethanolamine. 9. The method of claim 8, wherein the composition comprises 0.1% to 15% w/w of glycerin, 0.1% to 5% w/w of phenoxyethanol, and 0.01% to 5% w/w of triethanolamine. 10. The method of claim 1, wherein skin of the subject is treated and wherein the skin has a normal skin type. 11. A topical composition comprising saccharide isomerate, Alteromonas ferment extract, and a dermatologically acceptable vehicle wherein the composition is capable of moisturizing at least one of skin and hair, wherein the saccharide isomerate comprises an exopolysaccharide synthesized by Vibrio alginolyticus, and wherein the Alteromonas ferment extract comprises an exopolysaccharide from Kopara. 12. The composition of claim 11, wherein the composition comprises an effective amount of saccharide isomerate capable of increasing production of filaggrin, increasing skin moisture, increasing production of occluding, inhibiting TNFα production, and/or reducing oxidants, and/or wherein the composition comprises an effective amount of Alteromonas ferment extract capable of increasing production of hyaluronic acid, inhibiting hyaluronidase, and/or inhibiting elastase. 13. The composition of claim 11, wherein the composition comprises 0.001% to 2% w/w of saccharide isomerate and 0.0001% to 0.2% w/w of Alteromonas ferment extract. 14. The composition of claim 11, wherein the dermatologically acceptable vehicle comprises water. 15. The composition of claim 14, wherein the composition comprises 45% to 95% w/w of water. 16. The composition of claim 11, further comprising glycerin, phenoxyethanol, and triethanolamine. 17. The composition of claim 16, wherein the composition comprises 0.1% to 15% w/w of glycerin, 0.1% to 5% w/w of phenoxyethanol, and 0.01% to 5% w/w of triethanolamine. 18. The composition of claim 11, wherein the composition is formulated as at least one of a moisturizer, a mask, a freshener, or a cleanser. 19. The composition of claim 11, wherein the composition is formulated to treat a subject's skin, wherein the skin has a normal skin type. | The present invention relates generally to methods and compositions useful for application to skin and hair comprising saccharide isomerate, Alteromonas ferment extract, and a dermatologically acceptable vehicle, wherein the composition is capable of moisturizing and/or improving the appearance and/or condition of skin and/or hair.1. A method of treating a subject in need thereof comprising applying a topical composition comprising saccharide isomerate, Alteromonas ferment extract, and a dermatologically acceptable vehicle to skin, wherein at least one of filaggrin production is increased, skin moisture is increased, occludin production is increased, TNFα production is inhibited, an oxidant is reduced, hyaluronic acid production is increased, hyaluronidase is inhibited, or elastase is inhibited, wherein the saccharide isomerate comprises an exopolysaccharide synthesized by Vibrio alginolyticus, and wherein the Alteromonas ferment extract comprises an exopolysaccharide from Kopara. 2. The method of claim 1, wherein at least one of filaggrin production is increased, skin moisture is increased, occludin production is increased, TNFα production is inhibited, or an oxidant is reduced, and wherein at least one of hyaluronic acid production is increased, hyaluronidase is inhibited, or elastase is inhibited. 3. The method of claim 1, wherein the saccharide isomerate increases production of filaggrin, increases skin moisture, increases production of occluding, inhibits TNFα production, and/or reduces oxidants, and/or wherein the Alteromonas ferment extract increases production of hyaluronic acid, inhibits hyaluronidase, and/or inhibits elastase. 4. The method of claim 1, wherein the composition comprises 0.001% to 2% w/w of saccharide isomerate and 0.0001% to 0.2% w/w of Alteromonas ferment extract. 5. The method of claim 1, wherein the composition is formulated as at least one of a moisturizer, a mask, a freshener, or a cleanser. 6. The method of claim 1, wherein the dermatologically acceptable vehicle comprises water. The method of claim 6, wherein the composition comprises 45% to 95% w/w of water. 8. The method of claim 1, wherein the composition further comprises glycerin, phenoxyethanol, and triethanolamine. 9. The method of claim 8, wherein the composition comprises 0.1% to 15% w/w of glycerin, 0.1% to 5% w/w of phenoxyethanol, and 0.01% to 5% w/w of triethanolamine. 10. The method of claim 1, wherein skin of the subject is treated and wherein the skin has a normal skin type. 11. A topical composition comprising saccharide isomerate, Alteromonas ferment extract, and a dermatologically acceptable vehicle wherein the composition is capable of moisturizing at least one of skin and hair, wherein the saccharide isomerate comprises an exopolysaccharide synthesized by Vibrio alginolyticus, and wherein the Alteromonas ferment extract comprises an exopolysaccharide from Kopara. 12. The composition of claim 11, wherein the composition comprises an effective amount of saccharide isomerate capable of increasing production of filaggrin, increasing skin moisture, increasing production of occluding, inhibiting TNFα production, and/or reducing oxidants, and/or wherein the composition comprises an effective amount of Alteromonas ferment extract capable of increasing production of hyaluronic acid, inhibiting hyaluronidase, and/or inhibiting elastase. 13. The composition of claim 11, wherein the composition comprises 0.001% to 2% w/w of saccharide isomerate and 0.0001% to 0.2% w/w of Alteromonas ferment extract. 14. The composition of claim 11, wherein the dermatologically acceptable vehicle comprises water. 15. The composition of claim 14, wherein the composition comprises 45% to 95% w/w of water. 16. The composition of claim 11, further comprising glycerin, phenoxyethanol, and triethanolamine. 17. The composition of claim 16, wherein the composition comprises 0.1% to 15% w/w of glycerin, 0.1% to 5% w/w of phenoxyethanol, and 0.01% to 5% w/w of triethanolamine. 18. The composition of claim 11, wherein the composition is formulated as at least one of a moisturizer, a mask, a freshener, or a cleanser. 19. The composition of claim 11, wherein the composition is formulated to treat a subject's skin, wherein the skin has a normal skin type. | 1,600 |
671 | 14,430,875 | 1,644 | The present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; wherein the nanomatrix is 1 to 500 nm in size. At least one first and one second stimulatory agents are attached to the same or to separate mobile matrices. If the stimulatory agents are attached to separate nanomatrices, fine-tuning of nanomatrices for the stimulation of the T cells is possible. Closed cell culture systems also benefit from this method. | 1. The use of a nanomatrix for in-vitro polyclonal stimulation of T cells, the nanomatrix comprising
a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; wherein the nanomatrix is 1 to 500 nm in size. 2. An in vitro method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; wherein the nanomatrix is 1 to 500 nm in size. 3. The method according to claim 2, wherein at least one first and one second stimulatory agents are attached to the same matrix of mobile polymer chains. 4. The method according to claim 2, wherein at least one first and one second stimulatory agents are attached to separate matrices of mobile polymer chains. 5. The method according to claim 4, wherein the ratio of nanomatrices to cells is larger than 500:1 allowing fine-tuning of T cell stimulation. 6. The method according to any one of claims 2 to 5, wherein one stimulatory agent is an anti-CD3 antibody or fragment thereof. 7. The method according to any one of claims 1 to 6, wherein the second stimulatory agent is an anti-CD28 antibody. 8. The method according to any one of claims 2 to 7, wherein the matrix of mobile polymer chains consists of a polymer of dextran. 9. The method according to any one of claims 2 to 8, wherein the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the matrix of mobile polymer chains. 10. The method according to any one of claims 2 to 9, wherein the stimulatory agent is attached at high density with more than 25 μg per mg nanomatrix. 11. The method according to any one of claims 2 to 10, wherein the stimulated T cells are Treg cells. 12. A method for polyclonal stimulation of T cells in a closed cell culture system, the method comprising contacting a cell culture comprising a population of T cells within said closed cell culture system with a dosage of nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains; and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; wherein the nanomatrix is 1 to 500 nm in size, and wherein said dosage of nanomatrix is applied sterile to said closed cell culture system, and wherein said dosage depends on the volume of the cell culture in said closed cell culture system. 13. A method according to claim 12, wherein said volume of the cell culture can be determined by a balance or a camera system without affecting the sterility barrier. 14. A method according to claims 12 and 13, wherein determining and applying said dosage are performed automatically. 15. A pharmaceutical composition comprising a population of stimulated T cells produced according to the method of claims 12-14. | The present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; wherein the nanomatrix is 1 to 500 nm in size. At least one first and one second stimulatory agents are attached to the same or to separate mobile matrices. If the stimulatory agents are attached to separate nanomatrices, fine-tuning of nanomatrices for the stimulation of the T cells is possible. Closed cell culture systems also benefit from this method.1. The use of a nanomatrix for in-vitro polyclonal stimulation of T cells, the nanomatrix comprising
a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; wherein the nanomatrix is 1 to 500 nm in size. 2. An in vitro method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; wherein the nanomatrix is 1 to 500 nm in size. 3. The method according to claim 2, wherein at least one first and one second stimulatory agents are attached to the same matrix of mobile polymer chains. 4. The method according to claim 2, wherein at least one first and one second stimulatory agents are attached to separate matrices of mobile polymer chains. 5. The method according to claim 4, wherein the ratio of nanomatrices to cells is larger than 500:1 allowing fine-tuning of T cell stimulation. 6. The method according to any one of claims 2 to 5, wherein one stimulatory agent is an anti-CD3 antibody or fragment thereof. 7. The method according to any one of claims 1 to 6, wherein the second stimulatory agent is an anti-CD28 antibody. 8. The method according to any one of claims 2 to 7, wherein the matrix of mobile polymer chains consists of a polymer of dextran. 9. The method according to any one of claims 2 to 8, wherein the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the matrix of mobile polymer chains. 10. The method according to any one of claims 2 to 9, wherein the stimulatory agent is attached at high density with more than 25 μg per mg nanomatrix. 11. The method according to any one of claims 2 to 10, wherein the stimulated T cells are Treg cells. 12. A method for polyclonal stimulation of T cells in a closed cell culture system, the method comprising contacting a cell culture comprising a population of T cells within said closed cell culture system with a dosage of nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains; and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; wherein the nanomatrix is 1 to 500 nm in size, and wherein said dosage of nanomatrix is applied sterile to said closed cell culture system, and wherein said dosage depends on the volume of the cell culture in said closed cell culture system. 13. A method according to claim 12, wherein said volume of the cell culture can be determined by a balance or a camera system without affecting the sterility barrier. 14. A method according to claims 12 and 13, wherein determining and applying said dosage are performed automatically. 15. A pharmaceutical composition comprising a population of stimulated T cells produced according to the method of claims 12-14. | 1,600 |
672 | 14,649,421 | 1,631 | Methods and compositions are provided for the identification of a molecular diagnostic test for cancer. The test identifies cancer subtypes that have an up-regulation or a down-regulation in biomarker expression related to angiogenesis and vascular development. The present invention can be used to determine whether patients with cancer are clinically responsive or non-responsive to a therapeutic regimen prior to administration of any anti-angiogenic agent. This test may be used in different cancer types and with different drugs that directly or indirectly affect angiogenesis or angiogenesis signalling. In addition, the present invention may be used as a prognostic indicator for certain cancer types. In particular, the present invention is directed to the use of certain combinations of predictive markers, wherein the expression of the predictive markers correlates with responsiveness or non-responsiveness to a therapeutic regimen. | 1. A method for predicting a subject's responsiveness to an anti-angiogenic therapeutic agent comprising:
obtaining a test sample from a subject; measuring expression levels of a biomarker panel from the test sample obtained from the subject, wherein the biomarker panel comprises one or more biomarkers selected from Table 2C; determining a sample expression score for the biomarker panel; comparing the sample expression score to a threshold score; and classifying the subject as responsive or non-responsive to the anti-angiogenic therapeutic agent based on whether the sample expression score is above or below the threshold expression score. 2. The method of claim 1, wherein the biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4. 3. The method of claim 1, wherein the biomarker panel comprises the biomarkers listed in Table 2C. 4. The method of claim 1, wherein the sample expression score is determined by measuring an expression level for each biomarker and multiplying it by a corresponding weight, wherein an expression signature defines the weight for each biomarker. 5. The method of claim 1, wherein the expression level of each biomarker in the biomarker panel is measured using a microarray, quantitative PCR, or an immunoassay. 6. The method of claim 4, wherein the expression signature is derived by a method comprising:
isolating total RNA from a sample set of diseased tissue: hybridizing the isolated total RNA to a microarray to obtain a sample expression data set; selecting those probes on the microarray with a variability above a defined significance threshold to form a preliminary biomarker set; generating clusters of biomarkers within the preliminary biomarker set having a similar expression profile using a clustering algorithm; identifying the biological processes or biological pathways for each cluster of biomarkers; selecting the cluster corresponding to the biological process or biological pathway of interest; and defining an expression signature by analyzing the expression levels of the biomarkers in the selected cluster in a the sample set of diseased tissue using a supervised or unsupervised training algorithm. 7. The method of claim 6, wherein the expression signature is defined using a PLS classifier, a SVM classifier, a SDA classifier, or a DSDA classifier. 8. The method of claim 1, wherein the test sample is a cancer test sample. 9. The method of claim 8, wherein the cancer is ovarian cancer, prostate, breast cancer, colon cancer, or colorectal cancer. 10. The method of claim 1, wherein the anti-angiogenic therapeutic agent is a VEGF-pathway-targeted therapeutic agent, an angiopoietin-TIE2 pathway inhibitor, an endogenous angiogenic inhibitor, or an immunomodulatory agent. 11. The method of claim 10, wherein the VEGF pathway-targeted therapeutic agents include Bevacizumab (Avastin), Afibercept (VEGF Trap), IMC-1121B (Ramucirumab), Imatinib (Gleevec), Sorafenib (Nexavar), Gefitinib (Iressa), Sunitinib (Sutent), Erlotinib, Tivozinib, Cediranib (Recentin), Pazopanib (Votrient), BIBF 1120 (Vargatef), Dovitinib, Semaxanib (Sugen), Axitinib (AG013736), Vandetanib (Zactima), Nilotinib (Tasigna), Dasatinib (Sprycel), Vatalanib, Motesanib, ABT-869, TKI-258 or a combination thereof. 12. The method of claim 10, wherein the angiopoietin-TIE2 pathway inhibitor includes AMG-386, PF-4856884 CVX-060, CEP-11981, CE-245677, MEDI-3617, CVX-241, Trastuzumab (Herceptin) or a combination thereof. 13. The method of claim 10, wherein the endogenous angiogenic inhibitors include Thombospondin, Endostatin, Tumstatin, Canstatin, Arrestin, Angiostatin, Vasostatin, Interferon alpha or a combination thereof. 14. The method of claim 10, wherein the immunomodulatory agents include thalidomide and lenalidomide. 15. The method of claim 1, further comprising determining the subject's prognosis, wherein a classification as responsive to the anti-angiogenic therapeutic agent indicates a good prognosis, the good prognosis indicating at least an increased expected survival time as compared to a classification as non-responsive to the anti-angiogenic therapeutic agent. 16. A method for predicting subject responsiveness to anti-angiogenic therapeutic agents and providing treatment prognosis, the method comprising:
obtaining a test sample from a subject with a disease or disorder; measuring expression levels of a first biomarker panel from the test sample of diseased tissue obtained from the subject, wherein the biomarker panel comprises one or more biomarkers selected from Table 2C; determining a first sample expression score for the first biomarker panel; comparing the first sample expression score to a first threshold score classifying the subject as having a good prognosis or bad prognosis based on whether the first sample expression score is above or below the first threshold score, wherein a good prognosis indicates at least an increased expected survival time as compared to a bad prognosis and wherein, in the event of a bad prognosis; measuring expression levels of a second biomarker panel from the test sample of diseased tissue obtained from the subject, wherein the second biomarker panel comprises one or more biomarkers selected from Table 2A or Table 2B; determining a second sample expression score for the second biomarker panel; comparing the second sample expression score to a second threshold score; and classifying the subject as responsive or non-responsive to an anti-angiogenic therapeutic agent based on whether the second sample expression score is above or below the second threshold expression score. 17. The method of claim 16, wherein the first biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4, and the second biomarker panel comprises one or more of CCDC80, INHBA, THBS2, SFRP2, MMP2, PLAU, FAP, FN1, COL8A1, and RAB31. 18. The method of claim 16, wherein the first biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4, and the second biomarker panel comprises one or more of TMEM200A, GJB2, MMP13, GFPT2, POSTN, BICC1, CDH11, MRVI1, PMP22, and COL11A1. 19. The method of claim 16, wherein the first biomarker panel consists of the biomarkers in Table 2C, and the second biomarker panel consists of the biomakers in Table 2A or 2B. 20. The method of claim 16, wherein the expression level of each biomarker in the biomarker panel is measured using a microarray, quantitative PCR, or an immunoassay. 21. A method for diagnosing a subject as having a cancer, or being susceptible to developing a cancer, that is responsive to an anti-angiogenic therapeutic, the method comprising;
obtaining a test sample from a subject having or suspected of having a cancer; measuring expression levels of a biomarker panel from the test sample obtained from the subject, wherein the biomarker panel comprises one or more biomarkers selected from Table 2C; determining a sample expression score for the biomarker panel; comparing the sample expression score to a threshold score; and diagnosing the subject as having a cancer, or being susceptible to developing a cancer that is responsive to an anti-angiogenic therapeutic based on whether the sample expression score is above or below the threshold expression score. 22. The method of claim 21, wherein the biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4. 23. The method of claim 21, wherein the biomarker panel comprises the biomarkers listed in Table 2C. 24. The method of claim 21, wherein the sample expression score is determined by measuring an expression level for each biomarker and multiplying it by a corresponding weight, wherein an expression signature defines the weight for each biomarker. 25. The method of claim 24, wherein the expression signature is defined using a PLS classifier, a SVM classifier, a SDA classifier, or a DSDA classifier. 26. The method of, wherein the cancer is ovarian cancer, glioblastoma, breast cancer, prostate cancer, colorectal cancer or colon cancer. 27. The method of claim 21, wherein the expression level of each biomarker in the biomarker panel is measured using a microarray, quantitative PCR, or an immunoassay. | Methods and compositions are provided for the identification of a molecular diagnostic test for cancer. The test identifies cancer subtypes that have an up-regulation or a down-regulation in biomarker expression related to angiogenesis and vascular development. The present invention can be used to determine whether patients with cancer are clinically responsive or non-responsive to a therapeutic regimen prior to administration of any anti-angiogenic agent. This test may be used in different cancer types and with different drugs that directly or indirectly affect angiogenesis or angiogenesis signalling. In addition, the present invention may be used as a prognostic indicator for certain cancer types. In particular, the present invention is directed to the use of certain combinations of predictive markers, wherein the expression of the predictive markers correlates with responsiveness or non-responsiveness to a therapeutic regimen.1. A method for predicting a subject's responsiveness to an anti-angiogenic therapeutic agent comprising:
obtaining a test sample from a subject; measuring expression levels of a biomarker panel from the test sample obtained from the subject, wherein the biomarker panel comprises one or more biomarkers selected from Table 2C; determining a sample expression score for the biomarker panel; comparing the sample expression score to a threshold score; and classifying the subject as responsive or non-responsive to the anti-angiogenic therapeutic agent based on whether the sample expression score is above or below the threshold expression score. 2. The method of claim 1, wherein the biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4. 3. The method of claim 1, wherein the biomarker panel comprises the biomarkers listed in Table 2C. 4. The method of claim 1, wherein the sample expression score is determined by measuring an expression level for each biomarker and multiplying it by a corresponding weight, wherein an expression signature defines the weight for each biomarker. 5. The method of claim 1, wherein the expression level of each biomarker in the biomarker panel is measured using a microarray, quantitative PCR, or an immunoassay. 6. The method of claim 4, wherein the expression signature is derived by a method comprising:
isolating total RNA from a sample set of diseased tissue: hybridizing the isolated total RNA to a microarray to obtain a sample expression data set; selecting those probes on the microarray with a variability above a defined significance threshold to form a preliminary biomarker set; generating clusters of biomarkers within the preliminary biomarker set having a similar expression profile using a clustering algorithm; identifying the biological processes or biological pathways for each cluster of biomarkers; selecting the cluster corresponding to the biological process or biological pathway of interest; and defining an expression signature by analyzing the expression levels of the biomarkers in the selected cluster in a the sample set of diseased tissue using a supervised or unsupervised training algorithm. 7. The method of claim 6, wherein the expression signature is defined using a PLS classifier, a SVM classifier, a SDA classifier, or a DSDA classifier. 8. The method of claim 1, wherein the test sample is a cancer test sample. 9. The method of claim 8, wherein the cancer is ovarian cancer, prostate, breast cancer, colon cancer, or colorectal cancer. 10. The method of claim 1, wherein the anti-angiogenic therapeutic agent is a VEGF-pathway-targeted therapeutic agent, an angiopoietin-TIE2 pathway inhibitor, an endogenous angiogenic inhibitor, or an immunomodulatory agent. 11. The method of claim 10, wherein the VEGF pathway-targeted therapeutic agents include Bevacizumab (Avastin), Afibercept (VEGF Trap), IMC-1121B (Ramucirumab), Imatinib (Gleevec), Sorafenib (Nexavar), Gefitinib (Iressa), Sunitinib (Sutent), Erlotinib, Tivozinib, Cediranib (Recentin), Pazopanib (Votrient), BIBF 1120 (Vargatef), Dovitinib, Semaxanib (Sugen), Axitinib (AG013736), Vandetanib (Zactima), Nilotinib (Tasigna), Dasatinib (Sprycel), Vatalanib, Motesanib, ABT-869, TKI-258 or a combination thereof. 12. The method of claim 10, wherein the angiopoietin-TIE2 pathway inhibitor includes AMG-386, PF-4856884 CVX-060, CEP-11981, CE-245677, MEDI-3617, CVX-241, Trastuzumab (Herceptin) or a combination thereof. 13. The method of claim 10, wherein the endogenous angiogenic inhibitors include Thombospondin, Endostatin, Tumstatin, Canstatin, Arrestin, Angiostatin, Vasostatin, Interferon alpha or a combination thereof. 14. The method of claim 10, wherein the immunomodulatory agents include thalidomide and lenalidomide. 15. The method of claim 1, further comprising determining the subject's prognosis, wherein a classification as responsive to the anti-angiogenic therapeutic agent indicates a good prognosis, the good prognosis indicating at least an increased expected survival time as compared to a classification as non-responsive to the anti-angiogenic therapeutic agent. 16. A method for predicting subject responsiveness to anti-angiogenic therapeutic agents and providing treatment prognosis, the method comprising:
obtaining a test sample from a subject with a disease or disorder; measuring expression levels of a first biomarker panel from the test sample of diseased tissue obtained from the subject, wherein the biomarker panel comprises one or more biomarkers selected from Table 2C; determining a first sample expression score for the first biomarker panel; comparing the first sample expression score to a first threshold score classifying the subject as having a good prognosis or bad prognosis based on whether the first sample expression score is above or below the first threshold score, wherein a good prognosis indicates at least an increased expected survival time as compared to a bad prognosis and wherein, in the event of a bad prognosis; measuring expression levels of a second biomarker panel from the test sample of diseased tissue obtained from the subject, wherein the second biomarker panel comprises one or more biomarkers selected from Table 2A or Table 2B; determining a second sample expression score for the second biomarker panel; comparing the second sample expression score to a second threshold score; and classifying the subject as responsive or non-responsive to an anti-angiogenic therapeutic agent based on whether the second sample expression score is above or below the second threshold expression score. 17. The method of claim 16, wherein the first biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4, and the second biomarker panel comprises one or more of CCDC80, INHBA, THBS2, SFRP2, MMP2, PLAU, FAP, FN1, COL8A1, and RAB31. 18. The method of claim 16, wherein the first biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4, and the second biomarker panel comprises one or more of TMEM200A, GJB2, MMP13, GFPT2, POSTN, BICC1, CDH11, MRVI1, PMP22, and COL11A1. 19. The method of claim 16, wherein the first biomarker panel consists of the biomarkers in Table 2C, and the second biomarker panel consists of the biomakers in Table 2A or 2B. 20. The method of claim 16, wherein the expression level of each biomarker in the biomarker panel is measured using a microarray, quantitative PCR, or an immunoassay. 21. A method for diagnosing a subject as having a cancer, or being susceptible to developing a cancer, that is responsive to an anti-angiogenic therapeutic, the method comprising;
obtaining a test sample from a subject having or suspected of having a cancer; measuring expression levels of a biomarker panel from the test sample obtained from the subject, wherein the biomarker panel comprises one or more biomarkers selected from Table 2C; determining a sample expression score for the biomarker panel; comparing the sample expression score to a threshold score; and diagnosing the subject as having a cancer, or being susceptible to developing a cancer that is responsive to an anti-angiogenic therapeutic based on whether the sample expression score is above or below the threshold expression score. 22. The method of claim 21, wherein the biomarker panel comprises one or more of IGF2, SOX11, INS, CXCL17, SLC5A1, TMEM45A, CXCR2P1, MFAP2, MATN3, or RTP4. 23. The method of claim 21, wherein the biomarker panel comprises the biomarkers listed in Table 2C. 24. The method of claim 21, wherein the sample expression score is determined by measuring an expression level for each biomarker and multiplying it by a corresponding weight, wherein an expression signature defines the weight for each biomarker. 25. The method of claim 24, wherein the expression signature is defined using a PLS classifier, a SVM classifier, a SDA classifier, or a DSDA classifier. 26. The method of, wherein the cancer is ovarian cancer, glioblastoma, breast cancer, prostate cancer, colorectal cancer or colon cancer. 27. The method of claim 21, wherein the expression level of each biomarker in the biomarker panel is measured using a microarray, quantitative PCR, or an immunoassay. | 1,600 |
673 | 15,134,112 | 1,618 | Described herein are single and dual modality bisphosphonate conjugated imaging probes. Also described herein are methods of synthesizing and using the single and dual modality bisphosphonate conjugated imaging probes. | 1. An imaging probe comprising:
a bisphosphonate; and a positron emission tomography (PET) radionuclide, wherein the PET radionuclide is conjugated to the bisphosphonate. 2. The imaging probe of claim 1, wherein the PET radionuclide is directly conjugated to the bisphosphonate via N-succinimidyl-4-[18F]fluorobenzoate and an epoxy-containing linker. 3. The imaging probe of claim 1, wherein the PET radionuclide conjugate is synthesized from N-succinimidyl-4-[18F]fluorobenzoate bound to a bisphosphonate through an epoxy-containing linker. 4. The imaging probe of claim 1, wherein the PET radionuclide is selected from the group consisting of: 18F, 11C, 60Cu, 61Cu, 64Cu, 86Y, 124I, and 89Zr. 5. The imaging probe of claim 1, wherein the bisphosphonate is a nitrogen containing bisphosphonate. 6. The imaging probe of claim 1, wherein the bisphosphonate is a non-nitrogen containing bisphosphonate. 7. The imaging probe of claim 1, wherein the bisphosphonate is selected from the group consisting of risedronate, zoledronate, minodronate, pamidronate, neridronate, olpadronate, alendronate, ibandronate and analogues thereof. 8. The imaging probe of claim 1, wherein an alpha-hydroxyl of the bisphosphonate is substituted with H. 9. The imaging probe of claim 8, wherein the bisphosphonate is (2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid) or a salt thereof. 10. The imaging probe of claim 1, wherein the bisphosphonate is (1-hydroxy-2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid) or a salt thereof. 11. The imaging probe of claim 1, further comprising a fluorescent molecule, wherein the fluorescent molecule is conjugated to the bisphosphonate via an epoxy-containing linker and wherein the PET radionuclide is directly conjugated to the fluorescent molecule. 12. The imaging probe of claim 11, wherein the fluorescent molecule is a boron-dipyrromethene (BPDIPY) dye. 13. The imaging probe of claim 11, wherein the bisphosphonate is a nitrogen containing bisphosphonate. 14. The imaging probe of claim 11, wherein the bisphosphonate is a non nitrogen containing bisphosphonate. 15. The imaging probe of claim 11, wherein the bisphosphonate is selected from the group consisting of risedronate, zoledronate, minodronate, pamidronate, neridronate, olpadronate, alendronate, ibandronate and analogues thereof. 16. The imaging probe of claim 11, wherein the alpha-hydroxyl of the bisphosphonate is substituted with H. 17. The imaging probe of claim 16, wherein the bisphosphonate is (2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid or a salt thereof. 18. The imaging probe of claim 11, wherein the bisphosphonate is (1-hydroxy-2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid) or a salt thereof. 19. A method comprising:
administering an imaging probe to a subject in need thereof, wherein the imaging probe comprises:
a bisphosphonate; and
a positron emission tomography (PET) radionuclide, wherein the PET radionuclide is conjugated to the bisphosphonate; and
obtaining an image of at least a portion of the subject using PET scanning. 20. The method of claim 19, wherein the PET radionuclide is directly conjugated to the bisphosphonate via N-succinimidyl-4-[18F]fluorobenzoate and an epoxy-containing linker. 21. The method of claim 19, wherein the imaging probe further comprises a fluorescent molecule, wherein the fluorescent molecule is conjugated to the bisphosphonate via an epoxy-containing linker and wherein the PET radionuclide is directly conjugated to the fluorescent molecule. 22. The method of claim 19, wherein the PET radionuclide is selected from the group consisting of: 18F, 11C, 60Cu, 61Cu, 64Cu, 86Y, 124I, and 89Zr. 23. The method of claim 19, wherein the subject in need thereof has, is suspected of having, or is otherwise predisposed to having a bone related disease, wherein the bone related disease is selected from the group consisting of: multiple myeloma, bone metastasis, Paget's disease, steroid induced osteoporosis, osteosarcoma osteoporosis, osteopenia, heterotopic ossification, osteoarthritis, rheumatoid arthritis, a disorder characterized by high bone turnover, and any combination thereof. | Described herein are single and dual modality bisphosphonate conjugated imaging probes. Also described herein are methods of synthesizing and using the single and dual modality bisphosphonate conjugated imaging probes.1. An imaging probe comprising:
a bisphosphonate; and a positron emission tomography (PET) radionuclide, wherein the PET radionuclide is conjugated to the bisphosphonate. 2. The imaging probe of claim 1, wherein the PET radionuclide is directly conjugated to the bisphosphonate via N-succinimidyl-4-[18F]fluorobenzoate and an epoxy-containing linker. 3. The imaging probe of claim 1, wherein the PET radionuclide conjugate is synthesized from N-succinimidyl-4-[18F]fluorobenzoate bound to a bisphosphonate through an epoxy-containing linker. 4. The imaging probe of claim 1, wherein the PET radionuclide is selected from the group consisting of: 18F, 11C, 60Cu, 61Cu, 64Cu, 86Y, 124I, and 89Zr. 5. The imaging probe of claim 1, wherein the bisphosphonate is a nitrogen containing bisphosphonate. 6. The imaging probe of claim 1, wherein the bisphosphonate is a non-nitrogen containing bisphosphonate. 7. The imaging probe of claim 1, wherein the bisphosphonate is selected from the group consisting of risedronate, zoledronate, minodronate, pamidronate, neridronate, olpadronate, alendronate, ibandronate and analogues thereof. 8. The imaging probe of claim 1, wherein an alpha-hydroxyl of the bisphosphonate is substituted with H. 9. The imaging probe of claim 8, wherein the bisphosphonate is (2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid) or a salt thereof. 10. The imaging probe of claim 1, wherein the bisphosphonate is (1-hydroxy-2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid) or a salt thereof. 11. The imaging probe of claim 1, further comprising a fluorescent molecule, wherein the fluorescent molecule is conjugated to the bisphosphonate via an epoxy-containing linker and wherein the PET radionuclide is directly conjugated to the fluorescent molecule. 12. The imaging probe of claim 11, wherein the fluorescent molecule is a boron-dipyrromethene (BPDIPY) dye. 13. The imaging probe of claim 11, wherein the bisphosphonate is a nitrogen containing bisphosphonate. 14. The imaging probe of claim 11, wherein the bisphosphonate is a non nitrogen containing bisphosphonate. 15. The imaging probe of claim 11, wherein the bisphosphonate is selected from the group consisting of risedronate, zoledronate, minodronate, pamidronate, neridronate, olpadronate, alendronate, ibandronate and analogues thereof. 16. The imaging probe of claim 11, wherein the alpha-hydroxyl of the bisphosphonate is substituted with H. 17. The imaging probe of claim 16, wherein the bisphosphonate is (2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid or a salt thereof. 18. The imaging probe of claim 11, wherein the bisphosphonate is (1-hydroxy-2-(pyridin-4-yl)ethane-1,1-diyl)bis(phosphonic acid) or a salt thereof. 19. A method comprising:
administering an imaging probe to a subject in need thereof, wherein the imaging probe comprises:
a bisphosphonate; and
a positron emission tomography (PET) radionuclide, wherein the PET radionuclide is conjugated to the bisphosphonate; and
obtaining an image of at least a portion of the subject using PET scanning. 20. The method of claim 19, wherein the PET radionuclide is directly conjugated to the bisphosphonate via N-succinimidyl-4-[18F]fluorobenzoate and an epoxy-containing linker. 21. The method of claim 19, wherein the imaging probe further comprises a fluorescent molecule, wherein the fluorescent molecule is conjugated to the bisphosphonate via an epoxy-containing linker and wherein the PET radionuclide is directly conjugated to the fluorescent molecule. 22. The method of claim 19, wherein the PET radionuclide is selected from the group consisting of: 18F, 11C, 60Cu, 61Cu, 64Cu, 86Y, 124I, and 89Zr. 23. The method of claim 19, wherein the subject in need thereof has, is suspected of having, or is otherwise predisposed to having a bone related disease, wherein the bone related disease is selected from the group consisting of: multiple myeloma, bone metastasis, Paget's disease, steroid induced osteoporosis, osteosarcoma osteoporosis, osteopenia, heterotopic ossification, osteoarthritis, rheumatoid arthritis, a disorder characterized by high bone turnover, and any combination thereof. | 1,600 |
674 | 15,569,552 | 1,653 | The present disclosure provides a culture device for enumerating colonies of microorganisms. The device can comprise a base, a coversheet, and a nonporous spacer member disposed therebetween. The spacer member comprises an aperture that defines a growth compartment. At least one adhesive layer adhered to the base or the coversheet in the growth compartment. A cold water-soluble gelling agent and a dry oxygen-scavenging reagent are adhered to the at least one adhesive layer. The oxygen-scavenging reagent consists essentially of particles having a diameter of less than 106 microns. | 1. A culture device for enumerating colonies of microorganisms, the device comprising:
a base having opposing inner and outer surfaces; a coversheet having opposing inner and outer surfaces; a spacer member disposed between the base and the cover sheet, wherein the spacer member is coupled to the base or the coversheet, wherein the spacer member comprises an aperture that defines a shape and a depth of a growth compartment, wherein the spacer member and the growth compartment are disposed between the inner surface of the base and the inner surface of the coversheet; a first adhesive layer adhered to the base in the growth compartment or a second adhesive layer adhered to the coversheet in the growth compartment; an effective amount of a dry oxygen-scavenging reagent adhered to the first adhesive layer or the second adhesive layer; and a dry, cold-water-soluble gelling agent adhered to the first adhesive layer or the second adhesive layer; wherein the coversheet is coupled to the base or to the spacer member. 2. The culture device of claim 1, wherein the oxygen-scavenging reagent consists essentially of particles having a diameter of less than 106 microns. 3. The culture device of claim 1, further comprising an effective amount of a dry carbon dioxide-generating reagent adhered to the first adhesive layer or the second adhesive layer in the growth compartment. 4. The culture device of claim 1, wherein the spacer member is nonporous. 5. The culture device of claim 1, further comprising a dry nutrient to facilitate growth of a target microorganism, wherein the nutrient is adhered to the base or the coversheet in the growth compartment. 6. The culture device of claim 1, further comprising a first indicator reagent for detecting growth of a target anaerobic microorganism, wherein the first indicator reagent is adhered to the base or the coversheet in the growth compartment. 7. The culture device of claim 1, further comprising an effective amount of a selective agent to inhibit growth of a non-target microorganism, wherein the selective agent is adhered to the base or the coversheet in the growth compartment. 8. The culture device of claim 1, further comprising an effective amount of a reducing agent, wherein the reducing agent is adhered to the base or the coversheet in the growth compartment. 9. The culture device of claim 3, wherein the carbon dioxide-generating reagent comprises a metal carbonate. 10. The culture device of claim 1:
wherein the inner surface of the base has the first adhesive adhered thereto; wherein one or more first component disposed in the growth compartment is adhered to the first adhesive; wherein the first component is selected from the group consisting of the gelling agent, the oxygen-scavenging reagent, the buffer reagent, the nutrient, the indicator reagent, the selective agent, the reducing agent, and a combination of any two or more of the foregoing first components. 11. The culture device of claim 1:
wherein the inner surface of the coversheet has the second adhesive adhered thereto; wherein one or more third component disposed in the growth compartment is adhered to the second adhesive; wherein the third component is selected from the group consisting of the gelling agent, the oxygen-scavenging reagent, the buffer reagent, the nutrient, the indicator reagent, the selective agent, the reducing agent, and a combination of any two or more of the foregoing third components. 12. The culture device of claim 1, further comprising a second indicator reagent disposed in fluid communication with the growth compartment, wherein the second indicator reagent indicates a presence of non-target microorganisms and target microorganisms. 13. A method of detecting a microorganism in a sample, the method comprising:
placing the culture device of claim 1 into an open configuration that provides access to the growth compartment therein; placing a predefined volume of aqueous liquid into the growth compartment; placing a sample into the growth compartment; closing the culture device;
wherein placing the aqueous liquid and the sample into the growth compartment and closing the culture device comprises forming a semi-solid microbial culture medium enclosed by the base, the coversheet, and the spacer of the culture device;
incubating the culture device for a period of time sufficient to permit formation of a microbial colony in the culture medium; and detecting the microbial colony. 14. The method of claim 13, wherein closing the culture device comprises leaving an open fluid pathway from a gaseous environment outside the culture device to the semisolid microbial culture medium enclosed in the growth compartment. 15. The method of claim 13, wherein placing the sample into the growth compartment comprises placing an additive into the growth compartment, wherein the additive comprises an effective amount of a selective agent or an indicator for detecting microbial growth. 16. The method of claim 15, wherein the additive comprises a selective agent, wherein the effective amount of the selective agent substantially permits growth of lactic acid bacteria in the culture device and the effective amount of selective agent substantially inhibits growth of E. coli, S. aureus, C. sporogenes, C. perfringens, Bacteroides fragilis, Prevotella melaninogencia, and/or a Fusobacterium species. 17. The method of claim 13, wherein incubating the culture device for a period of time comprises incubating the culture device for the period of time in an aerobic atmosphere. 18. The method of claim 13 wherein, after incubating the culture device, detecting the microbial colony further comprises enumerating one or more optically-detectable colonies in the culture device. 19. The method of claim 13, wherein detecting the microbial colony comprises optically detecting a gas bubble proximate the colony in the growth compartment. 20. The method of claim 15, wherein enumerating one or more microbial colonies further comprises distinguishing carbon dioxide-producing colonies from non-carbon dioxide-producing colonies. | The present disclosure provides a culture device for enumerating colonies of microorganisms. The device can comprise a base, a coversheet, and a nonporous spacer member disposed therebetween. The spacer member comprises an aperture that defines a growth compartment. At least one adhesive layer adhered to the base or the coversheet in the growth compartment. A cold water-soluble gelling agent and a dry oxygen-scavenging reagent are adhered to the at least one adhesive layer. The oxygen-scavenging reagent consists essentially of particles having a diameter of less than 106 microns.1. A culture device for enumerating colonies of microorganisms, the device comprising:
a base having opposing inner and outer surfaces; a coversheet having opposing inner and outer surfaces; a spacer member disposed between the base and the cover sheet, wherein the spacer member is coupled to the base or the coversheet, wherein the spacer member comprises an aperture that defines a shape and a depth of a growth compartment, wherein the spacer member and the growth compartment are disposed between the inner surface of the base and the inner surface of the coversheet; a first adhesive layer adhered to the base in the growth compartment or a second adhesive layer adhered to the coversheet in the growth compartment; an effective amount of a dry oxygen-scavenging reagent adhered to the first adhesive layer or the second adhesive layer; and a dry, cold-water-soluble gelling agent adhered to the first adhesive layer or the second adhesive layer; wherein the coversheet is coupled to the base or to the spacer member. 2. The culture device of claim 1, wherein the oxygen-scavenging reagent consists essentially of particles having a diameter of less than 106 microns. 3. The culture device of claim 1, further comprising an effective amount of a dry carbon dioxide-generating reagent adhered to the first adhesive layer or the second adhesive layer in the growth compartment. 4. The culture device of claim 1, wherein the spacer member is nonporous. 5. The culture device of claim 1, further comprising a dry nutrient to facilitate growth of a target microorganism, wherein the nutrient is adhered to the base or the coversheet in the growth compartment. 6. The culture device of claim 1, further comprising a first indicator reagent for detecting growth of a target anaerobic microorganism, wherein the first indicator reagent is adhered to the base or the coversheet in the growth compartment. 7. The culture device of claim 1, further comprising an effective amount of a selective agent to inhibit growth of a non-target microorganism, wherein the selective agent is adhered to the base or the coversheet in the growth compartment. 8. The culture device of claim 1, further comprising an effective amount of a reducing agent, wherein the reducing agent is adhered to the base or the coversheet in the growth compartment. 9. The culture device of claim 3, wherein the carbon dioxide-generating reagent comprises a metal carbonate. 10. The culture device of claim 1:
wherein the inner surface of the base has the first adhesive adhered thereto; wherein one or more first component disposed in the growth compartment is adhered to the first adhesive; wherein the first component is selected from the group consisting of the gelling agent, the oxygen-scavenging reagent, the buffer reagent, the nutrient, the indicator reagent, the selective agent, the reducing agent, and a combination of any two or more of the foregoing first components. 11. The culture device of claim 1:
wherein the inner surface of the coversheet has the second adhesive adhered thereto; wherein one or more third component disposed in the growth compartment is adhered to the second adhesive; wherein the third component is selected from the group consisting of the gelling agent, the oxygen-scavenging reagent, the buffer reagent, the nutrient, the indicator reagent, the selective agent, the reducing agent, and a combination of any two or more of the foregoing third components. 12. The culture device of claim 1, further comprising a second indicator reagent disposed in fluid communication with the growth compartment, wherein the second indicator reagent indicates a presence of non-target microorganisms and target microorganisms. 13. A method of detecting a microorganism in a sample, the method comprising:
placing the culture device of claim 1 into an open configuration that provides access to the growth compartment therein; placing a predefined volume of aqueous liquid into the growth compartment; placing a sample into the growth compartment; closing the culture device;
wherein placing the aqueous liquid and the sample into the growth compartment and closing the culture device comprises forming a semi-solid microbial culture medium enclosed by the base, the coversheet, and the spacer of the culture device;
incubating the culture device for a period of time sufficient to permit formation of a microbial colony in the culture medium; and detecting the microbial colony. 14. The method of claim 13, wherein closing the culture device comprises leaving an open fluid pathway from a gaseous environment outside the culture device to the semisolid microbial culture medium enclosed in the growth compartment. 15. The method of claim 13, wherein placing the sample into the growth compartment comprises placing an additive into the growth compartment, wherein the additive comprises an effective amount of a selective agent or an indicator for detecting microbial growth. 16. The method of claim 15, wherein the additive comprises a selective agent, wherein the effective amount of the selective agent substantially permits growth of lactic acid bacteria in the culture device and the effective amount of selective agent substantially inhibits growth of E. coli, S. aureus, C. sporogenes, C. perfringens, Bacteroides fragilis, Prevotella melaninogencia, and/or a Fusobacterium species. 17. The method of claim 13, wherein incubating the culture device for a period of time comprises incubating the culture device for the period of time in an aerobic atmosphere. 18. The method of claim 13 wherein, after incubating the culture device, detecting the microbial colony further comprises enumerating one or more optically-detectable colonies in the culture device. 19. The method of claim 13, wherein detecting the microbial colony comprises optically detecting a gas bubble proximate the colony in the growth compartment. 20. The method of claim 15, wherein enumerating one or more microbial colonies further comprises distinguishing carbon dioxide-producing colonies from non-carbon dioxide-producing colonies. | 1,600 |
675 | 15,031,141 | 1,618 | The present disclosure is directed to methods and a diagnostic kit for producing pericyte information from a subject for identifying and/or treating a subject condition. In one aspect, a method for identifying a subject condition is provided. The method includes administering intravenously to a subject an effective amount of a solution comprising fluorescent markers to selectively label a plurality of pericytes in the subject's body. The method also includes acquiring fluorescence signals originating from labeled pericytes to produce pericyte information associated with tissues of the subject's body. The method further includes generating a report identifying a subject condition using the pericyte information. In some aspects, a treatment may be adapted using the identified subject condition. | 1. A method for identifying a subject condition, the method comprising:
administering intravenously to a subject an effective amount of an injectable solution comprising fluorescent markers to selectively label a plurality of pericytes in the subject's body; acquiring, using an imaging system, fluorescence signals originating from labeled pericytes to produce pericyte information associated with tissues of the subject's body; and generating a report identifying a subject condition using the pericyte information. 2. The method of claim 1, wherein the fluorescent markers comprise fluorescently conjugated Dextran markers. 3. The method of claim 1, wherein the fluorescent markers have a molecular weight in a range between 3 kiloDaltons and 70 kiloDaltons. 4. The method of claim 1, wherein the injectable solution comprises fluorescent markers diluted in an artificial cerebrospinal fluid with a concentration having values between 16 mg/Kg and 25 mg/Kg. 5. The method of claim 1, wherein the imaging system includes a confocal imaging system or a two-photon microscopy system configured to acquire the fluorescence signals from intravenously labeled pericytes. 6. The method of claim 1, wherein generating the report further includes determining a likelihood of vasospasmic activity using the pericyte information. 7. The method of claim 1, wherein the tissues of the subject's body include blood vessels. 8. The method of claim 1, wherein the subject condition includes a blood flow constriction. 9. The method of claim 1, wherein the subject condition includes a neural degeneration. 10. A method for treating a subject condition, the method comprising:
administering intravenously to a subject an effective amount of an injectable solution comprising fluorescent markers to selectively label a plurality of pericytes in the subject's body; acquiring, using an imaging system, fluorescence signals originating from labeled pericytes to produce pericyte information associated with tissues of the subject's body; identifying a subject condition using the pericyte information; and adapting a treatment using the identified subject condition. 11. The method of claim 10, wherein the fluorescent markers comprise fluorescently conjugated Dextran markers. 12. The method of claim 10, wherein the fluorescent markers have a molecular weight in a range between 3 kiloDaltons and 70 kiloDaltons. 13. The method of claim 10, wherein the injectable solution comprises fluorescent markers diluted in an artificial cerebrospinal fluid with a concentration having values between 16 mg/Kg and 25 mg/Kg. 14. The method of claim 10, wherein the imaging system includes a confocal imaging system or a two-photon microscopy system to acquire the fluorescence signals from intravenously labeled pericytes. 15. The method of claim 10, wherein the tissues of the subject's body include blood vessels. 16. The method of claim 10, wherein the treatment includes administration of at least one drug. 17. A kit for determining pericyte information associated with tissues of a subject, the kit comprising:
an injectable solution comprising fluorescent markers for selectively labeling pericytes in a subject's body; and a delivery apparatus configured for intravenously administering an effective amount of the injectable solution to a subject to selectively label a plurality of pericytes of the subject. 18. The diagnostic kit of claim 17, wherein the fluorescent markers comprise fluorescently conjugated Dextran markers. 19. The diagnostic kit of claim 17, wherein the fluorescent markers have a molecular weight in a range between 3 kiloDaltons and 70 kiloDaltons. 20. The diagnostic kit of claim 17, wherein the injectable solution comprises fluorescent markers diluted in an artificial cerebrospinal fluid with a concentration having values between 16 mg/Kg and 25 mg/Kg. | The present disclosure is directed to methods and a diagnostic kit for producing pericyte information from a subject for identifying and/or treating a subject condition. In one aspect, a method for identifying a subject condition is provided. The method includes administering intravenously to a subject an effective amount of a solution comprising fluorescent markers to selectively label a plurality of pericytes in the subject's body. The method also includes acquiring fluorescence signals originating from labeled pericytes to produce pericyte information associated with tissues of the subject's body. The method further includes generating a report identifying a subject condition using the pericyte information. In some aspects, a treatment may be adapted using the identified subject condition.1. A method for identifying a subject condition, the method comprising:
administering intravenously to a subject an effective amount of an injectable solution comprising fluorescent markers to selectively label a plurality of pericytes in the subject's body; acquiring, using an imaging system, fluorescence signals originating from labeled pericytes to produce pericyte information associated with tissues of the subject's body; and generating a report identifying a subject condition using the pericyte information. 2. The method of claim 1, wherein the fluorescent markers comprise fluorescently conjugated Dextran markers. 3. The method of claim 1, wherein the fluorescent markers have a molecular weight in a range between 3 kiloDaltons and 70 kiloDaltons. 4. The method of claim 1, wherein the injectable solution comprises fluorescent markers diluted in an artificial cerebrospinal fluid with a concentration having values between 16 mg/Kg and 25 mg/Kg. 5. The method of claim 1, wherein the imaging system includes a confocal imaging system or a two-photon microscopy system configured to acquire the fluorescence signals from intravenously labeled pericytes. 6. The method of claim 1, wherein generating the report further includes determining a likelihood of vasospasmic activity using the pericyte information. 7. The method of claim 1, wherein the tissues of the subject's body include blood vessels. 8. The method of claim 1, wherein the subject condition includes a blood flow constriction. 9. The method of claim 1, wherein the subject condition includes a neural degeneration. 10. A method for treating a subject condition, the method comprising:
administering intravenously to a subject an effective amount of an injectable solution comprising fluorescent markers to selectively label a plurality of pericytes in the subject's body; acquiring, using an imaging system, fluorescence signals originating from labeled pericytes to produce pericyte information associated with tissues of the subject's body; identifying a subject condition using the pericyte information; and adapting a treatment using the identified subject condition. 11. The method of claim 10, wherein the fluorescent markers comprise fluorescently conjugated Dextran markers. 12. The method of claim 10, wherein the fluorescent markers have a molecular weight in a range between 3 kiloDaltons and 70 kiloDaltons. 13. The method of claim 10, wherein the injectable solution comprises fluorescent markers diluted in an artificial cerebrospinal fluid with a concentration having values between 16 mg/Kg and 25 mg/Kg. 14. The method of claim 10, wherein the imaging system includes a confocal imaging system or a two-photon microscopy system to acquire the fluorescence signals from intravenously labeled pericytes. 15. The method of claim 10, wherein the tissues of the subject's body include blood vessels. 16. The method of claim 10, wherein the treatment includes administration of at least one drug. 17. A kit for determining pericyte information associated with tissues of a subject, the kit comprising:
an injectable solution comprising fluorescent markers for selectively labeling pericytes in a subject's body; and a delivery apparatus configured for intravenously administering an effective amount of the injectable solution to a subject to selectively label a plurality of pericytes of the subject. 18. The diagnostic kit of claim 17, wherein the fluorescent markers comprise fluorescently conjugated Dextran markers. 19. The diagnostic kit of claim 17, wherein the fluorescent markers have a molecular weight in a range between 3 kiloDaltons and 70 kiloDaltons. 20. The diagnostic kit of claim 17, wherein the injectable solution comprises fluorescent markers diluted in an artificial cerebrospinal fluid with a concentration having values between 16 mg/Kg and 25 mg/Kg. | 1,600 |
676 | 11,885,716 | 1,611 | A transdermal therapeutic system which is free of fibrous constituents, and a method for the production of such a transdermal therapeutic system A preparation containing active substance is applied by a printing method onto the pressure-sensitive adhesive layer of the transdermal therapeutic system. | 1. A method for producing a transdermal therapeutic system, comprising the steps of:
providing an active substance-impermeable carrier layer or an optionally present reservoir layer or matrix layer with a fibre-free pressure-sensitive adhesive layer; by a printing method, applying individually dosed portions of a flowable, active substance-containing preparation onto the pressure-sensitive adhesive layer, said flowable active substance-containing preparation comprising a polymer which is also a constituent of the fibre-free pressure-sensitive adhesive layer to form a composite laminate; and in a further step, applying an active substance-impermeable, fibre-free backing layer onto the pressure-sensitive adhesive layer, said pressure-sensitive adhesive layer having been provided with the active substance-containing preparation, said transdermal therapeutic systems being able to be singularised by at least one of cutting and punching prior to or following said step of applying the active substance-containing preparation, from the composite laminate having been formed by said last-mentioned step of applying the active substance-containing preparation. 2. The method according to claim 1, wherein said printing method is a pad printing method. 3. The method according to claim 1, wherein said printing method is a method comprising the step of transferring the active substance-containing preparation onto the fibre-free pressure-sensitive adhesive layer by a distributor plate of an application device, said distributor plate being provided with at least one passage. 4. The method according to claim 1, wherein said polymer is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 5. The method according to claim 1, comprising the step of adding auxiliary substances to adjust the viscosity of the flowable, active substance-containing preparation. 6. The method according to claim 1, wherein said backing layer is selected from the group consisting of completely or partially light-permeable backing layers, light-impermeable backing layers and skin-coloured backing layers. 7. The method according to claim 6, wherein said backing layer is lacquered skin-coloured. 8. The method according to claim 1, wherein said backing layer is free of fibres and comprises a material selected from the group consisting of polyethylene terephthalate, plasticised vinyl acetate-vinyl chloride copolymers, nylon, ethylene-vinyl acetate copolymers, plasticized polyvinyl chloride, polyurethane, polyvinylidene chloride, polypropylene, polyethylene and polyamide. 9. The method according to claim 1, wherein said pressure-sensitive adhesive layer is free of fibres and comprises a material which is selected from the group consisting of pressure-sensitive adhesive polymers based on at least one of acrylic acid and methacrylic acids, esters of at least one of acrylic acid and methacrylic acid, polyacrylates, isobutylene, polyvinyl acetate, ethylene-vinyl acetate, at least one of natural and synthetic rubbers, styrene-diene copolymers, and hot-melt adhesives, or which is produced on the basis of pressure-sensitive adhesive silicone polymers or polysiloxanes. 10. The method according to claim 9, wherein said material is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 11. The method according to claim 1, wherein said active substance is a liquid or solid at room temperature. 12. The method according to claim 1, wherein said active substance is selected from the group consisting of alprostadil, buprenorphine, bupropion, clonidine, dexamethasone, dextroamphetamine, diclofenac, dihydrotestosterone, estradiol, estradiol in combination with androgenic or progestinic active substances, fentanyl, flurbiprofen, lidocaine, methylphenidate, nicotine, nitroglycerin, rotigotin, salicylic acid, scopolamine, testosterone, tulobuterol and essential oils. 13. A transdermal therapeutic system (TTS) comprising an active substance-impermeable backing layer, at least one pressure-sensitive adhesive layer onto which an active substance-containing preparation has been applied, and a detachable protective layer, wherein said transdermal therapeutic system is free of fibrous constituents and the active substance-containing preparation comprises a polymer which is also a constituent of the fibre-free pressure-sensitive adhesive layer. 14. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system, or the backing layer and the pressure-sensitive adhesive layer of the transdermal therapeutic system, is/are permeable to light. 15. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system is completely or partially impermeable to light. 16. The transdermal therapeutic system according to claim 15, wherein said backing layer is completely or partially impermeable to light. 17. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system is skin-coloured. 18. The transdermal therapeutic system according to claim 17, wherein said backing layer is lacquered skin-coloured. 19. The transdermal therapeutic system according to claim 13, wherein said backing layer is free of fibres and comprises a material selected from the group consisting of polyethylene terephthalate, plasticised vinyl acetate-vinyl chloride copolymers, nylon, ethylene-vinyl acetate copolymers, plasticized polyvinyl chloride, polyurethane, polyvinylidene chloride, polypropylene, polyethylene and polyamide. 20. The transdermal therapeutic system according to claim 13, wherein said pressure-sensitive adhesive layer is free of fibres and comprises a material selected from the group consisting of pressure-sensitive adhesive polymers based on at least one of acrylic acid and methacrylic acids, esters of at least one of acrylic acid and methacrylic acid, polyacrylates, isobutylene, polyvinyl acetate, ethylene-vinyl acetate and at least one of natural and synthetic rubbers, or which pressure-sensitive adhesive layer is produced on the basis of pressure-sensitive adhesive silicone polymers or polysiloxanes. 21. The transdermal therapeutic system according to claim 19, wherein said material is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 22. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system further comprises at least one further reservoir layer or matrix layer, said at least one further reservoir layer or matrix layer being free of fibres and comprising a material which is selected from the group consisting of pressure-sensitive adhesive polymers based on at least one of acrylic acid and methacrylic acids, esters of at least one of acrylic acid and methacrylic acid, polyacrylates, isobutylene, polyvinyl acetate, ethylene-vinyl acetate, at least one of natural and synthetic rubbers, styrene-diene copolymers, and hot-melt adhesives, or which is produced on the basis of pressure-sensitive adhesive silicone polymers or polysiloxanes. 23. The transdermal therapeutic system according to claim 22, wherein said material is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 24. The transdermal therapeutic system according to claim 13, wherein said active substance is a liquid or a solid at room temperature. 25. The transdermal therapeutic system according to claim 13, wherein said active substance of said active substance-containing preparation is selected from the group consisting of alprostadil, buprenorphine, bupropion, clonidine, dexamethasone, dextroamphetamine, diclofenac, dihydrotestosterone, estradiol, estradiol in combination with androgenic or progestinic active substances, fentanyl, flurbiprofen, lidocaine, methylphenidate, nicotine, nitroglycerin, rotigotin, salicylic acid, scopolamine, testosterone, tulobuterol and essential oils. 26. The method according to claim 9, wherein said natural and synthetic rubbers are selected from the group consisting of acrylonitrile-butadiene rubber, butyl rubber and neoprene rubber, and wherein said styrene-diene copolymers are styrene-butadiene block copolymers. 27. The transdermal therapeutic system according to claim 22, wherein said natural and synthetic rubbers are selected from the group consisting of acrylonitrile-butadiene rubber, butyl rubber and neoprene rubber, and wherein said styrene-diene copolymers are styrene-butadiene block copolymers. | A transdermal therapeutic system which is free of fibrous constituents, and a method for the production of such a transdermal therapeutic system A preparation containing active substance is applied by a printing method onto the pressure-sensitive adhesive layer of the transdermal therapeutic system.1. A method for producing a transdermal therapeutic system, comprising the steps of:
providing an active substance-impermeable carrier layer or an optionally present reservoir layer or matrix layer with a fibre-free pressure-sensitive adhesive layer; by a printing method, applying individually dosed portions of a flowable, active substance-containing preparation onto the pressure-sensitive adhesive layer, said flowable active substance-containing preparation comprising a polymer which is also a constituent of the fibre-free pressure-sensitive adhesive layer to form a composite laminate; and in a further step, applying an active substance-impermeable, fibre-free backing layer onto the pressure-sensitive adhesive layer, said pressure-sensitive adhesive layer having been provided with the active substance-containing preparation, said transdermal therapeutic systems being able to be singularised by at least one of cutting and punching prior to or following said step of applying the active substance-containing preparation, from the composite laminate having been formed by said last-mentioned step of applying the active substance-containing preparation. 2. The method according to claim 1, wherein said printing method is a pad printing method. 3. The method according to claim 1, wherein said printing method is a method comprising the step of transferring the active substance-containing preparation onto the fibre-free pressure-sensitive adhesive layer by a distributor plate of an application device, said distributor plate being provided with at least one passage. 4. The method according to claim 1, wherein said polymer is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 5. The method according to claim 1, comprising the step of adding auxiliary substances to adjust the viscosity of the flowable, active substance-containing preparation. 6. The method according to claim 1, wherein said backing layer is selected from the group consisting of completely or partially light-permeable backing layers, light-impermeable backing layers and skin-coloured backing layers. 7. The method according to claim 6, wherein said backing layer is lacquered skin-coloured. 8. The method according to claim 1, wherein said backing layer is free of fibres and comprises a material selected from the group consisting of polyethylene terephthalate, plasticised vinyl acetate-vinyl chloride copolymers, nylon, ethylene-vinyl acetate copolymers, plasticized polyvinyl chloride, polyurethane, polyvinylidene chloride, polypropylene, polyethylene and polyamide. 9. The method according to claim 1, wherein said pressure-sensitive adhesive layer is free of fibres and comprises a material which is selected from the group consisting of pressure-sensitive adhesive polymers based on at least one of acrylic acid and methacrylic acids, esters of at least one of acrylic acid and methacrylic acid, polyacrylates, isobutylene, polyvinyl acetate, ethylene-vinyl acetate, at least one of natural and synthetic rubbers, styrene-diene copolymers, and hot-melt adhesives, or which is produced on the basis of pressure-sensitive adhesive silicone polymers or polysiloxanes. 10. The method according to claim 9, wherein said material is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 11. The method according to claim 1, wherein said active substance is a liquid or solid at room temperature. 12. The method according to claim 1, wherein said active substance is selected from the group consisting of alprostadil, buprenorphine, bupropion, clonidine, dexamethasone, dextroamphetamine, diclofenac, dihydrotestosterone, estradiol, estradiol in combination with androgenic or progestinic active substances, fentanyl, flurbiprofen, lidocaine, methylphenidate, nicotine, nitroglycerin, rotigotin, salicylic acid, scopolamine, testosterone, tulobuterol and essential oils. 13. A transdermal therapeutic system (TTS) comprising an active substance-impermeable backing layer, at least one pressure-sensitive adhesive layer onto which an active substance-containing preparation has been applied, and a detachable protective layer, wherein said transdermal therapeutic system is free of fibrous constituents and the active substance-containing preparation comprises a polymer which is also a constituent of the fibre-free pressure-sensitive adhesive layer. 14. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system, or the backing layer and the pressure-sensitive adhesive layer of the transdermal therapeutic system, is/are permeable to light. 15. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system is completely or partially impermeable to light. 16. The transdermal therapeutic system according to claim 15, wherein said backing layer is completely or partially impermeable to light. 17. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system is skin-coloured. 18. The transdermal therapeutic system according to claim 17, wherein said backing layer is lacquered skin-coloured. 19. The transdermal therapeutic system according to claim 13, wherein said backing layer is free of fibres and comprises a material selected from the group consisting of polyethylene terephthalate, plasticised vinyl acetate-vinyl chloride copolymers, nylon, ethylene-vinyl acetate copolymers, plasticized polyvinyl chloride, polyurethane, polyvinylidene chloride, polypropylene, polyethylene and polyamide. 20. The transdermal therapeutic system according to claim 13, wherein said pressure-sensitive adhesive layer is free of fibres and comprises a material selected from the group consisting of pressure-sensitive adhesive polymers based on at least one of acrylic acid and methacrylic acids, esters of at least one of acrylic acid and methacrylic acid, polyacrylates, isobutylene, polyvinyl acetate, ethylene-vinyl acetate and at least one of natural and synthetic rubbers, or which pressure-sensitive adhesive layer is produced on the basis of pressure-sensitive adhesive silicone polymers or polysiloxanes. 21. The transdermal therapeutic system according to claim 19, wherein said material is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 22. The transdermal therapeutic system according to claim 13, wherein said transdermal therapeutic system further comprises at least one further reservoir layer or matrix layer, said at least one further reservoir layer or matrix layer being free of fibres and comprising a material which is selected from the group consisting of pressure-sensitive adhesive polymers based on at least one of acrylic acid and methacrylic acids, esters of at least one of acrylic acid and methacrylic acid, polyacrylates, isobutylene, polyvinyl acetate, ethylene-vinyl acetate, at least one of natural and synthetic rubbers, styrene-diene copolymers, and hot-melt adhesives, or which is produced on the basis of pressure-sensitive adhesive silicone polymers or polysiloxanes. 23. The transdermal therapeutic system according to claim 22, wherein said material is selected from the group consisting of cationic copolymers based on dimethylaminoethyl methacrylate and neutral methacrylic esters, and neutral copolymers based on butyl methacrylate and methyl methacrylates. 24. The transdermal therapeutic system according to claim 13, wherein said active substance is a liquid or a solid at room temperature. 25. The transdermal therapeutic system according to claim 13, wherein said active substance of said active substance-containing preparation is selected from the group consisting of alprostadil, buprenorphine, bupropion, clonidine, dexamethasone, dextroamphetamine, diclofenac, dihydrotestosterone, estradiol, estradiol in combination with androgenic or progestinic active substances, fentanyl, flurbiprofen, lidocaine, methylphenidate, nicotine, nitroglycerin, rotigotin, salicylic acid, scopolamine, testosterone, tulobuterol and essential oils. 26. The method according to claim 9, wherein said natural and synthetic rubbers are selected from the group consisting of acrylonitrile-butadiene rubber, butyl rubber and neoprene rubber, and wherein said styrene-diene copolymers are styrene-butadiene block copolymers. 27. The transdermal therapeutic system according to claim 22, wherein said natural and synthetic rubbers are selected from the group consisting of acrylonitrile-butadiene rubber, butyl rubber and neoprene rubber, and wherein said styrene-diene copolymers are styrene-butadiene block copolymers. | 1,600 |
677 | 13,956,744 | 1,617 | Achieving enhanced scent expression in antiperspirant compositions and methods. | 1) A method of enhancing fragrance expression, comprising formulating an antiperspirant composition so that no more than 25% by weight of the composition, of the non-perfume and non-antiperspirant active ingredients, have a polarity between 3 MPa1/2 and 15 MPa1/2. 2) The method of claim 1, wherein the composition comprises less than 4%, by weight of the composition of non-perfume and non-antiperspirant ingredients with a polarity of 15 MPa1/2 or more. 3) The method of claim 1, wherein the composition comprises less than 2%, by weight of the composition of non-perfume and non-antiperspirant ingredients with a polarity of 15 MPa1/2 or more. 4) The method of claim 1, wherein the composition is substantially free of castor wax. 5) The method of claim 1, wherein the composition has a hardness of about 600 gram force or more. 6) The method of claim 1, wherein the composition comprises no more than 20% of non-perfume and non-antiperspirant ingredients with a polarity of between 3.0 MPa1/2 and 15 MPa1/2. 7) The method of claim 1, wherein the non-perfume and non-antiperspirant ingredients are selected from the group consisting of primary structurant, additional chassis ingredients, other optional ingredients, and combinations thereof. 8) The method of claim 7, wherein the additional chassis ingredients comprise an additional structurant, a solvent, a non-volatile organic fluid, or combinations thereof. 9) The method of claim 7, wherein the other optional ingredients comprise a fragrance complexing material, a microcapsule, or a combination thereof. 10) The method of claim 9, wherein the fragrance complexing material comprises beta cyclodextrin. 11) The method of claim 9, wherein the microcapsule comprises a polyacrylate microcapsule, a gelatin microcapsule, or a combination thereof. 12) A method of enhancing fragrance expression in a solid antiperspirant product, comprising formulating an antiperspirant composition comprising about 7 to about 20% of a primary structurant; about 10 to about 25% of an antiperspirant active; from 0% to 10% of talc; a perfume; and additional chassis ingredients; wherein about 15% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2. 13) The method of claim 12, wherein 12% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2. 14) The method of claim 12, wherein 8% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2 and the composition comprises less than 8% talc. 15) The method of claim 12, wherein 5% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2 and the composition comprises less than 8% talc. 16) The method of claim 12, wherein 3% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2 and the composition comprises less than about 6% talc. 17) The method of claim 12, wherein the composition is free of dipropylene glycol; free of added water; or the combination thereof. 18) The method of claim 12, wherein the composition has a hardness of about 600 gram force or more. 19) The method of claim 12, wherein the additional chassis ingredients are selected from the group consisting of an additional structurant, a solvent, a non-volatile organic fluid, and combinations thereof. 20) The method of claim 12, wherein the additional chassis ingredients are selected from the group consisting of cyclopentasiloxane, behenyl alcohol, PPG-14, petrolatum, mineral oil, dimethicone, polyethylene, ozokerite, and combinations thereof. | Achieving enhanced scent expression in antiperspirant compositions and methods.1) A method of enhancing fragrance expression, comprising formulating an antiperspirant composition so that no more than 25% by weight of the composition, of the non-perfume and non-antiperspirant active ingredients, have a polarity between 3 MPa1/2 and 15 MPa1/2. 2) The method of claim 1, wherein the composition comprises less than 4%, by weight of the composition of non-perfume and non-antiperspirant ingredients with a polarity of 15 MPa1/2 or more. 3) The method of claim 1, wherein the composition comprises less than 2%, by weight of the composition of non-perfume and non-antiperspirant ingredients with a polarity of 15 MPa1/2 or more. 4) The method of claim 1, wherein the composition is substantially free of castor wax. 5) The method of claim 1, wherein the composition has a hardness of about 600 gram force or more. 6) The method of claim 1, wherein the composition comprises no more than 20% of non-perfume and non-antiperspirant ingredients with a polarity of between 3.0 MPa1/2 and 15 MPa1/2. 7) The method of claim 1, wherein the non-perfume and non-antiperspirant ingredients are selected from the group consisting of primary structurant, additional chassis ingredients, other optional ingredients, and combinations thereof. 8) The method of claim 7, wherein the additional chassis ingredients comprise an additional structurant, a solvent, a non-volatile organic fluid, or combinations thereof. 9) The method of claim 7, wherein the other optional ingredients comprise a fragrance complexing material, a microcapsule, or a combination thereof. 10) The method of claim 9, wherein the fragrance complexing material comprises beta cyclodextrin. 11) The method of claim 9, wherein the microcapsule comprises a polyacrylate microcapsule, a gelatin microcapsule, or a combination thereof. 12) A method of enhancing fragrance expression in a solid antiperspirant product, comprising formulating an antiperspirant composition comprising about 7 to about 20% of a primary structurant; about 10 to about 25% of an antiperspirant active; from 0% to 10% of talc; a perfume; and additional chassis ingredients; wherein about 15% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2. 13) The method of claim 12, wherein 12% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2. 14) The method of claim 12, wherein 8% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2 and the composition comprises less than 8% talc. 15) The method of claim 12, wherein 5% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2 and the composition comprises less than 8% talc. 16) The method of claim 12, wherein 3% or less, by weight of the composition, of the additional chassis ingredients have a polarity greater than about 3.0 MPa1/2 and the composition comprises less than about 6% talc. 17) The method of claim 12, wherein the composition is free of dipropylene glycol; free of added water; or the combination thereof. 18) The method of claim 12, wherein the composition has a hardness of about 600 gram force or more. 19) The method of claim 12, wherein the additional chassis ingredients are selected from the group consisting of an additional structurant, a solvent, a non-volatile organic fluid, and combinations thereof. 20) The method of claim 12, wherein the additional chassis ingredients are selected from the group consisting of cyclopentasiloxane, behenyl alcohol, PPG-14, petrolatum, mineral oil, dimethicone, polyethylene, ozokerite, and combinations thereof. | 1,600 |
678 | 15,323,736 | 1,627 | Disclosed herein is an aqueous tissue clearing solution for making a biological material, such as a tissue or an organ of an animal, or a bio-engineered collagen scaffold transparent. | 1. An aqueous solution for rendering a biomaterial transparent, comprising:
a first active compound of formula (I)
or
a second active compound of formula (II)
or a combination thereof;
a sufficient amount of a solvent that is water or a salt balanced buffer solution for dissolving the first or second, or a combination of the active compounds therein and thereby forming the aqueous solution;
wherein:
R1, R2, R3, R4 and R8 are independently H, or C1-6 alkyl substituted with at least two —OH;
R5 is —CH2OCH3 or C1-3 alkyl optionally substituted with at least one —OH;
R6 and R7 are independently acetyl or C1-3 alkyl;
X1, X2, and X3 are independently a halogen selected from the group consisting of Cl, Br, and I;
Y is
or C1-3 alkyl substituted with at least one —OH;
the pH of the aqueous solution is less than 11; and
the osmolarity of the aqueous solution is from 200 to 3,500 mOSm/L. 2. The aqueous solution of claim 1, wherein the pH of the aqueous solution is between 6 to 9, and the osmolarity of the aqueous solution is from 250 to 1,000 mOSm/L. 3. The aqueous solution of claim 2, wherein the first active compound of formula (I) is any of the followings, 4. The aqueous solution of claim 3, wherein the first active compound of formula (I) has a concentration of at least 10% (w/v) in the aqueous solution. 5. The aqueous solution of claim 2, wherein the second active compound of formula (II) is any of the followings, 6. The aqueous solution of claim 5, wherein the second active compound of formula (II) has a concentration of at least 10% (w/v) in the aqueous solution. 7. The aqueous solution of claim 2, further comprising an anti-freezer or a humectant, and either of which is present in the aqueous solution in an amount of 5% to 30% (w/v). 8. The aqueous solution of claim 7, wherein the anti-freezer is sugar, glycerol or dimethyl sulfoxide (DMSO). 9. The aqueous solution of claim 8, wherein the anti-freezer is DMSO, provided that the biomaterial has a Keratin surface. 10. The aqueous solution of claim 7, wherein the humectant is hyaluronic acid or polyhydric alcohol. 11. A method for rendering a biomaterial transparent, comprising:
subjecting the biomaterial to the treatment of the aqueous solution of claim 1 for a sufficient period of time so as to render the biomaterial transparent. 12. The method of claim 11, wherein the pH of the aqueous solution is between 6 to 9, and the osmolarity of the aqueous solution is from 250 to 1,000 mOSm/L. 13. The method of claim 12, wherein the first active compound of formula (I) is any of the followings, 14. The method of claim 13, wherein the first active compound of formula (I) has a concentration of at least 10% (w/v) in the aqueous solution. 15. The method of claim 12, wherein the second active compound of formula (II) is any of the followings, 16. The method of claim 15, wherein the second active compound of formula (II) has a concentration of at least 10% (w/v) in the aqueous solution. 17. The method of claim 12, further comprising an anti-freezer or a humectant, and either of which is present in the aqueous solution in an amount of 5% to 30% (w/v). 18. The method of claim 17, wherein the anti-freezer is sugar, glycerol or dimethyl sulfoxide (DMSO). 19. The method of claim 18, wherein the anti-freezer is DMSO, provided that the tissue has a Keratin surface. 20. The method of claim 17, wherein the humectant is hyaluronic acid or polyhydric alcohol. 21. The method of claim 11, wherein the biomaterial is a tissue derived from an insect or a mammal, or a bio-engineered collagen scaffold. 22. The method of claim 21, wherein the tissue derived from a mammal is selected from brain, heart, stomach, pancreas, intestine, liver, lung, and ear. 23. The method of claim 21, wherein the tissue is the head of an insect. 24. The method of claim 21, wherein the biomaterial is pre-labeled with an imaging tracer that is a dye, a fluorescent protein, or an antibody conjugated with a fluorescent marker. | Disclosed herein is an aqueous tissue clearing solution for making a biological material, such as a tissue or an organ of an animal, or a bio-engineered collagen scaffold transparent.1. An aqueous solution for rendering a biomaterial transparent, comprising:
a first active compound of formula (I)
or
a second active compound of formula (II)
or a combination thereof;
a sufficient amount of a solvent that is water or a salt balanced buffer solution for dissolving the first or second, or a combination of the active compounds therein and thereby forming the aqueous solution;
wherein:
R1, R2, R3, R4 and R8 are independently H, or C1-6 alkyl substituted with at least two —OH;
R5 is —CH2OCH3 or C1-3 alkyl optionally substituted with at least one —OH;
R6 and R7 are independently acetyl or C1-3 alkyl;
X1, X2, and X3 are independently a halogen selected from the group consisting of Cl, Br, and I;
Y is
or C1-3 alkyl substituted with at least one —OH;
the pH of the aqueous solution is less than 11; and
the osmolarity of the aqueous solution is from 200 to 3,500 mOSm/L. 2. The aqueous solution of claim 1, wherein the pH of the aqueous solution is between 6 to 9, and the osmolarity of the aqueous solution is from 250 to 1,000 mOSm/L. 3. The aqueous solution of claim 2, wherein the first active compound of formula (I) is any of the followings, 4. The aqueous solution of claim 3, wherein the first active compound of formula (I) has a concentration of at least 10% (w/v) in the aqueous solution. 5. The aqueous solution of claim 2, wherein the second active compound of formula (II) is any of the followings, 6. The aqueous solution of claim 5, wherein the second active compound of formula (II) has a concentration of at least 10% (w/v) in the aqueous solution. 7. The aqueous solution of claim 2, further comprising an anti-freezer or a humectant, and either of which is present in the aqueous solution in an amount of 5% to 30% (w/v). 8. The aqueous solution of claim 7, wherein the anti-freezer is sugar, glycerol or dimethyl sulfoxide (DMSO). 9. The aqueous solution of claim 8, wherein the anti-freezer is DMSO, provided that the biomaterial has a Keratin surface. 10. The aqueous solution of claim 7, wherein the humectant is hyaluronic acid or polyhydric alcohol. 11. A method for rendering a biomaterial transparent, comprising:
subjecting the biomaterial to the treatment of the aqueous solution of claim 1 for a sufficient period of time so as to render the biomaterial transparent. 12. The method of claim 11, wherein the pH of the aqueous solution is between 6 to 9, and the osmolarity of the aqueous solution is from 250 to 1,000 mOSm/L. 13. The method of claim 12, wherein the first active compound of formula (I) is any of the followings, 14. The method of claim 13, wherein the first active compound of formula (I) has a concentration of at least 10% (w/v) in the aqueous solution. 15. The method of claim 12, wherein the second active compound of formula (II) is any of the followings, 16. The method of claim 15, wherein the second active compound of formula (II) has a concentration of at least 10% (w/v) in the aqueous solution. 17. The method of claim 12, further comprising an anti-freezer or a humectant, and either of which is present in the aqueous solution in an amount of 5% to 30% (w/v). 18. The method of claim 17, wherein the anti-freezer is sugar, glycerol or dimethyl sulfoxide (DMSO). 19. The method of claim 18, wherein the anti-freezer is DMSO, provided that the tissue has a Keratin surface. 20. The method of claim 17, wherein the humectant is hyaluronic acid or polyhydric alcohol. 21. The method of claim 11, wherein the biomaterial is a tissue derived from an insect or a mammal, or a bio-engineered collagen scaffold. 22. The method of claim 21, wherein the tissue derived from a mammal is selected from brain, heart, stomach, pancreas, intestine, liver, lung, and ear. 23. The method of claim 21, wherein the tissue is the head of an insect. 24. The method of claim 21, wherein the biomaterial is pre-labeled with an imaging tracer that is a dye, a fluorescent protein, or an antibody conjugated with a fluorescent marker. | 1,600 |
679 | 15,599,399 | 1,658 | A wound dressing composition comprising a chitosan and an oxidized cellulose. For example, the composition may be in the form of a sponge formed by freeze drying an aqueous dispersion of chitosan and oxidized regenerated cellulose (ORC). The composition is especially suitable for the treatment of chronic wounds. | 1. A wound dressing composition comprising a chitosan and an oxidized cellulose. 2. The wound dressing composition of claim 1, wherein the chitosan and the oxidized cellulose are intimately mixed in the wound dressing composition. 3. The wound dressing composition of claim 1, wherein the oxidized cellulose is in the form of dispersed fibers or powder. 4. The wound dressing composition of claim 1, wherein the oxidized cellulose and the chitosan are dispersed in a semi-solid or solid vehicle for topical application. 5. The wound dressing composition of claim 1, wherein the oxidized cellulose comprises oxidized regenerated cellulose (ORC). 6. The wound dressing composition of claim 1, wherein the oxidized cellulose and the chitosan together make up at least 25% by weight of the wound dressing composition on a dry weight basis. 7. The wound dressing composition of claim 6, wherein the oxidized cellulose and the chitosan together make up at least 50% by weight of the wound dressing composition on a dry weight basis. 8. The wound dressing composition of claim 1, wherein the wound dressing composition further comprises from about 0.01 to about 5% by weight on a dry weight basis of one or more wound healing therapeutic substances. 9. The wound dressing composition of claim 1, wherein the wound dressing composition is a flexible film. 10. The wound dressing composition of claim 1, wherein the chitosan and the oxidized cellulose are present in a weight ratio of chitosan to oxidized cellulose from 1:10 to 10:1. 11. The wound dressing composition of claim 10, wherein the weight ratio of chitosan to oxidized cellulose is in the range 1:4 to 4:1. 12. A wound dressing comprising the wound dressing composition of claim 1. 13. The wound dressing of claim 12, wherein the wound dressing is sterile and packaged in a microorganism-impermeable container. 14. A method of providing therapy to a wound, the method comprising applying the wound dressing composition of claim 1. 15. The method of claim 14, wherein the wound is a chronic wound. 16. The method of claim 15, wherein the chronic wound is selected from the group consisting of venous ulcers, decubitis ulcers and diabetic ulcers. 17. A method of separating cell growth factors from a biological sample or organism, the method comprising:
contacting the biological sample or organism with the wound dressing composition of claim 1 to bind the growth factors to the wound dressing composition, wherein the contacting the wound dressing composition is carried out in vivo or in vitro; and, removing the wound dressing composition from the sample or organism. 18. A method according to claim 17, further comprising recovering the bound growth factors from the wound dressing composition after removing the wound dressing composition from the sample or organism. 19. A method of preparing an active wound dressing material comprising the steps of:
contacting the wound dressing composition according to claim 1 with a biological medium containing cell growth factors to bind the cell growth factors to the material; and (ii) washing and drying the wound dressing material having the cell growth factors bound thereto to form the active wound dressing material. 20. A method according to claim 19, wherein the cell growth factors comprise platelet derived growth factor. | A wound dressing composition comprising a chitosan and an oxidized cellulose. For example, the composition may be in the form of a sponge formed by freeze drying an aqueous dispersion of chitosan and oxidized regenerated cellulose (ORC). The composition is especially suitable for the treatment of chronic wounds.1. A wound dressing composition comprising a chitosan and an oxidized cellulose. 2. The wound dressing composition of claim 1, wherein the chitosan and the oxidized cellulose are intimately mixed in the wound dressing composition. 3. The wound dressing composition of claim 1, wherein the oxidized cellulose is in the form of dispersed fibers or powder. 4. The wound dressing composition of claim 1, wherein the oxidized cellulose and the chitosan are dispersed in a semi-solid or solid vehicle for topical application. 5. The wound dressing composition of claim 1, wherein the oxidized cellulose comprises oxidized regenerated cellulose (ORC). 6. The wound dressing composition of claim 1, wherein the oxidized cellulose and the chitosan together make up at least 25% by weight of the wound dressing composition on a dry weight basis. 7. The wound dressing composition of claim 6, wherein the oxidized cellulose and the chitosan together make up at least 50% by weight of the wound dressing composition on a dry weight basis. 8. The wound dressing composition of claim 1, wherein the wound dressing composition further comprises from about 0.01 to about 5% by weight on a dry weight basis of one or more wound healing therapeutic substances. 9. The wound dressing composition of claim 1, wherein the wound dressing composition is a flexible film. 10. The wound dressing composition of claim 1, wherein the chitosan and the oxidized cellulose are present in a weight ratio of chitosan to oxidized cellulose from 1:10 to 10:1. 11. The wound dressing composition of claim 10, wherein the weight ratio of chitosan to oxidized cellulose is in the range 1:4 to 4:1. 12. A wound dressing comprising the wound dressing composition of claim 1. 13. The wound dressing of claim 12, wherein the wound dressing is sterile and packaged in a microorganism-impermeable container. 14. A method of providing therapy to a wound, the method comprising applying the wound dressing composition of claim 1. 15. The method of claim 14, wherein the wound is a chronic wound. 16. The method of claim 15, wherein the chronic wound is selected from the group consisting of venous ulcers, decubitis ulcers and diabetic ulcers. 17. A method of separating cell growth factors from a biological sample or organism, the method comprising:
contacting the biological sample or organism with the wound dressing composition of claim 1 to bind the growth factors to the wound dressing composition, wherein the contacting the wound dressing composition is carried out in vivo or in vitro; and, removing the wound dressing composition from the sample or organism. 18. A method according to claim 17, further comprising recovering the bound growth factors from the wound dressing composition after removing the wound dressing composition from the sample or organism. 19. A method of preparing an active wound dressing material comprising the steps of:
contacting the wound dressing composition according to claim 1 with a biological medium containing cell growth factors to bind the cell growth factors to the material; and (ii) washing and drying the wound dressing material having the cell growth factors bound thereto to form the active wound dressing material. 20. A method according to claim 19, wherein the cell growth factors comprise platelet derived growth factor. | 1,600 |
680 | 13,321,723 | 1,631 | A method ( 10 ) for forming novel signatures of biological data is provided. The method comprises ranking features based on a trend value, which is created based on multiple signatures identified by a pattern discovery method. Furthermore, a device ( 30 ) and a computer program product ( 40 ), performing the steps according to the method ( 10 ) is provided. Uses of the method, for statistically analyzing clinical data, designing assays based on multiple molecular signatures and interpreting assays based on multiple molecular signatures are also provided. | 1. A method (10) for forming novel signatures of biological data comprising a number of features (F={F1, F2, . . . , Fn}) based on analyzing a set of multiple signatures of biological data, said method comprising the steps of:
obtaining (110) a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times, and for each run (ri) of a number of runs ({r1, r2, . . . , rN}): forming (120 a) a first matrix (Mi) of size (F×F), wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and forming (120 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix (T) is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi); forming (130) a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T); sorting (140) each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and forming (150) a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking. 2. The method according to claim 1, wherein said signatures are molecular signatures. 3. The method according to claim 2, wherein said molecular signatures are chosen from the group comprising: nucleotide sequence, genetic variation, methylation status or genetic expression. 4. The method according to claim 1, wherein said feature subset selection is pattern discovery. 5. A device (30) for forming novel signatures of biological data, said device comprising:
a first unit (310) configured to obtain a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times; a second unit (320) for each run (r1), of a number of runs ({r1, r2, . . . , rN}), configured to: form (320 a) a first matrix (Mi) of size (F×F), wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and form (320 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix (T) is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi); a third unit (330) configured to form a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T); a fourth unit (340) configured to sort each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and a fifth unit (350) configured to form a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking, said units being operatively connected to each other. 6. A computer program product comprising a computer program (40) for processing by a computer, the computer program comprising:
a first code segment (410) configured to obtain a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times; a second code segment (420), for each run (ri) of a number of runs ({r1, r2, . . . , rN}), configured to: form (420 a) a first matrix (Mi) of size (F×F), wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and form (420 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix (T) is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi); a third code segment (430) configured to form a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T); a fourth code segment (440) configured to sort each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and a fifth code segment (450) configured to form a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking. 7. The computer program product according to claim 6, comprising code segments arranged, when run by a device having computer-processing properties, configured to perform all of the method steps of:
obtaining (110) a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times, and for each run (ri) of a number of runs ({r1, r2, . . . , rN}:
forming (120 a) a first matrix (Mi) of size (F×F) wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and
forming (120 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix T is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi);
forming (130) a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T);
sorting (140) each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and
forming (150) a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking. 8. (canceled) 9. (canceled) 10. (canceled) | A method ( 10 ) for forming novel signatures of biological data is provided. The method comprises ranking features based on a trend value, which is created based on multiple signatures identified by a pattern discovery method. Furthermore, a device ( 30 ) and a computer program product ( 40 ), performing the steps according to the method ( 10 ) is provided. Uses of the method, for statistically analyzing clinical data, designing assays based on multiple molecular signatures and interpreting assays based on multiple molecular signatures are also provided.1. A method (10) for forming novel signatures of biological data comprising a number of features (F={F1, F2, . . . , Fn}) based on analyzing a set of multiple signatures of biological data, said method comprising the steps of:
obtaining (110) a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times, and for each run (ri) of a number of runs ({r1, r2, . . . , rN}): forming (120 a) a first matrix (Mi) of size (F×F), wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and forming (120 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix (T) is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi); forming (130) a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T); sorting (140) each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and forming (150) a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking. 2. The method according to claim 1, wherein said signatures are molecular signatures. 3. The method according to claim 2, wherein said molecular signatures are chosen from the group comprising: nucleotide sequence, genetic variation, methylation status or genetic expression. 4. The method according to claim 1, wherein said feature subset selection is pattern discovery. 5. A device (30) for forming novel signatures of biological data, said device comprising:
a first unit (310) configured to obtain a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times; a second unit (320) for each run (r1), of a number of runs ({r1, r2, . . . , rN}), configured to: form (320 a) a first matrix (Mi) of size (F×F), wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and form (320 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix (T) is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi); a third unit (330) configured to form a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T); a fourth unit (340) configured to sort each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and a fifth unit (350) configured to form a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking, said units being operatively connected to each other. 6. A computer program product comprising a computer program (40) for processing by a computer, the computer program comprising:
a first code segment (410) configured to obtain a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times; a second code segment (420), for each run (ri) of a number of runs ({r1, r2, . . . , rN}), configured to: form (420 a) a first matrix (Mi) of size (F×F), wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and form (420 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix (T) is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi); a third code segment (430) configured to form a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T); a fourth code segment (440) configured to sort each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and a fifth code segment (450) configured to form a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking. 7. The computer program product according to claim 6, comprising code segments arranged, when run by a device having computer-processing properties, configured to perform all of the method steps of:
obtaining (110) a set of multiple signatures of biological data by running a feature subset selection algorithm on the biological data N times, and for each run (ri) of a number of runs ({r1, r2, . . . , rN}:
forming (120 a) a first matrix (Mi) of size (F×F) wherein each matrix element (i,j) of the first matrix (Mi) quantifies each corresponding feature-pair (FiFj) based on their co-occurrence in the analyzed signatures in run ri; and
forming (120 b) a second matrix (T) of size (F×N), wherein each element (T(i,j)) of the second matrix T is assigned the sum of the corresponding row (Fi) of the corresponding first matrix (Mi);
forming (130) a vector (C) of size (F) wherein each element (CFi) of said vector is assigned the average of the corresponding row (Ti) for all columns (1 to N) of the second matrix (T);
sorting (140) each value of vector (C) in descending order, resulting in ranking each feature (Fi) represented by each element of the vector (C) in descending order; and
forming (150) a novel set of signatures based on the ranked features by combining at least two features of signatures with similar ranking. 8. (canceled) 9. (canceled) 10. (canceled) | 1,600 |
681 | 14,430,255 | 1,612 | The present invention concerns an orodispersible self-supporting film free from hydrocolloids comprising: a) a film-forming substance consisting of a maltodextrin in an amount comprised between 40 and 80% by weight; b) a plasticizer in an amount comprised between 15 and 55% by weight; e) a surfactant System in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, characterised in that it contains a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 2 and 10% by weight where the percentages are calculated on the total weight of said film | 1. An orodispersible self-supporting film free from hydrocolloids comprising:
a) a film-forming substance consisting of a maltodextrin in an amount between 40 and 80% by weight; b) a plasticizer in an amount comprised between 15 and 55% by weight; e) a surfactant system in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, said orodispersible self-supporting film free from hydrocolloids further containing a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 2 and 10% by weight, where the percentages are calculated on the total weight of said film. 2. The orodispersible self-supporting film according to claim 1 in which the homopolymer or copolymer of vinyl acetate is selected from the group consisting of polyvinylpyrrolidone vinyl acetate, ethylene vinyl acetate and polyvinyl acetate. 3. The orodispersible self-supporting film according to claim 2 wherein the homopolymer or copolymer of vinyl acetate is polyvinyl acetate. 4. The orodispersible self-supporting film according to claim 3, wherein the polyvinyl acetate has a weight molecular weight between 5000 and 500000. 5. The orodispersible self-supporting film according to claim 4 wherein the polyvinyl acetate has a molecular weight comprised between 250000 and 450000. 6. The orodispersible self-supporting film according to claim 1, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 2.5 and 10% by weight based on the film total weight. 7. The orodispersible self-supporting film according to claim 6, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 3 and 10% by weight based on the film total weight. 8. The orodispersible self-supporting film according to claim 6, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 3 and 6% by weight based on the film total weight. 9. The orodispersible self-supporting film according to claim 8, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 3 and 5.5% by weight based on the film total weight. 10. The orodispersible self-supporting film according to claim 1, wherein the maltodextrin has a dextrose content, expressed in equivalents, of less than 50. 11. The orodispersible self-supporting film according to claim 10, wherein the dextrose content is between 11 and 40. 12. The orodispersible self-supporting film according to claim 1, wherein the plasticizer is selected from the class consisting of polyalcohols, citric acid esters, sebacic acid esters or mixtures thereof. 13. The orodispersible self-supporting film according to claim 12, wherein the plasticizer is selected from the class consisting of propylene glycol, glycerine, sorbitol, maltitol and mixtures thereof. 14. The orodispersible self-supporting film according to claim 1, wherein the surfactant system consists of one or more surfactants, preferably selected from the group consisting of sorbitan derivatives, sorbitol derivatives, esters of sucrose, fatty acid esters and mixtures thereof. 15. The orodispersible self-supporting film according to claim 1, wherein the active ingredient for food use is an active ingredient with a breath freshening action or it is indicated for oral hygiene or is a natural active ingredient suitable for nutritional supplementation. 16. The orodispersible self-supporting film according to claim 15, wherein said active ingredient for food use with a breath freshening action is selected from menthol and eugenol. 17. The orodispersible self-supporting film according to claim 15, wherein said natural active ingredient suitable for nutritional supplementation is chosen from mineral salts normally used for such purpose, and vitamins. 18. The orodispersible self-supporting film according to claim 17, wherein the vitamin is ascorbic acid. 19. The orodispersible self-supporting film according to claim 1, characterized in that the active ingredient for therapeutic use is chosen from active ingredients with essentially topical action. 20. The orodispersible self-supporting film according to claim 19, wherein the active ingredient for therapeutic use is chosen from antibacterial, antifungal, antiviral and disinfectants of the oral cavity. 21. The orodispersible self-supporting film according to claim 1, wherein the active ingredient for therapeutic use is selected from the class consisting of active ingredients with essentially systemic action. 22. The orodispersible self-supporting film according to claim 21, wherein the active ingredient with essentially systemic action is selected from the class consisting of anti-inflammatory, analgesic, antipsychotic, hypnotic, anxiolytic, muscle relaxant, antimigraine, antiparkinsonian, antihemetic, antihistaminic, beta blocker, anti-asthmatic, anti-hypertensive, antitussive agent. 23. The orodispersible self-supporting film according to claim 22 wherein the active ingredient is selected from the group consisting of:
Piroxicam, Ketoprofen, Diclofenac, Tramadol, Morphine, Nifedipine, Diazepam, Lorazepam, Alprazolam, Bromazepam, Triazolam, Lormetazolam, Zolpidem, Paracetamol, Selegiline, Atenolol, Salbutamol, Sumatriptan, Clozapine, Cetirizine, and their pharmaceutically acceptable salts. 24. The orodispersible self-supporting film according to claim 1, further containing one or more excipients selected from the group consisting of non-sticking agents, sweeteners, flavourings, colorants, preservatives, buffer systems and their mixtures. 25. The orodispersible self-supporting film according to claim 24, wherein the non-sticking agent is selected from the class consisting of colloidal silica and talc. 26. The orodispersible self-supporting film according to claim 1 for therapeutic or food use. 27. A process for the preparation of the self-supporting film according to claim 1, comprising the following steps:
i) dispersing maltodextrin, plasticizer, surfactant system, homopolymer or copolymer of vinyl acetate, and active ingredient for therapeutic or food in a polar solvent, ii) laminating on a silicone paper the mixture obtained in the previous step, iii) drying, iv) removing the silicone paper from the film obtained in the preceding step. | The present invention concerns an orodispersible self-supporting film free from hydrocolloids comprising: a) a film-forming substance consisting of a maltodextrin in an amount comprised between 40 and 80% by weight; b) a plasticizer in an amount comprised between 15 and 55% by weight; e) a surfactant System in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, characterised in that it contains a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 2 and 10% by weight where the percentages are calculated on the total weight of said film1. An orodispersible self-supporting film free from hydrocolloids comprising:
a) a film-forming substance consisting of a maltodextrin in an amount between 40 and 80% by weight; b) a plasticizer in an amount comprised between 15 and 55% by weight; e) a surfactant system in an amount comprised between 0.5 and 6% by weight; d) an active ingredient for food or therapeutic use in an amount between 0.05 and 30% by weight, said orodispersible self-supporting film free from hydrocolloids further containing a homopolymer or a copolymer of vinyl acetate in a quantity comprised between 2 and 10% by weight, where the percentages are calculated on the total weight of said film. 2. The orodispersible self-supporting film according to claim 1 in which the homopolymer or copolymer of vinyl acetate is selected from the group consisting of polyvinylpyrrolidone vinyl acetate, ethylene vinyl acetate and polyvinyl acetate. 3. The orodispersible self-supporting film according to claim 2 wherein the homopolymer or copolymer of vinyl acetate is polyvinyl acetate. 4. The orodispersible self-supporting film according to claim 3, wherein the polyvinyl acetate has a weight molecular weight between 5000 and 500000. 5. The orodispersible self-supporting film according to claim 4 wherein the polyvinyl acetate has a molecular weight comprised between 250000 and 450000. 6. The orodispersible self-supporting film according to claim 1, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 2.5 and 10% by weight based on the film total weight. 7. The orodispersible self-supporting film according to claim 6, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 3 and 10% by weight based on the film total weight. 8. The orodispersible self-supporting film according to claim 6, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 3 and 6% by weight based on the film total weight. 9. The orodispersible self-supporting film according to claim 8, wherein the vinylacetate homopolymer or copolymer is contained in an amount comprised between 3 and 5.5% by weight based on the film total weight. 10. The orodispersible self-supporting film according to claim 1, wherein the maltodextrin has a dextrose content, expressed in equivalents, of less than 50. 11. The orodispersible self-supporting film according to claim 10, wherein the dextrose content is between 11 and 40. 12. The orodispersible self-supporting film according to claim 1, wherein the plasticizer is selected from the class consisting of polyalcohols, citric acid esters, sebacic acid esters or mixtures thereof. 13. The orodispersible self-supporting film according to claim 12, wherein the plasticizer is selected from the class consisting of propylene glycol, glycerine, sorbitol, maltitol and mixtures thereof. 14. The orodispersible self-supporting film according to claim 1, wherein the surfactant system consists of one or more surfactants, preferably selected from the group consisting of sorbitan derivatives, sorbitol derivatives, esters of sucrose, fatty acid esters and mixtures thereof. 15. The orodispersible self-supporting film according to claim 1, wherein the active ingredient for food use is an active ingredient with a breath freshening action or it is indicated for oral hygiene or is a natural active ingredient suitable for nutritional supplementation. 16. The orodispersible self-supporting film according to claim 15, wherein said active ingredient for food use with a breath freshening action is selected from menthol and eugenol. 17. The orodispersible self-supporting film according to claim 15, wherein said natural active ingredient suitable for nutritional supplementation is chosen from mineral salts normally used for such purpose, and vitamins. 18. The orodispersible self-supporting film according to claim 17, wherein the vitamin is ascorbic acid. 19. The orodispersible self-supporting film according to claim 1, characterized in that the active ingredient for therapeutic use is chosen from active ingredients with essentially topical action. 20. The orodispersible self-supporting film according to claim 19, wherein the active ingredient for therapeutic use is chosen from antibacterial, antifungal, antiviral and disinfectants of the oral cavity. 21. The orodispersible self-supporting film according to claim 1, wherein the active ingredient for therapeutic use is selected from the class consisting of active ingredients with essentially systemic action. 22. The orodispersible self-supporting film according to claim 21, wherein the active ingredient with essentially systemic action is selected from the class consisting of anti-inflammatory, analgesic, antipsychotic, hypnotic, anxiolytic, muscle relaxant, antimigraine, antiparkinsonian, antihemetic, antihistaminic, beta blocker, anti-asthmatic, anti-hypertensive, antitussive agent. 23. The orodispersible self-supporting film according to claim 22 wherein the active ingredient is selected from the group consisting of:
Piroxicam, Ketoprofen, Diclofenac, Tramadol, Morphine, Nifedipine, Diazepam, Lorazepam, Alprazolam, Bromazepam, Triazolam, Lormetazolam, Zolpidem, Paracetamol, Selegiline, Atenolol, Salbutamol, Sumatriptan, Clozapine, Cetirizine, and their pharmaceutically acceptable salts. 24. The orodispersible self-supporting film according to claim 1, further containing one or more excipients selected from the group consisting of non-sticking agents, sweeteners, flavourings, colorants, preservatives, buffer systems and their mixtures. 25. The orodispersible self-supporting film according to claim 24, wherein the non-sticking agent is selected from the class consisting of colloidal silica and talc. 26. The orodispersible self-supporting film according to claim 1 for therapeutic or food use. 27. A process for the preparation of the self-supporting film according to claim 1, comprising the following steps:
i) dispersing maltodextrin, plasticizer, surfactant system, homopolymer or copolymer of vinyl acetate, and active ingredient for therapeutic or food in a polar solvent, ii) laminating on a silicone paper the mixture obtained in the previous step, iii) drying, iv) removing the silicone paper from the film obtained in the preceding step. | 1,600 |
682 | 15,273,276 | 1,627 | A topical composition for treating fungal skin infections with associated inflammation may include making or administering to the affected skin area an antifungal and steroid, such as a corticosteroid. Certain compositions may include antifungals such as itraconazole or econazole and corticosteroids such as fluticasone, fluocinonide, or clobetasol. The antifungal and steroid may be within a cream. In one example, a commercially available econazole nitrate cream may be used in conjunction with a commercially available fluocinonide cream or commercially available clobetasol cream to treat the affected skin area. | 1. A method of compounding a topical composition for treating a fungal skin condition and associated inflammation, the method comprising:
combining (a) fluticasone, or pharmaceutically acceptable salt, ester, or derivative thereof, (b) itraconazole, or pharmaceutically acceptable salt, ester, or derivative thereof, (c) meadowsweet flower extract in a polyethylene glycol ointment base, and (d) polyethylene glycol 300 MW, NF liquid, wherein itraconazole is combined in an amount between approximately 3% and approximately 8% by weight of the compounded topical composition, and wherein fluticasone is combined in an amount between approximately 0.5% and 2.5% by weight of the compounded topical composition. 2. (canceled) 3. The method of claim 2, wherein the meadowsweet flower extract in a polyethylene glycol ointment base and the polyethylene glycol 300 MW, NF liquid are combined at a ratio between approximately 8:1 and approximately 9:1 meadowsweet flower extract in a polyethylene glycol ointment base to polyethylene glycol 300 MW, NF liquid. 4. (canceled) 5. The method of claim 4, wherein the polyethylene glycol 300 MW, NF liquid is combined in an amount between 5% and 15% by weight of the compounded topical composition and the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount between 90% and 80% by weight of the compounded topical composition. 6. The method of claim 5, wherein itraconazole is combined in an amount approximately 5% by weight of the compounded topical composition, wherein fluticasone is combined in an amount approximately 1% by weight of the compounded topical composition, wherein the polyethylene glycol 300 MW, NF liquid is combined in an amount approximately 10% by weight of the compounded topical composition, and wherein the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount approximately 84% by weight of the compounded topical composition. 7. The method of claim 1, wherein the itraconazole and fluticasone are combined with the meadowsweet flower extract in a polyethylene glycol ointment base and the polyethylene glycol 300 MW, NF liquid or components thereof in the form of bulk powders. 8. A topical composition for treating a fungal skin condition and associated inflammation made by a process comprising:
combining (a) between approximately 3% and approximately 8% by weight itraconazole, or pharmaceutically acceptable salt, ester, or derivative thereof; (b) between approximately 0.5% and 2.5% by weight fluticasone, or pharmaceutically acceptable salt, ester, or derivative thereof; (c) meadowsweet flower extract in a polyethylene glycol ointment base; and (d) polyethylene glycol 300 MW, NF liquid. 9. (canceled) 10. The topical composition made by the process of claim 8, wherein the meadowsweet flower extract in a polyethylene glycol ointment base and the polyethylene glycol 300 MW, NF liquid are combined at a ratio between approximately 8:1 and approximately 9:1 meadowsweet flower extract in a polyethylene glycol ointment base to polyethylene glycol 300 MW, NF liquid. 11. (canceled) 12. The topical composition made by the process of claim 11, wherein the polyethylene glycol 300 MW, NF liquid is combined in an amount between 5% and 15% by weight of the topical composition and the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount between 90% and 80% by weight of the topical composition. 13. The topical composition made by the process of claim 12, wherein the itraconazole is combined in an amount approximately 5% by weight of the topical composition, the fluticasone is combined in an amount approximately 1% by weight of the topical composition, the polyethylene glycol 300 MW, NF liquid is combined in an amount, approximately 10% by weight of the topical composition, and the and the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount approximately 84% by weight of the topical composition. 14-20. (canceled) | A topical composition for treating fungal skin infections with associated inflammation may include making or administering to the affected skin area an antifungal and steroid, such as a corticosteroid. Certain compositions may include antifungals such as itraconazole or econazole and corticosteroids such as fluticasone, fluocinonide, or clobetasol. The antifungal and steroid may be within a cream. In one example, a commercially available econazole nitrate cream may be used in conjunction with a commercially available fluocinonide cream or commercially available clobetasol cream to treat the affected skin area.1. A method of compounding a topical composition for treating a fungal skin condition and associated inflammation, the method comprising:
combining (a) fluticasone, or pharmaceutically acceptable salt, ester, or derivative thereof, (b) itraconazole, or pharmaceutically acceptable salt, ester, or derivative thereof, (c) meadowsweet flower extract in a polyethylene glycol ointment base, and (d) polyethylene glycol 300 MW, NF liquid, wherein itraconazole is combined in an amount between approximately 3% and approximately 8% by weight of the compounded topical composition, and wherein fluticasone is combined in an amount between approximately 0.5% and 2.5% by weight of the compounded topical composition. 2. (canceled) 3. The method of claim 2, wherein the meadowsweet flower extract in a polyethylene glycol ointment base and the polyethylene glycol 300 MW, NF liquid are combined at a ratio between approximately 8:1 and approximately 9:1 meadowsweet flower extract in a polyethylene glycol ointment base to polyethylene glycol 300 MW, NF liquid. 4. (canceled) 5. The method of claim 4, wherein the polyethylene glycol 300 MW, NF liquid is combined in an amount between 5% and 15% by weight of the compounded topical composition and the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount between 90% and 80% by weight of the compounded topical composition. 6. The method of claim 5, wherein itraconazole is combined in an amount approximately 5% by weight of the compounded topical composition, wherein fluticasone is combined in an amount approximately 1% by weight of the compounded topical composition, wherein the polyethylene glycol 300 MW, NF liquid is combined in an amount approximately 10% by weight of the compounded topical composition, and wherein the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount approximately 84% by weight of the compounded topical composition. 7. The method of claim 1, wherein the itraconazole and fluticasone are combined with the meadowsweet flower extract in a polyethylene glycol ointment base and the polyethylene glycol 300 MW, NF liquid or components thereof in the form of bulk powders. 8. A topical composition for treating a fungal skin condition and associated inflammation made by a process comprising:
combining (a) between approximately 3% and approximately 8% by weight itraconazole, or pharmaceutically acceptable salt, ester, or derivative thereof; (b) between approximately 0.5% and 2.5% by weight fluticasone, or pharmaceutically acceptable salt, ester, or derivative thereof; (c) meadowsweet flower extract in a polyethylene glycol ointment base; and (d) polyethylene glycol 300 MW, NF liquid. 9. (canceled) 10. The topical composition made by the process of claim 8, wherein the meadowsweet flower extract in a polyethylene glycol ointment base and the polyethylene glycol 300 MW, NF liquid are combined at a ratio between approximately 8:1 and approximately 9:1 meadowsweet flower extract in a polyethylene glycol ointment base to polyethylene glycol 300 MW, NF liquid. 11. (canceled) 12. The topical composition made by the process of claim 11, wherein the polyethylene glycol 300 MW, NF liquid is combined in an amount between 5% and 15% by weight of the topical composition and the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount between 90% and 80% by weight of the topical composition. 13. The topical composition made by the process of claim 12, wherein the itraconazole is combined in an amount approximately 5% by weight of the topical composition, the fluticasone is combined in an amount approximately 1% by weight of the topical composition, the polyethylene glycol 300 MW, NF liquid is combined in an amount, approximately 10% by weight of the topical composition, and the and the meadowsweet flower extract in a polyethylene glycol ointment base is combined in an amount approximately 84% by weight of the topical composition. 14-20. (canceled) | 1,600 |
683 | 16,053,537 | 1,625 | The invention provides synthetic methods and synthetic intermediates that are useful for preparing the antibacterial compound: | 1. The compound: 2. A method for preparing the compound:
comprising, converting an amide of the formula:
to the compound. 3. A method for preparing the compound:
comprising, converting a chloride of the formula:
to the compound. | The invention provides synthetic methods and synthetic intermediates that are useful for preparing the antibacterial compound:1. The compound: 2. A method for preparing the compound:
comprising, converting an amide of the formula:
to the compound. 3. A method for preparing the compound:
comprising, converting a chloride of the formula:
to the compound. | 1,600 |
684 | 12,598,703 | 1,613 | The present invention relates to specific synergistic active antimicrobial compositions for hygiene disinfectant products like antimicrobial liquid or solid soaps, disinfectant cleansing solutions and disinfectant emulsions for the treatment of skin surface, specifically for the treatment of hand surface and/or for the treatment of technical surfaces like e.g. surgery equipment comprising a mixture comprising or consisting of
a) an antimicrobial active amount of 1,2-decanediol of formula 1:
and
b) an antimicrobial active amount of one or more compounds selected from the group consisting of ethanol, propan-1-ol, propan-2-ol; chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride. | 1. A cosmetic, pharmaceutical and/or household product preparation, comprising a mixture comprising or consisting of
a) an antimicrobial active amount of 1,2-decanediol of formula 1:
and
b) an antimicrobial active amount of one or more compounds selected from the group consisting of ethanol, propan-1-ol, propan-2-ol, chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride. 2. The preparation according to claim 1, additionally comprising or consisting of
c) an antimicrobial active amount of one or more compounds selected from the group consisting of mecetroniumetil sulfate, undecyleneamidopropyltrimonium methosulfate, (ethylendioxy)dimethanol, benzyl-C12-18-alkyldimethylammoniumchloride, didecyldimethylammonium chloride, N,N-didecyl-N-methyl-poly(oxethyl)ammonium propionate, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, N-dodecylpropan-1,3-diamin, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, clorofen, 2-biphenyl-2-ol, chlorocresol, hydrogen peroxide, acetic acid, peracetic acid, glutaral and formaldehyde. 3. The preparation according to claim 1, wherein the preparation is in the form of an alcoholic, glycolic and/or aqueous cleansing solution. 4. The preparation according to claim 1, wherein the preparation is in the form of a disinfectant liquid or solid soap. 5. The preparation according to claim 1, wherein the preparation is in the form of an oil in water or water in oil emulsion. 6. The preparation according to claim 1, wherein constituent a) and/or one or more compounds selected from constituent b) are present in the finished preparation in an amount which is capable of disinfecting skin surface. 7. The preparation according to claim 1, wherein constituent a) and/or one or more compounds selected from constituent b) are present in the finished preparation in an amount which is capable of disinfecting technical surfaces. 8. A method for the manufacture of a cosmetic, pharmaceutical and/or household product antimicrobial preparation comprising adding a mixture comprising or consisting of
a) an antimicrobial active amount of 1,2-decanediol of formula 1:
and
b) an antimicrobial active amount of one or more compounds selected from the group consisting of ethanol, propan-1-ol, propan-2-ol, chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride. 9. A method for preparing technical surfaces comprising applying a preparation according to claim 1 as an antimicrobial preparation for technical surfaces. 10. A method for disinfection of skin comprising or consisting of the step:
a) application of a preparation according to claim 1 to skin surface. 11. A method for disinfection of technical surfaces, comprising or consisting of the step:
a) application of a preparation according to claim 1 to technical surfaces. 12. A process for the production of a cosmetic and/or pharmaceutical preparation for disinfection of skin and/or a household product preparation for disinfection of technical surfaces comprising or consisting of the following steps:
a) providing 1,2-decanediol of formula 1:
b) providing one or more compounds selected from the group of ethanol, propan-1-ol, propan-2-ol, chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride, and
c) mixing one or more compounds provided in step a) and one or more compounds provided in step b) to form a preparation according to claim 1. 13. A process according to claim 12, additionally comprising or consisting of the steps:
d) providing one or more compounds selected from the group consisting of mecetroniumetil sulfate, undecyleneamidopropyltrimonium methosulfate, (ethylendioxy)dimethanol, benzyl-C12-18-alkyldimethylammoniumchloride, didecyldimethylammonium chloride, N,N-didecyl-N-methyl-poly(oxethyl)ammonium propionate, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, N-dodecylpropan-1,3-diamin, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, clorofen, 2-biphenyl-2-ol, chlorocresol, hydrogen peroxide, acetic acid, peracetic acid, glutaral and formaldehyde, e) mixing one or more compounds provided in step d) with 1,2-decanediol provided in step a) and one or more compounds provided in step b) to form a preparation according to claim 2. 14. The preparation according to claim 6, wherein the skin surface comprises a hand surface. 15. The method according to claim 10, wherein the preparation is applied to a hand surface. 16. The process according to claim 12, wherein the preparation is for disinfection of a hand surface. | The present invention relates to specific synergistic active antimicrobial compositions for hygiene disinfectant products like antimicrobial liquid or solid soaps, disinfectant cleansing solutions and disinfectant emulsions for the treatment of skin surface, specifically for the treatment of hand surface and/or for the treatment of technical surfaces like e.g. surgery equipment comprising a mixture comprising or consisting of
a) an antimicrobial active amount of 1,2-decanediol of formula 1:
and
b) an antimicrobial active amount of one or more compounds selected from the group consisting of ethanol, propan-1-ol, propan-2-ol; chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride.1. A cosmetic, pharmaceutical and/or household product preparation, comprising a mixture comprising or consisting of
a) an antimicrobial active amount of 1,2-decanediol of formula 1:
and
b) an antimicrobial active amount of one or more compounds selected from the group consisting of ethanol, propan-1-ol, propan-2-ol, chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride. 2. The preparation according to claim 1, additionally comprising or consisting of
c) an antimicrobial active amount of one or more compounds selected from the group consisting of mecetroniumetil sulfate, undecyleneamidopropyltrimonium methosulfate, (ethylendioxy)dimethanol, benzyl-C12-18-alkyldimethylammoniumchloride, didecyldimethylammonium chloride, N,N-didecyl-N-methyl-poly(oxethyl)ammonium propionate, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, N-dodecylpropan-1,3-diamin, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, clorofen, 2-biphenyl-2-ol, chlorocresol, hydrogen peroxide, acetic acid, peracetic acid, glutaral and formaldehyde. 3. The preparation according to claim 1, wherein the preparation is in the form of an alcoholic, glycolic and/or aqueous cleansing solution. 4. The preparation according to claim 1, wherein the preparation is in the form of a disinfectant liquid or solid soap. 5. The preparation according to claim 1, wherein the preparation is in the form of an oil in water or water in oil emulsion. 6. The preparation according to claim 1, wherein constituent a) and/or one or more compounds selected from constituent b) are present in the finished preparation in an amount which is capable of disinfecting skin surface. 7. The preparation according to claim 1, wherein constituent a) and/or one or more compounds selected from constituent b) are present in the finished preparation in an amount which is capable of disinfecting technical surfaces. 8. A method for the manufacture of a cosmetic, pharmaceutical and/or household product antimicrobial preparation comprising adding a mixture comprising or consisting of
a) an antimicrobial active amount of 1,2-decanediol of formula 1:
and
b) an antimicrobial active amount of one or more compounds selected from the group consisting of ethanol, propan-1-ol, propan-2-ol, chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride. 9. A method for preparing technical surfaces comprising applying a preparation according to claim 1 as an antimicrobial preparation for technical surfaces. 10. A method for disinfection of skin comprising or consisting of the step:
a) application of a preparation according to claim 1 to skin surface. 11. A method for disinfection of technical surfaces, comprising or consisting of the step:
a) application of a preparation according to claim 1 to technical surfaces. 12. A process for the production of a cosmetic and/or pharmaceutical preparation for disinfection of skin and/or a household product preparation for disinfection of technical surfaces comprising or consisting of the following steps:
a) providing 1,2-decanediol of formula 1:
b) providing one or more compounds selected from the group of ethanol, propan-1-ol, propan-2-ol, chlorhexidine digluconate, chloroxylenol, triclosan, triclocarban, benzethonium chloride, methylbenzethonium chloride and benzalkonium chloride, and
c) mixing one or more compounds provided in step a) and one or more compounds provided in step b) to form a preparation according to claim 1. 13. A process according to claim 12, additionally comprising or consisting of the steps:
d) providing one or more compounds selected from the group consisting of mecetroniumetil sulfate, undecyleneamidopropyltrimonium methosulfate, (ethylendioxy)dimethanol, benzyl-C12-18-alkyldimethylammoniumchloride, didecyldimethylammonium chloride, N,N-didecyl-N-methyl-poly(oxethyl)ammonium propionate, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, N-dodecylpropan-1,3-diamin, N-(3-aminopropyl)-N-dodecylpropan-1,3-diamin, clorofen, 2-biphenyl-2-ol, chlorocresol, hydrogen peroxide, acetic acid, peracetic acid, glutaral and formaldehyde, e) mixing one or more compounds provided in step d) with 1,2-decanediol provided in step a) and one or more compounds provided in step b) to form a preparation according to claim 2. 14. The preparation according to claim 6, wherein the skin surface comprises a hand surface. 15. The method according to claim 10, wherein the preparation is applied to a hand surface. 16. The process according to claim 12, wherein the preparation is for disinfection of a hand surface. | 1,600 |
685 | 15,350,506 | 1,632 | Disclosed herein are devices for detecting presence and/or amount of an analyte, such as glucose, in a urine sample when diluted in a toilet bowl containing water and methods of using same. The disclosed devices eliminate the need to handle urine samples or a device that has been contacted with a urine sample, and can be conveniently disposed of by flushing into a sewage or septic system. | 1. A method of detecting glucose in urine of a diabetic subject taking an amount of a sodium glucose cotransporter 2 (SGLT2) inhibitor, the method comprising:
placing in a toilet bowl containing water a device that comprises an absorbent substrate having one or more reagents in an amount sufficient to react with glucose present in the toilet bowl containing water and urine and produce a visually detectable color change in a detection region of the device or a visually detectable color change in the water of the toilet bowl; collecting urine from the diabetic subject in the toilet bowl; waiting a predetermined period of time; and visually determining presence or absence of a color change in the detection region of the device or a visually detectable color change in the water of the toilet bowl to obtain a test result, recording the test result; communicating the test result to a health care provider; and monitoring the effectiveness of the SGLT2 inhibitor based on the test result, wherein glucose is determined to be present in the urine where the detection region changes color or the water of the toilet bowl changes color, wherein the presence of glucose above a threshold level indicates that the SGLT2 inhibitor is effective in the diabetic subject, wherein the device is paper, and wherein the device further comprises a positive control region spatially separated from the detection region and comprising glucose and one or more reagents for detection of glucose. 2-6. (canceled) 7. The method of claim 1, wherein collecting urine from the subject in the toilet bowl is achieved by the subject directly urinating into the toilet bowl. 8. The method of claim 1, wherein the one or more reagents for detection of glucose comprise glucose oxidase, a peroxidase, and a chromogen. 9. The method of claim 1, wherein the device is comprised of a biodegradable material. 10. (canceled) 11. The method of claim 1, wherein the device is flushable. 12. The method of claim 1, wherein the predetermined period of time is 15 seconds to 3 minutes. 13. (canceled) 14. The method of claim 1, wherein the device is placed in the toilet bowl prior to collection of urine from the subject in the toilet bowl. 15. The method of claim 1, wherein the device is placed in the toilet bowl after collection of urine from the subject in the toilet bowl. 16. A device for detecting glucose in a toilet bowl containing water and urine, the device comprising: an absorbent substrate comprising a first detection region having one or more reagents in an amount sufficient to react with glucose present in the toilet bowl containing water and urine and produce a visually detectable color change in the first detection region or a visually detectable color change in the water of the toilet bowl. 17. The device of claim 16, wherein the device is comprised of a biodegradable material. 18. The device of claim 16, wherein the biodegradable material is paper. 19. The device of claim 18, wherein the paper is toilet paper. 20. The method of claim 16, wherein the device is flushable. 21. The method of claim 1 further comprising administering the SGLT2 inhibitor to the subject. | Disclosed herein are devices for detecting presence and/or amount of an analyte, such as glucose, in a urine sample when diluted in a toilet bowl containing water and methods of using same. The disclosed devices eliminate the need to handle urine samples or a device that has been contacted with a urine sample, and can be conveniently disposed of by flushing into a sewage or septic system.1. A method of detecting glucose in urine of a diabetic subject taking an amount of a sodium glucose cotransporter 2 (SGLT2) inhibitor, the method comprising:
placing in a toilet bowl containing water a device that comprises an absorbent substrate having one or more reagents in an amount sufficient to react with glucose present in the toilet bowl containing water and urine and produce a visually detectable color change in a detection region of the device or a visually detectable color change in the water of the toilet bowl; collecting urine from the diabetic subject in the toilet bowl; waiting a predetermined period of time; and visually determining presence or absence of a color change in the detection region of the device or a visually detectable color change in the water of the toilet bowl to obtain a test result, recording the test result; communicating the test result to a health care provider; and monitoring the effectiveness of the SGLT2 inhibitor based on the test result, wherein glucose is determined to be present in the urine where the detection region changes color or the water of the toilet bowl changes color, wherein the presence of glucose above a threshold level indicates that the SGLT2 inhibitor is effective in the diabetic subject, wherein the device is paper, and wherein the device further comprises a positive control region spatially separated from the detection region and comprising glucose and one or more reagents for detection of glucose. 2-6. (canceled) 7. The method of claim 1, wherein collecting urine from the subject in the toilet bowl is achieved by the subject directly urinating into the toilet bowl. 8. The method of claim 1, wherein the one or more reagents for detection of glucose comprise glucose oxidase, a peroxidase, and a chromogen. 9. The method of claim 1, wherein the device is comprised of a biodegradable material. 10. (canceled) 11. The method of claim 1, wherein the device is flushable. 12. The method of claim 1, wherein the predetermined period of time is 15 seconds to 3 minutes. 13. (canceled) 14. The method of claim 1, wherein the device is placed in the toilet bowl prior to collection of urine from the subject in the toilet bowl. 15. The method of claim 1, wherein the device is placed in the toilet bowl after collection of urine from the subject in the toilet bowl. 16. A device for detecting glucose in a toilet bowl containing water and urine, the device comprising: an absorbent substrate comprising a first detection region having one or more reagents in an amount sufficient to react with glucose present in the toilet bowl containing water and urine and produce a visually detectable color change in the first detection region or a visually detectable color change in the water of the toilet bowl. 17. The device of claim 16, wherein the device is comprised of a biodegradable material. 18. The device of claim 16, wherein the biodegradable material is paper. 19. The device of claim 18, wherein the paper is toilet paper. 20. The method of claim 16, wherein the device is flushable. 21. The method of claim 1 further comprising administering the SGLT2 inhibitor to the subject. | 1,600 |
686 | 14,979,263 | 1,612 | The invention relates to photocurable compositions comprising at least one dispersion of acrylic polymer particles. | 1. A nail composition comprising at least one photocrosslinkable compound, at least one photoinitiator and at least one dispersion of acrylic polymer particles, wherein the composition is photocurable, and wherein the at least one dispersion of acrylic polymer particles comprises particles of a C1-C4 alkyl (meth)acrylate polymer and at least one stabilizer selected from the group consisting of isobornyl (meth)acrylate homopolymers and statistical copolymers of isobornyl (meth)acrylate and of C1-C4 alkyl (meth)acrylate present in an isobornyl (meth)acrylate/C1-C4 alkyl (meth)acrylate weight ratio of greater than 4. 2. The nail composition of claim 1, wherein the at least one dispersion of acrylic polymer particles is present in an amount ranging from 0.1% to 20% by weight with respect to the total weight of the composition. 3. (canceled) 4. The nail composition of claim 1, wherein the polymer of the particles is a methyl acrylate and/or ethyl acrylate polymer. 5. The nail composition of claim 1, wherein the polymer of the particles comprises an ethylenically unsaturated acid monomer or the anhydride thereof. 6. The nail composition of claim 5, wherein the polymer of the particles comprises from 80% to 100% by weight of C1-C4 alkyl (meth)acrylate and from 0 to 20% by weight of ethylenically unsaturated acid monomer, relative to the total weight of the polymer. 7. The nail composition of claim 1, wherein the polymer of the particles is at least one selected from the group consisting of: methyl acrylate homopolymers; ethyl acrylate homopolymers; methyl acrylate/ethyl acrylate copolymers; methyl acrylate/ethyl acrylate/acrylic acid copolymers; methyl acrylate/ethyl acrylate/maleic anhydride copolymers; methyl acrylate/acrylic acid copolymers; ethyl acrylate/acrylic acid copolymers; methyl acrylate/maleic anhydride copolymers; and ethyl acrylate/maleic anhydride copolymers. 8. The nail composition of claim 1, wherein the polymer particles have an average size ranging from 100 nm to 250 nm. 9. The nail composition of claim 1, wherein the stabilizer is at least one selected from the group consisting of: isobornyl acrylate homopolymers; statistical copolymers of isobornyl acrylate/methyl acrylate; statistical copolymers of isobornyl acrylate/methyl acrylate/ethyl acrylate; and statistical copolymers of isobornyl methacrylate/methyl acrylate. 10. A nail composition set comprising the nail composition of claim 1. 11. (canceled) 12. (canceled) 13. (canceled) 14. The nail composition of claim 1, wherein the at least one dispersion of acrylic polymer is a dispersion of C1-C4 alkyl (meth)acrylate polymer particles stabilized with a stabilizer based on isobornyl (meth)acrylate polymer in a hydrocarbon oil. 15. The nail composition of claim 14, wherein the at least one dispersion of acrylic polymer particles is present in an amount ranging from 0.1% to 20% by weight with respect to the total weight of the composition | The invention relates to photocurable compositions comprising at least one dispersion of acrylic polymer particles.1. A nail composition comprising at least one photocrosslinkable compound, at least one photoinitiator and at least one dispersion of acrylic polymer particles, wherein the composition is photocurable, and wherein the at least one dispersion of acrylic polymer particles comprises particles of a C1-C4 alkyl (meth)acrylate polymer and at least one stabilizer selected from the group consisting of isobornyl (meth)acrylate homopolymers and statistical copolymers of isobornyl (meth)acrylate and of C1-C4 alkyl (meth)acrylate present in an isobornyl (meth)acrylate/C1-C4 alkyl (meth)acrylate weight ratio of greater than 4. 2. The nail composition of claim 1, wherein the at least one dispersion of acrylic polymer particles is present in an amount ranging from 0.1% to 20% by weight with respect to the total weight of the composition. 3. (canceled) 4. The nail composition of claim 1, wherein the polymer of the particles is a methyl acrylate and/or ethyl acrylate polymer. 5. The nail composition of claim 1, wherein the polymer of the particles comprises an ethylenically unsaturated acid monomer or the anhydride thereof. 6. The nail composition of claim 5, wherein the polymer of the particles comprises from 80% to 100% by weight of C1-C4 alkyl (meth)acrylate and from 0 to 20% by weight of ethylenically unsaturated acid monomer, relative to the total weight of the polymer. 7. The nail composition of claim 1, wherein the polymer of the particles is at least one selected from the group consisting of: methyl acrylate homopolymers; ethyl acrylate homopolymers; methyl acrylate/ethyl acrylate copolymers; methyl acrylate/ethyl acrylate/acrylic acid copolymers; methyl acrylate/ethyl acrylate/maleic anhydride copolymers; methyl acrylate/acrylic acid copolymers; ethyl acrylate/acrylic acid copolymers; methyl acrylate/maleic anhydride copolymers; and ethyl acrylate/maleic anhydride copolymers. 8. The nail composition of claim 1, wherein the polymer particles have an average size ranging from 100 nm to 250 nm. 9. The nail composition of claim 1, wherein the stabilizer is at least one selected from the group consisting of: isobornyl acrylate homopolymers; statistical copolymers of isobornyl acrylate/methyl acrylate; statistical copolymers of isobornyl acrylate/methyl acrylate/ethyl acrylate; and statistical copolymers of isobornyl methacrylate/methyl acrylate. 10. A nail composition set comprising the nail composition of claim 1. 11. (canceled) 12. (canceled) 13. (canceled) 14. The nail composition of claim 1, wherein the at least one dispersion of acrylic polymer is a dispersion of C1-C4 alkyl (meth)acrylate polymer particles stabilized with a stabilizer based on isobornyl (meth)acrylate polymer in a hydrocarbon oil. 15. The nail composition of claim 14, wherein the at least one dispersion of acrylic polymer particles is present in an amount ranging from 0.1% to 20% by weight with respect to the total weight of the composition | 1,600 |
687 | 14,910,577 | 1,662 | The present invention provides wheat grain comprising (1,3;1,4)-β-D-glucan (BG). The wheat grain is characterised by one or more of the following features; a BG content of at least 3% (w/w); the BG of the grain has a DP3/DP4 ratio between about 1.0 and about 2.0 or between about 1.0 and 2.3; and the BG is partially water soluble such that between 8.0% and about 25% or between about 10% and about 25% of the BG of the grain is water soluble. The present invention also provides uses of this grain. | 1. Wheat grain comprising (1,3;1,4)-β-D-glucan (BG), which is characterised by one or more or all of:
a) wherein the BG content of the grain is at least 3% (w/w);
b) wherein the BG of the grain has a DP3/DP4 ratio between about 1.0 and about 2.0 or between about 1.0 and 2.3; and
c) wherein the BG is partially water soluble such that between 8.0% and about 25% or between about 10% and about 25% of the BG of the grain is water soluble. 2. The grain of claim 1, wherein the BG content of the wheat grain is at least 4% (w/w), at least 5% (w/w), at least 6% (w/w), between 3% (w/w) and about 8% (w/w), between about 4% (w/w) and about 8% (w/w), between about 5% (w/w) and about 8% (w/w), about 4% (w/w), about 5% (w/w), about 6% (w/w), about 7% (w/w), or about 8% (w/w). 3. The grain of claim 1, wherein the BG comprises an increased proportion of water-soluble BG relative to a corresponding wild-type grain, as determined by treatment of a sample of wholemeal flour obtained from the grain with (i) 80% ethanol for 1 hour at 80° C., followed by (ii) solubilisation of BG in aqueous buffer for 2 hours at 37° C., and (iii) determination of the level of BG solubilised from the sample. 4. (canceled) 5. The grain of claim 1, wherein the BG of the grain has a DP3/DP4 ratio of less than about 2.5, preferably less than about 2.4, less than about 2.3, less than about 2.2, less than about 2.1, less than about 2.0, less than about 1.9, less than about 1.8, about 2.5, about 2,4, about 2.3, about 2.2, about 2.1, about 2.0, about 1.9, about 1.8, or between about 1.6 and about 2.5. 6. (canceled) 7. The grain of claim 1, which is transgenic, such as comprising one or more exogenous polynucleotides which encode one or more CslF polypeptides and/or a CslH polypeptide, a herbicide tolerance gene, or a polynucleotide which encodes a silencing RNA molecule. 8. (canceled) 9. (canceled) 10. (canceled) 11. The grain of claim 1, further comprising an exogenous CslH polypeptide. 12. The grain of claim 1 which is non-shrunken and/or has a weight of at least 25 mg, preferably at least about 30 mg or at least about 35 mg, or between about 30 mg and about 50 mg. 13. The grain of claim 1 which is capable of producing a wheat plant which is male and female fertile, or a wheat plant which is essentially the same in morphology and/or growth rate as a corresponding wild-type plant. 14. (canceled) 15. The grain of claim 1 comprising starch, wherein the starch of the grain has an amylose content of at least 60% (w/w), or at least 67 (w/w) or at least 70% (w/w) as a proportion of the extractable starch of the grain. 16. (canceled) 17. The grain of claim 1, wherein the starch content of the grain is at least 30%, preferably at least 35%, at least 40% or at least 45% as a percentage of the total grain weight. 18. The grain of claim 1, wherein the plant is hexaploid wheat, preferably Triticum aestivum. 19. The grain of claim 1, wherein the starch of the grain is characterised by one or more of properties selected from the group consisting of:
a. comprising at least 2% resistant starch; b. comprising a glycaemic index (GI) of less than 55; c. comprising less than 20% amylopectin as a proportion of the starch content of the grain; d. comprised in starch granules which have an altered morphology relative to wild-type wheat starch granules; e. comprised in starch granules that exhibit reduced granule birefringence under polarised light relative to wild-type wheat starch granules; f. when the grain is milled to flour, such flour exhibits reduced swelling volume; g. modified chain length distribution and/or branching frequency relative to wild-type wheat starch; h. delayed end of gelatinisation temperature and higher peak temperature; i. reduced viscosity (peak viscosity, pasting temperature); j. increased molecular weight of amylopectin; and k. modified % crystallinity or % A-type or B-type starch, relative to wild-type wheat starch granules or starch. 20. The grain of claim 1 which is processed so that it is no longer capable of germinating, such as kibbled, cracked, par-boiled, rolled, pearled, milled or ground grain. 21. (canceled) 22. A wheat plant which is capable of producing the grain of claim 1, which comprises one or more exogenous polynucleotides which encode a CslF6 polypeptide and/or a CslH polypeptide. 23. (canceled) 26. (canceled) 27. A composition comprising isolated wheat BG and arabinoxylan (AX) produced from the grain of claim 1, the BG and AX being soluble in aqueous media, and the BG having a DP3/DP4 ratio of less than 2.0 and predominantly a molecular weight of at least about 100 kDa. 28. (canceled) 29. A food ingredient that comprises the grain of claim 1 for sale. 30. (canceled) 31. (canceled) 32. (canceled) 33. (canceled) 34. (canceled) 35. A method of producing a wheat plant that produces grain according to claim 1 comprising the steps of (i) introducing one or more exogenous polynucleotides which encode one or more CslF polypeptides or a CslF polypeptide and a CslH polypeptide into a progenitor wheat cell, and (ii) producing a wheat plant from the wheat cell of (i). 36. (canceled) 37. (canceled) 38. A method of selecting a wheat plant, the method comprising (i) determining the amount of BG in grain obtained from each of at least two wheat plants, and (ii) selecting a plant from (i) which produces grain according to claim 1. 39-46. (canceled) 47. A method of producing at least partially purified BG or starch, comprising the steps of i) obtaining wheat grain according to claim 1, and ii) extracting the BG or starch from the grain, thereby producing the BG or starch. 48-53. (canceled) 54. A method of altering one or more physiological parameters in an animal, or of preventing or reducing the severity or incidence of a disease, the method comprising providing to the animal, the grain of claim 1, wherein the altered physiological parameter or reduced severity or incidence of disease is relative to providing to the animal the same amount of corresponding wild-type wheat grain, wheat plant, or composition or product made therefrom. 55-58. (canceled) | The present invention provides wheat grain comprising (1,3;1,4)-β-D-glucan (BG). The wheat grain is characterised by one or more of the following features; a BG content of at least 3% (w/w); the BG of the grain has a DP3/DP4 ratio between about 1.0 and about 2.0 or between about 1.0 and 2.3; and the BG is partially water soluble such that between 8.0% and about 25% or between about 10% and about 25% of the BG of the grain is water soluble. The present invention also provides uses of this grain.1. Wheat grain comprising (1,3;1,4)-β-D-glucan (BG), which is characterised by one or more or all of:
a) wherein the BG content of the grain is at least 3% (w/w);
b) wherein the BG of the grain has a DP3/DP4 ratio between about 1.0 and about 2.0 or between about 1.0 and 2.3; and
c) wherein the BG is partially water soluble such that between 8.0% and about 25% or between about 10% and about 25% of the BG of the grain is water soluble. 2. The grain of claim 1, wherein the BG content of the wheat grain is at least 4% (w/w), at least 5% (w/w), at least 6% (w/w), between 3% (w/w) and about 8% (w/w), between about 4% (w/w) and about 8% (w/w), between about 5% (w/w) and about 8% (w/w), about 4% (w/w), about 5% (w/w), about 6% (w/w), about 7% (w/w), or about 8% (w/w). 3. The grain of claim 1, wherein the BG comprises an increased proportion of water-soluble BG relative to a corresponding wild-type grain, as determined by treatment of a sample of wholemeal flour obtained from the grain with (i) 80% ethanol for 1 hour at 80° C., followed by (ii) solubilisation of BG in aqueous buffer for 2 hours at 37° C., and (iii) determination of the level of BG solubilised from the sample. 4. (canceled) 5. The grain of claim 1, wherein the BG of the grain has a DP3/DP4 ratio of less than about 2.5, preferably less than about 2.4, less than about 2.3, less than about 2.2, less than about 2.1, less than about 2.0, less than about 1.9, less than about 1.8, about 2.5, about 2,4, about 2.3, about 2.2, about 2.1, about 2.0, about 1.9, about 1.8, or between about 1.6 and about 2.5. 6. (canceled) 7. The grain of claim 1, which is transgenic, such as comprising one or more exogenous polynucleotides which encode one or more CslF polypeptides and/or a CslH polypeptide, a herbicide tolerance gene, or a polynucleotide which encodes a silencing RNA molecule. 8. (canceled) 9. (canceled) 10. (canceled) 11. The grain of claim 1, further comprising an exogenous CslH polypeptide. 12. The grain of claim 1 which is non-shrunken and/or has a weight of at least 25 mg, preferably at least about 30 mg or at least about 35 mg, or between about 30 mg and about 50 mg. 13. The grain of claim 1 which is capable of producing a wheat plant which is male and female fertile, or a wheat plant which is essentially the same in morphology and/or growth rate as a corresponding wild-type plant. 14. (canceled) 15. The grain of claim 1 comprising starch, wherein the starch of the grain has an amylose content of at least 60% (w/w), or at least 67 (w/w) or at least 70% (w/w) as a proportion of the extractable starch of the grain. 16. (canceled) 17. The grain of claim 1, wherein the starch content of the grain is at least 30%, preferably at least 35%, at least 40% or at least 45% as a percentage of the total grain weight. 18. The grain of claim 1, wherein the plant is hexaploid wheat, preferably Triticum aestivum. 19. The grain of claim 1, wherein the starch of the grain is characterised by one or more of properties selected from the group consisting of:
a. comprising at least 2% resistant starch; b. comprising a glycaemic index (GI) of less than 55; c. comprising less than 20% amylopectin as a proportion of the starch content of the grain; d. comprised in starch granules which have an altered morphology relative to wild-type wheat starch granules; e. comprised in starch granules that exhibit reduced granule birefringence under polarised light relative to wild-type wheat starch granules; f. when the grain is milled to flour, such flour exhibits reduced swelling volume; g. modified chain length distribution and/or branching frequency relative to wild-type wheat starch; h. delayed end of gelatinisation temperature and higher peak temperature; i. reduced viscosity (peak viscosity, pasting temperature); j. increased molecular weight of amylopectin; and k. modified % crystallinity or % A-type or B-type starch, relative to wild-type wheat starch granules or starch. 20. The grain of claim 1 which is processed so that it is no longer capable of germinating, such as kibbled, cracked, par-boiled, rolled, pearled, milled or ground grain. 21. (canceled) 22. A wheat plant which is capable of producing the grain of claim 1, which comprises one or more exogenous polynucleotides which encode a CslF6 polypeptide and/or a CslH polypeptide. 23. (canceled) 26. (canceled) 27. A composition comprising isolated wheat BG and arabinoxylan (AX) produced from the grain of claim 1, the BG and AX being soluble in aqueous media, and the BG having a DP3/DP4 ratio of less than 2.0 and predominantly a molecular weight of at least about 100 kDa. 28. (canceled) 29. A food ingredient that comprises the grain of claim 1 for sale. 30. (canceled) 31. (canceled) 32. (canceled) 33. (canceled) 34. (canceled) 35. A method of producing a wheat plant that produces grain according to claim 1 comprising the steps of (i) introducing one or more exogenous polynucleotides which encode one or more CslF polypeptides or a CslF polypeptide and a CslH polypeptide into a progenitor wheat cell, and (ii) producing a wheat plant from the wheat cell of (i). 36. (canceled) 37. (canceled) 38. A method of selecting a wheat plant, the method comprising (i) determining the amount of BG in grain obtained from each of at least two wheat plants, and (ii) selecting a plant from (i) which produces grain according to claim 1. 39-46. (canceled) 47. A method of producing at least partially purified BG or starch, comprising the steps of i) obtaining wheat grain according to claim 1, and ii) extracting the BG or starch from the grain, thereby producing the BG or starch. 48-53. (canceled) 54. A method of altering one or more physiological parameters in an animal, or of preventing or reducing the severity or incidence of a disease, the method comprising providing to the animal, the grain of claim 1, wherein the altered physiological parameter or reduced severity or incidence of disease is relative to providing to the animal the same amount of corresponding wild-type wheat grain, wheat plant, or composition or product made therefrom. 55-58. (canceled) | 1,600 |
688 | 14,039,114 | 1,611 | The invention provides for a method of treating inappetance-induced weight loss in one or more companion animals or livestock. The method provides for administering a therapeutically effective amount of a capromorelin-containing composition to the companion animal or livestock. Optionally, one or more flavoring agents or flavor-masking agents can be added to the capromorelin-containing composition to enhance or mask the flavoring of the composition for the companion animal or livestock. | 1. A method of treating inappetance in a companion animal, comprising the step of administering a therapeutically effective amount of a capromorelin-containing composition to the companion animal. 2. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is orally administered to the companion animal. 3. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 4. The method of claim 1, wherein the capromorelin-containing composition further comprises a flavoring agent or masking agent. 5. The method of claim 4, wherein the flavoring or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 6. The method of claim 5, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 7. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered using a method selected from the group consisting of a spray, a syringe, a pill, a tablet, an implant, a patch, and a film. 8. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is intravenously administered to the companion animal. 9. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered to the companion animal at least once per day. 10. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered to the companion animal at least twice per day. 11. The method of claim 1, wherein the therapeutically effective amount is administered for a treatment period of at least one day. 12. The method of claim 1, wherein the therapeutically effective amount is administered for a treatment period of at least one week. 13. The method of claim 1, wherein the therapeutically effective amount is administered for a treatment period of at least two weeks. 14. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered for a treatment period of at least one month. 15. The method of claim 1, wherein the capromorelin-containing composition comprises a concentration of from about 0.2 milligram to about 6 milligrams of capromorelin per kilogram of body weight of the companion animals. 16. The method of claim 1, wherein the capromorelin-containing composition comprises a concentration of from about 3 milligrams to about 6 milligrams of capromorelin per kilogram of body weight of the companion animals. 17. The method of claim 1, wherein the capromorelin-containing composition comprises a concentration of from about 3 milligrams to about 4.5 milligrams of capromorelin per kilogram of body weight of the companion animals. 18. The method of claim 1, wherein the companion animal is a cat. 19. The method of claim 1, wherein the companion animal is a dog. 20. The method of claim 1, wherein the companion animal is a horse. 21. The method of claim 1, wherein the therapeutically effective dose of the capromorelin-containing composition is administered in conjunction with a chemotherapeutic regimen. 22. The method of claim 1, wherein the administration of the capromorelin-containing composition is selected from the group consisting of oral administration, intramuscular administration, and subcutaneous administration. 23. The method of claim 1, wherein the capromorelin-containing composition is in a liquid dose form. 24. The method of claim 23, wherein the liquid dose form is either sprayed onto a food product or administered using a syringe. 25. The method of claim 1, wherein the capromorelin-containing composition is incorporated into a food product, a treat, or a chew. 26. The method of claim 1, wherein lean muscle mass in the companion animal is increased by at least 5% 27. The method of claim 1, wherein lean muscle mass in the companion animal is increased by at least 10%. 28. The method of claim 1, wherein the companion animal increases food intake by at least 50% when compared to no food intake. 29. The method of claim 1, wherein the companion animal increases food intake by at least 100% when compared to no food intake. 30. A method of increasing lean muscle mass and treating inappetance in a non-human animal comprising administering a therapeutically effective dose of a capromorelin composition to the non-human animal. 31. The method of claim 30, wherein the therapeutically effective dose of a capromorelin composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 32. The method of claim 30, wherein lean muscle mass in increased by at least 5% after the capromorelin composition is administered to the non-human animal. 33. The method of claim 30, wherein lean muscle mass in increased by at least 10% after the capromorelin composition is administered to the non-human animal. 34. The method of claim 30, wherein the therapeutically effective dose of the capromorelin composition is administered in conjunction with a chemotherapeutic regimen. 35. The method of claim 30, wherein the non-human animal is selected from the group consisting of a canine, a feline, and an equine. 36. The method of claim 30, wherein the non-human animal's food consumption is increased after the administration of the therapeutically effective dose of the capromorelin composition. 37. The method of claim 36, wherein the non-human animal's food consumption is increased by at least 50%, when compared to no food intake. 38. The method of claim 36, wherein the non-human animal's food consumption is increased by at least 100%. 39. The method of claim 30, wherein the capromorelin composition comprises a concentration of between about 0.2 milligram and about 6 milligrams of capromorelin per kilogram of body weight of the non-human animal. 40. The method of claim 30, wherein the capromorelin composition comprises one or more flavoring agents or masking agents. 41. The method of claim 40, wherein the flavoring agents or masking agents are selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, and a souring agent. 42. The method of claim 41, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, and a vanilla-comprising composition. 43. The method of claim 30, wherein the capromorelin composition further comprises at least one emulsifying agent. 44. The method of claim 30, wherein the administration of the capromorelin composition is selected from the group consisting of use of a syringe, a pill, an implant, a spray, an oral solution, a tablet, and a film. 45. The method of claim 30, wherein the capromorelin composition is administered at least once per day. 46. The method of claim 30, wherein the capromorelin composition is administered at least twice per day. 47. The method of claim 30, wherein the capromorelin composition is administered for at least one week. 48. The method of claim 30, wherein the capromorelin composition is administered for at least two weeks. 49. A method of treating inappetance-induced weight loss in a non-human animal comprising the steps of:
diagnosing the non-human animal as experiencing inappetance-induced weight loss; administering at least one dose of a capromorelin-containing composition to the non-human animal; and, observing at least one indicator or plasma marker of the non-human animal. 50. The method of claim 49, further comprising the step of administering further doses of the capromorelin-containing composition to the non-human animal to a point where the indicator or plasma marker either increases or decreases in response to the dose of capromorelin. 51. The method of claim 49, wherein the at least one plasma marker is selected from the group consisting of insulin-like growth factor-1, cortisol, a body weight of the non-human animal, and capromorelin. 52. The method of claim 51, wherein the insulin-like growth factor-1, cortisol and capromorelin are increased in an animal receiving the capromorelin-containing composition. 53. The method of claim 49, wherein the indicator is selected from the group consisting of amount of food consumption, weight gain, weight loss, and an increase in lean muscle mass. 54. The method of claim 54, wherein the indicator is weight loss and the capromorelin-containing composition is administered until the animal gains a sufficient amount of weight. 55. The method of claim 49, wherein the at least one dose of the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 56. The method of claim 49, wherein the dose of the capromorelin-containing composition comprises a concentration of between about 0.2 milligram and about 6 milligrams of capromorelin per kilogram of body weight of the non-human animal. 57. The method of claim 49, wherein at least one dose of the capromorelin-containing composition are administered to the non-human animal on a daily basis. 58. The method of claim 49, wherein at least two doses of the capromorelin-containing composition are administered to the non-human animal on a daily basis. 59. The method of claim 49, wherein the capromorelin-containing composition is administered for at least one week. 60. The method of claim 49, wherein the capromorelin-containing composition is administered for at least two weeks. 61. The method of claim 49, wherein the capromorelin-containing composition is administered via a route selected from the group consisting of an oral route and a parenteral route. 62. The method of claim 49, wherein the capromorelin-containing composition further comprises a flavoring agent or masking agent 63. The method of claim 62, wherein the flavoring or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 64. The method of claim 63, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 65. A method of treating weight loss in a companion animal, comprising the steps of:
determining that the companion animal is experiencing weight loss; administering at least one dose of a capromorelin-containing composition to the companion animal using a syringe; and, assessing one or more plasma markers or indicators of the companion animal. 66. The method of claim 65, wherein the one or more markers of the companion animal are selected from the group consisting of change in weight of the companion animal, change in food intake of the companion animal, and change in lean muscle mass. 67. The method of claim 65, further comprising the step of administering further doses of the capromorelin-containing composition to the non-human animal to a point where the indicator or plasma marker is at an appropriate level. 68. The method of claim 65, wherein the indicator is weight loss and the capromorelin-containing composition is administered until the animal gains a sufficient amount of weight. 69. The method of claim 65, wherein at least one dose of the capromorelin-containing composition are administered to the companion animal on a daily basis. 70. The method of claim 65, wherein at least two doses of the capromorelin-containing composition are administered to the companion animal on a daily basis. 71. The method of claim 65, wherein the one or more plasma markers are selected from the group consisting of insulin-like growth factor-1, cortisol, and capromorelin. 72. The method of claim 65, wherein the assessment of one or more plasma markers comprises determining a minimum amount of the marker in the blood stream of the animal. 73. The method of claim 65, further comprising the step of increasing or decreasing the dose of capromorelin based on the minimum amount of the plasma marker in the blood stream of the animal. 74. The method of claim 65, wherein assessing the one or more plasma markers or indicators occurs at least one time within first week after administration of the capromorelin-containing composition. 75. The method of claim 65, wherein the administration of the capromorelin-containing composition is selected from the group consisting of oral administration, intramuscular administration, and subcutaneous administration. 76. The method of claim 65, wherein the capromorelin-containing composition further comprises a flavoring agent or masking agent. 77. The method of claim 76, wherein the flavoring or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 78. The method of claim 77, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 79. A pharmaceutical composition for treatment of inappetance in a non-human animal comprising:
a therapeutically effective amount of capromorelin; and at least one carrier. 80. The pharmaceutical composition of claim 79, wherein the therapeutically effective amount of capromorelin comprises a concentration of between about 0.2 milligrams and about 6 milligrams of capromorelin per kilogram of body weight of the non-human animal. 81. The pharmaceutical composition of claim 79, further comprising a component selected from the group consisting of an emulsifying agent and a viscosifying agent. 82. The pharmaceutical composition of claim 79, further comprising a component selected from the group consisting of a flavoring agent, a masking agent, and combinations thereof. 83. The pharmaceutical composition of claim 82, wherein the flavoring agent or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 84. The pharmaceutical composition 83, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 85. The pharmaceutical composition of claim 79, wherein the pharmaceutical composition is contained within a syringe. 86. The pharmaceutical composition of claim 79, wherein the pharmaceutical composition is formulated for oral administration. 87. The pharmaceutical composition of claim 79, wherein the composition is in the form of a solution or a suspension. 88. The pharmaceutical formulation of claim 79, wherein the carrier comprises a salt solution. 89. The pharmaceutical formulation of claim 79, wherein the pharmaceutical composition is formulated for intravenous administration. 90. The pharmaceutical formulation of claim 79, wherein the pharmaceutical composition is formulated for subcutaneous administration. 91. The pharmaceutical formulation of claim 79, wherein the pharmaceutical composition is formulated as at least one of a film, a patch, a spray, an implant, and a suppository. 92. A method of treating inappetance-induced weight loss in a companion animal comprising the steps of:
orally administering a pharmaceutical composition comprising capromorelin to the companion animal using a syringe; and, determining effectiveness of the pharmaceutical composition by observing one or more plasma markers or indicators in the companion animal. 93. The method of claim 92, wherein the pharmaceutical composition is administered to the companion animal at least once per day. 94. The method of claim 92, wherein the pharmaceutical composition comprises a concentration of between about 0.2 milligram and 6 milligrams of capromorelin per kilogram of body weight of the companion animal or livestock. 95. The method of claim 92, wherein the one or more indicators are selected from the group consisting of a change in body weight of the companion animal and a change in food intake of the companion animal. 96. The method of claim 92, wherein administration of the pharmaceutical composition is selected from the group consisting of intravenous, intramuscular, and subcutaneous administration. 97. An oral pharmaceutical composition for treatment of inappetance in a companion animal comprising:
a therapeutically effective amount of capromorelin and a flavoring or masking agent, wherein the therapeutically effective amount of capromorelin comprises a concentration of between about 0.2 milligrams and about 6 milligrams of capromorelin per kilogram of body weight of the animal. 98. The oral pharmaceutical composition of claim 97, further comprising at least one of a flavoring agent or a masking agent. 99. The oral pharmaceutical composition of claim 98, wherein the flavoring agent or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, and a souring agent. 100. The oral pharmaceutical composition of claim 99, wherein the sweetening agent comprises at least one of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, and a vanilla-comprising composition. 101. The oral pharmaceutical composition of claim 97, wherein the pharmaceutical composition further comprises a carrier. 102. The oral pharmaceutical composition of claim 97, further comprising at least one of an emulsifying agent and a viscosifying agent. 103. The oral pharmaceutical formulation of claim 97, wherein the composition is integrated into a product selected from the group consisting of a treat, a chew, and a food product. 104. The oral pharmaceutical formulation of claim 97, wherein the companion animal is a cat or a dog. 105. A method of treating or preventing hepatic lipidosis in a companion animal, comprising the steps of administering a therapeutically effective amount of a capromorelin-containing composition to the companion animal. 106. The method of claim 105, wherein the companion animal is a cat. 107. The method of claim 105, wherein the capromorelin-containing composition is administered orally. 108. The method of claim 105, wherein the capromorelin-containing composition is administered to the companion animals at least once per day. 109. The method of claim 105, wherein the capromorelin-containing compositions administered to the companion animals at least twice per day. 110. The method of claim 105, wherein the capromorelin-containing composition comprises a concentration of from about 2 milligrams to about 4 milligrams of capromorelin per kilogram of body weight of the companion animals. 111. A kit to be used for treating inappetance in non-human animals, the kit comprising:
a syringe; a vessel comprising a pharmaceutical formulation, the pharmaceutical formulation comprising a capromorelin-containing composition; and directions instructing a user to withdraw about one dose of the pharmaceutical formulation from the vessel using the syringe and administer the pharmaceutical composition to the non-human animal using the syringe. 112. The kit of claim 111, wherein the pharmaceutical formulation is formulated to for oral administration. 113. The kit of claim 111, wherein the kit includes directions that the pharmaceutical formulation is to be administered to the non-human animals at least once per day. 114. A method of maintaining a body weight of a companion animal, the method comprising administering an amount of a capromorelin-containing composition and a flavoring or masking agent to a companion animal that is sufficient to maintain the body weight of the companion animal. 115. A pharmaceutical formulation comprising:
a carrier; and a capromorelin-containing composition, wherein the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 116. A pharmaceutical formulation comprising:
a carrier; and a capromorelin-containing composition, wherein the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 905 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around twenty-five minutes. 117. A method of increasing lean muscle mass in livestock, comprising the step of administering a capromorelin-containing composition to an animal in need thereof. 118. The method of claim 117, wherein the capromorelin-containing composition further comprises a flavoring or masking agent. 119. The method of claim 118, wherein the flavoring or masking agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 120. The method of claim 117, wherein the capromorelin-containing composition is mixed with feed. 121. The method of claim 120, wherein the capromorelin-containing composition is administered at least twice daily. 122. The method of claim 120, wherein the lean muscle mass of the animal is increased by at least 5%. 123. The method of claim 120, wherein the lean muscle mass of the animal is increased by at least 10%. 124. The method of claim 120, wherein the capromorelin-containing composition is administered for at least 1 week. 125. The method of claim 120, wherein the capromorelin-containing composition is administered for at least 2 weeks. 126. The method of claim 120, wherein the livestock is selected from the group consisting of bovine species, ovine species, porcine species, avian species, and animals used as a food source. 127. A method of maintaining food consumption comprising the steps of:
obtaining a liquid formulation of a capromorelin-containing composition comprising capromorelin and a flavoring or masking agent; spraying the liquid formulation on a food product; and feeding the food product to an animal. 128. The method of claim 127, wherein the method allows the animal to consume an amount of food that maintains the weight of the animal. 129. A pharmaceutical composition comprising:
a concentration of between about 0.2 milligrams and about 4 milligrams of capromorelin per kilogram of body weight of an animal; a flavoring or masking agent; and, a carrier. | The invention provides for a method of treating inappetance-induced weight loss in one or more companion animals or livestock. The method provides for administering a therapeutically effective amount of a capromorelin-containing composition to the companion animal or livestock. Optionally, one or more flavoring agents or flavor-masking agents can be added to the capromorelin-containing composition to enhance or mask the flavoring of the composition for the companion animal or livestock.1. A method of treating inappetance in a companion animal, comprising the step of administering a therapeutically effective amount of a capromorelin-containing composition to the companion animal. 2. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is orally administered to the companion animal. 3. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 4. The method of claim 1, wherein the capromorelin-containing composition further comprises a flavoring agent or masking agent. 5. The method of claim 4, wherein the flavoring or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 6. The method of claim 5, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 7. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered using a method selected from the group consisting of a spray, a syringe, a pill, a tablet, an implant, a patch, and a film. 8. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is intravenously administered to the companion animal. 9. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered to the companion animal at least once per day. 10. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered to the companion animal at least twice per day. 11. The method of claim 1, wherein the therapeutically effective amount is administered for a treatment period of at least one day. 12. The method of claim 1, wherein the therapeutically effective amount is administered for a treatment period of at least one week. 13. The method of claim 1, wherein the therapeutically effective amount is administered for a treatment period of at least two weeks. 14. The method of claim 1, wherein the therapeutically effective amount of the capromorelin-containing composition is administered for a treatment period of at least one month. 15. The method of claim 1, wherein the capromorelin-containing composition comprises a concentration of from about 0.2 milligram to about 6 milligrams of capromorelin per kilogram of body weight of the companion animals. 16. The method of claim 1, wherein the capromorelin-containing composition comprises a concentration of from about 3 milligrams to about 6 milligrams of capromorelin per kilogram of body weight of the companion animals. 17. The method of claim 1, wherein the capromorelin-containing composition comprises a concentration of from about 3 milligrams to about 4.5 milligrams of capromorelin per kilogram of body weight of the companion animals. 18. The method of claim 1, wherein the companion animal is a cat. 19. The method of claim 1, wherein the companion animal is a dog. 20. The method of claim 1, wherein the companion animal is a horse. 21. The method of claim 1, wherein the therapeutically effective dose of the capromorelin-containing composition is administered in conjunction with a chemotherapeutic regimen. 22. The method of claim 1, wherein the administration of the capromorelin-containing composition is selected from the group consisting of oral administration, intramuscular administration, and subcutaneous administration. 23. The method of claim 1, wherein the capromorelin-containing composition is in a liquid dose form. 24. The method of claim 23, wherein the liquid dose form is either sprayed onto a food product or administered using a syringe. 25. The method of claim 1, wherein the capromorelin-containing composition is incorporated into a food product, a treat, or a chew. 26. The method of claim 1, wherein lean muscle mass in the companion animal is increased by at least 5% 27. The method of claim 1, wherein lean muscle mass in the companion animal is increased by at least 10%. 28. The method of claim 1, wherein the companion animal increases food intake by at least 50% when compared to no food intake. 29. The method of claim 1, wherein the companion animal increases food intake by at least 100% when compared to no food intake. 30. A method of increasing lean muscle mass and treating inappetance in a non-human animal comprising administering a therapeutically effective dose of a capromorelin composition to the non-human animal. 31. The method of claim 30, wherein the therapeutically effective dose of a capromorelin composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 32. The method of claim 30, wherein lean muscle mass in increased by at least 5% after the capromorelin composition is administered to the non-human animal. 33. The method of claim 30, wherein lean muscle mass in increased by at least 10% after the capromorelin composition is administered to the non-human animal. 34. The method of claim 30, wherein the therapeutically effective dose of the capromorelin composition is administered in conjunction with a chemotherapeutic regimen. 35. The method of claim 30, wherein the non-human animal is selected from the group consisting of a canine, a feline, and an equine. 36. The method of claim 30, wherein the non-human animal's food consumption is increased after the administration of the therapeutically effective dose of the capromorelin composition. 37. The method of claim 36, wherein the non-human animal's food consumption is increased by at least 50%, when compared to no food intake. 38. The method of claim 36, wherein the non-human animal's food consumption is increased by at least 100%. 39. The method of claim 30, wherein the capromorelin composition comprises a concentration of between about 0.2 milligram and about 6 milligrams of capromorelin per kilogram of body weight of the non-human animal. 40. The method of claim 30, wherein the capromorelin composition comprises one or more flavoring agents or masking agents. 41. The method of claim 40, wherein the flavoring agents or masking agents are selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, and a souring agent. 42. The method of claim 41, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, and a vanilla-comprising composition. 43. The method of claim 30, wherein the capromorelin composition further comprises at least one emulsifying agent. 44. The method of claim 30, wherein the administration of the capromorelin composition is selected from the group consisting of use of a syringe, a pill, an implant, a spray, an oral solution, a tablet, and a film. 45. The method of claim 30, wherein the capromorelin composition is administered at least once per day. 46. The method of claim 30, wherein the capromorelin composition is administered at least twice per day. 47. The method of claim 30, wherein the capromorelin composition is administered for at least one week. 48. The method of claim 30, wherein the capromorelin composition is administered for at least two weeks. 49. A method of treating inappetance-induced weight loss in a non-human animal comprising the steps of:
diagnosing the non-human animal as experiencing inappetance-induced weight loss; administering at least one dose of a capromorelin-containing composition to the non-human animal; and, observing at least one indicator or plasma marker of the non-human animal. 50. The method of claim 49, further comprising the step of administering further doses of the capromorelin-containing composition to the non-human animal to a point where the indicator or plasma marker either increases or decreases in response to the dose of capromorelin. 51. The method of claim 49, wherein the at least one plasma marker is selected from the group consisting of insulin-like growth factor-1, cortisol, a body weight of the non-human animal, and capromorelin. 52. The method of claim 51, wherein the insulin-like growth factor-1, cortisol and capromorelin are increased in an animal receiving the capromorelin-containing composition. 53. The method of claim 49, wherein the indicator is selected from the group consisting of amount of food consumption, weight gain, weight loss, and an increase in lean muscle mass. 54. The method of claim 54, wherein the indicator is weight loss and the capromorelin-containing composition is administered until the animal gains a sufficient amount of weight. 55. The method of claim 49, wherein the at least one dose of the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 56. The method of claim 49, wherein the dose of the capromorelin-containing composition comprises a concentration of between about 0.2 milligram and about 6 milligrams of capromorelin per kilogram of body weight of the non-human animal. 57. The method of claim 49, wherein at least one dose of the capromorelin-containing composition are administered to the non-human animal on a daily basis. 58. The method of claim 49, wherein at least two doses of the capromorelin-containing composition are administered to the non-human animal on a daily basis. 59. The method of claim 49, wherein the capromorelin-containing composition is administered for at least one week. 60. The method of claim 49, wherein the capromorelin-containing composition is administered for at least two weeks. 61. The method of claim 49, wherein the capromorelin-containing composition is administered via a route selected from the group consisting of an oral route and a parenteral route. 62. The method of claim 49, wherein the capromorelin-containing composition further comprises a flavoring agent or masking agent 63. The method of claim 62, wherein the flavoring or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 64. The method of claim 63, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 65. A method of treating weight loss in a companion animal, comprising the steps of:
determining that the companion animal is experiencing weight loss; administering at least one dose of a capromorelin-containing composition to the companion animal using a syringe; and, assessing one or more plasma markers or indicators of the companion animal. 66. The method of claim 65, wherein the one or more markers of the companion animal are selected from the group consisting of change in weight of the companion animal, change in food intake of the companion animal, and change in lean muscle mass. 67. The method of claim 65, further comprising the step of administering further doses of the capromorelin-containing composition to the non-human animal to a point where the indicator or plasma marker is at an appropriate level. 68. The method of claim 65, wherein the indicator is weight loss and the capromorelin-containing composition is administered until the animal gains a sufficient amount of weight. 69. The method of claim 65, wherein at least one dose of the capromorelin-containing composition are administered to the companion animal on a daily basis. 70. The method of claim 65, wherein at least two doses of the capromorelin-containing composition are administered to the companion animal on a daily basis. 71. The method of claim 65, wherein the one or more plasma markers are selected from the group consisting of insulin-like growth factor-1, cortisol, and capromorelin. 72. The method of claim 65, wherein the assessment of one or more plasma markers comprises determining a minimum amount of the marker in the blood stream of the animal. 73. The method of claim 65, further comprising the step of increasing or decreasing the dose of capromorelin based on the minimum amount of the plasma marker in the blood stream of the animal. 74. The method of claim 65, wherein assessing the one or more plasma markers or indicators occurs at least one time within first week after administration of the capromorelin-containing composition. 75. The method of claim 65, wherein the administration of the capromorelin-containing composition is selected from the group consisting of oral administration, intramuscular administration, and subcutaneous administration. 76. The method of claim 65, wherein the capromorelin-containing composition further comprises a flavoring agent or masking agent. 77. The method of claim 76, wherein the flavoring or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 78. The method of claim 77, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 79. A pharmaceutical composition for treatment of inappetance in a non-human animal comprising:
a therapeutically effective amount of capromorelin; and at least one carrier. 80. The pharmaceutical composition of claim 79, wherein the therapeutically effective amount of capromorelin comprises a concentration of between about 0.2 milligrams and about 6 milligrams of capromorelin per kilogram of body weight of the non-human animal. 81. The pharmaceutical composition of claim 79, further comprising a component selected from the group consisting of an emulsifying agent and a viscosifying agent. 82. The pharmaceutical composition of claim 79, further comprising a component selected from the group consisting of a flavoring agent, a masking agent, and combinations thereof. 83. The pharmaceutical composition of claim 82, wherein the flavoring agent or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, a souring agent, and combinations thereof. 84. The pharmaceutical composition 83, wherein the sweetening agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 85. The pharmaceutical composition of claim 79, wherein the pharmaceutical composition is contained within a syringe. 86. The pharmaceutical composition of claim 79, wherein the pharmaceutical composition is formulated for oral administration. 87. The pharmaceutical composition of claim 79, wherein the composition is in the form of a solution or a suspension. 88. The pharmaceutical formulation of claim 79, wherein the carrier comprises a salt solution. 89. The pharmaceutical formulation of claim 79, wherein the pharmaceutical composition is formulated for intravenous administration. 90. The pharmaceutical formulation of claim 79, wherein the pharmaceutical composition is formulated for subcutaneous administration. 91. The pharmaceutical formulation of claim 79, wherein the pharmaceutical composition is formulated as at least one of a film, a patch, a spray, an implant, and a suppository. 92. A method of treating inappetance-induced weight loss in a companion animal comprising the steps of:
orally administering a pharmaceutical composition comprising capromorelin to the companion animal using a syringe; and, determining effectiveness of the pharmaceutical composition by observing one or more plasma markers or indicators in the companion animal. 93. The method of claim 92, wherein the pharmaceutical composition is administered to the companion animal at least once per day. 94. The method of claim 92, wherein the pharmaceutical composition comprises a concentration of between about 0.2 milligram and 6 milligrams of capromorelin per kilogram of body weight of the companion animal or livestock. 95. The method of claim 92, wherein the one or more indicators are selected from the group consisting of a change in body weight of the companion animal and a change in food intake of the companion animal. 96. The method of claim 92, wherein administration of the pharmaceutical composition is selected from the group consisting of intravenous, intramuscular, and subcutaneous administration. 97. An oral pharmaceutical composition for treatment of inappetance in a companion animal comprising:
a therapeutically effective amount of capromorelin and a flavoring or masking agent, wherein the therapeutically effective amount of capromorelin comprises a concentration of between about 0.2 milligrams and about 6 milligrams of capromorelin per kilogram of body weight of the animal. 98. The oral pharmaceutical composition of claim 97, further comprising at least one of a flavoring agent or a masking agent. 99. The oral pharmaceutical composition of claim 98, wherein the flavoring agent or masking agent is selected from the group consisting of a sweetening agent, a savory agent, a bittering agent, and a souring agent. 100. The oral pharmaceutical composition of claim 99, wherein the sweetening agent comprises at least one of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, and a vanilla-comprising composition. 101. The oral pharmaceutical composition of claim 97, wherein the pharmaceutical composition further comprises a carrier. 102. The oral pharmaceutical composition of claim 97, further comprising at least one of an emulsifying agent and a viscosifying agent. 103. The oral pharmaceutical formulation of claim 97, wherein the composition is integrated into a product selected from the group consisting of a treat, a chew, and a food product. 104. The oral pharmaceutical formulation of claim 97, wherein the companion animal is a cat or a dog. 105. A method of treating or preventing hepatic lipidosis in a companion animal, comprising the steps of administering a therapeutically effective amount of a capromorelin-containing composition to the companion animal. 106. The method of claim 105, wherein the companion animal is a cat. 107. The method of claim 105, wherein the capromorelin-containing composition is administered orally. 108. The method of claim 105, wherein the capromorelin-containing composition is administered to the companion animals at least once per day. 109. The method of claim 105, wherein the capromorelin-containing compositions administered to the companion animals at least twice per day. 110. The method of claim 105, wherein the capromorelin-containing composition comprises a concentration of from about 2 milligrams to about 4 milligrams of capromorelin per kilogram of body weight of the companion animals. 111. A kit to be used for treating inappetance in non-human animals, the kit comprising:
a syringe; a vessel comprising a pharmaceutical formulation, the pharmaceutical formulation comprising a capromorelin-containing composition; and directions instructing a user to withdraw about one dose of the pharmaceutical formulation from the vessel using the syringe and administer the pharmaceutical composition to the non-human animal using the syringe. 112. The kit of claim 111, wherein the pharmaceutical formulation is formulated to for oral administration. 113. The kit of claim 111, wherein the kit includes directions that the pharmaceutical formulation is to be administered to the non-human animals at least once per day. 114. A method of maintaining a body weight of a companion animal, the method comprising administering an amount of a capromorelin-containing composition and a flavoring or masking agent to a companion animal that is sufficient to maintain the body weight of the companion animal. 115. A pharmaceutical formulation comprising:
a carrier; and a capromorelin-containing composition, wherein the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 150 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around two hours. 116. A pharmaceutical formulation comprising:
a carrier; and a capromorelin-containing composition, wherein the capromorelin-containing composition comprises a sufficient amount of capromorelin to achieve a Cmax of around 905 nanograms of capromorelin or a metabolite thereof per milliliter of plasma at a Tmax of around twenty-five minutes. 117. A method of increasing lean muscle mass in livestock, comprising the step of administering a capromorelin-containing composition to an animal in need thereof. 118. The method of claim 117, wherein the capromorelin-containing composition further comprises a flavoring or masking agent. 119. The method of claim 118, wherein the flavoring or masking agent is selected from the group consisting of Thaumatin T200X, Stevia Rebaudioside A, MagnaSweet®, sucralose, Talin-Pure, OptisweetSD, a vanilla-comprising composition, and combinations thereof. 120. The method of claim 117, wherein the capromorelin-containing composition is mixed with feed. 121. The method of claim 120, wherein the capromorelin-containing composition is administered at least twice daily. 122. The method of claim 120, wherein the lean muscle mass of the animal is increased by at least 5%. 123. The method of claim 120, wherein the lean muscle mass of the animal is increased by at least 10%. 124. The method of claim 120, wherein the capromorelin-containing composition is administered for at least 1 week. 125. The method of claim 120, wherein the capromorelin-containing composition is administered for at least 2 weeks. 126. The method of claim 120, wherein the livestock is selected from the group consisting of bovine species, ovine species, porcine species, avian species, and animals used as a food source. 127. A method of maintaining food consumption comprising the steps of:
obtaining a liquid formulation of a capromorelin-containing composition comprising capromorelin and a flavoring or masking agent; spraying the liquid formulation on a food product; and feeding the food product to an animal. 128. The method of claim 127, wherein the method allows the animal to consume an amount of food that maintains the weight of the animal. 129. A pharmaceutical composition comprising:
a concentration of between about 0.2 milligrams and about 4 milligrams of capromorelin per kilogram of body weight of an animal; a flavoring or masking agent; and, a carrier. | 1,600 |
689 | 14,785,400 | 1,631 | The present application relates to a method for determining a risk score that indicates a risk that a clinical event will occur within a certain period of time. The risk score is based at least in part on a combination of inferred activities of two or more cellular signaling pathways in a tissue and/or cells and/or a body fluid of a subject. The cellular signaling pathways comprise a Wnt pathway, an ER pathway, an HH pathway, and/or an AR pathway. The risk score is defined such that the indicated risk that the clinical event will occur within the certain period of time decreases with an increasing P ER and increases with an increasing max(P Wnt , P HH ), wherein P ER , P wnt , and P HH denote the inferred activity of the ER pathway, the Wnt pathway, and the HH pathway, respectively. | 1. A method comprising:
inferring activity of two or more cellular signaling pathways in a tissue and/or cells and/or a body fluid of a subject based at least on the expression levels (20) of one or more target gene(s) of the cellular signaling pathways measured in an extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and determining a risk score that indicates a risk that a clinical event will occur within a certain period of time, wherein the risk score is based at least in part on a combination of the inferred activities, wherein the cellular signaling pathways comprise a Wnt pathway, an ER pathway, an HH pathway, and/or an AR pathway, wherein the cellular signaling pathways comprise the ER pathway, the Wnt pathway, and the HH pathway, and wherein the risk score is defined such that the indicated risk that the clinical event will occur within the certain period of time decreases with an increasing PER and increases with an increasing max(PWnt, PHH), wherein PER, PWnt, and PHH denote the inferred activity of the ER pathway, the Wnt pathway, and the HH pathway, respectively. 2. The method of claim 1, wherein the combination of the inferred activities comprises the expression
−α·P ER+β·max(P Wnt, PHH),
wherein α and β are positive constant scaling factors, and the indicated risk that the clinical event will take place within the certain period of time monotonically increases with an increasing value of the expression. 3. The method of claim 2, wherein the inferring comprises:
inferring activity of a Wnt pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the Wnt pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6, and FZD7, and/or inferring activity of an ER pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the ER pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: GREB1, PGR, XBP1, CAl2, SOD1, CTSD, IGFBP4, TFF1, SGK3, NRIP1, CELSR2, WISP2, and AP1B1, and/or inferring activity of an HH pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the HH pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN, and CTSL1, and/or inferring activity of an AR pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the AR pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: KLK2, PMEPA1, TMPRSS2, NKX3_1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR, and EAF2. 4. The method of claim 3, wherein the inferring is further based on:
expression levels (20) of at least one target gene of the Wnt pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A, and LECT2, and/or expression levels (20) of at least one target gene of the ER pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: RARA, MYC, DSCAM, EBAG9, COX7A2L, ERBB2, PISD, KRT19, HSPB1, TRIM25, PTMA, COL18A1, CDH26, NDUFV3, PRDM15, ATP5J, and ESR1, and/or expression levels (20) of at least one target gene of the HH pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: BCL2, FOXA2, FOXF1, H19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1, and TOM1, and/or expression levels (20) of at least one target gene of the AR pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1, and TACC2. 5. The method of claim 1, further comprising:
assigning the subject to at least one of a plurality of risk groups associated with different indicated risks that the clinical event will occur within the certain period of time, and/or deciding a treatment recommended for the subject based at least in part on the indicated risk that the clinical event will occur within the certain period of time. 6. The method of claim 5, comprising:
inferring activity of a Wnt pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the Wnt pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and/or inferring activity of an ER pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the ER pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and/or inferring activity of an HH pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the HH pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and/or inferring activity of an AR pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the AR pathway measured in the extracted sample of the tissue and/or the cells and/pr the body fluid of the subject. 7. The method of claim 6, wherein
the set of target genes of the Wnt pathway includes at least nine, preferably all target genes selected from the group consisting of: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6, and FZD7, and/or the set of target genes of the ER pathway includes at least nine, preferably all target genes selected from the group consisting of: GREB1, PGR, XBP1, CA12, SOD1, CTSD, IGFBP4, TFF1, SGK3, NRIP1, CELSR2, WISP2, and AP1B1, and/or the set of target genes of the HH pathway includes at least nine, preferably all target genes selected from the group consisting of: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN, and CTSL1, and/or the set of target genes of the AR pathway includes at least nine, preferably all target genes selected from the group consisting of: KLK2, PMEPA1, TMPRSS2, NKX3_1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR, and EAF2. 8. The method of claim 7, wherein
the set of target genes of the Wnt pathway further includes at least one target gene selected from the group consisting of: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A, and LECT2, and/or the set of target genes of the ER pathway further includes at least one target gene selected from the group consisting of: RARA, MYC, DSCAM, EBAG9, COX7A2L, ERBB2, PISD, KRT19, HSPB1, TRIM25, PTMA, COL18A1, CDH26, NDUFV3, PRDM15, ATP5J, and ESR1, and/or the set of target genes of the HH pathway further includes at least one target gene selected from the group consisting of: BCL2, FOXA2, FOXF1, H19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1, and TOM1, and/or the set of target genes of the AR pathway further includes at least one target gene selected from the group consisting of: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1, and TACC2. 9. The method of claim 8, further comprising combining the risk score and/or at least one of the inferred activities with one or more additional risk scores obtained from one or more additional prognostic tests to obtain a combined risk score, wherein the combined risk score indicates a risk that the clinical event will occur within the certain period of time. 10. The method of claim 9, wherein the clinical event is cancer, in particular, breast cancer. 11. An apparatus comprising a digital processor configured to perform a method as set forth in claim 1. 12. A non-transitory storage medium storing instructions that are executable by a digital processing device to perform a method as set forth in claim 1. 13. A computer program comprising program code means for causing a digital processing device to perform a method as set forth in claim 1. 14. A signal representing a risk score that indicates a risk that a clinical event will occur within a certain period of time, wherein the risk score results from performing a method as set forth in claim 1. | The present application relates to a method for determining a risk score that indicates a risk that a clinical event will occur within a certain period of time. The risk score is based at least in part on a combination of inferred activities of two or more cellular signaling pathways in a tissue and/or cells and/or a body fluid of a subject. The cellular signaling pathways comprise a Wnt pathway, an ER pathway, an HH pathway, and/or an AR pathway. The risk score is defined such that the indicated risk that the clinical event will occur within the certain period of time decreases with an increasing P ER and increases with an increasing max(P Wnt , P HH ), wherein P ER , P wnt , and P HH denote the inferred activity of the ER pathway, the Wnt pathway, and the HH pathway, respectively.1. A method comprising:
inferring activity of two or more cellular signaling pathways in a tissue and/or cells and/or a body fluid of a subject based at least on the expression levels (20) of one or more target gene(s) of the cellular signaling pathways measured in an extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and determining a risk score that indicates a risk that a clinical event will occur within a certain period of time, wherein the risk score is based at least in part on a combination of the inferred activities, wherein the cellular signaling pathways comprise a Wnt pathway, an ER pathway, an HH pathway, and/or an AR pathway, wherein the cellular signaling pathways comprise the ER pathway, the Wnt pathway, and the HH pathway, and wherein the risk score is defined such that the indicated risk that the clinical event will occur within the certain period of time decreases with an increasing PER and increases with an increasing max(PWnt, PHH), wherein PER, PWnt, and PHH denote the inferred activity of the ER pathway, the Wnt pathway, and the HH pathway, respectively. 2. The method of claim 1, wherein the combination of the inferred activities comprises the expression
−α·P ER+β·max(P Wnt, PHH),
wherein α and β are positive constant scaling factors, and the indicated risk that the clinical event will take place within the certain period of time monotonically increases with an increasing value of the expression. 3. The method of claim 2, wherein the inferring comprises:
inferring activity of a Wnt pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the Wnt pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6, and FZD7, and/or inferring activity of an ER pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the ER pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: GREB1, PGR, XBP1, CAl2, SOD1, CTSD, IGFBP4, TFF1, SGK3, NRIP1, CELSR2, WISP2, and AP1B1, and/or inferring activity of an HH pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the HH pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN, and CTSL1, and/or inferring activity of an AR pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the AR pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: KLK2, PMEPA1, TMPRSS2, NKX3_1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR, and EAF2. 4. The method of claim 3, wherein the inferring is further based on:
expression levels (20) of at least one target gene of the Wnt pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A, and LECT2, and/or expression levels (20) of at least one target gene of the ER pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: RARA, MYC, DSCAM, EBAG9, COX7A2L, ERBB2, PISD, KRT19, HSPB1, TRIM25, PTMA, COL18A1, CDH26, NDUFV3, PRDM15, ATP5J, and ESR1, and/or expression levels (20) of at least one target gene of the HH pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: BCL2, FOXA2, FOXF1, H19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1, and TOM1, and/or expression levels (20) of at least one target gene of the AR pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject selected from the group consisting of: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1, and TACC2. 5. The method of claim 1, further comprising:
assigning the subject to at least one of a plurality of risk groups associated with different indicated risks that the clinical event will occur within the certain period of time, and/or deciding a treatment recommended for the subject based at least in part on the indicated risk that the clinical event will occur within the certain period of time. 6. The method of claim 5, comprising:
inferring activity of a Wnt pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the Wnt pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and/or inferring activity of an ER pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the ER pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and/or inferring activity of an HH pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the HH pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the subject, and/or inferring activity of an AR pathway in the tissue and/or the cells and/or the body fluid of the subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the AR pathway measured in the extracted sample of the tissue and/or the cells and/pr the body fluid of the subject. 7. The method of claim 6, wherein
the set of target genes of the Wnt pathway includes at least nine, preferably all target genes selected from the group consisting of: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6, and FZD7, and/or the set of target genes of the ER pathway includes at least nine, preferably all target genes selected from the group consisting of: GREB1, PGR, XBP1, CA12, SOD1, CTSD, IGFBP4, TFF1, SGK3, NRIP1, CELSR2, WISP2, and AP1B1, and/or the set of target genes of the HH pathway includes at least nine, preferably all target genes selected from the group consisting of: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN, and CTSL1, and/or the set of target genes of the AR pathway includes at least nine, preferably all target genes selected from the group consisting of: KLK2, PMEPA1, TMPRSS2, NKX3_1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR, and EAF2. 8. The method of claim 7, wherein
the set of target genes of the Wnt pathway further includes at least one target gene selected from the group consisting of: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A, and LECT2, and/or the set of target genes of the ER pathway further includes at least one target gene selected from the group consisting of: RARA, MYC, DSCAM, EBAG9, COX7A2L, ERBB2, PISD, KRT19, HSPB1, TRIM25, PTMA, COL18A1, CDH26, NDUFV3, PRDM15, ATP5J, and ESR1, and/or the set of target genes of the HH pathway further includes at least one target gene selected from the group consisting of: BCL2, FOXA2, FOXF1, H19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1, and TOM1, and/or the set of target genes of the AR pathway further includes at least one target gene selected from the group consisting of: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1, and TACC2. 9. The method of claim 8, further comprising combining the risk score and/or at least one of the inferred activities with one or more additional risk scores obtained from one or more additional prognostic tests to obtain a combined risk score, wherein the combined risk score indicates a risk that the clinical event will occur within the certain period of time. 10. The method of claim 9, wherein the clinical event is cancer, in particular, breast cancer. 11. An apparatus comprising a digital processor configured to perform a method as set forth in claim 1. 12. A non-transitory storage medium storing instructions that are executable by a digital processing device to perform a method as set forth in claim 1. 13. A computer program comprising program code means for causing a digital processing device to perform a method as set forth in claim 1. 14. A signal representing a risk score that indicates a risk that a clinical event will occur within a certain period of time, wherein the risk score results from performing a method as set forth in claim 1. | 1,600 |
690 | 13,467,414 | 1,628 | The use of 25-OH D3 (calcifediol) to increase muscle strength, muscle function, or both is provided. Vitamin D3 (cholecalciferol) may optionally be used together with 25-OH D3. Forms and dosages of a pharmaceutical composition, as well as processes for manufacturing medicaments, are also disclosed. | 1. A pharmaceutical, nutraceutical, food supplement or food composition comprising 25-OH D3 in an amount sufficent to increase or retain or prevent the loss of muscle function or muscle strength in a human. 2. A composition according to claim 1 wherein the amounts are sufficient to increase muscle strength in a patient suffering from a muscle disease or condition selected from the group consisting of cancer or AIDS related cachexia, immobility due to a secondary condition, such as a stroke, atrophy, Muscle weakness, polymyositis, Amyotrophic lateral sclerosis, Botulism, Centronuclear myopathy, Myotubular myopathy, Dysautonomia, Charcot-Marie-Tooth, Hypokalemia, Motor neurone disease, Muscular dystrophy, Myotonic dystrophy, Myasthenia gravis, Progressive muscular atrophy, Spinal muscular atrophy, Cerebral palsy, Infectious mononucleosis, Herpes zoster, Vitamin D deficiency, Fibromyalgia, Celiac disease, Hypercortisolism (Cushing's syndrome), Hypocortisolism (Addison's disease), Primary hyperaldosteronism (Conn's syndrome), and Diarrhea. | The use of 25-OH D3 (calcifediol) to increase muscle strength, muscle function, or both is provided. Vitamin D3 (cholecalciferol) may optionally be used together with 25-OH D3. Forms and dosages of a pharmaceutical composition, as well as processes for manufacturing medicaments, are also disclosed.1. A pharmaceutical, nutraceutical, food supplement or food composition comprising 25-OH D3 in an amount sufficent to increase or retain or prevent the loss of muscle function or muscle strength in a human. 2. A composition according to claim 1 wherein the amounts are sufficient to increase muscle strength in a patient suffering from a muscle disease or condition selected from the group consisting of cancer or AIDS related cachexia, immobility due to a secondary condition, such as a stroke, atrophy, Muscle weakness, polymyositis, Amyotrophic lateral sclerosis, Botulism, Centronuclear myopathy, Myotubular myopathy, Dysautonomia, Charcot-Marie-Tooth, Hypokalemia, Motor neurone disease, Muscular dystrophy, Myotonic dystrophy, Myasthenia gravis, Progressive muscular atrophy, Spinal muscular atrophy, Cerebral palsy, Infectious mononucleosis, Herpes zoster, Vitamin D deficiency, Fibromyalgia, Celiac disease, Hypercortisolism (Cushing's syndrome), Hypocortisolism (Addison's disease), Primary hyperaldosteronism (Conn's syndrome), and Diarrhea. | 1,600 |
691 | 14,362,503 | 1,611 | The present invention relates to a method of disinfecting a surface and to an antimicrobial composition. Essential oils, which are used as antimicrobial actives, are also known for their strong odour; using high amounts of these gives a strong smell to the product that is not always appreciated by the consumer. It is therefore an object of the invention to provide an antimicrobial composition, having good anti-microbial properties, at very low levels of essential oil actives. The present inventors have achieved this using a synergistic combination of select anti-microbial actives and select hydrotropes. | 1. An antimicrobial composition comprising
(a) 0.01 to 2% of an essential oil active mixture of 0.02% to 0.2% thymol and 0.05% to 0.5% terpineol; and (b) 0.1% to 5% of a hydrotrope selected from the group consisting of sodium benzoate, sodium toluene sulphonate, sodium cumene sulphonate, sodium xylene sulphonate, sodium salicylate, sodium acetate, and mixtures thereof. 2. A composition as claimed in claim 1 wherein said hydrotrope is present in 0.1 to 20% by weight of the composition. 3. A composition as claimed in claim 1 wherein said hydrotrope is selected from the group consisting of sodium benzoate, sodium salicylate, sodium acetate, and mixtures thereof. 4. A soap bar composition comprising
(a) 40 to 80% total fatty matter; (b) an antimicrobial composition as claimed in claim 1; and (c) 10 to 20% water. 5. A soap bar composition as claimed in claim 4 comprising 0.1 to 5% of the hydrotrope. 6. A soap bar composition as claimed in claim 4 comprising an additional essential oil active eugenol. 7. A composition as claimed in claim 4, wherein the pH of the composition is between 5 and 11. 8. A method for providing an anti-microbial effect o skin comprising the steps of:
(a) applying a composition as claimed in claim 1 to skin, and (b) waiting for at least 15 seconds. 9. A method as claimed in claim 8, wherein the composition is wiped or rinsed from the substrate after step ‘b’. 10. Use of a composition according to claim 1 for reduction in microbial count in less than 5 minutes. | The present invention relates to a method of disinfecting a surface and to an antimicrobial composition. Essential oils, which are used as antimicrobial actives, are also known for their strong odour; using high amounts of these gives a strong smell to the product that is not always appreciated by the consumer. It is therefore an object of the invention to provide an antimicrobial composition, having good anti-microbial properties, at very low levels of essential oil actives. The present inventors have achieved this using a synergistic combination of select anti-microbial actives and select hydrotropes.1. An antimicrobial composition comprising
(a) 0.01 to 2% of an essential oil active mixture of 0.02% to 0.2% thymol and 0.05% to 0.5% terpineol; and (b) 0.1% to 5% of a hydrotrope selected from the group consisting of sodium benzoate, sodium toluene sulphonate, sodium cumene sulphonate, sodium xylene sulphonate, sodium salicylate, sodium acetate, and mixtures thereof. 2. A composition as claimed in claim 1 wherein said hydrotrope is present in 0.1 to 20% by weight of the composition. 3. A composition as claimed in claim 1 wherein said hydrotrope is selected from the group consisting of sodium benzoate, sodium salicylate, sodium acetate, and mixtures thereof. 4. A soap bar composition comprising
(a) 40 to 80% total fatty matter; (b) an antimicrobial composition as claimed in claim 1; and (c) 10 to 20% water. 5. A soap bar composition as claimed in claim 4 comprising 0.1 to 5% of the hydrotrope. 6. A soap bar composition as claimed in claim 4 comprising an additional essential oil active eugenol. 7. A composition as claimed in claim 4, wherein the pH of the composition is between 5 and 11. 8. A method for providing an anti-microbial effect o skin comprising the steps of:
(a) applying a composition as claimed in claim 1 to skin, and (b) waiting for at least 15 seconds. 9. A method as claimed in claim 8, wherein the composition is wiped or rinsed from the substrate after step ‘b’. 10. Use of a composition according to claim 1 for reduction in microbial count in less than 5 minutes. | 1,600 |
692 | 15,555,752 | 1,634 | The present application relates to the field of cancer, particular to hypoxic tumors. It was found that hypoxia is an important driver for hypermethylation of (promoters of) tumor suppressor genes. As this hypermethylation is a stable signature that is also present in circulating tumor DNA in peripheral blood, detecting this methylation pattern is a surrogate marker for tumor hypoxia. This can be used to adapt therapy as well. | 1-7. (canceled) 8. A method of determining tumor hypoxia via detecting hypermethylation in a cancer patient, comprising:
obtaining from a subject a tissue sample comprising circulating tumor DNA, wherein the circulating tumor DNA comprises one or more promoters of tumor suppressor genes; detecting whether methylation status of the one or more promoters of tumor suppressor genes is at least 3% higher compared to normoxic tissue, by determining methylation status of a normoxic tissue and methylation status of the one or more promoters of tumor suppressor genes. 9. The method of claim 8, wherein the methylation status is determined by measuring 5hmC level of the normoxic tissue and of the one or more promoters of tumor suppressor genes. 10. The method of claim 8, comprising detecting whether the methylation status of the one or more promoters of tumor suppressor genes is at least 6% higher compared to normoxic tissue. 11. The method of claim 8, comprising detecting whether the methylation status of the one or more promoters of tumor suppressor genes is at least 15% higher compared to normoxic tissue. 12. The method of claim 8, wherein the cancer is bladder, breast, colorectal, head and neck, kidney, lung adeno, lung squamous and uterine carcinoma, gliobastoma multiforme, or non-small cell lung cancer. 13. The method of claim 12, wherein the cancer is breast cancer. 14. The method of claim 8 wherein the tumor suppressor gene is HIC1, KDM6A, NF2, KDM5C, IGFBP2, ARNT2, PTEN, MGMT, ATM, MLH1, BRCA1, SEMA3B, TIMP3, THBD, or CLDN3. 15. The method of claim 8, further comprising after the detecting step adapting the dose or nature of cancer therapy to the level of hypermethylation. 16. The method of claim 8, wherein the tissue sample is obtained from a subject receiving cancer therapy. 17. A method of determining tumor hypoxia via detecting demethylation of one or more promoters of tumor suppressor genes in a cancer patient, comprising:
obtaining from a subject receiving cancer therapy a tissue sample comprising circulating tumor DNA, wherein the circulating tumor DNA comprises one or more promoters of tumor suppressor genes; detecting whether methylation status of the one or more promoters of tumor suppressor genes is at least 3% lower compared to normoxic tissue, by determining methylation status of a normoxic tissue and methylation status of the one or more promoters of tumor suppressor genes. 18. The method according to claim 17, further comprising after the detecting step adapting the dose or nature of the cancer therapy to the level of demethylation. 19. A kit for detecting tumor hypoxia, comprising primers or probes to detect hypermethylation or demethylation of one or more promoters of tumor suppressor genes. 20. The kit of claim 19 wherein the tumor suppressor gene is HIC1, KDM6A, NF2, KDM5C, IGFBP2, ARNT2, PTEN, MGMT, ATM, MLH1, BRCA1, SEMA3B, TIMP3, THBD, or CLDN3. | The present application relates to the field of cancer, particular to hypoxic tumors. It was found that hypoxia is an important driver for hypermethylation of (promoters of) tumor suppressor genes. As this hypermethylation is a stable signature that is also present in circulating tumor DNA in peripheral blood, detecting this methylation pattern is a surrogate marker for tumor hypoxia. This can be used to adapt therapy as well.1-7. (canceled) 8. A method of determining tumor hypoxia via detecting hypermethylation in a cancer patient, comprising:
obtaining from a subject a tissue sample comprising circulating tumor DNA, wherein the circulating tumor DNA comprises one or more promoters of tumor suppressor genes; detecting whether methylation status of the one or more promoters of tumor suppressor genes is at least 3% higher compared to normoxic tissue, by determining methylation status of a normoxic tissue and methylation status of the one or more promoters of tumor suppressor genes. 9. The method of claim 8, wherein the methylation status is determined by measuring 5hmC level of the normoxic tissue and of the one or more promoters of tumor suppressor genes. 10. The method of claim 8, comprising detecting whether the methylation status of the one or more promoters of tumor suppressor genes is at least 6% higher compared to normoxic tissue. 11. The method of claim 8, comprising detecting whether the methylation status of the one or more promoters of tumor suppressor genes is at least 15% higher compared to normoxic tissue. 12. The method of claim 8, wherein the cancer is bladder, breast, colorectal, head and neck, kidney, lung adeno, lung squamous and uterine carcinoma, gliobastoma multiforme, or non-small cell lung cancer. 13. The method of claim 12, wherein the cancer is breast cancer. 14. The method of claim 8 wherein the tumor suppressor gene is HIC1, KDM6A, NF2, KDM5C, IGFBP2, ARNT2, PTEN, MGMT, ATM, MLH1, BRCA1, SEMA3B, TIMP3, THBD, or CLDN3. 15. The method of claim 8, further comprising after the detecting step adapting the dose or nature of cancer therapy to the level of hypermethylation. 16. The method of claim 8, wherein the tissue sample is obtained from a subject receiving cancer therapy. 17. A method of determining tumor hypoxia via detecting demethylation of one or more promoters of tumor suppressor genes in a cancer patient, comprising:
obtaining from a subject receiving cancer therapy a tissue sample comprising circulating tumor DNA, wherein the circulating tumor DNA comprises one or more promoters of tumor suppressor genes; detecting whether methylation status of the one or more promoters of tumor suppressor genes is at least 3% lower compared to normoxic tissue, by determining methylation status of a normoxic tissue and methylation status of the one or more promoters of tumor suppressor genes. 18. The method according to claim 17, further comprising after the detecting step adapting the dose or nature of the cancer therapy to the level of demethylation. 19. A kit for detecting tumor hypoxia, comprising primers or probes to detect hypermethylation or demethylation of one or more promoters of tumor suppressor genes. 20. The kit of claim 19 wherein the tumor suppressor gene is HIC1, KDM6A, NF2, KDM5C, IGFBP2, ARNT2, PTEN, MGMT, ATM, MLH1, BRCA1, SEMA3B, TIMP3, THBD, or CLDN3. | 1,600 |
693 | 15,968,456 | 1,623 | wherein X is Cl or Br. A compound isolated from Actinomadura and having a chemical formula of C43H69ClN4O10S2 or C43H69BrN4O10S2 is also provided. Compositions including the compounds and methods of using the compounds to treat fungal infections including those such as Candida are also disclosed. | 1. A pharmaceutical composition comprising an effective amount of a compound of Formula I:
or pharmaceutically acceptable salt thereof for treating a fungal infection in a mammal, wherein X is Cl or Br; and
a pharmaceutically acceptable carrier. 2. The pharmaceutical composition of claim 1, formulated for oral, parenteral, nasal, or topical administration. 3. The pharmaceutical composition of claim 1, wherein the mammal is human. 4. The pharmaceutical composition of claim 1, further comprising amphotericin B, ketoconazole, terbinafine, nystatin, fluconazole, itraconazole, or nystatin. 5. The pharmaceutical composition of claim 1, wherein the compound of Formula I has the Formula IA:
wherein X is Cl or Br. 6. The pharmaceutical composition of claim 5, wherein the mammal is human. 7. The pharmaceutical composition of claim 1, wherein the compound of Formula I has the Formula IB: 8. The pharmaceutical composition of claim 7, wherein the mammal is human. 9. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida infection. 10. The pharmaceutical composition of claim 9, wherein the mammal is human. 11. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida albicans infection in a human. 12. A pharmaceutical composition comprising an effective amount of a compound of Formula I:
or pharmaceutically acceptable salt thereof for treating a fungal infection in a mammal, wherein X is Cl or Br; and
an isotonic aqueous solution. 13. The pharmaceutical composition of claim 12, formulated for parenteral administration. 14. The pharmaceutical composition of claim 12, wherein the mammal is human. 15. The pharmaceutical composition of claim 12, wherein the compound of Formula I has the Formula IA:
wherein X is Cl or Br. 16. The pharmaceutical composition of claim 12, wherein the compound of Formula I has the Formula IB: 17. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida infection. 18. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida albicans infection. 19. A pharmaceutical composition comprising an effective amount of a compound of Formula I:
or pharmaceutically acceptable salt thereof for treating a fungal infection in a mammal, wherein X is Cl or Br; and
one or more of cetyl esters wax, cetyl alcohol, white wax, glyceryl monostearate, propylene glycol monostearate, methyl stearate, benzyl alcohol, sodium lauryl sulfate, glycerin, mineral oil, carbomer, ethyl alcohol, acrylate adhesives, polyisobutylene adhesives, and silicone adhesives. 20. The pharmaceutical composition of claim 19, wherein the mammal is human. 21. The pharmaceutical composition of claim 19, further comprising amphotericin B, ketoconazole, terbinafine, nystatin, fluconazole, itraconazole, or nystatin. | wherein X is Cl or Br. A compound isolated from Actinomadura and having a chemical formula of C43H69ClN4O10S2 or C43H69BrN4O10S2 is also provided. Compositions including the compounds and methods of using the compounds to treat fungal infections including those such as Candida are also disclosed.1. A pharmaceutical composition comprising an effective amount of a compound of Formula I:
or pharmaceutically acceptable salt thereof for treating a fungal infection in a mammal, wherein X is Cl or Br; and
a pharmaceutically acceptable carrier. 2. The pharmaceutical composition of claim 1, formulated for oral, parenteral, nasal, or topical administration. 3. The pharmaceutical composition of claim 1, wherein the mammal is human. 4. The pharmaceutical composition of claim 1, further comprising amphotericin B, ketoconazole, terbinafine, nystatin, fluconazole, itraconazole, or nystatin. 5. The pharmaceutical composition of claim 1, wherein the compound of Formula I has the Formula IA:
wherein X is Cl or Br. 6. The pharmaceutical composition of claim 5, wherein the mammal is human. 7. The pharmaceutical composition of claim 1, wherein the compound of Formula I has the Formula IB: 8. The pharmaceutical composition of claim 7, wherein the mammal is human. 9. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida infection. 10. The pharmaceutical composition of claim 9, wherein the mammal is human. 11. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida albicans infection in a human. 12. A pharmaceutical composition comprising an effective amount of a compound of Formula I:
or pharmaceutically acceptable salt thereof for treating a fungal infection in a mammal, wherein X is Cl or Br; and
an isotonic aqueous solution. 13. The pharmaceutical composition of claim 12, formulated for parenteral administration. 14. The pharmaceutical composition of claim 12, wherein the mammal is human. 15. The pharmaceutical composition of claim 12, wherein the compound of Formula I has the Formula IA:
wherein X is Cl or Br. 16. The pharmaceutical composition of claim 12, wherein the compound of Formula I has the Formula IB: 17. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida infection. 18. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition comprises an effective amount of the compound or pharmaceutically acceptable salt thereof for treating a Candida albicans infection. 19. A pharmaceutical composition comprising an effective amount of a compound of Formula I:
or pharmaceutically acceptable salt thereof for treating a fungal infection in a mammal, wherein X is Cl or Br; and
one or more of cetyl esters wax, cetyl alcohol, white wax, glyceryl monostearate, propylene glycol monostearate, methyl stearate, benzyl alcohol, sodium lauryl sulfate, glycerin, mineral oil, carbomer, ethyl alcohol, acrylate adhesives, polyisobutylene adhesives, and silicone adhesives. 20. The pharmaceutical composition of claim 19, wherein the mammal is human. 21. The pharmaceutical composition of claim 19, further comprising amphotericin B, ketoconazole, terbinafine, nystatin, fluconazole, itraconazole, or nystatin. | 1,600 |
694 | 15,436,404 | 1,651 | Compositions suitable for oral administration to an animal comprising at least one immune stress-reducing enzyme in an amount effective to decrease the level of positive acute phase protein in an animal, increase the level of negative acute phase protein in an animal, and/or improve animal growth performance is provided, as are methods using such compositions. The compositions include animal feed compositions, liquid compositions other than animal feed, and solid compositions other than animal feed. | 1: A composition suitable for oral administration to an animal comprising an immune stress-reducing enzyme in an orally acceptable carrier, wherein said composition is selected from the group consisting of:
(i) an animal feed comprising an amount of said enzyme effective to decrease the level of positive acute phase protein in said animal, increase the level of negative acute phase protein in said animal, and/or improve animal growth performance; (ii) a liquid composition other than an animal feed comprising at least 40,000 IU enzyme/L; and (iii) a solid composition other than an animal feed comprising at least 40,000 IU enzyme/kg,
wherein:
(a) said enzyme is other than a β-mannanase-type hemicellulase or phospholipase, and
(b) if said enzyme comprises 1,3-β-glucanase, said composition is selected from the group consisting of (i) an animal feed comprising at least 20 IU 1,3-β-glucanase/kg feed; (ii) a liquid composition other than an animal feed comprising at least 155,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising at least 300,000 IU 1,3-β-glucanase/kg. 2-4. (canceled) 5: The composition of claim 1, wherein the composition is an animal feed that comprises an ingredient that induces an immune response in the animal and wherein the enzyme comprises an enzyme that degrades said ingredient. 6: The composition of claim 5, wherein said ingredient is an antigen displayed by a pathogenic microorganism. 7: The composition of claim 1, wherein the enzyme comprises an enzyme selected from the group consisting of β-glucosidase, xyloglucanase, DNAases, non-specific nucleases, RNAse L, dsRNA specific adenosine deaminase, CG specific restriction endonuclease, N-glycanases, endo enzymes, PNGases, carbohydrases, α-1,2-fucosidase, α-1,3-1,4-fucosidase, α-1,6-mannosidase, α-1,2-mannosidase, α-1,3-mannosidase, β-1,4-galactosidase, endo-β-N acetylglucosaminidase F (endo F), peptide-N—(N-acetyl-beta-glucosaminyl)asparagine amidase F (PNGase F), PNGase A, endo-β-N-acetylglucosarninidase H (endoH), endo D, endo C, α-N-acetylgalacosamidase, β-1,3-galactosidase, endo-N-acyl-neuraminidase (endo N), α-2,3-neuraminidase, α-2,6-neuraminidase, α-2,8-neuraminidase, β-N -acetylhexosarninidase, endo-β-N-galactosidase, endo-α-N-acetylglactosaminidase, endo-α-1,6-D-mannanase, arabinogalactanase, α-mannanase, α-mannosidase, sphingomyelinase, chitinase, chitin deacetylase, carbohydrate deacetylase, N-acetylglucosaminidase, phosphatidylserine decarboxylase, sulfatase, β-galactosidase, arabinanase, hyaluronidase, α-arabinofuranosidase, chondroitinase, glucocerebrosidase, methyl esterase, ferulic acid esterase, furuloyl esterase, acetyl esterase, and carbohydrate deacetylase. 8: The composition of claim 1, wherein the enzyme comprises 1,3-β-glucanase. 9: The composition of claim 8, wherein the composition is selected from the group consisting of (i) an animal feed comprising at least 30 IU 1,3-β-glucanase/kg feed; (ii) a liquid composition other than an animal feed comprising at least 230,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising at least 450,000 IU 1,3-β-glucanase/kg. 10: The composition of claim 1, wherein the enzyme comprises an enzyme selected from the group consisting of chitinase, xyloglucanase and arabinanase. 11: A composition suitable for oral administration to an animal comprising two or more immune stress-reducing enzymes, wherein said composition comprises at least one immune stress-reducing enzyme other than 1,4-β-mannanase and 1,3-β-glucanase, and wherein said composition is selected from the group consisting of:
(i) an animal feed comprising an amount of said immune stress-reducing enzymes effective to decrease the level of positive acute phase protein in said animal, increase the level of negative acute phase protein in said animal, and/or improve animal growth performance;
(ii) a liquid composition other than an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 40,000 IU enzyme/L; and
(iii) a solid composition other than an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 40,000 IU enzyme/kg. 12: The composition of claim 11, wherein said composition is an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 20 IU enzyme/kg feed. 13: The composition of claim 11, wherein the composition is a solid composition other than an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 80,000 IU enzyme/kg. 14: The composition of claim 11, wherein the composition is a solid composition other than an animal feed comprising at least 160,000 IU enzyme/kg. 15: The composition according to claim 11, comprising at least one of 1,4-β-mannanase and 1,3-β-glucanase. 16: The composition of claim 11, comprising at least one enzyme selected from the group consisting of β-glucosidase, xyloglucanase, DNAases, non-specific nucleases, RNAse L, dsRNA specific adenosine deaminase, CG specific restriction endonuclease, N-glycanases, endo enzymes, PNGases, carbohydrases, α-1,2-fucosidase, α-1,3-1,4-fucosidase, α-1,6-mannosidase, α-1,2-mannosidase, α-1,3-mannosidase, β-1,4-galactosidase, endo-β-N acetylglucosaminidase F (endo F), peptide-N—(N-acetyl-beta-glucosaminyl)asparagine amidase F (PNGase F), PNGase A, endo-β-N-acetylglucosarninidase H (endoH), endo D, endo C, α-N-acetylgalacosamidase, β-1,3-galactosidase, endo-N-acyl-neuraminidase (endo N), α-2,3-neuraminidase, α-2,6-neuraminidase, α-2,8-neuraminidase, β-N -acetylhexosarninidase, endo-β-N-galactosidase, endo-α-N-acetylglactosaminidase, endo-α-1,6-D-mannanase, arabinogalactanase, α-mannanase, α-mannosidase, sphingomyelinase, chitinase, chitin deacetylase, carbohydrate deacetylase, N-acetylglucosaminidase, phosphatidylserine decarboxylase, sulfatase, β-galactosidase, arabinanase, hyaluronidase, α-arabinofuranosidase, chondroitinase, glucocerebrosidase, methyl esterase, ferulic acid esterase, furuloyl esterase, acetyl esterase, and carbohydrate deacetylase. 17: The composition according to claim 11, wherein the composition is selected from the group consisting of (i) a composition comprising 1,4-β-mannanase and chitanase; (ii) a composition comprising 1,4-β-mannanase and xyloglucanase; (iii) a composition comprising 1,4-β-mannanase and arabinanase; (iv) a composition comprising 1,3-β-glucanase and chitanase; (v) a composition comprising 1,3-β-glucanase and xyloglucanase; (vi) a composition comprising 1,3-β-glucanase and arabinanase and (vii) a composition comprising 1,4-β-mannanase, 1,3-β-glucanase and arabinanase. 18: A composition suitable for oral administration to an animal comprising 1,4-β-mannanase and 1,3-β-glucanase, wherein said composition is selected from the group consisting of (i) an animal feed comprising 1,4-β-mannanase and at least 20 IU 1,3-β-glucanase/kg feed, (ii) a liquid composition other than an animal feed comprising 1,4-β-mannanase and at least 155,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising 1,4-β-mannanase and at least 300,000 IU 1,3-13-glucanase/kg. 19: The composition of claim 18, wherein the composition is selected from the group consisting of (i) an animal feed comprising 1,4-β-mannanase and at least 30 IU 1,3-β-glucanase/kg feed; (ii) a liquid composition other than an animal feed comprising 1,4-β-mannanase and at least 230,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising 1,4-β-mannanase and at least 450,000 IU 1,3-β-glucanase/kg. 20: The composition of claim 18, further comprising one or more additional immune stress-reducing enzymes. 21: A method of improving animal growth performance and/or reducing immune stress in an animal, comprising orally administering to said animal a composition according to claim 1. 22-26. (canceled) 27: The method of claim 21, wherein the composition comprises at least one enzyme selected from the group consisting of β-glucosidase, xyloglucanase, DNAases, non-specific nucleases, RNAse L, dsRNA specific adenosine deaminase, CG specific restriction endonuclease, N-glycanases, endo enzymes, PNGases, carbohydrases, α-1,2-fucosidase, α-1,3-1,4-fucosidase, α-1,6-mannosidase, α-1,2-mannosidase, α-1,3-mannosidase, β-1,4-galactosidase, endo-β-N acetylglucosaminidase F (endo F), peptide-N—(N-acetyl-beta-glucosaminyl)asparagine amidase F (PNGase F), PNGase A, endo-β-N-acetylglucosarninidase H (endoH), endo D, endo C, α-N-acetylgalacosamidase, β-1,3-galactosidase, endo-N-acyl-neuraminidase (endo N), α-2,3-neuraminidase, α-2,6-neuraminidase, α-2,8-neuraminidase, β-N -acetylhexosarninidase, endo-β-N-galactosidase, endo-α-N-acetylglactosaminidase, endo-α-1,6-D-mannanase, arabinogalactanase, α-mannanase, α-mannosidase, sphingomyelinase, chitinase, chitin deacetylase, carbohydrate deacetylase, N-acetylglucosaminidase, phosphatidylserine decarboxylase, sulfatase, β-galactosidase, arabinanase, hyaluronidase, α-arabinofuranosidase, chondroitinase, glucocerebrosidase, methyl esterase, ferulic acid esterase, furuloyl esterase, acetyl esterase, and carbohydrate deacetylase. 28. (canceled) 29: A method of preventing or treating infection associated with a pathogenic microorganism that displays an antigen, comprising orally administering to an animal in need thereof a composition according to claim 1, wherein the composition comprises at least one immune stress-reducing enzyme that degrades said antigen. 30. (canceled) | Compositions suitable for oral administration to an animal comprising at least one immune stress-reducing enzyme in an amount effective to decrease the level of positive acute phase protein in an animal, increase the level of negative acute phase protein in an animal, and/or improve animal growth performance is provided, as are methods using such compositions. The compositions include animal feed compositions, liquid compositions other than animal feed, and solid compositions other than animal feed.1: A composition suitable for oral administration to an animal comprising an immune stress-reducing enzyme in an orally acceptable carrier, wherein said composition is selected from the group consisting of:
(i) an animal feed comprising an amount of said enzyme effective to decrease the level of positive acute phase protein in said animal, increase the level of negative acute phase protein in said animal, and/or improve animal growth performance; (ii) a liquid composition other than an animal feed comprising at least 40,000 IU enzyme/L; and (iii) a solid composition other than an animal feed comprising at least 40,000 IU enzyme/kg,
wherein:
(a) said enzyme is other than a β-mannanase-type hemicellulase or phospholipase, and
(b) if said enzyme comprises 1,3-β-glucanase, said composition is selected from the group consisting of (i) an animal feed comprising at least 20 IU 1,3-β-glucanase/kg feed; (ii) a liquid composition other than an animal feed comprising at least 155,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising at least 300,000 IU 1,3-β-glucanase/kg. 2-4. (canceled) 5: The composition of claim 1, wherein the composition is an animal feed that comprises an ingredient that induces an immune response in the animal and wherein the enzyme comprises an enzyme that degrades said ingredient. 6: The composition of claim 5, wherein said ingredient is an antigen displayed by a pathogenic microorganism. 7: The composition of claim 1, wherein the enzyme comprises an enzyme selected from the group consisting of β-glucosidase, xyloglucanase, DNAases, non-specific nucleases, RNAse L, dsRNA specific adenosine deaminase, CG specific restriction endonuclease, N-glycanases, endo enzymes, PNGases, carbohydrases, α-1,2-fucosidase, α-1,3-1,4-fucosidase, α-1,6-mannosidase, α-1,2-mannosidase, α-1,3-mannosidase, β-1,4-galactosidase, endo-β-N acetylglucosaminidase F (endo F), peptide-N—(N-acetyl-beta-glucosaminyl)asparagine amidase F (PNGase F), PNGase A, endo-β-N-acetylglucosarninidase H (endoH), endo D, endo C, α-N-acetylgalacosamidase, β-1,3-galactosidase, endo-N-acyl-neuraminidase (endo N), α-2,3-neuraminidase, α-2,6-neuraminidase, α-2,8-neuraminidase, β-N -acetylhexosarninidase, endo-β-N-galactosidase, endo-α-N-acetylglactosaminidase, endo-α-1,6-D-mannanase, arabinogalactanase, α-mannanase, α-mannosidase, sphingomyelinase, chitinase, chitin deacetylase, carbohydrate deacetylase, N-acetylglucosaminidase, phosphatidylserine decarboxylase, sulfatase, β-galactosidase, arabinanase, hyaluronidase, α-arabinofuranosidase, chondroitinase, glucocerebrosidase, methyl esterase, ferulic acid esterase, furuloyl esterase, acetyl esterase, and carbohydrate deacetylase. 8: The composition of claim 1, wherein the enzyme comprises 1,3-β-glucanase. 9: The composition of claim 8, wherein the composition is selected from the group consisting of (i) an animal feed comprising at least 30 IU 1,3-β-glucanase/kg feed; (ii) a liquid composition other than an animal feed comprising at least 230,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising at least 450,000 IU 1,3-β-glucanase/kg. 10: The composition of claim 1, wherein the enzyme comprises an enzyme selected from the group consisting of chitinase, xyloglucanase and arabinanase. 11: A composition suitable for oral administration to an animal comprising two or more immune stress-reducing enzymes, wherein said composition comprises at least one immune stress-reducing enzyme other than 1,4-β-mannanase and 1,3-β-glucanase, and wherein said composition is selected from the group consisting of:
(i) an animal feed comprising an amount of said immune stress-reducing enzymes effective to decrease the level of positive acute phase protein in said animal, increase the level of negative acute phase protein in said animal, and/or improve animal growth performance;
(ii) a liquid composition other than an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 40,000 IU enzyme/L; and
(iii) a solid composition other than an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 40,000 IU enzyme/kg. 12: The composition of claim 11, wherein said composition is an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 20 IU enzyme/kg feed. 13: The composition of claim 11, wherein the composition is a solid composition other than an animal feed comprising at least one immune stress-reducing enzyme in an amount of at least 80,000 IU enzyme/kg. 14: The composition of claim 11, wherein the composition is a solid composition other than an animal feed comprising at least 160,000 IU enzyme/kg. 15: The composition according to claim 11, comprising at least one of 1,4-β-mannanase and 1,3-β-glucanase. 16: The composition of claim 11, comprising at least one enzyme selected from the group consisting of β-glucosidase, xyloglucanase, DNAases, non-specific nucleases, RNAse L, dsRNA specific adenosine deaminase, CG specific restriction endonuclease, N-glycanases, endo enzymes, PNGases, carbohydrases, α-1,2-fucosidase, α-1,3-1,4-fucosidase, α-1,6-mannosidase, α-1,2-mannosidase, α-1,3-mannosidase, β-1,4-galactosidase, endo-β-N acetylglucosaminidase F (endo F), peptide-N—(N-acetyl-beta-glucosaminyl)asparagine amidase F (PNGase F), PNGase A, endo-β-N-acetylglucosarninidase H (endoH), endo D, endo C, α-N-acetylgalacosamidase, β-1,3-galactosidase, endo-N-acyl-neuraminidase (endo N), α-2,3-neuraminidase, α-2,6-neuraminidase, α-2,8-neuraminidase, β-N -acetylhexosarninidase, endo-β-N-galactosidase, endo-α-N-acetylglactosaminidase, endo-α-1,6-D-mannanase, arabinogalactanase, α-mannanase, α-mannosidase, sphingomyelinase, chitinase, chitin deacetylase, carbohydrate deacetylase, N-acetylglucosaminidase, phosphatidylserine decarboxylase, sulfatase, β-galactosidase, arabinanase, hyaluronidase, α-arabinofuranosidase, chondroitinase, glucocerebrosidase, methyl esterase, ferulic acid esterase, furuloyl esterase, acetyl esterase, and carbohydrate deacetylase. 17: The composition according to claim 11, wherein the composition is selected from the group consisting of (i) a composition comprising 1,4-β-mannanase and chitanase; (ii) a composition comprising 1,4-β-mannanase and xyloglucanase; (iii) a composition comprising 1,4-β-mannanase and arabinanase; (iv) a composition comprising 1,3-β-glucanase and chitanase; (v) a composition comprising 1,3-β-glucanase and xyloglucanase; (vi) a composition comprising 1,3-β-glucanase and arabinanase and (vii) a composition comprising 1,4-β-mannanase, 1,3-β-glucanase and arabinanase. 18: A composition suitable for oral administration to an animal comprising 1,4-β-mannanase and 1,3-β-glucanase, wherein said composition is selected from the group consisting of (i) an animal feed comprising 1,4-β-mannanase and at least 20 IU 1,3-β-glucanase/kg feed, (ii) a liquid composition other than an animal feed comprising 1,4-β-mannanase and at least 155,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising 1,4-β-mannanase and at least 300,000 IU 1,3-13-glucanase/kg. 19: The composition of claim 18, wherein the composition is selected from the group consisting of (i) an animal feed comprising 1,4-β-mannanase and at least 30 IU 1,3-β-glucanase/kg feed; (ii) a liquid composition other than an animal feed comprising 1,4-β-mannanase and at least 230,000 IU 1,3-β-glucanase/L and (iii) a solid composition other than an animal feed comprising 1,4-β-mannanase and at least 450,000 IU 1,3-β-glucanase/kg. 20: The composition of claim 18, further comprising one or more additional immune stress-reducing enzymes. 21: A method of improving animal growth performance and/or reducing immune stress in an animal, comprising orally administering to said animal a composition according to claim 1. 22-26. (canceled) 27: The method of claim 21, wherein the composition comprises at least one enzyme selected from the group consisting of β-glucosidase, xyloglucanase, DNAases, non-specific nucleases, RNAse L, dsRNA specific adenosine deaminase, CG specific restriction endonuclease, N-glycanases, endo enzymes, PNGases, carbohydrases, α-1,2-fucosidase, α-1,3-1,4-fucosidase, α-1,6-mannosidase, α-1,2-mannosidase, α-1,3-mannosidase, β-1,4-galactosidase, endo-β-N acetylglucosaminidase F (endo F), peptide-N—(N-acetyl-beta-glucosaminyl)asparagine amidase F (PNGase F), PNGase A, endo-β-N-acetylglucosarninidase H (endoH), endo D, endo C, α-N-acetylgalacosamidase, β-1,3-galactosidase, endo-N-acyl-neuraminidase (endo N), α-2,3-neuraminidase, α-2,6-neuraminidase, α-2,8-neuraminidase, β-N -acetylhexosarninidase, endo-β-N-galactosidase, endo-α-N-acetylglactosaminidase, endo-α-1,6-D-mannanase, arabinogalactanase, α-mannanase, α-mannosidase, sphingomyelinase, chitinase, chitin deacetylase, carbohydrate deacetylase, N-acetylglucosaminidase, phosphatidylserine decarboxylase, sulfatase, β-galactosidase, arabinanase, hyaluronidase, α-arabinofuranosidase, chondroitinase, glucocerebrosidase, methyl esterase, ferulic acid esterase, furuloyl esterase, acetyl esterase, and carbohydrate deacetylase. 28. (canceled) 29: A method of preventing or treating infection associated with a pathogenic microorganism that displays an antigen, comprising orally administering to an animal in need thereof a composition according to claim 1, wherein the composition comprises at least one immune stress-reducing enzyme that degrades said antigen. 30. (canceled) | 1,600 |
695 | 15,238,014 | 1,655 | The present invention relates to processes and intermediates useful in the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia) and salts thereof, an S1P1 receptor modulator that is useful in the treatment of S1P1 receptor-associated disorders, for example, diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders (e.g., acute and chronic inflammatory conditions), cancer, and conditions characterized by an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic (e.g., as may occur in inflammation, tumor development and atherosclerosis). | 1. A process for preparing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia):
comprising the following steps:
a) hydrolyzing a compound of Formula (IIk):
wherein R3 is C1-C6 alkyl; in the presence of immobilized Candida antarctica lipase B and a hydrolyzing-step solvent to form said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia); and
b) contacting said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Formula (Ia)) with L-arginine or a salt thereof, in the presence of a contacting-step solvent and H2O to form said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia). 2. The process according to claim 1, wherein R3 is ethyl. 3. (canceled) 4. The process according to claim 1, wherein said hydrolyzing-step solvent comprises dimethylformamide (DMF), dimethylacetamide (DMA), dimethylsulfoxide (DMSO), tetrahydrofuran (THF), or acetonitrile. 5. The process according to claim 1, wherein said hydrolyzing-step solvent comprises acetonitrile. 6. The process according to claim 1, wherein: said compound of Formula (IIk) is:
and said hydrolyzing-step solvent comprises acetonitrile. 7. The process according to claim 1, wherein said hydrolyzing in step a), is conducted in the presence of a phosphate buffer at a pH of about 7.6 to about 8.0. 8. The process according to claim 7, wherein said phosphate buffer is a potassium phosphate buffer. 9. The process according to claim 1, wherein said hydrolyzing in step a), is conducted at a temperature of about 30° C. to about 55° C. 10. The process according to claim 1, wherein said hydrolyzing in step a), further comprises the step of isolating said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has an enantiomeric excess of about 95% or greater. 11. The process according to claim 1, wherein said contacting-step solvent comprises isopropyl alcohol. 12. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has a purity of about 95% or greater as determined by HPLC. 13. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has an enantiomeric excess of about 95% or greater. 14. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid wherein said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid is a crystalline form. 15. The process according to claim 14, wherein said crystalline form has a powder X-ray diffraction (PXRD) pattern substantially as shown in FIG. 5, a differential scanning calorimetry (DSC) trace substantially as shown in FIG. 6, a thermogravimetric analysis (TGA) profile substantially as shown in FIG. 7, or a dynamic vapor sorption (DVS) profile substantially as shown in FIG. 8. 16. (canceled) 17. (canceled) 18. (canceled) 19. (canceled) 20. (canceled) 21. (canceled) 22. (canceled) 23. (canceled) 24. (canceled) 25. (canceled) 26. (canceled) 27. (canceled) 28. (canceled) 29. (canceled) 30. (canceled) 31. (canceled) 32. (canceled) 33. (canceled) 34. (canceled) 35. (canceled) 36. (canceled) 37. (canceled) 38. (canceled) 39. (canceled) 40. (canceled) 41. (canceled) 42. (canceled) 43. (canceled) 44. (canceled) 45. (canceled) 46. (canceled) 47. (canceled) 48. (canceled) 49. (canceled) 50. (canceled) 51. (canceled) 52. (canceled) 53. (canceled) 54. (canceled) 55. (canceled) 56. (canceled) 57. (canceled) 58. (canceled) 59. (canceled) 60. (canceled) 61. (canceled) 62. A process of claim 1, further comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and a pharmaceutically acceptable carrier to form a pharmaceutical composition. 63. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has a purity of about 95% or greater as determined by HPLC and has an enantiomeric excess of about 95% or greater. 64. The process according to claim 63, further comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and a pharmaceutically acceptable carrier to form a pharmaceutical composition. 65. The process according to claim 6, wherein said hydrolyzing in step a), is conducted in the presence of a phosphate buffer at a pH of about 7.6 to about 8.0. 66. The process according to claim 65, wherein said phosphate buffer is a potassium phosphate buffer. 67. The process according to claim 66, wherein said hydrolyzing in step a), is conducted at a temperature of about 30° C. to about 55° C. 68. The process according to claim 67, wherein said contacting-step solvent comprises isopropyl alcohol. 69. The process according to claim 68, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has a purity of about 95% or greater as determined by HPLC and has an enantiomeric excess of about 95% or greater. 70. The process according to claim 69, further comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and a pharmaceutically acceptable carrier to form a pharmaceutical composition. | The present invention relates to processes and intermediates useful in the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia) and salts thereof, an S1P1 receptor modulator that is useful in the treatment of S1P1 receptor-associated disorders, for example, diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders (e.g., acute and chronic inflammatory conditions), cancer, and conditions characterized by an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic (e.g., as may occur in inflammation, tumor development and atherosclerosis).1. A process for preparing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia):
comprising the following steps:
a) hydrolyzing a compound of Formula (IIk):
wherein R3 is C1-C6 alkyl; in the presence of immobilized Candida antarctica lipase B and a hydrolyzing-step solvent to form said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia); and
b) contacting said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Formula (Ia)) with L-arginine or a salt thereof, in the presence of a contacting-step solvent and H2O to form said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (Ia). 2. The process according to claim 1, wherein R3 is ethyl. 3. (canceled) 4. The process according to claim 1, wherein said hydrolyzing-step solvent comprises dimethylformamide (DMF), dimethylacetamide (DMA), dimethylsulfoxide (DMSO), tetrahydrofuran (THF), or acetonitrile. 5. The process according to claim 1, wherein said hydrolyzing-step solvent comprises acetonitrile. 6. The process according to claim 1, wherein: said compound of Formula (IIk) is:
and said hydrolyzing-step solvent comprises acetonitrile. 7. The process according to claim 1, wherein said hydrolyzing in step a), is conducted in the presence of a phosphate buffer at a pH of about 7.6 to about 8.0. 8. The process according to claim 7, wherein said phosphate buffer is a potassium phosphate buffer. 9. The process according to claim 1, wherein said hydrolyzing in step a), is conducted at a temperature of about 30° C. to about 55° C. 10. The process according to claim 1, wherein said hydrolyzing in step a), further comprises the step of isolating said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has an enantiomeric excess of about 95% or greater. 11. The process according to claim 1, wherein said contacting-step solvent comprises isopropyl alcohol. 12. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has a purity of about 95% or greater as determined by HPLC. 13. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has an enantiomeric excess of about 95% or greater. 14. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid wherein said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid is a crystalline form. 15. The process according to claim 14, wherein said crystalline form has a powder X-ray diffraction (PXRD) pattern substantially as shown in FIG. 5, a differential scanning calorimetry (DSC) trace substantially as shown in FIG. 6, a thermogravimetric analysis (TGA) profile substantially as shown in FIG. 7, or a dynamic vapor sorption (DVS) profile substantially as shown in FIG. 8. 16. (canceled) 17. (canceled) 18. (canceled) 19. (canceled) 20. (canceled) 21. (canceled) 22. (canceled) 23. (canceled) 24. (canceled) 25. (canceled) 26. (canceled) 27. (canceled) 28. (canceled) 29. (canceled) 30. (canceled) 31. (canceled) 32. (canceled) 33. (canceled) 34. (canceled) 35. (canceled) 36. (canceled) 37. (canceled) 38. (canceled) 39. (canceled) 40. (canceled) 41. (canceled) 42. (canceled) 43. (canceled) 44. (canceled) 45. (canceled) 46. (canceled) 47. (canceled) 48. (canceled) 49. (canceled) 50. (canceled) 51. (canceled) 52. (canceled) 53. (canceled) 54. (canceled) 55. (canceled) 56. (canceled) 57. (canceled) 58. (canceled) 59. (canceled) 60. (canceled) 61. (canceled) 62. A process of claim 1, further comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and a pharmaceutically acceptable carrier to form a pharmaceutical composition. 63. The process according to claim 1, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has a purity of about 95% or greater as determined by HPLC and has an enantiomeric excess of about 95% or greater. 64. The process according to claim 63, further comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and a pharmaceutically acceptable carrier to form a pharmaceutical composition. 65. The process according to claim 6, wherein said hydrolyzing in step a), is conducted in the presence of a phosphate buffer at a pH of about 7.6 to about 8.0. 66. The process according to claim 65, wherein said phosphate buffer is a potassium phosphate buffer. 67. The process according to claim 66, wherein said hydrolyzing in step a), is conducted at a temperature of about 30° C. to about 55° C. 68. The process according to claim 67, wherein said contacting-step solvent comprises isopropyl alcohol. 69. The process according to claim 68, further comprising the step of isolating said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, wherein after said isolating, said L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid has a purity of about 95% or greater as determined by HPLC and has an enantiomeric excess of about 95% or greater. 70. The process according to claim 69, further comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and a pharmaceutically acceptable carrier to form a pharmaceutical composition. | 1,600 |
696 | 15,301,347 | 1,627 | The present invention relates to the use of compounds of the formula (I)
in which R 1 , R 2 , R 3 , n, X and Y have the meanings given in the description, for controlling animal pests, in particular insects and/or spider mites and/or nematodes, by drenching the soil, by drip application to the soil, by immersing roots, tubers or bulbs, or by soil injection or for treating seed. | 1. A method of using one or more N-arylamidine-substituted trifluoroethyl sulphoxide derivatives of formula (I)
where
n represents the number 0 or 1,
X, Y independently of one another
represent hydrogen, fluorine, chlorine, bromine, iodine, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy or aminothiocarbonyl,
or represent (C3-C6)-cycloalkyl, phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, pyrazolyl, triazolyl or tetrazolyl which is optionally mono- or disubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy or (C3-C6)-cycloalkyl,
R3 represents hydrogen, (C2-C4)-alkyl, cyano, (C1-C4)-haloalkyl, (C1-C4)-alkoxy-(C1-C4)-alkyl or (C1-C4)-cyanoalkyl,
or represents a 3- to 6-membered saturated, partially saturated or aromatic ring which may optionally contain one to two heteroatoms from the group consisting of O, S and N and which is optionally mono- to trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, iodine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl,
R1 and R2 independently of one another
represent hydrogen, cyano, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-cyanoalkyl, (C1-C4)-hydroxyalkyl, (C1-C4)-alkoxy-(C1-C4)-alkyl, (C2-C4)-alkenyl or (C2-C4)-alkynyl,
or represent (C1-C4)-alkylcarbonyl, (C1-C5)-alkoxycarbonyl, arylcarbonyl, thiophenylcarbonyl, pyridylcarbonyl, pyrimidylcarbonyl, thiazolylcarbonyl, pyrazolylcarbonyl, (C1-C4)-alkylsulphinyl, (C1-C4)-haloalkylsulphinyl, arylsulphinyl, aryl-(C1-C4)-alkylsulphinyl, hetarylsulphinyl, hetaryl-(C1-C4)-alkylsulphinyl, (C1-C4)-alkylsulphonyl, (C1-C4)-haloalkylsulphonyl, arylsulphonyl, aryl-(C1-C4)-alkylsulphonyl, hetarylsulphonyl, or hetaryl-(C1-C4)-alkylsulphonyl optionally mono- to trisubstituted independently of one another by fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-haloalkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy, (C1-C4)-alkylthio, (C1-C4)-alkylsulphinyl, (C1-C4)-alkylsulphonyl, (C1-C4)-alkylamino, di-(C1-C4)-alkylamino,
or represent a 3- to 6-membered saturated or aromatic ring which may optionally contain one to two heteroatoms from the group consisting of O, S and N, which may optionally be interrupted once or twice by C═O and which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl,
or represent —(CH2)m—R6 or —(CH2)m—O—R6, where R6 represents a 3- to 6-membered saturated, partially saturated or aromatic ring which may optionally contain one to two heteroatoms from the group consisting of O, S and N, which may optionally be interrupted once or twice by C═O and which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl, where m represents the number 1 or 2,
or
R1 and R2 together with the nitrogen atom to which they are attached may form a saturated or unsaturated 3- to 6-membered ring which is optionally mono- or tetrasubstituted by fluorine, chlorine, (C1-C4)-alkyl, (C1-C4)-alkoxy or (C1-C4)-haloalkyl and which may optionally contain a further heteroatom selected from the group consisting of sulphur, oxygen and nitrogen and/or at least one carbonyl group, or
R1 and R3 together with the atoms to which they are attached may form a saturated or unsaturated 5- to 6-membered ring which is optionally mono- or polysubstituted by methyl, ethyl, methoxy, ethoxy, fluorine, chlorine, trifluoromethyl, cyclopropyl, cyano, chlorocyclopropyl, fluorocyclopropyl, cyanocyclopropyl, methylcyclopropyl, (C2-C4)-alkanediyl, (C2-C4)-alkenediyl or butanedienyl (where butanedienyl may optionally be mono- or disubstituted by methyl, fluorine, chlorine, bromine, methoxy or trifluoromethyl and/or may optionally be interrupted by at least one oxygen or/and nitrogen atom) and which may optionally contain a further heteroatom selected from the group consisting of sulphur, oxygen and nitrogen and/or a carbonyl group,
for controlling animal pests, optionally insects and/or spider mites and/or nematodes, by drenching the soil, by drip application to the soil, by immersing roots, tubers or bulbs or by soil injection and/or for treating seed. 2. Method according to claim 1, wherein
n represents the number 0 or 1, X and Y independently of one another represent hydrogen, fluorine, chlorine, bromine, methyl, ethyl, trifluoromethyl, difluoromethyl, difluoromethoxy, trifluoromethoxy or OCH2CF3,
where X and Y represent in particular one of the following combinations X/Y: H/F, H/Cl, H/Br, H/methyl, H/ethyl, F/F, Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl, methyl/Br, Br/methyl, ethyl/ethyl, F/ethyl, ethyl/Cl, Cl/ethyl, ethyl/Br, Br/ethyl or H/trifluoromethyl,
R3 represents hydrogen, ethyl, propyl, cyano, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl, dichlorofluoromethyl, (2,2,2)-trifluoroethyl, 2-chloro-(2,2)-difluoroethyl, (2,2)-dichloro-2-fluoroethyl, (2,2,2)-trichloroethyl or pentafluoroethyl, or represents (C3-C6)-cycloalkyl, phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, pyrazolyl, triazolyl or tetrazolyl which is optionally mono- or disubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl, R1 represents hydrogen, methyl, ethyl, propyl, butyl, sec-butyl, isopropyl, tert-butyl, (2,2,2)-trifluoroethyl, (2,2)-difluoroethyl, methoxy, ethoxy, methoxymethyl, 2-methoxyethyl, cyanomethyl, 1-cyanoethyl or 2-cyanoethyl, or represents aryl which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, optionally phenyl which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, or represents —(CH2)m—R6, where R6 represents aryl which is optionally mono-, di- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, where m represents the number 1, optionally where R6 represents phenyl which is optionally mono-, di- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, R2 represents hydrogen, methyl, ethyl, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl, dichlorofluoromethyl, (2,2,2)-trifluoroethyl, (2,2)-difluoroethyl, 2-chloro-(2,2)-difluoroethyl, (2,2)-dichloro-2-fluoroethyl, (2,2,2)-trichloroethyl or pentafluoroethyl, or R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the group may optionally be mono- or disubstituted by methyl, ethyl, methoxy, ethoxy, fluorine, chlorine, trifluoromethyl, difluoromethyl, cyclopropyl, chlorocyclopropyl, fluorocyclopropyl, cyanocyclopropyl, methylcyclopropyl, and
where the arrow points to the remainder of the molecule. 3. Method according to claim 1, wherein
n represents the number 1, X and Y represent the following combinations X/Y: Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl or H/trifluoromethyl, R3 represents ethyl, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl or dichlorofluoromethyl, or represents cyclopropyl which may optionally be monosubstituted by fluorine, chlorine, cyano, methyl, trifluoromethyl or methoxy, or represents phenyl, pyridyl, pyrimidyl, pyrazinyl or pyridazinyl which is optionally monosubstituted by fluorine, chlorine, cyano, nitro, methyl, trifluoromethyl, methoxy, difluoromethoxy or trifluoromethoxy, R1 represents hydrogen, methyl or ethyl, R2 represents hydrogen, methyl, ethyl, (2,2)-difluoroethyl or (2,2,2)-trifluoroethyl, or R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the arrow points to the remainder of the molecule. 4. Method according to claim 1, wherein
n represents the number 1, X and Y represent the following combinations X/Y: Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl or H/trifluoromethyl, R3 represents ethyl, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl or dichlorofluoromethyl, or represents cyclopropyl which may optionally be monosubstituted by fluorine, chlorine, cyano, methyl, trifluoromethyl or methoxy, or represents phenyl, pyridyl, pyrimidyl, pyrazinyl or pyridazinyl which is optionally monosubstituted by fluorine, chlorine, cyano, nitro, methyl, trifluoromethyl, methoxy, difluoromethoxy or trifluoromethoxy, R1 represents hydrogen, methyl or ethyl, R2 represents hydrogen, methyl, ethyl, (2,2)-difluoroethyl or (2,2,2)-trifluoroethyl, or R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the arrow points to the remainder of the molecule. 5. Method according to claim 1, wherein
n represents the number 1, X and Y represent the following combinations X/Y: Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl or H/trifluoromethyl, R2 represents hydrogen, methyl, ethyl, (2,2)-difluoroethyl or (2,2,2)-trifluoroethyl, and R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the arrow points to the remainder of the molecule. 6. Method according to claim 1, wherein the compound of the formula (I) is its R enantiomer, with respect to the chiral sulphur atom, or is a mixture of the R enantiomer and the S enantiomer in which the proportion of the R enantiomer is at least 55% by weight, based on the enantiomer mixture. 7. Method according to claim 1, wherein the compound of the formula (I) is its S enantiomer, with respect to the chiral sulphur atom, or is a mixture of the S enantiomer and the R enantiomer in which the proportion of the S enantiomer is at least 55% by weight, based on the enantiomer mixture. 8. Method according to claim 1, where the plant to be treated is grown in an artificial growth substrate. 9. Method according to claim 1, where the plant to be treated is planted in a closed system. 10. Method according to claim 1, where the plants to be treated are selected from the group consisting of
vegetables, optionally fruiting vegetables or inflorescences, optionally bell peppers, chillies, tomatoes, aubergines, cucumbers, pumpkins, courgettes, broad beans, runner beans, dwarf beans, peas, artichokes, maize; leafy vegetables, head-forming lettuce, chicory, endives, various types of cress, rocket, lamb's lettuce, iceberg lettuce, leeks, spinach, Swiss chard; tuber, root or stem vegetables, optionally celeriac/celery, beetroot, carrots, radishes, horseradish, beets for human consumption scorzonera, asparagus, palm shoots, bamboo shoots; bulb vegetables, optionally onions, leek, fennel, garlic; Brassica vegetables, optionally cauliflower, broccoli, kohlrabi, red cabbage, white cabbage, curly kale, Savoy cabbage, Brussel sprouts, Chinese cabbage; citrus, optionally oranges, grapefruits, tangerines, lemons, limes, seville oranges, kumquats, satsumas; pome fruit, optionally apples, pears or quinces; stone fruit, optionally peaches, nectarines, cherries, plums, quetsch, apricots, almonds, pistachios, olives; grapevines, hops, tea or tropical crops, in particular mangoes, papayas, figs, pineapples, dates, bananas, durians, kaki fruit, coconuts, cacao, coffee, avocados, lychees, maracujas, guavas, almonds or nuts, optionally hazelnuts, walnuts, pistachios, cashew nuts, para nuts, pecan nuts, butternuts, chestnuts, hickory nuts, macadamia nuts, peanuts; soft fruit, optionally redcurrants, gooseberries, raspberries, blackberries, blueberries, strawberries, cranberries, including American cranberries, kiwi fruit; cut flowers, optionally roses, carnations, gerbera, lilies, marguerites, chrysanthemums, tulips, narcissi, anemones, poppies, amaryllis, dahlias, azaleas, mauves; bedding plants, pot plants or perennials, optionally roses, Tagetes, violas, geraniums, fuchsias, hibiscus, chrysanthemums, busy lizzies, cyclamen, African violet, sunflowers, begonias; bushes or conifers, optionally ficus, rhododendron, firs, spruces, pines, including umbrella pines, yews, juniper, oleander; spices, optionally aniseed, chilli pepper, paprika, pepper, vanilla, majoram, thyme, cloves, juniper berries, cinnamon, tarragon, coriander, saffron, ginger; cotton; soya beans. 11. Method according to claim 1 for treating seed. 12. Method according to claim 1 for treating seed of one or more transgenic plants. | The present invention relates to the use of compounds of the formula (I)
in which R 1 , R 2 , R 3 , n, X and Y have the meanings given in the description, for controlling animal pests, in particular insects and/or spider mites and/or nematodes, by drenching the soil, by drip application to the soil, by immersing roots, tubers or bulbs, or by soil injection or for treating seed.1. A method of using one or more N-arylamidine-substituted trifluoroethyl sulphoxide derivatives of formula (I)
where
n represents the number 0 or 1,
X, Y independently of one another
represent hydrogen, fluorine, chlorine, bromine, iodine, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy or aminothiocarbonyl,
or represent (C3-C6)-cycloalkyl, phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, pyrazolyl, triazolyl or tetrazolyl which is optionally mono- or disubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy or (C3-C6)-cycloalkyl,
R3 represents hydrogen, (C2-C4)-alkyl, cyano, (C1-C4)-haloalkyl, (C1-C4)-alkoxy-(C1-C4)-alkyl or (C1-C4)-cyanoalkyl,
or represents a 3- to 6-membered saturated, partially saturated or aromatic ring which may optionally contain one to two heteroatoms from the group consisting of O, S and N and which is optionally mono- to trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, iodine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl,
R1 and R2 independently of one another
represent hydrogen, cyano, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-cyanoalkyl, (C1-C4)-hydroxyalkyl, (C1-C4)-alkoxy-(C1-C4)-alkyl, (C2-C4)-alkenyl or (C2-C4)-alkynyl,
or represent (C1-C4)-alkylcarbonyl, (C1-C5)-alkoxycarbonyl, arylcarbonyl, thiophenylcarbonyl, pyridylcarbonyl, pyrimidylcarbonyl, thiazolylcarbonyl, pyrazolylcarbonyl, (C1-C4)-alkylsulphinyl, (C1-C4)-haloalkylsulphinyl, arylsulphinyl, aryl-(C1-C4)-alkylsulphinyl, hetarylsulphinyl, hetaryl-(C1-C4)-alkylsulphinyl, (C1-C4)-alkylsulphonyl, (C1-C4)-haloalkylsulphonyl, arylsulphonyl, aryl-(C1-C4)-alkylsulphonyl, hetarylsulphonyl, or hetaryl-(C1-C4)-alkylsulphonyl optionally mono- to trisubstituted independently of one another by fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-haloalkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy, (C1-C4)-alkylthio, (C1-C4)-alkylsulphinyl, (C1-C4)-alkylsulphonyl, (C1-C4)-alkylamino, di-(C1-C4)-alkylamino,
or represent a 3- to 6-membered saturated or aromatic ring which may optionally contain one to two heteroatoms from the group consisting of O, S and N, which may optionally be interrupted once or twice by C═O and which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl,
or represent —(CH2)m—R6 or —(CH2)m—O—R6, where R6 represents a 3- to 6-membered saturated, partially saturated or aromatic ring which may optionally contain one to two heteroatoms from the group consisting of O, S and N, which may optionally be interrupted once or twice by C═O and which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl, where m represents the number 1 or 2,
or
R1 and R2 together with the nitrogen atom to which they are attached may form a saturated or unsaturated 3- to 6-membered ring which is optionally mono- or tetrasubstituted by fluorine, chlorine, (C1-C4)-alkyl, (C1-C4)-alkoxy or (C1-C4)-haloalkyl and which may optionally contain a further heteroatom selected from the group consisting of sulphur, oxygen and nitrogen and/or at least one carbonyl group, or
R1 and R3 together with the atoms to which they are attached may form a saturated or unsaturated 5- to 6-membered ring which is optionally mono- or polysubstituted by methyl, ethyl, methoxy, ethoxy, fluorine, chlorine, trifluoromethyl, cyclopropyl, cyano, chlorocyclopropyl, fluorocyclopropyl, cyanocyclopropyl, methylcyclopropyl, (C2-C4)-alkanediyl, (C2-C4)-alkenediyl or butanedienyl (where butanedienyl may optionally be mono- or disubstituted by methyl, fluorine, chlorine, bromine, methoxy or trifluoromethyl and/or may optionally be interrupted by at least one oxygen or/and nitrogen atom) and which may optionally contain a further heteroatom selected from the group consisting of sulphur, oxygen and nitrogen and/or a carbonyl group,
for controlling animal pests, optionally insects and/or spider mites and/or nematodes, by drenching the soil, by drip application to the soil, by immersing roots, tubers or bulbs or by soil injection and/or for treating seed. 2. Method according to claim 1, wherein
n represents the number 0 or 1, X and Y independently of one another represent hydrogen, fluorine, chlorine, bromine, methyl, ethyl, trifluoromethyl, difluoromethyl, difluoromethoxy, trifluoromethoxy or OCH2CF3,
where X and Y represent in particular one of the following combinations X/Y: H/F, H/Cl, H/Br, H/methyl, H/ethyl, F/F, Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl, methyl/Br, Br/methyl, ethyl/ethyl, F/ethyl, ethyl/Cl, Cl/ethyl, ethyl/Br, Br/ethyl or H/trifluoromethyl,
R3 represents hydrogen, ethyl, propyl, cyano, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl, dichlorofluoromethyl, (2,2,2)-trifluoroethyl, 2-chloro-(2,2)-difluoroethyl, (2,2)-dichloro-2-fluoroethyl, (2,2,2)-trichloroethyl or pentafluoroethyl, or represents (C3-C6)-cycloalkyl, phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, pyrazolyl, triazolyl or tetrazolyl which is optionally mono- or disubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, (C1-C4)-alkyl, (C1-C4)-haloalkyl, (C1-C4)-alkoxy, (C1-C4)-haloalkoxy and (C3-C6)-cycloalkyl, R1 represents hydrogen, methyl, ethyl, propyl, butyl, sec-butyl, isopropyl, tert-butyl, (2,2,2)-trifluoroethyl, (2,2)-difluoroethyl, methoxy, ethoxy, methoxymethyl, 2-methoxyethyl, cyanomethyl, 1-cyanoethyl or 2-cyanoethyl, or represents aryl which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, optionally phenyl which is optionally mono- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, or represents —(CH2)m—R6, where R6 represents aryl which is optionally mono-, di- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, where m represents the number 1, optionally where R6 represents phenyl which is optionally mono-, di- or trisubstituted by identical or different substituents from the group consisting of fluorine, chlorine, bromine, cyano, nitro, methyl, ethyl, trifluoromethyl, vinyl, ethynyl, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, trifluoroethoxy and cyclopropyl, R2 represents hydrogen, methyl, ethyl, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl, dichlorofluoromethyl, (2,2,2)-trifluoroethyl, (2,2)-difluoroethyl, 2-chloro-(2,2)-difluoroethyl, (2,2)-dichloro-2-fluoroethyl, (2,2,2)-trichloroethyl or pentafluoroethyl, or R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the group may optionally be mono- or disubstituted by methyl, ethyl, methoxy, ethoxy, fluorine, chlorine, trifluoromethyl, difluoromethyl, cyclopropyl, chlorocyclopropyl, fluorocyclopropyl, cyanocyclopropyl, methylcyclopropyl, and
where the arrow points to the remainder of the molecule. 3. Method according to claim 1, wherein
n represents the number 1, X and Y represent the following combinations X/Y: Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl or H/trifluoromethyl, R3 represents ethyl, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl or dichlorofluoromethyl, or represents cyclopropyl which may optionally be monosubstituted by fluorine, chlorine, cyano, methyl, trifluoromethyl or methoxy, or represents phenyl, pyridyl, pyrimidyl, pyrazinyl or pyridazinyl which is optionally monosubstituted by fluorine, chlorine, cyano, nitro, methyl, trifluoromethyl, methoxy, difluoromethoxy or trifluoromethoxy, R1 represents hydrogen, methyl or ethyl, R2 represents hydrogen, methyl, ethyl, (2,2)-difluoroethyl or (2,2,2)-trifluoroethyl, or R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the arrow points to the remainder of the molecule. 4. Method according to claim 1, wherein
n represents the number 1, X and Y represent the following combinations X/Y: Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl or H/trifluoromethyl, R3 represents ethyl, trifluoromethyl, difluoromethyl, dichloromethyl, chloromethyl, trichloromethyl, difluorochloromethyl or dichlorofluoromethyl, or represents cyclopropyl which may optionally be monosubstituted by fluorine, chlorine, cyano, methyl, trifluoromethyl or methoxy, or represents phenyl, pyridyl, pyrimidyl, pyrazinyl or pyridazinyl which is optionally monosubstituted by fluorine, chlorine, cyano, nitro, methyl, trifluoromethyl, methoxy, difluoromethoxy or trifluoromethoxy, R1 represents hydrogen, methyl or ethyl, R2 represents hydrogen, methyl, ethyl, (2,2)-difluoroethyl or (2,2,2)-trifluoroethyl, or R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the arrow points to the remainder of the molecule. 5. Method according to claim 1, wherein
n represents the number 1, X and Y represent the following combinations X/Y: Cl/Cl, F/Cl, Br/Br, Br/Cl, Cl/Br, F/Br, methyl/methyl, F/methyl, methyl/Cl, Cl/methyl or H/trifluoromethyl, R2 represents hydrogen, methyl, ethyl, (2,2)-difluoroethyl or (2,2,2)-trifluoroethyl, and R1 and R3 together with the atoms to which they are attached represent one of the groups below
where the arrow points to the remainder of the molecule. 6. Method according to claim 1, wherein the compound of the formula (I) is its R enantiomer, with respect to the chiral sulphur atom, or is a mixture of the R enantiomer and the S enantiomer in which the proportion of the R enantiomer is at least 55% by weight, based on the enantiomer mixture. 7. Method according to claim 1, wherein the compound of the formula (I) is its S enantiomer, with respect to the chiral sulphur atom, or is a mixture of the S enantiomer and the R enantiomer in which the proportion of the S enantiomer is at least 55% by weight, based on the enantiomer mixture. 8. Method according to claim 1, where the plant to be treated is grown in an artificial growth substrate. 9. Method according to claim 1, where the plant to be treated is planted in a closed system. 10. Method according to claim 1, where the plants to be treated are selected from the group consisting of
vegetables, optionally fruiting vegetables or inflorescences, optionally bell peppers, chillies, tomatoes, aubergines, cucumbers, pumpkins, courgettes, broad beans, runner beans, dwarf beans, peas, artichokes, maize; leafy vegetables, head-forming lettuce, chicory, endives, various types of cress, rocket, lamb's lettuce, iceberg lettuce, leeks, spinach, Swiss chard; tuber, root or stem vegetables, optionally celeriac/celery, beetroot, carrots, radishes, horseradish, beets for human consumption scorzonera, asparagus, palm shoots, bamboo shoots; bulb vegetables, optionally onions, leek, fennel, garlic; Brassica vegetables, optionally cauliflower, broccoli, kohlrabi, red cabbage, white cabbage, curly kale, Savoy cabbage, Brussel sprouts, Chinese cabbage; citrus, optionally oranges, grapefruits, tangerines, lemons, limes, seville oranges, kumquats, satsumas; pome fruit, optionally apples, pears or quinces; stone fruit, optionally peaches, nectarines, cherries, plums, quetsch, apricots, almonds, pistachios, olives; grapevines, hops, tea or tropical crops, in particular mangoes, papayas, figs, pineapples, dates, bananas, durians, kaki fruit, coconuts, cacao, coffee, avocados, lychees, maracujas, guavas, almonds or nuts, optionally hazelnuts, walnuts, pistachios, cashew nuts, para nuts, pecan nuts, butternuts, chestnuts, hickory nuts, macadamia nuts, peanuts; soft fruit, optionally redcurrants, gooseberries, raspberries, blackberries, blueberries, strawberries, cranberries, including American cranberries, kiwi fruit; cut flowers, optionally roses, carnations, gerbera, lilies, marguerites, chrysanthemums, tulips, narcissi, anemones, poppies, amaryllis, dahlias, azaleas, mauves; bedding plants, pot plants or perennials, optionally roses, Tagetes, violas, geraniums, fuchsias, hibiscus, chrysanthemums, busy lizzies, cyclamen, African violet, sunflowers, begonias; bushes or conifers, optionally ficus, rhododendron, firs, spruces, pines, including umbrella pines, yews, juniper, oleander; spices, optionally aniseed, chilli pepper, paprika, pepper, vanilla, majoram, thyme, cloves, juniper berries, cinnamon, tarragon, coriander, saffron, ginger; cotton; soya beans. 11. Method according to claim 1 for treating seed. 12. Method according to claim 1 for treating seed of one or more transgenic plants. | 1,600 |
697 | 13,983,651 | 1,631 | The present invention relates to a method for stratifying a patient into a clinically relevant group comprising the identification of the probability of an alteration within one or more sets of molecular data from a patient sample in comparison to a database of molecular data of known phenotypes, the inference of the activity of a biological network on the basis of the probabilities, the identification of a network information flow probability for the patient via the probability of interactions in the network, the creation of multiple instances of network information flow for the patient sample and the calculation of the distance of the patient from other subjects in a patient database using multiple instances of the network information flow. The invention further relates to a biomedical marker or group of biomedical markers associated with a high likelihood of responsiveness of a subject to a cancer therapy wherein the biomedical marker or group of biomedical markers comprises altered biological pathway markers, as well as to an assay for detecting, diagnosing, graduating, monitoring or prognosticating a medical condition, or for detecting, diagnosing, monitoring or prognosticating the responsiveness of a subject to a therapy against said medical condition, in particular ovarian cancer. Furthermore, a corresponding clinical decision support system is provided. | 1. A method for stratifying a patient into a clinically relevant group, comprising with a computer performing the steps of:
obtaining datasets comprising one or more sets of molecular data from a patient sample; identifying the probability of an alteration within the one or more sets of molecular data in comparison to a database of molecular data of known phenotypes, preferably molecular data of the expression of one or more of the patient's genes; inferring the activity of a biological network on the basis of said probabilities; identifying a network information flow probability for said patient via the probability of interactions in said network based on said probability of altered molecular data; creating multiple instances of network information flow vectors for said patient sample by sampling from a full interaction probability distribution of the biological network; calculating the distance of said patient from other subjects in a patient database using the multiple instances of network information flow vectors; and assigning said patient to a clinically relevant group based on the outcome of the previous step. 2. The method of claim 1, wherein said molecular data comprise data on nonsense mutations, single nucleotide polymorphisms (SNP), copy number variations (CNV), splicing variations, variations of a regulatory sequence, small deletions, small insertions, small indels, gross deletions, gross insertions, complex genetic rearrangements, inter chromosomal rearrangements, intra chromosomal rearrangements, loss of heterozygosity, insertion of repeats, deletion of repeats, DNA methylation, histone methylation or acetylation states, gene and/or non-coding RNA expression and/or chromatin precipitation data revealing DNA binding sites or regions, preferably obtained by genome sequencing, immunohistochemistry, FISH, PCR-techniques and/or microarray-techniques. 3. The method of claim 1, wherein said comparison to a database of molecular data of known phenotypes is a comparison to a biological annotation database, a pathway database, a database on biological processes and/or a database on biological functions, preferably the National Cancer Institute Pathway interaction database, the KEGG pathway database, the BioCarta database, the Panther database, the Reactome database, and/or the DAVID database. 4. The method of claim 3, wherein the probability of an alteration within the one or more sets of molecular data is identified by estimating altered expression levels of individual genes in the network by integrating said molecular data using a probabilistic graphical model framework, preferably factor graphs. 5. The method of claim 3, wherein the probability of an alteration within the one or more sets of molecular data is identified by estimating altered copy number levels, altered methylation states, or altered gene function due to mutations of genomic loci or genomic regions in the network by integrating said molecular data using a probabilistic graphical model framework, preferably factor graphs. 6. The method of claim 1, wherein said interactions are interactions for genes or genomic loci with molecular alterations, preferably genes or genomic loci belonging to biological networks as defined in a pathway database. 7. The method of claim 1, wherein said creation of multiple instances of network information flow vectors is used for the generation of a distribution of sample information flow vectors, representing the information flow in a network for said patient. 8. The method of claim 7, wherein said distance of said patient from other subjects is calculated as the average of pairwise distance of sample information flow vectors in a given network. 9. The method of claim 8, wherein said pairwise distance of sample information flow vectors is calculated as the Euclidean distance between the sample information flow vectors in a given network, or as a weighted Euclidean distance, wherein the weights for each entry in the information flow vector are proportional to the depth of that interaction in the given network. 10. The method of claim 1, wherein said assignment of said patient to a clinically relevant group is performed with a clustering algorithm based on the pairwise distances of said patient with one, more or all subjects in a patient database. 11. The method of claim 1, wherein said patient database is a disease related database, preferably a cancer disease related database. 12. The method of claim 1, wherein said clinically relevant group is associated with a cancerous disease, preferably ovarian cancer, breast cancer, or prostate cancer, or with the likelihood of recurrence of a cancerous disease in a subject after a therapy, or wherein said clinically relevant group is associated with the likelihood of responsiveness of a subject to a therapy comprising one or more platinum based drugs. 13. A biomedical marker or group of biomedical markers for use in performing the method of claim 12 said biomedical marker or group of biomedical markers comprising at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or all markers selected from an altered endothelin pathway, an altered ceramide signaling pathway, an altered rapid glucocorticoid signaling pathway, an altered paxilin independent a4b1 and a4b7 pathway, an altered osteopontin pathway, an altered ILE signaling pathway, an altered telomerase pathway, an altered JNK signaling pathway in the CD4+TCR pathway, an altered PLK2- and PLK4-pathway, an altered EPO-signaling pathway, an altered p53-pathway, an altered VEGFR1- and VEGFR-2 signaling pathway, an altered VEGFR1-specific pathway, and an altered syndecan-1 signaling pathway, indicated in Table 1. 14. An assay for detecting, diagnosing, graduating, monitoring or prognosticating a medical condition, or for detecting, diagnosing, monitoring or prognosticating the responsiveness of a subject to a therapy against said medical condition, preferably cancer, more preferably ovarian cancer, comprising at least the steps of
(a) testing in a sample obtained from a subject for the alteration of a stratifying biomedical marker or group of biomedical markers as defined in claim 13; (b) testing in a control sample for alterations of the same marker or group of markers as in (a); (c) determining the difference in alterations of markers of steps (a) and (b); and (d) deciding on the presence or stage of a medical condition or the responsiveness of a subject to a therapy against said medical condition, preferably cancer, more preferably ovarian cancer, based on the results obtained in step (c). 15. A clinical decision support system comprising:
an input for providing datasets comprising multi-modality molecular profiling data from a patient; a computer program product for enabling a processor to carry out the method of claim 1 and for quantifying the degree of alteration of information flow of a biological network in said patient; and an output for outputting the assignment of a patient to a clinically relevant group, wherein said assignment of a patient to a clinically relevant groups is preferably visualized in the context of the information flow in the networks and other clinically relevant groups and/or healthy subjects. | The present invention relates to a method for stratifying a patient into a clinically relevant group comprising the identification of the probability of an alteration within one or more sets of molecular data from a patient sample in comparison to a database of molecular data of known phenotypes, the inference of the activity of a biological network on the basis of the probabilities, the identification of a network information flow probability for the patient via the probability of interactions in the network, the creation of multiple instances of network information flow for the patient sample and the calculation of the distance of the patient from other subjects in a patient database using multiple instances of the network information flow. The invention further relates to a biomedical marker or group of biomedical markers associated with a high likelihood of responsiveness of a subject to a cancer therapy wherein the biomedical marker or group of biomedical markers comprises altered biological pathway markers, as well as to an assay for detecting, diagnosing, graduating, monitoring or prognosticating a medical condition, or for detecting, diagnosing, monitoring or prognosticating the responsiveness of a subject to a therapy against said medical condition, in particular ovarian cancer. Furthermore, a corresponding clinical decision support system is provided.1. A method for stratifying a patient into a clinically relevant group, comprising with a computer performing the steps of:
obtaining datasets comprising one or more sets of molecular data from a patient sample; identifying the probability of an alteration within the one or more sets of molecular data in comparison to a database of molecular data of known phenotypes, preferably molecular data of the expression of one or more of the patient's genes; inferring the activity of a biological network on the basis of said probabilities; identifying a network information flow probability for said patient via the probability of interactions in said network based on said probability of altered molecular data; creating multiple instances of network information flow vectors for said patient sample by sampling from a full interaction probability distribution of the biological network; calculating the distance of said patient from other subjects in a patient database using the multiple instances of network information flow vectors; and assigning said patient to a clinically relevant group based on the outcome of the previous step. 2. The method of claim 1, wherein said molecular data comprise data on nonsense mutations, single nucleotide polymorphisms (SNP), copy number variations (CNV), splicing variations, variations of a regulatory sequence, small deletions, small insertions, small indels, gross deletions, gross insertions, complex genetic rearrangements, inter chromosomal rearrangements, intra chromosomal rearrangements, loss of heterozygosity, insertion of repeats, deletion of repeats, DNA methylation, histone methylation or acetylation states, gene and/or non-coding RNA expression and/or chromatin precipitation data revealing DNA binding sites or regions, preferably obtained by genome sequencing, immunohistochemistry, FISH, PCR-techniques and/or microarray-techniques. 3. The method of claim 1, wherein said comparison to a database of molecular data of known phenotypes is a comparison to a biological annotation database, a pathway database, a database on biological processes and/or a database on biological functions, preferably the National Cancer Institute Pathway interaction database, the KEGG pathway database, the BioCarta database, the Panther database, the Reactome database, and/or the DAVID database. 4. The method of claim 3, wherein the probability of an alteration within the one or more sets of molecular data is identified by estimating altered expression levels of individual genes in the network by integrating said molecular data using a probabilistic graphical model framework, preferably factor graphs. 5. The method of claim 3, wherein the probability of an alteration within the one or more sets of molecular data is identified by estimating altered copy number levels, altered methylation states, or altered gene function due to mutations of genomic loci or genomic regions in the network by integrating said molecular data using a probabilistic graphical model framework, preferably factor graphs. 6. The method of claim 1, wherein said interactions are interactions for genes or genomic loci with molecular alterations, preferably genes or genomic loci belonging to biological networks as defined in a pathway database. 7. The method of claim 1, wherein said creation of multiple instances of network information flow vectors is used for the generation of a distribution of sample information flow vectors, representing the information flow in a network for said patient. 8. The method of claim 7, wherein said distance of said patient from other subjects is calculated as the average of pairwise distance of sample information flow vectors in a given network. 9. The method of claim 8, wherein said pairwise distance of sample information flow vectors is calculated as the Euclidean distance between the sample information flow vectors in a given network, or as a weighted Euclidean distance, wherein the weights for each entry in the information flow vector are proportional to the depth of that interaction in the given network. 10. The method of claim 1, wherein said assignment of said patient to a clinically relevant group is performed with a clustering algorithm based on the pairwise distances of said patient with one, more or all subjects in a patient database. 11. The method of claim 1, wherein said patient database is a disease related database, preferably a cancer disease related database. 12. The method of claim 1, wherein said clinically relevant group is associated with a cancerous disease, preferably ovarian cancer, breast cancer, or prostate cancer, or with the likelihood of recurrence of a cancerous disease in a subject after a therapy, or wherein said clinically relevant group is associated with the likelihood of responsiveness of a subject to a therapy comprising one or more platinum based drugs. 13. A biomedical marker or group of biomedical markers for use in performing the method of claim 12 said biomedical marker or group of biomedical markers comprising at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or all markers selected from an altered endothelin pathway, an altered ceramide signaling pathway, an altered rapid glucocorticoid signaling pathway, an altered paxilin independent a4b1 and a4b7 pathway, an altered osteopontin pathway, an altered ILE signaling pathway, an altered telomerase pathway, an altered JNK signaling pathway in the CD4+TCR pathway, an altered PLK2- and PLK4-pathway, an altered EPO-signaling pathway, an altered p53-pathway, an altered VEGFR1- and VEGFR-2 signaling pathway, an altered VEGFR1-specific pathway, and an altered syndecan-1 signaling pathway, indicated in Table 1. 14. An assay for detecting, diagnosing, graduating, monitoring or prognosticating a medical condition, or for detecting, diagnosing, monitoring or prognosticating the responsiveness of a subject to a therapy against said medical condition, preferably cancer, more preferably ovarian cancer, comprising at least the steps of
(a) testing in a sample obtained from a subject for the alteration of a stratifying biomedical marker or group of biomedical markers as defined in claim 13; (b) testing in a control sample for alterations of the same marker or group of markers as in (a); (c) determining the difference in alterations of markers of steps (a) and (b); and (d) deciding on the presence or stage of a medical condition or the responsiveness of a subject to a therapy against said medical condition, preferably cancer, more preferably ovarian cancer, based on the results obtained in step (c). 15. A clinical decision support system comprising:
an input for providing datasets comprising multi-modality molecular profiling data from a patient; a computer program product for enabling a processor to carry out the method of claim 1 and for quantifying the degree of alteration of information flow of a biological network in said patient; and an output for outputting the assignment of a patient to a clinically relevant group, wherein said assignment of a patient to a clinically relevant groups is preferably visualized in the context of the information flow in the networks and other clinically relevant groups and/or healthy subjects. | 1,600 |
698 | 16,405,419 | 1,612 | A dental tool is provided which is capable of delivering an oligosaccharide containing composition to an oral biofilm, which can prevent and inhibit periodontal disease and in particular to devices and methods which mechanically and chemically disrupt biofilms formed by S. mutans. | 1. A dental tool comprising:
a substrate; and a composition comprising a complex carbohydrate, wherein said dental tool is selected from the group consisting of a floss, a tape, a floss pick, an interdental brush, and a pick, and wherein said complex carbohydrate is a β-glucan having a molecular weight of said β-glucan is 360 to 20,000. 2. (canceled) 3. (canceled) 4. (canceled) 5. The dental tool according to claim 14, wherein said β-glucan is at least one selected from the group consisting of oat glucan, cellulose, hydroxymethylcellulose, hydroxyethylcellulose and carboxymethylcellulose. 6. The dental tool according to claim 1, wherein said tool is a dental floss. 7. The dental tool according to claim 6, wherein said substrate is a polymer, wherein said polymer is coated or impregnated with said composition. 8. The dental tool according to claim 6, wherein said dental floss has a denier of 100 to 1350. 9. (canceled) 10. The dental tool according to claim 1, wherein said complex carbohydrate has a solubility in water of at least 10 mM in water at 23° C. 11. A method of preventing and/or inhibiting periodontal disease comprising mechanically and chemically disrupting a biofilm with a dental tool comprising a complex carbohydrate composition,
wherein said dental tool is selected from the group consisting of a floss, a tape, a floss pick, an interdental brush, and a pick, and wherein said complex carbohydrate is a β-glucan having a molecular weight of said β-glucan is 360 to 20,000. 12. The method according to claim 11, wherein said biofilm is located within an oral cavity of a mammal. 13. The method according to claim 12, wherein said mammal is a selected from the group consisting of human, canines, equine, bovine and felines. 14. (canceled) 15. (canceled) 16. (canceled) 17. The method according to claim 11, wherein said β-glucan is at least one selected from the group consisting of oat glucan, cellulose, hydroxymethylcellulose, hydroxyethylcellulose and carboxymethylcellulose. 18. The method according to claim 11, wherein said complex carbohydrate has a solubility in water of at least 10 mM in water at 23° C. 19. (canceled) 20. (canceled) 21. (canceled) 22. (canceled) 23. The dental tool according to claim 1, wherein said complex carbohydrate bonds to a glycosyltransferase expressed on a cell surface of S. mutans. 24. The dental tool according to claim 1, wherein said β-glucan is an oat glucan. 25. The dental tool according to claim 1, wherein said β-glucan is cellulose. 26. The dental tool according to claim 1, wherein said β-glucan is hydroxymethylcellulose. 27. The dental tool according to claim 1, wherein said β-glucan is hydroxyethylcellulose. 28. The dental tool according to claim 1, wherein said β-glucan is carboxymethylcellulose. 29. The method according to claim 11, wherein said β-glucan is an oat glucan. 30. The method according to claim 11, wherein said β-glucan is cellulose. 31. The method according to claim 11, wherein said β-glucan is hydroxymethylcellulose. 32. The method according to claim 11, wherein said β-glucan is hydroxyethylcellulose. 33. The method according to claim 11, wherein said β-glucan is carboxymethylcellulose. | A dental tool is provided which is capable of delivering an oligosaccharide containing composition to an oral biofilm, which can prevent and inhibit periodontal disease and in particular to devices and methods which mechanically and chemically disrupt biofilms formed by S. mutans.1. A dental tool comprising:
a substrate; and a composition comprising a complex carbohydrate, wherein said dental tool is selected from the group consisting of a floss, a tape, a floss pick, an interdental brush, and a pick, and wherein said complex carbohydrate is a β-glucan having a molecular weight of said β-glucan is 360 to 20,000. 2. (canceled) 3. (canceled) 4. (canceled) 5. The dental tool according to claim 14, wherein said β-glucan is at least one selected from the group consisting of oat glucan, cellulose, hydroxymethylcellulose, hydroxyethylcellulose and carboxymethylcellulose. 6. The dental tool according to claim 1, wherein said tool is a dental floss. 7. The dental tool according to claim 6, wherein said substrate is a polymer, wherein said polymer is coated or impregnated with said composition. 8. The dental tool according to claim 6, wherein said dental floss has a denier of 100 to 1350. 9. (canceled) 10. The dental tool according to claim 1, wherein said complex carbohydrate has a solubility in water of at least 10 mM in water at 23° C. 11. A method of preventing and/or inhibiting periodontal disease comprising mechanically and chemically disrupting a biofilm with a dental tool comprising a complex carbohydrate composition,
wherein said dental tool is selected from the group consisting of a floss, a tape, a floss pick, an interdental brush, and a pick, and wherein said complex carbohydrate is a β-glucan having a molecular weight of said β-glucan is 360 to 20,000. 12. The method according to claim 11, wherein said biofilm is located within an oral cavity of a mammal. 13. The method according to claim 12, wherein said mammal is a selected from the group consisting of human, canines, equine, bovine and felines. 14. (canceled) 15. (canceled) 16. (canceled) 17. The method according to claim 11, wherein said β-glucan is at least one selected from the group consisting of oat glucan, cellulose, hydroxymethylcellulose, hydroxyethylcellulose and carboxymethylcellulose. 18. The method according to claim 11, wherein said complex carbohydrate has a solubility in water of at least 10 mM in water at 23° C. 19. (canceled) 20. (canceled) 21. (canceled) 22. (canceled) 23. The dental tool according to claim 1, wherein said complex carbohydrate bonds to a glycosyltransferase expressed on a cell surface of S. mutans. 24. The dental tool according to claim 1, wherein said β-glucan is an oat glucan. 25. The dental tool according to claim 1, wherein said β-glucan is cellulose. 26. The dental tool according to claim 1, wherein said β-glucan is hydroxymethylcellulose. 27. The dental tool according to claim 1, wherein said β-glucan is hydroxyethylcellulose. 28. The dental tool according to claim 1, wherein said β-glucan is carboxymethylcellulose. 29. The method according to claim 11, wherein said β-glucan is an oat glucan. 30. The method according to claim 11, wherein said β-glucan is cellulose. 31. The method according to claim 11, wherein said β-glucan is hydroxymethylcellulose. 32. The method according to claim 11, wherein said β-glucan is hydroxyethylcellulose. 33. The method according to claim 11, wherein said β-glucan is carboxymethylcellulose. | 1,600 |
699 | 15,316,914 | 1,611 | Provided are ion complexes comprising treprostinil and an ion-exchange resin, pharmaceutical formulations based on such complexes, and methods of treating diseases and conditions using the ion complexes and pharmaceutical formulations. | 1. A composition comprising a) treprostinil or its derivative and b) an anion ion-exchange resin. 2. The composition of claim 1, wherein the treprostinil or its derivative form an ion complex with the anion exchange resin. 3. The composition of claim 1, wherein treprostinil is a free acid. 4. The composition of claim 1, wherein treprostinil is a pharmaceutically acceptable salt of treprostinil. 5. The composition of claim 1, wherein the ion-exchange resin comprises a bile acid sequestrant. 6. The composition of claim 5, wherein the bile acid sequestrant is selected from the group consisting of cholestyramine, colosevelam, and colestipol. 7. The composition of claim 6, wherein the ion-exchange resin is a cholestyramine resin. 8. The composition of claim 7, wherein a weight-to-weight ratio between treprostinil and the cholestyramine resin is from 1:2 to 2:1. 9. The composition of claim 7, prepared using an aqueous dispersion of treprostinil and the cholestyramine resin, wherein the dispersion has a concentration of treprostinil from 0.1 mg/ml to 100 mg/ml. 10. A pharmaceutical formulation comprising i) an ion complex formed between treprostinil or its derivative and an anion ion-exchange resin and ii) a pharmaceutically acceptable carrier. 11. The formulation of claim 10, wherein the formulation is a suspension. 12. The formulation of claim 10, wherein the formulation is a solid dosage form selected from tablets and capsules. 13. The formulation of claim 10, wherein the formulation is a liquid dosage form for oral delivery. 14. The formulation of claim 10, further comprising a water insoluble membrane coating on the ion complex. 15. The formulation of claim 14, wherein said coating comprises a polymer. 16. The formulation of claim 14, wherein said coating comprises cellulose acetate. 17. The formulation of claim 10, wherein said formulation further comprises a release retardant configured to prolong or modify a release of the treprostinil from the ion complex. 18. The formulation of claim 10, wherein the formulation is a controlled-release formulation providing a controlled release of the treprostinil over a time period ranging from 1 hour to 36 hours. 19. A method of treating pulmonary hypertension comprising administering to a subject in need thereof a therapeutically effective amount of the formulation of claim 10. 20. A method of preparing a treprostinil formulation comprising admixing an ion-exchange resin and a solution comprising treprostinil or its derivative to form a suspension comprising an ion complex of treprostinil or its derivative and the ion-exchange resin. | Provided are ion complexes comprising treprostinil and an ion-exchange resin, pharmaceutical formulations based on such complexes, and methods of treating diseases and conditions using the ion complexes and pharmaceutical formulations.1. A composition comprising a) treprostinil or its derivative and b) an anion ion-exchange resin. 2. The composition of claim 1, wherein the treprostinil or its derivative form an ion complex with the anion exchange resin. 3. The composition of claim 1, wherein treprostinil is a free acid. 4. The composition of claim 1, wherein treprostinil is a pharmaceutically acceptable salt of treprostinil. 5. The composition of claim 1, wherein the ion-exchange resin comprises a bile acid sequestrant. 6. The composition of claim 5, wherein the bile acid sequestrant is selected from the group consisting of cholestyramine, colosevelam, and colestipol. 7. The composition of claim 6, wherein the ion-exchange resin is a cholestyramine resin. 8. The composition of claim 7, wherein a weight-to-weight ratio between treprostinil and the cholestyramine resin is from 1:2 to 2:1. 9. The composition of claim 7, prepared using an aqueous dispersion of treprostinil and the cholestyramine resin, wherein the dispersion has a concentration of treprostinil from 0.1 mg/ml to 100 mg/ml. 10. A pharmaceutical formulation comprising i) an ion complex formed between treprostinil or its derivative and an anion ion-exchange resin and ii) a pharmaceutically acceptable carrier. 11. The formulation of claim 10, wherein the formulation is a suspension. 12. The formulation of claim 10, wherein the formulation is a solid dosage form selected from tablets and capsules. 13. The formulation of claim 10, wherein the formulation is a liquid dosage form for oral delivery. 14. The formulation of claim 10, further comprising a water insoluble membrane coating on the ion complex. 15. The formulation of claim 14, wherein said coating comprises a polymer. 16. The formulation of claim 14, wherein said coating comprises cellulose acetate. 17. The formulation of claim 10, wherein said formulation further comprises a release retardant configured to prolong or modify a release of the treprostinil from the ion complex. 18. The formulation of claim 10, wherein the formulation is a controlled-release formulation providing a controlled release of the treprostinil over a time period ranging from 1 hour to 36 hours. 19. A method of treating pulmonary hypertension comprising administering to a subject in need thereof a therapeutically effective amount of the formulation of claim 10. 20. A method of preparing a treprostinil formulation comprising admixing an ion-exchange resin and a solution comprising treprostinil or its derivative to form a suspension comprising an ion complex of treprostinil or its derivative and the ion-exchange resin. | 1,600 |
Subsets and Splits
No community queries yet
The top public SQL queries from the community will appear here once available.