Unnamed: 0
int64
0
350k
level_0
int64
0
351k
ApplicationNumber
int64
9.75M
96.1M
ArtUnit
int64
1.6k
3.99k
Abstract
stringlengths
1
8.37k
Claims
stringlengths
3
292k
abstract-claims
stringlengths
68
293k
TechCenter
int64
1.6k
3.9k
200
200
14,246,100
1,631
A method of estimating a likelihood of developing a disease is disclosed. The method comprises obtaining a set of gene sequences corresponding to the disease and a DNA sequence of a subject. The method further comprises, for each gene sequence of the set, searching over the DNA sequence for reoccurrences of the gene sequence, and calculating an average reoccurrence distance between adjacent reoccurrences of the gene sequence. The method further comprises estimating the likelihood of the subject to develop the disease, based on the calculated distances.
1. A method of estimating a likelihood of developing a disease, the method comprising performing the following operations on a data processor: obtaining a set of gene sequences corresponding to the disease; obtaining a DNA sequence of a subject; for each gene sequence of said set, searching over said DNA sequence for reoccurrences of said gene sequence, and calculating an average reoccurrence distance between adjacent reoccurrences of said gene sequence; and estimating the likelihood of said subject to develop the disease, based on said calculated distances. 2. The method of claim 1, wherein said likelihood is estimated based on a set average distance calculated over said set. 3. The method of claim 2, wherein said likelihood equals a reciprocal of said set average. 4. The method of claim 1, further comprising randomly selecting a starting position over said DNA sequence, wherein said searching is initiated at said selected starting position. 5. The method of claim 1, further comprising randomly selecting a plurality of starting positions over said DNA sequence, wherein said searching is initiated a respective plurality of times, each time at a different selected starting position. 6. The method of claim 5, wherein said searching is terminated when a predetermined number of reoccurrences is found. 7. The method of claim 5, wherein said predetermined number of reoccurrences is 1. 8. The method of claim 1, wherein said gene sequence is selected from the group consisting of an oncogene and a tumor suppressor gene. 9. The method of claim 1, wherein said disease is cancer. 10. A system for estimating a likelihood of developing a disease, the system comprising a data processor configured for: obtaining from a database a set of gene sequences corresponding to the disease; obtaining a DNA sequence of a subject; for each gene sequence of said set, searching over said DNA sequence for reoccurrences of said gene sequence, and calculating an average reoccurrence distance between adjacent reoccurrences of said gene sequence; and estimating the likelihood of said subject to develop the disease, based on said calculated distances. 11. A computer software product, comprising a non-volatile computer-readable medium in which program instructions are stored, which instructions, when read by a data processor, cause the data processor: to receive a DNA sequence of a subject, and a set of gene sequences corresponding to a disease; to search over said DNA sequence for reoccurrences of a gene sequence for each gene sequence of said set, and to calculate an average reoccurrence distance between adjacent reoccurrences of said gene sequence; and to estimate the likelihood of said subject to develop the disease, based on said calculated distances. 12. The product of claim 11, wherein said likelihood is estimated based on a set average distance calculated over said set. 13. The product of claim 12, wherein said likelihood equals a reciprocal of said set average. 14. The product of claim 11, wherein said instructions cause the data processor to randomly select a starting position over said DNA sequence, wherein said searching is initiated at said selected starting position. 15. The product of claim 11, wherein said instructions cause the data processor to randomly select a plurality of starting positions over said DNA sequence, wherein said searching is initiated a respective plurality of times, each time at a different selected starting position. 16. The product of claim 15, wherein said searching is terminated when a predetermined number of reoccurrences is found. 17. The product of claim 16, wherein said predetermined number of reoccurrences is 1. 18. The product of claim 11, wherein said gene sequence is selected from the group consisting of an oncogene and a tumor suppressor gene. 19. The product of claim 11, wherein said disease is cancer. 20. A method of constructing a database of disease related genes, the method comprising performing the following operations on a data processor: obtaining a DNA sequence of a subject identified as having a disease, and a set of gene sequences associated with said DNA sequence; for each of at least a few gene sequences in said set, calculating an average reoccurrence distance between adjacent occurrences of said gene in said DNA; for at least one subset of gene sequences, determining a correlation between said subset and said disease, based, at least in part, on average reoccurrence distances of genes in said subset. 21. The method of claim 20, wherein said correlation is determined based on a set average distance calculated over said subset. 22. The method of claim 21, wherein said correlation equals a reciprocal of said set average. 23. The method of claim 20, further comprising randomly selecting a starting position over said DNA sequence, wherein said searching is initiated at said selected starting position. 24. The method of claim 20, further comprising randomly selecting a plurality of starting positions over said DNA sequence, wherein said reoccurrences are searched a respective plurality of times, each time at a different selected starting position. 25. The method of claim 24, wherein said searching is terminated when a predetermined number of reoccurrences is found. 26. The method of claim 24, wherein said predetermined number of reoccurrences is 1. 27. The method of claim 20, wherein said gene sequence is selected from the group consisting of an oncogene and a tumor suppressor gene. 28. The method of claim 20, wherein said disease is cancer. 29. A computer software product, comprising a non-volatile computer-readable medium in which program instructions are stored, which instructions, when read by a data processor, cause the data processor: to obtain a DNA sequence of a subject identified as having a disease, and a set of gene sequences associated with said DNA sequence; to calculate, for each of at least a few gene sequences in said set, an average reoccurrence distance between adjacent occurrences of said gene in said DNA; to determine, for at least one subset of gene sequences, a correlation between said subset and said disease, based, at least in part, on average reoccurrence distances of genes in said subset.
A method of estimating a likelihood of developing a disease is disclosed. The method comprises obtaining a set of gene sequences corresponding to the disease and a DNA sequence of a subject. The method further comprises, for each gene sequence of the set, searching over the DNA sequence for reoccurrences of the gene sequence, and calculating an average reoccurrence distance between adjacent reoccurrences of the gene sequence. The method further comprises estimating the likelihood of the subject to develop the disease, based on the calculated distances.1. A method of estimating a likelihood of developing a disease, the method comprising performing the following operations on a data processor: obtaining a set of gene sequences corresponding to the disease; obtaining a DNA sequence of a subject; for each gene sequence of said set, searching over said DNA sequence for reoccurrences of said gene sequence, and calculating an average reoccurrence distance between adjacent reoccurrences of said gene sequence; and estimating the likelihood of said subject to develop the disease, based on said calculated distances. 2. The method of claim 1, wherein said likelihood is estimated based on a set average distance calculated over said set. 3. The method of claim 2, wherein said likelihood equals a reciprocal of said set average. 4. The method of claim 1, further comprising randomly selecting a starting position over said DNA sequence, wherein said searching is initiated at said selected starting position. 5. The method of claim 1, further comprising randomly selecting a plurality of starting positions over said DNA sequence, wherein said searching is initiated a respective plurality of times, each time at a different selected starting position. 6. The method of claim 5, wherein said searching is terminated when a predetermined number of reoccurrences is found. 7. The method of claim 5, wherein said predetermined number of reoccurrences is 1. 8. The method of claim 1, wherein said gene sequence is selected from the group consisting of an oncogene and a tumor suppressor gene. 9. The method of claim 1, wherein said disease is cancer. 10. A system for estimating a likelihood of developing a disease, the system comprising a data processor configured for: obtaining from a database a set of gene sequences corresponding to the disease; obtaining a DNA sequence of a subject; for each gene sequence of said set, searching over said DNA sequence for reoccurrences of said gene sequence, and calculating an average reoccurrence distance between adjacent reoccurrences of said gene sequence; and estimating the likelihood of said subject to develop the disease, based on said calculated distances. 11. A computer software product, comprising a non-volatile computer-readable medium in which program instructions are stored, which instructions, when read by a data processor, cause the data processor: to receive a DNA sequence of a subject, and a set of gene sequences corresponding to a disease; to search over said DNA sequence for reoccurrences of a gene sequence for each gene sequence of said set, and to calculate an average reoccurrence distance between adjacent reoccurrences of said gene sequence; and to estimate the likelihood of said subject to develop the disease, based on said calculated distances. 12. The product of claim 11, wherein said likelihood is estimated based on a set average distance calculated over said set. 13. The product of claim 12, wherein said likelihood equals a reciprocal of said set average. 14. The product of claim 11, wherein said instructions cause the data processor to randomly select a starting position over said DNA sequence, wherein said searching is initiated at said selected starting position. 15. The product of claim 11, wherein said instructions cause the data processor to randomly select a plurality of starting positions over said DNA sequence, wherein said searching is initiated a respective plurality of times, each time at a different selected starting position. 16. The product of claim 15, wherein said searching is terminated when a predetermined number of reoccurrences is found. 17. The product of claim 16, wherein said predetermined number of reoccurrences is 1. 18. The product of claim 11, wherein said gene sequence is selected from the group consisting of an oncogene and a tumor suppressor gene. 19. The product of claim 11, wherein said disease is cancer. 20. A method of constructing a database of disease related genes, the method comprising performing the following operations on a data processor: obtaining a DNA sequence of a subject identified as having a disease, and a set of gene sequences associated with said DNA sequence; for each of at least a few gene sequences in said set, calculating an average reoccurrence distance between adjacent occurrences of said gene in said DNA; for at least one subset of gene sequences, determining a correlation between said subset and said disease, based, at least in part, on average reoccurrence distances of genes in said subset. 21. The method of claim 20, wherein said correlation is determined based on a set average distance calculated over said subset. 22. The method of claim 21, wherein said correlation equals a reciprocal of said set average. 23. The method of claim 20, further comprising randomly selecting a starting position over said DNA sequence, wherein said searching is initiated at said selected starting position. 24. The method of claim 20, further comprising randomly selecting a plurality of starting positions over said DNA sequence, wherein said reoccurrences are searched a respective plurality of times, each time at a different selected starting position. 25. The method of claim 24, wherein said searching is terminated when a predetermined number of reoccurrences is found. 26. The method of claim 24, wherein said predetermined number of reoccurrences is 1. 27. The method of claim 20, wherein said gene sequence is selected from the group consisting of an oncogene and a tumor suppressor gene. 28. The method of claim 20, wherein said disease is cancer. 29. A computer software product, comprising a non-volatile computer-readable medium in which program instructions are stored, which instructions, when read by a data processor, cause the data processor: to obtain a DNA sequence of a subject identified as having a disease, and a set of gene sequences associated with said DNA sequence; to calculate, for each of at least a few gene sequences in said set, an average reoccurrence distance between adjacent occurrences of said gene in said DNA; to determine, for at least one subset of gene sequences, a correlation between said subset and said disease, based, at least in part, on average reoccurrence distances of genes in said subset.
1,600
201
201
15,270,327
1,611
The present disclosure relates to an oral, extended release pill containing a drug which is homogenously spread throughout a matrix. The pill can be prepared using a hot melt extrusion process and a forming unit. The formed pill meets regulatory guidelines for extended release formulations and can be abuse deterrent to parenteral administration due at least to particle size, viscosity, or purity limitations.
1. An oral, extended release pill comprising a drug and (i) 30 wt % to 50 wt % of polyethylene oxide having an average molecular weight between 50K Daltons and 150K Daltons; (ii) 8 wt % to 60 wt % of a controlled release agent; (iii) 0.2 wt % to 20 wt % polyethylene glycol; wherein the drug has an extended release profile, and wherein the pill is a formed, uniform extrudate having a uniform blend of the drug, matrix agent, controlled release agent and plasticizer, and is directly formed from an extrusion process. 2. The oral, extended release pill of claim 1, wherein the controlled release agent is selected from the group consisting of polyvinyl acetate, polyvinylpyrrolidone, ethylcellulose, hydroxypropyl methylcellulose, hydroxypropylcellulose, hydroxymethylcellulose, poly(meth)acrylic acid and derivatives, or combinations thereof. 3. The oral, extended release pill of claim 2, wherein the formulation contains 15 wt % to 45 wt % hydroxypropyl methylcellulose as the controlled release agent. 4. The oral, extended release pill of claim 2, wherein the formulation contains 29 wt % to 60 wt % of a combination of polyvinyl acetate and polyvinylpyrrolidone as the controlled release agent. 5. The oral, extended release pill of claim 1, wherein the polyethylene glycol has an average molecular weight of less than 10K Daltons. 6. The oral, extended release pill of claim 1, wherein the pill has at least 50 wt % of particles with a particle size greater than 0.5 mm following physical or mechanical manipulation of the pill. 7. The oral, extended release pill of claim 1, wherein the drug is present at 1 to 50 wt % of the pill. 8. The oral, extended release pill of claim 1, wherein the pill further comprises at least one preservative or antioxidant selected from the group consisting of silica, sodium laurel sulfate, citric acid, butylated hydroxytoluene (BHT), ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), erythorbic acid, hypophosphorous acid, lactobionic acid, monothioglycerol, potassium metabisulfite, propyl gallate, racemethionine, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, sodium sulfite, sodium thiosulfate, stannous chloride, sulfur dioxide and tocopherols. 9. The oral, extended release pill of claim 1, wherein between 20% and 40% of the drug is released from the pill after 1 hour of exposure to simulated gastric fluid, wherein between 35% and 55% of the drug is released from the pill after 2 hours of exposure to simulated gastric fluid, wherein between 55% and 75% of the drug is released from the pill after 4 hours of exposure to simulated gastric fluid, wherein between 70% and 90% of the drug is released from the pill after 6 hours of exposure to simulated gastric fluid, and wherein not less than 80% of the drug is released from the pill after 8 hours of exposure to simulated gastric fluid. 10. The oral, extended release pill of claim 1 directly formed from a hot melt extrusion process having a forming unit selected from the group consisting of a calendar, a rotary, and a chain forming machine.
The present disclosure relates to an oral, extended release pill containing a drug which is homogenously spread throughout a matrix. The pill can be prepared using a hot melt extrusion process and a forming unit. The formed pill meets regulatory guidelines for extended release formulations and can be abuse deterrent to parenteral administration due at least to particle size, viscosity, or purity limitations.1. An oral, extended release pill comprising a drug and (i) 30 wt % to 50 wt % of polyethylene oxide having an average molecular weight between 50K Daltons and 150K Daltons; (ii) 8 wt % to 60 wt % of a controlled release agent; (iii) 0.2 wt % to 20 wt % polyethylene glycol; wherein the drug has an extended release profile, and wherein the pill is a formed, uniform extrudate having a uniform blend of the drug, matrix agent, controlled release agent and plasticizer, and is directly formed from an extrusion process. 2. The oral, extended release pill of claim 1, wherein the controlled release agent is selected from the group consisting of polyvinyl acetate, polyvinylpyrrolidone, ethylcellulose, hydroxypropyl methylcellulose, hydroxypropylcellulose, hydroxymethylcellulose, poly(meth)acrylic acid and derivatives, or combinations thereof. 3. The oral, extended release pill of claim 2, wherein the formulation contains 15 wt % to 45 wt % hydroxypropyl methylcellulose as the controlled release agent. 4. The oral, extended release pill of claim 2, wherein the formulation contains 29 wt % to 60 wt % of a combination of polyvinyl acetate and polyvinylpyrrolidone as the controlled release agent. 5. The oral, extended release pill of claim 1, wherein the polyethylene glycol has an average molecular weight of less than 10K Daltons. 6. The oral, extended release pill of claim 1, wherein the pill has at least 50 wt % of particles with a particle size greater than 0.5 mm following physical or mechanical manipulation of the pill. 7. The oral, extended release pill of claim 1, wherein the drug is present at 1 to 50 wt % of the pill. 8. The oral, extended release pill of claim 1, wherein the pill further comprises at least one preservative or antioxidant selected from the group consisting of silica, sodium laurel sulfate, citric acid, butylated hydroxytoluene (BHT), ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), erythorbic acid, hypophosphorous acid, lactobionic acid, monothioglycerol, potassium metabisulfite, propyl gallate, racemethionine, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, sodium sulfite, sodium thiosulfate, stannous chloride, sulfur dioxide and tocopherols. 9. The oral, extended release pill of claim 1, wherein between 20% and 40% of the drug is released from the pill after 1 hour of exposure to simulated gastric fluid, wherein between 35% and 55% of the drug is released from the pill after 2 hours of exposure to simulated gastric fluid, wherein between 55% and 75% of the drug is released from the pill after 4 hours of exposure to simulated gastric fluid, wherein between 70% and 90% of the drug is released from the pill after 6 hours of exposure to simulated gastric fluid, and wherein not less than 80% of the drug is released from the pill after 8 hours of exposure to simulated gastric fluid. 10. The oral, extended release pill of claim 1 directly formed from a hot melt extrusion process having a forming unit selected from the group consisting of a calendar, a rotary, and a chain forming machine.
1,600
202
202
14,474,267
1,636
Disclosed are embodiments of methods, apparatus, systems, compositions, and articles of manufacture relating to identifying the source of bioparticles, such as bioparticles shed by an organism. In embodiments, a method may include collecting a sample of bioparticles from the environment, selecting from that sample the bioparticles most informative for identifying their source, and gathering data from those bioparticles to form bioparticle signatures; the bioparticle signatures may be processed into a multi-dimensional vector which may be compared to a multi-dimensional vector derived from a standard using a pattern recognition strategy. In embodiments, an apparatus may include a particle collection device to collect a sample, a transfer device to select bioparticles, and a detector that restricts the movement of the bioparticles; the restricted movement may be used to produce a bioparticle signature.
1. A method for identifying a biological source, the method comprising: collecting from the environment a plurality of bioparticles shed by a biological source organism; selecting one or more information-rich bioparticles from the plurality of bioparticles; restricting the movement of the one or more information-rich bioparticles; detecting a plurality of bioparticle signatures from the one or more information-rich bioparticles; processing the plurality of said bioparticle signatures into a multi-dimensional vector by combining bioparticle signatures; and identifying the biological source based upon comparison of said multi-dimensional vector to a standard multi-dimensional vector using a pattern recognition strategy. 2. The method of claim 1 wherein the biological source comprises a mammal. 3. The method of claim 1 wherein the biological source comprises one or more human beings. 4. The method of claim 1 wherein the plurality of bioparticles collected from the environment comprise an aerosol. 5. The method of claim 1 wherein the one or more information-rich bioparticles comprise material shed from the stratum corneum, hair or bacterial flora. 6. The method of claim 1 wherein detecting a plurality of bioparticle signatures from the one or more information-rich bioparticles comprises detecting a bioparticle signature from an intact bioparticle shed from stratum corneum and/or hair. 7. The method of claim 1 wherein restricting the movement of the information-rich bioparticle comprises restricting the movement of the information-rich bioparticle by dielectrophoresis. 8. The method of claim 1 wherein the standard multi-dimensional vector comprises a multi-dimensional vector derived from a bioparticle shed by a known biological source. 9. The method of claim 1 wherein the standard multi-dimensional vector comprises a multi-dimensional vector derived from one or more bioparticles collected from a known geographical location and identifying the biological source comprises identifying whether the information-rich bioparticles were shed by one or more organisms present at the known geographical location. 10. The method of claim 1, wherein, when the movement of the one or more bioparticles is restricted, a bioparticle signature is detected by surface plasmon resonance. 11. The method of claim 1 wherein identifying a biological source comprises identifying a geographic location or region associated with the biological source. 12. The method of claim 1 wherein the movement of the information-rich bioparticle is restricted by binding to a panel of compounds. 13. An apparatus for identifying a biological source organism from bioparticles shed by the organism into the environment, the apparatus comprising: a particle collector configured to collect a sample containing a plurality of bioparticles; a selection and concentration system coupled to the particle collector, configured to select one or more information-rich bioparticles from the plurality of bioparticles; a bioparticle signature detector coupled to the selection and concentration system, the bioparticle signature detector comprising means for restricting the movement of the information-rich bioparticles and means for detecting a plurality of bioparticle signatures therefrom; and a pattern recognition system linked to the bioparticle signature detector and configured to derive a multi-dimensional vector from the bioparticle signatures by combining the bioparticle signatures and to compare said multi-dimensional vector with a standard multi-dimensional vector in order to identify, from the one or more information-rich bioparticles, the biological source from which the bioparticles were shed. 14. The apparatus of claim 13 wherein the selection and concentration system comprises a portal configured to divert an information-rich bioparticle from a directional flow of a solution containing the plurality of bioparticles. 15. The apparatus of claim 14 wherein the portal is configured to divert the information-rich bioparticle with an electromagnetic field. 16. The apparatus of claim 13 wherein the bioparticle signature detector comprises a dielectrophoresis chamber for restricting movement of the one or more information-rich bioparticles. 17. The apparatus of claim 16 wherein the dielectrophoresis chamber comprises an element placed within an electromagnetic field. 18. The apparatus of claim 13 wherein the detector comprises a panel of compounds. 19. The apparatus of claim 13 wherein the means for detecting a plurality of bioparticle signatures comprises a surface plasmon resonance detector. 20. The apparatus of claim 19, wherein the surface plasmon resonance detector comprises a label-free surface plasmon resonance detector or a capillary electrophoresis surface plasmon resonance detector.
Disclosed are embodiments of methods, apparatus, systems, compositions, and articles of manufacture relating to identifying the source of bioparticles, such as bioparticles shed by an organism. In embodiments, a method may include collecting a sample of bioparticles from the environment, selecting from that sample the bioparticles most informative for identifying their source, and gathering data from those bioparticles to form bioparticle signatures; the bioparticle signatures may be processed into a multi-dimensional vector which may be compared to a multi-dimensional vector derived from a standard using a pattern recognition strategy. In embodiments, an apparatus may include a particle collection device to collect a sample, a transfer device to select bioparticles, and a detector that restricts the movement of the bioparticles; the restricted movement may be used to produce a bioparticle signature.1. A method for identifying a biological source, the method comprising: collecting from the environment a plurality of bioparticles shed by a biological source organism; selecting one or more information-rich bioparticles from the plurality of bioparticles; restricting the movement of the one or more information-rich bioparticles; detecting a plurality of bioparticle signatures from the one or more information-rich bioparticles; processing the plurality of said bioparticle signatures into a multi-dimensional vector by combining bioparticle signatures; and identifying the biological source based upon comparison of said multi-dimensional vector to a standard multi-dimensional vector using a pattern recognition strategy. 2. The method of claim 1 wherein the biological source comprises a mammal. 3. The method of claim 1 wherein the biological source comprises one or more human beings. 4. The method of claim 1 wherein the plurality of bioparticles collected from the environment comprise an aerosol. 5. The method of claim 1 wherein the one or more information-rich bioparticles comprise material shed from the stratum corneum, hair or bacterial flora. 6. The method of claim 1 wherein detecting a plurality of bioparticle signatures from the one or more information-rich bioparticles comprises detecting a bioparticle signature from an intact bioparticle shed from stratum corneum and/or hair. 7. The method of claim 1 wherein restricting the movement of the information-rich bioparticle comprises restricting the movement of the information-rich bioparticle by dielectrophoresis. 8. The method of claim 1 wherein the standard multi-dimensional vector comprises a multi-dimensional vector derived from a bioparticle shed by a known biological source. 9. The method of claim 1 wherein the standard multi-dimensional vector comprises a multi-dimensional vector derived from one or more bioparticles collected from a known geographical location and identifying the biological source comprises identifying whether the information-rich bioparticles were shed by one or more organisms present at the known geographical location. 10. The method of claim 1, wherein, when the movement of the one or more bioparticles is restricted, a bioparticle signature is detected by surface plasmon resonance. 11. The method of claim 1 wherein identifying a biological source comprises identifying a geographic location or region associated with the biological source. 12. The method of claim 1 wherein the movement of the information-rich bioparticle is restricted by binding to a panel of compounds. 13. An apparatus for identifying a biological source organism from bioparticles shed by the organism into the environment, the apparatus comprising: a particle collector configured to collect a sample containing a plurality of bioparticles; a selection and concentration system coupled to the particle collector, configured to select one or more information-rich bioparticles from the plurality of bioparticles; a bioparticle signature detector coupled to the selection and concentration system, the bioparticle signature detector comprising means for restricting the movement of the information-rich bioparticles and means for detecting a plurality of bioparticle signatures therefrom; and a pattern recognition system linked to the bioparticle signature detector and configured to derive a multi-dimensional vector from the bioparticle signatures by combining the bioparticle signatures and to compare said multi-dimensional vector with a standard multi-dimensional vector in order to identify, from the one or more information-rich bioparticles, the biological source from which the bioparticles were shed. 14. The apparatus of claim 13 wherein the selection and concentration system comprises a portal configured to divert an information-rich bioparticle from a directional flow of a solution containing the plurality of bioparticles. 15. The apparatus of claim 14 wherein the portal is configured to divert the information-rich bioparticle with an electromagnetic field. 16. The apparatus of claim 13 wherein the bioparticle signature detector comprises a dielectrophoresis chamber for restricting movement of the one or more information-rich bioparticles. 17. The apparatus of claim 16 wherein the dielectrophoresis chamber comprises an element placed within an electromagnetic field. 18. The apparatus of claim 13 wherein the detector comprises a panel of compounds. 19. The apparatus of claim 13 wherein the means for detecting a plurality of bioparticle signatures comprises a surface plasmon resonance detector. 20. The apparatus of claim 19, wherein the surface plasmon resonance detector comprises a label-free surface plasmon resonance detector or a capillary electrophoresis surface plasmon resonance detector.
1,600
203
203
14,499,191
1,627
The invention relates generally to ephedrine or pseudoephedrine compositions containing biocompatible organoleptic (food flavoring) excipients that would prevent the illicit manufacture of methamphetamine from ephedrine or pseudoephedrine.
1. A pharmaceutically acceptable composition, comprising (a) ephedrine or pseudoephedrine, and (b) a suitable amount of an organoleptic agent, wherein the organoleptic agent is effective in inhibiting the conversion of the ephedrine or pseudoephedrine to methamphetamine. 2. The composition of claim 1 wherein (a) the ephedrine or pseudoephedrine is pseudoephedrine, and (b) the organoleptic agent is selected from the group consisting of (i) a compound of Formula I wherein R1 to R4 are independently selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, C1-C10 alkoxy, C1-C10 alkoxyalkyl, C1-C10 alkylthio, and C1-C10 alkoxycarbonyl; and (ii) a compound of Formula II wherein R1 and R2 are independently selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, C1-C10 alkoxy, C1-C10 alkoxyalkyl, C1-C10 alkylthio, and C1-C10 alkoxycarbonyl; X and Y are independently selected from the group consisting of —(CH2)m—, —(CHR5)—, —O—, —N—, —NR6—, or —S—; Z is —(CHR7)—, —C(R8)═, or ═C(R9)—C(R10)═; subscript ‘m’ varies from 0 to 4; R5 is selected from the group consisting of C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, and C1-C10 alkoxycarbonyl; R6 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, and C1-C10 alkoxycarbonyl; R7 to R10 are independently selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, C1-C10 alkoxy, and C1-C10 alkoxycarbonyl. 3. The composition of claim 2 wherein the organoleptic agent is a compound of Formula I. 4. The composition of claim 3 wherein the mole ratio of the organoleptic agent to the pseudoephedrine is about 1:1. 5. The composition of claim 3 wherein the mole ratio of the organoleptic agent to the pseudoephedrine is about 2.5:1. 6. The composition of claim 3 wherein the mole ratio of the organoleptic agent to the pseudoephedrine is about 1:1 to about 2.5:1. 7. The composition of claim 3 wherein the pseudoephedrine and the organoleptic agent are present as a mixture. 8. The composition of claim 7 wherein the pseudoephedrine and the organoleptic agent were formed into a slurry or a solution. 9. The composition of claim 7 wherein the composition is a solid dosage form, which is covered with a coating agent. 10. The composition of claim 3 wherein in Formula I, R1 is acetyl, methoxy, or ethoxy, and R2 to R4 are independently hydrogen or methyl. 11. The composition of claim 3 wherein the compound of Formula I is selected from the group consisting of acetylpyrazine, 2-ethoxy-5-methylpyrazine, 2-ethoxy-6-methylpyrazine, 2-isobutyl-3-methyoxypyrazine, 2-methoxy-3-methylpyrazine, and 2,3,5-trimethylpyrazine. 12. The composition of claim 3 wherein, under the Li—NH3 conditions of Examples 1 and 3, not more than about 8 percent methamphetamine will be produced. 13. The composition of claim 12 wherein, under the Li—NH3 conditions of Examples 1 and 3, not more than about 4 percent methamphetamine will be produced. 14. The composition of claim 3 wherein, under the P—HI conditions of Examples 2 and 3, not more than about 27 percent methamphetamine will be produced. 15. The composition of claim 14 wherein, under the P—HI conditions of Examples 2 and 3, not more than about 13 percent methamphetamine will be produced. 16. The composition of claim 15 wherein, under the P—HI conditions of Examples 2 and 3, not more than about 6 percent methamphetamine will be produced. 17. The composition of claim 2 wherein the organoleptic agent is a compound of Formula II. 18. The composition of claim 17 wherein in Formula II, R1 and R2 are independently hydrogen, C1-C10 alkyl, C1-C10 acyl, or C1-C10 alkoxy; X is —(CH2)m— or —N—; Y is —(CH2)m—, —O—, —NR6—, or —S—; Z is —(CHR7)—, —C(R8)═, or ═C(R9)—C(R10)═; R6 to R10 are independently 19. The composition of claim 17 wherein in Formula II, R1 and R2 are independently hydrogen, C1-C10 alkyl, C1-C10 acyl, or C1-C10 alkoxy; X is —(CHR5)—; Y is —NR6—, —O—, or —S—; Z is —C(R8)═; R5 and R8 are independently hydrogen or C1-C10 alkyl. 20. A method of preventing the conversion of ephedrine or pseudoephedrine to methamphetamine, comprising formulating the pseudoephedrine or ephedrine with a suitable amount of an organoleptic agent, wherein the organoleptic agent is effective in chemically inhibiting the conversion of the ephedrine or pseudoephedrine to methamphetamine.
The invention relates generally to ephedrine or pseudoephedrine compositions containing biocompatible organoleptic (food flavoring) excipients that would prevent the illicit manufacture of methamphetamine from ephedrine or pseudoephedrine.1. A pharmaceutically acceptable composition, comprising (a) ephedrine or pseudoephedrine, and (b) a suitable amount of an organoleptic agent, wherein the organoleptic agent is effective in inhibiting the conversion of the ephedrine or pseudoephedrine to methamphetamine. 2. The composition of claim 1 wherein (a) the ephedrine or pseudoephedrine is pseudoephedrine, and (b) the organoleptic agent is selected from the group consisting of (i) a compound of Formula I wherein R1 to R4 are independently selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, C1-C10 alkoxy, C1-C10 alkoxyalkyl, C1-C10 alkylthio, and C1-C10 alkoxycarbonyl; and (ii) a compound of Formula II wherein R1 and R2 are independently selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, C1-C10 alkoxy, C1-C10 alkoxyalkyl, C1-C10 alkylthio, and C1-C10 alkoxycarbonyl; X and Y are independently selected from the group consisting of —(CH2)m—, —(CHR5)—, —O—, —N—, —NR6—, or —S—; Z is —(CHR7)—, —C(R8)═, or ═C(R9)—C(R10)═; subscript ‘m’ varies from 0 to 4; R5 is selected from the group consisting of C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, and C1-C10 alkoxycarbonyl; R6 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, and C1-C10 alkoxycarbonyl; R7 to R10 are independently selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 acyl, C1-C10 hydroxyalkyl, C1-C10 alkoxy, and C1-C10 alkoxycarbonyl. 3. The composition of claim 2 wherein the organoleptic agent is a compound of Formula I. 4. The composition of claim 3 wherein the mole ratio of the organoleptic agent to the pseudoephedrine is about 1:1. 5. The composition of claim 3 wherein the mole ratio of the organoleptic agent to the pseudoephedrine is about 2.5:1. 6. The composition of claim 3 wherein the mole ratio of the organoleptic agent to the pseudoephedrine is about 1:1 to about 2.5:1. 7. The composition of claim 3 wherein the pseudoephedrine and the organoleptic agent are present as a mixture. 8. The composition of claim 7 wherein the pseudoephedrine and the organoleptic agent were formed into a slurry or a solution. 9. The composition of claim 7 wherein the composition is a solid dosage form, which is covered with a coating agent. 10. The composition of claim 3 wherein in Formula I, R1 is acetyl, methoxy, or ethoxy, and R2 to R4 are independently hydrogen or methyl. 11. The composition of claim 3 wherein the compound of Formula I is selected from the group consisting of acetylpyrazine, 2-ethoxy-5-methylpyrazine, 2-ethoxy-6-methylpyrazine, 2-isobutyl-3-methyoxypyrazine, 2-methoxy-3-methylpyrazine, and 2,3,5-trimethylpyrazine. 12. The composition of claim 3 wherein, under the Li—NH3 conditions of Examples 1 and 3, not more than about 8 percent methamphetamine will be produced. 13. The composition of claim 12 wherein, under the Li—NH3 conditions of Examples 1 and 3, not more than about 4 percent methamphetamine will be produced. 14. The composition of claim 3 wherein, under the P—HI conditions of Examples 2 and 3, not more than about 27 percent methamphetamine will be produced. 15. The composition of claim 14 wherein, under the P—HI conditions of Examples 2 and 3, not more than about 13 percent methamphetamine will be produced. 16. The composition of claim 15 wherein, under the P—HI conditions of Examples 2 and 3, not more than about 6 percent methamphetamine will be produced. 17. The composition of claim 2 wherein the organoleptic agent is a compound of Formula II. 18. The composition of claim 17 wherein in Formula II, R1 and R2 are independently hydrogen, C1-C10 alkyl, C1-C10 acyl, or C1-C10 alkoxy; X is —(CH2)m— or —N—; Y is —(CH2)m—, —O—, —NR6—, or —S—; Z is —(CHR7)—, —C(R8)═, or ═C(R9)—C(R10)═; R6 to R10 are independently 19. The composition of claim 17 wherein in Formula II, R1 and R2 are independently hydrogen, C1-C10 alkyl, C1-C10 acyl, or C1-C10 alkoxy; X is —(CHR5)—; Y is —NR6—, —O—, or —S—; Z is —C(R8)═; R5 and R8 are independently hydrogen or C1-C10 alkyl. 20. A method of preventing the conversion of ephedrine or pseudoephedrine to methamphetamine, comprising formulating the pseudoephedrine or ephedrine with a suitable amount of an organoleptic agent, wherein the organoleptic agent is effective in chemically inhibiting the conversion of the ephedrine or pseudoephedrine to methamphetamine.
1,600
204
204
15,289,566
1,616
Methods of improving the growth of a plant by applying a plant growth effective amount of a plant growth composition that includes a hydrated aluminum magnesium silicate and at least one dispersant selected from the group consisting of a sucrose ester, a lignosulfonate, an alkylpolyglycoside, a naphthalenesulfonic acid formaldehyde condensate and a phosphate ester to plant propagation material in the absence of insect pest pressure.
1. A method of improving the growth of a plant comprising applying a plant growth effective amount of a plant growth composition comprising a hydrated aluminum-magnesium silicate and at least one dispersant selected from the group consisting of a sucrose ester, a limosulfonate, an alkylpolyglycoside, a naphthalenesulfonic acid formaldehyde condensate and a phosphate ester to plant propagation material in the absence of insect pest pressure. 2. The method of claim 1, wherein the plant growth composition comprises: a) about 1% to about 20% of hydrated aluminum-magnesium silicate and b) about 0.2% to about 20% of at least one dispersant selected from the group consisting of a sucrose ester, a lignosulfonate, an alkylpolyglycoside, a naphthalenesulfonic acid formaldehyde condensate and a phosphate ester; where all % are % by weight based upon the total weight of all components in the composition. 3. The method of claim 1, wherein the plant propagation material is selected from the group consisting of seeds, spores, bulbs, cuttings, sets, rhizomes, tubers, meristem tissue, plant cells, and combinations thereof. 4. The method of claim 1, wherein the plant propagation material comprises at least one seed. 5. The method of claim 1, wherein the plant growth composition is applied at a rate ranging from 100 g/ha to 500g /ha. 6. The method of claim 5, wherein the plant growth composition is applied at a rate ranging from 200 g/ha to 300 g/ha. 7. The method of claim 1, wherein the plant is selected from the group consisting of corn, cotton, soybean, sunflower, wheat, barley, rye, oat, and oilseed rape. 8. The method of claim 7, wherein the plant is cam. 9. The method of claim 1, wherein the plant growth composition further comprises a liquid fertilizer, 10. The method of claim 1, wherein the plant growth composition further comprises at least one of an anti-freeze agent, an anti-foam agent and a biocide.
Methods of improving the growth of a plant by applying a plant growth effective amount of a plant growth composition that includes a hydrated aluminum magnesium silicate and at least one dispersant selected from the group consisting of a sucrose ester, a lignosulfonate, an alkylpolyglycoside, a naphthalenesulfonic acid formaldehyde condensate and a phosphate ester to plant propagation material in the absence of insect pest pressure.1. A method of improving the growth of a plant comprising applying a plant growth effective amount of a plant growth composition comprising a hydrated aluminum-magnesium silicate and at least one dispersant selected from the group consisting of a sucrose ester, a limosulfonate, an alkylpolyglycoside, a naphthalenesulfonic acid formaldehyde condensate and a phosphate ester to plant propagation material in the absence of insect pest pressure. 2. The method of claim 1, wherein the plant growth composition comprises: a) about 1% to about 20% of hydrated aluminum-magnesium silicate and b) about 0.2% to about 20% of at least one dispersant selected from the group consisting of a sucrose ester, a lignosulfonate, an alkylpolyglycoside, a naphthalenesulfonic acid formaldehyde condensate and a phosphate ester; where all % are % by weight based upon the total weight of all components in the composition. 3. The method of claim 1, wherein the plant propagation material is selected from the group consisting of seeds, spores, bulbs, cuttings, sets, rhizomes, tubers, meristem tissue, plant cells, and combinations thereof. 4. The method of claim 1, wherein the plant propagation material comprises at least one seed. 5. The method of claim 1, wherein the plant growth composition is applied at a rate ranging from 100 g/ha to 500g /ha. 6. The method of claim 5, wherein the plant growth composition is applied at a rate ranging from 200 g/ha to 300 g/ha. 7. The method of claim 1, wherein the plant is selected from the group consisting of corn, cotton, soybean, sunflower, wheat, barley, rye, oat, and oilseed rape. 8. The method of claim 7, wherein the plant is cam. 9. The method of claim 1, wherein the plant growth composition further comprises a liquid fertilizer, 10. The method of claim 1, wherein the plant growth composition further comprises at least one of an anti-freeze agent, an anti-foam agent and a biocide.
1,600
205
205
15,049,946
1,639
The hemitartrate salt of a compound represented by the following structural formula: (Formula I Hemitartrate), which may be used in pharmaceutical applications, are disclosed. Particular single crystalline forms of the Formula (I) Hemitartrate are characterized by a variety of properties and physical measurements. As well, methods of producing crystalline Formula (I) Hemitartrate, and using it to inhibit glucosylceramide synthase or lowering glycosphingolipid concentrations in subjects to treat a number of diseases, are also discussed. Pharmaceutical compositions are also described.
1-93. (canceled) 94. A method of treating a subject with Gaucher disease who has been assessed as being a poor, intermediate or extensive P450 metabolizer, the method comprising administering to the subject a compound represented by the following structural formula or a pharmaceutically acceptable salt thereof, wherein if the subject is a poor metabolizer then the compound is administered in an effective amount at a first dose, and wherein if the subject is an intermediate or extensive metabolizer then the compound is administered in an adjusted effective amount at a second dose, wherein said second dose is greater than said first dose. 95. The method of claim 94, wherein said P450 is CYP2D6 or CYP3A4. 96. The method of claim 94, wherein said subject has been assessed as being a poor, intermediate or extensive metabolizer through genotyping. 97. The method of claim 96, wherein said subject has been assessed as being a poor metabolizer as a result of carrying two mutant alleles of the CYP2D6 gene which result in complete loss of enzyme activity. 98. The method of claim 94, wherein said subject has been assessed as being a poor metabolizer as a result of low expression of said P450. 99. The method of claim 94, wherein said subject has been assessed as being a poor metabolizer as a result of being treated with a drug that is an inhibitor of said P450. 100. The method of claim 99, wherein said subject possesses one reduced activity allele and one null allele of CYP2D6, or wherein said subject possesses at least one and no more than two normal functional alleles of CYP2D6. 101. The method of claim 99, wherein said drug is paroxetine, fluoxetine or quinidine. 102. The method of claim 94, wherein the compound is administered in a daily dose of from 25 milligrams to 150 milligrams. 103. The method of claim 94, wherein the compound is administered in a daily dose of from 50 milligrams to 100 milligrams. 104. The method of claim 96, wherein said subject has been assessed as being an intermediate metabolizer as a result of possessing one reduced activity allele and one null allele of the CYP2D6 gene. 105. The method of claim 96, wherein said subject has been assessed as being an extensive metabolizer as a result of possessing at least one and no more than two normal functional alleles of the CYP2D6 gene. 106. The method of claim 94, wherein the compound is administered at said second dose, this being a dose of greater than 100 milligrams per day. 107. The method of claim 106, wherein the compound is administered in a daily dose of from 100 milligrams to 300 milligrams. 108. The method of claim 106, wherein the compound is administered in a daily dose of greater than 150 milligrams per day. 109. The method of claim 104, wherein the compound is administered in a daily dose from 150 milligrams to 200 milligrams. 110. The method of claim 94, wherein said subject is a human patient. 111. The method of claim 94, wherein said Gaucher disease is type 1 Gaucher disease. 112. The method of claim 94, wherein the compound is represented by the hemitartrate salt of said structural formula.
The hemitartrate salt of a compound represented by the following structural formula: (Formula I Hemitartrate), which may be used in pharmaceutical applications, are disclosed. Particular single crystalline forms of the Formula (I) Hemitartrate are characterized by a variety of properties and physical measurements. As well, methods of producing crystalline Formula (I) Hemitartrate, and using it to inhibit glucosylceramide synthase or lowering glycosphingolipid concentrations in subjects to treat a number of diseases, are also discussed. Pharmaceutical compositions are also described.1-93. (canceled) 94. A method of treating a subject with Gaucher disease who has been assessed as being a poor, intermediate or extensive P450 metabolizer, the method comprising administering to the subject a compound represented by the following structural formula or a pharmaceutically acceptable salt thereof, wherein if the subject is a poor metabolizer then the compound is administered in an effective amount at a first dose, and wherein if the subject is an intermediate or extensive metabolizer then the compound is administered in an adjusted effective amount at a second dose, wherein said second dose is greater than said first dose. 95. The method of claim 94, wherein said P450 is CYP2D6 or CYP3A4. 96. The method of claim 94, wherein said subject has been assessed as being a poor, intermediate or extensive metabolizer through genotyping. 97. The method of claim 96, wherein said subject has been assessed as being a poor metabolizer as a result of carrying two mutant alleles of the CYP2D6 gene which result in complete loss of enzyme activity. 98. The method of claim 94, wherein said subject has been assessed as being a poor metabolizer as a result of low expression of said P450. 99. The method of claim 94, wherein said subject has been assessed as being a poor metabolizer as a result of being treated with a drug that is an inhibitor of said P450. 100. The method of claim 99, wherein said subject possesses one reduced activity allele and one null allele of CYP2D6, or wherein said subject possesses at least one and no more than two normal functional alleles of CYP2D6. 101. The method of claim 99, wherein said drug is paroxetine, fluoxetine or quinidine. 102. The method of claim 94, wherein the compound is administered in a daily dose of from 25 milligrams to 150 milligrams. 103. The method of claim 94, wherein the compound is administered in a daily dose of from 50 milligrams to 100 milligrams. 104. The method of claim 96, wherein said subject has been assessed as being an intermediate metabolizer as a result of possessing one reduced activity allele and one null allele of the CYP2D6 gene. 105. The method of claim 96, wherein said subject has been assessed as being an extensive metabolizer as a result of possessing at least one and no more than two normal functional alleles of the CYP2D6 gene. 106. The method of claim 94, wherein the compound is administered at said second dose, this being a dose of greater than 100 milligrams per day. 107. The method of claim 106, wherein the compound is administered in a daily dose of from 100 milligrams to 300 milligrams. 108. The method of claim 106, wherein the compound is administered in a daily dose of greater than 150 milligrams per day. 109. The method of claim 104, wherein the compound is administered in a daily dose from 150 milligrams to 200 milligrams. 110. The method of claim 94, wherein said subject is a human patient. 111. The method of claim 94, wherein said Gaucher disease is type 1 Gaucher disease. 112. The method of claim 94, wherein the compound is represented by the hemitartrate salt of said structural formula.
1,600
206
206
15,337,410
1,617
Disclosed are methods and compositions for the treatment or prevention of intestinal polyps or prevention of cancer in a patient who has been identified as being at risk for developing intestinal polyps or intestinal cancer. The disclosed methods and compositions include rapamycin, a rapamycin analog, or another such inhibitor of the target of rapamycin (TOR).
1. A method for preventing intestinal polyps or intestinal cancer in a patient comprising administering an effective amount of a composition comprising rapamycin or an analog thereof to a patient who has been identified as being at risk for developing intestinal polyps or intestinal cancer. 2. The method of claim 1, wherein the rapamycin or analog thereof is encased in a coating comprising cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate co-polymer, or a polymethacrylate-based copolymer selected from the group consisting of methyl acrylate-methacrylic acid copolymer, and a methyl methacrylate-methacrylic acid copolymer. 3. The method of claim 2, wherein the coating comprises Poly(methacrylic acid-co-ethyl acrylate) in a 1:1 ratio, Poly(methacrylic acid-co-ethyl acrylate) in a 1:1 ratio, Poly(methacrylic acid-co-methyl methacrylate) in a 1:1 ratio, Poly(methacrylic acid-co-methyl methacrylate) in a 1:2 ratio, Poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) in a 7:3:1 ratio, Poly(ethyl acrylate-co-methyl methacrylate-co-trimethylammonioethyl methacrylate chloride) in a 1:2:0.2 ratio, Poly(ethyl acrylate-co-methyl methacrylate-co-trimethylammonioethyl methacrylate chloride) in a 1:2:0.1 ratio, or Poly(butyl methacrylate-co-(2-dimethylaminoethyl) methacrylate-co-methyl methacrylate) in a 1:2:1 ratio, a naturally-derived polymer, or a synthetic polymer, or any combination thereof, wherein the naturally-derived polymer is selected from the group consisting of alginates and their various derivatives, chitosans and their various derivatives, carrageenans and their various analogues, celluloses, gums, gelatins, pectins, gellans, polyethyleneglycols (PEGs) and polyethyleneoxides (PEOs), acrylic acid homo- and copolymers with acrylates and methacrylates, homopolymers of acrylates and methacrylates, polyvinyl alcohol (PVOH), and polyvinyl pyrrolidone (PVP). 4.-5. (canceled) 6. The method of claim 3, wherein the patient has been diagnosed with one or more of the following: inflammatory bowel disease; an intestinal polyp or an adenoma; having a mutation that is known to cause increased WNT signaling; or having Familial Adenomatous Polyposis (FAP); and/or wherein the patient has a family history of intestinal polyps or intestinal cancer. 7.-11. (canceled) 12. The method of claim 6, wherein the composition comprises rapamycin or an analog thereof at a concentration of 0.001 mg to 30 mg total per dose, and wherein the composition comprising rapamycin or an analog of rapamycin further comprises 0.001% to 60% by weight of rapamycin or an analog of rapamycin. 13. (canceled) 14. The method of claim 12, wherein the average blood level of rapamycin in the subject is greater than 0.5 ng per mL whole blood after administration of the composition. 15. The method of claim 14, wherein the composition is administered orally, enterically, colonically, anally, intravenously, or dermally with a patch. 16. The method of claim 15, wherein the rapamycin or analog of rapamycin is administered in two or more doses. 17. The method of claim 16, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 0.5 to 30 days. 18. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 0.5 to 1 day. 19. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 3 days. 20. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 5 days. 21. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 7 days. 22. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 15 days. 23. The method of claim 17, wherein the subject is further administered a composition comprising a second active agent. 24. The method of claim 23, wherein the second active agent is metformin, celocoxib, eflornithine, sulindac, ursodeoxycholic acid, an anti-inflammatory agent, an anti-autoimmune agent, or a cytotoxic or cytostatic anti-cancer agent. 25. The method of claim 24, wherein the composition comprising rapamycin or an analog of rapamycin is administered at the same time as the composition comprising the second active agent. 26. The method of claim 24, wherein the composition comprising rapamycin or an analog of rapamycin is administered before or after the composition comprising the second active agent is administered, and wherein the interval of time between administration of the composition comprising rapamycin or an analog of rapamycin and the composition comprising the second active agent is 1 to 30 days. 27.-28. (canceled) 29. The method of claim 6, wherein the composition comprising rapamycin or an analog of rapamycin prevents intestinal polyps or intestinal cancer, prevents the development of new adenomas or polyps, decreases the number or severity of the adenomatous polyps, induces a reduction in size or number of existing adenomas or polyps, prevents the conversion of adenomas or polyps into adenocarcinomas and cancer tissue, or prevents the adenomas or polyps from converting into malignant cancer that spread into other bodily tissues, organs and blood systems in a patient that has been diagnosed as having intestinal adenomas, intestinal polyps or Familial Adenomatous Polyposis (FAP). 30. The method of claim 29, wherein the composition comprising rapamycin or an analog of rapamycin is comprised in a food or food additive.
Disclosed are methods and compositions for the treatment or prevention of intestinal polyps or prevention of cancer in a patient who has been identified as being at risk for developing intestinal polyps or intestinal cancer. The disclosed methods and compositions include rapamycin, a rapamycin analog, or another such inhibitor of the target of rapamycin (TOR).1. A method for preventing intestinal polyps or intestinal cancer in a patient comprising administering an effective amount of a composition comprising rapamycin or an analog thereof to a patient who has been identified as being at risk for developing intestinal polyps or intestinal cancer. 2. The method of claim 1, wherein the rapamycin or analog thereof is encased in a coating comprising cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate co-polymer, or a polymethacrylate-based copolymer selected from the group consisting of methyl acrylate-methacrylic acid copolymer, and a methyl methacrylate-methacrylic acid copolymer. 3. The method of claim 2, wherein the coating comprises Poly(methacrylic acid-co-ethyl acrylate) in a 1:1 ratio, Poly(methacrylic acid-co-ethyl acrylate) in a 1:1 ratio, Poly(methacrylic acid-co-methyl methacrylate) in a 1:1 ratio, Poly(methacrylic acid-co-methyl methacrylate) in a 1:2 ratio, Poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) in a 7:3:1 ratio, Poly(ethyl acrylate-co-methyl methacrylate-co-trimethylammonioethyl methacrylate chloride) in a 1:2:0.2 ratio, Poly(ethyl acrylate-co-methyl methacrylate-co-trimethylammonioethyl methacrylate chloride) in a 1:2:0.1 ratio, or Poly(butyl methacrylate-co-(2-dimethylaminoethyl) methacrylate-co-methyl methacrylate) in a 1:2:1 ratio, a naturally-derived polymer, or a synthetic polymer, or any combination thereof, wherein the naturally-derived polymer is selected from the group consisting of alginates and their various derivatives, chitosans and their various derivatives, carrageenans and their various analogues, celluloses, gums, gelatins, pectins, gellans, polyethyleneglycols (PEGs) and polyethyleneoxides (PEOs), acrylic acid homo- and copolymers with acrylates and methacrylates, homopolymers of acrylates and methacrylates, polyvinyl alcohol (PVOH), and polyvinyl pyrrolidone (PVP). 4.-5. (canceled) 6. The method of claim 3, wherein the patient has been diagnosed with one or more of the following: inflammatory bowel disease; an intestinal polyp or an adenoma; having a mutation that is known to cause increased WNT signaling; or having Familial Adenomatous Polyposis (FAP); and/or wherein the patient has a family history of intestinal polyps or intestinal cancer. 7.-11. (canceled) 12. The method of claim 6, wherein the composition comprises rapamycin or an analog thereof at a concentration of 0.001 mg to 30 mg total per dose, and wherein the composition comprising rapamycin or an analog of rapamycin further comprises 0.001% to 60% by weight of rapamycin or an analog of rapamycin. 13. (canceled) 14. The method of claim 12, wherein the average blood level of rapamycin in the subject is greater than 0.5 ng per mL whole blood after administration of the composition. 15. The method of claim 14, wherein the composition is administered orally, enterically, colonically, anally, intravenously, or dermally with a patch. 16. The method of claim 15, wherein the rapamycin or analog of rapamycin is administered in two or more doses. 17. The method of claim 16, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 0.5 to 30 days. 18. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 0.5 to 1 day. 19. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 3 days. 20. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 5 days. 21. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 7 days. 22. The method of claim 17, wherein the interval of time between administration of doses comprising rapamycin or an analog of rapamycin is 1 to 15 days. 23. The method of claim 17, wherein the subject is further administered a composition comprising a second active agent. 24. The method of claim 23, wherein the second active agent is metformin, celocoxib, eflornithine, sulindac, ursodeoxycholic acid, an anti-inflammatory agent, an anti-autoimmune agent, or a cytotoxic or cytostatic anti-cancer agent. 25. The method of claim 24, wherein the composition comprising rapamycin or an analog of rapamycin is administered at the same time as the composition comprising the second active agent. 26. The method of claim 24, wherein the composition comprising rapamycin or an analog of rapamycin is administered before or after the composition comprising the second active agent is administered, and wherein the interval of time between administration of the composition comprising rapamycin or an analog of rapamycin and the composition comprising the second active agent is 1 to 30 days. 27.-28. (canceled) 29. The method of claim 6, wherein the composition comprising rapamycin or an analog of rapamycin prevents intestinal polyps or intestinal cancer, prevents the development of new adenomas or polyps, decreases the number or severity of the adenomatous polyps, induces a reduction in size or number of existing adenomas or polyps, prevents the conversion of adenomas or polyps into adenocarcinomas and cancer tissue, or prevents the adenomas or polyps from converting into malignant cancer that spread into other bodily tissues, organs and blood systems in a patient that has been diagnosed as having intestinal adenomas, intestinal polyps or Familial Adenomatous Polyposis (FAP). 30. The method of claim 29, wherein the composition comprising rapamycin or an analog of rapamycin is comprised in a food or food additive.
1,600
207
207
15,082,658
1,651
The current technology is related to methods for rapidly determining the metabolic baseline and potential of living cells. Embodiments relate to measuring the activity of each of the two major energy-generating pathways within the cell: mitochondrial respiration and glycolysis, first under baseline conditions, and again after applying a stress to the cells to demand increased energy supply. In some embodiments the stress may be applied by exposing the cells to a combination of two chemical compounds: a mitochondrial uncoupler and an ATP synthase inhibitor. In one embodiment, the metabolic energy generating activity of the mitochondrial respiration pathway is determined by measuring the rate of oxygen consumption by the living cells, and the metabolic energy generating activity of the glycolysis pathway is determined from a measurement of extracellular acidification caused by secretion of protons from the cell. Other embodiments are related to an apparatus for determining a metabolic potential of a cell sample in a well of a multiwell plate.
1. A method of determining a metabolic potential of a cell sample, the method comprising the steps of: measuring an initial oxygen consumption rate and an initial extracellular acidification rate of the cell sample; thereafter, simultaneously administering to the cell sample a mitochondrial uncoupling agent and an ATP synthase inhibitor; thereafter, simultaneously measuring a subsequent oxygen consumption rate and a subsequent extracellular acidification rate of the cell sample; and determining the metabolic potential of the cell sample. 2. The method of claim 1, wherein at least one of the mitochondrial uncoupling agents comprises carbonyl cyanide p-trifluoromethoxyphenylhydrazone (“FCCP”), carbonyl cyanide-m-chlorophenylhydrazone (“CCCP”) or 2,4-dinitrophenol (“DNP”) or BAM15, and the ATP synthase inhibitor comprises oligomycin or 7-Chloro-5-(4-hydroxyphenyl)-1-methyl-3-(napthalen-2-ylmetyl)-4,5,-dihydro-1H-benzo[b][1,4]diazepin-2(3H)-one (Bz-423). 3. The method of claim 2, wherein the mitochondrial uncoupling agent comprises carbonyl cyanide p-trifluoromethoxyphenylhydrazone (“FCCP”) and the ATP synthase inhibitor comprises oligomycin. 4. The method of claim 1, wherein the cell sample comprises a plurality of cells disposed in a media. 5. The method of claim 4, wherein measuring the initial oxygen consumption rate comprises sensing a cell constituent disposed in the media. 6. The method of claim 4, wherein measuring the initial extracellular acidification rate of the cell sample comprises sensing a cell constituent disposed in the media. 7. The method of claim 4, wherein a concentration of the administered mitochondrial uncoupling agent in the media is in a range of about 0.1 μM to about 2.0 μM. 8. The method of claim 7, wherein the concentration of the administered mitochondrial uncoupling agent in the media is about 0.5 μM. 9. The method of claim 4, wherein a concentration of the administered ATP synthase inhibitor in the media is about 0.1 μM to about 2 μM. 10. The method of claim 9, wherein the concentration of the administered ATP synthase inhibitor in the media is about 1.0 μM. 11. The method of claim 1, further comprising the step of mixing the mitochondrial uncoupling agent and the ATP synthase inhibitor prior to administering to the cell sample. 12. The method of claim 1, further comprising the step of disposing the cell sample in a well of a multi-well plate prior to simultaneously measuring the initial oxygen consumption rate and the initial extracellular acidification rate of the cell sample. 13. The method of claim 12, wherein administering the mitochondrial uncoupling agent and the ATP synthase inhibitor comprises introducing the agent and the inhibitor simultaneously into the well from at least one port. 14. The method of claim 1, wherein determining the metabolic potential of the cell sample comprises (i) providing the initial oxygen consumption rate, the initial extracellular acidification rate, the subsequent oxygen consumption rate, and the subsequent extracellular acidification rate to a software program, and (ii) using the software program to calculate the metabolic potential. 15. The method of claim 1, wherein the initial oxygen consumption rate and an initial extracellular acidification rate of the cell sample are measured simultaneously. 16. An apparatus for determining a metabolic potential of a cell sample in a well of a multiwell plate, the apparatus comprising: (i) a stage adapted to support a multiwell plate; (ii) a sensor adapted to sense a cell constituent associated with the cell sample in a well of the multiwell plate; and (iii) a dispensing system adapted to introduce fluids into the well, wherein the stage, sensor, and dispensing system cooperate to: simultaneously measure an initial oxygen consumption rate and an initial extracellular acidification rate of the cell sample using the sensor; thereafter, use the dispensing system to simultaneously administer to the cell sample a mitochondrial uncoupling agent and an ATP synthase inhibitor; thereafter, simultaneously measure a subsequent oxygen consumption rate and a subsequent extracellular acidification rate of the cell sample using the sensor; and determine the metabolic potential of the cell sample. 17. The apparatus of claim 16, wherein the dispensing system comprises at least one port disposed above the well. 18. The apparatus of claim 16, wherein the sensor comprises an optical sensor. 19. The apparatus of claim 18, wherein the sensor is adapted to sense a fluorophore. 20. The apparatus of claim 16, further comprising a computer module and software adapted to calculate the metabolic potential based on information communicated to the computer module by the sensor.
The current technology is related to methods for rapidly determining the metabolic baseline and potential of living cells. Embodiments relate to measuring the activity of each of the two major energy-generating pathways within the cell: mitochondrial respiration and glycolysis, first under baseline conditions, and again after applying a stress to the cells to demand increased energy supply. In some embodiments the stress may be applied by exposing the cells to a combination of two chemical compounds: a mitochondrial uncoupler and an ATP synthase inhibitor. In one embodiment, the metabolic energy generating activity of the mitochondrial respiration pathway is determined by measuring the rate of oxygen consumption by the living cells, and the metabolic energy generating activity of the glycolysis pathway is determined from a measurement of extracellular acidification caused by secretion of protons from the cell. Other embodiments are related to an apparatus for determining a metabolic potential of a cell sample in a well of a multiwell plate.1. A method of determining a metabolic potential of a cell sample, the method comprising the steps of: measuring an initial oxygen consumption rate and an initial extracellular acidification rate of the cell sample; thereafter, simultaneously administering to the cell sample a mitochondrial uncoupling agent and an ATP synthase inhibitor; thereafter, simultaneously measuring a subsequent oxygen consumption rate and a subsequent extracellular acidification rate of the cell sample; and determining the metabolic potential of the cell sample. 2. The method of claim 1, wherein at least one of the mitochondrial uncoupling agents comprises carbonyl cyanide p-trifluoromethoxyphenylhydrazone (“FCCP”), carbonyl cyanide-m-chlorophenylhydrazone (“CCCP”) or 2,4-dinitrophenol (“DNP”) or BAM15, and the ATP synthase inhibitor comprises oligomycin or 7-Chloro-5-(4-hydroxyphenyl)-1-methyl-3-(napthalen-2-ylmetyl)-4,5,-dihydro-1H-benzo[b][1,4]diazepin-2(3H)-one (Bz-423). 3. The method of claim 2, wherein the mitochondrial uncoupling agent comprises carbonyl cyanide p-trifluoromethoxyphenylhydrazone (“FCCP”) and the ATP synthase inhibitor comprises oligomycin. 4. The method of claim 1, wherein the cell sample comprises a plurality of cells disposed in a media. 5. The method of claim 4, wherein measuring the initial oxygen consumption rate comprises sensing a cell constituent disposed in the media. 6. The method of claim 4, wherein measuring the initial extracellular acidification rate of the cell sample comprises sensing a cell constituent disposed in the media. 7. The method of claim 4, wherein a concentration of the administered mitochondrial uncoupling agent in the media is in a range of about 0.1 μM to about 2.0 μM. 8. The method of claim 7, wherein the concentration of the administered mitochondrial uncoupling agent in the media is about 0.5 μM. 9. The method of claim 4, wherein a concentration of the administered ATP synthase inhibitor in the media is about 0.1 μM to about 2 μM. 10. The method of claim 9, wherein the concentration of the administered ATP synthase inhibitor in the media is about 1.0 μM. 11. The method of claim 1, further comprising the step of mixing the mitochondrial uncoupling agent and the ATP synthase inhibitor prior to administering to the cell sample. 12. The method of claim 1, further comprising the step of disposing the cell sample in a well of a multi-well plate prior to simultaneously measuring the initial oxygen consumption rate and the initial extracellular acidification rate of the cell sample. 13. The method of claim 12, wherein administering the mitochondrial uncoupling agent and the ATP synthase inhibitor comprises introducing the agent and the inhibitor simultaneously into the well from at least one port. 14. The method of claim 1, wherein determining the metabolic potential of the cell sample comprises (i) providing the initial oxygen consumption rate, the initial extracellular acidification rate, the subsequent oxygen consumption rate, and the subsequent extracellular acidification rate to a software program, and (ii) using the software program to calculate the metabolic potential. 15. The method of claim 1, wherein the initial oxygen consumption rate and an initial extracellular acidification rate of the cell sample are measured simultaneously. 16. An apparatus for determining a metabolic potential of a cell sample in a well of a multiwell plate, the apparatus comprising: (i) a stage adapted to support a multiwell plate; (ii) a sensor adapted to sense a cell constituent associated with the cell sample in a well of the multiwell plate; and (iii) a dispensing system adapted to introduce fluids into the well, wherein the stage, sensor, and dispensing system cooperate to: simultaneously measure an initial oxygen consumption rate and an initial extracellular acidification rate of the cell sample using the sensor; thereafter, use the dispensing system to simultaneously administer to the cell sample a mitochondrial uncoupling agent and an ATP synthase inhibitor; thereafter, simultaneously measure a subsequent oxygen consumption rate and a subsequent extracellular acidification rate of the cell sample using the sensor; and determine the metabolic potential of the cell sample. 17. The apparatus of claim 16, wherein the dispensing system comprises at least one port disposed above the well. 18. The apparatus of claim 16, wherein the sensor comprises an optical sensor. 19. The apparatus of claim 18, wherein the sensor is adapted to sense a fluorophore. 20. The apparatus of claim 16, further comprising a computer module and software adapted to calculate the metabolic potential based on information communicated to the computer module by the sensor.
1,600
208
208
14,613,420
1,613
Products and methods are disclosed for reducing the production of unwanted odors from the pudendum.
1. A product for application to the pudendum of a user to reduce fishy odor, the product comprising at least 1% by weight of mandelic acid in a viscous lipophilic carrier having a non-zero yield stress, the product being physically associated with indicia specifying that the product should be applied to the entire pudendum to alleviate odor. 2. The product of claim 1, wherein the product comprises at least 2% by weight or greater mandelic acid, and further comprises about 1 weight percent or greater lactic acid. 3. The product of claim 1, where the product has an elevated yield stress allowing the product to readily adhere to substantially vertical human skin in an applied layer at least 2 millimeters thick without substantially flowing in response to gravitational force. 4. The product of claim 1, wherein the product has a pH of 4.5 or less and comprises from 2% to 30% mandelic acid. 5. The product of claim 1, wherein the container also holds at least one flexible wipe and wherein the product has a pH of about 5 or lower. 6. The product of claim 5, wherein the at least one flexible wipe is pretreated with the product. 7. The product of claim 1, wherein the indicia provide at least one of instructions that the product should be applied to pudendum after the pudendum has come in contact with a nitrogen-rich substance and instructions that the product should be applied to the pudendum after the pudendum has been exposed to a pH-elevating substance. 8. The product of claim 7, wherein the pH-elevating substance is at least one of semen, blood, menses, vaginal discharge, feces, soap and detergent. 9. The product of claim 7, wherein the nitrogen-rich substance is at least one of semen, blood, menses, feces, and vaginal discharge. 10. A wet wipe product for treating the pudendum of a user comprising a container having one or more flexible porous wipes impregnated with an aqueous solution, the solution having a pH between about 2.8 and 5, the solution comprising from 1% to 30% by weight of mandelic acid, the container further comprising indicia giving directions for use of the wipes on the pudendum. 11. The wet wipe product of claim 10, wherein a substantial portion of the mandelic acid is encapsulated for time release functionality. 12. A feminine wipe product for reducing the production of unwanted odor from the pudendum, the product comprising an openable enclosure containing at least one wipe pretreated with an acidifying composition, the acidifying composition comprising at least 1% by weight of a first alpha-hydroxy acid comprising at least 7 carbons and having a molecular weight in the range of about 135 to about 400, the alpha-hydroxy also acid having no more than one carboxylic acid group for every 7 carbons, the acidifying composition having a pH of from 3.5 to 4.5 and further comprising at least 50% by weight of a viscous lipophilic carrier. 13. The product of claim 12, wherein the first alpha-hydroxy acid comprises mandelic acid. 14. The product of claim 12, wherein the acidifying composition comprises 2% or more of the first alpha-hydroxy acid and about 0.5% or more of a second alpha-hydroxy acid. 15. A method for reducing or preventing fishy odor from the pudendum, comprising: (a) providing a user with a product comprising an acidifying composition having at least 1% by weight of mandelic acid, said acidifying composition having a pH between 2.8 and 5 in a viscous, lipophilic carrier, (b) providing directions to the user to apply the acidifying composition to the entire pudendum. 16. The method of claim 15, wherein the acidifying composition further comprises at least 1% by weight of a second carboxylic acid component, and wherein the pH of said acidifying composition is between 3.2 and 4.5. 17. The method of claim 16, wherein the second carboxylic acid component comprises at least 1% by weight lactic acid. 18. The method of claim 15 wherein said acidifying composition has a non-zero yield stress and wherein the directions are adapted to cause said acidifying composition to effectively remain in contact with the human body for a period of at least 10 minutes after applying said acidifying composition according to the directions. 19. The method of claim 15, wherein said acidifying composition comprises a lipophilic carrier. 20. The method of claim 19, wherein the product and the directions are adapted such that the composition, when applied according to the directions, will be effectively kept in contact with the pudendum for a period of at least 60 minutes, and wherein the pH on the skin in contact with the composition is between about 3.5 and 4.5 during a majority of the at least 60 minutes.
Products and methods are disclosed for reducing the production of unwanted odors from the pudendum.1. A product for application to the pudendum of a user to reduce fishy odor, the product comprising at least 1% by weight of mandelic acid in a viscous lipophilic carrier having a non-zero yield stress, the product being physically associated with indicia specifying that the product should be applied to the entire pudendum to alleviate odor. 2. The product of claim 1, wherein the product comprises at least 2% by weight or greater mandelic acid, and further comprises about 1 weight percent or greater lactic acid. 3. The product of claim 1, where the product has an elevated yield stress allowing the product to readily adhere to substantially vertical human skin in an applied layer at least 2 millimeters thick without substantially flowing in response to gravitational force. 4. The product of claim 1, wherein the product has a pH of 4.5 or less and comprises from 2% to 30% mandelic acid. 5. The product of claim 1, wherein the container also holds at least one flexible wipe and wherein the product has a pH of about 5 or lower. 6. The product of claim 5, wherein the at least one flexible wipe is pretreated with the product. 7. The product of claim 1, wherein the indicia provide at least one of instructions that the product should be applied to pudendum after the pudendum has come in contact with a nitrogen-rich substance and instructions that the product should be applied to the pudendum after the pudendum has been exposed to a pH-elevating substance. 8. The product of claim 7, wherein the pH-elevating substance is at least one of semen, blood, menses, vaginal discharge, feces, soap and detergent. 9. The product of claim 7, wherein the nitrogen-rich substance is at least one of semen, blood, menses, feces, and vaginal discharge. 10. A wet wipe product for treating the pudendum of a user comprising a container having one or more flexible porous wipes impregnated with an aqueous solution, the solution having a pH between about 2.8 and 5, the solution comprising from 1% to 30% by weight of mandelic acid, the container further comprising indicia giving directions for use of the wipes on the pudendum. 11. The wet wipe product of claim 10, wherein a substantial portion of the mandelic acid is encapsulated for time release functionality. 12. A feminine wipe product for reducing the production of unwanted odor from the pudendum, the product comprising an openable enclosure containing at least one wipe pretreated with an acidifying composition, the acidifying composition comprising at least 1% by weight of a first alpha-hydroxy acid comprising at least 7 carbons and having a molecular weight in the range of about 135 to about 400, the alpha-hydroxy also acid having no more than one carboxylic acid group for every 7 carbons, the acidifying composition having a pH of from 3.5 to 4.5 and further comprising at least 50% by weight of a viscous lipophilic carrier. 13. The product of claim 12, wherein the first alpha-hydroxy acid comprises mandelic acid. 14. The product of claim 12, wherein the acidifying composition comprises 2% or more of the first alpha-hydroxy acid and about 0.5% or more of a second alpha-hydroxy acid. 15. A method for reducing or preventing fishy odor from the pudendum, comprising: (a) providing a user with a product comprising an acidifying composition having at least 1% by weight of mandelic acid, said acidifying composition having a pH between 2.8 and 5 in a viscous, lipophilic carrier, (b) providing directions to the user to apply the acidifying composition to the entire pudendum. 16. The method of claim 15, wherein the acidifying composition further comprises at least 1% by weight of a second carboxylic acid component, and wherein the pH of said acidifying composition is between 3.2 and 4.5. 17. The method of claim 16, wherein the second carboxylic acid component comprises at least 1% by weight lactic acid. 18. The method of claim 15 wherein said acidifying composition has a non-zero yield stress and wherein the directions are adapted to cause said acidifying composition to effectively remain in contact with the human body for a period of at least 10 minutes after applying said acidifying composition according to the directions. 19. The method of claim 15, wherein said acidifying composition comprises a lipophilic carrier. 20. The method of claim 19, wherein the product and the directions are adapted such that the composition, when applied according to the directions, will be effectively kept in contact with the pudendum for a period of at least 60 minutes, and wherein the pH on the skin in contact with the composition is between about 3.5 and 4.5 during a majority of the at least 60 minutes.
1,600
209
209
14,283,073
1,611
A waterborne topical composition is designed specifically to address the treatment of acne vulgaris, rosacea, seborrheic dermatitis and other skin conditions. One composition contains effective amounts of essential components azelaic acid, niacinamide, and glycerin to create a rapidly penetrating and non-irritating compound. One composition contains effective amounts of essential components azelaic acid, niacinamide, and cyclodextran to create a rapidly penetrating and non-irritating compound.
1. A waterborne topical composition for the treatment of acne vulgaris, rosacea, seborrheic dermatitis or other skin conditions comprising effective amounts of azelaic acid, niacinamide, and wherein the composition demonstrates a penetration rate of at least 5% active ingredient/hr, within 2.5 hours of application to human skin. 2. The waterborne topical composition of claim 1 wherein the azelaic acid is present in an amount of up to 20 percent by weight further including cyclodextran. 3. The waterborne topical composition of claim 2 wherein the azelaic acid is present in an amount of at least 10 percent by weight and the cyclodextran is less than 6 percent by weight. 4. The waterborne topical composition of claim 1 wherein the and wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion further including cyclodextran. 5. The waterborne topical composition of claim 1 wherein the niacinamide is present in an amount of up to 10 percent by weight and further including cyclodextran in an amount less than 6 percent by weight. 6. The waterborne topical composition of claim 1 further including glycerin which is present in an amount of up to 10 percent by weight and further including cyclodextran. 7. The waterborne topical composition of claim 1 further comprising hyaluronic acid and/or a derivative thereof and further including hydroxypropyl beta. 8. The waterborne topical composition of claim 1 wherein the composition includes effective amounts of glycerin and wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion and further including hydroxypropyl beta. 9. The waterborne topical composition of claim 1 wherein the composition is essentially free of azelaic acid esters and further including hydroxypropyl beta. 10. The waterborne topical composition of claim 1 and further comprising one or more alcohols, a surfactant, isopropyl palmitate, sorbitol, and lactic acid; and further including hydroxypropyl beta. 11. The waterborne topical composition of claim 10 wherein the alcohol comprises cetyl alcohol, stearyl alcohol, and/or benzyl alcohol. 12. The waterborne topical composition of claim 10 wherein the surfactant comprises sodium lauryl sulfate. 13. The waterborne topical composition of claim 10 further including effective amounts of glycerin and wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion. 14. The waterborne topical composition of claim 10 wherein the composition is essentially free of azelaic acid esters. 15. A waterborne topical composition comprising: effective amounts of azelaic acid, wherein the azelaic acid is present in an amount of 4 percent to 20 percent by weight; effective amounts of niacinamide, wherein the niacinamide is present in an amount of 4 to 10 percent by weight; and effective amounts of cyclodextran, wherein the cyclodextran is present in an amount of 0.1 to 6 percent by weight. 16. The waterborne topical composition of claim 15 wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion. 17. The waterborne topical composition of claim 15 wherein the wherein the composition demonstrates a penetration rate of at least 5% active ingredient/hour, within 2.5 hours of application to human skin and the composition is a gel. 18. A waterborne topical composition for topical application of at least one active ingredient comprising effective amounts of azelaic acid, niacinamide, and hydroxypropyl beta. 19. The waterborne topical composition of claim 18 wherein the azelaic acid is present in an amount of 4 to 20 percent by weight, and the niacinamide is present in an amount of up to 10 percent by weight, and the hydroxypropyl beta is present in an amount of up to 6 percent by weight. 20. The waterborne topical composition of claim 19 wherein the composition demonstrates a penetration rate of at least 5% active ingredient/hour, within 2.5 hours of application to human skin.
A waterborne topical composition is designed specifically to address the treatment of acne vulgaris, rosacea, seborrheic dermatitis and other skin conditions. One composition contains effective amounts of essential components azelaic acid, niacinamide, and glycerin to create a rapidly penetrating and non-irritating compound. One composition contains effective amounts of essential components azelaic acid, niacinamide, and cyclodextran to create a rapidly penetrating and non-irritating compound.1. A waterborne topical composition for the treatment of acne vulgaris, rosacea, seborrheic dermatitis or other skin conditions comprising effective amounts of azelaic acid, niacinamide, and wherein the composition demonstrates a penetration rate of at least 5% active ingredient/hr, within 2.5 hours of application to human skin. 2. The waterborne topical composition of claim 1 wherein the azelaic acid is present in an amount of up to 20 percent by weight further including cyclodextran. 3. The waterborne topical composition of claim 2 wherein the azelaic acid is present in an amount of at least 10 percent by weight and the cyclodextran is less than 6 percent by weight. 4. The waterborne topical composition of claim 1 wherein the and wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion further including cyclodextran. 5. The waterborne topical composition of claim 1 wherein the niacinamide is present in an amount of up to 10 percent by weight and further including cyclodextran in an amount less than 6 percent by weight. 6. The waterborne topical composition of claim 1 further including glycerin which is present in an amount of up to 10 percent by weight and further including cyclodextran. 7. The waterborne topical composition of claim 1 further comprising hyaluronic acid and/or a derivative thereof and further including hydroxypropyl beta. 8. The waterborne topical composition of claim 1 wherein the composition includes effective amounts of glycerin and wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion and further including hydroxypropyl beta. 9. The waterborne topical composition of claim 1 wherein the composition is essentially free of azelaic acid esters and further including hydroxypropyl beta. 10. The waterborne topical composition of claim 1 and further comprising one or more alcohols, a surfactant, isopropyl palmitate, sorbitol, and lactic acid; and further including hydroxypropyl beta. 11. The waterborne topical composition of claim 10 wherein the alcohol comprises cetyl alcohol, stearyl alcohol, and/or benzyl alcohol. 12. The waterborne topical composition of claim 10 wherein the surfactant comprises sodium lauryl sulfate. 13. The waterborne topical composition of claim 10 further including effective amounts of glycerin and wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion. 14. The waterborne topical composition of claim 10 wherein the composition is essentially free of azelaic acid esters. 15. A waterborne topical composition comprising: effective amounts of azelaic acid, wherein the azelaic acid is present in an amount of 4 percent to 20 percent by weight; effective amounts of niacinamide, wherein the niacinamide is present in an amount of 4 to 10 percent by weight; and effective amounts of cyclodextran, wherein the cyclodextran is present in an amount of 0.1 to 6 percent by weight. 16. The waterborne topical composition of claim 15 wherein the waterborne composition is in the form of one of a liquid, emulsion and microemulsion. 17. The waterborne topical composition of claim 15 wherein the wherein the composition demonstrates a penetration rate of at least 5% active ingredient/hour, within 2.5 hours of application to human skin and the composition is a gel. 18. A waterborne topical composition for topical application of at least one active ingredient comprising effective amounts of azelaic acid, niacinamide, and hydroxypropyl beta. 19. The waterborne topical composition of claim 18 wherein the azelaic acid is present in an amount of 4 to 20 percent by weight, and the niacinamide is present in an amount of up to 10 percent by weight, and the hydroxypropyl beta is present in an amount of up to 6 percent by weight. 20. The waterborne topical composition of claim 19 wherein the composition demonstrates a penetration rate of at least 5% active ingredient/hour, within 2.5 hours of application to human skin.
1,600
210
210
14,692,422
1,633
The invention relates to methods of targeted drug delivery of antiviral compounds, including, chemical agents (like nucleoside analogs or protease inhibitors) and nucleic acid based drugs (like DNA vaccines, antisense oligonucleotides, ribozymes, catalytic DNA (DNAzymes) or RNA molecules, siRNAs or plasmids encoding thereof). Furthermore, the invention relates to targeted drug delivery of antiviral compounds to intracellular target sites within cells, tissues and organs, in particular to target sites within the central nervous system (CNS), into and across the blood-brain barrier, by targeting to internalizing uptake receptors present on these cells, tissues and organs. Thereto, the antiviral compounds, or the pharmaceutical acceptable carrier thereof, are conjugated to ligands that facilitate the specific binding to and internalization by these receptors.
1. A method for delivering a drug across the blood-central nervous system (CNS) barrier, comprising administering to a subject in need thereof an effective amount of a drug-encapsulating nanocontainer according to claim 21 or claim 22 such that the drug (a) is delivered across the blood-CNS barrier, (b) accumulates selectively in the subject's brain compared to tissues other than brain, and (c) accumulates to a greater extent in brain than does the drug encapsulated in a similarly administered control nanocontainer (i) which does not comprise a GR-binding conjugate or (ii) to which the polyethylene glycol alone is linked. 2. The method according to claim 1 wherein the blood-CNS barrier is the blood brain barrier. 3. The method according to claim 1 wherein said other tissue is one or more of heart, lung, liver, spleen and kidney. 4. The method of claim 1 wherein the lipid-polyethylene glycol is distearoylphosphatidylethanolamine-polyethylene glycol (DSPE-PEG)-maleimide (DSPE-PEG-MAL). 5. The method of claim 1 wherein the ligand is reduced glutathione. 6. The method of claim 1, wherein the polyethylene glycol has an average molecular weight of about 2000 or about 3400 Daltons. 7. The method of claim 1, wherein the conjugate has the formula: wherein R1 is the lipid-polyethylene glycol, and R2 is the ligand for a glutathione receptor. 8. The method of claim 7, wherein the ligand is reduced glutathione. 9. The method of claim 7, wherein R1 is DSPE-PEG. 10. The method of claim 7 wherein the polyethylene glycol has an average molecular weight of about 2000 or about 3400 Daltons. 11. The method of claim 1, wherein the nanocontainer is a nanoparticle, a liposome, a nanogel, a polyplex system or a lipoplex system. 12. The method according to claim 1, wherein the drug is an antiviral drug. 13. The method according to claim 12, wherein the drug is ribavirin. 14. A method for delivering a drug across the blood-CNS barrier, comprising administering to a subject in need thereof an effective dose of a drug-encapsulating nanocontainer that comprises a glutathione receptor-binding conjugate produced by reacting (a) a lipid-polyethylene glycol comprising a thiol-reactive group with (b) a GR ligand that comprises a maleimid-reactive thiol group; and such that the drug: (i) is delivered across the blood-CNS barrier; (ii) accumulates selectively in the subject's brain compared to tissues other than brain; and (iii) accumulates to a greater extent in the brain than does the drug encapsulated in a similarly administered control nanocontainer (A) which does not comprise a GR-binding conjugate or (B) to which the polyethylene glycol alone is linked. 15. The method according to claim 14 wherein the blood-CNS barrier is the blood brain barrier. 16. The method according to claim 14 wherein said other tissue is one or more of heart, lung, liver, spleen and kidney. 17. The method according to claim 14 wherein the thiol group in (b) is a maleimide-reactive thiol group. 18. The method of claim 17 wherein the lipid-polyethylene glycol of (a) is DSPE-PEG-MAL 19. The method of claim 14 wherein the ligand is reduced glutathione. 20. The method of claim 14, wherein the polyethylene glycol has an average molecular weight of about 2000 or about 3400 Daltons. 21. A conjugate comprising: a) a glutathione receptor (GR)—binding conjugate; and, b) at least one of a drug, and a nanocontainer comprising a drug; wherein the glutathione is conjugated to at least one of the agent and carrier in b). 22. A conjugate according to claim 21, wherein the conjugate is characterized by a lipid-polyethylene glycol linked to a thiol group of a GR ligand.
The invention relates to methods of targeted drug delivery of antiviral compounds, including, chemical agents (like nucleoside analogs or protease inhibitors) and nucleic acid based drugs (like DNA vaccines, antisense oligonucleotides, ribozymes, catalytic DNA (DNAzymes) or RNA molecules, siRNAs or plasmids encoding thereof). Furthermore, the invention relates to targeted drug delivery of antiviral compounds to intracellular target sites within cells, tissues and organs, in particular to target sites within the central nervous system (CNS), into and across the blood-brain barrier, by targeting to internalizing uptake receptors present on these cells, tissues and organs. Thereto, the antiviral compounds, or the pharmaceutical acceptable carrier thereof, are conjugated to ligands that facilitate the specific binding to and internalization by these receptors.1. A method for delivering a drug across the blood-central nervous system (CNS) barrier, comprising administering to a subject in need thereof an effective amount of a drug-encapsulating nanocontainer according to claim 21 or claim 22 such that the drug (a) is delivered across the blood-CNS barrier, (b) accumulates selectively in the subject's brain compared to tissues other than brain, and (c) accumulates to a greater extent in brain than does the drug encapsulated in a similarly administered control nanocontainer (i) which does not comprise a GR-binding conjugate or (ii) to which the polyethylene glycol alone is linked. 2. The method according to claim 1 wherein the blood-CNS barrier is the blood brain barrier. 3. The method according to claim 1 wherein said other tissue is one or more of heart, lung, liver, spleen and kidney. 4. The method of claim 1 wherein the lipid-polyethylene glycol is distearoylphosphatidylethanolamine-polyethylene glycol (DSPE-PEG)-maleimide (DSPE-PEG-MAL). 5. The method of claim 1 wherein the ligand is reduced glutathione. 6. The method of claim 1, wherein the polyethylene glycol has an average molecular weight of about 2000 or about 3400 Daltons. 7. The method of claim 1, wherein the conjugate has the formula: wherein R1 is the lipid-polyethylene glycol, and R2 is the ligand for a glutathione receptor. 8. The method of claim 7, wherein the ligand is reduced glutathione. 9. The method of claim 7, wherein R1 is DSPE-PEG. 10. The method of claim 7 wherein the polyethylene glycol has an average molecular weight of about 2000 or about 3400 Daltons. 11. The method of claim 1, wherein the nanocontainer is a nanoparticle, a liposome, a nanogel, a polyplex system or a lipoplex system. 12. The method according to claim 1, wherein the drug is an antiviral drug. 13. The method according to claim 12, wherein the drug is ribavirin. 14. A method for delivering a drug across the blood-CNS barrier, comprising administering to a subject in need thereof an effective dose of a drug-encapsulating nanocontainer that comprises a glutathione receptor-binding conjugate produced by reacting (a) a lipid-polyethylene glycol comprising a thiol-reactive group with (b) a GR ligand that comprises a maleimid-reactive thiol group; and such that the drug: (i) is delivered across the blood-CNS barrier; (ii) accumulates selectively in the subject's brain compared to tissues other than brain; and (iii) accumulates to a greater extent in the brain than does the drug encapsulated in a similarly administered control nanocontainer (A) which does not comprise a GR-binding conjugate or (B) to which the polyethylene glycol alone is linked. 15. The method according to claim 14 wherein the blood-CNS barrier is the blood brain barrier. 16. The method according to claim 14 wherein said other tissue is one or more of heart, lung, liver, spleen and kidney. 17. The method according to claim 14 wherein the thiol group in (b) is a maleimide-reactive thiol group. 18. The method of claim 17 wherein the lipid-polyethylene glycol of (a) is DSPE-PEG-MAL 19. The method of claim 14 wherein the ligand is reduced glutathione. 20. The method of claim 14, wherein the polyethylene glycol has an average molecular weight of about 2000 or about 3400 Daltons. 21. A conjugate comprising: a) a glutathione receptor (GR)—binding conjugate; and, b) at least one of a drug, and a nanocontainer comprising a drug; wherein the glutathione is conjugated to at least one of the agent and carrier in b). 22. A conjugate according to claim 21, wherein the conjugate is characterized by a lipid-polyethylene glycol linked to a thiol group of a GR ligand.
1,600
211
211
13,772,796
1,641
The present invention provides methods, kits, compositions, and devices for detecting Anti-Mullerian hormone (AMH) in whole blood samples. In certain embodiments, the methods, kits, compositions, and devices employ immunoassays that generate a colorimetric or fluorescent signal (e.g., using antibodies conjugated to gold nanoparticles or fluorescent particles) where the signal generated is proportional to the approximate concentration of AMH in a whole blood sample. In particular embodiments, the present invention provides quantitative or semi-quantitative lateral flow immunoassay devices and kits for detecting AMH at home (e.g., in order for women to estimate their ovarian age or diagnose polycystic ovarian syndrome).
1. A method of determining the approximate concentration of Anti-Mullerian hormone (AMH) in a whole blood sample comprising: a) contacting a whole blood sample from a subject with first antibodies specific for AMH under conditions such that a signal is generated that is proportional to the approximate concentration of AMH in said whole blood sample; and b) detecting the approximate level of said signal, thereby determining said approximate concentration of AMH in said whole blood sample. 2. The method of claim 1, wherein said first antibodies are labeled with first nanoparticles that produce a colorimetric or fluorescent signal when aggregated. 3. The method of claim 1, wherein said first nanoparticles comprise gold nanoparticles or fluorescent particles. 4. The method of claim 1, further comprising contacting said whole blood sample with a second antibodies specific for a non-AMH protein in whole blood. 5. The method of claim 1, wherein said second antibodies are labeled with first nanoparticles that produce a colorimetric or fluorescent signal when aggregated. 6. The method of claim 1, wherein said contacting is conducted on a membrane, wherein said membrane comprises: at least one test capture region which comprises third antibodies specific for said AMH or said first antibodies. 7. The method of claim 6, wherein said membrane further comprises: a control capture region which comprises fourth antibodies specific for said non-AMH protein or said second antibodies. 8. The method of claim 1, wherein said detecting said level of said signal comprises detecting the fluorescence absorbance level, the colorimetric intensity level, or the number of colorimetric symbols from, said signal. 9. The method of claim 1, further comprising comparing said approximate amount of said signal to reference signals of known AMH concentration in order to determine said approximate concentration of AMH in said whole blood sample. 10. The method of claim 1, wherein said whole blood sample has a volume of 1 drop of whole blood or less. 11. The method of claim 1, wherein said whole blood sample comprises oxygenated whole blood or a dried blood sample. 12. The method of claim 1, wherein said approximate concentration of AMH detected in said whole blood sample is greater than 3.5 ng/ml, wherein said subject is a female, and wherein said method further comprises at least one of the following steps: i) informing said subject that she has, or likely has, polycystic ovarian syndrome; ii) preparing and/or transmitting an electronic and/or paper report that indicates said subject has, or likely has, polycystic ovarian syndrome; iii) preparing and/or transmitting an electronic and/or paper report that said subject should be further evaluated for polycystic ovarian syndrome; iv) prescribing medication and/or surgical treatment to said subject to treat polycystic ovarian syndrome; and v) treating said subject with medication or surgical treatment directed toward alleviating polycystic ovarian syndrome. 13. A lateral flow immunoassay device for detecting Anti-Mullerian hormone (AMH) in whole blood comprising: a) a sample pad configured for receiving and transmitting a whole blood sample; b) a conjugate pad in contact with said sample pad and configured for receiving said whole blood sample from said sample pad, wherein said conjugate pad comprises: i) first antibodies specific for AMH, wherein said first antibodies are labeled with first nanoparticles that produce a first colorimetric or fluorescent signal when aggregated, ii) second antibodies specific for a non-AMH protein in whole blood, wherein said second antibodies are labeled with second nanoparticles that produce a second colorimetric signal when aggregated; c) a membrane in contact with said conjugate pad and configured to receive said whole blood sample from said conjugate pad, wherein said membrane comprises: i) at least one test capture region which comprises third antibodies specific for said AMH or said first antibodies, and d) a substrate, wherein said sample pad, said conjugate pad, and said membrane, are supported by said substrate. 14. The lateral flow immunoassay device of claim 13, wherein said membrane further comprises: ii) a control capture region which comprises fourth antibodies specific for said non-AMH protein or said second antibodies. 15. The lateral flow immunoassay device of claim 13, further comprising a wick component in contact with said membrane and configured to absorb excess whole blood sample. 16. The lateral flow immunoassay device of claim 13, wherein said conjugate pad, said membrane, and said wick component are attached to said substrate. 17. The lateral flow immunoassay device of claim 13, wherein said at least one test capture region comprises at least two test capture regions. 18. The lateral flow immunoassay device of claim 13, wherein said third antibodies are present in said at least one test capture region at an excess level compared to the maximum level of AMH that could be present in the amount of said whole blood that could reach said at least one test capture region. 19. The lateral flow immunoassay device of claim 13, wherein the intensity of said first colorimetric or fluorescent signal is proportional to the concentration of AMH present in said whole blood sample. 20. The lateral flow immunoassay device of claim 13, wherein said at least one test capture region is in the shape or a line, circle, or oval, and wherein said control capture region is in the shape of a line, circle, or oval. 21. The lateral flow immunoassay device of claim 13, further comprising a blood reservoir located on top of said sample pad, wherein said blood reservoir is configured to receive a dried blood sample. 22. A kit comprising: a) said lateral flow immunoassay device of claim 13, and b) at least one component selected from the group consisting of: i) at least one sterile lancet, ii) a gas impermeable foil bag, iii) a color chart, wherein said color chart allows a user of said lateral flow immunoassay to estimate the concentration of AMH in a whole blood sample tested on said lateral flow immunoassay device by comparison to said color chart, iv) a sterile gauze pad, v) a skin sterilization wipe, vi) printed instructions for collecting blood and applying it to said lateral flow immunoassay device, vii) printed instructions for interpreting said first colorimetric signal, viii) a piece of filter paper for collecting a dried blood sample, and ix) a container for housing said lateral flow immunoassay device. 23. A method of using a lateral flow immunoassay device for detecting Anti-Mullerian hormone (AMH) in a whole blood sample comprising: applying a whole blood sample from a subject to said sample pad of said lateral flow immunoassay device of claim 13 under conditions such that at least a portion of said whole blood sample migrates from said sample pad, through said conjugate pad to said at least one test capture region and said control capture region in said membrane thereby generating said first and second colorimetric or fluorescent signals, wherein said first colorimetric/fluorescent signal is proportional to the approximate concentration of AMH in said whole blood sample. 24. The method of claim 23, wherein said approximate concentration of AMH detected in said whole blood sample is greater than 3.5 ng/ml, wherein said subject is a female, and wherein said method further comprises at least one of the following steps: i) informing said subject that she has, or likely has, polycystic ovarian syndrome; ii) preparing and/or transmitting an electronic and/or paper report that indicates said subject has, or likely has, polycystic ovarian syndrome; iii) preparing and/or transmitting an electronic and/or paper report that said subject should be further evaluated for polycystic ovarian syndrome; iv) prescribing medication and/or surgical treatment to said subject to treat polycystic ovarian syndrome; and v) treating said subject with medication or surgical treatment directed toward alleviating polycystic ovarian syndrome.
The present invention provides methods, kits, compositions, and devices for detecting Anti-Mullerian hormone (AMH) in whole blood samples. In certain embodiments, the methods, kits, compositions, and devices employ immunoassays that generate a colorimetric or fluorescent signal (e.g., using antibodies conjugated to gold nanoparticles or fluorescent particles) where the signal generated is proportional to the approximate concentration of AMH in a whole blood sample. In particular embodiments, the present invention provides quantitative or semi-quantitative lateral flow immunoassay devices and kits for detecting AMH at home (e.g., in order for women to estimate their ovarian age or diagnose polycystic ovarian syndrome).1. A method of determining the approximate concentration of Anti-Mullerian hormone (AMH) in a whole blood sample comprising: a) contacting a whole blood sample from a subject with first antibodies specific for AMH under conditions such that a signal is generated that is proportional to the approximate concentration of AMH in said whole blood sample; and b) detecting the approximate level of said signal, thereby determining said approximate concentration of AMH in said whole blood sample. 2. The method of claim 1, wherein said first antibodies are labeled with first nanoparticles that produce a colorimetric or fluorescent signal when aggregated. 3. The method of claim 1, wherein said first nanoparticles comprise gold nanoparticles or fluorescent particles. 4. The method of claim 1, further comprising contacting said whole blood sample with a second antibodies specific for a non-AMH protein in whole blood. 5. The method of claim 1, wherein said second antibodies are labeled with first nanoparticles that produce a colorimetric or fluorescent signal when aggregated. 6. The method of claim 1, wherein said contacting is conducted on a membrane, wherein said membrane comprises: at least one test capture region which comprises third antibodies specific for said AMH or said first antibodies. 7. The method of claim 6, wherein said membrane further comprises: a control capture region which comprises fourth antibodies specific for said non-AMH protein or said second antibodies. 8. The method of claim 1, wherein said detecting said level of said signal comprises detecting the fluorescence absorbance level, the colorimetric intensity level, or the number of colorimetric symbols from, said signal. 9. The method of claim 1, further comprising comparing said approximate amount of said signal to reference signals of known AMH concentration in order to determine said approximate concentration of AMH in said whole blood sample. 10. The method of claim 1, wherein said whole blood sample has a volume of 1 drop of whole blood or less. 11. The method of claim 1, wherein said whole blood sample comprises oxygenated whole blood or a dried blood sample. 12. The method of claim 1, wherein said approximate concentration of AMH detected in said whole blood sample is greater than 3.5 ng/ml, wherein said subject is a female, and wherein said method further comprises at least one of the following steps: i) informing said subject that she has, or likely has, polycystic ovarian syndrome; ii) preparing and/or transmitting an electronic and/or paper report that indicates said subject has, or likely has, polycystic ovarian syndrome; iii) preparing and/or transmitting an electronic and/or paper report that said subject should be further evaluated for polycystic ovarian syndrome; iv) prescribing medication and/or surgical treatment to said subject to treat polycystic ovarian syndrome; and v) treating said subject with medication or surgical treatment directed toward alleviating polycystic ovarian syndrome. 13. A lateral flow immunoassay device for detecting Anti-Mullerian hormone (AMH) in whole blood comprising: a) a sample pad configured for receiving and transmitting a whole blood sample; b) a conjugate pad in contact with said sample pad and configured for receiving said whole blood sample from said sample pad, wherein said conjugate pad comprises: i) first antibodies specific for AMH, wherein said first antibodies are labeled with first nanoparticles that produce a first colorimetric or fluorescent signal when aggregated, ii) second antibodies specific for a non-AMH protein in whole blood, wherein said second antibodies are labeled with second nanoparticles that produce a second colorimetric signal when aggregated; c) a membrane in contact with said conjugate pad and configured to receive said whole blood sample from said conjugate pad, wherein said membrane comprises: i) at least one test capture region which comprises third antibodies specific for said AMH or said first antibodies, and d) a substrate, wherein said sample pad, said conjugate pad, and said membrane, are supported by said substrate. 14. The lateral flow immunoassay device of claim 13, wherein said membrane further comprises: ii) a control capture region which comprises fourth antibodies specific for said non-AMH protein or said second antibodies. 15. The lateral flow immunoassay device of claim 13, further comprising a wick component in contact with said membrane and configured to absorb excess whole blood sample. 16. The lateral flow immunoassay device of claim 13, wherein said conjugate pad, said membrane, and said wick component are attached to said substrate. 17. The lateral flow immunoassay device of claim 13, wherein said at least one test capture region comprises at least two test capture regions. 18. The lateral flow immunoassay device of claim 13, wherein said third antibodies are present in said at least one test capture region at an excess level compared to the maximum level of AMH that could be present in the amount of said whole blood that could reach said at least one test capture region. 19. The lateral flow immunoassay device of claim 13, wherein the intensity of said first colorimetric or fluorescent signal is proportional to the concentration of AMH present in said whole blood sample. 20. The lateral flow immunoassay device of claim 13, wherein said at least one test capture region is in the shape or a line, circle, or oval, and wherein said control capture region is in the shape of a line, circle, or oval. 21. The lateral flow immunoassay device of claim 13, further comprising a blood reservoir located on top of said sample pad, wherein said blood reservoir is configured to receive a dried blood sample. 22. A kit comprising: a) said lateral flow immunoassay device of claim 13, and b) at least one component selected from the group consisting of: i) at least one sterile lancet, ii) a gas impermeable foil bag, iii) a color chart, wherein said color chart allows a user of said lateral flow immunoassay to estimate the concentration of AMH in a whole blood sample tested on said lateral flow immunoassay device by comparison to said color chart, iv) a sterile gauze pad, v) a skin sterilization wipe, vi) printed instructions for collecting blood and applying it to said lateral flow immunoassay device, vii) printed instructions for interpreting said first colorimetric signal, viii) a piece of filter paper for collecting a dried blood sample, and ix) a container for housing said lateral flow immunoassay device. 23. A method of using a lateral flow immunoassay device for detecting Anti-Mullerian hormone (AMH) in a whole blood sample comprising: applying a whole blood sample from a subject to said sample pad of said lateral flow immunoassay device of claim 13 under conditions such that at least a portion of said whole blood sample migrates from said sample pad, through said conjugate pad to said at least one test capture region and said control capture region in said membrane thereby generating said first and second colorimetric or fluorescent signals, wherein said first colorimetric/fluorescent signal is proportional to the approximate concentration of AMH in said whole blood sample. 24. The method of claim 23, wherein said approximate concentration of AMH detected in said whole blood sample is greater than 3.5 ng/ml, wherein said subject is a female, and wherein said method further comprises at least one of the following steps: i) informing said subject that she has, or likely has, polycystic ovarian syndrome; ii) preparing and/or transmitting an electronic and/or paper report that indicates said subject has, or likely has, polycystic ovarian syndrome; iii) preparing and/or transmitting an electronic and/or paper report that said subject should be further evaluated for polycystic ovarian syndrome; iv) prescribing medication and/or surgical treatment to said subject to treat polycystic ovarian syndrome; and v) treating said subject with medication or surgical treatment directed toward alleviating polycystic ovarian syndrome.
1,600
212
212
13,147,905
1,643
A method for distinguishing prostate cancer from prostatic hypertrophy using the method for analyzing PSA and an analysis kit of PSA are provided. An object of the present invention can be solved by being brought into contact a lectin having an affinity for β-N-acetylgalactosamine residues and/or a lectin having an affinity for fucose α(1, 2) galactose residues with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectin. A method for distinguishing prostate cancer from prostatic hypertrophy can be provided by this method.
1. A method for analyzing PSA, characterized in that a lectin having an affinity for β-N-acetylgalactosamine residues is brought into contact with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectin. 2. The method for analyzing PSA according to claim 1, comprising the steps of (a) bringing into contact the lectin having an affinity for β-N-acetylgalactosamine residues with the sample possibly containing PSA, to separate PSA having an affinity for the lectin from PSA lacking an affinity for the lectin; and (b) determining the amount of PSA having an affinity for the lectin. 3. The method for analyzing PSA according to claim 2, wherein the amount of PSA having an affinity for the lectin is determined (1) by measuring an amount of separated PSA having an affinity for the lectin, (2) by measuring an amount of PSA in a sample before the separation and an amount of the separated PSA having an affinity for the lectin, or (3) by measuring an amount of PSA in a sample before the separation and an amount of the separated PSA lacking an affinity for the lectin. 4. The method for analyzing PSA according to claim 3, wherein the amount of PSA is determined by measuring total PSA or free PSA. 5. The method for analyzing PSA according to claim 1, wherein the lectin is Trichosanthes japonica agglutinin-II or Wisteria floribunda agglutinin. 6. The method for analyzing PSA according to claim 1, wherein the lectin further has an affinity for fucose α(1, 2) galactose residues. 7. The method for analyzing PSA according to claim 1, wherein the sample is obtained from a patient suspected of having prostate cancer. 8. The method for analyzing PSA according to claim 1, for diagnoses of the prostate cancer. 9. A method for analyzing PSA, characterized in that a lectin having an affinity for fucose α(1, 2) galactose residues is brought into contact with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectin. 10. The method for analyzing PSA according to claim 9, comprising the steps of (a) bringing into contact the lectin having an affinity for fucose α(1, 2) galactose residues with the sample possibly containing PSA, to separate PSA having an affinity for the lectin from PSA lacking an affinity for the lectin; and (b) determining the amount of PSA having an affinity for the lectin. 11. A method for analyzing PSA, characterized in that a lectin having an affinity for β-N-acetylgalactosamine residues and a lectin having an affinity for fucose α(1, 2) galactose residues are brought into contact with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectins. 12. The method for analyzing PSA according to claim 11, comprising the steps of (a) bringing the lectin having an affinity for β-N-acetylgalactosamine residues and the lectin having an affinity for fucose α(1, 2) galactose residues into contact with the sample possibly containing PSA, to separate PSA having an affinity for the lectins from PSA lacking an affinity for the lectins; and (b) determining the amount of PSA having an affinity for the lectins. 13. A method for distinguishing prostate cancer from prostatic hypertrophy, characterized in that the amount of PSA having an affinity for a lectin in a sample is analyzed by the method for analyzing PSA according to claim 1. 14. An analysis kit of PSA, comprising a lectin having an affinity for β-N-acetylgalactosamine residues and an anti-PSA antibody. 15. (canceled) 16. The analysis kit of PSA according to claim 14, wherein the lectin is Trichosanthes japonica agglutinin-II or Wisteria floribunda agglutinin. 17. The analysis kit of PSA according to claim 14, wherein the lectin further has an affinity for fucose α(1, 2) galactose residues. 18. An analysis kit of PSA, comprising a lectin having an affinity for fucose α(1, 2) galactose residues and an anti-PSA antibody. 19. An analysis kit of PSA, comprising a lectin having an affinity for β-N-acetylgalactosamine residues, and a lectin having an affinity for fucose α(1, 2) galactose residues and an anti-PSA antibody. 20. A PSA having β-N-acetylgalactosamine residues. 21. A PSA having fucose α(1, 2) galactose residues. 22. A PSA having β-N-acetylgalactosamine residues and fucose α(1, 2) galactose residues.
A method for distinguishing prostate cancer from prostatic hypertrophy using the method for analyzing PSA and an analysis kit of PSA are provided. An object of the present invention can be solved by being brought into contact a lectin having an affinity for β-N-acetylgalactosamine residues and/or a lectin having an affinity for fucose α(1, 2) galactose residues with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectin. A method for distinguishing prostate cancer from prostatic hypertrophy can be provided by this method.1. A method for analyzing PSA, characterized in that a lectin having an affinity for β-N-acetylgalactosamine residues is brought into contact with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectin. 2. The method for analyzing PSA according to claim 1, comprising the steps of (a) bringing into contact the lectin having an affinity for β-N-acetylgalactosamine residues with the sample possibly containing PSA, to separate PSA having an affinity for the lectin from PSA lacking an affinity for the lectin; and (b) determining the amount of PSA having an affinity for the lectin. 3. The method for analyzing PSA according to claim 2, wherein the amount of PSA having an affinity for the lectin is determined (1) by measuring an amount of separated PSA having an affinity for the lectin, (2) by measuring an amount of PSA in a sample before the separation and an amount of the separated PSA having an affinity for the lectin, or (3) by measuring an amount of PSA in a sample before the separation and an amount of the separated PSA lacking an affinity for the lectin. 4. The method for analyzing PSA according to claim 3, wherein the amount of PSA is determined by measuring total PSA or free PSA. 5. The method for analyzing PSA according to claim 1, wherein the lectin is Trichosanthes japonica agglutinin-II or Wisteria floribunda agglutinin. 6. The method for analyzing PSA according to claim 1, wherein the lectin further has an affinity for fucose α(1, 2) galactose residues. 7. The method for analyzing PSA according to claim 1, wherein the sample is obtained from a patient suspected of having prostate cancer. 8. The method for analyzing PSA according to claim 1, for diagnoses of the prostate cancer. 9. A method for analyzing PSA, characterized in that a lectin having an affinity for fucose α(1, 2) galactose residues is brought into contact with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectin. 10. The method for analyzing PSA according to claim 9, comprising the steps of (a) bringing into contact the lectin having an affinity for fucose α(1, 2) galactose residues with the sample possibly containing PSA, to separate PSA having an affinity for the lectin from PSA lacking an affinity for the lectin; and (b) determining the amount of PSA having an affinity for the lectin. 11. A method for analyzing PSA, characterized in that a lectin having an affinity for β-N-acetylgalactosamine residues and a lectin having an affinity for fucose α(1, 2) galactose residues are brought into contact with a sample possibly containing PSA, to determine an amount of PSA having an affinity for the lectins. 12. The method for analyzing PSA according to claim 11, comprising the steps of (a) bringing the lectin having an affinity for β-N-acetylgalactosamine residues and the lectin having an affinity for fucose α(1, 2) galactose residues into contact with the sample possibly containing PSA, to separate PSA having an affinity for the lectins from PSA lacking an affinity for the lectins; and (b) determining the amount of PSA having an affinity for the lectins. 13. A method for distinguishing prostate cancer from prostatic hypertrophy, characterized in that the amount of PSA having an affinity for a lectin in a sample is analyzed by the method for analyzing PSA according to claim 1. 14. An analysis kit of PSA, comprising a lectin having an affinity for β-N-acetylgalactosamine residues and an anti-PSA antibody. 15. (canceled) 16. The analysis kit of PSA according to claim 14, wherein the lectin is Trichosanthes japonica agglutinin-II or Wisteria floribunda agglutinin. 17. The analysis kit of PSA according to claim 14, wherein the lectin further has an affinity for fucose α(1, 2) galactose residues. 18. An analysis kit of PSA, comprising a lectin having an affinity for fucose α(1, 2) galactose residues and an anti-PSA antibody. 19. An analysis kit of PSA, comprising a lectin having an affinity for β-N-acetylgalactosamine residues, and a lectin having an affinity for fucose α(1, 2) galactose residues and an anti-PSA antibody. 20. A PSA having β-N-acetylgalactosamine residues. 21. A PSA having fucose α(1, 2) galactose residues. 22. A PSA having β-N-acetylgalactosamine residues and fucose α(1, 2) galactose residues.
1,600
213
213
14,139,519
1,645
A Clostridial toxin pharmaceutical composition comprising a Clostridial toxin, such as a botulinum toxin, wherein the Clostridial toxin present in the pharmaceutical composition is stabilized by a non-protein excipient such as a polyvinylpyrrolidone, a disaccharides, a trisaccharide, a polysaccharide, an alcohol, a metal, an amino acid, a surfactant and/or a polyethylene glycol.
1-8. (canceled) 9. A lyophilized or vacuum dried pharmaceutical composition comprising: (a) a botulinum toxin, wherein the botulinum toxin is not stabilized by a protein excipient, (b) polyvinylpyrrolidone, and (c) a disaccharide, wherein the potency of the botulinum toxin after reconstitution is at least 40% of the theoretical maximum potency of the botulinum toxin prior to lypophilization. 10. The pharmaceutical composition of claim 9, wherein the botulinum toxin is selected from the group consisting of the botulinum toxins types A, B, C, D, E, F, and G. 11. The pharmaceutical composition of claim 9, wherein the botulinum toxin is a botulinum toxin type A. 12. A pharmaceutical composition according to claim 9, wherein the potency of the botulinum toxin after reconstitution is at least 50% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 13. A pharmaceutical composition according to claim 12, wherein the potency of the botulinum toxin after reconstitution is at least 60% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 14. A pharmaceutical composition according to claim 13, wherein the potency of the botulinum toxin after reconstitution is at least 70% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 15. A lyophilized or vacuum dried pharmaceutical composition comprising: (a) a botulinum toxin, wherein the botulinum toxin is not stabilized by a protein excipient, (b) a compound selected from the group consisting of a first monosaccharide, a first disaccharide, a first trisaccharide, and a first alcohol made by reducing the first monosaccharide, and; (c) a compound selected from the group of compounds consisting of a second monosaccharide, a second disaccharide, a second trisaccharide, a metal, a second alcohol, and an amino acid, wherein the second monosaccharide, the second disaccharide and the second trisaccharide are different from respectively the first monosaccharide, the first disaccharide, and the first trisaccharide, wherein the potency of the botulinum toxin after reconstitution is at least 40% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 16. A lyophilized or vacuum dried pharmaceutical composition comprising: (a) a botulinum toxin, wherein the botulinum toxin is not stabilized by a protein excipient, (b) a polyethylene glycol, and; (c) a compound selected from the group of compounds consisting of a monosaccharide, a disaccharide, a trisaccharide, a metal, and an amino acid, wherein the potency of the botulinum toxin after reconstitution is at least 20% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 17. A pharmaceutical composition according to claim 16, wherein the potency of the botulinum toxin after reconstitution is at least 30% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 18. A pharmaceutical composition according to claim 17, wherein the potency of the botulinum toxin after reconstitution is at least 40% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 19. A pharmaceutical composition according to claim 18, wherein the potency of the botulinum toxin after reconstitution is at least 50% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 20. A pharmaceutical composition according to claim 19, wherein the potency of the botulinum toxin after reconstitution is at least 60% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 21. A pharmaceutical composition according to claim 20, wherein the potency of the botulinum toxin after reconstitution is at least 70% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 22. An animal protein free botulinum toxin pharmaceutical composition comprising: a botulinum toxin, a first compound selected from the group consisting of a first monosaccharide, a first disaccharide, and a first trisaccharide, and a second compound which is an amino acid, 23. A pharmaceutical composition according to claim 22, wherein the amino acid is methionine.
A Clostridial toxin pharmaceutical composition comprising a Clostridial toxin, such as a botulinum toxin, wherein the Clostridial toxin present in the pharmaceutical composition is stabilized by a non-protein excipient such as a polyvinylpyrrolidone, a disaccharides, a trisaccharide, a polysaccharide, an alcohol, a metal, an amino acid, a surfactant and/or a polyethylene glycol.1-8. (canceled) 9. A lyophilized or vacuum dried pharmaceutical composition comprising: (a) a botulinum toxin, wherein the botulinum toxin is not stabilized by a protein excipient, (b) polyvinylpyrrolidone, and (c) a disaccharide, wherein the potency of the botulinum toxin after reconstitution is at least 40% of the theoretical maximum potency of the botulinum toxin prior to lypophilization. 10. The pharmaceutical composition of claim 9, wherein the botulinum toxin is selected from the group consisting of the botulinum toxins types A, B, C, D, E, F, and G. 11. The pharmaceutical composition of claim 9, wherein the botulinum toxin is a botulinum toxin type A. 12. A pharmaceutical composition according to claim 9, wherein the potency of the botulinum toxin after reconstitution is at least 50% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 13. A pharmaceutical composition according to claim 12, wherein the potency of the botulinum toxin after reconstitution is at least 60% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 14. A pharmaceutical composition according to claim 13, wherein the potency of the botulinum toxin after reconstitution is at least 70% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 15. A lyophilized or vacuum dried pharmaceutical composition comprising: (a) a botulinum toxin, wherein the botulinum toxin is not stabilized by a protein excipient, (b) a compound selected from the group consisting of a first monosaccharide, a first disaccharide, a first trisaccharide, and a first alcohol made by reducing the first monosaccharide, and; (c) a compound selected from the group of compounds consisting of a second monosaccharide, a second disaccharide, a second trisaccharide, a metal, a second alcohol, and an amino acid, wherein the second monosaccharide, the second disaccharide and the second trisaccharide are different from respectively the first monosaccharide, the first disaccharide, and the first trisaccharide, wherein the potency of the botulinum toxin after reconstitution is at least 40% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 16. A lyophilized or vacuum dried pharmaceutical composition comprising: (a) a botulinum toxin, wherein the botulinum toxin is not stabilized by a protein excipient, (b) a polyethylene glycol, and; (c) a compound selected from the group of compounds consisting of a monosaccharide, a disaccharide, a trisaccharide, a metal, and an amino acid, wherein the potency of the botulinum toxin after reconstitution is at least 20% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 17. A pharmaceutical composition according to claim 16, wherein the potency of the botulinum toxin after reconstitution is at least 30% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 18. A pharmaceutical composition according to claim 17, wherein the potency of the botulinum toxin after reconstitution is at least 40% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 19. A pharmaceutical composition according to claim 18, wherein the potency of the botulinum toxin after reconstitution is at least 50% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 20. A pharmaceutical composition according to claim 19, wherein the potency of the botulinum toxin after reconstitution is at least 60% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 21. A pharmaceutical composition according to claim 20, wherein the potency of the botulinum toxin after reconstitution is at least 70% of the theoretical maximum potency of the botulinum toxin prior to lyophilization. 22. An animal protein free botulinum toxin pharmaceutical composition comprising: a botulinum toxin, a first compound selected from the group consisting of a first monosaccharide, a first disaccharide, and a first trisaccharide, and a second compound which is an amino acid, 23. A pharmaceutical composition according to claim 22, wherein the amino acid is methionine.
1,600
214
214
10,337,661
1,627
The present invention is a cosmetic or dermatological preparation or method of applying a cosmetic or dermatological preparation for the treatment or prophylaxis of skin damage or a skin condition, wherein the preparation includes creatine or a creatine derivative. The use of creatine or creatine derivatives in the cosmetic or dermatological preparations of the invention not only provides effective protection of the skin but also protects the preparations or the constituents of the preparations against harmful oxidation processes.
1. A method for the treatment or prophylaxis of the symptoms of skin damage or a skin condition, comprising applying a cosmetic or dermatological preparation comprising one or more compounds selected from the group consisting of creatine and creatine derivatives to the skin damage or skin condition. 2. The method as claimed in claim 1, said method for the treatment or prophylaxis of one or more manifestations from the group consisting of deficient, sensitive or hypoactive skin state or deficient, sensitive or hypoactive states of skin appendages; degenerative symptoms of the skin or skin appendages; environmentally induced or light-induced negative changes in the skin or the skin appendages; light-induced skin damage; pigmentation disorders; itching; horny layer barrier disorders; hair loss; inflammatory skin conditions; atopic eczema; seborrheic eczema; polymorphous photodermatosis; psoriasis; vitiligo; sensitive or irritated skin; deficiencies in the synthesis of collagen, hyaluronic acid and elastin; and deficiencies in intracellular DNA synthesis. 3. The method as claimed in claim 2, said method for the treatment or prophylaxis of polymorphous photodermatosis. 4. The method as claimed in claim 2, said method for the treatment or prophylaxis of environmentally-induced negative changes to the skin and the skin appendages caused by smoke, smog, reactive oxygen species and free radicals. 5. The method as claimed in claim 2, said method for the treatment or prophylaxis of itching. 6. The method as claimed in claim 2, said method for the treatment or prophylaxis of hair loss. 7. The method as claimed in claim 2, said method for the treatment or prophylaxis of deficiencies in the synthesis of one or more of collagen, hyaluronic acid and elastin. 8. The method as claimed in claim 2, said method for the treatment or prophylaxis of deficiencies in intracellular DNA synthesis. 9. The method as claimed in claim 2, said method for the treatment or prophylaxis of vitiligo. 10. The method as claimed in claim 1, wherein the preparation comprises a creatine derivative selected from the group consisting of creatine phosphate, creatine sulfate, creatine acetate, creatine ascorbate and esters of creatine and mono- or polyfunctional alcohols. 11. The method as claimed in claim 10, wherein the creatine derivative is creatine phosphate. 12. The method as claimed in claim 1, wherein the one or more compounds in the preparation selected from the group consisting of creatine and creatine derivatives are present in an amount from 0.001-0.49% by weight, based on the total weight of the preparation. 13. The method as claimed in claim 1, wherein the one or more compounds in the preparation selected from the group consisting of creatine and creatine derivatives are present in an amount from 10.1-30% by weight, based on the total weight of the preparation. 14. The method as claimed in claim 1, wherein the preparation further comprises one or more natural active ingredients and derivatives thereof selected from the group consisting of alpha-lipoic acid, phytoene, D-biotin, coenzyme Q10, alpha-glucosylrutin, carnitine, carnosine, isoflavone and taurine. 15. A cosmetic or dermatological preparation comprising one or more compounds selected from the group consisting of creatine and creatine derivatives for the treatment or prophylaxis of skin damage or a skin condition. 16. The preparation as claimed in claim 15, comprising a creatine derivative wherein the creatine derivative is selected from the group consisting of creatine phosphate, creatine sulfate, creatine acetate, creatine ascorbate and esters of creatine and mono- or polyfunctional alcohols. 17. The preparation as claimed in claim 16, wherein the creatine derivative is creatine phosphate. 18. The preparation as claimed in claim 15, wherein one or more compounds selected from the group consisting of creatine and creatine derivatives are present in an amount from 0.001-0.49% by weight, based on the total weight of the preparation. 19. The preparation as claimed in claim 15, wherein one or more compounds selected from the group consisting of creatine and creatine derivatives are present in an amount from 10.1-30% by weight, based on the total weight of the preparation. 20. The preparation as claimed in claim 15, wherein the preparation further comprises one or more natural active ingredients and derivatives thereof selected from the group consisting of alpha-lipoic acid, phytoene, D-biotin, coenzyme Q10, alpha-glucosylrutin, carnitine, carnosine, isoflavone and taurine.
The present invention is a cosmetic or dermatological preparation or method of applying a cosmetic or dermatological preparation for the treatment or prophylaxis of skin damage or a skin condition, wherein the preparation includes creatine or a creatine derivative. The use of creatine or creatine derivatives in the cosmetic or dermatological preparations of the invention not only provides effective protection of the skin but also protects the preparations or the constituents of the preparations against harmful oxidation processes.1. A method for the treatment or prophylaxis of the symptoms of skin damage or a skin condition, comprising applying a cosmetic or dermatological preparation comprising one or more compounds selected from the group consisting of creatine and creatine derivatives to the skin damage or skin condition. 2. The method as claimed in claim 1, said method for the treatment or prophylaxis of one or more manifestations from the group consisting of deficient, sensitive or hypoactive skin state or deficient, sensitive or hypoactive states of skin appendages; degenerative symptoms of the skin or skin appendages; environmentally induced or light-induced negative changes in the skin or the skin appendages; light-induced skin damage; pigmentation disorders; itching; horny layer barrier disorders; hair loss; inflammatory skin conditions; atopic eczema; seborrheic eczema; polymorphous photodermatosis; psoriasis; vitiligo; sensitive or irritated skin; deficiencies in the synthesis of collagen, hyaluronic acid and elastin; and deficiencies in intracellular DNA synthesis. 3. The method as claimed in claim 2, said method for the treatment or prophylaxis of polymorphous photodermatosis. 4. The method as claimed in claim 2, said method for the treatment or prophylaxis of environmentally-induced negative changes to the skin and the skin appendages caused by smoke, smog, reactive oxygen species and free radicals. 5. The method as claimed in claim 2, said method for the treatment or prophylaxis of itching. 6. The method as claimed in claim 2, said method for the treatment or prophylaxis of hair loss. 7. The method as claimed in claim 2, said method for the treatment or prophylaxis of deficiencies in the synthesis of one or more of collagen, hyaluronic acid and elastin. 8. The method as claimed in claim 2, said method for the treatment or prophylaxis of deficiencies in intracellular DNA synthesis. 9. The method as claimed in claim 2, said method for the treatment or prophylaxis of vitiligo. 10. The method as claimed in claim 1, wherein the preparation comprises a creatine derivative selected from the group consisting of creatine phosphate, creatine sulfate, creatine acetate, creatine ascorbate and esters of creatine and mono- or polyfunctional alcohols. 11. The method as claimed in claim 10, wherein the creatine derivative is creatine phosphate. 12. The method as claimed in claim 1, wherein the one or more compounds in the preparation selected from the group consisting of creatine and creatine derivatives are present in an amount from 0.001-0.49% by weight, based on the total weight of the preparation. 13. The method as claimed in claim 1, wherein the one or more compounds in the preparation selected from the group consisting of creatine and creatine derivatives are present in an amount from 10.1-30% by weight, based on the total weight of the preparation. 14. The method as claimed in claim 1, wherein the preparation further comprises one or more natural active ingredients and derivatives thereof selected from the group consisting of alpha-lipoic acid, phytoene, D-biotin, coenzyme Q10, alpha-glucosylrutin, carnitine, carnosine, isoflavone and taurine. 15. A cosmetic or dermatological preparation comprising one or more compounds selected from the group consisting of creatine and creatine derivatives for the treatment or prophylaxis of skin damage or a skin condition. 16. The preparation as claimed in claim 15, comprising a creatine derivative wherein the creatine derivative is selected from the group consisting of creatine phosphate, creatine sulfate, creatine acetate, creatine ascorbate and esters of creatine and mono- or polyfunctional alcohols. 17. The preparation as claimed in claim 16, wherein the creatine derivative is creatine phosphate. 18. The preparation as claimed in claim 15, wherein one or more compounds selected from the group consisting of creatine and creatine derivatives are present in an amount from 0.001-0.49% by weight, based on the total weight of the preparation. 19. The preparation as claimed in claim 15, wherein one or more compounds selected from the group consisting of creatine and creatine derivatives are present in an amount from 10.1-30% by weight, based on the total weight of the preparation. 20. The preparation as claimed in claim 15, wherein the preparation further comprises one or more natural active ingredients and derivatives thereof selected from the group consisting of alpha-lipoic acid, phytoene, D-biotin, coenzyme Q10, alpha-glucosylrutin, carnitine, carnosine, isoflavone and taurine.
1,600
215
215
14,772,961
1,611
Suggested is a cosmetic composition, comprising (a) at least one acetophenone derivative or a cosmetically or pharmaceutically acceptable salt thereof in a working amount of from 0.1 to 0.5 b.w.—calculated on the total composition, (b) at least one oil body or wax, and/or (c) at least one emulsifier and optionally (d) at least one active principle. The compositions show improved emulsion stability.
1. A cosmetic composition, comprising (a) at least one acetophenone derivative of formula (I) in which R1 stands for hydrogen or methyl, and R2 stands for hydrogen, hydroxyl or a —OCH3 group, or a cosmetically or pharmaceutically acceptable salt thereof in a working amount of from 0.1 to 0.5% b.w.—calculated on the total composition, (b) at least one oil body or wax, and/or (c) at least one emulsifier and optionally (d) at least one active principle. 2. The composition of claim 1, wherein the acetophenone derivatives selected from the group consisting of and mixtures thereof. 3. The composition of claim 2 comprising or consisting of t three of the compounds (Ia), (Ib) and (Ic). 4. The composition of claim 1, wherein the acetophenone derivatives are present in amounts of from about 0.1 to about 5% b.w.—calculated on the final composition. 5. The composition of claim 1, wherein the oil bodies (component b1) are selected from the group consisting of Guerbet alcohols based on fatty alcohols having 6 to 18 carbon atoms, esters of linear C6-C22-fatty acids with linear or branched C6-C22-fatty alcohols or esters of branched C6-C13-carboxylic acids with linear or branched C6-C22-fatty alcohols, esters of linear C6-C22-fatty acids with branched alcohols, esters of C18-C38-alkylhydroxy carboxylic acids with linear or branched C6-C22-fatty alcohols, esters of linear and/or branched fatty acids with polyhydric alcohols and/or Guerbet alcohols, triglycerides based on C6-C10-fatty acids, liquid mono-di-/triglyceride mixtures based on C6-C18-fatty acids, esters of C6-C22-fatty alcohols and/or Guerbet alcohols with aromatic carboxylic acids, esters of C2-C12-dicarboxylic acids with linear or branched alcohols having 1 to 22 carbon atoms or polyols having 2 to 10 carbon atoms and 2 to 6 hydroxyl groups, vegetable oils, branched primary alcohols, substituted cyclohexanes, linear and branched C6-C22-fatty alcohol carbonates, Guerbet carbonates, based on fatty alcohols having 6 to 18, preferably 8 to 10, carbon atoms, esters of benzoic acid with linear and/or branched C6-C22-alcohols, linear or branched, symmetrical or asymmetrical dialkyl ethers having 6 to 22 carbon atoms per alkyl group, ring-opening products of epoxidized fatty acid esters with polyols, silicone oils and/or aliphatic or naphthenic hydrocarbons, mineral oils and mixtures thereof. 6. The composition of claim 1, wherein the waxes (component b2) are selected from the group consisting of super fatting agents, consistency factors, pearlising waxes, natural waxes and mixtures thereof. 7. The composition of claim 1, wherein the emulsifiers (component c) are selected from the group consisting of non-ionic, anionic, cationic, or amphoteric emulsifiers and mixtures thereof. 8. The composition of claim 1, wherein the active principles (component d) are selected from the group consisting of abrasives, anti-acne agents, agents against ageing of the skin, anti-cellulitis agents, antidandruff agents, anti-inflammatory agents, irritation-preventing agents, irritation-inhibiting agents, antioxidants, astringents, perspiration-inhibiting agents, antiseptic agents, ant-statics, binders, buffers, carrier materials, chelating agents, cell stimulants, cleansing agents, care agents, depilatory agents, surface-active substances, deodorizing agents, antiperspirants, softeners, emulsifiers, enzymes, essential oils, fibres, film-forming agents, fixatives, foam-forming agents, foam stabilizers, substances for preventing foaming, foam boosters, gelling agents, gel-forming agents, hair care agents, hair-setting agents, hair-straightening agents, moisture-donating agents, moisturizing substances, moisture-retaining substances, bleaching agents, strengthening agents, stain-removing agents, optically brightening agents, impregnating agents, dirt-repellent agents, friction-reducing agents, lubricants, moisturizing creams, ointments, opacifying agents, plasticizing agents, covering agents, polish, gloss agents, polymers, powders, proteins, re-oiling agents, abrading agents, silicones, skin-soothing agents, skin-cleansing agents, skin care agents, skin-healing agents, skin-lightening agents, skin-protecting agents, skin-softening agents, hair promotion agents, cooling agents, skin-cooling agents, warming agents, skin-warming agents, stabilizers, UV-absorbing agents, UV filters, detergents, fabric conditioning agents, suspending agents, skin-tanning agents, thickeners, vitamins, oils, waxes, fats, phospholipids, saturated fatty acids, mono- or polyunsaturated fatty acids, -hydroxy acids, polyhydroxyfatty acids, liquefiers, dyestuffs, colour-protecting agents, pigments, anti-corrosives, aromas, flavouring substances, odoriferous substances, polyols, surfactants, electrolytes, organic solvents or silicone derivatives. 9. The composition of claim 1, wherein they comprise the components in the following amounts: (a) about 0.1 to about 0.5% b.w. acetophenone derivatives of formula (I); (b) about 99.9 to about 50% b.w. oil bodies and/or waxes; (c) 0.1 to about 25% b.w. emulsifiers; (d) 0 to about 25% b.w. active principles; on condition that the amounts add—optionally together with water and additional ingredients—to 100% b.w. 10. The composition of claim 1, wherein the product either contains water or is essentially free of water. 11. The composition of claim 1, wherein the product is an o/w or w/o or multiple o/w/o or w/o/w emulsion. 12. The composition of claim 1, wherein the product is a lotion, a cream or a stick. 13. A method of stabilizing an emulsion against separation, comprising the step of adding a working amount of at least one acetophenone derivative of formula (I) of claim 1 to the emulsion. 14. A method of stabilizing cosmetic compositions against separation, comprising the step of adding acetophenone derivatives according to formula (I) of claim 1 to a cosmetic composition as stabilizers against separation of the cosmetic composition. 15. A method of reducing the size of water or oil droplets in an emulsion, comprising the step of adding a working amount of at least one acetophenone derivative of formula (I) of claim 1 to the emulsion. 16. A method of reducing the size of water or oil droplets in an emulsion, comprising the step of adding acetophenone derivatives according to formula (I) of claim 1 to the emulsion. 17. A method of improving foam stability and viscosity of shampoos, comprising the step of adding a working amount of at least one acetophenone derivative of formula (I) of claim 1 to a shampoo. 18. A method of improving foam stability and viscosity of shampoos, comprising the step of adding acetophenone derivatives of formula (I) of claim 1 to a shampoo.
Suggested is a cosmetic composition, comprising (a) at least one acetophenone derivative or a cosmetically or pharmaceutically acceptable salt thereof in a working amount of from 0.1 to 0.5 b.w.—calculated on the total composition, (b) at least one oil body or wax, and/or (c) at least one emulsifier and optionally (d) at least one active principle. The compositions show improved emulsion stability.1. A cosmetic composition, comprising (a) at least one acetophenone derivative of formula (I) in which R1 stands for hydrogen or methyl, and R2 stands for hydrogen, hydroxyl or a —OCH3 group, or a cosmetically or pharmaceutically acceptable salt thereof in a working amount of from 0.1 to 0.5% b.w.—calculated on the total composition, (b) at least one oil body or wax, and/or (c) at least one emulsifier and optionally (d) at least one active principle. 2. The composition of claim 1, wherein the acetophenone derivatives selected from the group consisting of and mixtures thereof. 3. The composition of claim 2 comprising or consisting of t three of the compounds (Ia), (Ib) and (Ic). 4. The composition of claim 1, wherein the acetophenone derivatives are present in amounts of from about 0.1 to about 5% b.w.—calculated on the final composition. 5. The composition of claim 1, wherein the oil bodies (component b1) are selected from the group consisting of Guerbet alcohols based on fatty alcohols having 6 to 18 carbon atoms, esters of linear C6-C22-fatty acids with linear or branched C6-C22-fatty alcohols or esters of branched C6-C13-carboxylic acids with linear or branched C6-C22-fatty alcohols, esters of linear C6-C22-fatty acids with branched alcohols, esters of C18-C38-alkylhydroxy carboxylic acids with linear or branched C6-C22-fatty alcohols, esters of linear and/or branched fatty acids with polyhydric alcohols and/or Guerbet alcohols, triglycerides based on C6-C10-fatty acids, liquid mono-di-/triglyceride mixtures based on C6-C18-fatty acids, esters of C6-C22-fatty alcohols and/or Guerbet alcohols with aromatic carboxylic acids, esters of C2-C12-dicarboxylic acids with linear or branched alcohols having 1 to 22 carbon atoms or polyols having 2 to 10 carbon atoms and 2 to 6 hydroxyl groups, vegetable oils, branched primary alcohols, substituted cyclohexanes, linear and branched C6-C22-fatty alcohol carbonates, Guerbet carbonates, based on fatty alcohols having 6 to 18, preferably 8 to 10, carbon atoms, esters of benzoic acid with linear and/or branched C6-C22-alcohols, linear or branched, symmetrical or asymmetrical dialkyl ethers having 6 to 22 carbon atoms per alkyl group, ring-opening products of epoxidized fatty acid esters with polyols, silicone oils and/or aliphatic or naphthenic hydrocarbons, mineral oils and mixtures thereof. 6. The composition of claim 1, wherein the waxes (component b2) are selected from the group consisting of super fatting agents, consistency factors, pearlising waxes, natural waxes and mixtures thereof. 7. The composition of claim 1, wherein the emulsifiers (component c) are selected from the group consisting of non-ionic, anionic, cationic, or amphoteric emulsifiers and mixtures thereof. 8. The composition of claim 1, wherein the active principles (component d) are selected from the group consisting of abrasives, anti-acne agents, agents against ageing of the skin, anti-cellulitis agents, antidandruff agents, anti-inflammatory agents, irritation-preventing agents, irritation-inhibiting agents, antioxidants, astringents, perspiration-inhibiting agents, antiseptic agents, ant-statics, binders, buffers, carrier materials, chelating agents, cell stimulants, cleansing agents, care agents, depilatory agents, surface-active substances, deodorizing agents, antiperspirants, softeners, emulsifiers, enzymes, essential oils, fibres, film-forming agents, fixatives, foam-forming agents, foam stabilizers, substances for preventing foaming, foam boosters, gelling agents, gel-forming agents, hair care agents, hair-setting agents, hair-straightening agents, moisture-donating agents, moisturizing substances, moisture-retaining substances, bleaching agents, strengthening agents, stain-removing agents, optically brightening agents, impregnating agents, dirt-repellent agents, friction-reducing agents, lubricants, moisturizing creams, ointments, opacifying agents, plasticizing agents, covering agents, polish, gloss agents, polymers, powders, proteins, re-oiling agents, abrading agents, silicones, skin-soothing agents, skin-cleansing agents, skin care agents, skin-healing agents, skin-lightening agents, skin-protecting agents, skin-softening agents, hair promotion agents, cooling agents, skin-cooling agents, warming agents, skin-warming agents, stabilizers, UV-absorbing agents, UV filters, detergents, fabric conditioning agents, suspending agents, skin-tanning agents, thickeners, vitamins, oils, waxes, fats, phospholipids, saturated fatty acids, mono- or polyunsaturated fatty acids, -hydroxy acids, polyhydroxyfatty acids, liquefiers, dyestuffs, colour-protecting agents, pigments, anti-corrosives, aromas, flavouring substances, odoriferous substances, polyols, surfactants, electrolytes, organic solvents or silicone derivatives. 9. The composition of claim 1, wherein they comprise the components in the following amounts: (a) about 0.1 to about 0.5% b.w. acetophenone derivatives of formula (I); (b) about 99.9 to about 50% b.w. oil bodies and/or waxes; (c) 0.1 to about 25% b.w. emulsifiers; (d) 0 to about 25% b.w. active principles; on condition that the amounts add—optionally together with water and additional ingredients—to 100% b.w. 10. The composition of claim 1, wherein the product either contains water or is essentially free of water. 11. The composition of claim 1, wherein the product is an o/w or w/o or multiple o/w/o or w/o/w emulsion. 12. The composition of claim 1, wherein the product is a lotion, a cream or a stick. 13. A method of stabilizing an emulsion against separation, comprising the step of adding a working amount of at least one acetophenone derivative of formula (I) of claim 1 to the emulsion. 14. A method of stabilizing cosmetic compositions against separation, comprising the step of adding acetophenone derivatives according to formula (I) of claim 1 to a cosmetic composition as stabilizers against separation of the cosmetic composition. 15. A method of reducing the size of water or oil droplets in an emulsion, comprising the step of adding a working amount of at least one acetophenone derivative of formula (I) of claim 1 to the emulsion. 16. A method of reducing the size of water or oil droplets in an emulsion, comprising the step of adding acetophenone derivatives according to formula (I) of claim 1 to the emulsion. 17. A method of improving foam stability and viscosity of shampoos, comprising the step of adding a working amount of at least one acetophenone derivative of formula (I) of claim 1 to a shampoo. 18. A method of improving foam stability and viscosity of shampoos, comprising the step of adding acetophenone derivatives of formula (I) of claim 1 to a shampoo.
1,600
216
216
12,866,786
1,612
This invention relates to methods of treating early enamel lesions comprising applying an effective amount of a basic amino acid in free or salt form, together with fluoride to a patient in need thereof.
1. A method of treating early enamel caries comprising: applying an effective amount of a dentifrice composition to the oral cavity, the dentifrice composition having a base formulation comprising calcium carbonate, the dentifrice composition further comprising arginine, in free or salt form, and an effective amount of a fluoride ion source, wherein the arginine is provided in the amount of from 2 to 10 wt % based on the total weight of the dentifrice composition. 2. (canceled) 3. The method of claim 1 wherein the early enamel caries are detected by quantitative light-induced fluorescence (QLF) or electrical caries monitoring (ECM). 4. A method to improve the quantitative light-induced fluorescence (QLF) or electrical caries monitoring (ECM) value, correlating to early enamel lesions, comprising: applying an effective amount of a dentifrice composition to the oral cavity, the dentifrice composition having a base formulation comprising calcium carbonate, the dentifrice composition further comprising arginine, in free or salt form, and an effective amount of a fluoride ion source, wherein the arginine is provided in the amount of from 2 to 10 wt % based on the total weight of the dentifrice composition. 5-6. (canceled) 7. A method to reduce the size of existing early enamel lesions, comprising: applying an effective amount of a dentifrice composition to the oral cavity, the dentifrice composition having a base formulation comprising calcium carbonate, the dentifrice composition further comprising arginine, in free or salt form, and an effective amount of a fluoride ion source, wherein the arginine is provided in an amount of from 2 to 10 wt % based on the total weight of the dentifrice composition. 8-10. (canceled) 11. The method of claim 1 which is additionally effective to a. inhibit formation of dental caries, b. reduce or inhibit demineralization and promote remineralization of the teeth, c. reduce hypersensitivity of the teeth, d. reduce or inhibit gingivitis, e. promote healing of sores or cuts in the mouth, f. reduce levels of acid producing bacteria, g. increase relative levels of arginolytic bacteria, h. inhibit microbial biofilm formation in the oral cavi i. raise and/or maintain plaque pH at levels of at least about pH 5.5 following sugar challenge, j. reduce plaque accumulation, k. treat, relieve or reduce dry mouth, l. whiten teeth, m. promote systemic health, including cardiovascular health, n. reduce erosion of the teeth, o. immunize or protect the teeth against cariogenic bacteria, p. protect against cavities, and/or q. clean the teeth and oral cavity. 12-18. (canceled) 19. The method of claim 1, wherein the fluoride ion source is selected from stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 20. The method of claim 1, wherein the dentifrice composition further comprises a potassium ion source. 21. The method of claim 20, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 22. The method of claim 4, wherein the fluoride ion source is selected from stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 23. The method of claim 22, wherein the dentifrice composition further comprises a potassium ion source. 24. The method of claim 23, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 25. The method of claim 7, wherein the fluoride ion source is selected from stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 26. The method of claim 25, wherein the dentifrice composition further comprises a potassium ion source. 27. The method of claim 26, wherein said potassium ion source is selected from potassium nitrate and potassium chloride.
This invention relates to methods of treating early enamel lesions comprising applying an effective amount of a basic amino acid in free or salt form, together with fluoride to a patient in need thereof.1. A method of treating early enamel caries comprising: applying an effective amount of a dentifrice composition to the oral cavity, the dentifrice composition having a base formulation comprising calcium carbonate, the dentifrice composition further comprising arginine, in free or salt form, and an effective amount of a fluoride ion source, wherein the arginine is provided in the amount of from 2 to 10 wt % based on the total weight of the dentifrice composition. 2. (canceled) 3. The method of claim 1 wherein the early enamel caries are detected by quantitative light-induced fluorescence (QLF) or electrical caries monitoring (ECM). 4. A method to improve the quantitative light-induced fluorescence (QLF) or electrical caries monitoring (ECM) value, correlating to early enamel lesions, comprising: applying an effective amount of a dentifrice composition to the oral cavity, the dentifrice composition having a base formulation comprising calcium carbonate, the dentifrice composition further comprising arginine, in free or salt form, and an effective amount of a fluoride ion source, wherein the arginine is provided in the amount of from 2 to 10 wt % based on the total weight of the dentifrice composition. 5-6. (canceled) 7. A method to reduce the size of existing early enamel lesions, comprising: applying an effective amount of a dentifrice composition to the oral cavity, the dentifrice composition having a base formulation comprising calcium carbonate, the dentifrice composition further comprising arginine, in free or salt form, and an effective amount of a fluoride ion source, wherein the arginine is provided in an amount of from 2 to 10 wt % based on the total weight of the dentifrice composition. 8-10. (canceled) 11. The method of claim 1 which is additionally effective to a. inhibit formation of dental caries, b. reduce or inhibit demineralization and promote remineralization of the teeth, c. reduce hypersensitivity of the teeth, d. reduce or inhibit gingivitis, e. promote healing of sores or cuts in the mouth, f. reduce levels of acid producing bacteria, g. increase relative levels of arginolytic bacteria, h. inhibit microbial biofilm formation in the oral cavi i. raise and/or maintain plaque pH at levels of at least about pH 5.5 following sugar challenge, j. reduce plaque accumulation, k. treat, relieve or reduce dry mouth, l. whiten teeth, m. promote systemic health, including cardiovascular health, n. reduce erosion of the teeth, o. immunize or protect the teeth against cariogenic bacteria, p. protect against cavities, and/or q. clean the teeth and oral cavity. 12-18. (canceled) 19. The method of claim 1, wherein the fluoride ion source is selected from stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 20. The method of claim 1, wherein the dentifrice composition further comprises a potassium ion source. 21. The method of claim 20, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 22. The method of claim 4, wherein the fluoride ion source is selected from stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 23. The method of claim 22, wherein the dentifrice composition further comprises a potassium ion source. 24. The method of claim 23, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 25. The method of claim 7, wherein the fluoride ion source is selected from stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 26. The method of claim 25, wherein the dentifrice composition further comprises a potassium ion source. 27. The method of claim 26, wherein said potassium ion source is selected from potassium nitrate and potassium chloride.
1,600
217
217
15,204,826
1,617
The present invention relates to a methods of preparing active compounds complexed with lipids using aqueous systems that are free of organic solvents, and methods of using the complexes, e.g., in treating a disease in a subject. In some embodiments, the present invention comprises a composition comprising a complex comprising at least one active compound, e.g., a polyene antibiotic, an immunosuppressant agent such as tacrolimus or a taxane or taxane derivative, and one or more lipids. In some embodiments, the present invention provides a method comprising preparing a composition comprising a lipid complex comprising at least one active compound and at least one lipid and administering the composition to a subject. In certain embodiments the subject is a mammal. In certain preferred embodiments, the subject is human.
1. A method of treating a disease in a subject, comprising: a) using an aqueous system to prepare a composition comprising a complex, said complex comprising at least one active compound and at least one lipid; and b) administering said composition to a subject. 2. The method of claim 1, wherein said complex comprises a lipid compound suspension and wherein said aqueous system comprises a process comprising: a) preparing a suspension comprising said at least one active compound and said at least one lipid in a first aqueous medium at a pH between about pH 4.0 and pH 8.0; b) treating said suspension to form a lipid-compound suspension of defined particle size; c) lyophilizing the lipid-compound suspension of defined particle size to form lyophilized material; and d) reconstituting said lyophilized material with a second aqueous medium to obtain a suspension of lipid formulation of defined particle size, said defined particle size having a mean particle size of less than 5 microns. 3. The method of claim 1, wherein said at least one active compound is selected from the group consisting of amphotericin-B with deoxycholate, amphotericin B without deoxycholate, docetaxel, paclitaxel, tacrolimus, doxorubicin, Epirubicin, anthracyclines, and etoposide. 4. The method of claim 1, wherein said at least one lipid is selected from the group consisting of egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), soy phosphatidylcholine (SPC), hydrogenated soy phosphatidylcholine (HSPC), dimyristoylphosphatidylcholine (DMPC), dimyristoylphosphatidylglycerol (DMPG), dipalmitoylphosohatidylcholine (DPPC), disteroylphosphatidylglycerol (DSPG), dipalmitoylphosphatidylglycerol (DMPG), cholesterol (Chol), cholesterol sulfate and its salts (CS), cholesterol hemisuccinate and its salts (Chems), cholesterol phosphate and its salts (CP), cholesterylphosphocholine and other hydroxycholesterol or amino cholesterol derivatives, cholesteryl succinate, cholesteryl oleate, polyethylene glycol derivatives of cholesterol (cholesterol-PEG), coprostanol, cholestanol, cholestane, cholic acid, cortisol, corticosterone, hydrocortisone, and calciferol, monoglycerides, diglycerides, triglycerides, carbohydrate-based lipids selected from a group consisting of galactolipid, mannolipid, galactolecithin, β-sitosterol, stigmasterol, stigmastanol, lanosterol, α-spinasterol, lathosterol, campesterol, phosphatidylcholine, phosphatidylglycerol, phosphatidylethanolamine, phosphatidylserine, phosphatdylinositol, phosphatidic acid, and pegylated derivatives of distearoylphosphatidylglycerol, dipalmitoylphosphatidylglycerol, dimyristoylphosphatidylglycerol, and dioleoylphosphatidylglycerol. 5. The method of claim 1, wherein said at least one lipid comprises one or more of fatty acids selected from a group consisting of saturated or unsaturated fatty acids. 6. The method of claim 1, wherein said composition further comprises polyethylene glycol. 7. The method of claim 1, wherein said at least one lipid is selected from the group consisting of cholesterol or cholesterol sulfate and salts thereof, cholesterol hemisuccinate and salts thereof, cholesterol phosphate and salts thereof, and wherein said composition further comprises at least one phospholipid. 8. The method of claim 1, wherein said at least one lipid comprises a cholesterol or cholesterol derivative, wherein the mole ratio of active compound to cholesterol or cholesterol derivative is between about 1:1 and 1:10. 9. The method of claim 1, wherein said at least one lipid comprises hydrogenated soy phosphatidylcholine or soy phosphatidylcholine, wherein the mole ratio of active compound and hydrogenated soy phosphatidylcholine or soy phosphatidylcholine is between about 1:1 to about 1:90. 10. The method of claim 1, wherein said composition comprises active compound at a concentration of from about 0.5 mg/mL to about 25 mg/mL. 11. The method of claim 1, wherein said composition comprises a total lipid concentration of from 2.5% by weight to about 95% by weight. 12. The method of claim 1, wherein the molar ratio of active compound to lipid in said composition is between 1:10 to 1:100. 13. The method of claim 1, wherein the weight-to-weight ratio of total active compound to total lipid in said composition is between 1:10 to 1:60. 14. The method of claim 1, wherein said composition comprises a form selected from the group consisting of powder, solution, suspension, emulsion, micelle, liposome, lipidic particle, gel, and paste form. 15. The method of claim 14, wherein said composition comprises a plurality of micelles, wherein said micelles are in the form of monomeric, dimeric, polymeric or mixture of micelles and vesicles. 16. The method of claim 1, wherein said preparing of a composition comprising a complex comprises preparing said complex in a lyophilized form. 17. The method of claim 16, wherein said preparing said complex in a lyophilized form comprises using a cryoprotectant, wherein said cryoprotectant comprises one or more sugars selected from a group consisting of trehalose, maltose, lactose, sucrose, glucose, and dextran. 18. The method of claim 1, wherein, said composition comprises a tablet or a filled capsule, and optionally comprises an enteric coating material. 19. The method of claim 1, wherein said active compound is a partially water soluble or water insoluble drug. 20. The method of claim 1, wherein said administering comprises oral, intravenous, subcutaneous, parenteral, intraperitoneal, rectal, vaginal, and/or topical delivery of said lipidic composition to said subject. 21. A process for preparing a lipid formulation of an active compound, wherein said process comprises using an aqueous system to prepare a composition comprising a complex, said complex comprising at least one active compound and at least one lipid. 22. The process of claim 21, wherein said process is a process for preparing a lipid formulation of defined particle size, wherein said process comprises: a) preparing a suspension comprising at least one active compound and at least one lipid in a first aqueous medium at a pH between about pH 4.0 and pH 8.0; b) treating said suspension to form a lipid-compound suspension of defined particle size; c) lyophilizing the lipid-compound suspension of defined particle size to form lyophilized material; and d) reconstituting said lyophilized material with a second aqueous medium to obtain a suspension of lipid formulation of defined particle size, said defined particle size having a mean particle size of less than 5 microns. 23. The process of claim 22, wherein said first aqueous medium is water. 24. The process of claim 22, wherein said first aqueous medium and said second aqueous medium are different. 25. The process of claim 22, wherein said treating said suspension comprises extruding said suspension through a selected size aperture. 26. The process of claim 22, wherein said treating said suspension comprises high pressure split homogenization. 27. The process of claim 22 wherein said lyophilizing is in the presence of a cryoprotectant. 28. The process of claim 21, wherein said active compound comprises an active compound selected from the group consisting of a polyene antibiotic, a macrolide, an anti-cancer drug, and an immunosuppressant. 29. The process of claim 21, wherein said active compound comprises a compound selected from the group consisting of docetaxel, paclitaxel, doxorubicin, epirubicin, tamoxifen, endoxifen, etoposide, anthracyclines, amphotericin B, tacrolimus, and sacrolimus. 30. The process of claim 21, wherein said at least one lipid is selected from the group consisting of egg phosphatidylcholine, egg phosphatidylglycerol, soy phosphatidylcholine, hydrogenated soy phosphatidylcholine, dimyristoylphosphatidylcholine, dimyristoylphosphatidylglycerol, dipalmitoylphosohatidylcholine, disteroylphosphatidylglycerol, dipalmitoylphosphatidylglycerol, cholesterol, cholesterol sulfate and its salts, cholesterol hemisuccinate and its salts, cholesterol phosphate and its salts, cholesterylphosphocholine and other hydroxycholesterol or amino cholesterol derivatives, cholesteryl succinate, cholesteryl oleate, polyethylene glycol derivatives of cholesterol (cholesterol-PEG), coprostanol, cholestanol, cholestane, cholic acid, cortisol, corticosterone, hydrocortisone, and calciferol. 31. The process of claim 21, wherein said lipid formulation comprises cholesterol sulfate, and wherein the molar ratio of active compound to cholesterol sulfate in said suspension is in between about 1:1 to about 1:10. 32. The process of claim 22, wherein the composition mean particle size upon reconstitution is about 10-5000 nm. 33. The process of claim 21, wherein said at least one active compound exhibits poor solubility in water, alcohols, and halogenated hydrocarbon solvents. 34. The process of claim 22, wherein said suspension of lipid formulation of defined particle size comprises a suspension of liposomes and/or lipidic particles. 35. A method treating a cell with a lipidic composition comprising at least one active agent and at least one lipid, comprising: a) using an aqueous system to prepare a composition comprising a complex, said complex comprising at least one active compound and at least one lipid; and b) exposing said cell to said lipidic composition. 36. The method of claim 35, wherein said exposing said cell comprises exposing said cell to said lipidic composition in vivo. 37. The method of claim 35, wherein said subject is a mammal. 38. The method of claim 37, wherein said mammal is human.
The present invention relates to a methods of preparing active compounds complexed with lipids using aqueous systems that are free of organic solvents, and methods of using the complexes, e.g., in treating a disease in a subject. In some embodiments, the present invention comprises a composition comprising a complex comprising at least one active compound, e.g., a polyene antibiotic, an immunosuppressant agent such as tacrolimus or a taxane or taxane derivative, and one or more lipids. In some embodiments, the present invention provides a method comprising preparing a composition comprising a lipid complex comprising at least one active compound and at least one lipid and administering the composition to a subject. In certain embodiments the subject is a mammal. In certain preferred embodiments, the subject is human.1. A method of treating a disease in a subject, comprising: a) using an aqueous system to prepare a composition comprising a complex, said complex comprising at least one active compound and at least one lipid; and b) administering said composition to a subject. 2. The method of claim 1, wherein said complex comprises a lipid compound suspension and wherein said aqueous system comprises a process comprising: a) preparing a suspension comprising said at least one active compound and said at least one lipid in a first aqueous medium at a pH between about pH 4.0 and pH 8.0; b) treating said suspension to form a lipid-compound suspension of defined particle size; c) lyophilizing the lipid-compound suspension of defined particle size to form lyophilized material; and d) reconstituting said lyophilized material with a second aqueous medium to obtain a suspension of lipid formulation of defined particle size, said defined particle size having a mean particle size of less than 5 microns. 3. The method of claim 1, wherein said at least one active compound is selected from the group consisting of amphotericin-B with deoxycholate, amphotericin B without deoxycholate, docetaxel, paclitaxel, tacrolimus, doxorubicin, Epirubicin, anthracyclines, and etoposide. 4. The method of claim 1, wherein said at least one lipid is selected from the group consisting of egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), soy phosphatidylcholine (SPC), hydrogenated soy phosphatidylcholine (HSPC), dimyristoylphosphatidylcholine (DMPC), dimyristoylphosphatidylglycerol (DMPG), dipalmitoylphosohatidylcholine (DPPC), disteroylphosphatidylglycerol (DSPG), dipalmitoylphosphatidylglycerol (DMPG), cholesterol (Chol), cholesterol sulfate and its salts (CS), cholesterol hemisuccinate and its salts (Chems), cholesterol phosphate and its salts (CP), cholesterylphosphocholine and other hydroxycholesterol or amino cholesterol derivatives, cholesteryl succinate, cholesteryl oleate, polyethylene glycol derivatives of cholesterol (cholesterol-PEG), coprostanol, cholestanol, cholestane, cholic acid, cortisol, corticosterone, hydrocortisone, and calciferol, monoglycerides, diglycerides, triglycerides, carbohydrate-based lipids selected from a group consisting of galactolipid, mannolipid, galactolecithin, β-sitosterol, stigmasterol, stigmastanol, lanosterol, α-spinasterol, lathosterol, campesterol, phosphatidylcholine, phosphatidylglycerol, phosphatidylethanolamine, phosphatidylserine, phosphatdylinositol, phosphatidic acid, and pegylated derivatives of distearoylphosphatidylglycerol, dipalmitoylphosphatidylglycerol, dimyristoylphosphatidylglycerol, and dioleoylphosphatidylglycerol. 5. The method of claim 1, wherein said at least one lipid comprises one or more of fatty acids selected from a group consisting of saturated or unsaturated fatty acids. 6. The method of claim 1, wherein said composition further comprises polyethylene glycol. 7. The method of claim 1, wherein said at least one lipid is selected from the group consisting of cholesterol or cholesterol sulfate and salts thereof, cholesterol hemisuccinate and salts thereof, cholesterol phosphate and salts thereof, and wherein said composition further comprises at least one phospholipid. 8. The method of claim 1, wherein said at least one lipid comprises a cholesterol or cholesterol derivative, wherein the mole ratio of active compound to cholesterol or cholesterol derivative is between about 1:1 and 1:10. 9. The method of claim 1, wherein said at least one lipid comprises hydrogenated soy phosphatidylcholine or soy phosphatidylcholine, wherein the mole ratio of active compound and hydrogenated soy phosphatidylcholine or soy phosphatidylcholine is between about 1:1 to about 1:90. 10. The method of claim 1, wherein said composition comprises active compound at a concentration of from about 0.5 mg/mL to about 25 mg/mL. 11. The method of claim 1, wherein said composition comprises a total lipid concentration of from 2.5% by weight to about 95% by weight. 12. The method of claim 1, wherein the molar ratio of active compound to lipid in said composition is between 1:10 to 1:100. 13. The method of claim 1, wherein the weight-to-weight ratio of total active compound to total lipid in said composition is between 1:10 to 1:60. 14. The method of claim 1, wherein said composition comprises a form selected from the group consisting of powder, solution, suspension, emulsion, micelle, liposome, lipidic particle, gel, and paste form. 15. The method of claim 14, wherein said composition comprises a plurality of micelles, wherein said micelles are in the form of monomeric, dimeric, polymeric or mixture of micelles and vesicles. 16. The method of claim 1, wherein said preparing of a composition comprising a complex comprises preparing said complex in a lyophilized form. 17. The method of claim 16, wherein said preparing said complex in a lyophilized form comprises using a cryoprotectant, wherein said cryoprotectant comprises one or more sugars selected from a group consisting of trehalose, maltose, lactose, sucrose, glucose, and dextran. 18. The method of claim 1, wherein, said composition comprises a tablet or a filled capsule, and optionally comprises an enteric coating material. 19. The method of claim 1, wherein said active compound is a partially water soluble or water insoluble drug. 20. The method of claim 1, wherein said administering comprises oral, intravenous, subcutaneous, parenteral, intraperitoneal, rectal, vaginal, and/or topical delivery of said lipidic composition to said subject. 21. A process for preparing a lipid formulation of an active compound, wherein said process comprises using an aqueous system to prepare a composition comprising a complex, said complex comprising at least one active compound and at least one lipid. 22. The process of claim 21, wherein said process is a process for preparing a lipid formulation of defined particle size, wherein said process comprises: a) preparing a suspension comprising at least one active compound and at least one lipid in a first aqueous medium at a pH between about pH 4.0 and pH 8.0; b) treating said suspension to form a lipid-compound suspension of defined particle size; c) lyophilizing the lipid-compound suspension of defined particle size to form lyophilized material; and d) reconstituting said lyophilized material with a second aqueous medium to obtain a suspension of lipid formulation of defined particle size, said defined particle size having a mean particle size of less than 5 microns. 23. The process of claim 22, wherein said first aqueous medium is water. 24. The process of claim 22, wherein said first aqueous medium and said second aqueous medium are different. 25. The process of claim 22, wherein said treating said suspension comprises extruding said suspension through a selected size aperture. 26. The process of claim 22, wherein said treating said suspension comprises high pressure split homogenization. 27. The process of claim 22 wherein said lyophilizing is in the presence of a cryoprotectant. 28. The process of claim 21, wherein said active compound comprises an active compound selected from the group consisting of a polyene antibiotic, a macrolide, an anti-cancer drug, and an immunosuppressant. 29. The process of claim 21, wherein said active compound comprises a compound selected from the group consisting of docetaxel, paclitaxel, doxorubicin, epirubicin, tamoxifen, endoxifen, etoposide, anthracyclines, amphotericin B, tacrolimus, and sacrolimus. 30. The process of claim 21, wherein said at least one lipid is selected from the group consisting of egg phosphatidylcholine, egg phosphatidylglycerol, soy phosphatidylcholine, hydrogenated soy phosphatidylcholine, dimyristoylphosphatidylcholine, dimyristoylphosphatidylglycerol, dipalmitoylphosohatidylcholine, disteroylphosphatidylglycerol, dipalmitoylphosphatidylglycerol, cholesterol, cholesterol sulfate and its salts, cholesterol hemisuccinate and its salts, cholesterol phosphate and its salts, cholesterylphosphocholine and other hydroxycholesterol or amino cholesterol derivatives, cholesteryl succinate, cholesteryl oleate, polyethylene glycol derivatives of cholesterol (cholesterol-PEG), coprostanol, cholestanol, cholestane, cholic acid, cortisol, corticosterone, hydrocortisone, and calciferol. 31. The process of claim 21, wherein said lipid formulation comprises cholesterol sulfate, and wherein the molar ratio of active compound to cholesterol sulfate in said suspension is in between about 1:1 to about 1:10. 32. The process of claim 22, wherein the composition mean particle size upon reconstitution is about 10-5000 nm. 33. The process of claim 21, wherein said at least one active compound exhibits poor solubility in water, alcohols, and halogenated hydrocarbon solvents. 34. The process of claim 22, wherein said suspension of lipid formulation of defined particle size comprises a suspension of liposomes and/or lipidic particles. 35. A method treating a cell with a lipidic composition comprising at least one active agent and at least one lipid, comprising: a) using an aqueous system to prepare a composition comprising a complex, said complex comprising at least one active compound and at least one lipid; and b) exposing said cell to said lipidic composition. 36. The method of claim 35, wherein said exposing said cell comprises exposing said cell to said lipidic composition in vivo. 37. The method of claim 35, wherein said subject is a mammal. 38. The method of claim 37, wherein said mammal is human.
1,600
218
218
14,354,080
1,623
The invention relates to (among other things) a method comprising the steps of (a) administering to a patient a topoisomerase II-inhibiting amount of a long-acting topoisomerase II inhibitor; and (b) administering to the patient a topoisomerase I-inhibiting amount of a long-acting topoisomerase I inhibitor.
1. A method comprising the steps of: (a) administering to a patient a topoisomerase II-inhibiting amount of a long-acting topoisomerase II inhibitor; and (b) administering to the patient a topoisomerase I-inhibiting amount of a long-acting topoisomerase I inhibitor. 2. The method of claim 1, wherein the patient is suffering from a cancer. 3. The method of claim 2, wherein the cancer is a solid cancer. 4. The method of claim 3, wherein the solid cancer is selected from the group consisting of breast cancer, ovarian cancer, colon cancer, colorectal cancer, gastric cancer, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease and adrenocortical cancer. 5. The method of claim 2, wherein the cancer is ovarian cancer. 6. The method of claim 5, wherein the ovarian cancer is platinum-resistant ovarian cancer. 7. The method of claim 2, wherein the cancer is colorectal cancer. 8. The method of claim 2, is breast cancer. 9. The method of claim 2, wherein the cancer is selected from group consisting of lymphoma cancers, leukemia cancers, rhabdomyosarcoma and neuroblastoma. 10. The method of claim 1, wherein the patient is human. 11. The method of claim 1, wherein step (a) is carried out prior to step (b) being carried out. 12. The method of claim 1, wherein step (a) is carried out after step (b) is carried out. 13. The method of claim 1, wherein steps (a) and (b) are carried out simultaneously.
The invention relates to (among other things) a method comprising the steps of (a) administering to a patient a topoisomerase II-inhibiting amount of a long-acting topoisomerase II inhibitor; and (b) administering to the patient a topoisomerase I-inhibiting amount of a long-acting topoisomerase I inhibitor.1. A method comprising the steps of: (a) administering to a patient a topoisomerase II-inhibiting amount of a long-acting topoisomerase II inhibitor; and (b) administering to the patient a topoisomerase I-inhibiting amount of a long-acting topoisomerase I inhibitor. 2. The method of claim 1, wherein the patient is suffering from a cancer. 3. The method of claim 2, wherein the cancer is a solid cancer. 4. The method of claim 3, wherein the solid cancer is selected from the group consisting of breast cancer, ovarian cancer, colon cancer, colorectal cancer, gastric cancer, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease and adrenocortical cancer. 5. The method of claim 2, wherein the cancer is ovarian cancer. 6. The method of claim 5, wherein the ovarian cancer is platinum-resistant ovarian cancer. 7. The method of claim 2, wherein the cancer is colorectal cancer. 8. The method of claim 2, is breast cancer. 9. The method of claim 2, wherein the cancer is selected from group consisting of lymphoma cancers, leukemia cancers, rhabdomyosarcoma and neuroblastoma. 10. The method of claim 1, wherein the patient is human. 11. The method of claim 1, wherein step (a) is carried out prior to step (b) being carried out. 12. The method of claim 1, wherein step (a) is carried out after step (b) is carried out. 13. The method of claim 1, wherein steps (a) and (b) are carried out simultaneously.
1,600
219
219
13,427,582
1,645
Animal protein-free, solid-form Clostridial toxin pharmaceutical compositions comprising a Clostridial toxin active ingredient and at least two excipients.
1. An animal-protein free, solid-form Clostridial toxin pharmaceutical composition comprising a Clostridial toxin active ingredient, an effective amount of lactose, an effective amount of sucrose, and an effective amount of sodium chloride; and wherein after storage for one year the pharmaceutical composition exhibits a recovered potency of the Clostridial toxin active ingredient of at least 20% when reconstituted into a form suitable for injection. 2. The composition according to claim 1, wherein the storage temperature is below freezing. 3. The composition according to claim 1, wherein the storage temperature is at ambient temperatures. 4. The composition according to claim 1, wherein the storage temperature is below freezing and the recovered potency is at least 40%. 5. The composition according to claim 1, wherein the storage temperature is at ambient temperatures and the recovered potency is at least 40%.
Animal protein-free, solid-form Clostridial toxin pharmaceutical compositions comprising a Clostridial toxin active ingredient and at least two excipients.1. An animal-protein free, solid-form Clostridial toxin pharmaceutical composition comprising a Clostridial toxin active ingredient, an effective amount of lactose, an effective amount of sucrose, and an effective amount of sodium chloride; and wherein after storage for one year the pharmaceutical composition exhibits a recovered potency of the Clostridial toxin active ingredient of at least 20% when reconstituted into a form suitable for injection. 2. The composition according to claim 1, wherein the storage temperature is below freezing. 3. The composition according to claim 1, wherein the storage temperature is at ambient temperatures. 4. The composition according to claim 1, wherein the storage temperature is below freezing and the recovered potency is at least 40%. 5. The composition according to claim 1, wherein the storage temperature is at ambient temperatures and the recovered potency is at least 40%.
1,600
220
220
13,800,495
1,641
The present disclosure is directed to antibody-linked immuno-sedimentation agent, the antibody being linked to a sedimentation agent by a non-antigen binding region of the antibody, and a method of isolating a target from a sample using the antibody-linked immuno-sedimentation agent. The methods involve forming a mixture including a sample with an antibody linked immuno-sedimentation agent and red blood cells under conditions sufficient to form red blood cell rouleaux and allow antibody-antigen binding.
1. A method of isolating a target from a sample, the method comprising: a) forming a mixture comprising a sample, an antibody-linked immuno-sedimentation agent and red blood cells, wherein the antibody-linked immuno-sedimentation agent comprises 1) at least one sedimentation agent and 2) at least one antibody linked to the at least one sedimentation agent by a non-antigen binding region of the antibody; b) incubating the mixture under conditions sufficient to form a rouleaux and allow the antibody to bind to an antigen present in the mixture; and c) recovering a target from the mixture. 2. The method of claim 1, wherein the rouleaux is formed by adsorption of the at least one sedimentation agent onto the red blood cells of the rouleaux. 3. The method of claim 1, wherein the at least one antibody is linked to the at least one sedimentation agent prior to forming the mixture. 4. The method of claim 1, wherein the antibody binds a target antigen or a non-target antigen in the sample. 5. The method of claim 1, further comprising incubating the mixture under conditions sufficient to form a rouleaux network. 6. The method of claim 1, wherein the sample is selected from the group consisting of whole blood, blood plasma, blood stem cell isolates, platelet-rich plasma, fractionated blood, packed red blood cells, umbilical cord blood, bone marrow, bone-marrow aspirates, a buffy-coat layer, a leukapheresis isolate, a plasmapheresis isolate, a cell suspension, a cell culture medium, a cell mixture, a cell homogenate, a pancreas isolate, an organ homogenate, a saline solution, a Ringer's solution, a lactated Ringer's solution, a phosphate buffered saline solution, a viral culture medium, an oocyte mixture, a sperm, a gamete mixture, and combinations thereof. 7. The method of claim 1, wherein the mixture comprises red blood cells selected from the group consisting of endogenous red blood cells, exogenous red blood cells, and combinations thereof. 8. The method of claim 7, wherein the exogenous red blood cells are selected from the group consisting of homologous red blood cells, autologous red blood cells, heterologous red blood cells, and combinations thereof. 9. The method of claim 1, wherein the target comprises a multicellular organism, a single-cell organism, a cell, a subcellular component, a cellular fragment, a virus, a viroid, a virus-like target, a membrane-bound particle, a lipid particle, a macromolecule, a DNA, a RNA, a nucleic acid-like macromolecule, a peptide, a polypeptide, and a protein. 10. The method of claim 1, wherein the at least one sedimentation agent is selected from the group consisting of a soluble sedimentation agent, a partially soluble sedimentation agent, and combinations thereof. 11. The method of claim 1, wherein the at least one sedimentation agent comprises an insoluble sedimentation agent. 12. The method of claim 1, wherein the at least one sedimentation agent comprises a polymer. 13. The method of claim 12, wherein the polymer is selected from the group consisting of a biopolymer, a synthetic polymer, a modified biopolymer, and combinations thereof. 14. The method of claim 13, wherein the biopolymer is selected from the group consisting of a polysaccharide, a polypeptide, and combinations thereof. 15. The method of claim 14, wherein the polysaccharide is selected from the group consisting of a starch, dextran, cellulose, chitin, xanthum gum, a glycosaminoglycan, and combinations thereof. 16. The method of claim 15, wherein the glycosaminoglycan is selected from the group consisting of heparin, heparin sulfate, chondroitin sulfate, and combinations thereof. 17. The method of claim 14, wherein the polypeptide is selected from the group consisting of albumin, collagen, fibrinogen, immunoglobulin, and combinations thereof. 18. The method of claim 13, wherein the synthetic polymer is selected from the group consisting of polyoxyethylene, polyoxyethylene glycol, polyoxyethylene oxide, polyvinylpyrrolidone, polylactic acid, polyglycolic acid, polydioxanone, and combinations thereof. 19. The method of claim 13, wherein the modified biopolymer is selected from the group consisting of hydrolyzed collagen-gelatin, FICOLL, ethylene oxide modified starch, and combinations thereof. 20. The method claim of claim 13, wherein the modified biopolymer comprises an iodinated contrast media, wherein the iodinated contrast media is selected from the group consisting of diatrizoate, iopamidol, iohexol, ioxilan, iopromide, iodixanol, and ioxaglate. 21. The method of claim 19, wherein the ethylene oxide modified starch is selected from the group consisting of hydroxyethyl starch, pentastarch, and combinations thereof. 22. The method of claim 1, wherein the antibody is selected from the group consisting of a monoclonal antibody, a polyclonal antibody, and combinations thereof. 23. The method of claim 1, wherein the antibody comprises at least one of an antibody fragment, a chimeric antibody, a bifunctional antibody, and a bispecific antibody. 24. The method of claim 23, wherein the antibody fragment comprises an antigen-binding domain, an Fab, an F(ab′)2, an F(ab′). 25. The method of claim 1, wherein the sample comprises blood. 26. The method of claim 25, wherein the blood comprises whole blood, blood plasma, a blood stem cell isolate, platelet-rich plasma, fractionated blood, packed red blood cells, umbilical cord blood, bone marrow, a bone-marrow aspirate, and a buffy-coat layer. 27. The method of claim 1, wherein the incubating step is from about 10 minutes to about 120 minutes. 28. The method of claim 1, wherein the method is performed at a temperature between about 0° C. to about 45° C. 29. The method of claim 1, wherein the method is performed at a pH of between about 5.4 to about 9.4. 30. The method of claim 1, wherein recovering a target from the mixture comprises separating the rouleaux from the mixture. 31. The method of claim 5, wherein recovering a target from the mixture comprises separating the rouleaux network from the mixture. 32. The method of claim 1, wherein recovering a target from the mixture comprises subjecting the mixture to at least one of sedimentation, centrifugation, density gradient centrifugation, a magnetic field, an electrical field, cell lysis, chemical crosslinking, extraction, a phase transition, freeze-thaw cycling, evaporation, boiling, subliming, melting, crystallizing, plasticizing, filtration, and combinations thereof. 33. The method of claim 1, wherein the target is isolated by positive selection or negative selection. 34. The method of claim 1, wherein the target comprises at least one of a multicellular organism, a single-cell organism, a cell, a subcellular component, a cellular fragment, a virus, a virus-like target, a membrane-bound particle, a lipid particle, a macromolecule, a DNA, a RNA, and a protein. 35. The method of claim 1, wherein the antibody-linked immuno-sedimentation agent comprises antibodies directed to at least two different antigens and wherein the antibodies are linked to the sedimentation agent by a non-antigen binding region. 36. The method of claim 1, wherein the antibody-linked immuno-sedimentation agent comprises at least two different sedimentation agents. 37. The method of claim 1, further comprising adding a blocking agent to the mixture. 38. The method of claim 37, wherein the blocking agent is selected from the group consisting of human albumin, bovine serum albumin, fetal bovine serum, immunoglobulins, anti-CD16 antibodies, anti-Human CD32 (FcγRII), Fc receptor antibodies, and combinations thereof. 39. A method of isolating a target from a sample, the method comprising: a) forming a mixture comprising a sample, an antibody-linked immuno-sedimentation agent and red blood cells, wherein the antibody-linked immuno-sedimentation agent comprises 1) at least one sedimentation agent selected from the group consisting of polyethylene glycol, dextran, and hydroxyethyl starch; and 2) at least one antibody linked to the at least one sedimentation agent by a non-antigen binding region of the antibody, wherein the antibody is selected from the group consisting of an anti-CD3 antibody, an anti-CD19 antibody, an anti-insulin antibody, an anti-CD34 antibody, an anti-CD4 antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD36 antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-TCRγ/δ antibody, an anti-glycophorin A antibody, and an anti-GP120 antibody; b) incubating the mixture under conditions sufficient to form a rouleaux and allow the antibody to bind to an antigen present in the mixture; and c) recovering a target from the mixture. 40. An antibody-linked immuno-sedimentation agent comprising at least one sedimentation agent and at least one antibody linked to the at least one sedimentation agent, wherein the at least one antibody is linked to the at least one sedimentation agent by a non-antigen binding region. 41. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is selected from the group consisting of a monoclonal antibody, a polyclonal antibody, and combinations thereof. 42. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody comprises at least one of a chimeric antibody, a bifunctional antibody, a bispecific antibody, an IgA, an IgD, an IgE, an IgG, an IgM, an intact antibody, and an antibody fragment. 43. The antibody-linked immuno-sedimentation agent of claim 42, wherein the antibody fragment comprises at least one of an antigen binding region, an antibody hypervariable region, an antibody paratope, an antibody light chain, an antibody heavy chain, a paired antibody heavy and light chain, an Fab, an F(ab′)2, and an F(ab′). 44. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is linked to the at least one sedimentation agent by an Fc region, a hinge region, a light chain constant (CL) region, a heavy chain (CH 1) region, a light chain (CL) region and heavy chain (CH 1) region, a heavy chain (CH 2) region, and a heavy chain (CH 3) region. 45. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is directly linked to the at least one sedimentation agent. 46. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is indirectly linked to the at least one sedimentation agent. 47. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one sedimentation agent is selected from the group consisting of a soluble sedimentation agent, a partially soluble sedimentation agent, and combinations thereof. 48. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one sedimentation agent comprises an insoluble sedimentation agent. 49. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one sedimentation agent comprises a polymer. 50. The antibody-linked immuno-sedimentation agent of claim 49, wherein the polymer is selected from the group consisting of a biopolymer, a synthetic polymer, a modified biopolymer, and combinations thereof. 51. The antibody-linked immuno-sedimentation agent of claim 50, wherein the biopolymer comprises at least one of a polysaccharide and a polypeptide. 52. The antibody-linked immuno-sedimentation agent of claim 50, wherein the biopolymer comprises a polysaccharide. 53. The antibody-linked immuno-sedimentation agent of claim 51, wherein the polysaccharide comprises at least one of a starch, dextran, cellulose, chitin, xanthum gum, and a glycosaminoglycan. 54. The antibody-linked immuno-sedimentation agent of claim 53, wherein the glycosaminoglycan comprises at least one of heparin, heparin sulfate, and chondroitin sulfate. 55. The antibody-linked immuno-sedimentation agent of claim 51, wherein the polypeptide comprises at least one of albumin, collagen, fibrinogen, and immunoglobulin. 56. The antibody-linked immuno-sedimentation agent of claim 50, wherein the polymer comprises a synthetic polymer. 57. The antibody-linked immuno-sedimentation agent of claim 50, wherein the synthetic polymer comprises at least one of polyoxyethylene, polyoxyethylene glycol, polyoxyethylene oxide, polyvinylpyrrolidone, polylactic acid, polyglycolic acid, and polydioxanone. 58. The antibody-linked immuno-sedimentation agent of claim 50, wherein the modified biopolymer comprises at least one of hydrolyzed collagen-gelatin, FICOLL, and ethylene oxide modified starch. 59. The antibody-linked immuno-sedimentation agent of claim 50, wherein the modified biopolymer comprises an iodinated contrast media, wherein the iodated contrast media is selected from the group consisting of diatrizoate, iopamidol, iohexol, ioxilan, iopromide, iodixanol, and ioxaglate. 60. The antibody-linked immuno-sedimentation agent of claim 58, wherein the ethylene oxide modified starch comprises at least one of hydroxyethyl starch and pentastarch. 61. The antibody-linked immuno-sedimentation agent of claim 40, comprising antibodies directed to at least two different antigens and wherein the at least two different antibodies are linked to the sedimentation agent by a non-antigen binding region. 62. The antibody-linked immuno-sedimentation agent of claim 61, wherein the antibodies directed to at least two different antigens are linked to at least two different sedimentation agents by a non-antigen binding region. 63. The antibody-linked immuno-sedimentation agent of claim 40, comprising at least two different sedimentation agents. 64. An antibody-linked immuno-sedimentation agent comprising at least one sedimentation agent selected from the group consisting of polyethylene glycol, dextran, and hydroxyethyl starch and at least one antibody linked to the at least one sedimentation agent, wherein the at least one antibody is linked to the at least one sedimentation agent by a non-antigen binding region and wherein the at least one antibody is selected from the group consisting of an anti-CD3 antibody, an anti-CD19 antibody, an anti-insulin antibody, an anti-CD34 antibody, an anti-CD4 antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD36 antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-TCRγ/δ antibody, an anti-glycophorin A antibody, and an anti-GP120 antibody.
The present disclosure is directed to antibody-linked immuno-sedimentation agent, the antibody being linked to a sedimentation agent by a non-antigen binding region of the antibody, and a method of isolating a target from a sample using the antibody-linked immuno-sedimentation agent. The methods involve forming a mixture including a sample with an antibody linked immuno-sedimentation agent and red blood cells under conditions sufficient to form red blood cell rouleaux and allow antibody-antigen binding.1. A method of isolating a target from a sample, the method comprising: a) forming a mixture comprising a sample, an antibody-linked immuno-sedimentation agent and red blood cells, wherein the antibody-linked immuno-sedimentation agent comprises 1) at least one sedimentation agent and 2) at least one antibody linked to the at least one sedimentation agent by a non-antigen binding region of the antibody; b) incubating the mixture under conditions sufficient to form a rouleaux and allow the antibody to bind to an antigen present in the mixture; and c) recovering a target from the mixture. 2. The method of claim 1, wherein the rouleaux is formed by adsorption of the at least one sedimentation agent onto the red blood cells of the rouleaux. 3. The method of claim 1, wherein the at least one antibody is linked to the at least one sedimentation agent prior to forming the mixture. 4. The method of claim 1, wherein the antibody binds a target antigen or a non-target antigen in the sample. 5. The method of claim 1, further comprising incubating the mixture under conditions sufficient to form a rouleaux network. 6. The method of claim 1, wherein the sample is selected from the group consisting of whole blood, blood plasma, blood stem cell isolates, platelet-rich plasma, fractionated blood, packed red blood cells, umbilical cord blood, bone marrow, bone-marrow aspirates, a buffy-coat layer, a leukapheresis isolate, a plasmapheresis isolate, a cell suspension, a cell culture medium, a cell mixture, a cell homogenate, a pancreas isolate, an organ homogenate, a saline solution, a Ringer's solution, a lactated Ringer's solution, a phosphate buffered saline solution, a viral culture medium, an oocyte mixture, a sperm, a gamete mixture, and combinations thereof. 7. The method of claim 1, wherein the mixture comprises red blood cells selected from the group consisting of endogenous red blood cells, exogenous red blood cells, and combinations thereof. 8. The method of claim 7, wherein the exogenous red blood cells are selected from the group consisting of homologous red blood cells, autologous red blood cells, heterologous red blood cells, and combinations thereof. 9. The method of claim 1, wherein the target comprises a multicellular organism, a single-cell organism, a cell, a subcellular component, a cellular fragment, a virus, a viroid, a virus-like target, a membrane-bound particle, a lipid particle, a macromolecule, a DNA, a RNA, a nucleic acid-like macromolecule, a peptide, a polypeptide, and a protein. 10. The method of claim 1, wherein the at least one sedimentation agent is selected from the group consisting of a soluble sedimentation agent, a partially soluble sedimentation agent, and combinations thereof. 11. The method of claim 1, wherein the at least one sedimentation agent comprises an insoluble sedimentation agent. 12. The method of claim 1, wherein the at least one sedimentation agent comprises a polymer. 13. The method of claim 12, wherein the polymer is selected from the group consisting of a biopolymer, a synthetic polymer, a modified biopolymer, and combinations thereof. 14. The method of claim 13, wherein the biopolymer is selected from the group consisting of a polysaccharide, a polypeptide, and combinations thereof. 15. The method of claim 14, wherein the polysaccharide is selected from the group consisting of a starch, dextran, cellulose, chitin, xanthum gum, a glycosaminoglycan, and combinations thereof. 16. The method of claim 15, wherein the glycosaminoglycan is selected from the group consisting of heparin, heparin sulfate, chondroitin sulfate, and combinations thereof. 17. The method of claim 14, wherein the polypeptide is selected from the group consisting of albumin, collagen, fibrinogen, immunoglobulin, and combinations thereof. 18. The method of claim 13, wherein the synthetic polymer is selected from the group consisting of polyoxyethylene, polyoxyethylene glycol, polyoxyethylene oxide, polyvinylpyrrolidone, polylactic acid, polyglycolic acid, polydioxanone, and combinations thereof. 19. The method of claim 13, wherein the modified biopolymer is selected from the group consisting of hydrolyzed collagen-gelatin, FICOLL, ethylene oxide modified starch, and combinations thereof. 20. The method claim of claim 13, wherein the modified biopolymer comprises an iodinated contrast media, wherein the iodinated contrast media is selected from the group consisting of diatrizoate, iopamidol, iohexol, ioxilan, iopromide, iodixanol, and ioxaglate. 21. The method of claim 19, wherein the ethylene oxide modified starch is selected from the group consisting of hydroxyethyl starch, pentastarch, and combinations thereof. 22. The method of claim 1, wherein the antibody is selected from the group consisting of a monoclonal antibody, a polyclonal antibody, and combinations thereof. 23. The method of claim 1, wherein the antibody comprises at least one of an antibody fragment, a chimeric antibody, a bifunctional antibody, and a bispecific antibody. 24. The method of claim 23, wherein the antibody fragment comprises an antigen-binding domain, an Fab, an F(ab′)2, an F(ab′). 25. The method of claim 1, wherein the sample comprises blood. 26. The method of claim 25, wherein the blood comprises whole blood, blood plasma, a blood stem cell isolate, platelet-rich plasma, fractionated blood, packed red blood cells, umbilical cord blood, bone marrow, a bone-marrow aspirate, and a buffy-coat layer. 27. The method of claim 1, wherein the incubating step is from about 10 minutes to about 120 minutes. 28. The method of claim 1, wherein the method is performed at a temperature between about 0° C. to about 45° C. 29. The method of claim 1, wherein the method is performed at a pH of between about 5.4 to about 9.4. 30. The method of claim 1, wherein recovering a target from the mixture comprises separating the rouleaux from the mixture. 31. The method of claim 5, wherein recovering a target from the mixture comprises separating the rouleaux network from the mixture. 32. The method of claim 1, wherein recovering a target from the mixture comprises subjecting the mixture to at least one of sedimentation, centrifugation, density gradient centrifugation, a magnetic field, an electrical field, cell lysis, chemical crosslinking, extraction, a phase transition, freeze-thaw cycling, evaporation, boiling, subliming, melting, crystallizing, plasticizing, filtration, and combinations thereof. 33. The method of claim 1, wherein the target is isolated by positive selection or negative selection. 34. The method of claim 1, wherein the target comprises at least one of a multicellular organism, a single-cell organism, a cell, a subcellular component, a cellular fragment, a virus, a virus-like target, a membrane-bound particle, a lipid particle, a macromolecule, a DNA, a RNA, and a protein. 35. The method of claim 1, wherein the antibody-linked immuno-sedimentation agent comprises antibodies directed to at least two different antigens and wherein the antibodies are linked to the sedimentation agent by a non-antigen binding region. 36. The method of claim 1, wherein the antibody-linked immuno-sedimentation agent comprises at least two different sedimentation agents. 37. The method of claim 1, further comprising adding a blocking agent to the mixture. 38. The method of claim 37, wherein the blocking agent is selected from the group consisting of human albumin, bovine serum albumin, fetal bovine serum, immunoglobulins, anti-CD16 antibodies, anti-Human CD32 (FcγRII), Fc receptor antibodies, and combinations thereof. 39. A method of isolating a target from a sample, the method comprising: a) forming a mixture comprising a sample, an antibody-linked immuno-sedimentation agent and red blood cells, wherein the antibody-linked immuno-sedimentation agent comprises 1) at least one sedimentation agent selected from the group consisting of polyethylene glycol, dextran, and hydroxyethyl starch; and 2) at least one antibody linked to the at least one sedimentation agent by a non-antigen binding region of the antibody, wherein the antibody is selected from the group consisting of an anti-CD3 antibody, an anti-CD19 antibody, an anti-insulin antibody, an anti-CD34 antibody, an anti-CD4 antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD36 antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-TCRγ/δ antibody, an anti-glycophorin A antibody, and an anti-GP120 antibody; b) incubating the mixture under conditions sufficient to form a rouleaux and allow the antibody to bind to an antigen present in the mixture; and c) recovering a target from the mixture. 40. An antibody-linked immuno-sedimentation agent comprising at least one sedimentation agent and at least one antibody linked to the at least one sedimentation agent, wherein the at least one antibody is linked to the at least one sedimentation agent by a non-antigen binding region. 41. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is selected from the group consisting of a monoclonal antibody, a polyclonal antibody, and combinations thereof. 42. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody comprises at least one of a chimeric antibody, a bifunctional antibody, a bispecific antibody, an IgA, an IgD, an IgE, an IgG, an IgM, an intact antibody, and an antibody fragment. 43. The antibody-linked immuno-sedimentation agent of claim 42, wherein the antibody fragment comprises at least one of an antigen binding region, an antibody hypervariable region, an antibody paratope, an antibody light chain, an antibody heavy chain, a paired antibody heavy and light chain, an Fab, an F(ab′)2, and an F(ab′). 44. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is linked to the at least one sedimentation agent by an Fc region, a hinge region, a light chain constant (CL) region, a heavy chain (CH 1) region, a light chain (CL) region and heavy chain (CH 1) region, a heavy chain (CH 2) region, and a heavy chain (CH 3) region. 45. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is directly linked to the at least one sedimentation agent. 46. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one antibody linked to the at least one sedimentation agent is indirectly linked to the at least one sedimentation agent. 47. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one sedimentation agent is selected from the group consisting of a soluble sedimentation agent, a partially soluble sedimentation agent, and combinations thereof. 48. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one sedimentation agent comprises an insoluble sedimentation agent. 49. The antibody-linked immuno-sedimentation agent of claim 40, wherein the at least one sedimentation agent comprises a polymer. 50. The antibody-linked immuno-sedimentation agent of claim 49, wherein the polymer is selected from the group consisting of a biopolymer, a synthetic polymer, a modified biopolymer, and combinations thereof. 51. The antibody-linked immuno-sedimentation agent of claim 50, wherein the biopolymer comprises at least one of a polysaccharide and a polypeptide. 52. The antibody-linked immuno-sedimentation agent of claim 50, wherein the biopolymer comprises a polysaccharide. 53. The antibody-linked immuno-sedimentation agent of claim 51, wherein the polysaccharide comprises at least one of a starch, dextran, cellulose, chitin, xanthum gum, and a glycosaminoglycan. 54. The antibody-linked immuno-sedimentation agent of claim 53, wherein the glycosaminoglycan comprises at least one of heparin, heparin sulfate, and chondroitin sulfate. 55. The antibody-linked immuno-sedimentation agent of claim 51, wherein the polypeptide comprises at least one of albumin, collagen, fibrinogen, and immunoglobulin. 56. The antibody-linked immuno-sedimentation agent of claim 50, wherein the polymer comprises a synthetic polymer. 57. The antibody-linked immuno-sedimentation agent of claim 50, wherein the synthetic polymer comprises at least one of polyoxyethylene, polyoxyethylene glycol, polyoxyethylene oxide, polyvinylpyrrolidone, polylactic acid, polyglycolic acid, and polydioxanone. 58. The antibody-linked immuno-sedimentation agent of claim 50, wherein the modified biopolymer comprises at least one of hydrolyzed collagen-gelatin, FICOLL, and ethylene oxide modified starch. 59. The antibody-linked immuno-sedimentation agent of claim 50, wherein the modified biopolymer comprises an iodinated contrast media, wherein the iodated contrast media is selected from the group consisting of diatrizoate, iopamidol, iohexol, ioxilan, iopromide, iodixanol, and ioxaglate. 60. The antibody-linked immuno-sedimentation agent of claim 58, wherein the ethylene oxide modified starch comprises at least one of hydroxyethyl starch and pentastarch. 61. The antibody-linked immuno-sedimentation agent of claim 40, comprising antibodies directed to at least two different antigens and wherein the at least two different antibodies are linked to the sedimentation agent by a non-antigen binding region. 62. The antibody-linked immuno-sedimentation agent of claim 61, wherein the antibodies directed to at least two different antigens are linked to at least two different sedimentation agents by a non-antigen binding region. 63. The antibody-linked immuno-sedimentation agent of claim 40, comprising at least two different sedimentation agents. 64. An antibody-linked immuno-sedimentation agent comprising at least one sedimentation agent selected from the group consisting of polyethylene glycol, dextran, and hydroxyethyl starch and at least one antibody linked to the at least one sedimentation agent, wherein the at least one antibody is linked to the at least one sedimentation agent by a non-antigen binding region and wherein the at least one antibody is selected from the group consisting of an anti-CD3 antibody, an anti-CD19 antibody, an anti-insulin antibody, an anti-CD34 antibody, an anti-CD4 antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD36 antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-TCRγ/δ antibody, an anti-glycophorin A antibody, and an anti-GP120 antibody.
1,600
221
221
14,878,561
1,656
The disclosure describes methods for the purification of protein-enriched extracts to provide concentrates and isolates and methods for incorporation of such materials into products. The purification methods are adapted for removal of one or more of ash, metal salts, alkaloids, particulates, heavy metals, and other impurities and/or contaminants from extracts, as well as modifying the sensory characteristics (e.g., odor, color, and/or taste characteristics) of extracts. The methods generally include diafiltration, treatment with functionalized resins, and supercritical extraction. A protein composition in the form of a concentrate or isolate is provided, the protein composition including RuBisCO, F2 fraction proteins, or combination thereof extracted from a plant of the Nicotiana species, wherein the protein composition is characterized by one or more of: an ash content of less than about 15% by weight; a nicotine content of less than about 10 μg/g, and a heavy metal content of less than about 60 μg/g.
1.-27. (canceled) 28. A method for modifying the sensory characteristics of a protein-enriched material, the method comprising: a) receiving a plant-derived, protein-enriched material comprising RuBisCO, F2 fraction proteins, or a combination thereof, wherein the plant-derived, protein-enriched material exhibits an initial odor and taste; b) extracting components from the protein-enriched extract with a solvent while the solvent is in a supercritical state to provide a protein concentrate or isolate having an altered odor, taste, or combination thereof; and c) separating the protein concentrate or isolate from the unextracted residue. 29. The method of claim 28, wherein the plant-derived, protein-enriched material is in spray dried or freeze-dried form. 30. The method of claim 29, wherein the method further comprises moistening the plant-derived, protein-enriched material prior to the extracting step with water to provide a plant-derived, protein-enriched material having between about 1% and about 50% moisture by weight. 31. The method of claim 28, wherein the plant-derived, protein-enriched material comprises material from a plant of the Nicotiana species. 32. The method of claim 28, wherein the solvent comprises supercritical carbon dioxide. 33. The method of claim 28, wherein the solvent further comprises a modifier in an amount of up to about 30% by volume. 34. The method of claim 33, wherein the modifier is selected from the group consisting of ethanol, 1-butanol, isopropanol, methanol, 1-propanol, and mixtures thereof. 35. The method of claim 28, wherein the protein concentrate or isolate is characterized by a decrease in salty flavor as compared with the plant-derived, protein-enriched material. 36. The method of claim 28, wherein the protein concentrate or isolate is characterized by a decrease in nicotine as compared with the plant-derived, protein-enriched material. 37. The method of claim 28, wherein the plant-derived, protein-enriched material comprises at least about 40% RuBisCO, F2 fraction proteins, or a combination thereof on a dry weight basis. 38.-39. (canceled) 40. A method for modifying the content or sensory characteristics of a plant-derived, protein-enriched material, the method comprising receiving a plant-derived, protein-enriched material comprising RuBisCO, F2 fraction proteins, or a combination thereof, wherein the plant-derived, protein-enriched material further comprises one or more undesirable characteristics or impurities selected from the group consisting of ash, nicotine, heavy metals, odor, taste, or a combination thereof; and further comprising one or more of the following steps: a) dissolving at least a portion of the plant-derived, protein-enriched material in a solvent to form a solution and subjecting the plant-derived, protein-enriched material solution to diafiltration by passing the solution through one or more ultrafiltration membranes to afford a retentate comprising a protein concentrate or isolate having a reduced amount of ash, nicotine, or a combination thereof as compared with the plant-derived, protein-enriched material; b) dissolving at least a portion of the plant-derived, protein-enriched material in a solvent to form a solution, contacting the plant-derived, protein-enriched material solution with a functionalized resin to afford a treated solution comprising a protein concentrate or isolate having a reduced amount of one or more of copper, cadmium, lead, and nickel, and separating the treated solution from the functionalized resin; and c) extracting components from the protein-enriched extract with a solvent while the solvent is in a supercritical state to provide a protein concentrate or isolate having altered odor, taste, or a combination thereof, and separating the protein concentrate or isolate from the unextracted residue. 41. A protein composition in the form of a protein concentrate or isolate comprising RuBisCO, F2 fraction proteins, or a combination thereof extracted from a plant of the Nicotiana species, wherein the protein concentrate or isolate is characterized by one or more of the following: a) an ash content of less than about 15% by dry weight; b) a nicotine content of less than about 10 μg/g; and c) a heavy metal content of less than about 60 μg/g. 42. The protein composition of claim 41, comprising at least about 70% by dry weight of RuBisCO, F2 fraction proteins, or a combination thereof. 43. The protein composition of claim 41, comprising at least about 80% by dry weight of RuBisCO, F2 fraction proteins, or a combination thereof. 44. The protein composition of claim 41, comprising at least about 90% by dry weight of RuBisCO, F2 fraction proteins, or a combination thereof. 45. The protein composition of claim 41, having an ash content of less than about 10% by weight. 46. The protein composition of claim 41, having a nicotine content of less than about 5 μg/g. 47. The protein composition of claim 41, having a heavy metal content of less than about 50 μg/g. 48. The protein composition of claim 41, having a heavy metal content of less than about 40 μg/g. 49. The protein composition of claim 41, wherein the combined concentration of cadmium, copper, lead, and nickel is less than about 40 μg/g. 50. The protein composition of claim 41, wherein the combined concentration of cadmium, copper, lead, and nickel is less than about 20 μg/g. 51. The protein composition of claim 41, wherein the protein composition is odorless. 52. (canceled)
The disclosure describes methods for the purification of protein-enriched extracts to provide concentrates and isolates and methods for incorporation of such materials into products. The purification methods are adapted for removal of one or more of ash, metal salts, alkaloids, particulates, heavy metals, and other impurities and/or contaminants from extracts, as well as modifying the sensory characteristics (e.g., odor, color, and/or taste characteristics) of extracts. The methods generally include diafiltration, treatment with functionalized resins, and supercritical extraction. A protein composition in the form of a concentrate or isolate is provided, the protein composition including RuBisCO, F2 fraction proteins, or combination thereof extracted from a plant of the Nicotiana species, wherein the protein composition is characterized by one or more of: an ash content of less than about 15% by weight; a nicotine content of less than about 10 μg/g, and a heavy metal content of less than about 60 μg/g.1.-27. (canceled) 28. A method for modifying the sensory characteristics of a protein-enriched material, the method comprising: a) receiving a plant-derived, protein-enriched material comprising RuBisCO, F2 fraction proteins, or a combination thereof, wherein the plant-derived, protein-enriched material exhibits an initial odor and taste; b) extracting components from the protein-enriched extract with a solvent while the solvent is in a supercritical state to provide a protein concentrate or isolate having an altered odor, taste, or combination thereof; and c) separating the protein concentrate or isolate from the unextracted residue. 29. The method of claim 28, wherein the plant-derived, protein-enriched material is in spray dried or freeze-dried form. 30. The method of claim 29, wherein the method further comprises moistening the plant-derived, protein-enriched material prior to the extracting step with water to provide a plant-derived, protein-enriched material having between about 1% and about 50% moisture by weight. 31. The method of claim 28, wherein the plant-derived, protein-enriched material comprises material from a plant of the Nicotiana species. 32. The method of claim 28, wherein the solvent comprises supercritical carbon dioxide. 33. The method of claim 28, wherein the solvent further comprises a modifier in an amount of up to about 30% by volume. 34. The method of claim 33, wherein the modifier is selected from the group consisting of ethanol, 1-butanol, isopropanol, methanol, 1-propanol, and mixtures thereof. 35. The method of claim 28, wherein the protein concentrate or isolate is characterized by a decrease in salty flavor as compared with the plant-derived, protein-enriched material. 36. The method of claim 28, wherein the protein concentrate or isolate is characterized by a decrease in nicotine as compared with the plant-derived, protein-enriched material. 37. The method of claim 28, wherein the plant-derived, protein-enriched material comprises at least about 40% RuBisCO, F2 fraction proteins, or a combination thereof on a dry weight basis. 38.-39. (canceled) 40. A method for modifying the content or sensory characteristics of a plant-derived, protein-enriched material, the method comprising receiving a plant-derived, protein-enriched material comprising RuBisCO, F2 fraction proteins, or a combination thereof, wherein the plant-derived, protein-enriched material further comprises one or more undesirable characteristics or impurities selected from the group consisting of ash, nicotine, heavy metals, odor, taste, or a combination thereof; and further comprising one or more of the following steps: a) dissolving at least a portion of the plant-derived, protein-enriched material in a solvent to form a solution and subjecting the plant-derived, protein-enriched material solution to diafiltration by passing the solution through one or more ultrafiltration membranes to afford a retentate comprising a protein concentrate or isolate having a reduced amount of ash, nicotine, or a combination thereof as compared with the plant-derived, protein-enriched material; b) dissolving at least a portion of the plant-derived, protein-enriched material in a solvent to form a solution, contacting the plant-derived, protein-enriched material solution with a functionalized resin to afford a treated solution comprising a protein concentrate or isolate having a reduced amount of one or more of copper, cadmium, lead, and nickel, and separating the treated solution from the functionalized resin; and c) extracting components from the protein-enriched extract with a solvent while the solvent is in a supercritical state to provide a protein concentrate or isolate having altered odor, taste, or a combination thereof, and separating the protein concentrate or isolate from the unextracted residue. 41. A protein composition in the form of a protein concentrate or isolate comprising RuBisCO, F2 fraction proteins, or a combination thereof extracted from a plant of the Nicotiana species, wherein the protein concentrate or isolate is characterized by one or more of the following: a) an ash content of less than about 15% by dry weight; b) a nicotine content of less than about 10 μg/g; and c) a heavy metal content of less than about 60 μg/g. 42. The protein composition of claim 41, comprising at least about 70% by dry weight of RuBisCO, F2 fraction proteins, or a combination thereof. 43. The protein composition of claim 41, comprising at least about 80% by dry weight of RuBisCO, F2 fraction proteins, or a combination thereof. 44. The protein composition of claim 41, comprising at least about 90% by dry weight of RuBisCO, F2 fraction proteins, or a combination thereof. 45. The protein composition of claim 41, having an ash content of less than about 10% by weight. 46. The protein composition of claim 41, having a nicotine content of less than about 5 μg/g. 47. The protein composition of claim 41, having a heavy metal content of less than about 50 μg/g. 48. The protein composition of claim 41, having a heavy metal content of less than about 40 μg/g. 49. The protein composition of claim 41, wherein the combined concentration of cadmium, copper, lead, and nickel is less than about 40 μg/g. 50. The protein composition of claim 41, wherein the combined concentration of cadmium, copper, lead, and nickel is less than about 20 μg/g. 51. The protein composition of claim 41, wherein the protein composition is odorless. 52. (canceled)
1,600
222
222
15,072,965
1,612
Embodiments herein related to multi-component packaged food products which have been thermally processed to achieve commercial sterility in a package, including at least a first component and a second component in contact with (e.g., immersed in) the first component, wherein said first component is comprised of a substantially water-based, liquid, pasty or gelled food such as sauce, gel, gravy, jus or the like, and wherein the second component consists of one or more relatively hard, manufactured pieces that substantially retain their initial shape and hard or crunchy textural functionality for at least an expected shelf-life of the product whilst exposed to the moisture content of the first component.
1. A multi-component packaged food product which has been thermally processed to achieve commercial sterility in a package, including at least a first component and a second component in contact with the first component, wherein said first component is a substantially water-based, liquid, pasty or gelled food, and wherein the second component is one or more relatively hard, manufactured pieces, each piece including a matrix of inorganic material including calcium mineral salts. 2. The multi-component packaged food product of claim 1, wherein the pieces of the second component have an initial moisture content of less than 10.0% w/w prior to incorporation into said first component. 3. The multi-component packaged food product of claim 1, wherein prior to thermal processing, the difference in water activity between said first component and said pieces of the second component is in the range 0.10 to 0.20. 4. The multi-component packaged food product of claim 1, wherein the first component has a water activity of at least 0.9. 5. The multi-component packaged food product of claim 1, wherein the pieces of the second component pieces are compressed calcium mineral salts. 6. The multi-component packaged food product of claim 1, wherein the matrix of inorganic material includes a matrix of calcium hydroxyapatite. 7. The multi-component packaged food product of claim 6, wherein the solid phase pieces include one or more materials selected from the group consisting of nutraceutical actives, pharmaceutical actives, vitamins, functional minerals, dentifrices and chelating agents. 8. The multi-component packaged food product of claim 6, wherein the pieces include palatant substances incorporated therein or present thereon. 9. The multi-component packaged food product of claim 6, wherein the calcium hydroxyapatite is formulated and manufactured by dry blending of tri-calcium phosphate (TCP), calcium hydroxide (CH) and calcium sulphate (CS), preferably in a ratio of about 2TCP:1CH:1CS, subsequent addition of water and appropriate titration and neutralization using a food-grade acid to an about neutral pH value, followed by optional conditioning of the resulting slurry prior to drying and granulating. 10. A multi-component packaged food product which has been thermally processed to achieve commercial sterility in a package, including at least a wet phase component and a plurality of solid phase pieces, wherein said wet phase component consists of a wet phase comprising water and other edible substances and having a water activity of above 85 and which at ambient temperature has a liquid, viscous or gelled consistency, and wherein said solid phase pieces are at least partially surrounded by the wet phase component and include a manufactured matrix of inorganic and edible mineral salts having a bulk water activity value of 0.75 or below prior to thermal processing of the packaged food product. 11. The multi-component packaged food product of claim 10, wherein the solid phase pieces have an initial moisture content of less than 10.0% w/w prior to incorporation into said wet phase component. 12. The multi-component packaged food product of claim 10, wherein prior to thermal processing, the difference in water activity between said wet phase component and said solid phase pieces is in the range 0.10 to 0.20. 13. The multi-component packaged food product of claim 10, wherein the wet phase component has a water activity of at least 0.9. 14. The multi-component packaged food product of claim 10, wherein the solid phase pieces are compression tablets. 15. The multi-component packaged food product of claim 10, wherein the solid phase pieces include a matrix of inorganic material including calcium mineral salts. 16. The multi-component packaged food product of claim 15, wherein the calcium mineral salts include calcium hydroxyapatite. 17. The multi-component packaged food product of claim 16, wherein the solid phase pieces include one or more materials selected from the group consisting of nutraceutical actives, pharmaceutical actives, vitamins, functional minerals, dentifrices and chelating agents. 18. The multi-component packaged food product of claim 16, wherein the pieces include palatant substances incorporated therein or present thereon 19. The multi-component packaged food product of claim 16, wherein the calcium hydroxyapatite is formulated and manufactured by dry blending of tri-calcium phosphate (TCP), calcium hydroxide (CH) and calcium sulphate (CS), preferably in a ratio of about 2TCP:1CH:1CS, subsequent addition of water and appropriate titration and neutralization using a food-grade acid to an about neutral pH value, followed by optional conditioning of the resulting slurry prior to drying and granulating.
Embodiments herein related to multi-component packaged food products which have been thermally processed to achieve commercial sterility in a package, including at least a first component and a second component in contact with (e.g., immersed in) the first component, wherein said first component is comprised of a substantially water-based, liquid, pasty or gelled food such as sauce, gel, gravy, jus or the like, and wherein the second component consists of one or more relatively hard, manufactured pieces that substantially retain their initial shape and hard or crunchy textural functionality for at least an expected shelf-life of the product whilst exposed to the moisture content of the first component.1. A multi-component packaged food product which has been thermally processed to achieve commercial sterility in a package, including at least a first component and a second component in contact with the first component, wherein said first component is a substantially water-based, liquid, pasty or gelled food, and wherein the second component is one or more relatively hard, manufactured pieces, each piece including a matrix of inorganic material including calcium mineral salts. 2. The multi-component packaged food product of claim 1, wherein the pieces of the second component have an initial moisture content of less than 10.0% w/w prior to incorporation into said first component. 3. The multi-component packaged food product of claim 1, wherein prior to thermal processing, the difference in water activity between said first component and said pieces of the second component is in the range 0.10 to 0.20. 4. The multi-component packaged food product of claim 1, wherein the first component has a water activity of at least 0.9. 5. The multi-component packaged food product of claim 1, wherein the pieces of the second component pieces are compressed calcium mineral salts. 6. The multi-component packaged food product of claim 1, wherein the matrix of inorganic material includes a matrix of calcium hydroxyapatite. 7. The multi-component packaged food product of claim 6, wherein the solid phase pieces include one or more materials selected from the group consisting of nutraceutical actives, pharmaceutical actives, vitamins, functional minerals, dentifrices and chelating agents. 8. The multi-component packaged food product of claim 6, wherein the pieces include palatant substances incorporated therein or present thereon. 9. The multi-component packaged food product of claim 6, wherein the calcium hydroxyapatite is formulated and manufactured by dry blending of tri-calcium phosphate (TCP), calcium hydroxide (CH) and calcium sulphate (CS), preferably in a ratio of about 2TCP:1CH:1CS, subsequent addition of water and appropriate titration and neutralization using a food-grade acid to an about neutral pH value, followed by optional conditioning of the resulting slurry prior to drying and granulating. 10. A multi-component packaged food product which has been thermally processed to achieve commercial sterility in a package, including at least a wet phase component and a plurality of solid phase pieces, wherein said wet phase component consists of a wet phase comprising water and other edible substances and having a water activity of above 85 and which at ambient temperature has a liquid, viscous or gelled consistency, and wherein said solid phase pieces are at least partially surrounded by the wet phase component and include a manufactured matrix of inorganic and edible mineral salts having a bulk water activity value of 0.75 or below prior to thermal processing of the packaged food product. 11. The multi-component packaged food product of claim 10, wherein the solid phase pieces have an initial moisture content of less than 10.0% w/w prior to incorporation into said wet phase component. 12. The multi-component packaged food product of claim 10, wherein prior to thermal processing, the difference in water activity between said wet phase component and said solid phase pieces is in the range 0.10 to 0.20. 13. The multi-component packaged food product of claim 10, wherein the wet phase component has a water activity of at least 0.9. 14. The multi-component packaged food product of claim 10, wherein the solid phase pieces are compression tablets. 15. The multi-component packaged food product of claim 10, wherein the solid phase pieces include a matrix of inorganic material including calcium mineral salts. 16. The multi-component packaged food product of claim 15, wherein the calcium mineral salts include calcium hydroxyapatite. 17. The multi-component packaged food product of claim 16, wherein the solid phase pieces include one or more materials selected from the group consisting of nutraceutical actives, pharmaceutical actives, vitamins, functional minerals, dentifrices and chelating agents. 18. The multi-component packaged food product of claim 16, wherein the pieces include palatant substances incorporated therein or present thereon 19. The multi-component packaged food product of claim 16, wherein the calcium hydroxyapatite is formulated and manufactured by dry blending of tri-calcium phosphate (TCP), calcium hydroxide (CH) and calcium sulphate (CS), preferably in a ratio of about 2TCP:1CH:1CS, subsequent addition of water and appropriate titration and neutralization using a food-grade acid to an about neutral pH value, followed by optional conditioning of the resulting slurry prior to drying and granulating.
1,600
223
223
14,336,455
1,633
A mycological biopolymer product consisting entirely of fungal mycelium is made by inoculating a nutritive substrate with a selected fungus in a sealed environment except for a void space, which space is subsequently filled with a network of undifferentiated fungal mycelium. The environmental conditions for producing the mycological biopolymer product, i.e. a high carbon dioxide (CO 2 ) content (from 5% to 7% by volume) and an elevated temperature (from 85° F. to 95° F.), prevent full differentiation of the fungus into a mushroom. There are no stipe, cap, or spores produced. The biopolymer product grows into the void space of the tool, filling the space with an undifferentiated mycelium chitin-polymer, which is subsequently extracted from the substrate and dried.
1. A mycological biopolymer material characterized in being free of any stipe, cap, or spores. 2. A process of growing a mycological biopolymer material comprising the steps of providing a tool defining a cavity therein with an opening into said cavity; packing said cavity of the tool with nutritive substrate and a fungus; placing a lid on said tool to seal said cavity, said lid having only one outlet therein open to fresh air and defining a void space; allowing said fungus to grow mycelium within said cavity and to allow the mycelium to respirate within the tool thereby producing carbon dioxide while colonizing the nutritive substrate; allowing the produced carbon dioxide to diffuse out of said outlet in said lid to create a gradient of carbon dioxide; allowing the mycelia to grow along said gradient and to fill said void space without producing a stipe, cap or spore therein. 3. A process as set forth in claim 2 wherein the carbon dioxide constitutes from 3% to 7% of the environment within said void space. 4. A process as set forth in claim 3 wherein the environmental temperature is from 85° F. to 95° F. within the void space . 5. A process as set forth in claim 2 wherein said void space is disposed vertically above said substrate. 6. A process as set forth in claim 2 wherein said void space is disposed horizontally beside said substrate. 7. A process of growing a mycological biopolymer comprising the steps of filling a tool with a nutritive substrate and a fungus; placing a lid on the tool to seal the tool, said lid having only one outlet therein open to fresh air and defining a void space; incubation of the sealed tool at high temperatures and carbon dioxide concentrations to induce biopolymer growth into the void space wherein the environmental temperature is from 85° F. to 95° F. and carbon dioxide constitutes from 3% to 7% of the environment within the void space; and therefafter drying the produced mycological biopolymer. 8. A process as set forth in claim 7 further comprising the steps of suspending mats in the void space of the tool and growing the mycological biopolymer about the mats to incorporate the mats in the mycological biopolymer to increase tensile strength in said mycological biopolymer. 9. A process as set forth in claim 7 wherein said step of incubation at high temperatures and carbon dioxide concentration occurs for 5 to 14. 10. A process as set forth in claim 7 further comprising the step of applying at least one morphological modifier onto the surface of the mycological biopolymer to alter the morphology of the mycelia. 11. A process as set forth in claim 10 wherein said morphological modifier is at least one of a hormone, forskolin, calcium, and a calcium blocker. 12. A process as set forth in claim 7 further comprising the step of compressing the mycological biopolymer to predetermined dimensions after said step of incubation and thereafter incubating the compressed mycological biopolymer for an additional 0 to 72 hours to increase strength and density prior to said step of drying. 13. A process as set forth in claim 12 further comprising the step of shaping the dried mycological biopolymer to a predetermined shape. 14. A process as set forth in claim 7 further comprising the step of sandwiching the mycological biopolymer between a pair of laminates after said step of incubation and thereafter incubating the mycological biopolymer for an additional 12 to 72 hours to adhere the mycological biopolymer between and to said pair of laminates prior to said step of drying. 15. A product as made by the process of claim 7.
A mycological biopolymer product consisting entirely of fungal mycelium is made by inoculating a nutritive substrate with a selected fungus in a sealed environment except for a void space, which space is subsequently filled with a network of undifferentiated fungal mycelium. The environmental conditions for producing the mycological biopolymer product, i.e. a high carbon dioxide (CO 2 ) content (from 5% to 7% by volume) and an elevated temperature (from 85° F. to 95° F.), prevent full differentiation of the fungus into a mushroom. There are no stipe, cap, or spores produced. The biopolymer product grows into the void space of the tool, filling the space with an undifferentiated mycelium chitin-polymer, which is subsequently extracted from the substrate and dried.1. A mycological biopolymer material characterized in being free of any stipe, cap, or spores. 2. A process of growing a mycological biopolymer material comprising the steps of providing a tool defining a cavity therein with an opening into said cavity; packing said cavity of the tool with nutritive substrate and a fungus; placing a lid on said tool to seal said cavity, said lid having only one outlet therein open to fresh air and defining a void space; allowing said fungus to grow mycelium within said cavity and to allow the mycelium to respirate within the tool thereby producing carbon dioxide while colonizing the nutritive substrate; allowing the produced carbon dioxide to diffuse out of said outlet in said lid to create a gradient of carbon dioxide; allowing the mycelia to grow along said gradient and to fill said void space without producing a stipe, cap or spore therein. 3. A process as set forth in claim 2 wherein the carbon dioxide constitutes from 3% to 7% of the environment within said void space. 4. A process as set forth in claim 3 wherein the environmental temperature is from 85° F. to 95° F. within the void space . 5. A process as set forth in claim 2 wherein said void space is disposed vertically above said substrate. 6. A process as set forth in claim 2 wherein said void space is disposed horizontally beside said substrate. 7. A process of growing a mycological biopolymer comprising the steps of filling a tool with a nutritive substrate and a fungus; placing a lid on the tool to seal the tool, said lid having only one outlet therein open to fresh air and defining a void space; incubation of the sealed tool at high temperatures and carbon dioxide concentrations to induce biopolymer growth into the void space wherein the environmental temperature is from 85° F. to 95° F. and carbon dioxide constitutes from 3% to 7% of the environment within the void space; and therefafter drying the produced mycological biopolymer. 8. A process as set forth in claim 7 further comprising the steps of suspending mats in the void space of the tool and growing the mycological biopolymer about the mats to incorporate the mats in the mycological biopolymer to increase tensile strength in said mycological biopolymer. 9. A process as set forth in claim 7 wherein said step of incubation at high temperatures and carbon dioxide concentration occurs for 5 to 14. 10. A process as set forth in claim 7 further comprising the step of applying at least one morphological modifier onto the surface of the mycological biopolymer to alter the morphology of the mycelia. 11. A process as set forth in claim 10 wherein said morphological modifier is at least one of a hormone, forskolin, calcium, and a calcium blocker. 12. A process as set forth in claim 7 further comprising the step of compressing the mycological biopolymer to predetermined dimensions after said step of incubation and thereafter incubating the compressed mycological biopolymer for an additional 0 to 72 hours to increase strength and density prior to said step of drying. 13. A process as set forth in claim 12 further comprising the step of shaping the dried mycological biopolymer to a predetermined shape. 14. A process as set forth in claim 7 further comprising the step of sandwiching the mycological biopolymer between a pair of laminates after said step of incubation and thereafter incubating the mycological biopolymer for an additional 12 to 72 hours to adhere the mycological biopolymer between and to said pair of laminates prior to said step of drying. 15. A product as made by the process of claim 7.
1,600
224
224
13,813,705
1,663
The invention relates to endonucleases cleaving DNA target sequences from algae genomes, to appropriate vectors encoding such endonucleases, to cells or to algae modified by such vectors and to the use of these endonucleases and products derived therefrom for targeted genomic engineering in algae.
1-15. (canceled) 16. A method for targeted genomic engineering in an algal cell comprising introducing an endonuclease into the algal cell to induce a double-stranded cleavage at a site of interest in the genome of the algal cell. 17. The method of claim 16, comprising: providing an endonuclease capable of inducing a double-stranded cleavage at a site of interest in the genome of an algal cell; introducing the endonuclease into an algal cell; and isolating an algal cell having a modified targeted genomic site of interest. 18. The method of claim 16, wherein the endonuclease is introduced into the algal cell by electroporation or bombardment. 19. The method of claim 17, wherein the endonuclease is introduced into the algal cell by electroporation or bombardment. 20. The method of claim 16, wherein a targeted knock-out in algae is induced by the endonuclease at the site of interest in the genome. 21. The method of claim 16, wherein at least one transgene is inserted at the targeted genomic site of interest by introducing a template that is flanked by sequences sharing homology with the region surrounding the genomic DNA cleavage site of interest. 22. The method of claim 21, wherein the template comprises at least one transgene encoding a gene selected from the group consisting of quorum sensing, secretion of hydrocarbons, fatty acid composition, lipids accumulation, enhanced photosynthesis, pigments production, mercury volatilization, frustule composition or organization, and mitigation genes. 23. The method of claim 21, wherein the template comprises a nucleic acid encoding a selectable marker. 24. The method of claim 23, wherein the selectable marker is N-acetyltransferase 1 (Nat1) conferring the resistance to Nourseothricin. 25. The method of claim 22, wherein the transgene insertion does not modify expression of genes located in the vicinity of the target sequence. 26. The method of claim 21, wherein the template comprises multiple transgenes. 27. The method of claim 16, wherein the endonuclease is a meganuclease. 28. The method of claim 27, wherein the meganuclease is selected from homodimers, heterodimers, obligate heterodimers and single chain variants. 29. The method of claim 27, wherein the meganuclease is an engineered I-CreI. 30. The method of claim 17, wherein the endonuclease is an engineered zinc-finger binding domain fused to a restriction enzyme. 31. The method of claim 17, wherein the algal cell is selected from the group consisting of Amphora, Anabaena, Anikstrodesmis, Botryococcus, Chaetoceros, Chlamydomonas, Chlorella, Chlorococcum, Cyclotella, Cylindrotheca, Dunaliella, Emiliana, Euglena, Hematococcus, Isochrysis, Monochrysis, Monoraphidium, Nannochloris, Nannnochloropsis, Navicula, Nephrochloris, Nephroselmis, Nitzschia, Nodularia, Nostoc, Oochromonas, Oocystis, Oscillartoria, Pavlova, Phaeodactylum, Playtmonas, Pleurochrysis, Porhyra, Pseudoanabaena, Pyramimonas, Stichococcus, Synechococcus, Synechocystis, Tetraselmis, Thalassiosira, and Trichodesmium algal cells. 32. A targeted genome engineered algae obtained by the method of claim 16. 33. The targeted genome engineered algae of claim 32, comprising at least one transgene inserted into a targeted genomic site of interest. 34. The targeted genome engineered algae of claim 33, wherein the transgene encodes a gene selected from the group consisting of quorum sensing, secretion of hydrocarbons, fatty acid composition, lipids accumulation, enhanced photosynthesis, pigments production, mercury volatilization, frustule composition or organization, and mitigation genes. 35. An algae comprising a nucleic acid sequence encoding an endonuclease. 36. A method of increasing biofuel production comprising introducing an endonuclease into an algal cell to induce a double-stranded cleavage within a gene regulating the production of fatty acid and triacylglcerols in the genome of the algal cell, wherein the cleavage results in an increase of fatty acid and triacylglcerols in the algal cell. 37. The method according to claim 36 comprising: providing an endonuclease capable of inducing a double-stranded cleavage at a site of interest in the genome of an algal cell; introducing the endonuclease into an algal cell; and isolating an algal cell having a modified targeted genomic site of interest. 38. The method of claim 37, wherein the endonuclease is introduced into the algal cell by electroporation or bombardment.
The invention relates to endonucleases cleaving DNA target sequences from algae genomes, to appropriate vectors encoding such endonucleases, to cells or to algae modified by such vectors and to the use of these endonucleases and products derived therefrom for targeted genomic engineering in algae.1-15. (canceled) 16. A method for targeted genomic engineering in an algal cell comprising introducing an endonuclease into the algal cell to induce a double-stranded cleavage at a site of interest in the genome of the algal cell. 17. The method of claim 16, comprising: providing an endonuclease capable of inducing a double-stranded cleavage at a site of interest in the genome of an algal cell; introducing the endonuclease into an algal cell; and isolating an algal cell having a modified targeted genomic site of interest. 18. The method of claim 16, wherein the endonuclease is introduced into the algal cell by electroporation or bombardment. 19. The method of claim 17, wherein the endonuclease is introduced into the algal cell by electroporation or bombardment. 20. The method of claim 16, wherein a targeted knock-out in algae is induced by the endonuclease at the site of interest in the genome. 21. The method of claim 16, wherein at least one transgene is inserted at the targeted genomic site of interest by introducing a template that is flanked by sequences sharing homology with the region surrounding the genomic DNA cleavage site of interest. 22. The method of claim 21, wherein the template comprises at least one transgene encoding a gene selected from the group consisting of quorum sensing, secretion of hydrocarbons, fatty acid composition, lipids accumulation, enhanced photosynthesis, pigments production, mercury volatilization, frustule composition or organization, and mitigation genes. 23. The method of claim 21, wherein the template comprises a nucleic acid encoding a selectable marker. 24. The method of claim 23, wherein the selectable marker is N-acetyltransferase 1 (Nat1) conferring the resistance to Nourseothricin. 25. The method of claim 22, wherein the transgene insertion does not modify expression of genes located in the vicinity of the target sequence. 26. The method of claim 21, wherein the template comprises multiple transgenes. 27. The method of claim 16, wherein the endonuclease is a meganuclease. 28. The method of claim 27, wherein the meganuclease is selected from homodimers, heterodimers, obligate heterodimers and single chain variants. 29. The method of claim 27, wherein the meganuclease is an engineered I-CreI. 30. The method of claim 17, wherein the endonuclease is an engineered zinc-finger binding domain fused to a restriction enzyme. 31. The method of claim 17, wherein the algal cell is selected from the group consisting of Amphora, Anabaena, Anikstrodesmis, Botryococcus, Chaetoceros, Chlamydomonas, Chlorella, Chlorococcum, Cyclotella, Cylindrotheca, Dunaliella, Emiliana, Euglena, Hematococcus, Isochrysis, Monochrysis, Monoraphidium, Nannochloris, Nannnochloropsis, Navicula, Nephrochloris, Nephroselmis, Nitzschia, Nodularia, Nostoc, Oochromonas, Oocystis, Oscillartoria, Pavlova, Phaeodactylum, Playtmonas, Pleurochrysis, Porhyra, Pseudoanabaena, Pyramimonas, Stichococcus, Synechococcus, Synechocystis, Tetraselmis, Thalassiosira, and Trichodesmium algal cells. 32. A targeted genome engineered algae obtained by the method of claim 16. 33. The targeted genome engineered algae of claim 32, comprising at least one transgene inserted into a targeted genomic site of interest. 34. The targeted genome engineered algae of claim 33, wherein the transgene encodes a gene selected from the group consisting of quorum sensing, secretion of hydrocarbons, fatty acid composition, lipids accumulation, enhanced photosynthesis, pigments production, mercury volatilization, frustule composition or organization, and mitigation genes. 35. An algae comprising a nucleic acid sequence encoding an endonuclease. 36. A method of increasing biofuel production comprising introducing an endonuclease into an algal cell to induce a double-stranded cleavage within a gene regulating the production of fatty acid and triacylglcerols in the genome of the algal cell, wherein the cleavage results in an increase of fatty acid and triacylglcerols in the algal cell. 37. The method according to claim 36 comprising: providing an endonuclease capable of inducing a double-stranded cleavage at a site of interest in the genome of an algal cell; introducing the endonuclease into an algal cell; and isolating an algal cell having a modified targeted genomic site of interest. 38. The method of claim 37, wherein the endonuclease is introduced into the algal cell by electroporation or bombardment.
1,600
225
225
14,385,331
1,633
The disclosure provides non-human optogenetic animal models of depression. Specifically, non-human animals each expresses a light-responsive opsin in a neuron of the animal are provided. The animal models are useful for identifying agents and targets of therapeutic strategies for treatment of depression. Examples of using the non-human animals expressing light-responsive opsin including Halorhodopsin family of light-responsive chloride pumps and Channelrhodopsin family of light-responsive cation channel proteins are described.
1. A method for identifying a candidate agent treating depression in an individual, comprising: contacting a rodent that expresses an active optogenetic inhibitor of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with a test agent, and determining the effect of the test agent on a behavior of the rodent in a depression assay, wherein reduction in a depressive behavior of the rodent contacted with the test agent, compared to the behavior of a control rodent that has not been contacted with the test agent, indicates that the test agent is a candidate agent for treating depression. 2. The method of claim 1, wherein the active optogenetic inhibitor is a halorhodopsin (NpHR) polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the NpHR amino acid sequence set forth in SEQ ID NO:1. 3. The method of claim 2, wherein the NpHR is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the NpHR in a dopaminergic neuron. 4. The method of claim 2, wherein the NpHR comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 5. The method of claim 1, wherein said determining is carried out after, or concurrently with, exposing the VTA to light at a wavelength that activates the optogenetic inhibitor. 6. The method of claim 1, wherein said depression assay is a forced swim test, a tail suspension test, or a conditioned place aversion test. 7. A transgenic rodent comprising dopaminergic neurons of the ventral tegmental area that express an optogenetic inhibitor of neuronal function. 8. The transgenic rodent of claim 7, wherein the optogenetic inhibitor of neuronal function is a halorhodopsin (NpHR) polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the NpHR amino acid sequence set forth in SEQ ID NO:1. 9. The transgenic rodent of claim 8, wherein the NpHR is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the NpHR in a dopaminergic neuron. 10. The transgenic rodent of claim 8, wherein the NpHR comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 11. A method for identifying a candidate agent for treating depression in an individual, comprising: contacting a rodent that expresses an active optogenetic activator of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with a test agent, and determining the effect of the test agent on a behavior of the rodent in a depression assay, wherein reduction in a depressive behavior of the rodent contacted with the test agent, compared to the behavior of a control rodent that has not been contacted with the test agent, indicates that the test agent is a candidate agent for treating depression. 12. The method of claim 11, wherein said determining is carried out without exposing the VTA to light at a wavelength that activates the optogenetic inhibitor. 13. The method of claim 11, wherein the optogenetic activator of neuronal activity is a channelrhodopsin polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the channelrhodopsin amino acid sequence set forth in SEQ ID NO:5. 14. The method of claim 13, wherein the channelrhodopsin is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the channelrhodopsin in a dopaminergic neuron. 15. The method of claim 13, wherein the channelrhodopsin comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 16. A method for screening an agent for the ability to promote depression in an individual, the method comprising: contacting a rodent that expresses an active optogenetic activator of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with an agent, and determining the effect of the agent on the behavior of the rodent in a depression assay, wherein a depressive behavior of the rodent contacted with the agent, compared to the behavior of a control rodent that has not been contacted with the agent, indicates that the agent promotes depression. 17. The method of claim 16, wherein the active optogenetic activator is a channelrhodopsin. 18. The method of claim 16, wherein the active optogenetic activator of neuronal activity in VTA dopaminergic neurons is activated upon exposure of the VTA to light of an activating wavelength. 19. A method for screening an agent for the ability to promote depression in an individual, the method comprising: contacting a rodent that expresses an active optogenetic activator of neuronal activity in medial prefrontal cortex (mPFC) excitatory neurons with an agent, and determining the effect of the agent on the behavior of the rodent in a depression assay, wherein a depressive behavior of the rodent contacted with the agent, compared to the behavior of a control rodent that has not been contacted with the agent, indicates that the agent promotes depression. 20. The method according to claim 19, wherein the active optogenetic activator is a channelrhodopsin. 21. The method according to claim 19, wherein said contacting is carried out before or concurrently with exposing the dorsal raphe nucleus (DRN) to light of a wavelength that activates the optogenetic activator. 22. A method for identifying a candidate agent for treating an adverse psychological state in an individual, the method comprising: contacting a rodent that expresses an active optogenetic inhibitor of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with a test agent, and determining the effect of the test agent on a behavior of the rodent in a conditioned place aversion (CPA) test, wherein modulation in the CPA response behavior of the rodent contacted with the test agent, compared to the behavior of a control rodent that has not been contacted with the test agent, indicates that the test agent is a candidate agent for treating an adverse psychological state in an individual. 23. The method of claim 22, wherein the adverse psychological state is dysphoria, anhedonia, depression, suicidality, or anxiety. 24. The method of claim 22, wherein the active optogenetic inhibitor is a halorhodopsin (NpHR) polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the NpHR amino acid sequence set forth in SEQ ID NO:1. 25. The method of claim 24, wherein the NpHR is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the NpHR in a dopaminergic neuron. 26. The method of claim 24, wherein the NpHR comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 27. The method of claim 22, wherein said determining is carried out after, or concurrently with, exposing the VTA to light at a wavelength that activates the optogenetic inhibitor.
The disclosure provides non-human optogenetic animal models of depression. Specifically, non-human animals each expresses a light-responsive opsin in a neuron of the animal are provided. The animal models are useful for identifying agents and targets of therapeutic strategies for treatment of depression. Examples of using the non-human animals expressing light-responsive opsin including Halorhodopsin family of light-responsive chloride pumps and Channelrhodopsin family of light-responsive cation channel proteins are described.1. A method for identifying a candidate agent treating depression in an individual, comprising: contacting a rodent that expresses an active optogenetic inhibitor of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with a test agent, and determining the effect of the test agent on a behavior of the rodent in a depression assay, wherein reduction in a depressive behavior of the rodent contacted with the test agent, compared to the behavior of a control rodent that has not been contacted with the test agent, indicates that the test agent is a candidate agent for treating depression. 2. The method of claim 1, wherein the active optogenetic inhibitor is a halorhodopsin (NpHR) polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the NpHR amino acid sequence set forth in SEQ ID NO:1. 3. The method of claim 2, wherein the NpHR is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the NpHR in a dopaminergic neuron. 4. The method of claim 2, wherein the NpHR comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 5. The method of claim 1, wherein said determining is carried out after, or concurrently with, exposing the VTA to light at a wavelength that activates the optogenetic inhibitor. 6. The method of claim 1, wherein said depression assay is a forced swim test, a tail suspension test, or a conditioned place aversion test. 7. A transgenic rodent comprising dopaminergic neurons of the ventral tegmental area that express an optogenetic inhibitor of neuronal function. 8. The transgenic rodent of claim 7, wherein the optogenetic inhibitor of neuronal function is a halorhodopsin (NpHR) polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the NpHR amino acid sequence set forth in SEQ ID NO:1. 9. The transgenic rodent of claim 8, wherein the NpHR is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the NpHR in a dopaminergic neuron. 10. The transgenic rodent of claim 8, wherein the NpHR comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 11. A method for identifying a candidate agent for treating depression in an individual, comprising: contacting a rodent that expresses an active optogenetic activator of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with a test agent, and determining the effect of the test agent on a behavior of the rodent in a depression assay, wherein reduction in a depressive behavior of the rodent contacted with the test agent, compared to the behavior of a control rodent that has not been contacted with the test agent, indicates that the test agent is a candidate agent for treating depression. 12. The method of claim 11, wherein said determining is carried out without exposing the VTA to light at a wavelength that activates the optogenetic inhibitor. 13. The method of claim 11, wherein the optogenetic activator of neuronal activity is a channelrhodopsin polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the channelrhodopsin amino acid sequence set forth in SEQ ID NO:5. 14. The method of claim 13, wherein the channelrhodopsin is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the channelrhodopsin in a dopaminergic neuron. 15. The method of claim 13, wherein the channelrhodopsin comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 16. A method for screening an agent for the ability to promote depression in an individual, the method comprising: contacting a rodent that expresses an active optogenetic activator of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with an agent, and determining the effect of the agent on the behavior of the rodent in a depression assay, wherein a depressive behavior of the rodent contacted with the agent, compared to the behavior of a control rodent that has not been contacted with the agent, indicates that the agent promotes depression. 17. The method of claim 16, wherein the active optogenetic activator is a channelrhodopsin. 18. The method of claim 16, wherein the active optogenetic activator of neuronal activity in VTA dopaminergic neurons is activated upon exposure of the VTA to light of an activating wavelength. 19. A method for screening an agent for the ability to promote depression in an individual, the method comprising: contacting a rodent that expresses an active optogenetic activator of neuronal activity in medial prefrontal cortex (mPFC) excitatory neurons with an agent, and determining the effect of the agent on the behavior of the rodent in a depression assay, wherein a depressive behavior of the rodent contacted with the agent, compared to the behavior of a control rodent that has not been contacted with the agent, indicates that the agent promotes depression. 20. The method according to claim 19, wherein the active optogenetic activator is a channelrhodopsin. 21. The method according to claim 19, wherein said contacting is carried out before or concurrently with exposing the dorsal raphe nucleus (DRN) to light of a wavelength that activates the optogenetic activator. 22. A method for identifying a candidate agent for treating an adverse psychological state in an individual, the method comprising: contacting a rodent that expresses an active optogenetic inhibitor of neuronal activity in ventral tegmental area (VTA) dopaminergic neurons with a test agent, and determining the effect of the test agent on a behavior of the rodent in a conditioned place aversion (CPA) test, wherein modulation in the CPA response behavior of the rodent contacted with the test agent, compared to the behavior of a control rodent that has not been contacted with the test agent, indicates that the test agent is a candidate agent for treating an adverse psychological state in an individual. 23. The method of claim 22, wherein the adverse psychological state is dysphoria, anhedonia, depression, suicidality, or anxiety. 24. The method of claim 22, wherein the active optogenetic inhibitor is a halorhodopsin (NpHR) polypeptide comprising an amino acid sequence having at least about 95% amino acid sequence identity to the NpHR amino acid sequence set forth in SEQ ID NO:1. 25. The method of claim 24, wherein the NpHR is encoded by a nucleotide sequence that is operably linked to a promoter that provides for expression of the NpHR in a dopaminergic neuron. 26. The method of claim 24, wherein the NpHR comprises an endoplasmic reticulum export signal and a membrane trafficking signal. 27. The method of claim 22, wherein said determining is carried out after, or concurrently with, exposing the VTA to light at a wavelength that activates the optogenetic inhibitor.
1,600
226
226
13,639,752
1,612
The invention relates to novel pharmaceutical compositions highly dosed with biotin, as well as to the use thereof in treating visual impairments which are related, in particular, to optic atrophy.
1. A composition for oral administration, containing more than 20 mg of biotin per unit dose. 2. The composition as claimed in claim 1, characterized in that it is in the form of gel capsules, tablets (optionally film-coated), lozenges or pills. 3. The composition as claimed in claim 1, characterized in that it contains biotin and excipients, without any other active ingredient. 4. The composition as claimed in claim 1, characterized in that it contains excipients chosen from talc and microcrystalline cellulose. 5-8. (canceled) 9. A method for the treatment of a visual impairment or of a visual atrophy comprising administering biotin to a patient in need thereof. 10. The method for the treatment of a visual impairment as claimed in claim 9, wherein the amount of biotin administered to the patient is at least equal to 1 mg/kg/day. 11. The method for the treatment of a visual impairment as claimed in claim 9, wherein the visual impairment is related to leukoencephalopathy involving the following regions of the white matter of the brain: periventricular white matter, optic radiations, corticospinal tracts, cerebellar peduncles, as observed by brain MRI, and an elevation of the choline peak in the centrum semiovale, as observed by nuclear magnetic resonance spectroscopy. 12. A method for the treatment of a neurological pathological condition related to an energy metabolism disorder comprising administering biotin to a patient in need thereof.
The invention relates to novel pharmaceutical compositions highly dosed with biotin, as well as to the use thereof in treating visual impairments which are related, in particular, to optic atrophy.1. A composition for oral administration, containing more than 20 mg of biotin per unit dose. 2. The composition as claimed in claim 1, characterized in that it is in the form of gel capsules, tablets (optionally film-coated), lozenges or pills. 3. The composition as claimed in claim 1, characterized in that it contains biotin and excipients, without any other active ingredient. 4. The composition as claimed in claim 1, characterized in that it contains excipients chosen from talc and microcrystalline cellulose. 5-8. (canceled) 9. A method for the treatment of a visual impairment or of a visual atrophy comprising administering biotin to a patient in need thereof. 10. The method for the treatment of a visual impairment as claimed in claim 9, wherein the amount of biotin administered to the patient is at least equal to 1 mg/kg/day. 11. The method for the treatment of a visual impairment as claimed in claim 9, wherein the visual impairment is related to leukoencephalopathy involving the following regions of the white matter of the brain: periventricular white matter, optic radiations, corticospinal tracts, cerebellar peduncles, as observed by brain MRI, and an elevation of the choline peak in the centrum semiovale, as observed by nuclear magnetic resonance spectroscopy. 12. A method for the treatment of a neurological pathological condition related to an energy metabolism disorder comprising administering biotin to a patient in need thereof.
1,600
227
227
13,656,413
1,613
Pharmaceutical preparations, compositions, systems, and devices including medical devices and diagnostic or therapeutic agents, and methods to treat disease by modification of local tissue environment to modulate the therapeutic index of locally or systemically delivered therapeutic or diagnostic agents. Improved ability to reduce sympathetic nerve activity in the adventitia and perivascular tissues around arteries and veins in the body. Modulation of the local tissue environment around an artery to enable more effective denervation with or without a therapeutic agent. Modulation may include adjustment of the pH of the tissue.
1. A composition comprising a guanidine with pH>8. 2. The composition of claim 1, wherein the guanidine is guanethidine. 3. The composition of claim 2, wherein the guanethidine includes monosulfate, hemisulfate in a solution configured for denervation, or hemisulfate in a solution suitable for denervation. 4. (canceled) 5. (canceled) 6. The composition of claim 1, wherein the pH>9, or the pH>10. 7. (canceled) 8. The composition of claim 1, further comprising an alkaline buffer. 9. The composition of claim 8, wherein the alkaline buffer comprises NaOH, NaOH in a molar ratio to the guanidine concentration of 50% or greater, or NaOH in an equimolar or greater concentration to the guanidine. 10. (canceled) 11. (canceled) 12. (canceled) 13. The composition of claim 1, wherein the composition further comprises sodium chloride that is 0.7% to 0.9% of the solution. 14. (canceled) 15. The composition of claim 3, wherein the guanethidine monosulfate is in concentration of 0.1 mg/mL to 50 mg/mL, or in concentration of 1 mg/mL to 20 mg/mL. 16. (canceled) 17. A method for modulating local tissue physiology comprising the delivery of preparation comprising a liquid, gel, or semisolid into the tissue, wherein the preparation buffers the local tissue physiology by raising or lowering the pH of the local tissue. 18. (canceled) 19. The method of claim 17, wherein the preparation comprises a therapeutic agent that has its index effect at a physiological condition modulated by the delivery of such preparation, but not having an index effect at neutral physiological condition. 20. The method of claim 17, wherein the preparation further includes a therapeutic agent that has additional or enhanced index effect at a physiological condition modulated by the delivery of such preparation, but not having such additional or enhanced index effect at neutral physiological condition. 21. The method of claim 17, wherein the gel comprises a hydrogel that consumes protons as it resorbs in the tissue. 22. (canceled) 23. (canceled) 24. The method of claim 17, wherein the preparation includes guanethidine monosulfate, or has a pH>8, or includes guanethidine monosulfate and has a pH>8. 25. (canceled) 26. (canceled) 27. A method of creating renal denervation comprising the localized delivery of an acid or base with sufficiently low or high pH to create localized nerve damage or destruction. 28. A method of creating renal denervation comprising the localized delivery of a non-isotonic or non-isoosmolar solution that creates neuronal destruction while sparing other local tissues. 29. A method comprising the delivery of a preparation of guanethidine monosulfate at pH>8 or guanethidine hemisulfate at pH>8 into the renal artery adventitia and perivascular tissues which is effective in treating hypertension, in treating heart failure, in treating insulin resistance, in treating systemic inflammation, or in treating sleep apnea. 30. (canceled) 31. (canceled) 32. (canceled) 33. (canceled) 34. (canceled) 35. (canceled) 36. (canceled) 37. (canceled) 38. A method of creating renal denervation comprising the localized delivery of a first toxin entering cells through sodium channels, wherein such first toxin comprises one or more of: tetrodotoxin and batrachotoxin, a second toxin entering cells through potassium channels, wherein such second toxin comprises one or more of: maurotoxin, agitoxin, charybdotoxin, margatoxin, slotoxin, sycllatoxin and hefutoxin, and/or a third toxin entering cells through calcium channels, wherein such third toxin comprises one or more of: calciseptine, taicatoxin, calcicludine and PhTx3. 39. A method of creating renal denervation comprising the localized delivery of an agent, wherein the agent itself or a composition comprising such agent has a pH of at least 7, a pH of at most 11, a pH of at least 7 and at most 11, a pH of at least 8 and at most 10, a pH that is effective to denervate nerves to which such agent is delivered, or a pH that is adjusted to a level that is effective to denervate nerves to which such agent is delivered; wherein the agent itself or a composition comprising such agent has a pH of at most 7, a pH of at least 3, a pH of at most 7 and at least 3, a pH of at most 6 and at least 4, an acidic pH that is effective to denervate nerves to which such agent is delivered, or an acidic pH that is adjusted to a level that is effective to denervate nerves to which such agent is delivered; wherein tissue into which the agent is delivered is modulated to a pH of at least 7, a pH of at most 11, a pH of at least 7 and at most 11, a pH of at least 8 and at most 10, or to a pH that is effective to denervate nerves to which such agent is delivered; or wherein tissue into which the agent is delivered is modulated to a pH of at most 7, a pH of at least 3, a pH of at most 7 and at least 3, a pH of at most 6 and at least 4, an acidic pH that is effective to denervate nerves to which such agent is delivered. 40. The method of claim 39, wherein the agent comprises an adrenergic blocker, an androgenic inhibitor, an adrenergic stimulator, an Alpha-/beta-adrenergic blocker, an angiotensin converting enzyme (ACE) inhibitor, an ACE-receptor antagonist, a Beta blocker, a calcium channel blocker, an antiarrythmic of groups I-IV, an antiarrythmic, a cardiotonic, an alpha-2-agonists, a guanidine derivative, an imadazoline receptor agonist, a ganglion-blocking agent, nicotinic antagonist, ganglion-blocking agents, nicotinic antagonist, a MAOI inhibitor, an adrenergic uptake inhibitor, a tyrosine hydroxylase inhibitors, an alpha-1 blocker, a non-selective alpha blocker, a serotonin antagonist, an endothelin antagonist, a sclerosing agent, a sensory denervation agent, doxazosin, guanadrel, guanethidine, pheoxybenzamine, prazosin plus polythiazide, terazosin, methyldopa, clonidine, guanabenz, guanfacine, Labetalol, benazepril, catopril, enalapril, enalaprilat, fosinopril, lisinopril, moexipril, quinapril, ramipril, and combinations with calcium channel blockers and diuretics, losartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, esmolol, fimolol, pindolol, propranolol, penbatolol, metoprolol, nadolol, sotalol, Amiloride, amlodipine, bepridil, diltiazem, isradipine, nifedipine, verapamil, felodipine, nicardipine, nimodipine, bretylium, disopyramide, encamide, flecamide, lidocaine, mexiletine, moricizine, propafenone, procainamide, quinidine, tocamide, esmolol, propranolol, acebutolol, amiodarone, sotalol, verapamil, diltiazem, pindolol, bupranolol hydrochloride, trichlormethiazide, furosemide, prazosin hydrochloride, metoprolol tartrate, carteolol hydrochloride, oxprenolol hydrochloride, and propranolol hydrochloride, adenosine, digoxin; metildigoxin, caffeine, dopamine hydrochloride, dobutamine hydrochloride, octopamine hydrochloride, diprophylline, ubidecarenon, digitalis, capsaicin, anti-nerve growth factor, anti-dopamine beta-hydroxylase, anti-acetylcholinesterase, 6-hydroxyldopamine (6-OHDA), bretylium tosylate, guanacline, and N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4), OX7-SAP, 192-SAP, anti-dopamine beta-hydroxylase saporin (DBH-SAP), and anti-dopamine beta-hydroxylase immunotoxin (DHIT), phenol, ethanol, clonidine, guanfacine, methyldopa, betanidine, guanoxan, debrisoquine, guanoclor, guanazodine, guanoxabenz, moxonidine, relmenidine, mecamylamine, trimethaphan, pargyline, rescinnamine, reserpine, metirosine, prazosin, indoramin, trimazosin, doxazosin, urapidil, phentolamine, ketanserin, bosentan, ambrisentan, sitaxentan, quinacrine, chloroquine, sodium tetradecyl sulfate, ethanolamine oleate, sodium morrhuate, polidocanol, or a hypertonic solution. 41. (canceled) 42. (canceled) 43. The method of claim 39, wherein the tissue is modulated before, after or during the delivery of the agent to the tissue. 44. (canceled) 45. (canceled) 46. (canceled) 47. A method for enhancing the uptake of therapeutic agents into tissue comprising modulating pH of the tissue by creating a zone of the tissue having a center and an outer edge, and delivering a therapeutic agent into the zone, wherein the zone comprises a modulated pH as compared to a pre-modulation pH of the tissue prior to modulation or as compared to a neutral pH, wherein zone comprises a gradient of pH that is most modulated at the center of the zone and reduces to the pre-modulation pH of the tissue or to neutral pH at the outer edge of the zone, and wherein enhanced uptake of a therapeutic agent occurs in the zone as compared to uptake that would occur into tissue at the pre-modulation pH or at neutral pH. 48. (canceled) 49. The method of claim 47, wherein the therapeutic agent is delivered systemically and the modulating of the tissue pH enhances a buildup of the therapeutic agent in the zone or improves a therapeutic index in the zone. 50. (canceled) 51. (canceled) 52. (canceled) 53. (canceled) 54. (canceled) 55. The method of claim 47, wherein the therapeutic agent comprises guanethidine monosulfate or guanethidine hemisulfate. 56. (canceled) 57. The method of claim 47, wherein the modulated pH at least 7, at most 11, at least 7 and at most 11, at least 8 and at most 10, or a predetermined pH that is effective to denervate nerves to which such therapeutic agent is delivered. 58. The method of claim 39, wherein the agent is chosen from the following: guanethidine in a concentration ranging from 1 μg/mL to 50 mg/mL at pH of greater than 7, guanethidine in a concentration ranging from 1 mg/mL to 30 mg/mL at pH of greater than 7, a composition comprising guanethidine having a sodium chloride content between 0.7% and 0.9%, a composition comprising guanethidine having pH of about 9.5, a composition comprising guanethidine having pH that is adjusted to about 9.5 by buffering with an alkaline buffer agent, a composition comprising guanethidine having pH that is adjusted to about 9.5 by buffering with sodium hydroxide, or a composition comprising guanethidine having pH of between 8 and 11.
Pharmaceutical preparations, compositions, systems, and devices including medical devices and diagnostic or therapeutic agents, and methods to treat disease by modification of local tissue environment to modulate the therapeutic index of locally or systemically delivered therapeutic or diagnostic agents. Improved ability to reduce sympathetic nerve activity in the adventitia and perivascular tissues around arteries and veins in the body. Modulation of the local tissue environment around an artery to enable more effective denervation with or without a therapeutic agent. Modulation may include adjustment of the pH of the tissue.1. A composition comprising a guanidine with pH>8. 2. The composition of claim 1, wherein the guanidine is guanethidine. 3. The composition of claim 2, wherein the guanethidine includes monosulfate, hemisulfate in a solution configured for denervation, or hemisulfate in a solution suitable for denervation. 4. (canceled) 5. (canceled) 6. The composition of claim 1, wherein the pH>9, or the pH>10. 7. (canceled) 8. The composition of claim 1, further comprising an alkaline buffer. 9. The composition of claim 8, wherein the alkaline buffer comprises NaOH, NaOH in a molar ratio to the guanidine concentration of 50% or greater, or NaOH in an equimolar or greater concentration to the guanidine. 10. (canceled) 11. (canceled) 12. (canceled) 13. The composition of claim 1, wherein the composition further comprises sodium chloride that is 0.7% to 0.9% of the solution. 14. (canceled) 15. The composition of claim 3, wherein the guanethidine monosulfate is in concentration of 0.1 mg/mL to 50 mg/mL, or in concentration of 1 mg/mL to 20 mg/mL. 16. (canceled) 17. A method for modulating local tissue physiology comprising the delivery of preparation comprising a liquid, gel, or semisolid into the tissue, wherein the preparation buffers the local tissue physiology by raising or lowering the pH of the local tissue. 18. (canceled) 19. The method of claim 17, wherein the preparation comprises a therapeutic agent that has its index effect at a physiological condition modulated by the delivery of such preparation, but not having an index effect at neutral physiological condition. 20. The method of claim 17, wherein the preparation further includes a therapeutic agent that has additional or enhanced index effect at a physiological condition modulated by the delivery of such preparation, but not having such additional or enhanced index effect at neutral physiological condition. 21. The method of claim 17, wherein the gel comprises a hydrogel that consumes protons as it resorbs in the tissue. 22. (canceled) 23. (canceled) 24. The method of claim 17, wherein the preparation includes guanethidine monosulfate, or has a pH>8, or includes guanethidine monosulfate and has a pH>8. 25. (canceled) 26. (canceled) 27. A method of creating renal denervation comprising the localized delivery of an acid or base with sufficiently low or high pH to create localized nerve damage or destruction. 28. A method of creating renal denervation comprising the localized delivery of a non-isotonic or non-isoosmolar solution that creates neuronal destruction while sparing other local tissues. 29. A method comprising the delivery of a preparation of guanethidine monosulfate at pH>8 or guanethidine hemisulfate at pH>8 into the renal artery adventitia and perivascular tissues which is effective in treating hypertension, in treating heart failure, in treating insulin resistance, in treating systemic inflammation, or in treating sleep apnea. 30. (canceled) 31. (canceled) 32. (canceled) 33. (canceled) 34. (canceled) 35. (canceled) 36. (canceled) 37. (canceled) 38. A method of creating renal denervation comprising the localized delivery of a first toxin entering cells through sodium channels, wherein such first toxin comprises one or more of: tetrodotoxin and batrachotoxin, a second toxin entering cells through potassium channels, wherein such second toxin comprises one or more of: maurotoxin, agitoxin, charybdotoxin, margatoxin, slotoxin, sycllatoxin and hefutoxin, and/or a third toxin entering cells through calcium channels, wherein such third toxin comprises one or more of: calciseptine, taicatoxin, calcicludine and PhTx3. 39. A method of creating renal denervation comprising the localized delivery of an agent, wherein the agent itself or a composition comprising such agent has a pH of at least 7, a pH of at most 11, a pH of at least 7 and at most 11, a pH of at least 8 and at most 10, a pH that is effective to denervate nerves to which such agent is delivered, or a pH that is adjusted to a level that is effective to denervate nerves to which such agent is delivered; wherein the agent itself or a composition comprising such agent has a pH of at most 7, a pH of at least 3, a pH of at most 7 and at least 3, a pH of at most 6 and at least 4, an acidic pH that is effective to denervate nerves to which such agent is delivered, or an acidic pH that is adjusted to a level that is effective to denervate nerves to which such agent is delivered; wherein tissue into which the agent is delivered is modulated to a pH of at least 7, a pH of at most 11, a pH of at least 7 and at most 11, a pH of at least 8 and at most 10, or to a pH that is effective to denervate nerves to which such agent is delivered; or wherein tissue into which the agent is delivered is modulated to a pH of at most 7, a pH of at least 3, a pH of at most 7 and at least 3, a pH of at most 6 and at least 4, an acidic pH that is effective to denervate nerves to which such agent is delivered. 40. The method of claim 39, wherein the agent comprises an adrenergic blocker, an androgenic inhibitor, an adrenergic stimulator, an Alpha-/beta-adrenergic blocker, an angiotensin converting enzyme (ACE) inhibitor, an ACE-receptor antagonist, a Beta blocker, a calcium channel blocker, an antiarrythmic of groups I-IV, an antiarrythmic, a cardiotonic, an alpha-2-agonists, a guanidine derivative, an imadazoline receptor agonist, a ganglion-blocking agent, nicotinic antagonist, ganglion-blocking agents, nicotinic antagonist, a MAOI inhibitor, an adrenergic uptake inhibitor, a tyrosine hydroxylase inhibitors, an alpha-1 blocker, a non-selective alpha blocker, a serotonin antagonist, an endothelin antagonist, a sclerosing agent, a sensory denervation agent, doxazosin, guanadrel, guanethidine, pheoxybenzamine, prazosin plus polythiazide, terazosin, methyldopa, clonidine, guanabenz, guanfacine, Labetalol, benazepril, catopril, enalapril, enalaprilat, fosinopril, lisinopril, moexipril, quinapril, ramipril, and combinations with calcium channel blockers and diuretics, losartan, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, esmolol, fimolol, pindolol, propranolol, penbatolol, metoprolol, nadolol, sotalol, Amiloride, amlodipine, bepridil, diltiazem, isradipine, nifedipine, verapamil, felodipine, nicardipine, nimodipine, bretylium, disopyramide, encamide, flecamide, lidocaine, mexiletine, moricizine, propafenone, procainamide, quinidine, tocamide, esmolol, propranolol, acebutolol, amiodarone, sotalol, verapamil, diltiazem, pindolol, bupranolol hydrochloride, trichlormethiazide, furosemide, prazosin hydrochloride, metoprolol tartrate, carteolol hydrochloride, oxprenolol hydrochloride, and propranolol hydrochloride, adenosine, digoxin; metildigoxin, caffeine, dopamine hydrochloride, dobutamine hydrochloride, octopamine hydrochloride, diprophylline, ubidecarenon, digitalis, capsaicin, anti-nerve growth factor, anti-dopamine beta-hydroxylase, anti-acetylcholinesterase, 6-hydroxyldopamine (6-OHDA), bretylium tosylate, guanacline, and N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4), OX7-SAP, 192-SAP, anti-dopamine beta-hydroxylase saporin (DBH-SAP), and anti-dopamine beta-hydroxylase immunotoxin (DHIT), phenol, ethanol, clonidine, guanfacine, methyldopa, betanidine, guanoxan, debrisoquine, guanoclor, guanazodine, guanoxabenz, moxonidine, relmenidine, mecamylamine, trimethaphan, pargyline, rescinnamine, reserpine, metirosine, prazosin, indoramin, trimazosin, doxazosin, urapidil, phentolamine, ketanserin, bosentan, ambrisentan, sitaxentan, quinacrine, chloroquine, sodium tetradecyl sulfate, ethanolamine oleate, sodium morrhuate, polidocanol, or a hypertonic solution. 41. (canceled) 42. (canceled) 43. The method of claim 39, wherein the tissue is modulated before, after or during the delivery of the agent to the tissue. 44. (canceled) 45. (canceled) 46. (canceled) 47. A method for enhancing the uptake of therapeutic agents into tissue comprising modulating pH of the tissue by creating a zone of the tissue having a center and an outer edge, and delivering a therapeutic agent into the zone, wherein the zone comprises a modulated pH as compared to a pre-modulation pH of the tissue prior to modulation or as compared to a neutral pH, wherein zone comprises a gradient of pH that is most modulated at the center of the zone and reduces to the pre-modulation pH of the tissue or to neutral pH at the outer edge of the zone, and wherein enhanced uptake of a therapeutic agent occurs in the zone as compared to uptake that would occur into tissue at the pre-modulation pH or at neutral pH. 48. (canceled) 49. The method of claim 47, wherein the therapeutic agent is delivered systemically and the modulating of the tissue pH enhances a buildup of the therapeutic agent in the zone or improves a therapeutic index in the zone. 50. (canceled) 51. (canceled) 52. (canceled) 53. (canceled) 54. (canceled) 55. The method of claim 47, wherein the therapeutic agent comprises guanethidine monosulfate or guanethidine hemisulfate. 56. (canceled) 57. The method of claim 47, wherein the modulated pH at least 7, at most 11, at least 7 and at most 11, at least 8 and at most 10, or a predetermined pH that is effective to denervate nerves to which such therapeutic agent is delivered. 58. The method of claim 39, wherein the agent is chosen from the following: guanethidine in a concentration ranging from 1 μg/mL to 50 mg/mL at pH of greater than 7, guanethidine in a concentration ranging from 1 mg/mL to 30 mg/mL at pH of greater than 7, a composition comprising guanethidine having a sodium chloride content between 0.7% and 0.9%, a composition comprising guanethidine having pH of about 9.5, a composition comprising guanethidine having pH that is adjusted to about 9.5 by buffering with an alkaline buffer agent, a composition comprising guanethidine having pH that is adjusted to about 9.5 by buffering with sodium hydroxide, or a composition comprising guanethidine having pH of between 8 and 11.
1,600
228
228
15,425,713
1,613
A skin care composition in the form of a water-in-oil, high internal phase emulsion that provides an improved sensory experience when applied to keratinous tissue. The thick, creamy consistency of the emulsion provides a moisturization signal to the consumer, but the shear thinning behavior of the high internal phase emulsion in combination with the superabsorbent polymer in the internal aqueous phase provide a light, cool, silky feel. The composition includes a non-thickening, superabsorbent polymer in the aqueous phase, but does not form visible beads of water when applied to skin.
1. A personal care composition in the form of a water-in-oil, high internal phase emulsion, comprising: a. more than about 74%, by volume, of an internal aqueous phase; b. about 0.01% to about 3%, by weight, of a superabsorbent polymer (SAP) in the aqueous phase; and c. less than about 26%, by volume, of a continuous oil phase. 2. The composition of claim 1, further comprising a viscosity of between about 100 and 10,000 Pa-s. 3. The composition of claim 2, wherein the SAP does not substantially affect the viscosity of the composition. 4. The composition of claim 2, wherein the SAP does not change the viscosity of the composition by more than 10% as compared to an identical composition that does not include the SAP. 5. The composition of claim 2, wherein the viscosity of the personal care composition is substantially constant prior to the yield-point. 6. The composition of claim 1, wherein the internal aqueous phase is in the form of a multitude of droplets having a droplet size of between 10 and 250 microns. 7. The composition of claim 1, wherein the SAP is in the form of a multitude of particles that have a dry particle size of between about 2 microns and 100 microns. 8. The composition of claim 7, wherein the SAP has a swollen number-average particle size of between about 10 microns and about 250 microns. 9. The composition of claim 1, wherein the aqueous phase of the composition does not form visible beads when applied to a target portion of skin. 10. The composition of claim 1, wherein the personal care composition exhibits phase separation at a shear stress of between 10 and 100 Pa, according the Viscosity Method. 11. The composition of claim 1, further comprising about 0.01 to about 10% of an emulsifier. 12. The composition of claim 1, further comprising an active or agent selected from a group consisting of sugar amines, vitamins, oil control agents, humectants, emollients, phytosterols, hexamidine compounds, tightening agents, anti-wrinkle actives, anti-atrophy actives, flavonoids, N-acyl amino acid compounds, retinoids, peptides, particulate materials, anti-cellulite agents, desquamation actives, anti-acne actives, anti-oxidants, radical scavengers, conditioning agents, anti-inflammatory agents, tanning actives, skin lightening agents, botanical extracts, antimicrobial actives, antifungal actives, antibacterial actives, antiperspirant actives, sensates, preservatives, anti-dandruff actives, substantivity polymers, detersive surfactants, and combinations thereof. 13. The composition of claim 1 wherein the superabsorbent polymer is selected from the group consisting of sodium polyacrylate, sodium polyacrylate starch, sodium acrylates crosspolymer-2 and mixtures thereof. 14. The composition of claim 1, further comprising a silicone elastomer. 15. The composition of claim 14, wherein the silicone elastomer is a silicone elastomer gel. 16. A personal care composition in the form of a water-in-oil, high internal phase emulsion, comprising: a. more than about 74%, by volume, of an internal aqueous phase in the form of droplets having a droplet size of between about 10 and about 150 microns; b. about 0.1% to about 1%, by weight, of a superabsorbent polymer in the aqueous phase, the superabsorbent polymer being in the form of a multitude of particles having a number-average swollen particle size of between about 10 and about 150 microns; c. less than about 26%, by volume, of a continuous oil phase comprising at least one silicone elastomer; and d. a viscosity of between about 100 and about 10,000 Pa-s, wherein the aqueous phase does not form visible beads when applied to a target portion of skin. 17.-19. (canceled)
A skin care composition in the form of a water-in-oil, high internal phase emulsion that provides an improved sensory experience when applied to keratinous tissue. The thick, creamy consistency of the emulsion provides a moisturization signal to the consumer, but the shear thinning behavior of the high internal phase emulsion in combination with the superabsorbent polymer in the internal aqueous phase provide a light, cool, silky feel. The composition includes a non-thickening, superabsorbent polymer in the aqueous phase, but does not form visible beads of water when applied to skin.1. A personal care composition in the form of a water-in-oil, high internal phase emulsion, comprising: a. more than about 74%, by volume, of an internal aqueous phase; b. about 0.01% to about 3%, by weight, of a superabsorbent polymer (SAP) in the aqueous phase; and c. less than about 26%, by volume, of a continuous oil phase. 2. The composition of claim 1, further comprising a viscosity of between about 100 and 10,000 Pa-s. 3. The composition of claim 2, wherein the SAP does not substantially affect the viscosity of the composition. 4. The composition of claim 2, wherein the SAP does not change the viscosity of the composition by more than 10% as compared to an identical composition that does not include the SAP. 5. The composition of claim 2, wherein the viscosity of the personal care composition is substantially constant prior to the yield-point. 6. The composition of claim 1, wherein the internal aqueous phase is in the form of a multitude of droplets having a droplet size of between 10 and 250 microns. 7. The composition of claim 1, wherein the SAP is in the form of a multitude of particles that have a dry particle size of between about 2 microns and 100 microns. 8. The composition of claim 7, wherein the SAP has a swollen number-average particle size of between about 10 microns and about 250 microns. 9. The composition of claim 1, wherein the aqueous phase of the composition does not form visible beads when applied to a target portion of skin. 10. The composition of claim 1, wherein the personal care composition exhibits phase separation at a shear stress of between 10 and 100 Pa, according the Viscosity Method. 11. The composition of claim 1, further comprising about 0.01 to about 10% of an emulsifier. 12. The composition of claim 1, further comprising an active or agent selected from a group consisting of sugar amines, vitamins, oil control agents, humectants, emollients, phytosterols, hexamidine compounds, tightening agents, anti-wrinkle actives, anti-atrophy actives, flavonoids, N-acyl amino acid compounds, retinoids, peptides, particulate materials, anti-cellulite agents, desquamation actives, anti-acne actives, anti-oxidants, radical scavengers, conditioning agents, anti-inflammatory agents, tanning actives, skin lightening agents, botanical extracts, antimicrobial actives, antifungal actives, antibacterial actives, antiperspirant actives, sensates, preservatives, anti-dandruff actives, substantivity polymers, detersive surfactants, and combinations thereof. 13. The composition of claim 1 wherein the superabsorbent polymer is selected from the group consisting of sodium polyacrylate, sodium polyacrylate starch, sodium acrylates crosspolymer-2 and mixtures thereof. 14. The composition of claim 1, further comprising a silicone elastomer. 15. The composition of claim 14, wherein the silicone elastomer is a silicone elastomer gel. 16. A personal care composition in the form of a water-in-oil, high internal phase emulsion, comprising: a. more than about 74%, by volume, of an internal aqueous phase in the form of droplets having a droplet size of between about 10 and about 150 microns; b. about 0.1% to about 1%, by weight, of a superabsorbent polymer in the aqueous phase, the superabsorbent polymer being in the form of a multitude of particles having a number-average swollen particle size of between about 10 and about 150 microns; c. less than about 26%, by volume, of a continuous oil phase comprising at least one silicone elastomer; and d. a viscosity of between about 100 and about 10,000 Pa-s, wherein the aqueous phase does not form visible beads when applied to a target portion of skin. 17.-19. (canceled)
1,600
229
229
14,162,181
1,629
Provided is a compound useful in the prevention and treatment of non-alcoholic steatohepatitis. A prophylactic and/or therapeutic agent for non-alcoholic steatohepatitis which contains, as the active ingredient, 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same.
1. A method of treating non-alcoholic steatohepatitis comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof. 2. The method according to claim 1, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 3. A method of reducing the levels of cholesterol in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient with fatty liver. 4. A method of reducing the levels of triglyceride in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient with fatty liver. 5. A method of treating the inflammation in the liver with TNF-α or macrophage CD11c expression comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient with hepatic inflammation. 6. A method of treating liver damage comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof, wherein the levels of plasma alanine aminotransferase (ALT) is enhanced in the patient. 7. A method of inhibiting the development of lipid-droplet deposition in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof. 8. A method of inhibiting the development of fibrosis in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof. 9. The method according claim 3, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 10. The method according claim 4, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 11. The method according claim 5, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 12. The method according claim 6, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 13. The method according claim 7, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 14. The method according claim 8, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide.
Provided is a compound useful in the prevention and treatment of non-alcoholic steatohepatitis. A prophylactic and/or therapeutic agent for non-alcoholic steatohepatitis which contains, as the active ingredient, 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same.1. A method of treating non-alcoholic steatohepatitis comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof. 2. The method according to claim 1, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 3. A method of reducing the levels of cholesterol in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient with fatty liver. 4. A method of reducing the levels of triglyceride in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient with fatty liver. 5. A method of treating the inflammation in the liver with TNF-α or macrophage CD11c expression comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient with hepatic inflammation. 6. A method of treating liver damage comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof, wherein the levels of plasma alanine aminotransferase (ALT) is enhanced in the patient. 7. A method of inhibiting the development of lipid-droplet deposition in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof. 8. A method of inhibiting the development of fibrosis in the liver comprising: administering a drug comprising an effective amount of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same to a patient in need thereof. 9. The method according claim 3, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 10. The method according claim 4, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 11. The method according claim 5, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 12. The method according claim 6, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 13. The method according claim 7, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide. 14. The method according claim 8, wherein 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide, an acid-addition salt thereof, or a solvate of the same is monohydrochloride of 2-[4-[2-(benzimidazol-2-ylthio)ethyl]piperazin-1-yl]-N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]acetamide.
1,600
230
230
13,618,803
1,631
A system and method whereby acoustic signals can be classified and identified as to nature and location of the original signal. The system and method determine from an arbitrary set of signals a signature or other characterizing feature and distinguish signals associated with a plurality of conditions by means of dictionaries comprising atoms of signals.
1. A system for extracting, detecting, classifying, discriminating, localizing, and processing signals comprising: a. at least one transducer device operable to collect a plurality of signals; b. a processing device coupled to said transducer device, said processing device executing instructions for decomposing the signals into representational spaces matched with one another, each said representational space defining a multi-dimensional set of atoms, wherein the decomposing comprises performing simultaneous sparse approximations of the signals, each signal being thereby represented by a combination of atoms selected from said representational space, each selected atom being weighted by a corresponding coefficient; and, c. a computer memory coupled to said processor for storing said representational spaces. 2. The system as recited in claim 1, further comprising a detection unit coupled to said processing device, said detection unit selectively determining a match between a set of said signals responsive to respective representational spaces thereof. 3. The system as recited in claim 1, wherein said transducer device collects includes at least one sensor. 4. The system as recited in claim 1, wherein said processing device executes a Greedy Adaptive Discrimination (GAD) decomposition operating with a dictionary of prototype atoms. 5. The system as recited in claim 4, wherein the dictionary comprises a set of Gabor atoms together with Fourier and delta function bases. 6. The system as recited in claim 1, wherein the said atoms are selected from a random set of atoms. 7. The system as recited in claim 1, wherein each said atom selected for said representation of at least one signal is caused to be orthogonally related to at least one other atom in said representational space for said signal. 8. The system as recited in claim 1, wherein said processor device generates a set of test atoms based on said representational spaces, and defines a dictionary of said test atoms, a subsequently collected signal being decomposed in terms of test atoms selected from said dictionary, whereby a degree of similarity is determined for the subsequently collected signal relative to said representational spaces of previously collected signals. 9. The system as recited in claim 1, comprising a plurality of said transducer devices collecting signals through a plurality of channels, the signals of at least one said channel being alignment shifted in a predetermined measurement space relative to the signals of at least one other of said channels. 10. A method of signature extraction comprising the steps of: a. establishing two or more analysis sets, each set comprising at least one signal; b. decomposing the analysis sets into matched structure books, wherein the decomposing comprises executing a processing device to perform simultaneous sparse approximations of the signals of the analysis sets; each said structure book including at least one representation defining a multi-dimensional combination of coefficients and atoms; and, c. storing said structure hooks in a computer memory. 11. The method as recited in claim 10, further comprising actuating the processing device to produce a parametric mean structure book representative of a signature for each set of signals. 12. The method as recited in claim 10, wherein a separate simultaneous sparse approximation is performed for each analysis set. 13. The method as recited in claim 10, wherein a single simultaneous sparse approximation is performed for an aggregate collection of signals from a plurality of analysis sets. 14. The method as recited in claim 10, wherein at least one of the signals is alignment shifted in a predetermined measurement space relative to another of the signals before simultaneous sparse approximation. 15. The method as recited in claim 10, wherein the signals are collected at a plurality of different locations, the signals collected at each location determining at least one analysis set, a location specific signature being determined based on the resulting structure books. 16. A method of detecting, classifying, discriminating, and localizing signals comprising the steps of: a. collecting a set of signals; b. decomposing the sets of signals into matched structure books, wherein the decomposing comprises executing a processing device to perform simultaneous sparse approximations of the signals; each said structure book defining a multi-dimensional combination of coefficients and atoms; the simultaneous sparse approximation being executed with respect to a dictionary defined relative to previously determined signatures; and, c. comparing the resulting structure books to structure books of the previously determined si matures. 17. The method as recited in claim 16, wherein a single simultaneous sparse approximation is formed for an aggregate collection of signals from at least two groups of signals, each defining one of said analysis sets. 18. The method as recited in claim 16, wherein step c is accomplished by examining the values of the coefficients of the atoms in the resulting structure books compared to typical values for similar atoms of one or more known signatures. 19. The method as recited in claim 16, wherein the simultaneous sparse approximation is accomplished with respect to a dictionary comprising an aggregate collection of atoms present in previously extracted structure books of known signatures of interest. 20. The method as recited in claim 19, wherein the dictionary additionally includes the aggregate collection of all atoms selected during the sparse approximation that produced the previously extracted structure books of known signatures of interest, whether or not said atoms are present in said structure books of signatures of interest. 21. The method as recited in claim 16, wherein a plurality of signals are collected and at least one of the signals is alignment shifted in a predetermined measurement space relative to at least one other signal before the simultaneous sparse approximation operation. 22. The method as recited in claim 21, wherein a plurality of relative shifts are executed on at least one of the signals relative to another, a simultaneous sparse approximation operation being executed upon each relatively shifted signal set, and a preferred relative shift being determined based on the simultaneous sparse approximation structure books resulting respectively from the relative shifts. 23. The method as recited in claim 16, wherein: a. the collected signal or signals are sparsely approximated in aggregate with at least one previously established signal; and b. the structure books of the previously established signal or signals are compared to those of the newly collected signal or signals in order to classify the new signal or signals as either similar or different from the previous group. 24. The method as recited in claim 16, wherein a plurality of signals are collected at different locations and, subsequent to detection, time and phase information from the resulting structure books at different locations are used to extract directional information about the signals. 25. The method as recited in claim 16, wherein: a. a plurality of signals are collected at different locations; b. at least one signal from at least one first location is analyzed independently to detect signatures; c. at least one atom of one detected signature is reconstructed to produce a signature component; d. at least one other location channel is searched for the reconstructed signature component to determine relative time of arrival information therefor; and, e. signals for at least one location channel are time shifted relative to the signals for the first location before simultaneous sparse approximation relative to said signature component atoms. 26. The method as recited in claim 16, further comprising the step of reconstructing at least one component of a decomposed signal for restoration back to a signal domain thereof when collected. 27. The method as recited in claim 16, wherein the comparison of structure books is performed with respect to a subset of atoms containing fewer atoms than at least one structure book in said comparison. 28. The method as recited in claim 27, wherein said subset of atoms is determined by a comparison between the structure book atoms of at least two known signatures.
A system and method whereby acoustic signals can be classified and identified as to nature and location of the original signal. The system and method determine from an arbitrary set of signals a signature or other characterizing feature and distinguish signals associated with a plurality of conditions by means of dictionaries comprising atoms of signals.1. A system for extracting, detecting, classifying, discriminating, localizing, and processing signals comprising: a. at least one transducer device operable to collect a plurality of signals; b. a processing device coupled to said transducer device, said processing device executing instructions for decomposing the signals into representational spaces matched with one another, each said representational space defining a multi-dimensional set of atoms, wherein the decomposing comprises performing simultaneous sparse approximations of the signals, each signal being thereby represented by a combination of atoms selected from said representational space, each selected atom being weighted by a corresponding coefficient; and, c. a computer memory coupled to said processor for storing said representational spaces. 2. The system as recited in claim 1, further comprising a detection unit coupled to said processing device, said detection unit selectively determining a match between a set of said signals responsive to respective representational spaces thereof. 3. The system as recited in claim 1, wherein said transducer device collects includes at least one sensor. 4. The system as recited in claim 1, wherein said processing device executes a Greedy Adaptive Discrimination (GAD) decomposition operating with a dictionary of prototype atoms. 5. The system as recited in claim 4, wherein the dictionary comprises a set of Gabor atoms together with Fourier and delta function bases. 6. The system as recited in claim 1, wherein the said atoms are selected from a random set of atoms. 7. The system as recited in claim 1, wherein each said atom selected for said representation of at least one signal is caused to be orthogonally related to at least one other atom in said representational space for said signal. 8. The system as recited in claim 1, wherein said processor device generates a set of test atoms based on said representational spaces, and defines a dictionary of said test atoms, a subsequently collected signal being decomposed in terms of test atoms selected from said dictionary, whereby a degree of similarity is determined for the subsequently collected signal relative to said representational spaces of previously collected signals. 9. The system as recited in claim 1, comprising a plurality of said transducer devices collecting signals through a plurality of channels, the signals of at least one said channel being alignment shifted in a predetermined measurement space relative to the signals of at least one other of said channels. 10. A method of signature extraction comprising the steps of: a. establishing two or more analysis sets, each set comprising at least one signal; b. decomposing the analysis sets into matched structure books, wherein the decomposing comprises executing a processing device to perform simultaneous sparse approximations of the signals of the analysis sets; each said structure book including at least one representation defining a multi-dimensional combination of coefficients and atoms; and, c. storing said structure hooks in a computer memory. 11. The method as recited in claim 10, further comprising actuating the processing device to produce a parametric mean structure book representative of a signature for each set of signals. 12. The method as recited in claim 10, wherein a separate simultaneous sparse approximation is performed for each analysis set. 13. The method as recited in claim 10, wherein a single simultaneous sparse approximation is performed for an aggregate collection of signals from a plurality of analysis sets. 14. The method as recited in claim 10, wherein at least one of the signals is alignment shifted in a predetermined measurement space relative to another of the signals before simultaneous sparse approximation. 15. The method as recited in claim 10, wherein the signals are collected at a plurality of different locations, the signals collected at each location determining at least one analysis set, a location specific signature being determined based on the resulting structure books. 16. A method of detecting, classifying, discriminating, and localizing signals comprising the steps of: a. collecting a set of signals; b. decomposing the sets of signals into matched structure books, wherein the decomposing comprises executing a processing device to perform simultaneous sparse approximations of the signals; each said structure book defining a multi-dimensional combination of coefficients and atoms; the simultaneous sparse approximation being executed with respect to a dictionary defined relative to previously determined signatures; and, c. comparing the resulting structure books to structure books of the previously determined si matures. 17. The method as recited in claim 16, wherein a single simultaneous sparse approximation is formed for an aggregate collection of signals from at least two groups of signals, each defining one of said analysis sets. 18. The method as recited in claim 16, wherein step c is accomplished by examining the values of the coefficients of the atoms in the resulting structure books compared to typical values for similar atoms of one or more known signatures. 19. The method as recited in claim 16, wherein the simultaneous sparse approximation is accomplished with respect to a dictionary comprising an aggregate collection of atoms present in previously extracted structure books of known signatures of interest. 20. The method as recited in claim 19, wherein the dictionary additionally includes the aggregate collection of all atoms selected during the sparse approximation that produced the previously extracted structure books of known signatures of interest, whether or not said atoms are present in said structure books of signatures of interest. 21. The method as recited in claim 16, wherein a plurality of signals are collected and at least one of the signals is alignment shifted in a predetermined measurement space relative to at least one other signal before the simultaneous sparse approximation operation. 22. The method as recited in claim 21, wherein a plurality of relative shifts are executed on at least one of the signals relative to another, a simultaneous sparse approximation operation being executed upon each relatively shifted signal set, and a preferred relative shift being determined based on the simultaneous sparse approximation structure books resulting respectively from the relative shifts. 23. The method as recited in claim 16, wherein: a. the collected signal or signals are sparsely approximated in aggregate with at least one previously established signal; and b. the structure books of the previously established signal or signals are compared to those of the newly collected signal or signals in order to classify the new signal or signals as either similar or different from the previous group. 24. The method as recited in claim 16, wherein a plurality of signals are collected at different locations and, subsequent to detection, time and phase information from the resulting structure books at different locations are used to extract directional information about the signals. 25. The method as recited in claim 16, wherein: a. a plurality of signals are collected at different locations; b. at least one signal from at least one first location is analyzed independently to detect signatures; c. at least one atom of one detected signature is reconstructed to produce a signature component; d. at least one other location channel is searched for the reconstructed signature component to determine relative time of arrival information therefor; and, e. signals for at least one location channel are time shifted relative to the signals for the first location before simultaneous sparse approximation relative to said signature component atoms. 26. The method as recited in claim 16, further comprising the step of reconstructing at least one component of a decomposed signal for restoration back to a signal domain thereof when collected. 27. The method as recited in claim 16, wherein the comparison of structure books is performed with respect to a subset of atoms containing fewer atoms than at least one structure book in said comparison. 28. The method as recited in claim 27, wherein said subset of atoms is determined by a comparison between the structure book atoms of at least two known signatures.
1,600
231
231
14,929,111
1,615
Described are methods of preparing reduced 3,7-diamino-10H-phenothiazine (DAPTZ) compounds of formula: wherein: R 1 and R 9 are independently selected from: —H; C 1-4 alkyl; C 2-4 alkenyl; and halogenated C 1-4 alkyl; each of R 3NA and R 3NB is independently selected from: —H; C 1-4 alkyl; C 2-4 alkenyl; and halogenated C 1-4 alkyl; each of R 7NA and R 7NB is independently selected from: —H; C 1-4 alkyl; C 2-4 alkenyl; and halogenated C 1-4 alkyl; each of HX 1 and HX 2 is independently a protic acid; and pharmaceutically acceptable salts, solvates, and hydrates thereof. These methods are particularly useful for producing stable reduced forms, and with high purity. The stability and purity are especially relevant for pharmaceutical compositions for the treatment of disease. The compounds are useful for treatment of e.g. tauopathies, such as Alzheimer's disease, and also as prodrugs for the corresponding oxidized thioninium drugs.
1. A method of preparing a 3,7-diamino-10H-phenothiazine (DAPTZ) compound of formula (1): wherein: each of R1 and R9 is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R3NA and R3NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R7NA and R7NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; and each of HX1 and HX2 is independently a protic acid; from a corresponding ring amine-protected compound of formula (2): wherein RProt is a protecting group; said method comprising: ring amino deprotection (DP), wherein the protecting group RProt is removed from said corresponding ring amine-protected compound (2); and salt formation (SF), wherein the resulting deprotected compound reacts with a protic acid to form a salt. 2. A method of preparing a pharmaceutical composition comprising admixing the DAPTZ compound obtained according to a method of claim 1, and a pharmaceutically acceptable carrier or diluent. 3. A method of treatment or prophylaxis of a disease comprising administering to said subject a prophylactically or therapeutically effective amount of a compound of the following formula and pharmaceutically acceptable salts thereof: wherein: each of R1 and R9 is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R3NA and R3NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R7NA and R7NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of HX1 and HX2 is independently a protic acid. 4. A pharmaceutical composition comprising a therapeutically effective amount of a compound of the following formula and pharmaceutically acceptable salts thereof: wherein: each of R1 and R9 is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R3NA and R3NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R7NA and R7NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of HX1 and HX2 is independently a protic acid.
Described are methods of preparing reduced 3,7-diamino-10H-phenothiazine (DAPTZ) compounds of formula: wherein: R 1 and R 9 are independently selected from: —H; C 1-4 alkyl; C 2-4 alkenyl; and halogenated C 1-4 alkyl; each of R 3NA and R 3NB is independently selected from: —H; C 1-4 alkyl; C 2-4 alkenyl; and halogenated C 1-4 alkyl; each of R 7NA and R 7NB is independently selected from: —H; C 1-4 alkyl; C 2-4 alkenyl; and halogenated C 1-4 alkyl; each of HX 1 and HX 2 is independently a protic acid; and pharmaceutically acceptable salts, solvates, and hydrates thereof. These methods are particularly useful for producing stable reduced forms, and with high purity. The stability and purity are especially relevant for pharmaceutical compositions for the treatment of disease. The compounds are useful for treatment of e.g. tauopathies, such as Alzheimer's disease, and also as prodrugs for the corresponding oxidized thioninium drugs.1. A method of preparing a 3,7-diamino-10H-phenothiazine (DAPTZ) compound of formula (1): wherein: each of R1 and R9 is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R3NA and R3NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R7NA and R7NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; and each of HX1 and HX2 is independently a protic acid; from a corresponding ring amine-protected compound of formula (2): wherein RProt is a protecting group; said method comprising: ring amino deprotection (DP), wherein the protecting group RProt is removed from said corresponding ring amine-protected compound (2); and salt formation (SF), wherein the resulting deprotected compound reacts with a protic acid to form a salt. 2. A method of preparing a pharmaceutical composition comprising admixing the DAPTZ compound obtained according to a method of claim 1, and a pharmaceutically acceptable carrier or diluent. 3. A method of treatment or prophylaxis of a disease comprising administering to said subject a prophylactically or therapeutically effective amount of a compound of the following formula and pharmaceutically acceptable salts thereof: wherein: each of R1 and R9 is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R3NA and R3NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R7NA and R7NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of HX1 and HX2 is independently a protic acid. 4. A pharmaceutical composition comprising a therapeutically effective amount of a compound of the following formula and pharmaceutically acceptable salts thereof: wherein: each of R1 and R9 is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R3NA and R3NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of R7NA and R7NB is independently selected from: —H, C1-4alkyl, C2-4alkenyl, and halogenated C1-4alkyl; each of HX1 and HX2 is independently a protic acid.
1,600
232
232
14,931,316
1,613
The present invention relates to a nail composition containing at least one adhesive agent, at least one primary film former and at least one plasticizer.
1. A nail composition comprising: at least one adhesive agent, wherein the at least one adhesive agent is selected from the group consisting of radical or polycondensate synthetic polymers, polymers of natural origin, and mixtures thereof; at least one primary film former, wherein the at least one primary film former is a carboxyl functional acrylate having an acid value (number) ranging from about 40 to about 95 and having a Tg value ranging from about 40° C. to about 95° C.; and at least one plasticizer; wherein the composition has shine higher than about 60 GU at 20° and wherein the weight ratio of the at least one primary film former to the at least one plasticizer is greater than or equal to about 1:1. 2. The nail composition of claim 1, further comprising at least one solvent. 3. (canceled) 4. The nail composition of claim 1, further comprising at least one secondary film former. 5. The nail composition of claim 1, further comprising at least one silicone acrylate copolymer. 6. (canceled) 7. (canceled) 8. (canceled) 9. The nail composition of claim 4, wherein the at least one secondary film former is tosylamide epoxy resin. 10. (canceled) 11. The nail composition of claim 1, wherein the at least one plasticizer is selected from the group consisting of glycols and their ester derivatives, esters of acids carboxylic acids, and mixtures thereof. 12. The nail composition of claim 1, further comprising at least co-film former selected from the group consisting of polyurethanes, acrylic polymers, vinyl polymers, polyvinylbutyrals, alkyd resins, ketone/aldehyde resins, aryl sulfonamide formaldehyde resins, aryl-sulfonamide epoxy resins, ethyl tosylamide resins, and mixtures thereof. 13. The nail composition of claim 2, wherein the at least one solvent is selected from organic solvents, inorganic solvents, and mixtures thereof. 14. (canceled) 15. The nail composition of claim 1, further comprising at least one coloring agent. 16. A method of making up or protecting nails comprising applying the composition of claim 1 to nails in an amount sufficient to make up or protect the nails. 17. A method of making the composition of claim 1, comprising combining at least one adhesive agent, at least one primary film former, and at least one plasticizer in a nail composition. 18. The nail composition of claim 1, wherein the composition has shine higher than about 70 GU at 20°. 19. The nail composition of claim 1, wherein the weight ratio of the at least one primary film former to the at least one plasticizer is between about 1:2 and about 1:20. 20. The nail composition of claim 1, wherein the at least one plasticizer is ethyl tosylamide. 21. The nail composition of claim 1, wherein the primary film former is a styrene/acrylates copolymer. 22. The nail composition of claim 1, wherein the composition is a color coat which has a Young's modulus value lower than about 45 MPa. 23. The nail composition of claim 1, wherein the composition is a topcoat which has shine higher than about 70 GU at 20° and is transparent.
The present invention relates to a nail composition containing at least one adhesive agent, at least one primary film former and at least one plasticizer.1. A nail composition comprising: at least one adhesive agent, wherein the at least one adhesive agent is selected from the group consisting of radical or polycondensate synthetic polymers, polymers of natural origin, and mixtures thereof; at least one primary film former, wherein the at least one primary film former is a carboxyl functional acrylate having an acid value (number) ranging from about 40 to about 95 and having a Tg value ranging from about 40° C. to about 95° C.; and at least one plasticizer; wherein the composition has shine higher than about 60 GU at 20° and wherein the weight ratio of the at least one primary film former to the at least one plasticizer is greater than or equal to about 1:1. 2. The nail composition of claim 1, further comprising at least one solvent. 3. (canceled) 4. The nail composition of claim 1, further comprising at least one secondary film former. 5. The nail composition of claim 1, further comprising at least one silicone acrylate copolymer. 6. (canceled) 7. (canceled) 8. (canceled) 9. The nail composition of claim 4, wherein the at least one secondary film former is tosylamide epoxy resin. 10. (canceled) 11. The nail composition of claim 1, wherein the at least one plasticizer is selected from the group consisting of glycols and their ester derivatives, esters of acids carboxylic acids, and mixtures thereof. 12. The nail composition of claim 1, further comprising at least co-film former selected from the group consisting of polyurethanes, acrylic polymers, vinyl polymers, polyvinylbutyrals, alkyd resins, ketone/aldehyde resins, aryl sulfonamide formaldehyde resins, aryl-sulfonamide epoxy resins, ethyl tosylamide resins, and mixtures thereof. 13. The nail composition of claim 2, wherein the at least one solvent is selected from organic solvents, inorganic solvents, and mixtures thereof. 14. (canceled) 15. The nail composition of claim 1, further comprising at least one coloring agent. 16. A method of making up or protecting nails comprising applying the composition of claim 1 to nails in an amount sufficient to make up or protect the nails. 17. A method of making the composition of claim 1, comprising combining at least one adhesive agent, at least one primary film former, and at least one plasticizer in a nail composition. 18. The nail composition of claim 1, wherein the composition has shine higher than about 70 GU at 20°. 19. The nail composition of claim 1, wherein the weight ratio of the at least one primary film former to the at least one plasticizer is between about 1:2 and about 1:20. 20. The nail composition of claim 1, wherein the at least one plasticizer is ethyl tosylamide. 21. The nail composition of claim 1, wherein the primary film former is a styrene/acrylates copolymer. 22. The nail composition of claim 1, wherein the composition is a color coat which has a Young's modulus value lower than about 45 MPa. 23. The nail composition of claim 1, wherein the composition is a topcoat which has shine higher than about 70 GU at 20° and is transparent.
1,600
233
233
15,363,317
1,617
The present invention relates to a simple, robust and effective process for preparing a dry powder inhalation formulation containing at least one drug, preferably a highly active drug, more preferably Tiotropium bromide or Tiotropium bromide monohydrate, and a lactose carrier. The process is characterized in that a three-layered composition containing a drug layer in between layers of the lactose carrier is mixed and the obtained preblend is mixed with additional lactose carrier. The process combines a simple manufacturing procedure with an excellent content uniformity obtained over a wide range of mixing parameters.
1. A process for preparing a dry powder inhalation formulation containing at least one drug and a lactose carrier, comprising the steps: a) splitting the lactose carrier into a first, a second and a third portion, b) mixing the first and the second portion of the lactose carrier and the drug by i) placing the first portion of the lactose carrier in a mixer to obtain a first layer, ii) placing the entire drug on top of the first lactose layer to obtain a second layer, iii) placing the second portion of the lactose carrier on top of the second layer to obtain a third layer, iv) blending the three-layered composition obtained in step (iii) to obtain a preblend, v) optionally sifting the preblend, c) mixing the preblend obtained in step (b) and the third portion of the lactose carrier to obtain the dry powder inhalation formulation, and d) optionally sifting the dry powder inhalation formulation. 2. The process according to claim 1, wherein the lactose carrier is prepared by mixing coarse lactose and fine lactose. 3. The process according to claim 2, comprising the steps: a1) splitting the coarse lactose into a first, a second and a third fraction, a2) placing the first fraction of the coarse lactose in a mixer to obtain a first layer, a3) placing the entire fine lactose on top of the first layer to obtain a second layer, a4) placing the second fraction of the coarse lactose on top of the second layer to obtain a third layer, a5) blending the three-layered composition obtained in step (a4) to obtain a preblend, a6) optionally sifting the preblend, a7) mixing the preblend obtained in step (a5) or (a6) and the third fraction of the coarse lactose to obtain the lactose carrier, and a8) optionally sifting the lactose carrier. 4. The process according to claim 3, wherein the first, second and third fractions of the coarse lactose are equal. 5. The process according to claim 2, wherein the fine lactose content of the lactose carrier is 30% by weight or less, preferably 10-30% by weight, more preferred 20-30% by weight. 6. The process according to claim 1, wherein the amount of the first portion, the second portion and the third portion of the lactose carrier each is at least 10% by weight, based on the total weight of the lactose carrier. 7. The process according to claim 6, wherein the amount of the first portion of the lactose carrier is 10-70% by weight, the amount of the second portion of the lactose carrier is 10-20% by weight, and the amount of the third portion of the lactose carrier is 10-70% by weight, based on the total weight of the lactose carrier. 8. The process according to claim 6, wherein the weight ratio of the first portion/second portion/third portion of the lactose carrier is 7/2/1, 5/2/3, 5/1/4, 4/1/5 or 2/1/7. 9. The process according to claim 1, wherein the dry powder inhalation formulation consists of tiotropium bromide, preferably tiotropium bromide monohydrate, and the lactose carrier, preferably lactose monohydrate.
The present invention relates to a simple, robust and effective process for preparing a dry powder inhalation formulation containing at least one drug, preferably a highly active drug, more preferably Tiotropium bromide or Tiotropium bromide monohydrate, and a lactose carrier. The process is characterized in that a three-layered composition containing a drug layer in between layers of the lactose carrier is mixed and the obtained preblend is mixed with additional lactose carrier. The process combines a simple manufacturing procedure with an excellent content uniformity obtained over a wide range of mixing parameters.1. A process for preparing a dry powder inhalation formulation containing at least one drug and a lactose carrier, comprising the steps: a) splitting the lactose carrier into a first, a second and a third portion, b) mixing the first and the second portion of the lactose carrier and the drug by i) placing the first portion of the lactose carrier in a mixer to obtain a first layer, ii) placing the entire drug on top of the first lactose layer to obtain a second layer, iii) placing the second portion of the lactose carrier on top of the second layer to obtain a third layer, iv) blending the three-layered composition obtained in step (iii) to obtain a preblend, v) optionally sifting the preblend, c) mixing the preblend obtained in step (b) and the third portion of the lactose carrier to obtain the dry powder inhalation formulation, and d) optionally sifting the dry powder inhalation formulation. 2. The process according to claim 1, wherein the lactose carrier is prepared by mixing coarse lactose and fine lactose. 3. The process according to claim 2, comprising the steps: a1) splitting the coarse lactose into a first, a second and a third fraction, a2) placing the first fraction of the coarse lactose in a mixer to obtain a first layer, a3) placing the entire fine lactose on top of the first layer to obtain a second layer, a4) placing the second fraction of the coarse lactose on top of the second layer to obtain a third layer, a5) blending the three-layered composition obtained in step (a4) to obtain a preblend, a6) optionally sifting the preblend, a7) mixing the preblend obtained in step (a5) or (a6) and the third fraction of the coarse lactose to obtain the lactose carrier, and a8) optionally sifting the lactose carrier. 4. The process according to claim 3, wherein the first, second and third fractions of the coarse lactose are equal. 5. The process according to claim 2, wherein the fine lactose content of the lactose carrier is 30% by weight or less, preferably 10-30% by weight, more preferred 20-30% by weight. 6. The process according to claim 1, wherein the amount of the first portion, the second portion and the third portion of the lactose carrier each is at least 10% by weight, based on the total weight of the lactose carrier. 7. The process according to claim 6, wherein the amount of the first portion of the lactose carrier is 10-70% by weight, the amount of the second portion of the lactose carrier is 10-20% by weight, and the amount of the third portion of the lactose carrier is 10-70% by weight, based on the total weight of the lactose carrier. 8. The process according to claim 6, wherein the weight ratio of the first portion/second portion/third portion of the lactose carrier is 7/2/1, 5/2/3, 5/1/4, 4/1/5 or 2/1/7. 9. The process according to claim 1, wherein the dry powder inhalation formulation consists of tiotropium bromide, preferably tiotropium bromide monohydrate, and the lactose carrier, preferably lactose monohydrate.
1,600
234
234
14,376,981
1,619
Cosmetic and/or dermatological preparations based on a water-in-oil emulsion comprising at least two W/O emulsifiers which differ in their HLB value up to a maximum of 1 lead to sensorily acceptable preparations and at the same stable W/O emulsions.
1-17. (canceled) 18. A cosmetic and/or dermatological preparation, wherein the preparation is based on a water-in-oil emulsion comprising at least two W/O emulsifiers which differ in their HLB value by at most 1. 19. The preparation of claim 18, wherein the at least two W/O emulsifiers differ in their HLB value by at most 0.5. 20. The preparation of claim 18, wherein only two W/O emulsifiers are present. 21. The preparation of claim 18, wherein the at least two W/O emulsifiers comprise diisostearoyl polyglyceryl-3 dimer dilinoleate and polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate. 22. The preparation of claim 18, wherein the preparation comprises from 1% to 5.5% by weight of W/O emulsifiers, based on a total weight of the preparation. 23. The preparation of claim 22, wherein the preparation comprises from 1.5% to 3% by weight of W/O emulsifiers. 24. The preparation of claim 22, wherein the preparation comprises from 2% to 2.6% by weight of W/O emulsifiers. 25. The preparation of claim 21, wherein the preparation comprises from 0.1% to 2.5% by weight of polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate and from 0.1% to 3% by weight of diisostearoyl polyglyceryl-3 dimer dilinoleate, each based on a total weight of the preparation. 26. The preparation of claim 25, wherein the preparation comprises from 0.3% to 1.5% by weight of polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate and from 0.5% to 2% by weight of diisostearoyl polyglyceryl-3 dimer dilinoleate. 27. The preparation of claim 18, wherein the preparation further comprises a total of from 5% to 20% by weight, based on a total weight of the preparation, of at least one skin moisturizer. 28. The preparation of claim 27, wherein the at least one skin moisturizer comprises glycerol. 29. The preparation of claim 18, wherein the preparation further comprises from 10% to 30% by weight of one or more lipids, based on a total weight of the preparation. 30. The preparation of claim 18, wherein the one or more lipids are selected from paraffinum liquidum, isopropyl palmitate, C13-16 isoparaffin, cera microcristallina, cetyl palmitate, Vaseline (cera microcristallina+paraffinum liquidum) and natural oils. 31. The preparation of claim 18, wherein the preparation further comprises one or more powder raw materials. 32. The preparation of claim 31, wherein the one or more powder raw materials are present in a concentration of up to 5% by weight, based on a total weight of the preparation. 33. The preparation of claim 31, wherein the one or more powder raw materials comprise at least one of aluminum starch octenylsuccinate and talc. 34. The preparation of claim 18, wherein the preparation comprises from 40% to 80% by weight of water, based on a total weight of the preparation. 35. The preparation of claim 18, wherein the preparation comprises from 0% to less than 1% by weight, based on a total weight of the preparation, of polyethylene glycols and polyethylene glycol derivatives. 36. A method of improving the stability and/or sensorics of a W/O emulsion, wherein the method comprises including in the emulsion two W/O emulsifiers which differ in their HLB value by at most 1. 37. The method of claim 36, wherein the two W/O emulsifiers are diisostearoyl polyglyceryl-3 dimer dilinoleate and polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate.
Cosmetic and/or dermatological preparations based on a water-in-oil emulsion comprising at least two W/O emulsifiers which differ in their HLB value up to a maximum of 1 lead to sensorily acceptable preparations and at the same stable W/O emulsions.1-17. (canceled) 18. A cosmetic and/or dermatological preparation, wherein the preparation is based on a water-in-oil emulsion comprising at least two W/O emulsifiers which differ in their HLB value by at most 1. 19. The preparation of claim 18, wherein the at least two W/O emulsifiers differ in their HLB value by at most 0.5. 20. The preparation of claim 18, wherein only two W/O emulsifiers are present. 21. The preparation of claim 18, wherein the at least two W/O emulsifiers comprise diisostearoyl polyglyceryl-3 dimer dilinoleate and polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate. 22. The preparation of claim 18, wherein the preparation comprises from 1% to 5.5% by weight of W/O emulsifiers, based on a total weight of the preparation. 23. The preparation of claim 22, wherein the preparation comprises from 1.5% to 3% by weight of W/O emulsifiers. 24. The preparation of claim 22, wherein the preparation comprises from 2% to 2.6% by weight of W/O emulsifiers. 25. The preparation of claim 21, wherein the preparation comprises from 0.1% to 2.5% by weight of polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate and from 0.1% to 3% by weight of diisostearoyl polyglyceryl-3 dimer dilinoleate, each based on a total weight of the preparation. 26. The preparation of claim 25, wherein the preparation comprises from 0.3% to 1.5% by weight of polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate and from 0.5% to 2% by weight of diisostearoyl polyglyceryl-3 dimer dilinoleate. 27. The preparation of claim 18, wherein the preparation further comprises a total of from 5% to 20% by weight, based on a total weight of the preparation, of at least one skin moisturizer. 28. The preparation of claim 27, wherein the at least one skin moisturizer comprises glycerol. 29. The preparation of claim 18, wherein the preparation further comprises from 10% to 30% by weight of one or more lipids, based on a total weight of the preparation. 30. The preparation of claim 18, wherein the one or more lipids are selected from paraffinum liquidum, isopropyl palmitate, C13-16 isoparaffin, cera microcristallina, cetyl palmitate, Vaseline (cera microcristallina+paraffinum liquidum) and natural oils. 31. The preparation of claim 18, wherein the preparation further comprises one or more powder raw materials. 32. The preparation of claim 31, wherein the one or more powder raw materials are present in a concentration of up to 5% by weight, based on a total weight of the preparation. 33. The preparation of claim 31, wherein the one or more powder raw materials comprise at least one of aluminum starch octenylsuccinate and talc. 34. The preparation of claim 18, wherein the preparation comprises from 40% to 80% by weight of water, based on a total weight of the preparation. 35. The preparation of claim 18, wherein the preparation comprises from 0% to less than 1% by weight, based on a total weight of the preparation, of polyethylene glycols and polyethylene glycol derivatives. 36. A method of improving the stability and/or sensorics of a W/O emulsion, wherein the method comprises including in the emulsion two W/O emulsifiers which differ in their HLB value by at most 1. 37. The method of claim 36, wherein the two W/O emulsifiers are diisostearoyl polyglyceryl-3 dimer dilinoleate and polyglyceryl-4 diisostearate/polyhydroxystearate/sebacate.
1,600
235
235
14,264,983
1,611
Hydrogel compositions are provided (a) that have a continuous hydrophobic phase and a discontinuous hydrophilic phase, (b) that have a discontinuous hydrophilic phase and a continuous hydrophilic phase, or (c) that are entirely composed of a continuous hydrophilic phase. The hydrophobic phase, if present, is composed of a hydrophobic polymer, particularly a hydrophobic pressure-sensitive adhesive (PSA), a plasticizing elastomer, a tackifying resin, and an optional antioxidant. The discontinuous hydrophilic phase, if present, is composed of a crosslinked hydrophilic polymer, particularly a crosslinked cellulosic polymer such as crosslinked sodium carboxymethylcellulose. For those hydrogel compositions containing a continuous hydrophilic phase, the components of the phase include a cellulose ester composition or an acrylate polymer or copolymer, and a blend of a hydrophilic polymer and a complementary oligomer capable of hydrogen bonding thereto. Films prepared from hydrogel compositions containing or entirely composed of the aforementioned continuous hydrophilic phase can be made translucent, and may be prepared using either melt extrusion or solution casting. A preferred use of the hydrogel compositions is in wound dressings, although numerous other uses are possible as well.
1-83. (canceled) 84. A delivery system for topical administration of a locally active agent to the skin, the system comprising: a body-surface contacting layer comprised of an adhesive hydrogel composition positioned on a backing layer, the adhesive hydrogel composition comprising: (i) a water-swellable, water-insoluble polymer, wherein said polymer is insoluble in water within a selected pH range and is acrylate polymer or copolymer comprising monomers selected from the group consisting of acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate, and ethyl methacrylate; (ii) a blend of a hydrophilic polymer and a complementary oligomer, where the hydrophilic polymer is a poly(N-vinyl lactam) and the complementary oligomer is a low molecular weight polyalkylene glycol having a molecular weight of 300 to 600 daltons, (iii) a permeation enhancer, and (iv) a locally active agent. 85. The delivery system of claim 84, wherein the adhesive hydrogel composition is entirely comprised of a continuous hydrophilic phase. 86. The delivery system of claim 84, wherein the acrylate polymer or copolymer is a methacrylic acid-ethyl acrylate copolymer. 87. The delivery system of claim 86, wherein the copolymer is generally insoluble in aqueous fluids having a pH below 5.5. 88. The delivery system of claim 87, wherein the copolymer has a ratio of free carboxyl groups to ester groups of about 1:1. 89. The delivery system of claim 84, wherein the poly(N-vinyl lactam) is polyvinyl pyrrolidone. 90. The delivery system of claim 84, wherein the molecular weight of the hydrophilic polymer is in a range of approximately 500,000 to 1,500,000 daltons. 91. The delivery system of claim 84, wherein the polyalkylene glycol is polyethylene glycol. 92. The delivery system of claim 84, wherein the water-swellable, water-insoluble polymer represents about 2 weight percent to about 15 weight percent of the hydrogel composition. 93. The delivery system of claim 84, wherein the hydrophilic polymer-complementary oligomer blend represents about 17.5 weight percent to about 45 weight percent of the hydrogel composition. 94. The delivery system of claim 84, wherein the permeation enhancer is selected from the group consisting of sulfoxides, ethers, surfactants, alcohols, fatty acids, fatty acid esters, polyols, amides, terpenes, akanones, and organic acids. 95. The delivery system of claim 84, wherein the hydrogel composition comprises one or more additional additives. 96. The delivery system of claim 95, wherein the one or more additives comprises an antimicrobial agent. 97. The delivery system of claim 96, wherein the antimicrobial agent is selected from methyl paraben, propyl paraben, sodium benzoate, sorbic acid, imidurea, and combinations thereof. 98. The delivery system of claim 84, wherein the delivery system is monolithic. 99. The delivery system of claim 84, wherein the locally active agent is a cosmeceutical. 100. The delivery system of claim 85, comprising an additional low molecular weight plasticizer in addition to the complementary oligomer. 101. The delivery system of claim 84, further comprising a disposable release liner covering the adhesive hydrogel composition of the body-surface contacting layer.
Hydrogel compositions are provided (a) that have a continuous hydrophobic phase and a discontinuous hydrophilic phase, (b) that have a discontinuous hydrophilic phase and a continuous hydrophilic phase, or (c) that are entirely composed of a continuous hydrophilic phase. The hydrophobic phase, if present, is composed of a hydrophobic polymer, particularly a hydrophobic pressure-sensitive adhesive (PSA), a plasticizing elastomer, a tackifying resin, and an optional antioxidant. The discontinuous hydrophilic phase, if present, is composed of a crosslinked hydrophilic polymer, particularly a crosslinked cellulosic polymer such as crosslinked sodium carboxymethylcellulose. For those hydrogel compositions containing a continuous hydrophilic phase, the components of the phase include a cellulose ester composition or an acrylate polymer or copolymer, and a blend of a hydrophilic polymer and a complementary oligomer capable of hydrogen bonding thereto. Films prepared from hydrogel compositions containing or entirely composed of the aforementioned continuous hydrophilic phase can be made translucent, and may be prepared using either melt extrusion or solution casting. A preferred use of the hydrogel compositions is in wound dressings, although numerous other uses are possible as well.1-83. (canceled) 84. A delivery system for topical administration of a locally active agent to the skin, the system comprising: a body-surface contacting layer comprised of an adhesive hydrogel composition positioned on a backing layer, the adhesive hydrogel composition comprising: (i) a water-swellable, water-insoluble polymer, wherein said polymer is insoluble in water within a selected pH range and is acrylate polymer or copolymer comprising monomers selected from the group consisting of acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate, and ethyl methacrylate; (ii) a blend of a hydrophilic polymer and a complementary oligomer, where the hydrophilic polymer is a poly(N-vinyl lactam) and the complementary oligomer is a low molecular weight polyalkylene glycol having a molecular weight of 300 to 600 daltons, (iii) a permeation enhancer, and (iv) a locally active agent. 85. The delivery system of claim 84, wherein the adhesive hydrogel composition is entirely comprised of a continuous hydrophilic phase. 86. The delivery system of claim 84, wherein the acrylate polymer or copolymer is a methacrylic acid-ethyl acrylate copolymer. 87. The delivery system of claim 86, wherein the copolymer is generally insoluble in aqueous fluids having a pH below 5.5. 88. The delivery system of claim 87, wherein the copolymer has a ratio of free carboxyl groups to ester groups of about 1:1. 89. The delivery system of claim 84, wherein the poly(N-vinyl lactam) is polyvinyl pyrrolidone. 90. The delivery system of claim 84, wherein the molecular weight of the hydrophilic polymer is in a range of approximately 500,000 to 1,500,000 daltons. 91. The delivery system of claim 84, wherein the polyalkylene glycol is polyethylene glycol. 92. The delivery system of claim 84, wherein the water-swellable, water-insoluble polymer represents about 2 weight percent to about 15 weight percent of the hydrogel composition. 93. The delivery system of claim 84, wherein the hydrophilic polymer-complementary oligomer blend represents about 17.5 weight percent to about 45 weight percent of the hydrogel composition. 94. The delivery system of claim 84, wherein the permeation enhancer is selected from the group consisting of sulfoxides, ethers, surfactants, alcohols, fatty acids, fatty acid esters, polyols, amides, terpenes, akanones, and organic acids. 95. The delivery system of claim 84, wherein the hydrogel composition comprises one or more additional additives. 96. The delivery system of claim 95, wherein the one or more additives comprises an antimicrobial agent. 97. The delivery system of claim 96, wherein the antimicrobial agent is selected from methyl paraben, propyl paraben, sodium benzoate, sorbic acid, imidurea, and combinations thereof. 98. The delivery system of claim 84, wherein the delivery system is monolithic. 99. The delivery system of claim 84, wherein the locally active agent is a cosmeceutical. 100. The delivery system of claim 85, comprising an additional low molecular weight plasticizer in addition to the complementary oligomer. 101. The delivery system of claim 84, further comprising a disposable release liner covering the adhesive hydrogel composition of the body-surface contacting layer.
1,600
236
236
14,746,758
1,611
A composition is provided, wherein the composition comprises a water-swellable, water-insoluble polymer, a blend of a hydrophilic polymer with a complementary oligomer capable of hydrogen or electrostatic bonding to the hydrophilic polymer. The composition also includes a backing member. Active ingredients, such as a whitening agent, may be included. The composition finds utility as an oral dressing, for example, a tooth whitening composition that is applied to the teeth in need of whitening. The composition can be designed to be removed when the degree of whitening has been achieved or left in place and allowed to erode entirely. In certain embodiments, the composition is translucent. Methods for preparing and using the compositions are also disclosed.
1.-26. (canceled) 27. A method for preparing a system, the method comprising: (i) providing a hydrogel comprising a water-swellable, water-insoluble polymer, a blend of a hydrophilic polymer and a complementary oligomer capable of hydrogen bonding or electrostatic bonding to the hydrophilic polymer, and optionally, an active agent, and (ii) depositing the hydrogel on an erodible backing member that erodes in a moist environment at a slower rate than the hydrogel. 28. The method of claim 27, wherein the hydrogel comprises an active agent. 29. The method of claim 27, wherein the hydrogel provided in step (i) does not comprise an active agent, the method further comprising the step of loading an active agent onto the hydrogel that is deposited on the erodible backing layer. 30. The method of claim 27, wherein the depositing step comprises extruding the hydrogel directly onto the erodible backing member. 31. The method of claim 27, wherein the providing step comprises providing the hydrogel as an extrudate, and the depositing step comprises pressing the extrudate onto the erodible backing member. 32. The method of claim 27, wherein prior to the providing step, the hydrogel is prepared by mixing the water-swellable, water-insoluble polymer and the blend of the hydrophilic polymer and the complementary oligomer. 33. The method of claim 32, wherein the mixing is carried out at an elevated temperature. 34. The method of claim 32, wherein the hydrogel is prepared by mixing the water-swellable, water-insoluble polymer, the blend of the hydrophilic polymer and the complementary oligomer, and an organic solvent or water. 35. The method of claim 27, wherein the hydrogel is deposited on the erodible backing member at a thickness in a range of about 0.050 to 0.80 mm. 36. The method of claim 27, wherein the hydrogel in step (i) is provided as a solution in a volatile solvent, and the depositing step comprises casting the solution onto the erodible backing member to form a coated erodible backing member. 37. The method of claim 36, wherein the volatile solvent is selected from ethyl acetate and lower alkanols. 38. The method of claim 36, further comprising heating the coated erodible backing member to a temperature in a range of about 80 to 100° C. for about 1 to 4 hours. 39. The method of claim 27, wherein the erodible backing member is substantially non-tacky. 40. The method of claim 27, further comprising placing a removable release liner on the hydrogel that is deposited on the erodible backing member. 41. The method of claim 27, wherein the water-swellable, water-insoluble polymer is selected from the group consisting of a cellulose ester, alginic acid, and an acrylate polymer. 42. The method of claim 41, wherein the water-swellable, water-insoluble polymer is a cellulose ester selected from the group consisting of cellulose acetate, cellulose acetate propionate, cellulose acetate butyrate, cellulose propionate, cellulose butyrate, cellulose propionate butyrate, cellulose diacetate, and cellulose triacetate. 43. The method of claim 41, wherein the water-swellable, water-insoluble polymer is a cellulose ester comprised of a mixture of cellulose and cellulose ester monomer subunits. 44. The method of claim 41, wherein the water-swellable, water-insoluble polymer is an acrylate polymer selected from the group consisting of acrylate polymers and copolymers. 45. The method of claim 44, wherein the acrylate polymer is an acrylic acid or an acrylic acid ester polymer. 46. The method of claim 44, wherein the acrylate polymer is selected from polymers and copolymers of acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate, and ethyl methacrylate. 47. The method of claim 46, wherein the acrylate polymer is a copolymer of methacrylic acid and ethyl acrylate. 48. The method of claim 27, wherein the water-swellable, water-insoluble polymer is insoluble in aqueous liquids at a pH of less than 5.5. 49. The method of claim 27, wherein the hydrophilic polymer is selected from the group consisting of poly(N-vinyl lactams), poly(N-vinyl acrylamides), poly(N-alkylacrylamides), polyacrylic acids and polymethacrylic acids, poly(vinyl alcohol), polyvinylamine, and copolymers thereof. 50. The method of claim 49, wherein the hydrophilic polymer is a poly(N-vinyl lactam). 51. The method of claim 27, wherein the complementary oligomer is selected from the group consisting of polyhydridic alcohols, monomeric and oligomeric alkylene glycols, polyalkylene glycols, alkane diols and carbonic diacids. 52. The method of claim 51, wherein the complimentary oligomer is a polyethylene glycol having a molecular weight in a range from 200 to 600 daltons. 53. The method of claim 28 or 29, wherein the active agent is a tooth whitening agent.
A composition is provided, wherein the composition comprises a water-swellable, water-insoluble polymer, a blend of a hydrophilic polymer with a complementary oligomer capable of hydrogen or electrostatic bonding to the hydrophilic polymer. The composition also includes a backing member. Active ingredients, such as a whitening agent, may be included. The composition finds utility as an oral dressing, for example, a tooth whitening composition that is applied to the teeth in need of whitening. The composition can be designed to be removed when the degree of whitening has been achieved or left in place and allowed to erode entirely. In certain embodiments, the composition is translucent. Methods for preparing and using the compositions are also disclosed.1.-26. (canceled) 27. A method for preparing a system, the method comprising: (i) providing a hydrogel comprising a water-swellable, water-insoluble polymer, a blend of a hydrophilic polymer and a complementary oligomer capable of hydrogen bonding or electrostatic bonding to the hydrophilic polymer, and optionally, an active agent, and (ii) depositing the hydrogel on an erodible backing member that erodes in a moist environment at a slower rate than the hydrogel. 28. The method of claim 27, wherein the hydrogel comprises an active agent. 29. The method of claim 27, wherein the hydrogel provided in step (i) does not comprise an active agent, the method further comprising the step of loading an active agent onto the hydrogel that is deposited on the erodible backing layer. 30. The method of claim 27, wherein the depositing step comprises extruding the hydrogel directly onto the erodible backing member. 31. The method of claim 27, wherein the providing step comprises providing the hydrogel as an extrudate, and the depositing step comprises pressing the extrudate onto the erodible backing member. 32. The method of claim 27, wherein prior to the providing step, the hydrogel is prepared by mixing the water-swellable, water-insoluble polymer and the blend of the hydrophilic polymer and the complementary oligomer. 33. The method of claim 32, wherein the mixing is carried out at an elevated temperature. 34. The method of claim 32, wherein the hydrogel is prepared by mixing the water-swellable, water-insoluble polymer, the blend of the hydrophilic polymer and the complementary oligomer, and an organic solvent or water. 35. The method of claim 27, wherein the hydrogel is deposited on the erodible backing member at a thickness in a range of about 0.050 to 0.80 mm. 36. The method of claim 27, wherein the hydrogel in step (i) is provided as a solution in a volatile solvent, and the depositing step comprises casting the solution onto the erodible backing member to form a coated erodible backing member. 37. The method of claim 36, wherein the volatile solvent is selected from ethyl acetate and lower alkanols. 38. The method of claim 36, further comprising heating the coated erodible backing member to a temperature in a range of about 80 to 100° C. for about 1 to 4 hours. 39. The method of claim 27, wherein the erodible backing member is substantially non-tacky. 40. The method of claim 27, further comprising placing a removable release liner on the hydrogel that is deposited on the erodible backing member. 41. The method of claim 27, wherein the water-swellable, water-insoluble polymer is selected from the group consisting of a cellulose ester, alginic acid, and an acrylate polymer. 42. The method of claim 41, wherein the water-swellable, water-insoluble polymer is a cellulose ester selected from the group consisting of cellulose acetate, cellulose acetate propionate, cellulose acetate butyrate, cellulose propionate, cellulose butyrate, cellulose propionate butyrate, cellulose diacetate, and cellulose triacetate. 43. The method of claim 41, wherein the water-swellable, water-insoluble polymer is a cellulose ester comprised of a mixture of cellulose and cellulose ester monomer subunits. 44. The method of claim 41, wherein the water-swellable, water-insoluble polymer is an acrylate polymer selected from the group consisting of acrylate polymers and copolymers. 45. The method of claim 44, wherein the acrylate polymer is an acrylic acid or an acrylic acid ester polymer. 46. The method of claim 44, wherein the acrylate polymer is selected from polymers and copolymers of acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate, and ethyl methacrylate. 47. The method of claim 46, wherein the acrylate polymer is a copolymer of methacrylic acid and ethyl acrylate. 48. The method of claim 27, wherein the water-swellable, water-insoluble polymer is insoluble in aqueous liquids at a pH of less than 5.5. 49. The method of claim 27, wherein the hydrophilic polymer is selected from the group consisting of poly(N-vinyl lactams), poly(N-vinyl acrylamides), poly(N-alkylacrylamides), polyacrylic acids and polymethacrylic acids, poly(vinyl alcohol), polyvinylamine, and copolymers thereof. 50. The method of claim 49, wherein the hydrophilic polymer is a poly(N-vinyl lactam). 51. The method of claim 27, wherein the complementary oligomer is selected from the group consisting of polyhydridic alcohols, monomeric and oligomeric alkylene glycols, polyalkylene glycols, alkane diols and carbonic diacids. 52. The method of claim 51, wherein the complimentary oligomer is a polyethylene glycol having a molecular weight in a range from 200 to 600 daltons. 53. The method of claim 28 or 29, wherein the active agent is a tooth whitening agent.
1,600
237
237
15,633,492
1,618
A drug dosage form is provided in the form of a solid tablet which is greater than 50% by weight the local anesthetic agent. The local anesthetic agent may be selected from the group consisting of an aminoamide, an aminoester, and a combination thereof. The drug tablet may be in the form of a mini-tablet which is greater than 70 wt % drug, with the balance being excipient. For example, the anesthetic agent may include lidocaine, in a salt or base form, combined with binder and lubricant excipients. Implantable drug delivery devices including the tablets are also provided, e.g., one or more of the drug tablets may be contained in a biocompatible housing. The drug tablets may be substantially cylindrical with flat end faces, and the device may have from 10 to 100 drug tablets aligned in the housing with the flat end faces of adjacent tablets abutting one another.
1. A solid drug unit comprising: a drug in the form of a mini-tablet which is greater than 70% by weight the drug, with the balance being at least one excipient, wherein the mini-tablet is cylindrical with flat end faces, the length of the tablet exceeding its diameter so that the mini-tablet has an aspect ratio of greater than 1:1, and wherein the mini-tablet has a diameter from 1.0 mm to 3.2 mm and a length from 1.7 mm to 4.8 mm, such that the mini-tablet is sized and shaped for insertion through the urethra of a patient. 2. The solid drug unit of claim 1, wherein the mini-tablet has a diameter from 1.5 mm to 3.1 mm and a length from 2.0 mm to 4.5 mm. 3. The solid drug unit of claim 1, wherein the drug is between 70 wt % and 99 wt % of the mini-tablet. 4. The solid drug unit of claim 1, wherein the drug is an anesthetic agent, an antimicrobial agent, or a chemotherapeutic agent. 5. The solid drug unit of claim 4, wherein the excipient comprises a binder and a lubricant. 6. The solid drug unit of claim 1, wherein the drug comprises lidocaine. 7. The solid drug unit of claim 1, wherein the mini-tablet is between 85 wt % and 95 wt % lidocaine. 8. The solid drug unit of claim 1, wherein the friability of the mini-tablet is less than 2%. 9. The solid drug unit of claim 1, wherein the mini-tablet is sterilized. 10. The solid drug unit of claim 1, wherein the aspect ratio is from about 3:2 to about 5:2. 11. A solid drug unit for insertion into an intravesical device, comprising: a drug in the form of a mini-tablet which is greater than 70% by weight the drug, with the balance being at least one excipient, wherein the mini-tablet is cylindrical with end faces that are not convex, the length of the tablet exceeding its diameter so that the mini-tablet has an aspect ratio of greater than 1:1, and wherein the mini-tablet has a diameter from 1.0 mm to 3.2 mm and a length from 1.7 mm to 4.8 mm, such that the mini-tablet is sized and shaped for insertion through the urethra of a patient. 12. The solid drug unit of claim 11, wherein the mini-tablet has a diameter from 1.5 mm to 3.1 mm and a length from 2.0 mm to 4.5 mm. 13. The solid drug unit of claim 11, wherein the drug is between 70 wt % and 99 wt % of the mini-tablet. 14. The solid drug unit of claim 11, wherein the drug is an anesthetic agent, an antimicrobial agent, or a chemotherapeutic agent. 15. The solid drug unit of claim 14, wherein the excipient comprises a binder and a lubricant. 16. The solid drug unit of claim 11, wherein the drug comprises lidocaine. 17. The solid drug unit of claim 11, wherein the mini-tablet is between 85 wt % and 95 wt % lidocaine. 18. The solid drug unit of claim 11, wherein the friability of the mini-tablet is less than 2%. 19. The solid drug unit of claim 11, wherein the mini-tablet is sterilized. 20. The solid drug unit of claim 11, wherein the aspect ratio is from about 3:2 to about 5:2.
A drug dosage form is provided in the form of a solid tablet which is greater than 50% by weight the local anesthetic agent. The local anesthetic agent may be selected from the group consisting of an aminoamide, an aminoester, and a combination thereof. The drug tablet may be in the form of a mini-tablet which is greater than 70 wt % drug, with the balance being excipient. For example, the anesthetic agent may include lidocaine, in a salt or base form, combined with binder and lubricant excipients. Implantable drug delivery devices including the tablets are also provided, e.g., one or more of the drug tablets may be contained in a biocompatible housing. The drug tablets may be substantially cylindrical with flat end faces, and the device may have from 10 to 100 drug tablets aligned in the housing with the flat end faces of adjacent tablets abutting one another.1. A solid drug unit comprising: a drug in the form of a mini-tablet which is greater than 70% by weight the drug, with the balance being at least one excipient, wherein the mini-tablet is cylindrical with flat end faces, the length of the tablet exceeding its diameter so that the mini-tablet has an aspect ratio of greater than 1:1, and wherein the mini-tablet has a diameter from 1.0 mm to 3.2 mm and a length from 1.7 mm to 4.8 mm, such that the mini-tablet is sized and shaped for insertion through the urethra of a patient. 2. The solid drug unit of claim 1, wherein the mini-tablet has a diameter from 1.5 mm to 3.1 mm and a length from 2.0 mm to 4.5 mm. 3. The solid drug unit of claim 1, wherein the drug is between 70 wt % and 99 wt % of the mini-tablet. 4. The solid drug unit of claim 1, wherein the drug is an anesthetic agent, an antimicrobial agent, or a chemotherapeutic agent. 5. The solid drug unit of claim 4, wherein the excipient comprises a binder and a lubricant. 6. The solid drug unit of claim 1, wherein the drug comprises lidocaine. 7. The solid drug unit of claim 1, wherein the mini-tablet is between 85 wt % and 95 wt % lidocaine. 8. The solid drug unit of claim 1, wherein the friability of the mini-tablet is less than 2%. 9. The solid drug unit of claim 1, wherein the mini-tablet is sterilized. 10. The solid drug unit of claim 1, wherein the aspect ratio is from about 3:2 to about 5:2. 11. A solid drug unit for insertion into an intravesical device, comprising: a drug in the form of a mini-tablet which is greater than 70% by weight the drug, with the balance being at least one excipient, wherein the mini-tablet is cylindrical with end faces that are not convex, the length of the tablet exceeding its diameter so that the mini-tablet has an aspect ratio of greater than 1:1, and wherein the mini-tablet has a diameter from 1.0 mm to 3.2 mm and a length from 1.7 mm to 4.8 mm, such that the mini-tablet is sized and shaped for insertion through the urethra of a patient. 12. The solid drug unit of claim 11, wherein the mini-tablet has a diameter from 1.5 mm to 3.1 mm and a length from 2.0 mm to 4.5 mm. 13. The solid drug unit of claim 11, wherein the drug is between 70 wt % and 99 wt % of the mini-tablet. 14. The solid drug unit of claim 11, wherein the drug is an anesthetic agent, an antimicrobial agent, or a chemotherapeutic agent. 15. The solid drug unit of claim 14, wherein the excipient comprises a binder and a lubricant. 16. The solid drug unit of claim 11, wherein the drug comprises lidocaine. 17. The solid drug unit of claim 11, wherein the mini-tablet is between 85 wt % and 95 wt % lidocaine. 18. The solid drug unit of claim 11, wherein the friability of the mini-tablet is less than 2%. 19. The solid drug unit of claim 11, wherein the mini-tablet is sterilized. 20. The solid drug unit of claim 11, wherein the aspect ratio is from about 3:2 to about 5:2.
1,600
238
238
12,252,905
1,616
Cerium oxide nanoparticles (CeONP) can be used to treat or prevent neurodegenerative diseases, including for example Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, AIDS-related dementia, ALS, progressive supranuclear palsy, and encephalitis, as well as mitochondrial diseases and diseases associated with mitochondrial damage. In particular, CeONP having an average size of about 2 nm to about 100 nm can be administered in an amount sufficient to block production of hydroxyl or superoxide radicals, block free radical production by Aβ (1-42) , block Aβ (1-42) -induced neuronal death, block Aβ (1-42) -induced [Ca 2+ ] i dysfunction in neurons, block Aβ (1-42) -induced lipid peroxidation, decrease loss of dopaminergic neurotransmission, or reduce mitochondrial dysfunction in a cell. CeONP can also be effective in treating conditions involving toxic exposures to compounds that induce mitochondrial dysfunction, such as rotenone, cyanide, carbon monoxide, polychlorinated biphenyls (PCBs) and other mitochondrial toxins.
1. A method of treating or preventing a neurodegenerative disease comprising administering cerium oxide nanoparticles having an average particle diameter size of about 2 nm to about 100 nm in an amount sufficient to block production of hydroxyl or superoxide radicals, block free radical production by Aβ(1-42), block Aβ(1-42)-induced neuronal death, block Aβ(1-42)-induced [Ca2+]i dysfunction in neurons, or block Aβ(1-42)-induced lipid peroxidation in a cell. 2. The method according to claim 1, which is a method of preventing or treating neurodegenerative diseases characterized by oxidative stress and free radical production. 3. The method according to claim 2, which is a method of preventing or treating Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, AIDS-related dementia, ALS, progressive supranuclear palsy, or encephalitis. 4. The method according to claim 3, wherein the average particle size of cerium oxide nanoparticles ranges from about 10 nm to about 20 nm. 5. A method of treating or preventing a neurodegenerative disease comprising administering cerium oxide nanoparticles having an average particle diameter size of about 2 nm to about 100 nm in an amount sufficient to decrease loss of dopaminergic neurotransmission, to deter or prevent dopaminergic neuronal loss, or to protect dopaminergic neurons in a cell. 6. The method according to claim 5, wherein the neurodegenerative disease is Parkinson's Disease. 7. The method according to claim 6, wherein the average particle size of cerium oxide nanoparticles ranges from about 10 nm to about 20 nm. 8. A method of treating or preventing mitochondrial diseases or effects of mitochondrial toxins comprising administering cerium oxide nanoparticles having an average particle diameter size of about 2 nm to about 100 nm in an amount sufficient to reduce mitochondrial dysfunction in a cell. 9. The method according to claim 8, which is a method of treating or preventing effects of mitochondrial toxins resulting from exposure of a cell to rotenone, cyanide, carbon monoxide, or polychlorinated biphenyls (PCBs). 10. The method according to claim 8, wherein the average particle size of cerium oxide nanoparticles ranges from about 10 nm to about 20 nm. 11. The method according to claim 9, wherein the mitochondrial dysfunction is mitochondrial failure from inhibition of mitochondrial Complex I. 12. The method according to claim 9, wherein the mitochondrial dysfunction is caused by a decrease in mitochondrial membrane potential (MMP) and wherein the amount of cerium oxide nanoparticles is sufficient to increase MMP, deter or prevent a decrease in MMP, or preserve MMP in a cell. 13. Cerium oxide nanoparticles having an average particle diameter ranging from about 2 nm to about 100 nm in an amount sufficient to provide a single dose of particles to block production of hydroxyl or superoxide radicals, block free radical production by Aβ(1-42), block Aβ(1-42)-induced neuronal death, block Aβ(1-42)-induced [Ca2+]i dysfunction in neurons, or block Aβ(1-42)-induced lipid peroxidation in a cell; to decrease loss of dopaminergic neurotransmission, to deter or prevent dopaminergic neuronal loss, or to protect dopaminergic neurons in a cell; or to reduce mitochondrial dysfunction in a cell. 14. The cerium oxide nanoparticles according to claim 13, wherein the particles have a diameter ranging from about 10 nm to about 20 nm.
Cerium oxide nanoparticles (CeONP) can be used to treat or prevent neurodegenerative diseases, including for example Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, AIDS-related dementia, ALS, progressive supranuclear palsy, and encephalitis, as well as mitochondrial diseases and diseases associated with mitochondrial damage. In particular, CeONP having an average size of about 2 nm to about 100 nm can be administered in an amount sufficient to block production of hydroxyl or superoxide radicals, block free radical production by Aβ (1-42) , block Aβ (1-42) -induced neuronal death, block Aβ (1-42) -induced [Ca 2+ ] i dysfunction in neurons, block Aβ (1-42) -induced lipid peroxidation, decrease loss of dopaminergic neurotransmission, or reduce mitochondrial dysfunction in a cell. CeONP can also be effective in treating conditions involving toxic exposures to compounds that induce mitochondrial dysfunction, such as rotenone, cyanide, carbon monoxide, polychlorinated biphenyls (PCBs) and other mitochondrial toxins.1. A method of treating or preventing a neurodegenerative disease comprising administering cerium oxide nanoparticles having an average particle diameter size of about 2 nm to about 100 nm in an amount sufficient to block production of hydroxyl or superoxide radicals, block free radical production by Aβ(1-42), block Aβ(1-42)-induced neuronal death, block Aβ(1-42)-induced [Ca2+]i dysfunction in neurons, or block Aβ(1-42)-induced lipid peroxidation in a cell. 2. The method according to claim 1, which is a method of preventing or treating neurodegenerative diseases characterized by oxidative stress and free radical production. 3. The method according to claim 2, which is a method of preventing or treating Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, AIDS-related dementia, ALS, progressive supranuclear palsy, or encephalitis. 4. The method according to claim 3, wherein the average particle size of cerium oxide nanoparticles ranges from about 10 nm to about 20 nm. 5. A method of treating or preventing a neurodegenerative disease comprising administering cerium oxide nanoparticles having an average particle diameter size of about 2 nm to about 100 nm in an amount sufficient to decrease loss of dopaminergic neurotransmission, to deter or prevent dopaminergic neuronal loss, or to protect dopaminergic neurons in a cell. 6. The method according to claim 5, wherein the neurodegenerative disease is Parkinson's Disease. 7. The method according to claim 6, wherein the average particle size of cerium oxide nanoparticles ranges from about 10 nm to about 20 nm. 8. A method of treating or preventing mitochondrial diseases or effects of mitochondrial toxins comprising administering cerium oxide nanoparticles having an average particle diameter size of about 2 nm to about 100 nm in an amount sufficient to reduce mitochondrial dysfunction in a cell. 9. The method according to claim 8, which is a method of treating or preventing effects of mitochondrial toxins resulting from exposure of a cell to rotenone, cyanide, carbon monoxide, or polychlorinated biphenyls (PCBs). 10. The method according to claim 8, wherein the average particle size of cerium oxide nanoparticles ranges from about 10 nm to about 20 nm. 11. The method according to claim 9, wherein the mitochondrial dysfunction is mitochondrial failure from inhibition of mitochondrial Complex I. 12. The method according to claim 9, wherein the mitochondrial dysfunction is caused by a decrease in mitochondrial membrane potential (MMP) and wherein the amount of cerium oxide nanoparticles is sufficient to increase MMP, deter or prevent a decrease in MMP, or preserve MMP in a cell. 13. Cerium oxide nanoparticles having an average particle diameter ranging from about 2 nm to about 100 nm in an amount sufficient to provide a single dose of particles to block production of hydroxyl or superoxide radicals, block free radical production by Aβ(1-42), block Aβ(1-42)-induced neuronal death, block Aβ(1-42)-induced [Ca2+]i dysfunction in neurons, or block Aβ(1-42)-induced lipid peroxidation in a cell; to decrease loss of dopaminergic neurotransmission, to deter or prevent dopaminergic neuronal loss, or to protect dopaminergic neurons in a cell; or to reduce mitochondrial dysfunction in a cell. 14. The cerium oxide nanoparticles according to claim 13, wherein the particles have a diameter ranging from about 10 nm to about 20 nm.
1,600
239
239
13,462,466
1,628
Novel compositions including bepotastine besilate and a corticosteroid are provided, compositions including at least about 0.008% w/v benzalkonium chloride, and compositions including hydroxypropylmethyl cellulose E15 LV.
1-68. (canceled) 69. A method of treating at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis in a patient in need of such treatment, the method comprising nasally administering a pharmaceutical composition comprising a corticosteroid as a free form, pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof and bepotastine as a free base, pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof at a concentration ranging from 0.5% w/v to 8.00% w/v in aqueous solution to the patient in need thereof, in a dose regimen effective to treat at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis. 70. The method of claim 69 wherein bepotastine in the composition administered is at a concentration ranging from 2.00% w/v to 4.00% w/v and corticosteroid in the composition administered is at a concentration ranging from 0.01% w/v to 1% w/v. 71. The method of claim 69 wherein administration is from 1 time a day to 4 times a day. 72. The method of claim 69 wherein bepotastine in the composition administered is at a concentration of either 3.00% w/v or 4.00% w/v and administration is 1 time a day or at more than 12 hour intervals. 73. The method of claim 71 wherein the dose regimen is effective to treat allergic rhinitis. 74. The method of claim 69 wherein the composition administered comprises dibasic sodium phosphate heptahydrate at a concentration of 0.10% w/v to 1.00% w/v; sodium chloride at a concentration of 0.9% w/v with 0.5% bepotastine, 0.4% w/v with 2.00%-3.00% bepotastine, 0.3% w/v with 4.00% bepotastine, 0.2% w/v with 6.00% bepotastine, 0.1% w/v with 8.00% bepotastine; edetate disodium at a concentration of 0.005% w/v to 0.100% w/v; benzalkonium chloride at a concentration of 0.002% w/v to 0.200% w/v; and one of either: a blend of microcrystalline cellulose and carboxymethyl cellulose (AVICEL®) at a concentration of 0.5% w/v to 2.5% w/v and polyoxyethylene (20) sorbitan monooleate (polysorbate 80) at a concentration of 0.005% w/v to 0.050% w/v, or HPMC E15 LV at a concentration of 0.01% w/v to 1.00% w/v, citric acid monohydrate at a concentration of 0.10% w/v to 1.00% w/v, and a taste-making agent at a concentration of 0.01% w/v to 1.00% w/v. 75. The method of claim 69 wherein the composition administered comprises dibasic sodium phosphate heptahydrate at a concentration of 0.70% w/v, sodium chloride at a concentration of 0.30% w/v, edetate disodium at a concentration of 0.020% w/v, benzalkonium chloride at a concentration of 0.020% w/v, and one of either: AVICEL® at a concentration of 2.00% w/v and polysorbate 80 at a concentration of 0.015% w/v, or HPMC E15 LV at a concentration of 0.10% w/v, citric acid monohydrate at a concentration of 0.10% w/v, and sucralose at a concentration of 0.10% w/v. 76. The method of claim 69 wherein the pharmaceutically acceptable salt of bepotastine is besilate. 77. The method of claim 74 or claim 75 wherein AVICEL® is AVICEL® CL-611. 78. The method of claim 69 wherein the corticosteroid is selected from the group consisting of beclomethasone, beclomethasone dipropionate, mometasone furoate monohydrate, fluticasone propionate, fluticasone furoate, triamcinolone, triamcinolone acetonide, budesonide, budesonide free acid, ciclesonide, beclomethasone sodium, dexamethasone sodium, prednisolone acetate, and mixtures thereof. 79. A method to treat at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis, the method comprising nasally administering to a patient in need thereof a composition comprising bepotastine besilate and a corticosteroid free form, pharmaceutically acceptable salt, solvate, or physiologically functional derivative thereof, dibasic sodium phosphate heptahydrate, sodium chloride, edetate disodium, benzalkonium chloride, and one of either: a blend of microcrystalline cellulose and carboxymethyl cellulose (AVICEL®) and/or polyoxyethylene (20) sorbitan monooleate (polysorbate 80), or hydroxypropylmethyl cellulose (HPMC), citric acid monohydrate, and a taste making agent; under conditions to treat at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis. 80. The method of claim 79 wherein rhinitis includes acute rhinitis, chronic rhinitis, allergic rhinitis, seasonal allergic rhinitis, perennial allergic rhinitis, vasomotor rhinitis, infectious rhinitis, and atrophic rhinitis. 81. The method of claim 79 wherein the composition is formulated as a nasal spray, nasal drops, nasal droplets, or combinations thereof. 82. The method of claim 79 wherein the composition is nasally administered by a metered dose inhaler (MDI). 83. The method of claim 82 wherein the MDI is any of a breath-actuated MDI, a dry powder inhaler, a spacer/holding chambers in combination with a MDI, and a nebulizer. 84. The method of claim 82 wherein the composition is in a wet spray formulation or a dry spray formulation. 85. The method of claim 79 wherein the composition is nasally administered by a metered dose plunger spray pump.
Novel compositions including bepotastine besilate and a corticosteroid are provided, compositions including at least about 0.008% w/v benzalkonium chloride, and compositions including hydroxypropylmethyl cellulose E15 LV.1-68. (canceled) 69. A method of treating at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis in a patient in need of such treatment, the method comprising nasally administering a pharmaceutical composition comprising a corticosteroid as a free form, pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof and bepotastine as a free base, pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof at a concentration ranging from 0.5% w/v to 8.00% w/v in aqueous solution to the patient in need thereof, in a dose regimen effective to treat at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis. 70. The method of claim 69 wherein bepotastine in the composition administered is at a concentration ranging from 2.00% w/v to 4.00% w/v and corticosteroid in the composition administered is at a concentration ranging from 0.01% w/v to 1% w/v. 71. The method of claim 69 wherein administration is from 1 time a day to 4 times a day. 72. The method of claim 69 wherein bepotastine in the composition administered is at a concentration of either 3.00% w/v or 4.00% w/v and administration is 1 time a day or at more than 12 hour intervals. 73. The method of claim 71 wherein the dose regimen is effective to treat allergic rhinitis. 74. The method of claim 69 wherein the composition administered comprises dibasic sodium phosphate heptahydrate at a concentration of 0.10% w/v to 1.00% w/v; sodium chloride at a concentration of 0.9% w/v with 0.5% bepotastine, 0.4% w/v with 2.00%-3.00% bepotastine, 0.3% w/v with 4.00% bepotastine, 0.2% w/v with 6.00% bepotastine, 0.1% w/v with 8.00% bepotastine; edetate disodium at a concentration of 0.005% w/v to 0.100% w/v; benzalkonium chloride at a concentration of 0.002% w/v to 0.200% w/v; and one of either: a blend of microcrystalline cellulose and carboxymethyl cellulose (AVICEL®) at a concentration of 0.5% w/v to 2.5% w/v and polyoxyethylene (20) sorbitan monooleate (polysorbate 80) at a concentration of 0.005% w/v to 0.050% w/v, or HPMC E15 LV at a concentration of 0.01% w/v to 1.00% w/v, citric acid monohydrate at a concentration of 0.10% w/v to 1.00% w/v, and a taste-making agent at a concentration of 0.01% w/v to 1.00% w/v. 75. The method of claim 69 wherein the composition administered comprises dibasic sodium phosphate heptahydrate at a concentration of 0.70% w/v, sodium chloride at a concentration of 0.30% w/v, edetate disodium at a concentration of 0.020% w/v, benzalkonium chloride at a concentration of 0.020% w/v, and one of either: AVICEL® at a concentration of 2.00% w/v and polysorbate 80 at a concentration of 0.015% w/v, or HPMC E15 LV at a concentration of 0.10% w/v, citric acid monohydrate at a concentration of 0.10% w/v, and sucralose at a concentration of 0.10% w/v. 76. The method of claim 69 wherein the pharmaceutically acceptable salt of bepotastine is besilate. 77. The method of claim 74 or claim 75 wherein AVICEL® is AVICEL® CL-611. 78. The method of claim 69 wherein the corticosteroid is selected from the group consisting of beclomethasone, beclomethasone dipropionate, mometasone furoate monohydrate, fluticasone propionate, fluticasone furoate, triamcinolone, triamcinolone acetonide, budesonide, budesonide free acid, ciclesonide, beclomethasone sodium, dexamethasone sodium, prednisolone acetate, and mixtures thereof. 79. A method to treat at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis, the method comprising nasally administering to a patient in need thereof a composition comprising bepotastine besilate and a corticosteroid free form, pharmaceutically acceptable salt, solvate, or physiologically functional derivative thereof, dibasic sodium phosphate heptahydrate, sodium chloride, edetate disodium, benzalkonium chloride, and one of either: a blend of microcrystalline cellulose and carboxymethyl cellulose (AVICEL®) and/or polyoxyethylene (20) sorbitan monooleate (polysorbate 80), or hydroxypropylmethyl cellulose (HPMC), citric acid monohydrate, and a taste making agent; under conditions to treat at least one of rhinitis, mucosal inflammation associated with rhinitis, sinusitis, rhinosinusitis, and symptoms associated with rhinitis, mucosal inflammation associated with rhinitis, sinusitis, or rhinosinusitis. 80. The method of claim 79 wherein rhinitis includes acute rhinitis, chronic rhinitis, allergic rhinitis, seasonal allergic rhinitis, perennial allergic rhinitis, vasomotor rhinitis, infectious rhinitis, and atrophic rhinitis. 81. The method of claim 79 wherein the composition is formulated as a nasal spray, nasal drops, nasal droplets, or combinations thereof. 82. The method of claim 79 wherein the composition is nasally administered by a metered dose inhaler (MDI). 83. The method of claim 82 wherein the MDI is any of a breath-actuated MDI, a dry powder inhaler, a spacer/holding chambers in combination with a MDI, and a nebulizer. 84. The method of claim 82 wherein the composition is in a wet spray formulation or a dry spray formulation. 85. The method of claim 79 wherein the composition is nasally administered by a metered dose plunger spray pump.
1,600
240
240
13,114,617
1,616
The present invention is directed to a composition for the conditioning of a dental mineralized surface and/or a dental implant and/or for increasing the hydrophilicity of a dental implant having a metal containing surface. The composition comprises EDTA in a concentration of about 21-55% by weight and hydrogen peroxide at a concentration of about 2-4% by weight. The composition of the invention may be used for the conditioning of mineralized dental surfaces and dental implant surfaces in order to remove e.g. biofilm, debris, bacteria, bacterial toxins etc and/or for increasing the hydrophilicity of an implant having a metal containing surface. The invention is also directed to a kit comprising the composition of the invention and a device to apply the composition to a surface in need of conditioning.
1. A conditioning composition comprising ethylenediaminetetraacetic acid (EDTA) at a concentration in the range of 21-55% by weight and hydrogen peroxide (H2O2) at a concentration of 2-4% by weight. 2. A conditioning composition according to claim 1, wherein the concentration of EDTA is in the range of 21-27% by weight. 3. A conditioning composition according to claim 1, wherein the concentration of EDTA is in the range of 21.6-26.4 by weight. 4. A conditioning composition according to claim 1, wherein the concentration of hydrogen peroxide is about 3% by weight. 5. A conditioning composition according to claim 1, wherein the pH of the composition is in the range of 6-11.5. 6. A conditioning composition according to claim 1, wherein the pH of the composition is 9.0 or above. 7. A conditioning composition according to claim 1, further comprising a viscosity increasing agent. 8. A conditioning composition according to claim 7, wherein the viscosity increasing agent is selected from the group consisting of polysaccharides, proteins, glycoproteins and synthetic polymers. 9. A conditioning composition according to claim 7, wherein said viscosity increasing agent is carboxymethylcellulose, or a salt thereof. 10. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, and an aqueous solvent. 11. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, an aqueous solvent and a pH buffering agent. 12. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, an aqueous solvent and a viscosity increasing agent. 13. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, an aqueous solvent, a pH buffering agent and a viscosity increasing agent. 14. A conditioning composition according to claim 1, wherein the composition is a medicament. 15. A method for preventing and/or treating a periodontal disease, periimplantitis, caries and/or an infection and/or inflammation in the root canal comprising the administration of a therapeutically effective amount of the composition according to claim 1 to a subject, in need thereof. 16. A method according to claim 15, wherein said method comprises the conditioning of a mineralized dental surface and/or the surface of a dental implant with the conditioning composition according to claim 1. 17. A method for conditioning a mineralized dental surface and/or a surface of a dental implant comprising treating the surface with an effective amount of the conditioning composition according to claim 1. 18. A method for enabling and/or enhancing osseointegration of an implant comprising treating the implant with the conditioning composition according to claim 1. 19. A kit comprising a vial with the conditioning composition according to claim 1 and a device for the application to, and/or a device for cleaning, a mineralized dental surface and/or a surface of a dental implant. 20. A method according to claim 15, wherein the subject is a mammal.
The present invention is directed to a composition for the conditioning of a dental mineralized surface and/or a dental implant and/or for increasing the hydrophilicity of a dental implant having a metal containing surface. The composition comprises EDTA in a concentration of about 21-55% by weight and hydrogen peroxide at a concentration of about 2-4% by weight. The composition of the invention may be used for the conditioning of mineralized dental surfaces and dental implant surfaces in order to remove e.g. biofilm, debris, bacteria, bacterial toxins etc and/or for increasing the hydrophilicity of an implant having a metal containing surface. The invention is also directed to a kit comprising the composition of the invention and a device to apply the composition to a surface in need of conditioning.1. A conditioning composition comprising ethylenediaminetetraacetic acid (EDTA) at a concentration in the range of 21-55% by weight and hydrogen peroxide (H2O2) at a concentration of 2-4% by weight. 2. A conditioning composition according to claim 1, wherein the concentration of EDTA is in the range of 21-27% by weight. 3. A conditioning composition according to claim 1, wherein the concentration of EDTA is in the range of 21.6-26.4 by weight. 4. A conditioning composition according to claim 1, wherein the concentration of hydrogen peroxide is about 3% by weight. 5. A conditioning composition according to claim 1, wherein the pH of the composition is in the range of 6-11.5. 6. A conditioning composition according to claim 1, wherein the pH of the composition is 9.0 or above. 7. A conditioning composition according to claim 1, further comprising a viscosity increasing agent. 8. A conditioning composition according to claim 7, wherein the viscosity increasing agent is selected from the group consisting of polysaccharides, proteins, glycoproteins and synthetic polymers. 9. A conditioning composition according to claim 7, wherein said viscosity increasing agent is carboxymethylcellulose, or a salt thereof. 10. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, and an aqueous solvent. 11. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, an aqueous solvent and a pH buffering agent. 12. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, an aqueous solvent and a viscosity increasing agent. 13. A conditioning composition consisting essentially of EDTA at a concentration in the range of 21-55% by weight, hydrogen peroxide at a concentration of 2-4% by weight, an aqueous solvent, a pH buffering agent and a viscosity increasing agent. 14. A conditioning composition according to claim 1, wherein the composition is a medicament. 15. A method for preventing and/or treating a periodontal disease, periimplantitis, caries and/or an infection and/or inflammation in the root canal comprising the administration of a therapeutically effective amount of the composition according to claim 1 to a subject, in need thereof. 16. A method according to claim 15, wherein said method comprises the conditioning of a mineralized dental surface and/or the surface of a dental implant with the conditioning composition according to claim 1. 17. A method for conditioning a mineralized dental surface and/or a surface of a dental implant comprising treating the surface with an effective amount of the conditioning composition according to claim 1. 18. A method for enabling and/or enhancing osseointegration of an implant comprising treating the implant with the conditioning composition according to claim 1. 19. A kit comprising a vial with the conditioning composition according to claim 1 and a device for the application to, and/or a device for cleaning, a mineralized dental surface and/or a surface of a dental implant. 20. A method according to claim 15, wherein the subject is a mammal.
1,600
241
241
14,783,633
1,632
Provided herein are, inter alia, are media compositions useful for culturing neural cells. In particular, the compositions provided herein mimic important physiological conditions in the living brain and sustain neural activity. The media compositions provided herein improve the efficiency of human neuron maturation and promote synaptic function in long-term in vitro cultures.
1. A cell medium comprising: (a) Sodium chloride at a concentration of between about 70 and about 150 mM; (b) a neuroactive inorganic salt at a concentration of between about 0.000001 and about 10 mM; (c) Glycine at a concentration of between about 0.0001 and about 0.05 mM; (d) L-alanine at a concentration of between about 00001 and about 0.05 mM; and (e) L-serine at a concentration of between about 0.001 and about 0.03 mM. 2. The cell medium of claim 1, wherein the neuroactive inorganic salt is selected from the group consisting of Potassium Chloride, Calcium Chloride, Magnesium Sulfate, Magnesium Chloride, Ferric Nitrate, Zinc sulfate, Cupric sulfate, Ferric sulfate, and combinations thereof. 3. The cell medium of claim 1, further comprising L-aspartic acid at a concentration of between about 0.00001 and about 0.003 mM. 4. The cell medium of claim 1, further comprising L-glutamic acid at a concentration of between about 0.00001 and about 0.02 mM. 5. The cell medium of claim 1, further comprising a pH modulating agent, wherein the agent is an inorganic salt. 6. The cell medium of claim 5, wherein the inorganic salt is at a concentration of between about 0.001 and about 1 mM, and wherein the inorganic salt is selected from the group consisting of Sodium Phosphate dibasic, Sodium Phosphate monobasic, and combinations thereof. 7. The cell medium of claim 5, wherein the inorganic salt is at a concentration of between about 1 and about 35 mM, and wherein the inorganic salt is Sodium Bicarbonate. 8. The cell medium of claim 1, further comprising one or more amino acid, wherein each amino acid is at a concentration of between about 0.001 and about 1 mM. 9. The cell medium of claim 8, wherein the one or more amino acid is selected from the group consisting of L-Alanyl-L-Glutamine, L-Arginine hydrochloride, L-Asparagine-H2O, L-Cysteine hydrochloride-H2O, L-Cystine 2HCl, L-Histidine hydrochloride-H2O, L-Isoleucine, L-Leucine, L-Lysine hydrochloride, L-Methionine, L-Phenylalanine, L-Proline, L-Threonine, L-Tryptophan, L-Tyrosine disodium salt dihydrate, L-Valine, and combinations thereof. 10. The cell medium of claim 1, further comprising on or more vitamin, wherein each vitamin is present at a concentration of between about 0.00001 and about 1 mM. 11. The cell medium of claim 10, wherein the one or more vitamin is selected from the group consisting of Choline chloride, D-Calcium pantothenate (B5), Folic Acid (B9), i-Inositol, Niacinamide (B3), Pyridoxine hydrochloride, Thiamine hydrochloride, Vitamin B12 (cyanocobalamin), Riboflavin (B2), and combinations thereof. 12. The cell medium of claim 1, further comprising a supplemental agent selected from the group consisting of a protein, neurotrophic factor, steroid, hormone, fatty acid, lipid, vitamin, sulfate mineral, organic chemical compound, monosaccharide, nucleotide, and combinations thereof. 13. The cell medium of claim 1, further comprising an energetic substrate at a concentration of between 0.1 and about 5 mM. 14. The cell medium of claim 13, wherein the energetic substrate is selected from the group consisting of sugar, sodium pyruvate, and combinations thereof. 15. The cell medium of claim 1, further comprising a light sensitive agent. 16. The cell medium of claim 15, wherein the light sensitive agent is Riboflavin (B2) at a concentration of between about 0.0001 and about 0.0006 mM. 17. The cell medium of claim 15, wherein the light sensitive agent is HEPES at a concentration of between about 1 and about 10 mM. 18. The cell medium of claim 1, wherein the cell medium does not comprise serum. 19. The cell medium of claim 1, wherein the osmolarity of the medium is between about 280 and about 330 Osm/L. 20. A method of culturing a neuronal cell comprising contacting a neuronal cell with a medium comprising (a) Sodium chloride at a concentration of between about 70 and about 150 mM; (b) a neuroactive inorganic salt at a concentration of between about 0.000001 and about 10 mM; (c) Glycine at a concentration of between about 0.0001 and about 0.05 mM; (d) L-alanine at a concentration of between about 00001 and about 0.05 mM; and (e) L-serine at a concentration of between about 0.001 and about 0.03 mM. 21. The method of claim 20, wherein the neuronal cell is a primary neuronal cell. 22. The method of claim 20, wherein said neuronal cell is an iPSC-derived neuronal cell. 23. A mammalian cell cultured in a medium comprising (a) Sodium chloride at a concentration of between about 70 and about 150 mM; (b) a neuroactive inorganic salt at a concentration of between about 0.000001 and about 10 mM; (c) Glycine at a concentration of between about 0.0001 and about 0.05 mM; (d) L-alanine at a concentration of between about 00001 and about 0.05 mM; and (e) L-serine at a concentration of between about 0.001 and about 0.03 mM. 24. The mammalian cell of claim 23, wherein the mammalian cell is a neuronal cell.
Provided herein are, inter alia, are media compositions useful for culturing neural cells. In particular, the compositions provided herein mimic important physiological conditions in the living brain and sustain neural activity. The media compositions provided herein improve the efficiency of human neuron maturation and promote synaptic function in long-term in vitro cultures.1. A cell medium comprising: (a) Sodium chloride at a concentration of between about 70 and about 150 mM; (b) a neuroactive inorganic salt at a concentration of between about 0.000001 and about 10 mM; (c) Glycine at a concentration of between about 0.0001 and about 0.05 mM; (d) L-alanine at a concentration of between about 00001 and about 0.05 mM; and (e) L-serine at a concentration of between about 0.001 and about 0.03 mM. 2. The cell medium of claim 1, wherein the neuroactive inorganic salt is selected from the group consisting of Potassium Chloride, Calcium Chloride, Magnesium Sulfate, Magnesium Chloride, Ferric Nitrate, Zinc sulfate, Cupric sulfate, Ferric sulfate, and combinations thereof. 3. The cell medium of claim 1, further comprising L-aspartic acid at a concentration of between about 0.00001 and about 0.003 mM. 4. The cell medium of claim 1, further comprising L-glutamic acid at a concentration of between about 0.00001 and about 0.02 mM. 5. The cell medium of claim 1, further comprising a pH modulating agent, wherein the agent is an inorganic salt. 6. The cell medium of claim 5, wherein the inorganic salt is at a concentration of between about 0.001 and about 1 mM, and wherein the inorganic salt is selected from the group consisting of Sodium Phosphate dibasic, Sodium Phosphate monobasic, and combinations thereof. 7. The cell medium of claim 5, wherein the inorganic salt is at a concentration of between about 1 and about 35 mM, and wherein the inorganic salt is Sodium Bicarbonate. 8. The cell medium of claim 1, further comprising one or more amino acid, wherein each amino acid is at a concentration of between about 0.001 and about 1 mM. 9. The cell medium of claim 8, wherein the one or more amino acid is selected from the group consisting of L-Alanyl-L-Glutamine, L-Arginine hydrochloride, L-Asparagine-H2O, L-Cysteine hydrochloride-H2O, L-Cystine 2HCl, L-Histidine hydrochloride-H2O, L-Isoleucine, L-Leucine, L-Lysine hydrochloride, L-Methionine, L-Phenylalanine, L-Proline, L-Threonine, L-Tryptophan, L-Tyrosine disodium salt dihydrate, L-Valine, and combinations thereof. 10. The cell medium of claim 1, further comprising on or more vitamin, wherein each vitamin is present at a concentration of between about 0.00001 and about 1 mM. 11. The cell medium of claim 10, wherein the one or more vitamin is selected from the group consisting of Choline chloride, D-Calcium pantothenate (B5), Folic Acid (B9), i-Inositol, Niacinamide (B3), Pyridoxine hydrochloride, Thiamine hydrochloride, Vitamin B12 (cyanocobalamin), Riboflavin (B2), and combinations thereof. 12. The cell medium of claim 1, further comprising a supplemental agent selected from the group consisting of a protein, neurotrophic factor, steroid, hormone, fatty acid, lipid, vitamin, sulfate mineral, organic chemical compound, monosaccharide, nucleotide, and combinations thereof. 13. The cell medium of claim 1, further comprising an energetic substrate at a concentration of between 0.1 and about 5 mM. 14. The cell medium of claim 13, wherein the energetic substrate is selected from the group consisting of sugar, sodium pyruvate, and combinations thereof. 15. The cell medium of claim 1, further comprising a light sensitive agent. 16. The cell medium of claim 15, wherein the light sensitive agent is Riboflavin (B2) at a concentration of between about 0.0001 and about 0.0006 mM. 17. The cell medium of claim 15, wherein the light sensitive agent is HEPES at a concentration of between about 1 and about 10 mM. 18. The cell medium of claim 1, wherein the cell medium does not comprise serum. 19. The cell medium of claim 1, wherein the osmolarity of the medium is between about 280 and about 330 Osm/L. 20. A method of culturing a neuronal cell comprising contacting a neuronal cell with a medium comprising (a) Sodium chloride at a concentration of between about 70 and about 150 mM; (b) a neuroactive inorganic salt at a concentration of between about 0.000001 and about 10 mM; (c) Glycine at a concentration of between about 0.0001 and about 0.05 mM; (d) L-alanine at a concentration of between about 00001 and about 0.05 mM; and (e) L-serine at a concentration of between about 0.001 and about 0.03 mM. 21. The method of claim 20, wherein the neuronal cell is a primary neuronal cell. 22. The method of claim 20, wherein said neuronal cell is an iPSC-derived neuronal cell. 23. A mammalian cell cultured in a medium comprising (a) Sodium chloride at a concentration of between about 70 and about 150 mM; (b) a neuroactive inorganic salt at a concentration of between about 0.000001 and about 10 mM; (c) Glycine at a concentration of between about 0.0001 and about 0.05 mM; (d) L-alanine at a concentration of between about 00001 and about 0.05 mM; and (e) L-serine at a concentration of between about 0.001 and about 0.03 mM. 24. The mammalian cell of claim 23, wherein the mammalian cell is a neuronal cell.
1,600
242
242
12,866,655
1,612
This invention relates to oral care compositions comprising an effective amount of a basic amino acid in free or salt form and a low dosage of fluoride and to methods of using such compositions.
1. An oral care composition comprising: a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride ion source which comprises not greater than 0.1% by weight of the composition. 2. The composition according claim 1 wherein the basic amino acid is arginine. 3. The composition according to claim 1 wherein the basic amino acid is in salt form and selected from arginine phosphate, arginine bicarbonate, and arginine hydrochloride. 4. The composition according to claim 1, further comprising an anionic surfactant selected from sodium lauryl sulfate; sodium laureth sulfate; and mixtures thereof. 5. The composition according to claim 4 wherein the anionic surfactant is present in an amount of about 0.01 to about 10 weight % of the total composition. 6. The composition according to claim 5 wherein the anionic surfactant is present in an amount of about 0.3 to about 4.5 weight % of the total composition. 7. The composition according to claim 1, wherein the fluoride ion source is selected from sodium fluoride, sodium monofluorophosphate, stannous fluoride, potassium fluoride, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 8. The composition according to claim 1 further comprising an antibacterial agent. 9. The composition according to claim 8 wherein the antibacterial agent is triclosan. 10. The composition according to claim 1 further comprising an anionic polymer. 11. The composition according to claim 10 wherein the anionic polymer is a copolymer of methyl vinyl ether and maleic anhydride. 12. An oral care composition comprising a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride ion source that provides not greater than about 600 ppm of fluoride ions. 13. The composition according to claim 12 wherein the basic amino acid is arginine. 14. The composition according to claim 12 wherein the basic amino acid is in salt form and is selected from arginine phosphate, arginine bicarbonate, and arginine hydrochloride. 15. The composition according to claim 12 wherein the anionic surfactant is selected from sodium lauryl sulfate and sodium laureth sulfate and mixtures thereof. 16. The composition according to claim 12 wherein the anionic surfactant is present in an amount of about 0.01 to about 10 weight % of the total composition. 17. The composition according to claim 16 wherein the anionic surfactant is present in an amount of about 0.3 to about 4.5 weight % of the total composition. 18. The composition according to claim 12, wherein the fluoride ion source is selected from sodium fluoride, sodium monofluorophosphate, stannous fluoride, potassium fluoride, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 19. The composition according to claim 12 further comprising an antibacterial agent. 20. The composition according to claim 19 wherein the antibacterial agent is triclosan. 21. The composition according to claim 12 further comprising an anionic polymer. 22. The composition according to claim 21 wherein the anionic polymer is a copolymer of methyl vinyl ether and maleic anhydride. 23. A method to improve the oral health of a subject desiring a low fluoride dosage comprising administering to said subject an oral care composition comprising; a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride source that provides not greater than about 600 ppm of fluoride ions. 24. The method of claim 23 wherein the subject is in need of: a. reducing or inhibiting formation of dental caries, b. reducing, repairing or inhibiting early enamel lesions, c, reducing or inhibiting demineralization and promoting remineralization of the teeth, d. reducing hypersensitivity of the teeth, e. reducing or inhibiting gingivitis, f. promoting healing of sores or cuts in the mouth, g. reducing levels of acid producing bacteria, h. increasing relative levels of arginolytic bacteria, i. inhibiting microbial biofilm formation in the oral cavity, j. raising and/or maintaining plaque pH at levels of at least pH 55 following sugar challenge, k. reducing plaque accumulation, l. treating, relieving or reducing dry mouth, m. whitening teeth, n. promoting systemic health, including cardiovascular health, o. immunizing the teeth against cariogenic bacteria, p. reducing erosion of the teeth, and/or q. cleaning the teeth and oral cavity. 25. The composition of claim 1, further comprising a potassium ion source. 26. The composition of claim 25, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 27. The composition of claim 12, further comprising a potassium ion source. 28. The composition of claim 27, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 29. A method for promoting systemic health comprising administering to a subject in need thereof, an oral care composition comprising: a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride ion source that provides not greater than about 600 ppm of fluoride ions. 30. The method of claim 29, wherein the composition further comprises a potassium ion source. 31. The method of claim 30, wherein the potassium ion source is selected from potassium nitrate and potassium chloride.
This invention relates to oral care compositions comprising an effective amount of a basic amino acid in free or salt form and a low dosage of fluoride and to methods of using such compositions.1. An oral care composition comprising: a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride ion source which comprises not greater than 0.1% by weight of the composition. 2. The composition according claim 1 wherein the basic amino acid is arginine. 3. The composition according to claim 1 wherein the basic amino acid is in salt form and selected from arginine phosphate, arginine bicarbonate, and arginine hydrochloride. 4. The composition according to claim 1, further comprising an anionic surfactant selected from sodium lauryl sulfate; sodium laureth sulfate; and mixtures thereof. 5. The composition according to claim 4 wherein the anionic surfactant is present in an amount of about 0.01 to about 10 weight % of the total composition. 6. The composition according to claim 5 wherein the anionic surfactant is present in an amount of about 0.3 to about 4.5 weight % of the total composition. 7. The composition according to claim 1, wherein the fluoride ion source is selected from sodium fluoride, sodium monofluorophosphate, stannous fluoride, potassium fluoride, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 8. The composition according to claim 1 further comprising an antibacterial agent. 9. The composition according to claim 8 wherein the antibacterial agent is triclosan. 10. The composition according to claim 1 further comprising an anionic polymer. 11. The composition according to claim 10 wherein the anionic polymer is a copolymer of methyl vinyl ether and maleic anhydride. 12. An oral care composition comprising a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride ion source that provides not greater than about 600 ppm of fluoride ions. 13. The composition according to claim 12 wherein the basic amino acid is arginine. 14. The composition according to claim 12 wherein the basic amino acid is in salt form and is selected from arginine phosphate, arginine bicarbonate, and arginine hydrochloride. 15. The composition according to claim 12 wherein the anionic surfactant is selected from sodium lauryl sulfate and sodium laureth sulfate and mixtures thereof. 16. The composition according to claim 12 wherein the anionic surfactant is present in an amount of about 0.01 to about 10 weight % of the total composition. 17. The composition according to claim 16 wherein the anionic surfactant is present in an amount of about 0.3 to about 4.5 weight % of the total composition. 18. The composition according to claim 12, wherein the fluoride ion source is selected from sodium fluoride, sodium monofluorophosphate, stannous fluoride, potassium fluoride, sodium fluorosilicate, ammonium fluorosilicate, amine fluoride, ammonium fluoride, and combinations thereof. 19. The composition according to claim 12 further comprising an antibacterial agent. 20. The composition according to claim 19 wherein the antibacterial agent is triclosan. 21. The composition according to claim 12 further comprising an anionic polymer. 22. The composition according to claim 21 wherein the anionic polymer is a copolymer of methyl vinyl ether and maleic anhydride. 23. A method to improve the oral health of a subject desiring a low fluoride dosage comprising administering to said subject an oral care composition comprising; a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride source that provides not greater than about 600 ppm of fluoride ions. 24. The method of claim 23 wherein the subject is in need of: a. reducing or inhibiting formation of dental caries, b. reducing, repairing or inhibiting early enamel lesions, c, reducing or inhibiting demineralization and promoting remineralization of the teeth, d. reducing hypersensitivity of the teeth, e. reducing or inhibiting gingivitis, f. promoting healing of sores or cuts in the mouth, g. reducing levels of acid producing bacteria, h. increasing relative levels of arginolytic bacteria, i. inhibiting microbial biofilm formation in the oral cavity, j. raising and/or maintaining plaque pH at levels of at least pH 55 following sugar challenge, k. reducing plaque accumulation, l. treating, relieving or reducing dry mouth, m. whitening teeth, n. promoting systemic health, including cardiovascular health, o. immunizing the teeth against cariogenic bacteria, p. reducing erosion of the teeth, and/or q. cleaning the teeth and oral cavity. 25. The composition of claim 1, further comprising a potassium ion source. 26. The composition of claim 25, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 27. The composition of claim 12, further comprising a potassium ion source. 28. The composition of claim 27, wherein said potassium ion source is selected from potassium nitrate and potassium chloride. 29. A method for promoting systemic health comprising administering to a subject in need thereof, an oral care composition comprising: a. an effective amount of a basic amino acid, in free or salt form; and b. a fluoride ion source that provides not greater than about 600 ppm of fluoride ions. 30. The method of claim 29, wherein the composition further comprises a potassium ion source. 31. The method of claim 30, wherein the potassium ion source is selected from potassium nitrate and potassium chloride.
1,600
243
243
15,197,541
1,633
The present invention provides silk proteins, as well as nucleic acids encoding these proteins. The present invention also provides recombinant cells and/or organisms which synthesize silk proteins. Silk proteins of the invention can be used for a variety of purposes such as in the manufacture of personal care products, plastics, textiles, and biomedical products.
1-45. (canceled) 46. A silk polypeptide, wherein at least a portion of the polypeptide has a coiled coil structure, and wherein the polypeptide comprises an amino acid sequence which is at least 40% identical to any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63, and wherein the silk polypeptide is fused to at least one other polypeptide; or wherein the silk polypeptide is present in a silk fiber or copolymer and crosslinked to a surface of interest. 47. The silk polypeptide of claim 46, wherein i) the portion of the polypeptide that has a coiled coil structure comprises at least 10 copies of the heptad sequence abcdefg, and wherein at least 25% of the amino acids at positions a and d are alanine residues, or ii) the portion of the polypeptide that has a coiled coil structure comprises at least 10 copies of the heptad sequence abcdefg, and at least 25% of the amino acids at positions a, d and e are alanine residues. 48. The silk polypeptide of claim 46 which is fused to at least one other polypeptide. 49. The silk polypeptide of claim 46, wherein the silk polypeptide is present in a silk fiber or copolymer and crosslinked to a surface of interest. 50. A product comprising at least one silk polypeptide of claim 46. 51. The product of claim 50, wherein the product is selected from the group consisting of: a personal care product, textiles, plastics, and biomedical products. 52. A vector comprising: a polynucleotide which encodes a silk polypeptide, wherein at least a portion of the polypeptide has a coiled coil structure, and wherein the silk polypeptide comprises an amino acid sequence which is at least 40% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63; and a heterologous promoter operably linked to the polynucleotide. 53. A composition comprising the vector of claim 52, and one or more acceptable carriers. 54. A recombinant host cell comprising a polynucleotide which encodes a silk polypeptide, wherein at least a portion of the polypeptide has a coiled coil structure, and wherein the silk polypeptide comprises an amino acid sequence which is at least 40% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63; and wherein a) the polynucleotide is operably linked to a heterologous promoter, and/or b) the recombinant host cell is a bacterial, yeast or plant cell. 55. The recombinant host cell of claim 54, wherein the silk polypeptide comprises an amino acid sequence which is at least 50% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63. 56. The recombinant host cell of claim 54, wherein the silk polypeptide comprises an amino acid sequence which is at least 70% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63. 57. The recombinant host cell of claim 54, wherein the silk polypeptide comprises an amino acid sequence which is at least 80% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63. 58. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 40% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 59. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 50% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 60. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 70% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 61. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 80% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 62. The recombinant host cell of claim 54, wherein the portion of the silk polypeptide that has a coiled coil structure comprises at least 10 copies of the heptad sequence abcdefg, and wherein at least 25% of the amino acids at positions a and d are alanine residues. 63. The recombinant host cell of claim 54, wherein the portion of the silk polypeptide that has a coiled coil structure comprises at least 18 copies of the heptad sequence abcdefg, and wherein at least 25% of the amino acids at positions a and d are alanine residues. 64. The recombinant host cell of claim 54 which is a bacterial cell. 65. A process for preparing a silk polypeptide comprising cultivating the recombinant host cell of claim 54, under conditions which allow expression of the polynucleotide encoding the polypeptide, and recovering the expressed polypeptide.
The present invention provides silk proteins, as well as nucleic acids encoding these proteins. The present invention also provides recombinant cells and/or organisms which synthesize silk proteins. Silk proteins of the invention can be used for a variety of purposes such as in the manufacture of personal care products, plastics, textiles, and biomedical products.1-45. (canceled) 46. A silk polypeptide, wherein at least a portion of the polypeptide has a coiled coil structure, and wherein the polypeptide comprises an amino acid sequence which is at least 40% identical to any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63, and wherein the silk polypeptide is fused to at least one other polypeptide; or wherein the silk polypeptide is present in a silk fiber or copolymer and crosslinked to a surface of interest. 47. The silk polypeptide of claim 46, wherein i) the portion of the polypeptide that has a coiled coil structure comprises at least 10 copies of the heptad sequence abcdefg, and wherein at least 25% of the amino acids at positions a and d are alanine residues, or ii) the portion of the polypeptide that has a coiled coil structure comprises at least 10 copies of the heptad sequence abcdefg, and at least 25% of the amino acids at positions a, d and e are alanine residues. 48. The silk polypeptide of claim 46 which is fused to at least one other polypeptide. 49. The silk polypeptide of claim 46, wherein the silk polypeptide is present in a silk fiber or copolymer and crosslinked to a surface of interest. 50. A product comprising at least one silk polypeptide of claim 46. 51. The product of claim 50, wherein the product is selected from the group consisting of: a personal care product, textiles, plastics, and biomedical products. 52. A vector comprising: a polynucleotide which encodes a silk polypeptide, wherein at least a portion of the polypeptide has a coiled coil structure, and wherein the silk polypeptide comprises an amino acid sequence which is at least 40% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63; and a heterologous promoter operably linked to the polynucleotide. 53. A composition comprising the vector of claim 52, and one or more acceptable carriers. 54. A recombinant host cell comprising a polynucleotide which encodes a silk polypeptide, wherein at least a portion of the polypeptide has a coiled coil structure, and wherein the silk polypeptide comprises an amino acid sequence which is at least 40% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63; and wherein a) the polynucleotide is operably linked to a heterologous promoter, and/or b) the recombinant host cell is a bacterial, yeast or plant cell. 55. The recombinant host cell of claim 54, wherein the silk polypeptide comprises an amino acid sequence which is at least 50% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63. 56. The recombinant host cell of claim 54, wherein the silk polypeptide comprises an amino acid sequence which is at least 70% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63. 57. The recombinant host cell of claim 54, wherein the silk polypeptide comprises an amino acid sequence which is at least 80% identical to at least any one or more of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:56, SEQ ID NO:57; SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:62, or SEQ ID NO:63. 58. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 40% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 59. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 50% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 60. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 70% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 61. The recombinant host cell of claim 54, wherein the polynucleotide comprises a nucleic acid sequence which is at least 80% identical to any one or more of SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:70, SEQ ID NO:71, or SEQ ID NO:76. 62. The recombinant host cell of claim 54, wherein the portion of the silk polypeptide that has a coiled coil structure comprises at least 10 copies of the heptad sequence abcdefg, and wherein at least 25% of the amino acids at positions a and d are alanine residues. 63. The recombinant host cell of claim 54, wherein the portion of the silk polypeptide that has a coiled coil structure comprises at least 18 copies of the heptad sequence abcdefg, and wherein at least 25% of the amino acids at positions a and d are alanine residues. 64. The recombinant host cell of claim 54 which is a bacterial cell. 65. A process for preparing a silk polypeptide comprising cultivating the recombinant host cell of claim 54, under conditions which allow expression of the polynucleotide encoding the polypeptide, and recovering the expressed polypeptide.
1,600
244
244
14,523,242
1,615
A method of making a personal care composition, such as dentifrice, using compacted rheology modifiers to control viscosity development.
1. A method of producing a personal care composition comprising: a) forming in a mix tank a premix having a lower viscosity than a packaged personal care composition; b) adding compacted rheology modifiers to the premix; c) transferring the premix from the mix tank; d) packaging the premix to produce a personal care composition. 2. The method of claim 1, wherein rheology modifier is compacted using a roller compactor to produce compacted rheology modifier. 3. The method of claim 2, wherein the force employed for compaction ranges from about 300 kilograms force/linear centimeter roll width to about 4,000 kilograms force/linear centimeter roll. 4. The method of claim 2, wherein the rheology modifier average particle size ranges from about 0.5 μm to about 1000 μm. 5. The method of claim 2, wherein the compacted rheology modifier is ground to produce compacted rheology modifier particles. 6. The method of claim 5, wherein the compacted rheology modifier particle size ranges from about 44 μm to about 420 μm. 7. The method of claim 5, wherein the compacted rheology modifier particles are coated. 8. The method of claim 7, wherein the compacted rheology modifier particles are coated with at least one of silica or acid salt. 9. The method of claim 1, wherein the compacted rheology modifier is at least one of xanthan gum, carboxymethyl cellulose, carrageenan, carbomer, hydroxyethyl cellulose, guar gum, or thickening silica. 10. The method of claim 1, wherein as the premix is transferred from the mix tank it has a 30 second rate of hydration of about 0.1 l/s to about 60 l/s. 11. The method of claim 1, wherein the premix is under atmospheric conditions in the mix tank at a temperature of about 15° C. to about 55° C. for about 0 to 30 minutes at a System Suspension Ratio (SSR) of about 1.5 or greater. 12. The method of claim 1, wherein the compacted rheology modifier is added in an amount from about 0.1% to about 15% by weight of the premix. 13. The method of claim 1 wherein, a surfactant is added to the premix. 14. A method of producing a personal care composition comprising: a) forming in a mix tank a premix having a lower viscosity than a packaged personal care composition; b) compacting a rheology modifier; c) adding compacted rheology modifiers to the premix; d) transferring the premix from the mix tank; and e) packaging the premix to produce a personal care composition. 15. The method of claim 14, wherein a roller compactor, compacts the rheology modifier. 16. The method of claim 14, wherein as the premix is transferred from the mix tank it has a 30 second rate of hydration of about 0.1 l/s to about 60 l/s. 17. The method of claim 14, wherein the compacted rheology modifier is ground to produce compacted rheology modifier particles. 18. The method of claim 17, wherein the compacted rheology modifier particles are coated. 19. The method of claim 18, wherein the compacted rheology modifier particles are coated with at least one of silica or acid salt. 20. The method of claim 14, wherein the rheology modifier is at least one of xanthan gum, carboxymethyl cellulose, carrageenan, carbomer, hydroxyethyl cellulose, guar gum, or thickening silica.
A method of making a personal care composition, such as dentifrice, using compacted rheology modifiers to control viscosity development.1. A method of producing a personal care composition comprising: a) forming in a mix tank a premix having a lower viscosity than a packaged personal care composition; b) adding compacted rheology modifiers to the premix; c) transferring the premix from the mix tank; d) packaging the premix to produce a personal care composition. 2. The method of claim 1, wherein rheology modifier is compacted using a roller compactor to produce compacted rheology modifier. 3. The method of claim 2, wherein the force employed for compaction ranges from about 300 kilograms force/linear centimeter roll width to about 4,000 kilograms force/linear centimeter roll. 4. The method of claim 2, wherein the rheology modifier average particle size ranges from about 0.5 μm to about 1000 μm. 5. The method of claim 2, wherein the compacted rheology modifier is ground to produce compacted rheology modifier particles. 6. The method of claim 5, wherein the compacted rheology modifier particle size ranges from about 44 μm to about 420 μm. 7. The method of claim 5, wherein the compacted rheology modifier particles are coated. 8. The method of claim 7, wherein the compacted rheology modifier particles are coated with at least one of silica or acid salt. 9. The method of claim 1, wherein the compacted rheology modifier is at least one of xanthan gum, carboxymethyl cellulose, carrageenan, carbomer, hydroxyethyl cellulose, guar gum, or thickening silica. 10. The method of claim 1, wherein as the premix is transferred from the mix tank it has a 30 second rate of hydration of about 0.1 l/s to about 60 l/s. 11. The method of claim 1, wherein the premix is under atmospheric conditions in the mix tank at a temperature of about 15° C. to about 55° C. for about 0 to 30 minutes at a System Suspension Ratio (SSR) of about 1.5 or greater. 12. The method of claim 1, wherein the compacted rheology modifier is added in an amount from about 0.1% to about 15% by weight of the premix. 13. The method of claim 1 wherein, a surfactant is added to the premix. 14. A method of producing a personal care composition comprising: a) forming in a mix tank a premix having a lower viscosity than a packaged personal care composition; b) compacting a rheology modifier; c) adding compacted rheology modifiers to the premix; d) transferring the premix from the mix tank; and e) packaging the premix to produce a personal care composition. 15. The method of claim 14, wherein a roller compactor, compacts the rheology modifier. 16. The method of claim 14, wherein as the premix is transferred from the mix tank it has a 30 second rate of hydration of about 0.1 l/s to about 60 l/s. 17. The method of claim 14, wherein the compacted rheology modifier is ground to produce compacted rheology modifier particles. 18. The method of claim 17, wherein the compacted rheology modifier particles are coated. 19. The method of claim 18, wherein the compacted rheology modifier particles are coated with at least one of silica or acid salt. 20. The method of claim 14, wherein the rheology modifier is at least one of xanthan gum, carboxymethyl cellulose, carrageenan, carbomer, hydroxyethyl cellulose, guar gum, or thickening silica.
1,600
245
245
12,813,564
1,612
A composition in the form of an emulsion containing an aqueous phase, a fatty phase, a superabsorbent polymer, and an organic UV screening agent. Method for the treatment of a keratinous substance whereby the composition described is applied to the keratinous substance.
1. A composition in the form of an emulsion comprising an aqueous phase, a fatty phase, a superabsorbent polymer, and an organic UV screening agent. 2. The composition according to claim 1, wherein the superabsorbent polymer is provided in the form of particles having a number-average diameter ranging from 10 μm to 1000 μm. 3. The composition according to claim 1, wherein the superabsorbent polymer is provided in the form of particles having a number-average diameter of less than or equal to 100 μm. 4. The composition according to claim 1, wherein the superabsorbent polymer exhibits a water-absorbing capacity of 10 to 100 g/g. 5. The composition according to claim 1, wherein the superabsorbent polymer is chosen from crosslinked sodium polyacrylates, starches grafted by an acrylic polymer, hydrolysed starches grafted by an acrylic polymer, polymers based on starch, on gum and on cellulose derivative, and their blends. 6. The composition according to claim 1, wherein the superabsorbent polymer is chosen from crosslinked acrylic homo- or copolymers which are optionally neutralized. 7. The composition according to claim 1, wherein the superabsorbent polymer is chosen from crosslinked sodium polyacrylates. 8. The composition according to claim 1, wherein the superabsorbent polymer is provided in the form of spherical particles. 9. The composition according to claim 1, wherein the superabsorbent polymer is present in a content of 0.03 to 15% by weight with respect to the total weight of the composition. 10. The composition according to claim 1, comprising a lipophilic organic UV screening agent. 11. The composition according to claim 1, wherein the organic UV screening agent is chosen from para-aminobenzoic acid derivatives, salicylic derivatives, cinnamic derivatives, benzophenones or aminobenzophenones, anthranilic derivatives, dibenzoylmethane derivatives, β,β-diphenylacrylate derivatives, benzylidenecamphor derivatives, phenylbenzimidazole derivatives, benzotriazole derivatives, triazine derivatives, bisresorcinyltriazines, imidazoline derivatives, benzalmalonate derivatives, 4,4-diarylbutadiene derivatives, benzoxazole derivatives, merocyanines, diphenylbutadienemalonate or -malonitrile derivatives, chalcones and their mixtures. 12. The composition according to claim 1, wherein the organic UV screening agent is chosen from: salicylic derivatives, cinnamic derivatives, β,β-diphenylacrylate derivatives, dibenzoylmethane derivatives, triazine derivatives, benzotriazole derivatives, and their mixtures. 13. The composition according to claim 1, comprising a hydrophilic organic UV screening agent. 14. The composition according to claim 1, wherein the total amount of organic UV screening agent(s) is 0.05 to 30% by weight with respect to the total weight of the composition. 15. A method for the treatment of a keratinous substance, comprising applying the composition of claim 1 to the keratinous substance.
A composition in the form of an emulsion containing an aqueous phase, a fatty phase, a superabsorbent polymer, and an organic UV screening agent. Method for the treatment of a keratinous substance whereby the composition described is applied to the keratinous substance.1. A composition in the form of an emulsion comprising an aqueous phase, a fatty phase, a superabsorbent polymer, and an organic UV screening agent. 2. The composition according to claim 1, wherein the superabsorbent polymer is provided in the form of particles having a number-average diameter ranging from 10 μm to 1000 μm. 3. The composition according to claim 1, wherein the superabsorbent polymer is provided in the form of particles having a number-average diameter of less than or equal to 100 μm. 4. The composition according to claim 1, wherein the superabsorbent polymer exhibits a water-absorbing capacity of 10 to 100 g/g. 5. The composition according to claim 1, wherein the superabsorbent polymer is chosen from crosslinked sodium polyacrylates, starches grafted by an acrylic polymer, hydrolysed starches grafted by an acrylic polymer, polymers based on starch, on gum and on cellulose derivative, and their blends. 6. The composition according to claim 1, wherein the superabsorbent polymer is chosen from crosslinked acrylic homo- or copolymers which are optionally neutralized. 7. The composition according to claim 1, wherein the superabsorbent polymer is chosen from crosslinked sodium polyacrylates. 8. The composition according to claim 1, wherein the superabsorbent polymer is provided in the form of spherical particles. 9. The composition according to claim 1, wherein the superabsorbent polymer is present in a content of 0.03 to 15% by weight with respect to the total weight of the composition. 10. The composition according to claim 1, comprising a lipophilic organic UV screening agent. 11. The composition according to claim 1, wherein the organic UV screening agent is chosen from para-aminobenzoic acid derivatives, salicylic derivatives, cinnamic derivatives, benzophenones or aminobenzophenones, anthranilic derivatives, dibenzoylmethane derivatives, β,β-diphenylacrylate derivatives, benzylidenecamphor derivatives, phenylbenzimidazole derivatives, benzotriazole derivatives, triazine derivatives, bisresorcinyltriazines, imidazoline derivatives, benzalmalonate derivatives, 4,4-diarylbutadiene derivatives, benzoxazole derivatives, merocyanines, diphenylbutadienemalonate or -malonitrile derivatives, chalcones and their mixtures. 12. The composition according to claim 1, wherein the organic UV screening agent is chosen from: salicylic derivatives, cinnamic derivatives, β,β-diphenylacrylate derivatives, dibenzoylmethane derivatives, triazine derivatives, benzotriazole derivatives, and their mixtures. 13. The composition according to claim 1, comprising a hydrophilic organic UV screening agent. 14. The composition according to claim 1, wherein the total amount of organic UV screening agent(s) is 0.05 to 30% by weight with respect to the total weight of the composition. 15. A method for the treatment of a keratinous substance, comprising applying the composition of claim 1 to the keratinous substance.
1,600
246
246
15,004,646
1,644
Methods of modulating the occurrence of lymphangiogenesis in a subject are provided. In some instances, the method includes reducing antigen presenting cell function, such as cell trafficking to draining lymph nodes. In some instances, the method includes treating corneal transplant rejection in the subject. Aspects of the methods include administering to the subject an effective amount of an antagonist of angiopoietin-2 (Ang-2) to reduce the occurrence of lymphangiogenesis. In some cases, the method results in the improvement of graft survival in the subject. Also provided are methods of reducing immune cell activity in a sample, e.g., reducing antigen presenting cell trafficking to draining lymph nodes. Also provided are compositions, e.g., ophthalmic pharmaceutical compositions and kits that find use in the subject methods.
1. A method of reducing the occurrence of lymphangiogenesis in a corneal transplant subject, the method comprising administering to the subject an amount of an Ang-2 antagonist effective to reduce the occurrence of lymphangiogenesis in the subject. 2. The method according to claim 1, wherein the subject has a high risk of corneal transplant rejection. 3. The method according to claim 2, wherein the subject has an inflamed and vascularized graft bed. 4. The method according to claim 1, wherein the subject has a low risk of corneal transplant rejection. 5. The method according to claim 4, wherein the subject has an un-inflamed and avascular graft bed 6. The method according to claim 1, wherein the Ang-2 antagonist comprises a specific binding member that specifically binds to Ang-2. 7. The method according to claim 6, wherein the Ang-2 antagonist is selected from a neutralizing antibody or binding fragment thereof, a scaffolded protein binder, a small molecule and a peptide. 8. The method according to claim 1, wherein the Ang-2 antagonist selectively reduces expression of Ang-2. 9. The method according to claim 8, wherein the Ang-2 antagonist is a RNAi molecule. 10. The method according to claim 1, wherein the subject is human. 11. The method according to claim 1, further comprising transplanting corneal tissue in a graft bed of the subject. 12. The method according to claim 11, wherein the administering is performed prior to the transplanting. 13. The method according to claim 11, wherein the administering is performed during the transplanting. 14. The method according to claim 11, wherein the administering is performed after the transplanting. 15. The method according to claim 11, wherein the method comprises reducing or preventing lymphangiogenesis in the graft bed. 16. The method according to claim 11, wherein the method comprises reducing lymphangiogenesis around the grafting border between the transplanted tissue and the graft bed. 17. The method according to claim 1, wherein the method does not significantly inhibit blood vessel growth in the tissue. 18. The method according to claim 1, wherein the method comprises suppressing antigen presenting cell trafficking to draining lymph nodes in immune reaction. 19. The method according to claim 11, wherein the method comprises enhancing survival of the transplanted corneal tissue in the subject. 20. A kit, comprising: an ophthalmic pharmaceutical composition comprising an antagonist of Ang-2; and one or more components selected from a syringe suitable for intra-ocular injection, an eye numbing agent, a sterile dilution buffer and a sealed package configured to maintain the sterility of the ophthalmic pharmaceutical composition.
Methods of modulating the occurrence of lymphangiogenesis in a subject are provided. In some instances, the method includes reducing antigen presenting cell function, such as cell trafficking to draining lymph nodes. In some instances, the method includes treating corneal transplant rejection in the subject. Aspects of the methods include administering to the subject an effective amount of an antagonist of angiopoietin-2 (Ang-2) to reduce the occurrence of lymphangiogenesis. In some cases, the method results in the improvement of graft survival in the subject. Also provided are methods of reducing immune cell activity in a sample, e.g., reducing antigen presenting cell trafficking to draining lymph nodes. Also provided are compositions, e.g., ophthalmic pharmaceutical compositions and kits that find use in the subject methods.1. A method of reducing the occurrence of lymphangiogenesis in a corneal transplant subject, the method comprising administering to the subject an amount of an Ang-2 antagonist effective to reduce the occurrence of lymphangiogenesis in the subject. 2. The method according to claim 1, wherein the subject has a high risk of corneal transplant rejection. 3. The method according to claim 2, wherein the subject has an inflamed and vascularized graft bed. 4. The method according to claim 1, wherein the subject has a low risk of corneal transplant rejection. 5. The method according to claim 4, wherein the subject has an un-inflamed and avascular graft bed 6. The method according to claim 1, wherein the Ang-2 antagonist comprises a specific binding member that specifically binds to Ang-2. 7. The method according to claim 6, wherein the Ang-2 antagonist is selected from a neutralizing antibody or binding fragment thereof, a scaffolded protein binder, a small molecule and a peptide. 8. The method according to claim 1, wherein the Ang-2 antagonist selectively reduces expression of Ang-2. 9. The method according to claim 8, wherein the Ang-2 antagonist is a RNAi molecule. 10. The method according to claim 1, wherein the subject is human. 11. The method according to claim 1, further comprising transplanting corneal tissue in a graft bed of the subject. 12. The method according to claim 11, wherein the administering is performed prior to the transplanting. 13. The method according to claim 11, wherein the administering is performed during the transplanting. 14. The method according to claim 11, wherein the administering is performed after the transplanting. 15. The method according to claim 11, wherein the method comprises reducing or preventing lymphangiogenesis in the graft bed. 16. The method according to claim 11, wherein the method comprises reducing lymphangiogenesis around the grafting border between the transplanted tissue and the graft bed. 17. The method according to claim 1, wherein the method does not significantly inhibit blood vessel growth in the tissue. 18. The method according to claim 1, wherein the method comprises suppressing antigen presenting cell trafficking to draining lymph nodes in immune reaction. 19. The method according to claim 11, wherein the method comprises enhancing survival of the transplanted corneal tissue in the subject. 20. A kit, comprising: an ophthalmic pharmaceutical composition comprising an antagonist of Ang-2; and one or more components selected from a syringe suitable for intra-ocular injection, an eye numbing agent, a sterile dilution buffer and a sealed package configured to maintain the sterility of the ophthalmic pharmaceutical composition.
1,600
247
247
14,586,105
1,619
Disclosed are oil-in-water (O/W) emulsions comprising (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion. The disclosure also relates to hair styling and/or shaping compositions comprising the O/W emulsions, methods of making the emulsions, and methods of styling and/or shaping the hair.
1. An oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion. 2. The oil-in-water emulsion of claim 1, wherein the oil phase comprises at least one water-insoluble component chosen from volatile oils, non-volatile oils, UV agents, and antioxidants. 3. The oil-in-water emulsion of claim 2, wherein the at least one surfactant has an HLB value approximately the same as the HLB value of the water-insoluble component. 4. The oil-in-water emulsion of claim 2, wherein the at least one water-insoluble component is present in the oil-in-water emulsion in an amount ranging from about 0.1% to about 50% by weight, relative to the total weight of the emulsion. 5. The oil-in-water emulsion of claim 3, wherein the at least one surfactant is present in the emulsion in an amount ranging up to about 5% by weight, relative to the total weight of the emulsion. 6. The oil-in-water emulsion of claim 3, wherein the at least one surfactant is chosen from non-ionic and anionic surfactants. 7. The oil-in-water emulsion of claim 1, wherein the aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex is present in the oil-in-water emulsion in an amount ranging from about 0.5% to about 10% by weight as polymeric active material (dry weight basis), relative to the total weight of the emulsion. 8. The oil-in-water emulsion of claim 1, wherein the emulsion is stable. 9. A hair styling or hair shaping composition comprising the oil-in-water emulsion of claim 1. 10. The composition of claim 9, further comprising at least one additional component chosen from solvents, wax dispersions, oils, rheology modifiers, thickening agents, structuring agents, vitamins, plant extracts, or propellants. 11. The composition of claim 10, comprising at least one solvent. 12. The composition of claim 11, comprising at least one wax dispersion, wherein said wax dispersion comprises particles of natural and/or synthetic waxes dispersed in a surfactant mixture comprising nonionic and/or ionic surfactants. 13. A method of making an oil-in-water emulsion, said oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion, wherein the method comprises: combining the (a) the aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) the oil phase, and (c) the at least one surfactant, optionally mixing the combined components, and homogenizing the combined components under either low-pressure or high-pressure homogenization. 14. The method of claim 13, wherein the oil-in-water emulsion is stable. 15. The method according to claim 13, comprising mixing (a) the aqueous dispersion of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) the at least one oil phase, and (c) the at least one surfactant, prior to said homogenization. 16. The method according to claim 11, wherein the homogenization process is repeated from 2 to 10 times. 17. A method of styling or shaping the hair, said method comprising applying an oil-in-water emulsion to the hair, said oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion. 18. A method of styling and/or shaping the hair, said method comprising applying to the hair a composition comprising: (1) an oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion; and (2) at least one additional component chosen from solvents, wax dispersions, oils, rheology modifiers, thickening agents, structuring agents, propellants, vitamins, or plant extracts. 19. The method according to claim 18, wherein the composition comprises at least one solvent. 20. The method of claim 19, wherein the composition further comprises at least one wax dispersion, wherein said wax dispersion comprises particles of natural and/or synthetic waxes dispersed in a surfactant mixture comprising nonionic and/or ionic surfactants.
Disclosed are oil-in-water (O/W) emulsions comprising (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion. The disclosure also relates to hair styling and/or shaping compositions comprising the O/W emulsions, methods of making the emulsions, and methods of styling and/or shaping the hair.1. An oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion. 2. The oil-in-water emulsion of claim 1, wherein the oil phase comprises at least one water-insoluble component chosen from volatile oils, non-volatile oils, UV agents, and antioxidants. 3. The oil-in-water emulsion of claim 2, wherein the at least one surfactant has an HLB value approximately the same as the HLB value of the water-insoluble component. 4. The oil-in-water emulsion of claim 2, wherein the at least one water-insoluble component is present in the oil-in-water emulsion in an amount ranging from about 0.1% to about 50% by weight, relative to the total weight of the emulsion. 5. The oil-in-water emulsion of claim 3, wherein the at least one surfactant is present in the emulsion in an amount ranging up to about 5% by weight, relative to the total weight of the emulsion. 6. The oil-in-water emulsion of claim 3, wherein the at least one surfactant is chosen from non-ionic and anionic surfactants. 7. The oil-in-water emulsion of claim 1, wherein the aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex is present in the oil-in-water emulsion in an amount ranging from about 0.5% to about 10% by weight as polymeric active material (dry weight basis), relative to the total weight of the emulsion. 8. The oil-in-water emulsion of claim 1, wherein the emulsion is stable. 9. A hair styling or hair shaping composition comprising the oil-in-water emulsion of claim 1. 10. The composition of claim 9, further comprising at least one additional component chosen from solvents, wax dispersions, oils, rheology modifiers, thickening agents, structuring agents, vitamins, plant extracts, or propellants. 11. The composition of claim 10, comprising at least one solvent. 12. The composition of claim 11, comprising at least one wax dispersion, wherein said wax dispersion comprises particles of natural and/or synthetic waxes dispersed in a surfactant mixture comprising nonionic and/or ionic surfactants. 13. A method of making an oil-in-water emulsion, said oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion, wherein the method comprises: combining the (a) the aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) the oil phase, and (c) the at least one surfactant, optionally mixing the combined components, and homogenizing the combined components under either low-pressure or high-pressure homogenization. 14. The method of claim 13, wherein the oil-in-water emulsion is stable. 15. The method according to claim 13, comprising mixing (a) the aqueous dispersion of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) the at least one oil phase, and (c) the at least one surfactant, prior to said homogenization. 16. The method according to claim 11, wherein the homogenization process is repeated from 2 to 10 times. 17. A method of styling or shaping the hair, said method comprising applying an oil-in-water emulsion to the hair, said oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion. 18. A method of styling and/or shaping the hair, said method comprising applying to the hair a composition comprising: (1) an oil-in-water emulsion comprising: (a) an aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, (b) an oil phase, and (c) at least one surfactant, wherein the amount of the (a) aqueous dispersion of particles of at least one latex chosen from an acrylate latex, a polyurethane latex, or a silicone latex, in combination with the (c) at least one surfactant is sufficient to stabilize the oil-in-water emulsion; and wherein the (c) at least one surfactant is present in the oil-in-water emulsion in an amount that is not, by itself, sufficient to stabilize the emulsion; and (2) at least one additional component chosen from solvents, wax dispersions, oils, rheology modifiers, thickening agents, structuring agents, propellants, vitamins, or plant extracts. 19. The method according to claim 18, wherein the composition comprises at least one solvent. 20. The method of claim 19, wherein the composition further comprises at least one wax dispersion, wherein said wax dispersion comprises particles of natural and/or synthetic waxes dispersed in a surfactant mixture comprising nonionic and/or ionic surfactants.
1,600
248
248
15,147,198
1,617
Disclosed are compositions and methods related to the use of kinase inhibitors in treating macular degeneration and/or retinal vein occlusion.
1. An injectable composition, comprising a poorly water-soluble kinase inhibitor dissolved in a water-miscible organic solvent, wherein: the concentration of the kinase inhibitor in the composition is at least 200 μg/mL; and the concentration of the kinase inhibitor in the composition is at least 100 times higher than the solubility of the kinase inhibitor in water at 37° C. 2. The injectable composition of claim 1, wherein the concentration of the kinase inhibitor in the composition is at least 800 μg/mL. 3. The injectable composition of claim 1, wherein the concentration of the kinase inhibitor in the composition is at least 200 times higher than the solubility of the kinase inhibitor in water at 37° C. 4. The injectable composition of claim 1, wherein the solubility of the kinase inhibitor in water is less than 10 μg/mL. 5. The injectable composition of claim 1, wherein the organic solvent is ethanol. 6. The injectable composition claim 1, wherein the kinase inhibitor is selected from bafetinib, bosutinib, dasatinib, imatinib, nilotinib, ponatinib, radotinib, and SU6656. 7. The injectable composition of claim 1, wherein the kinase inhibitor is selected from apatinib, axitinib, cabozantinib, cediranib, crenolanib, foretinib, lenvatinib, linifanib, masitinib, motesanib, nintedanib, pazopanib, pegaptanib, regorafenib, semaxanib, sorafenib, sunitinib, tivozanib, toceranib, vandetanib, and vatalanib. 8. The injectable composition of claim 7, wherein the kinase inhibitor is sorafenib. 9. The injectable composition of claim 8, wherein: the kinase inhibitor is sorafenib; and the concentration of the kinase inhibitor is about 800 μg/mL to about 4000 μg/mL. 10. The injectable composition of claim 1, wherein the composition is a liquid. 11-14. (canceled) 15. A syringe comprising the injectable composition of claim 1. 16-19. (canceled) 20. The syringe of claim 15, further comprising a filter. 21. The syringe of claim 20, wherein the filter comprises pores with a pore size of about 0.2 μm to about 5 μm. 22. (canceled) 23. The syringe of claim 20, further comprising a needle, wherein the needle is about 0.25 inches long to about 1.0 inches long. 24-26. (canceled) 27. The syringe of claim 23, wherein the needle is about 28 gauge to about 33 gauge. 28. The syringe of claim 27, wherein the needle is 29 gauge, 30 gauge, 31 gauge, or 32 gauge. 29. A kit comprising a vial comprising the injectable composition of claim 1 and a syringe. 30. A method for preventing or treating an eye disease in a subject, comprising injecting the composition of claim 1 into the vitreous of an eye of the subject. 31. The method of claim 30, comprising injecting about 10 μL to about 100 μL of the composition into the vitreous of the eye. 32. The method of claim 31, comprising injecting about 50 μL of the composition into the vitreous of the eye. 33. The method of claim 30, comprising filtering the composition prior to injecting the composition into the eye. 34. (canceled) 35. The method of claim 30, wherein the subject is selected from rodents, lagomorphs, ovines, porcines, canines, felines, equines, bovines, and primates. 36. The method of claim 35, wherein the subject is a human. 37. The method of claim 30, wherein: the subject has age-related macular degeneration, dry macular degeneration, wet macular degeneration, geographic atrophy, vision loss, non-ischemic retinal vein occlusion, or ischemic retinal vein occlusion; or the subject is at risk of developing wet age-related macular degeneration, geographic atrophy, or vision loss. 38-47. (canceled)
Disclosed are compositions and methods related to the use of kinase inhibitors in treating macular degeneration and/or retinal vein occlusion.1. An injectable composition, comprising a poorly water-soluble kinase inhibitor dissolved in a water-miscible organic solvent, wherein: the concentration of the kinase inhibitor in the composition is at least 200 μg/mL; and the concentration of the kinase inhibitor in the composition is at least 100 times higher than the solubility of the kinase inhibitor in water at 37° C. 2. The injectable composition of claim 1, wherein the concentration of the kinase inhibitor in the composition is at least 800 μg/mL. 3. The injectable composition of claim 1, wherein the concentration of the kinase inhibitor in the composition is at least 200 times higher than the solubility of the kinase inhibitor in water at 37° C. 4. The injectable composition of claim 1, wherein the solubility of the kinase inhibitor in water is less than 10 μg/mL. 5. The injectable composition of claim 1, wherein the organic solvent is ethanol. 6. The injectable composition claim 1, wherein the kinase inhibitor is selected from bafetinib, bosutinib, dasatinib, imatinib, nilotinib, ponatinib, radotinib, and SU6656. 7. The injectable composition of claim 1, wherein the kinase inhibitor is selected from apatinib, axitinib, cabozantinib, cediranib, crenolanib, foretinib, lenvatinib, linifanib, masitinib, motesanib, nintedanib, pazopanib, pegaptanib, regorafenib, semaxanib, sorafenib, sunitinib, tivozanib, toceranib, vandetanib, and vatalanib. 8. The injectable composition of claim 7, wherein the kinase inhibitor is sorafenib. 9. The injectable composition of claim 8, wherein: the kinase inhibitor is sorafenib; and the concentration of the kinase inhibitor is about 800 μg/mL to about 4000 μg/mL. 10. The injectable composition of claim 1, wherein the composition is a liquid. 11-14. (canceled) 15. A syringe comprising the injectable composition of claim 1. 16-19. (canceled) 20. The syringe of claim 15, further comprising a filter. 21. The syringe of claim 20, wherein the filter comprises pores with a pore size of about 0.2 μm to about 5 μm. 22. (canceled) 23. The syringe of claim 20, further comprising a needle, wherein the needle is about 0.25 inches long to about 1.0 inches long. 24-26. (canceled) 27. The syringe of claim 23, wherein the needle is about 28 gauge to about 33 gauge. 28. The syringe of claim 27, wherein the needle is 29 gauge, 30 gauge, 31 gauge, or 32 gauge. 29. A kit comprising a vial comprising the injectable composition of claim 1 and a syringe. 30. A method for preventing or treating an eye disease in a subject, comprising injecting the composition of claim 1 into the vitreous of an eye of the subject. 31. The method of claim 30, comprising injecting about 10 μL to about 100 μL of the composition into the vitreous of the eye. 32. The method of claim 31, comprising injecting about 50 μL of the composition into the vitreous of the eye. 33. The method of claim 30, comprising filtering the composition prior to injecting the composition into the eye. 34. (canceled) 35. The method of claim 30, wherein the subject is selected from rodents, lagomorphs, ovines, porcines, canines, felines, equines, bovines, and primates. 36. The method of claim 35, wherein the subject is a human. 37. The method of claim 30, wherein: the subject has age-related macular degeneration, dry macular degeneration, wet macular degeneration, geographic atrophy, vision loss, non-ischemic retinal vein occlusion, or ischemic retinal vein occlusion; or the subject is at risk of developing wet age-related macular degeneration, geographic atrophy, or vision loss. 38-47. (canceled)
1,600
249
249
13,515,853
1,641
The invention relates to a substance determining apparatus and method for determining a substance within a fluid. Particles attach to the substance and bind to a binding surface ( 30 ), wherein a particle release curve being indicative of a release of bound particles from the binding surface ( 30 ) is determined, and a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding is determined based on the temporal behaviour of the particle release curve. The substance within the fluid is determined based on the part of the particle release curve. The substance can therefore be determined based on particles which are bound to the binding surface via a certain kind of binding, i.e. other kinds of binding substantially do not affect the determination of the substance, thereby improving the accuracy of determining the substance.
1. A substance determining apparatus for determining a substance within a fluid (3), the substance determining apparatus (19) comprising: particles (17) for being attached to the substance within the fluid (3), a binding surface (30) for binding the particles (17), if the particles (17) have been attached to the substance, wherein the particles are bindable to the binding surface (30) with different kinds of binding, a sensing unit (33) for sensing the particles (17) on the binding surface (30), wherein the sensing unit (33) is adapted to generate a temporal sensing signal depending on the bound particles, a particle release curve determination unit (42) for determining a particle release curve being indicative of a release of bound particles from the binding surface (30) depending on the generated temporal sensing signal, a binding determination unit (40) for determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, a substance determination unit (41) for determining the substance within the fluid based on the determined part of the particle release curve. 2. The substance determining apparatus as defined in claim 1, wherein the sensing unit (33) is adapted to generate a temporal sensing signal dependent on the bound particles integrated over a predefined region of the binding surface (30), wherein the particle release curve determination unit (42) is adapted to determine the generated temporal sensing signal as the particle release curve. 3. The substance determining apparatus as defined in claim 1, wherein the particle release curve determination unit (42) is adapted to determine lifetimes of the bindings of the particles on the binding surface (30) from the generated sensing signal, generate a histogram of the determined lifetimes, and determine the particle release curve depending on the generated histogram. 4. The substance determining apparatus as defined in claim 1, wherein the sensing unit (33) comprises a force applying unit (23, 24) for applying a force to the particles for putting bindings between the particles and the binding surface (30) under stress, while sensing the particles. 5. The substance determining apparatus as defined in claim 1, wherein the binding determination unit (40) is adapted to provide a first predefined fitting release curve having a first temporal behaviour being indicative of a first kind of binding and a second predefined fitting release curve having a second temporal behaviour being indicative of a second kind of binding, fit the first predefined fitting release curve and the second predefined fitting release curve to the determined particle release curve by fitting a linear combination of the first predefined fitting release curve and the second predefined fitting release curve to the determined particle release curve, determine one of the fitted first predefined release curve and the fitted second predefined release curve as the part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding. 6. The substance determining apparatus as defined in claim 1, wherein the binding determination unit (40) is adapted to determine time constants of the particle release curve as the temporal behaviour of the particle release curve, wherein the time constants are defined by reaction constants of the particles bound to the binding surface (30), and wherein the binding determination unit (40) is adapted to determine a part of the particle release curve, which changes with at least one of the determined time constants, as the part of the particle release curve caused by particles bound to the binding surface (30) via the predefined kind of binding. 7. The substance determining apparatus as defined in claim 6, wherein the sensing unit (33) comprises a force applying unit (23, 24) for applying a force to the particles for putting bindings between the particles and the binding surface (30) under stress, while sensing the particles, wherein the binding determination unit (40) is adapted to correct the time constants for the influence of the applied force based on the applied force and to determine a part of the particle release curve, which changes with at least one of the corrected time constants, as the part of the particle release curve caused by particles bound to the binding surface (30) via the predefined kind of binding. 8. The substance determining apparatus as defined in claim 6, wherein the particles are magnetic particles with magnetic properties, which magnetically influence each other, thereby influencing the time constants, wherein the sensing unit (33) is adapted to generate a temporal position signal being indicative of the positions of the particles, wherein the binding determination unit (40) is adapted to: determine the positions of the particles for different times from the generated temporal position signal, determine the magnetic particle-particle influence based on the determined positions and magnetic properties of the particles at the different times, correct the time constants depending on the determined magnetic particle-particle influence at the different times. determine a part of the particle release curve, which changes with at least one of the corrected time constants, as the part of the particle release curve caused by particles bound to the binding surface (30) via the predefined kind of binding. 9. The substance determining apparatus as defined in claim 8, wherein the sensing unit (33) comprises a light source (20) for generating radiation (28) for being directed to the binding surface (30) for generating an evanescent field and a light detector (21, 27) for detecting light (29, 31) from a detection plane (56) being parallel to the binding surface (30) or defined by the binding surface (30), wherein the detected light has been influenced by the particles in the detection plane (56) by influencing the evanescent field, wherein the position signal is generated depending on the detected influenced light and indicative of the positions of the particles within the detection plane (56). 10. The substance determining apparatus as defined in claim 9, wherein the light detector (21, 27) is adapted to move the detection plane (56) with respect to the binding surface (30) for generating the position signal indicative of height positions of the particles with respect to the binding surface (30). 11. A binding device for cooperating with an analyzing device for determining a substance within a fluid, the binding device (1) comprising particles (17) for being attached to the substance within the fluid (3), a binding surface (30) for binding the particles (17), if the particles (17) have been attached to the substance, wherein the particles are bindable to the binding surface with different kinds of binding, the analyzing device comprising: a sensing unit (33) for sensing the particles (17) on the binding surface (30), wherein the sensing unit is adapted to generate a temporal sensing signal depending on the bound particles, a particle release curve determination unit (42) for determining a particle release curve being indicative of a release of bound particles from the binding surface (30) depending on the generated temporal sensing signal, a binding determination unit (40) for determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, a substance determination unit (41) for determining the substance within the fluid based on the determined part of the particle release curve. 12. An analyzing device for cooperating with a binding device for determining a substance within a fluid, the binding device comprising: particles (17) for being attached to the substance within the fluid (3), a binding surface (30) for binding the particles (17), if the particles (17) have been attached to the substance, wherein the particles are bindable to the binding surface with different kinds of binding, the analyzing device comprising: a sensing unit (33) for sensing the particles (17) on the binding surface (30), wherein the sensing unit (33) is adapted to generate a temporal sensing signal depending on the bound particles, a particle release curve determination unit (42) for determining a particle release curve being indicative of a release of bound particles from the binding surface (30) depending on the generated temporal sensing signal, a binding determination unit (40) for determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, a substance determination unit (41) for determining the substance within the fluid based on the determined part of the particle release curve. 13. A substance determining method for determining a substance within a fluid, the substance determining method comprising: attaching particles (17) to the substance within the fluid, binding the particles to a binding surface (30), if the particles have been attached to the substance, wherein the particles are bound to the binding surface with different kinds of binding, sensing the particles (17) on the binding surface (30), wherein a temporal sensing signal is generated depending on the bound particles, determining a particle release curve being indicative of a release of bound particles from the binding surface depending on the generated temporal sensing signal, determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, determining the substance within the fluid based on the determined part of the particle release curve. 14. An analyzing method for cooperating with a binding method for determining a substance within a fluid, the binding method comprising: attaching particles (17) to the substance within the fluid, binding the particles to a binding surface (30), if the particles have been attached to the substance, wherein the particles are bound to the binding surface (30) with different kinds of binding, the analyzing method comprising: sensing the particles (17) on the binding surface (30), wherein a temporal sensing signal is generated depending on the bound particles, determining a particle release curve being indicative of a release of bound particles from the binding surface depending on the generated temporal sensing signal, determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, determining the substance within the fluid based on the determined part of the particle release curve. 15. An analyzing computer program for determining a substance within a fluid, the computer program comprising program code means for causing a substance determining apparatus as defined in claim 1 when the computer program is run on a computer controlling the analyzing device.
The invention relates to a substance determining apparatus and method for determining a substance within a fluid. Particles attach to the substance and bind to a binding surface ( 30 ), wherein a particle release curve being indicative of a release of bound particles from the binding surface ( 30 ) is determined, and a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding is determined based on the temporal behaviour of the particle release curve. The substance within the fluid is determined based on the part of the particle release curve. The substance can therefore be determined based on particles which are bound to the binding surface via a certain kind of binding, i.e. other kinds of binding substantially do not affect the determination of the substance, thereby improving the accuracy of determining the substance.1. A substance determining apparatus for determining a substance within a fluid (3), the substance determining apparatus (19) comprising: particles (17) for being attached to the substance within the fluid (3), a binding surface (30) for binding the particles (17), if the particles (17) have been attached to the substance, wherein the particles are bindable to the binding surface (30) with different kinds of binding, a sensing unit (33) for sensing the particles (17) on the binding surface (30), wherein the sensing unit (33) is adapted to generate a temporal sensing signal depending on the bound particles, a particle release curve determination unit (42) for determining a particle release curve being indicative of a release of bound particles from the binding surface (30) depending on the generated temporal sensing signal, a binding determination unit (40) for determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, a substance determination unit (41) for determining the substance within the fluid based on the determined part of the particle release curve. 2. The substance determining apparatus as defined in claim 1, wherein the sensing unit (33) is adapted to generate a temporal sensing signal dependent on the bound particles integrated over a predefined region of the binding surface (30), wherein the particle release curve determination unit (42) is adapted to determine the generated temporal sensing signal as the particle release curve. 3. The substance determining apparatus as defined in claim 1, wherein the particle release curve determination unit (42) is adapted to determine lifetimes of the bindings of the particles on the binding surface (30) from the generated sensing signal, generate a histogram of the determined lifetimes, and determine the particle release curve depending on the generated histogram. 4. The substance determining apparatus as defined in claim 1, wherein the sensing unit (33) comprises a force applying unit (23, 24) for applying a force to the particles for putting bindings between the particles and the binding surface (30) under stress, while sensing the particles. 5. The substance determining apparatus as defined in claim 1, wherein the binding determination unit (40) is adapted to provide a first predefined fitting release curve having a first temporal behaviour being indicative of a first kind of binding and a second predefined fitting release curve having a second temporal behaviour being indicative of a second kind of binding, fit the first predefined fitting release curve and the second predefined fitting release curve to the determined particle release curve by fitting a linear combination of the first predefined fitting release curve and the second predefined fitting release curve to the determined particle release curve, determine one of the fitted first predefined release curve and the fitted second predefined release curve as the part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding. 6. The substance determining apparatus as defined in claim 1, wherein the binding determination unit (40) is adapted to determine time constants of the particle release curve as the temporal behaviour of the particle release curve, wherein the time constants are defined by reaction constants of the particles bound to the binding surface (30), and wherein the binding determination unit (40) is adapted to determine a part of the particle release curve, which changes with at least one of the determined time constants, as the part of the particle release curve caused by particles bound to the binding surface (30) via the predefined kind of binding. 7. The substance determining apparatus as defined in claim 6, wherein the sensing unit (33) comprises a force applying unit (23, 24) for applying a force to the particles for putting bindings between the particles and the binding surface (30) under stress, while sensing the particles, wherein the binding determination unit (40) is adapted to correct the time constants for the influence of the applied force based on the applied force and to determine a part of the particle release curve, which changes with at least one of the corrected time constants, as the part of the particle release curve caused by particles bound to the binding surface (30) via the predefined kind of binding. 8. The substance determining apparatus as defined in claim 6, wherein the particles are magnetic particles with magnetic properties, which magnetically influence each other, thereby influencing the time constants, wherein the sensing unit (33) is adapted to generate a temporal position signal being indicative of the positions of the particles, wherein the binding determination unit (40) is adapted to: determine the positions of the particles for different times from the generated temporal position signal, determine the magnetic particle-particle influence based on the determined positions and magnetic properties of the particles at the different times, correct the time constants depending on the determined magnetic particle-particle influence at the different times. determine a part of the particle release curve, which changes with at least one of the corrected time constants, as the part of the particle release curve caused by particles bound to the binding surface (30) via the predefined kind of binding. 9. The substance determining apparatus as defined in claim 8, wherein the sensing unit (33) comprises a light source (20) for generating radiation (28) for being directed to the binding surface (30) for generating an evanescent field and a light detector (21, 27) for detecting light (29, 31) from a detection plane (56) being parallel to the binding surface (30) or defined by the binding surface (30), wherein the detected light has been influenced by the particles in the detection plane (56) by influencing the evanescent field, wherein the position signal is generated depending on the detected influenced light and indicative of the positions of the particles within the detection plane (56). 10. The substance determining apparatus as defined in claim 9, wherein the light detector (21, 27) is adapted to move the detection plane (56) with respect to the binding surface (30) for generating the position signal indicative of height positions of the particles with respect to the binding surface (30). 11. A binding device for cooperating with an analyzing device for determining a substance within a fluid, the binding device (1) comprising particles (17) for being attached to the substance within the fluid (3), a binding surface (30) for binding the particles (17), if the particles (17) have been attached to the substance, wherein the particles are bindable to the binding surface with different kinds of binding, the analyzing device comprising: a sensing unit (33) for sensing the particles (17) on the binding surface (30), wherein the sensing unit is adapted to generate a temporal sensing signal depending on the bound particles, a particle release curve determination unit (42) for determining a particle release curve being indicative of a release of bound particles from the binding surface (30) depending on the generated temporal sensing signal, a binding determination unit (40) for determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, a substance determination unit (41) for determining the substance within the fluid based on the determined part of the particle release curve. 12. An analyzing device for cooperating with a binding device for determining a substance within a fluid, the binding device comprising: particles (17) for being attached to the substance within the fluid (3), a binding surface (30) for binding the particles (17), if the particles (17) have been attached to the substance, wherein the particles are bindable to the binding surface with different kinds of binding, the analyzing device comprising: a sensing unit (33) for sensing the particles (17) on the binding surface (30), wherein the sensing unit (33) is adapted to generate a temporal sensing signal depending on the bound particles, a particle release curve determination unit (42) for determining a particle release curve being indicative of a release of bound particles from the binding surface (30) depending on the generated temporal sensing signal, a binding determination unit (40) for determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, a substance determination unit (41) for determining the substance within the fluid based on the determined part of the particle release curve. 13. A substance determining method for determining a substance within a fluid, the substance determining method comprising: attaching particles (17) to the substance within the fluid, binding the particles to a binding surface (30), if the particles have been attached to the substance, wherein the particles are bound to the binding surface with different kinds of binding, sensing the particles (17) on the binding surface (30), wherein a temporal sensing signal is generated depending on the bound particles, determining a particle release curve being indicative of a release of bound particles from the binding surface depending on the generated temporal sensing signal, determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, determining the substance within the fluid based on the determined part of the particle release curve. 14. An analyzing method for cooperating with a binding method for determining a substance within a fluid, the binding method comprising: attaching particles (17) to the substance within the fluid, binding the particles to a binding surface (30), if the particles have been attached to the substance, wherein the particles are bound to the binding surface (30) with different kinds of binding, the analyzing method comprising: sensing the particles (17) on the binding surface (30), wherein a temporal sensing signal is generated depending on the bound particles, determining a particle release curve being indicative of a release of bound particles from the binding surface depending on the generated temporal sensing signal, determining a part of the particle release curve caused by particles bound to the binding surface via a predefined kind of binding based on a temporal behaviour of the particle release curve, determining the substance within the fluid based on the determined part of the particle release curve. 15. An analyzing computer program for determining a substance within a fluid, the computer program comprising program code means for causing a substance determining apparatus as defined in claim 1 when the computer program is run on a computer controlling the analyzing device.
1,600
250
250
14,960,960
1,612
Disclosed herein are methods of promoting the aggregation and/or immune clearance of oral bacteria using oral care compositions comprising metal-amino acid complexes.
1. A method of promoting the aggregation and/or immune clearance of oral bacteria, the method comprising the step of administering to the oral cavity of a person an oral care composition comprising a metal-amino acid complex; wherein said person has demonstrated one or more of A. viscous, L. casei, S. oralis, V. parvula and F. nucleatum present in their oral cavities. 2. The method of claim 1, wherein the metal-amino acid complex is a zinc(II)-amino acid complex or a copper(II)-amino acid complex. 3. The method of claim 1, wherein the amino acid is a basic amino acid. 4. The method of claim 1, wherein the amino acid is selected from lysine, glycine, trimethylglycine and arginine. 5. The method of claim 1, wherein the metal-amino acid complex further comprises a halide. 6. The method of claim 1, wherein the complex is a zinc-lysine-chloride complex or a zinc-arginine chloride complex. 7. The method of claim 1, wherein the complex is a zinc lysine chloride complex having the chemical structure [Zn(C6H14N2O2)2Cl]+Cl−, either in solution of the cationic cation and the chloride anion, or in solid salt form, optionally in mono- or dihydrate form. 8. The method of claim 6, wherein the amount of zinc is 0.05-4% by weight. 9. The method of claim 1, wherein the composition is in the form of a toothpaste, gel, mouthwash, powder, cream, strip, or gum. 10. The method of claim 1, wherein the method is effective in treating diseases, disorders or conditions of the oral cavity or in disrupting the formation of plaque and bacterial biofilm. 11. A method of treating a disease, disorder, or condition of the oral cavity, comprising the step of administering to a patient in need thereof, an oral care composition comprising a metal-amino acid complex, to promote the aggregation and/or immune clearance of oral bacteria; wherein said patient has demonstrated one or more of A. viscous, L. casei, S. oralis, V. parvula and F. nucleatum present in their oral cavities. 12. The method of claim 11, wherein the metal-amino acid complex is a zinc(II)-amino acid complex or a copper(II)-amino acid complex. 13. The method of claim 11, wherein the amino acid is a basic amino acid. 14. The method of claim 11, wherein the amino acid is selected from lysine, glycine, trimethylglycine and arginine. 15. The method of claim 11, wherein the metal-amino acid complex further comprises a halide. 16. The method of claim 11, wherein the complex is a zinc-lysine-chloride complex or a zinc-arginine chloride complex. 17. The method of claim 11, wherein the complex is a zinc lysine chloride complex having the chemical structure [Zn(C6H14N2O2)2Cl]+Cl−, either in solution of the cationic cation and the chloride anion, or in solid salt form, optionally in mono- or dihydrate form. 18. The method of claim 17, wherein the amount of zinc is 0.05-4% by weight. 19. The method of claim 11, wherein the composition is in the form of a toothpaste, gel, mouthwash, powder, cream, strip, or gum. 20. The method of claim 11, wherein the disease, disorder or conditions of the oral cavity is selected from gingivitis, periodontitis, halitosis, cavity formation, enamel erosion, or an oral infection. 21. The method of claim 1, wherein said person is recovering from a treatment which predisposes the person to an oral bacterial infection.
Disclosed herein are methods of promoting the aggregation and/or immune clearance of oral bacteria using oral care compositions comprising metal-amino acid complexes.1. A method of promoting the aggregation and/or immune clearance of oral bacteria, the method comprising the step of administering to the oral cavity of a person an oral care composition comprising a metal-amino acid complex; wherein said person has demonstrated one or more of A. viscous, L. casei, S. oralis, V. parvula and F. nucleatum present in their oral cavities. 2. The method of claim 1, wherein the metal-amino acid complex is a zinc(II)-amino acid complex or a copper(II)-amino acid complex. 3. The method of claim 1, wherein the amino acid is a basic amino acid. 4. The method of claim 1, wherein the amino acid is selected from lysine, glycine, trimethylglycine and arginine. 5. The method of claim 1, wherein the metal-amino acid complex further comprises a halide. 6. The method of claim 1, wherein the complex is a zinc-lysine-chloride complex or a zinc-arginine chloride complex. 7. The method of claim 1, wherein the complex is a zinc lysine chloride complex having the chemical structure [Zn(C6H14N2O2)2Cl]+Cl−, either in solution of the cationic cation and the chloride anion, or in solid salt form, optionally in mono- or dihydrate form. 8. The method of claim 6, wherein the amount of zinc is 0.05-4% by weight. 9. The method of claim 1, wherein the composition is in the form of a toothpaste, gel, mouthwash, powder, cream, strip, or gum. 10. The method of claim 1, wherein the method is effective in treating diseases, disorders or conditions of the oral cavity or in disrupting the formation of plaque and bacterial biofilm. 11. A method of treating a disease, disorder, or condition of the oral cavity, comprising the step of administering to a patient in need thereof, an oral care composition comprising a metal-amino acid complex, to promote the aggregation and/or immune clearance of oral bacteria; wherein said patient has demonstrated one or more of A. viscous, L. casei, S. oralis, V. parvula and F. nucleatum present in their oral cavities. 12. The method of claim 11, wherein the metal-amino acid complex is a zinc(II)-amino acid complex or a copper(II)-amino acid complex. 13. The method of claim 11, wherein the amino acid is a basic amino acid. 14. The method of claim 11, wherein the amino acid is selected from lysine, glycine, trimethylglycine and arginine. 15. The method of claim 11, wherein the metal-amino acid complex further comprises a halide. 16. The method of claim 11, wherein the complex is a zinc-lysine-chloride complex or a zinc-arginine chloride complex. 17. The method of claim 11, wherein the complex is a zinc lysine chloride complex having the chemical structure [Zn(C6H14N2O2)2Cl]+Cl−, either in solution of the cationic cation and the chloride anion, or in solid salt form, optionally in mono- or dihydrate form. 18. The method of claim 17, wherein the amount of zinc is 0.05-4% by weight. 19. The method of claim 11, wherein the composition is in the form of a toothpaste, gel, mouthwash, powder, cream, strip, or gum. 20. The method of claim 11, wherein the disease, disorder or conditions of the oral cavity is selected from gingivitis, periodontitis, halitosis, cavity formation, enamel erosion, or an oral infection. 21. The method of claim 1, wherein said person is recovering from a treatment which predisposes the person to an oral bacterial infection.
1,600
251
251
14,401,791
1,634
The present disclosure describes methods, devices, reagents, and kits for the detection of one or more target molecules that may be present in a test sample. The described methods, devices, kits, and reagents facilitate the detection and quantification of a non-nucleic acid target (e.g., a protein target) in a test sample by detecting and quantifying a nucleic acid (i.e., an aptamer) where the aptamer-aptamer interactions are significantly reduced or eliminated while maintaining the aptamer-target interaction.
1-73. (canceled) 74. A method comprising: exposing an aptamer to a first solid support, wherein the aptamer comprises a first tag and the first solid support comprises a first capture element, and wherein the first tag has affinity for the first capture element; allowing the first tag to associate with the first capture element; washing the first solid support with one or more solutions that dissociate aggregated aptamers; contacting the aptamer with a test sample, wherein an aptamer-target affinity complex is formed if the target molecule is present in the test sample; removing one or more components not associated with the first solid support; attaching a second tag to the target molecule in the aptamer-target affinity complex, wherein the second tag has an affinity to a second capture element; releasing the aptamer-target affinity complex from said first solid support; exposing the released aptamer-target affinity complex to a second solid support comprising a second capture element and allowing the second tag to associate with said second capture element; removing one or more components not associated with the second solid support; and eluting the aptamer from the second solid support with one or more buffered solutions comprising a chaotropic salt. 75. The method of claim 74 further comprising the step of detecting the aptamer portion of said aptamer-target affinity complex. 76. The method of claim 75 further comprising quantifying the aptamer. 77. The method of claim 75 further comprising detecting the aptamer by hybridizing the aptamer to a third solid support, wherein the third solid support comprises a plurality of addressable features and wherein at least one of said features comprises at least capture element disposed thereon that is complementary to any sequence contained within the aptamer. 78. The method of claim 75, wherein the aptamer is detected and optionally quantified using a method selected from the group consisting of Q-PCR, MS, next-generation sequencing and hybridization. 79. The method of claim 78, wherein said Q-PCR is performed using TaqMan® PCR, an intercalating fluorescent dye during the PCR process, or a molecular beacon during the PCR process. 80. The method of claim 74, wherein the pH of the one or more solutions is about 11. 81. The method of claim 74, wherein the pH of the one or more buffered solutions is neutral. 82. The method of claim 74, wherein the chaotropic salt disrupts aptamer-target interactions. 83. The method of claim 74, wherein said chaotropic salt is selected from the group consisting of sodium perchlorate, lithium chloride, magnesium chloride and sodium chloride. 84. The method of claim 74, wherein the one or more of the buffered solutions comprises an organic solvent. 85. The method of claim 84, wherein the organic solvent is glycerol. 86. The method of claim 74, wherein the aptamer is a single-stranded nucleic acid or a double-stranded nucleic acid. 87. The method of claim 74, wherein the aptamer comprises DNA, RNA or both DNA and RNA. 88. The method of claim 74, wherein the aptamer-target affinity complex has a slow rate of dissociation. 89. The method of claim 74, wherein the rate of dissociation of the aptamer-target affinity complex (t1/2) is selected from the group consisting of >30 minutes, >60 minutes, >90 minutes, >120 minutes, >150 minutes, >180 minutes, >210 minutes, and >240 minutes. 90. The method of 74, wherein the aptamer comprises a detectable moiety is selected from the group consisting of a dye, a quantum dot, a radiolabel, an electrochemical functional group, and an enzyme plus a detectable enzyme substrate. 91. The method of claim 90, wherein the dye is a fluorescent dye. 92. The method of claim 74, wherein the aptamer comprises at least one C-5 modified nucleotide. 93. The method of claim 74, wherein the aptamer comprises at least one chemical modification comprising a chemical substitution at one or more positions independently selected from a ribose position, a deoxyribose position, a phosphate position, and a base position. 94. The method of claim 93, wherein the chemical modification is independently selected from the group consisting of a 2′-position sugar modification, a 2′-amino (2′-NH2), a 2′-fluoro (2′-F), a 2′-0-methyl (2′-OMe), a 5-position pyrimidine modification, an 8-position purine modification, a modification at a cytosine exocyclic amine, a substitution of 5-bromouracil, a substitution of 5-bromodeoxyuridine, a substitution of 5-bromodeoxycytidine, a backbone modification, methylation, a 3′ cap, and a 5′ cap. 95. The method of claim 74, wherein said target molecule is selected from the group consisting of a protein, a peptide, a carbohydrate, a polysaccharide, a glycoprotein, a hormone, a receptor, an antigen, an antibody, a virus, a substrate, a metabolite, a transition state analog, a cofactor, an inhibitor, a drug, a dye, a nutrient, a growth factor, a tissue, and a controlled substance. 96. The method claim 74, wherein the test sample is selected from the group consisting of blood, whole blood, leukocytes, peripheral blood mononuclear cells, plasma, serum, sputum, breath, urine, semen, saliva, meningeal fluid, amniotic fluid, glandular fluid, lymph fluid, nipple aspirate, bronchial aspirate, synovial fluid, joint aspirate, cells, a cellular extract, stool, tissue, a tissue extract, a tissue biopsy, and cerebrospinal fluid. 97. The method of claim 74, wherein the first tag and the second tag each comprises at least one component independently selected from the group consisting of a polynucleotide, a polypeptide, a peptide nucleic acid, a locked nucleic acid, an oligosaccharide, a polysaccharide, an antibody, an affibody, an antibody mimic, a cell receptor, a ligand, a lipid, biotin, avidin, streptavidin, Extravidin, neutravidin, Traptavidin, a metal, histidine, and any portion of any of these structures. 98. The method of claim 74, wherein said first capture element and said second capture element each comprises at least one component independently selected from a polynucleotide, a polypeptide, a peptide nucleic acid, a locked nucleic acid, an oligosaccharide, a polysaccharide, an antibody, an affibody, an antibody mimic, a cell receptor, a ligand, a lipid, biotin, avidin, streptavidin, Extravidin, neutravidin, Traptavidin, a metal, histidine, and any portion of any of these structures. 99. The method of claim 74, wherein the first tag comprises a releasable moiety. 100. The method of claim 99, wherein the releasable moiety comprises a photocleavable moiety. 101. The method of claim 74, wherein said first solid support and second solid support each is independently selected from the group consisting of a polymer bead, an agarose bead, a polystyrene bead, an acrylamide bead, a solid core bead, a porous bead, a paramagnetic bead, glass bead, controlled pore bead, a microtitre well, a cyclo-olefin copolymer substrate, a membrane, a plastic substrate, nylon, a Langmuir-Blodgett film, glass, a germanium substrate, a silicon substrate, a silicon wafer chip, a flow through chip, a microbead, a nanoparticle, a polytetrafluoroethylene substrate, a polystyrene substrate, a gallium arsenide substrate, a gold substrate, and a silver substrate. 102. A kit comprising: a) one or more aptamers, wherein each of the one or more aptamers has specific affinity for one or more targets; b) one or more solid supports; c) one or more partitioning reagents; d) one or more reagents for the release of an aptamer from an aptamer-target affinity complex; e) one or more buffer solutions comprising an organic solvent; and f) one or more buffer solutions comprising a chaotropic salt. 103. The kit of claim 102, wherein said organic solvent is glycerol. 104. The kit of claim 102, wherein said chaotropic salt is sodium perchlorate. 105. The kit of claim 102 further comprising a reagent to cleave a cleavable moiety of the one or more aptamers. 106. A method comprising: contacting an aptamer with a test sample, wherein an aptamer-target affinity complex is formed if the target molecule is present in the test sample, and wherein the aptamer is immobilized on a first solid support and washed with one or more solutions that dissociate aggregated aptamers; removing one or more components not associated with the first solid support; attaching a second tag to the target molecule in the aptamer-target affinity complex, wherein the second tag has an affinity to a second capture element; releasing the aptamer-target affinity complex from said first solid support; exposing the released aptamer-target affinity complex to a second solid support comprising a second capture element and allowing the second tag to associate with said second capture element; removing one or more components not associated with the second solid support; and eluting the aptamer from the second solid support with one or more solutions comprising a chaotropic salt.
The present disclosure describes methods, devices, reagents, and kits for the detection of one or more target molecules that may be present in a test sample. The described methods, devices, kits, and reagents facilitate the detection and quantification of a non-nucleic acid target (e.g., a protein target) in a test sample by detecting and quantifying a nucleic acid (i.e., an aptamer) where the aptamer-aptamer interactions are significantly reduced or eliminated while maintaining the aptamer-target interaction.1-73. (canceled) 74. A method comprising: exposing an aptamer to a first solid support, wherein the aptamer comprises a first tag and the first solid support comprises a first capture element, and wherein the first tag has affinity for the first capture element; allowing the first tag to associate with the first capture element; washing the first solid support with one or more solutions that dissociate aggregated aptamers; contacting the aptamer with a test sample, wherein an aptamer-target affinity complex is formed if the target molecule is present in the test sample; removing one or more components not associated with the first solid support; attaching a second tag to the target molecule in the aptamer-target affinity complex, wherein the second tag has an affinity to a second capture element; releasing the aptamer-target affinity complex from said first solid support; exposing the released aptamer-target affinity complex to a second solid support comprising a second capture element and allowing the second tag to associate with said second capture element; removing one or more components not associated with the second solid support; and eluting the aptamer from the second solid support with one or more buffered solutions comprising a chaotropic salt. 75. The method of claim 74 further comprising the step of detecting the aptamer portion of said aptamer-target affinity complex. 76. The method of claim 75 further comprising quantifying the aptamer. 77. The method of claim 75 further comprising detecting the aptamer by hybridizing the aptamer to a third solid support, wherein the third solid support comprises a plurality of addressable features and wherein at least one of said features comprises at least capture element disposed thereon that is complementary to any sequence contained within the aptamer. 78. The method of claim 75, wherein the aptamer is detected and optionally quantified using a method selected from the group consisting of Q-PCR, MS, next-generation sequencing and hybridization. 79. The method of claim 78, wherein said Q-PCR is performed using TaqMan® PCR, an intercalating fluorescent dye during the PCR process, or a molecular beacon during the PCR process. 80. The method of claim 74, wherein the pH of the one or more solutions is about 11. 81. The method of claim 74, wherein the pH of the one or more buffered solutions is neutral. 82. The method of claim 74, wherein the chaotropic salt disrupts aptamer-target interactions. 83. The method of claim 74, wherein said chaotropic salt is selected from the group consisting of sodium perchlorate, lithium chloride, magnesium chloride and sodium chloride. 84. The method of claim 74, wherein the one or more of the buffered solutions comprises an organic solvent. 85. The method of claim 84, wherein the organic solvent is glycerol. 86. The method of claim 74, wherein the aptamer is a single-stranded nucleic acid or a double-stranded nucleic acid. 87. The method of claim 74, wherein the aptamer comprises DNA, RNA or both DNA and RNA. 88. The method of claim 74, wherein the aptamer-target affinity complex has a slow rate of dissociation. 89. The method of claim 74, wherein the rate of dissociation of the aptamer-target affinity complex (t1/2) is selected from the group consisting of >30 minutes, >60 minutes, >90 minutes, >120 minutes, >150 minutes, >180 minutes, >210 minutes, and >240 minutes. 90. The method of 74, wherein the aptamer comprises a detectable moiety is selected from the group consisting of a dye, a quantum dot, a radiolabel, an electrochemical functional group, and an enzyme plus a detectable enzyme substrate. 91. The method of claim 90, wherein the dye is a fluorescent dye. 92. The method of claim 74, wherein the aptamer comprises at least one C-5 modified nucleotide. 93. The method of claim 74, wherein the aptamer comprises at least one chemical modification comprising a chemical substitution at one or more positions independently selected from a ribose position, a deoxyribose position, a phosphate position, and a base position. 94. The method of claim 93, wherein the chemical modification is independently selected from the group consisting of a 2′-position sugar modification, a 2′-amino (2′-NH2), a 2′-fluoro (2′-F), a 2′-0-methyl (2′-OMe), a 5-position pyrimidine modification, an 8-position purine modification, a modification at a cytosine exocyclic amine, a substitution of 5-bromouracil, a substitution of 5-bromodeoxyuridine, a substitution of 5-bromodeoxycytidine, a backbone modification, methylation, a 3′ cap, and a 5′ cap. 95. The method of claim 74, wherein said target molecule is selected from the group consisting of a protein, a peptide, a carbohydrate, a polysaccharide, a glycoprotein, a hormone, a receptor, an antigen, an antibody, a virus, a substrate, a metabolite, a transition state analog, a cofactor, an inhibitor, a drug, a dye, a nutrient, a growth factor, a tissue, and a controlled substance. 96. The method claim 74, wherein the test sample is selected from the group consisting of blood, whole blood, leukocytes, peripheral blood mononuclear cells, plasma, serum, sputum, breath, urine, semen, saliva, meningeal fluid, amniotic fluid, glandular fluid, lymph fluid, nipple aspirate, bronchial aspirate, synovial fluid, joint aspirate, cells, a cellular extract, stool, tissue, a tissue extract, a tissue biopsy, and cerebrospinal fluid. 97. The method of claim 74, wherein the first tag and the second tag each comprises at least one component independently selected from the group consisting of a polynucleotide, a polypeptide, a peptide nucleic acid, a locked nucleic acid, an oligosaccharide, a polysaccharide, an antibody, an affibody, an antibody mimic, a cell receptor, a ligand, a lipid, biotin, avidin, streptavidin, Extravidin, neutravidin, Traptavidin, a metal, histidine, and any portion of any of these structures. 98. The method of claim 74, wherein said first capture element and said second capture element each comprises at least one component independently selected from a polynucleotide, a polypeptide, a peptide nucleic acid, a locked nucleic acid, an oligosaccharide, a polysaccharide, an antibody, an affibody, an antibody mimic, a cell receptor, a ligand, a lipid, biotin, avidin, streptavidin, Extravidin, neutravidin, Traptavidin, a metal, histidine, and any portion of any of these structures. 99. The method of claim 74, wherein the first tag comprises a releasable moiety. 100. The method of claim 99, wherein the releasable moiety comprises a photocleavable moiety. 101. The method of claim 74, wherein said first solid support and second solid support each is independently selected from the group consisting of a polymer bead, an agarose bead, a polystyrene bead, an acrylamide bead, a solid core bead, a porous bead, a paramagnetic bead, glass bead, controlled pore bead, a microtitre well, a cyclo-olefin copolymer substrate, a membrane, a plastic substrate, nylon, a Langmuir-Blodgett film, glass, a germanium substrate, a silicon substrate, a silicon wafer chip, a flow through chip, a microbead, a nanoparticle, a polytetrafluoroethylene substrate, a polystyrene substrate, a gallium arsenide substrate, a gold substrate, and a silver substrate. 102. A kit comprising: a) one or more aptamers, wherein each of the one or more aptamers has specific affinity for one or more targets; b) one or more solid supports; c) one or more partitioning reagents; d) one or more reagents for the release of an aptamer from an aptamer-target affinity complex; e) one or more buffer solutions comprising an organic solvent; and f) one or more buffer solutions comprising a chaotropic salt. 103. The kit of claim 102, wherein said organic solvent is glycerol. 104. The kit of claim 102, wherein said chaotropic salt is sodium perchlorate. 105. The kit of claim 102 further comprising a reagent to cleave a cleavable moiety of the one or more aptamers. 106. A method comprising: contacting an aptamer with a test sample, wherein an aptamer-target affinity complex is formed if the target molecule is present in the test sample, and wherein the aptamer is immobilized on a first solid support and washed with one or more solutions that dissociate aggregated aptamers; removing one or more components not associated with the first solid support; attaching a second tag to the target molecule in the aptamer-target affinity complex, wherein the second tag has an affinity to a second capture element; releasing the aptamer-target affinity complex from said first solid support; exposing the released aptamer-target affinity complex to a second solid support comprising a second capture element and allowing the second tag to associate with said second capture element; removing one or more components not associated with the second solid support; and eluting the aptamer from the second solid support with one or more solutions comprising a chaotropic salt.
1,600
252
252
13,859,811
1,644
The production of soluble HLA class II molecules, as well as methods of using the soluble HLA class II molecules so produced, are described herein.
1. A method of producing isolated, HLA class II trimolecular complexes, wherein the isolated, HLA class II trimolecular complexes comprise a soluble, glycosylated alpha chain, a soluble, glycosylated beta chain, and a non-covalently associated, endogenously produced peptide ligand, the method comprising the steps of: inserting a first isolated nucleic acid segment and a second isolated nucleic acid segment into a mammalian cell line, the first isolated nucleic acid segment encoding a soluble form of an alpha chain of a HLA class II molecule having a first domain of a super secondary structural motif attached thereto, and the second isolated nucleic acid segment encoding a soluble form of a beta chain of the HLA class II molecule having a second domain of the super secondary structural motif attached thereto, wherein the mammalian cell line is a non-human mammalian cell line or a human cell line that does not express endogenous HLA class II, and wherein the mammalian cell line comprises glycosylation mechanisms required for glycosylation of proteins produced therein and chaperone complexes required for peptide ligand loading into HLA class II molecules; culturing the recombinant mammalian cell line under conditions that allow for expression of the soluble class II alpha and beta chains, association of the soluble class II alpha and beta chains through the first and second domains of the super secondary structural motif, glycosylation of the soluble class II alpha and beta chains, and loading of an antigen binding groove formed from the soluble class II alpha and beta chains with an endogenously produced, non-covalently associated peptide ligand, thereby producing soluble class II trimolecular complexes; and isolating the soluble class II trimolecular complexes secreted from the recombinant mammalian cell line, whereby each trimolecular complex so isolated comprises identical recombinant, individual soluble alpha and beta chain molecules of the HLA class II. 2. The method of claim 1, wherein the first and second isolated nucleic acid segments are present in a single recombinant vector. 3. The method of claim 1, wherein the first isolated nucleic acid segment is present in a first recombinant vector and the second isolated nucleic acid segment is present in a second recombinant vector. 4. The method of claim 1, wherein the super secondary structural motif attached to the alpha and beta chains is a leucine zipper motif sequence that acts as a tethering moiety for the alpha and beta chains. 5. A method of producing functionally active, individual soluble HLA class II trimolecular complexes that are purified substantially away from other proteins such that the individual soluble HLA class II trimolecular complexes maintain the physical, functional and antigenic integrity of the native HLA class II trimolecular complex, wherein each trimolecular complex comprises a recombinant, soluble alpha chain and a recombinant, soluble beta chain of an individual HLA class II molecule, and a peptide endogenously loaded in an antigen binding groove formed by the alpha and beta chains of the individual soluble HLA class II molecule, the method comprising the steps of: inserting a first isolated nucleic acid segment and a second isolated nucleic acid segment into a mammalian cell line, the first isolated nucleic acid segment encoding a soluble form of an alpha chain of a HLA class II molecule having a first domain of a super secondary structural motif attached thereto, and the second isolated nucleic acid segment encoding a soluble form of a beta chain of the HLA class II molecule having a second domain of the super secondary structural motif attached thereto, wherein the mammalian cell line is a non-human cell line or a human cell line that does not express endogenous HLA class II, and wherein the mammalian cell line comprises glycosylation mechanisms required for glycosylation of proteins produced therein and chaperone complexes required for peptide ligand loading into HLA class II molecules; culturing the recombinant mammalian cell line under conditions that allow for expression of the soluble class II alpha and beta chains, association of the soluble class II alpha and beta chains through the first and second domains of the super secondary structural motif, glycosylation of the soluble class II alpha and beta chains, and loading of an antigen binding groove formed from the soluble class II alpha and beta chains with an endogenously produced, non-covalently associated peptide ligand, thereby producing soluble HLA class II trimolecular complexes; and purifying the individual, soluble HLA class II trimolecular complexes substantially away from other proteins, wherein the individual soluble HLA class II trimolecular complexes maintain the physical, functional and antigenic integrity of the native HLA class II trimolecular complex, and wherein each trimolecular complex so purified comprises identical recombinant, individual soluble alpha and beta chain molecules of the HLA class II. 6. The method of claim 5, wherein the first and second isolated nucleic acid segments are present in a single recombinant vector. 7. The method of claim 5, wherein the first isolated nucleic acid segment is present in a first recombinant vector and the second isolated nucleic acid segment is present in a second recombinant vector. 8. The method of claim 5, wherein the super secondary structural motif attached to the alpha and beta chains is a leucine zipper motif sequence that acts as a tethering moiety for the alpha and beta chains. 9. A multimer of at least two soluble HLA class II trimolecular complexes, wherein each of the at least two soluble HLA class II trimolecular complexes comprises a soluble, glycosylated alpha chain attached to a soluble, glycosylated beta chain via a super secondary structure, and a non-covalently associated, endogenously produced peptide ligand disposed in an antigen binding groove formed by the association of the alpha and beta chains. 10. The multimer of claim 9, wherein the at least two soluble HLA class II trimolecular complexes comprises a tail attached thereto to aid in multimerization. 11. The multimer of claim 9, wherein the tail is selected from the group consisting of a biotinylation signal peptide tail, an immunoglobulin heavy chain tail, a TNF tail, an IgM tail, a leucine zipper, a Fos/Jun tail, and combinations thereof.
The production of soluble HLA class II molecules, as well as methods of using the soluble HLA class II molecules so produced, are described herein.1. A method of producing isolated, HLA class II trimolecular complexes, wherein the isolated, HLA class II trimolecular complexes comprise a soluble, glycosylated alpha chain, a soluble, glycosylated beta chain, and a non-covalently associated, endogenously produced peptide ligand, the method comprising the steps of: inserting a first isolated nucleic acid segment and a second isolated nucleic acid segment into a mammalian cell line, the first isolated nucleic acid segment encoding a soluble form of an alpha chain of a HLA class II molecule having a first domain of a super secondary structural motif attached thereto, and the second isolated nucleic acid segment encoding a soluble form of a beta chain of the HLA class II molecule having a second domain of the super secondary structural motif attached thereto, wherein the mammalian cell line is a non-human mammalian cell line or a human cell line that does not express endogenous HLA class II, and wherein the mammalian cell line comprises glycosylation mechanisms required for glycosylation of proteins produced therein and chaperone complexes required for peptide ligand loading into HLA class II molecules; culturing the recombinant mammalian cell line under conditions that allow for expression of the soluble class II alpha and beta chains, association of the soluble class II alpha and beta chains through the first and second domains of the super secondary structural motif, glycosylation of the soluble class II alpha and beta chains, and loading of an antigen binding groove formed from the soluble class II alpha and beta chains with an endogenously produced, non-covalently associated peptide ligand, thereby producing soluble class II trimolecular complexes; and isolating the soluble class II trimolecular complexes secreted from the recombinant mammalian cell line, whereby each trimolecular complex so isolated comprises identical recombinant, individual soluble alpha and beta chain molecules of the HLA class II. 2. The method of claim 1, wherein the first and second isolated nucleic acid segments are present in a single recombinant vector. 3. The method of claim 1, wherein the first isolated nucleic acid segment is present in a first recombinant vector and the second isolated nucleic acid segment is present in a second recombinant vector. 4. The method of claim 1, wherein the super secondary structural motif attached to the alpha and beta chains is a leucine zipper motif sequence that acts as a tethering moiety for the alpha and beta chains. 5. A method of producing functionally active, individual soluble HLA class II trimolecular complexes that are purified substantially away from other proteins such that the individual soluble HLA class II trimolecular complexes maintain the physical, functional and antigenic integrity of the native HLA class II trimolecular complex, wherein each trimolecular complex comprises a recombinant, soluble alpha chain and a recombinant, soluble beta chain of an individual HLA class II molecule, and a peptide endogenously loaded in an antigen binding groove formed by the alpha and beta chains of the individual soluble HLA class II molecule, the method comprising the steps of: inserting a first isolated nucleic acid segment and a second isolated nucleic acid segment into a mammalian cell line, the first isolated nucleic acid segment encoding a soluble form of an alpha chain of a HLA class II molecule having a first domain of a super secondary structural motif attached thereto, and the second isolated nucleic acid segment encoding a soluble form of a beta chain of the HLA class II molecule having a second domain of the super secondary structural motif attached thereto, wherein the mammalian cell line is a non-human cell line or a human cell line that does not express endogenous HLA class II, and wherein the mammalian cell line comprises glycosylation mechanisms required for glycosylation of proteins produced therein and chaperone complexes required for peptide ligand loading into HLA class II molecules; culturing the recombinant mammalian cell line under conditions that allow for expression of the soluble class II alpha and beta chains, association of the soluble class II alpha and beta chains through the first and second domains of the super secondary structural motif, glycosylation of the soluble class II alpha and beta chains, and loading of an antigen binding groove formed from the soluble class II alpha and beta chains with an endogenously produced, non-covalently associated peptide ligand, thereby producing soluble HLA class II trimolecular complexes; and purifying the individual, soluble HLA class II trimolecular complexes substantially away from other proteins, wherein the individual soluble HLA class II trimolecular complexes maintain the physical, functional and antigenic integrity of the native HLA class II trimolecular complex, and wherein each trimolecular complex so purified comprises identical recombinant, individual soluble alpha and beta chain molecules of the HLA class II. 6. The method of claim 5, wherein the first and second isolated nucleic acid segments are present in a single recombinant vector. 7. The method of claim 5, wherein the first isolated nucleic acid segment is present in a first recombinant vector and the second isolated nucleic acid segment is present in a second recombinant vector. 8. The method of claim 5, wherein the super secondary structural motif attached to the alpha and beta chains is a leucine zipper motif sequence that acts as a tethering moiety for the alpha and beta chains. 9. A multimer of at least two soluble HLA class II trimolecular complexes, wherein each of the at least two soluble HLA class II trimolecular complexes comprises a soluble, glycosylated alpha chain attached to a soluble, glycosylated beta chain via a super secondary structure, and a non-covalently associated, endogenously produced peptide ligand disposed in an antigen binding groove formed by the association of the alpha and beta chains. 10. The multimer of claim 9, wherein the at least two soluble HLA class II trimolecular complexes comprises a tail attached thereto to aid in multimerization. 11. The multimer of claim 9, wherein the tail is selected from the group consisting of a biotinylation signal peptide tail, an immunoglobulin heavy chain tail, a TNF tail, an IgM tail, a leucine zipper, a Fos/Jun tail, and combinations thereof.
1,600
253
253
15,415,754
1,652
Protein enriched micro-vesicles and methods of making and using the same are provided. Aspects of the methods include maintaining a cell having a membrane-associated protein comprising a first dimerization domain and a target protein having a second dimerization domain under conditions sufficient to produce a micro-vesicle from the cell, wherein the micro-vesicle includes the target protein. Also provided are cells, reagents and kits that find use in making the micro-vesicles, as well as methods of using the micro-vesicles, e.g., in research and therapeutic applications.
1.-34. (canceled) 35. A micro-vesicle comprising a membrane-associated protein comprising a first dimerization domain and a target protein comprising a second dimerization domain. 36. The micro-vesicle according to claim 35, wherein the membrane-associated protein and the target protein are present in a dimerized complex. 37. The micro-vesicle according to claim 36, wherein the first and second dimerization domains are specifically bound to each other in the dimerized complex. 38. The micro-vesicle according to claim 36, wherein first and second dimerization domains are bound to each other by a dimerization mediator. 39. The micro-vesicle according to claim 38, wherein the dimerization mediator is a modifiable dimerization mediator. 40. The micro-vesicle according to claim 35, wherein the micro-vesicle further comprises a micro-vesicle inducer. 41. The micro-vesicle according to claim 40, wherein the micro-vesicle inducer comprises a viral membrane fusion protein. 42. The micro-vesicle according to claim 41, wherein the viral membrane fusion protein is VSV-G. 43. The micro-vesicle according to claim 35, wherein the membrane-associated protein is selected from the group consisting of a myristoylated protein, a farnesylated protein, a membrane anchor protein, a transmembrane protein and membrane lipid protein. 44. The micro-vesicle according to claim 35, wherein the first and second dimerization domains are heterodimeric. 45. The micro-vesicle according to claim 35, wherein the first and second dimerization domains are homodimeric. 46. The micro-vesicle according to claim 35, wherein the first and second dimerization domains are selected from DmrA and DmrC domains, DmrB domains, DmrD domains, dimerization domains of the dihydrofolate reductase system, dimerization domains of TAg and p 53, and dimerization domains of SH2 and a PTRK protein. 47. The micro-vesicle according to claim 35, wherein the micro-vesicle further comprises a second target protein comprising a third dimerization domain. 48. A method of introducing a target protein into a cell, the method comprising: contacting the cell with a micro-vesicle comprising: (a) a membrane-associated protein comprising a first dimerization domain; and (b) a target protein comprising a second dimerization domain; under conditions sufficient for the micro-vesicle to fuse with the cell and introduce the target protein into the cell. 49-55. (canceled) 56. A kit, comprising: a first expression cassette comprising a coding sequence for a membrane-associated protein comprising a first dimerization domain; and a second expression cassette comprising a promoter and a second dimerization domain. 57-62. (canceled) 63. The micro-vesicle according to claim 35, wherein the target protein is a research protein. 64. The micro-vesicle according to claim 63, wherein the research protein is a genomic modification protein. 65. The micro-vesicle according to claim 64, wherein the genomic modification protein is selected from the group consisting of: CRE recombinases, meganucleases, Zinc-finger nucleases, CRISPR/Cas-9 nucleases and TAL effector nucleases.
Protein enriched micro-vesicles and methods of making and using the same are provided. Aspects of the methods include maintaining a cell having a membrane-associated protein comprising a first dimerization domain and a target protein having a second dimerization domain under conditions sufficient to produce a micro-vesicle from the cell, wherein the micro-vesicle includes the target protein. Also provided are cells, reagents and kits that find use in making the micro-vesicles, as well as methods of using the micro-vesicles, e.g., in research and therapeutic applications.1.-34. (canceled) 35. A micro-vesicle comprising a membrane-associated protein comprising a first dimerization domain and a target protein comprising a second dimerization domain. 36. The micro-vesicle according to claim 35, wherein the membrane-associated protein and the target protein are present in a dimerized complex. 37. The micro-vesicle according to claim 36, wherein the first and second dimerization domains are specifically bound to each other in the dimerized complex. 38. The micro-vesicle according to claim 36, wherein first and second dimerization domains are bound to each other by a dimerization mediator. 39. The micro-vesicle according to claim 38, wherein the dimerization mediator is a modifiable dimerization mediator. 40. The micro-vesicle according to claim 35, wherein the micro-vesicle further comprises a micro-vesicle inducer. 41. The micro-vesicle according to claim 40, wherein the micro-vesicle inducer comprises a viral membrane fusion protein. 42. The micro-vesicle according to claim 41, wherein the viral membrane fusion protein is VSV-G. 43. The micro-vesicle according to claim 35, wherein the membrane-associated protein is selected from the group consisting of a myristoylated protein, a farnesylated protein, a membrane anchor protein, a transmembrane protein and membrane lipid protein. 44. The micro-vesicle according to claim 35, wherein the first and second dimerization domains are heterodimeric. 45. The micro-vesicle according to claim 35, wherein the first and second dimerization domains are homodimeric. 46. The micro-vesicle according to claim 35, wherein the first and second dimerization domains are selected from DmrA and DmrC domains, DmrB domains, DmrD domains, dimerization domains of the dihydrofolate reductase system, dimerization domains of TAg and p 53, and dimerization domains of SH2 and a PTRK protein. 47. The micro-vesicle according to claim 35, wherein the micro-vesicle further comprises a second target protein comprising a third dimerization domain. 48. A method of introducing a target protein into a cell, the method comprising: contacting the cell with a micro-vesicle comprising: (a) a membrane-associated protein comprising a first dimerization domain; and (b) a target protein comprising a second dimerization domain; under conditions sufficient for the micro-vesicle to fuse with the cell and introduce the target protein into the cell. 49-55. (canceled) 56. A kit, comprising: a first expression cassette comprising a coding sequence for a membrane-associated protein comprising a first dimerization domain; and a second expression cassette comprising a promoter and a second dimerization domain. 57-62. (canceled) 63. The micro-vesicle according to claim 35, wherein the target protein is a research protein. 64. The micro-vesicle according to claim 63, wherein the research protein is a genomic modification protein. 65. The micro-vesicle according to claim 64, wherein the genomic modification protein is selected from the group consisting of: CRE recombinases, meganucleases, Zinc-finger nucleases, CRISPR/Cas-9 nucleases and TAL effector nucleases.
1,600
254
254
14,174,433
1,613
Disclosed is a method of providing cleanness to hair and/or scalp, for example, providing hair volume and/or providing less oiliness on hair and/or scalp, by applying a hair care composition comprising cationic surfactants, high melting point fatty compounds, metal pyrithione, and metal salts other than metal pyrithiones.
1. A method of providing cleanness to hair and/or scalp, comprising following steps: (i) after shampooing hair, applying to the hair and/or scalp an effective amount of a hair care composition; and (ii) then rinsing the hair; wherein the hair care composition comprises by weight: (a) from about 0.1% to about 10% of a cationic surfactant system; (b) from about 0.1% to about 20% of a high melting point fatty compound; (c) from about 0.01% to about5.0% of a metal pyrithione; (d) from about 0.05% to about 10% of a metal salt other than metal pyrithiones; and (e) an aqueous carrier. 2. The method of claim 1 wherein providing cleanness to hair and/or scalp means, providing hair volume and/or providing less oiliness on hair and/or scalp. 3. The method of claim 1 wherein the composition further comprising from about 0.01% to about 5.0% of polyquaternium-6. 4. The method of claim 3 wherein the polyquaternium-6 has a molecular weight of from about 800 g/mol to about 1,000,000 g/mol. 5. The method of claim 3 wherein the polyquaternium-6 has a molecular weight of from about 1,000 g/mol to about 500,000 g/mol. 6. The method of claim 3 wherein the weight ratio of polyquaternium-6 to the metal pyrithione is from about 1:1 to about 1:30. 7. The method of claim 3 wherein the weight ratio of polyquaternium-6 to the metal pyrithione is from about 1:2 to about 1:20. 8. The method of claim 3 wherein the weight ratio of polyquaternium-6 to the metal pyrithione is from about 1:5 to about 1:15. 9. The method of claim 1 wherein the cationic surfactant system is selected from: mono-long alkyl quaternized ammonium salt; a combination of mono-long alkyl quaternized ammonium salt and di-long alkyl quaternized ammonium salt; mono-long alkyl amidoamine; and a combination of mono-long alkyl amidoamine and di-long alkyl quaternized ammonium salt. 10. The method of claim 1 wherein the cationic surfactant system is a combination of a mono-long alkyl quaternized ammonium salt and a di-long alkyl quaternized ammonium salt. 11. The method of claim 1 wherein the composition is substantially free of anionic surfactants. 12. The method of claim 1 wherein the composition is substantially free of glucan gum, guar gum, and hydroxyethylcellulose.
Disclosed is a method of providing cleanness to hair and/or scalp, for example, providing hair volume and/or providing less oiliness on hair and/or scalp, by applying a hair care composition comprising cationic surfactants, high melting point fatty compounds, metal pyrithione, and metal salts other than metal pyrithiones.1. A method of providing cleanness to hair and/or scalp, comprising following steps: (i) after shampooing hair, applying to the hair and/or scalp an effective amount of a hair care composition; and (ii) then rinsing the hair; wherein the hair care composition comprises by weight: (a) from about 0.1% to about 10% of a cationic surfactant system; (b) from about 0.1% to about 20% of a high melting point fatty compound; (c) from about 0.01% to about5.0% of a metal pyrithione; (d) from about 0.05% to about 10% of a metal salt other than metal pyrithiones; and (e) an aqueous carrier. 2. The method of claim 1 wherein providing cleanness to hair and/or scalp means, providing hair volume and/or providing less oiliness on hair and/or scalp. 3. The method of claim 1 wherein the composition further comprising from about 0.01% to about 5.0% of polyquaternium-6. 4. The method of claim 3 wherein the polyquaternium-6 has a molecular weight of from about 800 g/mol to about 1,000,000 g/mol. 5. The method of claim 3 wherein the polyquaternium-6 has a molecular weight of from about 1,000 g/mol to about 500,000 g/mol. 6. The method of claim 3 wherein the weight ratio of polyquaternium-6 to the metal pyrithione is from about 1:1 to about 1:30. 7. The method of claim 3 wherein the weight ratio of polyquaternium-6 to the metal pyrithione is from about 1:2 to about 1:20. 8. The method of claim 3 wherein the weight ratio of polyquaternium-6 to the metal pyrithione is from about 1:5 to about 1:15. 9. The method of claim 1 wherein the cationic surfactant system is selected from: mono-long alkyl quaternized ammonium salt; a combination of mono-long alkyl quaternized ammonium salt and di-long alkyl quaternized ammonium salt; mono-long alkyl amidoamine; and a combination of mono-long alkyl amidoamine and di-long alkyl quaternized ammonium salt. 10. The method of claim 1 wherein the cationic surfactant system is a combination of a mono-long alkyl quaternized ammonium salt and a di-long alkyl quaternized ammonium salt. 11. The method of claim 1 wherein the composition is substantially free of anionic surfactants. 12. The method of claim 1 wherein the composition is substantially free of glucan gum, guar gum, and hydroxyethylcellulose.
1,600
255
255
14,342,889
1,627
The invention provides a method for treating ALS or suppressing the disease progress thereof, or treating symptoms caused by ALS or suppressing the disease progress thereof by administering an agent containing, as an active ingredient, 3-methyl-1-phenyl-2-pyrazolin-5-one or a physiologically acceptable salt thereof, to a patient that complies with specific criteria. The agent is administered by repeating a 14-day administration period and a 14-day drug holiday period, or by establishing an initial 14-day administration period and an initial 14-day drug holiday period and then repeating an administration period for 10 out of 14 days and a 14-day drug holiday period
1.-4. (canceled) 5. A method for treating amyotrophic lateral sclerosis or suppressing the disease progress thereof, or treating symptoms caused by amyotrophic lateral sclerosis or suppressing the disease progress thereof, which comprises administering an effective amount of 3-methyl-1-phenyl-2-pyrazolin-5-one or a physiologically acceptable salt thereof to a patient, wherein the administration is carried out by repeating a 14-day administration period and a 14-day drug holiday period, or by establishing an initial 14-day administration period and an initial 14-day drug holiday period and then repeating an administration period for 10 out of 14 days and a 14-day drug holiday period, and wherein the patient is any one of the following [1] to [7]: [1] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [2] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [3] a patient who scores two or more points from all items constituting the ALSFRS-R; [4] a patient whose % FVC is 80% or more; [5] a patient who scores two or more points from all items constituting the ALSFRS-R and whose % FVC is 80% or more; [6] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more; and [7] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more. 6. The method according to claim 5, wherein with regard to the administration period and the drug holiday period, an initial 14-day administration period and an initial 14-day drug holiday period are established, and thereafter, an administration period for 10 out of 14 days and a 14-day drug holiday period are repeated. 7. The method according to claim 5, wherein the symptoms caused by amyotrophic lateral sclerosis are a decrease in respiratory function, spoken language disorder, dysphagia, or limb movement disorder. 8. A method for treating amyotrophic lateral sclerosis or suppressing the disease progress thereof, or treating symptoms caused by amyotrophic lateral sclerosis or suppressing the disease progress thereof, which comprises a step of selecting any one of the following patients [1] to [7] from patients with amyotrophic lateral sclerosis and a step of administering an effective amount of 3-methyl-1-phenyl-2-pyrazolin-5-one or a physiologically acceptable salt thereof to the thus selected patient, wherein a 14-day administration period and a 14-day drug holiday period are repeated, or an initial 14-day administration period and an initial 14-day drug holiday period are established, and thereafter, an administration period for 10 out of 14 days and a 14-day drug holiday period are repeated: [1] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [2] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [3] a patient who scores two or more points from all items constituting the ALSFRS-R; [4] a patient whose % FVC is 80% or more; [5] a patient who scores two or more points from all items constituting the ALSFRS-R and whose % FVC is 80% or more; [6] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more; and [7] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more. 9. The method according to claim 8, wherein with regard to the administration period and the drug holiday period, an initial 14-day administration period and an initial 14-day drug holiday period are established, and thereafter, an administration period for 10 out of 14 days and a 14-day drug holiday period are repeated. 10. The method according to claim 8, wherein the symptoms caused by amyotrophic lateral sclerosis are a decrease in respiratory function, spoken language disorder, dysphagia, or limb movement disorder. 11.-13. (canceled)
The invention provides a method for treating ALS or suppressing the disease progress thereof, or treating symptoms caused by ALS or suppressing the disease progress thereof by administering an agent containing, as an active ingredient, 3-methyl-1-phenyl-2-pyrazolin-5-one or a physiologically acceptable salt thereof, to a patient that complies with specific criteria. The agent is administered by repeating a 14-day administration period and a 14-day drug holiday period, or by establishing an initial 14-day administration period and an initial 14-day drug holiday period and then repeating an administration period for 10 out of 14 days and a 14-day drug holiday period1.-4. (canceled) 5. A method for treating amyotrophic lateral sclerosis or suppressing the disease progress thereof, or treating symptoms caused by amyotrophic lateral sclerosis or suppressing the disease progress thereof, which comprises administering an effective amount of 3-methyl-1-phenyl-2-pyrazolin-5-one or a physiologically acceptable salt thereof to a patient, wherein the administration is carried out by repeating a 14-day administration period and a 14-day drug holiday period, or by establishing an initial 14-day administration period and an initial 14-day drug holiday period and then repeating an administration period for 10 out of 14 days and a 14-day drug holiday period, and wherein the patient is any one of the following [1] to [7]: [1] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [2] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [3] a patient who scores two or more points from all items constituting the ALSFRS-R; [4] a patient whose % FVC is 80% or more; [5] a patient who scores two or more points from all items constituting the ALSFRS-R and whose % FVC is 80% or more; [6] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more; and [7] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more. 6. The method according to claim 5, wherein with regard to the administration period and the drug holiday period, an initial 14-day administration period and an initial 14-day drug holiday period are established, and thereafter, an administration period for 10 out of 14 days and a 14-day drug holiday period are repeated. 7. The method according to claim 5, wherein the symptoms caused by amyotrophic lateral sclerosis are a decrease in respiratory function, spoken language disorder, dysphagia, or limb movement disorder. 8. A method for treating amyotrophic lateral sclerosis or suppressing the disease progress thereof, or treating symptoms caused by amyotrophic lateral sclerosis or suppressing the disease progress thereof, which comprises a step of selecting any one of the following patients [1] to [7] from patients with amyotrophic lateral sclerosis and a step of administering an effective amount of 3-methyl-1-phenyl-2-pyrazolin-5-one or a physiologically acceptable salt thereof to the thus selected patient, wherein a 14-day administration period and a 14-day drug holiday period are repeated, or an initial 14-day administration period and an initial 14-day drug holiday period are established, and thereafter, an administration period for 10 out of 14 days and a 14-day drug holiday period are repeated: [1] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [2] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria; [3] a patient who scores two or more points from all items constituting the ALSFRS-R; [4] a patient whose % FVC is 80% or more; [5] a patient who scores two or more points from all items constituting the ALSFRS-R and whose % FVC is 80% or more; [6] a patient who is determined to be “Definite ALS” or “Probable ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more; and [7] a patient who is determined to be “Definite ALS” according to the revised El Escorial (Airlie House) diagnostic criteria, scores two or more points from all items constituting the ALSFRS-R, and whose % FVC is 80% or more. 9. The method according to claim 8, wherein with regard to the administration period and the drug holiday period, an initial 14-day administration period and an initial 14-day drug holiday period are established, and thereafter, an administration period for 10 out of 14 days and a 14-day drug holiday period are repeated. 10. The method according to claim 8, wherein the symptoms caused by amyotrophic lateral sclerosis are a decrease in respiratory function, spoken language disorder, dysphagia, or limb movement disorder. 11.-13. (canceled)
1,600
256
256
15,680,113
1,612
The anesthetic chewing gum includes lidocaine HCL and prilocaine HCL as the anesthetic ingredients, as well as a chewing gum base. The anesthetic chewing gum may also include one or more sweeters, an anti-adherent, a lubricant, an opacifier, a glidant, a flavoring agent, and a flavor enhancer.
1. An anesthetic chewing gum, comprising a combination of an analgesic component and a chewing gum base component: the analgesic component consists of 2 mg lidocaine HCL and 2 mg prilocaine HCL; and the chewing gum base component comprises a chewing gum base. 2. The anesthetic chewing gum according to claim 1, wherein the chewing gum base comprises 436 mg of the chewing gum base. 3. The anesthetic chewing gum according to claim 2, further comprising: about 30 mg of a sweetener; about 10 mg of an anti-adherent; about 5 mg of a lubricant; about 5 mg of an opacifier; about 2.5 mg of a glidant; about 2.5 mg of a flavoring agent; and about 5 mg of a flavor enhancer. 4. The anesthetic chewing gum according to claim 2, wherein the chewing gum is a chewing gum tablet, wherein the tablet has a diameter of about 10 mm, a thickness of about 6 mm, and a weight of about 499 mg. 5. The anesthetic chewing gum according to claim 3, wherein the chewing gum tablet has a hardness of about 4 kg/cm2. 6. The anesthetic chewing gum according to claim 3, wherein the anti-adherent comprises talc. 7. The anesthetic chewing gum according to claim 3, wherein the lubricant comprises magnesium stearate. 8. The anesthetic chewing gum according to claim 3, wherein the opacifier comprises titanium dioxide. 9. The anesthetic chewing gum according to claim 3, wherein the glidant comprises colloidal silicon dioxide. 10. The anesthetic chewing gum according to claim 3, wherein the flavoring agent comprises peppermint oil. 11. The anesthetic chewing gum according to claim 3, wherein the flavor enhancer comprises menthol. 12. A method of making an anesthetic chewing gum, the steps consisting of: forming a plurality of separate lidocaine-HCL mixture portions by separately mixing an amount of lidocaine HCL with an amount of a secondary chewing gum ingredient, the secondary chewing gum ingredient being selected from the group consisting of a sweetener, an anti-adherent, a lubricant, an opacifier, a glidant, and a flavor enhancer; forming a plurality of separate prilocaine HCL mixture portions by separately mixing an amount of prilocaine HCL with an amount of one of the secondary chewing gum ingredients; mixing together each lidocaine HCL mixture portion and prilocaine HCL mixture portion having a same secondary chewing gum ingredient to form a plurality of lidocaine HCL/prilocaine HCL mixture portions; mixing the plurality of lidocaine HCL/prilocaine HCL mixture portions together to form a homogenized lidocaine HCL-prilocaine HCL mixture; combining the homogenized lidocaine HCL-prilocaine HCL mixture with a gum base to form a homogenized lidocaine HCL-prilocaine HCL gum base mixture; adding a flavoring agent to the homogenized lidocaine HCL-prilocaine HCL gum base mixture to form granules; drying the granules at room temperature; mixing the granules to form a final granule blend; and compressing the final granule blend into a plurality of chewing gum tablets, wherein each chewing gum tablet contains 2 mg of each of lidocaine HCL and prilocaine HCL. 13. The method of making an anesthetic chewing gum according to claim 12, wherein compressing the final granule blend comprises using a direct compression technique. 14. The method of making an anesthetic chewing gum according to claim 12, wherein the chewing gum tablets have an average diameter of about 10 mm, an average thickness of about 6 mm, and an average weight of about 499 mg. 15. The method of making an anesthetic chewing gum according to claim 12, wherein the chewing gum tablets have an average hardness of about 4 kg/cm2. 16. The anesthetic chewing gum according to claim 12, wherein the anti-adherent comprises talc. 17. The anesthetic chewing gum according to claim 12, wherein the lubricant comprises magnesium stearate. 18. The anesthetic chewing gum according to claim 12, wherein the opacifier comprises titanium dioxide. 19. The anesthetic chewing gum according to claim 12, wherein the glidant comprises colloidal silicon dioxide. 20. The anesthetic chewing gum according to claim 12, wherein the flavoring agent comprises peppermint oil.
The anesthetic chewing gum includes lidocaine HCL and prilocaine HCL as the anesthetic ingredients, as well as a chewing gum base. The anesthetic chewing gum may also include one or more sweeters, an anti-adherent, a lubricant, an opacifier, a glidant, a flavoring agent, and a flavor enhancer.1. An anesthetic chewing gum, comprising a combination of an analgesic component and a chewing gum base component: the analgesic component consists of 2 mg lidocaine HCL and 2 mg prilocaine HCL; and the chewing gum base component comprises a chewing gum base. 2. The anesthetic chewing gum according to claim 1, wherein the chewing gum base comprises 436 mg of the chewing gum base. 3. The anesthetic chewing gum according to claim 2, further comprising: about 30 mg of a sweetener; about 10 mg of an anti-adherent; about 5 mg of a lubricant; about 5 mg of an opacifier; about 2.5 mg of a glidant; about 2.5 mg of a flavoring agent; and about 5 mg of a flavor enhancer. 4. The anesthetic chewing gum according to claim 2, wherein the chewing gum is a chewing gum tablet, wherein the tablet has a diameter of about 10 mm, a thickness of about 6 mm, and a weight of about 499 mg. 5. The anesthetic chewing gum according to claim 3, wherein the chewing gum tablet has a hardness of about 4 kg/cm2. 6. The anesthetic chewing gum according to claim 3, wherein the anti-adherent comprises talc. 7. The anesthetic chewing gum according to claim 3, wherein the lubricant comprises magnesium stearate. 8. The anesthetic chewing gum according to claim 3, wherein the opacifier comprises titanium dioxide. 9. The anesthetic chewing gum according to claim 3, wherein the glidant comprises colloidal silicon dioxide. 10. The anesthetic chewing gum according to claim 3, wherein the flavoring agent comprises peppermint oil. 11. The anesthetic chewing gum according to claim 3, wherein the flavor enhancer comprises menthol. 12. A method of making an anesthetic chewing gum, the steps consisting of: forming a plurality of separate lidocaine-HCL mixture portions by separately mixing an amount of lidocaine HCL with an amount of a secondary chewing gum ingredient, the secondary chewing gum ingredient being selected from the group consisting of a sweetener, an anti-adherent, a lubricant, an opacifier, a glidant, and a flavor enhancer; forming a plurality of separate prilocaine HCL mixture portions by separately mixing an amount of prilocaine HCL with an amount of one of the secondary chewing gum ingredients; mixing together each lidocaine HCL mixture portion and prilocaine HCL mixture portion having a same secondary chewing gum ingredient to form a plurality of lidocaine HCL/prilocaine HCL mixture portions; mixing the plurality of lidocaine HCL/prilocaine HCL mixture portions together to form a homogenized lidocaine HCL-prilocaine HCL mixture; combining the homogenized lidocaine HCL-prilocaine HCL mixture with a gum base to form a homogenized lidocaine HCL-prilocaine HCL gum base mixture; adding a flavoring agent to the homogenized lidocaine HCL-prilocaine HCL gum base mixture to form granules; drying the granules at room temperature; mixing the granules to form a final granule blend; and compressing the final granule blend into a plurality of chewing gum tablets, wherein each chewing gum tablet contains 2 mg of each of lidocaine HCL and prilocaine HCL. 13. The method of making an anesthetic chewing gum according to claim 12, wherein compressing the final granule blend comprises using a direct compression technique. 14. The method of making an anesthetic chewing gum according to claim 12, wherein the chewing gum tablets have an average diameter of about 10 mm, an average thickness of about 6 mm, and an average weight of about 499 mg. 15. The method of making an anesthetic chewing gum according to claim 12, wherein the chewing gum tablets have an average hardness of about 4 kg/cm2. 16. The anesthetic chewing gum according to claim 12, wherein the anti-adherent comprises talc. 17. The anesthetic chewing gum according to claim 12, wherein the lubricant comprises magnesium stearate. 18. The anesthetic chewing gum according to claim 12, wherein the opacifier comprises titanium dioxide. 19. The anesthetic chewing gum according to claim 12, wherein the glidant comprises colloidal silicon dioxide. 20. The anesthetic chewing gum according to claim 12, wherein the flavoring agent comprises peppermint oil.
1,600
257
257
12,961,611
1,611
A wipe that contains a fibrous web on which is coated an antimicrobial composition is provided. The composition includes a botanical oil derived from a plant (e.g., thymol, carvacrol, etc.). Because the botanical oil is volatile and tends to evaporate and lose efficacy during storage and prior to use, a protein is also employed in the composition to enhance long term stability of the oil and, in turn, its antimicrobial efficacy. The protein is “film-forming” in the sense that it tends to form a substantially continuous film when coated onto a surface of the fibrous web. Because such proteins are typically stiff and brittle in nature, a continuous film would restrict the ability of the fibers to move and bend, thereby reducing web flexibility and drape. Thus, it is typically desired that the antimicrobial composition form a discontinuous coating on the fibrous web. In this regard, the present inventors have surprisingly discovered that the addition of an organopolysiloxane can help achieve such a discontinuous coating without adversely impacting the ability of the protein to stabilize the botanical oil. The organopolysiloxane may also enhance the softness and overall handfeel of the wipe.
1. A wipe comprising a web that includes a plurality of fibers, wherein the web is coated with an antimicrobial composition, the antimicrobial composition comprising a botanical oil that is at least partially encapsulated by a film-forming protein, wherein the composition forms a discontinuous coating on at least a portion of the fibers of the web. 2. The wipe of claim 1, wherein the botanical oil includes a monoterpene phenol. 3. The wipe of claim 2, wherein the monoterpene phenol is thymol, carvacol, or a mixture thereof. 4. The wipe of claim 1, wherein the botanical oil is derived from a plant oil extract. 5. The wipe of claim 1, wherein the botanical oil is synthesized. 6. The wipe of claim 1, wherein the protein is a plant protein. 7. The wipe of claim 6, wherein the plant protein is zein, corn gluten, wheat gluten, whey protein, soy protein, or a combination thereof. 8. The wipe of claim 6, wherein the plant protein is a soy protein. 9. The wipe of claim 1, wherein the antimicrobial composition further comprises an organopolysiloxane. 10. The wipe of claim 9, wherein the organopolysiloxane is a methylpolysiloxane. 11. The wipe of claim 10, wherein the methylpolysiloxane is polydimethylsiloxane. 12. The wipe of claim 9, wherein the organopolysiloxane contains a hydroxyl functional group, alkoxy functional group, amino functional group, or a combination thereof. 13. The wipe of claim 12, wherein the organopolysiloxane is hydroxy-terminated polydimethylsiloxane. 14. The wipe of claim 1, wherein the antimicrobial composition has a solvent content of less than about 5 wt. %. 15. The wipe of claim 1, wherein the antimicrobial composition comprises botanical oils in an amount of from about 0.05 wt. % to about 50 wt. %. 16. The wipe of claim 1, wherein the web is a nonwoven web. 17. The wipe of claim 1, wherein the fibers include absorbent fibers. 18. The wipe of claim 17, wherein the web is a composite of absorbent fibers and synthetic thermoplastic fibers. 19. The wipe of claim 1, wherein the antimicrobial composition further comprises a carbohydrate polymer. 20. The wipe of claim 19, wherein the carbohydrate biopolymer includes a chemically modified starch polymer. 21. The wipe of claim 20, wherein the chemically modified starch polymer is a hydroxyalkyl starch. 22. A method for forming an antimicrobial wipe, the method comprising: forming a coating solution that comprises a botanical oil, protein, and a solvent; applying the coating solution to a web that includes a plurality of fibers; and drying the coating solution to form an antimicrobial coating on the web. 23. The method of claim 22, wherein the monoterpene phenol is thymol, carvacol, or a mixture thereof. 24. The method of claim 22, wherein the protein is a plant protein. 25. The method of claim 22, wherein the coating solution further comprises an organopolysiloxane emulsion. 26. The method of claim 22, wherein the emulsion includes polydimethylsiloxane, modified polydimethylsiloxane, or a combination thereof. 27. The method of claim 22, wherein the antimicrobial coating has a solvent content of less than about 5 wt. %. 28. A method for inhibiting the growth of bacteria on a surface, the method comprising contacting the surface with a wipe that includes a web comprising a plurality of fibers, wherein the web is coated with an antimicrobial composition, the antimicrobial composition comprising a botanical oil that is at least partially encapsulated by a film-forming protein. 29. The method of claim 28, wherein the composition is in the form of a concentrate. 30. The method of claim 29, wherein prior to contacting the surface with the wipe, water is applied to the concentrate to initiate dispersion of the biopolymer and release the botanical oil. 31. An antimicrobial concentrate having a solvent content of about 5 wt. % or less, wherein the concentrate comprises at least one monoterpene phenol in an amount of from about 0.05 wt. % to about 50 wt. %, at least one film-forming protein in an amount of from about 5 wt. % to about 50 wt. %, and at least one organopolysiloxane in an amount of from about 10 wt. % to about 70 wt. %, and further wherein the protein at least partially encapsulates the monoterpene phenol. 32. The antimicrobial concentrate of claim 31, wherein the monoterpene phenol includes thymol, carvacol, or a mixture thereof. 33. The antimicrobial concentrate of claim 31, wherein the concentrate comprises at least one monoterpene phenol in an amount of from about 1 wt. % to about 40 wt. %, at least one film-forming protein in an amount of from about 10 wt. % to about 40 wt. %, and at least one organopolysiloxane in an amount of from about 20 wt. % to about 60 wt. %. 34. The antimicrobial concentrate of claim 31, further comprising at least one carbohydrate polymer in an amount of from about 5 wt. % to about 50 wt. %.
A wipe that contains a fibrous web on which is coated an antimicrobial composition is provided. The composition includes a botanical oil derived from a plant (e.g., thymol, carvacrol, etc.). Because the botanical oil is volatile and tends to evaporate and lose efficacy during storage and prior to use, a protein is also employed in the composition to enhance long term stability of the oil and, in turn, its antimicrobial efficacy. The protein is “film-forming” in the sense that it tends to form a substantially continuous film when coated onto a surface of the fibrous web. Because such proteins are typically stiff and brittle in nature, a continuous film would restrict the ability of the fibers to move and bend, thereby reducing web flexibility and drape. Thus, it is typically desired that the antimicrobial composition form a discontinuous coating on the fibrous web. In this regard, the present inventors have surprisingly discovered that the addition of an organopolysiloxane can help achieve such a discontinuous coating without adversely impacting the ability of the protein to stabilize the botanical oil. The organopolysiloxane may also enhance the softness and overall handfeel of the wipe.1. A wipe comprising a web that includes a plurality of fibers, wherein the web is coated with an antimicrobial composition, the antimicrobial composition comprising a botanical oil that is at least partially encapsulated by a film-forming protein, wherein the composition forms a discontinuous coating on at least a portion of the fibers of the web. 2. The wipe of claim 1, wherein the botanical oil includes a monoterpene phenol. 3. The wipe of claim 2, wherein the monoterpene phenol is thymol, carvacol, or a mixture thereof. 4. The wipe of claim 1, wherein the botanical oil is derived from a plant oil extract. 5. The wipe of claim 1, wherein the botanical oil is synthesized. 6. The wipe of claim 1, wherein the protein is a plant protein. 7. The wipe of claim 6, wherein the plant protein is zein, corn gluten, wheat gluten, whey protein, soy protein, or a combination thereof. 8. The wipe of claim 6, wherein the plant protein is a soy protein. 9. The wipe of claim 1, wherein the antimicrobial composition further comprises an organopolysiloxane. 10. The wipe of claim 9, wherein the organopolysiloxane is a methylpolysiloxane. 11. The wipe of claim 10, wherein the methylpolysiloxane is polydimethylsiloxane. 12. The wipe of claim 9, wherein the organopolysiloxane contains a hydroxyl functional group, alkoxy functional group, amino functional group, or a combination thereof. 13. The wipe of claim 12, wherein the organopolysiloxane is hydroxy-terminated polydimethylsiloxane. 14. The wipe of claim 1, wherein the antimicrobial composition has a solvent content of less than about 5 wt. %. 15. The wipe of claim 1, wherein the antimicrobial composition comprises botanical oils in an amount of from about 0.05 wt. % to about 50 wt. %. 16. The wipe of claim 1, wherein the web is a nonwoven web. 17. The wipe of claim 1, wherein the fibers include absorbent fibers. 18. The wipe of claim 17, wherein the web is a composite of absorbent fibers and synthetic thermoplastic fibers. 19. The wipe of claim 1, wherein the antimicrobial composition further comprises a carbohydrate polymer. 20. The wipe of claim 19, wherein the carbohydrate biopolymer includes a chemically modified starch polymer. 21. The wipe of claim 20, wherein the chemically modified starch polymer is a hydroxyalkyl starch. 22. A method for forming an antimicrobial wipe, the method comprising: forming a coating solution that comprises a botanical oil, protein, and a solvent; applying the coating solution to a web that includes a plurality of fibers; and drying the coating solution to form an antimicrobial coating on the web. 23. The method of claim 22, wherein the monoterpene phenol is thymol, carvacol, or a mixture thereof. 24. The method of claim 22, wherein the protein is a plant protein. 25. The method of claim 22, wherein the coating solution further comprises an organopolysiloxane emulsion. 26. The method of claim 22, wherein the emulsion includes polydimethylsiloxane, modified polydimethylsiloxane, or a combination thereof. 27. The method of claim 22, wherein the antimicrobial coating has a solvent content of less than about 5 wt. %. 28. A method for inhibiting the growth of bacteria on a surface, the method comprising contacting the surface with a wipe that includes a web comprising a plurality of fibers, wherein the web is coated with an antimicrobial composition, the antimicrobial composition comprising a botanical oil that is at least partially encapsulated by a film-forming protein. 29. The method of claim 28, wherein the composition is in the form of a concentrate. 30. The method of claim 29, wherein prior to contacting the surface with the wipe, water is applied to the concentrate to initiate dispersion of the biopolymer and release the botanical oil. 31. An antimicrobial concentrate having a solvent content of about 5 wt. % or less, wherein the concentrate comprises at least one monoterpene phenol in an amount of from about 0.05 wt. % to about 50 wt. %, at least one film-forming protein in an amount of from about 5 wt. % to about 50 wt. %, and at least one organopolysiloxane in an amount of from about 10 wt. % to about 70 wt. %, and further wherein the protein at least partially encapsulates the monoterpene phenol. 32. The antimicrobial concentrate of claim 31, wherein the monoterpene phenol includes thymol, carvacol, or a mixture thereof. 33. The antimicrobial concentrate of claim 31, wherein the concentrate comprises at least one monoterpene phenol in an amount of from about 1 wt. % to about 40 wt. %, at least one film-forming protein in an amount of from about 10 wt. % to about 40 wt. %, and at least one organopolysiloxane in an amount of from about 20 wt. % to about 60 wt. %. 34. The antimicrobial concentrate of claim 31, further comprising at least one carbohydrate polymer in an amount of from about 5 wt. % to about 50 wt. %.
1,600
258
258
14,912,013
1,612
An object of the present invention is to provide antimicrobial agents that do not contribute to the emergence of drug-resistant microorganisms, that present less side effects, and that can be combined with other medications while causing less adverse effects. The present invention provides antimicrobial agents containing at least one member selected from the group consisting of: at least one hydrogen isotope selected from the group consisting of hydrogen atom (H); 1 H, 2 H, 3 H, 4 H, 5 H, 6 H and 7 H; hydrogen molecule (H 2 ); a metal hydride; hydrogen ion (H + ); hydride ion (H − ); and atomic hydrogen. The antimicrobial agents of the present invention exhibit their antimicrobial action through the action of hydrogen, so they are less likely to induce emergence of drug-resistant microorganisms.
1-26. (canceled) 27. An antimicrobial agent comprising: at least one member selected from the group consisting of hydrogen atom (H); at least one hydrogen isotope selected from the group consisting of hydrogen atom (H); 1H, 2H, 3H, 4H, 5H, 6H and 7H; hydrogen molecule (H2); a metal hydride; hydrogen ion (H+); hydride ion (H−); and atomic hydrogen. 28. The antimicrobial agent according to claim 27, which contains a coral powder having the hydrogen molecule adsorbed thereon. 29. The antimicrobial agent according to claim 27, wherein the hydrogen molecule or metal hydride ionizes upon contact with water to generate a hydride ion. 30. The antimicrobial agent according to claim 27, wherein the metal hydride is a hydride of a metal of at least one species selected from Group 1, Group 2, Group 13 or Group 14 in the periodic table of elements. 31. The antimicrobial agent according to claim 30, wherein the metal hydride comprises calcium hydride. 32. The antimicrobial agent according to claim 27, which contains a reduction fired body of mollusk shell, livestock's bone, fish bone, calcified coral, coral calcium, calcium carbonate, silica, zeolite, or two or more combinations thereof. 33. The antimicrobial agent according to claim 27, wherein the fungus is a yeast-like fungus. 34. The antimicrobial agent according to claim 33, wherein the fungus is at least one yeast-like fungus selected from the group consisting of Candida albicans and Candida glabrata. 35. The antimicrobial agent according to claim 27, wherein the virus is at least one virus selected from the group consisting of viruses belonging to the family Orthomyxoviridae, viruses belonging to the family Paramyxoviridae, and viruses belonging to the family Adenoviridae. 36. The antimicrobial agent according to claim 35, wherein the virus is at least one virus selected from the group consisting of influenza viruses, RS viruses, and adenoviruses. 37. The antimicrobial agent according to claim 35, wherein the virus is an amantadine-resistant influenza virus or Tamiflu-resistant influenza virus. 38. The antimicrobial agent according to claim 27, which is in a gaseous form. 39. The antimicrobial agent according to claim 27, which is in an aerosol form. 40. The antimicrobial agent according to claim 27, which is in a liquid form. 41. The antimicrobial agent according to claim 27, which is in a solid form. 42. The antimicrobial agent according to claim 41, which is powdery. 43. The antimicrobial agent according to claim 42, which provides an approximate concentration of hydrogen when three grams of the antimicrobial agent is mixed with 20 milliliters of physiological saline. 44. A pharmaceutical composition containing the antimicrobial agent according to claim 27. 45. The pharmaceutical composition according to claim 44, which is to be used in combination with at least one other medication. 46. A microorganism control agent containing the antimicrobial agent according to claim 27. 47. A method for preventing or managing bacterial infection, comprising: administering to a subject in need thereof an effective amount of an antimicrobial agent comprising: a coral powder containing the hydrogen molecule adsorbed thereon. 48. The method according to claim 47, which is for preventing or managing infection with at least one bacterium belonging to Gram-negative bacilli, Gram-negative cocci, Gram-positive cocci, or Gram-positive bacilli. 49. A method for preventing or managing bacterial infection, comprising: administering to a subject in need thereof an effective amount of an antimicrobial agent comprising: a reduction fired body of mollusk shell, livestock's bone, fish bone, calcified coral, coral calcium, calcium carbonate, silica, zeolite, or two or more combinations thereof. 50. The method according to claim 49, wherein the bacterium is at least one bacterium selected from the group consisting of Escherichia coli, pathogenic Escherichia coli O157, Salmonella, Haemophilus influenzae, Vibrio parahaemolyticus, Enterococcus, Pneumococcus, Neisseria, Neisseria gonorrhoeae, Neisseria meningitidis, Staphylococcus aureus, Staphylococcus epidermidis, Group A Streptococcus, Group B Streptococcus, Group C/G Streptococcus, Listeria monocytogenes, Klebsiella pneumoniae, Shigella, Vibrio cholerae, B. cepacia, Citrobacter, and Serratia. 51. The method according to claim 49, wherein the bacterium is at least one bacterium selected from the group consisting of extended-spectrum β-lactamase (ESBL) producing Gram-negative bacilli, multidrug-resistant Pseudomonas aeruginosa (MDRP), New Delhi metallo-β-lactamase (NDM-1) producing Gram-negative bacilli, β-lactamase non-producing ampicillin-resistant (BLNAR) Haemophilus influenzae, methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), penicillin-resistant Streptococcus pneumoniae (PRSP), multidrug-resistant Acinetobacter (MDRA), Klebsiella pneumoniae carbapenemase producing bacterium (KPC), penicillinase-producing Neisseria gonorrhoeae (PPNG) and community-acquired infection type methicillin-resistant Staphylococcus aureus (CA-MRSA).
An object of the present invention is to provide antimicrobial agents that do not contribute to the emergence of drug-resistant microorganisms, that present less side effects, and that can be combined with other medications while causing less adverse effects. The present invention provides antimicrobial agents containing at least one member selected from the group consisting of: at least one hydrogen isotope selected from the group consisting of hydrogen atom (H); 1 H, 2 H, 3 H, 4 H, 5 H, 6 H and 7 H; hydrogen molecule (H 2 ); a metal hydride; hydrogen ion (H + ); hydride ion (H − ); and atomic hydrogen. The antimicrobial agents of the present invention exhibit their antimicrobial action through the action of hydrogen, so they are less likely to induce emergence of drug-resistant microorganisms.1-26. (canceled) 27. An antimicrobial agent comprising: at least one member selected from the group consisting of hydrogen atom (H); at least one hydrogen isotope selected from the group consisting of hydrogen atom (H); 1H, 2H, 3H, 4H, 5H, 6H and 7H; hydrogen molecule (H2); a metal hydride; hydrogen ion (H+); hydride ion (H−); and atomic hydrogen. 28. The antimicrobial agent according to claim 27, which contains a coral powder having the hydrogen molecule adsorbed thereon. 29. The antimicrobial agent according to claim 27, wherein the hydrogen molecule or metal hydride ionizes upon contact with water to generate a hydride ion. 30. The antimicrobial agent according to claim 27, wherein the metal hydride is a hydride of a metal of at least one species selected from Group 1, Group 2, Group 13 or Group 14 in the periodic table of elements. 31. The antimicrobial agent according to claim 30, wherein the metal hydride comprises calcium hydride. 32. The antimicrobial agent according to claim 27, which contains a reduction fired body of mollusk shell, livestock's bone, fish bone, calcified coral, coral calcium, calcium carbonate, silica, zeolite, or two or more combinations thereof. 33. The antimicrobial agent according to claim 27, wherein the fungus is a yeast-like fungus. 34. The antimicrobial agent according to claim 33, wherein the fungus is at least one yeast-like fungus selected from the group consisting of Candida albicans and Candida glabrata. 35. The antimicrobial agent according to claim 27, wherein the virus is at least one virus selected from the group consisting of viruses belonging to the family Orthomyxoviridae, viruses belonging to the family Paramyxoviridae, and viruses belonging to the family Adenoviridae. 36. The antimicrobial agent according to claim 35, wherein the virus is at least one virus selected from the group consisting of influenza viruses, RS viruses, and adenoviruses. 37. The antimicrobial agent according to claim 35, wherein the virus is an amantadine-resistant influenza virus or Tamiflu-resistant influenza virus. 38. The antimicrobial agent according to claim 27, which is in a gaseous form. 39. The antimicrobial agent according to claim 27, which is in an aerosol form. 40. The antimicrobial agent according to claim 27, which is in a liquid form. 41. The antimicrobial agent according to claim 27, which is in a solid form. 42. The antimicrobial agent according to claim 41, which is powdery. 43. The antimicrobial agent according to claim 42, which provides an approximate concentration of hydrogen when three grams of the antimicrobial agent is mixed with 20 milliliters of physiological saline. 44. A pharmaceutical composition containing the antimicrobial agent according to claim 27. 45. The pharmaceutical composition according to claim 44, which is to be used in combination with at least one other medication. 46. A microorganism control agent containing the antimicrobial agent according to claim 27. 47. A method for preventing or managing bacterial infection, comprising: administering to a subject in need thereof an effective amount of an antimicrobial agent comprising: a coral powder containing the hydrogen molecule adsorbed thereon. 48. The method according to claim 47, which is for preventing or managing infection with at least one bacterium belonging to Gram-negative bacilli, Gram-negative cocci, Gram-positive cocci, or Gram-positive bacilli. 49. A method for preventing or managing bacterial infection, comprising: administering to a subject in need thereof an effective amount of an antimicrobial agent comprising: a reduction fired body of mollusk shell, livestock's bone, fish bone, calcified coral, coral calcium, calcium carbonate, silica, zeolite, or two or more combinations thereof. 50. The method according to claim 49, wherein the bacterium is at least one bacterium selected from the group consisting of Escherichia coli, pathogenic Escherichia coli O157, Salmonella, Haemophilus influenzae, Vibrio parahaemolyticus, Enterococcus, Pneumococcus, Neisseria, Neisseria gonorrhoeae, Neisseria meningitidis, Staphylococcus aureus, Staphylococcus epidermidis, Group A Streptococcus, Group B Streptococcus, Group C/G Streptococcus, Listeria monocytogenes, Klebsiella pneumoniae, Shigella, Vibrio cholerae, B. cepacia, Citrobacter, and Serratia. 51. The method according to claim 49, wherein the bacterium is at least one bacterium selected from the group consisting of extended-spectrum β-lactamase (ESBL) producing Gram-negative bacilli, multidrug-resistant Pseudomonas aeruginosa (MDRP), New Delhi metallo-β-lactamase (NDM-1) producing Gram-negative bacilli, β-lactamase non-producing ampicillin-resistant (BLNAR) Haemophilus influenzae, methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), penicillin-resistant Streptococcus pneumoniae (PRSP), multidrug-resistant Acinetobacter (MDRA), Klebsiella pneumoniae carbapenemase producing bacterium (KPC), penicillinase-producing Neisseria gonorrhoeae (PPNG) and community-acquired infection type methicillin-resistant Staphylococcus aureus (CA-MRSA).
1,600
259
259
14,755,185
1,639
A method of treating mineral slurries to inhibit bacterial growth includes adding an effective amount of one or more of sodium N-methyldithiocarbamate or methylammonium monomethyldithiocarbamate to the mineral slurry to inhibit bacterial growth.
1. In a method of treating a mineral slurry to inhibit bacterial growth by adding an effective amount of a biocide to the mineral slurry, the improvement comprising adding an effective amount of one or more of sodium or potassium N-methyldithiocarbamate and methylammonium monomethyldithiocarbamate to the mineral slurry to inhibit bacterial growth. 2. The method claim 1, wherein the effective amount further comprises 0.04-0.45% by weight based on the total formulation. 3. The method of claim 1, wherein the mineral slurry further comprises a slurry containing water and one or more of a starch, a clay, calcium carbonate, or titanium dioxide. 4. The method of claim 3, wherein the clay is a kaolin clay and the calcium carbonate is a ground or precipitated calcium carbonate. 5. The method of claim 3, wherein the water is fresh water or reclaimed water. 6. The method of claim 1, further comprising adding an effective amount of an additional biocide selected from the group consisting of glutaraldehyde, 1,2 benzithiazolin-3-one, tetrahydro-3-5-methyl-2H-1,3,5-thiadizine-2-thione, isothiazoline, bronopol, 2,2-dibromo-3-nitrilopropionamide, or orthophenyl phenate. 7. In a method of treating water used in an oil field application to inhibit bacterial growth by adding an effective amount of a biocide to the water, the improvement comprising adding an effective amount of methylammonium monomethyldithiocarbamate to the water to inhibit bacterial growth. 8. The method claim 7, wherein the effective amount further comprises 200-4500 ppm weight based on the total formulation. 9. The method of claim 7, wherein the water further comprises oil drilling fluids, oil field waste water, drilling muds, workover or completion fluids, hydraulic fracturing fluids, slickwater fracturing fluids or borate crosslinked guar fluids. 10. In a method of treating adhesives, coatings, and high viscosity suspensions to inhibit bacterial growth by adding an effective amount of a biocide to the coatings, adhesives, and high viscosity suspensions, the improvement comprising adding an effective amount of one or more of sodium or potassium N-methyldithiocarbamate and methylammonium monomethyldithiocarbamate to one or more of the coatings, adhesives, and high viscosity suspensions to inhibit bacterial growth. 11. The method claim 10, wherein the effective amount further comprises 200-4500 ppm weight based on the total formulation. 12. The method of claim 10, further comprising adding an effective amount of an additional biocide to the one or more of the coatings, adhesives, and high viscosity suspensions, the biocide selected from the group consisting of glutaraldehyde, tetrahydro-3-5-methyl-2H-1,3,5-thiadizine-2-thione, 2,2-dibromo-3-nitrilopropionamide, bronopol, orthophenyl phenate, or tetra-kis-hydoxymethyly-phosphonium sulfate. 13. The method of claim 10, wherein the coatings comprise paper coatings. 14. The method of claim 10, wherein the adhesives comprise polyvinyl alcohol/polyvinyl acetate-based adhesives, starch based adhesives, and dextrin-based adhesives. 15. The method of claim 10, wherein the high viscosity suspensions comprise polymers and silica-polymer combinations. 16. The method of claim 7, further comprising adding an effective amount of an additional biocide selected from the group consisting of glutaraldehyde, 1,2 benzithiazolin-3-one, tetrahydro-3-5-methyl-2H-1,3,5-thiadizine-2-thione, isothiazoline, bronopol, 2,2-dibromo-3-nitrilopropionamide, or orthophenyl phenate. 17. The method of claim 7, wherein the water is fresh water or reclaimed water.
A method of treating mineral slurries to inhibit bacterial growth includes adding an effective amount of one or more of sodium N-methyldithiocarbamate or methylammonium monomethyldithiocarbamate to the mineral slurry to inhibit bacterial growth.1. In a method of treating a mineral slurry to inhibit bacterial growth by adding an effective amount of a biocide to the mineral slurry, the improvement comprising adding an effective amount of one or more of sodium or potassium N-methyldithiocarbamate and methylammonium monomethyldithiocarbamate to the mineral slurry to inhibit bacterial growth. 2. The method claim 1, wherein the effective amount further comprises 0.04-0.45% by weight based on the total formulation. 3. The method of claim 1, wherein the mineral slurry further comprises a slurry containing water and one or more of a starch, a clay, calcium carbonate, or titanium dioxide. 4. The method of claim 3, wherein the clay is a kaolin clay and the calcium carbonate is a ground or precipitated calcium carbonate. 5. The method of claim 3, wherein the water is fresh water or reclaimed water. 6. The method of claim 1, further comprising adding an effective amount of an additional biocide selected from the group consisting of glutaraldehyde, 1,2 benzithiazolin-3-one, tetrahydro-3-5-methyl-2H-1,3,5-thiadizine-2-thione, isothiazoline, bronopol, 2,2-dibromo-3-nitrilopropionamide, or orthophenyl phenate. 7. In a method of treating water used in an oil field application to inhibit bacterial growth by adding an effective amount of a biocide to the water, the improvement comprising adding an effective amount of methylammonium monomethyldithiocarbamate to the water to inhibit bacterial growth. 8. The method claim 7, wherein the effective amount further comprises 200-4500 ppm weight based on the total formulation. 9. The method of claim 7, wherein the water further comprises oil drilling fluids, oil field waste water, drilling muds, workover or completion fluids, hydraulic fracturing fluids, slickwater fracturing fluids or borate crosslinked guar fluids. 10. In a method of treating adhesives, coatings, and high viscosity suspensions to inhibit bacterial growth by adding an effective amount of a biocide to the coatings, adhesives, and high viscosity suspensions, the improvement comprising adding an effective amount of one or more of sodium or potassium N-methyldithiocarbamate and methylammonium monomethyldithiocarbamate to one or more of the coatings, adhesives, and high viscosity suspensions to inhibit bacterial growth. 11. The method claim 10, wherein the effective amount further comprises 200-4500 ppm weight based on the total formulation. 12. The method of claim 10, further comprising adding an effective amount of an additional biocide to the one or more of the coatings, adhesives, and high viscosity suspensions, the biocide selected from the group consisting of glutaraldehyde, tetrahydro-3-5-methyl-2H-1,3,5-thiadizine-2-thione, 2,2-dibromo-3-nitrilopropionamide, bronopol, orthophenyl phenate, or tetra-kis-hydoxymethyly-phosphonium sulfate. 13. The method of claim 10, wherein the coatings comprise paper coatings. 14. The method of claim 10, wherein the adhesives comprise polyvinyl alcohol/polyvinyl acetate-based adhesives, starch based adhesives, and dextrin-based adhesives. 15. The method of claim 10, wherein the high viscosity suspensions comprise polymers and silica-polymer combinations. 16. The method of claim 7, further comprising adding an effective amount of an additional biocide selected from the group consisting of glutaraldehyde, 1,2 benzithiazolin-3-one, tetrahydro-3-5-methyl-2H-1,3,5-thiadizine-2-thione, isothiazoline, bronopol, 2,2-dibromo-3-nitrilopropionamide, or orthophenyl phenate. 17. The method of claim 7, wherein the water is fresh water or reclaimed water.
1,600
260
260
13,446,594
1,628
The present disclosure comprises a natural method of feed supplement to beef cattle, with a maize extract obtained from the nixtamalization process with a high content of ferulic acid and its salts, for a period of 30 days in the final phase of the feedlot. Use of this dietary supplementation enables an increase in carcass yield and meat quality that is not negatively affected as with a commercial β-agonist. In addition to the above effects, the effective supplementation of ferulic acid to cattle is shown to have antioxidant effects on fresh meat during its shelf life, which can lead to great benefits for the industrial meat market.
1. A method for improving meat quality in beef cattle, which comprises administering a feed supplement comprising ferulic acid or a salt thereof in an amount from 100 to 250 mg. 2. The method according to claim 1, wherein said feed supplement comprises ferulic acid or a salt thereof in an amount from 150 to 200 mg. 3. The method according to claim 1, wherein said feed supplement is administered during the last phase of feedlot. 4. The method according to claim 3, wherein said feed supplement is administered for a period of 20 to 35 days. 5. The method according to claim 1, wherein said feed supplement increases carcass yield and improves the quality of the meat in terms of sensory tenderness and instrumental texture. 6. A food supplement for beef cattle comprising ferulic acid or a salt thereof in an amount from 100 to 250 mg. 7. The food supplement for beef cattle of claim 6, comprising ferulic acid or a salt thereof in an amount from 150 to 200 mg.
The present disclosure comprises a natural method of feed supplement to beef cattle, with a maize extract obtained from the nixtamalization process with a high content of ferulic acid and its salts, for a period of 30 days in the final phase of the feedlot. Use of this dietary supplementation enables an increase in carcass yield and meat quality that is not negatively affected as with a commercial β-agonist. In addition to the above effects, the effective supplementation of ferulic acid to cattle is shown to have antioxidant effects on fresh meat during its shelf life, which can lead to great benefits for the industrial meat market.1. A method for improving meat quality in beef cattle, which comprises administering a feed supplement comprising ferulic acid or a salt thereof in an amount from 100 to 250 mg. 2. The method according to claim 1, wherein said feed supplement comprises ferulic acid or a salt thereof in an amount from 150 to 200 mg. 3. The method according to claim 1, wherein said feed supplement is administered during the last phase of feedlot. 4. The method according to claim 3, wherein said feed supplement is administered for a period of 20 to 35 days. 5. The method according to claim 1, wherein said feed supplement increases carcass yield and improves the quality of the meat in terms of sensory tenderness and instrumental texture. 6. A food supplement for beef cattle comprising ferulic acid or a salt thereof in an amount from 100 to 250 mg. 7. The food supplement for beef cattle of claim 6, comprising ferulic acid or a salt thereof in an amount from 150 to 200 mg.
1,600
261
261
15,492,528
1,619
A topical mixture that produces nitric oxide and a method for using the topical mixture to increase the vasodilation of a bloodstream via transdermal absorption of the nitric oxide. The nitric oxide can then affect subcutaneous tissues. The systemic vasodilation of a mammal may be increased via a topical application of an appropriate nitric oxide producing substance.
1. A method for increasing vasodilation comprising: providing a mammal with an epidermal surface and a bloodstream having an initial systemic vasodilation of the bloodstream; providing a first medium comprising a nitrite compound; providing a second medium comprising at least two reducing agents; creating a mixture of the first medium and the second medium thereby initiating production of nitric oxide; applying the mixture onto a first location of the epidermal surface of the mammal; absorbing at least 500 ppm of nitric oxide transdermally within 10 minutes after the applying; and increasing the systemic vasodilation of the bloodstream at a second, untreated location of the mammal as compared to the initial systemic vasodilation. 2. The method of claim 1, wherein the epidermal surface is healthy skin. 3. The method of claim 1, wherein the mixture includes a salt buffer concentration for prolonging the production of nitric oxide, wherein the salt used in the salt buffer includes at least one salt selected from a group consisting of sodium chloride and calcium chloride. 4. The method of claim 1, further comprising treating the mammal for peripheral artery disease. 5. The method of claim 1, further comprising covering the epidermal surface with a barrier after applying the mixture. 6. The method of claim 1, wherein the first medium includes at least one nitrite selected from a group consisting of sodium nitrite and potassium nitrite. 7. The method of claim 1, wherein the second medium includes at least two reducing agents selected from a group consisting of citric acid, ascorbic acid, phytic acid, acetic acid, formic acid, and lactic acid. 8. The method of claim 1, wherein the first and second mediums further include a thickening agent derived from cellulose in an amount between 0.80% and 0.120%. 9. The method of claim 1, wherein the first medium further comprises sodium bicarbonate. 10. A method for treating systemic hemodynamic disorders comprising: providing a mammal with an epidermal surface and a bloodstream having a systemic hemodynamic disorder; providing a first medium comprising a nitrite compound and sodium bicarbonate; providing a second medium comprising a reducing agent; creating a mixture of the first medium and the second medium thereby initiating production of nitric oxide; applying the mixture onto a first location of the epidermal surface of the mammal; and increasing systemic vasodilation of the bloodstream at a second, unapplied location of the mammal to treat the systemic hemodynamic disorder. 11. The method of claim 10, wherein the systemic hemodynamic disorder is peripheral artery disease. 12. The method of claim 10, wherein the second medium includes at least two reducing agents selected from a group consisting of citric acid, ascorbic acid, phytic acid, acetic acid, formic acid, and lactic acid. 13. The method of claim 10, wherein the mixture includes a salt buffer concentration for prolonging the production of nitric oxide, wherein the salt used in the salt buffer includes at least one salt selected from a group consisting of sodium chloride and calcium chloride. 14. The method of claim 10, further comprising repeating the previous steps within three days. 15. A method for increasing vasodilation comprising: providing a mammal with an epidermal surface and a bloodstream having an initial systemic vasodilation of the bloodstream; providing a first medium comprising a nitrite compound; providing a second medium comprising at least two reducing agents; creating a mixture of the first medium and the second medium thereby initiating production of nitric oxide, wherein the mixture also includes a salt buffer for prolonging the production of nitric oxide, wherein the salt used in the salt buffer includes at least one salt selected from a group consisting of sodium chloride and calcium chloride; treating, by applying the mixture onto a first location of the epidermal surface of the mammal; absorbing an effective amount of nitric oxide transdermally; and increasing the systemic vasodilation of the bloodstream at a second, untreated location of the mammal as compared to the initial systemic vasodilation. 16. The method of claim 15, wherein the epidermal surface is healthy skin. 17. The method of claim 15, further comprising covering the epidermal surface with a barrier after applying the mixture. 18. The method of claim 15, wherein the first medium further comprises sodium bicarbonate. 19. The method of claim 15, wherein the first and second mediums further include a thickening agent derived from cellulose in an amount between 0.80% and 0.120%.
A topical mixture that produces nitric oxide and a method for using the topical mixture to increase the vasodilation of a bloodstream via transdermal absorption of the nitric oxide. The nitric oxide can then affect subcutaneous tissues. The systemic vasodilation of a mammal may be increased via a topical application of an appropriate nitric oxide producing substance.1. A method for increasing vasodilation comprising: providing a mammal with an epidermal surface and a bloodstream having an initial systemic vasodilation of the bloodstream; providing a first medium comprising a nitrite compound; providing a second medium comprising at least two reducing agents; creating a mixture of the first medium and the second medium thereby initiating production of nitric oxide; applying the mixture onto a first location of the epidermal surface of the mammal; absorbing at least 500 ppm of nitric oxide transdermally within 10 minutes after the applying; and increasing the systemic vasodilation of the bloodstream at a second, untreated location of the mammal as compared to the initial systemic vasodilation. 2. The method of claim 1, wherein the epidermal surface is healthy skin. 3. The method of claim 1, wherein the mixture includes a salt buffer concentration for prolonging the production of nitric oxide, wherein the salt used in the salt buffer includes at least one salt selected from a group consisting of sodium chloride and calcium chloride. 4. The method of claim 1, further comprising treating the mammal for peripheral artery disease. 5. The method of claim 1, further comprising covering the epidermal surface with a barrier after applying the mixture. 6. The method of claim 1, wherein the first medium includes at least one nitrite selected from a group consisting of sodium nitrite and potassium nitrite. 7. The method of claim 1, wherein the second medium includes at least two reducing agents selected from a group consisting of citric acid, ascorbic acid, phytic acid, acetic acid, formic acid, and lactic acid. 8. The method of claim 1, wherein the first and second mediums further include a thickening agent derived from cellulose in an amount between 0.80% and 0.120%. 9. The method of claim 1, wherein the first medium further comprises sodium bicarbonate. 10. A method for treating systemic hemodynamic disorders comprising: providing a mammal with an epidermal surface and a bloodstream having a systemic hemodynamic disorder; providing a first medium comprising a nitrite compound and sodium bicarbonate; providing a second medium comprising a reducing agent; creating a mixture of the first medium and the second medium thereby initiating production of nitric oxide; applying the mixture onto a first location of the epidermal surface of the mammal; and increasing systemic vasodilation of the bloodstream at a second, unapplied location of the mammal to treat the systemic hemodynamic disorder. 11. The method of claim 10, wherein the systemic hemodynamic disorder is peripheral artery disease. 12. The method of claim 10, wherein the second medium includes at least two reducing agents selected from a group consisting of citric acid, ascorbic acid, phytic acid, acetic acid, formic acid, and lactic acid. 13. The method of claim 10, wherein the mixture includes a salt buffer concentration for prolonging the production of nitric oxide, wherein the salt used in the salt buffer includes at least one salt selected from a group consisting of sodium chloride and calcium chloride. 14. The method of claim 10, further comprising repeating the previous steps within three days. 15. A method for increasing vasodilation comprising: providing a mammal with an epidermal surface and a bloodstream having an initial systemic vasodilation of the bloodstream; providing a first medium comprising a nitrite compound; providing a second medium comprising at least two reducing agents; creating a mixture of the first medium and the second medium thereby initiating production of nitric oxide, wherein the mixture also includes a salt buffer for prolonging the production of nitric oxide, wherein the salt used in the salt buffer includes at least one salt selected from a group consisting of sodium chloride and calcium chloride; treating, by applying the mixture onto a first location of the epidermal surface of the mammal; absorbing an effective amount of nitric oxide transdermally; and increasing the systemic vasodilation of the bloodstream at a second, untreated location of the mammal as compared to the initial systemic vasodilation. 16. The method of claim 15, wherein the epidermal surface is healthy skin. 17. The method of claim 15, further comprising covering the epidermal surface with a barrier after applying the mixture. 18. The method of claim 15, wherein the first medium further comprises sodium bicarbonate. 19. The method of claim 15, wherein the first and second mediums further include a thickening agent derived from cellulose in an amount between 0.80% and 0.120%.
1,600
262
262
14,352,740
1,653
The present invention provides, among other things methods of increasing cell density, viability and/or titer in a cell culture including steps of adding a composition comprising iron to the cell culture.
1. A method of increasing cell density, viability, and/or titer in a cell culture medium comprising steps of: (a) providing cells in a cell culture medium to start a cell culture process; and (b) adding a composition comprising iron to said cell culture medium during the cell culture process such that the concentration of iron in the cell culture medium is increased over the course of the cell culture process. 2. The method of claim 1, wherein the composition comprising iron is selected from the group consisting of FeSO4, Fe-citrate, Fe-transferrin, Fe-chloride, Fe-nitrate, Fe-EDTA, Fe(NO3)3, FeCl2, FeCl3 and combinations thereof. 3. The method of claim 1, wherein the composition comprising iron is added after day 0 of the cell culture process. 4. The method of claim 1, wherein the composition comprising iron is added on or after day 1 of the cell culture process. 5. The method of claim 1, wherein the composition comprising iron is added on or after day 3 of the cell culture process 6. (canceled) 7. The method of claim 1, wherein the composition comprising iron is added at multiple time points during the cell culture process. 8. The method of claim 1, wherein the concentration of iron in the cell culture medium after addition of the composition comprising iron ranges between 100 μM and 5 mM. 9. The method of claim 1, wherein the concentration of iron in the cell culture medium after addition of the composition comprising iron ranges between 300 μM and 1 mM. 10. (canceled) 11. The method of claim 1, wherein the cells are mammalian cells. 12. (canceled) 13. The method of claim 11, wherein the mammalian cells are CHO cells. 14-16. (canceled) 17. The method of claim 1, wherein the cell culture process is a large-scale production culture process. 18. The method of claim 17, wherein the volume of the cell culture medium is at least about 500 L. 19-20. (canceled) 21. The method of claim 1, wherein the cells carry a gene that encodes a recombinant protein. 22. The method of claim 21, wherein the recombinant protein is an antibody or fragment thereof. 23-30. (canceled) 31. The method of claim 1, wherein the method further comprises purifying the recombinant protein. 32. (canceled) 33. A method of preventing or delaying cell death in a cell culture, the method comprising a step of adding a composition comprising iron at one or more time points subsequent to the beginning of the cell culture process. 34. A method of inhibiting apoptosis in a cell culture, the method comprising a step of adding a composition comprising iron at one or more time points subsequent to the beginning of the cell culture process. 35. The method of claim 33, wherein the composition comprising iron is added after day 0. 36. The method of claim 33, wherein the composition comprising iron is added after day 3. 37. (canceled) 38. The method of claim 33, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 100 μM and 5 mM. 39. The method of claim 33, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 300 μM and 1 mM. 40-43. (canceled) 44. The method of claim 33, wherein the cell culture is a large-scale production culture. 45. The method of claim 44, wherein the volume of the cell culture is at least about 500 L. 46. The method of claim 33, wherein the composition comprising iron is selected from the group consisting of FeSO4, Fe-citrate, Fe-transferrin, Fe-chloride, Fe-nitrate, Fe-EDTA, Fe(NO3)3, FeCl2, FeCl3 and combinations thereof. 47-54. (canceled) 55. The method of claim 34, wherein the composition comprising iron is added after day 0. 56. The method of claim 34, wherein the composition comprising iron is added after day 3. 57. The method of claim 34, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 100 μM and 5 mM. 58. The method of claim 34, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 300 μM and 1 mM. 59. The method of claim 34, wherein the cell culture is a large-scale production culture. 60. The method of claim 59, wherein the volume of the cell culture is at least about 500 L. 61. The method of claim 34, wherein the composition comprising iron is selected from the group consisting of FeSO4, Fe-citrate, Fe-transferrin, Fe-chloride, Fe-nitrate, Fe-EDTA, Fe(NO3)3, FeCl2, FeCl3 and combinations thereof.
The present invention provides, among other things methods of increasing cell density, viability and/or titer in a cell culture including steps of adding a composition comprising iron to the cell culture.1. A method of increasing cell density, viability, and/or titer in a cell culture medium comprising steps of: (a) providing cells in a cell culture medium to start a cell culture process; and (b) adding a composition comprising iron to said cell culture medium during the cell culture process such that the concentration of iron in the cell culture medium is increased over the course of the cell culture process. 2. The method of claim 1, wherein the composition comprising iron is selected from the group consisting of FeSO4, Fe-citrate, Fe-transferrin, Fe-chloride, Fe-nitrate, Fe-EDTA, Fe(NO3)3, FeCl2, FeCl3 and combinations thereof. 3. The method of claim 1, wherein the composition comprising iron is added after day 0 of the cell culture process. 4. The method of claim 1, wherein the composition comprising iron is added on or after day 1 of the cell culture process. 5. The method of claim 1, wherein the composition comprising iron is added on or after day 3 of the cell culture process 6. (canceled) 7. The method of claim 1, wherein the composition comprising iron is added at multiple time points during the cell culture process. 8. The method of claim 1, wherein the concentration of iron in the cell culture medium after addition of the composition comprising iron ranges between 100 μM and 5 mM. 9. The method of claim 1, wherein the concentration of iron in the cell culture medium after addition of the composition comprising iron ranges between 300 μM and 1 mM. 10. (canceled) 11. The method of claim 1, wherein the cells are mammalian cells. 12. (canceled) 13. The method of claim 11, wherein the mammalian cells are CHO cells. 14-16. (canceled) 17. The method of claim 1, wherein the cell culture process is a large-scale production culture process. 18. The method of claim 17, wherein the volume of the cell culture medium is at least about 500 L. 19-20. (canceled) 21. The method of claim 1, wherein the cells carry a gene that encodes a recombinant protein. 22. The method of claim 21, wherein the recombinant protein is an antibody or fragment thereof. 23-30. (canceled) 31. The method of claim 1, wherein the method further comprises purifying the recombinant protein. 32. (canceled) 33. A method of preventing or delaying cell death in a cell culture, the method comprising a step of adding a composition comprising iron at one or more time points subsequent to the beginning of the cell culture process. 34. A method of inhibiting apoptosis in a cell culture, the method comprising a step of adding a composition comprising iron at one or more time points subsequent to the beginning of the cell culture process. 35. The method of claim 33, wherein the composition comprising iron is added after day 0. 36. The method of claim 33, wherein the composition comprising iron is added after day 3. 37. (canceled) 38. The method of claim 33, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 100 μM and 5 mM. 39. The method of claim 33, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 300 μM and 1 mM. 40-43. (canceled) 44. The method of claim 33, wherein the cell culture is a large-scale production culture. 45. The method of claim 44, wherein the volume of the cell culture is at least about 500 L. 46. The method of claim 33, wherein the composition comprising iron is selected from the group consisting of FeSO4, Fe-citrate, Fe-transferrin, Fe-chloride, Fe-nitrate, Fe-EDTA, Fe(NO3)3, FeCl2, FeCl3 and combinations thereof. 47-54. (canceled) 55. The method of claim 34, wherein the composition comprising iron is added after day 0. 56. The method of claim 34, wherein the composition comprising iron is added after day 3. 57. The method of claim 34, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 100 μM and 5 mM. 58. The method of claim 34, wherein the iron is added in an amount to effect a concentration in the cell culture medium ranging between 300 μM and 1 mM. 59. The method of claim 34, wherein the cell culture is a large-scale production culture. 60. The method of claim 59, wherein the volume of the cell culture is at least about 500 L. 61. The method of claim 34, wherein the composition comprising iron is selected from the group consisting of FeSO4, Fe-citrate, Fe-transferrin, Fe-chloride, Fe-nitrate, Fe-EDTA, Fe(NO3)3, FeCl2, FeCl3 and combinations thereof.
1,600
263
263
14,367,392
1,636
Human skin tissue sample methods and models for identifying and screening test agents as effective for providing skin tone benefits, methods for validating hypotheses for mechanisms driving skin pigmentation as well as methods for driving skin pigment levels in ex-vivo skin tissue. The method includes contacting a cultured human skin tissue sample with a test agent, generating a transcriptional profile from the sample, and comparing the results to a control to determine if the test agent is effective for providing a skin tone benefit.
1. A screening method for identifying a test agent as effective for providing a skin tone benefit, comprising: culturing a first human skin tissue sample, wherein the first human skin tissue sample comprises an epidermal layer and a dermal layer; contacting the human skin tissue sample with the test agent; generating a transcriptional profile from the first human skin tissue sample, wherein the transcriptional profile comprises data related to transcription of at least two genes selected from FIG. 10; and identifying the test agent as effective for providing a skin tone benefit when the at least two genes selected from FIG. 10 show a directionally appropriate increase or decrease in expression level in comparison to a control. 2. The screening method of claim 1, further comprising contacting a second human skin tissue sample with a benchmark tone agent to use as the control. 3. The screening method of claim 1 or claim 2, wherein at least one of the first and second human skin tissue samples is a human donor tissue sample. 4. The screening method of claim 3, further comprising removing a subcutaneous fat layer from the human donor tissue sample. 5. The screening method of any of claims 2 to 4, wherein the benchmark tone agent up regulates or down regulates, in the second human skin tissue sample, the expression level of at least 2 of the genes selected from FIG. 10 in a direction appropriate for providing a skin tone benefit. 6. The screening method of any preceding claim, wherein the human skin tissue sample is between a Fitzpatrick score of II and IV. 7. The screening method of any preceding claim, wherein the first human skin tissue sample is cultured at a temperature of from between 30° C. and 40° C. at a relative humidity of from 50% to 90% for a period of from 24 hours to 10 days. 8. The screening method of any preceding claim, wherein the first human tissue sample is stored from about 4° C. to about 10° C. prior to culturing. 9. The screening method of any preceding claim, further comprising placing the sample dermis-side down in an iso-osmotic solution to keep the dermis moist and the epidermis dry. 10. The screening method of any preceding claim, further comprising subjecting the first human skin tissue sample to one of an energy source or one or more compounds to mimic a skin tissue phenotype. 11. The screening method of any preceding claim, further comprising contacting the first human skin tissue sample with a paracrine agent either in a media or as applied topically. 12. The screening method of any preceding claim, wherein the test agent is at least one of Endothelin 1 and Stem Cell Factor. 13. The screening method of any preceding claim, further comprising formulating a cosmetic skin care composition comprising the test agent. 14. The screening method of any preceding claim, wherein the screening method is part of a tiered assay in which the test agent is subjected to at least one of an enzyme assay, a cell culture assay, and a skin equivalent assay prior to applying the test agent to the first human skin tissue sample. 15. The screening method of any preceding claim, further comprising changing a pigment level in an ex-vivo human skin tissue samples by the contacting the ex-vivo human skin tissue sample with at least one agent selected from the group consisting of Endothelin 1, Stem Cell Factor, Melanocyte Stimulating Hormone, Dobutamine, Forskolin, and ultraviolet light.
Human skin tissue sample methods and models for identifying and screening test agents as effective for providing skin tone benefits, methods for validating hypotheses for mechanisms driving skin pigmentation as well as methods for driving skin pigment levels in ex-vivo skin tissue. The method includes contacting a cultured human skin tissue sample with a test agent, generating a transcriptional profile from the sample, and comparing the results to a control to determine if the test agent is effective for providing a skin tone benefit.1. A screening method for identifying a test agent as effective for providing a skin tone benefit, comprising: culturing a first human skin tissue sample, wherein the first human skin tissue sample comprises an epidermal layer and a dermal layer; contacting the human skin tissue sample with the test agent; generating a transcriptional profile from the first human skin tissue sample, wherein the transcriptional profile comprises data related to transcription of at least two genes selected from FIG. 10; and identifying the test agent as effective for providing a skin tone benefit when the at least two genes selected from FIG. 10 show a directionally appropriate increase or decrease in expression level in comparison to a control. 2. The screening method of claim 1, further comprising contacting a second human skin tissue sample with a benchmark tone agent to use as the control. 3. The screening method of claim 1 or claim 2, wherein at least one of the first and second human skin tissue samples is a human donor tissue sample. 4. The screening method of claim 3, further comprising removing a subcutaneous fat layer from the human donor tissue sample. 5. The screening method of any of claims 2 to 4, wherein the benchmark tone agent up regulates or down regulates, in the second human skin tissue sample, the expression level of at least 2 of the genes selected from FIG. 10 in a direction appropriate for providing a skin tone benefit. 6. The screening method of any preceding claim, wherein the human skin tissue sample is between a Fitzpatrick score of II and IV. 7. The screening method of any preceding claim, wherein the first human skin tissue sample is cultured at a temperature of from between 30° C. and 40° C. at a relative humidity of from 50% to 90% for a period of from 24 hours to 10 days. 8. The screening method of any preceding claim, wherein the first human tissue sample is stored from about 4° C. to about 10° C. prior to culturing. 9. The screening method of any preceding claim, further comprising placing the sample dermis-side down in an iso-osmotic solution to keep the dermis moist and the epidermis dry. 10. The screening method of any preceding claim, further comprising subjecting the first human skin tissue sample to one of an energy source or one or more compounds to mimic a skin tissue phenotype. 11. The screening method of any preceding claim, further comprising contacting the first human skin tissue sample with a paracrine agent either in a media or as applied topically. 12. The screening method of any preceding claim, wherein the test agent is at least one of Endothelin 1 and Stem Cell Factor. 13. The screening method of any preceding claim, further comprising formulating a cosmetic skin care composition comprising the test agent. 14. The screening method of any preceding claim, wherein the screening method is part of a tiered assay in which the test agent is subjected to at least one of an enzyme assay, a cell culture assay, and a skin equivalent assay prior to applying the test agent to the first human skin tissue sample. 15. The screening method of any preceding claim, further comprising changing a pigment level in an ex-vivo human skin tissue samples by the contacting the ex-vivo human skin tissue sample with at least one agent selected from the group consisting of Endothelin 1, Stem Cell Factor, Melanocyte Stimulating Hormone, Dobutamine, Forskolin, and ultraviolet light.
1,600
264
264
14,345,695
1,627
Methods and compositions for treating acute suicidality by administration of buprenorphine are disclosed herein. An exemplary method is effected by administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, whereby the administration is ceased once the subject is no longer determined as having acute suicidality. The first therapeutically effective amount is less than 0.2 mg per day. A higher therapeutically effective amount may optionally be administered if the subject is determined as not fully responsive to the first therapeutically effective amount. Unit dosage forms of buprenorphine, comprising less than 0.2 mg (e.g., 0.1 mg) buprenorphine are also disclosed.
1-53. (canceled) 54. A method of treating acute suicidality in a subject in need thereof, the method being effected by administering to a subject determined as having acute suicidality a therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, thereby treating said acute suicidality in a subject in need thereof, wherein said subject is not afflicted by a depressive disorder. 55. The method of claim 54, wherein said subject is not afflicted by a borderline personality disorder. 56. The method of claim 54, comprising: (a) determining a presence of acute suicidality in a subject; and (b) administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during a first time period, said first therapeutically effective amount being less than 0.2 mg per day, and said first time period ranging from 3 days to 3 weeks; and (c) following said first time period, determining a responsiveness of the subject to said first therapeutically effective amount, to thereby determine if the subject is not fully responsive to said first therapeutically effective amount; and (d) if the subject is determined as not fully responsive to said first therapeutically effective amount, administering to the subject, during a second time period, a second therapeutically effective amount of buprenorphine, said second therapeutically effective amount being higher than said first therapeutically effective amount. 57. The method of claim 56, further comprising, following said administering said second therapeutically effective amount of buprenorphine during said second time period: (e) determining a responsiveness of the subject to said second therapeutically effective amount, to thereby determine if the subject is not fully responsive to said second therapeutically effective amount; and (f) if the subject is determined as not fully responsive to said second therapeutically effective amount, administering to the subject, during a third time period, a third therapeutically effective amount of buprenorphine, said third therapeutically effective amount being higher than said second therapeutically effective amount. 58. The method of claim 54, comprising: determining a presence of acute suicidality in a subject; and administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during a first time period, said first therapeutically effective amount being less than 0.2 mg per day. 59. The method of claim 54, wherein administering buprenorphine is effected for a total time period that ranges from one week to four weeks. 60. The method of claim 54, wherein said subject is afflicted by a disorder selected from the group consisting of a personality disorder, a psychosis, a substance abuse disorder, an anxiety disorder, an eating disorder, an attention deficit disorder, a tic disorder, a gender dysphoria, a dissociative disorder, a somatoform disorder, an impulse control disorder and an adjustment disorder. 61. The method of claim 54, wherein said subject is afflicted by a disorder selected from the group consisting of anorexia nervosa, a posttraumatic stress disorder, an adjustment disorder, schizophrenia, a borderline personality disorder, a narcissistic personality disorder, an antisocial personality disorder, an intermittent explosive disorder, an attention deficit disorder, a tic disorder, a panic disorder, a body dysmorphic disorder, a dissociative identity disorder, a social anxiety disorder, a substance abuse disorder, a bipolar disorder, and a gender dysphoria. 62. The method of claim 54, wherein said determining comprises measuring suicidality on a scale selected from the group consisting of a Beck Suicidal Ideation (BSI) scale, a Suicide Probability Scale (SPS), a Columbia-Suicide Severity Rating Scale (C-SSRS), and an Overt Aggression Scale Modified (OAS-M). 63. The method of claim 54, wherein said administering is effected by a route selected from the group consisting of sublingual administration and transdermal administration. 64. The method of claim 54, wherein said first therapeutically effective amount is in a range of from 0.02 to 0.15 mg per day. 65. The method of claim 54, wherein said second therapeutically effective amount is 0.5 mg per day or less. 66. A method of treating acute suicidality in a subject in need thereof, the method comprising: determining a presence of acute suicidality in a subject; and administering to a subject determined as having acute suicidality about 0.1 mg of buprenorphine, or a pharmaceutically acceptable salt thereof, twice per day during a first time period, said first time period ranging from 3 days to 3 weeks; and administering to the subject about 0.2 mg of buprenorphine, or a pharmaceutically acceptable salt thereof, twice per day during a second time period, said second time period ranging from 3 days to 3 weeks; and optionally administering to the subject more than 0.4 mg/day of buprenorphine, or a pharmaceutically acceptable salt thereof, during a third time period, thereby treating said acute suicidality in a subject in need thereof. 67. The method of claim 66, wherein administering more than 0.4 mg/day of buprenorphine is effected by administering about 0.3 mg of buprenorphine, or a pharmaceutically acceptable salt thereof, twice per day. 68. The method of claim 66, wherein administering buprenorphine is effected for a total time period that ranges from one week to four weeks. 69. The method of claim 66, wherein said subject is afflicted by a disorder selected from the group consisting of a mood disorder, a personality disorder, a psychosis, a substance abuse disorder, an anxiety disorder, an eating disorder, an attention deficit disorder, a tic disorder, a gender dysphoria, a dissociative disorder, a somatoform disorder, an impulse control disorder and an adjustment disorder. 70. The method of claim 66, wherein said subject is afflicted by a disorder selected from the group consisting of a major depressive disorder, anorexia nervosa, a posttraumatic stress disorder, an adjustment disorder, schizophrenia, a borderline personality disorder, a narcissistic personality disorder, an antisocial personality disorder, an intermittent explosive disorder, an attention deficit disorder, a tic disorder, a panic disorder, a body dysmorphic disorder, a dissociative identity disorder, a social anxiety disorder, a substance abuse disorder, a bipolar disorder, and a gender dysphoria. 71. The method of claim 66, wherein said subject is not afflicted by a depressive disorder. 72. The method of claim 66, wherein said subject is not afflicted by a borderline personality disorder. 73. The method of claim 66, wherein said determining comprises measuring suicidality on a scale selected from the group consisting of a Beck Suicidal Ideation (BSI) scale, a Suicide Probability Scale (SPS), a Columbia-Suicide Severity Rating Scale (C-SSRS), and an Overt Aggression Scale Modified (OAS-M). 74. The method of claim 66, wherein said administering is effected by a route selected from the group consisting of sublingual administration and transdermal administration. 75. A method of treating acute suicidality in a subject in need thereof, the method comprising: determining a presence of acute suicidality in a subject; and administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during a first time period, said first therapeutically effective amount being less than 0.2 mg per day, thereby treating said acute suicidality in a subject in need thereof. 76. The method of claim 75, comprising: (a) determining a presence of acute suicidality in a subject; and (b) administering to a subject determined as having acute suicidality said first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during said first time period, said first time period ranging from 3 days to 3 weeks; and (c) following said first time period, determining a responsiveness of the subject to said first therapeutically effective amount, to thereby determine if the subject is not fully responsive to said first therapeutically effective amount; and (d) if the subject is determined as not fully responsive to said first therapeutically effective amount, administering to the subject, during a second time period, a second therapeutically effective amount of buprenorphine, said second therapeutically effective amount being higher than said first therapeutically effective amount, thereby treating said acute suicidality in a subject in need thereof. 77. A pharmaceutical composition comprising buprenorphine, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, the composition being packaged in a packaging material and identified in print, in or on said packaging material, for use in the treatment of acute suicidality in a subject in need thereof. 78. The composition of claim 77, wherein said subject is not afflicted by a depressive disorder. 79. The composition of claim 77, wherein said subject is not afflicted by a borderline personality disorder. 80. The composition of claim 77, being formulated for an administration selected from the group consisting of sublingual administration and transdermal administration. 81. The composition of claim 77, comprising a unit dosage form, said unit dosage form comprising a therapeutically effective amount of said buprenorphine or a pharmaceutically acceptable salt thereof. 82. A pharmaceutical composition unit dosage form comprising buprenorphine, or a pharmaceutically acceptable salt thereof, in an amount of less than 0.2 mg buprenorphine. 83. The pharmaceutical composition unit dosage form of claim 82, wherein said amount is about 0.1 mg buprenorphine.
Methods and compositions for treating acute suicidality by administration of buprenorphine are disclosed herein. An exemplary method is effected by administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, whereby the administration is ceased once the subject is no longer determined as having acute suicidality. The first therapeutically effective amount is less than 0.2 mg per day. A higher therapeutically effective amount may optionally be administered if the subject is determined as not fully responsive to the first therapeutically effective amount. Unit dosage forms of buprenorphine, comprising less than 0.2 mg (e.g., 0.1 mg) buprenorphine are also disclosed.1-53. (canceled) 54. A method of treating acute suicidality in a subject in need thereof, the method being effected by administering to a subject determined as having acute suicidality a therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, thereby treating said acute suicidality in a subject in need thereof, wherein said subject is not afflicted by a depressive disorder. 55. The method of claim 54, wherein said subject is not afflicted by a borderline personality disorder. 56. The method of claim 54, comprising: (a) determining a presence of acute suicidality in a subject; and (b) administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during a first time period, said first therapeutically effective amount being less than 0.2 mg per day, and said first time period ranging from 3 days to 3 weeks; and (c) following said first time period, determining a responsiveness of the subject to said first therapeutically effective amount, to thereby determine if the subject is not fully responsive to said first therapeutically effective amount; and (d) if the subject is determined as not fully responsive to said first therapeutically effective amount, administering to the subject, during a second time period, a second therapeutically effective amount of buprenorphine, said second therapeutically effective amount being higher than said first therapeutically effective amount. 57. The method of claim 56, further comprising, following said administering said second therapeutically effective amount of buprenorphine during said second time period: (e) determining a responsiveness of the subject to said second therapeutically effective amount, to thereby determine if the subject is not fully responsive to said second therapeutically effective amount; and (f) if the subject is determined as not fully responsive to said second therapeutically effective amount, administering to the subject, during a third time period, a third therapeutically effective amount of buprenorphine, said third therapeutically effective amount being higher than said second therapeutically effective amount. 58. The method of claim 54, comprising: determining a presence of acute suicidality in a subject; and administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during a first time period, said first therapeutically effective amount being less than 0.2 mg per day. 59. The method of claim 54, wherein administering buprenorphine is effected for a total time period that ranges from one week to four weeks. 60. The method of claim 54, wherein said subject is afflicted by a disorder selected from the group consisting of a personality disorder, a psychosis, a substance abuse disorder, an anxiety disorder, an eating disorder, an attention deficit disorder, a tic disorder, a gender dysphoria, a dissociative disorder, a somatoform disorder, an impulse control disorder and an adjustment disorder. 61. The method of claim 54, wherein said subject is afflicted by a disorder selected from the group consisting of anorexia nervosa, a posttraumatic stress disorder, an adjustment disorder, schizophrenia, a borderline personality disorder, a narcissistic personality disorder, an antisocial personality disorder, an intermittent explosive disorder, an attention deficit disorder, a tic disorder, a panic disorder, a body dysmorphic disorder, a dissociative identity disorder, a social anxiety disorder, a substance abuse disorder, a bipolar disorder, and a gender dysphoria. 62. The method of claim 54, wherein said determining comprises measuring suicidality on a scale selected from the group consisting of a Beck Suicidal Ideation (BSI) scale, a Suicide Probability Scale (SPS), a Columbia-Suicide Severity Rating Scale (C-SSRS), and an Overt Aggression Scale Modified (OAS-M). 63. The method of claim 54, wherein said administering is effected by a route selected from the group consisting of sublingual administration and transdermal administration. 64. The method of claim 54, wherein said first therapeutically effective amount is in a range of from 0.02 to 0.15 mg per day. 65. The method of claim 54, wherein said second therapeutically effective amount is 0.5 mg per day or less. 66. A method of treating acute suicidality in a subject in need thereof, the method comprising: determining a presence of acute suicidality in a subject; and administering to a subject determined as having acute suicidality about 0.1 mg of buprenorphine, or a pharmaceutically acceptable salt thereof, twice per day during a first time period, said first time period ranging from 3 days to 3 weeks; and administering to the subject about 0.2 mg of buprenorphine, or a pharmaceutically acceptable salt thereof, twice per day during a second time period, said second time period ranging from 3 days to 3 weeks; and optionally administering to the subject more than 0.4 mg/day of buprenorphine, or a pharmaceutically acceptable salt thereof, during a third time period, thereby treating said acute suicidality in a subject in need thereof. 67. The method of claim 66, wherein administering more than 0.4 mg/day of buprenorphine is effected by administering about 0.3 mg of buprenorphine, or a pharmaceutically acceptable salt thereof, twice per day. 68. The method of claim 66, wherein administering buprenorphine is effected for a total time period that ranges from one week to four weeks. 69. The method of claim 66, wherein said subject is afflicted by a disorder selected from the group consisting of a mood disorder, a personality disorder, a psychosis, a substance abuse disorder, an anxiety disorder, an eating disorder, an attention deficit disorder, a tic disorder, a gender dysphoria, a dissociative disorder, a somatoform disorder, an impulse control disorder and an adjustment disorder. 70. The method of claim 66, wherein said subject is afflicted by a disorder selected from the group consisting of a major depressive disorder, anorexia nervosa, a posttraumatic stress disorder, an adjustment disorder, schizophrenia, a borderline personality disorder, a narcissistic personality disorder, an antisocial personality disorder, an intermittent explosive disorder, an attention deficit disorder, a tic disorder, a panic disorder, a body dysmorphic disorder, a dissociative identity disorder, a social anxiety disorder, a substance abuse disorder, a bipolar disorder, and a gender dysphoria. 71. The method of claim 66, wherein said subject is not afflicted by a depressive disorder. 72. The method of claim 66, wherein said subject is not afflicted by a borderline personality disorder. 73. The method of claim 66, wherein said determining comprises measuring suicidality on a scale selected from the group consisting of a Beck Suicidal Ideation (BSI) scale, a Suicide Probability Scale (SPS), a Columbia-Suicide Severity Rating Scale (C-SSRS), and an Overt Aggression Scale Modified (OAS-M). 74. The method of claim 66, wherein said administering is effected by a route selected from the group consisting of sublingual administration and transdermal administration. 75. A method of treating acute suicidality in a subject in need thereof, the method comprising: determining a presence of acute suicidality in a subject; and administering to a subject determined as having acute suicidality a first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during a first time period, said first therapeutically effective amount being less than 0.2 mg per day, thereby treating said acute suicidality in a subject in need thereof. 76. The method of claim 75, comprising: (a) determining a presence of acute suicidality in a subject; and (b) administering to a subject determined as having acute suicidality said first therapeutically effective amount of buprenorphine, or a pharmaceutically acceptable salt thereof, during said first time period, said first time period ranging from 3 days to 3 weeks; and (c) following said first time period, determining a responsiveness of the subject to said first therapeutically effective amount, to thereby determine if the subject is not fully responsive to said first therapeutically effective amount; and (d) if the subject is determined as not fully responsive to said first therapeutically effective amount, administering to the subject, during a second time period, a second therapeutically effective amount of buprenorphine, said second therapeutically effective amount being higher than said first therapeutically effective amount, thereby treating said acute suicidality in a subject in need thereof. 77. A pharmaceutical composition comprising buprenorphine, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, the composition being packaged in a packaging material and identified in print, in or on said packaging material, for use in the treatment of acute suicidality in a subject in need thereof. 78. The composition of claim 77, wherein said subject is not afflicted by a depressive disorder. 79. The composition of claim 77, wherein said subject is not afflicted by a borderline personality disorder. 80. The composition of claim 77, being formulated for an administration selected from the group consisting of sublingual administration and transdermal administration. 81. The composition of claim 77, comprising a unit dosage form, said unit dosage form comprising a therapeutically effective amount of said buprenorphine or a pharmaceutically acceptable salt thereof. 82. A pharmaceutical composition unit dosage form comprising buprenorphine, or a pharmaceutically acceptable salt thereof, in an amount of less than 0.2 mg buprenorphine. 83. The pharmaceutical composition unit dosage form of claim 82, wherein said amount is about 0.1 mg buprenorphine.
1,600
265
265
14,363,538
1,653
In accordance with the purpose(s) of the present disclosure, as embodied and broadly described herein, embodiments of the present disclosure, in one aspect, relate to TMJ implantation materials and implants (e.g., temporomandi bular joint (TMJ) methods of making TMJ implantation materials and implants, methods of forming a TMJ implantation material or an implant, and the like.
1. A temporomandibular joint (TMJ) disc, comprising: a TMJ scaffold structure having a plurality of holes in the scaffold structure. 2. The TMJ disc of claim 1, wherein the TMJ scaffold structure includes one or more populations of cells, wherein the population of cells is selected from the group consisting of: a stem cell, an endothelial cell, a smooth muscle cell, a fibroblast, and a combination thereof. 3. The TMJ disc of claim 1, wherein the population of cells is a homogeneous population of cells. 4. The TMJ disc of claim 1, wherein the population of cells is a heterogenous population of cells. 5. The TMJ disc of claim 1, wherein a portion of the cells are located in the holes in the scaffold structure. 6. The TMJ disc of claim 5, wherein the holes can have a diameter of about 1 to 500 micrometers. 7. The TMJ disc of claim 5, wherein one or more pairs of holes are spaced by about 1 micrometer to 10 millimeters. 8. The TMJ disc of claim 5, wherein a portion of the holes are through the scaffold structure. 9. The TMJ disc of claim 5, wherein a portion of the holes are only partially through the scaffold structure. 10. The TMJ disc of claim 1, wherein the TMJ scaffold structure is a porcine TMJ scaffold structure. 11. A temporomandibular joint (TMJ) disc, comprising: a freeze-dried TMJ scaffold structure having a plurality of holes in the scaffold structure. 12. The TMJ disc of claim 11, wherein the holes can have a diameter of about 1 to 1000 micrometers. 13. The TMJ disc of claim 11, wherein one or more pairs of holes are spaced by about 1 micrometer to 10 millimeters. 14. The TMJ disc of claim 11, wherein a portion of the holes are through the scaffold structure. 15. The TMJ disc of claim 11, wherein a portion of the holes are only partially through the scaffold structure. 16. The TMJ disc of claim 11, wherein the freeze-dried TMJ scaffold structure is acellular. 17. The TMJ disc of claim 16, wherein the freeze-dried TMJ scaffold structure is a porcine freeze-dried TMJ scaffold structure. 18. A method of forming a temporomandibular joint (TMJ) disc, comprising the steps of: obtaining a precursor TMJ scaffold structure; decellularizing the precursor TMJ scaffold structure to produce a decellularized TMJ scaffold structure; freeze drying the decellularized TMJ scaffold structure to form a freeze-dried TMJ scaffold structure; and drilling a plurality of drilled holes in the decellularized TMJ scaffold structure. 19. The method of claim 18, further comprising: culturing the freeze-dried TMJ scaffold structure under conditions whereby the freeze-dried TMJ scaffold structure is colonized by a population of cells, wherein a portion of the cells are in the holes; and thawing the freeze-dried TMJ scaffold structure to form a TMJ scaffold structure. 20. The method of claim 19, wherein during the thawing process a portion of the cells enter the interior of the TMJ scaffold structure. 21. The method of claim 18, wherein the decellularized TMJ scaffold structure is substantially free of non-TMJ tissue. 22. The method of claim 18, wherein decellularizing includes contacting the precursor TMJ scaffold structure with sodium dodecyl sulphate or sodium chloride. 23. The method of claim 18, wherein the cells are selected from the group consisting of: a stem cell, an endothelial cell, a smooth muscle cell, a fibroblast, and a combination thereof. 24. The method of claim 18, wherein the population of cells is a homogeneous population of cells. 25. The method of claim 18, wherein the population of cells is a heterogenous population of cells. 26. The method of claim 18, wherein the holes can have a diameter of about 1 to 500 micrometers. 27. The method of claim 18, wherein one or more pairs of holes are spaced by about 1 micrometer to 10 millimeters. 28. The method of claim 18, wherein a portion of the holes are through the scaffold structure. 29. The method of claim 18, wherein a portion of the holes are only partially through the scaffold structure. 30. The method of claim 18, wherein the TMJ scaffold structure is a porcine TMJ scaffold structure.
In accordance with the purpose(s) of the present disclosure, as embodied and broadly described herein, embodiments of the present disclosure, in one aspect, relate to TMJ implantation materials and implants (e.g., temporomandi bular joint (TMJ) methods of making TMJ implantation materials and implants, methods of forming a TMJ implantation material or an implant, and the like.1. A temporomandibular joint (TMJ) disc, comprising: a TMJ scaffold structure having a plurality of holes in the scaffold structure. 2. The TMJ disc of claim 1, wherein the TMJ scaffold structure includes one or more populations of cells, wherein the population of cells is selected from the group consisting of: a stem cell, an endothelial cell, a smooth muscle cell, a fibroblast, and a combination thereof. 3. The TMJ disc of claim 1, wherein the population of cells is a homogeneous population of cells. 4. The TMJ disc of claim 1, wherein the population of cells is a heterogenous population of cells. 5. The TMJ disc of claim 1, wherein a portion of the cells are located in the holes in the scaffold structure. 6. The TMJ disc of claim 5, wherein the holes can have a diameter of about 1 to 500 micrometers. 7. The TMJ disc of claim 5, wherein one or more pairs of holes are spaced by about 1 micrometer to 10 millimeters. 8. The TMJ disc of claim 5, wherein a portion of the holes are through the scaffold structure. 9. The TMJ disc of claim 5, wherein a portion of the holes are only partially through the scaffold structure. 10. The TMJ disc of claim 1, wherein the TMJ scaffold structure is a porcine TMJ scaffold structure. 11. A temporomandibular joint (TMJ) disc, comprising: a freeze-dried TMJ scaffold structure having a plurality of holes in the scaffold structure. 12. The TMJ disc of claim 11, wherein the holes can have a diameter of about 1 to 1000 micrometers. 13. The TMJ disc of claim 11, wherein one or more pairs of holes are spaced by about 1 micrometer to 10 millimeters. 14. The TMJ disc of claim 11, wherein a portion of the holes are through the scaffold structure. 15. The TMJ disc of claim 11, wherein a portion of the holes are only partially through the scaffold structure. 16. The TMJ disc of claim 11, wherein the freeze-dried TMJ scaffold structure is acellular. 17. The TMJ disc of claim 16, wherein the freeze-dried TMJ scaffold structure is a porcine freeze-dried TMJ scaffold structure. 18. A method of forming a temporomandibular joint (TMJ) disc, comprising the steps of: obtaining a precursor TMJ scaffold structure; decellularizing the precursor TMJ scaffold structure to produce a decellularized TMJ scaffold structure; freeze drying the decellularized TMJ scaffold structure to form a freeze-dried TMJ scaffold structure; and drilling a plurality of drilled holes in the decellularized TMJ scaffold structure. 19. The method of claim 18, further comprising: culturing the freeze-dried TMJ scaffold structure under conditions whereby the freeze-dried TMJ scaffold structure is colonized by a population of cells, wherein a portion of the cells are in the holes; and thawing the freeze-dried TMJ scaffold structure to form a TMJ scaffold structure. 20. The method of claim 19, wherein during the thawing process a portion of the cells enter the interior of the TMJ scaffold structure. 21. The method of claim 18, wherein the decellularized TMJ scaffold structure is substantially free of non-TMJ tissue. 22. The method of claim 18, wherein decellularizing includes contacting the precursor TMJ scaffold structure with sodium dodecyl sulphate or sodium chloride. 23. The method of claim 18, wherein the cells are selected from the group consisting of: a stem cell, an endothelial cell, a smooth muscle cell, a fibroblast, and a combination thereof. 24. The method of claim 18, wherein the population of cells is a homogeneous population of cells. 25. The method of claim 18, wherein the population of cells is a heterogenous population of cells. 26. The method of claim 18, wherein the holes can have a diameter of about 1 to 500 micrometers. 27. The method of claim 18, wherein one or more pairs of holes are spaced by about 1 micrometer to 10 millimeters. 28. The method of claim 18, wherein a portion of the holes are through the scaffold structure. 29. The method of claim 18, wherein a portion of the holes are only partially through the scaffold structure. 30. The method of claim 18, wherein the TMJ scaffold structure is a porcine TMJ scaffold structure.
1,600
266
266
14,775,829
1,617
The present disclosure relates to controlling insect damage in a plant. In one embodiment, the methods include applying insecticide and at least one fertilizer in-furrow at planting. In particular examples, methods of controlling cutworms and/or rootworms in corn include applying tefluthrin and at least one pop-up fertilizer in-furrow at planting.
1. A method of controlling insect damage in a plant, the method comprising: applying, at planting ±5 days, a a pyrethroid insecticide and a pop-up fertilizer. 2. The method of claim 1, wherein the pyrethroid insecticide includes at least one of permethrin, cypermethrin, fenvalerate, esfenvalerate, deltamethrin, cyhalothrin, lambda-cyhalothrin, bifenthrin, fenpropathrin, cyfluthrin, tefluthrin, ethofenprox, natural pyrethrin, tetramethrin, s-bioallethrin, fenfluthrin, prallethrin and 5-benzyl-3-furylmethyl-(E)-(1R,3S)-2,2-dimethyl-3-(2-oxothiolan-3-ylidene-methyl)cyclopropane carboxylate. 3. The method of claim 2, wherein the pyrethroid insecticide includes tefluthrin. 4. The method of claim 3, wherein the tefluthrin is in the form of Force CS insecticide. 5. The method of claim 1, wherein the pop-up fertilizer is an N:P:K ratio fertilizer, wherein P is in the range of 12-40. 6. The method of claim 1, wherein the pop-up fertilizer is an N:P:K ratio fertilizer, wherein N is in the ratio range of 4 to 12; P is in the ratio range of 12-40; and K is in the ratio range of 1-10. 7. The method of claim 1, wherein the pyrethroid insecticide and the pop-up fertilizer are applied as an insecticidal composition. 8. The method of claim 1, further including forming the pyrethroid insecticide and the pop-up fertilizer into an insecticidal composition prior to applying the composition, wherein forming includes: depositing the pyrethroid insecticide, in liquid form, into a first reservoir; depositing the pop-up fertilizer, in liquid form, into a second reservoir; and pumping to a mixing chamber the pyrethroid insecticide and the fertilizer, thereby forming the insecticidal composition, prior to application. 9. The method of claim 8, wherein the pyrethroid insecticide further includes at least one phosphated arylphenol alkoxylate surfactant having an average degree of alkoxylation of from 4-8; and at least one alkylamine alkoxylate surfactant having an average degree of alkoxylation of from 2-12. 10. The method of claim 7, wherein the pyrethroid insecticide in the first reservoir is Force CS insecticide. 11. The method of claim 1, wherein the applying is on the same day as planting. 12. The method of claim 1, wherein the applying is in the same pass as planting. 13. The method of claim 1, wherein the applying is in-furrow. 14. The method of claim 1, wherein the insect damage is damage from an insect chosen from at least one of a cutworm, a seed maggot, a wireworm, a grub, and a rootworm. 15. The method of claim 1, wherein the plant is chosen from at least one of a corn plant, a cotton plant, a soybean, and a vegetable plant. 16. A method of controlling at least one of cutworms and rootworms in corn, the method comprising: applying, in-furrow at planting, an insecticide composition comprising tefluthrin; and a pop-up fertilizer having an N:P:K ratio, wherein N is in the ratio range of 4 to 12; P is in the ratio range of 12-40; and K is in the ratio range of 1-10. 17. The method of claim 16, wherein the insecticide composition is formed in the same pass as application, wherein forming includes pumping the tefluthrin from a first reservoir to a mixing chamber, and pumping the fertilizer from a second reservoir to a mixing chamber, thereby forming the insecticide composition. 18. The method of claim 1, wherein the pyrethroid (Al) is applied in the range of about 0.1 lb Al/acre to about 0.3 lb Al/acre. 19. The method of claim 1, wherein the pop-up fertilizer is applied in the range of about 3 lb nutrients/acre to about 50 lb nutrients/acre. 20. The method of claim 1, wherein the pyrethroid and the fertilizer nutrients are applied in a weight ratio in the range of 1:30 to 1:500.
The present disclosure relates to controlling insect damage in a plant. In one embodiment, the methods include applying insecticide and at least one fertilizer in-furrow at planting. In particular examples, methods of controlling cutworms and/or rootworms in corn include applying tefluthrin and at least one pop-up fertilizer in-furrow at planting.1. A method of controlling insect damage in a plant, the method comprising: applying, at planting ±5 days, a a pyrethroid insecticide and a pop-up fertilizer. 2. The method of claim 1, wherein the pyrethroid insecticide includes at least one of permethrin, cypermethrin, fenvalerate, esfenvalerate, deltamethrin, cyhalothrin, lambda-cyhalothrin, bifenthrin, fenpropathrin, cyfluthrin, tefluthrin, ethofenprox, natural pyrethrin, tetramethrin, s-bioallethrin, fenfluthrin, prallethrin and 5-benzyl-3-furylmethyl-(E)-(1R,3S)-2,2-dimethyl-3-(2-oxothiolan-3-ylidene-methyl)cyclopropane carboxylate. 3. The method of claim 2, wherein the pyrethroid insecticide includes tefluthrin. 4. The method of claim 3, wherein the tefluthrin is in the form of Force CS insecticide. 5. The method of claim 1, wherein the pop-up fertilizer is an N:P:K ratio fertilizer, wherein P is in the range of 12-40. 6. The method of claim 1, wherein the pop-up fertilizer is an N:P:K ratio fertilizer, wherein N is in the ratio range of 4 to 12; P is in the ratio range of 12-40; and K is in the ratio range of 1-10. 7. The method of claim 1, wherein the pyrethroid insecticide and the pop-up fertilizer are applied as an insecticidal composition. 8. The method of claim 1, further including forming the pyrethroid insecticide and the pop-up fertilizer into an insecticidal composition prior to applying the composition, wherein forming includes: depositing the pyrethroid insecticide, in liquid form, into a first reservoir; depositing the pop-up fertilizer, in liquid form, into a second reservoir; and pumping to a mixing chamber the pyrethroid insecticide and the fertilizer, thereby forming the insecticidal composition, prior to application. 9. The method of claim 8, wherein the pyrethroid insecticide further includes at least one phosphated arylphenol alkoxylate surfactant having an average degree of alkoxylation of from 4-8; and at least one alkylamine alkoxylate surfactant having an average degree of alkoxylation of from 2-12. 10. The method of claim 7, wherein the pyrethroid insecticide in the first reservoir is Force CS insecticide. 11. The method of claim 1, wherein the applying is on the same day as planting. 12. The method of claim 1, wherein the applying is in the same pass as planting. 13. The method of claim 1, wherein the applying is in-furrow. 14. The method of claim 1, wherein the insect damage is damage from an insect chosen from at least one of a cutworm, a seed maggot, a wireworm, a grub, and a rootworm. 15. The method of claim 1, wherein the plant is chosen from at least one of a corn plant, a cotton plant, a soybean, and a vegetable plant. 16. A method of controlling at least one of cutworms and rootworms in corn, the method comprising: applying, in-furrow at planting, an insecticide composition comprising tefluthrin; and a pop-up fertilizer having an N:P:K ratio, wherein N is in the ratio range of 4 to 12; P is in the ratio range of 12-40; and K is in the ratio range of 1-10. 17. The method of claim 16, wherein the insecticide composition is formed in the same pass as application, wherein forming includes pumping the tefluthrin from a first reservoir to a mixing chamber, and pumping the fertilizer from a second reservoir to a mixing chamber, thereby forming the insecticide composition. 18. The method of claim 1, wherein the pyrethroid (Al) is applied in the range of about 0.1 lb Al/acre to about 0.3 lb Al/acre. 19. The method of claim 1, wherein the pop-up fertilizer is applied in the range of about 3 lb nutrients/acre to about 50 lb nutrients/acre. 20. The method of claim 1, wherein the pyrethroid and the fertilizer nutrients are applied in a weight ratio in the range of 1:30 to 1:500.
1,600
267
267
14,082,662
1,617
An aqueous liquid composition comprising: (a) 2-amino-3-(4-bromobenzoyl)phenylacetic acid or its pharmacologically acceptable salt or hydrate; (b) a non-ionic surfactant; and (c) a preservative; wherein at least 90 percent of the original amount of said 2-amino-3-(4-bromobenzoyl)phenylacetic acid or pharmacologically acceptable salt or hydrate remains in the composition after storage at 60° C., for 4 weeks. The composition can satisfy the preservative efficacy of US Pharmacopeia 35 (2012). The composition is useful for the treatment of ocular or nasal inflammation of diverse etiology.
1. An aqueous liquid composition comprising: (a) 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof; (b) a non-ionic surfactant; and (c) water; wherein the non-ionic surfactant is selected from the group consisting of copolymers of polyoxyethylene and polyoxypropylene, fatty acid esters of polyethylene glycol, polyethylene glycol mono(alkylaryl)ethers, polyethoxylated triglycerides, polyethoxylated hydrogenated triglycerides, and mixtures thereof, and at least 90 percent of the original amount of said 2-amino-3-(4-bromobenzoyl)phenylacetic acid or pharmacologically acceptable salt thereof or hydrate thereof remains in the composition after storage at 60° C., for 4 weeks. 2. The composition of claim 1, further comprising an ophthalmically acceptable preservative selected from the group consisting of chlorhexidine, polyaminopropyl biguanide (“PAPB”), polyquaternium-1, polyquaternium-42, perborate, and mixtures thereof. 3. An aqueous liquid composition comprising: (a) 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof; (b) a non-ionic surfactant; (c) water; and (d) an ophthalmically acceptable preservative; wherein the non-ionic surfactant is a polyethylene glycol poly(alkylaryl)ether; and wherein the ophthalmically acceptable preservative is selected from the group consisting of chlorhexidine, polyaminopropyl biguanide (“PAPB”), polyquaternium-1, polyquaternium-42, perborate, and mixtures thereof. 4. The composition of claim 3, wherein the polyethylene glycol poly(alkylaryl)ether is tyloxapol. 5. The composition of claim 1, wherein the non-ionic surfactant is selected from the group consisting of poloxamer 407, polyoxyl 40 stearate, octoxynol 40, and polyethylene glycol 40 castor oil. 6. The composition of claim 1, wherein the concentration of the non-ionic surfactant is in the range from 0.005 w/v % to 0.12 w/v %. 7. The composition of claim 3, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 8. The composition of claim 1, wherein the composition does not include any polyol. 9. The composition of claim 8, wherein the polyol is mannitol. 10. A method for inhibiting decrease in preservative efficacy of a preservative in an aqueous composition that comprises 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof, which method comprises incorporating a non-ionic surfactant into an aqueous liquid preparation comprising 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof and a preservative. 11. The method of claim 10, wherein the preservative efficacy satisfies the preservative efficacy acceptance criteria of US Pharmacopeia 35 (2012). 12. The composition of claim 2, wherein the non-ionic surfactant is selected from the group consisting of poloxamer 407, polyoxyl 40 stearate, octoxynol 40, and polyethylene glycol 40 castor oil. 13. The composition of claim 3, wherein the concentration of the non-ionic surfactant is in the range from 0.005 w/v % to 0.12 w/v %. 14. The composition of claim 5, wherein the concentration of the non-ionic surfactant is in the range from 0.005 w/v % to 0.12 w/v %. 15. The composition of claim 1, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 16. The composition of claim 5, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 17. The composition of claim 12, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 18. The composition of claim 6, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 19. The composition of claim 2, wherein the composition does not include any polyol. 20. The composition of claim 3, wherein the composition does not include any polyol.
An aqueous liquid composition comprising: (a) 2-amino-3-(4-bromobenzoyl)phenylacetic acid or its pharmacologically acceptable salt or hydrate; (b) a non-ionic surfactant; and (c) a preservative; wherein at least 90 percent of the original amount of said 2-amino-3-(4-bromobenzoyl)phenylacetic acid or pharmacologically acceptable salt or hydrate remains in the composition after storage at 60° C., for 4 weeks. The composition can satisfy the preservative efficacy of US Pharmacopeia 35 (2012). The composition is useful for the treatment of ocular or nasal inflammation of diverse etiology.1. An aqueous liquid composition comprising: (a) 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof; (b) a non-ionic surfactant; and (c) water; wherein the non-ionic surfactant is selected from the group consisting of copolymers of polyoxyethylene and polyoxypropylene, fatty acid esters of polyethylene glycol, polyethylene glycol mono(alkylaryl)ethers, polyethoxylated triglycerides, polyethoxylated hydrogenated triglycerides, and mixtures thereof, and at least 90 percent of the original amount of said 2-amino-3-(4-bromobenzoyl)phenylacetic acid or pharmacologically acceptable salt thereof or hydrate thereof remains in the composition after storage at 60° C., for 4 weeks. 2. The composition of claim 1, further comprising an ophthalmically acceptable preservative selected from the group consisting of chlorhexidine, polyaminopropyl biguanide (“PAPB”), polyquaternium-1, polyquaternium-42, perborate, and mixtures thereof. 3. An aqueous liquid composition comprising: (a) 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof; (b) a non-ionic surfactant; (c) water; and (d) an ophthalmically acceptable preservative; wherein the non-ionic surfactant is a polyethylene glycol poly(alkylaryl)ether; and wherein the ophthalmically acceptable preservative is selected from the group consisting of chlorhexidine, polyaminopropyl biguanide (“PAPB”), polyquaternium-1, polyquaternium-42, perborate, and mixtures thereof. 4. The composition of claim 3, wherein the polyethylene glycol poly(alkylaryl)ether is tyloxapol. 5. The composition of claim 1, wherein the non-ionic surfactant is selected from the group consisting of poloxamer 407, polyoxyl 40 stearate, octoxynol 40, and polyethylene glycol 40 castor oil. 6. The composition of claim 1, wherein the concentration of the non-ionic surfactant is in the range from 0.005 w/v % to 0.12 w/v %. 7. The composition of claim 3, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 8. The composition of claim 1, wherein the composition does not include any polyol. 9. The composition of claim 8, wherein the polyol is mannitol. 10. A method for inhibiting decrease in preservative efficacy of a preservative in an aqueous composition that comprises 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof, which method comprises incorporating a non-ionic surfactant into an aqueous liquid preparation comprising 2-amino-3-(4-bromobenzoyl)phenylacetic acid or a pharmacologically acceptable salt thereof or a hydrate thereof and a preservative. 11. The method of claim 10, wherein the preservative efficacy satisfies the preservative efficacy acceptance criteria of US Pharmacopeia 35 (2012). 12. The composition of claim 2, wherein the non-ionic surfactant is selected from the group consisting of poloxamer 407, polyoxyl 40 stearate, octoxynol 40, and polyethylene glycol 40 castor oil. 13. The composition of claim 3, wherein the concentration of the non-ionic surfactant is in the range from 0.005 w/v % to 0.12 w/v %. 14. The composition of claim 5, wherein the concentration of the non-ionic surfactant is in the range from 0.005 w/v % to 0.12 w/v %. 15. The composition of claim 1, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 16. The composition of claim 5, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 17. The composition of claim 12, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 18. The composition of claim 6, wherein the pH of the aqueous liquid preparation is within a range of 7.3 to 8.5. 19. The composition of claim 2, wherein the composition does not include any polyol. 20. The composition of claim 3, wherein the composition does not include any polyol.
1,600
268
268
12,682,582
1,623
One or more complex lipids including gangliosides to achieve particular health benefits including maintaining or increasing cognitive development or maintaining or increasing growth in a foetal, infant or child subject.
1-28. (canceled) 29. A method for maintaining or increasing growth of a foetal, infant, or child subject, the method comprising administering one or more complex lipids to a foetal, infant, or child subject in need thereof, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 30. The method of claim 29, wherein the one or more complex lipids are administered to a mother during gestation and the growth is brain weight of a foetal subject or brain ganglioside content of a foetal subject. 31. The method of claim 29, wherein the one or more complex lipids are administered to an infant or child subject and the growth is one or more of body weight, body length, and bone mineral density. 32. A method for maintaining or increasing cognitive development of a foetal, infant, or child subject by administering a one or more complex lipids to a foetal, infant, or child subject in need thereof, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 33. The method of claim 32, wherein the one or more complex lipids are administered to a mother during gestation and the cognitive development is brain weight of a foetal subject or brain ganglioside content of a foetal subject. 34. A method for maintaining or increasing growth or maintaining or increasing cognitive development of a foetal subject, the method comprising providing a pregnant mother with one or more complex lipids and informing the mother that the one or more complex lipids will maintain or increase growth or maintain or increase cognitive development of the foetal subject, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 35. A method for maintaining or increasing growth or maintaining or increasing cognitive development in a subject, the method comprising providing a subject with one or more complex lipids and informing the subject that the one or more complex lipids will maintain or increase growth or maintain or increase cognitive development of the subject, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 36. The method of claim 29, wherein the one or more complex lipids comprises one or more phospholipids, one or more sphingolipids, one or more sphingomyelins or derivatives thereof, one or more ceramides, one or more cerebrosides, one or more gangliosides, or any combination of any two or more thereof. 37. The method of claim 36, wherein the one or more gangliosides comprises GM3, GD3, or a mixture of at least GM3 and GD3. 38. The method of claim 29, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg gangliosides per, 100 g or about 5 mg to about 20 mg gangliosides per 100 g. 39. (canceled) 40. The method of claim 29, wherein the one or more complex lipids comprises a milk fat extract. 41. The method of claim 40, wherein the milk fat extract comprises about 15% to about 99% by weight total lipid, about 1% to about 80% by weight phospholipid, about 1% to about 25% by weight phosphatidylcholine, about 0.1% to about 15% by weight phosphatidylinositol, about 0.1% to about 15% by weight phosphatidylserine, about 1% to about 30% by weight phosphatidylethanolamine, about 0.5% to about 25% by weight sphingomyelin, and about 0.1 to about 10% by weight ganglioside. 42. The method of claim 41, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg milk fat extract per 100 g, at least about 5 mg milk fat extract per 100 g, or at least about 7.5 mg milk fat extract per 100 g. 43. (canceled) 44. (canceled) 45. The method of claim 32, wherein the one or more complex lipids comprises one or more phospholipids, one or more sphingolipids, one or more sphingomyelins or derivatives thereof, one or more ceramides, one or more cerebrosides, one or more gangliosides, or any combination of any two or more thereof. 46. The method of claim 45, wherein the one or more gangliosides comprises GM3, GD3, or a mixture of at least GM3 and GD3. 47. The method of claim 32, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg gangliosides per 100 g or about 5 mg to about 20 mg gangliosides per 100 g. 48. The method of claim 29, wherein the one or more complex lipids comprises a milk fat extract. 49. The method of claim 48, wherein the milk fat extract comprises about 15% to about 99% by weight total lipid, about 1% to about 80% by weight phospholipid, about 1% to about 25% by weight phosphatidylcholine, about 0.1% to about 15% by weight phosphatidylinositol, about 0.1% to about 15% by weight phosphatidylserine, about 1% to about 30% by weight phosphatidylethanolamine, about 0.5% to about 25% by weight sphingomyelin, and about 0.1 to about 10% by weight ganglioside. 50. The method of claim 49, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg milk fat extract per 100 g, at least about 5 mg milk fat extract per 100 g, or at least about 7.5 mg milk fat extract per 100 g. 51. The method of claim 29, wherein the one or more complex lipids comprises (a) about 15 to about 25% w/w lipid, about 5 to about 15% w/w phospholipid, and about 0.1 to about 1% w/w ganglioside, or (b) about 15 to about 25% w/w lipid, about 5 to about 15% w/w phospholipid, about 1 to about 5% w/w phosphatidylcholine, about 0.1 to about 2% w/w phosphatidylinositol, about 0.5 to about 2% w/w phosphatidylserine, about 1.5 to about 6% w/w phosphatidylethanolamine, about 1 to about 5% w/w sphingomyelin, and about 0.1 to about 1% w/w ganglioside, or (c) about 25 to about 45% w/w lipid, about 10 to about 25% w/w phospholipid, and about 0.1 to about 2.0% w/w ganglioside, or (d) about 25 to about 45% w/w lipid, about 10 to about 25% w/w phospholipid, about 1 to about 5% w/w phosphatidylcholine, about 0.1 to 2% w/w phosphatidylinositol, about 0.5 to about 2% w/w phosphatidylserine, about 1.5 to about 6% w/w phosphatidylethanolamine, about 1 to about 5% w/w sphingomyelin, and about 0.1 to about 2.0% w/w ganglioside, or (e) about 12 to about 32% w/w lipid, about 5 to about 25% w/w phospholipid, and about 0.1 to about 2.0% w/w ganglioside, or (f) about 12 to about 32% w/w lipid, about 5 to about 25% w/w phospholipid, about 2 to about 8% w/w phosphatidylcholine, about 0.5 to 3% w/w phosphatidylinositol, about 1 to about 3.5% w/w phosphatidylserine, about 1 to about 10% w/w phosphatidylethanolamine, about 1 to about 8% w/w sphingomyelin, and about 0.5 to about 2.5% w/w ganglioside, or (g) about 80 to about 99% w/w lipid, about 20 to about 75% w/w phospholipid, and about 0.5 to about 5% w/w ganglioside, or (h) about 80 to about 99% w/w lipid, about 20 to about 75% w/w phospholipid, about 2 to about 22% w/w phosphatidylcholine, about 1 to about 10% w/w phosphatidylinositol, about 1 to about 10% w/w phosphatidylserine, about 5 to about 30% w/w phosphatidylethanolamine, about 1 to about 20% w/w sphingomyelin, and about 0.5 to about 5% w/w ganglioside, or (i) about 90 to about 99% w/w lipid, about 20 to about 40% w/w phospholipid, and about 0.5 to about 5% w/w ganglioside, or (j) about 80 to about 99% w/w lipid, about 60 to about 80% w/w phospholipid, and about 0.5 to about 5% w/w ganglioside, or (k) about 15 to about 45% w/w lipid, about 8 to about 25% w/w phospholipid, and about 0.1 to about 5% w/w ganglioside, or (l) about 15 to about 45% w/w lipid, about 8 to about 25% w/w phospholipid, about 1 to about 5% w/w phosphatidylcholine, about 1 to about 5% w/w phosphatidylinositol, about 2 to about 8% w/w phosphatidylserine, about 2 to about 8% w/w phosphatidylethanolamine, about 0.5 to about 5% w/w sphingomyelin, and about 0.1 to about 5% w/w ganglioside, or (m) about 50 to about 99% w/w lipid, about 15 to about 60% w/w phospholipid, and about 1 to about 10% w/w ganglioside, or (n) about 50 to about 99% w/w lipid, about 15 to about 60% w/w phospholipid, about 1 to about 10% w/w phosphatidylcholine, about 1 to about 15% w/w phosphatidylinositol, about 1 to about 20% w/w phosphatidylserine, about 1 to about 20% w/w phosphatidylethanolamine, about 1 to about 10% w/w sphingomyelin, and about 0.1 to about 10% w/w ganglioside.
One or more complex lipids including gangliosides to achieve particular health benefits including maintaining or increasing cognitive development or maintaining or increasing growth in a foetal, infant or child subject.1-28. (canceled) 29. A method for maintaining or increasing growth of a foetal, infant, or child subject, the method comprising administering one or more complex lipids to a foetal, infant, or child subject in need thereof, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 30. The method of claim 29, wherein the one or more complex lipids are administered to a mother during gestation and the growth is brain weight of a foetal subject or brain ganglioside content of a foetal subject. 31. The method of claim 29, wherein the one or more complex lipids are administered to an infant or child subject and the growth is one or more of body weight, body length, and bone mineral density. 32. A method for maintaining or increasing cognitive development of a foetal, infant, or child subject by administering a one or more complex lipids to a foetal, infant, or child subject in need thereof, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 33. The method of claim 32, wherein the one or more complex lipids are administered to a mother during gestation and the cognitive development is brain weight of a foetal subject or brain ganglioside content of a foetal subject. 34. A method for maintaining or increasing growth or maintaining or increasing cognitive development of a foetal subject, the method comprising providing a pregnant mother with one or more complex lipids and informing the mother that the one or more complex lipids will maintain or increase growth or maintain or increase cognitive development of the foetal subject, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 35. A method for maintaining or increasing growth or maintaining or increasing cognitive development in a subject, the method comprising providing a subject with one or more complex lipids and informing the subject that the one or more complex lipids will maintain or increase growth or maintain or increase cognitive development of the subject, wherein the one or more complex lipids comprises at least about 0.1% gangliosides w/w on a dry basis. 36. The method of claim 29, wherein the one or more complex lipids comprises one or more phospholipids, one or more sphingolipids, one or more sphingomyelins or derivatives thereof, one or more ceramides, one or more cerebrosides, one or more gangliosides, or any combination of any two or more thereof. 37. The method of claim 36, wherein the one or more gangliosides comprises GM3, GD3, or a mixture of at least GM3 and GD3. 38. The method of claim 29, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg gangliosides per, 100 g or about 5 mg to about 20 mg gangliosides per 100 g. 39. (canceled) 40. The method of claim 29, wherein the one or more complex lipids comprises a milk fat extract. 41. The method of claim 40, wherein the milk fat extract comprises about 15% to about 99% by weight total lipid, about 1% to about 80% by weight phospholipid, about 1% to about 25% by weight phosphatidylcholine, about 0.1% to about 15% by weight phosphatidylinositol, about 0.1% to about 15% by weight phosphatidylserine, about 1% to about 30% by weight phosphatidylethanolamine, about 0.5% to about 25% by weight sphingomyelin, and about 0.1 to about 10% by weight ganglioside. 42. The method of claim 41, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg milk fat extract per 100 g, at least about 5 mg milk fat extract per 100 g, or at least about 7.5 mg milk fat extract per 100 g. 43. (canceled) 44. (canceled) 45. The method of claim 32, wherein the one or more complex lipids comprises one or more phospholipids, one or more sphingolipids, one or more sphingomyelins or derivatives thereof, one or more ceramides, one or more cerebrosides, one or more gangliosides, or any combination of any two or more thereof. 46. The method of claim 45, wherein the one or more gangliosides comprises GM3, GD3, or a mixture of at least GM3 and GD3. 47. The method of claim 32, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg gangliosides per 100 g or about 5 mg to about 20 mg gangliosides per 100 g. 48. The method of claim 29, wherein the one or more complex lipids comprises a milk fat extract. 49. The method of claim 48, wherein the milk fat extract comprises about 15% to about 99% by weight total lipid, about 1% to about 80% by weight phospholipid, about 1% to about 25% by weight phosphatidylcholine, about 0.1% to about 15% by weight phosphatidylinositol, about 0.1% to about 15% by weight phosphatidylserine, about 1% to about 30% by weight phosphatidylethanolamine, about 0.5% to about 25% by weight sphingomyelin, and about 0.1 to about 10% by weight ganglioside. 50. The method of claim 49, wherein the one or more complex lipids are administered in a composition comprising at least about 2 mg milk fat extract per 100 g, at least about 5 mg milk fat extract per 100 g, or at least about 7.5 mg milk fat extract per 100 g. 51. The method of claim 29, wherein the one or more complex lipids comprises (a) about 15 to about 25% w/w lipid, about 5 to about 15% w/w phospholipid, and about 0.1 to about 1% w/w ganglioside, or (b) about 15 to about 25% w/w lipid, about 5 to about 15% w/w phospholipid, about 1 to about 5% w/w phosphatidylcholine, about 0.1 to about 2% w/w phosphatidylinositol, about 0.5 to about 2% w/w phosphatidylserine, about 1.5 to about 6% w/w phosphatidylethanolamine, about 1 to about 5% w/w sphingomyelin, and about 0.1 to about 1% w/w ganglioside, or (c) about 25 to about 45% w/w lipid, about 10 to about 25% w/w phospholipid, and about 0.1 to about 2.0% w/w ganglioside, or (d) about 25 to about 45% w/w lipid, about 10 to about 25% w/w phospholipid, about 1 to about 5% w/w phosphatidylcholine, about 0.1 to 2% w/w phosphatidylinositol, about 0.5 to about 2% w/w phosphatidylserine, about 1.5 to about 6% w/w phosphatidylethanolamine, about 1 to about 5% w/w sphingomyelin, and about 0.1 to about 2.0% w/w ganglioside, or (e) about 12 to about 32% w/w lipid, about 5 to about 25% w/w phospholipid, and about 0.1 to about 2.0% w/w ganglioside, or (f) about 12 to about 32% w/w lipid, about 5 to about 25% w/w phospholipid, about 2 to about 8% w/w phosphatidylcholine, about 0.5 to 3% w/w phosphatidylinositol, about 1 to about 3.5% w/w phosphatidylserine, about 1 to about 10% w/w phosphatidylethanolamine, about 1 to about 8% w/w sphingomyelin, and about 0.5 to about 2.5% w/w ganglioside, or (g) about 80 to about 99% w/w lipid, about 20 to about 75% w/w phospholipid, and about 0.5 to about 5% w/w ganglioside, or (h) about 80 to about 99% w/w lipid, about 20 to about 75% w/w phospholipid, about 2 to about 22% w/w phosphatidylcholine, about 1 to about 10% w/w phosphatidylinositol, about 1 to about 10% w/w phosphatidylserine, about 5 to about 30% w/w phosphatidylethanolamine, about 1 to about 20% w/w sphingomyelin, and about 0.5 to about 5% w/w ganglioside, or (i) about 90 to about 99% w/w lipid, about 20 to about 40% w/w phospholipid, and about 0.5 to about 5% w/w ganglioside, or (j) about 80 to about 99% w/w lipid, about 60 to about 80% w/w phospholipid, and about 0.5 to about 5% w/w ganglioside, or (k) about 15 to about 45% w/w lipid, about 8 to about 25% w/w phospholipid, and about 0.1 to about 5% w/w ganglioside, or (l) about 15 to about 45% w/w lipid, about 8 to about 25% w/w phospholipid, about 1 to about 5% w/w phosphatidylcholine, about 1 to about 5% w/w phosphatidylinositol, about 2 to about 8% w/w phosphatidylserine, about 2 to about 8% w/w phosphatidylethanolamine, about 0.5 to about 5% w/w sphingomyelin, and about 0.1 to about 5% w/w ganglioside, or (m) about 50 to about 99% w/w lipid, about 15 to about 60% w/w phospholipid, and about 1 to about 10% w/w ganglioside, or (n) about 50 to about 99% w/w lipid, about 15 to about 60% w/w phospholipid, about 1 to about 10% w/w phosphatidylcholine, about 1 to about 15% w/w phosphatidylinositol, about 1 to about 20% w/w phosphatidylserine, about 1 to about 20% w/w phosphatidylethanolamine, about 1 to about 10% w/w sphingomyelin, and about 0.1 to about 10% w/w ganglioside.
1,600
269
269
14,172,604
1,612
A method is provided of making a medicinal delivery system which satiates a craving in an individual when the medicinal delivery system is administered orally to the individual. A coating composition is applied on a saliva-soluble powder to establish a coated powder, the coating composition featuring an at least partially solubilized craving satiation medicinal compound. The coated powder is combined with an edible carrier base to establish a medicinal delivery system that rapidly releases medicine and buffer preferably followed by slower, sustained release.
1. A method of making a medicinal delivery system that delivers medicine which satiates a craving in an individual in need of craving relief when the medicinal delivery system is administered orally to the individual, comprising: applying a coating composition on a saliva-soluble powder to establish a coated powder, the coating composition comprising an at least partially solubilized craving-satiation medicinal compound; and combining the coated powder with a buffer and an edible carrier base to establish a medicinal delivery system. 2. (canceled) 3. (canceled) 4. (canceled) 5. The method of claim 1, wherein the saliva-soluble powder comprises a bulk sweetener. 6. (canceled) 7. (canceled) 8. (canceled) 9. (canceled) 10. (canceled) 11. (canceled) 12. A method of satiating a craving in an individual, comprising: administering a medicinal delivery system made according to the method of claim 1 to an individual in need of craving-satiation when a craving is experienced. 13. (canceled) 14. (canceled) 15. (canceled) 16. (canceled) 17. (canceled) 18. (canceled) 19. (canceled) 20. (canceled) 21. (canceled) 22. (canceled) 23. (canceled) 24. A method of satiating a smoking craving in an individual, comprising: administering a medicinal delivery system made according to the method of claim 17 to an individual in need of a smoking-craving satiation.
A method is provided of making a medicinal delivery system which satiates a craving in an individual when the medicinal delivery system is administered orally to the individual. A coating composition is applied on a saliva-soluble powder to establish a coated powder, the coating composition featuring an at least partially solubilized craving satiation medicinal compound. The coated powder is combined with an edible carrier base to establish a medicinal delivery system that rapidly releases medicine and buffer preferably followed by slower, sustained release.1. A method of making a medicinal delivery system that delivers medicine which satiates a craving in an individual in need of craving relief when the medicinal delivery system is administered orally to the individual, comprising: applying a coating composition on a saliva-soluble powder to establish a coated powder, the coating composition comprising an at least partially solubilized craving-satiation medicinal compound; and combining the coated powder with a buffer and an edible carrier base to establish a medicinal delivery system. 2. (canceled) 3. (canceled) 4. (canceled) 5. The method of claim 1, wherein the saliva-soluble powder comprises a bulk sweetener. 6. (canceled) 7. (canceled) 8. (canceled) 9. (canceled) 10. (canceled) 11. (canceled) 12. A method of satiating a craving in an individual, comprising: administering a medicinal delivery system made according to the method of claim 1 to an individual in need of craving-satiation when a craving is experienced. 13. (canceled) 14. (canceled) 15. (canceled) 16. (canceled) 17. (canceled) 18. (canceled) 19. (canceled) 20. (canceled) 21. (canceled) 22. (canceled) 23. (canceled) 24. A method of satiating a smoking craving in an individual, comprising: administering a medicinal delivery system made according to the method of claim 17 to an individual in need of a smoking-craving satiation.
1,600
270
270
13,412,051
1,632
The present application relates to methods and compositions for treating diseased or damaged cardiac tissue comprising regenerative cells harvested from donor cardiac tissue. In one embodiment, regenerative cells are harvested from an allogeneic source and after administration result in increased viability and/or functional improvement of damaged or diseased cardiac tissue.
1. A method for the reduction of teratoma formation following the delivery of non-self cells to a first subject, comprising: delivering to a first subject a population of regenerative cells, wherein said regenerative cells are isolated from a tissue source harvested from a second subject, wherein said regenerative cells express one or more factors that reduce teratoma formation, wherein said at least a portion of said regenerative cells engraft into a target tissue of said first subject after delivery to said subject, wherein said engraftment persists for a time period ranging from about 1 week to about 6 weeks, wherein during said period of engraftment at least a portion of said regenerative cells are destroyed by the immune system of said first subject, and wherein said engraftment of a portion of said regenerative cells, said period of engraftment, and said destruction of said regenerative cells reduces teratoma formation. 2. The method of claim 1, wherein the reduction in teratoma formation is in comparison to teratoma formation after delivery of embryonic cells to a subject. 3. The method of claim 1, wherein said second subject is an adult. 4. The method of claim 1, wherein during said period of engraftment, said regenerative cells induce endogenous cells to express one or more factors that reduce teratoma formation. 5. The method of claim 1, wherein delivery of said regenerative cells is for the purpose of repairing a damaged or diseased tissue of said first subject. 6. The method of claim 5, wherein said damaged or diseased tissue of said first subject comprises damaged or diseased cardiac tissue and wherein said population of regenerative cells comprises cardiac stem cells. 7. The method of claim 6, wherein said cardiac stem cells are selected from the group consisting of cardiospheres, cardiosphere-derived cells, and a subsequent generation of cardiospheres. 8. The method of claim 1, wherein the regenerative cells express one or more stem cell markers selected from the group consisting of c-kit, CD90, and sca-1. 9. The method of claim 1, wherein the regenerative cells express one or more endothelial cell markers selected from the group consisting of KDR, flk-1, CD31, von Willebrand factor, Ve-cadherin, and smooth muscle alpha actin. 10. The method of claim 1, wherein the regenerative cells express one or more of said stem cell markers or one or more of said endothelial cell markers, but are not selected for based on the expression of said one or more expressed markers. 11. The method of claim 1, wherein said isolated regenerative cells are expanded in culture prior to delivery. 12. The method of claim 11, wherein said isolated regenerative cells generate teratoma-reducing factors in culture. 13. The method of claim 12, further comprising isolating said teratoma-reducing factors from said culture. 14. The method of claim 13, further comprising delivering said isolated teratoma-reducing factors from said culture to said first subject. 15. The method of claim 14, wherein delivery of said isolated teratoma-reducing factors is prior to, concurrently with, or after delivery of said regenerative cells. 16. The method of claim 15, wherein delivery of said isolated teratoma-reducing factors is at multiple time points throughout the period of engraftment of said regenerative cells. 17. The method of claim 1, wherein expression of said factors comprises cell-surface expression. 18. The method of claim 1, wherein expression of said factors comprises release of said factors from the cells. 19. A method of treating a first subject having damaged cardiac tissue with allogeneic cells from a second subject, the method comprising: obtaining a plurality of cells harvested from the cardiac tissue of a second subject, wherein said second subject is an adult, wherein said cells were have been expanded in culture to yield a population of cardiosphere-derived cells (CDCs); wherein said CDCs are not pluripotent and are committed to differentiating into cardiac tissue; administering between about 1×106 and about 100×106 of said CDCs said a first subject, wherein said administered CDCs generate one or more cytokines, chemokines or diffusible factors, wherein, after administration, at least a portion of said administered CDCs engraft into the cardiac tissue of said first subject; and wherein said one or more of said generated cytokines, chemokines or diffusible factors or said engraftment improves the function of said damaged cardiac tissue, thereby treating said first subject. 20. The method of claim 19, wherein said method reduces the risk of producing undesired tissue growth in said first subject as compared to treatment with embryonic cells.
The present application relates to methods and compositions for treating diseased or damaged cardiac tissue comprising regenerative cells harvested from donor cardiac tissue. In one embodiment, regenerative cells are harvested from an allogeneic source and after administration result in increased viability and/or functional improvement of damaged or diseased cardiac tissue.1. A method for the reduction of teratoma formation following the delivery of non-self cells to a first subject, comprising: delivering to a first subject a population of regenerative cells, wherein said regenerative cells are isolated from a tissue source harvested from a second subject, wherein said regenerative cells express one or more factors that reduce teratoma formation, wherein said at least a portion of said regenerative cells engraft into a target tissue of said first subject after delivery to said subject, wherein said engraftment persists for a time period ranging from about 1 week to about 6 weeks, wherein during said period of engraftment at least a portion of said regenerative cells are destroyed by the immune system of said first subject, and wherein said engraftment of a portion of said regenerative cells, said period of engraftment, and said destruction of said regenerative cells reduces teratoma formation. 2. The method of claim 1, wherein the reduction in teratoma formation is in comparison to teratoma formation after delivery of embryonic cells to a subject. 3. The method of claim 1, wherein said second subject is an adult. 4. The method of claim 1, wherein during said period of engraftment, said regenerative cells induce endogenous cells to express one or more factors that reduce teratoma formation. 5. The method of claim 1, wherein delivery of said regenerative cells is for the purpose of repairing a damaged or diseased tissue of said first subject. 6. The method of claim 5, wherein said damaged or diseased tissue of said first subject comprises damaged or diseased cardiac tissue and wherein said population of regenerative cells comprises cardiac stem cells. 7. The method of claim 6, wherein said cardiac stem cells are selected from the group consisting of cardiospheres, cardiosphere-derived cells, and a subsequent generation of cardiospheres. 8. The method of claim 1, wherein the regenerative cells express one or more stem cell markers selected from the group consisting of c-kit, CD90, and sca-1. 9. The method of claim 1, wherein the regenerative cells express one or more endothelial cell markers selected from the group consisting of KDR, flk-1, CD31, von Willebrand factor, Ve-cadherin, and smooth muscle alpha actin. 10. The method of claim 1, wherein the regenerative cells express one or more of said stem cell markers or one or more of said endothelial cell markers, but are not selected for based on the expression of said one or more expressed markers. 11. The method of claim 1, wherein said isolated regenerative cells are expanded in culture prior to delivery. 12. The method of claim 11, wherein said isolated regenerative cells generate teratoma-reducing factors in culture. 13. The method of claim 12, further comprising isolating said teratoma-reducing factors from said culture. 14. The method of claim 13, further comprising delivering said isolated teratoma-reducing factors from said culture to said first subject. 15. The method of claim 14, wherein delivery of said isolated teratoma-reducing factors is prior to, concurrently with, or after delivery of said regenerative cells. 16. The method of claim 15, wherein delivery of said isolated teratoma-reducing factors is at multiple time points throughout the period of engraftment of said regenerative cells. 17. The method of claim 1, wherein expression of said factors comprises cell-surface expression. 18. The method of claim 1, wherein expression of said factors comprises release of said factors from the cells. 19. A method of treating a first subject having damaged cardiac tissue with allogeneic cells from a second subject, the method comprising: obtaining a plurality of cells harvested from the cardiac tissue of a second subject, wherein said second subject is an adult, wherein said cells were have been expanded in culture to yield a population of cardiosphere-derived cells (CDCs); wherein said CDCs are not pluripotent and are committed to differentiating into cardiac tissue; administering between about 1×106 and about 100×106 of said CDCs said a first subject, wherein said administered CDCs generate one or more cytokines, chemokines or diffusible factors, wherein, after administration, at least a portion of said administered CDCs engraft into the cardiac tissue of said first subject; and wherein said one or more of said generated cytokines, chemokines or diffusible factors or said engraftment improves the function of said damaged cardiac tissue, thereby treating said first subject. 20. The method of claim 19, wherein said method reduces the risk of producing undesired tissue growth in said first subject as compared to treatment with embryonic cells.
1,600
271
271
13,139,809
1,631
Exemplary embodiments of the present disclosure relate generally to methods, computer-accessible medium and systems for assembling haplotype and/or genotype sequences of at least one genome, which can be based upon, e.g., consistent layouts of short sequence reads and long-range genome related data. For example, a processing arrangement can be configured to perform a procedure including, e.g., obtaining randomly located short sequence reads, using at least one score function in combination with constraints based on, e.g., the long range data, generating a layout of randomly located short sequence reads such that the layout is globally optimal with respect to the score function, obtained through searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints, generating a whole and/or a part of a genome wide haplotype sequence and/or genotype sequence, and converting a globally optimal layout into one or more consensus sequences.
1. A computer-accessible medium having stored thereon computer executable instructions for assembling at least one haplotype sequence or genotype sequence of at least one genome, wherein, when the executable instructions are executed by a processing arrangement, the processing arrangement is configured to perform at least one procedure comprising: (a) obtaining a plurality of randomly located short sequence reads; (b) using at least one score function in combination with constraints based on long range information associated with the at least one genome, generating a layout of all of or a subset of randomly located short sequence reads such that the generated layout is globally optimal with respect to the at least one score function while substantially satisfying the constraints, wherein the at least one score-function is derived from short-range overlap relations among the randomly located short sequence reads; (c) searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints; (d) generating at least one of a whole or at least one part of at least one genome wide haplotype sequence or genotype sequence of the at least one genome; and (e) converting a globally optimal layout into one or more consensus sequences so as to substantially satisfy the constraints. 2. The computer-accessible medium of claim 1, wherein the processing arrangement is further configured to obtain the randomly located short sequence reads using at least one of Sanger chemistry, sequencing-by-synthesis, sequencing-by-hybridization or sequencing-by-ligation. 3. The computer-accessible medium of claim 1, wherein the process arrangement is further configured to obtain the randomly located short sequence reads using at least one method having at least one error, wherein the at least one error is at least one of: incorrect base-calls, missing bases, inserted bases or homopolymeric compression. 4. The computer-accessible medium of claim 1, wherein the at least one genome includes genomes from at least one of at least one of a plurality of diseased cells or non-diseased cells, at least one individual organism, at least one population, or at least one ecological system. 5. The computer-accessible medium of claim 1, wherein the randomly located non-contextual sequence reads are effectuated by a score-function encoding particular long-range information. 6. The computer-accessible medium of claim 1, wherein the particular information includes long-range information. 7. The computer-accessible medium of claim 5, wherein the long-range information is obtained from at least one of at least one of a plurality of diseased or non-diseased cells, at least one individual organism, at least one population, or at least one ecological system. 8. The computer-accessible medium of claim 5, wherein the long-range information is obtained from at least one of a mathematical model, existing data, genomic single-molecules, or genomic materials amplified or modified in a particular manner. 9. The computer-accessible medium of claim 8, wherein the mathematical model is at least one of Bayesian or empirical Bayesian. 10. The computer-accessible medium of claim 8, wherein the long-range information is obtained from at least one of randomly sheared single molecules or targeted genomic single-molecules. 11. The computer-accessible medium of claim 10, wherein the long-range information is a physical map that is at least one of an ordered restriction map, a probe map, or a base-distribution map. 12. The computer-accessible medium of claim 8, wherein the long-range information is obtained from amplified clones that are analyzed by at least one of restriction activities or an end sequencing procedure. 13. The computer-accessible medium of claim 8, wherein the long-range information is obtained from existing data that includes at least one of (i) a reference haplotype or genotype whole-genome sequence, (ii) a reference collection of phased, unphased, haplotyped or genotyped sequence contigs, (iii) population-wide whole-genome sequences, or (iv) population-wide collections of phased, unphased, haplotyped or genotyped sequence-contigs. 14. The computer-accessible medium of claim 1, wherein the processing arrangement is further configured to store the sequence reads in a tree-type of a data structure having paths that are usable to organize possible arrangements, and wherein the sequence reads are configured to be overlayed and taking into account the overlaps, containments, and overhangs among consecutive sequence reads in an overlay along at least one path in the arrangement. 15. The computer-accessible medium of claim 14, wherein the processing arrangement is further configured to evaluate overlays along the at least one path by utilizing a score function. 16. The computer-accessible medium of claim 14, wherein the processing arrangement is further configured to use the score-function to identify the at least one path having relatively low score values with respect to a rank order of the score values of all of the at least one path or plausible bounds. 17. The computer-accessible medium of claim 15, wherein the processing arrangement is further configured to evaluate the score-function with respect to at least one of the overlaps, containment and overhangs among at least one of a single pair or a local collection of pairs of the sequence reads. 18. The computer-accessible medium of claim 17, wherein the processing arrangement is further configured to evaluate the overlaps, containments or overhangs among the sequence reads with unknown orientations, locations, and haplotypic identities. 19. The computer-accessible medium of claim 17, wherein the processing arrangement is further configured to evaluate the overlaps, containments or overhangs occurring among the sequence reads from different sequencing technologies, and wherein each different sequencing technology has a respective separate process for performing erroneous reads. 20. The computer-accessible medium of claim 19, wherein the processing arrangement is further configured to determine thresholds below which the detected overlaps, containments or overhangs are to be discarded from a further consideration. 21. The computer-accessible medium of claim 20, wherein the processing arrangement is further configured to determine the values of at least one of the thresholds using at least one of a Bayesian method or an empirical Bayesian method. 22. The computer-accessible medium of claim 20, wherein the processing arrangement is further configured to determine the values of at least one of the thresholds using a procedure for controlling false discovery rates. 23. The computer-accessible medium of claim 15, wherein the processing arrangement is further configured to evaluate the score-function based on a consistency of the score-function with respect to particular long-range information. 24. The computer-accessible medium of claim 23, wherein the processing arrangement is further configured to evaluate the score-function based on a consistency of the score-function with respect to the particular long-range information by determining a local alignment with an alignment score. 25. The computer-accessible medium of claim 24, wherein the processing arrangement is further configured to determine the score-function using a dynamic programming procedure for a local alignment with an alignment score. 26. The computer-accessible medium of claim 25, wherein the processing arrangement is further configured to determine the score-function by using at least one alignment score parameter obtained by at least one of a learning procedure, heuristics or a Bayesian-based design. 27. The computer-accessible medium of claim 14, wherein the processing arrangement is further configured to select at least one of relatively best scoring arrangements of the sequence reads to determine a corresponding multiple sequence alignment that is combinable to generate the at least one whole-genome haplotypic sequence. 28. The computer-accessible medium of claim 1, wherein the randomly located short sequence reads are generated using at least one of Sanger chemistry, sequencing-by-synthesis, sequencing-by-hybridization or sequencing-by-ligation. 29. The computer-accessible medium of claim 1, wherein the randomly located short sequence reads are generated using at least one method having at least one error, and wherein the at least one error is at least one of: incorrect base-calls, missing bases, inserted bases or homopolymeric compression. 30. The computer-accessible medium of claim 1, wherein the short-range comprises approximately 1 Kb-10 Kb of information associated with the at least one genome. 31. The computer-accessible medium of claim 1, wherein the long-range comprises approximately 10 Kb-200 mb of information associated with the at least one genome. 32. The computer-accessible medium of claim 1, wherein the searching procedure is performed simultaneously with a score and constraint dependent pruning procedure. 33. The computer-accessible medium of claim 32, wherein the globally optimal layout is based on a consistency with respect to satisfying the constraints. 34. A process for assembling at least one haplotype sequence of at least one genome, comprising: (a) obtaining a plurality of randomly located short sequence reads; (b) using at least one score function in combination with constraints based on long range information associated with the at least one genome, generating a layout of all of or a subset of randomly located short sequence reads such that the generated layout is globally optimal with respect to the at least one score function while substantially satisfying the constraints, wherein the at least one score-function is derived from short-range overlap relations among the randomly located short sequence reads; (c) searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints; (d) generating at least one of a whole or at least one part of at least one genome wide haplotype sequence or genotype sequence of the at least one genome; and (e) converting a globally optimal layout into one or more consensus sequences so as to substantially satisfy the constraints. 35. The process of claim 34, further comprising at least one of displaying or storing at least one of the particular information or the at least one genome wide haplotype sequence in a storage arrangement in at least one of a user-accessible format or a user-readable format. 36. A system for assembling at least one haplotype sequence of at least one genome, comprising a processing arrangement which, when executed, is configured to perform at least one procedure comprising: (a) obtaining a plurality of randomly located short sequence reads; (b) using at least one score function in combination with constraints based on long range information associated with the at least one genome, generating a layout of all of or a subset of randomly located short sequence reads such that the generated layout is globally optimal with respect to the at least one score function while substantially satisfying the constraints, wherein the at least one score-function is derived from short-range overlap relations among the randomly located short sequence reads; (c) searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints; (d) generating at least one of a whole or at least one part of at least one genome wide haplotype sequence or genotype sequence of the at least one genome; and (e) converting a globally optimal layout into one or more consensus sequences so as to substantially satisfy the constraints.
Exemplary embodiments of the present disclosure relate generally to methods, computer-accessible medium and systems for assembling haplotype and/or genotype sequences of at least one genome, which can be based upon, e.g., consistent layouts of short sequence reads and long-range genome related data. For example, a processing arrangement can be configured to perform a procedure including, e.g., obtaining randomly located short sequence reads, using at least one score function in combination with constraints based on, e.g., the long range data, generating a layout of randomly located short sequence reads such that the layout is globally optimal with respect to the score function, obtained through searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints, generating a whole and/or a part of a genome wide haplotype sequence and/or genotype sequence, and converting a globally optimal layout into one or more consensus sequences.1. A computer-accessible medium having stored thereon computer executable instructions for assembling at least one haplotype sequence or genotype sequence of at least one genome, wherein, when the executable instructions are executed by a processing arrangement, the processing arrangement is configured to perform at least one procedure comprising: (a) obtaining a plurality of randomly located short sequence reads; (b) using at least one score function in combination with constraints based on long range information associated with the at least one genome, generating a layout of all of or a subset of randomly located short sequence reads such that the generated layout is globally optimal with respect to the at least one score function while substantially satisfying the constraints, wherein the at least one score-function is derived from short-range overlap relations among the randomly located short sequence reads; (c) searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints; (d) generating at least one of a whole or at least one part of at least one genome wide haplotype sequence or genotype sequence of the at least one genome; and (e) converting a globally optimal layout into one or more consensus sequences so as to substantially satisfy the constraints. 2. The computer-accessible medium of claim 1, wherein the processing arrangement is further configured to obtain the randomly located short sequence reads using at least one of Sanger chemistry, sequencing-by-synthesis, sequencing-by-hybridization or sequencing-by-ligation. 3. The computer-accessible medium of claim 1, wherein the process arrangement is further configured to obtain the randomly located short sequence reads using at least one method having at least one error, wherein the at least one error is at least one of: incorrect base-calls, missing bases, inserted bases or homopolymeric compression. 4. The computer-accessible medium of claim 1, wherein the at least one genome includes genomes from at least one of at least one of a plurality of diseased cells or non-diseased cells, at least one individual organism, at least one population, or at least one ecological system. 5. The computer-accessible medium of claim 1, wherein the randomly located non-contextual sequence reads are effectuated by a score-function encoding particular long-range information. 6. The computer-accessible medium of claim 1, wherein the particular information includes long-range information. 7. The computer-accessible medium of claim 5, wherein the long-range information is obtained from at least one of at least one of a plurality of diseased or non-diseased cells, at least one individual organism, at least one population, or at least one ecological system. 8. The computer-accessible medium of claim 5, wherein the long-range information is obtained from at least one of a mathematical model, existing data, genomic single-molecules, or genomic materials amplified or modified in a particular manner. 9. The computer-accessible medium of claim 8, wherein the mathematical model is at least one of Bayesian or empirical Bayesian. 10. The computer-accessible medium of claim 8, wherein the long-range information is obtained from at least one of randomly sheared single molecules or targeted genomic single-molecules. 11. The computer-accessible medium of claim 10, wherein the long-range information is a physical map that is at least one of an ordered restriction map, a probe map, or a base-distribution map. 12. The computer-accessible medium of claim 8, wherein the long-range information is obtained from amplified clones that are analyzed by at least one of restriction activities or an end sequencing procedure. 13. The computer-accessible medium of claim 8, wherein the long-range information is obtained from existing data that includes at least one of (i) a reference haplotype or genotype whole-genome sequence, (ii) a reference collection of phased, unphased, haplotyped or genotyped sequence contigs, (iii) population-wide whole-genome sequences, or (iv) population-wide collections of phased, unphased, haplotyped or genotyped sequence-contigs. 14. The computer-accessible medium of claim 1, wherein the processing arrangement is further configured to store the sequence reads in a tree-type of a data structure having paths that are usable to organize possible arrangements, and wherein the sequence reads are configured to be overlayed and taking into account the overlaps, containments, and overhangs among consecutive sequence reads in an overlay along at least one path in the arrangement. 15. The computer-accessible medium of claim 14, wherein the processing arrangement is further configured to evaluate overlays along the at least one path by utilizing a score function. 16. The computer-accessible medium of claim 14, wherein the processing arrangement is further configured to use the score-function to identify the at least one path having relatively low score values with respect to a rank order of the score values of all of the at least one path or plausible bounds. 17. The computer-accessible medium of claim 15, wherein the processing arrangement is further configured to evaluate the score-function with respect to at least one of the overlaps, containment and overhangs among at least one of a single pair or a local collection of pairs of the sequence reads. 18. The computer-accessible medium of claim 17, wherein the processing arrangement is further configured to evaluate the overlaps, containments or overhangs among the sequence reads with unknown orientations, locations, and haplotypic identities. 19. The computer-accessible medium of claim 17, wherein the processing arrangement is further configured to evaluate the overlaps, containments or overhangs occurring among the sequence reads from different sequencing technologies, and wherein each different sequencing technology has a respective separate process for performing erroneous reads. 20. The computer-accessible medium of claim 19, wherein the processing arrangement is further configured to determine thresholds below which the detected overlaps, containments or overhangs are to be discarded from a further consideration. 21. The computer-accessible medium of claim 20, wherein the processing arrangement is further configured to determine the values of at least one of the thresholds using at least one of a Bayesian method or an empirical Bayesian method. 22. The computer-accessible medium of claim 20, wherein the processing arrangement is further configured to determine the values of at least one of the thresholds using a procedure for controlling false discovery rates. 23. The computer-accessible medium of claim 15, wherein the processing arrangement is further configured to evaluate the score-function based on a consistency of the score-function with respect to particular long-range information. 24. The computer-accessible medium of claim 23, wherein the processing arrangement is further configured to evaluate the score-function based on a consistency of the score-function with respect to the particular long-range information by determining a local alignment with an alignment score. 25. The computer-accessible medium of claim 24, wherein the processing arrangement is further configured to determine the score-function using a dynamic programming procedure for a local alignment with an alignment score. 26. The computer-accessible medium of claim 25, wherein the processing arrangement is further configured to determine the score-function by using at least one alignment score parameter obtained by at least one of a learning procedure, heuristics or a Bayesian-based design. 27. The computer-accessible medium of claim 14, wherein the processing arrangement is further configured to select at least one of relatively best scoring arrangements of the sequence reads to determine a corresponding multiple sequence alignment that is combinable to generate the at least one whole-genome haplotypic sequence. 28. The computer-accessible medium of claim 1, wherein the randomly located short sequence reads are generated using at least one of Sanger chemistry, sequencing-by-synthesis, sequencing-by-hybridization or sequencing-by-ligation. 29. The computer-accessible medium of claim 1, wherein the randomly located short sequence reads are generated using at least one method having at least one error, and wherein the at least one error is at least one of: incorrect base-calls, missing bases, inserted bases or homopolymeric compression. 30. The computer-accessible medium of claim 1, wherein the short-range comprises approximately 1 Kb-10 Kb of information associated with the at least one genome. 31. The computer-accessible medium of claim 1, wherein the long-range comprises approximately 10 Kb-200 mb of information associated with the at least one genome. 32. The computer-accessible medium of claim 1, wherein the searching procedure is performed simultaneously with a score and constraint dependent pruning procedure. 33. The computer-accessible medium of claim 32, wherein the globally optimal layout is based on a consistency with respect to satisfying the constraints. 34. A process for assembling at least one haplotype sequence of at least one genome, comprising: (a) obtaining a plurality of randomly located short sequence reads; (b) using at least one score function in combination with constraints based on long range information associated with the at least one genome, generating a layout of all of or a subset of randomly located short sequence reads such that the generated layout is globally optimal with respect to the at least one score function while substantially satisfying the constraints, wherein the at least one score-function is derived from short-range overlap relations among the randomly located short sequence reads; (c) searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints; (d) generating at least one of a whole or at least one part of at least one genome wide haplotype sequence or genotype sequence of the at least one genome; and (e) converting a globally optimal layout into one or more consensus sequences so as to substantially satisfy the constraints. 35. The process of claim 34, further comprising at least one of displaying or storing at least one of the particular information or the at least one genome wide haplotype sequence in a storage arrangement in at least one of a user-accessible format or a user-readable format. 36. A system for assembling at least one haplotype sequence of at least one genome, comprising a processing arrangement which, when executed, is configured to perform at least one procedure comprising: (a) obtaining a plurality of randomly located short sequence reads; (b) using at least one score function in combination with constraints based on long range information associated with the at least one genome, generating a layout of all of or a subset of randomly located short sequence reads such that the generated layout is globally optimal with respect to the at least one score function while substantially satisfying the constraints, wherein the at least one score-function is derived from short-range overlap relations among the randomly located short sequence reads; (c) searching coupled with score and constraint dependent pruning to determine the globally optimal layout substantially satisfying the constraints; (d) generating at least one of a whole or at least one part of at least one genome wide haplotype sequence or genotype sequence of the at least one genome; and (e) converting a globally optimal layout into one or more consensus sequences so as to substantially satisfy the constraints.
1,600
272
272
15,065,083
1,627
The invention relates to the compounds and methods for inhibiting the Cyclin-Dependent Kinase Inhibitor (CDKI) pathway. More particularly, the invention relates to compounds and methods for inhibiting the CDKI pathway for studies of and intervention in senescence-related and other CDKI-related diseases. The invention provides new compounds having improved solubility and/or potency, and methods for their use. In various aspects, the invention relates to the treatment of cancer. The invention provides methods for chemoprevention and prevention of tumor recurrence or metastasis. The invention further provides diagnostic techniques for treatment for certain cancer types. The invention utilizes specific inhibitors of CDK8/19 and/or measurement of CDK8 levels in a patient.
1-39. (canceled) 40. A method for treating a cancer in a patient that is a tumor that expresses β-catenin, comprising administering to the patient a specific inhibitor of CDK8/19. 41. A method for chemoprotecting a patient at risk for developing cancer, comprising administering to the patient a specific inhibitor of CDK8/19. 42. A method for preventing cancer recurrence or metastasis in a cancer patient who has undergone debulking treatment for a tumor, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19 following debulking. 43. The method according to claim 42, wherein the patient is an ovarian or breast cancer patient. 44. A method for preventing tumor invasion, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19. 45. A method for treating a cancer patient comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19 in combination with a DNA damaging agent. 46. The method according to claim 45, wherein the patient is an ovarian or breast cancer patient. 47. A method for inhibiting tumor-promoting factor secretion by one or more of fibroblasts and stromal cells in a patient, wherein the secretion is induced by a DNA-damaging agent, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19. 48. A method for improving the efficacy of adjuvant therapy given to a patient in combination with surgery, comprising administering a small molecule compound that specifically inhibits CDK8/19 in combination with the adjuvant therapy. 49. A method for treating breast cancer in a patient, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19. 50. A method for determining whether a cancer patient is suitable to be treated with a DNA-damaging agent, the method comprising determining whether one or more of CDK8, CCNC and CDK19 is overexpressed in a tumor sample from the patient. 51. The method according to claim 50, wherein it is determined whether CDK8 is overexpressed in the tumor sample. 52. The method according to claim 50, wherein the patient is an ovarian or breast cancer patient. 53. A method for determining the likelihood of recurrence or metastasis of a tumor in a cancer patient, comprising determining whether one or more of CDK8, CCNC and CDK19 is overexpresssed in a tumor sample from the patient. 54. The method according to claim 53, wherein it is determined whether CDK8 is overexpressed in the tumor sample. 55. The method according to claim 53, wherein the patient is an ovarian or breast cancer patient. 56. A method for determining whether a surgical cancer patient will benefit from adjuvant therapy, comprising determining whether one or more of CDK8, CCNC and CDK19 is overexpressed in a tumor sample from the patient. 57. The method according to claim 56, wherein it is determined whether CDK8 is overexpressed in the tumor sample. 58. The method according to claim 56, wherein the patient is an ovarian or breast cancer patient.
The invention relates to the compounds and methods for inhibiting the Cyclin-Dependent Kinase Inhibitor (CDKI) pathway. More particularly, the invention relates to compounds and methods for inhibiting the CDKI pathway for studies of and intervention in senescence-related and other CDKI-related diseases. The invention provides new compounds having improved solubility and/or potency, and methods for their use. In various aspects, the invention relates to the treatment of cancer. The invention provides methods for chemoprevention and prevention of tumor recurrence or metastasis. The invention further provides diagnostic techniques for treatment for certain cancer types. The invention utilizes specific inhibitors of CDK8/19 and/or measurement of CDK8 levels in a patient.1-39. (canceled) 40. A method for treating a cancer in a patient that is a tumor that expresses β-catenin, comprising administering to the patient a specific inhibitor of CDK8/19. 41. A method for chemoprotecting a patient at risk for developing cancer, comprising administering to the patient a specific inhibitor of CDK8/19. 42. A method for preventing cancer recurrence or metastasis in a cancer patient who has undergone debulking treatment for a tumor, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19 following debulking. 43. The method according to claim 42, wherein the patient is an ovarian or breast cancer patient. 44. A method for preventing tumor invasion, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19. 45. A method for treating a cancer patient comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19 in combination with a DNA damaging agent. 46. The method according to claim 45, wherein the patient is an ovarian or breast cancer patient. 47. A method for inhibiting tumor-promoting factor secretion by one or more of fibroblasts and stromal cells in a patient, wherein the secretion is induced by a DNA-damaging agent, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19. 48. A method for improving the efficacy of adjuvant therapy given to a patient in combination with surgery, comprising administering a small molecule compound that specifically inhibits CDK8/19 in combination with the adjuvant therapy. 49. A method for treating breast cancer in a patient, comprising administering to the patient a small molecule compound that specifically inhibits CDK8/19. 50. A method for determining whether a cancer patient is suitable to be treated with a DNA-damaging agent, the method comprising determining whether one or more of CDK8, CCNC and CDK19 is overexpressed in a tumor sample from the patient. 51. The method according to claim 50, wherein it is determined whether CDK8 is overexpressed in the tumor sample. 52. The method according to claim 50, wherein the patient is an ovarian or breast cancer patient. 53. A method for determining the likelihood of recurrence or metastasis of a tumor in a cancer patient, comprising determining whether one or more of CDK8, CCNC and CDK19 is overexpresssed in a tumor sample from the patient. 54. The method according to claim 53, wherein it is determined whether CDK8 is overexpressed in the tumor sample. 55. The method according to claim 53, wherein the patient is an ovarian or breast cancer patient. 56. A method for determining whether a surgical cancer patient will benefit from adjuvant therapy, comprising determining whether one or more of CDK8, CCNC and CDK19 is overexpressed in a tumor sample from the patient. 57. The method according to claim 56, wherein it is determined whether CDK8 is overexpressed in the tumor sample. 58. The method according to claim 56, wherein the patient is an ovarian or breast cancer patient.
1,600
273
273
13,701,619
1,619
The present invention relates to a coating particle containing a nuclear particle covered with a coating layer, and in the coating particle, the coating layer is a layer containing hydroxyalkyl cellulose and a binder.
1. A coating particle comprising a nuclear particle covered with a coating layer, wherein the coating layer is a layer comprising hydroxyalkyl cellulose and a binder. 2. The coating particle according to claim 1, wherein the binder is at least one selected from the group consisting of polyalkylene glycol, polyalkylene glycol higher fatty acid ester, higher fatty acid, higher alcohol, higher alcohol ester and natural wax. 3. The coating particle according to claim 1, wherein the coating layer further comprises an elution controlling base and/or silica. 4. A method for producing a coating particle, comprising a first step of dry-coating a nuclear particle with hydroxyalkyl cellulose and a binder. 5. The method for producing a coating particle according to claim 4, further comprising a second step of dry-coating the particle obtained in the first step with an elution controlling base and a binder. 6. The method for producing a coating particle according to claim 4, further comprising a third step of overcoating the particle obtained in the first step with silica. 7. The method for producing a coating particle according to claim 4, further comprising a second step of dry-coating the particle obtained in the first step with an elution controlling base and a binder, and a third step of overcoating the particle obtained in the second step with silica.
The present invention relates to a coating particle containing a nuclear particle covered with a coating layer, and in the coating particle, the coating layer is a layer containing hydroxyalkyl cellulose and a binder.1. A coating particle comprising a nuclear particle covered with a coating layer, wherein the coating layer is a layer comprising hydroxyalkyl cellulose and a binder. 2. The coating particle according to claim 1, wherein the binder is at least one selected from the group consisting of polyalkylene glycol, polyalkylene glycol higher fatty acid ester, higher fatty acid, higher alcohol, higher alcohol ester and natural wax. 3. The coating particle according to claim 1, wherein the coating layer further comprises an elution controlling base and/or silica. 4. A method for producing a coating particle, comprising a first step of dry-coating a nuclear particle with hydroxyalkyl cellulose and a binder. 5. The method for producing a coating particle according to claim 4, further comprising a second step of dry-coating the particle obtained in the first step with an elution controlling base and a binder. 6. The method for producing a coating particle according to claim 4, further comprising a third step of overcoating the particle obtained in the first step with silica. 7. The method for producing a coating particle according to claim 4, further comprising a second step of dry-coating the particle obtained in the first step with an elution controlling base and a binder, and a third step of overcoating the particle obtained in the second step with silica.
1,600
274
274
12,554,497
1,699
The present invention is directed to a biomarker and kit for diagnosing, monitoring and/or staging Alzheimer's disease comprising redox-reactive autoantibodies. The present invention is also directed to a method for diagnosing, monitoring and/or staging Alzheimer's disease which comprises conducting a blood test using the same.
1. A method for diagnosing, monitoring and/or staging Alzheimer's disease which comprises conducting a blood test for redox-reactive autoantibodies. 2. A method in accordance with claim 1 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 3. A method in accordance with claim 1 wherein the autoantibodies are autoantibodies which bind to phospholipids. 4. A method in accordance with claim 3 wherein the phospholipids are at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine. 5. A method according to claim 1 wherein the blood test comprises an assay that can detect antiphospholipid autoantibodies. 6. A method in accordance with claim 5 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 7. A method in accordance with claim 5 wherein the autoantibodies bind to at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine. 8. A biomarker for diagnosing, monitoring and/or staging Alzheimer's disease which comprises redox-reactive autoantibodies. 9. A biomarker in accordance with claim 8 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 10. A biomarker in accordance with claim 8 wherein the autoantibodies are autoantibodies which bind to phospholipids. 11. A biomarker in accordance with claim 8 wherein the phospholipids are at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine. 12. A kit for diagnosing, monitoring and/or staging Alzheimer's disease which comprises an assay which can detect antiphospholipid autoantibodies, 13. A kit in accordance with claim 12 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 14. A kit in accordance with claim 12 wherein the autoantibodies bind to at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine.
The present invention is directed to a biomarker and kit for diagnosing, monitoring and/or staging Alzheimer's disease comprising redox-reactive autoantibodies. The present invention is also directed to a method for diagnosing, monitoring and/or staging Alzheimer's disease which comprises conducting a blood test using the same.1. A method for diagnosing, monitoring and/or staging Alzheimer's disease which comprises conducting a blood test for redox-reactive autoantibodies. 2. A method in accordance with claim 1 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 3. A method in accordance with claim 1 wherein the autoantibodies are autoantibodies which bind to phospholipids. 4. A method in accordance with claim 3 wherein the phospholipids are at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine. 5. A method according to claim 1 wherein the blood test comprises an assay that can detect antiphospholipid autoantibodies. 6. A method in accordance with claim 5 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 7. A method in accordance with claim 5 wherein the autoantibodies bind to at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine. 8. A biomarker for diagnosing, monitoring and/or staging Alzheimer's disease which comprises redox-reactive autoantibodies. 9. A biomarker in accordance with claim 8 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 10. A biomarker in accordance with claim 8 wherein the autoantibodies are autoantibodies which bind to phospholipids. 11. A biomarker in accordance with claim 8 wherein the phospholipids are at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine. 12. A kit for diagnosing, monitoring and/or staging Alzheimer's disease which comprises an assay which can detect antiphospholipid autoantibodies, 13. A kit in accordance with claim 12 wherein the autoantibodies are at least one of IgG, IgM, IgA, IgE, and IgD. 14. A kit in accordance with claim 12 wherein the autoantibodies bind to at least one of phosphatidylserine, cardiolipin, phosphatidylethanolamine and phosphatidylcholine.
1,600
275
275
15,025,445
1,618
An extruded water-soluble article includes a water-soluble polymer having an extrusion temperature of 90 to 150 C., a plasticizer, and one or more active agents in a total amount of 0.1% to 50% by weight of the article. Combining the active agent(s) with a plasticizer prior to melt-blending with the water-soluble polymer prevents phase separation of the active agent and the polymer. Articles include films, fibers, rods, bars and the like. Such articles may be placed in an absorbent article such as a bandage, a medical drape, a wipe, a sheet, a diaper, a pant, or a pad.
1. An extruded water-soluble article comprising: a water-soluble, polymer having an extrusion temperature of 90 to 150° C.; a plasticizer; and one or more thermal active agents in a total amount of 0.1% to 50% by weight of the article, wherein the one or more thermal active agents are completely dissolvable/dispersible in the plasticizer; wherein the combination of the one or more thermal active agents and the plasticizer is a homogeneous mixture/solution; and wherein a homogeneous blend, comprising the polymer and the homogeneous mixture/solution, has an extrusion temperature of 50 to 125° C. 2. The extruded water-soluble article of claim 1, wherein the polymer is selected from the group consisting of polyvinyl alcohol (PVOH), polyethylene oxide (PEO), polyethylene glycol (PEG), polyacylate (acid), polyacylamide, polyester, thermoplastic starch, polyolefin copolymer; and a combination thereof. 3. The extruded water soluble article of claim 1, wherein the plasticizer is selected from glycerin, PEG-400, PEG-800, PEG-1000, and low molecular weight polypropylene oxide/polyethylene oxide-based copolymers. 4. The extruded water-soluble article of claim 1, wherein the polymer is an amorphous vinyl alcohol matrix. 5. The extruded water-soluble article of claim 1, wherein the extrusion temperature is 90 to 125° C. 6. The extruded water-soluble article of claim 1, wherein the one or more thermal active agents are in a total amount of 1% to 20% by weight of the article. 7. The extruded water-soluble article of claim 1 further comprising up to 50% thermoplastic starch by weight. 8. The extruded water-soluble article of claim 1 further comprising up to 30% by weight of ethylene vinyl acetate. 9. The extruded water-soluble article of claim 1, wherein the article is a mono-layer or multi-layer film. 10. The extruded water-soluble article of claim 9, wherein the film has a water dissolution speed from 5 seconds to 30 minutes as determined by the Dissolution Test of the present disclosure. 11. The extruded water-soluble article of claim 9, wherein the film has a basis weight of 5 gsm to 500 gsm. 12. The extruded water-soluble article of claim 9, wherein the film has an elongation of 5% to 500% according to the Tensile Test of the present disclosure. 13. The extruded water-soluble article of claim 1, wherein the one or more thermal active agents are menthol, a menthol derivative thereof, or a combination thereof. 14. The extruded water-soluble article of claim 1 wherein the one or more thermal active agents are configured to either stimulate human sensory receptors or change the temperature of the skin upon contact therewith. 15. The extruded water-soluble article of claim 1 wherein the one or more thermal active agents comprise a temperature agent selected from menthol, menthol derivatives, xylitol, capsaicin, polyols, urea, self-heating zeolite or alkali metal-compounds, magnesium chloride, magnesium sulfate and combinations thereof. 16. A personal absorbent article comprising: an absorbent member sandwiched between a water-impermeable backsheet and a water-permeable liner, wherein the liner has a body-facing surface and an opposite outward-facing surface; and a film attached to either the outward-facing surface of the liner or a surface of the absorbent member that is adjacent the liner, wherein the film comprises: a water-soluble, polymer having an extrusion temperature of 90 to 150° C.; a plasticizer; and one or more thermal active agents in a total amount of 0.1% to 50% by weight of the article, wherein the one or more thermal active ingredients are dissolvable/dispersible in the plasticizer; wherein the combination of the one or more thermal active agents and the plasticizer is a homogeneous mixture/solution; and wherein a homogeneous blend comprising the polymer and the homogeneous mixture/solution has an extrusion temperature of 50 to 125° C. 17. The personal absorbent article of claim 16, wherein the personal absorbent article is a bandage, a medical drape, a wipe, a sheet, a diaper, a pant, or a pad. 18. A method of making an extruded article comprising the following steps: a) prepare a mixture/solution comprising a thermoplastic plasticizer and one or more thermal active agents; b) form a homogenous blend having an extrusion and a processing temperature of 90 to 125° C., the homogeneous blend comprising the mixture/solution and an amorphous, water-soluble polyvinyl alcohol, wherein the homogeneous blend comprises 0.1% to 40% by weight of the one or more thermal active agents; and c) extrude the homogeneous blend to form a film. 19. The method of claim 18, wherein the homogenous blend can be extruded directly onto a carrier substrate, forming a bond therebetween. 20. The method of claim 18, wherein one or more secondary active agents are co-extruded with the homogeneous blend.
An extruded water-soluble article includes a water-soluble polymer having an extrusion temperature of 90 to 150 C., a plasticizer, and one or more active agents in a total amount of 0.1% to 50% by weight of the article. Combining the active agent(s) with a plasticizer prior to melt-blending with the water-soluble polymer prevents phase separation of the active agent and the polymer. Articles include films, fibers, rods, bars and the like. Such articles may be placed in an absorbent article such as a bandage, a medical drape, a wipe, a sheet, a diaper, a pant, or a pad.1. An extruded water-soluble article comprising: a water-soluble, polymer having an extrusion temperature of 90 to 150° C.; a plasticizer; and one or more thermal active agents in a total amount of 0.1% to 50% by weight of the article, wherein the one or more thermal active agents are completely dissolvable/dispersible in the plasticizer; wherein the combination of the one or more thermal active agents and the plasticizer is a homogeneous mixture/solution; and wherein a homogeneous blend, comprising the polymer and the homogeneous mixture/solution, has an extrusion temperature of 50 to 125° C. 2. The extruded water-soluble article of claim 1, wherein the polymer is selected from the group consisting of polyvinyl alcohol (PVOH), polyethylene oxide (PEO), polyethylene glycol (PEG), polyacylate (acid), polyacylamide, polyester, thermoplastic starch, polyolefin copolymer; and a combination thereof. 3. The extruded water soluble article of claim 1, wherein the plasticizer is selected from glycerin, PEG-400, PEG-800, PEG-1000, and low molecular weight polypropylene oxide/polyethylene oxide-based copolymers. 4. The extruded water-soluble article of claim 1, wherein the polymer is an amorphous vinyl alcohol matrix. 5. The extruded water-soluble article of claim 1, wherein the extrusion temperature is 90 to 125° C. 6. The extruded water-soluble article of claim 1, wherein the one or more thermal active agents are in a total amount of 1% to 20% by weight of the article. 7. The extruded water-soluble article of claim 1 further comprising up to 50% thermoplastic starch by weight. 8. The extruded water-soluble article of claim 1 further comprising up to 30% by weight of ethylene vinyl acetate. 9. The extruded water-soluble article of claim 1, wherein the article is a mono-layer or multi-layer film. 10. The extruded water-soluble article of claim 9, wherein the film has a water dissolution speed from 5 seconds to 30 minutes as determined by the Dissolution Test of the present disclosure. 11. The extruded water-soluble article of claim 9, wherein the film has a basis weight of 5 gsm to 500 gsm. 12. The extruded water-soluble article of claim 9, wherein the film has an elongation of 5% to 500% according to the Tensile Test of the present disclosure. 13. The extruded water-soluble article of claim 1, wherein the one or more thermal active agents are menthol, a menthol derivative thereof, or a combination thereof. 14. The extruded water-soluble article of claim 1 wherein the one or more thermal active agents are configured to either stimulate human sensory receptors or change the temperature of the skin upon contact therewith. 15. The extruded water-soluble article of claim 1 wherein the one or more thermal active agents comprise a temperature agent selected from menthol, menthol derivatives, xylitol, capsaicin, polyols, urea, self-heating zeolite or alkali metal-compounds, magnesium chloride, magnesium sulfate and combinations thereof. 16. A personal absorbent article comprising: an absorbent member sandwiched between a water-impermeable backsheet and a water-permeable liner, wherein the liner has a body-facing surface and an opposite outward-facing surface; and a film attached to either the outward-facing surface of the liner or a surface of the absorbent member that is adjacent the liner, wherein the film comprises: a water-soluble, polymer having an extrusion temperature of 90 to 150° C.; a plasticizer; and one or more thermal active agents in a total amount of 0.1% to 50% by weight of the article, wherein the one or more thermal active ingredients are dissolvable/dispersible in the plasticizer; wherein the combination of the one or more thermal active agents and the plasticizer is a homogeneous mixture/solution; and wherein a homogeneous blend comprising the polymer and the homogeneous mixture/solution has an extrusion temperature of 50 to 125° C. 17. The personal absorbent article of claim 16, wherein the personal absorbent article is a bandage, a medical drape, a wipe, a sheet, a diaper, a pant, or a pad. 18. A method of making an extruded article comprising the following steps: a) prepare a mixture/solution comprising a thermoplastic plasticizer and one or more thermal active agents; b) form a homogenous blend having an extrusion and a processing temperature of 90 to 125° C., the homogeneous blend comprising the mixture/solution and an amorphous, water-soluble polyvinyl alcohol, wherein the homogeneous blend comprises 0.1% to 40% by weight of the one or more thermal active agents; and c) extrude the homogeneous blend to form a film. 19. The method of claim 18, wherein the homogenous blend can be extruded directly onto a carrier substrate, forming a bond therebetween. 20. The method of claim 18, wherein one or more secondary active agents are co-extruded with the homogeneous blend.
1,600
276
276
14,353,833
1,613
The invention provides a method of treating rhinitis. The method comprises administering an effective amount of a pharmaceutical composition comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP) formulated for nasal administration. The invention also provides a pharmaceutical product comprising a DA-DKP containing composition.
1. A method of treating rhinitis comprising administering an effective amount of a pharmaceutical composition comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP), to an animal in need thereof. 2. The method of claim 1, wherein the rhinitis is allergic rhinitis. 3. The method of claim 1, wherein the effective amount of the DA-DKP in the composition is from about 100 μg to about 3000 μg per day. 4. The method of claim 1, wherein the effective amount of the DA-DKP in the composition is from about from about 500 μg to about 1500 μg per day. 5. The method of claim 1, wherein administration of the composition comprising DA-DKP, is commenced within 24 hours of diagnosis of rhinitis. 6. The method of claim 1, wherein administration of the composition comprising DA-DKP, is commenced at the appearance of one or more early signs of, or a predisposition to develop, rhinitis. 7. The method of claim 6, wherein the one or more early signs of rhinitis are selected from the group consisting of rhinorrhea, nasal congestion, nasal itching and sneezing. 8. The method of claim 1, wherein the DA-DKP is in a composition prepared by removing albumin from a solution of a human serum albumin composition. 9. The method of claim 8, wherein the step of removing the albumin comprises treating the human serum albumin composition by a separation method selected from the group consisting of ultrafiltration, sucrose gradient centrifugation, chromatography, salt precipitation, and sonication. 10. The method of claim 9, wherein the step of removing comprises passing the human serum albumin composition over an ultarfiltration membrane with a molecular weight cut off that retains the albumin, and wherein the resulting filtrate comprises DA-DKP. 11. The method of claim 10, wherein the ultrafiltration membrane has molecular weight cutoff of less than 50 kDa, less than 40 kDa, less than 30 kDA, less than 20 kDa, less than 10 kDa, less than 5 kDa or less than 3 kDA. 12. The method of claim 1, wherein the composition comprising DA-DKP is administered in combination with a second drug suitable for treating rhinitis. 13. The method of claim 12, wherein the second drug suitable for treating rhinitis is selected from the group consisting of antihistamines, decongestants, anti-inflammatories, mast cell stabilizers, leukotriene modifiers and IgE blockers. 14. A pharmaceutical product, comprising a DA-DKP containing composition formulated for administration by a route selected from the group consisting of inhalation, insufflation and nasal administration to the nose. 15. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition is formulated for administration by inhalation is packaged in a device selected from the group consisting of insufflators, nebulizers, pressurized packs, squeeze bottle, a syringe, a dropper, a spray device, an atomizer device, and an aerosolizer. 16. The pharmaceutical product of claim 15, wherein the pressurized pack comprises a propellant selected from the group consisting of dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, and carbon dioxide. 17. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition formulated for administration by inhalation or insufflation comprises a powder mix of the DA-DKP containing composition and a powder base. 18. The pharmaceutical product of claim 17, wherein the powder mix is in a dosage form selected from the group consisting of capsules, cartridges, gelatin packs and blister packs. 19. The pharmaceutical product of claim 18, wherein the powder mix is delivered by a device selected from the group consisting of an inhalator, insufflator and metered-dose inhaler. 20. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition formulated for nasal administration is in a form of drops or sprays. 21. The pharmaceutical product of claim 20, wherein the DA-DKP comprises from about 0.1% (w/v) to about 10% (w/v) of the composition. 22. The pharmaceutical product of claim 20, wherein the drops or sprays are contained within an intranasal delivery system. 23. The pharmaceutical product of claim 22, wherein the intranasal delivery system comprises an atomizing device. 24. The pharmaceutical product of claim 23, wherein the atomizing device comprises a bottle and a pump. 25. The pharmaceutical product of claim 24, wherein the pump is a metered dose pump. 26. The pharmaceutical product of claim 25, wherein the metered dose pump delivers an intranasal volume of the DA-DKP containing composition of about 0.15 ml per pump. 27. The pharmaceutical product of claim 20, further comprising an aqueous or non-aqueous base comprising one or more agents selected from the group consisting of dispersing agents, solubilizing agents, and suspending agents. 28. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition formulated for nasal administration is in a form selected from the group consisting of ointments, gels and creams. 29. The pharmaceutical product of claim 28, wherein the DA-DKP containing composition further comprises excipients selected from the group consisting of animal fats, vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycol, silicones, bentonites, silicic acid, talc, zinc oxide and mixtures thereof. 30. The pharmaceutical product of claim 14, further comprising an absorption or permeation enhancer. 31. The pharmaceutical product of claim 14, further comprising a thickening agent or viscosity enhancer to increase the residence time of the DA-DKP containing composition in the nose. 32. The pharmaceutical product of claim 14, further comprising a pharmaceutically-acceptable carrier. 33. The pharmaceutical product of claim 14, wherein the DA-DKP is prepared by removing albumin from a solution of a human serum albumin composition. 34. The pharmaceutical product of claim 33, wherein the step of removing the albumin comprises treating the human serum albumin composition by a separation method selected from the group consisting of ultrafiltration, sucrose gradient centrifugation, chromatography, salt precipitation, and sonication. 35. The pharmaceutical product of claim 34, wherein the step of removing comprises passing the human serum albumin composition over an ultarfiltration membrane with a molecular weight cut off that retains the albumin, and wherein the resulting filtrate comprises DA-DKP. 36. The pharmaceutical product of claim 35, wherein the ultrafiltration membrane has molecular weight cutoff of less than 50 kDa, less than 40 kDa, less than 30 kDA, less than 20 kDa, less than 10 kDa, less than 5 kDa or less than 3 kDA.
The invention provides a method of treating rhinitis. The method comprises administering an effective amount of a pharmaceutical composition comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP) formulated for nasal administration. The invention also provides a pharmaceutical product comprising a DA-DKP containing composition.1. A method of treating rhinitis comprising administering an effective amount of a pharmaceutical composition comprising a diketopiperazine with amino acid side chains of aspartic acid and alanine (DA-DKP), to an animal in need thereof. 2. The method of claim 1, wherein the rhinitis is allergic rhinitis. 3. The method of claim 1, wherein the effective amount of the DA-DKP in the composition is from about 100 μg to about 3000 μg per day. 4. The method of claim 1, wherein the effective amount of the DA-DKP in the composition is from about from about 500 μg to about 1500 μg per day. 5. The method of claim 1, wherein administration of the composition comprising DA-DKP, is commenced within 24 hours of diagnosis of rhinitis. 6. The method of claim 1, wherein administration of the composition comprising DA-DKP, is commenced at the appearance of one or more early signs of, or a predisposition to develop, rhinitis. 7. The method of claim 6, wherein the one or more early signs of rhinitis are selected from the group consisting of rhinorrhea, nasal congestion, nasal itching and sneezing. 8. The method of claim 1, wherein the DA-DKP is in a composition prepared by removing albumin from a solution of a human serum albumin composition. 9. The method of claim 8, wherein the step of removing the albumin comprises treating the human serum albumin composition by a separation method selected from the group consisting of ultrafiltration, sucrose gradient centrifugation, chromatography, salt precipitation, and sonication. 10. The method of claim 9, wherein the step of removing comprises passing the human serum albumin composition over an ultarfiltration membrane with a molecular weight cut off that retains the albumin, and wherein the resulting filtrate comprises DA-DKP. 11. The method of claim 10, wherein the ultrafiltration membrane has molecular weight cutoff of less than 50 kDa, less than 40 kDa, less than 30 kDA, less than 20 kDa, less than 10 kDa, less than 5 kDa or less than 3 kDA. 12. The method of claim 1, wherein the composition comprising DA-DKP is administered in combination with a second drug suitable for treating rhinitis. 13. The method of claim 12, wherein the second drug suitable for treating rhinitis is selected from the group consisting of antihistamines, decongestants, anti-inflammatories, mast cell stabilizers, leukotriene modifiers and IgE blockers. 14. A pharmaceutical product, comprising a DA-DKP containing composition formulated for administration by a route selected from the group consisting of inhalation, insufflation and nasal administration to the nose. 15. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition is formulated for administration by inhalation is packaged in a device selected from the group consisting of insufflators, nebulizers, pressurized packs, squeeze bottle, a syringe, a dropper, a spray device, an atomizer device, and an aerosolizer. 16. The pharmaceutical product of claim 15, wherein the pressurized pack comprises a propellant selected from the group consisting of dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, and carbon dioxide. 17. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition formulated for administration by inhalation or insufflation comprises a powder mix of the DA-DKP containing composition and a powder base. 18. The pharmaceutical product of claim 17, wherein the powder mix is in a dosage form selected from the group consisting of capsules, cartridges, gelatin packs and blister packs. 19. The pharmaceutical product of claim 18, wherein the powder mix is delivered by a device selected from the group consisting of an inhalator, insufflator and metered-dose inhaler. 20. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition formulated for nasal administration is in a form of drops or sprays. 21. The pharmaceutical product of claim 20, wherein the DA-DKP comprises from about 0.1% (w/v) to about 10% (w/v) of the composition. 22. The pharmaceutical product of claim 20, wherein the drops or sprays are contained within an intranasal delivery system. 23. The pharmaceutical product of claim 22, wherein the intranasal delivery system comprises an atomizing device. 24. The pharmaceutical product of claim 23, wherein the atomizing device comprises a bottle and a pump. 25. The pharmaceutical product of claim 24, wherein the pump is a metered dose pump. 26. The pharmaceutical product of claim 25, wherein the metered dose pump delivers an intranasal volume of the DA-DKP containing composition of about 0.15 ml per pump. 27. The pharmaceutical product of claim 20, further comprising an aqueous or non-aqueous base comprising one or more agents selected from the group consisting of dispersing agents, solubilizing agents, and suspending agents. 28. The pharmaceutical product of claim 14, wherein the DA-DKP containing composition formulated for nasal administration is in a form selected from the group consisting of ointments, gels and creams. 29. The pharmaceutical product of claim 28, wherein the DA-DKP containing composition further comprises excipients selected from the group consisting of animal fats, vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycol, silicones, bentonites, silicic acid, talc, zinc oxide and mixtures thereof. 30. The pharmaceutical product of claim 14, further comprising an absorption or permeation enhancer. 31. The pharmaceutical product of claim 14, further comprising a thickening agent or viscosity enhancer to increase the residence time of the DA-DKP containing composition in the nose. 32. The pharmaceutical product of claim 14, further comprising a pharmaceutically-acceptable carrier. 33. The pharmaceutical product of claim 14, wherein the DA-DKP is prepared by removing albumin from a solution of a human serum albumin composition. 34. The pharmaceutical product of claim 33, wherein the step of removing the albumin comprises treating the human serum albumin composition by a separation method selected from the group consisting of ultrafiltration, sucrose gradient centrifugation, chromatography, salt precipitation, and sonication. 35. The pharmaceutical product of claim 34, wherein the step of removing comprises passing the human serum albumin composition over an ultarfiltration membrane with a molecular weight cut off that retains the albumin, and wherein the resulting filtrate comprises DA-DKP. 36. The pharmaceutical product of claim 35, wherein the ultrafiltration membrane has molecular weight cutoff of less than 50 kDa, less than 40 kDa, less than 30 kDA, less than 20 kDa, less than 10 kDa, less than 5 kDa or less than 3 kDA.
1,600
277
277
14,790,247
1,643
Isolated monoclonal antibodies which bind to human CD27 and related antibody-based compositions and molecules are disclosed. Also disclosed are therapeutic and diagnostic methods for using the antibodies.
1. A method for inducing or enhancing an immune response against an antigen in a subject comprising administering to the subject a monoclonal antibody which binds to human CD27, in an amount effective to induce or enhance an immune response against an antigen, wherein the antibody comprises a heavy chain variable region CDR1 comprising SEQ ID NO: 38; a heavy chain variable region CDR2 comprising SEQ ID NO: 39; a heavy chain variable region CDR3 comprising SEQ ID NO: 40; a light chain variable region CDR1 comprising SEQ ID NO: 44; a light chain variable region CDR2 comprising SEQ ID NO: 45; and a light chain variable region CDR3 comprising SEQ ID NO: 46. 2. A method of inhibiting growth of CD27 expressing cells comprising contacting the cells with a monoclonal antibody which binds to human CD27, in an amount effective to inhibit growth of CD27 expressing cells, wherein the antibody comprises a heavy chain variable region CDR1 comprising SEQ ID NO: 38; a heavy chain variable region CDR2 comprising SEQ ID NO: 39; a heavy chain variable region CDR3 comprising SEQ ID NO: 40; a light chain variable region CDR1 comprising SEQ ID NO: 44; a light chain variable region CDR2 comprising SEQ ID NO: 45; and a light chain variable region CDR3 comprising SEQ ID NO: 46. 3. A method of inhibiting the binding of CD70 to CD27 on cells in a subject having a disorder by administering to the subject a monoclonal antibody which binds to human CD27, wherein the antibody comprises a heavy chain variable region CDR1 comprising SEQ ID NO: 38; a heavy chain variable region CDR2 comprising SEQ ID NO: 39; a heavy chain variable region CDR3 comprising SEQ ID NO: 40; a light chain variable region CDR1 comprising SEQ ID NO: 44; a light chain variable region CDR2 comprising SEQ ID NO: 45; and a light chain variable region CDR3 comprising SEQ ID NO: 46. 4. The method of any one of claims 1-3, wherein the antibody comprises a heavy chain variable region comprising SEQ ID NO:37. 5. The method of any one of claims 1-3, wherein the antibody comprises a light chain variable region comprising SEQ ID NO:43. 6. The method of any one of claims 1-3, wherein the antibody comprises a heavy chain variable region comprising SEQ ID NO:37 and a light chain variable region comprising SEQ ID NO:43. 7. The method of claim 1, wherein the immune response is an antigen-specific T cell response.
Isolated monoclonal antibodies which bind to human CD27 and related antibody-based compositions and molecules are disclosed. Also disclosed are therapeutic and diagnostic methods for using the antibodies.1. A method for inducing or enhancing an immune response against an antigen in a subject comprising administering to the subject a monoclonal antibody which binds to human CD27, in an amount effective to induce or enhance an immune response against an antigen, wherein the antibody comprises a heavy chain variable region CDR1 comprising SEQ ID NO: 38; a heavy chain variable region CDR2 comprising SEQ ID NO: 39; a heavy chain variable region CDR3 comprising SEQ ID NO: 40; a light chain variable region CDR1 comprising SEQ ID NO: 44; a light chain variable region CDR2 comprising SEQ ID NO: 45; and a light chain variable region CDR3 comprising SEQ ID NO: 46. 2. A method of inhibiting growth of CD27 expressing cells comprising contacting the cells with a monoclonal antibody which binds to human CD27, in an amount effective to inhibit growth of CD27 expressing cells, wherein the antibody comprises a heavy chain variable region CDR1 comprising SEQ ID NO: 38; a heavy chain variable region CDR2 comprising SEQ ID NO: 39; a heavy chain variable region CDR3 comprising SEQ ID NO: 40; a light chain variable region CDR1 comprising SEQ ID NO: 44; a light chain variable region CDR2 comprising SEQ ID NO: 45; and a light chain variable region CDR3 comprising SEQ ID NO: 46. 3. A method of inhibiting the binding of CD70 to CD27 on cells in a subject having a disorder by administering to the subject a monoclonal antibody which binds to human CD27, wherein the antibody comprises a heavy chain variable region CDR1 comprising SEQ ID NO: 38; a heavy chain variable region CDR2 comprising SEQ ID NO: 39; a heavy chain variable region CDR3 comprising SEQ ID NO: 40; a light chain variable region CDR1 comprising SEQ ID NO: 44; a light chain variable region CDR2 comprising SEQ ID NO: 45; and a light chain variable region CDR3 comprising SEQ ID NO: 46. 4. The method of any one of claims 1-3, wherein the antibody comprises a heavy chain variable region comprising SEQ ID NO:37. 5. The method of any one of claims 1-3, wherein the antibody comprises a light chain variable region comprising SEQ ID NO:43. 6. The method of any one of claims 1-3, wherein the antibody comprises a heavy chain variable region comprising SEQ ID NO:37 and a light chain variable region comprising SEQ ID NO:43. 7. The method of claim 1, wherein the immune response is an antigen-specific T cell response.
1,600
278
278
14,423,062
1,634
The present invention pertains to a method for isolation and purification of nucleic acids from a stabilized sample or portion or fraction thereof, wherein the sample stabilization involved the use of at least one formaldehyde releaser and wherein the isolation of the nucleic acids from the stabilized sample or portion or fraction thereof involves the use of at least one cationic detergent during lysis.
1. A method for isolating nucleic acids from a stabilized sample or portion or fraction thereof, wherein the sample stabilization involved the use of at least one formaldehyde releaser, comprising: (a) lysing the stabilized sample or portion or fraction thereof in the presence of at least one cationic detergent to provide a lysed sample, and (b) isolating nucleic acids from the lysed sample. 2. The method according to claim 1, wherein the cationic detergent is selected from the following group of cationic detergents: a) a cationic compound of the general formula (1): Y′R1R2R3R4X−  (1) wherein Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched Ci-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; b) a detergent comprising under the used lysis conditions a charged quaternary ammonium cation as polar head group; c) a cationic detergent obtained in a compostion comprising (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C20 alkyl residue, C6-C26 aryl residue or C6-C26 aralkyl residue, preferably H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt; d) a cationic detergent obtained from an amino surfactant selected from the group consisting of the protonated forms of dodecylamine, N-methyldodecylamine, N,N-dimethyldodecylamjne, N,N-dimethyldodecylamine N oxide and 4-tetradecylaniline; e) a cationic detergent comprising a permanently charged quaternary ammonium cation as polar head group; and/or f) a cationic detergent selected from the group consisting of cetyl trimethyl ammonium bromide (CTAB), tetra decyl trimethyl ammonium bromide (TTAB) and dodecyl trimethyl ammonium bromide (DTRB) or the corresponding compounds comprising a chloride instead of the bromide. 3. The method according to claim 1, wherein step (a) comprises contacting the stabilized sample or portion or fraction thereof with a) a lysis composition comprising (i) a cationic compound of the general formula (1): Y+R1R2R3R4X−  (1) wherein Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched C1-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; and (ii) at least one proton donor; or b) a lysis composition comprising (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt. 4. The method according to claim 1, further comprising: obtaining the stabilized sample or portion or fraction thereof prior to step (a), wherein step (a) comprises contacting the stabilized sample or portion or fraction thereof with at least one cationic detergent to provide a lysed sample; 5. The method according to claim 4, wherein step (a) comprises: (a)(1)obtaining cells from the stabilized sample; and (a)(2)contacting the cells with at least one cationic detergent for lysis and providing to provide a lysed sample; and 6. The method according to claim 4, wherein step b(a) has one or more of the following characteristics: i) step (a) comprises incubating the composition comprising the stabilized sample or portion or fraction of the stabilized sample, the at least one cationic detergent and optionally one or more further lysis agents to provide the lysed sample; ii) step (a) comprises obtaining a nucleic acid containing portion from the lysed sample and subjecting said nucleic acid containing portion to the nucleic acid isolation step (b); and/or iii) the concentration of the cationic detergent in the lysis composition that is obtained when contacting the stabilized sample or portion or fraction thereof with the cationic detergent and optionally one or more further lysis agents is selected from a range of 0.25% (w/v) to 30% (w/v), or 0.5% (w/v) to 15% (w/v). 7. The method according to claim 4, wherein step e-cb) has one or more of the following characteristics: i) step (b) comprises isolating nucleic acids from the lysed sample or from a nucleic acid containing portion obtained from the lysed sample; ii) step (b) comprises contacting the lysed sample or the nucleic acid containing portion obtained from the lysed sample with one or more additional lysing agents thereby providing a lysis mixture; iii) step (b) comprises using a nucleic acid binding solid phase; and/or step (b) comprises using ef-at least one chaotropic salt and/or alcohol. 8. The method according to claim 4, wherein step (b) comprises the following steps: i) contacting the lysed sample or a nucleic acid containing portion obtained from the lysed sample with (aa) at least one chaotropic agent, (bb) at least one proteolytic enzyme, and/or (cc) one or more salts, thereby providing a lysis mixture; ii) binding nucleic acids contained in the lysis mixture to a nucleic acid binding solid phase, wherein in step ii) optionally the binding conditions are adjusted by adding a binding composition; iii) separating the solid phase with the bound nucleic acids from the remaining sample; and iv) optionally washing the nucleic acids; and v) optionally eluting nucleic acids from the solid phase. 9. The method according to claim 1, wherein the formaldehyde releaser used for stabilization of the sample has one or more of the following characteristics: a) the formaldehyde releaser is a chemical fixative; b) the formaldehyde releaser is selected from the group consisting of diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1 aza-3,7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1 aza-3,7dioxabicyclo[3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1 aza-3,7dioxabicyclo[3.3.0]octane, quaternary adamantine and any combination of the foregoing; c) the formaldehyde releaser is a heterocyclic urea, diazolidinyl urea and/or imidazolidinyl urea; and/or d) the formaldehyde releaser is diazolidinyl urea. 10. The method according to claim 1, wherein the sample was stabilized additionally using one or more of the following or wherein the sample was stabilized using a stabilization composition additionally comprising one or more of the following: a) one or more enzyme inhibitors, wherein said enzyme inhibitor has one or more of the following characteristics: i) the enzyme inhibitor is a nuclease inhibitor; ii) the enzyme inhibitor is selected from the group consisting of: dithiothreitol (DTT), iodoacetamide, iodoacetic acid, heparin, chitosan, cobalt chloride, diethyl pyrocarbonate, ethanol, aurintricarboxylic acid (ATA), glyceraldehydes, sodium fluoride, ethylenediamine tetraacetic acid (EDTA), formamtde, vanadyl-ribonucleoside complexes, macaloid, hydroxylamine-oxygen-cupric ion, bentonite, ammonium sulfate, beta-mercaptoethanol, cysteine, dithioerythritol, tris(2-carboxyethyl) phosphene hydrochloride, a divalent cation and any combination of the foregoing; and/or iii) the enzyme inhibitor is aurintricarboxylic acid; b) one or more metabolic inhibitors having one or more of the following characteristics: i) the metabolic inhibitor is selected from the group consisting of: dihydroxyacetone phosphate, glyceraldehyde 3-phosphate, 1,3-bisphosphoglycerate, 3-phosphoglycerate, 2-phosphoglycerate, phosphoenolpyruvate, pyruvate and glycerate dihydroxyacetate, sodium fluoride, K2C2O4 and any combination of the foregoing; ii) the metabolic inhibitor is glyceraldehyde; iii) the metabolic inhibitor is sodium fluoride; and/or iv) the metabolic inhibitor is a combination of glyceraldehyde and sodium fluoride; and/or c) one or more metal ion chelators wherein the metal ion chelator is selected from the group consisting of ethylene glycol tetraacetic acid (EGTA), 1,2-bis-(o-Aminophenoxy)-ethane-N′,N′,-N′,N′-tetraacetic acid tetraacetoxy-Methyl ester (BAPTA-AM), dietyldithiocarbamate (DEDTC), ethylenediaminetetraacetic acid (EDTA), dicarboxymethyl-glutamic acid, nitrilotriacetic acid (NTA), ethylenediaminedisuccinic acid (EDDS), EDTA, citrat and any combination of the foregoing, preferably the metal chelator is EDTA. 11. The method according to claim 1, wherein the sample was stabilized using a stabilization composition comprising: a) a formaldehyde releaser agent selected from a chemical fixative that contains urea; and at least one, two or preferably all of the following components: b) an enzyme inhibitor; a metabolic inhibitor; and c) a metal ion chelator. 12. The method according to claim 1, wherein the sample or portion or fraction thereof has one or more of the following characteristics: a) it comprises cells; b) it is selected from the group consisting of whole blood, plasma, serum, lymphatic fluid, urine, liquor, ascites, milk, stool, bronchial lavage, saliva, bone marrow aspirates, amniotic fluid, semen/seminal fluid, swabs/smears, body fluids, body secretions, nasal secretions, vaginal secretions, wound secretions and excretions, cell suspensions, cell culture and cell culture supernatants; c) it is a cell-free, cell-depleted or cell containing body fluid sample; and/or d) it is whole blood. 13. The method according to claim 1, having one or more of the following characteristics: a) the sample is stabilized by mixing the sample with the stabilization composition directly after and/or during the collection of the sample, thereby providing a stabilized sample; b) step (a) comprises isolating cells arc isolated from the stabilized sample, and step (b) comprises isolating nucleic acids from the cells; c) step (b) comprises or the method further comprises isolating nucleic acids from a cell-free or cell-reduced portion of the stabilized sample; and/or d) the method further comprises processing and/or analyzing the isolated nucleic acids in a further step; and/or e) the method further comprises analyzing the isolated nucleic acids to identify, detect, screen for, monitor or exclude a disease or infection. 14. The method according to claim 1, wherein the sample is blood, the blood stabilization involves the use of at least one formaldehyde releaser and at least one anticoagulant, step (a) comprises: (a)(1) obtaining the stabilized blood sample or a portion or fraction thereof wherein the portion or fraction of the stabilised blood sample is selected from blood cells; and (a)(2) contacting the stabilized blood sample or portion or fraction thereof with at least one cationic to provide a lysed sample; and the nucleic acids isolated in step (b) compris or consist of RNA. 15. The method according to claim 14, wherein the formaldehyde releaser is selected from a heterocyclic urea, diazolidinyl urea and/or imidazolidinyl urea, and in step a)(2), the cationic detergent is selected from the group consisting of: a) a cationic compound of the general formula (1): Y+R1R2R3R4X−  (1) wherein, Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched C1-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; b) a detergent comprising under the used lysis conditions a charged quaternary ammonium cation as polar head group; c) a cationic detergent obtained in a composition comprising (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C20 alkyl residue, C6-C26 aryl residue or C6-C26 aralkyl residue, preferably H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt; d) a cationic detergent obtained from an amino surfactant selected from the group consisting of the protonated forms of dodecylamine, N-methyldodecylamine, N,N-dimethyldodecylamjne, N,N-dimethyldodecylamine N oxide and 4-tetradecylaniline; e) a cationic detergent comprising a permanently charged quaternary ammonium cation as polar head group; and/or f) a cationic detergent selected from the group consisting of cetyl trimethyl ammonium bromide (CTAB), tetra decyl trimethyl ammonium bromide (TTAB) and dodecyl trimethyl ammonium bromide (DTRB) or the corresponding compounds comprising a chloride instead of the bromide; or step (a)(1) uses a lysis composition comprising: (i) a cationic compound of the general formula (1): Y+R1R2R3R4X−  (1) wherein Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched C1-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; and (ii) at least one proton donor; or (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt; step (a) further comprises incubating the composition comprising the stabilized sample or portion or fraction of the stabilized sample, the at least one cationic detergent and optionally one or more further lysis agents to provide the lysed sample; and step (b) comprises the following steps: i. contacting the lysed sample or a nucleic acid containing portion obtained from the lysed sample with one or more additional lysing agents thereby providing a lysis mixture; optionally removing DNA from the lysis mixture; ii. adding alcohol to the lysis mixture to adjust the binding conditions and binding RNA to a nucleic acid binding solid phase; iii. separating the solid phase with the bound RNA from the remaining sample; and iv. optionally washing the RNA and v. optionally eluting RNA from the solid phase. 16. The method according to claim 3, wherein Y represents nitrogen. 17. The method according to claim 8, wherein the chaotropic agent is a chaotropic salt and/or wherein said binding compositions comprises a chaotropic salt and/or alcohol. 18. The method according to claim 11, wherein the formaldehyde releaser agent is diazolidinyl urea and/or imidazolidinyl urea. 19. The method according to claim 11, wherein the metabolic inhibitor is glyceraldehyde and/or sodium fluoride. 20. The method according to claim 15, wherein step (b) i. comprises contacting the lysed sample or the nucleic acid containing portion obtained from the lysed sample with (aa) at least one chaotropic agent, (bb) at least one proteolytic enzyme; and/or (cc) one or more salts.
The present invention pertains to a method for isolation and purification of nucleic acids from a stabilized sample or portion or fraction thereof, wherein the sample stabilization involved the use of at least one formaldehyde releaser and wherein the isolation of the nucleic acids from the stabilized sample or portion or fraction thereof involves the use of at least one cationic detergent during lysis.1. A method for isolating nucleic acids from a stabilized sample or portion or fraction thereof, wherein the sample stabilization involved the use of at least one formaldehyde releaser, comprising: (a) lysing the stabilized sample or portion or fraction thereof in the presence of at least one cationic detergent to provide a lysed sample, and (b) isolating nucleic acids from the lysed sample. 2. The method according to claim 1, wherein the cationic detergent is selected from the following group of cationic detergents: a) a cationic compound of the general formula (1): Y′R1R2R3R4X−  (1) wherein Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched Ci-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; b) a detergent comprising under the used lysis conditions a charged quaternary ammonium cation as polar head group; c) a cationic detergent obtained in a compostion comprising (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C20 alkyl residue, C6-C26 aryl residue or C6-C26 aralkyl residue, preferably H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt; d) a cationic detergent obtained from an amino surfactant selected from the group consisting of the protonated forms of dodecylamine, N-methyldodecylamine, N,N-dimethyldodecylamjne, N,N-dimethyldodecylamine N oxide and 4-tetradecylaniline; e) a cationic detergent comprising a permanently charged quaternary ammonium cation as polar head group; and/or f) a cationic detergent selected from the group consisting of cetyl trimethyl ammonium bromide (CTAB), tetra decyl trimethyl ammonium bromide (TTAB) and dodecyl trimethyl ammonium bromide (DTRB) or the corresponding compounds comprising a chloride instead of the bromide. 3. The method according to claim 1, wherein step (a) comprises contacting the stabilized sample or portion or fraction thereof with a) a lysis composition comprising (i) a cationic compound of the general formula (1): Y+R1R2R3R4X−  (1) wherein Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched C1-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; and (ii) at least one proton donor; or b) a lysis composition comprising (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt. 4. The method according to claim 1, further comprising: obtaining the stabilized sample or portion or fraction thereof prior to step (a), wherein step (a) comprises contacting the stabilized sample or portion or fraction thereof with at least one cationic detergent to provide a lysed sample; 5. The method according to claim 4, wherein step (a) comprises: (a)(1)obtaining cells from the stabilized sample; and (a)(2)contacting the cells with at least one cationic detergent for lysis and providing to provide a lysed sample; and 6. The method according to claim 4, wherein step b(a) has one or more of the following characteristics: i) step (a) comprises incubating the composition comprising the stabilized sample or portion or fraction of the stabilized sample, the at least one cationic detergent and optionally one or more further lysis agents to provide the lysed sample; ii) step (a) comprises obtaining a nucleic acid containing portion from the lysed sample and subjecting said nucleic acid containing portion to the nucleic acid isolation step (b); and/or iii) the concentration of the cationic detergent in the lysis composition that is obtained when contacting the stabilized sample or portion or fraction thereof with the cationic detergent and optionally one or more further lysis agents is selected from a range of 0.25% (w/v) to 30% (w/v), or 0.5% (w/v) to 15% (w/v). 7. The method according to claim 4, wherein step e-cb) has one or more of the following characteristics: i) step (b) comprises isolating nucleic acids from the lysed sample or from a nucleic acid containing portion obtained from the lysed sample; ii) step (b) comprises contacting the lysed sample or the nucleic acid containing portion obtained from the lysed sample with one or more additional lysing agents thereby providing a lysis mixture; iii) step (b) comprises using a nucleic acid binding solid phase; and/or step (b) comprises using ef-at least one chaotropic salt and/or alcohol. 8. The method according to claim 4, wherein step (b) comprises the following steps: i) contacting the lysed sample or a nucleic acid containing portion obtained from the lysed sample with (aa) at least one chaotropic agent, (bb) at least one proteolytic enzyme, and/or (cc) one or more salts, thereby providing a lysis mixture; ii) binding nucleic acids contained in the lysis mixture to a nucleic acid binding solid phase, wherein in step ii) optionally the binding conditions are adjusted by adding a binding composition; iii) separating the solid phase with the bound nucleic acids from the remaining sample; and iv) optionally washing the nucleic acids; and v) optionally eluting nucleic acids from the solid phase. 9. The method according to claim 1, wherein the formaldehyde releaser used for stabilization of the sample has one or more of the following characteristics: a) the formaldehyde releaser is a chemical fixative; b) the formaldehyde releaser is selected from the group consisting of diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1 aza-3,7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1 aza-3,7dioxabicyclo[3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1 aza-3,7dioxabicyclo[3.3.0]octane, quaternary adamantine and any combination of the foregoing; c) the formaldehyde releaser is a heterocyclic urea, diazolidinyl urea and/or imidazolidinyl urea; and/or d) the formaldehyde releaser is diazolidinyl urea. 10. The method according to claim 1, wherein the sample was stabilized additionally using one or more of the following or wherein the sample was stabilized using a stabilization composition additionally comprising one or more of the following: a) one or more enzyme inhibitors, wherein said enzyme inhibitor has one or more of the following characteristics: i) the enzyme inhibitor is a nuclease inhibitor; ii) the enzyme inhibitor is selected from the group consisting of: dithiothreitol (DTT), iodoacetamide, iodoacetic acid, heparin, chitosan, cobalt chloride, diethyl pyrocarbonate, ethanol, aurintricarboxylic acid (ATA), glyceraldehydes, sodium fluoride, ethylenediamine tetraacetic acid (EDTA), formamtde, vanadyl-ribonucleoside complexes, macaloid, hydroxylamine-oxygen-cupric ion, bentonite, ammonium sulfate, beta-mercaptoethanol, cysteine, dithioerythritol, tris(2-carboxyethyl) phosphene hydrochloride, a divalent cation and any combination of the foregoing; and/or iii) the enzyme inhibitor is aurintricarboxylic acid; b) one or more metabolic inhibitors having one or more of the following characteristics: i) the metabolic inhibitor is selected from the group consisting of: dihydroxyacetone phosphate, glyceraldehyde 3-phosphate, 1,3-bisphosphoglycerate, 3-phosphoglycerate, 2-phosphoglycerate, phosphoenolpyruvate, pyruvate and glycerate dihydroxyacetate, sodium fluoride, K2C2O4 and any combination of the foregoing; ii) the metabolic inhibitor is glyceraldehyde; iii) the metabolic inhibitor is sodium fluoride; and/or iv) the metabolic inhibitor is a combination of glyceraldehyde and sodium fluoride; and/or c) one or more metal ion chelators wherein the metal ion chelator is selected from the group consisting of ethylene glycol tetraacetic acid (EGTA), 1,2-bis-(o-Aminophenoxy)-ethane-N′,N′,-N′,N′-tetraacetic acid tetraacetoxy-Methyl ester (BAPTA-AM), dietyldithiocarbamate (DEDTC), ethylenediaminetetraacetic acid (EDTA), dicarboxymethyl-glutamic acid, nitrilotriacetic acid (NTA), ethylenediaminedisuccinic acid (EDDS), EDTA, citrat and any combination of the foregoing, preferably the metal chelator is EDTA. 11. The method according to claim 1, wherein the sample was stabilized using a stabilization composition comprising: a) a formaldehyde releaser agent selected from a chemical fixative that contains urea; and at least one, two or preferably all of the following components: b) an enzyme inhibitor; a metabolic inhibitor; and c) a metal ion chelator. 12. The method according to claim 1, wherein the sample or portion or fraction thereof has one or more of the following characteristics: a) it comprises cells; b) it is selected from the group consisting of whole blood, plasma, serum, lymphatic fluid, urine, liquor, ascites, milk, stool, bronchial lavage, saliva, bone marrow aspirates, amniotic fluid, semen/seminal fluid, swabs/smears, body fluids, body secretions, nasal secretions, vaginal secretions, wound secretions and excretions, cell suspensions, cell culture and cell culture supernatants; c) it is a cell-free, cell-depleted or cell containing body fluid sample; and/or d) it is whole blood. 13. The method according to claim 1, having one or more of the following characteristics: a) the sample is stabilized by mixing the sample with the stabilization composition directly after and/or during the collection of the sample, thereby providing a stabilized sample; b) step (a) comprises isolating cells arc isolated from the stabilized sample, and step (b) comprises isolating nucleic acids from the cells; c) step (b) comprises or the method further comprises isolating nucleic acids from a cell-free or cell-reduced portion of the stabilized sample; and/or d) the method further comprises processing and/or analyzing the isolated nucleic acids in a further step; and/or e) the method further comprises analyzing the isolated nucleic acids to identify, detect, screen for, monitor or exclude a disease or infection. 14. The method according to claim 1, wherein the sample is blood, the blood stabilization involves the use of at least one formaldehyde releaser and at least one anticoagulant, step (a) comprises: (a)(1) obtaining the stabilized blood sample or a portion or fraction thereof wherein the portion or fraction of the stabilised blood sample is selected from blood cells; and (a)(2) contacting the stabilized blood sample or portion or fraction thereof with at least one cationic to provide a lysed sample; and the nucleic acids isolated in step (b) compris or consist of RNA. 15. The method according to claim 14, wherein the formaldehyde releaser is selected from a heterocyclic urea, diazolidinyl urea and/or imidazolidinyl urea, and in step a)(2), the cationic detergent is selected from the group consisting of: a) a cationic compound of the general formula (1): Y+R1R2R3R4X−  (1) wherein, Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched C1-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; b) a detergent comprising under the used lysis conditions a charged quaternary ammonium cation as polar head group; c) a cationic detergent obtained in a composition comprising (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C20 alkyl residue, C6-C26 aryl residue or C6-C26 aralkyl residue, preferably H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt; d) a cationic detergent obtained from an amino surfactant selected from the group consisting of the protonated forms of dodecylamine, N-methyldodecylamine, N,N-dimethyldodecylamjne, N,N-dimethyldodecylamine N oxide and 4-tetradecylaniline; e) a cationic detergent comprising a permanently charged quaternary ammonium cation as polar head group; and/or f) a cationic detergent selected from the group consisting of cetyl trimethyl ammonium bromide (CTAB), tetra decyl trimethyl ammonium bromide (TTAB) and dodecyl trimethyl ammonium bromide (DTRB) or the corresponding compounds comprising a chloride instead of the bromide; or step (a)(1) uses a lysis composition comprising: (i) a cationic compound of the general formula (1): Y+R1R2R3R4X−  (1) wherein Y represents nitrogen or phosphor, R1R2R3 and R4 independently, represent a branched or unbranched C1-C20-alkyl group, a C6-C20aryl group and/or a C6-C26 aralkyl group; X− represents an anion of an inorganic or organic, mono- or polybasic acid; and (ii) at least one proton donor; or (i) an amino surfactant having the following formula (2): R1R2R3N(O)x   (2) wherein, R1 and R2 each independently is H, C1-C6 alkyl residue, C6-C12 aryl residue or C6-C12 aralkyl residue, R3 is C1-C20 alkyl group, C6-C26 aryl residue or C6-C26 aralkyl residue, X is an integer of 0 and 1 and (ii) an acid or acid salt; step (a) further comprises incubating the composition comprising the stabilized sample or portion or fraction of the stabilized sample, the at least one cationic detergent and optionally one or more further lysis agents to provide the lysed sample; and step (b) comprises the following steps: i. contacting the lysed sample or a nucleic acid containing portion obtained from the lysed sample with one or more additional lysing agents thereby providing a lysis mixture; optionally removing DNA from the lysis mixture; ii. adding alcohol to the lysis mixture to adjust the binding conditions and binding RNA to a nucleic acid binding solid phase; iii. separating the solid phase with the bound RNA from the remaining sample; and iv. optionally washing the RNA and v. optionally eluting RNA from the solid phase. 16. The method according to claim 3, wherein Y represents nitrogen. 17. The method according to claim 8, wherein the chaotropic agent is a chaotropic salt and/or wherein said binding compositions comprises a chaotropic salt and/or alcohol. 18. The method according to claim 11, wherein the formaldehyde releaser agent is diazolidinyl urea and/or imidazolidinyl urea. 19. The method according to claim 11, wherein the metabolic inhibitor is glyceraldehyde and/or sodium fluoride. 20. The method according to claim 15, wherein step (b) i. comprises contacting the lysed sample or the nucleic acid containing portion obtained from the lysed sample with (aa) at least one chaotropic agent, (bb) at least one proteolytic enzyme; and/or (cc) one or more salts.
1,600
279
279
14,399,336
1,628
Novel sterol derivatives, the preparation method thereof, pharmaceutical compositions containing them and use thereof for treating diseases involving transformed astrocyte cells or for treating malignant haemopathies. The treatment of glioblastoma multiforme, as well as of other cancers, such as lymphomas, neuroblastomas and melanomas is also described.
1-22. (canceled) 23. Compound of formula (I) having a 7beta-hydroxycholesterol basic structure in which A represents an —(R1)n— group in which R1 is an amino acid residue bound by its C-terminal end and n=1 or 2, each R1 being identical or different, in which the N-terminal end of said amino acid can be substituted with a —C(O)—R2 group in which R2 is a mono- or polycyclic C6-C14 arylalkyl group or a mono- or polycyclic C5-C14 heteroarylalkyl group that can comprise one or more heteroatoms, which may be identical or different, which is unsubstituted or substituted with at least one linear or branched C1-C4 alkyl group; a mono- or polycyclic C6-C14 arylalkyloxy group or a mono- or polycyclic C5-C14 heteroarylalkyloxy group that can comprise one or more heteroatoms, which may be identical or different, which are unsubstituted or substituted with at least one linear or branched C1-C4 alkyl group, or a —C(O)—NH—R4 or —C(S)—NH—R4 group in which R4 is hydrogen; a C1-C12 alkyl group, linear or branched, unsubstituted or substituted with at least one group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl; a C6-C14 aryl group, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or at least one group selected from OR, NHR and SR, as defined above; an acyl group; a formyl group; a sulphonyl group; a sulphinyl group; or else R4 represents an allyl group or a sugar residue; a —C(O)—OR5 group in which R5 is a C1-C12 alkyl, linear or branched, unsubstituted or substituted with at least one group selected from OR, NHR and SR, as defined above; a —C(O)—R6 group in which R6 is a saturated C5-C14 heterocycle comprising 1 or 2 heteroatoms, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or at least one group selected from OR, NHR and SR, as defined above; or a linear or branched C1-C12 alkyl group unsubstituted or substituted with a group selected from OR, NHR and SR, as defined above; a C6-C14 aryl group or a C5-C14 heteroaryl group, which are unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or with at least one group selected from OR, NHR and SR, as defined above; or else a sugar residue. B represents a —C(O)—R7 group in which R7 is hydrogen; a C1-C12, preferably C1-C6, alkyl, linear or branched, unsubstituted or substituted with at least one group selected from OR, NHR and SR, as defined above; a C6-C14 aryl group, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or at least one group selected from OR, NHR and SR, as defined above; or R7 represents OR8, in which R8 is a linear or branched, C1-C12, preferably C1-C6, alkyl, provided that: when B is —C(O)—R7 and A is —C(O)—R6, then R6 and R7 are not simultaneously methyl, propyl or an unsubstituted phenyl group; when B is —C(O)—R7 where R7 is OR8 and R8 is ethyl, then A is different from —C(O)—OR5 where R5 is ethyl. 24. Compound of formula (I) according to claim 23, in which at least one of the following conditions is fulfilled: A represents an —(R1)n— group in which R1 is an amino acid residue and n=1 or 2; A represents an —(R1)n— group in which R1 is an amino acid residue, n=1 or 2 and the N-terminal end of said amino acid is substituted with an arylalkoxycarbonyl group, in particular benzyloxycarbonyl; A represents an alanyl radical linked to a glycinyl radical, optionally substituted on its N-terminal end with an arylalkoxycarbonyl group, in particular benzyloxycarbonyl; A represents a methionyl radical linked to a glycinyl radical, optionally substituted on its N-terminal end with an arylalkoxycarbonyl group, in particular benzyloxycarbonyl. 25. Compound of formula (I) according to claim 23, in which A represents a —C(O)—R6 group in which R6 is a 2,2-dimethyl-1,3-dioxolane group; or a linear or branched C1-C6 alkyl group unsubstituted or substituted with a group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl; a C6-C14 aryl group or a C5-C14 heteroaryl group, which are unsubstituted or substituted with at least one linear or branched C1-C6 alkyl, or with at least one group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl; or else a sugar residue. 26. Compound of formula (I) according to claim 23, in which B represents an acyl group in which the alkyl group is C1-C6, in particular acetyl, or an alkoxycarbonyl group in which the alkyl group is C1-C6, in particular a tert-butoxycarbonyl group. 27. Compound of formula (I) according to claim 23, in which B can also, in particular, represent a C1-C6 alkyl group, unsubstituted or substituted with at least one group selected from OR, NHR and SR, as defined above; or a C6-C14 aryl group, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or with at least one group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl. 28. Compound of formula (I), characterized in that it is selected from the following compounds: 7-((tert-butoxycarbonyl)oxy)-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl 2-(2-(((benzyloxy)carbonyl)amino)-acetamido)propanoate (molecule 1.a); 7-acetoxy-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl 2-(2-(((benzyloxy)carbonyl)amino)-acetamido)propanoate (molecule 1.b); 7-((tert-butoxycarbonyl)oxy)-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl 2,2-dimethyl-1,3-dioxolane-4-carboxylate (molecule 2.a); 7-acetoxy-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]-phenanthren-3-yl 2,2-dimethyl-1,3-dioxolane-4-carboxylate (molecule 2.b). 29. Method for the preparation of a compound of formula (I) according to claim 23, characterized in that it comprises the following steps: protection of the hydroxyl function in position 3 of cholesterol with a protective group, introduction of a ketone function in position 7, reduction of the ketone function to a hydroxyl function, introduction of a protective group on the hydroxyl function in position 7, corresponding to the B group, and deprotection of the hydroxyl function in position 3. 30. Method according to claim 29, characterized in that, after deprotection, the hydroxyl function in position 3 can be substituted with the desired A group. 31. Pharmaceutical composition comprising at least one compound of formula (I) according to claim 23 and a pharmaceutically acceptable vehicle. 32. Pharmaceutical composition according to claim 31, characterized in that it consists of a liposome comprising at least one compound of formula (I), alone or in combination with another active ingredient. 33. Pharmaceutical composition according to claim 31, characterized in that it consists of an alcoholic solution comprising at least one compound of formula (I), alone or in combination with another active ingredient. 34. Pharmaceutical composition according to claim 31, characterized in that it is suitable for administration by oral route. 35. Pharmaceutical composition according to claim 34, for administration by oral route, selected from tablets, capsules, powders, granules, solutions, emulsions, oral suspensions, drops, syrups, complexes of compounds of formula (I) with biliary salts and combinations of compounds of formula (I) with phospholipids, in liposomal or non-liposomal form. 36. Pharmaceutical composition according to claim 31, characterized in that said compound of formula (I) is used as the only active ingredient, or in combination with an anti-cancer agent. 37. A method of treating a disease involving transformed astrocytes, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23. 38. The method according to claim 37, wherein the disease is glioblastoma multiforme. 39. The method according to claim 37, wherein said administering is a sequential treatment comprising at least one step of administering a first compound of formula (I) and at least one step of administering a second compound of formula (I), different from the first. 40. A method of treating malignant haemopathies, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23. 41. The method according to claim 40, wherein the malignant haemopathies are of the myeloid type. 42. The method according to claim 40, wherein the malignant haemopathies are lymphomas. 43. A method of treating neuroblastomas, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23. 44. A method of treating melanomas, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23.
Novel sterol derivatives, the preparation method thereof, pharmaceutical compositions containing them and use thereof for treating diseases involving transformed astrocyte cells or for treating malignant haemopathies. The treatment of glioblastoma multiforme, as well as of other cancers, such as lymphomas, neuroblastomas and melanomas is also described.1-22. (canceled) 23. Compound of formula (I) having a 7beta-hydroxycholesterol basic structure in which A represents an —(R1)n— group in which R1 is an amino acid residue bound by its C-terminal end and n=1 or 2, each R1 being identical or different, in which the N-terminal end of said amino acid can be substituted with a —C(O)—R2 group in which R2 is a mono- or polycyclic C6-C14 arylalkyl group or a mono- or polycyclic C5-C14 heteroarylalkyl group that can comprise one or more heteroatoms, which may be identical or different, which is unsubstituted or substituted with at least one linear or branched C1-C4 alkyl group; a mono- or polycyclic C6-C14 arylalkyloxy group or a mono- or polycyclic C5-C14 heteroarylalkyloxy group that can comprise one or more heteroatoms, which may be identical or different, which are unsubstituted or substituted with at least one linear or branched C1-C4 alkyl group, or a —C(O)—NH—R4 or —C(S)—NH—R4 group in which R4 is hydrogen; a C1-C12 alkyl group, linear or branched, unsubstituted or substituted with at least one group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl; a C6-C14 aryl group, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or at least one group selected from OR, NHR and SR, as defined above; an acyl group; a formyl group; a sulphonyl group; a sulphinyl group; or else R4 represents an allyl group or a sugar residue; a —C(O)—OR5 group in which R5 is a C1-C12 alkyl, linear or branched, unsubstituted or substituted with at least one group selected from OR, NHR and SR, as defined above; a —C(O)—R6 group in which R6 is a saturated C5-C14 heterocycle comprising 1 or 2 heteroatoms, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or at least one group selected from OR, NHR and SR, as defined above; or a linear or branched C1-C12 alkyl group unsubstituted or substituted with a group selected from OR, NHR and SR, as defined above; a C6-C14 aryl group or a C5-C14 heteroaryl group, which are unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or with at least one group selected from OR, NHR and SR, as defined above; or else a sugar residue. B represents a —C(O)—R7 group in which R7 is hydrogen; a C1-C12, preferably C1-C6, alkyl, linear or branched, unsubstituted or substituted with at least one group selected from OR, NHR and SR, as defined above; a C6-C14 aryl group, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or at least one group selected from OR, NHR and SR, as defined above; or R7 represents OR8, in which R8 is a linear or branched, C1-C12, preferably C1-C6, alkyl, provided that: when B is —C(O)—R7 and A is —C(O)—R6, then R6 and R7 are not simultaneously methyl, propyl or an unsubstituted phenyl group; when B is —C(O)—R7 where R7 is OR8 and R8 is ethyl, then A is different from —C(O)—OR5 where R5 is ethyl. 24. Compound of formula (I) according to claim 23, in which at least one of the following conditions is fulfilled: A represents an —(R1)n— group in which R1 is an amino acid residue and n=1 or 2; A represents an —(R1)n— group in which R1 is an amino acid residue, n=1 or 2 and the N-terminal end of said amino acid is substituted with an arylalkoxycarbonyl group, in particular benzyloxycarbonyl; A represents an alanyl radical linked to a glycinyl radical, optionally substituted on its N-terminal end with an arylalkoxycarbonyl group, in particular benzyloxycarbonyl; A represents a methionyl radical linked to a glycinyl radical, optionally substituted on its N-terminal end with an arylalkoxycarbonyl group, in particular benzyloxycarbonyl. 25. Compound of formula (I) according to claim 23, in which A represents a —C(O)—R6 group in which R6 is a 2,2-dimethyl-1,3-dioxolane group; or a linear or branched C1-C6 alkyl group unsubstituted or substituted with a group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl; a C6-C14 aryl group or a C5-C14 heteroaryl group, which are unsubstituted or substituted with at least one linear or branched C1-C6 alkyl, or with at least one group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl; or else a sugar residue. 26. Compound of formula (I) according to claim 23, in which B represents an acyl group in which the alkyl group is C1-C6, in particular acetyl, or an alkoxycarbonyl group in which the alkyl group is C1-C6, in particular a tert-butoxycarbonyl group. 27. Compound of formula (I) according to claim 23, in which B can also, in particular, represent a C1-C6 alkyl group, unsubstituted or substituted with at least one group selected from OR, NHR and SR, as defined above; or a C6-C14 aryl group, unsubstituted or substituted with at least one linear or branched C1-C6 alkyl or with at least one group selected from OR, NHR and SR, where R represents hydrogen, a linear C1-C12 alkyl or an unsubstituted C6-C14 aryl. 28. Compound of formula (I), characterized in that it is selected from the following compounds: 7-((tert-butoxycarbonyl)oxy)-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl 2-(2-(((benzyloxy)carbonyl)amino)-acetamido)propanoate (molecule 1.a); 7-acetoxy-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl 2-(2-(((benzyloxy)carbonyl)amino)-acetamido)propanoate (molecule 1.b); 7-((tert-butoxycarbonyl)oxy)-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl 2,2-dimethyl-1,3-dioxolane-4-carboxylate (molecule 2.a); 7-acetoxy-10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]-phenanthren-3-yl 2,2-dimethyl-1,3-dioxolane-4-carboxylate (molecule 2.b). 29. Method for the preparation of a compound of formula (I) according to claim 23, characterized in that it comprises the following steps: protection of the hydroxyl function in position 3 of cholesterol with a protective group, introduction of a ketone function in position 7, reduction of the ketone function to a hydroxyl function, introduction of a protective group on the hydroxyl function in position 7, corresponding to the B group, and deprotection of the hydroxyl function in position 3. 30. Method according to claim 29, characterized in that, after deprotection, the hydroxyl function in position 3 can be substituted with the desired A group. 31. Pharmaceutical composition comprising at least one compound of formula (I) according to claim 23 and a pharmaceutically acceptable vehicle. 32. Pharmaceutical composition according to claim 31, characterized in that it consists of a liposome comprising at least one compound of formula (I), alone or in combination with another active ingredient. 33. Pharmaceutical composition according to claim 31, characterized in that it consists of an alcoholic solution comprising at least one compound of formula (I), alone or in combination with another active ingredient. 34. Pharmaceutical composition according to claim 31, characterized in that it is suitable for administration by oral route. 35. Pharmaceutical composition according to claim 34, for administration by oral route, selected from tablets, capsules, powders, granules, solutions, emulsions, oral suspensions, drops, syrups, complexes of compounds of formula (I) with biliary salts and combinations of compounds of formula (I) with phospholipids, in liposomal or non-liposomal form. 36. Pharmaceutical composition according to claim 31, characterized in that said compound of formula (I) is used as the only active ingredient, or in combination with an anti-cancer agent. 37. A method of treating a disease involving transformed astrocytes, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23. 38. The method according to claim 37, wherein the disease is glioblastoma multiforme. 39. The method according to claim 37, wherein said administering is a sequential treatment comprising at least one step of administering a first compound of formula (I) and at least one step of administering a second compound of formula (I), different from the first. 40. A method of treating malignant haemopathies, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23. 41. The method according to claim 40, wherein the malignant haemopathies are of the myeloid type. 42. The method according to claim 40, wherein the malignant haemopathies are lymphomas. 43. A method of treating neuroblastomas, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23. 44. A method of treating melanomas, comprising administering to a subject in need thereof an effective amount of a compound of formula (I) according to claim 23.
1,600
280
280
10,633,629
1,633
Method and compositions for improving DNA polymerase and reverse transcriptase reactions are provided. Addition of anti-foam reagents to the reactions improves fluid handling, especially of small volumes and allows enhanced accuracy of optical detection, without substantially inhibiting enzymatic activity.
1. A method for detecting a target nucleic acid in a sample, comprising the step of amplifying the target nucleic acid using a polymerase chain reaction, wherein said polymerase chain reaction is carried out in the presence of an effective amount of at least one anti-foam reagent that does not substantially inhibit the action of the polymerase. 2. The method according to claim 1, wherein said polymerase chain reaction is a quantitative polymerase chain reaction. 3. The method according to claim 2, wherein said polymerase chain reaction is a reverse transcriptase polymerase chain reaction 4. The method according to claim 1, further comprising detecting the product of said polymerase chain reaction by optical detection. 5. The method according to claim 4, comprising detecting said product using a probe labeled with a detectable label. 6. The method according to claim 5, wherein said detectable label is a fluorescent dye. 7. The method according to claim 4, comprising detecting said product using a fluorescent nucleic acid-binding dye. 8. The method according to any of claim 1, wherein said polymerase chain reaction is carried out in the presence of an effective amount of at least two anti-foam reagents. 9. The method according to claim 1, wherein said anti-foam agent is selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 10. The method according to claim 8, wherein said at least two anti-foam reagents are selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 11. A composition for amplifying a target nucleic acid, comprising (a) at least one primer molecule that hybridizes to the target nucleic acid; (b) nucleotide triphosphates (c) a thermostable DNA polymerase (d) a detergent; and (e) an effective amount of at least one anti-foam reagent that does not substantially inhibit the action of said thermostable DNA polymerase. 12. A composition according to claim 11, comprising at least two anti-foam reagents. 13. A composition according to claim 11 wherein said anti-foam agent is selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 14. The composition according to claim 12, wherein said at least two anti-foam reagents are selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 15. The method according to claim 1 wherein said polymerase chain reaction is carried out in a sample chamber of a device comprising a plurality of said sample chambers. 16. The method according to claim 15, wherein each of a plurality of said sample chambers of said device contains reagents suitable for detecting a target nucleic acid. 17. The method according to claim 16, wherein a plurality of sample chambers of said device contains reagents suitable for detecting different target nucleic acids. 18. The method according to claim 17, further comprising detecting the amplified products in said sample chambers by optical detection. 19. The method according to claim 18, comprising detecting said amplified products using a probe labeled with a detectable label. 20. The method according to claim 19, wherein said detectable label is a fluorescent dye. 21. The method according to claim 18, comprising detecting said amplified products using a fluorescent nucleic acid-binding dye.
Method and compositions for improving DNA polymerase and reverse transcriptase reactions are provided. Addition of anti-foam reagents to the reactions improves fluid handling, especially of small volumes and allows enhanced accuracy of optical detection, without substantially inhibiting enzymatic activity.1. A method for detecting a target nucleic acid in a sample, comprising the step of amplifying the target nucleic acid using a polymerase chain reaction, wherein said polymerase chain reaction is carried out in the presence of an effective amount of at least one anti-foam reagent that does not substantially inhibit the action of the polymerase. 2. The method according to claim 1, wherein said polymerase chain reaction is a quantitative polymerase chain reaction. 3. The method according to claim 2, wherein said polymerase chain reaction is a reverse transcriptase polymerase chain reaction 4. The method according to claim 1, further comprising detecting the product of said polymerase chain reaction by optical detection. 5. The method according to claim 4, comprising detecting said product using a probe labeled with a detectable label. 6. The method according to claim 5, wherein said detectable label is a fluorescent dye. 7. The method according to claim 4, comprising detecting said product using a fluorescent nucleic acid-binding dye. 8. The method according to any of claim 1, wherein said polymerase chain reaction is carried out in the presence of an effective amount of at least two anti-foam reagents. 9. The method according to claim 1, wherein said anti-foam agent is selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 10. The method according to claim 8, wherein said at least two anti-foam reagents are selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 11. A composition for amplifying a target nucleic acid, comprising (a) at least one primer molecule that hybridizes to the target nucleic acid; (b) nucleotide triphosphates (c) a thermostable DNA polymerase (d) a detergent; and (e) an effective amount of at least one anti-foam reagent that does not substantially inhibit the action of said thermostable DNA polymerase. 12. A composition according to claim 11, comprising at least two anti-foam reagents. 13. A composition according to claim 11 wherein said anti-foam agent is selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 14. The composition according to claim 12, wherein said at least two anti-foam reagents are selected from the group consisting of 1520-US, AF, FG-10, O-30, SE-15, and Antifoam B. 15. The method according to claim 1 wherein said polymerase chain reaction is carried out in a sample chamber of a device comprising a plurality of said sample chambers. 16. The method according to claim 15, wherein each of a plurality of said sample chambers of said device contains reagents suitable for detecting a target nucleic acid. 17. The method according to claim 16, wherein a plurality of sample chambers of said device contains reagents suitable for detecting different target nucleic acids. 18. The method according to claim 17, further comprising detecting the amplified products in said sample chambers by optical detection. 19. The method according to claim 18, comprising detecting said amplified products using a probe labeled with a detectable label. 20. The method according to claim 19, wherein said detectable label is a fluorescent dye. 21. The method according to claim 18, comprising detecting said amplified products using a fluorescent nucleic acid-binding dye.
1,600
281
281
14,407,498
1,627
The invention relates to a pharmaceutical composition comprising a compound selected from the group consisting of desethylamiodarone and pharmaceutically acceptable salts, hydrates and solvates thereof, together with pharmaceutically acceptable excipients, vehicle and/or carrier, as well as the pharmaceutical composition for use in the treatment and prevention of atrial fibrillation with fewer side effects than its parent compound.
1. A pharmaceutical composition comprising a compound selected from the group consisting of desethylamiodarone and pharmaceutically acceptable salts, hydrates and solvates thereof, together with pharmaceutically acceptable excipients, vehicle and/or carrier, for use in the treatment and/or prevention of atrial fibrillation by oral administration. 2. The composition for use according to claim 1, wherein the composition is administered orally, sublingually, or buccally. 3. The composition for use according to claim 1, wherein the composition is administered chronically. 4. The composition for use according to claim 1, wherein the composition is administered once a day. 5. A method for the treatment and/or prevention of atrial fibrillation, said method comprising orally administering to a human or animal a pharmaceutical composition comprising a compound selected from the group consisting of desethylamiodarone and pharmaceutically acceptable salts, hydrates and solvates thereof, together with pharmaceutically acceptable excipients, vehicle and/or carrier. 6. The method of claim 5, wherein the composition is administered sublingually, buccally, or by swallowing. 7. The method of claim 5, wherein the composition is administered chronically. 8. The method of claim 5, wherein the composition is administered once a day. 9. The method of claim 5, wherein the human or animal suffers from atrial fibrillation.
The invention relates to a pharmaceutical composition comprising a compound selected from the group consisting of desethylamiodarone and pharmaceutically acceptable salts, hydrates and solvates thereof, together with pharmaceutically acceptable excipients, vehicle and/or carrier, as well as the pharmaceutical composition for use in the treatment and prevention of atrial fibrillation with fewer side effects than its parent compound.1. A pharmaceutical composition comprising a compound selected from the group consisting of desethylamiodarone and pharmaceutically acceptable salts, hydrates and solvates thereof, together with pharmaceutically acceptable excipients, vehicle and/or carrier, for use in the treatment and/or prevention of atrial fibrillation by oral administration. 2. The composition for use according to claim 1, wherein the composition is administered orally, sublingually, or buccally. 3. The composition for use according to claim 1, wherein the composition is administered chronically. 4. The composition for use according to claim 1, wherein the composition is administered once a day. 5. A method for the treatment and/or prevention of atrial fibrillation, said method comprising orally administering to a human or animal a pharmaceutical composition comprising a compound selected from the group consisting of desethylamiodarone and pharmaceutically acceptable salts, hydrates and solvates thereof, together with pharmaceutically acceptable excipients, vehicle and/or carrier. 6. The method of claim 5, wherein the composition is administered sublingually, buccally, or by swallowing. 7. The method of claim 5, wherein the composition is administered chronically. 8. The method of claim 5, wherein the composition is administered once a day. 9. The method of claim 5, wherein the human or animal suffers from atrial fibrillation.
1,600
282
282
15,279,939
1,643
The present invention relates to novel methods, compositions and kits useful for the treatment of pulmonary diseases such as pulmonary arterial hypertension. In particular, aerosolisable compositions of iloprost are provided which are for use in inhalation therapy. Their administration is by bolus inhalation, which is patient-friendly, effective, and well tolerated. Bolus inhalation may, for example, be achieved using an efficient nebuliser based on the vibrating mesh technology.
1-21. (canceled) 22. A method for delivering an active ingredient selected from iloprost and salts thereof, to a patient in need thereof comprising the pulmonary administration of a pharmaceutical composition to the patient, wherein an amount of the composition administered comprises an effective single dose from 1.5 μg to 5.0 μg of the active ingredient and is provided using an inhaler configured to provide the dose in aerosolised form for bolus inhalation within a period of less than 2 minutes. 23. The method of claim 22, wherein the amount of the composition has a strength of 10 μg/mL or more. 24. The method of claim 22, wherein the effective single dose is 2.5 μg and the amount of the composition has a strength of 10 μg/mL. 25. The method of claim 22, wherein the effective single dose is 5 μg and the amount of the composition has a strength of 20 μg/mL. 26. The method of claim 22, wherein the composition is formulated as an aqueous solution further comprising one or more of: (a) a cosolvent; (b) an isotonising agent; (c) a pH-adjusting agent; and (d) a stabilising agent. 27. A pharmaceutical kit comprising: (a) a composition comprising an active ingredient selected from iloprost and salts thereof; and (b) an inhaler adapted for providing an amount of the composition comprising an effective single dose from 1.5 μg to about 5.0 μg of the active ingredient in aerosolised form for bolus inhalation within a period of less than 2 minutes, and wherein the inhaler is a vibrating mesh nebuliser. 28. The pharmaceutical kit of claim 27, wherein the inhaler is adapted to emit the composition at an output rate of 0.5 mL/min or more. 29. The pharmaceutical kit of claim 27, wherein the inhaler is adapted to emit the composition at a rate of at least 5 μg of iloprost per minute. 30. The pharmaceutical kit of claim 27, wherein the inhaler is adapted to emit, per inhalation phase, a predetermined volume of aerosol-free air immediately following a predetermined volume of the aerosol. 31. The pharmaceutical kit of claim 27, wherein the composition is formulated as an aqueous solution further comprising one or more of: (a) a cosolvent; (b) an isotonising agent; (c) a pH-adjusting agent; and (d) a stabilising agent. 32. The pharmaceutical kit of claim 31, further comprising an electronic data storage means which is capable of controlling the operation of the inhaler. 33. The pharmaceutical kit of claim 32, wherein the data storage means is a programmable chip card.
The present invention relates to novel methods, compositions and kits useful for the treatment of pulmonary diseases such as pulmonary arterial hypertension. In particular, aerosolisable compositions of iloprost are provided which are for use in inhalation therapy. Their administration is by bolus inhalation, which is patient-friendly, effective, and well tolerated. Bolus inhalation may, for example, be achieved using an efficient nebuliser based on the vibrating mesh technology.1-21. (canceled) 22. A method for delivering an active ingredient selected from iloprost and salts thereof, to a patient in need thereof comprising the pulmonary administration of a pharmaceutical composition to the patient, wherein an amount of the composition administered comprises an effective single dose from 1.5 μg to 5.0 μg of the active ingredient and is provided using an inhaler configured to provide the dose in aerosolised form for bolus inhalation within a period of less than 2 minutes. 23. The method of claim 22, wherein the amount of the composition has a strength of 10 μg/mL or more. 24. The method of claim 22, wherein the effective single dose is 2.5 μg and the amount of the composition has a strength of 10 μg/mL. 25. The method of claim 22, wherein the effective single dose is 5 μg and the amount of the composition has a strength of 20 μg/mL. 26. The method of claim 22, wherein the composition is formulated as an aqueous solution further comprising one or more of: (a) a cosolvent; (b) an isotonising agent; (c) a pH-adjusting agent; and (d) a stabilising agent. 27. A pharmaceutical kit comprising: (a) a composition comprising an active ingredient selected from iloprost and salts thereof; and (b) an inhaler adapted for providing an amount of the composition comprising an effective single dose from 1.5 μg to about 5.0 μg of the active ingredient in aerosolised form for bolus inhalation within a period of less than 2 minutes, and wherein the inhaler is a vibrating mesh nebuliser. 28. The pharmaceutical kit of claim 27, wherein the inhaler is adapted to emit the composition at an output rate of 0.5 mL/min or more. 29. The pharmaceutical kit of claim 27, wherein the inhaler is adapted to emit the composition at a rate of at least 5 μg of iloprost per minute. 30. The pharmaceutical kit of claim 27, wherein the inhaler is adapted to emit, per inhalation phase, a predetermined volume of aerosol-free air immediately following a predetermined volume of the aerosol. 31. The pharmaceutical kit of claim 27, wherein the composition is formulated as an aqueous solution further comprising one or more of: (a) a cosolvent; (b) an isotonising agent; (c) a pH-adjusting agent; and (d) a stabilising agent. 32. The pharmaceutical kit of claim 31, further comprising an electronic data storage means which is capable of controlling the operation of the inhaler. 33. The pharmaceutical kit of claim 32, wherein the data storage means is a programmable chip card.
1,600
283
283
13,265,756
1,699
A bioperformance enhancing adjuvant of Formula (I) R 1 O[BO] n [AO] m R 2 where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R 1 is optionally substituted C 4-20 alkyl or optionally substituted C 4-20 alkenyl; and R 2 is hydrogen or optionally substituted C 1-3 alkyl.
1. (canceled) 2. A compound of formula (I) R1O[BO]n[AO]mR2  (I) where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R1 is C4-20 alkyl or C4-20 alkenyl; and R2 is hydrogen or C1-3 alkyl; provided that the compound of formula (I) is not a compound of formula (Ia) R3O[CH2CH(C2H5)O]a[C2H4O]bH  (Ia) where R3 is n-butyl, n-hexyl, n-octyl or n-decyl; a is 1, 2, 3 or 4; and b is from 9.7 to 10.2. 3. A compound of formula (I) as claimed in claim 2 where R1 is C6-18 alkyl or C6-18 alkenyl. 4. A compound of formula (I) as claimed in claim 3 where R1 is oleyl. 5. A compound of formula (I) as claimed in claim 2 where R2 is hydrogen or C1-2 alkyl. 6. A compound of formula (I) as claimed in claim 2 which is a compound of formula (Ib): R1O[BO]n[PO]m′[EO]m″R2  (Ib) where (m′+m″=m). 7. A compound of formula (I) as claimed in claim 2 where n is from 2 to 8. 8. A compound of formula (I) as claimed in claim 2 where m is from 5 to 15. 9. A compound of formula (I) as claimed in claim 2 where m=0. 10. (canceled) 11. An agrochemical composition which comprises at least one pesticide selected from the group consisting of insecticides, fungicides, herbicides, acaricides, nematicides, biocides and mixtures thereof, and which additionally comprises a compound of formula (I) as a bioperformance enhancing adjuvant R1O[BO]n[AO]mR2  (I) where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R1 is C4-20 alkyl or C4-20 alkenyl; and R2 is hydrogen or C1-3 alkyl. 12. A method of enhancing the bioperformance of an agrochemical composition containing at least one pesticide selected from the group consisting of insecticides, fungicides, herbicides, acaricides, nematicides, biocides and mixtures thereof, which comprises applying the composition along with a bioperformance enhancing amount of an adjuvant comprising a compound of formula (I) R1O[BO]n[AO]mR2  (I) where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R1 is C4-20 alkyl or C4-20 alkenyl; and R2 is hydrogen or C1-3 alkyl.
A bioperformance enhancing adjuvant of Formula (I) R 1 O[BO] n [AO] m R 2 where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R 1 is optionally substituted C 4-20 alkyl or optionally substituted C 4-20 alkenyl; and R 2 is hydrogen or optionally substituted C 1-3 alkyl.1. (canceled) 2. A compound of formula (I) R1O[BO]n[AO]mR2  (I) where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R1 is C4-20 alkyl or C4-20 alkenyl; and R2 is hydrogen or C1-3 alkyl; provided that the compound of formula (I) is not a compound of formula (Ia) R3O[CH2CH(C2H5)O]a[C2H4O]bH  (Ia) where R3 is n-butyl, n-hexyl, n-octyl or n-decyl; a is 1, 2, 3 or 4; and b is from 9.7 to 10.2. 3. A compound of formula (I) as claimed in claim 2 where R1 is C6-18 alkyl or C6-18 alkenyl. 4. A compound of formula (I) as claimed in claim 3 where R1 is oleyl. 5. A compound of formula (I) as claimed in claim 2 where R2 is hydrogen or C1-2 alkyl. 6. A compound of formula (I) as claimed in claim 2 which is a compound of formula (Ib): R1O[BO]n[PO]m′[EO]m″R2  (Ib) where (m′+m″=m). 7. A compound of formula (I) as claimed in claim 2 where n is from 2 to 8. 8. A compound of formula (I) as claimed in claim 2 where m is from 5 to 15. 9. A compound of formula (I) as claimed in claim 2 where m=0. 10. (canceled) 11. An agrochemical composition which comprises at least one pesticide selected from the group consisting of insecticides, fungicides, herbicides, acaricides, nematicides, biocides and mixtures thereof, and which additionally comprises a compound of formula (I) as a bioperformance enhancing adjuvant R1O[BO]n[AO]mR2  (I) where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R1 is C4-20 alkyl or C4-20 alkenyl; and R2 is hydrogen or C1-3 alkyl. 12. A method of enhancing the bioperformance of an agrochemical composition containing at least one pesticide selected from the group consisting of insecticides, fungicides, herbicides, acaricides, nematicides, biocides and mixtures thereof, which comprises applying the composition along with a bioperformance enhancing amount of an adjuvant comprising a compound of formula (I) R1O[BO]n[AO]mR2  (I) where BO is butylene oxide; and each AO is independently propylene oxide or ethylene oxide; n is from 1 to 12; m is from 0 to 20; R1 is C4-20 alkyl or C4-20 alkenyl; and R2 is hydrogen or C1-3 alkyl.
1,600
284
284
15,685,257
1,644
This disclosure relates to immunogens and monoclonal antibodies useful in the identification and/or treatment of cancer cells, including those of the dog. In one example, chimeric anti-canine CD20 antibodies are provided. The antibodies can be used therapeutically to treat lymphoma in dogs.
1-20. (canceled) 21. An isolated monoclonal antibody comprising at least one set of variable region amino acid sequences selected from the group consisting of: a light chain variable region (LC-V) comprising the sequence of DIVMTQAAPSVPVTPGESVSISCRSX1KX2LLHRX3X4NTYLYWFLQRPGQSPQL LIYRMSNLASGVPDRFSGSGSGTAFTLRISRVEAEDVGVYYCMQHLEFPFTFG GGTKLEIK (SEQ ID NO.:17) and a heavy chain variable region (HC-V) comprising the sequence of EVQLQQSGPELVKPGASVKISCKASGYTFTDYYMNWVKQSHGKSLEWIGDIN PNNGDTSYNQKFKGKAPLTVDKSSSTAYMEVRSLTSEDSAVYFCARGGVLR YPYYYVMDYWGQGTSVTVSS (SEQ ID NO.: 11); where X1 is any amino acid other than asparagine (N) when X2 is serine (S) or threonine (T); where X2 is any amino acid other than S or T when X1 is N; where X3 is an amino acid selected from the group consisting of alanine (A), glutamic acid (E), phenylalanine (F), histidine (H), isoleucine (I), lysine (K), leucine (L), proline (P), glutamine (Q), arginine (R), threonine (T), valine (V) and tyrosine (Y), when X4 is glycine (G); and where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and tryptophan (W), when X3 is N; an LC-V comprising the sequence of DIVMTQAAPSVPVTPGESVSISCRSX1KX2LLHRX3X4NTYLYWFLQRPGQSPQL LIYRMSNLASGVPDRFSGSGSGTAFTLRISRVEAEDVGVYYCMQHLEFPFTFG GGTKLEIK (SEQ ID NO.:17) and a HC-V comprising the sequence of EVQLQQSGPELVKPGASVKISCKASGYTFTDYYMNWVKQSHGKSLEWIGDIN PNX3X4DTSYNQKFKGKAPLTVDKSSSTAYMEVRSLTSEDSAVYFCARGGVLR YPYYYVMDYWGQGTSVTVSS (SEQ ID NO.: 18), where X1 is any amino acid other than N when X2 is S or T; where X2 is any amino acid other than S or T when X1 is N; where X3 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, T, V and Y, when X4 is G; and where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and W, when X3 is N; an LC-V comprising the sequence of DIVMTQAAP SVPVTPGESVSISCRSNKSLLHRNGNTYLYWFLQRPGQSPQLLI YRMSNLASGVPDRFSGSGSGTAFTLRISRVEAEDVGVYYCMQHLEFPFTFGG GTKLEIK (SEQ ID NO.: 9) and a HC-V comprising the sequence of EVQLQQSGPELVKPGASVKISCKASGYTFTDYYMNWVKQSHGKSLEWIGDIN PNX3X4DTSYNQKFKGKAPLTVDKSSSTAYMEVRSLTSEDSAVYFCARGGVLR YPYYYVMDYWGQGTSVTVSS (SEQ ID NO.: 18); where X3 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, T, V and Y, when X4 is G; and where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and W, when X3 is N; and, an LC-V comprising the sequence of DIVMTQSQKFMSRSVGDRVSVTCKASQNVGPNVAWYQQRPGQSPKPLIYSAS YRYSGVPDRFTGSGSGTDFTLTISNVQSEDLAEYFCQQYNNYPYTFGGGTKLE IK (SEQ ID NO.: 13) and a HC-V comprising the sequence of EVQLQQSGAELVRPGASVKLSCTASGFNIKDDYMHWVKQRPEQGLEWIGWI X5X6EX3X4HTKYASKFQGKATITADTSSNTAYLQLSSLTSEDTAVYYCTSLRH YYGSSYVSPHYYWGQGTTLTVSS (SEQ ID NO.: 19); where X3 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, T, V and Y, when X4 is G; where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and W, when X3 is N; where X5 is any amino acid other than aspartic acid (D); and where X6 is any amino acid other than P; and wherein the antibody binds canine CD20 with an affinity (Kd) of at least 20 nM. 22. The monoclonal antibody of claim 21, wherein the antibody comprises at least one light chain immunoglobulin constant region amino acid sequence from canine, and at least one heavy chain immunoglobulin constant region amino acid sequence from canine. 23. The monoclonal antibody of claim 22, wherein the at least one light chain immunoglobulin constant region amino acid sequence from canine comprises an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 55) RNDAQPAVYLFQPSPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, (SEQ ID NO.: 78) RTDAQPAVYLFQPSPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, (SEQ ID NO.: 79) RNDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, and (SEQ ID NO.: 80) RTDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDTG IQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKSFQ RSECQRVD; and wherein the at least one heavy chain immunoglobulin constant region amino acid sequence from canine comprises an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 57) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 81) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEPAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 82) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFAGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, and (SEQ ID NO.: 83) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEAAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK. 24. A method for treating a canine CD20-expressing lymphoma in a canine animal comprising administering to the animal at least one effective dose of the monoclonal antibody of claim 21. 25. The method of claim 24, wherein the monoclonal antibody is administered in a dosage amount of about 1 to 50 mg/kg of body weight of the animal. 26. The method of claim 24, wherein multiple doses are administered to the animal. 27. The method of claim 24, wherein the monoclonal antibody is administered in conjunction with one or more chemotherapeutic agents. 28. An isolated monoclonal antibody comprising a set of amino acid sequences selected from the group consisting of: a. a heavy chain (H) complementary determining region (CDR) 1 (CDRH1) selected from the group consisting of DYGML (SEQ ID NO.: 20) and GFTFSDY (SEQ ID NO.: 21), a CDRH2 selected from the group consisting of YISSGSSTIYYADRVKG (SEQ ID NO.: 22) and SSGSST (SEQ ID NO.: 23), a CDRH3 comprising the sequence of GTFAY (SEQ ID NO.: 24), a light chain (L) CDR1 (CDRL1) selected from the group consisting of RSSQSLIYNNGNTYLH (SEQ ID NO.: 25), SQSLIYNNGNTY (SEQ ID NO.: 26), RSSQSLIYNKGNTYLH (SEQ ID NO.: 70), SQSLIYNKGNTY (SEQ ID NO.: 71), RSSQSLIYNNKNTYLH (SEQ ID NO.: 72), SQSLIYNNKNTY (SEQ ID NO.: 73), RSSQSLIYNNQNTYLH (SEQ ID NO.: 74), SQSLIYNNQNTY (SEQ ID NO.: 75), RSSQSLIYNNANTYLH (SEQ ID NO.: 76), and SQSLIYNNANTY (SEQ ID NO.: 77), a CDRL2 selected from the group consisting of KVSNRFS (SEQ ID NO.: 27) and KVS (SEQ ID NO.: 28), and a CDRL3 selected from the group consisting of SQSTHVPFT (SEQ ID NO.: 29) and STHVPF (SEQ ID NO.: 30); b. a CDRH1 selected from the group consisting of DDYMEI (SEQ ID NO.: 31) and GFNIKDD (SEQ ID NO.: 32), a CDRH2 selected from the group consisting of WIDPENGHTKYASKFQG (SEQ ID NO.: 33) and DPENGH (SEQ ID NO.: 34), a CDRH3 comprising the sequence of LRHYYGSSYVSPHYY (SEQ ID NO.: 35), a CDRL1 selected from the group consisting of KASQNVGPNVA (SEQ ID NO.: 37) and SQNVGPN (SEQ ID NO.: 38), a CDRL2 selected from the group consisting of SASYRYS (SEQ ID NO.: 39) and SAS (SEQ ID NO.: 40), and a CDRL3 selected from the group consisting of QQYNNYPYT (SEQ ID NO.: 41) and YNNYPY (SEQ ID NO.: 42); and c. a CDRH1 selected from the group consisting of DYYMN (SEQ ID NO.: 43) and GYTFTDY (SEQ ID NO.: 44), a CDRH2 selected from the group consisting of DINPNNGDTSYNQKFKG (SEQ ID NO.: 45) and NPNNGD (SEQ ID NO.: 46), a CDRH3 comprising the sequence of GGVLRYPYYYVMDY (SEQ ID NO.: 47), a CDRL1 selected from the group consisting of RSNKSLLHRNGNTYLY (SEQ ID NO.: 49) and NKSLLHRNGNTY (SEQ ID NO.: 50), a CDRL2 selected from the group consisting of RMSNLAS (SEQ ID NO.: 51) and RMS (SEQ ID NO.: 52), and a CDRL3 selected from the group consisting of MQHLEFPFT (SEQ ID NO.: 53) and HLEFPF (SEQ ID NO.: 54); wherein the antibody comprises at least one heavy chain polypeptide comprising a CDRH1, a CDRH2, and a CDRH3 sequentially, and at least one light chain polypeptide comprising a CDRL1, a CDRL2, and a CDRL3 sequentially; wherein the at least one heavy chain polypeptide comprises framework regions from canine; wherein the at least one light chain polypeptide comprises framework regions from canine; and wherein the antibody binds canine CD20 with an affinity (Kd) of at least 20 nM. 29. The monoclonal antibody of claim 28, the antibody further comprising at least one light chain immunoglobulin constant region amino acid sequence from canine comprising an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 55) RNDAQPAVYLFQPSPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, (SEQ ID NO.: 78) RTDAQPAVYLFQP SPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQD TGIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIK SFQRSECQRVD, (SEQ ID NO.: 79) RNDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD,  and (SEQ ID NO.: 80) RTDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD; and wherein the antibody further comprises at least one heavy chain immunoglobulin constant region amino acid sequence from canine comprising an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 57) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 81) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEPAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 82) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFAGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, and (SEQ ID NO.: 83) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEAAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK. 30. A method for treating a canine CD20-expressing lymphoma in a canine animal comprising administering to the animal at least one effective dose of the monoclonal antibody of claim 28. 31. The method of claim 30, wherein the monoclonal antibody is administered in a dosage amount of about 1 to 50 mg/kg of body weight of the animal. 32. The method of claim 30, wherein multiple doses are administered to the animal. 33. The method of claim 30, wherein the monoclonal antibody is administered in conjunction with one or more chemotherapeutic agents.
This disclosure relates to immunogens and monoclonal antibodies useful in the identification and/or treatment of cancer cells, including those of the dog. In one example, chimeric anti-canine CD20 antibodies are provided. The antibodies can be used therapeutically to treat lymphoma in dogs.1-20. (canceled) 21. An isolated monoclonal antibody comprising at least one set of variable region amino acid sequences selected from the group consisting of: a light chain variable region (LC-V) comprising the sequence of DIVMTQAAPSVPVTPGESVSISCRSX1KX2LLHRX3X4NTYLYWFLQRPGQSPQL LIYRMSNLASGVPDRFSGSGSGTAFTLRISRVEAEDVGVYYCMQHLEFPFTFG GGTKLEIK (SEQ ID NO.:17) and a heavy chain variable region (HC-V) comprising the sequence of EVQLQQSGPELVKPGASVKISCKASGYTFTDYYMNWVKQSHGKSLEWIGDIN PNNGDTSYNQKFKGKAPLTVDKSSSTAYMEVRSLTSEDSAVYFCARGGVLR YPYYYVMDYWGQGTSVTVSS (SEQ ID NO.: 11); where X1 is any amino acid other than asparagine (N) when X2 is serine (S) or threonine (T); where X2 is any amino acid other than S or T when X1 is N; where X3 is an amino acid selected from the group consisting of alanine (A), glutamic acid (E), phenylalanine (F), histidine (H), isoleucine (I), lysine (K), leucine (L), proline (P), glutamine (Q), arginine (R), threonine (T), valine (V) and tyrosine (Y), when X4 is glycine (G); and where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and tryptophan (W), when X3 is N; an LC-V comprising the sequence of DIVMTQAAPSVPVTPGESVSISCRSX1KX2LLHRX3X4NTYLYWFLQRPGQSPQL LIYRMSNLASGVPDRFSGSGSGTAFTLRISRVEAEDVGVYYCMQHLEFPFTFG GGTKLEIK (SEQ ID NO.:17) and a HC-V comprising the sequence of EVQLQQSGPELVKPGASVKISCKASGYTFTDYYMNWVKQSHGKSLEWIGDIN PNX3X4DTSYNQKFKGKAPLTVDKSSSTAYMEVRSLTSEDSAVYFCARGGVLR YPYYYVMDYWGQGTSVTVSS (SEQ ID NO.: 18), where X1 is any amino acid other than N when X2 is S or T; where X2 is any amino acid other than S or T when X1 is N; where X3 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, T, V and Y, when X4 is G; and where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and W, when X3 is N; an LC-V comprising the sequence of DIVMTQAAP SVPVTPGESVSISCRSNKSLLHRNGNTYLYWFLQRPGQSPQLLI YRMSNLASGVPDRFSGSGSGTAFTLRISRVEAEDVGVYYCMQHLEFPFTFGG GTKLEIK (SEQ ID NO.: 9) and a HC-V comprising the sequence of EVQLQQSGPELVKPGASVKISCKASGYTFTDYYMNWVKQSHGKSLEWIGDIN PNX3X4DTSYNQKFKGKAPLTVDKSSSTAYMEVRSLTSEDSAVYFCARGGVLR YPYYYVMDYWGQGTSVTVSS (SEQ ID NO.: 18); where X3 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, T, V and Y, when X4 is G; and where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and W, when X3 is N; and, an LC-V comprising the sequence of DIVMTQSQKFMSRSVGDRVSVTCKASQNVGPNVAWYQQRPGQSPKPLIYSAS YRYSGVPDRFTGSGSGTDFTLTISNVQSEDLAEYFCQQYNNYPYTFGGGTKLE IK (SEQ ID NO.: 13) and a HC-V comprising the sequence of EVQLQQSGAELVRPGASVKLSCTASGFNIKDDYMHWVKQRPEQGLEWIGWI X5X6EX3X4HTKYASKFQGKATITADTSSNTAYLQLSSLTSEDTAVYYCTSLRH YYGSSYVSPHYYWGQGTTLTVSS (SEQ ID NO.: 19); where X3 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, T, V and Y, when X4 is G; where X4 is an amino acid selected from the group consisting of A, E, F, H, I, K, L, P, Q, R, V, Y, and W, when X3 is N; where X5 is any amino acid other than aspartic acid (D); and where X6 is any amino acid other than P; and wherein the antibody binds canine CD20 with an affinity (Kd) of at least 20 nM. 22. The monoclonal antibody of claim 21, wherein the antibody comprises at least one light chain immunoglobulin constant region amino acid sequence from canine, and at least one heavy chain immunoglobulin constant region amino acid sequence from canine. 23. The monoclonal antibody of claim 22, wherein the at least one light chain immunoglobulin constant region amino acid sequence from canine comprises an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 55) RNDAQPAVYLFQPSPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, (SEQ ID NO.: 78) RTDAQPAVYLFQPSPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, (SEQ ID NO.: 79) RNDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, and (SEQ ID NO.: 80) RTDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDTG IQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKSFQ RSECQRVD; and wherein the at least one heavy chain immunoglobulin constant region amino acid sequence from canine comprises an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 57) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 81) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEPAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 82) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFAGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, and (SEQ ID NO.: 83) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEAAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK. 24. A method for treating a canine CD20-expressing lymphoma in a canine animal comprising administering to the animal at least one effective dose of the monoclonal antibody of claim 21. 25. The method of claim 24, wherein the monoclonal antibody is administered in a dosage amount of about 1 to 50 mg/kg of body weight of the animal. 26. The method of claim 24, wherein multiple doses are administered to the animal. 27. The method of claim 24, wherein the monoclonal antibody is administered in conjunction with one or more chemotherapeutic agents. 28. An isolated monoclonal antibody comprising a set of amino acid sequences selected from the group consisting of: a. a heavy chain (H) complementary determining region (CDR) 1 (CDRH1) selected from the group consisting of DYGML (SEQ ID NO.: 20) and GFTFSDY (SEQ ID NO.: 21), a CDRH2 selected from the group consisting of YISSGSSTIYYADRVKG (SEQ ID NO.: 22) and SSGSST (SEQ ID NO.: 23), a CDRH3 comprising the sequence of GTFAY (SEQ ID NO.: 24), a light chain (L) CDR1 (CDRL1) selected from the group consisting of RSSQSLIYNNGNTYLH (SEQ ID NO.: 25), SQSLIYNNGNTY (SEQ ID NO.: 26), RSSQSLIYNKGNTYLH (SEQ ID NO.: 70), SQSLIYNKGNTY (SEQ ID NO.: 71), RSSQSLIYNNKNTYLH (SEQ ID NO.: 72), SQSLIYNNKNTY (SEQ ID NO.: 73), RSSQSLIYNNQNTYLH (SEQ ID NO.: 74), SQSLIYNNQNTY (SEQ ID NO.: 75), RSSQSLIYNNANTYLH (SEQ ID NO.: 76), and SQSLIYNNANTY (SEQ ID NO.: 77), a CDRL2 selected from the group consisting of KVSNRFS (SEQ ID NO.: 27) and KVS (SEQ ID NO.: 28), and a CDRL3 selected from the group consisting of SQSTHVPFT (SEQ ID NO.: 29) and STHVPF (SEQ ID NO.: 30); b. a CDRH1 selected from the group consisting of DDYMEI (SEQ ID NO.: 31) and GFNIKDD (SEQ ID NO.: 32), a CDRH2 selected from the group consisting of WIDPENGHTKYASKFQG (SEQ ID NO.: 33) and DPENGH (SEQ ID NO.: 34), a CDRH3 comprising the sequence of LRHYYGSSYVSPHYY (SEQ ID NO.: 35), a CDRL1 selected from the group consisting of KASQNVGPNVA (SEQ ID NO.: 37) and SQNVGPN (SEQ ID NO.: 38), a CDRL2 selected from the group consisting of SASYRYS (SEQ ID NO.: 39) and SAS (SEQ ID NO.: 40), and a CDRL3 selected from the group consisting of QQYNNYPYT (SEQ ID NO.: 41) and YNNYPY (SEQ ID NO.: 42); and c. a CDRH1 selected from the group consisting of DYYMN (SEQ ID NO.: 43) and GYTFTDY (SEQ ID NO.: 44), a CDRH2 selected from the group consisting of DINPNNGDTSYNQKFKG (SEQ ID NO.: 45) and NPNNGD (SEQ ID NO.: 46), a CDRH3 comprising the sequence of GGVLRYPYYYVMDY (SEQ ID NO.: 47), a CDRL1 selected from the group consisting of RSNKSLLHRNGNTYLY (SEQ ID NO.: 49) and NKSLLHRNGNTY (SEQ ID NO.: 50), a CDRL2 selected from the group consisting of RMSNLAS (SEQ ID NO.: 51) and RMS (SEQ ID NO.: 52), and a CDRL3 selected from the group consisting of MQHLEFPFT (SEQ ID NO.: 53) and HLEFPF (SEQ ID NO.: 54); wherein the antibody comprises at least one heavy chain polypeptide comprising a CDRH1, a CDRH2, and a CDRH3 sequentially, and at least one light chain polypeptide comprising a CDRL1, a CDRL2, and a CDRL3 sequentially; wherein the at least one heavy chain polypeptide comprises framework regions from canine; wherein the at least one light chain polypeptide comprises framework regions from canine; and wherein the antibody binds canine CD20 with an affinity (Kd) of at least 20 nM. 29. The monoclonal antibody of claim 28, the antibody further comprising at least one light chain immunoglobulin constant region amino acid sequence from canine comprising an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 55) RNDAQPAVYLFQPSPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD, (SEQ ID NO.: 78) RTDAQPAVYLFQP SPDQLHTGSASVVCLLNSFYPKDINVKWKVDGVIQD TGIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIK SFQRSECQRVD, (SEQ ID NO.: 79) RNDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD,  and (SEQ ID NO.: 80) RTDAQPAVYLFQPSPDQLHTGSASVVCLLSSFYPKDINVKWKVDGVIQDT GIQESVTEQDKDSTYSLSSTLTMSSTEYLSHELYSCEITHKSLPSTLIKS FQRSECQRVD; and wherein the antibody further comprises at least one heavy chain immunoglobulin constant region amino acid sequence from canine comprising an amino acid sequence selected from the group consisting of: (SEQ ID NO.: 57) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 81) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEPAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, (SEQ ID NO.: 82) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEMLGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFAGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK, and (SEQ ID NO.: 83) ASTTAPSVFPLAPSCGSTSGSTVALACLVSGYFPEPVTVSWNSGSLTSGV HTFPSVLQSSGLYSLSSMVTVPSSRWPSETFTCNVAHPASKTKVDKPVPK RENGRVPRPPDCPKCPAPEAAGGPSVFIFPPKPKDTLLIARTPEVTCVVV DLDPEDPEVQISWFVDGKQMQTAKTQPREEQFNGTYRVVSVLPIGHQDWL KGKQFTCKVNNKALPSPIERTISKARGQAHQPSVYVLPPSREELSKNTVS LTCLIKDFFPPDIDVEWQSNGQQEPESKYRTTPPQLDEDGSYFLYSKLSV DKSRWQRGDTFICAVMHEALHNHYTQKSLSHSPGK. 30. A method for treating a canine CD20-expressing lymphoma in a canine animal comprising administering to the animal at least one effective dose of the monoclonal antibody of claim 28. 31. The method of claim 30, wherein the monoclonal antibody is administered in a dosage amount of about 1 to 50 mg/kg of body weight of the animal. 32. The method of claim 30, wherein multiple doses are administered to the animal. 33. The method of claim 30, wherein the monoclonal antibody is administered in conjunction with one or more chemotherapeutic agents.
1,600
285
285
14,678,749
1,699
Described are methods of making organic compounds by metathesis chemistry. The methods of the invention are particularly useful for making industrially-important organic compounds beginning with starting compositions derived from renewable feedstocks, such as natural oils. The methods make use of a cross-metathesis step with an olefin compound to produce functionalized alkene intermediates having a pre-determined double bond position. Once isolated, the functionalized alkene intermediate can be self-metathesized or cross-metathesized (e.g., with a second functionalized alkene) to produce the desired organic compound or a precursor thereto. The method may be used to make bifunctional organic compounds, such as diacids, diesters, dicarboxylate salts, acid/esters, acid/amines, acid/alcohols, acid/aldehydes, acid/ketones, acid/halides, acid/nitriles, ester/amines, ester/alcohols, ester/aldehydes, ester/ketones, ester/halides, ester/nitriles, and the like.
1-90. (canceled) 91. A method comprising: providing (a) triglycerides and (b) short-chain olefins; cross-metathesizing the triglycerides and the short-chain olefins in the presence of a metathesis catalyst to provide cross-metathesis products comprising olefins and ester-functionalized alkenes; separating the olefins in the cross-metathesis products and the ester-functionalized alkenes in the cross-metathesis products to provide separated olefins and separated ester-functionalized alkenes; and transesterifying the separated ester-functionalized alkenes in the presence of an alcohol to provide a transesterified product comprising glycerol and free ester-functionalized alkenes. 92. The method of claim 91, further comprising: separating spent metathesis catalyst in the cross-metathesis products to provide separated spent metathesis catalyst. 93. The method of claim 91, further comprising: separating the glycerol in the transesterified product and the free ester-functionalized alkenes in the transesterified product to provide separated glycerol and separated free ester-functionalized alkenes. 94. The method of claim 93, further comprising: self-metathesizing the separated free-ester functionalized alkenes in the presence of second metathesis catalyst to provide a self-metathesis product comprising diester alkenes. 95. The method of claim 94, further comprising: hydrogenating the diester alkenes in the self-metathesis product. 96. The method of claim 91, wherein the triglycerides is derived from a natural oil. 97. The method of claim 96, wherein the natural oil is a plant-based oil or animal fat. 98. The method of claim 97, wherein the plaint-based oil is selected from the group consisting of: canola oil, rapeseed oil, coconut oil, corn oil, cottonseed oil, olive oil, palm oil, peanut oil, safflower oil, sesame oil, soybean oil, sunflower oil, linseed oil, palm kernel oil, tung oil, castor oil, and mixtures thereof. 99. The method of claim 97, wherein the animal fat is selected from the group consisting of: lard, tallow, chicken fat, fish oil, and mixtures thereof. 100. The method of claim 91, wherein the alcohol is a monohydric alcohol. 101. The method of claim 100, wherein the monohydric alcohol is selected from the group consisting of: methanol, ethanol, propanol, butanol, and mixtures thereof. 102. The method of claim 100, wherein the monohydric alcohol is methanol. 103. The method of claim 91, wherein the short-chain olefins are short-chain internal olefins. 104. The method of claim 91, wherein the short-chain internal olefins are symmetric short-chain internal olefins. 105. The method of claim 104, wherein the short-chain internal olefins comprise 2-butene. 106. The method of claim 91, wherein the short-chain olefins are short-chain alpha-olefins. 107. The method of claim 106, wherein the short-chain alpha-olefins are selected from the group consisting of: 1-propene, 1-butene, 1-pentene, 1-hexene, 1-heptene, 1-octene, 1-nonene, and mixtures thereof.
Described are methods of making organic compounds by metathesis chemistry. The methods of the invention are particularly useful for making industrially-important organic compounds beginning with starting compositions derived from renewable feedstocks, such as natural oils. The methods make use of a cross-metathesis step with an olefin compound to produce functionalized alkene intermediates having a pre-determined double bond position. Once isolated, the functionalized alkene intermediate can be self-metathesized or cross-metathesized (e.g., with a second functionalized alkene) to produce the desired organic compound or a precursor thereto. The method may be used to make bifunctional organic compounds, such as diacids, diesters, dicarboxylate salts, acid/esters, acid/amines, acid/alcohols, acid/aldehydes, acid/ketones, acid/halides, acid/nitriles, ester/amines, ester/alcohols, ester/aldehydes, ester/ketones, ester/halides, ester/nitriles, and the like.1-90. (canceled) 91. A method comprising: providing (a) triglycerides and (b) short-chain olefins; cross-metathesizing the triglycerides and the short-chain olefins in the presence of a metathesis catalyst to provide cross-metathesis products comprising olefins and ester-functionalized alkenes; separating the olefins in the cross-metathesis products and the ester-functionalized alkenes in the cross-metathesis products to provide separated olefins and separated ester-functionalized alkenes; and transesterifying the separated ester-functionalized alkenes in the presence of an alcohol to provide a transesterified product comprising glycerol and free ester-functionalized alkenes. 92. The method of claim 91, further comprising: separating spent metathesis catalyst in the cross-metathesis products to provide separated spent metathesis catalyst. 93. The method of claim 91, further comprising: separating the glycerol in the transesterified product and the free ester-functionalized alkenes in the transesterified product to provide separated glycerol and separated free ester-functionalized alkenes. 94. The method of claim 93, further comprising: self-metathesizing the separated free-ester functionalized alkenes in the presence of second metathesis catalyst to provide a self-metathesis product comprising diester alkenes. 95. The method of claim 94, further comprising: hydrogenating the diester alkenes in the self-metathesis product. 96. The method of claim 91, wherein the triglycerides is derived from a natural oil. 97. The method of claim 96, wherein the natural oil is a plant-based oil or animal fat. 98. The method of claim 97, wherein the plaint-based oil is selected from the group consisting of: canola oil, rapeseed oil, coconut oil, corn oil, cottonseed oil, olive oil, palm oil, peanut oil, safflower oil, sesame oil, soybean oil, sunflower oil, linseed oil, palm kernel oil, tung oil, castor oil, and mixtures thereof. 99. The method of claim 97, wherein the animal fat is selected from the group consisting of: lard, tallow, chicken fat, fish oil, and mixtures thereof. 100. The method of claim 91, wherein the alcohol is a monohydric alcohol. 101. The method of claim 100, wherein the monohydric alcohol is selected from the group consisting of: methanol, ethanol, propanol, butanol, and mixtures thereof. 102. The method of claim 100, wherein the monohydric alcohol is methanol. 103. The method of claim 91, wherein the short-chain olefins are short-chain internal olefins. 104. The method of claim 91, wherein the short-chain internal olefins are symmetric short-chain internal olefins. 105. The method of claim 104, wherein the short-chain internal olefins comprise 2-butene. 106. The method of claim 91, wherein the short-chain olefins are short-chain alpha-olefins. 107. The method of claim 106, wherein the short-chain alpha-olefins are selected from the group consisting of: 1-propene, 1-butene, 1-pentene, 1-hexene, 1-heptene, 1-octene, 1-nonene, and mixtures thereof.
1,600
286
286
15,641,451
1,617
A method for the treatment of a local pigmentation disorder comprises applying to skin affected by the disorder a transdermal therapeutic system which comprises 4-n-butylresorcinol as an active ingredient.
1. A method for the treatment of a local pigmentation disorder, wherein the method comprises applying to skin affected by the disorder a transdermal therapeutic system which comprises 4-n-butylresorcinol as an active ingredient. 2. The method of claim 1, wherein the local pigmentation disorder comprises local hyperpigmentation. 3. The method of claim 1, wherein the local pigmentation disorder comprises age spots. 4. The method of claim 1, wherein the local pigmentation disorder comprises freckles. 5. The method of claim 1, wherein the local pigmentation disorder comprises post-inflammatory hyperpigmentation. 6. The method of claim 1, wherein the local pigmentation disorder comprises melasma. 7. The method of claim 1, wherein the transdermal therapeutic system is employed in the form of a self-adhesive flat bandage. 8. The method of claim 1, wherein the transdermal therapeutic system is employed in the form of at least one of a cosmetic plaster and a medicinal plaster. 9. The method of claim 1, wherein the transdermal therapeutic system is employed in the form of a matrix system. 10. The method of claim 9, wherein the matrix is selected from nonpolar matrices based on synthetic and natural rubber, polar wet adhesive films based on polyacrylic acid/polyvinyl alcohol, nonpolar matrices based on polyacrylic acid copolymers, polar anhydrous gel matrices based on polyacrylic acid/polyvinylpyrrolidone, nonpolar polyisobutylene matrices, and polar water gel matrices based on agar agar/polyacrylic acid. 11. The method of claim 9, wherein the matrix is selected from polyisobutylene matrices. 12. The method of claim 9, wherein the matrix is selected from water gel matrices based on agar agar/polyacrylic acid. 13. The method of claim 1, wherein the transdermal therapeutic system comprises from 0.001% to 10% by weight of 4-n-butylresorcinol, based on a total weight of the system. 14. The method of claim 1, wherein the transdermal therapeutic system comprises from 0.01% to 1% by weight of 4-n-butylresorcinol. 15. The method of claim 11, wherein the matrix has a layer thickness of from 0.15 mm to 1.00 mm. 16. The method of claim 12, wherein the matrix has a layer thickness of from 0.15 mm to 1.00 mm. 17. The method of claim 11, wherein the matrix has a layer thickness of from 0.20 mm to 0.50 mm. 18. The method of claim 12, wherein the matrix has a layer thickness of from 0.20 mm to 0.50 mm. 19. The method of claim 1, wherein the transdermal therapeutic system comprises from 0.001% to 10% by weight of one or more penetration accelerators. 20. The method of claim 19, wherein the one or more penetration accelerators comprise at least one of isopropyl palmitate and isopropyl myristate.
A method for the treatment of a local pigmentation disorder comprises applying to skin affected by the disorder a transdermal therapeutic system which comprises 4-n-butylresorcinol as an active ingredient.1. A method for the treatment of a local pigmentation disorder, wherein the method comprises applying to skin affected by the disorder a transdermal therapeutic system which comprises 4-n-butylresorcinol as an active ingredient. 2. The method of claim 1, wherein the local pigmentation disorder comprises local hyperpigmentation. 3. The method of claim 1, wherein the local pigmentation disorder comprises age spots. 4. The method of claim 1, wherein the local pigmentation disorder comprises freckles. 5. The method of claim 1, wherein the local pigmentation disorder comprises post-inflammatory hyperpigmentation. 6. The method of claim 1, wherein the local pigmentation disorder comprises melasma. 7. The method of claim 1, wherein the transdermal therapeutic system is employed in the form of a self-adhesive flat bandage. 8. The method of claim 1, wherein the transdermal therapeutic system is employed in the form of at least one of a cosmetic plaster and a medicinal plaster. 9. The method of claim 1, wherein the transdermal therapeutic system is employed in the form of a matrix system. 10. The method of claim 9, wherein the matrix is selected from nonpolar matrices based on synthetic and natural rubber, polar wet adhesive films based on polyacrylic acid/polyvinyl alcohol, nonpolar matrices based on polyacrylic acid copolymers, polar anhydrous gel matrices based on polyacrylic acid/polyvinylpyrrolidone, nonpolar polyisobutylene matrices, and polar water gel matrices based on agar agar/polyacrylic acid. 11. The method of claim 9, wherein the matrix is selected from polyisobutylene matrices. 12. The method of claim 9, wherein the matrix is selected from water gel matrices based on agar agar/polyacrylic acid. 13. The method of claim 1, wherein the transdermal therapeutic system comprises from 0.001% to 10% by weight of 4-n-butylresorcinol, based on a total weight of the system. 14. The method of claim 1, wherein the transdermal therapeutic system comprises from 0.01% to 1% by weight of 4-n-butylresorcinol. 15. The method of claim 11, wherein the matrix has a layer thickness of from 0.15 mm to 1.00 mm. 16. The method of claim 12, wherein the matrix has a layer thickness of from 0.15 mm to 1.00 mm. 17. The method of claim 11, wherein the matrix has a layer thickness of from 0.20 mm to 0.50 mm. 18. The method of claim 12, wherein the matrix has a layer thickness of from 0.20 mm to 0.50 mm. 19. The method of claim 1, wherein the transdermal therapeutic system comprises from 0.001% to 10% by weight of one or more penetration accelerators. 20. The method of claim 19, wherein the one or more penetration accelerators comprise at least one of isopropyl palmitate and isopropyl myristate.
1,600
287
287
13,515,925
1,656
The present invention relates to a method for producing a hydrolysate of from lignocellulose-containing material, comprising pre-treatment with low temperature, hydrolysis and fermentation, wherein hydrolysis is performed by contacting the lignocellulose-containing material with an enzyme composition comprising at least 10% xylanase enzyme protein w/w%.
1-15. (canceled) 16. A process for producing a hydrolysate of a lignocellulosic material comprising a) subjecting the lignocellulosic material to a pretreatment at a temperature between 165° C. and 175° C., and b) subjecting the pretreated lignocellulosic material to the action of hydrolytic enzymes to produce a hydrolysate, wherein the hydrolytic enzymes comprise cellulytic enzymes and a xylanase, and wherein said xylanase is present in an amount of at least 10% of the total amount hydrolytic enzyme protein. 17. The process according to claim 16, wherein the hydrolytic enzymes comprise a cellulase system derived from Trichoderma reesei. 18. The process according to claim 16, wherein the xylanase has at least 90% identity with the sequence shown as SEQ ID NO:1. 19. The process according to claim 16, wherein the xylanase is present in an amount of at least 15% of the total amount of hydrolytic enzyme protein. 20. The process according to claim 16, wherein the hydrolytic enzymes comprise a family 61 glycoside hydrolase in an amount of at least 5% of the total amount of hydrolytic enzyme protein. 21. The process according to claim 16, wherein the hydrolytic enzymes comprise a beta-glucosidase in an amount of at least 1% of the total amount of hydrolytic enzyme protein. 22. The process according to claim 16, wherein the lignocellulose-containing material originates from materials selected from the group consisting of corn stover, corn fiber, hard wood, such as poplar and birch, soft wood, cereal straw, such as, wheat straw, switch grass, rice hulls, municipal solid waste, industrial organic waste, office paper, and mixtures thereof. 23. The process according to claim 16, wherein the pretreatment in step (a) is carried out using an organic or inorganic acid selected from the group consisting of sulphuric acid, acetic acid, citric acid, tartaric acid, succinic acid, and mixtures thereof. 24. The process according to claim 16, wherein the pretreatment in step (a) is carried out using from 0.1 to 2.5 wt. % acid. 25. The process according to claim 16, wherein the pretreatment is performed at a temperature of around 170° C. 26. The process according to claim 16, wherein the pretreatment is performed as steam explosion. 27. The process according to claim 16, further comprising fermenting the hydrolysate of step (b) to produce a fermentation product. 28. The process according to claim 27, wherein the hydrolysis and fermentation are simultaneous. 29. The process according to claim 27, further comprising recovery of the fermentation product.
The present invention relates to a method for producing a hydrolysate of from lignocellulose-containing material, comprising pre-treatment with low temperature, hydrolysis and fermentation, wherein hydrolysis is performed by contacting the lignocellulose-containing material with an enzyme composition comprising at least 10% xylanase enzyme protein w/w%.1-15. (canceled) 16. A process for producing a hydrolysate of a lignocellulosic material comprising a) subjecting the lignocellulosic material to a pretreatment at a temperature between 165° C. and 175° C., and b) subjecting the pretreated lignocellulosic material to the action of hydrolytic enzymes to produce a hydrolysate, wherein the hydrolytic enzymes comprise cellulytic enzymes and a xylanase, and wherein said xylanase is present in an amount of at least 10% of the total amount hydrolytic enzyme protein. 17. The process according to claim 16, wherein the hydrolytic enzymes comprise a cellulase system derived from Trichoderma reesei. 18. The process according to claim 16, wherein the xylanase has at least 90% identity with the sequence shown as SEQ ID NO:1. 19. The process according to claim 16, wherein the xylanase is present in an amount of at least 15% of the total amount of hydrolytic enzyme protein. 20. The process according to claim 16, wherein the hydrolytic enzymes comprise a family 61 glycoside hydrolase in an amount of at least 5% of the total amount of hydrolytic enzyme protein. 21. The process according to claim 16, wherein the hydrolytic enzymes comprise a beta-glucosidase in an amount of at least 1% of the total amount of hydrolytic enzyme protein. 22. The process according to claim 16, wherein the lignocellulose-containing material originates from materials selected from the group consisting of corn stover, corn fiber, hard wood, such as poplar and birch, soft wood, cereal straw, such as, wheat straw, switch grass, rice hulls, municipal solid waste, industrial organic waste, office paper, and mixtures thereof. 23. The process according to claim 16, wherein the pretreatment in step (a) is carried out using an organic or inorganic acid selected from the group consisting of sulphuric acid, acetic acid, citric acid, tartaric acid, succinic acid, and mixtures thereof. 24. The process according to claim 16, wherein the pretreatment in step (a) is carried out using from 0.1 to 2.5 wt. % acid. 25. The process according to claim 16, wherein the pretreatment is performed at a temperature of around 170° C. 26. The process according to claim 16, wherein the pretreatment is performed as steam explosion. 27. The process according to claim 16, further comprising fermenting the hydrolysate of step (b) to produce a fermentation product. 28. The process according to claim 27, wherein the hydrolysis and fermentation are simultaneous. 29. The process according to claim 27, further comprising recovery of the fermentation product.
1,600
288
288
15,106,279
1,612
An oral care composition comprising: arginine in free or salt form; and zinc ions, wherein the zinc ions are present in the composition at a concentration of from 0.0001 mols/100 g to 0.0150 mols/100 g, based on the total weight of the composition.
1. An oral care composition comprising: a) arginine in free or salt form; and b) zinc ions, wherein the zinc ions are present in the composition at a concentration selected from the group consisting of from 0.0001 mols/100 g to 0.015 mols/100 g from 0.00025 mols/100 g, to 0.005 mols/100 g, from 0.0004 mols/100 g to 0.0015 mols/100 g, from 0.0005 mols/100 g to 0.001 mols/100 g, and from 0.00052 mols/100 g to 0.00062 mols/100 g, based on the total weight of the composition. 2. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.00025 mols/100 g to 0.005 mols/100 g, based on the total weight of the composition. 3. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.0004 mols/100 g to 0.0015 mols/100 g, based on the total weight of the composition. 4. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.0005 mols/100 g to 0.001 mols/100 g based on the total weight of the composition. 5. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.00052 mols/100 g to 0.00062 mols/100 g, based on the total weight of the composition. 6. The oral care composition of claim 1, wherein the arginine is present in the composition at a concentration of from 0.1 weight % to 1 weight %, based on the total weight of the composition. 7. The oral care composition of claim 1, wherein the arginine is present in the composition at a concentration of from 0.5 weight % to 0.8 weight %, based on the total weight of the composition. 8. The oral care composition of claim 1, wherein the zinc ions are provided by at least one zinc compound chosen from zinc oxide, zinc lactate, zinc chloride, zinc citrate, zinc acetate, zinc borate, zinc butyrate, zinc carbonate, zinc formate, zinc gluconate, zinc glycerate, zinc glycolate, zinc phosphate, zinc picolinate, zinc proprionate, zinc salicylate, zinc silicate, zinc stearate, zinc tartrate, zinc undecylenate, zinc phosphate, zinc ricinoleate, zinc nitrate, and zinc sulfate. 9. The oral care composition of claim 1, wherein the zinc ions are provided by a zinc compound selected from group consisting of zinc, oxide, zinc lactate, zinc chloride and zinc citrate. 10. The oral care composition of claim 1, wherein the arginine is present as free arginine. 11. The oral care composition of claim 1, wherein the arginine is present as an arginine salt selected from the group consisting of arginine bicarbonate, arginine hydroxide, arginine carbonate, arginine phosphate and mixtures thereof. 12. The oral care composition of claim 1, wherein the arginine is present within a 2-4 amino acid peptide. 13. The oral care composition of claim 1, wherein the composition further comprises an orally acceptable carrier. 14. The oral care composition of claim 1, wherein the composition is a toothpaste, a gel, a mouthwash, a mouthrinse, a lozenge, a spray, a gum, a tablet, or a film. 15. The oral care composition of claim 1, wherein the composition is a mouthwash. 16. The oral care composition of claim 1, wherein the composition is a toothpaste. 17. A method of reducing oral malodor in an oral cavity of a subject, the method comprising applying the oral care composition of claim 1 to the oral cavity. 18. (canceled)
An oral care composition comprising: arginine in free or salt form; and zinc ions, wherein the zinc ions are present in the composition at a concentration of from 0.0001 mols/100 g to 0.0150 mols/100 g, based on the total weight of the composition.1. An oral care composition comprising: a) arginine in free or salt form; and b) zinc ions, wherein the zinc ions are present in the composition at a concentration selected from the group consisting of from 0.0001 mols/100 g to 0.015 mols/100 g from 0.00025 mols/100 g, to 0.005 mols/100 g, from 0.0004 mols/100 g to 0.0015 mols/100 g, from 0.0005 mols/100 g to 0.001 mols/100 g, and from 0.00052 mols/100 g to 0.00062 mols/100 g, based on the total weight of the composition. 2. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.00025 mols/100 g to 0.005 mols/100 g, based on the total weight of the composition. 3. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.0004 mols/100 g to 0.0015 mols/100 g, based on the total weight of the composition. 4. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.0005 mols/100 g to 0.001 mols/100 g based on the total weight of the composition. 5. The oral care composition of claim 1, wherein the zinc ions are present in the composition at a concentration of from 0.00052 mols/100 g to 0.00062 mols/100 g, based on the total weight of the composition. 6. The oral care composition of claim 1, wherein the arginine is present in the composition at a concentration of from 0.1 weight % to 1 weight %, based on the total weight of the composition. 7. The oral care composition of claim 1, wherein the arginine is present in the composition at a concentration of from 0.5 weight % to 0.8 weight %, based on the total weight of the composition. 8. The oral care composition of claim 1, wherein the zinc ions are provided by at least one zinc compound chosen from zinc oxide, zinc lactate, zinc chloride, zinc citrate, zinc acetate, zinc borate, zinc butyrate, zinc carbonate, zinc formate, zinc gluconate, zinc glycerate, zinc glycolate, zinc phosphate, zinc picolinate, zinc proprionate, zinc salicylate, zinc silicate, zinc stearate, zinc tartrate, zinc undecylenate, zinc phosphate, zinc ricinoleate, zinc nitrate, and zinc sulfate. 9. The oral care composition of claim 1, wherein the zinc ions are provided by a zinc compound selected from group consisting of zinc, oxide, zinc lactate, zinc chloride and zinc citrate. 10. The oral care composition of claim 1, wherein the arginine is present as free arginine. 11. The oral care composition of claim 1, wherein the arginine is present as an arginine salt selected from the group consisting of arginine bicarbonate, arginine hydroxide, arginine carbonate, arginine phosphate and mixtures thereof. 12. The oral care composition of claim 1, wherein the arginine is present within a 2-4 amino acid peptide. 13. The oral care composition of claim 1, wherein the composition further comprises an orally acceptable carrier. 14. The oral care composition of claim 1, wherein the composition is a toothpaste, a gel, a mouthwash, a mouthrinse, a lozenge, a spray, a gum, a tablet, or a film. 15. The oral care composition of claim 1, wherein the composition is a mouthwash. 16. The oral care composition of claim 1, wherein the composition is a toothpaste. 17. A method of reducing oral malodor in an oral cavity of a subject, the method comprising applying the oral care composition of claim 1 to the oral cavity. 18. (canceled)
1,600
289
289
14,437,812
1,632
Several embodiments disclosed herein relate generally to compositions of stem cells and methods of utilizing said compositions for repair, regeneration, and/or functional recovery of damaged tissue. In some embodiments the compositions comprise therapeutic populations of stem cells depleted of cells expressing one or more specific markers, and methods of using the depleted therapeutic cell populations to repair and/or regenerate diseased or damaged tissue.
1-18. (canceled) 19. A composition of therapeutic cells derived from donor cardiac tissue comprising a population of cells substantially depleted of cells expressing the CD90 cell marker. 19-50. (canceled) 51. The composition of claim 19, wherein said therapeutic population of cells is depleted of at least 90%, 95% or 99% of cells expressing the CD90 marker. 52. The composition of claim 19, wherein said therapeutic population of cells is entirely depleted of cells expressing the CD90 marker. 53. The composition of claim 19, wherein therapeutic population of cells of cells comprises cardiosphere-derived cells (CDCs). 54. The composition of claim 53, wherein the CDCs are about 5 to about 35 microns in diameter and are suitable for systemic delivery to a recipient in need of repair of damaged or diseased cardiac tissue. 55. The composition of claim 19, wherein the therapeutic population of cells further comprises cardiac cells. 56. The composition of claim 55, wherein said cardiac cells express at least one of the following cardiac markers: myosin heavy chain, myosin light chain, alpha sarcomeric actin, Troponin-T, nkx2.5, and GATA-4. 57. The composition of claim 19, wherein the therapeutic population of cells further comprises endothelial cells. 58. The composition of claim 57, wherein said endothelial cells express at least one of the following endothelial markers: CD105, KDR, flk-1, CD31, von Willebrand factor, Ve-cadherin, and smooth muscle alpha actin. 59. The composition of claim 19, wherein said therapeutic population of cells is further substantially depleted of cells expressing the c-kit stem cell marker. 60. The composition of claim 59, wherein said therapeutic population of cells is depleted of at least 90%, 95% or 99% of cells expressing the c-kit stem cell marker. 61. The composition of claim 19, wherein said therapeutic population of cells comprises cardiospheres. 62. A method of treating a first subject having damaged cardiac tissue with allogeneic cells from a second subject, the method comprising: obtaining a population of cardiosphere-derived cells (CDCs) depleted of one or more subpopulation of cells, wherein a plurality of cells were harvested from the cardiac tissue of a second subject to generate said depleted CDCs, wherein said second subject is an adult, wherein said plurality of harvested cells were cultured to yield a population of CDCs; wherein said population of CDCs had one or more subpopulation of cells specifically depleted from the population of CDCs, thereby generating said depleted CDCs; and administering between about 1×106 and about 100×106 of said depleted CDCs to said first subject, wherein said administered depleted CDCs improve the function and/or viability of said damaged cardiac tissue, thereby treating said first subject. 63. The method of claim 62, wherein said depleted CDCs are substantially depleted of CDCs expressing the CD90 cell marker. 64. The method of claim 63, wherein said depleted CDCs are depleted of at least 90%, 95% or 99% of CDCs expressing the CD90 marker. 65. The method of claim 63, wherein said depleted CDCs are further substantially depleted of stem cells expressing the c-kit marker. 66. The method of claim 65, wherein said depleted CDCs are depleted of at least 90%, 95% or 99% of CDCs expressing the c-kit marker.
Several embodiments disclosed herein relate generally to compositions of stem cells and methods of utilizing said compositions for repair, regeneration, and/or functional recovery of damaged tissue. In some embodiments the compositions comprise therapeutic populations of stem cells depleted of cells expressing one or more specific markers, and methods of using the depleted therapeutic cell populations to repair and/or regenerate diseased or damaged tissue.1-18. (canceled) 19. A composition of therapeutic cells derived from donor cardiac tissue comprising a population of cells substantially depleted of cells expressing the CD90 cell marker. 19-50. (canceled) 51. The composition of claim 19, wherein said therapeutic population of cells is depleted of at least 90%, 95% or 99% of cells expressing the CD90 marker. 52. The composition of claim 19, wherein said therapeutic population of cells is entirely depleted of cells expressing the CD90 marker. 53. The composition of claim 19, wherein therapeutic population of cells of cells comprises cardiosphere-derived cells (CDCs). 54. The composition of claim 53, wherein the CDCs are about 5 to about 35 microns in diameter and are suitable for systemic delivery to a recipient in need of repair of damaged or diseased cardiac tissue. 55. The composition of claim 19, wherein the therapeutic population of cells further comprises cardiac cells. 56. The composition of claim 55, wherein said cardiac cells express at least one of the following cardiac markers: myosin heavy chain, myosin light chain, alpha sarcomeric actin, Troponin-T, nkx2.5, and GATA-4. 57. The composition of claim 19, wherein the therapeutic population of cells further comprises endothelial cells. 58. The composition of claim 57, wherein said endothelial cells express at least one of the following endothelial markers: CD105, KDR, flk-1, CD31, von Willebrand factor, Ve-cadherin, and smooth muscle alpha actin. 59. The composition of claim 19, wherein said therapeutic population of cells is further substantially depleted of cells expressing the c-kit stem cell marker. 60. The composition of claim 59, wherein said therapeutic population of cells is depleted of at least 90%, 95% or 99% of cells expressing the c-kit stem cell marker. 61. The composition of claim 19, wherein said therapeutic population of cells comprises cardiospheres. 62. A method of treating a first subject having damaged cardiac tissue with allogeneic cells from a second subject, the method comprising: obtaining a population of cardiosphere-derived cells (CDCs) depleted of one or more subpopulation of cells, wherein a plurality of cells were harvested from the cardiac tissue of a second subject to generate said depleted CDCs, wherein said second subject is an adult, wherein said plurality of harvested cells were cultured to yield a population of CDCs; wherein said population of CDCs had one or more subpopulation of cells specifically depleted from the population of CDCs, thereby generating said depleted CDCs; and administering between about 1×106 and about 100×106 of said depleted CDCs to said first subject, wherein said administered depleted CDCs improve the function and/or viability of said damaged cardiac tissue, thereby treating said first subject. 63. The method of claim 62, wherein said depleted CDCs are substantially depleted of CDCs expressing the CD90 cell marker. 64. The method of claim 63, wherein said depleted CDCs are depleted of at least 90%, 95% or 99% of CDCs expressing the CD90 marker. 65. The method of claim 63, wherein said depleted CDCs are further substantially depleted of stem cells expressing the c-kit marker. 66. The method of claim 65, wherein said depleted CDCs are depleted of at least 90%, 95% or 99% of CDCs expressing the c-kit marker.
1,600
290
290
14,443,862
1,639
The invention relates to a hydrodispersion, comprising thickeners, waxes, and oils. The waxes and the overall preparation have specific melting ranges that ensure a pleasant, sensorily light profile and good care effects.
1.-15. (canceled) 16. A hydrodispersion, wherein the hydrodispersion comprises, based on a total mass of the dispersion: (a) water, (b) from 0.05% to 5% by weight of at least one thickener, (c) from 2% to 20% by weight of one or more waxes, at least one wax having a melting point Tonset below 30° C., and (d) from 0% to 20% by weight of one or more oils. 17. The hydrodispersion of claim 16, wherein the hydrodispersion exhibits at least one melting range between 5° C. and 30° C. by DSC. 18. The hydrodispersion of claim 16, wherein (b) comprises at least one substance selected from homopolymers or copolymers of acrylic acid and/or acrylamide and derivatives thereof, organic natural compounds, organic modified natural substances, organic fully synthetic compounds. 19. The hydrodispersion of claim 18, wherein (b) comprises at least one substance selected from homopolymers or copolymers of acrylic acid and/or acrylamide and derivatives thereof, agar-agar, carrageenan, tragacanth, gum arabic, alginates, pectins, polyoses, guar flour, carob seed flour, starch, dextrins, gelatins, casein, xanthan gum, carboxymethyl cellulose, hydroxyethyl and/or hydroxypropyl cellulose, microcrystalline cellulose, polyacrylic and/or polymethacrylic compounds, vinyl polymers, polycarboxylic acids, polyethers, polyimines, polyamides, polyurethanes, AMPS copolymers, acrylic acid/VP Crosspolymer, PVP, acrylamide/ammonium acrylate copolymer. 20. The hydrodispersion of claim 16, wherein (b) comprises sodium polyacrylate. 21. The hydrodispersion of claim 16, wherein from 0.1% to 2% by weight of (b) are present. 22. The hydrodispersion of claim 16, wherein from 60% to 95% by weight of (a) are present. 23. The hydrodispersion of claim 21, wherein from 70% to 85% by weight of (a) are present. 24. The hydrodispersion of claim 16, wherein the at least one wax having a Tonset below 30° C. is selected from fats (triglycerides), mono- and diglycerides, natural and synthetic waxes, fatty alcohols, wax alcohols, fatty acids, esters of fatty alcohols and fatty acids, fatty acid amides, and any combinations of these substances. 25. The hydrodispersion of claim 16, wherein the at least one wax having a Tonset below 30° C. comprises at least one of Shorea stenoptera seed butter, hydrogenated vegetable oil, hydrogenated coco-glycerides, Butyrospermum parkii butter, Theobroma cacao (cocoa) seed butter, mango butter, methyl palmitate, hydrogenated palm kernel glycerides, hydrogenated palm glycerides, Acacia decurrens/jojoba/sunflower seed wax polyglyceryl-3 esters, cetearyl nonanoate, cetearyl alcohol. 26. The hydrodispersion of claim 16, wherein the hydrodispersion comprises more than 0.5% by weight of (d). 27. The hydrodispersion of claim 26, wherein the hydrodispersion comprises from 1% to 10% by weight of (d). 28. The hydrodispersion of claim 26, wherein a concentration of (c) is greater than a concentration of (d). 29. The hydrodispersion of claim 16, wherein the hydrodispersion does not contain any anionic emulsifiers or surfactants. 30. The hydrodispersion of claim 16, wherein the hydrodispersion does not contain any cationic emulsifiers or surfactants. 31. The hydrodispersion of claim 16, wherein the hydrodispersion does not contain any anionic or cationic emulsifiers or surfactants. 32. A cosmetic or dermatological composition, wherein the composition comprises or consists of the hydrodispersion of claim 16. 33. The composition of claim 32, wherein the composition is a skincare cream. 34. The composition of claim 32, wherein the composition is an eye cream. 35. A method of skincare, wherein the method comprises applying to skin in need of skincare the composition of claim 32.
The invention relates to a hydrodispersion, comprising thickeners, waxes, and oils. The waxes and the overall preparation have specific melting ranges that ensure a pleasant, sensorily light profile and good care effects.1.-15. (canceled) 16. A hydrodispersion, wherein the hydrodispersion comprises, based on a total mass of the dispersion: (a) water, (b) from 0.05% to 5% by weight of at least one thickener, (c) from 2% to 20% by weight of one or more waxes, at least one wax having a melting point Tonset below 30° C., and (d) from 0% to 20% by weight of one or more oils. 17. The hydrodispersion of claim 16, wherein the hydrodispersion exhibits at least one melting range between 5° C. and 30° C. by DSC. 18. The hydrodispersion of claim 16, wherein (b) comprises at least one substance selected from homopolymers or copolymers of acrylic acid and/or acrylamide and derivatives thereof, organic natural compounds, organic modified natural substances, organic fully synthetic compounds. 19. The hydrodispersion of claim 18, wherein (b) comprises at least one substance selected from homopolymers or copolymers of acrylic acid and/or acrylamide and derivatives thereof, agar-agar, carrageenan, tragacanth, gum arabic, alginates, pectins, polyoses, guar flour, carob seed flour, starch, dextrins, gelatins, casein, xanthan gum, carboxymethyl cellulose, hydroxyethyl and/or hydroxypropyl cellulose, microcrystalline cellulose, polyacrylic and/or polymethacrylic compounds, vinyl polymers, polycarboxylic acids, polyethers, polyimines, polyamides, polyurethanes, AMPS copolymers, acrylic acid/VP Crosspolymer, PVP, acrylamide/ammonium acrylate copolymer. 20. The hydrodispersion of claim 16, wherein (b) comprises sodium polyacrylate. 21. The hydrodispersion of claim 16, wherein from 0.1% to 2% by weight of (b) are present. 22. The hydrodispersion of claim 16, wherein from 60% to 95% by weight of (a) are present. 23. The hydrodispersion of claim 21, wherein from 70% to 85% by weight of (a) are present. 24. The hydrodispersion of claim 16, wherein the at least one wax having a Tonset below 30° C. is selected from fats (triglycerides), mono- and diglycerides, natural and synthetic waxes, fatty alcohols, wax alcohols, fatty acids, esters of fatty alcohols and fatty acids, fatty acid amides, and any combinations of these substances. 25. The hydrodispersion of claim 16, wherein the at least one wax having a Tonset below 30° C. comprises at least one of Shorea stenoptera seed butter, hydrogenated vegetable oil, hydrogenated coco-glycerides, Butyrospermum parkii butter, Theobroma cacao (cocoa) seed butter, mango butter, methyl palmitate, hydrogenated palm kernel glycerides, hydrogenated palm glycerides, Acacia decurrens/jojoba/sunflower seed wax polyglyceryl-3 esters, cetearyl nonanoate, cetearyl alcohol. 26. The hydrodispersion of claim 16, wherein the hydrodispersion comprises more than 0.5% by weight of (d). 27. The hydrodispersion of claim 26, wherein the hydrodispersion comprises from 1% to 10% by weight of (d). 28. The hydrodispersion of claim 26, wherein a concentration of (c) is greater than a concentration of (d). 29. The hydrodispersion of claim 16, wherein the hydrodispersion does not contain any anionic emulsifiers or surfactants. 30. The hydrodispersion of claim 16, wherein the hydrodispersion does not contain any cationic emulsifiers or surfactants. 31. The hydrodispersion of claim 16, wherein the hydrodispersion does not contain any anionic or cationic emulsifiers or surfactants. 32. A cosmetic or dermatological composition, wherein the composition comprises or consists of the hydrodispersion of claim 16. 33. The composition of claim 32, wherein the composition is a skincare cream. 34. The composition of claim 32, wherein the composition is an eye cream. 35. A method of skincare, wherein the method comprises applying to skin in need of skincare the composition of claim 32.
1,600
291
291
14,685,510
1,636
The present invention relates to targeted genome editing in eukaryotic cells or organisms. More particularly, the present invention relates to a composition for cleaving a target DNA in eukaryotic cells or organisms comprising a guide RNA specific for the target DNA and Cas protein-encoding nucleic acid or Cas protein, and use thereof.
1-57. (canceled) 58. A composition for cleaving a target nucleic acid sequence in a mammalian cell, the composition comprising a Type II Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas system in the mammalian cell, wherein the Type II CRISPR/Cas system comprises: a) a nucleic acid encoding a Cas9 polypeptide, wherein the Cas9 polypeptide comprises a nuclear localization signal, and b) a chimeric guide RNA comprising a CRISPR RNA (crRNA) portion fused to a trans activating crRNA (tracrRNA) portion, wherein the Cas9 polypeptide and the chimeric guide RNA form a Cas9/RNA complex in the mammalian cell and wherein the crRNA portion has sufficient sequence complementarity to the target nucleic acid sequence in the mammalian cell to allow the Cas9 polypeptide to mediate double stranded cleavage at the target nucleic acid sequence. 59. The composition of claim 58, wherein the nuclear localization signal is located at the C terminus of the Cas9 polypeptide. 60. The composition of claim 58, wherein the mammalian cell is a human cell. 61. The composition of claim 58, wherein the nucleic acid encoding the Cas 9 polypeptide is codon-optimized for expression in mammalian cells. 62. The composition of claim 58, wherein the chimeric guide RNA is in vitro transcribed RNA. 63. The composition of claim 58, wherein the target nucleic acid sequence is a genomic sequence located at its endogenous site in the genome of the mammalian cell. 64. The composition of claim 58, wherein the Cas9 polypeptide is a Streptococcus pyogenes Cas9 polypeptide. 65. The composition of claim 58, wherein the target nucleic acid sequence consists of 20 nucleotides complementary to the crRNA portion of the chimeric guide RNA and a trinucleotide protospacer adjacent motif (PAM), and wherein the PAM consists of the trinucleotide 5′-NGG-3′. 66. A method of introducing a site-specific, double-stranded break at a target nucleic acid sequence in a mammalian cell, the method comprising introducing into the mammalian cell a Type II Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas system, wherein the CRISPR/Cas system comprises: a) a nucleic acid encoding a Cas9 polypeptide, wherein the Cas9 polypeptide comprises a nuclear localization signal, and b) a chimeric guide RNA comprising a CRISPR RNA (crRNA) portion fused to a trans activating crRNA (tracrRNA) portion, wherein the Cas9 polypeptide and the chimeric guide RNA form a Cas9/RNA complex in the mammalian cell and wherein the crRNA portion has sufficient sequence complementarity to the target nucleic acid sequence in the mammalian cell to allow the Cas9 polypeptide to mediate double stranded cleavage at the target sequence. 67. The method of claim 66, wherein the nuclear localization signal is located at the C terminus of the Cas9 polypeptide. 68. The method of claim 66, wherein the mammalian cell is a human cell. 69. The method of claim 66, wherein the nucleic acid encoding the Cas 9 polypeptide is codon-optimized for expression in mammalian cells. 70. The method of claim 66, wherein the target nucleic acid sequence is a genomic sequence located at its endogenous site in the genome of the mammalian cell. 71. The method of claim 66, wherein the chimeric guide RNA is transcribed in vitro before introduction into the mammalian cell. 72. The method of claim 66, wherein the Cas9 polypeptide is a Streptococcus pyogenes Cas9 polypeptide. 73. The method of claim 66, wherein the target nucleic acid sequence consists of 20 nucleotides complementary to the crRNA portion of the chimeric guide RNA and a trinucleotide protospacer adjacent motif (PAM), wherein the PAM consists of the trinucleotide 5′-NGG-3′. 74. The method of claim 66, wherein the nucleic acid encoding the Cas9 protein is introduced into the mammalian cell before introducing the chimeric guide RNA into the mammalian cell. 75. A Type II Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas system for site-specific, double stranded cleavage of a target nucleic acid sequence in a mammalian cell, the CRISPR/Cas system comprising: a) a Cas9 polypeptide, wherein the Cas9 polypeptide comprises a nuclear localization signal, and b) a chimeric guide RNA comprising a CRISPR RNA (crRNA) portion and a trans-activating crRNA (tracrRNA) portion, wherein the Cas9 polypeptide and the chimeric guide RNA form a Cas9/RNA complex in the mammalian cell and wherein the crRNA portion has sufficient sequence complementarity to the target nucleic acid sequence in the mammalian cell to allow the Cas9 polypeptide to mediate double stranded cleavage at the target sequence. 76. The CRISPR/Cas system of claim 75, wherein the nuclear localization signal is located at the C terminus of the Cas9 polypeptide. 77. The CRISPR/Cas system of claim 75, wherein the mammalian cell is a human cell. 78. The CRISPR/Cas system of claim 75, wherein the nucleic acid encoding the Cas 9 polypeptide is codon-optimized for expression in mammalian cells. 79. The CRISPR/Cas system of claim 75, wherein the chimeric guide RNA is in vitro transcribed RNA. 80. The CRISPR/Cas system of claim 75, wherein the target nucleic acid sequence is a genomic sequence located at its endogenous site in the genome of the mammalian cell. 81. The CRISPR/Cas system of claim 75, wherein the Cas9 polypeptide is a Streptococcus pyogenes Cas9 polypeptide.
The present invention relates to targeted genome editing in eukaryotic cells or organisms. More particularly, the present invention relates to a composition for cleaving a target DNA in eukaryotic cells or organisms comprising a guide RNA specific for the target DNA and Cas protein-encoding nucleic acid or Cas protein, and use thereof.1-57. (canceled) 58. A composition for cleaving a target nucleic acid sequence in a mammalian cell, the composition comprising a Type II Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas system in the mammalian cell, wherein the Type II CRISPR/Cas system comprises: a) a nucleic acid encoding a Cas9 polypeptide, wherein the Cas9 polypeptide comprises a nuclear localization signal, and b) a chimeric guide RNA comprising a CRISPR RNA (crRNA) portion fused to a trans activating crRNA (tracrRNA) portion, wherein the Cas9 polypeptide and the chimeric guide RNA form a Cas9/RNA complex in the mammalian cell and wherein the crRNA portion has sufficient sequence complementarity to the target nucleic acid sequence in the mammalian cell to allow the Cas9 polypeptide to mediate double stranded cleavage at the target nucleic acid sequence. 59. The composition of claim 58, wherein the nuclear localization signal is located at the C terminus of the Cas9 polypeptide. 60. The composition of claim 58, wherein the mammalian cell is a human cell. 61. The composition of claim 58, wherein the nucleic acid encoding the Cas 9 polypeptide is codon-optimized for expression in mammalian cells. 62. The composition of claim 58, wherein the chimeric guide RNA is in vitro transcribed RNA. 63. The composition of claim 58, wherein the target nucleic acid sequence is a genomic sequence located at its endogenous site in the genome of the mammalian cell. 64. The composition of claim 58, wherein the Cas9 polypeptide is a Streptococcus pyogenes Cas9 polypeptide. 65. The composition of claim 58, wherein the target nucleic acid sequence consists of 20 nucleotides complementary to the crRNA portion of the chimeric guide RNA and a trinucleotide protospacer adjacent motif (PAM), and wherein the PAM consists of the trinucleotide 5′-NGG-3′. 66. A method of introducing a site-specific, double-stranded break at a target nucleic acid sequence in a mammalian cell, the method comprising introducing into the mammalian cell a Type II Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas system, wherein the CRISPR/Cas system comprises: a) a nucleic acid encoding a Cas9 polypeptide, wherein the Cas9 polypeptide comprises a nuclear localization signal, and b) a chimeric guide RNA comprising a CRISPR RNA (crRNA) portion fused to a trans activating crRNA (tracrRNA) portion, wherein the Cas9 polypeptide and the chimeric guide RNA form a Cas9/RNA complex in the mammalian cell and wherein the crRNA portion has sufficient sequence complementarity to the target nucleic acid sequence in the mammalian cell to allow the Cas9 polypeptide to mediate double stranded cleavage at the target sequence. 67. The method of claim 66, wherein the nuclear localization signal is located at the C terminus of the Cas9 polypeptide. 68. The method of claim 66, wherein the mammalian cell is a human cell. 69. The method of claim 66, wherein the nucleic acid encoding the Cas 9 polypeptide is codon-optimized for expression in mammalian cells. 70. The method of claim 66, wherein the target nucleic acid sequence is a genomic sequence located at its endogenous site in the genome of the mammalian cell. 71. The method of claim 66, wherein the chimeric guide RNA is transcribed in vitro before introduction into the mammalian cell. 72. The method of claim 66, wherein the Cas9 polypeptide is a Streptococcus pyogenes Cas9 polypeptide. 73. The method of claim 66, wherein the target nucleic acid sequence consists of 20 nucleotides complementary to the crRNA portion of the chimeric guide RNA and a trinucleotide protospacer adjacent motif (PAM), wherein the PAM consists of the trinucleotide 5′-NGG-3′. 74. The method of claim 66, wherein the nucleic acid encoding the Cas9 protein is introduced into the mammalian cell before introducing the chimeric guide RNA into the mammalian cell. 75. A Type II Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas system for site-specific, double stranded cleavage of a target nucleic acid sequence in a mammalian cell, the CRISPR/Cas system comprising: a) a Cas9 polypeptide, wherein the Cas9 polypeptide comprises a nuclear localization signal, and b) a chimeric guide RNA comprising a CRISPR RNA (crRNA) portion and a trans-activating crRNA (tracrRNA) portion, wherein the Cas9 polypeptide and the chimeric guide RNA form a Cas9/RNA complex in the mammalian cell and wherein the crRNA portion has sufficient sequence complementarity to the target nucleic acid sequence in the mammalian cell to allow the Cas9 polypeptide to mediate double stranded cleavage at the target sequence. 76. The CRISPR/Cas system of claim 75, wherein the nuclear localization signal is located at the C terminus of the Cas9 polypeptide. 77. The CRISPR/Cas system of claim 75, wherein the mammalian cell is a human cell. 78. The CRISPR/Cas system of claim 75, wherein the nucleic acid encoding the Cas 9 polypeptide is codon-optimized for expression in mammalian cells. 79. The CRISPR/Cas system of claim 75, wherein the chimeric guide RNA is in vitro transcribed RNA. 80. The CRISPR/Cas system of claim 75, wherein the target nucleic acid sequence is a genomic sequence located at its endogenous site in the genome of the mammalian cell. 81. The CRISPR/Cas system of claim 75, wherein the Cas9 polypeptide is a Streptococcus pyogenes Cas9 polypeptide.
1,600
292
292
14,556,510
1,611
A controlled-release pharmaceutical composition including first and second groups of microparticles, each of the microparticles including a core including tamsulosin or pharmaceutically acceptable salts thereof, a controlled-release polymer coating layer formed on the core, and an enteric polymer outer layer formed on the controlled-release polymer coating layer, wherein the average thickness of the controlled-release polymer coating layer is different in each of the first and second groups of microparticles, and an oral formulation including the same, are provided. This pharmaceutical composition can easily control the extent of release of an active ingredient depending on changes in pH in the intestinal tract and the release pattern of the active ingredient in the small intestine, thus preventing the active ingredient from being rapidly transferred into the blood to thereby minimize side-effects, and maintaining the effective blood concentration of the active ingredient for a predetermined period of time. Furthermore, this composition can shield the bitter taste of the active ingredient even when exposed to the inside of the mouth, thus increasing the therapeutic effects for patients upon oral administration.
1-20. (canceled) 21. A method for controlling tamsulosin release so that upon oral administration, 30% or less of tamsulosin is released for 2 hours and the rest of tamsulosin is gradually released for 6 hours or longer, which comprises: administering a pharmaceutical composition which comprises a first group of microparticles and a second group of microparticles, each of the microparticles comprising: a core comprising tamsulosin or pharmaceutically acceptable salts thereof; a controlled-release polymer coating layer formed on the core; and an enteric polymer outer layer formed on the controlled-release polymer coating layer, wherein an average thickness of the controlled-release polymer coating layer is different between the first group of microparticles and the second group of microparticles, said controlled-release polymer coating layer comprises a water-insoluble polymer an ethyl acrylate/methyl methacrylate/trimethylammonioethyl methacrylate chloride copolymer, said enteric polymer outer layer comprises methacrylic acid/ethyl acrylate copolymer; wherein said first group of microparticles has an average thickness of the controlled-release polymer coating layer of 1˜20 μm, and wherein an average thickness ratio of the controlled-release polymer coating layer in the first group of microparticles and the second group of microparticles is 1:1.2˜10, a weight ratio of the first group of microparticles and the second group of microparticles is adjusted so that a total weight ratio of tamsulosin or pharmaceutically acceptable salts thereof comprised in the first group of microparticles and the second group of microparticles falls in a range of 1:0.1˜10, the controlled-release polymer coating layer of each of the groups of microparticles has a same polymer composition; and said core is formed by mixing tamsulosin or pharmaceutically acceptable salts thereof with an inert seed, or by incorporating tamsulosin or pharmaceutically acceptable salts thereof in the inert seed. 22. The method of claim 21, wherein the pharmaceutical composition further comprises a third group of microparticles, each of said microparticles in said third group comprising: a core including tamsulosin or pharmaceutically acceptable salts thereof; a controlled-release polymer coating layer formed on the core; and an enteric polymer outer layer formed on the controlled-release polymer coating layer; wherein an average thickness of the controlled-release polymer coating layer of the third group of microparticles is different from the average thickness of the controlled-release polymer coating layer of the first group of microparticles and the second group of microparticles, said controlled-release polymer coating layer comprises a water-insoluble polymer an ethyl acrylate/methyl methacrylate/trimethylammonioethyl methacrylate chloride copolymer, said enteric polymer outer layer comprises methacrylic acid/ethyl acrylate copolymer, the controlled-release polymer coating layer of each of the groups of microparticles has a same polymer composition, and wherein an average thickness ratio of the controlled-release polymer coating layer in the second group of microparticles and the third group of microparticles is 1:1.2˜5. 23. The method of claim 22, wherein a weight ratio of the first group of microparticles, the second group of microparticles, and the third group of microparticles is adjusted so that a total weight ratio of tamsulosin or pharmaceutically acceptable salts thereof included in the first group of microparticles, the second group of microparticles, and the third group of microparticles falls in a range of 1:0.1˜10:0.1˜10. 24. The method of claim 21, wherein an average diameter of each of the groups of microparticles is 10˜4500 μm. 25. The method of claim 21, wherein the controlled-release polymer coating layer further comprises one or more selected from the group consisting of: water-soluble polymers comprising methyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinyl alcohol, polyethylene glycol, and polypropylene glycol; enteric polymers comprising hydroxypropyl methylcellulose acetate succinate, hydroxypropyl methylcellulose phthalate, hydroxymethyl ethylcellulose phthalate, cellulose acetate phthalate, cellulose acetate succinate, cellulose acetate maleate, cellulose benzoate phthalate, cellulose propionate phthalate, methylcellulose phthalate, carboxylmethyl ethylcellulose, ethylhydroxyethyl cellulose phthalate, a styrene/acrylic acid copolymer, a methyl acrylate/acrylic acid copolymer, a methyl acrylate/methacrylic acid copolymer, a butyl acrylate/styrene.acrylic acid copolymer, a methacrylic acid/ethyl acrylate copolymer, a methyl acrylate.methacrylic acid/octyl acrylate copolymer, a vinyl acetate/maleic anhydride copolymer, an ethylene/maleic anhydride copolymer, a vinyl butyl ether/maleic anhydride copolymer, an acrylonitrile/methyl acrylate.maleic anhydride copolymer, a butyl acrylate/styrene.maleic anhydride copolymer, polyvinyl alcohol phthalate, polyvinylacetal phthalate, polyvinylbutyrate phthalate, and polyvinylacetoacetal phthalate; and gastric polymers comprising polyvinylacetal diethylamino acetate, and a methyl methacrylate/butyl methacrylate/dimethylaminoethyl methacrylate copolymer. 26. The method of claim 21, wherein the enteric polymer outer layer further comprises one or more selected from the group consisting of: water-insoluble polymers comprising ethylcellulose, an ethyl acrylate/methyl methacrylate/trimethylammonioethyl methacrylate chloride copolymer, a methyl methacrylate/ethyl acrylate copolymer, and polyvinylacetate; water-soluble polymers comprising methylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinyl alcohol, polyethylene glycol, and polypropylene glycol; and gastric polymers comprising polyvinylacetal diethylamino acetate and a methyl methacrylate/butyl methacrylate/dimethylaminoethyl methacrylate copolymer. 27. The method of claim 21, wherein one or more of the core, the controlled-release polymer coating layer, and the enteric polymer outer layer further comprise one or more selected from the group consisting of a saccharide, a sugar alcohol, a water-soluble polymer, a water-insoluble polymer, an enteric polymer, a gastric polymer, a colorant, a flavorant, a sweetener, a surfactant, a lubricant, a stabilizer, an antioxidant, a foaming agent, paraffin, wax, an antifoaming agent, and a plasticizer. 28. The method of claim 27, wherein the plasticizer is one or more selected from the group consisting of triethyl citrate, dibutyl phthalate, diethyl phthalate, dibutyl sebacate, diethyl sebacate, tributyl citrate, acetyl triethyl citrate, propylene glycol, triacetin, polyethylene glycol, cetyl alcohol, stearyl alcohol, and cetostearyl alcohol. 29. The method of claim 21, wherein the pharmaceutical composition has a dissolution pattern in which, according to a dissolution test method of Korean Pharmacopoeia 8th revision (KP VIII) (a second paddle method: 100 rotations per min, 500 ml of dissolution solutions at pH 1.2 and pH 7.2): a) with respect to a dissolution solution at pH 1.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 1˜30 wt % within 2 h, based on a total weight thereof; and b) with respect to a dissolution solution at pH 7.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 10˜60 wt % within 30 min, 30˜80 wt % within 1 h, and 50 wt % or more within 4 h, based on the total weight thereof. 30. The method of claim 21, wherein the pharmaceutical composition has a dissolution pattern in which, according to a dissolution test method of Korean Pharmacopoeia 8th revision (KP VIII) (a second paddle method: 100 rotations per min, 500 ml of dissolution solutions at pH 1.2 and pH 7.2): a) with respect to a dissolution solution at pH 1.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 5˜15 wt % within 2 h, based on a total weight thereof; and b) with respect to a dissolution solution at pH 7.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 17˜55 wt % within 30 min, 30˜75 wt % within 1 h, and 80 wt % or more within 4 h, based on the total weight thereof. 31. The method of claim 21, wherein the pharmaceutical composition is administered in form of an oral formulation. 32. The method of claim 31, wherein the oral formulation is a capsule, a normal tablet, a double layer tablet, a chewable tablet, an orally disintegrating tablet, a dry syrup formulation, a syrup, a jelly formulation, or a granule.
A controlled-release pharmaceutical composition including first and second groups of microparticles, each of the microparticles including a core including tamsulosin or pharmaceutically acceptable salts thereof, a controlled-release polymer coating layer formed on the core, and an enteric polymer outer layer formed on the controlled-release polymer coating layer, wherein the average thickness of the controlled-release polymer coating layer is different in each of the first and second groups of microparticles, and an oral formulation including the same, are provided. This pharmaceutical composition can easily control the extent of release of an active ingredient depending on changes in pH in the intestinal tract and the release pattern of the active ingredient in the small intestine, thus preventing the active ingredient from being rapidly transferred into the blood to thereby minimize side-effects, and maintaining the effective blood concentration of the active ingredient for a predetermined period of time. Furthermore, this composition can shield the bitter taste of the active ingredient even when exposed to the inside of the mouth, thus increasing the therapeutic effects for patients upon oral administration.1-20. (canceled) 21. A method for controlling tamsulosin release so that upon oral administration, 30% or less of tamsulosin is released for 2 hours and the rest of tamsulosin is gradually released for 6 hours or longer, which comprises: administering a pharmaceutical composition which comprises a first group of microparticles and a second group of microparticles, each of the microparticles comprising: a core comprising tamsulosin or pharmaceutically acceptable salts thereof; a controlled-release polymer coating layer formed on the core; and an enteric polymer outer layer formed on the controlled-release polymer coating layer, wherein an average thickness of the controlled-release polymer coating layer is different between the first group of microparticles and the second group of microparticles, said controlled-release polymer coating layer comprises a water-insoluble polymer an ethyl acrylate/methyl methacrylate/trimethylammonioethyl methacrylate chloride copolymer, said enteric polymer outer layer comprises methacrylic acid/ethyl acrylate copolymer; wherein said first group of microparticles has an average thickness of the controlled-release polymer coating layer of 1˜20 μm, and wherein an average thickness ratio of the controlled-release polymer coating layer in the first group of microparticles and the second group of microparticles is 1:1.2˜10, a weight ratio of the first group of microparticles and the second group of microparticles is adjusted so that a total weight ratio of tamsulosin or pharmaceutically acceptable salts thereof comprised in the first group of microparticles and the second group of microparticles falls in a range of 1:0.1˜10, the controlled-release polymer coating layer of each of the groups of microparticles has a same polymer composition; and said core is formed by mixing tamsulosin or pharmaceutically acceptable salts thereof with an inert seed, or by incorporating tamsulosin or pharmaceutically acceptable salts thereof in the inert seed. 22. The method of claim 21, wherein the pharmaceutical composition further comprises a third group of microparticles, each of said microparticles in said third group comprising: a core including tamsulosin or pharmaceutically acceptable salts thereof; a controlled-release polymer coating layer formed on the core; and an enteric polymer outer layer formed on the controlled-release polymer coating layer; wherein an average thickness of the controlled-release polymer coating layer of the third group of microparticles is different from the average thickness of the controlled-release polymer coating layer of the first group of microparticles and the second group of microparticles, said controlled-release polymer coating layer comprises a water-insoluble polymer an ethyl acrylate/methyl methacrylate/trimethylammonioethyl methacrylate chloride copolymer, said enteric polymer outer layer comprises methacrylic acid/ethyl acrylate copolymer, the controlled-release polymer coating layer of each of the groups of microparticles has a same polymer composition, and wherein an average thickness ratio of the controlled-release polymer coating layer in the second group of microparticles and the third group of microparticles is 1:1.2˜5. 23. The method of claim 22, wherein a weight ratio of the first group of microparticles, the second group of microparticles, and the third group of microparticles is adjusted so that a total weight ratio of tamsulosin or pharmaceutically acceptable salts thereof included in the first group of microparticles, the second group of microparticles, and the third group of microparticles falls in a range of 1:0.1˜10:0.1˜10. 24. The method of claim 21, wherein an average diameter of each of the groups of microparticles is 10˜4500 μm. 25. The method of claim 21, wherein the controlled-release polymer coating layer further comprises one or more selected from the group consisting of: water-soluble polymers comprising methyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinyl alcohol, polyethylene glycol, and polypropylene glycol; enteric polymers comprising hydroxypropyl methylcellulose acetate succinate, hydroxypropyl methylcellulose phthalate, hydroxymethyl ethylcellulose phthalate, cellulose acetate phthalate, cellulose acetate succinate, cellulose acetate maleate, cellulose benzoate phthalate, cellulose propionate phthalate, methylcellulose phthalate, carboxylmethyl ethylcellulose, ethylhydroxyethyl cellulose phthalate, a styrene/acrylic acid copolymer, a methyl acrylate/acrylic acid copolymer, a methyl acrylate/methacrylic acid copolymer, a butyl acrylate/styrene.acrylic acid copolymer, a methacrylic acid/ethyl acrylate copolymer, a methyl acrylate.methacrylic acid/octyl acrylate copolymer, a vinyl acetate/maleic anhydride copolymer, an ethylene/maleic anhydride copolymer, a vinyl butyl ether/maleic anhydride copolymer, an acrylonitrile/methyl acrylate.maleic anhydride copolymer, a butyl acrylate/styrene.maleic anhydride copolymer, polyvinyl alcohol phthalate, polyvinylacetal phthalate, polyvinylbutyrate phthalate, and polyvinylacetoacetal phthalate; and gastric polymers comprising polyvinylacetal diethylamino acetate, and a methyl methacrylate/butyl methacrylate/dimethylaminoethyl methacrylate copolymer. 26. The method of claim 21, wherein the enteric polymer outer layer further comprises one or more selected from the group consisting of: water-insoluble polymers comprising ethylcellulose, an ethyl acrylate/methyl methacrylate/trimethylammonioethyl methacrylate chloride copolymer, a methyl methacrylate/ethyl acrylate copolymer, and polyvinylacetate; water-soluble polymers comprising methylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinyl alcohol, polyethylene glycol, and polypropylene glycol; and gastric polymers comprising polyvinylacetal diethylamino acetate and a methyl methacrylate/butyl methacrylate/dimethylaminoethyl methacrylate copolymer. 27. The method of claim 21, wherein one or more of the core, the controlled-release polymer coating layer, and the enteric polymer outer layer further comprise one or more selected from the group consisting of a saccharide, a sugar alcohol, a water-soluble polymer, a water-insoluble polymer, an enteric polymer, a gastric polymer, a colorant, a flavorant, a sweetener, a surfactant, a lubricant, a stabilizer, an antioxidant, a foaming agent, paraffin, wax, an antifoaming agent, and a plasticizer. 28. The method of claim 27, wherein the plasticizer is one or more selected from the group consisting of triethyl citrate, dibutyl phthalate, diethyl phthalate, dibutyl sebacate, diethyl sebacate, tributyl citrate, acetyl triethyl citrate, propylene glycol, triacetin, polyethylene glycol, cetyl alcohol, stearyl alcohol, and cetostearyl alcohol. 29. The method of claim 21, wherein the pharmaceutical composition has a dissolution pattern in which, according to a dissolution test method of Korean Pharmacopoeia 8th revision (KP VIII) (a second paddle method: 100 rotations per min, 500 ml of dissolution solutions at pH 1.2 and pH 7.2): a) with respect to a dissolution solution at pH 1.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 1˜30 wt % within 2 h, based on a total weight thereof; and b) with respect to a dissolution solution at pH 7.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 10˜60 wt % within 30 min, 30˜80 wt % within 1 h, and 50 wt % or more within 4 h, based on the total weight thereof. 30. The method of claim 21, wherein the pharmaceutical composition has a dissolution pattern in which, according to a dissolution test method of Korean Pharmacopoeia 8th revision (KP VIII) (a second paddle method: 100 rotations per min, 500 ml of dissolution solutions at pH 1.2 and pH 7.2): a) with respect to a dissolution solution at pH 1.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 5˜15 wt % within 2 h, based on a total weight thereof; and b) with respect to a dissolution solution at pH 7.2, the tamsulosin or pharmaceutically acceptable salts thereof are dissolved in an amount of 17˜55 wt % within 30 min, 30˜75 wt % within 1 h, and 80 wt % or more within 4 h, based on the total weight thereof. 31. The method of claim 21, wherein the pharmaceutical composition is administered in form of an oral formulation. 32. The method of claim 31, wherein the oral formulation is a capsule, a normal tablet, a double layer tablet, a chewable tablet, an orally disintegrating tablet, a dry syrup formulation, a syrup, a jelly formulation, or a granule.
1,600
293
293
14,359,385
1,636
An in vitro method for screening for candidate compounds for preventing and/or attenuating skin ageing, and/or hydrating skin, includes: a) contacting a test compound with a sample of papillary fibroblasts; b) measuring the expression of a gene selected from PDPN, CCRL1 and NTN1, in the papillary fibroblasts; and c) selecting compounds for which an activation of at least 1.5 fold of the expression of at least one of the genes is measured in the treated papillary fibroblasts compared with untreated papillary fibroblasts. Another in vitro method includes: a) contacting a test compound with a sample of reticular fibroblasts; b) measuring the expression of a gene selected from MGP, PPP1R14A and TGM2, in the reticular fibroblasts; and c) selecting compounds for which an activation of at most 1.0 fold of the expression of at least one of the genes is measured in the treated reticular fibroblasts compared with untreated reticular fibroblasts.
1. In vitro method for screening for candidate compounds for preventing and/or attenuating ageing of the skin, and/or for hydrating the skin, comprising the following steps: a. bringing at least one test compound in contact with a sample of papillary fibroblasts; b. measuring the expression of at least one gene selected from PDPN, CCRL1 and NTN1, in said papillary fibroblastss; c. selecting the compounds for which an activation of at least 1.5 fold of the expression of at least one of said genes is measured in the papillary fibroblasts treated in a. compared with the untreated papillary fibroblasts. 2. In vitro method for screening for candidate compounds for preventing and/or attenuating ageing of the skin, and/or for hydrating the skin, comprising the following steps: a. bringing at least one test compound in contact with a sample of reticular fibroblasts ; b. measuring the expression of at least one gene selected from TGM2, MGP and PPP1R14A, in said reticular fibroblasts; c. selecting the compounds for which an activation of at most 1.0 fold of the expression of at least one of said genes is measured in the reticular fibroblasts treated in a. compared with the untreated reticular fibroblasts. 3. In vitro method for detecting the presence of papillary fibroblasts and of reticular fibroblasts in a biological sample, comprising the step of measuring the expression of at least one gene selected in the group consisting of PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 in said fibroblasts. 4. In vitro method for monitoring the differentiation of papillary fibroblasts into reticular fibroblasts in a biological sample, comprising the step of measuring the expression of at least one gene selected in the group consisting of PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 in said fibroblasts. 5. Method according to claim 1, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 6. Method according to claim, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 7. Method according to claim, characterized in that the test compounds are chosen from botanical extracts. 8. Method according to claim 2, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 9. Method according to claim 2, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 10. Method according to claim 2, characterized in that the test compounds are chosen from botanical extracts. 11. Method according to claim 3, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 12. Method according to claim 3, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 13. Method according to claim 3, characterized in that the test compounds are chosen from botanical extracts. 14. Method according to claim 4, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 15. Method according to claim 4, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 16. Method according to claim 4, characterized in that the test compounds are chosen from botanical extracts. 17. Method according to claim 5, characterized in that the test compounds are chosen from botanical extracts. 18. Method according to claim 6, characterized in that the test compounds are chosen from botanical extracts.
An in vitro method for screening for candidate compounds for preventing and/or attenuating skin ageing, and/or hydrating skin, includes: a) contacting a test compound with a sample of papillary fibroblasts; b) measuring the expression of a gene selected from PDPN, CCRL1 and NTN1, in the papillary fibroblasts; and c) selecting compounds for which an activation of at least 1.5 fold of the expression of at least one of the genes is measured in the treated papillary fibroblasts compared with untreated papillary fibroblasts. Another in vitro method includes: a) contacting a test compound with a sample of reticular fibroblasts; b) measuring the expression of a gene selected from MGP, PPP1R14A and TGM2, in the reticular fibroblasts; and c) selecting compounds for which an activation of at most 1.0 fold of the expression of at least one of the genes is measured in the treated reticular fibroblasts compared with untreated reticular fibroblasts.1. In vitro method for screening for candidate compounds for preventing and/or attenuating ageing of the skin, and/or for hydrating the skin, comprising the following steps: a. bringing at least one test compound in contact with a sample of papillary fibroblasts; b. measuring the expression of at least one gene selected from PDPN, CCRL1 and NTN1, in said papillary fibroblastss; c. selecting the compounds for which an activation of at least 1.5 fold of the expression of at least one of said genes is measured in the papillary fibroblasts treated in a. compared with the untreated papillary fibroblasts. 2. In vitro method for screening for candidate compounds for preventing and/or attenuating ageing of the skin, and/or for hydrating the skin, comprising the following steps: a. bringing at least one test compound in contact with a sample of reticular fibroblasts ; b. measuring the expression of at least one gene selected from TGM2, MGP and PPP1R14A, in said reticular fibroblasts; c. selecting the compounds for which an activation of at most 1.0 fold of the expression of at least one of said genes is measured in the reticular fibroblasts treated in a. compared with the untreated reticular fibroblasts. 3. In vitro method for detecting the presence of papillary fibroblasts and of reticular fibroblasts in a biological sample, comprising the step of measuring the expression of at least one gene selected in the group consisting of PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 in said fibroblasts. 4. In vitro method for monitoring the differentiation of papillary fibroblasts into reticular fibroblasts in a biological sample, comprising the step of measuring the expression of at least one gene selected in the group consisting of PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 in said fibroblasts. 5. Method according to claim 1, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 6. Method according to claim, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 7. Method according to claim, characterized in that the test compounds are chosen from botanical extracts. 8. Method according to claim 2, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 9. Method according to claim 2, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 10. Method according to claim 2, characterized in that the test compounds are chosen from botanical extracts. 11. Method according to claim 3, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 12. Method according to claim 3, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 13. Method according to claim 3, characterized in that the test compounds are chosen from botanical extracts. 14. Method according to claim 4, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the mRNA of the corresponding gene. 15. Method according to claim 4, characterized in that the expression of at least one gene selected from PDPN, MGP, CCRL1, PPP1R14A, NTN1 and TGM2 is measured by quantifying the protein encoded by the corresponding gene. 16. Method according to claim 4, characterized in that the test compounds are chosen from botanical extracts. 17. Method according to claim 5, characterized in that the test compounds are chosen from botanical extracts. 18. Method according to claim 6, characterized in that the test compounds are chosen from botanical extracts.
1,600
294
294
13,882,566
1,632
Provided herein are methods of inducing psychosis in animals using light-responsive opsins and methods of identifying or screening compounds that may be useful in treating psychosis.
1. A non-human animal comprising a light-responsive opsin expressed on the cell membrane of a subset of layer V pyramidal neurons in the prefrontal cortex, wherein light activation of the opsin induces depolarization of the membrane and induces psychosis of the animal. 2. The animal of claim 1, wherein the subset of layer V pyramidal neurons have a single large apical dendrite. 3. The animal of claim 1, wherein the opsin is selected from the group consisting of ChR2, VChR1, and DChR. 4. (canceled) 5. A prefrontal cortex tissue slice comprising a subset of layer V pyramidal neurons, wherein a light-responsive opsin is expressed on the cell membrane of the apical dendrites in layer V pyramidal neurons, and wherein light activation of the opsin induces depolarization of the membrane. 6. The prefrontal cortex tissue slice of claim 5, wherein the subset of layer V pyramidal neurons have a single large apical dendrite. 7. The prefrontal cortex tissue slice of claim 5, wherein the opsin is selected from the group consisting of ChR2, VChR1, and DChR. 8. The prefrontal cortex tissue of claim 5, wherein the opsin is selected from the group consisting of SFO, SSFO, C1V1, C1V1-E122T, C1V1-E162T, and C1V1-E122T/E162T. 9. A method of inducing psychosis in a non-human animal, comprising activating a light-responsive opsin by light, wherein the light-responsive opsin is expressed on the cell membrane of a subset of layer V pyramidal neurons in the prefrontal cortex in the animal, and wherein the light activation of the opsin induces depolarization of the cell membrane. 10. The method of claim 9, wherein the subset of layer V pyramidal neurons have a single large apical dendrite. 11. The method of claim 9, wherein the opsin is selected from the group consisting of ChR2, VChR1, and DChR. 12. The method of claim 9, wherein the opsin is selected from the group consisting of SFO, SSFO, C1V1, C1V1-E122T, C1V1-E162T, and C1V1-E122T/E162T. 13. A method of identifying a candidate compound for treating psychosis, the method comprising measuring a psychotic state of a non-human animal before and after administering the compound to the prefrontal cortex of the animal, wherein the psychotic state is induced by light activation of a light-responsive opsin expressed on the cell membrane of a layer of V pyramidal neurons in the animal, and activation of the opsin induces depolarization of the membrane; wherein an improvement in one or more of psychotic state measurements after the administration of the compound indicates that the compound is a candidate for treating psychosis. 14. The method of claim 13, wherein the psychotic state measurement is a behavioral measurement. 15. The method of claim 13, wherein the psychotic state measurement is a cellular measurement. 16. The method of claim 9, further comprising a step of administering a D2 agonist to the animal before administration of the compound. 17. A method of identifying a candidate compound for treating psychosis, the method comprising: measuring a psychotic state of a prefrontal cortex tissue slice before and after incubating the tissue slice with the compound, wherein the prefrontal cortex tissue slice comprises a layer of V pyramidal neurons and a light-responsive opsin is expressed on the cell membrane of the layer of V pyramidal neurons, wherein the psychotic state is induced by the membrane depolarization of the neurons induced by activation of the light-responsive opsin; wherein an improvement in one or more of a psychotic state readouts after incubation with the compound indicates that the compound is a candidate for treating psychosis. 18. The method of claim 17, wherein the psychotic state measurement is a cellular measurement. 19. The method of claim 17, further comprising a step of incubating a D2 agonist with the prefrontal cortex tissue slice before incubation with the compound. 20. A method comprising: providing optical stimulation to a target neuron population that expresses a light-responsive opsin; measuring a first electrical pattern of the target neuron population in response to the optical stimulation; introducing a drug, known to induce psychosis, to the target neuron population; providing optical stimulation to the target neuron population measuring a subsequent electrical pattern of the target neuron population in response to the optical stimulation; and comparing the first electrical pattern and the subsequent electrical pattern. 21. The method of claim 20, further including identifying a subset of neurons associated with psychosis. 22. The method of claim 21, wherein the subset of neurons is a subset of level 5 pyramidal neurons. 23. The method of claim 21, wherein the target neuron cell population is in a patient. 24. The method of claim 23, further including providing a potential treatment to the patient and observing a third electrical pattern in response to light during and after the potential treatment. 25. The method of claim 24, further including comparing the third electrical pattern to the first and second electrical patterns and assessing the efficacy of the potential treatment. 26. The method of claim 20, further comprising elevating activity within a Thy1-expressing subset of prefrontal cortical neurons. 27-32. (canceled) 33. A method comprising: modifying a target neuron population with a light-responsive molecule, the neurons of the target neuron population having a single, large apical dendrite; providing light to the target neuron population, the light activating the light-responsive molecule; and introducing a drug to the target neuron population; the drug causing the membrane potential of the neurons to remain elevated after removal of the light. 34. The method of claim 33, wherein the light-responsive molecule excites the target neuron population in response to light. 35. The method of claim 34, wherein the light-responsive molecule is ChR2. 36. The method of claim 33, wherein the target neuron population is a subset of layer V pyramidal neurons. 37. The method of claim 33, wherein the elevation of the membrane potential inhibits firing of the cell in response to a stimulus. 38. The method of claim 33, wherein the elevation of the membrane potential results in firing after a depolarizing current is removed. 39. The method of claim 33, wherein the apical dendrite extends into superficial layers of the brain. 40. The method of claim 33, further including determining the source of the elevated membrane potential. 41. The method of claim 33, wherein the drug induces psychosis. 42. The method of claim 33, wherein L-type calcium channels of the target neuron population are involved in an activity-dependent depolarization. 43-49. (canceled) 50. The non-human animal of claim 1, wherein the opsin comprises an amino acid sequence having at least 90% amino acid sequence identity to the amino acid sequence set forth in one of SEQ ID NOs:1-7.
Provided herein are methods of inducing psychosis in animals using light-responsive opsins and methods of identifying or screening compounds that may be useful in treating psychosis.1. A non-human animal comprising a light-responsive opsin expressed on the cell membrane of a subset of layer V pyramidal neurons in the prefrontal cortex, wherein light activation of the opsin induces depolarization of the membrane and induces psychosis of the animal. 2. The animal of claim 1, wherein the subset of layer V pyramidal neurons have a single large apical dendrite. 3. The animal of claim 1, wherein the opsin is selected from the group consisting of ChR2, VChR1, and DChR. 4. (canceled) 5. A prefrontal cortex tissue slice comprising a subset of layer V pyramidal neurons, wherein a light-responsive opsin is expressed on the cell membrane of the apical dendrites in layer V pyramidal neurons, and wherein light activation of the opsin induces depolarization of the membrane. 6. The prefrontal cortex tissue slice of claim 5, wherein the subset of layer V pyramidal neurons have a single large apical dendrite. 7. The prefrontal cortex tissue slice of claim 5, wherein the opsin is selected from the group consisting of ChR2, VChR1, and DChR. 8. The prefrontal cortex tissue of claim 5, wherein the opsin is selected from the group consisting of SFO, SSFO, C1V1, C1V1-E122T, C1V1-E162T, and C1V1-E122T/E162T. 9. A method of inducing psychosis in a non-human animal, comprising activating a light-responsive opsin by light, wherein the light-responsive opsin is expressed on the cell membrane of a subset of layer V pyramidal neurons in the prefrontal cortex in the animal, and wherein the light activation of the opsin induces depolarization of the cell membrane. 10. The method of claim 9, wherein the subset of layer V pyramidal neurons have a single large apical dendrite. 11. The method of claim 9, wherein the opsin is selected from the group consisting of ChR2, VChR1, and DChR. 12. The method of claim 9, wherein the opsin is selected from the group consisting of SFO, SSFO, C1V1, C1V1-E122T, C1V1-E162T, and C1V1-E122T/E162T. 13. A method of identifying a candidate compound for treating psychosis, the method comprising measuring a psychotic state of a non-human animal before and after administering the compound to the prefrontal cortex of the animal, wherein the psychotic state is induced by light activation of a light-responsive opsin expressed on the cell membrane of a layer of V pyramidal neurons in the animal, and activation of the opsin induces depolarization of the membrane; wherein an improvement in one or more of psychotic state measurements after the administration of the compound indicates that the compound is a candidate for treating psychosis. 14. The method of claim 13, wherein the psychotic state measurement is a behavioral measurement. 15. The method of claim 13, wherein the psychotic state measurement is a cellular measurement. 16. The method of claim 9, further comprising a step of administering a D2 agonist to the animal before administration of the compound. 17. A method of identifying a candidate compound for treating psychosis, the method comprising: measuring a psychotic state of a prefrontal cortex tissue slice before and after incubating the tissue slice with the compound, wherein the prefrontal cortex tissue slice comprises a layer of V pyramidal neurons and a light-responsive opsin is expressed on the cell membrane of the layer of V pyramidal neurons, wherein the psychotic state is induced by the membrane depolarization of the neurons induced by activation of the light-responsive opsin; wherein an improvement in one or more of a psychotic state readouts after incubation with the compound indicates that the compound is a candidate for treating psychosis. 18. The method of claim 17, wherein the psychotic state measurement is a cellular measurement. 19. The method of claim 17, further comprising a step of incubating a D2 agonist with the prefrontal cortex tissue slice before incubation with the compound. 20. A method comprising: providing optical stimulation to a target neuron population that expresses a light-responsive opsin; measuring a first electrical pattern of the target neuron population in response to the optical stimulation; introducing a drug, known to induce psychosis, to the target neuron population; providing optical stimulation to the target neuron population measuring a subsequent electrical pattern of the target neuron population in response to the optical stimulation; and comparing the first electrical pattern and the subsequent electrical pattern. 21. The method of claim 20, further including identifying a subset of neurons associated with psychosis. 22. The method of claim 21, wherein the subset of neurons is a subset of level 5 pyramidal neurons. 23. The method of claim 21, wherein the target neuron cell population is in a patient. 24. The method of claim 23, further including providing a potential treatment to the patient and observing a third electrical pattern in response to light during and after the potential treatment. 25. The method of claim 24, further including comparing the third electrical pattern to the first and second electrical patterns and assessing the efficacy of the potential treatment. 26. The method of claim 20, further comprising elevating activity within a Thy1-expressing subset of prefrontal cortical neurons. 27-32. (canceled) 33. A method comprising: modifying a target neuron population with a light-responsive molecule, the neurons of the target neuron population having a single, large apical dendrite; providing light to the target neuron population, the light activating the light-responsive molecule; and introducing a drug to the target neuron population; the drug causing the membrane potential of the neurons to remain elevated after removal of the light. 34. The method of claim 33, wherein the light-responsive molecule excites the target neuron population in response to light. 35. The method of claim 34, wherein the light-responsive molecule is ChR2. 36. The method of claim 33, wherein the target neuron population is a subset of layer V pyramidal neurons. 37. The method of claim 33, wherein the elevation of the membrane potential inhibits firing of the cell in response to a stimulus. 38. The method of claim 33, wherein the elevation of the membrane potential results in firing after a depolarizing current is removed. 39. The method of claim 33, wherein the apical dendrite extends into superficial layers of the brain. 40. The method of claim 33, further including determining the source of the elevated membrane potential. 41. The method of claim 33, wherein the drug induces psychosis. 42. The method of claim 33, wherein L-type calcium channels of the target neuron population are involved in an activity-dependent depolarization. 43-49. (canceled) 50. The non-human animal of claim 1, wherein the opsin comprises an amino acid sequence having at least 90% amino acid sequence identity to the amino acid sequence set forth in one of SEQ ID NOs:1-7.
1,600
295
295
15,531,254
1,633
The invention encompasses methods for generating stable exosome formulations and encompasses stable exosome formulations. The exosome formulations encompass stable liquid exosome formulations and stable lyophilized exosome formulations. In some embodiments, the exosome formulations can be generated by ultrafiltration and diafiltration. The exosome formulations can be suitable for administration to a human.
1. A stable lyophilized exosome formulation suitable for administration to a human comprising at least 106 exosomes, wherein the wherein the lyophilized exosomes maintain at least 90% of the levels of miR-210 RNA and miR-146A of the exosome formulation pre-lyophilization. 2. The stable lyophilized exosome formulation of claim 1, wherein the exosome formulation comprises at least 108 exosomes. 3. The stable lyophilized exosome formulation of claim 1, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs). 4. The stable lyophilized exosome formulation of claim 3, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs) by ultrafiltration and diafiltration. 5. A stable lyophilized exosome formulation suitable for administration to a human comprising at least 106 exosomes, wherein the lyophilized exosomes maintain at least 90% of the level of in vitro biological activity of the exosome preparation pre-lyophilization. 6. The stable lyophilized exosome formulation of claim 5, wherein the exosome formulation comprises at least 108 exosomes. 7. The stable lyophilized exosome formulation of claim 5, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs). 8. The stable lyophilized exosome formulation of claim 7, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs) by ultrafiltration and diafiltration. 9. The stable lyophilized exosome formulation of claim 1, wherein the lyophilized exosomes maintain at least 90% of the level of in vivo biological activity of the exosome preparation pre-lyophilization. 10. The stable lyophilized exosome formulation of claim 9, wherein the exosome formulation comprises at least 108 exosomes. 11. The stable lyophilized exosome formulation of claim 9, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs). 12. The stable lyophilized exosome formulation of claim 11, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs) by ultrafiltration and diafiltration. 13. A stable liquid cardiosphere-derived cell (CDC) exosome formulation suitable for administration to a human comprising at least 106 CDC exosomes, wherein the exosome formulation maintains at least 75% of the levels of miR-210 RNA and miR-146A of the starting exosome formulation for 30 days at 4° C. 14. The stable liquid CDC exosome formulation of claim 13, wherein the exosome formulation comprises at least 108 exosomes. 15. The stable liquid CDC exosome formulation of claim 13, wherein the exosome formulation is generated from CDCs by ultrafiltration and diafiltration.
The invention encompasses methods for generating stable exosome formulations and encompasses stable exosome formulations. The exosome formulations encompass stable liquid exosome formulations and stable lyophilized exosome formulations. In some embodiments, the exosome formulations can be generated by ultrafiltration and diafiltration. The exosome formulations can be suitable for administration to a human.1. A stable lyophilized exosome formulation suitable for administration to a human comprising at least 106 exosomes, wherein the wherein the lyophilized exosomes maintain at least 90% of the levels of miR-210 RNA and miR-146A of the exosome formulation pre-lyophilization. 2. The stable lyophilized exosome formulation of claim 1, wherein the exosome formulation comprises at least 108 exosomes. 3. The stable lyophilized exosome formulation of claim 1, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs). 4. The stable lyophilized exosome formulation of claim 3, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs) by ultrafiltration and diafiltration. 5. A stable lyophilized exosome formulation suitable for administration to a human comprising at least 106 exosomes, wherein the lyophilized exosomes maintain at least 90% of the level of in vitro biological activity of the exosome preparation pre-lyophilization. 6. The stable lyophilized exosome formulation of claim 5, wherein the exosome formulation comprises at least 108 exosomes. 7. The stable lyophilized exosome formulation of claim 5, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs). 8. The stable lyophilized exosome formulation of claim 7, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs) by ultrafiltration and diafiltration. 9. The stable lyophilized exosome formulation of claim 1, wherein the lyophilized exosomes maintain at least 90% of the level of in vivo biological activity of the exosome preparation pre-lyophilization. 10. The stable lyophilized exosome formulation of claim 9, wherein the exosome formulation comprises at least 108 exosomes. 11. The stable lyophilized exosome formulation of claim 9, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs). 12. The stable lyophilized exosome formulation of claim 11, wherein the exosome formulation is generated from cardiosphere-derived cells (CDCs) by ultrafiltration and diafiltration. 13. A stable liquid cardiosphere-derived cell (CDC) exosome formulation suitable for administration to a human comprising at least 106 CDC exosomes, wherein the exosome formulation maintains at least 75% of the levels of miR-210 RNA and miR-146A of the starting exosome formulation for 30 days at 4° C. 14. The stable liquid CDC exosome formulation of claim 13, wherein the exosome formulation comprises at least 108 exosomes. 15. The stable liquid CDC exosome formulation of claim 13, wherein the exosome formulation is generated from CDCs by ultrafiltration and diafiltration.
1,600
296
296
15,575,076
1,699
The invention discloses a process for the oxidation of limonene, comprising the reaction of limonene with hydrogen peroxide in the presence of a catalyst containing atoms and/or ions of at least one metal, selected from the group consisting of molybdenum, tungsten, scandium, vanadium, titanium, lanthanum, zirconium, praseodymium, neodymium, samarium, europium, terbium, dysprosium, erbium or ytterbium, characterised in that the molecular weight of the catalyst is less than 2,000 g/mol and that the reaction is performed at a pH value of more than 7.5.
1. A process for the oxidation of limonene, comprising the reaction of limonene with hydrogen peroxide in the presence of a catalyst containing atoms and/or ions of at least one metal selected from the group consisting of molybdenum, tungsten, scandium, vanadium, titanium, lanthanum, zirconium, praseodymium, neodymium, samarium, europium, terbium, dysprosium, erbium or ytterbium and mixtures thereof, wherein the molecular weight of the catalyst is less than 2,000 g/mol and the reaction is performed at a pH value of more than 7.5. 2. The process of claim 1, wherein the catalyst contains atoms and/or ions of at least one metal selected from the group consisting of molybdenum, tungsten, scandium, vanadium, titanium and lanthanum and mixtures thereof. 3. The process of claim 1, wherein the catalyst is selected from the group consisting of sodium molybdate, sodium molybdate dihydrate, sodium tungstate, sodium tungstate dihydrate and lanthanum nitrate and mixtures thereof. 4. The process of claim 1, wherein the reaction is performed in at least one organic solvent. 5. The process of claim 4, wherein the solvent is selected from the group consisting of C1-C8 alcohols and amides and mixtures thereof. 6. The process of claim 1, wherein the pH value is more than 8. 7. The process of claim 1, wherein the temperature is from 25 to 90° C. 8. The process of claim 1, wherein the pH value is more than 8, and the temperature is in the range from 40° C. to 90° C. 9. The process of claim 1, wherein the pH value is more than 9, and the temperature is in the range from 40° C. to 90° C. 10. The process of claim 1, wherein the amount of catalyst used is 1 to 50 mole percent, based on limonene. 11. The process of claim 1, wherein 2-10 molar equivalents of hydrogen peroxide per 1 mole limonene are used. 12. A process for the production of hydroxy derivatives of limonene, comprising the following steps: (a) oxidation of limonene according to claim 1, and (b) reaction of the mixture obtained in step (a) with a reducing agent. 13. The process of claim 12, performed at a temperature of 25 to 90° C. at a pH value of more than 7.5. 14. The process of claim 12, additionally comprising separating the reaction product. 15. A process for the production of an aroma composition, comprising adding at least one peroxide derivative of limonene prepared according to claim 1. 16. The process of claim 12, wherein the metal is selected from the group consisting of molybdenum and tungsten and mixtures thereof. 17. The process according to claim 14, wherein the reaction product is separated by distillation. 18. The process according to claim 15, wherein two or more peroxide derivatives of limonene are added.
The invention discloses a process for the oxidation of limonene, comprising the reaction of limonene with hydrogen peroxide in the presence of a catalyst containing atoms and/or ions of at least one metal, selected from the group consisting of molybdenum, tungsten, scandium, vanadium, titanium, lanthanum, zirconium, praseodymium, neodymium, samarium, europium, terbium, dysprosium, erbium or ytterbium, characterised in that the molecular weight of the catalyst is less than 2,000 g/mol and that the reaction is performed at a pH value of more than 7.5.1. A process for the oxidation of limonene, comprising the reaction of limonene with hydrogen peroxide in the presence of a catalyst containing atoms and/or ions of at least one metal selected from the group consisting of molybdenum, tungsten, scandium, vanadium, titanium, lanthanum, zirconium, praseodymium, neodymium, samarium, europium, terbium, dysprosium, erbium or ytterbium and mixtures thereof, wherein the molecular weight of the catalyst is less than 2,000 g/mol and the reaction is performed at a pH value of more than 7.5. 2. The process of claim 1, wherein the catalyst contains atoms and/or ions of at least one metal selected from the group consisting of molybdenum, tungsten, scandium, vanadium, titanium and lanthanum and mixtures thereof. 3. The process of claim 1, wherein the catalyst is selected from the group consisting of sodium molybdate, sodium molybdate dihydrate, sodium tungstate, sodium tungstate dihydrate and lanthanum nitrate and mixtures thereof. 4. The process of claim 1, wherein the reaction is performed in at least one organic solvent. 5. The process of claim 4, wherein the solvent is selected from the group consisting of C1-C8 alcohols and amides and mixtures thereof. 6. The process of claim 1, wherein the pH value is more than 8. 7. The process of claim 1, wherein the temperature is from 25 to 90° C. 8. The process of claim 1, wherein the pH value is more than 8, and the temperature is in the range from 40° C. to 90° C. 9. The process of claim 1, wherein the pH value is more than 9, and the temperature is in the range from 40° C. to 90° C. 10. The process of claim 1, wherein the amount of catalyst used is 1 to 50 mole percent, based on limonene. 11. The process of claim 1, wherein 2-10 molar equivalents of hydrogen peroxide per 1 mole limonene are used. 12. A process for the production of hydroxy derivatives of limonene, comprising the following steps: (a) oxidation of limonene according to claim 1, and (b) reaction of the mixture obtained in step (a) with a reducing agent. 13. The process of claim 12, performed at a temperature of 25 to 90° C. at a pH value of more than 7.5. 14. The process of claim 12, additionally comprising separating the reaction product. 15. A process for the production of an aroma composition, comprising adding at least one peroxide derivative of limonene prepared according to claim 1. 16. The process of claim 12, wherein the metal is selected from the group consisting of molybdenum and tungsten and mixtures thereof. 17. The process according to claim 14, wherein the reaction product is separated by distillation. 18. The process according to claim 15, wherein two or more peroxide derivatives of limonene are added.
1,600
297
297
14,881,714
1,611
This invention relates to an improved crystallization process of hydrogenated castor oil structurant which results in (i) a dispersion having a high concentration of the structurant, and (ii) a crystal habit/form that results in a higher yield stress in the final product, imparting high stability, for a given amount structurant. In addition, the resulting personal care product is consumer desired, as less structurant residue is observed on hair after the use of the personal care product.
1. A personal care composition comprising a. from about 5% to about 25 wt % of an detersive surfactant; b. from about 1% to about 20% wt % of hydrogenated castor oil premix composition by weight of the personal care composition wherein the hydrogenated castor oil premix composition comprises: i. from about 0.30% to about 4% of hydrogenated castor oil by weight of the premix composition; ii. from about 15% to about 40% of one or more surfactants, by weight of the premix composition; iii. from about 60% to about 80% a aqueous carrier by weight of the premix composition; c. from about 80% to about 95% of an aqueous carrier by weight of the personal care composition; wherein the personal care composition comprises from about 0.03% to about 1% of hydrogenated castor oil by weight of the personal care composition, and wherein the personal care composition comprises crystals and wherein from about 80 weight % to about 100 weight % of the crystals have a fiber shape, and wherein from about 80% to about 100% of the fiber shaped crystals are longer than about 5 micrometers. 2. The personal care composition of claim 1, wherein the detersive surfactant is an anionic surfactant selected from the group consisting of SLE1S and SLS. 3. The personal care composition of claim 1, further comprising a gel network. 4. The personal care composition of claim 1, further comprising a cationic polymer. 5. The personal care composition of claim 1, further comprising a conditioning agent. 6. A method of making a personal care composition comprising the steps of: a. preparing a premix composition by 1. mixing i. from about 0.30% to about 4% of hydrogenated castor oil by weight of the premix composition, wherein the hydrogenated castor oil comprises of particles of which 90% by weight of the total hydrogenated castor oil consists of particle with size of less than 60 micrometers; ii. from about 15% to about 40% of one or more surfactants, by weight of the premix composition; iii. from about 60% to about 80% of an aqueous carrier by weight of the premix composition; 2. adjusting the pH of the premix composition to about 5 to about 12; 3. heating the premix composition to 65-84° C.; 4. cooling the premix composition to a temperature of about 60° C. to about 20° C. b. forming a personal care composition by combining the pre-mix with 1. from about 5% to about 25% of detersive surfactant by weight of the personal care composition; 2. from about 80% to about 95% of an aqueous carrier by weight of the personal care composition; wherein the personal care composition comprises from about 0.03% to about 1% of hydrogenated castor oil by weight of the personal care composition, and wherein the personal care composition comprises crystals and wherein from about 80 weight % to about 100 weight % of the resulting crystals have a fiber shape, and wherein from about 80% to about 100% of the fiber shaped crystals are longer than about 5 micrometers. 6. A method according to claim 6, wherein the personal care composition comprises from about 0.05% to about 0.5% of hydrogenated castor oil by weight of the personal care composition. 7. A method according to claim 7, wherein the personal care composition comprises from about 0.05% to about 0.15% of hydrogenated castor oil by weight of the personal care composition. 8. The personal care composition of claim 6, wherein from about 80% to about 100% of the fiber shaped crystals have a length of from about 5 micrometers to about 80 micrometers. 9. A method according to claim 9, wherein from about 80% to about 100% of the fiber shaped crystals have a length of from about 10 micrometers to about 50 micrometers. 10. A method according to claim 10, wherein from about 80% to about 100% of the fiber shaped crystals have a length of from about 20 micrometers to about 40 micrometers. 11. A method according to claim 6, wherein the cooling of the mixture to a temperature of 50° C. to about 20° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 10° C. per minute to about 1° C. per minute. 12. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 45° C. to about 25° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 10° C. per minute to about 1° C. per minute. 13. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 40° C. to about 30° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 10° C. per minute to about 1° C. per minute. 14. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 50° C. to about 20° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 5° C. per minute to about 1° C. per minute. 15. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 50° C. to about 20° C. is performed by passing the mixture through a cooling device and collecting it in a different vessel. 16. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 50° C. to about 20° C. is performed by passing the mixture through a cooling device and recirculating it back into the same vessel until the desired vessel temperature is achieved. 17. The method of claim 6, wherein the personal care composition further comprises from about 0.1 to about 0.25 by weight of the personal care composition, of a cationic guar polymer. 18. The method of claim 6, wherein the personal care composition further comprises from about from about 0.1 to about 1.5 by weight, of a conditioning agent. 19. The method of claim 6, wherein the personal care composition comprises from about 20% to about 35% by weight of the premix composition of a detersive surfactant, wherein the detersive surfactant is an anionic surfactant. 20. The method of claim 20, wherein the anionic surfactant SLE1S. 21. The method of claim 20, wherein the anionic surfactant SLS. 22. The method of claim 6, wherein the step of preparing of the premix composition further comprises the formation of the crystals. 24. A personal care composition comprising a. from about 5% to about 25% by weight of the personal are composition of a detersive surfactant; b. from about 0.30% to about 4% of hydrogenated castor oil by weight of the personal care composition; c. an aqueous carrier; wherein the personal care composition comprises crystals and wherein from about 80 weight % to about 100 weight % of the crystals have a fiber shape, and wherein from about 80% to about 100% of the fiber shaped crystals are longer than about 10 micrometers. 25. The personal care composition of claim 24, comprising from about 0.03% to about 1% of hydrogenated castor oil by weight of the personal care composition
This invention relates to an improved crystallization process of hydrogenated castor oil structurant which results in (i) a dispersion having a high concentration of the structurant, and (ii) a crystal habit/form that results in a higher yield stress in the final product, imparting high stability, for a given amount structurant. In addition, the resulting personal care product is consumer desired, as less structurant residue is observed on hair after the use of the personal care product.1. A personal care composition comprising a. from about 5% to about 25 wt % of an detersive surfactant; b. from about 1% to about 20% wt % of hydrogenated castor oil premix composition by weight of the personal care composition wherein the hydrogenated castor oil premix composition comprises: i. from about 0.30% to about 4% of hydrogenated castor oil by weight of the premix composition; ii. from about 15% to about 40% of one or more surfactants, by weight of the premix composition; iii. from about 60% to about 80% a aqueous carrier by weight of the premix composition; c. from about 80% to about 95% of an aqueous carrier by weight of the personal care composition; wherein the personal care composition comprises from about 0.03% to about 1% of hydrogenated castor oil by weight of the personal care composition, and wherein the personal care composition comprises crystals and wherein from about 80 weight % to about 100 weight % of the crystals have a fiber shape, and wherein from about 80% to about 100% of the fiber shaped crystals are longer than about 5 micrometers. 2. The personal care composition of claim 1, wherein the detersive surfactant is an anionic surfactant selected from the group consisting of SLE1S and SLS. 3. The personal care composition of claim 1, further comprising a gel network. 4. The personal care composition of claim 1, further comprising a cationic polymer. 5. The personal care composition of claim 1, further comprising a conditioning agent. 6. A method of making a personal care composition comprising the steps of: a. preparing a premix composition by 1. mixing i. from about 0.30% to about 4% of hydrogenated castor oil by weight of the premix composition, wherein the hydrogenated castor oil comprises of particles of which 90% by weight of the total hydrogenated castor oil consists of particle with size of less than 60 micrometers; ii. from about 15% to about 40% of one or more surfactants, by weight of the premix composition; iii. from about 60% to about 80% of an aqueous carrier by weight of the premix composition; 2. adjusting the pH of the premix composition to about 5 to about 12; 3. heating the premix composition to 65-84° C.; 4. cooling the premix composition to a temperature of about 60° C. to about 20° C. b. forming a personal care composition by combining the pre-mix with 1. from about 5% to about 25% of detersive surfactant by weight of the personal care composition; 2. from about 80% to about 95% of an aqueous carrier by weight of the personal care composition; wherein the personal care composition comprises from about 0.03% to about 1% of hydrogenated castor oil by weight of the personal care composition, and wherein the personal care composition comprises crystals and wherein from about 80 weight % to about 100 weight % of the resulting crystals have a fiber shape, and wherein from about 80% to about 100% of the fiber shaped crystals are longer than about 5 micrometers. 6. A method according to claim 6, wherein the personal care composition comprises from about 0.05% to about 0.5% of hydrogenated castor oil by weight of the personal care composition. 7. A method according to claim 7, wherein the personal care composition comprises from about 0.05% to about 0.15% of hydrogenated castor oil by weight of the personal care composition. 8. The personal care composition of claim 6, wherein from about 80% to about 100% of the fiber shaped crystals have a length of from about 5 micrometers to about 80 micrometers. 9. A method according to claim 9, wherein from about 80% to about 100% of the fiber shaped crystals have a length of from about 10 micrometers to about 50 micrometers. 10. A method according to claim 10, wherein from about 80% to about 100% of the fiber shaped crystals have a length of from about 20 micrometers to about 40 micrometers. 11. A method according to claim 6, wherein the cooling of the mixture to a temperature of 50° C. to about 20° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 10° C. per minute to about 1° C. per minute. 12. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 45° C. to about 25° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 10° C. per minute to about 1° C. per minute. 13. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 40° C. to about 30° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 10° C. per minute to about 1° C. per minute. 14. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 50° C. to about 20° C. is performed in a vessel by cooling the walls of the vessel while mixing and wherein the rate of cooling is from about 5° C. per minute to about 1° C. per minute. 15. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 50° C. to about 20° C. is performed by passing the mixture through a cooling device and collecting it in a different vessel. 16. A method according to claim 6, wherein the cooling of the mixture to a temperature of about 50° C. to about 20° C. is performed by passing the mixture through a cooling device and recirculating it back into the same vessel until the desired vessel temperature is achieved. 17. The method of claim 6, wherein the personal care composition further comprises from about 0.1 to about 0.25 by weight of the personal care composition, of a cationic guar polymer. 18. The method of claim 6, wherein the personal care composition further comprises from about from about 0.1 to about 1.5 by weight, of a conditioning agent. 19. The method of claim 6, wherein the personal care composition comprises from about 20% to about 35% by weight of the premix composition of a detersive surfactant, wherein the detersive surfactant is an anionic surfactant. 20. The method of claim 20, wherein the anionic surfactant SLE1S. 21. The method of claim 20, wherein the anionic surfactant SLS. 22. The method of claim 6, wherein the step of preparing of the premix composition further comprises the formation of the crystals. 24. A personal care composition comprising a. from about 5% to about 25% by weight of the personal are composition of a detersive surfactant; b. from about 0.30% to about 4% of hydrogenated castor oil by weight of the personal care composition; c. an aqueous carrier; wherein the personal care composition comprises crystals and wherein from about 80 weight % to about 100 weight % of the crystals have a fiber shape, and wherein from about 80% to about 100% of the fiber shaped crystals are longer than about 10 micrometers. 25. The personal care composition of claim 24, comprising from about 0.03% to about 1% of hydrogenated castor oil by weight of the personal care composition
1,600
298
298
14,681,301
1,631
Some embodiments are directed to an apparatus for monitoring patient orientation. A reusable sensor is configured to detect inclination angles of an anterior region of the patient. An elongated disposable attachment device is configured for removable attachment to both the sensor and the patient's anterior region such that the direction of elongation of the attachment device extends along the patient's sternum. The attachment device includes a sensor attachment portion configured for removable attachment to the sensor such that the sensor is prevented from direct contact with the patient, and a patient attachment portion configured for removable attachment to the patient's anterior region.
1. An apparatus for monitoring one or more conditions of a patient, comprising: a reusable sensor configured to sense the one or more conditions of the patient; and a disposable attachment device configured for removable attachment to both the sensor and the patient's anterior region, the attachment device including: a pouch configured for removable attachment to the sensor such that the sensor is prevented from direct contact with the patient, the pouch being configured to dispose the sensor therein to fully encapsulate the sensor, the pouch comprising a sealable opening to facilitate insertion of the sensor into the pouch, a patient attachment portion configured for removable attachment to the patient's anterior region, and a first connector attached to the pouch and a second connector attached to the patient attachment portion, the first connector and the second connector being configured to detachably mate to each other. 2. The apparatus for monitoring one or more conditions of a patient according to claim 1, further comprising a wireless transmitter configured to be in communication with the sensor to receive measurements from the sensor and to transmit the measurements wirelessly to an external device. 3. The apparatus for monitoring one or more conditions of a patient according to claim 2, wherein the pouch is configured to fully encapsulate the wireless transmitter. 4. The apparatus for monitoring one or more conditions of a patient according to claim 3, further comprising a sensor strip, wherein the sensor and wireless transmitter are disposed on or in the sensor strip. 5. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein a first end of the pouch comprises a pivoting arm to open the pouch to facilitate insertion of the sensor into the pouch. 6. The apparatus for monitoring one or more conditions of a patient according to claim 1, further comprising a battery receiver disposed in the pouch and configured to house a battery to provide power to the reusable sensor. 7. The apparatus for monitoring one or more conditions of a patient according to claim 6, further comprising a sensor strip with which the sensor is provided, wherein the sensor strip is configured to detachably mate with the battery receiver to allow the battery receiver to transmit power to the sensor. 8. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein the patient attachment portion comprises two separate, physically distinct and non-integral attachment portions that are each individually removably attachable to the pouch. 9. The apparatus for monitoring one or more conditions of a patient according to claim 1, further comprising a controller that is spaced apart from and physically unconnected with the sensor, wherein the transmitter wirelessly transmits signals between the sensor and the controller. 10. The apparatus for monitoring one or more conditions of a patient according to claim 9, wherein the controller controls the sensor to vary the frequency of measurements. 11. The apparatus for monitoring one or more conditions of a patient according to claim 9, wherein the controller provides an alert in response to signals received from the sensor. 12. The apparatus for monitoring one or more conditions of a patient according to claim 11, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle falls below a certain angle. 13. The apparatus for monitoring one or more conditions of a patient according to claim 12, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle falls below 30 degrees. 14. The apparatus for monitoring one or more conditions of a patient according to claim 11, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle indicates that the patient is attempting to stand. 15. The apparatus for monitoring one or more conditions of a patient according to claim 11, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle exceeds a certain angle. 16. The apparatus for monitoring one or more conditions of a patient according to claim 9, wherein the controller is configured to monitor the one or more conditions of the patient, including at least two of heart rate, respiratory rate, respiratory depth, GI motility, sleep analysis, and seizure detection. 17. The apparatus for monitoring one or more conditions of a patient according to claim 16, wherein the controller is configured to control an aspect of sensing of at least one of the monitored conditions based on at least one of another of the monitored conditions. 18. The apparatus for monitoring one or more conditions of a patient according to claim 17, wherein the controller is configured to affect the frequency of monitoring of patient incline based on data of at least one of the other monitored conditions. 19. The apparatus for monitoring one or more conditions of a patient according to claim 18, wherein the controller is configured to affect the frequency of monitoring of patient incline based on data of at least one of heart rate, respiratory rate, and respiratory depth that indicates that the patient is asleep. 20. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein the patient attachment portion is configured for removable attachment to skin of the patient at a sternum of the patient. 21. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein the pouch is formed of a deformable material. 22. An apparatus for monitoring conditions of a patient, comprising: a reusable sensor configured to sense plural conditions of the patient to monitor at least two of heart rate, respiratory rate, respiratory depth, GI motility, sleep analysis, and seizure detection; and a disposable attachment device configured for removable attachment to both the sensor and the patient's anterior region, the attachment device including: a pouch configured for removable attachment to the sensor such that the sensor is prevented from direct contact with the patient, the pouch being configured to dispose the sensor therein to fully encapsulate the sensor, the pouch comprising a sealable opening to facilitate insertion of the sensor into the pouch, and a patient attachment portion attached to the pouch and configured for removable attachment to the patient's anterior region. 23. The apparatus for monitoring one or more conditions of a patient according to claim 22, further comprising a wireless transmitter configured to be in communication with the sensor to receive measurements from the sensor and to transmit the measurements wirelessly to an external device. 24. The apparatus for monitoring one or more conditions of a patient according to claim 23, further comprising a controller that is spaced apart from and physically unconnected with the sensor and configured to wirelessly receive sensor measurements from the transmitter. 25. The apparatus for monitoring one or more conditions of a patient according to claim 24, wherein the controller is configured to allow a user to select which of the plural conditions of the patient to monitor. 26. The apparatus for monitoring one or more conditions of a patient according to claim 25, further comprising a display configured to indicate the patient conditions that are being monitored. 27. The apparatus for monitoring one or more conditions of a patient according to claim 24, wherein the controller is configured to generate an alert in response to the received sensor measurements. 28. The apparatus for monitoring one or more conditions of a patient according to claim 27, wherein the controller is configured to generate an alert in response to the received sensor measurements and a comparison with information obtained from a user input. 29. The apparatus for monitoring one or more conditions of a patient according to claim 28, wherein the user input comprises an incline angle. 30. The apparatus for monitoring one or more conditions of a patient according to claim 28, wherein the user input comprises a heart rate limit.
Some embodiments are directed to an apparatus for monitoring patient orientation. A reusable sensor is configured to detect inclination angles of an anterior region of the patient. An elongated disposable attachment device is configured for removable attachment to both the sensor and the patient's anterior region such that the direction of elongation of the attachment device extends along the patient's sternum. The attachment device includes a sensor attachment portion configured for removable attachment to the sensor such that the sensor is prevented from direct contact with the patient, and a patient attachment portion configured for removable attachment to the patient's anterior region.1. An apparatus for monitoring one or more conditions of a patient, comprising: a reusable sensor configured to sense the one or more conditions of the patient; and a disposable attachment device configured for removable attachment to both the sensor and the patient's anterior region, the attachment device including: a pouch configured for removable attachment to the sensor such that the sensor is prevented from direct contact with the patient, the pouch being configured to dispose the sensor therein to fully encapsulate the sensor, the pouch comprising a sealable opening to facilitate insertion of the sensor into the pouch, a patient attachment portion configured for removable attachment to the patient's anterior region, and a first connector attached to the pouch and a second connector attached to the patient attachment portion, the first connector and the second connector being configured to detachably mate to each other. 2. The apparatus for monitoring one or more conditions of a patient according to claim 1, further comprising a wireless transmitter configured to be in communication with the sensor to receive measurements from the sensor and to transmit the measurements wirelessly to an external device. 3. The apparatus for monitoring one or more conditions of a patient according to claim 2, wherein the pouch is configured to fully encapsulate the wireless transmitter. 4. The apparatus for monitoring one or more conditions of a patient according to claim 3, further comprising a sensor strip, wherein the sensor and wireless transmitter are disposed on or in the sensor strip. 5. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein a first end of the pouch comprises a pivoting arm to open the pouch to facilitate insertion of the sensor into the pouch. 6. The apparatus for monitoring one or more conditions of a patient according to claim 1, further comprising a battery receiver disposed in the pouch and configured to house a battery to provide power to the reusable sensor. 7. The apparatus for monitoring one or more conditions of a patient according to claim 6, further comprising a sensor strip with which the sensor is provided, wherein the sensor strip is configured to detachably mate with the battery receiver to allow the battery receiver to transmit power to the sensor. 8. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein the patient attachment portion comprises two separate, physically distinct and non-integral attachment portions that are each individually removably attachable to the pouch. 9. The apparatus for monitoring one or more conditions of a patient according to claim 1, further comprising a controller that is spaced apart from and physically unconnected with the sensor, wherein the transmitter wirelessly transmits signals between the sensor and the controller. 10. The apparatus for monitoring one or more conditions of a patient according to claim 9, wherein the controller controls the sensor to vary the frequency of measurements. 11. The apparatus for monitoring one or more conditions of a patient according to claim 9, wherein the controller provides an alert in response to signals received from the sensor. 12. The apparatus for monitoring one or more conditions of a patient according to claim 11, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle falls below a certain angle. 13. The apparatus for monitoring one or more conditions of a patient according to claim 12, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle falls below 30 degrees. 14. The apparatus for monitoring one or more conditions of a patient according to claim 11, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle indicates that the patient is attempting to stand. 15. The apparatus for monitoring one or more conditions of a patient according to claim 11, wherein the controller provides an alarm indication if the sensor detects that the patient's incline angle exceeds a certain angle. 16. The apparatus for monitoring one or more conditions of a patient according to claim 9, wherein the controller is configured to monitor the one or more conditions of the patient, including at least two of heart rate, respiratory rate, respiratory depth, GI motility, sleep analysis, and seizure detection. 17. The apparatus for monitoring one or more conditions of a patient according to claim 16, wherein the controller is configured to control an aspect of sensing of at least one of the monitored conditions based on at least one of another of the monitored conditions. 18. The apparatus for monitoring one or more conditions of a patient according to claim 17, wherein the controller is configured to affect the frequency of monitoring of patient incline based on data of at least one of the other monitored conditions. 19. The apparatus for monitoring one or more conditions of a patient according to claim 18, wherein the controller is configured to affect the frequency of monitoring of patient incline based on data of at least one of heart rate, respiratory rate, and respiratory depth that indicates that the patient is asleep. 20. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein the patient attachment portion is configured for removable attachment to skin of the patient at a sternum of the patient. 21. The apparatus for monitoring one or more conditions of a patient according to claim 1, wherein the pouch is formed of a deformable material. 22. An apparatus for monitoring conditions of a patient, comprising: a reusable sensor configured to sense plural conditions of the patient to monitor at least two of heart rate, respiratory rate, respiratory depth, GI motility, sleep analysis, and seizure detection; and a disposable attachment device configured for removable attachment to both the sensor and the patient's anterior region, the attachment device including: a pouch configured for removable attachment to the sensor such that the sensor is prevented from direct contact with the patient, the pouch being configured to dispose the sensor therein to fully encapsulate the sensor, the pouch comprising a sealable opening to facilitate insertion of the sensor into the pouch, and a patient attachment portion attached to the pouch and configured for removable attachment to the patient's anterior region. 23. The apparatus for monitoring one or more conditions of a patient according to claim 22, further comprising a wireless transmitter configured to be in communication with the sensor to receive measurements from the sensor and to transmit the measurements wirelessly to an external device. 24. The apparatus for monitoring one or more conditions of a patient according to claim 23, further comprising a controller that is spaced apart from and physically unconnected with the sensor and configured to wirelessly receive sensor measurements from the transmitter. 25. The apparatus for monitoring one or more conditions of a patient according to claim 24, wherein the controller is configured to allow a user to select which of the plural conditions of the patient to monitor. 26. The apparatus for monitoring one or more conditions of a patient according to claim 25, further comprising a display configured to indicate the patient conditions that are being monitored. 27. The apparatus for monitoring one or more conditions of a patient according to claim 24, wherein the controller is configured to generate an alert in response to the received sensor measurements. 28. The apparatus for monitoring one or more conditions of a patient according to claim 27, wherein the controller is configured to generate an alert in response to the received sensor measurements and a comparison with information obtained from a user input. 29. The apparatus for monitoring one or more conditions of a patient according to claim 28, wherein the user input comprises an incline angle. 30. The apparatus for monitoring one or more conditions of a patient according to claim 28, wherein the user input comprises a heart rate limit.
1,600
299
299
14,898,284
1,633
Methods of making vaccinia viruses for gene-directed prodrug therapy are disclosed and their use in the treatment of disease is provided. In particular, the anti-tumor effects of vaccinia viruses that are modified to express a prodrug-activating enzyme are disclosed.
1.-21. (canceled) 22. A vaccinia virus which is capable of expressing a glutamate carboxypeptidase within a cell. 23. The vaccinia virus according to claim 22, wherein the glutamate carboxypeptidase is CPG2 or a mutant, variant or homologue thereof. 24. The vaccinia virus according to claim 23, wherein the glutamate carboxypeptidase comprises an amino acid sequence which has at least 80% identity to SEQ ID NO: 2. 25. The vaccinia virus according to claim 23, wherein the vaccinia virus comprises a CPG2 expression cassette insert in a vaccinia virus virulence gene. 26. The vaccinia virus according to claim 25, wherein the vaccinia virus virulence gene is vaccinia thymidine kinase (vTK). 27. A two component system for gene directed enzyme prodrug therapy which comprises: (a) a vaccinia virus according to claim 22; and (b) a prodrug which can be converted into an active drug by a glutamate carboxypeptidase. 28. The two component system according to claim 27, wherein the prodrug comprises; a glutamic acid group or a glutamic acid analogue group, wherein the glutamic acid group or the glutamic acid analogue is connected via N to a linker which is connected to a cytotoxic agent. 29. The two component system according to claim 28, wherein the linker comprises a functional group selected from: carbonyl, carbamate, urea, and equivalents. 30. The two component system according to claim 28, wherein the cytotoxic agent comprises a nitrogen mustard or a nitrogen mustard analogue. 31. The two component system according to claim 30, wherein the prodrug is N-(4-[bis(2-iodoethyl)amino]phenoxycarbonyl)-L-glutamic acid or a salt thereof. 32. A method of killing a neoplastic cell, the method comprising treating the neoplastic cell with: a vaccinia virus according to claim 22 and a prodrug which is capable of being converted into an active form by a glutamate carboxypeptidase. 33. The method of killing a neoplastic cell according to claim 32, wherein the prodrug comprises a glutamic acid group or a glutamic acid analogue group, wherein the glutamic acid group or the glutamic acid analogue is connected via N to a linker which is connected to a cytotoxic agent. 34. The method of killing a neoplastic cell according to claim 33, wherein the linker comprises a functional group selected from: carbonyl, carbamate, urea, and equivalents. 35. The method of killing a neoplastic cell according to claim 33, wherein the cytotoxic agent comprises a nitrogen mustard or a nitrogen mustard analogue. 36. The method of killing a neoplastic cell according to claim 35, wherein the prodrug is N-(4-[bis(2-iodoethyl)amino]phenoxycarbonyl)-L-glutamic acid or a salt thereof. 37. A method of treating a tumour in a patient in need of treatment, the method comprising administering to the patient: a vaccinia virus which is capable of expressing a glutamate carboxypeptidase in a cell; and, a prodrug which is capable of being converted into an active form by the glutamate carboxypeptidase. 38. The method of claim 37, wherein the vaccinia virus is genetically modified to have enhanced tumour selectivity. 39. The method of claim 37, wherein the vaccinia virus has one or more inactivated virulence genes. 40. The method of claim 39, wherein the one or more inactivated virulence genes include vaccinia thymidine kinase (vTK). 41. The method of claim 40, wherein the inactivated vTK gene comprises a CPG2 expression cassette insert. 42. The method of claim 40, wherein the inactivated vTK gene comprises a luciferase expression cassette insert. 43. The method of claim 39, wherein the one or more inactivated virulence genes include vaccinia growth factor (VGF). 44. The method of claim 37, wherein the glutamate carboxypeptidase is CPG2 or a mutant, variant or homologue thereof. 45. The method of claim 23, wherein the CPG2 comprises an amino acid sequence which has at least 80% identity to SEQ ID NO: 2. 46. The method of claim 37, wherein the tumor is a solid tumor. 47. The method of claim 37, wherein the tumor is selected from head and neck cancer, colorectal cancer, lung cancer, melanoma, or breast cancer, cervix cancer, CNS cancer, ovarian cancer, kidney cancer, leukemia, brain tumor, or prostate cancer.
Methods of making vaccinia viruses for gene-directed prodrug therapy are disclosed and their use in the treatment of disease is provided. In particular, the anti-tumor effects of vaccinia viruses that are modified to express a prodrug-activating enzyme are disclosed.1.-21. (canceled) 22. A vaccinia virus which is capable of expressing a glutamate carboxypeptidase within a cell. 23. The vaccinia virus according to claim 22, wherein the glutamate carboxypeptidase is CPG2 or a mutant, variant or homologue thereof. 24. The vaccinia virus according to claim 23, wherein the glutamate carboxypeptidase comprises an amino acid sequence which has at least 80% identity to SEQ ID NO: 2. 25. The vaccinia virus according to claim 23, wherein the vaccinia virus comprises a CPG2 expression cassette insert in a vaccinia virus virulence gene. 26. The vaccinia virus according to claim 25, wherein the vaccinia virus virulence gene is vaccinia thymidine kinase (vTK). 27. A two component system for gene directed enzyme prodrug therapy which comprises: (a) a vaccinia virus according to claim 22; and (b) a prodrug which can be converted into an active drug by a glutamate carboxypeptidase. 28. The two component system according to claim 27, wherein the prodrug comprises; a glutamic acid group or a glutamic acid analogue group, wherein the glutamic acid group or the glutamic acid analogue is connected via N to a linker which is connected to a cytotoxic agent. 29. The two component system according to claim 28, wherein the linker comprises a functional group selected from: carbonyl, carbamate, urea, and equivalents. 30. The two component system according to claim 28, wherein the cytotoxic agent comprises a nitrogen mustard or a nitrogen mustard analogue. 31. The two component system according to claim 30, wherein the prodrug is N-(4-[bis(2-iodoethyl)amino]phenoxycarbonyl)-L-glutamic acid or a salt thereof. 32. A method of killing a neoplastic cell, the method comprising treating the neoplastic cell with: a vaccinia virus according to claim 22 and a prodrug which is capable of being converted into an active form by a glutamate carboxypeptidase. 33. The method of killing a neoplastic cell according to claim 32, wherein the prodrug comprises a glutamic acid group or a glutamic acid analogue group, wherein the glutamic acid group or the glutamic acid analogue is connected via N to a linker which is connected to a cytotoxic agent. 34. The method of killing a neoplastic cell according to claim 33, wherein the linker comprises a functional group selected from: carbonyl, carbamate, urea, and equivalents. 35. The method of killing a neoplastic cell according to claim 33, wherein the cytotoxic agent comprises a nitrogen mustard or a nitrogen mustard analogue. 36. The method of killing a neoplastic cell according to claim 35, wherein the prodrug is N-(4-[bis(2-iodoethyl)amino]phenoxycarbonyl)-L-glutamic acid or a salt thereof. 37. A method of treating a tumour in a patient in need of treatment, the method comprising administering to the patient: a vaccinia virus which is capable of expressing a glutamate carboxypeptidase in a cell; and, a prodrug which is capable of being converted into an active form by the glutamate carboxypeptidase. 38. The method of claim 37, wherein the vaccinia virus is genetically modified to have enhanced tumour selectivity. 39. The method of claim 37, wherein the vaccinia virus has one or more inactivated virulence genes. 40. The method of claim 39, wherein the one or more inactivated virulence genes include vaccinia thymidine kinase (vTK). 41. The method of claim 40, wherein the inactivated vTK gene comprises a CPG2 expression cassette insert. 42. The method of claim 40, wherein the inactivated vTK gene comprises a luciferase expression cassette insert. 43. The method of claim 39, wherein the one or more inactivated virulence genes include vaccinia growth factor (VGF). 44. The method of claim 37, wherein the glutamate carboxypeptidase is CPG2 or a mutant, variant or homologue thereof. 45. The method of claim 23, wherein the CPG2 comprises an amino acid sequence which has at least 80% identity to SEQ ID NO: 2. 46. The method of claim 37, wherein the tumor is a solid tumor. 47. The method of claim 37, wherein the tumor is selected from head and neck cancer, colorectal cancer, lung cancer, melanoma, or breast cancer, cervix cancer, CNS cancer, ovarian cancer, kidney cancer, leukemia, brain tumor, or prostate cancer.
1,600