Unnamed: 0
int64
0
274k
ApplicationNumber
int64
9.75M
96.1M
ArtUnit
int64
1.6k
3.99k
Abstract
stringlengths
7
8.37k
Claims
stringlengths
3
292k
abstract-claims
stringlengths
75
293k
TechCenter
int64
1.6k
3.9k
300
14,463,687
1,662
Compositions and methods are provided for genome modification of a target sequence in the genome of a plant or plant cell. The methods and compositions employ a guide RNA/Cas endonuclease system to provide an effective system for modifying or altering target sites within the genome of a plant, plant cell or seed. Also provided are compositions and methods employing a guide polynucleotide/Cas endonuclease system for genome modification of a nucleotide sequence in the genome of a cell or organism, for gene editing, and/or for inserting or deleting a polynucleotide of interest into or from the genome of a cell or organism. Once a genomic target site is identified, a variety of methods can be employed to further modify the target sites such that they contain a variety of polynucleotides of interest. Breeding methods and methods for selecting plants utilizing a two component RNA guide and Cas endonuclease system are also disclosed. Compositions and methods are also provided for editing a nucleotide sequence in the genome of a cell.
1. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising: a) obtaining a first plant comprising at least one Cas endonuclease capable of introducing a double strand break at a target site in the plant genome; b) obtaining a second plant comprising a guide RNA that is capable of forming a complex with the Cas endonuclease of (a); c) crossing the first plant of (a) with the second plant of (b); d) evaluating the progeny of (c) for an alteration in the target site; and, e) selecting a progeny plant that possesses the desired alteration of said target site. 2. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising selecting at least one progeny plant that comprises an alteration at a target site in its plant genome, wherein said progeny plant was obtained by crossing a first plant comprising at least one a Cas endonuclease with a second plant comprising a guide RNA, wherein said Cas endonuclease is capable of introducing a double strand break at said target site. 3. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising: a) obtaining a first plant comprising at least one Cas endonuclease capable of introducing a double strand break at a target site in the plant genome; b) obtaining a second plant comprising a guide RNA and a donor DNA, wherein said guide RNA is capable of forming a complex with the Cas endonuclease of (a), wherein said donor DNA comprises a polynucleotide of interest; c) crossing the first plant of (a) with the second plant of (b); d) evaluating the progeny of (c) for an alteration in the target site; and, e) selecting a progeny plant that comprises the polynucleotide of interest inserted at said target site. 4. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising selecting at least one progeny plant that comprises an alteration at a target site in its plant genome, wherein said progeny plant was obtained by crossing a first plant expressing at least one Cas endonuclease to a second plant comprising a guide RNA and a donor DNA, wherein said Cas endonuclease is capable of introducing a double strand break at said target site, wherein said donor DNA comprises a polynucleotide of interest. 5. A method for modifying a target site in the genome of a plant cell, the method comprising providing a guide RNA to a plant cell having a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site. 6. A method for modifying a target site in the genome of a plant cell, the method comprising providing a guide RNA and a Cas endonuclease to said plant cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site. 7. A method for modifying a target site in the genome of a plant cell, the method comprising providing a guide RNA and a donor DNA to a plant cell having a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site, wherein said donor DNA comprises a polynucleotide of interest. 8. A method for modifying a target site in the genome of a plant cell, the method comprising: a) providing to a plant cell a guide RNA and a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site; and, b) identifying at least one plant cell that has a modification at said target, wherein the modification includes at least one deletion or substitution of one or more nucleotides in said target site. 9. A method for modifying a target DNA sequence in the genome of a plant cell, the method comprising: a) providing to a plant cell a first recombinant DNA construct capable of expressing a guide RNA and a second recombinant DNA construct capable of expressing a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site; and, b) identifying at least one plant cell that has a modification at said target, wherein the modification includes at least one deletion or substitution of one or more nucleotides in said target site. 10. A method for introducing a polynucleotide of Interest into a target site in the genome of a plant cell, the method comprising: a) providing to a plant cell a first recombinant DNA construct capable of expressing a guide RNA and a second recombinant DNA construct capable of expressing a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site; b) contacting the plant cell of (a) with a donor DNA comprising a polynucleotide of Interest; and, c) identifying at least one plant cell from (b) comprising in its genome the polynucleotide of Interest integrated at said target site. 11. The method of claim 5, wherein the guide RNA is introduced directly by particle bombardment. 12. The method of claim 5, wherein the guide RNA is introduced via particle bombardment or Agrobacterium transformation of a recombinant DNA construct comprising the corresponding guide DNA operably linked to a plant U6 polymerase III promoter. 13. The method of claim 1, wherein the Cas endonuclease gene is a plant optimized Cas9 endonuclease. 14. The method of claim 1, wherein the Cas endonuclease gene is operably linked to a SV40 nuclear targeting signal upstream of the Cas codon region and a VirD2 nuclear localization signal downstream of the Cas codon region. 15. The method of claim 1, wherein the plant is a monocot or a dicot. 16. The method of claim 15, wherein the monocot is selected from the group consisting of maize, rice, sorghum, rye, barley, wheat, millet, oats, sugarcane, turfgrass, or switchgrass. 17. The method of claim 16, wherein the dicot is selected from the group consisting of soybean, canola, alfalfa, sunflower, cotton, tobacco, peanut, potato, tobacco, Arabidopsis, or safflower. 18. The method of claim 1, wherein the target site is located in the gene sequence of an acetolactate synthase (ALS) gene, an Enolpyruvylshikimate Phosphate Synthase Gene (ESPSP) gene, a male fertility (MS45, MS26 or MSCA1). 19. A plant or seed produced by the method of claim 5. 20. A plant comprising a recombinant DNA construct, said recombinant DNA construct comprising a promoter operably linked to a nucleotide sequence encoding a plant optimized Cas9 endonuclease, wherein said plant optimized Cas9 endonuclease is capable of binding to and creating a double strand break in a genomic target sequence said plant genome. 21. A plant comprising a recombinant DNA construct and a guide RNA, wherein said recombinant DNA construct comprises a promoter operably linked to a nucleotide sequence encoding a plant optimized Cas9 endonuclease, wherein said plant optimized Cas9 endonuclease and guide RNA are capable of forming a complex and creating a double strand break in a genomic target sequence said plant genome. 22. A recombinant DNA construct comprising a promoter operably linked to a nucleotide sequence encoding a plant optimized Cas9 endonuclease, wherein said plant optimized Cas9 endonuclease is capable of binding to and creating a double strand break in a genomic target sequence said plant genome. 23. A recombinant DNA construct comprising a promoter operably linked to a nucleotide sequence expressing a guide RNA, wherein said guide RNA is capable of forming a complex with a plant optimized Cas9 endonuclease, and wherein said complex is capable of binding to and creating a double strand break in a genomic target sequence said plant genome. 24. A method for selecting a male sterile plant, the method comprising selecting at least one progeny plant that comprises an alteration at a genomic target site located in a male fertility gene locus, wherein said progeny plant is obtained by crossing a first plant expressing a Cas9 endonuclease to a second plant comprising a guide RNA, wherein said Cas endonuclease is capable of introducing a double strand break at said genomic target site, 25. A method for producing a male sterile plant, the method comprising: a) obtaining a first plant comprising at least one Cas endonuclease capable of introducing a double strand break at a genomic target site located in a male fertility gene locus in the plant genome; b) obtaining a second plant comprising a guide RNA that is capable of forming a complex with the Cas endonuclease of (a); c) crossing the first plant of (a) with the second plant of (b); d) evaluating the progeny of (c) for an alteration in the target site; and, e) selecting a progeny plant that is male sterile. 26. The method of claim 24, wherein the male fertility gene is selected from the group consisting of MS26, MS45 and MSCA1. 27. The method of claim 24, wherein the plant is a monocot or a dicot. 28. The method of claim 27, wherein the monocot is selected from the group consisting of maize, rice, sorghum, rye, barley, wheat, millet, oats, sugarcane, turfgrass, or switchgrass. 29. A method for editing a nucleotide sequence in the genome of a cell, the method comprising introducing at least one guide RNA, at least one polynucleotide modification template and at least one Cas endonuclease into a cell, wherein the Cas endonuclease introduces a double-strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification of said nucleotide sequence. 30. The method of claim 29, wherein the cell is a plant cell. 31. The method of claim 29 wherein the nucleotide sequence is a promoter, a regulatory sequence or a gene of interest of interest. 32. The method of claim 31 wherein the gene of interest is an enolpyruvylshikimate-3-phosphate synthase (EPSPS) gene or an acetolactate synthase (ALS) gene. 33. The method of claim 30 wherein the plant cell is a monocot or dicot plant cell. 34. A method for producing an epsps mutant plant, the method comprising: a) providing a guide RNA, a polynucleotide modification template and at least one Cas endonuclease to a plant cell, wherein the Cas endonuclease introduces a double strand break at a target site within an epsps genomic sequence in the plant genome, wherein said polynucleotide modification template comprises at least one nucleotide modification of said epsps genomic sequence; b) obtaining a plant from the plant cell of (a); c) evaluating the plant of (b) for the presence of said at least one nucleotide modification; and, d) selecting a progeny plant that shows tolerance to glyphosate. 35. A method for producing an epsps mutant plant, the method comprising: a) providing a guide RNA, a polynucleotide modification template and at least one Cas endonuclease to a plant cell, wherein the Cas endonuclease introduces a double strand break at a target site within an epsps genomic sequence in the plant genome, wherein said polynucleotide modification template comprises at least one nucleotide modification of said epsps genomic sequence; b) obtaining a plant from the plant cell of (a); c) evaluating the plant of (b) for the presence of said at least one nucleotide modification; and, d) screening a progeny plant of (c) that is void of said guide RNA and Cas endonuclease. 36. The method of claim 35 further comprising selecting a plant that shows resistance to glyphosate. 37. A plant, plant cell or seed produced by the method of claim 29. 38. The method of claim 29, wherein the Cas endonuclease is a Cas9 endonuclease. 39. The method of claim 38, wherein the Cas9 endonuclease is expressed by SEQ ID NO: 5. 40. The method of claim 38 wherein the Cas9 endonuclease is encoded by any one of SEQ ID NOs: 1, 124, 212, 213, 214, 215, 216, 193 or nucleotides 2037-6329 of SEQ ID NO: 5, or any functional fragment thereof. 41. The plant or plant cell of claim 37, wherein said plant cell shows resistance to glyphosate. 42. A plant cell comprising a modified nucleotide sequence, wherein the modified nucleotide sequence was produced by providing a guide RNA, a polynucleotide modification template and at least one Cas endonuclease to a plant cell, wherein the Cas endonuclease is capable of introducing a double-strand break at a target site in the plant genome, wherein said polynucleotide modification template comprises at least one nucleotide modification of said nucleotide sequence. 43. The method of claim 29, wherein the at least one nucleotide modification is not a modification at said target site. 44. A method for producing a male sterile plant, the method comprising: a) providing to a plant cell a guide RNA and a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site located in or near a male fertility gene; b) identifying at least one plant cell that has a modification in said male fertility gene, wherein the modification includes at least one deletion, insertion, or substitution of one or more nucleotides in said male sterility gene; and, c) obtaining a plant from the plant cell of b). 45. The method of claim 44, further comprising selecting a progeny plant from the plant of c) wherein said progeny plant is male sterile. 46. The method of claim 44, wherein the male fertility gene is selected from the group consisting of MS26, MS45 and MSCA1. 47. A plant comprising at least one altered target site, wherein the at least one altered target site originated from a corresponding target site that was recognized and cleaved by a guideRNA/Cas endonuclease system, and wherein the at least one altered target site is in a genomic region of interest that extends from the target sequence set forth in SEQ ID NO: 229 to the target site set forth in SEQ ID NO: 235. 48. The plant of claim 47, wherein the at least one altered target site has an alteration selected from the group consisting of (i) replacement of at least one nucleotide, (ii) a deletion of at least one nucleotide, (iii) an insertion of at least one nucleotide, and (iv) any combination of (i)-(iii). 49. The plant of claim 47, wherein the at least one altered target site comprises a recombinant DNA molecule. 50. The plant of claim 47, wherein the plant comprises at least two altered target sites, wherein each of the altered target site originated from corresponding target site that was recognized and cleaved by a guideRNA/Cas endonuclease system, wherein the corresponding target site is selected from the group consisting of SEQ ID NOs: 229, 230, 231, 232, 233, 234, 235 and 236. 51. A method for editing a nucleotide sequence in the genome of a cell, the method comprising providing a guide polynucleotide, a Cas endonuclease, and optionally a polynucleotide modification template, to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification of said nucleotide sequence. 52. The method of claim 51, wherein the nucleotide sequence in the genome of a cell is selected from the group consisting of a promoter sequence, a terminator sequence, a regulatory element sequence, a splice site, a coding sequence, a polyubiquitination site, an intron site and an intron enhancing motif. 53. A method for editing a promoter sequence in the genome of a cell, the method comprising providing a guide polynucleotide, a polynucleotide modification template and at least one Cas endonuclease to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification of said promoter sequence to be edited. 54. A method for replacing a first promoter sequence in a cell, the method comprising providing a guide RNA, a polynucleotide modification template, and a Cas endonuclease to said cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises a second promoter or second promoter fragment that is different from said first promoter sequence. 55. The method of claim 54, wherein the replacement of the first promoter sequence results in any one of the following, or any one combination of the following: an increased promoter activity, an increased promoter tissue specificity, a decreased promoter activity, a decreased promoter tissue specificity, a new promoter activity, an inducible promoter activity, an extended window of gene expression, or a modification of the timing or developmental progress of gene expression in the same cell layer or other cell layer. 56. The method of claim 54, wherein the first promoter sequence is selected from the group consisting of Zea mays ARGOS 8 promoter, a soybean EPSPS1 promoter, a maize EPSPS promoter, maize NPK1 promoter, wherein the second promoter sequence is selected from the group consisting of a Zea mays GOS2 PRO:GOS2-intron promoter, a soybean ubiquitin promoter, a stress inducible maize RAB17 promoter, a Zea mays-PEPC1 promoter, a Zea mays Ubiquitin promoter, a Zea mays-Rootmet2 promoter, a rice actin promoter, a sorghum RCC3 promoter, a Zea mays-GOS2 promoter, a Zea mays-ACO2 promoter, and a Zea mays oleosin promoter. 57. A method for deleting a promoter sequence in the genome of a cell, the method comprising providing a guide polynucleotide, a Cas endonuclease to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break in at least one target site located inside or outside said promoter sequence. 58. A method for inserting a promoter or a promoter element in the genome of a cell, the method comprising providing a guide polynucleotide, a polynucleotide modification template comprising the promoter or the promoter element, and a Cas endonuclease to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell. 59. The method of claim 58, wherein the insertion of the promoter or promoter element results in any one of the following, or any one combination of the following: an increased promoter activity, an increased promoter tissue specificity, a decreased promoter activity, a decreased promoter tissue specificity, a new promoter activity, an inducible promoter activity, an extended window of gene expression, a modification of the timing or developmental progress of gene expression, a mutation of DNA binding elements, or an addition of DNA binding elements. 60. A method for editing a Zinc Finger transcription factor, the method comprising providing a guide polynucleotide, a Cas endonuclease, and optionally a polynucleotide modification template, to a cell, wherein the Cas endonuclease introduces a double-strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification or deletion of said Zinc Finger transcription factor, wherein the deletion or modification of said Zinc Finger transcription factor results in the creation of a dominant negative Zinc Finger transcription factor mutant. 61. A method for creating a fusion protein, the method comprising introducing a guide polynucleotide, a Cas endonuclease, and a polynucleotide modification template, into a cell, wherein the Cas endonuclease introduces a double-strand break at a target site located inside or outside a first coding sequence in the genome of said cell, wherein said polynucleotide modification template comprises a second coding sequence encoding a protein of interest, wherein the protein fusion results in any one of the following, or any one combination of the following: a targeting of the fusion protein to the chloroplast of said cell, an increased protein activity, an increased protein functionality, a decreased protein activity, a decreased protein functionality, a new protein functionality, a modified protein functionality, a new protein localization, a new timing of protein expression, a modified protein expression pattern, a chimeric protein, or a modified protein with dominant phenotype functionality. 62. A method for producing in a plant a complex trait locus comprising at least two altered target sequences in a genomic region of interest, said method comprising: (a) selecting a genomic region in a plant, wherein the genomic region comprises a first target sequence and a second target sequence; (b) contacting at least one plant cell with at least a first guide polynucleotide, a second polynucleotide, and optionally at least one Donor DNA, and a Cas endonuclease, wherein the first and second guide polynucleotide and the Cas endonuclease can form a complex that enables the Cas endonuclease to introduce a double strand break in at least a first and a second target sequence; (c) identifying a cell from (b) comprising a first alteration at the first target sequence and a second alteration at the second target sequence; and (d) recovering a first fertile plant from the cell of (c) said fertile plant comprising the first alteration and the second alteration, wherein the first alteration and the second alteration are physically linked. 63. A method for producing in a plant a complex trait locus comprising at least two altered target sequences in a genomic region of interest, said method comprising: (a) selecting a genomic region in a plant, wherein the genomic region comprises a first target sequence and a second target sequence; (b) contacting at least one plant cell with a first guide polynucleotide, a Cas endonuclease, and optionally a first Donor DNA, wherein the first guide polynucleotide and the Cas endonuclease can form a complex that enables the Cas endonuclease to introduce a double strand break a first target sequence; (c) identifying a cell from (b) comprising a first alteration at the first target sequence; (d) recovering a first fertile plant from the cell of (c), said first fertile plant comprising the first alteration; (e) contacting at least one plant cell with a second guide polynucleotide, a Cas endonuclease, and optionally a second Donor DNA; (f) identifying a cell from (e) comprising a second alteration at the second target sequence; (g) recovering a second fertile plant from the cell of (f), said second fertile plant comprising the second alteration; and, (h) obtaining a fertile progeny plant from the second fertile plant of (g), said fertile progeny plant comprising the first alteration and the second alteration, wherein the first alteration and the second alteration are physically linked. 64. The method of claim 29, wherein the editing of said nucleotide sequence renders said nucleotide sequence capable of conferring herbicide resistance to said cell. 65. A method for producing an acetolactate synthase (ALS) mutant plant, the method comprising: a) obtaining a plant or a seed thereof, wherein the plant or the seed comprises a modification in an endogenous ALS gene, the modification generated by a Cas endonuclease, a guide RNA and a polynucleotide modification template, wherein the plant or the seed is resistant to sulphonylurea; and, b) producing a progeny plant that is void of said guide RNA and Cas endonuclease. 66. A method of generating a sulphonylurea resistant plant, the method comprising providing a plant cell wherein its endogenous chromosomal ALS gene by has been modified through a guide RNA/Cas endonuclease system to produce a sulphonylurea resistant ALS protein and growing a plant from said maize plant cell, wherein said plant is resistant to sulphonylurea.
Compositions and methods are provided for genome modification of a target sequence in the genome of a plant or plant cell. The methods and compositions employ a guide RNA/Cas endonuclease system to provide an effective system for modifying or altering target sites within the genome of a plant, plant cell or seed. Also provided are compositions and methods employing a guide polynucleotide/Cas endonuclease system for genome modification of a nucleotide sequence in the genome of a cell or organism, for gene editing, and/or for inserting or deleting a polynucleotide of interest into or from the genome of a cell or organism. Once a genomic target site is identified, a variety of methods can be employed to further modify the target sites such that they contain a variety of polynucleotides of interest. Breeding methods and methods for selecting plants utilizing a two component RNA guide and Cas endonuclease system are also disclosed. Compositions and methods are also provided for editing a nucleotide sequence in the genome of a cell.1. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising: a) obtaining a first plant comprising at least one Cas endonuclease capable of introducing a double strand break at a target site in the plant genome; b) obtaining a second plant comprising a guide RNA that is capable of forming a complex with the Cas endonuclease of (a); c) crossing the first plant of (a) with the second plant of (b); d) evaluating the progeny of (c) for an alteration in the target site; and, e) selecting a progeny plant that possesses the desired alteration of said target site. 2. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising selecting at least one progeny plant that comprises an alteration at a target site in its plant genome, wherein said progeny plant was obtained by crossing a first plant comprising at least one a Cas endonuclease with a second plant comprising a guide RNA, wherein said Cas endonuclease is capable of introducing a double strand break at said target site. 3. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising: a) obtaining a first plant comprising at least one Cas endonuclease capable of introducing a double strand break at a target site in the plant genome; b) obtaining a second plant comprising a guide RNA and a donor DNA, wherein said guide RNA is capable of forming a complex with the Cas endonuclease of (a), wherein said donor DNA comprises a polynucleotide of interest; c) crossing the first plant of (a) with the second plant of (b); d) evaluating the progeny of (c) for an alteration in the target site; and, e) selecting a progeny plant that comprises the polynucleotide of interest inserted at said target site. 4. A method for selecting a plant comprising an altered target site in its plant genome, the method comprising selecting at least one progeny plant that comprises an alteration at a target site in its plant genome, wherein said progeny plant was obtained by crossing a first plant expressing at least one Cas endonuclease to a second plant comprising a guide RNA and a donor DNA, wherein said Cas endonuclease is capable of introducing a double strand break at said target site, wherein said donor DNA comprises a polynucleotide of interest. 5. A method for modifying a target site in the genome of a plant cell, the method comprising providing a guide RNA to a plant cell having a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site. 6. A method for modifying a target site in the genome of a plant cell, the method comprising providing a guide RNA and a Cas endonuclease to said plant cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site. 7. A method for modifying a target site in the genome of a plant cell, the method comprising providing a guide RNA and a donor DNA to a plant cell having a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site, wherein said donor DNA comprises a polynucleotide of interest. 8. A method for modifying a target site in the genome of a plant cell, the method comprising: a) providing to a plant cell a guide RNA and a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site; and, b) identifying at least one plant cell that has a modification at said target, wherein the modification includes at least one deletion or substitution of one or more nucleotides in said target site. 9. A method for modifying a target DNA sequence in the genome of a plant cell, the method comprising: a) providing to a plant cell a first recombinant DNA construct capable of expressing a guide RNA and a second recombinant DNA construct capable of expressing a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site; and, b) identifying at least one plant cell that has a modification at said target, wherein the modification includes at least one deletion or substitution of one or more nucleotides in said target site. 10. A method for introducing a polynucleotide of Interest into a target site in the genome of a plant cell, the method comprising: a) providing to a plant cell a first recombinant DNA construct capable of expressing a guide RNA and a second recombinant DNA construct capable of expressing a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at said target site; b) contacting the plant cell of (a) with a donor DNA comprising a polynucleotide of Interest; and, c) identifying at least one plant cell from (b) comprising in its genome the polynucleotide of Interest integrated at said target site. 11. The method of claim 5, wherein the guide RNA is introduced directly by particle bombardment. 12. The method of claim 5, wherein the guide RNA is introduced via particle bombardment or Agrobacterium transformation of a recombinant DNA construct comprising the corresponding guide DNA operably linked to a plant U6 polymerase III promoter. 13. The method of claim 1, wherein the Cas endonuclease gene is a plant optimized Cas9 endonuclease. 14. The method of claim 1, wherein the Cas endonuclease gene is operably linked to a SV40 nuclear targeting signal upstream of the Cas codon region and a VirD2 nuclear localization signal downstream of the Cas codon region. 15. The method of claim 1, wherein the plant is a monocot or a dicot. 16. The method of claim 15, wherein the monocot is selected from the group consisting of maize, rice, sorghum, rye, barley, wheat, millet, oats, sugarcane, turfgrass, or switchgrass. 17. The method of claim 16, wherein the dicot is selected from the group consisting of soybean, canola, alfalfa, sunflower, cotton, tobacco, peanut, potato, tobacco, Arabidopsis, or safflower. 18. The method of claim 1, wherein the target site is located in the gene sequence of an acetolactate synthase (ALS) gene, an Enolpyruvylshikimate Phosphate Synthase Gene (ESPSP) gene, a male fertility (MS45, MS26 or MSCA1). 19. A plant or seed produced by the method of claim 5. 20. A plant comprising a recombinant DNA construct, said recombinant DNA construct comprising a promoter operably linked to a nucleotide sequence encoding a plant optimized Cas9 endonuclease, wherein said plant optimized Cas9 endonuclease is capable of binding to and creating a double strand break in a genomic target sequence said plant genome. 21. A plant comprising a recombinant DNA construct and a guide RNA, wherein said recombinant DNA construct comprises a promoter operably linked to a nucleotide sequence encoding a plant optimized Cas9 endonuclease, wherein said plant optimized Cas9 endonuclease and guide RNA are capable of forming a complex and creating a double strand break in a genomic target sequence said plant genome. 22. A recombinant DNA construct comprising a promoter operably linked to a nucleotide sequence encoding a plant optimized Cas9 endonuclease, wherein said plant optimized Cas9 endonuclease is capable of binding to and creating a double strand break in a genomic target sequence said plant genome. 23. A recombinant DNA construct comprising a promoter operably linked to a nucleotide sequence expressing a guide RNA, wherein said guide RNA is capable of forming a complex with a plant optimized Cas9 endonuclease, and wherein said complex is capable of binding to and creating a double strand break in a genomic target sequence said plant genome. 24. A method for selecting a male sterile plant, the method comprising selecting at least one progeny plant that comprises an alteration at a genomic target site located in a male fertility gene locus, wherein said progeny plant is obtained by crossing a first plant expressing a Cas9 endonuclease to a second plant comprising a guide RNA, wherein said Cas endonuclease is capable of introducing a double strand break at said genomic target site, 25. A method for producing a male sterile plant, the method comprising: a) obtaining a first plant comprising at least one Cas endonuclease capable of introducing a double strand break at a genomic target site located in a male fertility gene locus in the plant genome; b) obtaining a second plant comprising a guide RNA that is capable of forming a complex with the Cas endonuclease of (a); c) crossing the first plant of (a) with the second plant of (b); d) evaluating the progeny of (c) for an alteration in the target site; and, e) selecting a progeny plant that is male sterile. 26. The method of claim 24, wherein the male fertility gene is selected from the group consisting of MS26, MS45 and MSCA1. 27. The method of claim 24, wherein the plant is a monocot or a dicot. 28. The method of claim 27, wherein the monocot is selected from the group consisting of maize, rice, sorghum, rye, barley, wheat, millet, oats, sugarcane, turfgrass, or switchgrass. 29. A method for editing a nucleotide sequence in the genome of a cell, the method comprising introducing at least one guide RNA, at least one polynucleotide modification template and at least one Cas endonuclease into a cell, wherein the Cas endonuclease introduces a double-strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification of said nucleotide sequence. 30. The method of claim 29, wherein the cell is a plant cell. 31. The method of claim 29 wherein the nucleotide sequence is a promoter, a regulatory sequence or a gene of interest of interest. 32. The method of claim 31 wherein the gene of interest is an enolpyruvylshikimate-3-phosphate synthase (EPSPS) gene or an acetolactate synthase (ALS) gene. 33. The method of claim 30 wherein the plant cell is a monocot or dicot plant cell. 34. A method for producing an epsps mutant plant, the method comprising: a) providing a guide RNA, a polynucleotide modification template and at least one Cas endonuclease to a plant cell, wherein the Cas endonuclease introduces a double strand break at a target site within an epsps genomic sequence in the plant genome, wherein said polynucleotide modification template comprises at least one nucleotide modification of said epsps genomic sequence; b) obtaining a plant from the plant cell of (a); c) evaluating the plant of (b) for the presence of said at least one nucleotide modification; and, d) selecting a progeny plant that shows tolerance to glyphosate. 35. A method for producing an epsps mutant plant, the method comprising: a) providing a guide RNA, a polynucleotide modification template and at least one Cas endonuclease to a plant cell, wherein the Cas endonuclease introduces a double strand break at a target site within an epsps genomic sequence in the plant genome, wherein said polynucleotide modification template comprises at least one nucleotide modification of said epsps genomic sequence; b) obtaining a plant from the plant cell of (a); c) evaluating the plant of (b) for the presence of said at least one nucleotide modification; and, d) screening a progeny plant of (c) that is void of said guide RNA and Cas endonuclease. 36. The method of claim 35 further comprising selecting a plant that shows resistance to glyphosate. 37. A plant, plant cell or seed produced by the method of claim 29. 38. The method of claim 29, wherein the Cas endonuclease is a Cas9 endonuclease. 39. The method of claim 38, wherein the Cas9 endonuclease is expressed by SEQ ID NO: 5. 40. The method of claim 38 wherein the Cas9 endonuclease is encoded by any one of SEQ ID NOs: 1, 124, 212, 213, 214, 215, 216, 193 or nucleotides 2037-6329 of SEQ ID NO: 5, or any functional fragment thereof. 41. The plant or plant cell of claim 37, wherein said plant cell shows resistance to glyphosate. 42. A plant cell comprising a modified nucleotide sequence, wherein the modified nucleotide sequence was produced by providing a guide RNA, a polynucleotide modification template and at least one Cas endonuclease to a plant cell, wherein the Cas endonuclease is capable of introducing a double-strand break at a target site in the plant genome, wherein said polynucleotide modification template comprises at least one nucleotide modification of said nucleotide sequence. 43. The method of claim 29, wherein the at least one nucleotide modification is not a modification at said target site. 44. A method for producing a male sterile plant, the method comprising: a) providing to a plant cell a guide RNA and a Cas endonuclease, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site located in or near a male fertility gene; b) identifying at least one plant cell that has a modification in said male fertility gene, wherein the modification includes at least one deletion, insertion, or substitution of one or more nucleotides in said male sterility gene; and, c) obtaining a plant from the plant cell of b). 45. The method of claim 44, further comprising selecting a progeny plant from the plant of c) wherein said progeny plant is male sterile. 46. The method of claim 44, wherein the male fertility gene is selected from the group consisting of MS26, MS45 and MSCA1. 47. A plant comprising at least one altered target site, wherein the at least one altered target site originated from a corresponding target site that was recognized and cleaved by a guideRNA/Cas endonuclease system, and wherein the at least one altered target site is in a genomic region of interest that extends from the target sequence set forth in SEQ ID NO: 229 to the target site set forth in SEQ ID NO: 235. 48. The plant of claim 47, wherein the at least one altered target site has an alteration selected from the group consisting of (i) replacement of at least one nucleotide, (ii) a deletion of at least one nucleotide, (iii) an insertion of at least one nucleotide, and (iv) any combination of (i)-(iii). 49. The plant of claim 47, wherein the at least one altered target site comprises a recombinant DNA molecule. 50. The plant of claim 47, wherein the plant comprises at least two altered target sites, wherein each of the altered target site originated from corresponding target site that was recognized and cleaved by a guideRNA/Cas endonuclease system, wherein the corresponding target site is selected from the group consisting of SEQ ID NOs: 229, 230, 231, 232, 233, 234, 235 and 236. 51. A method for editing a nucleotide sequence in the genome of a cell, the method comprising providing a guide polynucleotide, a Cas endonuclease, and optionally a polynucleotide modification template, to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification of said nucleotide sequence. 52. The method of claim 51, wherein the nucleotide sequence in the genome of a cell is selected from the group consisting of a promoter sequence, a terminator sequence, a regulatory element sequence, a splice site, a coding sequence, a polyubiquitination site, an intron site and an intron enhancing motif. 53. A method for editing a promoter sequence in the genome of a cell, the method comprising providing a guide polynucleotide, a polynucleotide modification template and at least one Cas endonuclease to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification of said promoter sequence to be edited. 54. A method for replacing a first promoter sequence in a cell, the method comprising providing a guide RNA, a polynucleotide modification template, and a Cas endonuclease to said cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises a second promoter or second promoter fragment that is different from said first promoter sequence. 55. The method of claim 54, wherein the replacement of the first promoter sequence results in any one of the following, or any one combination of the following: an increased promoter activity, an increased promoter tissue specificity, a decreased promoter activity, a decreased promoter tissue specificity, a new promoter activity, an inducible promoter activity, an extended window of gene expression, or a modification of the timing or developmental progress of gene expression in the same cell layer or other cell layer. 56. The method of claim 54, wherein the first promoter sequence is selected from the group consisting of Zea mays ARGOS 8 promoter, a soybean EPSPS1 promoter, a maize EPSPS promoter, maize NPK1 promoter, wherein the second promoter sequence is selected from the group consisting of a Zea mays GOS2 PRO:GOS2-intron promoter, a soybean ubiquitin promoter, a stress inducible maize RAB17 promoter, a Zea mays-PEPC1 promoter, a Zea mays Ubiquitin promoter, a Zea mays-Rootmet2 promoter, a rice actin promoter, a sorghum RCC3 promoter, a Zea mays-GOS2 promoter, a Zea mays-ACO2 promoter, and a Zea mays oleosin promoter. 57. A method for deleting a promoter sequence in the genome of a cell, the method comprising providing a guide polynucleotide, a Cas endonuclease to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break in at least one target site located inside or outside said promoter sequence. 58. A method for inserting a promoter or a promoter element in the genome of a cell, the method comprising providing a guide polynucleotide, a polynucleotide modification template comprising the promoter or the promoter element, and a Cas endonuclease to a cell, wherein said guide RNA and Cas endonuclease are capable of forming a complex that enables the Cas endonuclease to introduce a double strand break at a target site in the genome of said cell. 59. The method of claim 58, wherein the insertion of the promoter or promoter element results in any one of the following, or any one combination of the following: an increased promoter activity, an increased promoter tissue specificity, a decreased promoter activity, a decreased promoter tissue specificity, a new promoter activity, an inducible promoter activity, an extended window of gene expression, a modification of the timing or developmental progress of gene expression, a mutation of DNA binding elements, or an addition of DNA binding elements. 60. A method for editing a Zinc Finger transcription factor, the method comprising providing a guide polynucleotide, a Cas endonuclease, and optionally a polynucleotide modification template, to a cell, wherein the Cas endonuclease introduces a double-strand break at a target site in the genome of said cell, wherein said polynucleotide modification template comprises at least one nucleotide modification or deletion of said Zinc Finger transcription factor, wherein the deletion or modification of said Zinc Finger transcription factor results in the creation of a dominant negative Zinc Finger transcription factor mutant. 61. A method for creating a fusion protein, the method comprising introducing a guide polynucleotide, a Cas endonuclease, and a polynucleotide modification template, into a cell, wherein the Cas endonuclease introduces a double-strand break at a target site located inside or outside a first coding sequence in the genome of said cell, wherein said polynucleotide modification template comprises a second coding sequence encoding a protein of interest, wherein the protein fusion results in any one of the following, or any one combination of the following: a targeting of the fusion protein to the chloroplast of said cell, an increased protein activity, an increased protein functionality, a decreased protein activity, a decreased protein functionality, a new protein functionality, a modified protein functionality, a new protein localization, a new timing of protein expression, a modified protein expression pattern, a chimeric protein, or a modified protein with dominant phenotype functionality. 62. A method for producing in a plant a complex trait locus comprising at least two altered target sequences in a genomic region of interest, said method comprising: (a) selecting a genomic region in a plant, wherein the genomic region comprises a first target sequence and a second target sequence; (b) contacting at least one plant cell with at least a first guide polynucleotide, a second polynucleotide, and optionally at least one Donor DNA, and a Cas endonuclease, wherein the first and second guide polynucleotide and the Cas endonuclease can form a complex that enables the Cas endonuclease to introduce a double strand break in at least a first and a second target sequence; (c) identifying a cell from (b) comprising a first alteration at the first target sequence and a second alteration at the second target sequence; and (d) recovering a first fertile plant from the cell of (c) said fertile plant comprising the first alteration and the second alteration, wherein the first alteration and the second alteration are physically linked. 63. A method for producing in a plant a complex trait locus comprising at least two altered target sequences in a genomic region of interest, said method comprising: (a) selecting a genomic region in a plant, wherein the genomic region comprises a first target sequence and a second target sequence; (b) contacting at least one plant cell with a first guide polynucleotide, a Cas endonuclease, and optionally a first Donor DNA, wherein the first guide polynucleotide and the Cas endonuclease can form a complex that enables the Cas endonuclease to introduce a double strand break a first target sequence; (c) identifying a cell from (b) comprising a first alteration at the first target sequence; (d) recovering a first fertile plant from the cell of (c), said first fertile plant comprising the first alteration; (e) contacting at least one plant cell with a second guide polynucleotide, a Cas endonuclease, and optionally a second Donor DNA; (f) identifying a cell from (e) comprising a second alteration at the second target sequence; (g) recovering a second fertile plant from the cell of (f), said second fertile plant comprising the second alteration; and, (h) obtaining a fertile progeny plant from the second fertile plant of (g), said fertile progeny plant comprising the first alteration and the second alteration, wherein the first alteration and the second alteration are physically linked. 64. The method of claim 29, wherein the editing of said nucleotide sequence renders said nucleotide sequence capable of conferring herbicide resistance to said cell. 65. A method for producing an acetolactate synthase (ALS) mutant plant, the method comprising: a) obtaining a plant or a seed thereof, wherein the plant or the seed comprises a modification in an endogenous ALS gene, the modification generated by a Cas endonuclease, a guide RNA and a polynucleotide modification template, wherein the plant or the seed is resistant to sulphonylurea; and, b) producing a progeny plant that is void of said guide RNA and Cas endonuclease. 66. A method of generating a sulphonylurea resistant plant, the method comprising providing a plant cell wherein its endogenous chromosomal ALS gene by has been modified through a guide RNA/Cas endonuclease system to produce a sulphonylurea resistant ALS protein and growing a plant from said maize plant cell, wherein said plant is resistant to sulphonylurea.
1,600
301
14,412,189
1,619
The invention relates to the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and magnesium or a salt or complex thereof. The invention also relates to the combination of a sulforaphane or a derivative thereof and magnesium or a salt or complex thereof. The invention also relates to the combination of a broccoli extract or powder and magnesium or a salt or complex thereof. The invention provides compositions and methods relating to these combinations.
1-15. (canceled) 16. An orally administrable composition comprising a broccoli extract or powder and magnesium or a salt or complex thereof. 17. The orally administrable composition of claim 16, wherein the broccoli extract or powder comprises glucoraphanin in an amount of about 1 to about 75% w/w. 18. The orally administrable composition of claim 16, wherein the broccoli extract or powder comprises myrosinase. 19. The orally administrable composition of claim 16, further comprising an enzyme potentiator. 20. The orally administration composition of claim 16, wherein the enzyme potentiator comprises ascorbic acid. 21. The orally administrable composition of claim 16, wherein the composition comprises an enteric-coated dosage form. 22. The orally administrable composition of claim 16, further comprising one or more additional components selected from the group consisting of: vitamin k2, quercetin, an aminosugar, a glycosaminoglycan, avocado/soybean unsaponifiable, coffee fruit, magnesium, silymarin, proanthocyanidins, ursolic acid, curcumin, phytosterols, phytostanols, and maitake, shiitake, and/or reishi mushroom. 23. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and reducing secondary recurrences of a disease or condition associated with the endothelium or cardiovascular system, comprising administering to a subject in need thereof the orally administrable composition of claim 16. 24. A method of decreasing levels or decreasing gene expression of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject, comprising administering to the subject in need thereof the orally administrable composition of claim 16. 25. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a disease or condition associated with elevated levels of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject in need thereof, comprising administering to the subject in need thereof the orally administrable composition of claim 16. 26. An orally administrable composition comprising sulforaphane or a derivative thereof and magnesium or a salt or complex thereof. 27. The orally administrable composition of claim 26, further comprising one or more additional components selected from the group consisting of: vitamin k2, quercetin, an aminosugar, a glycosaminoglycan, avocado/soybean unsaponifiable, coffee fruit, magnesium, silymarin, proanthocyanidins, ursolic acid, curcumin, phytosterols, phytostanols, and maitake, shiitake, and/or reishi mushroom. 28. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and reducing secondary recurrences of a disease or condition associated with the endothelium or cardiovascular system, comprising administering to a subject in need thereof the orally administrable composition of claim 26. 29. A method of decreasing levels or decreasing gene expression of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject, comprising administering to the subject in need thereof the orally administrable composition of claim 26. 30. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a disease or condition associated with elevated levels of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject in need thereof, comprising administering to the subject in need thereof the orally administrable composition of claim 26.
The invention relates to the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and magnesium or a salt or complex thereof. The invention also relates to the combination of a sulforaphane or a derivative thereof and magnesium or a salt or complex thereof. The invention also relates to the combination of a broccoli extract or powder and magnesium or a salt or complex thereof. The invention provides compositions and methods relating to these combinations.1-15. (canceled) 16. An orally administrable composition comprising a broccoli extract or powder and magnesium or a salt or complex thereof. 17. The orally administrable composition of claim 16, wherein the broccoli extract or powder comprises glucoraphanin in an amount of about 1 to about 75% w/w. 18. The orally administrable composition of claim 16, wherein the broccoli extract or powder comprises myrosinase. 19. The orally administrable composition of claim 16, further comprising an enzyme potentiator. 20. The orally administration composition of claim 16, wherein the enzyme potentiator comprises ascorbic acid. 21. The orally administrable composition of claim 16, wherein the composition comprises an enteric-coated dosage form. 22. The orally administrable composition of claim 16, further comprising one or more additional components selected from the group consisting of: vitamin k2, quercetin, an aminosugar, a glycosaminoglycan, avocado/soybean unsaponifiable, coffee fruit, magnesium, silymarin, proanthocyanidins, ursolic acid, curcumin, phytosterols, phytostanols, and maitake, shiitake, and/or reishi mushroom. 23. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and reducing secondary recurrences of a disease or condition associated with the endothelium or cardiovascular system, comprising administering to a subject in need thereof the orally administrable composition of claim 16. 24. A method of decreasing levels or decreasing gene expression of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject, comprising administering to the subject in need thereof the orally administrable composition of claim 16. 25. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a disease or condition associated with elevated levels of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject in need thereof, comprising administering to the subject in need thereof the orally administrable composition of claim 16. 26. An orally administrable composition comprising sulforaphane or a derivative thereof and magnesium or a salt or complex thereof. 27. The orally administrable composition of claim 26, further comprising one or more additional components selected from the group consisting of: vitamin k2, quercetin, an aminosugar, a glycosaminoglycan, avocado/soybean unsaponifiable, coffee fruit, magnesium, silymarin, proanthocyanidins, ursolic acid, curcumin, phytosterols, phytostanols, and maitake, shiitake, and/or reishi mushroom. 28. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and reducing secondary recurrences of a disease or condition associated with the endothelium or cardiovascular system, comprising administering to a subject in need thereof the orally administrable composition of claim 26. 29. A method of decreasing levels or decreasing gene expression of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject, comprising administering to the subject in need thereof the orally administrable composition of claim 26. 30. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a disease or condition associated with elevated levels of interleukin-8 (IL-8) and/or monocyte chemoattractant protein-1 (MCP-1) in a subject in need thereof, comprising administering to the subject in need thereof the orally administrable composition of claim 26.
1,600
302
14,550,713
1,639
Provided herein, among other things, is a method comprising: (a) obtaining a mixture of multiple sets of oligonucleotides, wherein the oligonucleotides within each set each comprise a terminal indexer sequence can be assembled to produce a synthon; and (b) hybridizing the oligonucleotide mixture to an array, thereby spatially-separating the different sets of oligonucleotides from one another. Other embodiments are also provided.
1. A method comprising: (a) obtaining a mixture of multiple sets of oligonucleotides, wherein the oligonucleotides within each set each comprise a terminal indexer sequence and can be assembled to produce a synthon; and (b) hybridizing the oligonucleotide mixture to an array, thereby spatially-separating the different sets of oligonucleotides from one another. 2. The method of claim 1, wherein the oligonucleotides are single-stranded oligonucleotides. 3. The method of claim 1, wherein the mixture comprises double-stranded oligonucleotides. 4. The method of claim 1, wherein the oligonucleotides are single stranded and comprise a 3′ hairpin. 5. The method of claim 4, comprising: contacting the array with a solution comprising a polymerase and nucleotides, thereby extending the hairpin and producing, for each feature bound by the oligonucleotides, a set of double-stranded extension products. 6. The method of claim 1, wherein the oligonucleotides hybridize directly to oligonucleotides that are immobilized on the array. 7. The method of claim 1, wherein the oligonucleotides hybridize via an adaptor to oligonucleotides that are immobilized on the array. 8. The method of claim 7, wherein the method comprises: contacting the array with a solution comprising a polymerase and nucleotides, thereby extending the adaptor and producing, for each feature bound by the oligonucleotides, a set of double-stranded extension products. 9. The method of claim 1, further comprising (c) contacting the array with a solution, thereby producing, for each feature bound by the oligonucleotides, a discrete droplet comprising one or more features. 10. The method of claim 9, further comprising placing an immiscible liquid over the droplets, thereby producing, for each feature bound by the oligonucleotides, a discrete reaction chamber defined by a droplet. 11. The method of claim 10, further comprising incubating the array under conditions by which a synthon is assembled in each of the reaction chambers. 12. The method of claim 11, wherein the droplets comprise double-stranded oligonucleotides or double-stranded extension products, and the solution comprises a Type IIs restriction endonuclease, a DNA ligase and ATP, wherein the products of digestion of the double-stranded oligonucleotides or double-stranded extension products by the Type IIs restriction endonuclease are ligated to one another in a defined order by the DNA ligase in the discrete reaction chambers, thereby producing a synthon. 13. The method of claim 11, wherein the oligonucleotides are single-stranded oligonucleotides and the method comprises: cleaving the terminal indexer sequence from the oligonucleotides to release assembly sequences from at least some of the oligonucleotides; and assembling the synthon from the assembly sequences by polymerase chain assembly or by ligation. 14. The method of claim 13 wherein the cleaving the terminal indexer sequence from the oligonucleotide comprises cleaving a photocleavable linkage. 15. The method of claim 1, wherein the oligonucleotides are double-stranded oligonucleotides that comprise staggered photocleavable or chemically cleavable linkages, and wherein the method comprises cleaving said staggered cleavable linkages using light or a chemical treatment to produce fragments that are ligatable to one another in order. 16. The method of claim 11, further comprising separating the synthons from the array. 17. A composition comprising multiple sets of oligonucleotides, wherein the oligonucleotides within each set comprise: (i) a terminal indexer sequence and (ii) an assembly sequence, wherein the terminal indexer sequence and the assembly sequence are separated by a photocleavable or chemically cleavable linker and the assembly sequences of each set of oligonucleotides can be assembled to produce a synthon. 18. The composition of claim 17, wherein the assembly sequences of each set comprise overlapping complementary sequences that can be ligated directly to one another after cleavage of the terminal indexer sequences. 19. The composition of claim 17, wherein the assembly sequences of each set comprise overlapping complementary sequences that can be assembled by polymerase chain assembly after cleavage of the terminal indexer sequences. 20. An apparatus comprising: a planar support, a plurality of spatially distinct droplets on a surface of the planar support, and an immiscible liquid covering the droplets, wherein the apparatus comprises a plurality of reaction chambers defined by the droplets, and each reaction chamber comprises a different synthon.
Provided herein, among other things, is a method comprising: (a) obtaining a mixture of multiple sets of oligonucleotides, wherein the oligonucleotides within each set each comprise a terminal indexer sequence can be assembled to produce a synthon; and (b) hybridizing the oligonucleotide mixture to an array, thereby spatially-separating the different sets of oligonucleotides from one another. Other embodiments are also provided.1. A method comprising: (a) obtaining a mixture of multiple sets of oligonucleotides, wherein the oligonucleotides within each set each comprise a terminal indexer sequence and can be assembled to produce a synthon; and (b) hybridizing the oligonucleotide mixture to an array, thereby spatially-separating the different sets of oligonucleotides from one another. 2. The method of claim 1, wherein the oligonucleotides are single-stranded oligonucleotides. 3. The method of claim 1, wherein the mixture comprises double-stranded oligonucleotides. 4. The method of claim 1, wherein the oligonucleotides are single stranded and comprise a 3′ hairpin. 5. The method of claim 4, comprising: contacting the array with a solution comprising a polymerase and nucleotides, thereby extending the hairpin and producing, for each feature bound by the oligonucleotides, a set of double-stranded extension products. 6. The method of claim 1, wherein the oligonucleotides hybridize directly to oligonucleotides that are immobilized on the array. 7. The method of claim 1, wherein the oligonucleotides hybridize via an adaptor to oligonucleotides that are immobilized on the array. 8. The method of claim 7, wherein the method comprises: contacting the array with a solution comprising a polymerase and nucleotides, thereby extending the adaptor and producing, for each feature bound by the oligonucleotides, a set of double-stranded extension products. 9. The method of claim 1, further comprising (c) contacting the array with a solution, thereby producing, for each feature bound by the oligonucleotides, a discrete droplet comprising one or more features. 10. The method of claim 9, further comprising placing an immiscible liquid over the droplets, thereby producing, for each feature bound by the oligonucleotides, a discrete reaction chamber defined by a droplet. 11. The method of claim 10, further comprising incubating the array under conditions by which a synthon is assembled in each of the reaction chambers. 12. The method of claim 11, wherein the droplets comprise double-stranded oligonucleotides or double-stranded extension products, and the solution comprises a Type IIs restriction endonuclease, a DNA ligase and ATP, wherein the products of digestion of the double-stranded oligonucleotides or double-stranded extension products by the Type IIs restriction endonuclease are ligated to one another in a defined order by the DNA ligase in the discrete reaction chambers, thereby producing a synthon. 13. The method of claim 11, wherein the oligonucleotides are single-stranded oligonucleotides and the method comprises: cleaving the terminal indexer sequence from the oligonucleotides to release assembly sequences from at least some of the oligonucleotides; and assembling the synthon from the assembly sequences by polymerase chain assembly or by ligation. 14. The method of claim 13 wherein the cleaving the terminal indexer sequence from the oligonucleotide comprises cleaving a photocleavable linkage. 15. The method of claim 1, wherein the oligonucleotides are double-stranded oligonucleotides that comprise staggered photocleavable or chemically cleavable linkages, and wherein the method comprises cleaving said staggered cleavable linkages using light or a chemical treatment to produce fragments that are ligatable to one another in order. 16. The method of claim 11, further comprising separating the synthons from the array. 17. A composition comprising multiple sets of oligonucleotides, wherein the oligonucleotides within each set comprise: (i) a terminal indexer sequence and (ii) an assembly sequence, wherein the terminal indexer sequence and the assembly sequence are separated by a photocleavable or chemically cleavable linker and the assembly sequences of each set of oligonucleotides can be assembled to produce a synthon. 18. The composition of claim 17, wherein the assembly sequences of each set comprise overlapping complementary sequences that can be ligated directly to one another after cleavage of the terminal indexer sequences. 19. The composition of claim 17, wherein the assembly sequences of each set comprise overlapping complementary sequences that can be assembled by polymerase chain assembly after cleavage of the terminal indexer sequences. 20. An apparatus comprising: a planar support, a plurality of spatially distinct droplets on a surface of the planar support, and an immiscible liquid covering the droplets, wherein the apparatus comprises a plurality of reaction chambers defined by the droplets, and each reaction chamber comprises a different synthon.
1,600
303
14,120,309
1,618
Described herein are methods of delivering a nanoparticle to the brain of a subject by administering to the subject a nanoparticle having a nanoparticle core and a targeting agent. A variety of targeting agents may serve to promote delivery of the described nanoparticle. For example, the targeting agent may include a ligand specific for a receptor expressed by brain endothelial cells and a linker that connects the ligand to the external surface of the nanoparticle core. Additionally, the linker can promote disassociation of the ligand from the nanoparticle when inside a cell.
1. A method of delivering a nanoparticle to the brain of a subject comprising administering to the subject a nanoparticle having a nanoparticle core and a targeting agent, wherein said targeting agent includes a ligand specific for a receptor expressed by brain endothelial cells and a linker that connects the ligand to the nanoparticle core, wherein said linker causes dissociation of the ligand from the nanoparticle when inside a brain endothelial cell, and wherein said ligand is conjugated to the external surface of the nanoparticle core through the linker. 2. The method of claim 1, wherein: the surface of the nanoparticle core comprises any one of cationic mucic acid polymers (cMAP), poly(lactic-co-glycolic acid) (PLGA), chitosan, synthetic polymers such as polyethyleneimine, dendrimers, gold, or iron oxide; the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises a nitrophenyl boronic acid when unbound to the nanoparticle and a forms a nitrophenyl boronic ester when bound to the nanoparticle. 3. The method of claim 2, wherein the nanoparticle core comprises cationic mucic acid polymer (cMAP) having the structure: wherein m is any whole number between 5 and 50. 4. The method of claim 3, wherein m is any one of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. 5. The method of claim 1, wherein the linker of the targeting agent further comprises a polyethylene glycol (PEG) polymer between said nitrophenyl boronic acid and said ligand. 6. The method of claim 5, wherein the linker has the structure: where n is any whole number between 2 and 2000 and R is a functional group selected from a primary amine, azide, alcohol, thiol, aldehyde, or carboxylic acid. 7. The method of claim 6, wherein the targeting agent is and n is any whole number between 2 and 2000. 8. The method of claim 7, wherein n is any whole number from about 110 to about 150. 9. The method of claim 1, wherein: the surface of the nanoparticle core comprises poly(lactic-co-glycolic acid) (PLGA) polymers, the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises a PEG polymer. 10. The method of claim 9, wherein the PEG polymer is conjugated to the ligand via a disulfide bond or a polypeptide having an enzyme cleavage site. 11. The method of claim 10, wherein the nanoparticle core comprises PLGA having the structure: wherein x and y are independently any whole number between 5 and 500. 12. The method of claim 11, wherein x and y are independently any one of 40, 45, 50, 55, or 60. 13. The method of claim 12, wherein the nanoparticle core comprising PLGA is conjugated to a PEG linker and has the structure: and z is any whole number between 2 and 2000 and x and y are independently any whole number between 5 and 500, and R is selected from a primary amine, azide, alcohol, thiol, aldehyde, or carboxylic acid. 14. The method of claim 13, wherein z is any whole number from about 110 to about 150 and x and y are 50. 15. The method of claim 14, wherein the targeting agent is and z is any whole number from about 110 to about 150 and x and y are 50. 16. The method of claim 1, wherein: the surface of the nanoparticle core comprises any one of cationic mucic acid polymers (cMAP), poly(lactic-co-glycolic acid) (PLGA), chitosan, synthetic polymers such as polyethyleneimine, dendrimers, gold, or iron oxide; the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises a diamino ketal conjugated to PEG. 17. The method of claim 1, wherein the linker includes a disulfide bond that can be reduced to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 18. The method of claim 1, wherein the linker includes a polypeptide or chemical bond that can be enzymatically cleaved to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 19. The method of claim 1, wherein the linker includes a hydrolyzable chemical bond that can be disrupted at low pH to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 20. The method of claim 1, wherein the linker includes a chemical bond having a pKa that can be disrupted at low pH to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 21. The method of claim 19, wherein low pH is a value from about 6.8 to about 2.0. 22. The method of claim 21, wherein low pH is a value from about 5.5 to about 2.5. 23. The method of claim 21, wherein low pH is a value from about 5.5 to about 4.0. 24. The method of claim 19, wherein the surface of the nanoparticle core comprises poly(lactic-co-glycolic acid) (PLGA). 25. The method of claim 19, wherein the surface of the nanoparticle core comprises cationic mucic acid polymers (cMAP). 26. The method of claim 19, wherein the surface of the nanoparticle core comprises gold. 27. The method of claim 1, wherein: the surface of the nanoparticle core comprises the surface of the nanoparticle core comprises any one of cationic mucic acid polymers (cMAP), poly(lactic-co-glycolic acid) (PLGA), chitosan, synthetic polymers such as polyethyleneimine, dendrimers, gold, or iron oxide; the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like-growth factor receptor 1; a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises an acid-cleavable chemical bond selected from an orthoester, acetal, ketal, imine, or hydrazone, that is conjugated to PEG. 28. The method of claim 1, wherein the nanoparticle comprises less than 200 targeting agents conjugated to its surface. 29. The method of claim 1, wherein the nanoparticle comprises less than 20 targeting agents conjugated to its surface. 30. The method of claim 1, wherein the nanoparticle comprises less than 5 targeting agents conjugated to its surface. 31. The method of claim 1, wherein the nanoparticle comprises a single targeting agent conjugated to its surface. 32. The method of claim 1, wherein the nanoparticle has a size of from about 40 nm to about 100 nm as measured by dynamic light scattering (DLS). 33. The method of claim 32, wherein the nanoparticle has a size of from about 50 nm to about 70 nm as measured by dynamic light scattering (DLS). 34. The method of claim 33, wherein the nanoparticle has a size of 55 nm, 56 nm, 57 nm, 58 nm, 59 nm, 60 nm, 61 nm, 62 nm, 63 nm, 64 nm, 65 nm, 66 nm, 67 nm, 68 nm, or 69 nm as measured by dynamic light scattering (DLS). 35. The method of claim 1, wherein the nanoparticle has an average zeta potential of from about −0.5 mV to about −15.0 mV as measured by phase analysis light scattering. 36. The method of claim 35, wherein the nanoparticle has an average zeta potential of −5.0, −5.1, −5.2, −5.3, −5.4, −5.5, −5.6, −5.7, −5.8, −5.9, −6.0, −6.1, −6.2, −6.3, −6.4, −6.5, −6.6, −6.7, −6.8, −6.9, −7.0, −7.1, −7.2, −7.3, −7.4, −7.5, −7.6, −7.7, −7.8, −7.9, or −8.0 mV as measured by phase analysis light scattering. 37. The method of claim 1, wherein the nanoparticle further comprises a therapeutic agent. 38. The method of claim 37, wherein the therapeutic agent is effective against a neurological disorder. 39. The method of claim 38, wherein the therapeutic agent is serotonin or dopamine. 40. The method of claim 1, wherein the nanoparticle further comprises an imaging agent. 41. The method of claim 40, wherein the imaging agent is Cu-64. 42. The method of claim 1, wherein the nanoparticle includes a first targeting agent and a second targeting agent, wherein the second targeting agent comprises: a linker that is not amenable to disassociation from the nanoparticle core when inside of a brain endothelial cell, and a ligand that targets the particle to a specific cell in the brain. 43. A kit for producing a nanoparticle targeted for delivery to the brain comprising cationic mucic acid polymers (cMAP) or poly(lactic-co-glycolic acid) (PLGA); a targeting agent specific for a receptor expressed by brain endothelial cells, wherein said targeting agent includes a ligand that is conjugated to a linker that causes dissociation of the ligand from the nanoparticle when inside a brain endothelial cell; and instructions for assembling the nanoparticle. 44. The kit of claim 43, wherein the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and wherein the linker comprises a nitrophenyl boronic acid when unbound to the nanoparticle and a forms a nitrophenyl boronic ester when bound to the nanoparticle. 45. The kit of claim 44, further comprising a therapeutic agent or an imaging agent.
Described herein are methods of delivering a nanoparticle to the brain of a subject by administering to the subject a nanoparticle having a nanoparticle core and a targeting agent. A variety of targeting agents may serve to promote delivery of the described nanoparticle. For example, the targeting agent may include a ligand specific for a receptor expressed by brain endothelial cells and a linker that connects the ligand to the external surface of the nanoparticle core. Additionally, the linker can promote disassociation of the ligand from the nanoparticle when inside a cell.1. A method of delivering a nanoparticle to the brain of a subject comprising administering to the subject a nanoparticle having a nanoparticle core and a targeting agent, wherein said targeting agent includes a ligand specific for a receptor expressed by brain endothelial cells and a linker that connects the ligand to the nanoparticle core, wherein said linker causes dissociation of the ligand from the nanoparticle when inside a brain endothelial cell, and wherein said ligand is conjugated to the external surface of the nanoparticle core through the linker. 2. The method of claim 1, wherein: the surface of the nanoparticle core comprises any one of cationic mucic acid polymers (cMAP), poly(lactic-co-glycolic acid) (PLGA), chitosan, synthetic polymers such as polyethyleneimine, dendrimers, gold, or iron oxide; the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises a nitrophenyl boronic acid when unbound to the nanoparticle and a forms a nitrophenyl boronic ester when bound to the nanoparticle. 3. The method of claim 2, wherein the nanoparticle core comprises cationic mucic acid polymer (cMAP) having the structure: wherein m is any whole number between 5 and 50. 4. The method of claim 3, wherein m is any one of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. 5. The method of claim 1, wherein the linker of the targeting agent further comprises a polyethylene glycol (PEG) polymer between said nitrophenyl boronic acid and said ligand. 6. The method of claim 5, wherein the linker has the structure: where n is any whole number between 2 and 2000 and R is a functional group selected from a primary amine, azide, alcohol, thiol, aldehyde, or carboxylic acid. 7. The method of claim 6, wherein the targeting agent is and n is any whole number between 2 and 2000. 8. The method of claim 7, wherein n is any whole number from about 110 to about 150. 9. The method of claim 1, wherein: the surface of the nanoparticle core comprises poly(lactic-co-glycolic acid) (PLGA) polymers, the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises a PEG polymer. 10. The method of claim 9, wherein the PEG polymer is conjugated to the ligand via a disulfide bond or a polypeptide having an enzyme cleavage site. 11. The method of claim 10, wherein the nanoparticle core comprises PLGA having the structure: wherein x and y are independently any whole number between 5 and 500. 12. The method of claim 11, wherein x and y are independently any one of 40, 45, 50, 55, or 60. 13. The method of claim 12, wherein the nanoparticle core comprising PLGA is conjugated to a PEG linker and has the structure: and z is any whole number between 2 and 2000 and x and y are independently any whole number between 5 and 500, and R is selected from a primary amine, azide, alcohol, thiol, aldehyde, or carboxylic acid. 14. The method of claim 13, wherein z is any whole number from about 110 to about 150 and x and y are 50. 15. The method of claim 14, wherein the targeting agent is and z is any whole number from about 110 to about 150 and x and y are 50. 16. The method of claim 1, wherein: the surface of the nanoparticle core comprises any one of cationic mucic acid polymers (cMAP), poly(lactic-co-glycolic acid) (PLGA), chitosan, synthetic polymers such as polyethyleneimine, dendrimers, gold, or iron oxide; the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises a diamino ketal conjugated to PEG. 17. The method of claim 1, wherein the linker includes a disulfide bond that can be reduced to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 18. The method of claim 1, wherein the linker includes a polypeptide or chemical bond that can be enzymatically cleaved to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 19. The method of claim 1, wherein the linker includes a hydrolyzable chemical bond that can be disrupted at low pH to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 20. The method of claim 1, wherein the linker includes a chemical bond having a pKa that can be disrupted at low pH to cause dissociation of the ligand from the nanoparticle when the nanoparticle is inside a brain endothelial cell. 21. The method of claim 19, wherein low pH is a value from about 6.8 to about 2.0. 22. The method of claim 21, wherein low pH is a value from about 5.5 to about 2.5. 23. The method of claim 21, wherein low pH is a value from about 5.5 to about 4.0. 24. The method of claim 19, wherein the surface of the nanoparticle core comprises poly(lactic-co-glycolic acid) (PLGA). 25. The method of claim 19, wherein the surface of the nanoparticle core comprises cationic mucic acid polymers (cMAP). 26. The method of claim 19, wherein the surface of the nanoparticle core comprises gold. 27. The method of claim 1, wherein: the surface of the nanoparticle core comprises the surface of the nanoparticle core comprises any one of cationic mucic acid polymers (cMAP), poly(lactic-co-glycolic acid) (PLGA), chitosan, synthetic polymers such as polyethyleneimine, dendrimers, gold, or iron oxide; the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like-growth factor receptor 1; a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and the linker comprises an acid-cleavable chemical bond selected from an orthoester, acetal, ketal, imine, or hydrazone, that is conjugated to PEG. 28. The method of claim 1, wherein the nanoparticle comprises less than 200 targeting agents conjugated to its surface. 29. The method of claim 1, wherein the nanoparticle comprises less than 20 targeting agents conjugated to its surface. 30. The method of claim 1, wherein the nanoparticle comprises less than 5 targeting agents conjugated to its surface. 31. The method of claim 1, wherein the nanoparticle comprises a single targeting agent conjugated to its surface. 32. The method of claim 1, wherein the nanoparticle has a size of from about 40 nm to about 100 nm as measured by dynamic light scattering (DLS). 33. The method of claim 32, wherein the nanoparticle has a size of from about 50 nm to about 70 nm as measured by dynamic light scattering (DLS). 34. The method of claim 33, wherein the nanoparticle has a size of 55 nm, 56 nm, 57 nm, 58 nm, 59 nm, 60 nm, 61 nm, 62 nm, 63 nm, 64 nm, 65 nm, 66 nm, 67 nm, 68 nm, or 69 nm as measured by dynamic light scattering (DLS). 35. The method of claim 1, wherein the nanoparticle has an average zeta potential of from about −0.5 mV to about −15.0 mV as measured by phase analysis light scattering. 36. The method of claim 35, wherein the nanoparticle has an average zeta potential of −5.0, −5.1, −5.2, −5.3, −5.4, −5.5, −5.6, −5.7, −5.8, −5.9, −6.0, −6.1, −6.2, −6.3, −6.4, −6.5, −6.6, −6.7, −6.8, −6.9, −7.0, −7.1, −7.2, −7.3, −7.4, −7.5, −7.6, −7.7, −7.8, −7.9, or −8.0 mV as measured by phase analysis light scattering. 37. The method of claim 1, wherein the nanoparticle further comprises a therapeutic agent. 38. The method of claim 37, wherein the therapeutic agent is effective against a neurological disorder. 39. The method of claim 38, wherein the therapeutic agent is serotonin or dopamine. 40. The method of claim 1, wherein the nanoparticle further comprises an imaging agent. 41. The method of claim 40, wherein the imaging agent is Cu-64. 42. The method of claim 1, wherein the nanoparticle includes a first targeting agent and a second targeting agent, wherein the second targeting agent comprises: a linker that is not amenable to disassociation from the nanoparticle core when inside of a brain endothelial cell, and a ligand that targets the particle to a specific cell in the brain. 43. A kit for producing a nanoparticle targeted for delivery to the brain comprising cationic mucic acid polymers (cMAP) or poly(lactic-co-glycolic acid) (PLGA); a targeting agent specific for a receptor expressed by brain endothelial cells, wherein said targeting agent includes a ligand that is conjugated to a linker that causes dissociation of the ligand from the nanoparticle when inside a brain endothelial cell; and instructions for assembling the nanoparticle. 44. The kit of claim 43, wherein the ligand is any one of transferrin, an antibody specific for the transferrin receptor, a polypeptide that specifically binds to the transferrin receptor, insulin, an antibody specific for the insulin receptor, a polypeptide that specifically binds to the insulin receptor, insulin-like growth factor 1, an antibody specific for the insulin-like growth factor receptor 1, a polypeptide that specifically binds to the insulin-like growth factor receptor 1, apolipoprotein E, angiopep-2, an antibody specific for low density lipoprotein receptor or lipoprotein receptor-related protein, a polypeptide that specifically binds to low density lipoprotein receptor or lipoprotein receptor-related protein; an antibody specific for diphtheria toxin receptor, or a polypeptide that specifically binds to diphtheria toxin receptor; and wherein the linker comprises a nitrophenyl boronic acid when unbound to the nanoparticle and a forms a nitrophenyl boronic ester when bound to the nanoparticle. 45. The kit of claim 44, further comprising a therapeutic agent or an imaging agent.
1,600
304
15,758,593
1,653
Provided is a method for preparing a glycoprotein sugar chain including: an isolation step of acting a sugar chain-isolating enzyme on a sample which contains a glycoprotein fixed to a solid phase in a container to obtain an isolated product which contains a sugar chain; and a labeling step of adding a labeling reagent to the isolated product in the container to obtain a labeled product which contains a labeled substance of the sugar chain.
1-22. (canceled) 23. A method for preparing a glycoprotein sugar chain, comprising: an isolation step of acting a sugar chain-isolating enzyme on a sample which contains a glycoprotein fixed to a solid phase in a container to obtain an isolated product which contains a sugar chain; and a labeling step of adding a labeling reagent to the isolated product in the container to obtain a labeled product which contains a labeled substance of the sugar chain, wherein the glycoprotein is an antibody, and the solid phase includes a ligand selected from the group consisting of protein A, protein G, protein L, protein H, protein D, and protein Arp, in the surface thereof. 24. The method for preparing a glycoprotein sugar chain according to claim 23, wherein the isolation step is performed in an open system and under heating conditions. 25. The method of preparing a glycoprotein sugar chain according to claim 23, wherein the labeling reagent contains 2-aminobenzamide, a reducing agent, and a solvent. 26. The method of preparing a glycoprotein sugar chain according to claim 25, wherein the reducing agent is picoline borane. 27. The method of preparing a glycoprotein sugar chain according to claim 25, wherein the solvent contains a protic compound. 28. The method of preparing a glycoprotein sugar chain according to claim 27, wherein the solvent further contains an aprotic compound having a boiling point higher than that of the protic compound. 29. The method of preparing a glycoprotein sugar chain according to claim 23, further comprising: a separation step of performing solid-liquid separation after the isolation step to obtain a separate liquid which contains the isolated product. 30. The method of preparing a glycoprotein sugar chain according to claim 23, further comprising: a separation step of performing solid-liquid separation after the labeling step to obtain a separate liquid which contains the labeled substance of the sugar chain. 31. A method for preparing a glycoprotein sugar chain, comprising: an isolation step of acting a sugar chain-isolating enzyme on a sample which contains a glycoprotein fixed to a solid phase in a container to obtain an isolated product which contains a sugar chain; and a labeling step of adding a labeling reagent to the isolated product in the container to obtain a labeled product which contains a labeled substance of the sugar chain, wherein wherein the isolation step is performed in the presence of a deglycosylation promoter containing an acid-derived anionic surfactant. 32. The method for preparing a glycoprotein sugar chain according to claim 31, further comprising: a pre-treatment step of bringing a pre-treatment agent containing a surfactant into contact with the sample before the isolation step. 33. The method for preparing a glycoprotein sugar chain according to claim 31, wherein the acid-derived anionic surfactant is selected from the group consisting of a carboxylic acid type anionic surfactant, a sulfonic acid type anionic surfactant, a sulfuric acid ester type anionic surfactant, and a phosphoric acid ester type anionic surfactant. 34. A kit for preparing a glycoprotein sugar chain, comprising: a solid phase for fixing a glycoprotein; a container for isolating and labeling a sugar chain by holding the solid phase; and a sugar chain-isolating enzyme, wherein the solid phase includes a ligand selected from the group consisting of protein A, protein G, protein L, protein H, protein D, and protein Arp, in the surface thereof. 35. A kit for preparing a glycoprotein sugar chain, comprising: a solid phase for fixing a glycoprotein; a container for isolating and labeling a sugar chain by holding the solid phase; a deglycosylation promoter which contains an acid-derived anionic surfactant; and a sugar chain-isolating enzyme. 36. A device for preparing a glycoprotein sugar chain comprising: a container holding portion which holds a container in which a sample that contains a glycoprotein fixed to a solid phase is accommodated; and a reagent introducing unit which introduces a reagent into the container, wherein the glycoprotein is an antibody, and the solid phase includes a ligand selected from the group consisting of protein A, protein G, protein L, protein H, protein D, and protein Alp, in the surface thereof, and the reagent introducing unit includes a sugar chain-isolating enzyme introducing unit which introduces a sugar chain-isolating enzyme into the container and a labeling reagent introducing unit which introduces a labeling reagent into the container. 37. The device for preparing a glycoprotein sugar chain according to claim 36, further comprising: a solid-liquid separating unit which performs solid-liquid separation on the contents in the container. 38. A device for preparing a glycoprotein sugar chain comprising: a container holding portion which holds a container in which a sample that contains a glycoprotein fixed to a solid phase is accommodated; and a reagent introducing unit which introduces a reagent into the container, wherein the reagent introducing unit includes a sugar chain-isolating enzyme introducing unit which introduces a sugar chain-isolating enzyme into the container, a labeling reagent introducing unit which introduces a labeling reagent into the container, and a deglycosylation promoter introducing unit which introduces a deglycosylation promoter which contains an acid-derived anionic surfactant into the container.
Provided is a method for preparing a glycoprotein sugar chain including: an isolation step of acting a sugar chain-isolating enzyme on a sample which contains a glycoprotein fixed to a solid phase in a container to obtain an isolated product which contains a sugar chain; and a labeling step of adding a labeling reagent to the isolated product in the container to obtain a labeled product which contains a labeled substance of the sugar chain.1-22. (canceled) 23. A method for preparing a glycoprotein sugar chain, comprising: an isolation step of acting a sugar chain-isolating enzyme on a sample which contains a glycoprotein fixed to a solid phase in a container to obtain an isolated product which contains a sugar chain; and a labeling step of adding a labeling reagent to the isolated product in the container to obtain a labeled product which contains a labeled substance of the sugar chain, wherein the glycoprotein is an antibody, and the solid phase includes a ligand selected from the group consisting of protein A, protein G, protein L, protein H, protein D, and protein Arp, in the surface thereof. 24. The method for preparing a glycoprotein sugar chain according to claim 23, wherein the isolation step is performed in an open system and under heating conditions. 25. The method of preparing a glycoprotein sugar chain according to claim 23, wherein the labeling reagent contains 2-aminobenzamide, a reducing agent, and a solvent. 26. The method of preparing a glycoprotein sugar chain according to claim 25, wherein the reducing agent is picoline borane. 27. The method of preparing a glycoprotein sugar chain according to claim 25, wherein the solvent contains a protic compound. 28. The method of preparing a glycoprotein sugar chain according to claim 27, wherein the solvent further contains an aprotic compound having a boiling point higher than that of the protic compound. 29. The method of preparing a glycoprotein sugar chain according to claim 23, further comprising: a separation step of performing solid-liquid separation after the isolation step to obtain a separate liquid which contains the isolated product. 30. The method of preparing a glycoprotein sugar chain according to claim 23, further comprising: a separation step of performing solid-liquid separation after the labeling step to obtain a separate liquid which contains the labeled substance of the sugar chain. 31. A method for preparing a glycoprotein sugar chain, comprising: an isolation step of acting a sugar chain-isolating enzyme on a sample which contains a glycoprotein fixed to a solid phase in a container to obtain an isolated product which contains a sugar chain; and a labeling step of adding a labeling reagent to the isolated product in the container to obtain a labeled product which contains a labeled substance of the sugar chain, wherein wherein the isolation step is performed in the presence of a deglycosylation promoter containing an acid-derived anionic surfactant. 32. The method for preparing a glycoprotein sugar chain according to claim 31, further comprising: a pre-treatment step of bringing a pre-treatment agent containing a surfactant into contact with the sample before the isolation step. 33. The method for preparing a glycoprotein sugar chain according to claim 31, wherein the acid-derived anionic surfactant is selected from the group consisting of a carboxylic acid type anionic surfactant, a sulfonic acid type anionic surfactant, a sulfuric acid ester type anionic surfactant, and a phosphoric acid ester type anionic surfactant. 34. A kit for preparing a glycoprotein sugar chain, comprising: a solid phase for fixing a glycoprotein; a container for isolating and labeling a sugar chain by holding the solid phase; and a sugar chain-isolating enzyme, wherein the solid phase includes a ligand selected from the group consisting of protein A, protein G, protein L, protein H, protein D, and protein Arp, in the surface thereof. 35. A kit for preparing a glycoprotein sugar chain, comprising: a solid phase for fixing a glycoprotein; a container for isolating and labeling a sugar chain by holding the solid phase; a deglycosylation promoter which contains an acid-derived anionic surfactant; and a sugar chain-isolating enzyme. 36. A device for preparing a glycoprotein sugar chain comprising: a container holding portion which holds a container in which a sample that contains a glycoprotein fixed to a solid phase is accommodated; and a reagent introducing unit which introduces a reagent into the container, wherein the glycoprotein is an antibody, and the solid phase includes a ligand selected from the group consisting of protein A, protein G, protein L, protein H, protein D, and protein Alp, in the surface thereof, and the reagent introducing unit includes a sugar chain-isolating enzyme introducing unit which introduces a sugar chain-isolating enzyme into the container and a labeling reagent introducing unit which introduces a labeling reagent into the container. 37. The device for preparing a glycoprotein sugar chain according to claim 36, further comprising: a solid-liquid separating unit which performs solid-liquid separation on the contents in the container. 38. A device for preparing a glycoprotein sugar chain comprising: a container holding portion which holds a container in which a sample that contains a glycoprotein fixed to a solid phase is accommodated; and a reagent introducing unit which introduces a reagent into the container, wherein the reagent introducing unit includes a sugar chain-isolating enzyme introducing unit which introduces a sugar chain-isolating enzyme into the container, a labeling reagent introducing unit which introduces a labeling reagent into the container, and a deglycosylation promoter introducing unit which introduces a deglycosylation promoter which contains an acid-derived anionic surfactant into the container.
1,600
305
13,320,629
1,643
The invention relates to modulating the SIRPα-CD47 interaction in order to treat hematological cancer and compounds therefor. In some embodiments, there is provided methods and uses of SIRPα polypeptides, fragments and fusion proteins for treating hematological cancer, preferably human acute myeloid leukemia.
1. A method for treating cancer cells or tumours that are CD47+, comprising modulating the interaction between human Sirpα and human CD47. 2. The method of claim 1, wherein the interaction between human Sirpα and human CD47 is modulated by administering a therapeutically effective amount of a polypeptide capable of binding to the extracellular domain of human CD47. 3. The method of claim 2, wherein the polypeptide comprises soluble human Sirpα, or a CD47-binding fragment thereof. 4. The method of claim 3, wherein the polypeptide is the extracellular domain of human Sirpα. 5. The method of claim 3, wherein the polypeptide is fused to a second protein. 6. The method of claim 5, wherein the second protein is the Fc portion of IgG. 7. The method of claim 6, wherein the resulting fusion protein is SEQ ID NO. 13. 8. The method of claim 1, wherein the modulation results in desupression of macrophages. 9. The method of claim 1 wherein the cancer cells or tumour are associated with a hematological cancer. 10. The method of claim 9, wherein the hematological cancer is a leukemia selected from acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, myeloproliferative disorder/neoplasm and myelodysplastic syndrome. 11. The method of claim 10, wherein the leukemia is human acute myeloid leukemia. 12. The method of claim 9, wherein the hematological cancer is a lymphoma or myeloma selected from Hodgkin's lymphoma, both indolent and aggressive non-Hodgkin's lymphoma, Burkitt's lymphoma, follicular lymphoma (small cell and large cell), multiple myeloma (MM), giant cell myeloma, heavy-chain myeloma, and light chain or Bence-Jones myeloma. 13-24. (canceled) 25. A method of determining genetic polymorphisms in humans affecting survival to a cancer with cancer cells or tumours that are CD47+, comprising: a. sequencing the Sirpα gene from a plurality of humans having the cancer; b. determining nucleotide differences in the Sirpα gene within the plurality of humans; and c. correlating the nucleotide differences with survival to determine relevant polymorphisms. 26. The method of claim 25, wherein the nucleotide differences result in amino acid differences. 27. A method of prognosing likelihood of survival to a cancer with cancer cells or tumours that are CD47+, comprising: a. sequencing the Sirpα gene from the recipient; and b. determining whether the relevant polymorphisms of claim 23 exist. 28. The method of claim 27, wherein the nucleotide differences result in amino acid differences. 29. The method of claim 28, wherein the amino acid differences is at least one of: a. replacement of at least one of residues at positions 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 2 with corresponding residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1; or b. deletion of at least one of residues 129 and 130 of SEQ ID NO. 2. 30. The method of claim 2, wherein the polypeptide is selected from the group consisting of: a. a polypeptide consisting of the amino acid sequence of SEQ ID NO. 1; b. a polypeptide consisting of a CD47-binding fragment of the amino acid sequence of SEQ ID NO. 1, wherein the fragment comprises at least one of residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1; and c. a CD47-binding variant of one of the polypeptide in a) and b) with up to 1 amino acid insertion, deletion or substitution for every 7 amino acids in length of the polypeptide, wherein the polypeptide comprises at least one of residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1. 31. The method of claim 2, wherein the polypeptide is selected from the group consisting of: a) a polypeptide consisting of the amino acid sequence of SEQ ID NO. 2; b) a polypeptide consisting of a CD47-binding fragment of the amino acid sequence of SEQ ID NO. 2; and c) a CD47-binding variant of one of the polypeptide in a) and b) with up to 1 amino acid insertion, deletion or substitution for every 7 amino acids in length of the polypeptide; wherein: i. at least one of residues at positions 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 2 in the polypeptide is replaced with corresponding residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1; or ii. at least one of residues 129 and 130 of SEQ ID NO. 2 in the polypeptide is deleted. 32. The method of claim 2, wherein the polypeptide is selected from the group consisting of: a. a polypeptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NOS. 4-7; b. a polypeptide consisting of a CD47-binding fragment of an amino acid sequence selected from the group consisting of SEQ ID NOS. 4-7; and c. a CD47-binding variant of one of the polypeptide in a) and b) with up to 1 amino acid insertion, deletion or substitution for every 7 amino acids in length of the polypeptide. 33-53. (canceled)
The invention relates to modulating the SIRPα-CD47 interaction in order to treat hematological cancer and compounds therefor. In some embodiments, there is provided methods and uses of SIRPα polypeptides, fragments and fusion proteins for treating hematological cancer, preferably human acute myeloid leukemia.1. A method for treating cancer cells or tumours that are CD47+, comprising modulating the interaction between human Sirpα and human CD47. 2. The method of claim 1, wherein the interaction between human Sirpα and human CD47 is modulated by administering a therapeutically effective amount of a polypeptide capable of binding to the extracellular domain of human CD47. 3. The method of claim 2, wherein the polypeptide comprises soluble human Sirpα, or a CD47-binding fragment thereof. 4. The method of claim 3, wherein the polypeptide is the extracellular domain of human Sirpα. 5. The method of claim 3, wherein the polypeptide is fused to a second protein. 6. The method of claim 5, wherein the second protein is the Fc portion of IgG. 7. The method of claim 6, wherein the resulting fusion protein is SEQ ID NO. 13. 8. The method of claim 1, wherein the modulation results in desupression of macrophages. 9. The method of claim 1 wherein the cancer cells or tumour are associated with a hematological cancer. 10. The method of claim 9, wherein the hematological cancer is a leukemia selected from acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, myeloproliferative disorder/neoplasm and myelodysplastic syndrome. 11. The method of claim 10, wherein the leukemia is human acute myeloid leukemia. 12. The method of claim 9, wherein the hematological cancer is a lymphoma or myeloma selected from Hodgkin's lymphoma, both indolent and aggressive non-Hodgkin's lymphoma, Burkitt's lymphoma, follicular lymphoma (small cell and large cell), multiple myeloma (MM), giant cell myeloma, heavy-chain myeloma, and light chain or Bence-Jones myeloma. 13-24. (canceled) 25. A method of determining genetic polymorphisms in humans affecting survival to a cancer with cancer cells or tumours that are CD47+, comprising: a. sequencing the Sirpα gene from a plurality of humans having the cancer; b. determining nucleotide differences in the Sirpα gene within the plurality of humans; and c. correlating the nucleotide differences with survival to determine relevant polymorphisms. 26. The method of claim 25, wherein the nucleotide differences result in amino acid differences. 27. A method of prognosing likelihood of survival to a cancer with cancer cells or tumours that are CD47+, comprising: a. sequencing the Sirpα gene from the recipient; and b. determining whether the relevant polymorphisms of claim 23 exist. 28. The method of claim 27, wherein the nucleotide differences result in amino acid differences. 29. The method of claim 28, wherein the amino acid differences is at least one of: a. replacement of at least one of residues at positions 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 2 with corresponding residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1; or b. deletion of at least one of residues 129 and 130 of SEQ ID NO. 2. 30. The method of claim 2, wherein the polypeptide is selected from the group consisting of: a. a polypeptide consisting of the amino acid sequence of SEQ ID NO. 1; b. a polypeptide consisting of a CD47-binding fragment of the amino acid sequence of SEQ ID NO. 1, wherein the fragment comprises at least one of residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1; and c. a CD47-binding variant of one of the polypeptide in a) and b) with up to 1 amino acid insertion, deletion or substitution for every 7 amino acids in length of the polypeptide, wherein the polypeptide comprises at least one of residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1. 31. The method of claim 2, wherein the polypeptide is selected from the group consisting of: a) a polypeptide consisting of the amino acid sequence of SEQ ID NO. 2; b) a polypeptide consisting of a CD47-binding fragment of the amino acid sequence of SEQ ID NO. 2; and c) a CD47-binding variant of one of the polypeptide in a) and b) with up to 1 amino acid insertion, deletion or substitution for every 7 amino acids in length of the polypeptide; wherein: i. at least one of residues at positions 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 2 in the polypeptide is replaced with corresponding residues 31, 32, 34, 37, 74, 77, 83, 84, 86, 87, 90, 91, 96, 100, 102, 114, 118, 126 of SEQ ID NO. 1; or ii. at least one of residues 129 and 130 of SEQ ID NO. 2 in the polypeptide is deleted. 32. The method of claim 2, wherein the polypeptide is selected from the group consisting of: a. a polypeptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NOS. 4-7; b. a polypeptide consisting of a CD47-binding fragment of an amino acid sequence selected from the group consisting of SEQ ID NOS. 4-7; and c. a CD47-binding variant of one of the polypeptide in a) and b) with up to 1 amino acid insertion, deletion or substitution for every 7 amino acids in length of the polypeptide. 33-53. (canceled)
1,600
306
14,672,789
1,641
Apparatus is disclosed for identifying at least a first target condition in a human or animal body. The apparatus comprises one or more test portions for identifying a first analyte in a biological sample from the body, the first analyte providing a marker of the first target condition, and a second analyte in the biological sample, the second analyte being different from the first analyte. The apparatus is configured to identify the first target condition in the body based on the identification of both the first and second analytes. In one embodiment, the first target condition is pregnancy, the first analyte is human chorionic gonadotropin (hCG) and the second analyte is luteinizing hormone (LH).
1. A pregnancy test device for identifying pregnancy in a human or animal body, the device comprising: one or more test portions for identifying: human chorionic gonadotropin (hCG) in the biological sample; and luteinizing hormone (LH) in the biological sample; and a processor adapted to: identify a level of the hCG in the biological sample; identify a level of the LH in the biological sample; determine which of a plurality of discrete LH ranges the identified level of LH falls within, wherein a different hCG threshold level is associated with each one of the LH ranges; select the hCG threshold level that is associated with the LH range in which the identified level of LH falls within; identify pregnancy in the body if the identified level of hCG is above the selected hCG threshold level. 2. The pregnancy test device of claim 1, wherein the plurality of discrete LH ranges comprises first and second LH ranges defined by a first LH threshold level; wherein: the first LH range includes all LH values below the first LH threshold level; and the second LH range includes all LH values above the first LH threshold level; and wherein: a first hCG threshold level is associated with the first LH range; and a second hCG threshold level is associated with the second LH range. 3. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.0 and 2.0 IU/L. 3. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.3 and 1.9 IU/L. 4. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.3 and 1.8 IU/L. 5. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.3 and 1.7 IU/L. 6. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.4 and 1.6 IU/L. 7. The pregnancy test device of claim 2, wherein the first hCG threshold level is about 1.5 IU/L. 8. The pregnancy test device of claim 2, wherein the difference between the first and second hCG threshold levels is at least 5 IU/L. 9. The pregnancy test device of claim 2, wherein the difference between the first and second hCG threshold levels is at least 10 IU/L. 10. The pregnancy test device of claim 2, wherein the difference between the first and second hCG threshold levels is at least 15 IU/L. 11. The pregnancy test device of claim 2, wherein the first LH threshold level is greater than 5 IU/L. 12. The pregnancy test device of claim 2, wherein the first LH threshold level is greater than 10 IU/L. 13. The pregnancy test device of claim 2, wherein the first LH threshold level is greater than 15 IU/L. 14. The pregnancy test device of claim 2, wherein the first LH threshold level is about 20 IU/L. 15. The pregnancy test device of claim 2, wherein: the first LH threshold level is greater than 5 IU/L; and the first hCG threshold level is between 1.0 and 2.0 IU/L. 16. The pregnancy test device of claim 2, wherein: the first LH threshold level is greater than 10 IU/L; and the first hCG threshold level is between 1.3 and 2.0 IU/L. 17. The pregnancy test device of claim 2, wherein: the first LH threshold level is greater than 15 IU/L; and the first hCG threshold level is between 1.4 and 2.0 IU/L. 18. The pregnancy test device of claim 2, wherein: the first LH threshold level is about 20 IU/L; and the first hCG threshold level is about 1.5 IU/L. 19. The pregnancy test device of claim 1, wherein the plurality of discrete LH ranges comprises first, second and third LH ranges defined by a first LH threshold level and a second LH threshold level, the second LH threshold level being higher than the first LH threshold level; wherein: the first LH range includes all LH values below the first LH threshold level; the second LH range includes all LH values between the first LH threshold level and the second LH threshold level; and the third LH range includes all LH values above the second LH threshold level; and wherein: a first hCG threshold level is associated with the first LH range; a second hCG threshold level is associated with the second LH range; and a third hCG threshold level is associated with the third LH range. 20. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.0 and 2.0 IU/L. 21. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.3 and 1.9 IU/L. 22. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.3 and 1.8 IU/L. 23. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.3 and 1.7 IU/L. 24. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.4 and 1.6 IU/L. 25. The pregnancy test device of claim 19, wherein the first hCG threshold level is about 1.5 IU/L. 26. The pregnancy test device of claim 19, wherein the difference between the first and second hCG threshold levels is at least 2 IU/L. 27. The pregnancy test device of claim 19, wherein the difference between the first and second hCG threshold levels is at least 3 IU/L. 28. The pregnancy test device of claim 19, wherein the difference between the first and second hCG threshold levels is at least 5 IU/L. 29. The pregnancy test device of claim 19, wherein the difference between the second and third hCG threshold levels is at least 5 IU/L. 30. The pregnancy test device of claim 19, wherein the difference between the second and third hCG threshold levels is at least 10 IU/L. 31. The pregnancy test device of claim 19, wherein the difference between the second and third hCG threshold levels is at least 15 IU/L. 32. The pregnancy test device of claim 19, wherein the first LH threshold level is greater than 5 IU/L. 33. The pregnancy test device of claim 19, wherein the first LH threshold level is greater than 10 IU/L. 34. The pregnancy test device of claim 19, wherein the first LH threshold level is greater than 15 IU/L. 35. The pregnancy test device of claim 19, wherein the first LH threshold level is about 15 IU/L. 36. The pregnancy test device of claim 19, wherein the second LH threshold level is greater than 20 IU/L. 37. The pregnancy test device of claim 19, wherein the second LH threshold level is greater than 25 IU/L. 38. The pregnancy test device of claim 19, wherein the second LH threshold level is greater than 30 IU/L. 39. The pregnancy test device of claim 19, wherein the second LH threshold level is about 30 IU/L. 40. The pregnancy test device of claim 19, wherein: the first LH threshold level is between 10 IU/L and 20 IU/L; the second LH threshold level is between 20 IU/L and 40 IU/L; and the first hCG threshold level is between 1.0 and 2.0 IU/L. 41. The pregnancy test device of claim 19, wherein: the first LH threshold level is between 10 IU/L and 20 IU/L; the second LH threshold level is between 20 IU/L and 40 IU/L; and the first hCG threshold level is between 1.0 and 2.0 IU/L; the second hCG threshold level is between 5 and 10 IU/L; and the third HCG threshold level is at least 20 IU/L. 42. The pregnancy test device of claim 19, wherein: the first LH threshold level is about 15 IU/L; the second LH threshold level is about 30 IU/L; the first hCG threshold level is about 1.5 IU/L the second hCG threshold level is about 5 IU/L; and the third hCG threshold level is about 20 IU/L.
Apparatus is disclosed for identifying at least a first target condition in a human or animal body. The apparatus comprises one or more test portions for identifying a first analyte in a biological sample from the body, the first analyte providing a marker of the first target condition, and a second analyte in the biological sample, the second analyte being different from the first analyte. The apparatus is configured to identify the first target condition in the body based on the identification of both the first and second analytes. In one embodiment, the first target condition is pregnancy, the first analyte is human chorionic gonadotropin (hCG) and the second analyte is luteinizing hormone (LH).1. A pregnancy test device for identifying pregnancy in a human or animal body, the device comprising: one or more test portions for identifying: human chorionic gonadotropin (hCG) in the biological sample; and luteinizing hormone (LH) in the biological sample; and a processor adapted to: identify a level of the hCG in the biological sample; identify a level of the LH in the biological sample; determine which of a plurality of discrete LH ranges the identified level of LH falls within, wherein a different hCG threshold level is associated with each one of the LH ranges; select the hCG threshold level that is associated with the LH range in which the identified level of LH falls within; identify pregnancy in the body if the identified level of hCG is above the selected hCG threshold level. 2. The pregnancy test device of claim 1, wherein the plurality of discrete LH ranges comprises first and second LH ranges defined by a first LH threshold level; wherein: the first LH range includes all LH values below the first LH threshold level; and the second LH range includes all LH values above the first LH threshold level; and wherein: a first hCG threshold level is associated with the first LH range; and a second hCG threshold level is associated with the second LH range. 3. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.0 and 2.0 IU/L. 3. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.3 and 1.9 IU/L. 4. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.3 and 1.8 IU/L. 5. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.3 and 1.7 IU/L. 6. The pregnancy test device of claim 2, wherein the first hCG threshold level is between 1.4 and 1.6 IU/L. 7. The pregnancy test device of claim 2, wherein the first hCG threshold level is about 1.5 IU/L. 8. The pregnancy test device of claim 2, wherein the difference between the first and second hCG threshold levels is at least 5 IU/L. 9. The pregnancy test device of claim 2, wherein the difference between the first and second hCG threshold levels is at least 10 IU/L. 10. The pregnancy test device of claim 2, wherein the difference between the first and second hCG threshold levels is at least 15 IU/L. 11. The pregnancy test device of claim 2, wherein the first LH threshold level is greater than 5 IU/L. 12. The pregnancy test device of claim 2, wherein the first LH threshold level is greater than 10 IU/L. 13. The pregnancy test device of claim 2, wherein the first LH threshold level is greater than 15 IU/L. 14. The pregnancy test device of claim 2, wherein the first LH threshold level is about 20 IU/L. 15. The pregnancy test device of claim 2, wherein: the first LH threshold level is greater than 5 IU/L; and the first hCG threshold level is between 1.0 and 2.0 IU/L. 16. The pregnancy test device of claim 2, wherein: the first LH threshold level is greater than 10 IU/L; and the first hCG threshold level is between 1.3 and 2.0 IU/L. 17. The pregnancy test device of claim 2, wherein: the first LH threshold level is greater than 15 IU/L; and the first hCG threshold level is between 1.4 and 2.0 IU/L. 18. The pregnancy test device of claim 2, wherein: the first LH threshold level is about 20 IU/L; and the first hCG threshold level is about 1.5 IU/L. 19. The pregnancy test device of claim 1, wherein the plurality of discrete LH ranges comprises first, second and third LH ranges defined by a first LH threshold level and a second LH threshold level, the second LH threshold level being higher than the first LH threshold level; wherein: the first LH range includes all LH values below the first LH threshold level; the second LH range includes all LH values between the first LH threshold level and the second LH threshold level; and the third LH range includes all LH values above the second LH threshold level; and wherein: a first hCG threshold level is associated with the first LH range; a second hCG threshold level is associated with the second LH range; and a third hCG threshold level is associated with the third LH range. 20. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.0 and 2.0 IU/L. 21. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.3 and 1.9 IU/L. 22. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.3 and 1.8 IU/L. 23. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.3 and 1.7 IU/L. 24. The pregnancy test device of claim 19, wherein the first hCG threshold level is between 1.4 and 1.6 IU/L. 25. The pregnancy test device of claim 19, wherein the first hCG threshold level is about 1.5 IU/L. 26. The pregnancy test device of claim 19, wherein the difference between the first and second hCG threshold levels is at least 2 IU/L. 27. The pregnancy test device of claim 19, wherein the difference between the first and second hCG threshold levels is at least 3 IU/L. 28. The pregnancy test device of claim 19, wherein the difference between the first and second hCG threshold levels is at least 5 IU/L. 29. The pregnancy test device of claim 19, wherein the difference between the second and third hCG threshold levels is at least 5 IU/L. 30. The pregnancy test device of claim 19, wherein the difference between the second and third hCG threshold levels is at least 10 IU/L. 31. The pregnancy test device of claim 19, wherein the difference between the second and third hCG threshold levels is at least 15 IU/L. 32. The pregnancy test device of claim 19, wherein the first LH threshold level is greater than 5 IU/L. 33. The pregnancy test device of claim 19, wherein the first LH threshold level is greater than 10 IU/L. 34. The pregnancy test device of claim 19, wherein the first LH threshold level is greater than 15 IU/L. 35. The pregnancy test device of claim 19, wherein the first LH threshold level is about 15 IU/L. 36. The pregnancy test device of claim 19, wherein the second LH threshold level is greater than 20 IU/L. 37. The pregnancy test device of claim 19, wherein the second LH threshold level is greater than 25 IU/L. 38. The pregnancy test device of claim 19, wherein the second LH threshold level is greater than 30 IU/L. 39. The pregnancy test device of claim 19, wherein the second LH threshold level is about 30 IU/L. 40. The pregnancy test device of claim 19, wherein: the first LH threshold level is between 10 IU/L and 20 IU/L; the second LH threshold level is between 20 IU/L and 40 IU/L; and the first hCG threshold level is between 1.0 and 2.0 IU/L. 41. The pregnancy test device of claim 19, wherein: the first LH threshold level is between 10 IU/L and 20 IU/L; the second LH threshold level is between 20 IU/L and 40 IU/L; and the first hCG threshold level is between 1.0 and 2.0 IU/L; the second hCG threshold level is between 5 and 10 IU/L; and the third HCG threshold level is at least 20 IU/L. 42. The pregnancy test device of claim 19, wherein: the first LH threshold level is about 15 IU/L; the second LH threshold level is about 30 IU/L; the first hCG threshold level is about 1.5 IU/L the second hCG threshold level is about 5 IU/L; and the third hCG threshold level is about 20 IU/L.
1,600
307
14,863,600
1,619
A composition for applying to skin in need of treatment for skin barrier restoration, hydration and moisturization, the composition comprising extracts of Ampelopsis grossedentata and Albizia julibrissin . Also provided are methods for treating skin in need of skin barrier treatment, methods of treating skin to reduce the signs of aging and methods of treating skin to reduce the signs of inflammation with a composition comprising extracts of Ampelopsis grossedentata and Albizia julibrissin.
1. A method of improving the barrier function of skin, said method comprising applying to the skin a topical composition comprising an extract of Ampelopsis grossedentata, an extract of Albizia julibrissin and a topical carrier. 2. The method of claim 1, wherein the extracts are polar extracts prepared using polar solvents selected from the group of methanol, ethanol, isopropyl alcohol, n-butanol, propylene glycol, water, and mixtures thereof. 3. The method of claim 1, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:10 to 10:1. 4. The method claim 2, wherein the polar extract of Ampelopsis grossedentata is a leaf extract. 5. The method of claim 2, wherein the polar extract of Albizia julibrissin is a flower extract. 6. The method of claim 1 wherein the composition further comprises a material selected from the group consisting of surfactants, chelating agents, emollients, humectants, conditioners, preservatives, opacifiers, fragrances, and combinations of two or more thereof. 7. The method of claim 1 wherein said composition is a skin care composition in a form selected from the group consisting of lotions, creams, serums, gels, sticks, sprays, ointments, liquid washes, soap bars, shampoos, hair conditioners, pastes, foams, powders, mousses, shaving creams, hydrogels, film-forming products, fluid on wipes, fluid on facial masks, and combinations of two or more thereof. 8. The method of claim 2, wherein the polar extract of Ampelopsis grossedentata is made by extracting with ethanol. 9. The method of claim 2, wherein the polar extract of Albizia julibrissin is made by extracting with ethanol. 10. The method of claim 2, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Ampelopsis grossedentata. 11. The method of claim 2, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Albizia julibrissin. 12. The method of claim 3, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:7 to 7:1. 13. The method of claim 12, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:5 to 5:1. 14. The method of claim 13, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is 1:1. 15. The method of claim 1, wherein the total amount of the combination of the Ampelopsis grossedentata extract and the Albizia julibrissin extract is from about 0.0005% to about 30% by weight of the composition. 16. The method of claim 1, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 17. The method of claim 16, wherein the amount of the Ampelopsis grossedentata extract is from about 0.01% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.01% to about 5% by weight of the composition. 18. The method of claim 1, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 19. The method of claim 1, wherein the amount of the Ampelopsis grossedentata extract is from about 0.1% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.1% to about 5% by weight of the composition. 20. A method for moisturizing the skin, said method comprising applying to the skin a topical composition comprising a polar extract of Ampelopsis grossedentata, a polar extract of Albizia julibrissin and a topical carrier. 21. The method of claim 20, wherein the extracts are polar extracts prepared using polar solvents selected from the group of methanol, ethanol, isopropyl alcohol, n-butanol, propylene glycol, water, and mixtures thereof. 22. The method of claim 20, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:10 to 10:1. 23. The method claim 21, wherein the polar extract of Ampelopsis grossedentata is a leaf extract. 24. The method of claim 21, wherein the polar extract of Albizia julibrissin is a flower extract. 25. The method of claim 20 wherein the composition further comprises a material selected from the group consisting of surfactants, chelating agents, emollients, humectants, conditioners, preservatives, opaciflers, fragrances, and combinations of two or more thereof. 26. The method of claim 20 wherein said composition is a skin care composition in a form selected from the group consisting of lotions, creams, serums, gels, sticks, sprays, ointments, liquid washes, soap bars, shampoos, hair conditioners, pastes, foams, powders, mousses, shaving creams, hydrogels, film-forming products, fluid on wipes, fluid on facial masks, and combinations of two or more thereof. 27. The method of claim 21, wherein the polar extract of Ampelopsis grossedentata is made by extracting with ethanol. 28. The method of claim 21, wherein the polar extract of Albizia julibrissin is made by extracting with ethanol. 29. The method of claim 21, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Ampelopsis grossedentata. 30. The method of claim 21, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Albizia julibrissin. 31. The method of claim 22, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:7 to 7:1. 32. The method of claim 31, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:5 to 5:1. 33. The method of claim 32, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is 1:1. 34. The method of claim 20, wherein the total amount of the combination of the Ampelopsis grossedentata extract and the Albizia julibrissin extract is from about 0.0005% to about 30% by weight of the composition. 35. The method of claim 20, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 36. The method of claim 35, wherein the amount of the Ampelopsis grossedentata extract is from about 0.01% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.01% to about 5% by weight of the composition. 37. The method of claim 20, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 38. The method of claim 20, wherein the amount of the Ampelopsis grossedentata extract is from about 0.1% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.1% to about 5% by weight of the composition.
A composition for applying to skin in need of treatment for skin barrier restoration, hydration and moisturization, the composition comprising extracts of Ampelopsis grossedentata and Albizia julibrissin . Also provided are methods for treating skin in need of skin barrier treatment, methods of treating skin to reduce the signs of aging and methods of treating skin to reduce the signs of inflammation with a composition comprising extracts of Ampelopsis grossedentata and Albizia julibrissin.1. A method of improving the barrier function of skin, said method comprising applying to the skin a topical composition comprising an extract of Ampelopsis grossedentata, an extract of Albizia julibrissin and a topical carrier. 2. The method of claim 1, wherein the extracts are polar extracts prepared using polar solvents selected from the group of methanol, ethanol, isopropyl alcohol, n-butanol, propylene glycol, water, and mixtures thereof. 3. The method of claim 1, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:10 to 10:1. 4. The method claim 2, wherein the polar extract of Ampelopsis grossedentata is a leaf extract. 5. The method of claim 2, wherein the polar extract of Albizia julibrissin is a flower extract. 6. The method of claim 1 wherein the composition further comprises a material selected from the group consisting of surfactants, chelating agents, emollients, humectants, conditioners, preservatives, opacifiers, fragrances, and combinations of two or more thereof. 7. The method of claim 1 wherein said composition is a skin care composition in a form selected from the group consisting of lotions, creams, serums, gels, sticks, sprays, ointments, liquid washes, soap bars, shampoos, hair conditioners, pastes, foams, powders, mousses, shaving creams, hydrogels, film-forming products, fluid on wipes, fluid on facial masks, and combinations of two or more thereof. 8. The method of claim 2, wherein the polar extract of Ampelopsis grossedentata is made by extracting with ethanol. 9. The method of claim 2, wherein the polar extract of Albizia julibrissin is made by extracting with ethanol. 10. The method of claim 2, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Ampelopsis grossedentata. 11. The method of claim 2, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Albizia julibrissin. 12. The method of claim 3, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:7 to 7:1. 13. The method of claim 12, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:5 to 5:1. 14. The method of claim 13, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is 1:1. 15. The method of claim 1, wherein the total amount of the combination of the Ampelopsis grossedentata extract and the Albizia julibrissin extract is from about 0.0005% to about 30% by weight of the composition. 16. The method of claim 1, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 17. The method of claim 16, wherein the amount of the Ampelopsis grossedentata extract is from about 0.01% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.01% to about 5% by weight of the composition. 18. The method of claim 1, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 19. The method of claim 1, wherein the amount of the Ampelopsis grossedentata extract is from about 0.1% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.1% to about 5% by weight of the composition. 20. A method for moisturizing the skin, said method comprising applying to the skin a topical composition comprising a polar extract of Ampelopsis grossedentata, a polar extract of Albizia julibrissin and a topical carrier. 21. The method of claim 20, wherein the extracts are polar extracts prepared using polar solvents selected from the group of methanol, ethanol, isopropyl alcohol, n-butanol, propylene glycol, water, and mixtures thereof. 22. The method of claim 20, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:10 to 10:1. 23. The method claim 21, wherein the polar extract of Ampelopsis grossedentata is a leaf extract. 24. The method of claim 21, wherein the polar extract of Albizia julibrissin is a flower extract. 25. The method of claim 20 wherein the composition further comprises a material selected from the group consisting of surfactants, chelating agents, emollients, humectants, conditioners, preservatives, opaciflers, fragrances, and combinations of two or more thereof. 26. The method of claim 20 wherein said composition is a skin care composition in a form selected from the group consisting of lotions, creams, serums, gels, sticks, sprays, ointments, liquid washes, soap bars, shampoos, hair conditioners, pastes, foams, powders, mousses, shaving creams, hydrogels, film-forming products, fluid on wipes, fluid on facial masks, and combinations of two or more thereof. 27. The method of claim 21, wherein the polar extract of Ampelopsis grossedentata is made by extracting with ethanol. 28. The method of claim 21, wherein the polar extract of Albizia julibrissin is made by extracting with ethanol. 29. The method of claim 21, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Ampelopsis grossedentata. 30. The method of claim 21, wherein said composition comprises from about 0.01% to about 2% of the polar extract of Albizia julibrissin. 31. The method of claim 22, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:7 to 7:1. 32. The method of claim 31, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is between 1:5 to 5:1. 33. The method of claim 32, wherein the weight ratio in the composition of the extracts of Ampelopsis grossedentata to Albizia julibrissin is 1:1. 34. The method of claim 20, wherein the total amount of the combination of the Ampelopsis grossedentata extract and the Albizia julibrissin extract is from about 0.0005% to about 30% by weight of the composition. 35. The method of claim 20, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 36. The method of claim 35, wherein the amount of the Ampelopsis grossedentata extract is from about 0.01% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.01% to about 5% by weight of the composition. 37. The method of claim 20, wherein the amount of the Ampelopsis grossedentata extract is from about 0.001% to about 10% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.001% to about 10% by weight of the composition. 38. The method of claim 20, wherein the amount of the Ampelopsis grossedentata extract is from about 0.1% to about 5% by weight of the composition and the amount of Albizia julibrissin extract is from about 0.1% to about 5% by weight of the composition.
1,600
308
15,213,444
1,625
The invention relates to compounds of formula (I) where R 1 , R 2 , R 3 , R 4 are each independently selected from —(C 1 -C 12 )-alkyl, —(C 3 -C 12 )-cycloalkyl, —(C 3 -C 12 )-heterocycloalkyl, —(C 6 -C 20 )-aryl, —(C 3 -C 20 )-heteroaryl; at least one of the R 1 , R 2 , R 3 , R 4 radicals is a —(C 3 -C 20 )-heteroaryl radical; and R 1 , R 2 , R 3 , R 4 , if they are —(C 1 -C 12 )-alkyl, —(C 3 -C 12 )-cycloalkyl, —(C 3 -C 12 )-heterocycloalkyl, —(C 6 -C 20 )-aryl or —(C 3 -C 20 )-heteroaryl, may each independently be substituted by one or more substituents selected from —(C 1 -C 12 )-alkyl, —(C 3 -C 12 )-cycloalkyl, —(C 3 -C 12 )-heterocycloalkyl, —O—(C 1 -C 12 )-alkyl, —O—(C 1 -C 12 )-alkyl-(C 6 -C 20 )-aryl, —O—(C 3 -C 12 )-cycloalkyl, —S—(C 1 -C 12 )-alkyl, —S—(C 3 -C 12 )-cycloalkyl, —COO—(C 1 -C 12 )-alkyl, —COO—(C 3 -C 12 )-cycloalkyl, —CONH—(C 1 -C 12 )-alkyl, —CONH—(C 3 -C 12 )-cycloalkyl, —CO—(C 1 -C 12 )-alkyl, —CO—(C 3 -C 12 )-cycloalkyl, —N—[(C 1 -C 12 )-alkyl] 2 , —(C 6 -C 20 )-aryl, —(C 6 -C 20 )-aryl-(C 1 -C 12 )-alkyl, —(C 6 -C 20 )-aryl-O—(C 1 -C 12 )-alkyl, —(C 3 -C 20 )-heteroaryl, —(C 3 -C 20 )-heteroaryl-(C 1 -C 12 )-alkyl, —(C 3 -C 20 )-heteroaryl-O-(C 1 -C 12 )-alkyl, —COOH, —OH, —SO 3 H, —NH 2 , halogen; and to the use thereof as ligands in alkoxycarbonylation.
1. Compound of formula (I) where R1, R2, R3, R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl, —(C3-C20)-heteroaryl; at least one of the R1, R2, R3, R4 radicals is a —(C3-C20)-heteroaryl radical; and R1, R2, R3, R4, if they are —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl or —(C3-C20)-heteroaryl, may each independently be substituted by one or more substituents selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —O—(C1-C12)-alkyl, —O—(C1-C12)-alkyl-(C6-C20)-aryl, —O—(C3-C12)-cycloalkyl, —S—(C1-C12)-alkyl, —S—(C3-C12)-cycloalkyl, —COO—(C1-C12)-alkyl, —COO—(C3-C12)-cycloalkyl, —CONH—(C1-C12)-alkyl, —CONH—(C3-C12)-cycloalkyl, —CO—(C1-C12)-alkyl, —CO—(C3-C12)-cycloalkyl, —N—[(C1-C12)-alkyl]2, —(C6-C20)-aryl, —(C6-C20)-aryl-(C1-C12)-alkyl, —(C6-C20)-aryl-O—(C1-C12)-alkyl, —(C3-C20)-heteroaryl, —(C3-C20)-heteroaryl-(C1-C12)-alkyl, —(C3-C20)-heteroaryl-O—(C1-C12)-alkyl, —COOH, —OH, —SO3H, —NH2, halogen. 2. Compound according to claim 1, where at least two of the R1, R2, R3, R4 radicals are a —(C3-C20)-heteroaryl radical. 3. Compound according to claim 1, where the R1 and R3 radicals are each a —(C3-C20)-heteroaryl radical. 4. Compound according to claim 1, where the R1 and R3 radicals are each a —(C3-C20)-heteroaryl radical; and R2 and R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C 12)-heterocycloalkyl, —(C6-C20)-aryl. 5. Compound according to claim 1, where the R1 and R3 radicals are each a —(C3-C20)-heteroaryl radical; and R2 and R4 are each a —(C1-C12)-alkyl radical. 6. Compound according to claim 1, where R1, R2, R3, R4, if they are a heteroaryl radical, are each independently selected from furyl, thienyl, pyrrolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, imidazolyl, pyrazolyl, furazanyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidyl, pyrazinyl, benzofuranyl, indolyl, isoindolyl, benzimidazolyl, quinolyl, isoquinolyl. 7. Compound according to claim 1, of the formula (1) 8. Complex comprising Pd and a compound according to claim 1. 9. Process comprising the following process steps: a) initially charging an ethylenically unsaturated compound; b) adding a compound of formula (I) where R1, R2, R3, R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl, —(C3-C20)-heteroaryl; at least one of the R1, R2, R3, R4 radicals is a —(C3-C20)-heteroaryl radical; and R1, R2, R3, R4, if they are —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)—heterocycloalkyl, —(C6-C20)-aryl or —(C3-C20)-heteroaryl, may each independently be substituted by one or more substituents selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —O—(C1-C12)-alkyl, —O—(C1-C12)-alkyl-(C6-C20)-aryl, —O—(C3-C12)-cycloalkyl, —S—(C1-C12)-alkyl, —S—(C3-C12)-cycloalkyl, —COO—(C1-C12)-alkyl, —COO—(C3-C12)-cycloalkyl, —CONH—(C1-C12)-alkyl, —CONH—(C3-C12)-cycloalkyl, —CO—(C1-C12)-alkyl, —CO—(C3-C12)-cycloalkyl, —N—[(C1-C12)-alkyl]2, —(C6-C20)-aryl, —(C6-C20)-aryl-(C1-C12)-alkyl, —(C6-C20)-aryl-O—(C1-C12)-alkyl, —(C3-C20)-heteroaryl, —(C3-C20)-heteroaryl-(C1-C12)-alkyl, —(C3-C20)-heteroaryl-O—(C1-C12)-alkyl, —COOH, —OH, —SO3H, —NH2, halogen and a compound comprising Pd, or adding a complex according to claim 8; c) adding an alcohol; d) feeding in CO; e) heating the reaction mixture, with conversion of the ethylenically unsaturated compound to an ester. 10. Process according to claim 9, wherein the ethylenically unsaturated compound comprises 2 to 30 carbon atoms and optionally one or more functional groups selected from carboxyl, thiocarboxyl, sulpho, sulphinyl, carboxylic anhydride, imide, carboxylic ester, sulphonic ester, carbamoyl, sulphamoyl, cyano, carbonyl, carbonothioyl, hydroxyl, sulphhydryl, amino, ether, thioether, aryl, heteroaryl or silyl groups and/or halogen substituents. 11. Process according to claim 9, wherein the ethylenically unsaturated compound is selected from ethene, propene, 1-butene, cis- and/or trans-2-butene, isobutene, 1,3-butadiene, 1-pentene, cis- and/or trans-2-pentene, 2-methyl-1-butene, 3-methyl-1-butene, 2-methyl-2-butene, hexene, tetramethylethylene, heptene, 1-octene, 2-octene, di-n-butene, and mixtures thereof. 12. Process according to claim 9, wherein the ethylenically unsaturated compound comprises 6 to 22 carbon atoms. 13. Process according to claim 9, wherein the compound comprising Pd in process step b) is selected from palladium dichloride, palladium(II) acetylacetonate, palladium(II) acetate, dichloro(1,5-cyclooctadiene)palladium(II), bis(dibenzylideneacetone)palladium, bis(acetonitrile)dichloropalladium(II), palladium(cinnamyl) dichloride. 14. Process according to claim 9, wherein the alcohol in process step c) is selected from methanol, ethanol, 1-propanol, 1-butanol, 1-pentanol, 1-hexanol, 2-propanol, tent-butanol, 3-pentanol, cyclohexanol, phenol, and mixtures thereof. 15. A process for catalysis of an alkoxycarbonylation reaction, comprising: introducing a compound of formula (I) where Rl, R2, R3, R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl, —(C3-C20)-heteroaryl; at least one of the R1, R2, R3, R4 radicals is a —(C3-C70)-heteroaryl radical; and R1, R2, R3, R4, if they are —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl or —(C3-C20)-heteroaryl, may each independently be substituted by one or more substituents selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —O—(C1-C12)-alkyl, —O—(C1C12)-alkyl-(C6-C20)-aryl, —O—(C3-C12)-cycloalkyl, —S—(C1-C12)-alkyl, —S—(C3-C12)-cycloalkyl, —COO—(C1-C12)-alkyl, —COO—(C3-C12)-cycloalkyl, —CONH—(C1-C12)-alkyl, —CONH—(C3-C12)-cycloalkyl, —CO—(C1-C12)-alkyl, —CO—(C3-C12)-cycloalkyl, —N—[(C1-C12)-alkyl]2, —(C6-C20)-aryl, —(C6-C20)-aryl-(C1-C12)-alkyl, —(C6-C20)-aryl-O—(C1-C12)-alkyl, —(C3-C20)-heteroaryl, —(C3-C20)-heteroaryl-(C1-C12)-alkyl, —(C3-C20)-heteroaryl-O—(C1-C12)-alkyl, —COOH, —OH, —SO3H, —NH2, halogen or a complex according to claim 8.
The invention relates to compounds of formula (I) where R 1 , R 2 , R 3 , R 4 are each independently selected from —(C 1 -C 12 )-alkyl, —(C 3 -C 12 )-cycloalkyl, —(C 3 -C 12 )-heterocycloalkyl, —(C 6 -C 20 )-aryl, —(C 3 -C 20 )-heteroaryl; at least one of the R 1 , R 2 , R 3 , R 4 radicals is a —(C 3 -C 20 )-heteroaryl radical; and R 1 , R 2 , R 3 , R 4 , if they are —(C 1 -C 12 )-alkyl, —(C 3 -C 12 )-cycloalkyl, —(C 3 -C 12 )-heterocycloalkyl, —(C 6 -C 20 )-aryl or —(C 3 -C 20 )-heteroaryl, may each independently be substituted by one or more substituents selected from —(C 1 -C 12 )-alkyl, —(C 3 -C 12 )-cycloalkyl, —(C 3 -C 12 )-heterocycloalkyl, —O—(C 1 -C 12 )-alkyl, —O—(C 1 -C 12 )-alkyl-(C 6 -C 20 )-aryl, —O—(C 3 -C 12 )-cycloalkyl, —S—(C 1 -C 12 )-alkyl, —S—(C 3 -C 12 )-cycloalkyl, —COO—(C 1 -C 12 )-alkyl, —COO—(C 3 -C 12 )-cycloalkyl, —CONH—(C 1 -C 12 )-alkyl, —CONH—(C 3 -C 12 )-cycloalkyl, —CO—(C 1 -C 12 )-alkyl, —CO—(C 3 -C 12 )-cycloalkyl, —N—[(C 1 -C 12 )-alkyl] 2 , —(C 6 -C 20 )-aryl, —(C 6 -C 20 )-aryl-(C 1 -C 12 )-alkyl, —(C 6 -C 20 )-aryl-O—(C 1 -C 12 )-alkyl, —(C 3 -C 20 )-heteroaryl, —(C 3 -C 20 )-heteroaryl-(C 1 -C 12 )-alkyl, —(C 3 -C 20 )-heteroaryl-O-(C 1 -C 12 )-alkyl, —COOH, —OH, —SO 3 H, —NH 2 , halogen; and to the use thereof as ligands in alkoxycarbonylation.1. Compound of formula (I) where R1, R2, R3, R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl, —(C3-C20)-heteroaryl; at least one of the R1, R2, R3, R4 radicals is a —(C3-C20)-heteroaryl radical; and R1, R2, R3, R4, if they are —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl or —(C3-C20)-heteroaryl, may each independently be substituted by one or more substituents selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —O—(C1-C12)-alkyl, —O—(C1-C12)-alkyl-(C6-C20)-aryl, —O—(C3-C12)-cycloalkyl, —S—(C1-C12)-alkyl, —S—(C3-C12)-cycloalkyl, —COO—(C1-C12)-alkyl, —COO—(C3-C12)-cycloalkyl, —CONH—(C1-C12)-alkyl, —CONH—(C3-C12)-cycloalkyl, —CO—(C1-C12)-alkyl, —CO—(C3-C12)-cycloalkyl, —N—[(C1-C12)-alkyl]2, —(C6-C20)-aryl, —(C6-C20)-aryl-(C1-C12)-alkyl, —(C6-C20)-aryl-O—(C1-C12)-alkyl, —(C3-C20)-heteroaryl, —(C3-C20)-heteroaryl-(C1-C12)-alkyl, —(C3-C20)-heteroaryl-O—(C1-C12)-alkyl, —COOH, —OH, —SO3H, —NH2, halogen. 2. Compound according to claim 1, where at least two of the R1, R2, R3, R4 radicals are a —(C3-C20)-heteroaryl radical. 3. Compound according to claim 1, where the R1 and R3 radicals are each a —(C3-C20)-heteroaryl radical. 4. Compound according to claim 1, where the R1 and R3 radicals are each a —(C3-C20)-heteroaryl radical; and R2 and R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C 12)-heterocycloalkyl, —(C6-C20)-aryl. 5. Compound according to claim 1, where the R1 and R3 radicals are each a —(C3-C20)-heteroaryl radical; and R2 and R4 are each a —(C1-C12)-alkyl radical. 6. Compound according to claim 1, where R1, R2, R3, R4, if they are a heteroaryl radical, are each independently selected from furyl, thienyl, pyrrolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, imidazolyl, pyrazolyl, furazanyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidyl, pyrazinyl, benzofuranyl, indolyl, isoindolyl, benzimidazolyl, quinolyl, isoquinolyl. 7. Compound according to claim 1, of the formula (1) 8. Complex comprising Pd and a compound according to claim 1. 9. Process comprising the following process steps: a) initially charging an ethylenically unsaturated compound; b) adding a compound of formula (I) where R1, R2, R3, R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl, —(C3-C20)-heteroaryl; at least one of the R1, R2, R3, R4 radicals is a —(C3-C20)-heteroaryl radical; and R1, R2, R3, R4, if they are —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)—heterocycloalkyl, —(C6-C20)-aryl or —(C3-C20)-heteroaryl, may each independently be substituted by one or more substituents selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —O—(C1-C12)-alkyl, —O—(C1-C12)-alkyl-(C6-C20)-aryl, —O—(C3-C12)-cycloalkyl, —S—(C1-C12)-alkyl, —S—(C3-C12)-cycloalkyl, —COO—(C1-C12)-alkyl, —COO—(C3-C12)-cycloalkyl, —CONH—(C1-C12)-alkyl, —CONH—(C3-C12)-cycloalkyl, —CO—(C1-C12)-alkyl, —CO—(C3-C12)-cycloalkyl, —N—[(C1-C12)-alkyl]2, —(C6-C20)-aryl, —(C6-C20)-aryl-(C1-C12)-alkyl, —(C6-C20)-aryl-O—(C1-C12)-alkyl, —(C3-C20)-heteroaryl, —(C3-C20)-heteroaryl-(C1-C12)-alkyl, —(C3-C20)-heteroaryl-O—(C1-C12)-alkyl, —COOH, —OH, —SO3H, —NH2, halogen and a compound comprising Pd, or adding a complex according to claim 8; c) adding an alcohol; d) feeding in CO; e) heating the reaction mixture, with conversion of the ethylenically unsaturated compound to an ester. 10. Process according to claim 9, wherein the ethylenically unsaturated compound comprises 2 to 30 carbon atoms and optionally one or more functional groups selected from carboxyl, thiocarboxyl, sulpho, sulphinyl, carboxylic anhydride, imide, carboxylic ester, sulphonic ester, carbamoyl, sulphamoyl, cyano, carbonyl, carbonothioyl, hydroxyl, sulphhydryl, amino, ether, thioether, aryl, heteroaryl or silyl groups and/or halogen substituents. 11. Process according to claim 9, wherein the ethylenically unsaturated compound is selected from ethene, propene, 1-butene, cis- and/or trans-2-butene, isobutene, 1,3-butadiene, 1-pentene, cis- and/or trans-2-pentene, 2-methyl-1-butene, 3-methyl-1-butene, 2-methyl-2-butene, hexene, tetramethylethylene, heptene, 1-octene, 2-octene, di-n-butene, and mixtures thereof. 12. Process according to claim 9, wherein the ethylenically unsaturated compound comprises 6 to 22 carbon atoms. 13. Process according to claim 9, wherein the compound comprising Pd in process step b) is selected from palladium dichloride, palladium(II) acetylacetonate, palladium(II) acetate, dichloro(1,5-cyclooctadiene)palladium(II), bis(dibenzylideneacetone)palladium, bis(acetonitrile)dichloropalladium(II), palladium(cinnamyl) dichloride. 14. Process according to claim 9, wherein the alcohol in process step c) is selected from methanol, ethanol, 1-propanol, 1-butanol, 1-pentanol, 1-hexanol, 2-propanol, tent-butanol, 3-pentanol, cyclohexanol, phenol, and mixtures thereof. 15. A process for catalysis of an alkoxycarbonylation reaction, comprising: introducing a compound of formula (I) where Rl, R2, R3, R4 are each independently selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl, —(C3-C20)-heteroaryl; at least one of the R1, R2, R3, R4 radicals is a —(C3-C70)-heteroaryl radical; and R1, R2, R3, R4, if they are —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —(C6-C20)-aryl or —(C3-C20)-heteroaryl, may each independently be substituted by one or more substituents selected from —(C1-C12)-alkyl, —(C3-C12)-cycloalkyl, —(C3-C12)-heterocycloalkyl, —O—(C1-C12)-alkyl, —O—(C1C12)-alkyl-(C6-C20)-aryl, —O—(C3-C12)-cycloalkyl, —S—(C1-C12)-alkyl, —S—(C3-C12)-cycloalkyl, —COO—(C1-C12)-alkyl, —COO—(C3-C12)-cycloalkyl, —CONH—(C1-C12)-alkyl, —CONH—(C3-C12)-cycloalkyl, —CO—(C1-C12)-alkyl, —CO—(C3-C12)-cycloalkyl, —N—[(C1-C12)-alkyl]2, —(C6-C20)-aryl, —(C6-C20)-aryl-(C1-C12)-alkyl, —(C6-C20)-aryl-O—(C1-C12)-alkyl, —(C3-C20)-heteroaryl, —(C3-C20)-heteroaryl-(C1-C12)-alkyl, —(C3-C20)-heteroaryl-O—(C1-C12)-alkyl, —COOH, —OH, —SO3H, —NH2, halogen or a complex according to claim 8.
1,600
309
13,805,981
1,611
This invention relates to disinfectant compositions comprising a first active ingredient comprising a source of acetyl radicals, and a second active ingredient comprising a source of peroxygen, the composition being soluble in water to provide a solution of peracetic acid.
1. A disinfectant composition comprising a first active ingredient comprising a source of acetyl radicals, and a second active ingredient comprising a source of peroxygen, the composition being soluble in water to provide a solution of peracetic acid. 2. A disinfectant composition according to claim 1 in tablet form. 3. A disinfectant composition according to claim 1 wherein the disinfectant composition is held in a container. 4. A disinfectant composition according to claim 1 wherein the container is a sachet or a pouch. 5. A disinfectant composition according to claim 4 wherein the container is formed of a water soluble material. 6. A disinfectant composition according to claim 5 wherein the water soluble material is polyvinyl alcohol. 7. A disinfectant composition according to claim 1, further comprising a third active ingredient comprising a pH modifier. 8. A disinfectant composition according to claim 1, wherein the first active ingredient comprises tetra-acetyl ethylenediamine (TAED). 9. A disinfectant composition according to claim 8 wherein the TAED is uncoated TAED. 10. A disinfectant composition according to claim 1, wherein the first active ingredient is present in the disinfectant composition in an amount in the range of 1 wt % to 50 wt %. 11. A disinfectant composition according to claim 10 wherein the first active ingredient is present in the disinfectant composition in an amount in the range of 20 wt % to 40 wt %. 12. A disinfectant composition according to claim 11 wherein the first active ingredient is present in the disinfectant composition in an amount in the range of 22 wt % to 24 wt %. 13. A disinfectant composition according to claim 1, wherein the second active ingredient is selected from the group consisting of sodium, potassium or ammonium percarbonate; sodium, potassium or ammonium perborate; sodium, potassium or ammonium persulphate; sodium, potassium or ammonium perphosphate; and carbamide peroxide. 14. A disinfectant composition according to claim 13 wherein the second active ingredient is sodium percarbonate or sodium perborate. 15. A disinfectant composition according to claim 14 wherein the sodium perborate is sodium perborate monohydrate. 16. A disinfectant composition according to claim 1, wherein the second active ingredient is present in the disinfectant composition in an amount in the range of 1 wt % to 100 wt %. 17. A disinfectant composition according to claim 16 wherein the second active ingredient is present in the disinfectant composition in an amount in the range of 40 wt % to 60 wt %. 18. A disinfectant composition according to claim 17 wherein the second active ingredient is present in the disinfectant composition in an amount of the range of 44 wt % to 46 wt %. 19. A disinfectant composition according to claim 7 wherein the third active ingredient comprises an anhydrous organic acid. 20. A disinfectant composition according to claim 19 wherein the third active ingredient is one or more selected from citric acid, fumaric acid, tartaric acid, adipic acid. 21. A disinfectant composition according to claim 20 wherein the third active ingredient comprises anhydrous citric acid. 22. A disinfectant composition according to claim 7, wherein the third active ingredient is present in the disinfectant composition in an amount in the range of 1 wt % to 50 wt %. 23. A disinfectant composition according to claim 22 wherein the third active ingredient is present in the disinfectant composition in an amount in the range of 20 wt % to 40 wt %. 24. A disinfectant composition according to claim 23 wherein the third active ingredient is present in the disinfectant composition in an amount in the range of 25 wt % to 29 wt %. 25. A disinfectant composition according to claim 1, wherein the disinfectant composition further comprises a chelating agent. 26. A disinfectant composition according to claim 25 wherein the chelating agent comprises a phosphonate or ethylenediamine tetra acetic acid (EDTA). 27. A disinfectant composition according to claim 26 wherein the chelating agent is disodium EDTA or dipotassium EDTA. 28. A disinfectant composition according to claim 25 wherein the chelating agent is present in the disinfectant composition in an amount in the range of 0.01 wt % to 1 wt %. 29. A disinfectant composition according to claim 28 wherein the chelating agent is present in the disinfectant composition in an amount of 0.05 wt % to 0.15 wt %. 30. A disinfectant composition according to claim 29 wherein the chelating agent is present in the disinfectant composition in an amount in the range of 0.09 wt % to 0.11 wt %. 31. A disinfectant composition according to claim 1, further comprising a surfactant. 32. A disinfectant composition according to claim 31 wherein the surfactant comprises an anionic surfactant. 33. A disinfectant composition according to claim 32 wherein the anionic surfactant is selected from the group consisting of Fatty Alcohol Sulphates, Fatty Alcohol Phosphates, fatty alcohol benzene sulphonates, soaps (sodium and potassium salts of fatty acids), fatty alcohol ether sulphates, olefin sulphonates, lignosulphonates, phosphate esters and sarcosinates. 34. A disinfectant composition according to claim 33 wherein the anionic surfactant is sodium lauryl sulphate. 35. A disinfectant composition according to claim 31 wherein the surfactant comprises a non-ionic surfactant. 36. A disinfectant composition according to claim 35 wherein the non-ionic surfactant is selected from the group consisting of ethoxylated and propoxylated fatty alcohols, ethoxylated and propoxylated alkyl phenols, fatty acid esters, Polyethylene glycol esters, alkyl glucosides, glyceryl and polyglyceryl esters, ethoxylated/propoxylated copolymers, ethoxylated thiols, glucose and sucrose esters, sugar esters, sorbitan esters, ethoxylated glucose and sucrose esters, ethoxylated sugar esters, ethoxylated sorbitan esters, linear and branched fatty alcohol pyrrolidone derivatives (N-substituted). 37. A disinfectant composition according to claim 36 wherein the non-ionic surfactant comprises a fatty alcohol derivative. 38. A disinfectant composition according to claim 37 wherein the fatty alcohol derivative is a fatty alcohol glucoside, for example a C10 fatty alcohol glucoside. 39. A disinfectant composition according to claim 35 wherein the non-ionic surfactant comprises an N-substituted pyrrolidone for example a C8-N-substituted pyrrolidone (or n-octyl pyrrolidone). 40. A disinfectant composition according to claim 35 wherein the surfactant is present in the disinfectant composition in an amount in the range of 0.01 wt % to 0.5 wt %. 41. A disinfectant composition according to claim 40 wherein the surfactant is present in the disinfectant composition in an amount in the range of 0.1 wt % to 0.3 wt %. 42. A disinfectant composition according to claim 41 wherein the surfactant is present in the disinfectant composition in amount of 0.15 wt % to 0.25 wt %. 43. A disinfectant composition according to claim 1, wherein the disinfectant composition further includes a corrosion inhibitor. 44. A disinfectant composition according to claim 43 wherein the corrosion inhibitor comprises a triazole compound. 45. A disinfectant composition according to claim 43 wherein the corrosion inhibitor is present in the disinfectant composition in an amount in the range of 0.01 wt % to 0.5 wt %. 46. A disinfectant system comprising a disinfectant composition according to claim 1, and a water soluble container in which the disinfectant composition is held. 47. A disinfectant system according to claim 46 wherein the container comprises a sachet or pouch formed of a water soluble thermoplastic film. 48. A disinfectant system according to claim 47 wherein the water soluble film is formed of polyvinyl alcohol. 49. A process for the preparation of a disinfectant composition according to claim 7, the process comprising the steps of: i) preparing a liquid composition of a non-ionic surfactant(s), optionally including an inert carrier, and a corrosion inhibitor; ii) preparing a powder composition of the first active ingredient, second active ingredient, third active ingredient, an anionic surfactant and a chelating agent; and iii) adding the liquid composition to the powder composition to provide a homogeneous blend. 50. A process according to claim 49 wherein the powder composition is prepared by blending the components consecutively in the following order: first active ingredient, second active ingredient, third active ingredient, anionic surfactant and chelating agent.
This invention relates to disinfectant compositions comprising a first active ingredient comprising a source of acetyl radicals, and a second active ingredient comprising a source of peroxygen, the composition being soluble in water to provide a solution of peracetic acid.1. A disinfectant composition comprising a first active ingredient comprising a source of acetyl radicals, and a second active ingredient comprising a source of peroxygen, the composition being soluble in water to provide a solution of peracetic acid. 2. A disinfectant composition according to claim 1 in tablet form. 3. A disinfectant composition according to claim 1 wherein the disinfectant composition is held in a container. 4. A disinfectant composition according to claim 1 wherein the container is a sachet or a pouch. 5. A disinfectant composition according to claim 4 wherein the container is formed of a water soluble material. 6. A disinfectant composition according to claim 5 wherein the water soluble material is polyvinyl alcohol. 7. A disinfectant composition according to claim 1, further comprising a third active ingredient comprising a pH modifier. 8. A disinfectant composition according to claim 1, wherein the first active ingredient comprises tetra-acetyl ethylenediamine (TAED). 9. A disinfectant composition according to claim 8 wherein the TAED is uncoated TAED. 10. A disinfectant composition according to claim 1, wherein the first active ingredient is present in the disinfectant composition in an amount in the range of 1 wt % to 50 wt %. 11. A disinfectant composition according to claim 10 wherein the first active ingredient is present in the disinfectant composition in an amount in the range of 20 wt % to 40 wt %. 12. A disinfectant composition according to claim 11 wherein the first active ingredient is present in the disinfectant composition in an amount in the range of 22 wt % to 24 wt %. 13. A disinfectant composition according to claim 1, wherein the second active ingredient is selected from the group consisting of sodium, potassium or ammonium percarbonate; sodium, potassium or ammonium perborate; sodium, potassium or ammonium persulphate; sodium, potassium or ammonium perphosphate; and carbamide peroxide. 14. A disinfectant composition according to claim 13 wherein the second active ingredient is sodium percarbonate or sodium perborate. 15. A disinfectant composition according to claim 14 wherein the sodium perborate is sodium perborate monohydrate. 16. A disinfectant composition according to claim 1, wherein the second active ingredient is present in the disinfectant composition in an amount in the range of 1 wt % to 100 wt %. 17. A disinfectant composition according to claim 16 wherein the second active ingredient is present in the disinfectant composition in an amount in the range of 40 wt % to 60 wt %. 18. A disinfectant composition according to claim 17 wherein the second active ingredient is present in the disinfectant composition in an amount of the range of 44 wt % to 46 wt %. 19. A disinfectant composition according to claim 7 wherein the third active ingredient comprises an anhydrous organic acid. 20. A disinfectant composition according to claim 19 wherein the third active ingredient is one or more selected from citric acid, fumaric acid, tartaric acid, adipic acid. 21. A disinfectant composition according to claim 20 wherein the third active ingredient comprises anhydrous citric acid. 22. A disinfectant composition according to claim 7, wherein the third active ingredient is present in the disinfectant composition in an amount in the range of 1 wt % to 50 wt %. 23. A disinfectant composition according to claim 22 wherein the third active ingredient is present in the disinfectant composition in an amount in the range of 20 wt % to 40 wt %. 24. A disinfectant composition according to claim 23 wherein the third active ingredient is present in the disinfectant composition in an amount in the range of 25 wt % to 29 wt %. 25. A disinfectant composition according to claim 1, wherein the disinfectant composition further comprises a chelating agent. 26. A disinfectant composition according to claim 25 wherein the chelating agent comprises a phosphonate or ethylenediamine tetra acetic acid (EDTA). 27. A disinfectant composition according to claim 26 wherein the chelating agent is disodium EDTA or dipotassium EDTA. 28. A disinfectant composition according to claim 25 wherein the chelating agent is present in the disinfectant composition in an amount in the range of 0.01 wt % to 1 wt %. 29. A disinfectant composition according to claim 28 wherein the chelating agent is present in the disinfectant composition in an amount of 0.05 wt % to 0.15 wt %. 30. A disinfectant composition according to claim 29 wherein the chelating agent is present in the disinfectant composition in an amount in the range of 0.09 wt % to 0.11 wt %. 31. A disinfectant composition according to claim 1, further comprising a surfactant. 32. A disinfectant composition according to claim 31 wherein the surfactant comprises an anionic surfactant. 33. A disinfectant composition according to claim 32 wherein the anionic surfactant is selected from the group consisting of Fatty Alcohol Sulphates, Fatty Alcohol Phosphates, fatty alcohol benzene sulphonates, soaps (sodium and potassium salts of fatty acids), fatty alcohol ether sulphates, olefin sulphonates, lignosulphonates, phosphate esters and sarcosinates. 34. A disinfectant composition according to claim 33 wherein the anionic surfactant is sodium lauryl sulphate. 35. A disinfectant composition according to claim 31 wherein the surfactant comprises a non-ionic surfactant. 36. A disinfectant composition according to claim 35 wherein the non-ionic surfactant is selected from the group consisting of ethoxylated and propoxylated fatty alcohols, ethoxylated and propoxylated alkyl phenols, fatty acid esters, Polyethylene glycol esters, alkyl glucosides, glyceryl and polyglyceryl esters, ethoxylated/propoxylated copolymers, ethoxylated thiols, glucose and sucrose esters, sugar esters, sorbitan esters, ethoxylated glucose and sucrose esters, ethoxylated sugar esters, ethoxylated sorbitan esters, linear and branched fatty alcohol pyrrolidone derivatives (N-substituted). 37. A disinfectant composition according to claim 36 wherein the non-ionic surfactant comprises a fatty alcohol derivative. 38. A disinfectant composition according to claim 37 wherein the fatty alcohol derivative is a fatty alcohol glucoside, for example a C10 fatty alcohol glucoside. 39. A disinfectant composition according to claim 35 wherein the non-ionic surfactant comprises an N-substituted pyrrolidone for example a C8-N-substituted pyrrolidone (or n-octyl pyrrolidone). 40. A disinfectant composition according to claim 35 wherein the surfactant is present in the disinfectant composition in an amount in the range of 0.01 wt % to 0.5 wt %. 41. A disinfectant composition according to claim 40 wherein the surfactant is present in the disinfectant composition in an amount in the range of 0.1 wt % to 0.3 wt %. 42. A disinfectant composition according to claim 41 wherein the surfactant is present in the disinfectant composition in amount of 0.15 wt % to 0.25 wt %. 43. A disinfectant composition according to claim 1, wherein the disinfectant composition further includes a corrosion inhibitor. 44. A disinfectant composition according to claim 43 wherein the corrosion inhibitor comprises a triazole compound. 45. A disinfectant composition according to claim 43 wherein the corrosion inhibitor is present in the disinfectant composition in an amount in the range of 0.01 wt % to 0.5 wt %. 46. A disinfectant system comprising a disinfectant composition according to claim 1, and a water soluble container in which the disinfectant composition is held. 47. A disinfectant system according to claim 46 wherein the container comprises a sachet or pouch formed of a water soluble thermoplastic film. 48. A disinfectant system according to claim 47 wherein the water soluble film is formed of polyvinyl alcohol. 49. A process for the preparation of a disinfectant composition according to claim 7, the process comprising the steps of: i) preparing a liquid composition of a non-ionic surfactant(s), optionally including an inert carrier, and a corrosion inhibitor; ii) preparing a powder composition of the first active ingredient, second active ingredient, third active ingredient, an anionic surfactant and a chelating agent; and iii) adding the liquid composition to the powder composition to provide a homogeneous blend. 50. A process according to claim 49 wherein the powder composition is prepared by blending the components consecutively in the following order: first active ingredient, second active ingredient, third active ingredient, anionic surfactant and chelating agent.
1,600
310
12,698,224
1,631
There is provided a computer system for visualizing and editing single molecule fragments and one or more previously-produced single molecule assemblies or “contigs.” The present visualization and editing system allows a user to visualize large data sets resulting from single molecule map assembly operations, and to rapidly discern important features while errors and other discrepancies are conveniently resolved. The system includes one or more connectors connecting each to one or more databases capable of storing a diverse array of biomedical information, in addition to the single molecule data against which a user may validate the prior alignment and assembly. Embodiments described herein are thus useful in studies of macromolecules such as DNA, RNA, peptides and proteins. The visualization and editing system may be implemented and deployed over a computer network, and may be ergonomically optimized to facilitate user interaction.
1. A computer system for validating single molecule assemblies, the computer system comprising: (a) a first database comprising single molecule data, the single molecule data comprising image data derived from optical mapping of a single molecule assembly; (b) a second database comprising biomedical data associated with the single molecule assembly; (c) a first database connector communicatively linked to the first database, and a second database connector communicatively linked to the second database; (d) a user interface programmatically linked to the first database connector and the second database connector, the user interface displaying the single molecule data from the first database and the biomedical data from the second database, the user interface programmed to display the single molecule data alongside the biomedical data, to provide horizontal and vertical scaling of the single molecule image data, to receive user input commands for interacting with the single molecule data and biomedical data, and to delete the single molecule data from the first database upon receipt of a command from the user input device; and (e) a user input device communicatively linked to the user interface, the user input device transmitting at least one user input command for interacting with the single molecule data. 2. The computer system of claim 1, wherein the single molecule assembly comprising a first single molecule fragment and a second molecule fragment, the first database associating the single molecule data of the single molecule fragment with the single molecule data of the second molecule fragment. 3. The computer system of claim 2, wherein the user interface further programmed to display the first molecule data in a first row and the second molecule data in a second row, the first row and the second row being proximately located on the user interface. 4. The computer system of claim 3, wherein the user interface further programmed to differentiate the first molecule data from the second molecule data using a color code. 5. The computer system of claim 2, wherein the user interface further programmed to merge the first single molecule fragment with the second molecule fragment as a merged molecule data and to store the merged molecule data in the first database upon receipt of a command from the user input device. 6. (canceled) 7. The computer system of claim 1, wherein the single molecule data further comprising at least one restriction map. 8. The computer system of claim 1, further comprising a computer network and at least one database server, wherein the first database connector and the second database connector are communicatively linked to the first database and second database by the computer network, and the first database and the second database being stored on at least one database server. 9. The computer system of claim 1, wherein the biomedical data is genomic data, and wherein the single molecule assemblies are DNA molecules. 10. The computer system of claim 1, wherein the biomedical data is proteomics data, and wherein the single molecule assemblies are protein molecules. 11. The computer system of claim 1, wherein the user interface is a graphical user interface, the graphical user interface being programmed to display a window for displaying the single molecule data alongside the biomedical data. 12. The computer system of claim 11, wherein the user interface is further programmed to differentiate single molecule data from the biomedical data using color coding. 13. The computer system of claim 1, further comprising a software application, wherein the user interface is implemented as a plug-in for the software application. 14. The computer system of claim 1, wherein the user interface further programmed to include a command line for receiving user input commands. 15. The computer system of claim 1, further comprising a third database and a third database connector communicatively linked to the third database, the third database comprising genetic annotation data associated with the single molecule data; and wherein the user interface further programmed to display the genetic annotation data. 16. The computer system of claim 15, wherein the genetic annotation data is displayed along side the single molecule data and the biomedical data. 17. The computer system of claim 1, wherein the biomedical data is selected from the group consisting of: genes, sequence coverage, sequence tagged side (STS) markers, single-nucleotide polymorphism (SNP) sites, CpG islands, chromosome banding, guanine-cytosine (GC) content, chromosome banding, amino acid sequences of the encoded proteins, primary and tertiary structures of encoded proteins, and molecules or agents that potentially interact with DNA molecules or encoded proteins. 18. The computer system of claim 1, further comprising a computer network and at least one database server, wherein the second database connector is communicatively linked to the second database by the computer network, and the second database is stored on the least one database server. 19. The computer system of claim 8 or 18, wherein the computer network is the internet. 20. The computer system of claim 8 or 18, wherein the computer network is a local area network. 21. The computer system of claim 1, wherein the first database and second database are relational databases. 22. The computer system of claim 1, wherein the first database and second database are object databases.
There is provided a computer system for visualizing and editing single molecule fragments and one or more previously-produced single molecule assemblies or “contigs.” The present visualization and editing system allows a user to visualize large data sets resulting from single molecule map assembly operations, and to rapidly discern important features while errors and other discrepancies are conveniently resolved. The system includes one or more connectors connecting each to one or more databases capable of storing a diverse array of biomedical information, in addition to the single molecule data against which a user may validate the prior alignment and assembly. Embodiments described herein are thus useful in studies of macromolecules such as DNA, RNA, peptides and proteins. The visualization and editing system may be implemented and deployed over a computer network, and may be ergonomically optimized to facilitate user interaction.1. A computer system for validating single molecule assemblies, the computer system comprising: (a) a first database comprising single molecule data, the single molecule data comprising image data derived from optical mapping of a single molecule assembly; (b) a second database comprising biomedical data associated with the single molecule assembly; (c) a first database connector communicatively linked to the first database, and a second database connector communicatively linked to the second database; (d) a user interface programmatically linked to the first database connector and the second database connector, the user interface displaying the single molecule data from the first database and the biomedical data from the second database, the user interface programmed to display the single molecule data alongside the biomedical data, to provide horizontal and vertical scaling of the single molecule image data, to receive user input commands for interacting with the single molecule data and biomedical data, and to delete the single molecule data from the first database upon receipt of a command from the user input device; and (e) a user input device communicatively linked to the user interface, the user input device transmitting at least one user input command for interacting with the single molecule data. 2. The computer system of claim 1, wherein the single molecule assembly comprising a first single molecule fragment and a second molecule fragment, the first database associating the single molecule data of the single molecule fragment with the single molecule data of the second molecule fragment. 3. The computer system of claim 2, wherein the user interface further programmed to display the first molecule data in a first row and the second molecule data in a second row, the first row and the second row being proximately located on the user interface. 4. The computer system of claim 3, wherein the user interface further programmed to differentiate the first molecule data from the second molecule data using a color code. 5. The computer system of claim 2, wherein the user interface further programmed to merge the first single molecule fragment with the second molecule fragment as a merged molecule data and to store the merged molecule data in the first database upon receipt of a command from the user input device. 6. (canceled) 7. The computer system of claim 1, wherein the single molecule data further comprising at least one restriction map. 8. The computer system of claim 1, further comprising a computer network and at least one database server, wherein the first database connector and the second database connector are communicatively linked to the first database and second database by the computer network, and the first database and the second database being stored on at least one database server. 9. The computer system of claim 1, wherein the biomedical data is genomic data, and wherein the single molecule assemblies are DNA molecules. 10. The computer system of claim 1, wherein the biomedical data is proteomics data, and wherein the single molecule assemblies are protein molecules. 11. The computer system of claim 1, wherein the user interface is a graphical user interface, the graphical user interface being programmed to display a window for displaying the single molecule data alongside the biomedical data. 12. The computer system of claim 11, wherein the user interface is further programmed to differentiate single molecule data from the biomedical data using color coding. 13. The computer system of claim 1, further comprising a software application, wherein the user interface is implemented as a plug-in for the software application. 14. The computer system of claim 1, wherein the user interface further programmed to include a command line for receiving user input commands. 15. The computer system of claim 1, further comprising a third database and a third database connector communicatively linked to the third database, the third database comprising genetic annotation data associated with the single molecule data; and wherein the user interface further programmed to display the genetic annotation data. 16. The computer system of claim 15, wherein the genetic annotation data is displayed along side the single molecule data and the biomedical data. 17. The computer system of claim 1, wherein the biomedical data is selected from the group consisting of: genes, sequence coverage, sequence tagged side (STS) markers, single-nucleotide polymorphism (SNP) sites, CpG islands, chromosome banding, guanine-cytosine (GC) content, chromosome banding, amino acid sequences of the encoded proteins, primary and tertiary structures of encoded proteins, and molecules or agents that potentially interact with DNA molecules or encoded proteins. 18. The computer system of claim 1, further comprising a computer network and at least one database server, wherein the second database connector is communicatively linked to the second database by the computer network, and the second database is stored on the least one database server. 19. The computer system of claim 8 or 18, wherein the computer network is the internet. 20. The computer system of claim 8 or 18, wherein the computer network is a local area network. 21. The computer system of claim 1, wherein the first database and second database are relational databases. 22. The computer system of claim 1, wherein the first database and second database are object databases.
1,600
311
15,052,781
1,631
Methods and apparatus are disclosed herein for encoding human readable text conveying a non-genetic message into nucleic acid sequences with a substantially reduced probability of biological impact and decoding such text from nucleic acid sequences. In one embodiment, each symbol of a symbol set of human readable symbols uniquely maps to a respective codon identifier. Mapping may ensure that each symbol will not map to a codon identifier that generates an amino acid residue which has a single-letter abbreviation that is the equivalent to the respective symbol. Synthetic nucleic acid sequences comprising such human readable text, and recombinant or synthetic cells comprising such sequences are provided, as well as methods of identifying cells, organisms, or samples containing such sequences.
1. A method of generating a sequence of codon identifiers corresponding to a sequence of human readable symbols, and assigned according to a coding scheme to convey a non-genetic message in a human reference language, the method comprising: (i) receiving the sequence of human readable symbols at a memory module; (ii) loading a human readable symbol map within the memory module, wherein the human readable symbol map is configured to determine a codon identifier that maps to each human readable symbol within the sequence, wherein the human readable symbol map is further configured to map a human readable symbol with a frequency of occurrence that is less than one percent within a reference language to a start codon, and wherein the symbol map is further configured to map a human readable symbol with a frequency of occurrence that is greater than five percent within the reference language to a stop codon and no symbol is coded for by A TG; and (iii) outputting a sequence of codon identifiers corresponding to each human readable symbol within the sequence, (iv) synthesizing a nucleic acid with the sequence of step (iii). 2. A method of generating a recombinant or synthetic organism or cell, said method comprising: (i) generating a nucleic acid sequence of claim 1; and (ii) introducing said nucleic acid sequence into a recombinant or synthetic organism. 3. The method of claim 1 wherein said set of human readable symbols comprises a watermark that allows the authentication or identification of said recombinant or synthetic organism comprising said watermark. 4. The method of claim 3 wherein said watermark is a copyright notice, a trademark, a company identifier, a name, a phrase, a sentence, a quotation, genetic information, unique identifying information, data, or a combination of any thereof. 5. The method of claim 1 wherein the synthetic nucleic acid sequence further comprises an all-6 reading frame stop codon containing sequence 5′ to a first codon identifier in the sequence and/or an all-6 reading frame stop codon containing sequence 3′ to the last codon identifier in the sequence. 6. A recombinant or synthetic organism or cell comprising the synthetic nucleic acid sequence of claim 1, wherein said nucleic acid sequence has a length of 0.1 kb to 40 kb. 7. The recombinant or synthetic organism or cell of claim 6, wherein said recombinant or synthetic cell is a prokaryotic cell, a eukaryotic cell, an archaei cell, a virus, a bacterial cell, a yeast cell, a fungal cell, an algal cell, an animal cell, or a plant cell. 8. The recombinant or synthetic organism or cell of claim 7 wherein the cell is a bacterial cell. 9. The recombinant or synthetic organism or cell of claim 6, wherein the codon identifiers: (i) do not correspond to sequence of a gene or other biologically active sequence; (ii) correspond to one or more letters, one or more numbers, one or more spaces, one or more punctuation marks, one or more mathematical symbols, one or more typographical characters, one or more new lines, or a combination of any thereof; or (iii) consist of three nucleotides. 10. The recombinant or synthetic organism or cell of claim 9, wherein the nucleic acid with the sequence of step (iii) further comprises an all-6 reading frame stop codon containing sequence 5′ to a first codon identifier in the sequence and/or an all-6 reading frame stop codon containing sequence 3′ to the last codon identifier in the sequence. 11. The recombinant or synthetic organism or cell of claim 10 wherein the all-6 reading frame stop codon is SEQ ID NO: 1. 12. The recombinant or synthetic organism or cell of claim 2 wherein the reference language is selected from the group consisting of: English, Spanish, French, Italian, and German. 13. The recombinant or synthetic organism or cell of claim 12 wherein the reference language is English. 14. The recombinant or synthetic organism or cell of claim 2 wherein: a. said set of human readable symbols comprises a watermark that allows the authentication or identification of said recombinant or synthetic organism comprising said watermark; b. the recombinant or synthetic organism or cell is a prokaryotic cell or bacterial cell; and c. the reference language is selected from the group consisting of: English, Spanish, French, Italian, and German. 15. An apparatus for transforming a sequence of codon identifiers into a sequence of claim 1, the apparatus comprising: (i) a processor adapted to execute instructions; and (ii) a storage module, wherein the storage module comprises a data structure for mapping codon identifiers into human readable symbols, and a set of instructions which, when executed by the processor, generate a human readable symbol for each codon identifier read from a sequence of codon identifiers, wherein the human readable symbol generated is based at least in part upon the data structure; wherein the data structure is configured to map a start codon to a human readable symbol with a frequency of occurrence within a reference language that is less than one percent, and wherein the data structure is further configured to map a plurality of stop codons to human readable symbols with frequencies of occurrence within the reference language that are greater than five percent, and no symbol is coded for by ATG. 16. The apparatus of claim 15, wherein the data structure or mapping function, respectively, does not map a codon identifier to a single letter representation of an amino acid residue normally assigned to that codon identifier in the standard genetic code. 17. The apparatus of claim 15, wherein the sequence of codon identifiers comprises an all-6 reading frame stop codon containing sequence 5′ to a first codon identifier in the sequence and/or an all-6 reading frame stop codon containing sequence 3′ to the last codon identifier in the sequence.
Methods and apparatus are disclosed herein for encoding human readable text conveying a non-genetic message into nucleic acid sequences with a substantially reduced probability of biological impact and decoding such text from nucleic acid sequences. In one embodiment, each symbol of a symbol set of human readable symbols uniquely maps to a respective codon identifier. Mapping may ensure that each symbol will not map to a codon identifier that generates an amino acid residue which has a single-letter abbreviation that is the equivalent to the respective symbol. Synthetic nucleic acid sequences comprising such human readable text, and recombinant or synthetic cells comprising such sequences are provided, as well as methods of identifying cells, organisms, or samples containing such sequences.1. A method of generating a sequence of codon identifiers corresponding to a sequence of human readable symbols, and assigned according to a coding scheme to convey a non-genetic message in a human reference language, the method comprising: (i) receiving the sequence of human readable symbols at a memory module; (ii) loading a human readable symbol map within the memory module, wherein the human readable symbol map is configured to determine a codon identifier that maps to each human readable symbol within the sequence, wherein the human readable symbol map is further configured to map a human readable symbol with a frequency of occurrence that is less than one percent within a reference language to a start codon, and wherein the symbol map is further configured to map a human readable symbol with a frequency of occurrence that is greater than five percent within the reference language to a stop codon and no symbol is coded for by A TG; and (iii) outputting a sequence of codon identifiers corresponding to each human readable symbol within the sequence, (iv) synthesizing a nucleic acid with the sequence of step (iii). 2. A method of generating a recombinant or synthetic organism or cell, said method comprising: (i) generating a nucleic acid sequence of claim 1; and (ii) introducing said nucleic acid sequence into a recombinant or synthetic organism. 3. The method of claim 1 wherein said set of human readable symbols comprises a watermark that allows the authentication or identification of said recombinant or synthetic organism comprising said watermark. 4. The method of claim 3 wherein said watermark is a copyright notice, a trademark, a company identifier, a name, a phrase, a sentence, a quotation, genetic information, unique identifying information, data, or a combination of any thereof. 5. The method of claim 1 wherein the synthetic nucleic acid sequence further comprises an all-6 reading frame stop codon containing sequence 5′ to a first codon identifier in the sequence and/or an all-6 reading frame stop codon containing sequence 3′ to the last codon identifier in the sequence. 6. A recombinant or synthetic organism or cell comprising the synthetic nucleic acid sequence of claim 1, wherein said nucleic acid sequence has a length of 0.1 kb to 40 kb. 7. The recombinant or synthetic organism or cell of claim 6, wherein said recombinant or synthetic cell is a prokaryotic cell, a eukaryotic cell, an archaei cell, a virus, a bacterial cell, a yeast cell, a fungal cell, an algal cell, an animal cell, or a plant cell. 8. The recombinant or synthetic organism or cell of claim 7 wherein the cell is a bacterial cell. 9. The recombinant or synthetic organism or cell of claim 6, wherein the codon identifiers: (i) do not correspond to sequence of a gene or other biologically active sequence; (ii) correspond to one or more letters, one or more numbers, one or more spaces, one or more punctuation marks, one or more mathematical symbols, one or more typographical characters, one or more new lines, or a combination of any thereof; or (iii) consist of three nucleotides. 10. The recombinant or synthetic organism or cell of claim 9, wherein the nucleic acid with the sequence of step (iii) further comprises an all-6 reading frame stop codon containing sequence 5′ to a first codon identifier in the sequence and/or an all-6 reading frame stop codon containing sequence 3′ to the last codon identifier in the sequence. 11. The recombinant or synthetic organism or cell of claim 10 wherein the all-6 reading frame stop codon is SEQ ID NO: 1. 12. The recombinant or synthetic organism or cell of claim 2 wherein the reference language is selected from the group consisting of: English, Spanish, French, Italian, and German. 13. The recombinant or synthetic organism or cell of claim 12 wherein the reference language is English. 14. The recombinant or synthetic organism or cell of claim 2 wherein: a. said set of human readable symbols comprises a watermark that allows the authentication or identification of said recombinant or synthetic organism comprising said watermark; b. the recombinant or synthetic organism or cell is a prokaryotic cell or bacterial cell; and c. the reference language is selected from the group consisting of: English, Spanish, French, Italian, and German. 15. An apparatus for transforming a sequence of codon identifiers into a sequence of claim 1, the apparatus comprising: (i) a processor adapted to execute instructions; and (ii) a storage module, wherein the storage module comprises a data structure for mapping codon identifiers into human readable symbols, and a set of instructions which, when executed by the processor, generate a human readable symbol for each codon identifier read from a sequence of codon identifiers, wherein the human readable symbol generated is based at least in part upon the data structure; wherein the data structure is configured to map a start codon to a human readable symbol with a frequency of occurrence within a reference language that is less than one percent, and wherein the data structure is further configured to map a plurality of stop codons to human readable symbols with frequencies of occurrence within the reference language that are greater than five percent, and no symbol is coded for by ATG. 16. The apparatus of claim 15, wherein the data structure or mapping function, respectively, does not map a codon identifier to a single letter representation of an amino acid residue normally assigned to that codon identifier in the standard genetic code. 17. The apparatus of claim 15, wherein the sequence of codon identifiers comprises an all-6 reading frame stop codon containing sequence 5′ to a first codon identifier in the sequence and/or an all-6 reading frame stop codon containing sequence 3′ to the last codon identifier in the sequence.
1,600
312
14,105,489
1,627
The present application relates to personal care compositions including perfumes and sulfur-based perfume raw materials, and methods for making and using the personal care compositions to resist consumer fragrance habituation.
1. A personal care composition comprising a perfume, the perfume comprising, based on total perfume weight, from about 0.000001% to about 10%, of a perfume raw material comprising a sulfur atom, such that the perfume raw material resists the fragrance habituation of a consumer to the personal care composition. 2. The personal care composition of claim 1 comprising from about 0.001% to about 0.1%, of the perfume raw material. 3. The personal care composition of claim 1, wherein the perfume raw material further comprises one or more of an oxygen atom and a nitrogen atom. 4. The personal care composition of claim 1 exhibits an anti-habituating effect on a consumer. 5. A personal care composition comprising a perfume, the perfume comprising, based on total perfume weight, a perfume raw material selected from the group consisting of: a. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiol moiety; b. from about 0.0000001% to about 10%, of a perfume raw material comprising a sulfide moiety; c. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiazole moiety; d. from about 0.0000001% to about 10%, of a perfume raw material comprising an oxathiane moiety; e. from about 0.00000005% to about 5%, of a perfume raw material comprising an isothiocyanate; f. from about 0.000001% to about 10%, of a perfume raw material comprising a oxygen, sulfur, and nitrogen; and g. mixtures thereof; such that the perfume raw material resists the fragrance habituation of a consumer to the personal care composition. 6. The personal care composition of claim 5, wherein the perfume raw material is selected from the group consisting of: a. from about 0.00001% to about 1%, of the perfume raw material comprising the thiol moiety; b. from about 0.00001% to about 1%, of the perfume raw material comprising the sulfide moiety; c. from about 0.0005% to about 1%, of the perfume raw material comprising the thiazole moiety; d. from about 0.00001% to about 1%, of the perfume raw material comprising the oxathiane moiety; e. from about 0.000001% to about 1%, of the perfume raw material comprising the isothiocyanate; and f. from about 0.0005% to about 1%, of the perfume raw material comprising the oxygen, sulfur, and nitrogen. 7. The personal care composition of claim 6, wherein the perfume raw material is selected from the group consisting of: a. from about 0.000025% to about 0.8%, of the perfume raw material comprising the thiol moiety; b. from about 0.000025% to about 0.5%, of the perfume raw material comprising the sulfide moiety; c. from about 0.001% to about 0.1%, of the perfume raw material comprising the thiazole moiety; d. from about 0.000025% to about 0.8%, of the perfume raw material comprising the oxathiane moiety; e. from about 0.000005% to about 0.5%, of the perfume raw material comprising the isothiocyanate; and f. from about 0.001% to about 0.1%, of the perfume raw material comprising the oxygen, sulfur, and nitrogen. 8. The personal care composition of claim 5, wherein: the thiol moiety is selected from the group consisting of 5-methyl-5-sulfanylhexan-3-one; 2-(4-methyl-1-cyclohex-3-enyl)propane-2-thiol; 5-methyl-2-(2-sulfanylpropan-2-yl)cyclohexan-1-one; 4,7,7-trimethyl-6-thiabicyclo[3.2.1]octane; 4-methoxy-2-methylbutane-2-thiol; methanethiol; ethanethiol; prop-2-ene-1-thiol; propane-2-thiol; 2-methylpropane-2-thiol; propane-1-thiol; butane-2-thiol; butane-1-thiol; 2-methylpropane-1-thiol; methyldisulfanylmethane; 2-methylbutane-2-thiol; 3-methylbutane-2-thiol; 3-methylbutane-2-thiol; pentane-2-thiol; pentane-1-thiol; 2-methylbutane-1-thiol; cyclopentanethiol; 3-methyldisulfanylprop-1-ene; methylsulfanyldisulfanylmethane; 1-methyldisulfanylpropane; ethane-1,2-dithiol; 1-(methyldisulfanyl)prop-1-ene; 3-sulfanylbutan-2-one; ethyldisulfanylethane; hexane-1-thiol; 1-ethyldisulfanylpropane; thiophene-2-thiol; propane-1,3-dithiol; 3-sulfanylpentan-2-one; 2-propan-2-yldisulfanylpropane; butane-1,4-dithiol; benzenethiol; ethyl sulfanyldisulfanylethane; 3-methylsulfanyldisulfanylprop-1-ene; 1-methyl sulfanyldisulfanylpropane; butane-2,3-dithiol; 4-methyl-4-sulfanylpentan-2-one; 3-prop-2-enyldisulfanylprop-1-ene; 1-methoxyhexane-3-thiol; ethyl 2-sulfanylpropanoate; 1-(prop-2-enyldisulfanyl)propane; 1-propyldisulfanylpropane; 1-(4-hydroxy-3-methoxyphenyl)ethanone butane-1,3-dithiol; 1-propyldisulfanylprop-1-ene; 2-methylbenzenethiol; thiophen-2-ylmethanethiol; 3-sulfanylbutan-2-ol; phenylmethanethiol pentane-1,5-dithiol; 2-ethylbenzenethiol; 3-prop-2-enylsulfanyldisulfanylprop-1-ene; methyldisulfanyldisulfanylmethane; 1-propylsulfanyldisulfanylpropane; 2,7,7-trimethylbicyclo[3.1.1]heptane-2-thiol; 2,6-dimethylbenzenethiol; 2-phenylethanethiol; hexane-1,6-dithiol; 2-(methyldisulfanylmethyl)furan; pyridin-2-ylmethanethiol; 2-methoxybenzenethiol; (7,7-dimethyl-2-bicyclo[3.1.1]heptanyl)methanethiol; methyldisulfanylbenzene; 1-butyldisulfanylbutane; (4-methoxyphenyl)methanethiol; 2-sulfanylpropanoic acid; ethyl 2-methyldisulfanylpropanoate; (2E)-3,7-dimethylocta-2,6-diene-1-thiol; 3,7-dimethylocta-2,6-diene-1-thiol; pyrazin-2-ylmethanethiol; methyldisulfanylmethylbenzene; 2-methyl-5-(1-sulfanylpropan-2-yl)cyclohexane-1-thiol; octane-1,8-dithiol; 2-pyrazin-2-ylethanethiol; naphthalene-2-thiol; 2-oxo-3-sulfanylpropanoic acid; 2-thiophen-2-yldisulfanylthiophene; cyclohexyldisulfanylcyclohexane; 2-(furan-2-ylmethyldisulfanylmethyl)furan; phenyldisulfanylbenzene; benzyldisulfanylmethylbenzene; 8-Hydroxy-5-quinolinesulfonic acid; bis(3-methylbutyl) 2-sulfanylbutanedioate; 2-aminoethanesulfonic acid; 2-phenyl-3H-benzimidazole-5-sulfonic acid; 2-methyl-2-sulfanylpentan-1-01; and mixtures thereof; the sulfide moiety is selected from the group consisting of 1-butylsulfanylbutane; ethyl 3-methyl sulfanylpropanoate; 2-(methylsulfanylmethyl)furan; methylsulfanylmethane; methylsulfanylethane; 3-methylsulfanylprop-1-ene; S-methyl ethanethioate; ethylsulfanylethane; 1-methylsulfanylpropane; S-ethyl ethanethioate; 1-methylsulfanylbutane; 2-propan-2-ylsulfanylpropane; bis(methylsulfanyl)methane; 1-ethylsulfanylpropane; thiolane; 1-propylsulfanylpropane; 1-ethylsulfanylbutane; S-ethyl propanethioate; S-methyl butanethioate; S-methyl 3-methylbutanethioate; 3-methylsulfanylpropanal; 3-prop-2-enylsulfanylprop-1-ene; methyl 2-methylsulfanylacetate; S-prop-2-enyl propanethioate; 1-methylsulfanylbutan-2-one; 4-methyl sulfanylbutan-2-one; 3-methylsulfanylpropan-1-am; 2,4,6-trimethyl-1,3,5-trithiane; 3-methylsulfanylbutanal; 2-methyl-1,3-thiazolidine; 2-methyl-4,5-dihydro-1,3-thiazole; ethyl 2-methylsulfanylacetate; methyl 3-methylsulfanylpropanoate; S-propan-2-yl 3-methylbutanethioate; 4-methyl-4-methylsulfanylpentan-2-one; 2-methyl-1,3-dithiolane; methyl 2-methylsulfanylbutanoate; S-methyl furan-2-carbothioate; S-propan-2-yl 3-methylbut-2-enethioate; thiolan-3-one; 3,5-diethyl-1,2,4-trithiolane; methylsulfanylmethylbenzene; 3-methyl sulfanylpropan-1-ol; 2-(propan-2-ylsulfanylmethyl)furan; 2-methyl-5-methylsulfanylfuran; S-(furan-2-ylmethyl)methanethioate; 1,2,4-trithiolane; 2-methylthiolan-3-one; 4-methylsulfanylbutan-1-ol; S-butan-2-yl 3-methylbutanethioate; S-butan-2-yl 3-methylbut-2-enethioate; S-(furan-2-ylmethyl)ethanethioate; 2-propyl-1,3-thiazolidine; 3-methyl-1,1-bis(methylsulfanyl)butane; 3-ethylsulfanylpropan-1-ol; S-methyl benzenecarbothioate; 3,5-dimethyl-1,2,4-trithiolane; S-butan-2-yl 2-methylbutanethioate; methylsulfanylbenzene; 1-pentylsulfanylpentane; (2R,4S)-2-methyl-4-propyl-1,3-oxathiane; 2-methyl-4-propyl-1,3-oxathiane; ethyl 2-methyl-2-methylsulfanylpropanoate; S-(furan-2-ylmethyl)propanethioate; 4,7,7-trimethyl-6-thiabicyclo[3.2.1]octane; 3-methyl-1,2,4-trithiane; methyl sulfanylmethyl hexanoate; 1-(4,5-dihydro-1,3-thiazol-2-yl)ethanone; 3-methylsulfanylpropanoic acid; 5-methylsulfanyl-2-(methylsulfanylmethyl)pent-2-enal; 4,5-dimethyl-2-(2-methylpropyl)-2,5-dihydro-1,3-thiazole; 3-methylsulfanylhexan-1-ol; 2-methyl-4,5-dihydrofuran-3-thiol acetate; 4-(3-oxobutylsulfanyl)butan-2-one; 3-methylsulfanylbutanoic acid; 2-methylsulfanylpyrazine; 2-methyl-3-methylsulfanylpyrazine; 2-(furan-2-ylmethylsulfanylmethyl)furan; 2-(methylsulfanylmethyl)pyrazine; 3,5-di(propan-2-yl)-1,2,4-trithiolane; 2-methylsulfanylphenol; 2-methyl-3-methylsulfanylpyrazine; ethyl 3-(furan-2-ylmethylsulfanyl)propanoate; 2,2,4,4,6,6-hexamethyl-1,3,5-trithiane; 2-methyl-5,7-dihydrothieno[3,4-d]pyrimidine; 2-amino-4-methylsulfanylbutanoic acid; (2S)-2-amino-4-methylsulfanylbutanoic acid; 2′,3a-dimethylspiro[6,6a-dihydro-5H-[1,3]dithiolo[4,5-b]furan-2,3′-oxolane]; 2,5-dimethyl-1,4-dithiane-2,5-diol; Methyl 2-thiofuroate; and mixtures thereof; the thiazole moiety is selected from the group consisting of 2-(2-methylpropyl)-1,3-thiazole; 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 4-methyl-2-propan-2-yl-1,3-thiazole; 1-(1,3-thiazol-2-yl)ethanone; 2,4,5-Trimethylthiazole; 2-isopropyl-4-methylthiazole; 4-vinyl-5-methylthiazole; 2,4-Dimethyl-5-acetylthiazole 1,3-thiazole; 4-methyl-1,3-thiazole; 2,4-dimethyl-1,3-thiazole; 4,5-dimethyl-1,3-thiazole; 2,5-dimethyl-1,3-thiazole; 5-ethenyl-4-methyl-1,3-thiazole; 2-ethyl-4-methyl-1,3-thiazole; 4-ethyl-2-methyl-1,3-thiazole; 2-propyl-1,3-thiazole; 2,4,5-trimethyl-1,3-thiazole; 2-ethyl-1,3-thiazole; 2-ethoxy-1,3-thiazole; 2-butan-2-yl-1,3-thiazole; 5-methoxy-2-methyl-1,3-thiazole; 2-ethyl-4,5-dimethyl-1,3-thiazole; 1,3-benzothiazole; 2,5-diethyl-4-methyl-1,3-thiazole; 1-(1,3-thiazol-2-yl)propan-1-one; 4,5-dimethyl-2-(2-methylpropyl)-1,3-thiazole; 2-methyl-1,3-benzothiazole; 1-(2,4-dimethyl-1,3-thiazol-5-yl)ethanone; 4-methyl-2-propan-2-yl-1,3-thiazole; and mixtures thereof; the oxathiane moiety is selected from the group consisting of (2R,4S)-2-methyl-4-propyl-1,3-oxathiane, 2-methyl-4-propyl-1,3-oxathiane, 2-pentyl-4-propyl-1,3-oxathiane; and mixtures thereof; and the perfume raw material comprising oxygen, sulfur, and nitrogen is selected from the group consisting of 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 1-(1,3-thiazol-2-yl)ethanone; 6-methyl-7-Oxa-1-thia-4-azaspiro[4.4]nonane; 2-[(furan-2-ylmethyl)sulfanyl]-5-methylpyrazine; 2,4-Dimethyl-5-acetylthiazole; 2-ethoxy-1,3-thiazole; 5-methoxy-2-methyl-1,3-thiazole; 1-(4,5-dihydro-1,3-thiazol-2-yl)ethanone; 1-(1,3-thiazol-2-yl)propan-1-one; 1-(2,4-dimethyl-1,3-thiazol-5-yl)ethanone; 2-amino-4-methylsulfanylbutanoic acid; (2S)-2-amino-4-methylsulfanylbutanoic acid; 8-Hydroxy-5-quinolinesulfonic acid; 2-aminoethanesulfonic acid; 2-phenyl-3H-benzimidazole-5-sulfonic acid; and mixtures thereof. 9. The personal care composition of claim 8, wherein: the thiol moiety is selected from the group consisting of 5-methyl-5-sulfanylhexan-3-one; 2-(4-methyl-1-cyclohex-3-enyl)propane-2-thiol; 5-methyl-2-(2-sulfanylpropan-2-yl)cyclohexan-1-one; 4,7,7-trimethyl-6-thiabicyclo[3.2.1]octane; 4-methoxy-2-methylbutane-2-thiol; and mixtures thereof; the sulfide moiety is selected from the group consisting of 1-butylsulfanylbutane; ethyl 3-methylsulfanylpropanoate; 2-(methylsulfanylmethyl)furan; and mixtures thereof; the thiazole moiety is selected from the group consisting of 2-(2-methylpropyl)-1,3-thiazole; 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 4-methyl-2-propan-2-yl-1,3-thiazole; 4-methyl-2-propan-2-yl-1,3-thiazole; 1-(1,3-thiazol-2-yl)ethanone; and mixtures thereof; the oxathiane moiety is (2R,4S)-2-methyl-4-propyl-1,3-oxathiane; and the perfume raw material comprising oxygen, sulfur, and nitrogen is selected from the group consisting of 2-(4-methyl-1,3-thiazol-5-yl)ethanol, 1-(1,3-thiazol-2-yl)ethanone; 6-methyl-7-Oxa-1-thia-4-azaspiro[4.4]nonane; and mixtures thereof. 10. The personal care composition of claim 5, wherein the perfume raw materials are: (a) 5-mercapto-5-methyl-3-hexanone; p-mentha-8-thiol-3-one; and 1-para-menthene-8-thiol; or (b) 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 2-(2-methylpropyl)-1,3-thiazol; and 4-methyl-2-propan-2-yl-1-,3-thiazole. 11. The personal care composition of claim 5, wherein the perfume raw material is 2-methyl-3-methylsulfanylpyrazine. 12. The personal care composition of claim 5 exhibits an anti-habituating effect on a consumer. 13. The personal care composition of 5 is a body wash. 14. The personal care composition of claim 13, wherein the body wash is a single-phase body wash or a multi-phase body wash. 15. The personal care composition of claim 5 is a shampoo or lotion. 16. The personal care composition of claim 5 is a bar soap. 17. The personal care composition of claim 5, wherein the personal care composition further comprises a perfume delivery system configured to deliver the perfume and perfume raw materials. 18. The personal care composition of claim 5, further comprising a perfume raw material comprising a nitrogen atom. 19. The personal care composition of claim 18, wherein the perfume raw material comprising a nitrogen atom is (NE)-N-[(6E)-2,4,4,7-tetramethylnona-6,8-dien-3-ylidene]hydroxylamine). 20. A method of resisting the fragrance habituation of a personal care composition, the method comprising: forming a personal care composition comprising a perfume, the perfume comprising, based on total perfume weight, a perfume raw material selected from the group consisting of: a. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiol moiety; b. from about 0.0000001% to about 10%, of a perfume raw material comprising a sulfide moiety; c. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiazole moiety; d. from about 0.0000001% to about 10%, of a perfume raw material comprising a oxathiane moiety; e. from about 0.00000005% to about 5%, of a perfume raw material comprising an isothiocyanate; f. from about 0.000001% to about 10%, of a perfume raw material comprising a sulfur, oxygen and nitrogen; and g. mixtures thereof.
The present application relates to personal care compositions including perfumes and sulfur-based perfume raw materials, and methods for making and using the personal care compositions to resist consumer fragrance habituation.1. A personal care composition comprising a perfume, the perfume comprising, based on total perfume weight, from about 0.000001% to about 10%, of a perfume raw material comprising a sulfur atom, such that the perfume raw material resists the fragrance habituation of a consumer to the personal care composition. 2. The personal care composition of claim 1 comprising from about 0.001% to about 0.1%, of the perfume raw material. 3. The personal care composition of claim 1, wherein the perfume raw material further comprises one or more of an oxygen atom and a nitrogen atom. 4. The personal care composition of claim 1 exhibits an anti-habituating effect on a consumer. 5. A personal care composition comprising a perfume, the perfume comprising, based on total perfume weight, a perfume raw material selected from the group consisting of: a. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiol moiety; b. from about 0.0000001% to about 10%, of a perfume raw material comprising a sulfide moiety; c. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiazole moiety; d. from about 0.0000001% to about 10%, of a perfume raw material comprising an oxathiane moiety; e. from about 0.00000005% to about 5%, of a perfume raw material comprising an isothiocyanate; f. from about 0.000001% to about 10%, of a perfume raw material comprising a oxygen, sulfur, and nitrogen; and g. mixtures thereof; such that the perfume raw material resists the fragrance habituation of a consumer to the personal care composition. 6. The personal care composition of claim 5, wherein the perfume raw material is selected from the group consisting of: a. from about 0.00001% to about 1%, of the perfume raw material comprising the thiol moiety; b. from about 0.00001% to about 1%, of the perfume raw material comprising the sulfide moiety; c. from about 0.0005% to about 1%, of the perfume raw material comprising the thiazole moiety; d. from about 0.00001% to about 1%, of the perfume raw material comprising the oxathiane moiety; e. from about 0.000001% to about 1%, of the perfume raw material comprising the isothiocyanate; and f. from about 0.0005% to about 1%, of the perfume raw material comprising the oxygen, sulfur, and nitrogen. 7. The personal care composition of claim 6, wherein the perfume raw material is selected from the group consisting of: a. from about 0.000025% to about 0.8%, of the perfume raw material comprising the thiol moiety; b. from about 0.000025% to about 0.5%, of the perfume raw material comprising the sulfide moiety; c. from about 0.001% to about 0.1%, of the perfume raw material comprising the thiazole moiety; d. from about 0.000025% to about 0.8%, of the perfume raw material comprising the oxathiane moiety; e. from about 0.000005% to about 0.5%, of the perfume raw material comprising the isothiocyanate; and f. from about 0.001% to about 0.1%, of the perfume raw material comprising the oxygen, sulfur, and nitrogen. 8. The personal care composition of claim 5, wherein: the thiol moiety is selected from the group consisting of 5-methyl-5-sulfanylhexan-3-one; 2-(4-methyl-1-cyclohex-3-enyl)propane-2-thiol; 5-methyl-2-(2-sulfanylpropan-2-yl)cyclohexan-1-one; 4,7,7-trimethyl-6-thiabicyclo[3.2.1]octane; 4-methoxy-2-methylbutane-2-thiol; methanethiol; ethanethiol; prop-2-ene-1-thiol; propane-2-thiol; 2-methylpropane-2-thiol; propane-1-thiol; butane-2-thiol; butane-1-thiol; 2-methylpropane-1-thiol; methyldisulfanylmethane; 2-methylbutane-2-thiol; 3-methylbutane-2-thiol; 3-methylbutane-2-thiol; pentane-2-thiol; pentane-1-thiol; 2-methylbutane-1-thiol; cyclopentanethiol; 3-methyldisulfanylprop-1-ene; methylsulfanyldisulfanylmethane; 1-methyldisulfanylpropane; ethane-1,2-dithiol; 1-(methyldisulfanyl)prop-1-ene; 3-sulfanylbutan-2-one; ethyldisulfanylethane; hexane-1-thiol; 1-ethyldisulfanylpropane; thiophene-2-thiol; propane-1,3-dithiol; 3-sulfanylpentan-2-one; 2-propan-2-yldisulfanylpropane; butane-1,4-dithiol; benzenethiol; ethyl sulfanyldisulfanylethane; 3-methylsulfanyldisulfanylprop-1-ene; 1-methyl sulfanyldisulfanylpropane; butane-2,3-dithiol; 4-methyl-4-sulfanylpentan-2-one; 3-prop-2-enyldisulfanylprop-1-ene; 1-methoxyhexane-3-thiol; ethyl 2-sulfanylpropanoate; 1-(prop-2-enyldisulfanyl)propane; 1-propyldisulfanylpropane; 1-(4-hydroxy-3-methoxyphenyl)ethanone butane-1,3-dithiol; 1-propyldisulfanylprop-1-ene; 2-methylbenzenethiol; thiophen-2-ylmethanethiol; 3-sulfanylbutan-2-ol; phenylmethanethiol pentane-1,5-dithiol; 2-ethylbenzenethiol; 3-prop-2-enylsulfanyldisulfanylprop-1-ene; methyldisulfanyldisulfanylmethane; 1-propylsulfanyldisulfanylpropane; 2,7,7-trimethylbicyclo[3.1.1]heptane-2-thiol; 2,6-dimethylbenzenethiol; 2-phenylethanethiol; hexane-1,6-dithiol; 2-(methyldisulfanylmethyl)furan; pyridin-2-ylmethanethiol; 2-methoxybenzenethiol; (7,7-dimethyl-2-bicyclo[3.1.1]heptanyl)methanethiol; methyldisulfanylbenzene; 1-butyldisulfanylbutane; (4-methoxyphenyl)methanethiol; 2-sulfanylpropanoic acid; ethyl 2-methyldisulfanylpropanoate; (2E)-3,7-dimethylocta-2,6-diene-1-thiol; 3,7-dimethylocta-2,6-diene-1-thiol; pyrazin-2-ylmethanethiol; methyldisulfanylmethylbenzene; 2-methyl-5-(1-sulfanylpropan-2-yl)cyclohexane-1-thiol; octane-1,8-dithiol; 2-pyrazin-2-ylethanethiol; naphthalene-2-thiol; 2-oxo-3-sulfanylpropanoic acid; 2-thiophen-2-yldisulfanylthiophene; cyclohexyldisulfanylcyclohexane; 2-(furan-2-ylmethyldisulfanylmethyl)furan; phenyldisulfanylbenzene; benzyldisulfanylmethylbenzene; 8-Hydroxy-5-quinolinesulfonic acid; bis(3-methylbutyl) 2-sulfanylbutanedioate; 2-aminoethanesulfonic acid; 2-phenyl-3H-benzimidazole-5-sulfonic acid; 2-methyl-2-sulfanylpentan-1-01; and mixtures thereof; the sulfide moiety is selected from the group consisting of 1-butylsulfanylbutane; ethyl 3-methyl sulfanylpropanoate; 2-(methylsulfanylmethyl)furan; methylsulfanylmethane; methylsulfanylethane; 3-methylsulfanylprop-1-ene; S-methyl ethanethioate; ethylsulfanylethane; 1-methylsulfanylpropane; S-ethyl ethanethioate; 1-methylsulfanylbutane; 2-propan-2-ylsulfanylpropane; bis(methylsulfanyl)methane; 1-ethylsulfanylpropane; thiolane; 1-propylsulfanylpropane; 1-ethylsulfanylbutane; S-ethyl propanethioate; S-methyl butanethioate; S-methyl 3-methylbutanethioate; 3-methylsulfanylpropanal; 3-prop-2-enylsulfanylprop-1-ene; methyl 2-methylsulfanylacetate; S-prop-2-enyl propanethioate; 1-methylsulfanylbutan-2-one; 4-methyl sulfanylbutan-2-one; 3-methylsulfanylpropan-1-am; 2,4,6-trimethyl-1,3,5-trithiane; 3-methylsulfanylbutanal; 2-methyl-1,3-thiazolidine; 2-methyl-4,5-dihydro-1,3-thiazole; ethyl 2-methylsulfanylacetate; methyl 3-methylsulfanylpropanoate; S-propan-2-yl 3-methylbutanethioate; 4-methyl-4-methylsulfanylpentan-2-one; 2-methyl-1,3-dithiolane; methyl 2-methylsulfanylbutanoate; S-methyl furan-2-carbothioate; S-propan-2-yl 3-methylbut-2-enethioate; thiolan-3-one; 3,5-diethyl-1,2,4-trithiolane; methylsulfanylmethylbenzene; 3-methyl sulfanylpropan-1-ol; 2-(propan-2-ylsulfanylmethyl)furan; 2-methyl-5-methylsulfanylfuran; S-(furan-2-ylmethyl)methanethioate; 1,2,4-trithiolane; 2-methylthiolan-3-one; 4-methylsulfanylbutan-1-ol; S-butan-2-yl 3-methylbutanethioate; S-butan-2-yl 3-methylbut-2-enethioate; S-(furan-2-ylmethyl)ethanethioate; 2-propyl-1,3-thiazolidine; 3-methyl-1,1-bis(methylsulfanyl)butane; 3-ethylsulfanylpropan-1-ol; S-methyl benzenecarbothioate; 3,5-dimethyl-1,2,4-trithiolane; S-butan-2-yl 2-methylbutanethioate; methylsulfanylbenzene; 1-pentylsulfanylpentane; (2R,4S)-2-methyl-4-propyl-1,3-oxathiane; 2-methyl-4-propyl-1,3-oxathiane; ethyl 2-methyl-2-methylsulfanylpropanoate; S-(furan-2-ylmethyl)propanethioate; 4,7,7-trimethyl-6-thiabicyclo[3.2.1]octane; 3-methyl-1,2,4-trithiane; methyl sulfanylmethyl hexanoate; 1-(4,5-dihydro-1,3-thiazol-2-yl)ethanone; 3-methylsulfanylpropanoic acid; 5-methylsulfanyl-2-(methylsulfanylmethyl)pent-2-enal; 4,5-dimethyl-2-(2-methylpropyl)-2,5-dihydro-1,3-thiazole; 3-methylsulfanylhexan-1-ol; 2-methyl-4,5-dihydrofuran-3-thiol acetate; 4-(3-oxobutylsulfanyl)butan-2-one; 3-methylsulfanylbutanoic acid; 2-methylsulfanylpyrazine; 2-methyl-3-methylsulfanylpyrazine; 2-(furan-2-ylmethylsulfanylmethyl)furan; 2-(methylsulfanylmethyl)pyrazine; 3,5-di(propan-2-yl)-1,2,4-trithiolane; 2-methylsulfanylphenol; 2-methyl-3-methylsulfanylpyrazine; ethyl 3-(furan-2-ylmethylsulfanyl)propanoate; 2,2,4,4,6,6-hexamethyl-1,3,5-trithiane; 2-methyl-5,7-dihydrothieno[3,4-d]pyrimidine; 2-amino-4-methylsulfanylbutanoic acid; (2S)-2-amino-4-methylsulfanylbutanoic acid; 2′,3a-dimethylspiro[6,6a-dihydro-5H-[1,3]dithiolo[4,5-b]furan-2,3′-oxolane]; 2,5-dimethyl-1,4-dithiane-2,5-diol; Methyl 2-thiofuroate; and mixtures thereof; the thiazole moiety is selected from the group consisting of 2-(2-methylpropyl)-1,3-thiazole; 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 4-methyl-2-propan-2-yl-1,3-thiazole; 1-(1,3-thiazol-2-yl)ethanone; 2,4,5-Trimethylthiazole; 2-isopropyl-4-methylthiazole; 4-vinyl-5-methylthiazole; 2,4-Dimethyl-5-acetylthiazole 1,3-thiazole; 4-methyl-1,3-thiazole; 2,4-dimethyl-1,3-thiazole; 4,5-dimethyl-1,3-thiazole; 2,5-dimethyl-1,3-thiazole; 5-ethenyl-4-methyl-1,3-thiazole; 2-ethyl-4-methyl-1,3-thiazole; 4-ethyl-2-methyl-1,3-thiazole; 2-propyl-1,3-thiazole; 2,4,5-trimethyl-1,3-thiazole; 2-ethyl-1,3-thiazole; 2-ethoxy-1,3-thiazole; 2-butan-2-yl-1,3-thiazole; 5-methoxy-2-methyl-1,3-thiazole; 2-ethyl-4,5-dimethyl-1,3-thiazole; 1,3-benzothiazole; 2,5-diethyl-4-methyl-1,3-thiazole; 1-(1,3-thiazol-2-yl)propan-1-one; 4,5-dimethyl-2-(2-methylpropyl)-1,3-thiazole; 2-methyl-1,3-benzothiazole; 1-(2,4-dimethyl-1,3-thiazol-5-yl)ethanone; 4-methyl-2-propan-2-yl-1,3-thiazole; and mixtures thereof; the oxathiane moiety is selected from the group consisting of (2R,4S)-2-methyl-4-propyl-1,3-oxathiane, 2-methyl-4-propyl-1,3-oxathiane, 2-pentyl-4-propyl-1,3-oxathiane; and mixtures thereof; and the perfume raw material comprising oxygen, sulfur, and nitrogen is selected from the group consisting of 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 1-(1,3-thiazol-2-yl)ethanone; 6-methyl-7-Oxa-1-thia-4-azaspiro[4.4]nonane; 2-[(furan-2-ylmethyl)sulfanyl]-5-methylpyrazine; 2,4-Dimethyl-5-acetylthiazole; 2-ethoxy-1,3-thiazole; 5-methoxy-2-methyl-1,3-thiazole; 1-(4,5-dihydro-1,3-thiazol-2-yl)ethanone; 1-(1,3-thiazol-2-yl)propan-1-one; 1-(2,4-dimethyl-1,3-thiazol-5-yl)ethanone; 2-amino-4-methylsulfanylbutanoic acid; (2S)-2-amino-4-methylsulfanylbutanoic acid; 8-Hydroxy-5-quinolinesulfonic acid; 2-aminoethanesulfonic acid; 2-phenyl-3H-benzimidazole-5-sulfonic acid; and mixtures thereof. 9. The personal care composition of claim 8, wherein: the thiol moiety is selected from the group consisting of 5-methyl-5-sulfanylhexan-3-one; 2-(4-methyl-1-cyclohex-3-enyl)propane-2-thiol; 5-methyl-2-(2-sulfanylpropan-2-yl)cyclohexan-1-one; 4,7,7-trimethyl-6-thiabicyclo[3.2.1]octane; 4-methoxy-2-methylbutane-2-thiol; and mixtures thereof; the sulfide moiety is selected from the group consisting of 1-butylsulfanylbutane; ethyl 3-methylsulfanylpropanoate; 2-(methylsulfanylmethyl)furan; and mixtures thereof; the thiazole moiety is selected from the group consisting of 2-(2-methylpropyl)-1,3-thiazole; 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 4-methyl-2-propan-2-yl-1,3-thiazole; 4-methyl-2-propan-2-yl-1,3-thiazole; 1-(1,3-thiazol-2-yl)ethanone; and mixtures thereof; the oxathiane moiety is (2R,4S)-2-methyl-4-propyl-1,3-oxathiane; and the perfume raw material comprising oxygen, sulfur, and nitrogen is selected from the group consisting of 2-(4-methyl-1,3-thiazol-5-yl)ethanol, 1-(1,3-thiazol-2-yl)ethanone; 6-methyl-7-Oxa-1-thia-4-azaspiro[4.4]nonane; and mixtures thereof. 10. The personal care composition of claim 5, wherein the perfume raw materials are: (a) 5-mercapto-5-methyl-3-hexanone; p-mentha-8-thiol-3-one; and 1-para-menthene-8-thiol; or (b) 2-(4-methyl-1,3-thiazol-5-yl)ethanol; 2-(2-methylpropyl)-1,3-thiazol; and 4-methyl-2-propan-2-yl-1-,3-thiazole. 11. The personal care composition of claim 5, wherein the perfume raw material is 2-methyl-3-methylsulfanylpyrazine. 12. The personal care composition of claim 5 exhibits an anti-habituating effect on a consumer. 13. The personal care composition of 5 is a body wash. 14. The personal care composition of claim 13, wherein the body wash is a single-phase body wash or a multi-phase body wash. 15. The personal care composition of claim 5 is a shampoo or lotion. 16. The personal care composition of claim 5 is a bar soap. 17. The personal care composition of claim 5, wherein the personal care composition further comprises a perfume delivery system configured to deliver the perfume and perfume raw materials. 18. The personal care composition of claim 5, further comprising a perfume raw material comprising a nitrogen atom. 19. The personal care composition of claim 18, wherein the perfume raw material comprising a nitrogen atom is (NE)-N-[(6E)-2,4,4,7-tetramethylnona-6,8-dien-3-ylidene]hydroxylamine). 20. A method of resisting the fragrance habituation of a personal care composition, the method comprising: forming a personal care composition comprising a perfume, the perfume comprising, based on total perfume weight, a perfume raw material selected from the group consisting of: a. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiol moiety; b. from about 0.0000001% to about 10%, of a perfume raw material comprising a sulfide moiety; c. from about 0.0000001% to about 10%, of a perfume raw material comprising a thiazole moiety; d. from about 0.0000001% to about 10%, of a perfume raw material comprising a oxathiane moiety; e. from about 0.00000005% to about 5%, of a perfume raw material comprising an isothiocyanate; f. from about 0.000001% to about 10%, of a perfume raw material comprising a sulfur, oxygen and nitrogen; and g. mixtures thereof.
1,600
313
14,445,434
1,613
A method of improving the look and feel of skin by using a two-step regimen that includes applying an underlying layer of a skin care composition and applying an overlying layer of a finisher composition on top of the underlying skin care composition layer. The finisher composition is an oil-in-water emulsion comprising a continuous aqueous phase, a dispersed oil phase, and from 10 to 25 wt % of substantially spherical particles. The oil phase includes non-volatile oil that is at least 50 wt % of a liquid UV agent. The spherical particles can be starch particles, silicone elastomer particles or combinations of these. The finisher composition is generally free of glycerin or other humectants.
1. A method of improving the look and feel of skin, comprising: a. applying an underlying layer of a skin care composition to a target portion of skin; and b. applying an overlying layer of a finisher composition on top of the underlying skin care composition layer, wherein the finisher composition is an oil-in-water emulsion comprising i. a continuous aqueous phase that include from about 20 to 85% by weight of the finisher composition of water, ii. a dispersed oil phase comprising a non-volatile oil, the non-volatile oil comprising a liquid UV agent present at an amount of at least 50% by weight of the non-volatile oil, iii. from 10 to 25%, by weight of substantially spherical particles selected from the group consisting of starch particles, silicone elastomer particles and combinations thereof, wherein the particles have a particle size of from about 2 to 40 microns and a weight ratio of the non-volatile oil to the particles is from about 1:10 to about 1:1, iv. optionally, from about 1 to 20 wt % of a volatile oil, and v. wherein the finisher composition is substantially free of glycerin. 2. The finisher composition of claim 1, wherein the weight ratio of the non-volatile oil to the particles is from about 1:5 to about 4:5. 3. The finisher composition of claim 2, wherein the weight ratio of non-volatile oil to the particles is from about 1:4 to about 3:5 4. The finisher composition of claim 1, wherein the liquid UV agent is present at an amount of at least 90% by weight of the non-volatile oil. 5. The finisher composition of claim 4, wherein the non-volatile oil consists of the liquid UV agent. 6. The finisher composition of claim 1, where the liquid UV agent further comprises an oil soluble solid UV agent. 7. The finisher composition of claim 1, wherein the liquid UV agent comprises Ethylhexyl Dimethyl PABA, Ethylhexyl Methoxycinnamate (octinoxate), Ethylhexyl Salicylate (octisalate), Homosalate, Isoamyl p-Methoxycinnamate (amiloxate), Menthyl Anthranilate (meradimate), Octocrylene, Polysilicone-15 (diethylbenzylidene malonate dimethicone), Benzophenone-3 (oxybenzone), Benzophenone-9 (dioxybenzone), Bis-Ethylhexyloxyphenol Methoxyphenyl Triazine (bemotrizinol), Butyl Methoxydibenzoylmethane (avobenzone), Diethylamino Hydroxybenzoyl Hexyl Benzoate, Diethylhexyl Butamido Triazone (iscotrizinol), Drometrizole Trisiloxane, Ethylhexyl Triazone (octyl triazone), 4-Methylbenzylidene Camphor (enacamene) or a combination of these. 8. The finisher composition of claim 1, wherein the substantially spherical particles are silicone elastomer particles and the particles are present at an amount of from about 14 wt % to about 20 wt %. 9. The finisher composition of claim 1, wherein the substantially spherical particles are starch particles selected from the group consisting of tapioca starch, corn starch, potato starch, glyceryl starch, aluminum starch octenylsuccinate, calcium starch octenylsuccinate, polymethylsilsesquioxane coated tapioca starch, and combinations thereof. 10. The finisher composition of claim 9, wherein the substantially spherical starch particles have a mean particle size of from 5 to 30 microns. 11. The composition of claim 1, further comprising a chroma of less than about 10 according to the Chroma Method. 12. The composition of claim 1, further comprising a chroma of less than about 6 according to the Chroma Method. 13. The finisher composition of claim 1, wherein the composition is substantially free of humectants. 14. A method of improving the look and feel of skin, comprising: a. applying an underlying layer of a skin care composition to a target portion of skin; and b. applying an overlying layer of a finisher composition on top of the underlying skin care composition layer, wherein the finisher composition is an oil-in-water emulsion comprising i. a continuous aqueous phase comprising from about 20 to 85 wt % of water, ii. a dispersed oil phase comprising a non-volatile oil, wherein the non-volatile oil comprises a liquid UV agent present at an amount of at least 50% by weight of the non-volatile oil, iii. from about 10 to 25 wt % of substantially spherical particles selected from the group consisting of starch particles, silicone elastomer particles, and combinations thereof, wherein the particles have a mean particle size of from about 2 to 40 microns and wherein a weight ratio of the non-volatile oil to the particles is from about 1:10 to about 1:1, iv. less than 1 wt % of a pigment, and v. a contrast ratio of less than about 20 according to the Contrast Ratio method. 15. The composition of claim 14, wherein the composition has a contrast ratio of less than about 10. 16. The composition of claim 14, wherein the composition is substantially free of pigments. 17. The composition of claim 14, wherein the composition includes less than 4% non-spherical particles. 18. The composition of claim 14, wherein the underlying layer of skin care composition comprises a humectant. 19. The composition of claim 18, wherein the humectant is glycerin.
A method of improving the look and feel of skin by using a two-step regimen that includes applying an underlying layer of a skin care composition and applying an overlying layer of a finisher composition on top of the underlying skin care composition layer. The finisher composition is an oil-in-water emulsion comprising a continuous aqueous phase, a dispersed oil phase, and from 10 to 25 wt % of substantially spherical particles. The oil phase includes non-volatile oil that is at least 50 wt % of a liquid UV agent. The spherical particles can be starch particles, silicone elastomer particles or combinations of these. The finisher composition is generally free of glycerin or other humectants.1. A method of improving the look and feel of skin, comprising: a. applying an underlying layer of a skin care composition to a target portion of skin; and b. applying an overlying layer of a finisher composition on top of the underlying skin care composition layer, wherein the finisher composition is an oil-in-water emulsion comprising i. a continuous aqueous phase that include from about 20 to 85% by weight of the finisher composition of water, ii. a dispersed oil phase comprising a non-volatile oil, the non-volatile oil comprising a liquid UV agent present at an amount of at least 50% by weight of the non-volatile oil, iii. from 10 to 25%, by weight of substantially spherical particles selected from the group consisting of starch particles, silicone elastomer particles and combinations thereof, wherein the particles have a particle size of from about 2 to 40 microns and a weight ratio of the non-volatile oil to the particles is from about 1:10 to about 1:1, iv. optionally, from about 1 to 20 wt % of a volatile oil, and v. wherein the finisher composition is substantially free of glycerin. 2. The finisher composition of claim 1, wherein the weight ratio of the non-volatile oil to the particles is from about 1:5 to about 4:5. 3. The finisher composition of claim 2, wherein the weight ratio of non-volatile oil to the particles is from about 1:4 to about 3:5 4. The finisher composition of claim 1, wherein the liquid UV agent is present at an amount of at least 90% by weight of the non-volatile oil. 5. The finisher composition of claim 4, wherein the non-volatile oil consists of the liquid UV agent. 6. The finisher composition of claim 1, where the liquid UV agent further comprises an oil soluble solid UV agent. 7. The finisher composition of claim 1, wherein the liquid UV agent comprises Ethylhexyl Dimethyl PABA, Ethylhexyl Methoxycinnamate (octinoxate), Ethylhexyl Salicylate (octisalate), Homosalate, Isoamyl p-Methoxycinnamate (amiloxate), Menthyl Anthranilate (meradimate), Octocrylene, Polysilicone-15 (diethylbenzylidene malonate dimethicone), Benzophenone-3 (oxybenzone), Benzophenone-9 (dioxybenzone), Bis-Ethylhexyloxyphenol Methoxyphenyl Triazine (bemotrizinol), Butyl Methoxydibenzoylmethane (avobenzone), Diethylamino Hydroxybenzoyl Hexyl Benzoate, Diethylhexyl Butamido Triazone (iscotrizinol), Drometrizole Trisiloxane, Ethylhexyl Triazone (octyl triazone), 4-Methylbenzylidene Camphor (enacamene) or a combination of these. 8. The finisher composition of claim 1, wherein the substantially spherical particles are silicone elastomer particles and the particles are present at an amount of from about 14 wt % to about 20 wt %. 9. The finisher composition of claim 1, wherein the substantially spherical particles are starch particles selected from the group consisting of tapioca starch, corn starch, potato starch, glyceryl starch, aluminum starch octenylsuccinate, calcium starch octenylsuccinate, polymethylsilsesquioxane coated tapioca starch, and combinations thereof. 10. The finisher composition of claim 9, wherein the substantially spherical starch particles have a mean particle size of from 5 to 30 microns. 11. The composition of claim 1, further comprising a chroma of less than about 10 according to the Chroma Method. 12. The composition of claim 1, further comprising a chroma of less than about 6 according to the Chroma Method. 13. The finisher composition of claim 1, wherein the composition is substantially free of humectants. 14. A method of improving the look and feel of skin, comprising: a. applying an underlying layer of a skin care composition to a target portion of skin; and b. applying an overlying layer of a finisher composition on top of the underlying skin care composition layer, wherein the finisher composition is an oil-in-water emulsion comprising i. a continuous aqueous phase comprising from about 20 to 85 wt % of water, ii. a dispersed oil phase comprising a non-volatile oil, wherein the non-volatile oil comprises a liquid UV agent present at an amount of at least 50% by weight of the non-volatile oil, iii. from about 10 to 25 wt % of substantially spherical particles selected from the group consisting of starch particles, silicone elastomer particles, and combinations thereof, wherein the particles have a mean particle size of from about 2 to 40 microns and wherein a weight ratio of the non-volatile oil to the particles is from about 1:10 to about 1:1, iv. less than 1 wt % of a pigment, and v. a contrast ratio of less than about 20 according to the Contrast Ratio method. 15. The composition of claim 14, wherein the composition has a contrast ratio of less than about 10. 16. The composition of claim 14, wherein the composition is substantially free of pigments. 17. The composition of claim 14, wherein the composition includes less than 4% non-spherical particles. 18. The composition of claim 14, wherein the underlying layer of skin care composition comprises a humectant. 19. The composition of claim 18, wherein the humectant is glycerin.
1,600
314
15,322,881
1,646
A method for treating ocular-disorders which, involve angiogenesis or neovascularization as part of their pathologic process. These types of retinal disorders can be categorized as (a) retinal vascular disorders (such as diabetic retinopathy, retinal vein occlusions and retinopathy of prematurity), (b) subretinal neovascular disorders (such as neovascular age-related macular degeneration, ocular histoplasmosis, pathologic myopia) (c) intraocular tumors (such as ocular melanoma, ocular lymphoma and retino-blastoma). The treatment for these ocular disorder involves the administration of an effective amount of human chorionic gonadotropin (hCG) antagonists, luteinizing hormone (LH) antagonists or hCG/LH receptor antagonists, optionally in combination with at least one additional bioactive agent.
1. A method for reducing the likelihood or inhibiting the progression of an ocular disorder in a patient in need comprising administering to said patient an effective amount of a hCG/LH antagonist, optionally in combination with an additional bioactive agent effective for ameliorating the effects of said ocular disorder. 2. The method of claim 1 in which said hCG/LH antagonist is a small molecule entity having a molecular weight of less than 150 kDa. 3. The method of claim 1 in which said hCG/LH antagonist is a small molecule entity having a molecular weight of from 20 to 80 kDa. 4. The method of claim 3 in which said hCG/LH antagonist is applied topically to the eye. 5. The method of claim 1 in which said hCG/LH antagonist is a small molecule entity having a molecular weight of 30-50 kDa. 6. The method of claim 5 in which said hCG/LH antagonist is applied topically to the eye. 7. The method according to claim 2 wherein said hCG/LH antagonist is a deglycosylated hCG, a mutant hCG or an hCG mutant which has also been deglycosylated. 8. The method according to claim 2 wherein said hCG/LH antagonist is deglycosylated hCG. 9. The method according to claim 2 wherein said hCG/LH antagonist is a mutant hCG. 10. The method according to claim 2 wherein said hCG/LH antagonist is a mutant hCG which is also deglycosylated. 11. The method according to claim 2 wherein said deglycosylated hCG comprises less N-linked carbohydrates than naturally occurring hCG. 12. The method according to claim 8 wherein said deglycosylated hCG has at least 55% less carbohydrates than naturally occurring hCG. 13. The method according to claim 8 wherein said deglycosylated hCG has at least 95% less carbohydrates than naturally occurring hCG. 14. The method according to claim 1 wherein said hCG/LH antagonist is coadministered in combination with an additional bioactive agent. 15. The method according to claim 14 wherein said additional bioactive agent is at least one agent selected from the group consisting of ranibizumab, aflibercept, pegatanib, verteporfin, triamcinolone and bevacizumab. 16. The method according to claim 14 wherein the ocular disorder is a retinal disorder. 17. The method according to claim 1 wherein said ocular disorder is selected from the group consisting of diabetic retinopathy, retinal vein occlusions, and retinopathy of prematurity. 18. The method according to claim 1 wherein said ocular disorder is selected from a group of subretinal neovascular disorders selected from the group consisting of macular degeneration, ocular histoplasmosis, and pathologic myopia. 19. The method according to claim 1 wherein said ocular disorder is selected from the group consisting of intraocular tumors which are NOT known to secrete hCG or LH. 20. The method according to claim 1 wherein said hCG/LH antagonists are hCG/LH antibodies. 21. A pharmaceutical composition comprising an effective amount of an hCG/LH antagonist in combination with at least one agent selected from the groups consisting of ranibizumab, aflibercept, pegatanib, verteporfin, triamcinolone and bevacizumab, in combination with a carrier, additive and/or excipient. 22. A pharmaceutical composition comprising an ophthalmic solution of a small molecule hCG/LH antagonist having a molecular weight of from 20 to 80 kDa. 23. The pharmaceutical composition of claim 22 in which said small molecule hCG/LH antagonist has a molecular weight of 30-50 kDa. 24. A pharmaceutical composition comprising a topical application eye drop solution of a small molecule hCG/LH antagonist having a molecular weight of from 20 to 80 kDa. 25. The pharmaceutical composition of claim 24 in which said small molecule hCG/LH antagonist has a molecular weight of 30-50 kDa.
A method for treating ocular-disorders which, involve angiogenesis or neovascularization as part of their pathologic process. These types of retinal disorders can be categorized as (a) retinal vascular disorders (such as diabetic retinopathy, retinal vein occlusions and retinopathy of prematurity), (b) subretinal neovascular disorders (such as neovascular age-related macular degeneration, ocular histoplasmosis, pathologic myopia) (c) intraocular tumors (such as ocular melanoma, ocular lymphoma and retino-blastoma). The treatment for these ocular disorder involves the administration of an effective amount of human chorionic gonadotropin (hCG) antagonists, luteinizing hormone (LH) antagonists or hCG/LH receptor antagonists, optionally in combination with at least one additional bioactive agent.1. A method for reducing the likelihood or inhibiting the progression of an ocular disorder in a patient in need comprising administering to said patient an effective amount of a hCG/LH antagonist, optionally in combination with an additional bioactive agent effective for ameliorating the effects of said ocular disorder. 2. The method of claim 1 in which said hCG/LH antagonist is a small molecule entity having a molecular weight of less than 150 kDa. 3. The method of claim 1 in which said hCG/LH antagonist is a small molecule entity having a molecular weight of from 20 to 80 kDa. 4. The method of claim 3 in which said hCG/LH antagonist is applied topically to the eye. 5. The method of claim 1 in which said hCG/LH antagonist is a small molecule entity having a molecular weight of 30-50 kDa. 6. The method of claim 5 in which said hCG/LH antagonist is applied topically to the eye. 7. The method according to claim 2 wherein said hCG/LH antagonist is a deglycosylated hCG, a mutant hCG or an hCG mutant which has also been deglycosylated. 8. The method according to claim 2 wherein said hCG/LH antagonist is deglycosylated hCG. 9. The method according to claim 2 wherein said hCG/LH antagonist is a mutant hCG. 10. The method according to claim 2 wherein said hCG/LH antagonist is a mutant hCG which is also deglycosylated. 11. The method according to claim 2 wherein said deglycosylated hCG comprises less N-linked carbohydrates than naturally occurring hCG. 12. The method according to claim 8 wherein said deglycosylated hCG has at least 55% less carbohydrates than naturally occurring hCG. 13. The method according to claim 8 wherein said deglycosylated hCG has at least 95% less carbohydrates than naturally occurring hCG. 14. The method according to claim 1 wherein said hCG/LH antagonist is coadministered in combination with an additional bioactive agent. 15. The method according to claim 14 wherein said additional bioactive agent is at least one agent selected from the group consisting of ranibizumab, aflibercept, pegatanib, verteporfin, triamcinolone and bevacizumab. 16. The method according to claim 14 wherein the ocular disorder is a retinal disorder. 17. The method according to claim 1 wherein said ocular disorder is selected from the group consisting of diabetic retinopathy, retinal vein occlusions, and retinopathy of prematurity. 18. The method according to claim 1 wherein said ocular disorder is selected from a group of subretinal neovascular disorders selected from the group consisting of macular degeneration, ocular histoplasmosis, and pathologic myopia. 19. The method according to claim 1 wherein said ocular disorder is selected from the group consisting of intraocular tumors which are NOT known to secrete hCG or LH. 20. The method according to claim 1 wherein said hCG/LH antagonists are hCG/LH antibodies. 21. A pharmaceutical composition comprising an effective amount of an hCG/LH antagonist in combination with at least one agent selected from the groups consisting of ranibizumab, aflibercept, pegatanib, verteporfin, triamcinolone and bevacizumab, in combination with a carrier, additive and/or excipient. 22. A pharmaceutical composition comprising an ophthalmic solution of a small molecule hCG/LH antagonist having a molecular weight of from 20 to 80 kDa. 23. The pharmaceutical composition of claim 22 in which said small molecule hCG/LH antagonist has a molecular weight of 30-50 kDa. 24. A pharmaceutical composition comprising a topical application eye drop solution of a small molecule hCG/LH antagonist having a molecular weight of from 20 to 80 kDa. 25. The pharmaceutical composition of claim 24 in which said small molecule hCG/LH antagonist has a molecular weight of 30-50 kDa.
1,600
315
13,645,871
1,628
A compound of formula (I) is described and use thereof as fragrance, especially as lily-of-the-valley fragrance and/or as an agent for increasing the substantivity and/or the retention of a fragrance preparation and/or as a fixative.
1. A compound selected from the group consisting of a compound of formula (Ia) and a compound of formula (Ib) 2. A mixture, comprising a compound of formula (Ia) and a compound of formula (Ib), as defined in claim 1. 3. A mixture according to claim 2, wherein the mass ratio of the compound of formula (Ia) to the compound of formula (Ib) is in the range from 0.01 to 99.99 and/or the total proportion of compounds of formulas (Ia) and (Ib) is greater than 0.01 wt. 4. (canceled) 5. (canceled) 6. A fragrance preparation, comprising: a compound of formula (I) and an additional fragrance that is not a compound of formula (I). 7. A fragrance preparation according to claim 6, wherein the amount of a compound of formula (Ia) and/or a compound of formula (Ib) is in the range from 0.0001 to 40 wt relative to the total weight of the fragrance preparation. 8. A fragrance preparation according to claim 6 comprising an additional fragrance with an odor note of lily of the valley. 9. A fragrance preparation according to claim 6, wherein (i) the amount of the compound of formula (Ia) is sufficient to endow the fragrance preparation with an odor note of lily of the valley, and/or (ii) an additional fragrance of the fragrance preparation imparts an odor note of lily of the valley, wherein the amount of the compound of formula (Ia) is sufficient, in comparison with a comparative fragrance preparation of otherwise identical composition without the compound of formula (Ia), to modify and/or to intensify the odor note of lily of the valley, and/or (iii) the amount of the compound of formula (Ia) is sufficient to endow the fragrance preparation with an impression selected from the group consisting of natural, caring, complex and radiant and/or (iv) the amount of the compound of formula (Ia) is sufficient to modify and/or to intensify an impression selected from the group consisting of natural, caring, complex and radiant in comparison with a comparative fragrance preparation of otherwise identical composition without the compound of formula (Ia) and/or (v) the amount of the compound of formula (Ib) is sufficient to endow the fragrance preparation with an odor note of lily of the valley, and/or (vi) an additional fragrance of the fragrance preparation imparts an odor note of lily of the valley, wherein the amount of the compound of formula (Ib) is sufficient, in comparison with a comparative fragrance preparation of otherwise identical composition without the compound of formula (Ib), to modify and/or to intensify the odor note of lily of the valley. 10. A perfumed product comprising a compound of formula (I) as defined in claim 1 in a sensorially effective amount. 11. A perfumed product according to claim 10, wherein the amount of a compound of formula (Ia) and/or a compound of formula (Ib) is in the range from 0.0001 to 5 wt %, relative to the total weight of the product. 12. A perfumed product according to claim 10, selected from the group consisting of washing and cleaning products, hygiene products, and care products. 13. A method for of providing skin, hair, surfaces, or textile fibers with an odor note of lily of the valley comprising applying a fragrance preparation according to claim 6 to the skin, hair, surfaces, or textile fibers. 14. A method for producing the compound of formula (I) comprising an acid-catalyzed reaction of an acetal of formula (II), wherein R is a linear or branched, saturated or unsaturated alkyl group with 1 to 10 carbon atoms. 15. An acetal of formula (II) wherein R is a linear or branched, saturated or unsaturated alkyl group with 1 to 10 carbon atoms. 16. A method for producing a compound of formula (Ia) or a compound of formula (Ib) as defined in claim 1, or a mixture of compounds of formula (Ia) and (Ib), comprising the use of d-(+) limonene as educt for producing the compound of formula (Ia), of 1-(−) limonene as educt for producing the compound of formula (Ib) as, or of a mixture containing d-(+) limonene and 1-(−) limonene as educt for producing a mixture containing the compound of formula (Ia) and the compound of formula (Ib). 17. A method for producing a compound of formula (I) as defined in claim 6 comprising pyrolysis of the spirolactone of formula (VIII) to form the compound of formula (I) 18. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a compound according to claim 1. a compound of formula (I), 19. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a mixture according to claim 2. 20. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a fragrance preparation according to claim 6. 21. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a fragrance preparation according to claim 7. 22. A method for increasing the sub stantivity and/or the retention of a fragrance comprising combining the fragrance with a compound of formula (I) as defined in claim 6.
A compound of formula (I) is described and use thereof as fragrance, especially as lily-of-the-valley fragrance and/or as an agent for increasing the substantivity and/or the retention of a fragrance preparation and/or as a fixative.1. A compound selected from the group consisting of a compound of formula (Ia) and a compound of formula (Ib) 2. A mixture, comprising a compound of formula (Ia) and a compound of formula (Ib), as defined in claim 1. 3. A mixture according to claim 2, wherein the mass ratio of the compound of formula (Ia) to the compound of formula (Ib) is in the range from 0.01 to 99.99 and/or the total proportion of compounds of formulas (Ia) and (Ib) is greater than 0.01 wt. 4. (canceled) 5. (canceled) 6. A fragrance preparation, comprising: a compound of formula (I) and an additional fragrance that is not a compound of formula (I). 7. A fragrance preparation according to claim 6, wherein the amount of a compound of formula (Ia) and/or a compound of formula (Ib) is in the range from 0.0001 to 40 wt relative to the total weight of the fragrance preparation. 8. A fragrance preparation according to claim 6 comprising an additional fragrance with an odor note of lily of the valley. 9. A fragrance preparation according to claim 6, wherein (i) the amount of the compound of formula (Ia) is sufficient to endow the fragrance preparation with an odor note of lily of the valley, and/or (ii) an additional fragrance of the fragrance preparation imparts an odor note of lily of the valley, wherein the amount of the compound of formula (Ia) is sufficient, in comparison with a comparative fragrance preparation of otherwise identical composition without the compound of formula (Ia), to modify and/or to intensify the odor note of lily of the valley, and/or (iii) the amount of the compound of formula (Ia) is sufficient to endow the fragrance preparation with an impression selected from the group consisting of natural, caring, complex and radiant and/or (iv) the amount of the compound of formula (Ia) is sufficient to modify and/or to intensify an impression selected from the group consisting of natural, caring, complex and radiant in comparison with a comparative fragrance preparation of otherwise identical composition without the compound of formula (Ia) and/or (v) the amount of the compound of formula (Ib) is sufficient to endow the fragrance preparation with an odor note of lily of the valley, and/or (vi) an additional fragrance of the fragrance preparation imparts an odor note of lily of the valley, wherein the amount of the compound of formula (Ib) is sufficient, in comparison with a comparative fragrance preparation of otherwise identical composition without the compound of formula (Ib), to modify and/or to intensify the odor note of lily of the valley. 10. A perfumed product comprising a compound of formula (I) as defined in claim 1 in a sensorially effective amount. 11. A perfumed product according to claim 10, wherein the amount of a compound of formula (Ia) and/or a compound of formula (Ib) is in the range from 0.0001 to 5 wt %, relative to the total weight of the product. 12. A perfumed product according to claim 10, selected from the group consisting of washing and cleaning products, hygiene products, and care products. 13. A method for of providing skin, hair, surfaces, or textile fibers with an odor note of lily of the valley comprising applying a fragrance preparation according to claim 6 to the skin, hair, surfaces, or textile fibers. 14. A method for producing the compound of formula (I) comprising an acid-catalyzed reaction of an acetal of formula (II), wherein R is a linear or branched, saturated or unsaturated alkyl group with 1 to 10 carbon atoms. 15. An acetal of formula (II) wherein R is a linear or branched, saturated or unsaturated alkyl group with 1 to 10 carbon atoms. 16. A method for producing a compound of formula (Ia) or a compound of formula (Ib) as defined in claim 1, or a mixture of compounds of formula (Ia) and (Ib), comprising the use of d-(+) limonene as educt for producing the compound of formula (Ia), of 1-(−) limonene as educt for producing the compound of formula (Ib) as, or of a mixture containing d-(+) limonene and 1-(−) limonene as educt for producing a mixture containing the compound of formula (Ia) and the compound of formula (Ib). 17. A method for producing a compound of formula (I) as defined in claim 6 comprising pyrolysis of the spirolactone of formula (VIII) to form the compound of formula (I) 18. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a compound according to claim 1. a compound of formula (I), 19. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a mixture according to claim 2. 20. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a fragrance preparation according to claim 6. 21. A method for mediating, intensifying and/or modifying an odor note of lily of the valley to a substance comprising bringing the substance into contact with a fragrance preparation according to claim 7. 22. A method for increasing the sub stantivity and/or the retention of a fragrance comprising combining the fragrance with a compound of formula (I) as defined in claim 6.
1,600
316
15,742,512
1,618
The present invention relates to a method of in vivo imaging and in particular to a method for the evaluation of in vivo images of beta amyloid (Aβ) distribution in the brain of a subject suspected of having Alzheimer's disease (AD). The method of the present invention provides more detailed information to the clinician as compared with prior art methods, facilitating identification of those subjects who will benefit most from disease modifying therapies.
1. A method for staging beta amyloid (Aβ) pathology in a subject's brain wherein said method comprises: (i) obtaining an in vivo image of said subject's brain using a Aβ imaging agent; (ii) determining from said in vivo image the uptake of said Aβ imaging agent in a cortical region of said brain; and, (iii) determining from said in vivo image the uptake of said Aβ imaging agent in a striatal region of said brain; wherein positive uptake of said Aβ imaging agent in said cortical region and negative uptake of said Aβ imaging agent in said striatal region indicates Thal Phase 3 Aβ pathology and wherein positive uptake of said Aβ imaging agent in said cortical region and positive uptake of said Aβ imaging agent in said striatal region indicates Thal Phase 4 or 5 Aβ pathology. 2. The method as defined in claim 1 wherein said subject is a mammalian subject. 3. The method as defined in claim 1 wherein said subject is a human subject. 4. The method as defined in claim 3 wherein said subject is suspected of having Alzheimer's disease. 5. The method as defined in claim 1 wherein uptake of said Aβ imaging agent in any one of the frontal/anterior cingulate, the posterior cingulate/precuneus, the insula and the lateral temporal lobe is taken to indicate uptake in said cortical region. 6. The method as defined in claim 1 wherein uptake of said Aβ imaging agent at level of the head of the caudate nucleus and putamen is taken to indicate uptake in said striatal region. 7. The method as defined in claim 1 wherein said Aβ imaging agent is either a positron emission tomography (PET) imaging agent or a single photon emission tomography (SPECT) imaging agent. 8. The method as defined in claim 1 wherein said Aβ imaging agent is a PET imaging agent. 9. The method as defined in claim 8 wherein said PET imaging agent comprises a compound radiolabelled with 11C or 18F. 10. The method as defined in claim 9 wherein said PET imaging agent comprises a compound radiolabelled with 11C. 11. The method as defined in claim 10 wherein said PET imaging agent is 11C-PIB: 12. The method as defined in claim 9 wherein said PET imaging agent comprises a compound radiolabelled with 18F. 13. The method as defined in claim 12 wherein said PET imaging agent is selected from one of the following compounds: 14. The method as defined in claim 1 wherein said Aβ imaging agent is a SPECT imaging agent. 15. The method as defined in claim 14 wherein said SPECT imaging agent comprises a compound radiolabelled with 123I or 125I. 16. The method as defined in claim 15 wherein said SPECT imaging agent is: 17. A method for treatment of Alzheimer's disease (AD) wherein said method comprises the method as defined in claim 1 and the further steps of: (iv) selecting those subjects in whom Thal Phase 3 Aβ pathology or greater has been indicated; (v) treating those subjects selected in step (iii) with an AD therapy. 18. A method for the evaluation of the effects of an experimental AD therapy wherein said method comprises carrying out the method as defined in claim 1 on a group of subjects to whom said experimental AD therapy has been given. 19. A method to determine inclusion of subjects into a clinical trial for the evaluation of an Aβ imaging agent comprising the method as defined in claim 1 and admitting those subjects having positive uptake of said Aβ imaging agent in said cortical region and negative uptake of said Aβ imaging agent in said striatal region into said clinical trial.
The present invention relates to a method of in vivo imaging and in particular to a method for the evaluation of in vivo images of beta amyloid (Aβ) distribution in the brain of a subject suspected of having Alzheimer's disease (AD). The method of the present invention provides more detailed information to the clinician as compared with prior art methods, facilitating identification of those subjects who will benefit most from disease modifying therapies.1. A method for staging beta amyloid (Aβ) pathology in a subject's brain wherein said method comprises: (i) obtaining an in vivo image of said subject's brain using a Aβ imaging agent; (ii) determining from said in vivo image the uptake of said Aβ imaging agent in a cortical region of said brain; and, (iii) determining from said in vivo image the uptake of said Aβ imaging agent in a striatal region of said brain; wherein positive uptake of said Aβ imaging agent in said cortical region and negative uptake of said Aβ imaging agent in said striatal region indicates Thal Phase 3 Aβ pathology and wherein positive uptake of said Aβ imaging agent in said cortical region and positive uptake of said Aβ imaging agent in said striatal region indicates Thal Phase 4 or 5 Aβ pathology. 2. The method as defined in claim 1 wherein said subject is a mammalian subject. 3. The method as defined in claim 1 wherein said subject is a human subject. 4. The method as defined in claim 3 wherein said subject is suspected of having Alzheimer's disease. 5. The method as defined in claim 1 wherein uptake of said Aβ imaging agent in any one of the frontal/anterior cingulate, the posterior cingulate/precuneus, the insula and the lateral temporal lobe is taken to indicate uptake in said cortical region. 6. The method as defined in claim 1 wherein uptake of said Aβ imaging agent at level of the head of the caudate nucleus and putamen is taken to indicate uptake in said striatal region. 7. The method as defined in claim 1 wherein said Aβ imaging agent is either a positron emission tomography (PET) imaging agent or a single photon emission tomography (SPECT) imaging agent. 8. The method as defined in claim 1 wherein said Aβ imaging agent is a PET imaging agent. 9. The method as defined in claim 8 wherein said PET imaging agent comprises a compound radiolabelled with 11C or 18F. 10. The method as defined in claim 9 wherein said PET imaging agent comprises a compound radiolabelled with 11C. 11. The method as defined in claim 10 wherein said PET imaging agent is 11C-PIB: 12. The method as defined in claim 9 wherein said PET imaging agent comprises a compound radiolabelled with 18F. 13. The method as defined in claim 12 wherein said PET imaging agent is selected from one of the following compounds: 14. The method as defined in claim 1 wherein said Aβ imaging agent is a SPECT imaging agent. 15. The method as defined in claim 14 wherein said SPECT imaging agent comprises a compound radiolabelled with 123I or 125I. 16. The method as defined in claim 15 wherein said SPECT imaging agent is: 17. A method for treatment of Alzheimer's disease (AD) wherein said method comprises the method as defined in claim 1 and the further steps of: (iv) selecting those subjects in whom Thal Phase 3 Aβ pathology or greater has been indicated; (v) treating those subjects selected in step (iii) with an AD therapy. 18. A method for the evaluation of the effects of an experimental AD therapy wherein said method comprises carrying out the method as defined in claim 1 on a group of subjects to whom said experimental AD therapy has been given. 19. A method to determine inclusion of subjects into a clinical trial for the evaluation of an Aβ imaging agent comprising the method as defined in claim 1 and admitting those subjects having positive uptake of said Aβ imaging agent in said cortical region and negative uptake of said Aβ imaging agent in said striatal region into said clinical trial.
1,600
317
15,310,195
1,623
A medicament for accelerated wound healing is proposed containing derivatives of the formula (I), in which R 1 stands for hydrogen or methyl.
1. A drug containing a derivative of the formula (I) in which R1 denotes hydrogen or methyl, for acceleration of wound healing. 2. The drug according to claim 1, wherein said drug is Sandalore®. 3. The drug according to claim 1, wherein said drug is Brahmanol®. 4. A method for acceleration of wound healing, comprising using the drug according to claim 1 for activation of the olfactory receptor OR2AT4. 5. A method for acceleration of wound healing, comprising using the drug according to claim 1 for stimulating the proliferation and migration of cells. 6. A method for acceleration of wound healing, comprising using the drug according to claim 1 for stimulating the phosphorylation of MAP kinases. 7. A method for acceleration of wound healing, comprising using the drug according to claim 1 for stimulating the expression and secretion of interleukin IL-1α. 8. The drug according to claim 1, in a form for topical use. 9. The drug according to claim 1, wherein said drug is in the form of lotions, creams, emulsions, gels, ointments, or sprays, or corresponding dressing materials are impregnated or coated therewith. 10. The drug according to claim 1, wherein said drug contains the active ingredient of the formula (I) in amounts of 0.001 to approx. 2 wt. %. 11. A method for identifying active ingredients that promote wound healing, comprising (a) providing a keratinocyte culture that contains the olfactory receptor OR2AT4, (b) adding the active ingredient to be tested to the culture, and (c) determining the change in intracellular calcium concentration.
A medicament for accelerated wound healing is proposed containing derivatives of the formula (I), in which R 1 stands for hydrogen or methyl.1. A drug containing a derivative of the formula (I) in which R1 denotes hydrogen or methyl, for acceleration of wound healing. 2. The drug according to claim 1, wherein said drug is Sandalore®. 3. The drug according to claim 1, wherein said drug is Brahmanol®. 4. A method for acceleration of wound healing, comprising using the drug according to claim 1 for activation of the olfactory receptor OR2AT4. 5. A method for acceleration of wound healing, comprising using the drug according to claim 1 for stimulating the proliferation and migration of cells. 6. A method for acceleration of wound healing, comprising using the drug according to claim 1 for stimulating the phosphorylation of MAP kinases. 7. A method for acceleration of wound healing, comprising using the drug according to claim 1 for stimulating the expression and secretion of interleukin IL-1α. 8. The drug according to claim 1, in a form for topical use. 9. The drug according to claim 1, wherein said drug is in the form of lotions, creams, emulsions, gels, ointments, or sprays, or corresponding dressing materials are impregnated or coated therewith. 10. The drug according to claim 1, wherein said drug contains the active ingredient of the formula (I) in amounts of 0.001 to approx. 2 wt. %. 11. A method for identifying active ingredients that promote wound healing, comprising (a) providing a keratinocyte culture that contains the olfactory receptor OR2AT4, (b) adding the active ingredient to be tested to the culture, and (c) determining the change in intracellular calcium concentration.
1,600
318
14,572,664
1,611
An avermectin-based topical formulation is disclosed which is useful for prevention and treatment of head lice ( Pediculus humanus capitis ). This topical formulation may be formulated as a shampoo-condition which comprises an effective amount of avermectin, solubilizers, suspending agents, preservatives, nonionic surfactants, humectants, a silicone compound, and water. Also disclosed are methods of using the topical formulations disclosed within this specification to treat either a susceptible or treatment-resistant strain of lice, as well as uses in the manufacture of a medicament for treating or preventing a lice infestation from a susceptible or treatment-resistant strain in a human patient.
1. A topical formulation comprising about 0.1% to about 2.0% by weight ivermectin, a solubilizer, about 20% to 30% by weight olive oil, one or more one paraben preservatives, a non-ionic surfactant, and about 30% to 40% by weight water. 2. The topical formulation of claim 1, comprising about 0.5% by weight ivermectin. 3. The topical formulation of claim 1, further comprising shea butter. 4. The topical formulation of claim 1, wherein said solubilizer comprises polysorbate 80, cetyl acetate, acetylated lanolin alcohol or a combination thereof. 5. The topical formulation of claim 1, comprising methylparaben, propylparaben, ethylparaben, butylparaben, isobutylparaben, isopropylparaben, or benzylparaben, or their sodium salts thereof, or any combination thereof. 6. The topical formulation of claim 1, wherein said nonionic surfactant is selected from the group consisting of oleyl alcohol, lanolin alcohol, sorbitan tristerate, bees wax, erucyl alcohol, ricinolyl alcohol, arachidyl alcohol, capryl alcohol, capric alcohol, behenyl alcohol, lauryl alcohol, myristyl alcohol, cetyl alcohol, stearyl alcohol, isostearyl alcohol, oleyl alcohol, palmitoleyl alcohol, linoleyl alcohol, elaidyl alcohol, elaidolinoleyl alcohol, linolenyl alcohol, elaidolinolenyl alcohol, glycerine, glyceryl triacetate, sorbitol, xylitol, maltitol, polydextrose, quillaia, lactic acid, urea, and combinations thereof. 7. The topical formulation of claim 1, comprising oleyl alcohol, lanolin alcohol, acetylated lanolin alcohol, sorbitan tristearate, or a combination thereof. 8. The topical formulation of claim 1, further comprising a conditioner. 9. The topical formulation of claim 8, wherein said conditioner is selected from the group consisting of cyclomethicone, dimethicone, hexamethyldisiloxane, octamethyltrisiloxane, decamethyltetrasiloxane, dodecamethylpentasiloxane, polydimethylsiloxanes, and combinations thereof. 10. The topical formulation of claim 1, formulated as a cream, gel, or pomade. 11. The topical formulation of claim 1 prepared according to a method comprising: (a) preparing a first solution by dissolving ivermectin in the solubilizer; and (b) adding to the first solution a second solution comprising the olive oil, the one or more one paraben preservatives, and the non-ionic surfactant; and (c) dispersing the oil mixture of (b) in water. 12. The topical formulation of claim 11, wherein the solubilizer phase comprises polysorbate 80, cetyl acetate, acetylated lanolin alcohol or a combination thereof. 13. A method of treating a head lice infestation in a human patient comprising topically administering to said patient the topical formulation of claim 1 in one dosage or multiple dosages. 14. The method of claim 13, wherein said human patient is a child. 15. The method of claim 13, wherein at least about 60 mL of the topical formulation is applied to said human patient. 16. The method of claim 13, wherein about 1 mL to about 100 mL of the topical formulation is applied to said human patient. 17. The method of claim 13, wherein said topical formulation is left on the scalp for about 1 to about 60 minutes. 18. The method of claim 17, wherein said topical formulation is left on the scalp for about 3 minutes, about 5 minutes, or about 10 minutes 19. The method of claim 1 wherein the topical formulation is administered as a single dose. 20. The method of claim 1 wherein the topical formulation is administered as an initial dosage with one or two additional dosages.
An avermectin-based topical formulation is disclosed which is useful for prevention and treatment of head lice ( Pediculus humanus capitis ). This topical formulation may be formulated as a shampoo-condition which comprises an effective amount of avermectin, solubilizers, suspending agents, preservatives, nonionic surfactants, humectants, a silicone compound, and water. Also disclosed are methods of using the topical formulations disclosed within this specification to treat either a susceptible or treatment-resistant strain of lice, as well as uses in the manufacture of a medicament for treating or preventing a lice infestation from a susceptible or treatment-resistant strain in a human patient.1. A topical formulation comprising about 0.1% to about 2.0% by weight ivermectin, a solubilizer, about 20% to 30% by weight olive oil, one or more one paraben preservatives, a non-ionic surfactant, and about 30% to 40% by weight water. 2. The topical formulation of claim 1, comprising about 0.5% by weight ivermectin. 3. The topical formulation of claim 1, further comprising shea butter. 4. The topical formulation of claim 1, wherein said solubilizer comprises polysorbate 80, cetyl acetate, acetylated lanolin alcohol or a combination thereof. 5. The topical formulation of claim 1, comprising methylparaben, propylparaben, ethylparaben, butylparaben, isobutylparaben, isopropylparaben, or benzylparaben, or their sodium salts thereof, or any combination thereof. 6. The topical formulation of claim 1, wherein said nonionic surfactant is selected from the group consisting of oleyl alcohol, lanolin alcohol, sorbitan tristerate, bees wax, erucyl alcohol, ricinolyl alcohol, arachidyl alcohol, capryl alcohol, capric alcohol, behenyl alcohol, lauryl alcohol, myristyl alcohol, cetyl alcohol, stearyl alcohol, isostearyl alcohol, oleyl alcohol, palmitoleyl alcohol, linoleyl alcohol, elaidyl alcohol, elaidolinoleyl alcohol, linolenyl alcohol, elaidolinolenyl alcohol, glycerine, glyceryl triacetate, sorbitol, xylitol, maltitol, polydextrose, quillaia, lactic acid, urea, and combinations thereof. 7. The topical formulation of claim 1, comprising oleyl alcohol, lanolin alcohol, acetylated lanolin alcohol, sorbitan tristearate, or a combination thereof. 8. The topical formulation of claim 1, further comprising a conditioner. 9. The topical formulation of claim 8, wherein said conditioner is selected from the group consisting of cyclomethicone, dimethicone, hexamethyldisiloxane, octamethyltrisiloxane, decamethyltetrasiloxane, dodecamethylpentasiloxane, polydimethylsiloxanes, and combinations thereof. 10. The topical formulation of claim 1, formulated as a cream, gel, or pomade. 11. The topical formulation of claim 1 prepared according to a method comprising: (a) preparing a first solution by dissolving ivermectin in the solubilizer; and (b) adding to the first solution a second solution comprising the olive oil, the one or more one paraben preservatives, and the non-ionic surfactant; and (c) dispersing the oil mixture of (b) in water. 12. The topical formulation of claim 11, wherein the solubilizer phase comprises polysorbate 80, cetyl acetate, acetylated lanolin alcohol or a combination thereof. 13. A method of treating a head lice infestation in a human patient comprising topically administering to said patient the topical formulation of claim 1 in one dosage or multiple dosages. 14. The method of claim 13, wherein said human patient is a child. 15. The method of claim 13, wherein at least about 60 mL of the topical formulation is applied to said human patient. 16. The method of claim 13, wherein about 1 mL to about 100 mL of the topical formulation is applied to said human patient. 17. The method of claim 13, wherein said topical formulation is left on the scalp for about 1 to about 60 minutes. 18. The method of claim 17, wherein said topical formulation is left on the scalp for about 3 minutes, about 5 minutes, or about 10 minutes 19. The method of claim 1 wherein the topical formulation is administered as a single dose. 20. The method of claim 1 wherein the topical formulation is administered as an initial dosage with one or two additional dosages.
1,600
319
13,688,078
1,618
This disclosure describes example antimicrobial compositions that may be used in combination with IV port cleansing caps, protective caps, or nasal decolonizer devices. According to another implementations, the disclosure describes that the antimicrobial composition may provide an indication that it has come into contact with a contaminant by bubbling or foam on a surface that is being cleaned. According to another implementation, the disclosure describes that the antimicrobial composition may leave a residual film or barrier to inhibit the recontamination of a surface that has been cleaned.
1. A method comprising: identifying a surface to be decontaminated; applying a medical applicator to the surface, wherein the medical applicator comprises: a cap device comprising a cylindrical cavity, the cylindrical cavity having a foam insert or a nasal decolonizer device comprising a reservoir; and an antimicrobial composition within the foam insert of the cap device or reservoir of the nasal decolonizer device, wherein the antimicrobial composition comprises water, an alcohol, a peroxide or peroxide-generating agent and a chelating agent; and receiving an indication that the antimicrobial composition has come in contact with one or more contaminants on the surface. 2. The method as recited in claim 1, wherein the one or more contaminants comprises at least one of one or more of a bacterium, a spore, a parasite, a virus, bodily fluid or a combination thereof. 3. The method as recited in claim 2, wherein the one or more contaminants includes staphylococcus aureus (MRSA), Pseudomonas aeruginosa or other resistant bacteria. 4. The method as recited in claim 1, wherein the antimicrobial composition further comprises: about 5 to about 50 mg/ml of ethlyenediaminetetraacetic acid (EDTA); at most about 70% ethanol, by volume; at most about 7.5% hydrogen peroxide, by volume; and water. 5. The method as recited in claim 1, wherein the antimicrobial composition is in the form of a liquid or gel. 6. The method as recited in claim 1, wherein the chelating agent is present in a quantity to remain on a surface after the water, the alcohol and the peroxide have dispersed. 7. The method as recited in claim 1, wherein receiving an indication that the antimicrobial composition has come in contact with one or more contaminant on the surface comprises foaming or bubbling. 8. The method as recited in claim 1, wherein the surface to be decontaminated comprises human tissue or an area on a medical device. 9. A method of providing a barrier against contamination comprising: identifying a surface to be protected; applying an antimicrobial agent to the surface, wherein the antimicrobial agent comprises water, a low molecular weight alcohol, a peroxide or peroxide-generating agent and a chelating agent; wherein the chelating agent is present in an amount to remain on the surface after the water, a low molecular weight alcohol, a peroxide or peroxide-generating agent have evaporated. 10. The method as recited in claim 9, wherein the surface to be protected comprises human tissue or an area on a medical device. 11. The method as recited in claim 9, wherein the antimicrobial agent further comprises: about 5 to about 50 mg/ml of ethlyenediaminetetraacetic acid (EDTA); at most about 70% ethanol, by volume; at most about 7.5% hydrogen peroxide, by volume; and water. 12. The method as recited in claim 9, wherein the antimicrobial agent is in the form of a liquid or gel. 13. The method as recited in claim 9, wherein the chelating agent remaining on the surface inhibits the reproductive capabilities of bacteria, spores, fungi, parasites and viruses. 14. A method of giving a visual indication of one or more contaminants on a surface comprising: determining one or more surfaces to be decontaminated; applying an antimicrobial agent to the surface, wherein the antimicrobial agent comprises water, a low molecular weight alcohol, a peroxide or peroxide-generating agent and a chelating agent; and receiving an indication that the antimicrobial agent has come in contact with one or more contaminant on the surface. 15. The method as recited in claim 14, wherein the surface to be decontaminated comprises human tissue or an area on a medical device. 16. The method as recited in claim 14, wherein the antimicrobial agent further comprises: about 5 to about 50 mg/ml of ethlyenediaminetetraacetic acid (EDTA); at most about 70% ethanol, by volume; at most about 7.5% hydrogen peroxide, by volume; and water. 17. The method as recited in claim 14, wherein receiving an indication that the antimicrobial agent has come in contact with one or more contaminant on the surface comprises a visual indication such as foaming or bubbling on the surface. 18. The method as recited in claim 14, wherein the one or more contaminants comprises at least one of one or more bacteria, one or more spores, one or more parasites, one or more viruses, one or more bodily fluids. 19. The method as recited in claim 18, wherein the one or more contaminants includes staphylococcus aureus (MRSA), Pseudomonas aeruginosa and other resistant bacteria. 20. The method as recited in claim 14, wherein the amount chelating agent present in the antimicrobial agent does not inhibit the indication that the antimicrobial agent has come in contact with one or more contaminant on the surface.
This disclosure describes example antimicrobial compositions that may be used in combination with IV port cleansing caps, protective caps, or nasal decolonizer devices. According to another implementations, the disclosure describes that the antimicrobial composition may provide an indication that it has come into contact with a contaminant by bubbling or foam on a surface that is being cleaned. According to another implementation, the disclosure describes that the antimicrobial composition may leave a residual film or barrier to inhibit the recontamination of a surface that has been cleaned.1. A method comprising: identifying a surface to be decontaminated; applying a medical applicator to the surface, wherein the medical applicator comprises: a cap device comprising a cylindrical cavity, the cylindrical cavity having a foam insert or a nasal decolonizer device comprising a reservoir; and an antimicrobial composition within the foam insert of the cap device or reservoir of the nasal decolonizer device, wherein the antimicrobial composition comprises water, an alcohol, a peroxide or peroxide-generating agent and a chelating agent; and receiving an indication that the antimicrobial composition has come in contact with one or more contaminants on the surface. 2. The method as recited in claim 1, wherein the one or more contaminants comprises at least one of one or more of a bacterium, a spore, a parasite, a virus, bodily fluid or a combination thereof. 3. The method as recited in claim 2, wherein the one or more contaminants includes staphylococcus aureus (MRSA), Pseudomonas aeruginosa or other resistant bacteria. 4. The method as recited in claim 1, wherein the antimicrobial composition further comprises: about 5 to about 50 mg/ml of ethlyenediaminetetraacetic acid (EDTA); at most about 70% ethanol, by volume; at most about 7.5% hydrogen peroxide, by volume; and water. 5. The method as recited in claim 1, wherein the antimicrobial composition is in the form of a liquid or gel. 6. The method as recited in claim 1, wherein the chelating agent is present in a quantity to remain on a surface after the water, the alcohol and the peroxide have dispersed. 7. The method as recited in claim 1, wherein receiving an indication that the antimicrobial composition has come in contact with one or more contaminant on the surface comprises foaming or bubbling. 8. The method as recited in claim 1, wherein the surface to be decontaminated comprises human tissue or an area on a medical device. 9. A method of providing a barrier against contamination comprising: identifying a surface to be protected; applying an antimicrobial agent to the surface, wherein the antimicrobial agent comprises water, a low molecular weight alcohol, a peroxide or peroxide-generating agent and a chelating agent; wherein the chelating agent is present in an amount to remain on the surface after the water, a low molecular weight alcohol, a peroxide or peroxide-generating agent have evaporated. 10. The method as recited in claim 9, wherein the surface to be protected comprises human tissue or an area on a medical device. 11. The method as recited in claim 9, wherein the antimicrobial agent further comprises: about 5 to about 50 mg/ml of ethlyenediaminetetraacetic acid (EDTA); at most about 70% ethanol, by volume; at most about 7.5% hydrogen peroxide, by volume; and water. 12. The method as recited in claim 9, wherein the antimicrobial agent is in the form of a liquid or gel. 13. The method as recited in claim 9, wherein the chelating agent remaining on the surface inhibits the reproductive capabilities of bacteria, spores, fungi, parasites and viruses. 14. A method of giving a visual indication of one or more contaminants on a surface comprising: determining one or more surfaces to be decontaminated; applying an antimicrobial agent to the surface, wherein the antimicrobial agent comprises water, a low molecular weight alcohol, a peroxide or peroxide-generating agent and a chelating agent; and receiving an indication that the antimicrobial agent has come in contact with one or more contaminant on the surface. 15. The method as recited in claim 14, wherein the surface to be decontaminated comprises human tissue or an area on a medical device. 16. The method as recited in claim 14, wherein the antimicrobial agent further comprises: about 5 to about 50 mg/ml of ethlyenediaminetetraacetic acid (EDTA); at most about 70% ethanol, by volume; at most about 7.5% hydrogen peroxide, by volume; and water. 17. The method as recited in claim 14, wherein receiving an indication that the antimicrobial agent has come in contact with one or more contaminant on the surface comprises a visual indication such as foaming or bubbling on the surface. 18. The method as recited in claim 14, wherein the one or more contaminants comprises at least one of one or more bacteria, one or more spores, one or more parasites, one or more viruses, one or more bodily fluids. 19. The method as recited in claim 18, wherein the one or more contaminants includes staphylococcus aureus (MRSA), Pseudomonas aeruginosa and other resistant bacteria. 20. The method as recited in claim 14, wherein the amount chelating agent present in the antimicrobial agent does not inhibit the indication that the antimicrobial agent has come in contact with one or more contaminant on the surface.
1,600
320
12,956,058
1,632
Devices and methods for treating defects in peripheral nerves are provided. The devices can include acellular arterial tissue matrices that facilitate regrowth of nerve tissue across a gap or defect in a peripheral nerve.
1. A method for treating a nerve, comprising: selecting a peripheral nerve having a gap across a portion of its length; and positioning an arterial tissue matrix across the gap, wherein substantially all of the native cells have been removed from the tissue matrix. 2. The method of claim 1, wherein the arterial tissue matrix is a porcine arterial tissue matrix. 3. The method of claim 2, wherein the matrix is produced from a pig that has been genetically modified to have reduced expression of α-1,3-galactose. 4. The method of claim 3, wherein the pig lacks expression of α-galactosyltransferase. 5. The method of claim 2, further including treating the matrix to remove α-1,3-galactose moieties. 6. The method of claim 1, further including placing stem cells within the tissue matrix. 7. The method of claim 6, wherein the stem cells are mesenchymal stem cells. 8. The method of claim 1, wherein the arterial tissue matrix forms a conduit through which a peripheral nerve can grow when the conduit is implanted across a defect in the peripheral nerve. 9. The method of claim 1, wherein the arterial tissue matrix allows at least 50% functional recovery of a muscle innervated by the nerve. 10. The method of claim 9, wherein the arterial tissue matrix allows at least 60% functional recovery of a muscle innervated by the nerve. 11. The method of claim 10, wherein the arterial tissue matrix allows at least 70% functional recovery of a muscle innervated by the nerve. 12. The method of claim 11, wherein the arterial tissue matrix allows at least 80% functional recovery of a muscle innervated by the nerve. 13. The method of any one of claim 9-12, wherein functional recovery is quantified using the size of a muscle innervated by a treated nerve. 14. The method of claim 13, wherein functional recovery is measured by comparing the dry weight or volume of a muscle innervated by the nerve after recovery with the dry weight of a corresponding muscle either before a defect occurred or on an opposing, unaffected limb. 15. The method of claim 1, wherein the gap is greater than 1 cm in length. 16. The method of claim 1, wherein the gap is greater than 2 cm in length. 17. A device for treating a nerve, comprising: an arterial tissue matrix, wherein substantially all of the native cells have been removed. 18. The device of claim 17, wherein the arterial tissue matrix is a porcine arterial tissue matrix. 19. The device of claim 18, wherein the matrix is produced from a pig that has been genetically modified to have reduced expression of α-1,3-galactose. 20. The device of claim 19, wherein the pig lacks expression of α-galactosyltransferase. 21. The device of claim 18, wherein the matrix has been treated to remove α-1,3-galactose moieties. 22. The device of claim 17, further including stem cells. 23. The device of claim 22, wherein the stem cells are mesenchymal stem cells. 24. The device of claim 17, wherein the arterial tissue matrix forms a conduit through which a peripheral nerve can grow when the conduit is implanted across a defect in the peripheral nerve. 25. The device of claim 17, wherein the arterial tissue matrix is capable of supporting peripheral nerve regeneration across a gap greater than 1 cm. 26. The device of claim 17, wherein the arterial tissue matrix includes glycosaminoglycans and elastins present in intact arterial tissue from which the tissue matrix is derived. 27. The device of claim 17, wherein the arterial tissue matrix includes an intact basement membrane. 28. The device of claim 17, wherein the arterial tissue matrix allows at least 50% functional recovery of a muscle innervated by the nerve. 29. The device of claim 28, wherein the arterial tissue matrix allows at least 60% functional recovery of a muscle innervated by the nerve. 30. The device of claim 29, wherein the arterial tissue matrix allows at least 70% functional recovery of a muscle innervated by the nerve. 31. The device of claim 30, wherein the arterial tissue matrix allows at least 80% functional recovery of a muscle innervated by the nerve. 32. The device of any one of claim 27-31, wherein functional recovery is quantified using the size of a muscle innervated by a treated nerve. 33. The device of claim 32, wherein functional recovery is measured by comparing the dry weight or volume of a muscle innervated by the nerve after recovery with the dry weight of a corresponding muscle either before a defect occurred or on an opposing, unaffected limb.
Devices and methods for treating defects in peripheral nerves are provided. The devices can include acellular arterial tissue matrices that facilitate regrowth of nerve tissue across a gap or defect in a peripheral nerve.1. A method for treating a nerve, comprising: selecting a peripheral nerve having a gap across a portion of its length; and positioning an arterial tissue matrix across the gap, wherein substantially all of the native cells have been removed from the tissue matrix. 2. The method of claim 1, wherein the arterial tissue matrix is a porcine arterial tissue matrix. 3. The method of claim 2, wherein the matrix is produced from a pig that has been genetically modified to have reduced expression of α-1,3-galactose. 4. The method of claim 3, wherein the pig lacks expression of α-galactosyltransferase. 5. The method of claim 2, further including treating the matrix to remove α-1,3-galactose moieties. 6. The method of claim 1, further including placing stem cells within the tissue matrix. 7. The method of claim 6, wherein the stem cells are mesenchymal stem cells. 8. The method of claim 1, wherein the arterial tissue matrix forms a conduit through which a peripheral nerve can grow when the conduit is implanted across a defect in the peripheral nerve. 9. The method of claim 1, wherein the arterial tissue matrix allows at least 50% functional recovery of a muscle innervated by the nerve. 10. The method of claim 9, wherein the arterial tissue matrix allows at least 60% functional recovery of a muscle innervated by the nerve. 11. The method of claim 10, wherein the arterial tissue matrix allows at least 70% functional recovery of a muscle innervated by the nerve. 12. The method of claim 11, wherein the arterial tissue matrix allows at least 80% functional recovery of a muscle innervated by the nerve. 13. The method of any one of claim 9-12, wherein functional recovery is quantified using the size of a muscle innervated by a treated nerve. 14. The method of claim 13, wherein functional recovery is measured by comparing the dry weight or volume of a muscle innervated by the nerve after recovery with the dry weight of a corresponding muscle either before a defect occurred or on an opposing, unaffected limb. 15. The method of claim 1, wherein the gap is greater than 1 cm in length. 16. The method of claim 1, wherein the gap is greater than 2 cm in length. 17. A device for treating a nerve, comprising: an arterial tissue matrix, wherein substantially all of the native cells have been removed. 18. The device of claim 17, wherein the arterial tissue matrix is a porcine arterial tissue matrix. 19. The device of claim 18, wherein the matrix is produced from a pig that has been genetically modified to have reduced expression of α-1,3-galactose. 20. The device of claim 19, wherein the pig lacks expression of α-galactosyltransferase. 21. The device of claim 18, wherein the matrix has been treated to remove α-1,3-galactose moieties. 22. The device of claim 17, further including stem cells. 23. The device of claim 22, wherein the stem cells are mesenchymal stem cells. 24. The device of claim 17, wherein the arterial tissue matrix forms a conduit through which a peripheral nerve can grow when the conduit is implanted across a defect in the peripheral nerve. 25. The device of claim 17, wherein the arterial tissue matrix is capable of supporting peripheral nerve regeneration across a gap greater than 1 cm. 26. The device of claim 17, wherein the arterial tissue matrix includes glycosaminoglycans and elastins present in intact arterial tissue from which the tissue matrix is derived. 27. The device of claim 17, wherein the arterial tissue matrix includes an intact basement membrane. 28. The device of claim 17, wherein the arterial tissue matrix allows at least 50% functional recovery of a muscle innervated by the nerve. 29. The device of claim 28, wherein the arterial tissue matrix allows at least 60% functional recovery of a muscle innervated by the nerve. 30. The device of claim 29, wherein the arterial tissue matrix allows at least 70% functional recovery of a muscle innervated by the nerve. 31. The device of claim 30, wherein the arterial tissue matrix allows at least 80% functional recovery of a muscle innervated by the nerve. 32. The device of any one of claim 27-31, wherein functional recovery is quantified using the size of a muscle innervated by a treated nerve. 33. The device of claim 32, wherein functional recovery is measured by comparing the dry weight or volume of a muscle innervated by the nerve after recovery with the dry weight of a corresponding muscle either before a defect occurred or on an opposing, unaffected limb.
1,600
321
15,891,852
1,632
Compositions and surgical methods are provided for repairing damaged avascular zones, including intervertebral disc, in a patient in need thereof.
1. A method of surgery for repairing a degenerative cartilage in a patient utilizing an organic active ingredient, the method comprising: administering a therapeutically effective amount of one or more peptides to the degenerative cartilage, wherein the one or more peptides is selected from the group consisting of: a growth factor, a cytokine, an integrin, a cadherin, and a peptide known to promote angiogenesis, vasculogenesis, or arteriogenesis; preparing a blood-derived compound using a method comprising: harvesting marrow blood from the patient; separating a blood composition from the marrow blood; culturing the blood composition in a culture medium under a selective pressure of about 1% to about 10% oxygen and a pH of about 6.6 to about 7.0 for 1-28 days; selecting mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; generating the blood-derived compound by suspending the selected mesenchymal stem cells in blood plasma harvested from the patient; and returning a therapeutically effective amount of the blood derived compound to the patient at the degenerative cartilage. 2. The method of claim 1, wherein the one or more peptides are selected from the group consisting of: VEGF-A, PIGF, VEGF-B, VEGF-C, VEGF-D, TGF-β, Ang-1, Ang-2, IGF, HGF, FGF, Tie2, PDGF, CCL2, Alpha-V Beta-5, Alpha-5 Beta-1, VE-cadherin, PECAM-1, plasminogen activator, nitrogen oxide synthase, and combinations thereof. 3. The method of claim 1, wherein the growth factor is selected from the group consisting of: TGF-β, FGF, PDGF, IGF, and combinations thereof. 4. The method of claim 1, wherein harvesting the marrow blood further comprises: utilizing a needle aspirate technique to draw the marrow blood from the posterior superior iliac spine. 5. A method for treating an avascular zone in a patient in need thereof, the method comprising: culturing nucleated cells harvested from the patient in need thereof in a culture medium under a selective pressure of about 1% to about 10% oxygen for 1-28 days; selecting viable mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; and providing the selected, viable mesenchymal stem cells for implantation in the avascular zone. 6. The method of claim 5, wherein the selective pressure is about 3 to about 7% oxygen. 7. The method of claim 5, wherein the selective pressure further comprises about 2% to about 10% carbon dioxide. 8. The method of claim 7, wherein the selective pressure further comprises a pH of about 6.6 to about 7.0. 9. The method of claim 8, wherein the pH is about 6.6 to about 6.8. 10. The method of claim 5, wherein the selective pressure further comprises an osmolarity of about 350 mOsm to about 600 mOsm. 11. The method of claim 5, wherein the culture medium is supplemented with about 10 to about 20% human platelet cell lysate. 12. The method of claim 5, further comprising: implanting the selected, viable mesenchymal stem cells; implanting platelet cell lysate prior to, during, or after implanting the selected, viable mesenchymal stem cells; wherein the implanting of the platelet cell lysate and the selected, viable mesenchymal stem cells is in the avascular zone. 13. The method of claim 5, further comprising: implanting the selected, viable mesenchymal stem cells; implanting platelets prior to, during, or after implanting the selected, viable mesenchymal stem cells; wherein the implanting of the platelets and the selected, viable mesenchymal stem cells is in the avascular zone. 14. The method of claim 13, wherein the platelets are harvested from the patient in need thereof. 15. The method of claim 14, wherein the platelets are treated with thrombin and calcium chloride 1-7 days prior to implanting. 16. The method of claim 15, wherein the amount of thrombin is about 28.56 U/ml and the amount of calcium chloride is about 2.86 mg/ml. 17. The method of claim 14, wherein the platelets are treated with a composition selected from the group consisting of thrombin, calcium chloride or its salts, thromboxane A2, adenosine triphosphate, arachidonate, and combinations thereof. 18. The method of claim 5, further comprising: administering one or more compounds selected from the group consisting of growth factors, cytokines, integrins, cadherins, molecules or drugs that promote angiogenesis, molecules or drugs that promote vasculogenesis, molecules or drugs that promote aerteriogenesis, and combinations thereof. 19. The method of claim 18, wherein the one or more compounds is selected from a group consisting of VEGF-A, PIGF, VEGF-B, VEGF-C, VEGF-D, TGF-β, Ang-1, Ang-2, IGF, HGF, FGF, Tie2, PDGF, CCL2, Alpha-V Beta-5, Alpha-5 Beta-1, VE-cadherin, PECAM-1, plasminogen activator, nitrogen oxide synthase, and combinations thereof. 20. The method of claim 5, further comprising: administering one or more growth factors before, during, or after implanting the selected, viable mesenchymal stem cells in the avascular zone. 21. The method of claim 20, wherein the one or more growth factors is selected from the group consisting of: TGF-β, FGF, PDGF, IGF, and combinations thereof. 22. The method of claim 5, wherein the culturing is for about two to about five passages. 23. A method for treating a degenerative intervertebral disc in a patient in need thereof, the method comprising: culturing nucleated cells harvested from the patient in need thereof in a culture medium under a selective pressure of about 1% to about 10% oxygen for 1-28 days; selecting viable mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; and providing the selected, viable mesenchymal stem cells for implantation in an intervertebral disc. 24. A method for treating a degenerative intervertebral disc in a patient in need thereof, the method comprising: culturing nucleated cells harvested from the patient in need thereof in a culture medium under a selective pressure of about 1% to about 10% oxygen for 1-28 days; selecting viable mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; and implanting the selected, viable mesenchymal stem cells in an intervertebral disc.
Compositions and surgical methods are provided for repairing damaged avascular zones, including intervertebral disc, in a patient in need thereof.1. A method of surgery for repairing a degenerative cartilage in a patient utilizing an organic active ingredient, the method comprising: administering a therapeutically effective amount of one or more peptides to the degenerative cartilage, wherein the one or more peptides is selected from the group consisting of: a growth factor, a cytokine, an integrin, a cadherin, and a peptide known to promote angiogenesis, vasculogenesis, or arteriogenesis; preparing a blood-derived compound using a method comprising: harvesting marrow blood from the patient; separating a blood composition from the marrow blood; culturing the blood composition in a culture medium under a selective pressure of about 1% to about 10% oxygen and a pH of about 6.6 to about 7.0 for 1-28 days; selecting mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; generating the blood-derived compound by suspending the selected mesenchymal stem cells in blood plasma harvested from the patient; and returning a therapeutically effective amount of the blood derived compound to the patient at the degenerative cartilage. 2. The method of claim 1, wherein the one or more peptides are selected from the group consisting of: VEGF-A, PIGF, VEGF-B, VEGF-C, VEGF-D, TGF-β, Ang-1, Ang-2, IGF, HGF, FGF, Tie2, PDGF, CCL2, Alpha-V Beta-5, Alpha-5 Beta-1, VE-cadherin, PECAM-1, plasminogen activator, nitrogen oxide synthase, and combinations thereof. 3. The method of claim 1, wherein the growth factor is selected from the group consisting of: TGF-β, FGF, PDGF, IGF, and combinations thereof. 4. The method of claim 1, wherein harvesting the marrow blood further comprises: utilizing a needle aspirate technique to draw the marrow blood from the posterior superior iliac spine. 5. A method for treating an avascular zone in a patient in need thereof, the method comprising: culturing nucleated cells harvested from the patient in need thereof in a culture medium under a selective pressure of about 1% to about 10% oxygen for 1-28 days; selecting viable mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; and providing the selected, viable mesenchymal stem cells for implantation in the avascular zone. 6. The method of claim 5, wherein the selective pressure is about 3 to about 7% oxygen. 7. The method of claim 5, wherein the selective pressure further comprises about 2% to about 10% carbon dioxide. 8. The method of claim 7, wherein the selective pressure further comprises a pH of about 6.6 to about 7.0. 9. The method of claim 8, wherein the pH is about 6.6 to about 6.8. 10. The method of claim 5, wherein the selective pressure further comprises an osmolarity of about 350 mOsm to about 600 mOsm. 11. The method of claim 5, wherein the culture medium is supplemented with about 10 to about 20% human platelet cell lysate. 12. The method of claim 5, further comprising: implanting the selected, viable mesenchymal stem cells; implanting platelet cell lysate prior to, during, or after implanting the selected, viable mesenchymal stem cells; wherein the implanting of the platelet cell lysate and the selected, viable mesenchymal stem cells is in the avascular zone. 13. The method of claim 5, further comprising: implanting the selected, viable mesenchymal stem cells; implanting platelets prior to, during, or after implanting the selected, viable mesenchymal stem cells; wherein the implanting of the platelets and the selected, viable mesenchymal stem cells is in the avascular zone. 14. The method of claim 13, wherein the platelets are harvested from the patient in need thereof. 15. The method of claim 14, wherein the platelets are treated with thrombin and calcium chloride 1-7 days prior to implanting. 16. The method of claim 15, wherein the amount of thrombin is about 28.56 U/ml and the amount of calcium chloride is about 2.86 mg/ml. 17. The method of claim 14, wherein the platelets are treated with a composition selected from the group consisting of thrombin, calcium chloride or its salts, thromboxane A2, adenosine triphosphate, arachidonate, and combinations thereof. 18. The method of claim 5, further comprising: administering one or more compounds selected from the group consisting of growth factors, cytokines, integrins, cadherins, molecules or drugs that promote angiogenesis, molecules or drugs that promote vasculogenesis, molecules or drugs that promote aerteriogenesis, and combinations thereof. 19. The method of claim 18, wherein the one or more compounds is selected from a group consisting of VEGF-A, PIGF, VEGF-B, VEGF-C, VEGF-D, TGF-β, Ang-1, Ang-2, IGF, HGF, FGF, Tie2, PDGF, CCL2, Alpha-V Beta-5, Alpha-5 Beta-1, VE-cadherin, PECAM-1, plasminogen activator, nitrogen oxide synthase, and combinations thereof. 20. The method of claim 5, further comprising: administering one or more growth factors before, during, or after implanting the selected, viable mesenchymal stem cells in the avascular zone. 21. The method of claim 20, wherein the one or more growth factors is selected from the group consisting of: TGF-β, FGF, PDGF, IGF, and combinations thereof. 22. The method of claim 5, wherein the culturing is for about two to about five passages. 23. A method for treating a degenerative intervertebral disc in a patient in need thereof, the method comprising: culturing nucleated cells harvested from the patient in need thereof in a culture medium under a selective pressure of about 1% to about 10% oxygen for 1-28 days; selecting viable mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; and providing the selected, viable mesenchymal stem cells for implantation in an intervertebral disc. 24. A method for treating a degenerative intervertebral disc in a patient in need thereof, the method comprising: culturing nucleated cells harvested from the patient in need thereof in a culture medium under a selective pressure of about 1% to about 10% oxygen for 1-28 days; selecting viable mesenchymal stem cells capable of growth in the culture medium under the selective pressure of about 1% to about 10% oxygen; and implanting the selected, viable mesenchymal stem cells in an intervertebral disc.
1,600
322
13,120,112
1,619
The invention relates to a pH-dependent controlled release pharmaceutical composition, comprising at least one pharmaceutical active ingredient, with the exception of opioids, wherein the core is coated at least by one coating layer, controlling the release of the pharmaceutical composition, wherein the coating layer comprises a polymer mixture of i) 40-95% by weight, based on dry weight of the polymer mixture, of at least one water insoluble essentially neutral vinyl polymer or copolymer, and ii) 5-60% by weight, based on dry weight of the polymer mixture, of at least one anionic polymer or copolymer, which is insoluble in a buffered medium below pH 4.0 and soluble at least in the range from pH 7.0 to pH 8.0, characterized in that the coating layer further comprises 110 to 250% by weight of a non-porous inert lubricant, 1 to 35% by weight of at least one neutral cellulosic compound and 1 to 25% by weight of at least one emulsifier, each calculated on dry weight of the polymer mixture.
1. A pH-dependent controlled release pharmaceutical composition, comprising: a core comprising at least one pharmaceutical active ingredient, with the exception of opioids, wherein the core is coated by at least one coating layer controlling the release of the pharmaceutical composition, the at least one coating layer comprises (A) a polymer mixture of i) 40-95% by weight, based on dry weight of the polymer mixture, of at least one water insoluble essentially neutral vinyl polymer or copolymer, and ii) 5-60% by weight, based on dry weight of the polymer mixture, of at least one anionic polymer or copolymer, which is insoluble in a buffered medium below pH 4.0 and soluble at least in the range from pH 7.0 to pH 8.0, (B) 110 to 250% by weight of a non-porous inert lubricant, (C) 1 to 35% by weight of at least one neutral cellulosic compound and (D) 1 to 25% by weight of at least one emulsifier, and (B), (C), and (D) are each calculated on dry weight of the polymer mixture. 2. The controlled release pharmaceutical composition according to claim 1, wherein the non-porous inert lubricant is a layered silica component, a pigment, or a stearate compound. 3. The controlled release pharmaceutical composition according to claim 2, wherein the inert lubricant is talc. 4. The controlled release pharmaceutical composition according to claim 2, wherein the inert lubricant is Ca- or Mg-stearate. 5. The controlled release pharmaceutical composition according to claim 1, wherein the water insoluble essentially neutral vinyl polymer or copolymer is a copolymer comprising at least one free-radical polymerized unit of more than 95 up to 100% by weight of at least one C1- to C4-alkyl ester of acrylic or of methacrylic acid, and less than 5% by weight of acrylic or methacrylic acid. 6. The controlled release pharmaceutical composition according to claim 1, wherein the water insoluble essentially neutral polymer is a polyvinyl acetate polymer or copolymer. 7. The controlled release pharmaceutical composition according to claim 1, wherein the water soluble anionic polymer is a (meth)acrylate copolymer comprising free-radical polymerized units of 25 to 95% by weight of at least one C1- to C4-alkyl ester of acrylic or of methacrylic acid, and 5 to 75% by weight of at least one (meth)acrylate monomer having an anionic group. 8. The controlled release pharmaceutical composition according to claim 7, wherein the water soluble anionic polymer comprises at least one free-radical polymerized unit of 10 to 30% by weight methyl methacrylate, 50 to 70% by weight methyl acrylate, and 5 to 15% by weight methacrylic acid. 9. The controlled release pharmaceutical composition according to claim 1, wherein the neutral cellulosic compound is hydroxypropylmethylcellulose. 10. The controlled release pharmaceutical composition according to claim 1, wherein the emulsifier is a nonionic emulsifier. 11. The controlled release pharmaceutical composition according to claim 10, wherein the emulsifier is polyoxyethylene derivative of a sorbitan ester. 12. The controlled release pharmaceutical composition according to claim 10, wherein the emulsifier is a polyethoxy sorbitan monooleate. 13. The controlled release pharmaceutical composition according to claim 1, wherein, under in-vitro conditions according to USP paddle, 100 rpm, buffered at pH 6.8 in a medium with and without an addition of 40% (v/v) ethanol, the controlled release pharmaceutical composition has: when the pharmaceutical active ingredient is released to a degree of less than 20% without the addition of 40% (v/v) ethanol, a difference in the release rate with the addition of 40% (v/v) ethanol is not more than plus or minus 15% of a corresponding release value without 40% (v/v) ethanol, and when the pharmaceutical active ingredient is released to a degree of 20-80% without the addition of 40% (v/v) ethanol, the difference in the release rate with the addition of 40% (v/v) ethanol is not more than plus or minus 30% of the corresponding release value without 40% (v/v) ethanol. 14. The controlled release pharmaceutical composition according to claim 1, wherein the pharmaceutical active ingredient is metoprolol or a pharmaceutically acceptable salt of metoprolol. 15. The controlled release pharmaceutical composition according to claim 1, wherein the controlled release pharmaceutical composition is in a form of pellet comprised in a multiparticulate pharmaceutical form selected from the group consisting of a compressed tablet, a capsule, a sachet, an effervescent tablet, and a reconstitutable powder. 16. The controlled release pharmaceutical composition according to claim 1, comprising at least one of a sub coat and a top coat. 17. The controlled release pharmaceutical composition according to claim 1, wherein the controlled release pharmaceutical composition is present in a form of at least one coated pellet with an overall average diameter in the range of from 100 to 5000 μm. 18. The controlled release pharmaceutical composition according to claim 17, wherein the at least one coated pellet has an overall average diameter in the range between 100 to 700 μm. 19. The controlled release pharmaceutical composition according to claim 17, wherein the at least one coated pellet has an overall average diameter in the range between 1400 to 5000 μm. 20. The controlled release pharmaceutical composition according to claim 19, wherein the coating layer is present in an amount of at least 30% by weight relative to a weight of core. 21. The controlled release pharmaceutical composition according to claim 1, wherein the active ingredient is released to a degree of 10% or less in simulated gastric fluid pH 1.2 with or without an addition of 40% ethanol (v/v) within 2 hours. 22. A process for preparing a controlled release pharmaceutical composition according to claim 1, comprising: direct compressing the controlled release pharmaceutical composition, compressing at least one dry, wet or sintered granule of the controlled release pharmaceutical composition, and subsequent rounding off, wet or dry granulating or direct pelleting the controlled release pharmaceutical composition, or binding at least one powder (powder layering) onto at least one active ingredient-free bead or neutral core (nonpareilles) or at least one active ingredient-containing particle and by applying the polymer coating in a spray process or by fluidized bed granulation. 23. A method for reducing a risk of enhanced or reduced release of the included pharmaceutical active ingredient after oral ingestion by simultaneous or subsequent consumption of ethanol containing drinks, comprising: administering the controlled release pharmaceutical composition of claim 1 to a subject in need thereof.
The invention relates to a pH-dependent controlled release pharmaceutical composition, comprising at least one pharmaceutical active ingredient, with the exception of opioids, wherein the core is coated at least by one coating layer, controlling the release of the pharmaceutical composition, wherein the coating layer comprises a polymer mixture of i) 40-95% by weight, based on dry weight of the polymer mixture, of at least one water insoluble essentially neutral vinyl polymer or copolymer, and ii) 5-60% by weight, based on dry weight of the polymer mixture, of at least one anionic polymer or copolymer, which is insoluble in a buffered medium below pH 4.0 and soluble at least in the range from pH 7.0 to pH 8.0, characterized in that the coating layer further comprises 110 to 250% by weight of a non-porous inert lubricant, 1 to 35% by weight of at least one neutral cellulosic compound and 1 to 25% by weight of at least one emulsifier, each calculated on dry weight of the polymer mixture.1. A pH-dependent controlled release pharmaceutical composition, comprising: a core comprising at least one pharmaceutical active ingredient, with the exception of opioids, wherein the core is coated by at least one coating layer controlling the release of the pharmaceutical composition, the at least one coating layer comprises (A) a polymer mixture of i) 40-95% by weight, based on dry weight of the polymer mixture, of at least one water insoluble essentially neutral vinyl polymer or copolymer, and ii) 5-60% by weight, based on dry weight of the polymer mixture, of at least one anionic polymer or copolymer, which is insoluble in a buffered medium below pH 4.0 and soluble at least in the range from pH 7.0 to pH 8.0, (B) 110 to 250% by weight of a non-porous inert lubricant, (C) 1 to 35% by weight of at least one neutral cellulosic compound and (D) 1 to 25% by weight of at least one emulsifier, and (B), (C), and (D) are each calculated on dry weight of the polymer mixture. 2. The controlled release pharmaceutical composition according to claim 1, wherein the non-porous inert lubricant is a layered silica component, a pigment, or a stearate compound. 3. The controlled release pharmaceutical composition according to claim 2, wherein the inert lubricant is talc. 4. The controlled release pharmaceutical composition according to claim 2, wherein the inert lubricant is Ca- or Mg-stearate. 5. The controlled release pharmaceutical composition according to claim 1, wherein the water insoluble essentially neutral vinyl polymer or copolymer is a copolymer comprising at least one free-radical polymerized unit of more than 95 up to 100% by weight of at least one C1- to C4-alkyl ester of acrylic or of methacrylic acid, and less than 5% by weight of acrylic or methacrylic acid. 6. The controlled release pharmaceutical composition according to claim 1, wherein the water insoluble essentially neutral polymer is a polyvinyl acetate polymer or copolymer. 7. The controlled release pharmaceutical composition according to claim 1, wherein the water soluble anionic polymer is a (meth)acrylate copolymer comprising free-radical polymerized units of 25 to 95% by weight of at least one C1- to C4-alkyl ester of acrylic or of methacrylic acid, and 5 to 75% by weight of at least one (meth)acrylate monomer having an anionic group. 8. The controlled release pharmaceutical composition according to claim 7, wherein the water soluble anionic polymer comprises at least one free-radical polymerized unit of 10 to 30% by weight methyl methacrylate, 50 to 70% by weight methyl acrylate, and 5 to 15% by weight methacrylic acid. 9. The controlled release pharmaceutical composition according to claim 1, wherein the neutral cellulosic compound is hydroxypropylmethylcellulose. 10. The controlled release pharmaceutical composition according to claim 1, wherein the emulsifier is a nonionic emulsifier. 11. The controlled release pharmaceutical composition according to claim 10, wherein the emulsifier is polyoxyethylene derivative of a sorbitan ester. 12. The controlled release pharmaceutical composition according to claim 10, wherein the emulsifier is a polyethoxy sorbitan monooleate. 13. The controlled release pharmaceutical composition according to claim 1, wherein, under in-vitro conditions according to USP paddle, 100 rpm, buffered at pH 6.8 in a medium with and without an addition of 40% (v/v) ethanol, the controlled release pharmaceutical composition has: when the pharmaceutical active ingredient is released to a degree of less than 20% without the addition of 40% (v/v) ethanol, a difference in the release rate with the addition of 40% (v/v) ethanol is not more than plus or minus 15% of a corresponding release value without 40% (v/v) ethanol, and when the pharmaceutical active ingredient is released to a degree of 20-80% without the addition of 40% (v/v) ethanol, the difference in the release rate with the addition of 40% (v/v) ethanol is not more than plus or minus 30% of the corresponding release value without 40% (v/v) ethanol. 14. The controlled release pharmaceutical composition according to claim 1, wherein the pharmaceutical active ingredient is metoprolol or a pharmaceutically acceptable salt of metoprolol. 15. The controlled release pharmaceutical composition according to claim 1, wherein the controlled release pharmaceutical composition is in a form of pellet comprised in a multiparticulate pharmaceutical form selected from the group consisting of a compressed tablet, a capsule, a sachet, an effervescent tablet, and a reconstitutable powder. 16. The controlled release pharmaceutical composition according to claim 1, comprising at least one of a sub coat and a top coat. 17. The controlled release pharmaceutical composition according to claim 1, wherein the controlled release pharmaceutical composition is present in a form of at least one coated pellet with an overall average diameter in the range of from 100 to 5000 μm. 18. The controlled release pharmaceutical composition according to claim 17, wherein the at least one coated pellet has an overall average diameter in the range between 100 to 700 μm. 19. The controlled release pharmaceutical composition according to claim 17, wherein the at least one coated pellet has an overall average diameter in the range between 1400 to 5000 μm. 20. The controlled release pharmaceutical composition according to claim 19, wherein the coating layer is present in an amount of at least 30% by weight relative to a weight of core. 21. The controlled release pharmaceutical composition according to claim 1, wherein the active ingredient is released to a degree of 10% or less in simulated gastric fluid pH 1.2 with or without an addition of 40% ethanol (v/v) within 2 hours. 22. A process for preparing a controlled release pharmaceutical composition according to claim 1, comprising: direct compressing the controlled release pharmaceutical composition, compressing at least one dry, wet or sintered granule of the controlled release pharmaceutical composition, and subsequent rounding off, wet or dry granulating or direct pelleting the controlled release pharmaceutical composition, or binding at least one powder (powder layering) onto at least one active ingredient-free bead or neutral core (nonpareilles) or at least one active ingredient-containing particle and by applying the polymer coating in a spray process or by fluidized bed granulation. 23. A method for reducing a risk of enhanced or reduced release of the included pharmaceutical active ingredient after oral ingestion by simultaneous or subsequent consumption of ethanol containing drinks, comprising: administering the controlled release pharmaceutical composition of claim 1 to a subject in need thereof.
1,600
323
14,773,088
1,649
This invention discloses methods and compositions for the treatment of demyelinating disorders. Specifically, the invention relates to the use guanabenz or guanabenz derivative for treating demyelinating disorders.
1. A method of increasing viability of a neuronal cell in a subject suffering from a demyelinating disorder, comprising administering to the subject an effective amount of a compound of Formula I: or a derivative or pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, and R5 are independently hydrogen, deuterium, halogen, haloalkyl, alkyl, alkoxy, hydroxyl, aryl, or aryloxy, thereby increasing viability of a neuronal cell in said subject. 2. A method of treating, reducing severity, reducing relapse, or delaying onset of a symptom of a demyelinating disorder in a subject in need thereof, comprising the step of administering to the subject in need thereof an effective amount of a compound of Formula I: or a derivative or pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, and R5 are independently hydrogen, deuterium, halogen, haloalkyl, alkyl, alkoxy, hydroxyl, aryl, or aryloxy, thereby treating a symptom of a demyelinating disorder in said subject. 3-6. (canceled) 7. The method of claim 2, wherein R1 and R5 are not hydrogen and/or wherein R1 and R5 are halogen. 8. (canceled) 9. The method of claim 2, wherein the compound of Formula I is of Formula II or pharmaceutically acceptable salt thereof: 10. (canceled) 11. The method of claim 2, wherein the demyelinating disorder is selected from the group consisting of: multiple sclerosis, acute disseminated encephalomyelitis, periventricular leukomalacia, periventricular white matter injury, Tabes Dorsalis, Devic's disease, optic neuritis, progressive multifocal leukoencephalopathy, transverse myelitis, chronic inflammatory demyelinating polyneuropathy, anti-MAG peripheral neuropathy, adrenoleukodystrophy, adrenomyeloneuropathy, Guillain-Barré Syndrome, central pontine myelinolysis, diffuse white matter injury, inherited demyelinating diseases such as leukodystrophy, and Charcot Marie Tooth Disease. 12. The method of claim 2, wherein a symptom of a demyelinating disorder is selected from the group consisting of: fatigue, somatosensory dysfunction, tingling, pain, numbness, balance problems, problems with walking, changes in vision, depression, emotional changes, mood swings, impaired cognition, muscle dysfunction, impaired muscle coordination, sexual impairment, speech impairment, swallowing impairment, bladder dysfunction, bowel dysfunction. 13. The method of claim 2, wherein about 1 mg to about 64 mg of the compound is administered to the subject. 14. (canceled) 15. The method of claim 2, wherein the compound is administered orally. 16. The method of claim 1, wherein the neuronal cell is a neuron. 17-26. (canceled) 27. The method of claim 2, wherein the administration of the compound is combined with one or more of glatiramer acetate, dimethyl fumerate (BG-12), fingolimod (FTY720), interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, dalfampridine, teriflunomide, and/or daclizumab. 28-36. (canceled) 37. The method of claim 2, wherein the compound of Formula I is guanabenz. 38. The method of claim 37, wherein about 64 mg/day of said guanabenz is administered to said subject. 39-40. (canceled) 41. A composition comprising a compound of Formula I: or a derivative or pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, and R5 are independently hydrogen, deuterium, halogen, haloalkyl, alkyl, alkoxy, hydroxyl, aryl, or aryloxy, and at least one other agent useful for the treatment of a demyelinating disorder. 42. The composition of claim 41, wherein said compound of Formula I is guanabenz. 43. (canceled) 44. The composition of claim 41, wherein the composition is formulated for subcutaneous or intravenous injection. 45-46. (canceled) 47. The composition of claim 41, wherein said at least one other agent is glatiramer acetate, dimethyl fumerate (BG-12), fingolimod (FTY720), interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, dalfampridine, teriflunomide, and/or daclizumab. 48-56. (canceled) 57. The composition of claim 41, wherein R1 and R5 are not hydrogen and/or wherein R1 and R5 are halogen. 58. The composition of claim 41, wherein the compound of Formula I is of Formula II or pharmaceutically acceptable salt thereof: 59. The method of claim 2, wherein the compound of Formula I is: or a pharmaceutically acceptable salt thereof. 60. The composition of claim 41, wherein the compound of Formula I is: or a pharmaceutically acceptable salt thereof.
This invention discloses methods and compositions for the treatment of demyelinating disorders. Specifically, the invention relates to the use guanabenz or guanabenz derivative for treating demyelinating disorders.1. A method of increasing viability of a neuronal cell in a subject suffering from a demyelinating disorder, comprising administering to the subject an effective amount of a compound of Formula I: or a derivative or pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, and R5 are independently hydrogen, deuterium, halogen, haloalkyl, alkyl, alkoxy, hydroxyl, aryl, or aryloxy, thereby increasing viability of a neuronal cell in said subject. 2. A method of treating, reducing severity, reducing relapse, or delaying onset of a symptom of a demyelinating disorder in a subject in need thereof, comprising the step of administering to the subject in need thereof an effective amount of a compound of Formula I: or a derivative or pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, and R5 are independently hydrogen, deuterium, halogen, haloalkyl, alkyl, alkoxy, hydroxyl, aryl, or aryloxy, thereby treating a symptom of a demyelinating disorder in said subject. 3-6. (canceled) 7. The method of claim 2, wherein R1 and R5 are not hydrogen and/or wherein R1 and R5 are halogen. 8. (canceled) 9. The method of claim 2, wherein the compound of Formula I is of Formula II or pharmaceutically acceptable salt thereof: 10. (canceled) 11. The method of claim 2, wherein the demyelinating disorder is selected from the group consisting of: multiple sclerosis, acute disseminated encephalomyelitis, periventricular leukomalacia, periventricular white matter injury, Tabes Dorsalis, Devic's disease, optic neuritis, progressive multifocal leukoencephalopathy, transverse myelitis, chronic inflammatory demyelinating polyneuropathy, anti-MAG peripheral neuropathy, adrenoleukodystrophy, adrenomyeloneuropathy, Guillain-Barré Syndrome, central pontine myelinolysis, diffuse white matter injury, inherited demyelinating diseases such as leukodystrophy, and Charcot Marie Tooth Disease. 12. The method of claim 2, wherein a symptom of a demyelinating disorder is selected from the group consisting of: fatigue, somatosensory dysfunction, tingling, pain, numbness, balance problems, problems with walking, changes in vision, depression, emotional changes, mood swings, impaired cognition, muscle dysfunction, impaired muscle coordination, sexual impairment, speech impairment, swallowing impairment, bladder dysfunction, bowel dysfunction. 13. The method of claim 2, wherein about 1 mg to about 64 mg of the compound is administered to the subject. 14. (canceled) 15. The method of claim 2, wherein the compound is administered orally. 16. The method of claim 1, wherein the neuronal cell is a neuron. 17-26. (canceled) 27. The method of claim 2, wherein the administration of the compound is combined with one or more of glatiramer acetate, dimethyl fumerate (BG-12), fingolimod (FTY720), interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, dalfampridine, teriflunomide, and/or daclizumab. 28-36. (canceled) 37. The method of claim 2, wherein the compound of Formula I is guanabenz. 38. The method of claim 37, wherein about 64 mg/day of said guanabenz is administered to said subject. 39-40. (canceled) 41. A composition comprising a compound of Formula I: or a derivative or pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, and R5 are independently hydrogen, deuterium, halogen, haloalkyl, alkyl, alkoxy, hydroxyl, aryl, or aryloxy, and at least one other agent useful for the treatment of a demyelinating disorder. 42. The composition of claim 41, wherein said compound of Formula I is guanabenz. 43. (canceled) 44. The composition of claim 41, wherein the composition is formulated for subcutaneous or intravenous injection. 45-46. (canceled) 47. The composition of claim 41, wherein said at least one other agent is glatiramer acetate, dimethyl fumerate (BG-12), fingolimod (FTY720), interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, dalfampridine, teriflunomide, and/or daclizumab. 48-56. (canceled) 57. The composition of claim 41, wherein R1 and R5 are not hydrogen and/or wherein R1 and R5 are halogen. 58. The composition of claim 41, wherein the compound of Formula I is of Formula II or pharmaceutically acceptable salt thereof: 59. The method of claim 2, wherein the compound of Formula I is: or a pharmaceutically acceptable salt thereof. 60. The composition of claim 41, wherein the compound of Formula I is: or a pharmaceutically acceptable salt thereof.
1,600
324
14,110,549
1,619
Multilayer thin film devices that include a bioactive agent for elution to the surrounding tissue upon administration to a subject are provided. The multilayer thin film devices are useful as medical devices, such as ocular devices. Also provided are methods and kits for localized delivery of a bioactive agent to a tissue of a subject, and methods of preparing the subject devices. The multilayer thin film medical device includes a first layer, a bioactive agent and a second layer. The first and the second layers may be porous or non-porous. The devices have a furled structure, suitable for administration to a subject.
1-53. (canceled) 54. A method of localized delivery of a bioactive agent to a tissue, the method comprising: positioning in a subject a multilayer thin film medical device comprising: a first layer comprising a polymer and a pore forming agent; a bioactive agent; and a second layer in contact with the bioactive agent; wherein the bioactive agent is positioned between the first layer and the second layer, and wherein the multilayer thin film structure is in a furled state; unfurling the furled multilayer thin film structure upon contact with a hydrating liquid in the subject to produce an unfurled multilayer thin film structure; dissolving the pore forming agent from the first layer of the unfurled multilayer thin film structure to produce a porous first layer; and releasing the bioactive agent from the medical device through the porous first layer to locally deliver the bioactive agent to the tissue. 55. The method of claim 54, wherein the unfurling and dissolving occurs simultaneously. 56. The method of claim 54, wherein the unfurling and dissolving occurs sequentially. 57. The method of claim 54, wherein the releasing locally delivers an effective amount of the bioactive agent over an extended period of time. 58. The method of claim 54, wherein the releasing of the bioactive agent from the medical device is substantially zero order. 59. The method of claim 54, wherein the positioning is in the eye(s) of the subject. 60. The method of claim 54, wherein the positioning comprises injecting the furled device into the eye(s) of the subject. 61. The method of claim 60, wherein the injecting comprises intravitreal injection. 62. The method of claim 60, wherein the injecting comprises subretinal injection. 63. The method of claim 54, wherein the bioactive agent is a protein therapeutic, a small molecule drug, a large molecule drug or an aptamer. 64. The method of claim 63, wherein the bioactive agent is a protein therapeutic selected from ranibizumab, bevacizumab, trastuzumab, rituximab, gentuzumab ozogamicin and cetuximab. 65. The method of claim 54, wherein the polymer is a biodegradable polymer. 66. The method of claim 65, wherein the biodegradable polymer is selected from the group consisting of a PLA, PGA, PCL, PLGA, PLCL, and combinations thereof. 67. The method of claim 54, wherein the pore forming agent is selected from the group consisting of gelatin, PEG, chitosan, or agarose. 68. The method of claim 54, wherein the second layer is porous or non-porous. 69. The method of claim 54, wherein the ratio of biodegradable polymer to pore forming agent in the first layer of the furled multilayer thin film structure is between about 1:5 and about 99:1. 70. The method of claim 69, wherein the ratio of biodegradable polymer to pore forming agent in the first layer of the furled multilayer thin film structure is about 9:1. 71. The method of claim 54, wherein the % porosity of the porous first layer of the unfurled multilayer thin film structure is between about 20% and about 0.01%. 72. The method of claim 54, wherein the porous first layer of the unfurled multilayer thin film structure has an average pore size of 30 μm or less. 73. The method of claim 72, wherein the porous first layer of the unfurled multilayer thin film structure has an average pore size of 10 μm or less. 74. The method of claim 54, wherein the unfurled multilayer thin film structure has an area between 0.7 mm2 and 2000 mm2. 75. The method of claim 54, wherein the multilayer thin film medical device further comprises a third nanostructured porous layer positioned between the first layer and the reservoir of the bioactive agent, wherein the third nanostructured porous layer comprises a biodegradable polymer. 76. The method of claim 75, wherein the third nanostructured porous layer has an average pore size of between about 1 nm and about 100 nm. 77. The method of claim 75, wherein the biodegradable polymer is selected from the group consisting of a PLA, PGA, PCL, PLGA, PLCL, and combinations thereof. 78. The method of claim 54, wherein the second layer is biodegradable. 79-137. (canceled)
Multilayer thin film devices that include a bioactive agent for elution to the surrounding tissue upon administration to a subject are provided. The multilayer thin film devices are useful as medical devices, such as ocular devices. Also provided are methods and kits for localized delivery of a bioactive agent to a tissue of a subject, and methods of preparing the subject devices. The multilayer thin film medical device includes a first layer, a bioactive agent and a second layer. The first and the second layers may be porous or non-porous. The devices have a furled structure, suitable for administration to a subject.1-53. (canceled) 54. A method of localized delivery of a bioactive agent to a tissue, the method comprising: positioning in a subject a multilayer thin film medical device comprising: a first layer comprising a polymer and a pore forming agent; a bioactive agent; and a second layer in contact with the bioactive agent; wherein the bioactive agent is positioned between the first layer and the second layer, and wherein the multilayer thin film structure is in a furled state; unfurling the furled multilayer thin film structure upon contact with a hydrating liquid in the subject to produce an unfurled multilayer thin film structure; dissolving the pore forming agent from the first layer of the unfurled multilayer thin film structure to produce a porous first layer; and releasing the bioactive agent from the medical device through the porous first layer to locally deliver the bioactive agent to the tissue. 55. The method of claim 54, wherein the unfurling and dissolving occurs simultaneously. 56. The method of claim 54, wherein the unfurling and dissolving occurs sequentially. 57. The method of claim 54, wherein the releasing locally delivers an effective amount of the bioactive agent over an extended period of time. 58. The method of claim 54, wherein the releasing of the bioactive agent from the medical device is substantially zero order. 59. The method of claim 54, wherein the positioning is in the eye(s) of the subject. 60. The method of claim 54, wherein the positioning comprises injecting the furled device into the eye(s) of the subject. 61. The method of claim 60, wherein the injecting comprises intravitreal injection. 62. The method of claim 60, wherein the injecting comprises subretinal injection. 63. The method of claim 54, wherein the bioactive agent is a protein therapeutic, a small molecule drug, a large molecule drug or an aptamer. 64. The method of claim 63, wherein the bioactive agent is a protein therapeutic selected from ranibizumab, bevacizumab, trastuzumab, rituximab, gentuzumab ozogamicin and cetuximab. 65. The method of claim 54, wherein the polymer is a biodegradable polymer. 66. The method of claim 65, wherein the biodegradable polymer is selected from the group consisting of a PLA, PGA, PCL, PLGA, PLCL, and combinations thereof. 67. The method of claim 54, wherein the pore forming agent is selected from the group consisting of gelatin, PEG, chitosan, or agarose. 68. The method of claim 54, wherein the second layer is porous or non-porous. 69. The method of claim 54, wherein the ratio of biodegradable polymer to pore forming agent in the first layer of the furled multilayer thin film structure is between about 1:5 and about 99:1. 70. The method of claim 69, wherein the ratio of biodegradable polymer to pore forming agent in the first layer of the furled multilayer thin film structure is about 9:1. 71. The method of claim 54, wherein the % porosity of the porous first layer of the unfurled multilayer thin film structure is between about 20% and about 0.01%. 72. The method of claim 54, wherein the porous first layer of the unfurled multilayer thin film structure has an average pore size of 30 μm or less. 73. The method of claim 72, wherein the porous first layer of the unfurled multilayer thin film structure has an average pore size of 10 μm or less. 74. The method of claim 54, wherein the unfurled multilayer thin film structure has an area between 0.7 mm2 and 2000 mm2. 75. The method of claim 54, wherein the multilayer thin film medical device further comprises a third nanostructured porous layer positioned between the first layer and the reservoir of the bioactive agent, wherein the third nanostructured porous layer comprises a biodegradable polymer. 76. The method of claim 75, wherein the third nanostructured porous layer has an average pore size of between about 1 nm and about 100 nm. 77. The method of claim 75, wherein the biodegradable polymer is selected from the group consisting of a PLA, PGA, PCL, PLGA, PLCL, and combinations thereof. 78. The method of claim 54, wherein the second layer is biodegradable. 79-137. (canceled)
1,600
325
13,132,840
1,632
Compositions and methods are provided for repairing damaged avascular zones, including intervertebral disc, in a patient in need thereof.
1. A method for treating a degenerative intervertebral disc in a patient in need thereof comprising: harvesting stem cells from the patient in need thereof; culturing the harvested stem cells under selective pressure that corresponds to the environment of a damaged intervertebral disc; and implanting the selected stem cells in a posterior disc annulus of the degenerative intervertebral disc. 2. The method of claim 1 wherein the selective pressure include culturing the harvested stem cells in from about 3 to about 10% oxygen. 3. The method of claim 2 wherein the selective pressure includes culturing the harvested stem cells in from about 3 to about 7% oxygen. 4. The method of claim 1 wherein the selective pressure include culturing the harvested stem cells in from about 2 to about 10% carbon dioxide. 5. The method of claim 2 further comprising the selective pressure including culturing the harvested stem cells in from about 2 to about 10% carbon dioxide. 6. The method of claim 1 further comprising: harvesting NP cells from the patient in need thereof; and co-culturing the NP cells with the harvested stem cells under selective pressure. 7. The method of claim 1 wherein the stem cells are mesenchymal stem cells. 8. The method of claim 1 further comprising: harvesting platelets from the patient in need thereof; and implanting the platelets prior to, during or after implanting selected stem cells; wherein the implanting of both platelets and stem cells is in the posterior disc annulus. 9. The method of claim 8 wherein the platelets are treated with thrombin and calcium chloride 1-7 days prior to implanting in posterior disc annulus. 10. The method of claim 9 wherein the amount of thrombin is 28.56 U/ml and the amount of calcium chloride is 2.86 mg/ml. 11. The method of claim 8 wherein the platelets are treated with thrombin, calcium chloride or its salts, thromboxane A2, adenosine triphosphate and arichidonate. 12. The method of claim 1 further comprising administering one or more compounds selected from the group consisting of growth factors, cytokines, integrins cadherins, molecules or drugs that promote angiogenesis, molecules or drugs that promote vasculogenesis and molecules or drugs that promote aerteriogenesis. 13. The method of claim 12 wherein the compound is VEGF-A, PIGF, VEGF-B, VEGF-C, VEGF-D, TGF-β, Ang-1, Ang-2, IGF, HGF, FGF, Tie2, PDGF, CCL2, Alpha-V Beta-5, Alpha-5 Beta-1, VE-cadherin, PECAM-1, plasminogen activator, or nitrogen oxide synthase. 14. The method of claim 1 further comprising administering one or more growth factors before, during or after implanting selected stem cells in the posterior disc annulus. 15. The method of claim 14 wherein the one or more growth factor is selected from the group consisting of: TGF-β, FGF, PDGF, and IGF. 16. The method of claim 1 wherein the culturing of harvested stem cells is performed in a basal culture media made from a D-MEM base. 17. A pharmaceutical composition comprising stem cells, platelets, one or more growth factors, and a pharmaceutical carrier or diluent. 18. The composition of claim 17 wherein the stem cells are mesenchymal stem cells. 19. The composition of claim 17 wherein the stem cells have been selected for their capacity for viability and expansion under hypoxic conditions. 20. The compositions of claim 17 wherein the stem cells have been selected for their capacity for viability and expansion under culture conditions comparable to a damaged intervertebral disc. 21. A method for treating an avascular zone in a patient in need thereof comprising: harvesting stem cells from the patient in need thereof; culturing the harvested stem cells under selective pressure that corresponds to the environment of a damaged or aged avascular zone; and implanting the selected stem cells in the avascular zone. 22. The method of claim 21 wherein the avascular zone is in a patient's shoulder. 23. The method of claim 21 wherein the avascular zone is in a patient's hip.
Compositions and methods are provided for repairing damaged avascular zones, including intervertebral disc, in a patient in need thereof.1. A method for treating a degenerative intervertebral disc in a patient in need thereof comprising: harvesting stem cells from the patient in need thereof; culturing the harvested stem cells under selective pressure that corresponds to the environment of a damaged intervertebral disc; and implanting the selected stem cells in a posterior disc annulus of the degenerative intervertebral disc. 2. The method of claim 1 wherein the selective pressure include culturing the harvested stem cells in from about 3 to about 10% oxygen. 3. The method of claim 2 wherein the selective pressure includes culturing the harvested stem cells in from about 3 to about 7% oxygen. 4. The method of claim 1 wherein the selective pressure include culturing the harvested stem cells in from about 2 to about 10% carbon dioxide. 5. The method of claim 2 further comprising the selective pressure including culturing the harvested stem cells in from about 2 to about 10% carbon dioxide. 6. The method of claim 1 further comprising: harvesting NP cells from the patient in need thereof; and co-culturing the NP cells with the harvested stem cells under selective pressure. 7. The method of claim 1 wherein the stem cells are mesenchymal stem cells. 8. The method of claim 1 further comprising: harvesting platelets from the patient in need thereof; and implanting the platelets prior to, during or after implanting selected stem cells; wherein the implanting of both platelets and stem cells is in the posterior disc annulus. 9. The method of claim 8 wherein the platelets are treated with thrombin and calcium chloride 1-7 days prior to implanting in posterior disc annulus. 10. The method of claim 9 wherein the amount of thrombin is 28.56 U/ml and the amount of calcium chloride is 2.86 mg/ml. 11. The method of claim 8 wherein the platelets are treated with thrombin, calcium chloride or its salts, thromboxane A2, adenosine triphosphate and arichidonate. 12. The method of claim 1 further comprising administering one or more compounds selected from the group consisting of growth factors, cytokines, integrins cadherins, molecules or drugs that promote angiogenesis, molecules or drugs that promote vasculogenesis and molecules or drugs that promote aerteriogenesis. 13. The method of claim 12 wherein the compound is VEGF-A, PIGF, VEGF-B, VEGF-C, VEGF-D, TGF-β, Ang-1, Ang-2, IGF, HGF, FGF, Tie2, PDGF, CCL2, Alpha-V Beta-5, Alpha-5 Beta-1, VE-cadherin, PECAM-1, plasminogen activator, or nitrogen oxide synthase. 14. The method of claim 1 further comprising administering one or more growth factors before, during or after implanting selected stem cells in the posterior disc annulus. 15. The method of claim 14 wherein the one or more growth factor is selected from the group consisting of: TGF-β, FGF, PDGF, and IGF. 16. The method of claim 1 wherein the culturing of harvested stem cells is performed in a basal culture media made from a D-MEM base. 17. A pharmaceutical composition comprising stem cells, platelets, one or more growth factors, and a pharmaceutical carrier or diluent. 18. The composition of claim 17 wherein the stem cells are mesenchymal stem cells. 19. The composition of claim 17 wherein the stem cells have been selected for their capacity for viability and expansion under hypoxic conditions. 20. The compositions of claim 17 wherein the stem cells have been selected for their capacity for viability and expansion under culture conditions comparable to a damaged intervertebral disc. 21. A method for treating an avascular zone in a patient in need thereof comprising: harvesting stem cells from the patient in need thereof; culturing the harvested stem cells under selective pressure that corresponds to the environment of a damaged or aged avascular zone; and implanting the selected stem cells in the avascular zone. 22. The method of claim 21 wherein the avascular zone is in a patient's shoulder. 23. The method of claim 21 wherein the avascular zone is in a patient's hip.
1,600
326
14,238,439
1,635
Disclosed are oligomeric compounds which are useful for hybridizing to a complementary nucleic acid, including but not limited, to nucleic acids in a cell. The hybridization results in modulation of the amount activity or expression of the target nucleic acid in a cell.
1.-272. (canceled) 273. A oligomeric compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides, wherein the modified oligonucleotide has a modification motif comprising: a 5′-region consisting of 2-8 linked 5′-region nucleosides, each independently selected from among a modified nucleoside and an unmodified deoxynucleoside, provided that at least one 5′-region nucleoside is a modified nucleoside and wherein the 3′-most 5′-region nucleoside is a modified nucleoside; a 3′-region consisting of 2-8 linked 3′-region nucleosides, each independently selected from among a modified nucleoside and an unmodified deoxynucleoside, provided that at least one 3′-region nucleoside is a modified nucleoside and wherein the 5′-most 3′-region nucleoside is a modified nucleoside; and a central region between the 5′-region and the 3′-region consisting of 6-12 linked central region nucleosides, each independently selected from among: a modified nucleoside and an unmodified deoxynucleoside, wherein the 5′-most central region nucleoside is an unmodified deoxynucleoside and the 3′-most central region nucleoside is an unmodified deoxynucleoside; wherein the modified oligonucleotide has a nucleobase sequence complementary to the nucleobase sequence of a target region of a target nucleic acid. 274. The oligomeric compound of claim 273, wherein the 5′-region has a motif selected from among: AB, ABB, AAA, BBB, BBBAA, AAB, BAA, BBAA, AABB, AAAB, ABBW, ABBWW, ABBB, ABBBB, ABAB, ABABAB, ABABBB, ABABAA, AAABB, AAAABB, AABB, AAAAB, AABBB, ABBBB, BBBBB, AAABW, AAAAA, and BBBBAA; wherein the 3′-region has a motif selected from among: BBA, AAB, AAA, BBB, BBAA, AABB, WBBA, WAAB, BBBA, BBBBA, BBBB, BBBBBA, ABBBBB, BBAAA, AABBB, BBBAA, BBBBA, BBBBB, BABA, AAAAA, BBAAAA, AABBBB, BAAAA, and ABBBB; wherein the central region has a nucleoside motif selected from among: DDDDDD, DDDDDDD, DDDDDDDD, DDDDDDDDD, DDDDDDDDDD, DDDDDDDDD, DXDDDDDDD, DDXDDDDDD, DDDXDDDDD, DDDDXDDDD, DDDDDXDDD, DDDDDDXDD, DDDDDDDXD, DXXDDDDDD, DDDDDDXXD, DDXXDDDDD, DDDXXDDDD, DDDDXXDDD, DDDDDXXDD, DXDDDDDXD, DXDDDDXDD, DXDDDXDDD, DXDDXDDDD, DXDXDDDDD, DDXDDDDXD, DDXDDDXDD, DDXDDXDDD, DDXDXDDDD, DDDXDDDXD, DDDXDDXDD, DDDXDXDDD, DDDDXDDXD, DDDDXDXDD, and DDDDDXDXD, DDDDDDDD, DXDDDDDD, DDXDDDDD, DDDXDDDD, DDDDXDDD, DDDDDXDD, DDDDDDXD, DXDDDDXD, DXDDDXDD, DXDDXDDD, DXDXDDDD, DXXDDDDD, DDXXDDDD, DDXDXDDD, DDXDDXDD, DXDDDDXD, DDDXXDDD, DDDXDXDD, DDDXDDXD, DDDDXXDD, DDDDXDXD, and DDDDDXXD, DXDDDDD, DDXDDDD, DDDXDDD, DDDDXDD, DDDDDXD, DXDDDXD, DXDDXDD, DXDXDDD, DXXDDDD, DDXXDDD, DDXDXDD, DDXDDXD, DDDXXDD, DDDXDXD, and DDDDXXD, DXDDDD, DDXDDD, DDDXDD, DDDDXD, DXXDDD, DXDXDD, DXDDXD, DDXXDD, DDXDXD, and DDDXXD; and wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type, each W is a modified nucleoside of a first type, a second type, or a third type, each D is an unmodified deoxynucleoside, and each X is a modified nucleoside or a modified nucleobase. 275. The oligomeric compound of claim 274, wherein the 5′-region has a motif selected from among: AB, ABB, AAA, BBB, BBBAA, AAB, BAA, BBAA, AABB, ABBW, ABBWW, ABBB, ABBBB, ABAB, ABABAB, ABABBB, ABABAA, AAABB, AAAABB, AABB, AAAAB, AABBB, ABBBB, BBBBB, AAABW, and BBBBAA; and wherein the 3′-region has a BBA motif. 276. The oligomeric compound of claim 274, wherein each A nucleoside comprises a bicyclic sugar moiety selected from among: cEt, cMOE, LNA, α-LNA, ENA and 2′-thio LNA. 277. The oligomeric compound of claim 274, wherein each A nucleoside comprises a cEt. 278. The oligomeric compound of claim 276, wherein each B nucleoside comprises a 2′-substituted sugar moiety comprising a 2′-substituent selected from among: a halogen, OCH3, OCF3, OCH2CH3, OCH2CF3, OCH2—CH═CH2, O(CH2)2—OCH3, O(CH2)2—O(CH2)2—N(CH3)2, OCH2C(═O)—N(H)CH3, OCH2C(═O)—N(H)—(CH2)2—N(CH3)2, and OCH2—N(H)—C(═NH)NH2. 279. The oligomeric compound of claim 278, wherein each B nucleoside comprises a 2′-substituted sugar moiety comprising a 2′-substituent selected from among: F, OCH3, O(CH2)2—OCH3. 280. The oligomeric compound of claim 274, wherein one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2′-MOE sugar moiety, and W comprises a 2-thio-thymidine nucleobase. 281. The oligomeric compound of claim 274, wherein one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2′-MOE sugar moiety, and W comprises FHNA. 282. The oligomeric compound of claim 274, wherein one of A or B comprises cEt, another of A or B comprises a 2′-modified sugar moiety, and W comprises a 2-thio-thymidine nucleobase. 283. The oligomeric compound of claim 274, wherein one of A or B comprises cEt, another of A or B comprises a 2′-modified sugar moiety, and W comprises FHNA. 284. The oligomeric compound of claim 274, wherein each A comprises MOE, each B comprises cEt, and each W is selected from among cEt or FHNA. 285. The oligomeric compound of claim 284, wherein each W comprises cEt. 286. The oligomeric compound of claim 284, wherein each W comprises 2-thio-thymidine. 287. The oligomeric compound of claim 284, wherein each W comprises FHNA. 288. The oligomeric compound of claim 274 comprising at least one modified internucleoside linkage. 289. The oligomeric compound of claim 288, wherein each internucleoside linkage is a modified internucleoside linkage. 290. The oligomeric compound of claim 289 comprising at least one phosphorothioate internucleoside linkage. 291. The oligomeric compound of claim 288 comprising at least one methylphosphonate internucleoside linkage. 292. The oligomeric compound of claim 275, wherein each A nucleoside comprises a bicyclic sugar moiety selected from among: cEt and LNA and each B nucleoside comprises a 2′-MOE.
Disclosed are oligomeric compounds which are useful for hybridizing to a complementary nucleic acid, including but not limited, to nucleic acids in a cell. The hybridization results in modulation of the amount activity or expression of the target nucleic acid in a cell.1.-272. (canceled) 273. A oligomeric compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides, wherein the modified oligonucleotide has a modification motif comprising: a 5′-region consisting of 2-8 linked 5′-region nucleosides, each independently selected from among a modified nucleoside and an unmodified deoxynucleoside, provided that at least one 5′-region nucleoside is a modified nucleoside and wherein the 3′-most 5′-region nucleoside is a modified nucleoside; a 3′-region consisting of 2-8 linked 3′-region nucleosides, each independently selected from among a modified nucleoside and an unmodified deoxynucleoside, provided that at least one 3′-region nucleoside is a modified nucleoside and wherein the 5′-most 3′-region nucleoside is a modified nucleoside; and a central region between the 5′-region and the 3′-region consisting of 6-12 linked central region nucleosides, each independently selected from among: a modified nucleoside and an unmodified deoxynucleoside, wherein the 5′-most central region nucleoside is an unmodified deoxynucleoside and the 3′-most central region nucleoside is an unmodified deoxynucleoside; wherein the modified oligonucleotide has a nucleobase sequence complementary to the nucleobase sequence of a target region of a target nucleic acid. 274. The oligomeric compound of claim 273, wherein the 5′-region has a motif selected from among: AB, ABB, AAA, BBB, BBBAA, AAB, BAA, BBAA, AABB, AAAB, ABBW, ABBWW, ABBB, ABBBB, ABAB, ABABAB, ABABBB, ABABAA, AAABB, AAAABB, AABB, AAAAB, AABBB, ABBBB, BBBBB, AAABW, AAAAA, and BBBBAA; wherein the 3′-region has a motif selected from among: BBA, AAB, AAA, BBB, BBAA, AABB, WBBA, WAAB, BBBA, BBBBA, BBBB, BBBBBA, ABBBBB, BBAAA, AABBB, BBBAA, BBBBA, BBBBB, BABA, AAAAA, BBAAAA, AABBBB, BAAAA, and ABBBB; wherein the central region has a nucleoside motif selected from among: DDDDDD, DDDDDDD, DDDDDDDD, DDDDDDDDD, DDDDDDDDDD, DDDDDDDDD, DXDDDDDDD, DDXDDDDDD, DDDXDDDDD, DDDDXDDDD, DDDDDXDDD, DDDDDDXDD, DDDDDDDXD, DXXDDDDDD, DDDDDDXXD, DDXXDDDDD, DDDXXDDDD, DDDDXXDDD, DDDDDXXDD, DXDDDDDXD, DXDDDDXDD, DXDDDXDDD, DXDDXDDDD, DXDXDDDDD, DDXDDDDXD, DDXDDDXDD, DDXDDXDDD, DDXDXDDDD, DDDXDDDXD, DDDXDDXDD, DDDXDXDDD, DDDDXDDXD, DDDDXDXDD, and DDDDDXDXD, DDDDDDDD, DXDDDDDD, DDXDDDDD, DDDXDDDD, DDDDXDDD, DDDDDXDD, DDDDDDXD, DXDDDDXD, DXDDDXDD, DXDDXDDD, DXDXDDDD, DXXDDDDD, DDXXDDDD, DDXDXDDD, DDXDDXDD, DXDDDDXD, DDDXXDDD, DDDXDXDD, DDDXDDXD, DDDDXXDD, DDDDXDXD, and DDDDDXXD, DXDDDDD, DDXDDDD, DDDXDDD, DDDDXDD, DDDDDXD, DXDDDXD, DXDDXDD, DXDXDDD, DXXDDDD, DDXXDDD, DDXDXDD, DDXDDXD, DDDXXDD, DDDXDXD, and DDDDXXD, DXDDDD, DDXDDD, DDDXDD, DDDDXD, DXXDDD, DXDXDD, DXDDXD, DDXXDD, DDXDXD, and DDDXXD; and wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type, each W is a modified nucleoside of a first type, a second type, or a third type, each D is an unmodified deoxynucleoside, and each X is a modified nucleoside or a modified nucleobase. 275. The oligomeric compound of claim 274, wherein the 5′-region has a motif selected from among: AB, ABB, AAA, BBB, BBBAA, AAB, BAA, BBAA, AABB, ABBW, ABBWW, ABBB, ABBBB, ABAB, ABABAB, ABABBB, ABABAA, AAABB, AAAABB, AABB, AAAAB, AABBB, ABBBB, BBBBB, AAABW, and BBBBAA; and wherein the 3′-region has a BBA motif. 276. The oligomeric compound of claim 274, wherein each A nucleoside comprises a bicyclic sugar moiety selected from among: cEt, cMOE, LNA, α-LNA, ENA and 2′-thio LNA. 277. The oligomeric compound of claim 274, wherein each A nucleoside comprises a cEt. 278. The oligomeric compound of claim 276, wherein each B nucleoside comprises a 2′-substituted sugar moiety comprising a 2′-substituent selected from among: a halogen, OCH3, OCF3, OCH2CH3, OCH2CF3, OCH2—CH═CH2, O(CH2)2—OCH3, O(CH2)2—O(CH2)2—N(CH3)2, OCH2C(═O)—N(H)CH3, OCH2C(═O)—N(H)—(CH2)2—N(CH3)2, and OCH2—N(H)—C(═NH)NH2. 279. The oligomeric compound of claim 278, wherein each B nucleoside comprises a 2′-substituted sugar moiety comprising a 2′-substituent selected from among: F, OCH3, O(CH2)2—OCH3. 280. The oligomeric compound of claim 274, wherein one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2′-MOE sugar moiety, and W comprises a 2-thio-thymidine nucleobase. 281. The oligomeric compound of claim 274, wherein one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2′-MOE sugar moiety, and W comprises FHNA. 282. The oligomeric compound of claim 274, wherein one of A or B comprises cEt, another of A or B comprises a 2′-modified sugar moiety, and W comprises a 2-thio-thymidine nucleobase. 283. The oligomeric compound of claim 274, wherein one of A or B comprises cEt, another of A or B comprises a 2′-modified sugar moiety, and W comprises FHNA. 284. The oligomeric compound of claim 274, wherein each A comprises MOE, each B comprises cEt, and each W is selected from among cEt or FHNA. 285. The oligomeric compound of claim 284, wherein each W comprises cEt. 286. The oligomeric compound of claim 284, wherein each W comprises 2-thio-thymidine. 287. The oligomeric compound of claim 284, wherein each W comprises FHNA. 288. The oligomeric compound of claim 274 comprising at least one modified internucleoside linkage. 289. The oligomeric compound of claim 288, wherein each internucleoside linkage is a modified internucleoside linkage. 290. The oligomeric compound of claim 289 comprising at least one phosphorothioate internucleoside linkage. 291. The oligomeric compound of claim 288 comprising at least one methylphosphonate internucleoside linkage. 292. The oligomeric compound of claim 275, wherein each A nucleoside comprises a bicyclic sugar moiety selected from among: cEt and LNA and each B nucleoside comprises a 2′-MOE.
1,600
327
15,418,017
1,617
The invention relates to a composition, especially a cosmetic composition, including water, at least one film-forming agent, at least one volumizing agent, and an emulsification system comprising (i) about 2% to about 6% of low HLB emulsifier(s) (HLB value less than 8), (ii) about 2% to about 8% of intermediate HLB emulsifier(s) (HLB value of 8 to 16), and (iii) about 1% to about 8% of high HLB emulsifier(s) (HLB value greater than 16), with the compositions further including at least one colorant and/or excluding wax, as well as to methods of using such compositions.
1. A composition comprising water, at least one dispersion of film forming particles in aqueous phase, at least one volumizing agent, and an emulsification system comprising about 2% to about 6% of low HLB emulsifier(s), about 2% to about 8% of intermediate HLB emulsifier(s), and about 1% to about 8% of high HLB emulsifier(s) by weight with respect to the total weight of the composition. 2. The composition of claim 1, wherein the composition is a mascara. 3. The composition of claim 1, wherein the composition is wax-free. 4. The composition of claim 1, further comprising at least one colorant. 5. The composition of claim 1, wherein the at least one volumizing agent is a filler. 6. The composition of claim 5, wherein the at least one volumizing agent is polytetrafluoroethylene. 7. The composition of claim 1, wherein the at least one volumizing agent is shea butter. 8. The composition of claim 7, wherein the shea butter is present in the composition in an amount of about 2% to about 10% by weight with respect to the total weight of the composition. 9. The composition of claim 1, wherein water is present in the composition in an amount of about 30% to about 60% by weight with respect to the total weight of the composition. 10. The composition of claim 1, in the form of an oil-in-water emulsion. 11. The composition of claim 9, in the form of an oil-in-water emulsion 12. The composition of claim 1, wherein the low HLB emulsifier(s) and the intermediate HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:4. 13. The composition of claim 1, wherein the low HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:3. 14. The composition of claim 1, wherein the intermediate HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 20:1 to 1:3. 15. The composition of claim 1, wherein: the low HLB emulsifier(s) and the intermediate HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:4; the low HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:3; and the intermediate HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 5:1 to 1:4. 16. The composition of claim 15, wherein the composition is wax-free. 17. The composition of claim 15, wherein the composition is a mascara. 18. The composition of claim 1, further comprising at least one wax in an amount ranging from about 0.1% to about 5% by weight based on the total weight of the composition. 19. The composition of claim 15, further comprising at least one wax in an amount ranging from about 0.1% to about 5% by weight based on the total weight of the composition. 20. A method of making up eyelashes comprising applying the composition of claim 1 to the eyelashes.
The invention relates to a composition, especially a cosmetic composition, including water, at least one film-forming agent, at least one volumizing agent, and an emulsification system comprising (i) about 2% to about 6% of low HLB emulsifier(s) (HLB value less than 8), (ii) about 2% to about 8% of intermediate HLB emulsifier(s) (HLB value of 8 to 16), and (iii) about 1% to about 8% of high HLB emulsifier(s) (HLB value greater than 16), with the compositions further including at least one colorant and/or excluding wax, as well as to methods of using such compositions.1. A composition comprising water, at least one dispersion of film forming particles in aqueous phase, at least one volumizing agent, and an emulsification system comprising about 2% to about 6% of low HLB emulsifier(s), about 2% to about 8% of intermediate HLB emulsifier(s), and about 1% to about 8% of high HLB emulsifier(s) by weight with respect to the total weight of the composition. 2. The composition of claim 1, wherein the composition is a mascara. 3. The composition of claim 1, wherein the composition is wax-free. 4. The composition of claim 1, further comprising at least one colorant. 5. The composition of claim 1, wherein the at least one volumizing agent is a filler. 6. The composition of claim 5, wherein the at least one volumizing agent is polytetrafluoroethylene. 7. The composition of claim 1, wherein the at least one volumizing agent is shea butter. 8. The composition of claim 7, wherein the shea butter is present in the composition in an amount of about 2% to about 10% by weight with respect to the total weight of the composition. 9. The composition of claim 1, wherein water is present in the composition in an amount of about 30% to about 60% by weight with respect to the total weight of the composition. 10. The composition of claim 1, in the form of an oil-in-water emulsion. 11. The composition of claim 9, in the form of an oil-in-water emulsion 12. The composition of claim 1, wherein the low HLB emulsifier(s) and the intermediate HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:4. 13. The composition of claim 1, wherein the low HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:3. 14. The composition of claim 1, wherein the intermediate HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 20:1 to 1:3. 15. The composition of claim 1, wherein: the low HLB emulsifier(s) and the intermediate HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:4; the low HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 3:1 to 1:3; and the intermediate HLB emulsifier(s) and the high HLB emulsifier(s) are present in the compositions of the present invention in a weight ratio of from 5:1 to 1:4. 16. The composition of claim 15, wherein the composition is wax-free. 17. The composition of claim 15, wherein the composition is a mascara. 18. The composition of claim 1, further comprising at least one wax in an amount ranging from about 0.1% to about 5% by weight based on the total weight of the composition. 19. The composition of claim 15, further comprising at least one wax in an amount ranging from about 0.1% to about 5% by weight based on the total weight of the composition. 20. A method of making up eyelashes comprising applying the composition of claim 1 to the eyelashes.
1,600
328
14,806,487
1,658
The present invention provides a modified atrial natriuretic peptide that exhibits prolonged duration in blood and maintains cGMP elevating activity. The present invention provides a modified peptide in which at least one sugar substance is linked directly through a glycosidic bond or via a linker structure to at least one hANP peptide, or a pharmaceutically acceptable salt thereof, a medicament comprising the modified peptide or the salt thereof as an active ingredient, etc.
1. A modified peptide in which at least one sugar substance is linked directly through a glycosidic bond or via a linker structure to at least one hANP peptide, or a pharmaceutically acceptable salt thereof. 2. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked directly through a glycosidic bond or via a linker structure to at least one of the N terminus of the hANP peptide, the C terminus of the hANP peptide, and the side chain of at least one amino acid constituting the peptide. 3. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the hANP peptide is hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 4. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is selected from at least one type of monosaccharide, disaccharide, trisaccharide, and glycochain of 4 or more monosaccharides bonded through glycosidic bonds, and when a plurality of sugar substances are contained in one molecule, the sugar substances may be the same as or different from each other. 5. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is a glycochain of 4 or more monosaccharides bonded through glycosidic bonds. 6. The modified peptide according to claim 5 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is a glycoprotein-derived N-linked glycochain or O-linked glycochain, or an altered glycochain thereof. wherein Gxx is GlcNAc, Glc, or Man (hereinafter, glycochains having the above structure are referred to as “AG(9)”, “AG(9-Glc)”, and “AG(9-Man)”, respectively, according to the type of GXX), and “O/N-L” represents binding to the linker structure or the hANP peptide through an O-glycosidic bond or a N-glycosidic bond. 10. The modified peptide according to claim 9 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is a glycochain comprising a glycochain structure represented by the following formula: wherein Gxx is GlcNAc, Glc, or Man (hereinafter, glycochains having the above structure are referred to as “SG”, “SG(Glc)”, and “SG(Man)”, respectively, according to the type of GXX), and “O/N-L” represents binding to the linker structure or the hANP peptide through an O-glycosidic bond or a N-glycosidic bond. 11. The modified peptide according to claim 7 or a pharmaceutically acceptable salt thereof, wherein in the sugar substance, Gxx is GlcNAc. 12. The modified peptide according to claim 11 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is SG. 13. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein 10 or less sugar substances are linked to one hANP peptide. 14. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein 1, 2, or 3 sugar substances are linked to one hANP peptide, 15. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein one molecule contains a divalent or higher hANP peptide. 16. The modified peptide according to claim 3 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked via a linker structure to the hANP peptide, and the linker structure is a chemical structure that has a linking chain of 3 or more atoms and is bonded at at least one site to the reducing end of the sugar substance through a glycosidic bond and bonded at at least one site to the hANP peptide. 17. The modified peptide according to claim 16 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked to either the N terminus or the C terminus, or both, of the hANP peptide via a linker structure. 18. The modified peptide according to claim 17 or a pharmaceutically acceptable salt thereof, wherein the linker structure is a structure having a linking chain of 15 or less atoms. 19. The modified peptide according to claim 18 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is SG-hANP(1-28) (compound 2-1), hANP(1-28)-SG (compound 2-2), SG-hANP(1-28)-SG (compound 2-7), AG(9)-hANP(1-28) (compound 2-10), SG-triazole-hANP(1-28) (compound 2-12), SG-thioacetamide-hANP(1-28) (compound 2-25), or AG(5)-hANP(1-28) (compound 2-26), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 20. The modified peptide according to claim 16 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one structure selected from a polyoxyalkylene chain, an amino acid, and an oligopeptide chain consisting of 2 or more amino acids. 21. The modified peptide according to claim 20 or a pharmaceutically acceptable salt thereof, wherein the polyoxyalkylene chain, the amino acid, and/or the oligopeptide chain contained in the linker structure is bonded through an amide bond to the N terminus and/or the C terminus of the hANP peptide. 22. The modified peptide according to claim 21 or a pharmaceutically acceptable salt thereof, wherein the polyoxyalkylene chain is PEG. 23. The modified peptide according to claim 21 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is SG-PEG(3)-(SG-)Asn-hANP(1-28) (compound 2-16), AG(9)-(AG(9)-)Asn-PEG(3)-hANP(1-28) (compound 2-21), AG(7)-(AG(7)-)Asn-PEG(3)-hANP(1-28) (compound 2-22), SG-PEG(3)-hANP(1-28)-PEG(3)-SG (compound 2-24), SG-(SG-) Asn-PEG(11)-hANP(1-28) (compound 2-27), SG-(SG-)Asn-PEG(11)-PEG(11)-hANP(1-28) (compound 2-28), SG-PEG(3)-hANP(1-28) (compound 2-29), SG-PEG(11)-hANP(1-28) (compound 2-30), SG-*(SG-)Gln-Mal-PEG(3)-hANP(1-28) (compound 2-31), SG-(SG-)Gln-PEG(3)-Mal-hANP(1-28) (compound 2-32), SG-(SG-)Asn-(Ser-Gly)3-hANP(1-28) (compound 2-36), or SG-(SG-)Asn-Gly6-hANP(1-28) (compound 2-37), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 24. The modified peptide according to claim 20 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a functional group on the side chain selected from an amino acid having an amino group on the side chain, an amino acid having SH on the side chain, an amino acid having a carboxyl group on the side chain, an amino acid having a hydroxy group on the side chain, and an amino acid having phenol on the side chain and is linked at the side chain of the amino acid having a functional group on the side chain to the sugar substance or the hANP peptide. 25. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having an amino group on the side chain and has a structure of the following general formula (C) in which the sugar substance is linked to the side chain of the amino acid having an amino group on the side chain: wherein GLY represents the sugar substance; Le, represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and N-(AA) represents a nitrogen atom derived from the side chain amino group of the amino acid having an amino group on the side chain. 26. The modified peptide according to claim 25 or a pharmaceutically acceptable salt thereof, wherein the side chain amino group and the α amino group of the amino acid having an amino group on the side chain form amide bonds with the α carboxyl groups of other amino acids. 27. The modified peptide according to claim 25 or a pharmaceutically acceptable salt thereof, wherein the amino acid having an amino group on the side chain is Lys. 28. The modified peptide according to claim 27 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is SG-(SG-)Lys-Gly-hANP(1-28) (compound 2-14), [(SG-)Cys-Gly]3-hANP(1-28) (compound 2-15), SG-Mal-(SG-Mal-)Lys-[SG-Mal-(SG-Mal)Lys-]Lys-PEG(3)-hANP(1-28) (compound 2-19), [SG2-Mal-(SG2-Mal-)Lys-[SG2-Mal-(SG2-Mal-)-Lys-]Lys-PEG(3)-hANP(1-28) (compound 2-20), SG-Mal-(SG-Mal-)Lys-hANP(1-28) (compound 2-33), SG-thioacetamide-(SG-thioacetamide-)Lys-PEG-(3)-hANP(1-28) (compound 2-34), or SG-(SG-)Lys-PEG(3)-hANP(1-28) (compound 2-35), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 29. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a SH group on the side chain and has a structure of the following general formula in which the sugar substance is linked to the side chain of the amino acid having an SH group on the side chain: wherein GLY represents the sugar substance; Lg represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and S represents a sulfur atom derived from the side chain SH group of the amino acid having a SH group on the side chain. 30. The modified peptide according to claim 29 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a SH group on the side chain is Cys. 31. The modified peptide according to claim 30 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is [(SG-)Cys-Gly]5-hANP(1-28) (compound 2-17), [(SG2-)Cys-Gly]5-hANP(1-28) (compound 2-18), or SG-Mal-(SG-Mal-)Lys-[SG-Mal-(SG-Mal-)Lys-]Lys-PEG(11)-hANP(1-28) (compound 2-23), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 32. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a carboxyl group on the side chain and has a structure of the following general formula in which the sugar substance is linked to the side chain of the amino acid having carboxylic acid on the side chain: wherein GLY represents the sugar substance; Lg represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and CO represents CO derived from the side chain of the amino acid having carboxylic acid on the side chain. 33. The modified peptide according to claim 32 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is bonded through a N-glycosidic bond to both of the side chain carboxyl group and the α carboxyl group of the amino acid having a carboxyl group on the side chain and bonded to another linker structure or the hANP peptide via the α amino group of said amino acid. 34. The modified peptide according to claim 32 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a carboxylic acid group on the side chain is Glu, Gln, Asp, or Asn. 35. The modified peptide according to claim 34 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is (SG-)Asn-hANP(1-28) (compound 2-3), (SG-)Asn-hANP(2-28) (compound 2-4), (SG-)Asn-hANP(3-28) (compound 2-8), SG-(SG-)Asn-hANP(1-28) (compound 2-9), or SG-(SG-)Asn-PEG(3)-hANP(1-28) (compound 2-13), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 36. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having phenol on the side chain and has a structure of the following general formula in which the sugar substance is linked to the side chain of the amino acid having phenol on the side chain: wherein GLY represents the sugar substance; Lg represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and the phenol group represents a phenol group derived from the side chain of the amino acid having a phenol group on the side chain. 37. The modified peptide according to claim 36 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a phenol group on the side chain is Tyr. 38. The modified peptide according to claim 37 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is hANP(1-27)-(SG-)Tyr (compound 2-6), or is derived from the modified peptide by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 39. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a hydroxy group on the side chain and has a structure of the following general formula in which the sugar substance is bonded through an O-glycosidic bond to the side chain of the amino acid having a hydroxy group on the side chain: wherein GLY represents the sugar substance; and O represents an oxygen atom derived from the side chain hydroxy group of the amino acid having a hydroxy group on the side chain. 40. The modified peptide according to claim 39 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a hydroxy group on the side chain is Ser. 41. The modified peptide according to claim 40 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is (SG-)Ser-hANP(2-28) (compound 2-5), or is derived from the modified peptide by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 42. The modified peptide claim 16 or a pharmaceutically acceptable salt thereof, wherein the modified peptide has one or two SG molecules as the sugar substance and one hANP(1-28) (SEQ ID NO: 1) as the hANP peptide, and the SG is linked to the N terminus of the hANP(1-28) via a linker structure having a linking chain of 10 or less atoms. 43. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide has a structure represented by the formula of the following compound 2-1, 2-3, 2-10, 2-11, 2-12, 2-13, 2-14, 2-15, 2-16, 2-25, 2-26, 2-27, 2-29, or 2-30: wherein hANP is hANP(1-28) consisting of the amino acid sequence of SEQ ID NO: 1 and is bonded at the N terminus of the amino acid sequence to the linker structure through an amide bond. 44. The salt of the modified peptide according to claim 42, wherein the pharmaceutically acceptable salt is trifluoroacetate or an acetate. 45. The modified peptide according to claim 3 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked to the side chain of an amino acid in the hANP peptide, and the linked amino acid is an amino acid other than amino acids at amino acid positions 7 to 23 of SEQ ID NO: 1 contained in the hANP peptide. 46. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide or the pharmaceutically acceptable salt thereof exhibits a prolonged duration in blood compared with unmodified hANP(1-28) and maintains cGMP elevating activity. 47. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide or the pharmaceutically acceptable salt thereof has resistance to the degradation of the hANP peptide by neutral endopeptidase. 48. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide or the pharmaceutically acceptable salt thereof exhibits 3 or more times the water solubility of unmodified hANP(1-28). 49. A medicament comprising a modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof. 50. The medicament according to claim 48, wherein the medicament is an agent for treating or alleviating a cardiovascular disease. 51. A method for treating or alleviating a cardiovascular disease, comprising administering an effective amount of a modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof. 52. A method for producing a modified peptide according to claim 1, comprising the step of linking a hANP peptide, a sugar substance, and, if necessary, a linker molecule and an acceptor compound. 53. The method according to claim 52, further comprising the step of transferring a glycochain to a GlcNAc compound, a Glc compound, or a Man compound by use of Endo-M or a mutant enzyme thereof.
The present invention provides a modified atrial natriuretic peptide that exhibits prolonged duration in blood and maintains cGMP elevating activity. The present invention provides a modified peptide in which at least one sugar substance is linked directly through a glycosidic bond or via a linker structure to at least one hANP peptide, or a pharmaceutically acceptable salt thereof, a medicament comprising the modified peptide or the salt thereof as an active ingredient, etc.1. A modified peptide in which at least one sugar substance is linked directly through a glycosidic bond or via a linker structure to at least one hANP peptide, or a pharmaceutically acceptable salt thereof. 2. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked directly through a glycosidic bond or via a linker structure to at least one of the N terminus of the hANP peptide, the C terminus of the hANP peptide, and the side chain of at least one amino acid constituting the peptide. 3. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the hANP peptide is hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 4. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is selected from at least one type of monosaccharide, disaccharide, trisaccharide, and glycochain of 4 or more monosaccharides bonded through glycosidic bonds, and when a plurality of sugar substances are contained in one molecule, the sugar substances may be the same as or different from each other. 5. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is a glycochain of 4 or more monosaccharides bonded through glycosidic bonds. 6. The modified peptide according to claim 5 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is a glycoprotein-derived N-linked glycochain or O-linked glycochain, or an altered glycochain thereof. wherein Gxx is GlcNAc, Glc, or Man (hereinafter, glycochains having the above structure are referred to as “AG(9)”, “AG(9-Glc)”, and “AG(9-Man)”, respectively, according to the type of GXX), and “O/N-L” represents binding to the linker structure or the hANP peptide through an O-glycosidic bond or a N-glycosidic bond. 10. The modified peptide according to claim 9 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is a glycochain comprising a glycochain structure represented by the following formula: wherein Gxx is GlcNAc, Glc, or Man (hereinafter, glycochains having the above structure are referred to as “SG”, “SG(Glc)”, and “SG(Man)”, respectively, according to the type of GXX), and “O/N-L” represents binding to the linker structure or the hANP peptide through an O-glycosidic bond or a N-glycosidic bond. 11. The modified peptide according to claim 7 or a pharmaceutically acceptable salt thereof, wherein in the sugar substance, Gxx is GlcNAc. 12. The modified peptide according to claim 11 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is SG. 13. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein 10 or less sugar substances are linked to one hANP peptide. 14. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein 1, 2, or 3 sugar substances are linked to one hANP peptide, 15. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein one molecule contains a divalent or higher hANP peptide. 16. The modified peptide according to claim 3 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked via a linker structure to the hANP peptide, and the linker structure is a chemical structure that has a linking chain of 3 or more atoms and is bonded at at least one site to the reducing end of the sugar substance through a glycosidic bond and bonded at at least one site to the hANP peptide. 17. The modified peptide according to claim 16 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked to either the N terminus or the C terminus, or both, of the hANP peptide via a linker structure. 18. The modified peptide according to claim 17 or a pharmaceutically acceptable salt thereof, wherein the linker structure is a structure having a linking chain of 15 or less atoms. 19. The modified peptide according to claim 18 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is SG-hANP(1-28) (compound 2-1), hANP(1-28)-SG (compound 2-2), SG-hANP(1-28)-SG (compound 2-7), AG(9)-hANP(1-28) (compound 2-10), SG-triazole-hANP(1-28) (compound 2-12), SG-thioacetamide-hANP(1-28) (compound 2-25), or AG(5)-hANP(1-28) (compound 2-26), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 20. The modified peptide according to claim 16 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one structure selected from a polyoxyalkylene chain, an amino acid, and an oligopeptide chain consisting of 2 or more amino acids. 21. The modified peptide according to claim 20 or a pharmaceutically acceptable salt thereof, wherein the polyoxyalkylene chain, the amino acid, and/or the oligopeptide chain contained in the linker structure is bonded through an amide bond to the N terminus and/or the C terminus of the hANP peptide. 22. The modified peptide according to claim 21 or a pharmaceutically acceptable salt thereof, wherein the polyoxyalkylene chain is PEG. 23. The modified peptide according to claim 21 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is SG-PEG(3)-(SG-)Asn-hANP(1-28) (compound 2-16), AG(9)-(AG(9)-)Asn-PEG(3)-hANP(1-28) (compound 2-21), AG(7)-(AG(7)-)Asn-PEG(3)-hANP(1-28) (compound 2-22), SG-PEG(3)-hANP(1-28)-PEG(3)-SG (compound 2-24), SG-(SG-) Asn-PEG(11)-hANP(1-28) (compound 2-27), SG-(SG-)Asn-PEG(11)-PEG(11)-hANP(1-28) (compound 2-28), SG-PEG(3)-hANP(1-28) (compound 2-29), SG-PEG(11)-hANP(1-28) (compound 2-30), SG-*(SG-)Gln-Mal-PEG(3)-hANP(1-28) (compound 2-31), SG-(SG-)Gln-PEG(3)-Mal-hANP(1-28) (compound 2-32), SG-(SG-)Asn-(Ser-Gly)3-hANP(1-28) (compound 2-36), or SG-(SG-)Asn-Gly6-hANP(1-28) (compound 2-37), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 24. The modified peptide according to claim 20 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a functional group on the side chain selected from an amino acid having an amino group on the side chain, an amino acid having SH on the side chain, an amino acid having a carboxyl group on the side chain, an amino acid having a hydroxy group on the side chain, and an amino acid having phenol on the side chain and is linked at the side chain of the amino acid having a functional group on the side chain to the sugar substance or the hANP peptide. 25. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having an amino group on the side chain and has a structure of the following general formula (C) in which the sugar substance is linked to the side chain of the amino acid having an amino group on the side chain: wherein GLY represents the sugar substance; Le, represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and N-(AA) represents a nitrogen atom derived from the side chain amino group of the amino acid having an amino group on the side chain. 26. The modified peptide according to claim 25 or a pharmaceutically acceptable salt thereof, wherein the side chain amino group and the α amino group of the amino acid having an amino group on the side chain form amide bonds with the α carboxyl groups of other amino acids. 27. The modified peptide according to claim 25 or a pharmaceutically acceptable salt thereof, wherein the amino acid having an amino group on the side chain is Lys. 28. The modified peptide according to claim 27 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is SG-(SG-)Lys-Gly-hANP(1-28) (compound 2-14), [(SG-)Cys-Gly]3-hANP(1-28) (compound 2-15), SG-Mal-(SG-Mal-)Lys-[SG-Mal-(SG-Mal)Lys-]Lys-PEG(3)-hANP(1-28) (compound 2-19), [SG2-Mal-(SG2-Mal-)Lys-[SG2-Mal-(SG2-Mal-)-Lys-]Lys-PEG(3)-hANP(1-28) (compound 2-20), SG-Mal-(SG-Mal-)Lys-hANP(1-28) (compound 2-33), SG-thioacetamide-(SG-thioacetamide-)Lys-PEG-(3)-hANP(1-28) (compound 2-34), or SG-(SG-)Lys-PEG(3)-hANP(1-28) (compound 2-35), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 29. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a SH group on the side chain and has a structure of the following general formula in which the sugar substance is linked to the side chain of the amino acid having an SH group on the side chain: wherein GLY represents the sugar substance; Lg represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and S represents a sulfur atom derived from the side chain SH group of the amino acid having a SH group on the side chain. 30. The modified peptide according to claim 29 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a SH group on the side chain is Cys. 31. The modified peptide according to claim 30 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is [(SG-)Cys-Gly]5-hANP(1-28) (compound 2-17), [(SG2-)Cys-Gly]5-hANP(1-28) (compound 2-18), or SG-Mal-(SG-Mal-)Lys-[SG-Mal-(SG-Mal-)Lys-]Lys-PEG(11)-hANP(1-28) (compound 2-23), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 32. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a carboxyl group on the side chain and has a structure of the following general formula in which the sugar substance is linked to the side chain of the amino acid having carboxylic acid on the side chain: wherein GLY represents the sugar substance; Lg represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and CO represents CO derived from the side chain of the amino acid having carboxylic acid on the side chain. 33. The modified peptide according to claim 32 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is bonded through a N-glycosidic bond to both of the side chain carboxyl group and the α carboxyl group of the amino acid having a carboxyl group on the side chain and bonded to another linker structure or the hANP peptide via the α amino group of said amino acid. 34. The modified peptide according to claim 32 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a carboxylic acid group on the side chain is Glu, Gln, Asp, or Asn. 35. The modified peptide according to claim 34 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is (SG-)Asn-hANP(1-28) (compound 2-3), (SG-)Asn-hANP(2-28) (compound 2-4), (SG-)Asn-hANP(3-28) (compound 2-8), SG-(SG-)Asn-hANP(1-28) (compound 2-9), or SG-(SG-)Asn-PEG(3)-hANP(1-28) (compound 2-13), or is derived from any of these modified peptides by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 36. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having phenol on the side chain and has a structure of the following general formula in which the sugar substance is linked to the side chain of the amino acid having phenol on the side chain: wherein GLY represents the sugar substance; Lg represents a structure on the glycochain side in the linker structure and may be linear or have two or more branches; GLY and L are bonded through an O- or N-glycosidic bond; when Lg is branched, the same number of GLY as the number of branch ends is capable of being linked thereto; and the phenol group represents a phenol group derived from the side chain of the amino acid having a phenol group on the side chain. 37. The modified peptide according to claim 36 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a phenol group on the side chain is Tyr. 38. The modified peptide according to claim 37 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is hANP(1-27)-(SG-)Tyr (compound 2-6), or is derived from the modified peptide by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 39. The modified peptide according to claim 24 or a pharmaceutically acceptable salt thereof, wherein the linker structure comprises at least one amino acid having a hydroxy group on the side chain and has a structure of the following general formula in which the sugar substance is bonded through an O-glycosidic bond to the side chain of the amino acid having a hydroxy group on the side chain: wherein GLY represents the sugar substance; and O represents an oxygen atom derived from the side chain hydroxy group of the amino acid having a hydroxy group on the side chain. 40. The modified peptide according to claim 39 or a pharmaceutically acceptable salt thereof, wherein the amino acid having a hydroxy group on the side chain is Ser. 41. The modified peptide according to claim 40 or a pharmaceutically acceptable salt thereof, wherein the modified peptide is (SG-)Ser-hANP(2-28) (compound 2-5), or is derived from the modified peptide by the replacement of the sugar substance with SG, SG(Glc), SG(Man), AG(5), AG(5-Glc), AG(5-Man), AG(7), AG(7-Glc), AG(7-Man), AG(9), AG(9-Glc), AG(9-Man), or GlcNAc and/or the replacement of the hANP peptide with hANP(1-28), hANP(2-28), hANP(3-28), hANP(1-27), hANP(2-27), or hANP(3-27). 42. The modified peptide claim 16 or a pharmaceutically acceptable salt thereof, wherein the modified peptide has one or two SG molecules as the sugar substance and one hANP(1-28) (SEQ ID NO: 1) as the hANP peptide, and the SG is linked to the N terminus of the hANP(1-28) via a linker structure having a linking chain of 10 or less atoms. 43. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide has a structure represented by the formula of the following compound 2-1, 2-3, 2-10, 2-11, 2-12, 2-13, 2-14, 2-15, 2-16, 2-25, 2-26, 2-27, 2-29, or 2-30: wherein hANP is hANP(1-28) consisting of the amino acid sequence of SEQ ID NO: 1 and is bonded at the N terminus of the amino acid sequence to the linker structure through an amide bond. 44. The salt of the modified peptide according to claim 42, wherein the pharmaceutically acceptable salt is trifluoroacetate or an acetate. 45. The modified peptide according to claim 3 or a pharmaceutically acceptable salt thereof, wherein the sugar substance is linked to the side chain of an amino acid in the hANP peptide, and the linked amino acid is an amino acid other than amino acids at amino acid positions 7 to 23 of SEQ ID NO: 1 contained in the hANP peptide. 46. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide or the pharmaceutically acceptable salt thereof exhibits a prolonged duration in blood compared with unmodified hANP(1-28) and maintains cGMP elevating activity. 47. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide or the pharmaceutically acceptable salt thereof has resistance to the degradation of the hANP peptide by neutral endopeptidase. 48. The modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof, wherein the modified peptide or the pharmaceutically acceptable salt thereof exhibits 3 or more times the water solubility of unmodified hANP(1-28). 49. A medicament comprising a modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof. 50. The medicament according to claim 48, wherein the medicament is an agent for treating or alleviating a cardiovascular disease. 51. A method for treating or alleviating a cardiovascular disease, comprising administering an effective amount of a modified peptide according to claim 1 or a pharmaceutically acceptable salt thereof. 52. A method for producing a modified peptide according to claim 1, comprising the step of linking a hANP peptide, a sugar substance, and, if necessary, a linker molecule and an acceptor compound. 53. The method according to claim 52, further comprising the step of transferring a glycochain to a GlcNAc compound, a Glc compound, or a Man compound by use of Endo-M or a mutant enzyme thereof.
1,600
329
15,157,656
1,631
A hemodialysis patient data acquisition and management system resides on a host computer which receives information from one or more non-invasive, optical blood monitors associated with a hemodialysis system. When a patient is undergoing hemodialysis treatment, a sensor assembly monitors the patient's blood flowing through the hemodialysis system and a controller for the blood monitor generates data which includes at least an identification code for the patient undergoing the treatment on the respective system, and non-invasively determined blood data taken at the onset of the scheduled treatment, such as initial Hgb, HCT, and SAT values. A host computer communicates with the one or more optical blood monitors, preferably via a wireless network, and the patient's session commencement data is downloaded to the host computer. The host computer includes a patient database containing historical session commencement data for a plurality of patients, as well as screen displays for displaying historical data for individual patients, such as Hgb trends. The system also preferably provides a predictive algorithm for the patient's Hgb at the patient's next scheduled hemodialysis treatment session. The preferred system also includes software that provides a recommended dose of the anemia management drug for the patient.
1. A system for hemodialysis patient data acquisition and management, comprising: a hemodialysis system, configured to draw blood from a patient, pass the drawn blood through extracorporeal tubing and through a dialyzer, and return the dialyzed blood through extracorporeal tubing to the patient; a non-invasive blood monitor, the blood monitor comprising: a sensor assembly, configured to monitor the patient's blood flowing through the hemodialysis system, and a controller, configured to generate session data for the patient which includes an identification code for the patient and blood measurement data for the patient taken at the onset of the treatment session; and a host computer, configured to receive session data from the non-invasive blood monitor, the host computer comprising: a patient database comprising historical session data for a plurality of patients, and a display screen, configured to display historical session data for a selected patient of the plurality of patients. 2. The system according to claim 1, wherein the non-invasive blood monitor is an optical blood monitor congiured to monitor the patient's blood flowing through the hemodialysis system in real-time. 3. The system according to claim 2, wherein the sensor assembly comprises: a blood chamber having an inlet and an outlet which are connected inline with extracorporeal tubing for the hemodialysis system, and a fluid passageway through which the blood drawn from the patient flows; a first photo emitter, configured to emit light at a first wavelength to pass through the blood chamber and the drawn blood flowing through the blood chamber; a second photo emitter, configured to emit light at a second wavelength to pass through the blood chamber and the drawn blood flowing through the blood chamber; and at least one photo detector, configured to detect the intensity of light at each wavelength after it passes through the blood chamber and the drawn blood; wherein the controller is further configured to determine hematocrit values from the detected intensity of the light at the first and second wavelengths using a ratiometric model. 4. The system according to claim 3, wherein the sensor assembly further comprises: a third photo emitter, configured to emit light at a third wavelength to pass through the blood chamber and the drawn blood flowing through the blood chamber; and a photo detector corresponding to the third photo emitter, configured to detect the intensity of light at the third wavelength after it has passed through the blood chamber and the blood flowing through the blood chamber; wherein the controller is further configured to determine an oxygen saturation value for the patient using a ratiometric model. 5. The system according to claim I, wherein the host computer is configured to communicate with multiple non-invasive blood monitors corresponding to multiple hemodialysis systems. 6. The system according to claim 5, wherein the communications between the multiple non-invasive blood monitors and the host computer are wireless communications. 7. The system according to claim 1, wherein the session data for the patient comprises an initial hematocrit value and an initial oxygen saturation value. 8. The system according to claim 7, wherein the session data for the patient further comprises an initial hemoglobin value. 9. The system according to claim 1, wherein the host computer comprises an input interface for receiving manually-input patient session data. 10. The system according to claim 1, wherein displaying the historical session data for the selected patient comprises displaying a graphical depiction illustrating a minimum desired hemoglobin value, a maximum desired hemoglobin value for the patient, and a historical trend line of initial hemoglobin values from a series of previous hemodialysis treatment sessions for the patient. 11. The system according to claim 1, wherein the non-invasive blood monitor further comprises a display screen configured to display real-time hematocrit values determined by the controller. 12. The system according to claim 1, wherein the host computer is configured to determine the patient's expected initial hemoglobin level for the patient's next scheduled treatment session based on a predictive model. 13. The system according to claim 1, wherein the host computer is configured to output a therapeutic care recommendation based on historical patient session data and predicted future patient session data. 14. The system according to claim 13, wherein the host computer is further configured to determine a dosage recommendation for administration of an anemia management drug for the patient based on historical hemoglobin values for the patient stored in the patient database and based on a predicted hemoglobin value for the patient at a future treatment session. 15. The system according to claim 14, wherein the historical hemoglobin values upon which the dosage recommendation is based includes at least three historical data points of hemoglobin corresponding to the patient for at least three prior hemodialysis treatment sessions.
A hemodialysis patient data acquisition and management system resides on a host computer which receives information from one or more non-invasive, optical blood monitors associated with a hemodialysis system. When a patient is undergoing hemodialysis treatment, a sensor assembly monitors the patient's blood flowing through the hemodialysis system and a controller for the blood monitor generates data which includes at least an identification code for the patient undergoing the treatment on the respective system, and non-invasively determined blood data taken at the onset of the scheduled treatment, such as initial Hgb, HCT, and SAT values. A host computer communicates with the one or more optical blood monitors, preferably via a wireless network, and the patient's session commencement data is downloaded to the host computer. The host computer includes a patient database containing historical session commencement data for a plurality of patients, as well as screen displays for displaying historical data for individual patients, such as Hgb trends. The system also preferably provides a predictive algorithm for the patient's Hgb at the patient's next scheduled hemodialysis treatment session. The preferred system also includes software that provides a recommended dose of the anemia management drug for the patient.1. A system for hemodialysis patient data acquisition and management, comprising: a hemodialysis system, configured to draw blood from a patient, pass the drawn blood through extracorporeal tubing and through a dialyzer, and return the dialyzed blood through extracorporeal tubing to the patient; a non-invasive blood monitor, the blood monitor comprising: a sensor assembly, configured to monitor the patient's blood flowing through the hemodialysis system, and a controller, configured to generate session data for the patient which includes an identification code for the patient and blood measurement data for the patient taken at the onset of the treatment session; and a host computer, configured to receive session data from the non-invasive blood monitor, the host computer comprising: a patient database comprising historical session data for a plurality of patients, and a display screen, configured to display historical session data for a selected patient of the plurality of patients. 2. The system according to claim 1, wherein the non-invasive blood monitor is an optical blood monitor congiured to monitor the patient's blood flowing through the hemodialysis system in real-time. 3. The system according to claim 2, wherein the sensor assembly comprises: a blood chamber having an inlet and an outlet which are connected inline with extracorporeal tubing for the hemodialysis system, and a fluid passageway through which the blood drawn from the patient flows; a first photo emitter, configured to emit light at a first wavelength to pass through the blood chamber and the drawn blood flowing through the blood chamber; a second photo emitter, configured to emit light at a second wavelength to pass through the blood chamber and the drawn blood flowing through the blood chamber; and at least one photo detector, configured to detect the intensity of light at each wavelength after it passes through the blood chamber and the drawn blood; wherein the controller is further configured to determine hematocrit values from the detected intensity of the light at the first and second wavelengths using a ratiometric model. 4. The system according to claim 3, wherein the sensor assembly further comprises: a third photo emitter, configured to emit light at a third wavelength to pass through the blood chamber and the drawn blood flowing through the blood chamber; and a photo detector corresponding to the third photo emitter, configured to detect the intensity of light at the third wavelength after it has passed through the blood chamber and the blood flowing through the blood chamber; wherein the controller is further configured to determine an oxygen saturation value for the patient using a ratiometric model. 5. The system according to claim I, wherein the host computer is configured to communicate with multiple non-invasive blood monitors corresponding to multiple hemodialysis systems. 6. The system according to claim 5, wherein the communications between the multiple non-invasive blood monitors and the host computer are wireless communications. 7. The system according to claim 1, wherein the session data for the patient comprises an initial hematocrit value and an initial oxygen saturation value. 8. The system according to claim 7, wherein the session data for the patient further comprises an initial hemoglobin value. 9. The system according to claim 1, wherein the host computer comprises an input interface for receiving manually-input patient session data. 10. The system according to claim 1, wherein displaying the historical session data for the selected patient comprises displaying a graphical depiction illustrating a minimum desired hemoglobin value, a maximum desired hemoglobin value for the patient, and a historical trend line of initial hemoglobin values from a series of previous hemodialysis treatment sessions for the patient. 11. The system according to claim 1, wherein the non-invasive blood monitor further comprises a display screen configured to display real-time hematocrit values determined by the controller. 12. The system according to claim 1, wherein the host computer is configured to determine the patient's expected initial hemoglobin level for the patient's next scheduled treatment session based on a predictive model. 13. The system according to claim 1, wherein the host computer is configured to output a therapeutic care recommendation based on historical patient session data and predicted future patient session data. 14. The system according to claim 13, wherein the host computer is further configured to determine a dosage recommendation for administration of an anemia management drug for the patient based on historical hemoglobin values for the patient stored in the patient database and based on a predicted hemoglobin value for the patient at a future treatment session. 15. The system according to claim 14, wherein the historical hemoglobin values upon which the dosage recommendation is based includes at least three historical data points of hemoglobin corresponding to the patient for at least three prior hemodialysis treatment sessions.
1,600
330
14,674,536
1,616
A sunscreen composition including a polymer composition that contains a UV-absorbing polyglycerol having a UV-absorbing chromophore chemically bound thereto, and a cosmetically-acceptable topical carrier that contains at least one percent by weight of the sunscreen composition of a non-UV-absorbing polyglycerol, based on total weight of the sunscreen composition.
1. A sunscreen composition comprising: a polymer composition comprising a UV-absorbing polyglycerol comprising a UV-absorbing chromophore chemically bound thereto; and a cosmetically-acceptable topical carrier comprising at least one percent of a non-UV-absorbing polyglycerol, based on the total weight of said sunscreen composition. 2. The sunscreen composition of claim 1, wherein said cosmetically-acceptable topical carrier further comprises a hydrophobic diluent. 3. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol is soluble in said hydrophobic diluent. 4. The sunscreen composition of claim 1, wherein the sum of the weight of said non-UV-absorbing polyglycerol and said UV-absorbing polyglycerol is at least twelve percent, based on the total weight of said sunscreen composition. 5. The sunscreen composition of claim 1, wherein the sum of the weight of said non-UV-radiation absorbing polyglycerol and said UV-absorbing polyglycerol is at least twenty percent, based on the total weight of said sunscreen composition. 6. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol comprises a hydrophobic moiety. 7. The sunscreen composition of claim 1, wherein said non-UV-absorbing polyglycerol comprises a hydrophobic moiety. 8. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol comprises a hydrophobic moiety and said non-UV-absorbing polyglycerol comprises a hydrophobic moiety. 9. The sunscreen composition of claim 8, wherein said hydrophobic moiety has an average number of carbon atoms that is from 8 to 30. 10. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol comprises a first hydrophobic moiety having a first average number of carbon atoms and said non-UV-absorbing polyglycerol comprises a second hydrophobic moiety having a second average number of carbon atoms, wherein said first average number of carbon atoms and said second average number of carbon atoms are within 10 carbon atoms of one another. 11. The sunscreen composition of claim 10, wherein said first average number of carbon atoms and said second average number of carbon atoms are within 2 carbon atoms of one another. 12. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol has a first average degree of glycerol polymerization and said non-UV-absorbing polyglycerol has a second average degree of glycerol polymerization, and wherein said first average degree of glycerol polymerization and said second average degree of glycerol polymerization are each from about 2 to about 20. 13. The sunscreen composition of claim 12, wherein said first average degree of glycerol polymerization and said second average degree of glycerol polymerization are within about 2 units of each other. 14. The sunscreen composition of claim 1, comprising about two percent or more of said non-UV-absorbing polyglycerol and about ten percent or more of said UV-absorbing polyglycerol. 15. The sunscreen composition of claim 1 wherein said UV-absorbing chromophore is selected from the group consisting of a transesterification product of 3-(3-(2H-benzo[d][1,2,3]triazol-2-yl)-5-(tert-butyl)-4-hydroxyphenyl)propanoic acid with polyethylene glycol 300, benzenepropanoic acid, 3-(2H-benzotriazol-2-yl)-5-(1,1-dimethylethyl)-4-hydroxy-, C7-9-branched and linear alkyl esters, 6-octyl-2-(4-(4,6-di([1,1′-biphenyl]-4-yl)-1,3,5-triazin-2-yl)-3-hydroxyphenoxy)propanoate, 3-(3-(2H-benzo[d][1,2,3]triazol-2-yl)-5-(tert-butyl)-4-hydroxyphenyl)propanoic acid, and polyethylene glycol ester of 3-[3-(2H-1,2,3-benzotriazol-2-yl)-5-tert-butyl-4-hydroxyphenyl]propanoate. 16. The sunscreen composition of claim 1 wherein said UV-absorbing chromophore is 3-(3-(2H-benzo[d][1,2,3]triazol-2-yl)-5-(tert-butyl)-4-hydroxyphenyl)propanoic acid. 17. The sunscreen composition of claim 1 wherein said UV-absorbing chromophore is polyethylene glycol ester of 3-[3-(2H-1,2,3-benzotriazol-2-yl)-5-tert-butyl-4-hydroxyphenyl]propanoate 18. The sunscreen composition of claim 1 comprising from about 5 percent to about 50 percent by weight of said UV-absorbing polyglycerol. 19. The sunscreen composition of claim 1 comprising from about 10 percent to about 30 percent by weight of said UV-absorbing polyglycerol. 20. The sunscreen composition of claim 1 comprising about 10 percent or more of said UV-absorbing polyglycerol. 21. The sunscreen composition of claim 1 comprising about 25 percent or more of said UV-absorbing polyglycerol. 22. The sunscreen composition of claim 1 wherein said sunscreen composition is essentially free of non-polymeric UV-absorbing sunscreen agents. 23. The sunscreen composition of claim 1 wherein said sunscreen composition is essentially free of a sunscreen agent other than said UV-absorbing polyglycerol. 24. The sunscreen composition of claim 1 comprising a sunscreen agent selected from the group consisting of a UV-absorbing polymer other than said UV-absorbing polyglycerol, a non-polymeric UV-absorbing sunscreen agent and non-UV-absorbing, light-scattering particles.
A sunscreen composition including a polymer composition that contains a UV-absorbing polyglycerol having a UV-absorbing chromophore chemically bound thereto, and a cosmetically-acceptable topical carrier that contains at least one percent by weight of the sunscreen composition of a non-UV-absorbing polyglycerol, based on total weight of the sunscreen composition.1. A sunscreen composition comprising: a polymer composition comprising a UV-absorbing polyglycerol comprising a UV-absorbing chromophore chemically bound thereto; and a cosmetically-acceptable topical carrier comprising at least one percent of a non-UV-absorbing polyglycerol, based on the total weight of said sunscreen composition. 2. The sunscreen composition of claim 1, wherein said cosmetically-acceptable topical carrier further comprises a hydrophobic diluent. 3. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol is soluble in said hydrophobic diluent. 4. The sunscreen composition of claim 1, wherein the sum of the weight of said non-UV-absorbing polyglycerol and said UV-absorbing polyglycerol is at least twelve percent, based on the total weight of said sunscreen composition. 5. The sunscreen composition of claim 1, wherein the sum of the weight of said non-UV-radiation absorbing polyglycerol and said UV-absorbing polyglycerol is at least twenty percent, based on the total weight of said sunscreen composition. 6. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol comprises a hydrophobic moiety. 7. The sunscreen composition of claim 1, wherein said non-UV-absorbing polyglycerol comprises a hydrophobic moiety. 8. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol comprises a hydrophobic moiety and said non-UV-absorbing polyglycerol comprises a hydrophobic moiety. 9. The sunscreen composition of claim 8, wherein said hydrophobic moiety has an average number of carbon atoms that is from 8 to 30. 10. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol comprises a first hydrophobic moiety having a first average number of carbon atoms and said non-UV-absorbing polyglycerol comprises a second hydrophobic moiety having a second average number of carbon atoms, wherein said first average number of carbon atoms and said second average number of carbon atoms are within 10 carbon atoms of one another. 11. The sunscreen composition of claim 10, wherein said first average number of carbon atoms and said second average number of carbon atoms are within 2 carbon atoms of one another. 12. The sunscreen composition of claim 1, wherein said UV-absorbing polyglycerol has a first average degree of glycerol polymerization and said non-UV-absorbing polyglycerol has a second average degree of glycerol polymerization, and wherein said first average degree of glycerol polymerization and said second average degree of glycerol polymerization are each from about 2 to about 20. 13. The sunscreen composition of claim 12, wherein said first average degree of glycerol polymerization and said second average degree of glycerol polymerization are within about 2 units of each other. 14. The sunscreen composition of claim 1, comprising about two percent or more of said non-UV-absorbing polyglycerol and about ten percent or more of said UV-absorbing polyglycerol. 15. The sunscreen composition of claim 1 wherein said UV-absorbing chromophore is selected from the group consisting of a transesterification product of 3-(3-(2H-benzo[d][1,2,3]triazol-2-yl)-5-(tert-butyl)-4-hydroxyphenyl)propanoic acid with polyethylene glycol 300, benzenepropanoic acid, 3-(2H-benzotriazol-2-yl)-5-(1,1-dimethylethyl)-4-hydroxy-, C7-9-branched and linear alkyl esters, 6-octyl-2-(4-(4,6-di([1,1′-biphenyl]-4-yl)-1,3,5-triazin-2-yl)-3-hydroxyphenoxy)propanoate, 3-(3-(2H-benzo[d][1,2,3]triazol-2-yl)-5-(tert-butyl)-4-hydroxyphenyl)propanoic acid, and polyethylene glycol ester of 3-[3-(2H-1,2,3-benzotriazol-2-yl)-5-tert-butyl-4-hydroxyphenyl]propanoate. 16. The sunscreen composition of claim 1 wherein said UV-absorbing chromophore is 3-(3-(2H-benzo[d][1,2,3]triazol-2-yl)-5-(tert-butyl)-4-hydroxyphenyl)propanoic acid. 17. The sunscreen composition of claim 1 wherein said UV-absorbing chromophore is polyethylene glycol ester of 3-[3-(2H-1,2,3-benzotriazol-2-yl)-5-tert-butyl-4-hydroxyphenyl]propanoate 18. The sunscreen composition of claim 1 comprising from about 5 percent to about 50 percent by weight of said UV-absorbing polyglycerol. 19. The sunscreen composition of claim 1 comprising from about 10 percent to about 30 percent by weight of said UV-absorbing polyglycerol. 20. The sunscreen composition of claim 1 comprising about 10 percent or more of said UV-absorbing polyglycerol. 21. The sunscreen composition of claim 1 comprising about 25 percent or more of said UV-absorbing polyglycerol. 22. The sunscreen composition of claim 1 wherein said sunscreen composition is essentially free of non-polymeric UV-absorbing sunscreen agents. 23. The sunscreen composition of claim 1 wherein said sunscreen composition is essentially free of a sunscreen agent other than said UV-absorbing polyglycerol. 24. The sunscreen composition of claim 1 comprising a sunscreen agent selected from the group consisting of a UV-absorbing polymer other than said UV-absorbing polyglycerol, a non-polymeric UV-absorbing sunscreen agent and non-UV-absorbing, light-scattering particles.
1,600
331
12,735,595
1,635
The invention relates to cationic siRNAs, characterized in that they are double-stranded RNA fragments, grafted to the ends of which are oligocations, the number of cationic charges grafted being comparable to or greater than that of the anionic charges carried by the internucleoside phosphates of the RNA strands.
1. A cationic siRNA, characterized in that it is a fragment of double-stranded RNA, at the ends of which are grafted oligocations, the number of grafted cationic charges being comparable to or greater than that of the anionic charges borne by the internucleoside phosphates of the RNA strands. 2. The cationic siRNA as claimed in claim 1, characterized in that the total sum of the cationic charges of the grafted oligocations minus the total sum of the anionic charges of the internucleoside phosphate groups is from −30 to +50. 3. The cationic siRNA as claimed in claim 1, characterized in that the strand complementarity is total or partial in the double helix. 4. The cationic siRNA as claimed in claim 1, characterized in that each oligoribonucleotide has a single-strand extension at the 3′ end of the double helix. 5. The cationic siRNA as claimed in claim 1, characterized in that the oligocations are grafted at the 3′ and/or 5′ end of the sense strand and/or at the 3′ end of the antisense strand. 6. The cationic siRNA as claimed in claim 1, characterized in that said fragments are formed from double-stranded RNA 15 to 30 nucleotides long, in particular from 19 to 30 nucleotides long, at the ends of which are grafted, by covalent bonding, 1 to 3 oligocations, the total number of cationic charges of which is comparable to or greater than the number of anionic charges borne by the RNA. 7. The cationic siRNA as claimed in claim 1, characterized in that at least one strand corresponds to formula (I) Ni-Aj or Ai-Ni  (I) in which Ni represents an i-mer oligoribonucleotide, with i=15 to 30 and in particular 19 to 30, and also any chemical modifications or substitutions that conserve the A form of the double helix of the siRNA, Aj represents a j-mer oligocation, with j=1 to 50, A corresponding to formula (II) —HPO3—R1—(X—R2)n1—X—R3—O—  (II), with R1, R2 and R3, which may be identical or different, representing a lower alkylene radical, X being NH or NC(NH2)2, and n1=2 to 20, or to formula (III) —HPO3—R4—CH(R5X1)—R6—O—,  (III) with R4, R5 and R6, which may be identical or different, representing a lower alkylene radical and X1 being chosen from putrescine, spermidine or spermine, or to formula (IV) —HPO3—R7-(aa)n2—R8—O—,  (IV) with R7 and R8, which may be identical or different, representing a lower alkylene radical, (aa)n2 being a peptide containing natural amino acids with cationic side chains, such as arginine, lysine, ornithine, histidine or diaminopropionic acid and n2=2 to 20. 8. The cationic siRNA as claimed in claim 1, characterized in that the oligocations are oligoamines. 9. The cationic siRNA as claimed in claim 8, characterized in that the oligoamines are chosen from the group comprising spermine, spermidine or putrescine. 10. The cationic siRNA as claimed in claim 9, characterized in that it is an oligonucleotide-oligospermine of structure 11. The cationic siRNA as claimed in claim 1, for use as a medicament. 12. A pharmaceutical composition, characterized in that it contains an effective amount of at least one cationic siRNA, as claimed in claim 1, in combination with a pharmaceutically inert vehicle, a cationic lipid or a cationic polymer. 13. Application of the cationic siRNA as claimed in claim 1, in molecular biology and functional genomics. 14. A process for the automated synthesis of a strand of cationic siRNA as claimed in claim 1, characterized in that it comprises: the sequential coupling from 3′ to 5′ of 19 to 30 ribonucleotides to a solid support, preceded and/or followed by the sequential coupling of cationic synthons, the strand corresponding to formula (VI) dT-dT-(N)i-Aj or Aj-dT-dT-(N)i  (VI). 15. The process as claimed in claim 14, characterized in that the cationic synthons are phosphoramidite oligoamines and correspond: to formula (VII) P(OR9)(N(R10)2)—O—R1—(X—R2)n1—X—R3—O-Prot,  (VII) in which R1, R2, R3 and n1 are as defined above, X is a protected group NH or NC(NH2)2, R9 is —CH2CH2CN or a lower alkylene radical, R19 is a lower alkylene radical, or —N(R10)2 is a pyrrolidino, piperidino or morpholino radical and Prot is a protecting group used in oligonucleotide synthesis, such as DMT or MMT; or to formula (VIII) P(OR9)(N(R10)2)—O—R4—CH(R5X1)-R6—O-Prot,  (VIII) in which R4, R5 and R6, which may be identical or different, represent a lower alkylene radical, X1 is a putrescine, spermidine or spermine with suitable protecting groups, and R9 and R10 are as defined above; or to formula (IX) P(OR9)(N(R10)2)—O—R7-(aa)n2—R8—O-Prot,  (IX) in which R7, R8, R9, R10, n2 and Prot are as defined above, (aa)n2 is a peptide containing natural amino acids with protected cationic side chains, such as arginine, lysine, ornithine, histidine or diaminopropionic acid, and n2=2 to 20.
The invention relates to cationic siRNAs, characterized in that they are double-stranded RNA fragments, grafted to the ends of which are oligocations, the number of cationic charges grafted being comparable to or greater than that of the anionic charges carried by the internucleoside phosphates of the RNA strands.1. A cationic siRNA, characterized in that it is a fragment of double-stranded RNA, at the ends of which are grafted oligocations, the number of grafted cationic charges being comparable to or greater than that of the anionic charges borne by the internucleoside phosphates of the RNA strands. 2. The cationic siRNA as claimed in claim 1, characterized in that the total sum of the cationic charges of the grafted oligocations minus the total sum of the anionic charges of the internucleoside phosphate groups is from −30 to +50. 3. The cationic siRNA as claimed in claim 1, characterized in that the strand complementarity is total or partial in the double helix. 4. The cationic siRNA as claimed in claim 1, characterized in that each oligoribonucleotide has a single-strand extension at the 3′ end of the double helix. 5. The cationic siRNA as claimed in claim 1, characterized in that the oligocations are grafted at the 3′ and/or 5′ end of the sense strand and/or at the 3′ end of the antisense strand. 6. The cationic siRNA as claimed in claim 1, characterized in that said fragments are formed from double-stranded RNA 15 to 30 nucleotides long, in particular from 19 to 30 nucleotides long, at the ends of which are grafted, by covalent bonding, 1 to 3 oligocations, the total number of cationic charges of which is comparable to or greater than the number of anionic charges borne by the RNA. 7. The cationic siRNA as claimed in claim 1, characterized in that at least one strand corresponds to formula (I) Ni-Aj or Ai-Ni  (I) in which Ni represents an i-mer oligoribonucleotide, with i=15 to 30 and in particular 19 to 30, and also any chemical modifications or substitutions that conserve the A form of the double helix of the siRNA, Aj represents a j-mer oligocation, with j=1 to 50, A corresponding to formula (II) —HPO3—R1—(X—R2)n1—X—R3—O—  (II), with R1, R2 and R3, which may be identical or different, representing a lower alkylene radical, X being NH or NC(NH2)2, and n1=2 to 20, or to formula (III) —HPO3—R4—CH(R5X1)—R6—O—,  (III) with R4, R5 and R6, which may be identical or different, representing a lower alkylene radical and X1 being chosen from putrescine, spermidine or spermine, or to formula (IV) —HPO3—R7-(aa)n2—R8—O—,  (IV) with R7 and R8, which may be identical or different, representing a lower alkylene radical, (aa)n2 being a peptide containing natural amino acids with cationic side chains, such as arginine, lysine, ornithine, histidine or diaminopropionic acid and n2=2 to 20. 8. The cationic siRNA as claimed in claim 1, characterized in that the oligocations are oligoamines. 9. The cationic siRNA as claimed in claim 8, characterized in that the oligoamines are chosen from the group comprising spermine, spermidine or putrescine. 10. The cationic siRNA as claimed in claim 9, characterized in that it is an oligonucleotide-oligospermine of structure 11. The cationic siRNA as claimed in claim 1, for use as a medicament. 12. A pharmaceutical composition, characterized in that it contains an effective amount of at least one cationic siRNA, as claimed in claim 1, in combination with a pharmaceutically inert vehicle, a cationic lipid or a cationic polymer. 13. Application of the cationic siRNA as claimed in claim 1, in molecular biology and functional genomics. 14. A process for the automated synthesis of a strand of cationic siRNA as claimed in claim 1, characterized in that it comprises: the sequential coupling from 3′ to 5′ of 19 to 30 ribonucleotides to a solid support, preceded and/or followed by the sequential coupling of cationic synthons, the strand corresponding to formula (VI) dT-dT-(N)i-Aj or Aj-dT-dT-(N)i  (VI). 15. The process as claimed in claim 14, characterized in that the cationic synthons are phosphoramidite oligoamines and correspond: to formula (VII) P(OR9)(N(R10)2)—O—R1—(X—R2)n1—X—R3—O-Prot,  (VII) in which R1, R2, R3 and n1 are as defined above, X is a protected group NH or NC(NH2)2, R9 is —CH2CH2CN or a lower alkylene radical, R19 is a lower alkylene radical, or —N(R10)2 is a pyrrolidino, piperidino or morpholino radical and Prot is a protecting group used in oligonucleotide synthesis, such as DMT or MMT; or to formula (VIII) P(OR9)(N(R10)2)—O—R4—CH(R5X1)-R6—O-Prot,  (VIII) in which R4, R5 and R6, which may be identical or different, represent a lower alkylene radical, X1 is a putrescine, spermidine or spermine with suitable protecting groups, and R9 and R10 are as defined above; or to formula (IX) P(OR9)(N(R10)2)—O—R7-(aa)n2—R8—O-Prot,  (IX) in which R7, R8, R9, R10, n2 and Prot are as defined above, (aa)n2 is a peptide containing natural amino acids with protected cationic side chains, such as arginine, lysine, ornithine, histidine or diaminopropionic acid, and n2=2 to 20.
1,600
332
14,913,132
1,627
The present invention relates to an indole compound represented by formula (1), a pharmaceutically acceptable salt or isomer thereof, a composition for prevention or treatment of necrosis and necrosis-associated diseases, and a method for preparing the composition, the composition comprising the indole compound or the pharmaceutically acceptable salt or isomer thereof as an active ingredient.
1. An indole compound of the following Formula (1) or a pharmaceutically acceptable salt or isomer thereof: in which m denotes a number of 1 to 3; n denotes a number of 0 to 2; R1 represents hydrogen, C1-C6-alkyl, —(CH2)n—C3-C6-cycloalkyl or —(CH2)n-heterocyclyl, wherein heterocyclyl is a 4- to 8-membered ring having 1 to 3 heteroatoms selected from N, O and S; R2 represents C1-C6-alkyl or —(CH2)n-A-R6, wherein A represents C4-C8-cycloalkyl, or represents 4- to 8-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N, O and S, or represents 6- to 10-membered aryl, R6 represents hydrogen, C1-C6-alkyl, halogen, hydroxy, nitrile, nitro, —C(O)—R7 or —SO2R7, and R7 represents C1-C6-alkyl or allyl, or represents 6- to 10-membered aryl, or represents 4- to 8-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N and S, and is optionally substituted with oxo; R3 represents hydrogen, halogen, hydroxy, —O—R7, —NH—R7 or —(CH2)n—R7; R4 represents hydrogen or XR8R9, wherein X represents CH or N, R8 and R9 independently of one another represent hydrogen or Z—R10, Z represents —(CH2)n—, —C(O)—, —C(O)(CH2)n— or —(CH2)nC(O)—, R10 represents hydrogen, amino, C3-C6-cycloalkyl or —(NH)rC(═NH)NH2, or represents 4- to 8-membered heteroaryl or heterocyclyl each of which has 1 to 3 heteroatoms selected from N, O and S, and r denotes a number of 0 or 1; R5 represents hydrogen, hydroxy or C1-C6-alkoxy, or represents 6- to 10-membered aryl or 6- to 10-membered aryl-C1-C6-alkyloxy, or represents —(CH2)n-4- to 9-membered heterocyclyl which has 1 to 4 heteroatoms selected from N, O and S, and optionally contains oxo; R4 and R5 may be connected with an atom(s) to which they are attached to form the following structure: wherein R5 and R8 are the same as defined above; and where alkyl, alkoxy, aryl, cycloalkyl, heterocycle and heteroaryl may be optionally substituted, and the substituents are one or more selected from the group consisting of hydroxy, halogen, nitrile, amino, C1-C6-alkylamino, di(C1-C6-alkyl)amino, carboxy, C1-C6-alkyl, halogeno-C1-C6-alkyl, C1-C6-alkoxy, aryl-C1-C6-alkoxy and oxo. 2. The compound according to claim 1, wherein m denotes a number of 1 to 3; n denotes a number of 0 to 2; R1 represents hydrogen, C1-C6-alkyl, —(CH2)n—C3-C6-cycloalkyl or —(CH2)n-heterocyclyl, wherein heterocyclyl is a 4- to 8-membered ring having 1 to 3 heteroatoms selected from N, O and S; R2 represents C1-C6-alkyl or —(CH2)n-A-R6, wherein A represents C4-C6-cycloalkyl, or represents 5- or 6-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N, O and S, or represents 6- to 10-membered aryl, R6 represents hydrogen, C1-C6-alkyl, halogen, hydroxy, —C(O)—R7 or —SO2R7, and R7 represents C1-C6-alkyl, or represents 6- to 10-membered aryl, or represents 5- or 6-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N and S, and is optionally substituted with oxo; R3 represents hydrogen, halogen, —O—R7, —NH—R7 or —(CH2)n—R7; R4 represents hydrogen or XR8R9, wherein X represents CH or N, R8 and R9 independently of one another represent hydrogen or Z—R10, Z represents —(CH2)n—, —C(O)—, —C(O)(CH2)n— or —(CH2)nC(O)—, R10 represents hydrogen, amino, C3-C6-cycloalkyl or —(NH)rC(═NH)NH2, or represents 4- to 8-membered heteroaryl or heterocyclyl each of which has 1 to 3 heteroatoms selected from N, O and S, and is optionally substituted with amino, and r denotes a number of 0 or 1; R5 represents hydrogen, hydroxy or C1-C6-alkoxy, or represents 6- to 10-membered aryl or 6- to 10-membered aryl-C1-C6-alkyloxy, or represents —(CH2)n-4- to 9-membered heterocyclyl which has 1 to 4 heteroatoms selected from N, O and S, and optionally contains oxo and is optionally substituted with halogeno-C1-C6-alkyl; and R4 and R5 may be connected with an atom(s) to which they are attached to form the following structure: wherein R5 and R8 are the same as defined above. 3. The compound according to claim 1, which is the following Formula (1a) or (1b): X, m, n, R1, R2, R3, R5, R8 and R9 are the same as defined in claim 1. 4. The compound according to claim 3, wherein the ring of Formula (1b) is cyclohexyl, pyrrolidine or piperidine. 5. The compound according to claim 1, wherein R1 is hydrogen, C1-C6-alkyl or 5- or 6-membered heterocyclyl having 1 to 3 heteroatoms selected from N, O and S. 6. The compound according to claim 5, wherein R1 is hydrogen, isopentyl or tetrahydropyran. 7. The compound according to claim 1, wherein R2 is C1-C6-alkyl or —(CH2)n-A-R6, wherein n denotes a number of 0 to 2, A represents C4-C6-cycloalkyl, or represents 5- or 6-membered heterocyclyl having 1 or 2 heteroatoms selected from N and O, or represents phenyl, R6 represents hydrogen, —C(O)—R7 or —SO2R7, and R7 represents C1-C3-alkyl. 8. The compound according to claim 7, wherein R2 is isopentyl, cyclopentyl, benzyl, tetrahydropyran, tetrahydropyran-4-ylmethyl, 1-acetyl-piperidine, tetrahydropyran-2-ylmethyl or piperidin-4-ylmethyl. 9. The compound according to claim 1, wherein R3 is hydrogen, halogen or —(CH2)n—R7, wherein n denotes a number of 0 to 2, R7 represents C1-C6-alkyl, or represents 5- or 6-membered heterocyclyl which has 1 or 2 heteroatoms selected from N and S, and is optionally substituted with oxo. 10. The compound according to claim 9, wherein R3 is hydrogen, methyl, chloro or 1,1-dioxothiomorpholin-4-ylmethyl. 11. The compound according to claim 1, wherein R5 is hydrogen, hydroxyl, C1-C6-alkoxy, 6- to 10-membered aryl, 6- to 10-membered aryl-C1-C3-alkyloxy or —(CH2)n-4- to 9-membered heterocyclyl which has 1 to 4 heteroatoms selected from N, O and S, and optionally contains oxo and is optionally substituted with halogeno-C1-C3-alkyl, wherein n denotes a number of 0 to 2. 12. The compound according to claim 11, wherein R5 is hydrogen, hydroxy, benzyloxy, 1,1-dioxothiomorpholine, 2-oxopiperazine, 3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl, morpholine or phenyl. 13. The compound according to claim 1, wherein R8 is hydrogen or Z—R10, wherein Z represents —(CH2)n—, —C(O)—, —C(O)(CH2)n— or —(CH2)nC(O)—, n denotes a number of 0 to 2, R10 represents hydrogen, amino, C4-C6-cycloalkyl or —(NH)rC(═NH)NH2, or represents 5- or 6-membered heteroaryl or heterocyclyl each of which has 1 to 3 heteroatoms selected from N and S, and is optionally substituted with amino, and r denotes a number of 0 or 1. 14. The compound according to claim 1, wherein R8 is hydrogen, cyclohexyl-ethyl, 2-amino-pyridin-3-ylmethyl, pyrrolidine, 3-amino-triazol-5-carbonyl, aminomethyl-carbonyl, NH2(NH═)C—, NH2(NH═)C—NH—CH2—C(O)—, 2-amino-thiazol-4-ylmethyl, cyclopentyl-methyl, NH2(NH═)C—NH—C(O)—CH2— or acetyl. 15. The compound according to claim 1, which is selected from the following group: 4-(5-Chloro-7-cyclopentylamino-1H-indol-2-ylmethyl)-piperazin-2-one; 4-{2-[5-Chloro-7-(piperidin-4-ylamino)-1H-indol-2-yl]-ethyl}-piperazin-2-one; 4-{2-[7-(1-Acetyl-piperidin-4-ylamino)-5-chloro-1H-indol-2-yl]-ethyl}-piperazin-2-one; (5-Chloro-7-cyclopentylamino-1H-indol-2-yl)-methanol; 2-(5-Chloro-7-cyclopentylamino-1H-indol-2-yl)-ethanol; 4-{5-Methyl-7-[(piperidin-4-ylmethyl)-amino]-1H-indol-2-ylmethyl}-piperazin-2-one; [2-(1,1-Dioxothiomorpholin-4-ylmethyl)-5-methyl-1H-indol-7-yl]-(tetrahydropyran-4-ylmethyl)-amine; Cyclopentyl-[2-(1,1-dioxothiomorpholin-4-ylmethyl)-5-methyl-1H-indol-7-yl]-amine; 4-[5-Methyl-7-(tetrahydropyran-4-ylmethylamino)-1H-indol-2-ylmethyl)-piperazin-2-one; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(tetrahydrofuran-2-ylmethyl)-amine; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-2-ylmethyl)-amine; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(1-methanesulfonyl-piperidin-4-yl)-amine; 1-(4-{5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-piperidin-1-yl)-ethanone; 4-[2-(7-Benzylamino-5-chloro-1H-indol-2-yl)-ethyl]-piperazin-2-one; 4-(2-{5-Chloro-7-[(tetrahydrofuran-2-ylmethyl)-amino]-1H-indol-2-yl)-ethyl]-piperazin-2-one; 4-(2-{5-Chloro-7-[(tetrahydropyran-2-ylmethyl)-amino]-1H-indol-2-yl)-ethyl]-piperazin-2-one; 4-{5-Chloro-7-[(tetrahydropyran-4-ylmethyl)-amino]-1H-indol-2-ylmethyl}-piperazin-2-one; {5-Chloro-2-[2-(1,1-dioxothiomorpholin-4-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-4-yl)-amine; {5-Chloro-2-[2-(1,1-dioxothiomorpholin-4-yl)-ethyl]-1H-indol-7-yl}-cyclopentyl-amine; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-cyclopentyl-amine; {5-Chloro-2-[2-(1,1-dioxothiomorpholin-4-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-4-ylmethyl)-amine; 4-(2-{5-Chloro-7-[(tetrahydropyran-4-ylmethyl)-amino]-1H-indol-2-yl}-ethyl)-piperazin-2-one; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-4-ylmethyl)-amine; [5-Chloro-2-(2-morpholin-4-yl-ethyl)-1H-indol-7-yl-(tetrahydropyran-4-ylmethyl)-amine; Cyclopentyl-(5-methyl-2-morpholin-4-ylmethyl-1H-indol-7-yl)-amine; (2-Cyclohexyl-5-methyl-1H-indol-7-yl)-cyclopentyl-amine; [2-((R)-1-amino-2-phenyl-ethyl)-5-methyl-1H-indol-7-yl]-cyclopentyl-amine; {2-[(R)-1-(2-cyclohexyl-ethylamino)-2-phenyl-ethyl]-5-methyl-1H-indol-7-yl}-cyclopentylamine; Benzyl-{5-chloro-2-[(R)-2-phenyl-1-(pyrrolidin-3-ylamino)-ethyl]-1H-indol-7-yl}-amine; 2-Amino-N—[(R)-1-(7-benzylamino-5-chloro-1H-indol-2-yl)-2-phenylethyl]-acetamide; N—[(R)-1-(7-Benzylamino-5-chloro-1H-indol-2-yl)-2-phenyl-ethyl]-guanidine; {2-[(R)-1-(cyclohexylmethyl-amino)-2-phenyl-ethyl]-5-methyl-1H-indol-7-yl}-cyclopentyl-amine; (2-{(S)-1-[(2-amino-pyridin-3-ylmethyl)-amino]-2-phenyl-ethyl}-5-chloro-1H-indol-7-yl)-(3-methyl-butyl)-amine; 3-Amino-4H-[1,2,4]triazol-4-carboxilic acid [(S)-1-(7-benzylamino-5-chloro-1H-indol-2-yl)-2-phenyl-ethyl]-amide; 2-Amino-N—{(S)-1-[5-chloro-7-(3-methyl-butylamino)-1H-indol-2-yl]-2-phenyl-ethyl}-acetamide; N—[(R)-1-(7-benzylamino-5-chloro-1H-indol-2-yl)-2-phenyl-ethyl]-2-guanidino-acetamide; (S)-2-Amino-2-(5-chloro-7-cyclopentylamino-1H-indol-2-yl)-ethanol; ((S)-5-chloro-2-pyrrolidin-2-yl-1H-indol-7-yl)-bis-(3-methyl-butyl)-amine; (2S,4R)-4-benzyloxy-2-(5-chloro-7-cyclopentylamino-1H-indol-2-yl)-pyrrolidin-1-carboxamidine; (S)-2-{7-[bis-(3-methyl-butyl)-amino]-5-chloro-1H-indol-2-yl)-pyrrolidin-1-carboxamidine; (S)-2-(5-chloro-7-cyclopentylamino-1H-indol-2-yl)-pyrrolidin-1-carboxamidine; [2-((2S,4R)-4-benzyloxy-[1,3′]bipyrrolidin-2-yl)-5-chloro-1H-indol-7-yl)-cyclopentyl-amine; ((S)-[1,3′]bipyrrolidin-2-yl-5-chloro-1H-indol-7-yl)-cyclopentyl-amine; {2-[(S)-1-(2-amino-thiazol-4-ylmethyl)-pyrrolidin-2-yl]-5-chloro-1H-indol-7-yl)-cyclopentyl-amine; [5-Chloro-2-((S)-1-cyclopentylmethyl-pyrrolidin-2-yl)-1H-indol-7-yl]-bis-(3-methyl-butyl)-amine; ((S)-[1,3′]bipyrrolidin-2-yl-5-chloro-1H-indol-7-yl)-bis-(3-methyl-butyl)-amine; N-[2-((S)-2-{7-[bis-(3-methyl-butyl)-amino]-5-chloro-1H-indol-2-yl}-pyrrolidin-1-yl)-acetyl]-guanidine; (5-Chloro-2-piperidin-4-yl-1H-indol-7-yl)-cyclopentyl-amine; (5-Chloro-2-piperidin-4-yl-1H-indol-7-yl)-(tetrahydropyran-4-yl)-amine; Cyclopentyl-(5-methyl-2-piperidin-4-yl-1H-indol-7-yl)-amine; 1-[4-(7-Cyclopentylamino-5-methyl-1H-indol-2-yl)-piperidin-1-yl]-ethanone; (5-Methyl-2-piperidin-4-yl-1H-indol-7-yl)-(tetrahydropyran-4-yl)-amine; (5-Methyl-2-piperidin-4-yl-1H-indol-7-yl)-bis-(tetrahydropyran-4-yl)-amine; 1-{4-[5-Methyl-7-(tetrahydropyran-4-ylamino)-1H-indol-2-yl]-piperidin-1-yl]-ethanone; (2-Cyclohexyl-5-methyl-1H-indol-7-yl)-cyclopentyl-amine; [5-(1,1-Dioxo-thiomorpholin-4-ylmethyl)-2-piperidin-4-yl-1H-indol-7-yl]-(tetrahydropyran-4-yl)-amine; and 1-{4-[5-(1,1-Dioxo-thiomorpholin-4-ylmethyl)-7-(tetrahydropyran-4-ylamino)-1H-indol-2-yl]-piperidin-1-yl}-ethanone. 16. A composition for the prevention or treatment of necrosis and necrosis-associated diseases, which comprises a therapeutically effective amount of the compounds of Formula (1), a pharmaceutically acceptable salt or isomer thereof as defined in claim 1 as an active ingredient together with a pharmaceutically acceptable carrier or diluent. 17. The composition according to claim 16, wherein the necrosis and necrosis-associated diseases are selected from the group consisting of acute/chronic hepatic disease, neurodegenerative disease, ischemic disease, diabetes, pancreatitis, bacterial/viral sepsis, necrotizing procolitis, cystic fibrosis, rheumatoid arthritis, degenerative arthritis, nephropathy, bacterial infection, viral infection, multiple sclerosis, leukemia, lymphoma, neonatal respiratory distress syndrome, asphyxia, tuberculosis, endometriosis, angiasthenia, psoriasis, chilblain, steroid treatment complications, gangrene, pressure sores, hemoglobinuria, burns, hyperthermia, Crohn's disease, celiac disease, compartment syndrome, spinal cord injury, glomerulonephritis, muscular dystrophy, metabolic inherited disease, mycoplasmal disease, anthrax, Andersen's disease, congenital mitochondrial disease, phenylketonuria, placental infarction, syphilis, and aseptic necrosis. 18. The composition according to claim 16, wherein the necrosis and necrosis-associated diseases are caused by a drug and toxic substance, and are selected from the group consisting of the necrosis associated with alcoholism, the exposure to, or administration or self-administration of, cocaine, drug, antibiotic, anti-cancer agent, adriamycin, puromycin, bleomycin, NSAID, cyclosporine, chemical toxin, poison gas, agrochemical, heavy metal, or injury due to the exposure to radioactivity/UV and associated necrosis thereof. 19. The composition according to claim 16, which is for hepatoprotection, hepatic functional improvement, and prevention or treatment of hepatic disease. 20. The composition according to claim 19, wherein the hepatic disease is selected from the group consisting of liver transplantation, alcoholic or non-alcoholic fatty liver, hepatic fibrosis, hepatocirrhosis and hepatitis caused by virus or drug. 21. The composition according to claim 19, wherein the hepatic disease is alcoholic acute/chronic hepatic disease. 22. The composition according to claim 19, wherein the hepatic disease is fatty acid-induced fatty liver or acute/chronic hepatic disease derived from fatty liver. 23. The composition according to claim 19, wherein the hepatic disease is mediated by reactive oxygen species (ROS). 24. The composition according to claim 19, wherein the hepatic disease is mediated by heavy metal. 25. The composition according to claim 16, which is co-administered with a prophylactic or therapeutic agent for drug-induced necrosis and necrosis-associated diseases. 26. The composition according to claim 25, wherein the prophylactic or therapeutic agent for drug-induced necrosis and necrosis-associated diseases is selected from the group consisting of antibiotic, anti-cancer agent, anti-viral agent, anti-infective, anti-inflammatory agent, anti-coagulant, lipid-improving agent, cell death inhibitor, anti-hypertensive agent, anti-diabetic/anti-obesity agent, therapeutic agent for cardiovascular disease, therapeutic agent for neurodegenerative disease, anti-aging agent and therapeutic agent for metabolic disease. 27. The composition according to claim 19, which is co-administered with an agent selected from the group consisting of hepatocyte regeneration promoter, hepatic functional adjuvant, anti-viral agent, immunosuppressant and fibrosis inhibitor. 28. The composition according to claim 17, wherein the neurodegenerative disease is dementia, Parkinson's disease or Huntington's disease. 29. The composition according to claim 17, wherein the ischemic disease is cardiac disease, reperfusion injury, ischemic stroke or ischemic injury. 30. The composition according to claim 17, wherein the diabetes is caused by a pancreatic cell-destroying substance, diabetic complications or diabetic vascular disease. 31. The composition according to claim 30, wherein the diabetes is mediated by virus, hyperglycemia, fatty acid, diet, toxin or streptozotocin. 32. A method of preparing a composition for the prevention or treatment of necrosis and necrosis-associated diseases, which comprises the step of mixing the compound of Formula (1), a pharmaceutically acceptable salt or isomer thereof as defined in claim 1 as an active ingredient together with a pharmaceutically acceptable carrier.
The present invention relates to an indole compound represented by formula (1), a pharmaceutically acceptable salt or isomer thereof, a composition for prevention or treatment of necrosis and necrosis-associated diseases, and a method for preparing the composition, the composition comprising the indole compound or the pharmaceutically acceptable salt or isomer thereof as an active ingredient.1. An indole compound of the following Formula (1) or a pharmaceutically acceptable salt or isomer thereof: in which m denotes a number of 1 to 3; n denotes a number of 0 to 2; R1 represents hydrogen, C1-C6-alkyl, —(CH2)n—C3-C6-cycloalkyl or —(CH2)n-heterocyclyl, wherein heterocyclyl is a 4- to 8-membered ring having 1 to 3 heteroatoms selected from N, O and S; R2 represents C1-C6-alkyl or —(CH2)n-A-R6, wherein A represents C4-C8-cycloalkyl, or represents 4- to 8-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N, O and S, or represents 6- to 10-membered aryl, R6 represents hydrogen, C1-C6-alkyl, halogen, hydroxy, nitrile, nitro, —C(O)—R7 or —SO2R7, and R7 represents C1-C6-alkyl or allyl, or represents 6- to 10-membered aryl, or represents 4- to 8-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N and S, and is optionally substituted with oxo; R3 represents hydrogen, halogen, hydroxy, —O—R7, —NH—R7 or —(CH2)n—R7; R4 represents hydrogen or XR8R9, wherein X represents CH or N, R8 and R9 independently of one another represent hydrogen or Z—R10, Z represents —(CH2)n—, —C(O)—, —C(O)(CH2)n— or —(CH2)nC(O)—, R10 represents hydrogen, amino, C3-C6-cycloalkyl or —(NH)rC(═NH)NH2, or represents 4- to 8-membered heteroaryl or heterocyclyl each of which has 1 to 3 heteroatoms selected from N, O and S, and r denotes a number of 0 or 1; R5 represents hydrogen, hydroxy or C1-C6-alkoxy, or represents 6- to 10-membered aryl or 6- to 10-membered aryl-C1-C6-alkyloxy, or represents —(CH2)n-4- to 9-membered heterocyclyl which has 1 to 4 heteroatoms selected from N, O and S, and optionally contains oxo; R4 and R5 may be connected with an atom(s) to which they are attached to form the following structure: wherein R5 and R8 are the same as defined above; and where alkyl, alkoxy, aryl, cycloalkyl, heterocycle and heteroaryl may be optionally substituted, and the substituents are one or more selected from the group consisting of hydroxy, halogen, nitrile, amino, C1-C6-alkylamino, di(C1-C6-alkyl)amino, carboxy, C1-C6-alkyl, halogeno-C1-C6-alkyl, C1-C6-alkoxy, aryl-C1-C6-alkoxy and oxo. 2. The compound according to claim 1, wherein m denotes a number of 1 to 3; n denotes a number of 0 to 2; R1 represents hydrogen, C1-C6-alkyl, —(CH2)n—C3-C6-cycloalkyl or —(CH2)n-heterocyclyl, wherein heterocyclyl is a 4- to 8-membered ring having 1 to 3 heteroatoms selected from N, O and S; R2 represents C1-C6-alkyl or —(CH2)n-A-R6, wherein A represents C4-C6-cycloalkyl, or represents 5- or 6-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N, O and S, or represents 6- to 10-membered aryl, R6 represents hydrogen, C1-C6-alkyl, halogen, hydroxy, —C(O)—R7 or —SO2R7, and R7 represents C1-C6-alkyl, or represents 6- to 10-membered aryl, or represents 5- or 6-membered heterocyclyl or heteroaryl each of which has 1 to 3 heteroatoms selected from N and S, and is optionally substituted with oxo; R3 represents hydrogen, halogen, —O—R7, —NH—R7 or —(CH2)n—R7; R4 represents hydrogen or XR8R9, wherein X represents CH or N, R8 and R9 independently of one another represent hydrogen or Z—R10, Z represents —(CH2)n—, —C(O)—, —C(O)(CH2)n— or —(CH2)nC(O)—, R10 represents hydrogen, amino, C3-C6-cycloalkyl or —(NH)rC(═NH)NH2, or represents 4- to 8-membered heteroaryl or heterocyclyl each of which has 1 to 3 heteroatoms selected from N, O and S, and is optionally substituted with amino, and r denotes a number of 0 or 1; R5 represents hydrogen, hydroxy or C1-C6-alkoxy, or represents 6- to 10-membered aryl or 6- to 10-membered aryl-C1-C6-alkyloxy, or represents —(CH2)n-4- to 9-membered heterocyclyl which has 1 to 4 heteroatoms selected from N, O and S, and optionally contains oxo and is optionally substituted with halogeno-C1-C6-alkyl; and R4 and R5 may be connected with an atom(s) to which they are attached to form the following structure: wherein R5 and R8 are the same as defined above. 3. The compound according to claim 1, which is the following Formula (1a) or (1b): X, m, n, R1, R2, R3, R5, R8 and R9 are the same as defined in claim 1. 4. The compound according to claim 3, wherein the ring of Formula (1b) is cyclohexyl, pyrrolidine or piperidine. 5. The compound according to claim 1, wherein R1 is hydrogen, C1-C6-alkyl or 5- or 6-membered heterocyclyl having 1 to 3 heteroatoms selected from N, O and S. 6. The compound according to claim 5, wherein R1 is hydrogen, isopentyl or tetrahydropyran. 7. The compound according to claim 1, wherein R2 is C1-C6-alkyl or —(CH2)n-A-R6, wherein n denotes a number of 0 to 2, A represents C4-C6-cycloalkyl, or represents 5- or 6-membered heterocyclyl having 1 or 2 heteroatoms selected from N and O, or represents phenyl, R6 represents hydrogen, —C(O)—R7 or —SO2R7, and R7 represents C1-C3-alkyl. 8. The compound according to claim 7, wherein R2 is isopentyl, cyclopentyl, benzyl, tetrahydropyran, tetrahydropyran-4-ylmethyl, 1-acetyl-piperidine, tetrahydropyran-2-ylmethyl or piperidin-4-ylmethyl. 9. The compound according to claim 1, wherein R3 is hydrogen, halogen or —(CH2)n—R7, wherein n denotes a number of 0 to 2, R7 represents C1-C6-alkyl, or represents 5- or 6-membered heterocyclyl which has 1 or 2 heteroatoms selected from N and S, and is optionally substituted with oxo. 10. The compound according to claim 9, wherein R3 is hydrogen, methyl, chloro or 1,1-dioxothiomorpholin-4-ylmethyl. 11. The compound according to claim 1, wherein R5 is hydrogen, hydroxyl, C1-C6-alkoxy, 6- to 10-membered aryl, 6- to 10-membered aryl-C1-C3-alkyloxy or —(CH2)n-4- to 9-membered heterocyclyl which has 1 to 4 heteroatoms selected from N, O and S, and optionally contains oxo and is optionally substituted with halogeno-C1-C3-alkyl, wherein n denotes a number of 0 to 2. 12. The compound according to claim 11, wherein R5 is hydrogen, hydroxy, benzyloxy, 1,1-dioxothiomorpholine, 2-oxopiperazine, 3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl, morpholine or phenyl. 13. The compound according to claim 1, wherein R8 is hydrogen or Z—R10, wherein Z represents —(CH2)n—, —C(O)—, —C(O)(CH2)n— or —(CH2)nC(O)—, n denotes a number of 0 to 2, R10 represents hydrogen, amino, C4-C6-cycloalkyl or —(NH)rC(═NH)NH2, or represents 5- or 6-membered heteroaryl or heterocyclyl each of which has 1 to 3 heteroatoms selected from N and S, and is optionally substituted with amino, and r denotes a number of 0 or 1. 14. The compound according to claim 1, wherein R8 is hydrogen, cyclohexyl-ethyl, 2-amino-pyridin-3-ylmethyl, pyrrolidine, 3-amino-triazol-5-carbonyl, aminomethyl-carbonyl, NH2(NH═)C—, NH2(NH═)C—NH—CH2—C(O)—, 2-amino-thiazol-4-ylmethyl, cyclopentyl-methyl, NH2(NH═)C—NH—C(O)—CH2— or acetyl. 15. The compound according to claim 1, which is selected from the following group: 4-(5-Chloro-7-cyclopentylamino-1H-indol-2-ylmethyl)-piperazin-2-one; 4-{2-[5-Chloro-7-(piperidin-4-ylamino)-1H-indol-2-yl]-ethyl}-piperazin-2-one; 4-{2-[7-(1-Acetyl-piperidin-4-ylamino)-5-chloro-1H-indol-2-yl]-ethyl}-piperazin-2-one; (5-Chloro-7-cyclopentylamino-1H-indol-2-yl)-methanol; 2-(5-Chloro-7-cyclopentylamino-1H-indol-2-yl)-ethanol; 4-{5-Methyl-7-[(piperidin-4-ylmethyl)-amino]-1H-indol-2-ylmethyl}-piperazin-2-one; [2-(1,1-Dioxothiomorpholin-4-ylmethyl)-5-methyl-1H-indol-7-yl]-(tetrahydropyran-4-ylmethyl)-amine; Cyclopentyl-[2-(1,1-dioxothiomorpholin-4-ylmethyl)-5-methyl-1H-indol-7-yl]-amine; 4-[5-Methyl-7-(tetrahydropyran-4-ylmethylamino)-1H-indol-2-ylmethyl)-piperazin-2-one; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(tetrahydrofuran-2-ylmethyl)-amine; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-2-ylmethyl)-amine; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(1-methanesulfonyl-piperidin-4-yl)-amine; 1-(4-{5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-piperidin-1-yl)-ethanone; 4-[2-(7-Benzylamino-5-chloro-1H-indol-2-yl)-ethyl]-piperazin-2-one; 4-(2-{5-Chloro-7-[(tetrahydrofuran-2-ylmethyl)-amino]-1H-indol-2-yl)-ethyl]-piperazin-2-one; 4-(2-{5-Chloro-7-[(tetrahydropyran-2-ylmethyl)-amino]-1H-indol-2-yl)-ethyl]-piperazin-2-one; 4-{5-Chloro-7-[(tetrahydropyran-4-ylmethyl)-amino]-1H-indol-2-ylmethyl}-piperazin-2-one; {5-Chloro-2-[2-(1,1-dioxothiomorpholin-4-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-4-yl)-amine; {5-Chloro-2-[2-(1,1-dioxothiomorpholin-4-yl)-ethyl]-1H-indol-7-yl}-cyclopentyl-amine; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-cyclopentyl-amine; {5-Chloro-2-[2-(1,1-dioxothiomorpholin-4-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-4-ylmethyl)-amine; 4-(2-{5-Chloro-7-[(tetrahydropyran-4-ylmethyl)-amino]-1H-indol-2-yl}-ethyl)-piperazin-2-one; {5-Chloro-2-[2-(3-trifluoromethyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-1-yl)-ethyl]-1H-indol-7-yl}-(tetrahydropyran-4-ylmethyl)-amine; [5-Chloro-2-(2-morpholin-4-yl-ethyl)-1H-indol-7-yl-(tetrahydropyran-4-ylmethyl)-amine; Cyclopentyl-(5-methyl-2-morpholin-4-ylmethyl-1H-indol-7-yl)-amine; (2-Cyclohexyl-5-methyl-1H-indol-7-yl)-cyclopentyl-amine; [2-((R)-1-amino-2-phenyl-ethyl)-5-methyl-1H-indol-7-yl]-cyclopentyl-amine; {2-[(R)-1-(2-cyclohexyl-ethylamino)-2-phenyl-ethyl]-5-methyl-1H-indol-7-yl}-cyclopentylamine; Benzyl-{5-chloro-2-[(R)-2-phenyl-1-(pyrrolidin-3-ylamino)-ethyl]-1H-indol-7-yl}-amine; 2-Amino-N—[(R)-1-(7-benzylamino-5-chloro-1H-indol-2-yl)-2-phenylethyl]-acetamide; N—[(R)-1-(7-Benzylamino-5-chloro-1H-indol-2-yl)-2-phenyl-ethyl]-guanidine; {2-[(R)-1-(cyclohexylmethyl-amino)-2-phenyl-ethyl]-5-methyl-1H-indol-7-yl}-cyclopentyl-amine; (2-{(S)-1-[(2-amino-pyridin-3-ylmethyl)-amino]-2-phenyl-ethyl}-5-chloro-1H-indol-7-yl)-(3-methyl-butyl)-amine; 3-Amino-4H-[1,2,4]triazol-4-carboxilic acid [(S)-1-(7-benzylamino-5-chloro-1H-indol-2-yl)-2-phenyl-ethyl]-amide; 2-Amino-N—{(S)-1-[5-chloro-7-(3-methyl-butylamino)-1H-indol-2-yl]-2-phenyl-ethyl}-acetamide; N—[(R)-1-(7-benzylamino-5-chloro-1H-indol-2-yl)-2-phenyl-ethyl]-2-guanidino-acetamide; (S)-2-Amino-2-(5-chloro-7-cyclopentylamino-1H-indol-2-yl)-ethanol; ((S)-5-chloro-2-pyrrolidin-2-yl-1H-indol-7-yl)-bis-(3-methyl-butyl)-amine; (2S,4R)-4-benzyloxy-2-(5-chloro-7-cyclopentylamino-1H-indol-2-yl)-pyrrolidin-1-carboxamidine; (S)-2-{7-[bis-(3-methyl-butyl)-amino]-5-chloro-1H-indol-2-yl)-pyrrolidin-1-carboxamidine; (S)-2-(5-chloro-7-cyclopentylamino-1H-indol-2-yl)-pyrrolidin-1-carboxamidine; [2-((2S,4R)-4-benzyloxy-[1,3′]bipyrrolidin-2-yl)-5-chloro-1H-indol-7-yl)-cyclopentyl-amine; ((S)-[1,3′]bipyrrolidin-2-yl-5-chloro-1H-indol-7-yl)-cyclopentyl-amine; {2-[(S)-1-(2-amino-thiazol-4-ylmethyl)-pyrrolidin-2-yl]-5-chloro-1H-indol-7-yl)-cyclopentyl-amine; [5-Chloro-2-((S)-1-cyclopentylmethyl-pyrrolidin-2-yl)-1H-indol-7-yl]-bis-(3-methyl-butyl)-amine; ((S)-[1,3′]bipyrrolidin-2-yl-5-chloro-1H-indol-7-yl)-bis-(3-methyl-butyl)-amine; N-[2-((S)-2-{7-[bis-(3-methyl-butyl)-amino]-5-chloro-1H-indol-2-yl}-pyrrolidin-1-yl)-acetyl]-guanidine; (5-Chloro-2-piperidin-4-yl-1H-indol-7-yl)-cyclopentyl-amine; (5-Chloro-2-piperidin-4-yl-1H-indol-7-yl)-(tetrahydropyran-4-yl)-amine; Cyclopentyl-(5-methyl-2-piperidin-4-yl-1H-indol-7-yl)-amine; 1-[4-(7-Cyclopentylamino-5-methyl-1H-indol-2-yl)-piperidin-1-yl]-ethanone; (5-Methyl-2-piperidin-4-yl-1H-indol-7-yl)-(tetrahydropyran-4-yl)-amine; (5-Methyl-2-piperidin-4-yl-1H-indol-7-yl)-bis-(tetrahydropyran-4-yl)-amine; 1-{4-[5-Methyl-7-(tetrahydropyran-4-ylamino)-1H-indol-2-yl]-piperidin-1-yl]-ethanone; (2-Cyclohexyl-5-methyl-1H-indol-7-yl)-cyclopentyl-amine; [5-(1,1-Dioxo-thiomorpholin-4-ylmethyl)-2-piperidin-4-yl-1H-indol-7-yl]-(tetrahydropyran-4-yl)-amine; and 1-{4-[5-(1,1-Dioxo-thiomorpholin-4-ylmethyl)-7-(tetrahydropyran-4-ylamino)-1H-indol-2-yl]-piperidin-1-yl}-ethanone. 16. A composition for the prevention or treatment of necrosis and necrosis-associated diseases, which comprises a therapeutically effective amount of the compounds of Formula (1), a pharmaceutically acceptable salt or isomer thereof as defined in claim 1 as an active ingredient together with a pharmaceutically acceptable carrier or diluent. 17. The composition according to claim 16, wherein the necrosis and necrosis-associated diseases are selected from the group consisting of acute/chronic hepatic disease, neurodegenerative disease, ischemic disease, diabetes, pancreatitis, bacterial/viral sepsis, necrotizing procolitis, cystic fibrosis, rheumatoid arthritis, degenerative arthritis, nephropathy, bacterial infection, viral infection, multiple sclerosis, leukemia, lymphoma, neonatal respiratory distress syndrome, asphyxia, tuberculosis, endometriosis, angiasthenia, psoriasis, chilblain, steroid treatment complications, gangrene, pressure sores, hemoglobinuria, burns, hyperthermia, Crohn's disease, celiac disease, compartment syndrome, spinal cord injury, glomerulonephritis, muscular dystrophy, metabolic inherited disease, mycoplasmal disease, anthrax, Andersen's disease, congenital mitochondrial disease, phenylketonuria, placental infarction, syphilis, and aseptic necrosis. 18. The composition according to claim 16, wherein the necrosis and necrosis-associated diseases are caused by a drug and toxic substance, and are selected from the group consisting of the necrosis associated with alcoholism, the exposure to, or administration or self-administration of, cocaine, drug, antibiotic, anti-cancer agent, adriamycin, puromycin, bleomycin, NSAID, cyclosporine, chemical toxin, poison gas, agrochemical, heavy metal, or injury due to the exposure to radioactivity/UV and associated necrosis thereof. 19. The composition according to claim 16, which is for hepatoprotection, hepatic functional improvement, and prevention or treatment of hepatic disease. 20. The composition according to claim 19, wherein the hepatic disease is selected from the group consisting of liver transplantation, alcoholic or non-alcoholic fatty liver, hepatic fibrosis, hepatocirrhosis and hepatitis caused by virus or drug. 21. The composition according to claim 19, wherein the hepatic disease is alcoholic acute/chronic hepatic disease. 22. The composition according to claim 19, wherein the hepatic disease is fatty acid-induced fatty liver or acute/chronic hepatic disease derived from fatty liver. 23. The composition according to claim 19, wherein the hepatic disease is mediated by reactive oxygen species (ROS). 24. The composition according to claim 19, wherein the hepatic disease is mediated by heavy metal. 25. The composition according to claim 16, which is co-administered with a prophylactic or therapeutic agent for drug-induced necrosis and necrosis-associated diseases. 26. The composition according to claim 25, wherein the prophylactic or therapeutic agent for drug-induced necrosis and necrosis-associated diseases is selected from the group consisting of antibiotic, anti-cancer agent, anti-viral agent, anti-infective, anti-inflammatory agent, anti-coagulant, lipid-improving agent, cell death inhibitor, anti-hypertensive agent, anti-diabetic/anti-obesity agent, therapeutic agent for cardiovascular disease, therapeutic agent for neurodegenerative disease, anti-aging agent and therapeutic agent for metabolic disease. 27. The composition according to claim 19, which is co-administered with an agent selected from the group consisting of hepatocyte regeneration promoter, hepatic functional adjuvant, anti-viral agent, immunosuppressant and fibrosis inhibitor. 28. The composition according to claim 17, wherein the neurodegenerative disease is dementia, Parkinson's disease or Huntington's disease. 29. The composition according to claim 17, wherein the ischemic disease is cardiac disease, reperfusion injury, ischemic stroke or ischemic injury. 30. The composition according to claim 17, wherein the diabetes is caused by a pancreatic cell-destroying substance, diabetic complications or diabetic vascular disease. 31. The composition according to claim 30, wherein the diabetes is mediated by virus, hyperglycemia, fatty acid, diet, toxin or streptozotocin. 32. A method of preparing a composition for the prevention or treatment of necrosis and necrosis-associated diseases, which comprises the step of mixing the compound of Formula (1), a pharmaceutically acceptable salt or isomer thereof as defined in claim 1 as an active ingredient together with a pharmaceutically acceptable carrier.
1,600
333
15,497,414
1,625
Provided herein are compounds and pharmaceutical compositions of formula I where R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are as described herein. Also provided pharmaceutically acceptable salts or stereoisomers of these compounds. In addition methods are provided for inhibiting the binding of an integrin to treat various pathophysiological conditions.
1: A compound of formula I having a chemical structure of wherein R1 is R2 and R3 are independently hydrogen or C1-4 alkyl; R4 is H or C1-4 alkyl; R5 is phenyl, aryl, heterocyclyl or aralkyl any of which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryloxy, oxo, halogen, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl; R6 is C1-4 alkyl, halogen, phenyl, aryl, or heterocycyl any of which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryl, aryloxy, halogen, oxo, acetyl, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl; R7 is H or C1-4 alkyl; R8, R9 and R10 are independently hydrogen, halogen, C1-4 alkyl, C3-6 cycloalkyl, or hydroxyl; or a pharmaceutically acceptable salt or stereoisomer thereof. 2: The compound of claim 1, wherein R2 and R3 are each hydrogen. 3: The compound of claim 1, wherein R4 is hydrogen; methyl, ethyl or t-butyl. 4: The compound of claim 1, wherein R7 is hydrogen, methyl or ethyl. 5: The compound of claim 1, wherein R8 and R9 are independently hydrogen or methyl. 6: The compound of claim 1, wherein R10 is hydroxyl. 7: The compound of claim 1, wherein the steroisomer is of the (S)-configuration. 8: The compound of claim 1, wherein the compound of formula I is the compound of formula I A having a chemical structure of wherein, R2, R3, R4, R5, R6, R7, R8, R9 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 9: The compound of claim 1, wherein the compound of formula I is the compound of formula I B having a chemical structure of wherein, R2, R3, R4, R5, R6, R7, R9 and R10 are as defined in claim 1; or the pharmaceutically acceptable salt or stereoisomers thereof. 10: The compound of claim 1, wherein the compound of formula I is the compound of formula I C having a chemical structure of wherein, R2, R3, R4, R5, R6, R7 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 11: The compound of claim 1, wherein the compound of formula I is the compound of formula I D having a chemical structure of wherein, R2, R3, R4, R5, R6, R7 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 12: The compound of claim 1, wherein the compound of formula I is the compound of formula IE having a chemical structure of wherein, R2, R3, R4, R5, R6, R7 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 13: The compound of claim 1, wherein the compound of formula I is the compound of formula IF having a chemical structure of wherein, R2, R3, R4, R5, R6, R9 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 14: The compound of claim 1, wherein the pharmaceutically acceptable salt is the mono or disodium sodium salt 15: A pharmaceutical composition, comprising at least one compound of claim 1 and one or more pharmaceutically acceptable carriers. 16: A method for treating a pathophysiological condition mediated by α4 integrins i.e. α4β1, α4β7 or mixed α4β1 and α4β7 integrin in a subject in need of such treatment, comprising: administering to the subject a pharmacologically effective amount of the pharmaceutical composition of claim 15. 17: The method of claim 16, wherein the pathophysiological condition is hematopoietic stem cell transplant therapy, sickle cell disease, dry eye, atherosclerosis, rheumatoid arthritis, asthma, allergy, multiple sclerosis, lupus, inflammatory bowel disease, graft rejection, contact hypersensitivity, stroke, pulmonary arterial hypertension, diabetes or cancer. 18: A method for inhibiting integrin binding in a cell associated with a pathophysiological condition, comprising: contacting the cell with one or more compounds of claim 1. 19: The method of claim 18, wherein the integrin is α4β1 and/or α4β7 integrin. 20: The method of claim 18, wherein the pathophysiological condition is a cancer. 21: A compound of formula I having a chemical structure of wherein R1 is R2 and R3 are independently hydrogen; R4 is hydrogen; methyl, ethyl or t-butyl; R5 is phenyl, aryl, heterocyclyl or aralkyl which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryloxy, oxo, halogen, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl R6 is C1-4 alkyl, halogen, phenyl, aryl or heterocycyl which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryl, aryloxy, halogen, oxo, acetyl, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl R7 is hydrogen; methyl or ethyl R8 and R9 are independently hydrogen or methyl and or a pharmaceutically acceptable salt or stereoisomers thereof. 22: A compound that is: ethyl (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(biphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(2-oxo-3-phenylpyridin-1(2H)-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(2′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoate; ethyl 3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-3-yl)phenyl)propanoate; ethyl (S)-3-(3′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrazin-2-yl)phenyl)propanoate, ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,3′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-oxo-1-phenyl-1,6-dihydropyridin-3-yl)propanoate; ethyl (S)-3-(2′,6′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl 3-(4′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(2′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethyl)biphenyl-3-yl)propanoate; ethyl (S)-3-(5,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoate; ethyl (S)-3-(2′,5′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrimidin-5-yl)phenyl)propanoate; ethyl (S)-3-(2′,6′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylfuran-2-yl)propanoate; ethyl (S)-3-(3-(2,5-dimethyl-1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3′-chloro-4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(2′,3′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5′-methoxy-2′-methylbiphenyl-3-yl)propanoate; ethyl 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(2,2,2-trifluoroethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′-ethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,4′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,3′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4′-fluoro-3′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(1-phenyl-1H-pyrrol-2-yl)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(5′-chloro-2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,3′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoate; ethyl (S)-3-(2′-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl 3-(2′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[b]pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-4-methyl-5-oxo-4,5-dihydrothieno[3,2-b]pyridin-6-yl)ureido)propanoate; ethyl (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-indazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluoro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(3-(1-ethyl-1H-indol-6-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(2′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-(3-methoxyphenyl)thiophen-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(4-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiophen-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3′-chloro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(5-chlorothiophen-2-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(4-methylthiophen-3-yl)phenyl)propanoate; ethyl (S)-3-(2′,4′-difluoro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(4-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-pyrrol-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(6-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(1-methyl-1H-pyrazol-4-yl)phenyl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(5-hydroxy-2-methyl-3-oxo-2,3-dihydropyridazin-4-yl)ureido)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3′-chloro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(furan-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-methoxyphenyl)thiophen-2-yl)propanoate; ethyl (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-phenylpyridin-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethyl biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydro indolizin-6-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-(trifluoromethoxy)phenyl)thiophen-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(3,5-dimethylisoxazol-4-yl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(5-(2,5-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(tri fluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoate; ethyl (S)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(tri fluoromethyl)biphenyl-3-yl)propanoate; ethyl (S)-3-(5-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(3-fluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl 3-(3′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(4′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiazol-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridazin-3-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylpyridin-3-yl)propanoate; (S)-ethyl 3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(biphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6′-methoxypyridin-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-bromophenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate, ethyl 3-(3-bromophenyl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate, ethyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid (S)-3-(biphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(2-oxo-3-phenylpyridin-1(2H)-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(2′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoic acid; 3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-3-yl)phenyl)propanoic acid; (S)-3-(3′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethyl)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,3′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-oxo-1-phenyl-1,6-dihydropyridin-3-yl)propanoic acid; (S)-3-(2′,6′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(4′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(2′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethyl)biphenyl-3-yl)propanoic acid; (S)-3-(5,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoic acid; (S)-3-(2′,5′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrimidin-5-yl)phenyl)propanoic acid; (S)-3-(2′,6′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylfuran-2-yl)propanoic acid; (S)-3-(3-(2,5-dimethyl-1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′-chloro-4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(2′,3′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5′-methoxy-2′-methylbiphenyl-3-yl)propanoic acid; 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(2,22-trifluoroethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′-ethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,4′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,3′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4′-fluoro-3′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(1-phenyl-1H-pyrrol-2-yl)propanoic acid; (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(5′-chloro-2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,3′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoic acid; (S)-3-(2′-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; 3-(2′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[b]pyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-4-methyl-5-oxo-4,5-dihydrothieno[3,2-b]pyridin-6-yl)ureido)propanoic acid; (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-indazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluoro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoic acid; (S)-3-(3-(1-ethyl-1H-indol-6-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(2′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-(3-methoxyphenyl)thiophen-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(4-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiophen-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3′-chloro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-4′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(5-chlorothiophen-2-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(4-methylthiophen-3-yl)phenyl)propanoic acid; (S)-3-(2′,4′-difluoro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoic acid; (S)-3-(4-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-pyrrol-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(6-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(1-methyl-1H-pyrazol-4-yl)phenyl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(5-hydroxy-2-methyl-3-oxo-2,3-dihydropyridazin-4-yl)ureido)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3′-chloro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(furan-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-methoxyphenyl)thiophen-2-yl)propanoic acid; (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoic acid 1; (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-phenylpyridin-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoic acid; (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-(trifluoromethoxy)phenyl)thiophen-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(3,5-dimethylisoxazol-4-yl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoic acid; (S)-3-(5-(2,5-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoic acid; (S)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoromethyl)biphenyl-3-yl)propanoic acid; (S)-3-(5-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(3-fluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl-6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; 3-(3′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(4′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiazol-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridazin-3-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylpyridin-3-yl)propanoic acid; (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(biphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6′-methoxypyridin-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrazin-2-yl)phenyl)propanoic acid, 3-(3-bromophenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido) propanoic acid, 3-(3-bromophenyl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido) propanoic acid, sodium (S)-3-(biphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl) ureido)propanoate; sodium (S)-3-(biphenyl-3-yl)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(2-oxo-3-phenylpyridin-1(2H)-yl)phenyl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoate; sodium 3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,5′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methyl biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-3-yl)phenyl)propanoate; sodium (S)-3-(3′,5′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′,5′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethyl)biphenyl-3-yl)propanoate; sodium (S)-3-(3′,4′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,3′-dimethylbiphenyl-3-yl)propanoate; sodium 3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-oxo-1-phenyl-1,6-dihydropyridin-3-yl)propanoate; sodium (S)-3-(2′,6′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,4′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium 3-(4′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(2′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethyl)biphenyl-3-yl)propanoate; sodium (S)-3-(5,6-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoate; sodium (S)-3-(2′,5′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrimidin-5-yl)phenyl)propanoate; sodium (S)-3-(2′,6′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylfuran-2-yl)propanoate; sodium (S)-3-(3-(2,5-dimethyl-1H-pyrrol-1-yl)phenyl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′-chloro-4′-fluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(2′,3′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′,5′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5′-methoxy-2′-methylbiphenyl-3-yl)propanoate; sodium 3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(2,2,2-trifluoroethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-ethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′,4′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(2′,3′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-fluoro-3′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(1-phenyl-1H-pyrrol-2-yl)propanoate; sodium (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-methyl biphenyl-3-yl)propanoate; sodium (S)-3-(3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(5′-chloro-2′,4′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,3′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(6-methoxypyridin-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium 3-(2′,4′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[b]pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-methyl-7-oxido-5-oxo-4,5-dihydrothieno[3,2-b]pyridin-6-yl)ureido)propanoate; sodium (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-indazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluoro-6-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenyl thiophen-2-yl)propanoate; sodium (S)-3-(3-(1-ethyl-1H-indol-6-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium 3-(2′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(3-methoxyphenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(3-chlorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-ethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiophen-2-yl)phenyl)propanoate; sodium (S)-3-(6-methoxy-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-chloro-6-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methoxy-4′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,6′-dichlorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(5-chlorothiophen-2-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(4-methylthiophen-3-yl)phenyl)propanoate; sodium (S)-3-(2′,4′-difluoro-6-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-methoxy-6-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-(2,6-dimethylphenyl)thiophen-2-yl)propanoate; sodium (S)-3-(3-(1H-pyrrol-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(6-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-1H-pyrazol-4-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(2-methyl-5-oxido-3-oxo-2,3-dihydropyridazin-4-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3′-chloro-6-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(6-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(furan-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(3-methoxyphenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-phenylpyridin-2-yl)propanoate; sodium (S)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-methoxy-5-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(7-oxido-5-oxo-1,2,3,5-tetrahydro indolizin-6-yl)ureido)propanoate; sodium (S)-3-(4′-methyl-2,3′-bithiophen-5-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-2,3′-bithiophen-5-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(4-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-(trifluoromethoxy)phenyl)thiophen-2-yl)propanoate; sodium (S)-3-(4-methoxy-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-fluoro-3′-trifluoromethoxybiphenyl-3-yl)propanoate; sodium (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(3,5-dimethylisoxazol-4-yl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-fluoro-2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoate; sodium (S)-3-(5-(2,5-difluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoro methoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl biphenyl-4-yl)propanoate; sodium (S)-3-(5-methoxybiphenyl-3-yl)-3-(3-(7-oxido-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoro methyl)biphenyl-3-yl)propanoate; sodium (S)-3-(5-(3-chlorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(3-fluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium 3-(3′-acetylbiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium 3-(4′-acetylbiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenyl thiophen-2-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenyl thiophen-2-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-oxido-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiazol-2-yl)phenyl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl biphenyl-4-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6′-dimethylbiphenyl-4-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-2-yl)phenyl)propanoate; sodium (S)-3-(3-(6-methoxypyridazin-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylpyridin-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(biphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6′-methoxypyridin-2-yl)phenyl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrazin-2-yl)phenyl)propanoate, sodium 3-(3-bromophenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl) ureido)propanoate, or sodium 3-(3-bromophenyl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate.
Provided herein are compounds and pharmaceutical compositions of formula I where R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are as described herein. Also provided pharmaceutically acceptable salts or stereoisomers of these compounds. In addition methods are provided for inhibiting the binding of an integrin to treat various pathophysiological conditions.1: A compound of formula I having a chemical structure of wherein R1 is R2 and R3 are independently hydrogen or C1-4 alkyl; R4 is H or C1-4 alkyl; R5 is phenyl, aryl, heterocyclyl or aralkyl any of which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryloxy, oxo, halogen, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl; R6 is C1-4 alkyl, halogen, phenyl, aryl, or heterocycyl any of which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryl, aryloxy, halogen, oxo, acetyl, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl; R7 is H or C1-4 alkyl; R8, R9 and R10 are independently hydrogen, halogen, C1-4 alkyl, C3-6 cycloalkyl, or hydroxyl; or a pharmaceutically acceptable salt or stereoisomer thereof. 2: The compound of claim 1, wherein R2 and R3 are each hydrogen. 3: The compound of claim 1, wherein R4 is hydrogen; methyl, ethyl or t-butyl. 4: The compound of claim 1, wherein R7 is hydrogen, methyl or ethyl. 5: The compound of claim 1, wherein R8 and R9 are independently hydrogen or methyl. 6: The compound of claim 1, wherein R10 is hydroxyl. 7: The compound of claim 1, wherein the steroisomer is of the (S)-configuration. 8: The compound of claim 1, wherein the compound of formula I is the compound of formula I A having a chemical structure of wherein, R2, R3, R4, R5, R6, R7, R8, R9 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 9: The compound of claim 1, wherein the compound of formula I is the compound of formula I B having a chemical structure of wherein, R2, R3, R4, R5, R6, R7, R9 and R10 are as defined in claim 1; or the pharmaceutically acceptable salt or stereoisomers thereof. 10: The compound of claim 1, wherein the compound of formula I is the compound of formula I C having a chemical structure of wherein, R2, R3, R4, R5, R6, R7 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 11: The compound of claim 1, wherein the compound of formula I is the compound of formula I D having a chemical structure of wherein, R2, R3, R4, R5, R6, R7 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 12: The compound of claim 1, wherein the compound of formula I is the compound of formula IE having a chemical structure of wherein, R2, R3, R4, R5, R6, R7 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 13: The compound of claim 1, wherein the compound of formula I is the compound of formula IF having a chemical structure of wherein, R2, R3, R4, R5, R6, R9 and R10 are as defined in claim 1; or a pharmaceutically acceptable salt or stereoisomers thereof. 14: The compound of claim 1, wherein the pharmaceutically acceptable salt is the mono or disodium sodium salt 15: A pharmaceutical composition, comprising at least one compound of claim 1 and one or more pharmaceutically acceptable carriers. 16: A method for treating a pathophysiological condition mediated by α4 integrins i.e. α4β1, α4β7 or mixed α4β1 and α4β7 integrin in a subject in need of such treatment, comprising: administering to the subject a pharmacologically effective amount of the pharmaceutical composition of claim 15. 17: The method of claim 16, wherein the pathophysiological condition is hematopoietic stem cell transplant therapy, sickle cell disease, dry eye, atherosclerosis, rheumatoid arthritis, asthma, allergy, multiple sclerosis, lupus, inflammatory bowel disease, graft rejection, contact hypersensitivity, stroke, pulmonary arterial hypertension, diabetes or cancer. 18: A method for inhibiting integrin binding in a cell associated with a pathophysiological condition, comprising: contacting the cell with one or more compounds of claim 1. 19: The method of claim 18, wherein the integrin is α4β1 and/or α4β7 integrin. 20: The method of claim 18, wherein the pathophysiological condition is a cancer. 21: A compound of formula I having a chemical structure of wherein R1 is R2 and R3 are independently hydrogen; R4 is hydrogen; methyl, ethyl or t-butyl; R5 is phenyl, aryl, heterocyclyl or aralkyl which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryloxy, oxo, halogen, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl R6 is C1-4 alkyl, halogen, phenyl, aryl or heterocycyl which is substituted with one or more of hydrogen, C1-4 alkyl, alkoxy, aryl, aryloxy, halogen, oxo, acetyl, haloalkoxy, —CF3, hydroxyl, —OCF3, aryl, —OCF2H, —OCF2CF2H, —O(C3-6 cycloalkyl), —OCH2CF3, thioalkoxy, dialkylamino, C3-6 cycloalkyl or haloalkyl R7 is hydrogen; methyl or ethyl R8 and R9 are independently hydrogen or methyl and or a pharmaceutically acceptable salt or stereoisomers thereof. 22: A compound that is: ethyl (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(biphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(2-oxo-3-phenylpyridin-1(2H)-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(2′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoate; ethyl 3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-3-yl)phenyl)propanoate; ethyl (S)-3-(3′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrazin-2-yl)phenyl)propanoate, ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,3′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-oxo-1-phenyl-1,6-dihydropyridin-3-yl)propanoate; ethyl (S)-3-(2′,6′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl 3-(4′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(2′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethyl)biphenyl-3-yl)propanoate; ethyl (S)-3-(5,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoate; ethyl (S)-3-(2′,5′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrimidin-5-yl)phenyl)propanoate; ethyl (S)-3-(2′,6′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylfuran-2-yl)propanoate; ethyl (S)-3-(3-(2,5-dimethyl-1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3′-chloro-4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(2′,3′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5′-methoxy-2′-methylbiphenyl-3-yl)propanoate; ethyl 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(2,2,2-trifluoroethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′-ethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,4′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,3′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4′-fluoro-3′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(1-phenyl-1H-pyrrol-2-yl)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(5′-chloro-2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,3′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoate; ethyl (S)-3-(2′-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl 3-(2′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[b]pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-4-methyl-5-oxo-4,5-dihydrothieno[3,2-b]pyridin-6-yl)ureido)propanoate; ethyl (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-indazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluoro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(3-(1-ethyl-1H-indol-6-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(2′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-(3-methoxyphenyl)thiophen-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(4-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiophen-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3′-chloro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(5-chlorothiophen-2-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(4-methylthiophen-3-yl)phenyl)propanoate; ethyl (S)-3-(2′,4′-difluoro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(4-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(1H-pyrrol-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(6-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(1-methyl-1H-pyrazol-4-yl)phenyl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(5-hydroxy-2-methyl-3-oxo-2,3-dihydropyridazin-4-yl)ureido)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3′-chloro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(furan-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-methoxyphenyl)thiophen-2-yl)propanoate; ethyl (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-phenylpyridin-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethyl biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydro indolizin-6-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; ethyl (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-(trifluoromethoxy)phenyl)thiophen-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; ethyl (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(3,5-dimethylisoxazol-4-yl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(5-(2,5-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(tri fluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoate; ethyl (S)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(tri fluoromethyl)biphenyl-3-yl)propanoate; ethyl (S)-3-(5-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(5-(3-fluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl 3-(3′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(4′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiazol-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoate; ethyl (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridazin-3-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylpyridin-3-yl)propanoate; (S)-ethyl 3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; tert-butyl (S)-3-(biphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6′-methoxypyridin-2-yl)phenyl)propanoate; ethyl (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; ethyl (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; ethyl 3-(3-bromophenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate, ethyl 3-(3-bromophenyl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; ethyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate, ethyl (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid (S)-3-(biphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(2-oxo-3-phenylpyridin-1(2H)-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(2′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoic acid; 3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-3-yl)phenyl)propanoic acid; (S)-3-(3′,5′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethyl)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,5′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,3′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-oxo-1-phenyl-1,6-dihydropyridin-3-yl)propanoic acid; (S)-3-(2′,6′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,4′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(4′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(2′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethyl)biphenyl-3-yl)propanoic acid; (S)-3-(5,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoic acid; (S)-3-(2′,5′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrimidin-5-yl)phenyl)propanoic acid; (S)-3-(2′,6′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylfuran-2-yl)propanoic acid; (S)-3-(3-(2,5-dimethyl-1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′-chloro-4′-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(2′,3′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(3′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5′-methoxy-2′-methylbiphenyl-3-yl)propanoic acid; 3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(2,22-trifluoroethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′-ethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,4′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,3′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4′-fluoro-3′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(1-phenyl-1H-pyrrol-2-yl)propanoic acid; (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(5′-chloro-2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,3′-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoic acid; (S)-3-(2′-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; 3-(2′,4′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[b]pyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-4-methyl-5-oxo-4,5-dihydrothieno[3,2-b]pyridin-6-yl)ureido)propanoic acid; (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-indazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluoro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoic acid; (S)-3-(3-(1-ethyl-1H-indol-6-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(2′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-(3-methoxyphenyl)thiophen-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(4-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-ethyl-4-hydroxy-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiophen-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3′-chloro-6-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-4′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(5-chlorothiophen-2-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(4-methylthiophen-3-yl)phenyl)propanoic acid; (S)-3-(2′,4′-difluoro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoic acid; (S)-3-(4-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(1H-pyrrol-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(6-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(1-methyl-1H-pyrazol-4-yl)phenyl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(5-hydroxy-2-methyl-3-oxo-2,3-dihydropyridazin-4-yl)ureido)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3′-chloro-6-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-hydroxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(furan-3-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-methoxyphenyl)thiophen-2-yl)propanoic acid; (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoic acid 1; (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-phenylpyridin-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-[2,3′-bithiophen]-5-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoic acid; (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoic acid; (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-(trifluoromethoxy)phenyl)thiophen-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoic acid; (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(3,5-dimethylisoxazol-4-yl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2-fluorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoic acid; (S)-3-(5-(2,5-difluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoic acid; (S)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)-3-(5-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoromethyl)biphenyl-3-yl)propanoic acid; (S)-3-(5-(3-chlorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(5-(3-fluorophenyl)thiophen-2-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl-6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; 3-(3′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; 3-(4′-acetylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenylthiophen-2-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoic acid; (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-hydroxy-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiazol-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-4-yl)propanoic acid; (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridazin-3-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylpyridin-3-yl)propanoic acid; (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(biphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6′-methoxypyridin-2-yl)phenyl)propanoic acid; (S)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoic acid; (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoic acid; (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrazin-2-yl)phenyl)propanoic acid, 3-(3-bromophenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido) propanoic acid, 3-(3-bromophenyl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido) propanoic acid, sodium (S)-3-(biphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl) ureido)propanoate; sodium (S)-3-(biphenyl-3-yl)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(2-oxo-3-phenylpyridin-1(2H)-yl)phenyl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methylbiphenyl-3-yl)propanoate; sodium 3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,5′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methyl biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-3-yl)phenyl)propanoate; sodium (S)-3-(3′,5′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′,5′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethyl)biphenyl-3-yl)propanoate; sodium (S)-3-(3′,4′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,3′-dimethylbiphenyl-3-yl)propanoate; sodium 3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-oxo-1-phenyl-1,6-dihydropyridin-3-yl)propanoate; sodium (S)-3-(2′,6′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,4′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium 3-(4′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(2′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethyl)biphenyl-3-yl)propanoate; sodium (S)-3-(5,6-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6-methoxypyridin-3-yl)phenyl)propanoate; sodium (S)-3-(2′,5′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrimidin-5-yl)phenyl)propanoate; sodium (S)-3-(2′,6′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1-ethyl-5-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′,4′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylfuran-2-yl)propanoate; sodium (S)-3-(3-(2,5-dimethyl-1H-pyrrol-1-yl)phenyl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′-chloro-4′-fluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(2′,3′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(3′,5′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5′-methoxy-2′-methylbiphenyl-3-yl)propanoate; sodium 3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(2,2,2-trifluoroethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-ethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′,4′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(2′,3′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-fluoro-3′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(1-phenyl-1H-pyrrol-2-yl)propanoate; sodium (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-methyl biphenyl-3-yl)propanoate; sodium (S)-3-(3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium 3-(5′-chloro-2′,4′-difluorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,3′-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-pyrrol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(6-methoxypyridin-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium 3-(2′,4′-dichlorobiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[b]pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(4-methyl-7-oxido-5-oxo-4,5-dihydrothieno[3,2-b]pyridin-6-yl)ureido)propanoate; sodium (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(benzo[d][1,3]dioxol-5-yl)phenyl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-indazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluoro-6-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenyl thiophen-2-yl)propanoate; sodium (S)-3-(3-(1-ethyl-1H-indol-6-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium 3-(2′,5′-dichlorobiphenyl-3-yl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(3-methoxyphenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(3-chlorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(1-ethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiophen-2-yl)phenyl)propanoate; sodium (S)-3-(6-methoxy-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-chloro-6-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methoxy-4′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,6′-dichlorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(5-chlorothiophen-2-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(4-methylthiophen-3-yl)phenyl)propanoate; sodium (S)-3-(2′,4′-difluoro-6-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-methoxy-6-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-(2,6-dimethylphenyl)thiophen-2-yl)propanoate; sodium (S)-3-(3-(1H-pyrrol-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6,6′-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(6-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-1H-pyrazol-4-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(2-methyl-5-oxido-3-oxo-2,3-dihydropyridazin-4-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3′,6-dimethoxybiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3′-chloro-6-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-chlorobiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(6-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(furan-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(2,4-difluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(3-methoxyphenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′,5-dimethoxybiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(4-fluorobiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-phenylpyridin-2-yl)propanoate; sodium (S)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methylbiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-fluoro-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methoxy-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(2′,4′-difluoro-5-methoxybiphenyl-3-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3′-methoxy-5-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,6′-dimethylbiphenyl-3-yl)-3-(3-(7-oxido-5-oxo-1,2,3,5-tetrahydro indolizin-6-yl)ureido)propanoate; sodium (S)-3-(4′-methyl-2,3′-bithiophen-5-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methyl-2,3′-bithiophen-5-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-5-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(2,6-dimethylphenyl)thiophen-2-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(4-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4,4′-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(4-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,5,6′-trimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(3-(trifluoromethoxy)phenyl)thiophen-2-yl)propanoate; sodium (S)-3-(4-methoxy-2′,6′-dimethylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-fluoro-3′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-fluoro-3′-methoxybiphenyl-3-yl)-3-(3-(4-hydroxy-1,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-fluoro-3′-trifluoromethoxybiphenyl-3-yl)propanoate; sodium (S)-3-(5-fluoro-3′-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(3,5-dimethylisoxazol-4-yl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(3,5-dimethylisoxazol-4-yl)phenyl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2-fluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-fluoro-2′-methylbiphenyl-3-yl)propanoate; sodium (S)-3-(6-fluoro-2′-methylbiphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylthiophen-2-yl)propanoate; sodium (S)-3-(5-(2,5-difluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoro methoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl biphenyl-4-yl)propanoate; sodium (S)-3-(5-methoxybiphenyl-3-yl)-3-(3-(7-oxido-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-(trifluoro methyl)biphenyl-3-yl)propanoate; sodium (S)-3-(5-(3-chlorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(5-(3-fluorophenyl)thiophen-2-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-di hydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(6-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium 3-(3′-acetylbiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium 3-(4′-acetylbiphenyl-3-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydro pyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4-phenyl thiophen-2-yl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenyl thiophen-2-yl)propanoate; sodium (S)-3-(2′,4′-difluorobiphenyl-3-yl)-3-(3-(7-oxido-5-oxo-1,2,3,5-tetrahydroindolizin-6-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(thiazol-2-yl)phenyl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-methyl-3′-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,4′,5-trifluorobiphenyl-3-yl)propanoate; sodium (S)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3′-methoxy-6-(trifluoromethoxy)biphenyl-3-yl)propanoate; sodium (S)-3-(2′,6′-dimethylbiphenyl-4-yl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′-methyl biphenyl-4-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(2′,6′-dimethylbiphenyl-4-yl)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyridin-2-yl)phenyl)propanoate; sodium (S)-3-(3-(6-methoxypyridazin-3-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-phenylpyridin-3-yl)propanoate; sodium (S)-3-(3-(1,6-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(5-fluoro-2′,6′-dimethylbiphenyl-3-yl)propanoate; sodium (S)-3-(biphenyl-4-yl)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(6′-methoxypyridin-2-yl)phenyl)propanoate; sodium (S)-3-(3-(1,5-dimethyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(4′-methoxybiphenyl-3-yl)propanoate; sodium (S)-3-(3-(1H-imidazol-1-yl)phenyl)-3-(3-(1-methyl-4-oxido-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate; sodium (S)-3-(3-(4-hydroxy-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)-3-(3-(pyrazin-2-yl)phenyl)propanoate, sodium 3-(3-bromophenyl)-3-(3-(4-hydroxy-1,5-dimethyl-2-oxo-1,2-dihydro pyridin-3-yl) ureido)propanoate, or sodium 3-(3-bromophenyl)-3-(3-(1-ethyl-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl)ureido)propanoate.
1,600
334
12,903,521
1,632
The present disclosure provides methods of generating germ layers from stem cells comprising culturing the stem cells in a culture medium having an osmolality less than 340 mOsm/kg. The present disclosure also includes a method to generate different cell lineages from the germ layers as well as to detect them by immunological methods. The present disclosure further provides methods for the generation, isolation, cultivation and propagation of committed progenitor cells and for the production of differentiated cells from the three germ layers. The present disclosure also provides culture media for use in inducing the three germ layers.
1. A method of generating a population of germ layer progenitor cells comprising: a) dissociating pluripotent stem cells into clusters or single cells; b) culturing the dissociated cells from a) in culture media with an osmolality of 260 to 340 mOsm/kg; and c) dissociating the cells of b) and plating the cells onto coated culture dishes and culturing for at least 1 day in the culture media to produce germ layer progenitor cells. 2. The method of claim 1, wherein b) comprises i) culturing the dissociated cells from a) in a microwell device for about 24 hours to form aggregates and continuing the culture in the microwell device for more than 24 hours in the culture media followed by releasing the aggregates and adhering onto coated culture dishes and culturing in the culture media for at least 1 day; ii) culturing the dissociated cells from a) in the culture media in a microwell device for about 24 hours to form aggregates, releasing the aggregates from the microwell device, followed by culturing the released aggregates in suspension in the culture media for at least 1 day, dissociating and adhering the aggregates onto coated culture dishes and culturing in the culture media for at least 1 day; or iii) culturing the dissociated cells from a) in suspension in the culture media for at least 1 day followed by dissociating the cells and adhering onto coated culture dishes and culturing in the culture media for at least 1 day. 3. The method of claim 2, wherein the aggregates comprise embryoid bodies. 4. The method of claim 3, wherein the embryoid bodies comprise 500 to 20,000 cells. 5. The method of claim 1, wherein b) comprises adhering the dissociated cells from a) onto coated culture dishes or feeders and culturing for at least 3 days in the culture media. 6. The method of claim 1, wherein the culture media comprises Dulbecco's minimal essential medium and further comprises vitamins, trace elements, selenium, insulin, lipids, β-mercaptoethanol, non-essential amino acids, antibiotics, bFGF, B27, N2 or mixtures thereof. 7. The method of claim 1, wherein the pluripotent stem cells are mammalian pluripotent stem cells. 8. The method of claim 7, wherein the pluripotent stem cells are human pluripotent stem cells. 9. The method of claim 1, wherein the pluripotent stem cells are induced pluripotent stem cells or embryonic stem cells. 10. The method of claim 1, wherein the germ layer is ectodermal, endodermal and/or mesodermal. 11. The method of claim 2, wherein the osmolality of the culture media is 260 to 280 mOsm/kg for inducing ectodermal progenitor cells. 12. The method of claim 5, wherein the osmolality of the culture media is 270 to 320 mOsm/kg for inducing ectodermal progenitor cells. 13. A method of maintaining single neural progenitor cells for at least 3 passages in culture media with an osmolality of 260-340 mOsm/kg comprising generating ectodermal progenitor cells according to the method of claim 11; dissociating the ectodermal progenitor cells from the adhered cultures; plating and culturing said progenitor cells in the culture media for at least 1 day. 14. The method of claim 13, wherein the cells are dissociated with a buffered solution comprising a Ca2+ and Mg2+ free buffer solution comprising 1×PBS or 1×Hank's Buffered Salt Solution with a pH range of pH 7.0 to pH 8.0. 15. The method of claim 13, wherein the neural progenitor cells are further differentiated to form neurons, astrocytes and/or oligodendrocytes. 16. The method of claim 2, wherein the osmolality of the culture media is 290-340 mOsm/kg for inducing endodermal and/or mesodermal progenitor cells. 17. The method of claim 5, wherein the osmolality of the culture media is 320-340 mOsm/kg for inducing endodermal and/or mesodermal progenitor cells. 18. A method of maintaining single mesodermal and/or endodermal progenitor cells for at least 3 passages in culture media with an osmolality of 320-340 mOsm/kg comprising generating mesodermal and/or endodermal progenitor cells according to the method of claim 16; dissociating the mesodermal and/or endodermal progenitor cells from adherent cultures; and plating and culturing said progenitor cells. 19. The method of claim 18, wherein the mesodermal and endodermal progenitor cells are further differentiated to form mesenchymal stem cells, chondrocytes, cardiomyocytes, hematopoietic stem cells, skeletal muscle cells, pancreatic cells or liver cells. 20. A method of screening germ layer cells comprising (a) preparing a culture of ectodermal, endodermal or mesodermal germ layer cells according to the method of claim 1; (b) treating the germ layer cells with a test agent or agents; and (c) subjecting the treated germ layer cells to analysis.
The present disclosure provides methods of generating germ layers from stem cells comprising culturing the stem cells in a culture medium having an osmolality less than 340 mOsm/kg. The present disclosure also includes a method to generate different cell lineages from the germ layers as well as to detect them by immunological methods. The present disclosure further provides methods for the generation, isolation, cultivation and propagation of committed progenitor cells and for the production of differentiated cells from the three germ layers. The present disclosure also provides culture media for use in inducing the three germ layers.1. A method of generating a population of germ layer progenitor cells comprising: a) dissociating pluripotent stem cells into clusters or single cells; b) culturing the dissociated cells from a) in culture media with an osmolality of 260 to 340 mOsm/kg; and c) dissociating the cells of b) and plating the cells onto coated culture dishes and culturing for at least 1 day in the culture media to produce germ layer progenitor cells. 2. The method of claim 1, wherein b) comprises i) culturing the dissociated cells from a) in a microwell device for about 24 hours to form aggregates and continuing the culture in the microwell device for more than 24 hours in the culture media followed by releasing the aggregates and adhering onto coated culture dishes and culturing in the culture media for at least 1 day; ii) culturing the dissociated cells from a) in the culture media in a microwell device for about 24 hours to form aggregates, releasing the aggregates from the microwell device, followed by culturing the released aggregates in suspension in the culture media for at least 1 day, dissociating and adhering the aggregates onto coated culture dishes and culturing in the culture media for at least 1 day; or iii) culturing the dissociated cells from a) in suspension in the culture media for at least 1 day followed by dissociating the cells and adhering onto coated culture dishes and culturing in the culture media for at least 1 day. 3. The method of claim 2, wherein the aggregates comprise embryoid bodies. 4. The method of claim 3, wherein the embryoid bodies comprise 500 to 20,000 cells. 5. The method of claim 1, wherein b) comprises adhering the dissociated cells from a) onto coated culture dishes or feeders and culturing for at least 3 days in the culture media. 6. The method of claim 1, wherein the culture media comprises Dulbecco's minimal essential medium and further comprises vitamins, trace elements, selenium, insulin, lipids, β-mercaptoethanol, non-essential amino acids, antibiotics, bFGF, B27, N2 or mixtures thereof. 7. The method of claim 1, wherein the pluripotent stem cells are mammalian pluripotent stem cells. 8. The method of claim 7, wherein the pluripotent stem cells are human pluripotent stem cells. 9. The method of claim 1, wherein the pluripotent stem cells are induced pluripotent stem cells or embryonic stem cells. 10. The method of claim 1, wherein the germ layer is ectodermal, endodermal and/or mesodermal. 11. The method of claim 2, wherein the osmolality of the culture media is 260 to 280 mOsm/kg for inducing ectodermal progenitor cells. 12. The method of claim 5, wherein the osmolality of the culture media is 270 to 320 mOsm/kg for inducing ectodermal progenitor cells. 13. A method of maintaining single neural progenitor cells for at least 3 passages in culture media with an osmolality of 260-340 mOsm/kg comprising generating ectodermal progenitor cells according to the method of claim 11; dissociating the ectodermal progenitor cells from the adhered cultures; plating and culturing said progenitor cells in the culture media for at least 1 day. 14. The method of claim 13, wherein the cells are dissociated with a buffered solution comprising a Ca2+ and Mg2+ free buffer solution comprising 1×PBS or 1×Hank's Buffered Salt Solution with a pH range of pH 7.0 to pH 8.0. 15. The method of claim 13, wherein the neural progenitor cells are further differentiated to form neurons, astrocytes and/or oligodendrocytes. 16. The method of claim 2, wherein the osmolality of the culture media is 290-340 mOsm/kg for inducing endodermal and/or mesodermal progenitor cells. 17. The method of claim 5, wherein the osmolality of the culture media is 320-340 mOsm/kg for inducing endodermal and/or mesodermal progenitor cells. 18. A method of maintaining single mesodermal and/or endodermal progenitor cells for at least 3 passages in culture media with an osmolality of 320-340 mOsm/kg comprising generating mesodermal and/or endodermal progenitor cells according to the method of claim 16; dissociating the mesodermal and/or endodermal progenitor cells from adherent cultures; and plating and culturing said progenitor cells. 19. The method of claim 18, wherein the mesodermal and endodermal progenitor cells are further differentiated to form mesenchymal stem cells, chondrocytes, cardiomyocytes, hematopoietic stem cells, skeletal muscle cells, pancreatic cells or liver cells. 20. A method of screening germ layer cells comprising (a) preparing a culture of ectodermal, endodermal or mesodermal germ layer cells according to the method of claim 1; (b) treating the germ layer cells with a test agent or agents; and (c) subjecting the treated germ layer cells to analysis.
1,600
335
14,334,462
1,634
A system for fast DNA sequencing by amplification of genetic material within microreactors, denaturing, demulsifying, and then sequencing the material, while retaining it in a PCR/sequencing zone by a magnetic field. One embodiment includes sequencing nucleic acids on a microchip that includes a microchannel flow channel in the microchip. The nucleic acids are isolated and hybridized to magnetic nanoparticles or to magnetic polystyrene-coated beads. Microreactor droplets are formed in the microchannel flow channel. The microreactor droplets containing the nucleic acids and the magnetic nanoparticles are retained in a magnetic trap in the microchannel flow channel and sequenced.
1. A method of sequencing nucleic acids on a microchip, comprising the steps of: providing a microchannel flow channel in the microchip; isolating the nucleic acids; hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads; forming microreactors in said microchannel flow channel, said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads; positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel, and sequencing the nucleic acids. 2. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads comprises hybridizing the nucleic acids to magnetic nanoparticles. 3. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads comprises hybridizing the nucleic acids to magnetic-cored optically discrete nanoparticles. 4. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads comprises hybridizing the nucleic acids to magnetic polystyrene-coated beads. 5. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of sequencing the nucleic acids comprises pyrosequencing the nucleic acids. 6. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of sequencing the nucleic acids comprises four-color sequencing the nucleic acids by synthesis. 7. The method of sequencing nucleic acids on a microchip of claim 1 wherein the nucleic acids being analyzed include virions, capsids, or individual cells containing the nucleic acids and wherein said step of isolating the nucleic acids comprises lysing the virions, capsids, or individual cells containing the nucleic acids by acoustic wave lysing, or by heating, or by optical heating, or by chemical lysing, or by laser-induced lysing, followed by hybridizing to the magnetic beads. 8. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel comprises moving the nucleic acids in a hydrophobic carrier fluid to said magnetic trap. 9. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel comprises forcing the nucleic acid into a water-oil emulsification hydrophobic carrier fluid and moving the nucleic acid in said water-oil emulsification hydrophobic carrier fluid to said magnetic trap. 10. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel comprises forcing the nucleic acid into liquid slugs in a carrier fluid and moving the nucleic acid in said liquid slugs to said magnetic trap. 11. The method of sequencing nucleic acids on a microchip of claim 1 including the step of flowing reagent over the nucleic acids in said magnetic trap. 12. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of providing a microchannel flow channel in the microchip comprises providing a microchannel flow channel cross section aspect ratio, width and depth, that prevents said magnetic nanoparticles or magnetic polystyrene-coated beads from vertical stacking. 13. The method of sequencing nucleic acids on a microchip of claim 1 including the step of placing a cover over said microchip during said steps of isolating the nucleic acids; hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads; positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel, and sequencing the nucleic acids. 14. A method of fast DNA sequencing by amplification of genetic material within microreactors, comprising the steps of: providing a microchannel flow channel, hybridizing the genetic material to magnetic nanoparticles or to magnetic polystyrene-coated beads, forming the microreactors in said microchannel flow channel, and retaining the genetic material in a PCR/Sequencing Zone by a magnetic field. 15. The method of fast DNA sequencing by amplification of genetic material within microreactors of claim 14 wherein the method encounters reaction products or excess reagents, including the step of holding the genetic material in said PCR/Sequencing Zone by said magnetic field and washing away the reaction products or the excess reagents or removing the reaction products or the excess reagents by chemical degradation. 16. The method of fast DNA sequencing by amplification of genetic material within microreactors of claim 14 including the step of turning off the magnetic field permitting the genetic material to be flushed from said PCR/Sequencing Zone to waste or to archival storage.
A system for fast DNA sequencing by amplification of genetic material within microreactors, denaturing, demulsifying, and then sequencing the material, while retaining it in a PCR/sequencing zone by a magnetic field. One embodiment includes sequencing nucleic acids on a microchip that includes a microchannel flow channel in the microchip. The nucleic acids are isolated and hybridized to magnetic nanoparticles or to magnetic polystyrene-coated beads. Microreactor droplets are formed in the microchannel flow channel. The microreactor droplets containing the nucleic acids and the magnetic nanoparticles are retained in a magnetic trap in the microchannel flow channel and sequenced.1. A method of sequencing nucleic acids on a microchip, comprising the steps of: providing a microchannel flow channel in the microchip; isolating the nucleic acids; hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads; forming microreactors in said microchannel flow channel, said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads; positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel, and sequencing the nucleic acids. 2. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads comprises hybridizing the nucleic acids to magnetic nanoparticles. 3. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads comprises hybridizing the nucleic acids to magnetic-cored optically discrete nanoparticles. 4. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads comprises hybridizing the nucleic acids to magnetic polystyrene-coated beads. 5. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of sequencing the nucleic acids comprises pyrosequencing the nucleic acids. 6. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of sequencing the nucleic acids comprises four-color sequencing the nucleic acids by synthesis. 7. The method of sequencing nucleic acids on a microchip of claim 1 wherein the nucleic acids being analyzed include virions, capsids, or individual cells containing the nucleic acids and wherein said step of isolating the nucleic acids comprises lysing the virions, capsids, or individual cells containing the nucleic acids by acoustic wave lysing, or by heating, or by optical heating, or by chemical lysing, or by laser-induced lysing, followed by hybridizing to the magnetic beads. 8. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel comprises moving the nucleic acids in a hydrophobic carrier fluid to said magnetic trap. 9. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel comprises forcing the nucleic acid into a water-oil emulsification hydrophobic carrier fluid and moving the nucleic acid in said water-oil emulsification hydrophobic carrier fluid to said magnetic trap. 10. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel comprises forcing the nucleic acid into liquid slugs in a carrier fluid and moving the nucleic acid in said liquid slugs to said magnetic trap. 11. The method of sequencing nucleic acids on a microchip of claim 1 including the step of flowing reagent over the nucleic acids in said magnetic trap. 12. The method of sequencing nucleic acids on a microchip of claim 1 wherein said step of providing a microchannel flow channel in the microchip comprises providing a microchannel flow channel cross section aspect ratio, width and depth, that prevents said magnetic nanoparticles or magnetic polystyrene-coated beads from vertical stacking. 13. The method of sequencing nucleic acids on a microchip of claim 1 including the step of placing a cover over said microchip during said steps of isolating the nucleic acids; hybridizing the nucleic acids to magnetic nanoparticles or to magnetic polystyrene-coated beads; positioning said microreactors containing the nucleic acids and said magnetic nanoparticles or magnetic polystyrene-coated beads in a magnetic trap in said microchannel flow channel, and sequencing the nucleic acids. 14. A method of fast DNA sequencing by amplification of genetic material within microreactors, comprising the steps of: providing a microchannel flow channel, hybridizing the genetic material to magnetic nanoparticles or to magnetic polystyrene-coated beads, forming the microreactors in said microchannel flow channel, and retaining the genetic material in a PCR/Sequencing Zone by a magnetic field. 15. The method of fast DNA sequencing by amplification of genetic material within microreactors of claim 14 wherein the method encounters reaction products or excess reagents, including the step of holding the genetic material in said PCR/Sequencing Zone by said magnetic field and washing away the reaction products or the excess reagents or removing the reaction products or the excess reagents by chemical degradation. 16. The method of fast DNA sequencing by amplification of genetic material within microreactors of claim 14 including the step of turning off the magnetic field permitting the genetic material to be flushed from said PCR/Sequencing Zone to waste or to archival storage.
1,600
336
13,582,372
1,657
The invention relates to the determination of resistances of microorganisms which produce β-lactamases, in particular “extended spectrum β-lactamases” (ESBL). The invention provides a method whereby the microbial resistance can be measured very simply and quickly by means of the catalytic effect of the microbially produced β-lactamases on β-lactam antibiotics, which consists in a hydrolytic cleavage of the β-lactam ring. The method determines the resistance of the bacteria a few hours after a suitable substrate, either a β-lactam antibiotic or a customized β-lactam derivative, has been added to a suspension of the microbes, by direct mass spectrometric measurement of the substrate breakdown caused by the β-lactamases.
1. Method for the determination of a β-lactam resistance of microbes based on the production of β-lactamases by the microbes, wherein the microbes are brought together with a substrate and the enzymatic breakdown of the substrate by the β-lactamases of the microbes is measured mass spectrometrically by acquiring a mass spectrum of the remaining substrate and the breakdown product. 2. Method according to claim 1, wherein the molecules of the substrate comprises a β-lactam ring. 3. Method according to claim 2, wherein the substrate comprises a β-lactam antibiotic or a β-lactam derivative. 4. Method according to claim 1, wherein the substrate has a molecular weight between approximately 700 and 1200 atomic mass units. 5. Method according to claim 1, wherein the substrate has only a weak antibiotic effect. 6. Method according to claim 1, wherein the molecules of the substrate have an anchor group which can be used to extract them from solutions. 7. Method according to claim 6, wherein the anchor group is a biotin group or a 6-His tag. 8. Method according to claim 1, wherein the breakdown of several types of substrate is measured simultaneously. 9. Method according to claim 8, wherein the different types of substrate are customized such that their breakdown pattern makes it possible to identify the different classes of β-lactamases. 10. Method according to claim 9, wherein the different substrates around the β-lactam ring imitate the steric forms of different antibiotics. 11. Method according to claim 1, wherein the reaction speeds of the breakdown of the substrates is measured. 12. Method according to claims 1, wherein the microbes have been obtained from blood or a blood culture. 13. Method according to claim 1, wherein the quantities of the remaining substrate and its breakdown product are measured mass spectrometrically with ionization by matrix-assisted laser desorption. 14. Method for the determination of a β-lactam resistance of microbes based on the production of β-lactamases by the microbes comprising the steps: (a) Adding the microbes to a solution of at least one substrate which can be broken down by the β-lactamases, (b) Incubating the solution at a specified temperature for a specified time, (c) Separating the solution with the remaining substrate and its breakdown product from the microbes, and (d) Acquiring a mass spectrum of the solution. 15. Pack of consumables (kit) for the mass spectrometric determination of a microbial β-lactam resistance based on the production of β-lactamases by the microbes, wherein the pack provides substrates which can by enzymatically broken down by the microbial β-lactamases and a matrix substance for ionization by matrix-assisted laser desorption. 16. Program for the evaluation of the mass spectra which have been acquired with the method according to claim 1.
The invention relates to the determination of resistances of microorganisms which produce β-lactamases, in particular “extended spectrum β-lactamases” (ESBL). The invention provides a method whereby the microbial resistance can be measured very simply and quickly by means of the catalytic effect of the microbially produced β-lactamases on β-lactam antibiotics, which consists in a hydrolytic cleavage of the β-lactam ring. The method determines the resistance of the bacteria a few hours after a suitable substrate, either a β-lactam antibiotic or a customized β-lactam derivative, has been added to a suspension of the microbes, by direct mass spectrometric measurement of the substrate breakdown caused by the β-lactamases.1. Method for the determination of a β-lactam resistance of microbes based on the production of β-lactamases by the microbes, wherein the microbes are brought together with a substrate and the enzymatic breakdown of the substrate by the β-lactamases of the microbes is measured mass spectrometrically by acquiring a mass spectrum of the remaining substrate and the breakdown product. 2. Method according to claim 1, wherein the molecules of the substrate comprises a β-lactam ring. 3. Method according to claim 2, wherein the substrate comprises a β-lactam antibiotic or a β-lactam derivative. 4. Method according to claim 1, wherein the substrate has a molecular weight between approximately 700 and 1200 atomic mass units. 5. Method according to claim 1, wherein the substrate has only a weak antibiotic effect. 6. Method according to claim 1, wherein the molecules of the substrate have an anchor group which can be used to extract them from solutions. 7. Method according to claim 6, wherein the anchor group is a biotin group or a 6-His tag. 8. Method according to claim 1, wherein the breakdown of several types of substrate is measured simultaneously. 9. Method according to claim 8, wherein the different types of substrate are customized such that their breakdown pattern makes it possible to identify the different classes of β-lactamases. 10. Method according to claim 9, wherein the different substrates around the β-lactam ring imitate the steric forms of different antibiotics. 11. Method according to claim 1, wherein the reaction speeds of the breakdown of the substrates is measured. 12. Method according to claims 1, wherein the microbes have been obtained from blood or a blood culture. 13. Method according to claim 1, wherein the quantities of the remaining substrate and its breakdown product are measured mass spectrometrically with ionization by matrix-assisted laser desorption. 14. Method for the determination of a β-lactam resistance of microbes based on the production of β-lactamases by the microbes comprising the steps: (a) Adding the microbes to a solution of at least one substrate which can be broken down by the β-lactamases, (b) Incubating the solution at a specified temperature for a specified time, (c) Separating the solution with the remaining substrate and its breakdown product from the microbes, and (d) Acquiring a mass spectrum of the solution. 15. Pack of consumables (kit) for the mass spectrometric determination of a microbial β-lactam resistance based on the production of β-lactamases by the microbes, wherein the pack provides substrates which can by enzymatically broken down by the microbial β-lactamases and a matrix substance for ionization by matrix-assisted laser desorption. 16. Program for the evaluation of the mass spectra which have been acquired with the method according to claim 1.
1,600
337
15,067,901
1,618
A method of magnetic resonance imaging a target tissue in a subject includes a step of administering a first Eu 2+ -containing complex to the subject. The first Eu 2+ -containing complex has a reduction potential that is more negative than a reduction potential for a selected compound present in the target tissue. A first set of images of the target tissue in the subject is then taken by T 1 -weighted magnetic resonance imaging.
1. A method of magnetic resonance imaging a target tissue in a subject, the method comprising: a) administering a first Eu2+-containing complex to the subject, the first Eu2+-containing complex having a reduction potential that is more negative than a reduction potential for a selected compound present in the target tissue; and b) taking a first set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging. 2. The method of claim 1 wherein the first Eu2+-containing complex includes a europium metal ion (Eu2+) and a multi-dentate ligand selected from the group consisting of cryptands and thiacryptands and if necessary counter-ions to maintain charge neutrality. 3. The method of claim 2 wherein the multi-dentate ligand is described by formula I: wherein: Y1, Y2, Y3, Y4, Y5, and Y6 are each independently O or S; R1, R2, R3 are each independently H, C1-12 alkyl, C1-12 alkynyl, C1-12 alkenyl, C1-12 fluoroalkyl, Cl, F, Br, nitro, cyano, or C6-14 aryl, C5-14 hetereoaryl, or 5 and 6 membered rings formed by combining R1 on adjacent carbon atoms or R2 and R3 on adjacent carbon atoms, ═O by combining R1, R2, or R3 on the same carbon atom, ═S by combining R1, R2, or R3 on the same carbon atom, or ═NR by combining R1, R2, or R3 on the same carbon atom; and R is H or C1-12 alkyl. 4. The method of claim 2 wherein R1, R2, or R3 are each independently H, phenyl, or biphenyl. In some refinements, R2 and R3 are hydrogen and one of the R1 is not hydrogen. In other refinements, R2 and R3 are hydrogen and two of the R1 are not hydrogen. 5. The method of claim 1 further comprising c) administering a second Eu2+-containing complex to the subject, the second Eu2+-containing complex having a reduction potential that is more positive than a reduction potential for the selected compound present; and d) taking a second set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging. 6. The method of claim 5 further comprising comparing the first set of images and the second set of images to identify regions that possess different concentrations of the selected compound. 7. The method of claim 5 wherein the first Eu2+-containing complex and the second Eu2+-containing complex to the subject are administered by intravenous, intraperitoneal, or subcutaneous injection. 8. The method of claim 5 wherein the second Eu2+-containing complex includes a europium metal ion (Eu2+) and a multi-dentate ligand selected from the group consisting of cryptands and thiacryptands and if necessary counter-ions to maintain charge neutrality. 9. The method of claim 8 wherein the multi-dentate ligand is described by formula I: wherein: Y1, Y2, Y3, Y4, Y5, and Y6 are each independently O or S; R1, R2, R3 are each independently H, C1-12 alkyl, C1-12 alkynyl, C1-12 alkenyl, C1-12 fluoroalkyl, Cl, F, Br, nitro, cyano, or C6-14 aryl, C5-14 hetereoaryl, or 5 and 6 membered rings formed by combining R1 on adjacent carbon atoms or R2 and R3 on adjacent carbon atoms, ═O by combining R1, R2, or R3 on the same carbon atom, ═S by combining R1, R2, or R3 on the same carbon atom, or ═NR by combining R1, R2, or R3 on the same carbon atom; and R is H or C1-12 alkyl. 10. The method of claim 1 wherein the target tissue is necrotic tissue. 11. A method of magnetic resonance imaging a target tissue in a subject, the method comprising: a) administering a first Eu2+-containing complex to the subject, the first Eu2+-containing complex having a reduction potential that is more negative than a reduction potential for a selected compound present in the target tissue; b) taking a first set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging; c) administering a second Eu2+-containing complex to the subject, the second Eu2+-containing complex having a reduction potential that is more positive than a reduction potential for the selected compound present; d) taking a second set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging; and e) comparing the first set of images and the second set of images to identify regions that possess different concentrations of the selected compound. 12. The method of claim 11 wherein the first Eu2+-containing complex and the second Eu2+-containing complex each independently include a europium metal ion (Eu2+) and a multi-dentate ligand selected from the group consisting of cryptands and thiacryptands and if necessary counter-ions to maintain charge neutrality. 13. The method of claim 12 wherein the multi-dentate ligand is described by formula I: wherein: Y1, Y2, Y3, Y4, Y5, and Y6 are each independently O or S; R1, R2, R3 are each independently H, C1-12 alkyl, C1-12 alkynyl, C1-12 alkenyl, C1-12 fluoroalkyl, Cl, F, Br, nitro, cyano, or C6-14 aryl, C5-14 hetereoaryl, or 5 and 6 membered rings formed by combining R1 on adjacent carbon atoms or R2 and R3 on adjacent carbon atoms, ═O by combining R1, R2, or R3 on the same carbon atom, ═S by combining R1, R2, or R3 on the same carbon atom, or ═NR by combining R1, R2, or R3 on the same carbon atom; and R is H or C1-12 alkyl. 14. The method of claim 13 wherein R1, R2, or R3 are each independently H, phenyl, or biphenyl. In some refinements, R2 and R3 are hydrogen and one of the R1 is not hydrogen. In other refinements, R2 and R3 are hydrogen and two of the R1 are not hydrogen. 15. The method of claim 11 wherein the a first Eu2+-containing complex and the second Eu2+-containing complex to the subject are administered by intravenous, intraperitoneal, or subcutaneous injection. 16. The method of claim 11 wherein the target tissue is necrotic tissue.
A method of magnetic resonance imaging a target tissue in a subject includes a step of administering a first Eu 2+ -containing complex to the subject. The first Eu 2+ -containing complex has a reduction potential that is more negative than a reduction potential for a selected compound present in the target tissue. A first set of images of the target tissue in the subject is then taken by T 1 -weighted magnetic resonance imaging.1. A method of magnetic resonance imaging a target tissue in a subject, the method comprising: a) administering a first Eu2+-containing complex to the subject, the first Eu2+-containing complex having a reduction potential that is more negative than a reduction potential for a selected compound present in the target tissue; and b) taking a first set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging. 2. The method of claim 1 wherein the first Eu2+-containing complex includes a europium metal ion (Eu2+) and a multi-dentate ligand selected from the group consisting of cryptands and thiacryptands and if necessary counter-ions to maintain charge neutrality. 3. The method of claim 2 wherein the multi-dentate ligand is described by formula I: wherein: Y1, Y2, Y3, Y4, Y5, and Y6 are each independently O or S; R1, R2, R3 are each independently H, C1-12 alkyl, C1-12 alkynyl, C1-12 alkenyl, C1-12 fluoroalkyl, Cl, F, Br, nitro, cyano, or C6-14 aryl, C5-14 hetereoaryl, or 5 and 6 membered rings formed by combining R1 on adjacent carbon atoms or R2 and R3 on adjacent carbon atoms, ═O by combining R1, R2, or R3 on the same carbon atom, ═S by combining R1, R2, or R3 on the same carbon atom, or ═NR by combining R1, R2, or R3 on the same carbon atom; and R is H or C1-12 alkyl. 4. The method of claim 2 wherein R1, R2, or R3 are each independently H, phenyl, or biphenyl. In some refinements, R2 and R3 are hydrogen and one of the R1 is not hydrogen. In other refinements, R2 and R3 are hydrogen and two of the R1 are not hydrogen. 5. The method of claim 1 further comprising c) administering a second Eu2+-containing complex to the subject, the second Eu2+-containing complex having a reduction potential that is more positive than a reduction potential for the selected compound present; and d) taking a second set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging. 6. The method of claim 5 further comprising comparing the first set of images and the second set of images to identify regions that possess different concentrations of the selected compound. 7. The method of claim 5 wherein the first Eu2+-containing complex and the second Eu2+-containing complex to the subject are administered by intravenous, intraperitoneal, or subcutaneous injection. 8. The method of claim 5 wherein the second Eu2+-containing complex includes a europium metal ion (Eu2+) and a multi-dentate ligand selected from the group consisting of cryptands and thiacryptands and if necessary counter-ions to maintain charge neutrality. 9. The method of claim 8 wherein the multi-dentate ligand is described by formula I: wherein: Y1, Y2, Y3, Y4, Y5, and Y6 are each independently O or S; R1, R2, R3 are each independently H, C1-12 alkyl, C1-12 alkynyl, C1-12 alkenyl, C1-12 fluoroalkyl, Cl, F, Br, nitro, cyano, or C6-14 aryl, C5-14 hetereoaryl, or 5 and 6 membered rings formed by combining R1 on adjacent carbon atoms or R2 and R3 on adjacent carbon atoms, ═O by combining R1, R2, or R3 on the same carbon atom, ═S by combining R1, R2, or R3 on the same carbon atom, or ═NR by combining R1, R2, or R3 on the same carbon atom; and R is H or C1-12 alkyl. 10. The method of claim 1 wherein the target tissue is necrotic tissue. 11. A method of magnetic resonance imaging a target tissue in a subject, the method comprising: a) administering a first Eu2+-containing complex to the subject, the first Eu2+-containing complex having a reduction potential that is more negative than a reduction potential for a selected compound present in the target tissue; b) taking a first set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging; c) administering a second Eu2+-containing complex to the subject, the second Eu2+-containing complex having a reduction potential that is more positive than a reduction potential for the selected compound present; d) taking a second set of images of the target tissue in the subject by T1-weighted magnetic resonance imaging; and e) comparing the first set of images and the second set of images to identify regions that possess different concentrations of the selected compound. 12. The method of claim 11 wherein the first Eu2+-containing complex and the second Eu2+-containing complex each independently include a europium metal ion (Eu2+) and a multi-dentate ligand selected from the group consisting of cryptands and thiacryptands and if necessary counter-ions to maintain charge neutrality. 13. The method of claim 12 wherein the multi-dentate ligand is described by formula I: wherein: Y1, Y2, Y3, Y4, Y5, and Y6 are each independently O or S; R1, R2, R3 are each independently H, C1-12 alkyl, C1-12 alkynyl, C1-12 alkenyl, C1-12 fluoroalkyl, Cl, F, Br, nitro, cyano, or C6-14 aryl, C5-14 hetereoaryl, or 5 and 6 membered rings formed by combining R1 on adjacent carbon atoms or R2 and R3 on adjacent carbon atoms, ═O by combining R1, R2, or R3 on the same carbon atom, ═S by combining R1, R2, or R3 on the same carbon atom, or ═NR by combining R1, R2, or R3 on the same carbon atom; and R is H or C1-12 alkyl. 14. The method of claim 13 wherein R1, R2, or R3 are each independently H, phenyl, or biphenyl. In some refinements, R2 and R3 are hydrogen and one of the R1 is not hydrogen. In other refinements, R2 and R3 are hydrogen and two of the R1 are not hydrogen. 15. The method of claim 11 wherein the a first Eu2+-containing complex and the second Eu2+-containing complex to the subject are administered by intravenous, intraperitoneal, or subcutaneous injection. 16. The method of claim 11 wherein the target tissue is necrotic tissue.
1,600
338
14,310,568
1,616
A composition directed to comprising from about 25% to about 60% a pyrithione or polyvalent metal salt of a pyrithione; from about 0.01 to about 1% of a cationic polymer; from about 0.01% to about 2.0% of an anionic surfactant wherein the cationic polymer has a molecular weight from about 100,000 to about 2,000,000.
1. A composition comprising: a) from about 25% to about 60% of a pyrithione or polyvalent metal salt of a pyrithione; b) from about 0.01 to about 1% of a cationic polymer; c) from about 0.01% to about 2.0% of an anionic surfactant; wherein the cationic polymer has a molecular weight from about 100,000 to about 2,000,000. 2. A composition according to claim 1 wherein the pyrithione or polyvalent metal salt of a pyrithione is from about 30% to about 50%. 3. A composition according to claim 1 wherein the pyrithione or polyvalent metal salt of a pyrithione is zinc pyrithione. 4. A composition according to claim 1 wherein the cationic polymer is from about 0.05% to about 0.3%. 5. A composition according to claim 1 wherein the anionic surfactant is from about 0.08% to about 1.25%. 6. A composition according to claim 1 wherein the anionic surfactant is a sodium polynaphthalenesulfonate. 7. A composition according to claim 1 wherein the cationic polymer is from about 200,000 to about 700,000. 8. A composition according to claim 1 wherein the cationic polymer is from about 300,000 to about 500,000. 9. A composition according to claim 1 wherein the composition has a settling rate of less than 2.1 mm/day. 10. A composition according to claim 1 wherein the cationic polymer is selected from the group consisting of galactomanan, modified galactommans, cellulosics, modified cellulosics, starches, and mixtures thereof. 11. A composition according to claim 1 wherein the galactoman comprises fenugreek gum (galactose:mannose ratio 1:1), guar gum (ratio 1:2), tara gum (ratio 1:3), locust bean (ratio 1:4), cassia (ratio 1:5) and mixtures thereof. 12. A composition according to claim 1 wherein the starch is selected from the group consisting of corn, rice, potato, tapioca, and mixtures thereof. 13. A composition according to claim 1 wherein the synthetic polymer is selected from the group consisting of a copolymer of acrylamide and 1,3-Propanediaminium,N-[2-[[[dimethyl[3-[(2-methyl-1-oxo-2-propenyl)amino]propyl]ammonio]acetyl]amino]ethyl]2-hydroxy-N,N,N′,N′,N′-pentamethyl-, trichloride, a trimethylammoniopropylmethacrylamide chloride-N-Acrylamide copolymer, polydiallyldimethylammonium chloride and mixtures thereof. 14. A composition according to claim 1 wherein the composition further comprises a dispersant. 15. A composition according to claim 1 wherein the composition further comprises an inorganic salt. 16. A composition according to claim 1 wherein the composition further comprises a polymeric thickener. 17. A composition according to claim 16 wherein the polymeric thickener is selected from the group consisting of carbomers, modified carbomers, septic polyacrylates, EGDS, PEG-150 Distearate, polyethylene, PEG XM 7, PEGXM14, PEGXM23, glycerin, thixin and mixtures thereof. 18. A process of making of a composition comprising a) from about 25 to about 70 wt % of a pyrithione or polyvalent metal salt of a pyrithione; b) from about 0.01 to about 1 wt % of a cationic polymer with a molecular weight of about 100,000 to about 10,000,000 and a charge density of 0.1 to 7.0 meq; c) from about 0.01 to 2 wt % of an anionic surfactant; d) water to 100 wt %, comprising the steps of: 1. mixing the pyrithione or polyvalent metal salt of a pyrithione, the anionic surfactant and the water using a high speed dispersing equipment or a media mill, and 2. adding a cationic polymer to a final concentration of said cationic polymer of about 0.01% to about 1% of the total composition, wherein the cationic polymer has a molecular weight range of about 100,000 to about 10,000,000 and charge density of about 0.1 to 7 meq. 19. A process of making of a composition comprising a) from about 25 to about 70 wt % of a pyrithione or polyvalent metal salt of a pyrithione; b) from about 0.01 to about 1 wt % of a cationic polymer with molecular weight of about 100,000 to about 10,000,000 and charge density of about 0.1 to 7.0 meq; c) from about 0.01 to about 2 wt % of an anionic surfactant; d) water to 100 wt %, comprising the step of mixing the pyrithione or polyvalent metal salt of a pyrithione, the anionic surfactant, the cationic polymer and the water using a high speed dispersing equipment or a media mill.
A composition directed to comprising from about 25% to about 60% a pyrithione or polyvalent metal salt of a pyrithione; from about 0.01 to about 1% of a cationic polymer; from about 0.01% to about 2.0% of an anionic surfactant wherein the cationic polymer has a molecular weight from about 100,000 to about 2,000,000.1. A composition comprising: a) from about 25% to about 60% of a pyrithione or polyvalent metal salt of a pyrithione; b) from about 0.01 to about 1% of a cationic polymer; c) from about 0.01% to about 2.0% of an anionic surfactant; wherein the cationic polymer has a molecular weight from about 100,000 to about 2,000,000. 2. A composition according to claim 1 wherein the pyrithione or polyvalent metal salt of a pyrithione is from about 30% to about 50%. 3. A composition according to claim 1 wherein the pyrithione or polyvalent metal salt of a pyrithione is zinc pyrithione. 4. A composition according to claim 1 wherein the cationic polymer is from about 0.05% to about 0.3%. 5. A composition according to claim 1 wherein the anionic surfactant is from about 0.08% to about 1.25%. 6. A composition according to claim 1 wherein the anionic surfactant is a sodium polynaphthalenesulfonate. 7. A composition according to claim 1 wherein the cationic polymer is from about 200,000 to about 700,000. 8. A composition according to claim 1 wherein the cationic polymer is from about 300,000 to about 500,000. 9. A composition according to claim 1 wherein the composition has a settling rate of less than 2.1 mm/day. 10. A composition according to claim 1 wherein the cationic polymer is selected from the group consisting of galactomanan, modified galactommans, cellulosics, modified cellulosics, starches, and mixtures thereof. 11. A composition according to claim 1 wherein the galactoman comprises fenugreek gum (galactose:mannose ratio 1:1), guar gum (ratio 1:2), tara gum (ratio 1:3), locust bean (ratio 1:4), cassia (ratio 1:5) and mixtures thereof. 12. A composition according to claim 1 wherein the starch is selected from the group consisting of corn, rice, potato, tapioca, and mixtures thereof. 13. A composition according to claim 1 wherein the synthetic polymer is selected from the group consisting of a copolymer of acrylamide and 1,3-Propanediaminium,N-[2-[[[dimethyl[3-[(2-methyl-1-oxo-2-propenyl)amino]propyl]ammonio]acetyl]amino]ethyl]2-hydroxy-N,N,N′,N′,N′-pentamethyl-, trichloride, a trimethylammoniopropylmethacrylamide chloride-N-Acrylamide copolymer, polydiallyldimethylammonium chloride and mixtures thereof. 14. A composition according to claim 1 wherein the composition further comprises a dispersant. 15. A composition according to claim 1 wherein the composition further comprises an inorganic salt. 16. A composition according to claim 1 wherein the composition further comprises a polymeric thickener. 17. A composition according to claim 16 wherein the polymeric thickener is selected from the group consisting of carbomers, modified carbomers, septic polyacrylates, EGDS, PEG-150 Distearate, polyethylene, PEG XM 7, PEGXM14, PEGXM23, glycerin, thixin and mixtures thereof. 18. A process of making of a composition comprising a) from about 25 to about 70 wt % of a pyrithione or polyvalent metal salt of a pyrithione; b) from about 0.01 to about 1 wt % of a cationic polymer with a molecular weight of about 100,000 to about 10,000,000 and a charge density of 0.1 to 7.0 meq; c) from about 0.01 to 2 wt % of an anionic surfactant; d) water to 100 wt %, comprising the steps of: 1. mixing the pyrithione or polyvalent metal salt of a pyrithione, the anionic surfactant and the water using a high speed dispersing equipment or a media mill, and 2. adding a cationic polymer to a final concentration of said cationic polymer of about 0.01% to about 1% of the total composition, wherein the cationic polymer has a molecular weight range of about 100,000 to about 10,000,000 and charge density of about 0.1 to 7 meq. 19. A process of making of a composition comprising a) from about 25 to about 70 wt % of a pyrithione or polyvalent metal salt of a pyrithione; b) from about 0.01 to about 1 wt % of a cationic polymer with molecular weight of about 100,000 to about 10,000,000 and charge density of about 0.1 to 7.0 meq; c) from about 0.01 to about 2 wt % of an anionic surfactant; d) water to 100 wt %, comprising the step of mixing the pyrithione or polyvalent metal salt of a pyrithione, the anionic surfactant, the cationic polymer and the water using a high speed dispersing equipment or a media mill.
1,600
339
15,234,084
1,627
A topical composition comprising about 5 wt % to about 12.5 wt % of metronidazole or a pharmacologically acceptable salt thereof in a non-aqueous vehicle. The composition may be used in the treatment of skin damage due to inflammatory skin conditions; thermal, chemical or electrical burns; infections or radiation treatment. One advantage of the composition is that topical administration of metronidazole results in a primarily local effect, and thus, side effects observed from systemic administration are avoided.
1. A method of relieving pain and/or promoting wound healing in a patient due a pilonidal sinus wound, hidradenitis or pressure sore wounds, the method comprising: applying to the damaged skin area a topical composition, wherein the topical composition consists essentially of metronidazole in a therapeutically effective concentration of 10 wt % to treat the pilonidal sinus wound, hidradenitis or pressure sore wounds and in a pharmacologically acceptable non-aqueous vehicle. 2. The method of claim 1, wherein said vehicle is an organic vehicle. 3. The method of claim 1, wherein the vehicle comprises at least one hydrocarbon compound. 4. The method of claim 1, wherein the vehicle comprises a mixture of at least two semi-solid saturated hydrocarbon compounds. 5. The method of claim 1, wherein the vehicle comprises white petrolatum (USP). 6. The method of claim 1, wherein said topical composition is in the form of an ointment, lotion, gel, foam or cream. 7. The method of claim 1, wherein metronidazole is applied at a dosage for each application from about 125 mg to about 1250 mg. 8. The method of claim 7, wherein the dosage of metronidazole for each application is from about 125 mg to about 375 mg. 9. The method of claim 8, wherein the dosage of metronidazole for each application is about 250 mg. 10. The method of claim 1, wherein said topical composition is applied between from 2 to 4 times daily. 11. The method of claim 1, wherein said topical composition has a fluffy texture. 12. The method of claim 11, wherein said topical composition is obtained by passing metronidazole and white petrolatum through an ointment mill. 13. A topical composition for treatment of a pilonidal sinus wound, hidradenitis and pressure sore wounds, wherein the topical composition consists essentially of metronidazole in a therapeutically effective concentration of 10 wt % to treat the a pilonidal sinus wound, hidradenitis and pressure sore wounds and in a pharmacologically acceptable non-aqueous vehicle. 14. The topical composition of claim 13, wherein the composition is in the form of an ointment, lotion, gel, foam or cream.
A topical composition comprising about 5 wt % to about 12.5 wt % of metronidazole or a pharmacologically acceptable salt thereof in a non-aqueous vehicle. The composition may be used in the treatment of skin damage due to inflammatory skin conditions; thermal, chemical or electrical burns; infections or radiation treatment. One advantage of the composition is that topical administration of metronidazole results in a primarily local effect, and thus, side effects observed from systemic administration are avoided.1. A method of relieving pain and/or promoting wound healing in a patient due a pilonidal sinus wound, hidradenitis or pressure sore wounds, the method comprising: applying to the damaged skin area a topical composition, wherein the topical composition consists essentially of metronidazole in a therapeutically effective concentration of 10 wt % to treat the pilonidal sinus wound, hidradenitis or pressure sore wounds and in a pharmacologically acceptable non-aqueous vehicle. 2. The method of claim 1, wherein said vehicle is an organic vehicle. 3. The method of claim 1, wherein the vehicle comprises at least one hydrocarbon compound. 4. The method of claim 1, wherein the vehicle comprises a mixture of at least two semi-solid saturated hydrocarbon compounds. 5. The method of claim 1, wherein the vehicle comprises white petrolatum (USP). 6. The method of claim 1, wherein said topical composition is in the form of an ointment, lotion, gel, foam or cream. 7. The method of claim 1, wherein metronidazole is applied at a dosage for each application from about 125 mg to about 1250 mg. 8. The method of claim 7, wherein the dosage of metronidazole for each application is from about 125 mg to about 375 mg. 9. The method of claim 8, wherein the dosage of metronidazole for each application is about 250 mg. 10. The method of claim 1, wherein said topical composition is applied between from 2 to 4 times daily. 11. The method of claim 1, wherein said topical composition has a fluffy texture. 12. The method of claim 11, wherein said topical composition is obtained by passing metronidazole and white petrolatum through an ointment mill. 13. A topical composition for treatment of a pilonidal sinus wound, hidradenitis and pressure sore wounds, wherein the topical composition consists essentially of metronidazole in a therapeutically effective concentration of 10 wt % to treat the a pilonidal sinus wound, hidradenitis and pressure sore wounds and in a pharmacologically acceptable non-aqueous vehicle. 14. The topical composition of claim 13, wherein the composition is in the form of an ointment, lotion, gel, foam or cream.
1,600
340
15,395,233
1,644
Embodiments of the present invention provide for the facile generation of a stable recombinant fusion polypeptides with intrinsic fluorescent properties. The recombinant antibodies may be suitable for qualitative and/or quantitative immunofluorescence analysis. Generally, the fluorescent polypeptides include a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus.
1-18. (canceled) 19. A polynucleotide that encodes the polypeptide comprising: a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus. 20. A cell comprising the polynucleotide of claim 19. 21-22. (canceled) 23. The cell of claim 20 wherein the cell comprises a microbe. 24. The cell of claim 20 wherein the cell comprises a tumor cell. 25. A method comprising introducing into a cell a polynucleotide that encodes a polypeptide that comprises: a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus. 26. The method of claim 25 wherein the cell comprises a tumor cell. 27-51. (canceled) 52. A method of making a fusion polypeptide, the method comprising: creating an expression vector that comprises a polynucleotide operably linked to a promoter, wherein the polynucleotide encodes an fusion polypeptide comprising: a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus; introducing the expression vector into a host cell; and growing the host cell comprising the expression vector in conditions effective for the host cell to express the fusion polypeptide.
Embodiments of the present invention provide for the facile generation of a stable recombinant fusion polypeptides with intrinsic fluorescent properties. The recombinant antibodies may be suitable for qualitative and/or quantitative immunofluorescence analysis. Generally, the fluorescent polypeptides include a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus.1-18. (canceled) 19. A polynucleotide that encodes the polypeptide comprising: a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus. 20. A cell comprising the polynucleotide of claim 19. 21-22. (canceled) 23. The cell of claim 20 wherein the cell comprises a microbe. 24. The cell of claim 20 wherein the cell comprises a tumor cell. 25. A method comprising introducing into a cell a polynucleotide that encodes a polypeptide that comprises: a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus. 26. The method of claim 25 wherein the cell comprises a tumor cell. 27-51. (canceled) 52. A method of making a fusion polypeptide, the method comprising: creating an expression vector that comprises a polynucleotide operably linked to a promoter, wherein the polynucleotide encodes an fusion polypeptide comprising: a fluorescent domain comprising a C-terminus and an N-terminus; a first antibody domain covalently linked to the C-terminus; and a second antibody domain covalently linked to the N-terminus; introducing the expression vector into a host cell; and growing the host cell comprising the expression vector in conditions effective for the host cell to express the fusion polypeptide.
1,600
341
14,886,238
1,662
A high protein (up to 40%) tetraploid oat variety is provided that is suitable for large scale oat production using standard farming practices. A tetraploid oat variety includes one or more of lodging resistance, shattering resistance, erect growth habit, and seeds similar to traditional cultivated hexaploid oat, A. sativa . The tetraploid oat variety of the invention can also include a stable fatty acid profile, high iron content, high folic acid content, or high free essential amino acid content. A tetraploid oat variety may be used as foundational seed in a plant breeding program for development of lines and varieties with high protein content. Oat products produced from the tetraploid oat variety of the invention are also included as well as resultant oat foodstuffs such as high protein granola bars, hot cereals food stuffs, cold cereal foodstuffs, snackbars, cookies, gluten-free products, snacks, muffins, pancake mix and the like.
1. A plant of a tetraploid oat variety that is cultivatable using one or more standard farming practices. 2. The plant of claim 1, wherein the standard farming practices include mechanical planting, mechanical harvesting, and/or mechanical milling. 3. The plant of claim 1, wherein the plant is Avena magna. 4. The plant of claim 1, wherein the variety has one or more of the following traits being comparable or superior to Avena sativa variety ‘Leggett’: stature, shattering resistance, lodging resistance, plant height, time to maturity, ease of dehulling, and seed yield. 5. The plant of claim 1, comprising a seed protein content of greater than 14%. 6. The plant of claim 1, comprising a seed protein content of greater than 18%. 7. The plant of claim 1, comprising a seed oleic/linoleic ratio greater than 1. 8. The plant of claim 1, comprising a seed oleic/linoleic ratio that is superior to that of A. sativa ‘Leggett’. 9. The plant of claim 1, comprising a seed iron content that is superior to that of A. sativa ‘Leggett’. 10. The plant of claim 1, comprising a seed folic acid content that is superior to that of A. sativa ‘Leggett’. 11. The plant of claim 1, comprising a seed protein content that is superior to that of A. sativa ‘Leggett’. 12. The plant of claim 1, comprising a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 13. An oat plant produced from growing seed of the plant of claim 1. 14. A progeny plant of the plant of claim 1 that is cultivatable using one or more standard farming practices. 15. A progeny plant of claim 14, further comprising a seed protein content, a seed oleic/linoleic ratio, a seed iron content, a seed folic acid content, or a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 16. A progeny plant of claim 13 having a lineage comprising a second oat plant. 17. The plant of claim 1, wherein the variety is 96.5.6, 96.5.34, 96.5.55, or 100.2.35, or a descendant thereof. 18. An oat ingredient derived from a plant of a tetraploid oat variety that is cultivatable using one or more standard farming practices. 19. A composition comprising the oat ingredient of claim 18. 20. A product comprising the composition of claim 19. 21. A plant of an oat variety having a seed protein content greater than about 18%. 22. The plant of claim 21, wherein the plant is A. magna. 23. The plant of claim 22, wherein the oat variety is cultivatable using one or more standard farming techniques. 24. The plant of claim 23, wherein the standard farming practices include mechanical planting, mechanical harvesting, and/or mechanical milling. 25. The plant of claim 21, wherein the oat variety is a tetraploid. 26. The plant of claim 21, comprising a seed oleic/linoleic ratio greater than 1. 27. The plant of claim 21, comprising a seed oleic/linoleic ratio that is superior to that of A. sativa ‘Leggett’. 28. The plant of claim 21, comprising a seed iron content that is superior to that of A. sativa ‘Leggett’. 29. The plant of claim 21, comprising a seed folic acid content that is superior to that of A. sativa ‘Leggett’. 30. The plant of claim 21, comprising a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 31. The plant claim 21, wherein said variety has one or more of the following traits being comparable or superior to A. sativa ‘Leggett’: stature, shattering resistance, lodging resistance, plat height, time to maturity, ease of dehulling, and seed yield. 32. A progeny plant of the plant of claim 21 that is cultivatable using one or more standard farming practices. 33. A progeny plant of claim 32, further comprising a seed protein content, a seed oleic/linoleic ratio, a seed iron content, a seed folic acid content, or a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 34. A progeny plant of claim 32 having a lineage comprising a second oat plant. 35. The plant of claim 21, wherein the variety is 96.5.6, 96.5.34, 96.5.55, or 100.2.35, or a descendant thereof. 36. An oat ingredient derived from a plant of an oat variety having a seed protein content of greater than about 18%. 37. A composition comprising the oat ingredient of claim 36. 38. A product comprising the composition of claim 37. 39. A plant of the species Avena magna ssp domestica. 40. The plant of claim 39, which is fertile with wild Avena magna. 41. A population of plants comprising a plurality of the plant of claim 39.
A high protein (up to 40%) tetraploid oat variety is provided that is suitable for large scale oat production using standard farming practices. A tetraploid oat variety includes one or more of lodging resistance, shattering resistance, erect growth habit, and seeds similar to traditional cultivated hexaploid oat, A. sativa . The tetraploid oat variety of the invention can also include a stable fatty acid profile, high iron content, high folic acid content, or high free essential amino acid content. A tetraploid oat variety may be used as foundational seed in a plant breeding program for development of lines and varieties with high protein content. Oat products produced from the tetraploid oat variety of the invention are also included as well as resultant oat foodstuffs such as high protein granola bars, hot cereals food stuffs, cold cereal foodstuffs, snackbars, cookies, gluten-free products, snacks, muffins, pancake mix and the like.1. A plant of a tetraploid oat variety that is cultivatable using one or more standard farming practices. 2. The plant of claim 1, wherein the standard farming practices include mechanical planting, mechanical harvesting, and/or mechanical milling. 3. The plant of claim 1, wherein the plant is Avena magna. 4. The plant of claim 1, wherein the variety has one or more of the following traits being comparable or superior to Avena sativa variety ‘Leggett’: stature, shattering resistance, lodging resistance, plant height, time to maturity, ease of dehulling, and seed yield. 5. The plant of claim 1, comprising a seed protein content of greater than 14%. 6. The plant of claim 1, comprising a seed protein content of greater than 18%. 7. The plant of claim 1, comprising a seed oleic/linoleic ratio greater than 1. 8. The plant of claim 1, comprising a seed oleic/linoleic ratio that is superior to that of A. sativa ‘Leggett’. 9. The plant of claim 1, comprising a seed iron content that is superior to that of A. sativa ‘Leggett’. 10. The plant of claim 1, comprising a seed folic acid content that is superior to that of A. sativa ‘Leggett’. 11. The plant of claim 1, comprising a seed protein content that is superior to that of A. sativa ‘Leggett’. 12. The plant of claim 1, comprising a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 13. An oat plant produced from growing seed of the plant of claim 1. 14. A progeny plant of the plant of claim 1 that is cultivatable using one or more standard farming practices. 15. A progeny plant of claim 14, further comprising a seed protein content, a seed oleic/linoleic ratio, a seed iron content, a seed folic acid content, or a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 16. A progeny plant of claim 13 having a lineage comprising a second oat plant. 17. The plant of claim 1, wherein the variety is 96.5.6, 96.5.34, 96.5.55, or 100.2.35, or a descendant thereof. 18. An oat ingredient derived from a plant of a tetraploid oat variety that is cultivatable using one or more standard farming practices. 19. A composition comprising the oat ingredient of claim 18. 20. A product comprising the composition of claim 19. 21. A plant of an oat variety having a seed protein content greater than about 18%. 22. The plant of claim 21, wherein the plant is A. magna. 23. The plant of claim 22, wherein the oat variety is cultivatable using one or more standard farming techniques. 24. The plant of claim 23, wherein the standard farming practices include mechanical planting, mechanical harvesting, and/or mechanical milling. 25. The plant of claim 21, wherein the oat variety is a tetraploid. 26. The plant of claim 21, comprising a seed oleic/linoleic ratio greater than 1. 27. The plant of claim 21, comprising a seed oleic/linoleic ratio that is superior to that of A. sativa ‘Leggett’. 28. The plant of claim 21, comprising a seed iron content that is superior to that of A. sativa ‘Leggett’. 29. The plant of claim 21, comprising a seed folic acid content that is superior to that of A. sativa ‘Leggett’. 30. The plant of claim 21, comprising a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 31. The plant claim 21, wherein said variety has one or more of the following traits being comparable or superior to A. sativa ‘Leggett’: stature, shattering resistance, lodging resistance, plat height, time to maturity, ease of dehulling, and seed yield. 32. A progeny plant of the plant of claim 21 that is cultivatable using one or more standard farming practices. 33. A progeny plant of claim 32, further comprising a seed protein content, a seed oleic/linoleic ratio, a seed iron content, a seed folic acid content, or a seed free essential amino acid content that is superior to that of A. sativa ‘Leggett’. 34. A progeny plant of claim 32 having a lineage comprising a second oat plant. 35. The plant of claim 21, wherein the variety is 96.5.6, 96.5.34, 96.5.55, or 100.2.35, or a descendant thereof. 36. An oat ingredient derived from a plant of an oat variety having a seed protein content of greater than about 18%. 37. A composition comprising the oat ingredient of claim 36. 38. A product comprising the composition of claim 37. 39. A plant of the species Avena magna ssp domestica. 40. The plant of claim 39, which is fertile with wild Avena magna. 41. A population of plants comprising a plurality of the plant of claim 39.
1,600
342
12,086,453
1,612
A method for preparing a product for treating the skin and mucous membranes comprises the steps of making available a suitable quantity of trichloracetic acid, making available a suitable quantity of hydrogen peroxide, making a first mixture of the trichloracetic acid and the hydrogen peroxide, making available a determinate quantity of the basic compound able to achieve a buffer effect of the trichloracetic acid comprised in the first mixture, and adding the basic compound.
1. A method for preparing a product for treating the skin and mucous membranes, comprising the following steps: a) to make available a suitable quantity of trichloracetic acid (CCl3COOH or TCA); b) to make available a suitable quantity of hydrogen peroxide (H2O2); c) to make a first mixture of said trichloracetic acid with said hydrogen peroxide; d) to make available a determinate quantity of basic compound, able to achieve a buffer effect of said trichloracetic acid comprised in said first mixture; e) to add said basic compound to said first mixture in order to buffer said first mixture comprising said trichloracetic acid CCl3COOH. 2. The method as in claim 1, wherein said basic compound is at least a compound chosen from a group comprising ammonia, triethanolamine and suchlike, or mixture thereof. 3. The method as in claim 1, wherein said product obtained from step e) has a pH comprised between about 2.3 and 2.6. 4. The method as in claim 1, wherein said trichloracetic acid is supplied substantially pure in said step a). 5. The method as in claim 1, wherein said trichloracetic acid of said step a) is obtained by means of a step of chlorination of acetic acid or chloracetic acid or a mixture thereof. 6. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration comprised between about 15 and 50% w/w. 7. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration comprised between about 25 and 40% w/w. 8. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration comprised between about 30 and 35% w/w. 9. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration of about 33% w/w. 10. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration comprised between about 85 and 50% w/w. 11. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration comprised between about 75 and 60% w/w. 12. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration comprised between about 70 and 65% w/w. 13. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration of about 67% w/w. 14-18. (canceled) 19. A cream, an ointment, a liquid, a gel, an aerosol and suchlike comprising a product obtainable from a method as in claim 1. 20. The method as in claim 1, further comprising a gelling step of the product obtained in step e), by using a thickening agent. 21. The method as in claim 20, wherein the gelled product has a pH greater than the starting pH of the product subjected to gelling. 22. The method as in claim 21, wherein the gelled product has a pH between about 5 and 6. 23. A cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19 having a pH greater than the starting pH of said product. 24. A cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 23 having a pH between about 5 and 6. 25. The use of a cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19 for the therapeutic treatment of pathologies of the skin and mucous membranes, such as acne, juvenile acne, scars resulting from pathologies such as juvenile acne, chicken pox and suchlike, damage caused by the sun, sun freckles, pre-cancerous lesions, problems of pigmentation, damage deriving from recent depressed scars, atrophy, hypertrophy, retraction and alteration of the scarring of every part of the body, including the gums, damage caused by ageing, photo-ageing of the skin, wrinkles, sagging of the skin, drooping of the breasts and suchlike. 26. The use of a cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19, for the production of a medication for the therapeutic application for the treatment of pathologies of the skin and mucous membranes such as acne, juvenile acne, scars resulting from pathologies such as juvenile acne, chicken pox and suchlike, damage caused by the sun, freckles, pre-cancerous lesions, problems of pigmentation, damage deriving from recent depressed scars, atrophy, hypertrophy, retraction and alteration of the scarring of every part of the body, including the gums, damage caused by ageing, photo-ageing of the skin, wrinkles, sagging of the skin, drooping of the breasts and suchlike. 27. The use of a cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19 for the cosmetic treatment of pathologies of the skin and mucous membranes, such as acne, juvenile acne, scars resulting from pathologies such as juvenile acne, chicken pox and suchlike, damage caused by the sun, sun freckles, pre-cancerous lesions, problems of pigmentation, damage deriving from recent depressed scars, atrophy, hypertrophy, retraction and alteration of the scarring of every part of the body, including the gums, damage caused by ageing, photo-ageing of the skin, wrinkles, sagging of the skin, drooping of the breasts and suchlike.
A method for preparing a product for treating the skin and mucous membranes comprises the steps of making available a suitable quantity of trichloracetic acid, making available a suitable quantity of hydrogen peroxide, making a first mixture of the trichloracetic acid and the hydrogen peroxide, making available a determinate quantity of the basic compound able to achieve a buffer effect of the trichloracetic acid comprised in the first mixture, and adding the basic compound.1. A method for preparing a product for treating the skin and mucous membranes, comprising the following steps: a) to make available a suitable quantity of trichloracetic acid (CCl3COOH or TCA); b) to make available a suitable quantity of hydrogen peroxide (H2O2); c) to make a first mixture of said trichloracetic acid with said hydrogen peroxide; d) to make available a determinate quantity of basic compound, able to achieve a buffer effect of said trichloracetic acid comprised in said first mixture; e) to add said basic compound to said first mixture in order to buffer said first mixture comprising said trichloracetic acid CCl3COOH. 2. The method as in claim 1, wherein said basic compound is at least a compound chosen from a group comprising ammonia, triethanolamine and suchlike, or mixture thereof. 3. The method as in claim 1, wherein said product obtained from step e) has a pH comprised between about 2.3 and 2.6. 4. The method as in claim 1, wherein said trichloracetic acid is supplied substantially pure in said step a). 5. The method as in claim 1, wherein said trichloracetic acid of said step a) is obtained by means of a step of chlorination of acetic acid or chloracetic acid or a mixture thereof. 6. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration comprised between about 15 and 50% w/w. 7. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration comprised between about 25 and 40% w/w. 8. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration comprised between about 30 and 35% w/w. 9. The method as in claim 1, wherein said trichloracetic acid in said first mixture is in a concentration of about 33% w/w. 10. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration comprised between about 85 and 50% w/w. 11. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration comprised between about 75 and 60% w/w. 12. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration comprised between about 70 and 65% w/w. 13. The method as in claim 1, wherein said hydrogen peroxide in said first mixture is in a concentration of about 67% w/w. 14-18. (canceled) 19. A cream, an ointment, a liquid, a gel, an aerosol and suchlike comprising a product obtainable from a method as in claim 1. 20. The method as in claim 1, further comprising a gelling step of the product obtained in step e), by using a thickening agent. 21. The method as in claim 20, wherein the gelled product has a pH greater than the starting pH of the product subjected to gelling. 22. The method as in claim 21, wherein the gelled product has a pH between about 5 and 6. 23. A cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19 having a pH greater than the starting pH of said product. 24. A cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 23 having a pH between about 5 and 6. 25. The use of a cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19 for the therapeutic treatment of pathologies of the skin and mucous membranes, such as acne, juvenile acne, scars resulting from pathologies such as juvenile acne, chicken pox and suchlike, damage caused by the sun, sun freckles, pre-cancerous lesions, problems of pigmentation, damage deriving from recent depressed scars, atrophy, hypertrophy, retraction and alteration of the scarring of every part of the body, including the gums, damage caused by ageing, photo-ageing of the skin, wrinkles, sagging of the skin, drooping of the breasts and suchlike. 26. The use of a cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19, for the production of a medication for the therapeutic application for the treatment of pathologies of the skin and mucous membranes such as acne, juvenile acne, scars resulting from pathologies such as juvenile acne, chicken pox and suchlike, damage caused by the sun, freckles, pre-cancerous lesions, problems of pigmentation, damage deriving from recent depressed scars, atrophy, hypertrophy, retraction and alteration of the scarring of every part of the body, including the gums, damage caused by ageing, photo-ageing of the skin, wrinkles, sagging of the skin, drooping of the breasts and suchlike. 27. The use of a cream, an ointment, a liquid, a gel, an aerosol and suchlike as in claim 19 for the cosmetic treatment of pathologies of the skin and mucous membranes, such as acne, juvenile acne, scars resulting from pathologies such as juvenile acne, chicken pox and suchlike, damage caused by the sun, sun freckles, pre-cancerous lesions, problems of pigmentation, damage deriving from recent depressed scars, atrophy, hypertrophy, retraction and alteration of the scarring of every part of the body, including the gums, damage caused by ageing, photo-ageing of the skin, wrinkles, sagging of the skin, drooping of the breasts and suchlike.
1,600
343
14,353,443
1,653
The invention relates to a process for the conversion of lignocellulose into an organic acid including an alkaline pretreatment step and a fermentation step, wherein liquid phase obtained in the fermentation step is recycled to the alkaline pretreatment step and/or the fermentation step. Organic acid is recovered as its magnesium of calcium salt from solid phase obtained in the fermentation step.
1. A process for the conversion of lignocellulose into an organic acid, the process comprising the following steps: a) pretreating a feed comprising lignocellulose material with an alkaline agent comprising a divalent cation, wherein the divalent cation is a calcium or magnesium cation, in the presence of water at a pretreatment temperature, to obtain an aqueous slurry of alkaline pretreated lignocellulose material; b) supplying at least part of the slurry of alkaline pretreated lignocellulose material to a fermentation zone and subjecting the pretreated lignocellulose material, in the fermentation zone in the presence of a hydrolytic enzyme and a micro-organism that is able to convert saccharides into an organic acid, to enzymatic hydrolysis and fermentation to obtain a fermentation broth comprising insoluble lignocellulose, precipitated and dissolved salt of the organic acid with the divalent cation, and enzyme; c) discharging fermentation broth obtained in step (b) from the fermentation zone; d) separating from the fermentation broth a liquid phase comprising the dissolved salt of the organic acid and a solid phase comprising insoluble lignocellulose and the precipitated salt of the organic acid; and e) recycling at least part of the liquid phase to alkaline pre-treatment step a) and/or to the fermentation zone. 2. A process according to claim 1, wherein at least part of the liquid phase is recycled to step a). 3. A process according to claim 2, wherein at least 50 vol % of the liquid phase is recycled to step a). 4. A process according to any one of the preceding claim 1, wherein the fermentation zone comprises at least two fermentors in series and wherein in step c) fermentation broth is discharged from the last fermentor in series. 5. A process according to claim 1, further comprising washing the solid phase obtained in step d) with water at a temperature in the range of from 10 to 50° C. to obtain washed solid phase and wash water and supplying at least part of the wash water to step a). 6. A process according to claim 1, further comprising recovering the salt of the organic acid from the solid phase. 7. A process according to claim 6, comprising extracting the solid phase obtained in step d), optionally after washing, with water at a temperature in the range of from 60 to 100° C. to obtain a solution of the salt of the organic acid and an extracted solid phase. 8. A process according to claim 7 further comprising cooling the solution to obtain precipitated salt of the organic acid and recovering the precipitated salt in solid form. 9. A process according to claim 6, further comprising a fermentation step wherein the salt of organic acid is fermented into a fermentation product and the fermentation product is recovered as product. 10. A process according to claim 9, wherein a further acid is added during the fermentation step and a calcium or magnesium salt of the further acid is formed as co-product in the fermentation step. 11. A process according to claim 7, further comprising feeding the extracted solid phase to a power generator to generate electricity and waste heat. 12. A process according to claim 11, wherein the feed comprises paper comprising calcium carbonate and wherein calcium oxide is formed in the power generator and wherein calcium oxide formed in the power generator is recycled to alkaline pretreatment step a) as alkaline agent. 13. A process according to claim 11, wherein the waste heat generated is used to achieve the pretreatment temperature in step a). 14. A process according to claim 1, wherein the micro-organism is a lactic-acid producing micro-organism and the organic acid is lactic acid. 15. A process according to claim 1, wherein the feed comprises protein. 16. A process according to claim 15, wherein amino acids and/or peptides are recovered from the liquid phase separated from the fermentation broth.
The invention relates to a process for the conversion of lignocellulose into an organic acid including an alkaline pretreatment step and a fermentation step, wherein liquid phase obtained in the fermentation step is recycled to the alkaline pretreatment step and/or the fermentation step. Organic acid is recovered as its magnesium of calcium salt from solid phase obtained in the fermentation step.1. A process for the conversion of lignocellulose into an organic acid, the process comprising the following steps: a) pretreating a feed comprising lignocellulose material with an alkaline agent comprising a divalent cation, wherein the divalent cation is a calcium or magnesium cation, in the presence of water at a pretreatment temperature, to obtain an aqueous slurry of alkaline pretreated lignocellulose material; b) supplying at least part of the slurry of alkaline pretreated lignocellulose material to a fermentation zone and subjecting the pretreated lignocellulose material, in the fermentation zone in the presence of a hydrolytic enzyme and a micro-organism that is able to convert saccharides into an organic acid, to enzymatic hydrolysis and fermentation to obtain a fermentation broth comprising insoluble lignocellulose, precipitated and dissolved salt of the organic acid with the divalent cation, and enzyme; c) discharging fermentation broth obtained in step (b) from the fermentation zone; d) separating from the fermentation broth a liquid phase comprising the dissolved salt of the organic acid and a solid phase comprising insoluble lignocellulose and the precipitated salt of the organic acid; and e) recycling at least part of the liquid phase to alkaline pre-treatment step a) and/or to the fermentation zone. 2. A process according to claim 1, wherein at least part of the liquid phase is recycled to step a). 3. A process according to claim 2, wherein at least 50 vol % of the liquid phase is recycled to step a). 4. A process according to any one of the preceding claim 1, wherein the fermentation zone comprises at least two fermentors in series and wherein in step c) fermentation broth is discharged from the last fermentor in series. 5. A process according to claim 1, further comprising washing the solid phase obtained in step d) with water at a temperature in the range of from 10 to 50° C. to obtain washed solid phase and wash water and supplying at least part of the wash water to step a). 6. A process according to claim 1, further comprising recovering the salt of the organic acid from the solid phase. 7. A process according to claim 6, comprising extracting the solid phase obtained in step d), optionally after washing, with water at a temperature in the range of from 60 to 100° C. to obtain a solution of the salt of the organic acid and an extracted solid phase. 8. A process according to claim 7 further comprising cooling the solution to obtain precipitated salt of the organic acid and recovering the precipitated salt in solid form. 9. A process according to claim 6, further comprising a fermentation step wherein the salt of organic acid is fermented into a fermentation product and the fermentation product is recovered as product. 10. A process according to claim 9, wherein a further acid is added during the fermentation step and a calcium or magnesium salt of the further acid is formed as co-product in the fermentation step. 11. A process according to claim 7, further comprising feeding the extracted solid phase to a power generator to generate electricity and waste heat. 12. A process according to claim 11, wherein the feed comprises paper comprising calcium carbonate and wherein calcium oxide is formed in the power generator and wherein calcium oxide formed in the power generator is recycled to alkaline pretreatment step a) as alkaline agent. 13. A process according to claim 11, wherein the waste heat generated is used to achieve the pretreatment temperature in step a). 14. A process according to claim 1, wherein the micro-organism is a lactic-acid producing micro-organism and the organic acid is lactic acid. 15. A process according to claim 1, wherein the feed comprises protein. 16. A process according to claim 15, wherein amino acids and/or peptides are recovered from the liquid phase separated from the fermentation broth.
1,600
344
14,419,010
1,631
The invention relates to the identification of microbes in a sample by calculating the similarities between a mass spectrum of the sample and all reference spectra in a spectral library; it particularly concerns the detection of microbe mixtures. Microbe mixtures are probably present if several microbe species which are not closely related to each other are among the score list containing the most similar reference spectra. Methods are proposed which (1) operate with a list of the relationships, (2) determine the similarity between the reference spectra of the different microbe species of the score list, or (3) always carry out a mixture analysis in accordance with document DE 10 2009 007 266 A1 from the score list, with the generation of combination spectra, and only afterwards check the similarity of the combined spectra and thus the relationship between the microbe strains concerned.
1. Method for the recognition of microbe mixtures in the mass-spectrometric identification of a microbe sample, with compilation of a score list of the reference spectra in a library which are most similar to the sample spectrum, wherein, if two or more microbe species are present in the score list, the relationships between the microbe strains are determined with the aid of an exclusion list, and the microbe sample is indicated as a microbe mixture if the relationship is below a specified degree. 2. Method for the recognition of microbe mixtures in the mass-spectrometric identification of a microbe sample, with compilation of a score list of the reference spectra in a library which are similar to the sample spectrum, wherein, if two or more identical microbe species are present in the score list, the similarities between the reference spectra of the different microbe species are investigated, and the microbe sample is indicated as a microbe mixture if the similarities are below a specified similarity threshold. 3. Method according to claim 1 or 2, wherein, if a microbe mixture has been indicated, a mixture analysis using combination spectra is carried out. 4. Method for the detection and identification of microbe mixtures in the mass-spectrometric identification of a microbe sample, with compilation of a score list of the reference spectra in a library which are similar to the sample spectrum, comprising the steps a) forming combination spectra from the reference spectra of the different microbe species in the score list, b) computing the similarities between these combination spectra and the sample spectrum, c) selecting the combination spectra whose similarity to the sample spectrum is better than the similarities between the sample spectrum and the pure reference spectra, d) computing the similarities between the reference spectra combined for these selected combination spectra, e) indicating a mixture if the similarities of the combined reference spectra are below a specified similarity threshold, and f) indicating the microbe species of the mixture as those whose combinations of reference spectra demonstrate the best similarities to the sample spectrum. 5. Method according to claim 4, wherein the specified similarity thresholds are determined experimentally. 6. Method according to claim 1, wherein the reference spectra of the score list are differentiated from the other reference spectra in the library by a specified similarity threshold. 7. Method according to claim 1, wherein the score list contains a specified number of most similar reference spectra. 8. Method according to claim 1, wherein the score list contains all the reference spectra up to the reference spectrum where a specified jump takes place in the similarity score in relation to the next most similar reference spectrum. 9. Method according to claim 2 wherein, if a microbe mixture has been indicated, a mixture analysis using combination spectra is carried out. 10. Method according to claim 2, wherein the specified similarity thresholds are determined experimentally. 11. Method according to claim 2, wherein the reference spectra of the score list are differentiated from the other reference spectra in the library by a specified similarity threshold. 12. Method according to claim 2, wherein the score list contains a specified number of most similar reference spectra. 13. Method according to claim 2, wherein the score list contains all the reference spectra up to the reference spectrum where a specified jump takes place in the similarity score in relation to the next most similar reference spectrum.
The invention relates to the identification of microbes in a sample by calculating the similarities between a mass spectrum of the sample and all reference spectra in a spectral library; it particularly concerns the detection of microbe mixtures. Microbe mixtures are probably present if several microbe species which are not closely related to each other are among the score list containing the most similar reference spectra. Methods are proposed which (1) operate with a list of the relationships, (2) determine the similarity between the reference spectra of the different microbe species of the score list, or (3) always carry out a mixture analysis in accordance with document DE 10 2009 007 266 A1 from the score list, with the generation of combination spectra, and only afterwards check the similarity of the combined spectra and thus the relationship between the microbe strains concerned.1. Method for the recognition of microbe mixtures in the mass-spectrometric identification of a microbe sample, with compilation of a score list of the reference spectra in a library which are most similar to the sample spectrum, wherein, if two or more microbe species are present in the score list, the relationships between the microbe strains are determined with the aid of an exclusion list, and the microbe sample is indicated as a microbe mixture if the relationship is below a specified degree. 2. Method for the recognition of microbe mixtures in the mass-spectrometric identification of a microbe sample, with compilation of a score list of the reference spectra in a library which are similar to the sample spectrum, wherein, if two or more identical microbe species are present in the score list, the similarities between the reference spectra of the different microbe species are investigated, and the microbe sample is indicated as a microbe mixture if the similarities are below a specified similarity threshold. 3. Method according to claim 1 or 2, wherein, if a microbe mixture has been indicated, a mixture analysis using combination spectra is carried out. 4. Method for the detection and identification of microbe mixtures in the mass-spectrometric identification of a microbe sample, with compilation of a score list of the reference spectra in a library which are similar to the sample spectrum, comprising the steps a) forming combination spectra from the reference spectra of the different microbe species in the score list, b) computing the similarities between these combination spectra and the sample spectrum, c) selecting the combination spectra whose similarity to the sample spectrum is better than the similarities between the sample spectrum and the pure reference spectra, d) computing the similarities between the reference spectra combined for these selected combination spectra, e) indicating a mixture if the similarities of the combined reference spectra are below a specified similarity threshold, and f) indicating the microbe species of the mixture as those whose combinations of reference spectra demonstrate the best similarities to the sample spectrum. 5. Method according to claim 4, wherein the specified similarity thresholds are determined experimentally. 6. Method according to claim 1, wherein the reference spectra of the score list are differentiated from the other reference spectra in the library by a specified similarity threshold. 7. Method according to claim 1, wherein the score list contains a specified number of most similar reference spectra. 8. Method according to claim 1, wherein the score list contains all the reference spectra up to the reference spectrum where a specified jump takes place in the similarity score in relation to the next most similar reference spectrum. 9. Method according to claim 2 wherein, if a microbe mixture has been indicated, a mixture analysis using combination spectra is carried out. 10. Method according to claim 2, wherein the specified similarity thresholds are determined experimentally. 11. Method according to claim 2, wherein the reference spectra of the score list are differentiated from the other reference spectra in the library by a specified similarity threshold. 12. Method according to claim 2, wherein the score list contains a specified number of most similar reference spectra. 13. Method according to claim 2, wherein the score list contains all the reference spectra up to the reference spectrum where a specified jump takes place in the similarity score in relation to the next most similar reference spectrum.
1,600
345
11,322,156
1,627
The invention relates to medicine and is directed at developing means and methods for narcological practice and for preventive measures in order to reduce the risk of falling sick with such menacing modem diseases like dependence diseases, in particular alcoholism, narcomania and toximania. Said invention provides for using already known chemical compounds, i.e. vicinal dithioglycols for developing medical agents exhibiting properties for preventing contraction or development or reducing the speed of pathological processes forming a base for dependence diseases, in particular withdrawal syndromes. The inventive substances reduce or block pharmacological effects resulting from ethanol injection and/or narcotics, thereby directly influencing pathogenic mechanisms of the dependence diseases, in particular alcoholism. It is stated that said agents are particularly efficient when they are perorally administered. Said invention is accompanied by experimental material which proves the large perspective of the inventive agents and of a prophylactic method which provides for the use thereof before and after consuming alcohol-containing beverages or narcotics. The use of the invention makes it possible to reduce the degree of falling sick with alcoholism and narcomania and to reduce the toxic action of alcohols during the household use thereof.
1. A method for the prevention of beginnings and/or for the prevention of development and/or for reducing the rate of development of pathological processes caused by the use of alcohol-containing beverages and/or substances having an addictive potential, comprising (a) providing a preparation comprising at least one vicinal dithioglycol of general chemical formula (1): R1CH(SH)CH(SH)R2  (1)where R1 is selected from the groups: (—H) or (—COOH), or (—SO3H); R2 is selected from the groups: (—H) or (—COOH), or (—OH), or (—CH2—COOH), or (—CH2—OH), or (—CH2—SO3H), or (—CH2—O—CH2—SO3H); or salts of derivatives thereof, comprising salt forming groups (—OH) or (—COOH), or (—SO3H); and (b) administering the preparation to a subject by peroral or nasal administration in an effective amount. 2. The method according to claim 1, wherein the substance having an addictive potential is a narcotic substance. 3. The method according to claim 1, wherein the preparation is used for attaining at least one effect selected from the group comprising: decreasing the rate of dependence formation; decreasing the rate of withdrawal syndrome formation; blocking or inhibiting the development of somatovegetative symptoms of withdrawal syndrome; blocking or inhibiting the development of psychoneurological symptoms of withdrawal syndrome; blocking or inhibiting the formation of morbid attraction to alcohol and/or PASs; blocking or inhibiting the formation of the quantitative control loss symptom; inhibition of tolerance development; inhibition of the development of organ pathologies against high tolerance background; inhibition of the development of somatoneurological disorders against high tolerance background; inhibition of the development of vegetovascular, vegetoasthenic disorders against high tolerance background; inhibition of the development of psychopathy-like or psychic disorders against high tolerance background; reducing the reinforcing properties of ethanol and/or other ingested PASs; reducing the rate of development of organ pathologies, particularly of the brain, liver, myocardium; reducing the rate of development of somatoneurological disorders; inhibition of the development of psychopathy-like or psychic disorders; reducing the rate of lowering of cognitive-mnestic functions, in particular, preventing the origination of alcoholism amnesia. 4. The method according to claim 1, wherein the preparation contains a pharmaceutically suitable vehicle. 5. The method according to claim 4, wherein said vehicle comprises auxiliary substances selected from the group consisting of a binder, a filler, a lubricant, a disintegrant, a wetting agent, an inert solvent, a surfactant, a dispersant, a suspending agent, an emulsifying agent, an edible oil, flavorings, a food colorant or mixtures thereof. 6. The method according to claim 4, wherein the preparation is made in a form for topical administration via the oral mucosa, wherein the above-said vehicle comprises an ingredient selected from the group consisting of a flavoring agent, a carbohydrate, acacia, tragacanth, gelatin, glycerin and mixtures thereof. 7. The method according to claim 4, wherein the preparation is made in a form for nasal administration, wherein the above-said vehicle comprises an ingredient selected from the group consisting of a dispersing agent, a solubilizing agent, a suspending agent or mixtures thereof. 8. The method according to claim 4, wherein the preparation is made in a form for administration by inhalation, wherein the above-said vehicle comprises a propellant. 9. The method according to claim 1, which provides for administering the preparation comprising a vicinal dithioglycol of general formula (1), ensuring ingress of the preparation into the organism in an effective amount, wherein said amount is from about 5 mg to about 10 g per day. 10. The method according to claim 9, wherein said amount is from about 50 mg to about 2 g per day. 11. The method according to claim 10, wherein said amount is from about 100 mg to about 1000 mg per day. 12. The method according to claim 1, wherein the preparation further comprises at least one source of metal ions for maintaining the water-electrolyte balance and/or normal level of enzymatic activity and/or of neuroregulation and excitation or inhibition transmission systems. 13. The method according to claim 12, wherein said metal is a bivalent metal, such as calcium, magnesium, manganese, etc. 14. The method according to claim 1, wherein the preparation further comprises at least one ingredient selected from the group consisting of a vitamin, a vitamin precursor, a vitamin derivative, an organic acid, an organic acid derivative, a lipid, a physiologically active peptide, an amino acid, an amino acid derivative, an enzyme, an enzyme derivative, an enzyme precursor, a coenzyme, a coenzyme derivative, a coenzyme precursor, a carbohydrate, a protein, and mixtures thereof. 15. The method according to claim 1, wherein the preparation is intended for preventive or curative-preventive purposes in a condition wherein the origination or development of alcoholism and/or of dependence on psychoactive substances can take place. 16. A method for the treatment or prevention of conditions or pathological processes in which the formation of tetrahydroisoquinolines (THIQs) can take place in the organism, which method provides for preparing a composition comprising as at least one vicinal dithioglycol of general formula (1) for blocking and/or preventing the development and/or for reducing the rate of development of pathological processes caused by using alcohol-containing beverages and/or substances having an addictive potential. 17. The method according to claim 16, wherein said composition is used for attaining at least one effect selected from the group comprising: decreasing the rate of dependence formation; decreasing the rate of withdrawal syndrome formation; blocking or inhibiting the development of somatovegetative symptoms of withdrawal syndrome; blocking or inhibiting the development of psychoneurological symptoms of withdrawal syndrome; blocking or inhibiting the formation of morbid attraction to alcohol and/or PASs; blocking or inhibiting the formation of the quantitative control loss symptom; inhibition of tolerance development; inhibition of the development of organ pathologies against high tolerance background; inhibition of the development of somatoneurological disorders against high tolerance background; inhibition of the development of vegetovascular, vegetoasthenic disorders against high tolerance background; inhibition of the development of psychopathy-like or psychic disorders against high tolerance background; reducing the reinforcing properties of ethanol and/or other ingested PASs; reducing the rate of development of organ pathologies, particularly of the brain, liver, myocardium; reducing the rate of development of somatoneurological disorders; inhibition of the development of psychopathy-like or psychic disorders; reducing the rate of lowering of cognitive-mnestic functions, in particular, preventing the origination of alcoholism amnesia. 18. The use according to claim 17, wherein said vicinal dithioglycol is meso-2,3-dimercaptosuccinic acid (DMSA) and/or sodium 2,3-dimercaptopropane-1-sulfonate (DMPS). 19. A method for preventing the origination or development of alcoholism and/or of dependence on the intake of substances having an addictive potential, which method provides for administering at least one vicinal dithioglycol of general formula (1) as the agent for these purposes. 20. The method according to claim 19, wherein said vicinal dithioglycol is meso-2,3-dimercaptosuccinic acid (DMSA) and/or sodium 2,3-dimercaptopropane-1-sulfonate (DMPS).
The invention relates to medicine and is directed at developing means and methods for narcological practice and for preventive measures in order to reduce the risk of falling sick with such menacing modem diseases like dependence diseases, in particular alcoholism, narcomania and toximania. Said invention provides for using already known chemical compounds, i.e. vicinal dithioglycols for developing medical agents exhibiting properties for preventing contraction or development or reducing the speed of pathological processes forming a base for dependence diseases, in particular withdrawal syndromes. The inventive substances reduce or block pharmacological effects resulting from ethanol injection and/or narcotics, thereby directly influencing pathogenic mechanisms of the dependence diseases, in particular alcoholism. It is stated that said agents are particularly efficient when they are perorally administered. Said invention is accompanied by experimental material which proves the large perspective of the inventive agents and of a prophylactic method which provides for the use thereof before and after consuming alcohol-containing beverages or narcotics. The use of the invention makes it possible to reduce the degree of falling sick with alcoholism and narcomania and to reduce the toxic action of alcohols during the household use thereof.1. A method for the prevention of beginnings and/or for the prevention of development and/or for reducing the rate of development of pathological processes caused by the use of alcohol-containing beverages and/or substances having an addictive potential, comprising (a) providing a preparation comprising at least one vicinal dithioglycol of general chemical formula (1): R1CH(SH)CH(SH)R2  (1)where R1 is selected from the groups: (—H) or (—COOH), or (—SO3H); R2 is selected from the groups: (—H) or (—COOH), or (—OH), or (—CH2—COOH), or (—CH2—OH), or (—CH2—SO3H), or (—CH2—O—CH2—SO3H); or salts of derivatives thereof, comprising salt forming groups (—OH) or (—COOH), or (—SO3H); and (b) administering the preparation to a subject by peroral or nasal administration in an effective amount. 2. The method according to claim 1, wherein the substance having an addictive potential is a narcotic substance. 3. The method according to claim 1, wherein the preparation is used for attaining at least one effect selected from the group comprising: decreasing the rate of dependence formation; decreasing the rate of withdrawal syndrome formation; blocking or inhibiting the development of somatovegetative symptoms of withdrawal syndrome; blocking or inhibiting the development of psychoneurological symptoms of withdrawal syndrome; blocking or inhibiting the formation of morbid attraction to alcohol and/or PASs; blocking or inhibiting the formation of the quantitative control loss symptom; inhibition of tolerance development; inhibition of the development of organ pathologies against high tolerance background; inhibition of the development of somatoneurological disorders against high tolerance background; inhibition of the development of vegetovascular, vegetoasthenic disorders against high tolerance background; inhibition of the development of psychopathy-like or psychic disorders against high tolerance background; reducing the reinforcing properties of ethanol and/or other ingested PASs; reducing the rate of development of organ pathologies, particularly of the brain, liver, myocardium; reducing the rate of development of somatoneurological disorders; inhibition of the development of psychopathy-like or psychic disorders; reducing the rate of lowering of cognitive-mnestic functions, in particular, preventing the origination of alcoholism amnesia. 4. The method according to claim 1, wherein the preparation contains a pharmaceutically suitable vehicle. 5. The method according to claim 4, wherein said vehicle comprises auxiliary substances selected from the group consisting of a binder, a filler, a lubricant, a disintegrant, a wetting agent, an inert solvent, a surfactant, a dispersant, a suspending agent, an emulsifying agent, an edible oil, flavorings, a food colorant or mixtures thereof. 6. The method according to claim 4, wherein the preparation is made in a form for topical administration via the oral mucosa, wherein the above-said vehicle comprises an ingredient selected from the group consisting of a flavoring agent, a carbohydrate, acacia, tragacanth, gelatin, glycerin and mixtures thereof. 7. The method according to claim 4, wherein the preparation is made in a form for nasal administration, wherein the above-said vehicle comprises an ingredient selected from the group consisting of a dispersing agent, a solubilizing agent, a suspending agent or mixtures thereof. 8. The method according to claim 4, wherein the preparation is made in a form for administration by inhalation, wherein the above-said vehicle comprises a propellant. 9. The method according to claim 1, which provides for administering the preparation comprising a vicinal dithioglycol of general formula (1), ensuring ingress of the preparation into the organism in an effective amount, wherein said amount is from about 5 mg to about 10 g per day. 10. The method according to claim 9, wherein said amount is from about 50 mg to about 2 g per day. 11. The method according to claim 10, wherein said amount is from about 100 mg to about 1000 mg per day. 12. The method according to claim 1, wherein the preparation further comprises at least one source of metal ions for maintaining the water-electrolyte balance and/or normal level of enzymatic activity and/or of neuroregulation and excitation or inhibition transmission systems. 13. The method according to claim 12, wherein said metal is a bivalent metal, such as calcium, magnesium, manganese, etc. 14. The method according to claim 1, wherein the preparation further comprises at least one ingredient selected from the group consisting of a vitamin, a vitamin precursor, a vitamin derivative, an organic acid, an organic acid derivative, a lipid, a physiologically active peptide, an amino acid, an amino acid derivative, an enzyme, an enzyme derivative, an enzyme precursor, a coenzyme, a coenzyme derivative, a coenzyme precursor, a carbohydrate, a protein, and mixtures thereof. 15. The method according to claim 1, wherein the preparation is intended for preventive or curative-preventive purposes in a condition wherein the origination or development of alcoholism and/or of dependence on psychoactive substances can take place. 16. A method for the treatment or prevention of conditions or pathological processes in which the formation of tetrahydroisoquinolines (THIQs) can take place in the organism, which method provides for preparing a composition comprising as at least one vicinal dithioglycol of general formula (1) for blocking and/or preventing the development and/or for reducing the rate of development of pathological processes caused by using alcohol-containing beverages and/or substances having an addictive potential. 17. The method according to claim 16, wherein said composition is used for attaining at least one effect selected from the group comprising: decreasing the rate of dependence formation; decreasing the rate of withdrawal syndrome formation; blocking or inhibiting the development of somatovegetative symptoms of withdrawal syndrome; blocking or inhibiting the development of psychoneurological symptoms of withdrawal syndrome; blocking or inhibiting the formation of morbid attraction to alcohol and/or PASs; blocking or inhibiting the formation of the quantitative control loss symptom; inhibition of tolerance development; inhibition of the development of organ pathologies against high tolerance background; inhibition of the development of somatoneurological disorders against high tolerance background; inhibition of the development of vegetovascular, vegetoasthenic disorders against high tolerance background; inhibition of the development of psychopathy-like or psychic disorders against high tolerance background; reducing the reinforcing properties of ethanol and/or other ingested PASs; reducing the rate of development of organ pathologies, particularly of the brain, liver, myocardium; reducing the rate of development of somatoneurological disorders; inhibition of the development of psychopathy-like or psychic disorders; reducing the rate of lowering of cognitive-mnestic functions, in particular, preventing the origination of alcoholism amnesia. 18. The use according to claim 17, wherein said vicinal dithioglycol is meso-2,3-dimercaptosuccinic acid (DMSA) and/or sodium 2,3-dimercaptopropane-1-sulfonate (DMPS). 19. A method for preventing the origination or development of alcoholism and/or of dependence on the intake of substances having an addictive potential, which method provides for administering at least one vicinal dithioglycol of general formula (1) as the agent for these purposes. 20. The method according to claim 19, wherein said vicinal dithioglycol is meso-2,3-dimercaptosuccinic acid (DMSA) and/or sodium 2,3-dimercaptopropane-1-sulfonate (DMPS).
1,600
346
13,622,263
1,645
Methods for generating immune responses using adenovirus vectors that allow multiple vaccinations with the same adenovirus vector and vaccinations in individuals with preexisting immunity to adenovirus are provided.
1.-36. (canceled) 37. A composition for immunizing a subject suffering from or at risk of a condition, the composition comprising: a replication defective adenovirus vector comprising a deletion in the E2b region and one or more nucleic acids encoding a variant of an antigen associated with said condition or a fragment thereof. 38. The method of claim 37, wherein the antigen is a cancer target antigen. 39. The method of claim 37, wherein the antigen is CEA. 40. The method of claim 37, wherein the antigen is Her2/neu. 41. The method of claim 37, wherein the antigen is MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A10, or MAGE-A12. 42. The method of claim 37, wherein the antigen is HPV E6 or HPV E7. 43. The method of claim 37, wherein the antigen is MUC1 or MUC2. 44. The method of claim 37, wherein the antigen is selected from the group consisting of WT1, p53, BAGE, DAM-6, DAM-10, GAGE-1, GAGE-2, GAGE-8, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7B, NA88-A, NY-ESO-1, MART-1, MC1R, Gp100, PSA, PSM, Tyrosinase, TRP-1, TRP-2, ART-4, CAMEL, Cyp-B, BRCA1, hTERT, hTRT, iCE, PRAME, P15, RU1, RU2, SART-1, SART-3, WT1, AFP, β-catenin/m, Caspase-8/m, CDK-4/m, ELF2M, GnT-V, G250, HSP70-2M, HST-2, KIAA0205, MUM-1, MUM-2, MUM-3, Myosin/m, RAGE, SART-2, TRP-2/INT2, 707-AP, Annexin II, CDC27/m, TPI/mbcr-abl, ETV6/AML, LDLR/FUT, Pml/RARα, TEL/AML1, influenza virus haemagglutinin, influenza virus neuraminidase, influenza virus matrix protein M1, and HIV-gag. 45. The composition of claim 37, wherein the replication defective adenovirus vector further comprises a deletion in the E1 region. 46. The composition of claim 37, wherein the one or more nucleic acids are inserted in the E1 region. 47. The composition of claim 37, wherein the replication defective adenovirus vector further comprises a deletion in the E3 region. 48. The composition of claim 37, wherein the one or more nucleic acids are inserted in the E3 region. 49. The composition of claim 37, wherein the replication defective adenovirus vector is derived from Adenovirus serotype 5. 50. The composition of claim 37, wherein the composition comprises the replication defective adenovirus vector at a concentration of at least 1010 virus particles/ml. 51. The composition of claim 37, wherein the replication defective adenovirus vector is not helper dependent. 52. The composition of claim 37, further comprising an immunostimulant. 53. The composition of claim 37, further comprising a nucleic acid encoding an immunostimulant in a second vector, wherein the second vector comprises a deletion in the E2b region. 54. The composition of claim 53, wherein the second vector further comprises a deletion in the E 1 region. 55. The composition of claim 53, wherein the second vector further comprises a deletion in the E3 region. 56. The composition of claim 53, wherein the second vector is derived from Adenovirus serotype 5. 57. The composition of claim 52 or 53, wherein the immunostimulant is selected from the group consisting of granulocyte macrophage colony-stimulating factor (GM-CSF), granulocyte-colony stimulating factor (G-CSF), interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), interleukin-2 (IL-2), IL-7, IL-12, IL-4, IL-5, IL-6, IL-10, and IL-12. 58. The composition of claim 44, wherein the influenza virus selected from the group consisting of: H5N1, H3N2, H9N1, H1N1, H2N2, H7N7, H1N2, H9N2, H7N2, H7N3, and H10N7. 59. The composition of claim 37, wherein the composition is suitable for intramuscular, subcutaneous, or intradermal administration. 60. The composition of claim 37, wherein the one or more nucleic acids further encode an immunological fusion partner. 61. The composition of claim 60, wherein the immunological fusion partner comprises a Mycobacterium tuberculosis-derived Ra12 fragment, a protein D derivative, or LYTA. 62. A method of inducing an antigen-specific immune, immunoprotective, or immunotherapeutic response in a subject suffering from or at risk of a condition comprising: administering the composition of claim 37 to said subject.
Methods for generating immune responses using adenovirus vectors that allow multiple vaccinations with the same adenovirus vector and vaccinations in individuals with preexisting immunity to adenovirus are provided.1.-36. (canceled) 37. A composition for immunizing a subject suffering from or at risk of a condition, the composition comprising: a replication defective adenovirus vector comprising a deletion in the E2b region and one or more nucleic acids encoding a variant of an antigen associated with said condition or a fragment thereof. 38. The method of claim 37, wherein the antigen is a cancer target antigen. 39. The method of claim 37, wherein the antigen is CEA. 40. The method of claim 37, wherein the antigen is Her2/neu. 41. The method of claim 37, wherein the antigen is MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A10, or MAGE-A12. 42. The method of claim 37, wherein the antigen is HPV E6 or HPV E7. 43. The method of claim 37, wherein the antigen is MUC1 or MUC2. 44. The method of claim 37, wherein the antigen is selected from the group consisting of WT1, p53, BAGE, DAM-6, DAM-10, GAGE-1, GAGE-2, GAGE-8, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7B, NA88-A, NY-ESO-1, MART-1, MC1R, Gp100, PSA, PSM, Tyrosinase, TRP-1, TRP-2, ART-4, CAMEL, Cyp-B, BRCA1, hTERT, hTRT, iCE, PRAME, P15, RU1, RU2, SART-1, SART-3, WT1, AFP, β-catenin/m, Caspase-8/m, CDK-4/m, ELF2M, GnT-V, G250, HSP70-2M, HST-2, KIAA0205, MUM-1, MUM-2, MUM-3, Myosin/m, RAGE, SART-2, TRP-2/INT2, 707-AP, Annexin II, CDC27/m, TPI/mbcr-abl, ETV6/AML, LDLR/FUT, Pml/RARα, TEL/AML1, influenza virus haemagglutinin, influenza virus neuraminidase, influenza virus matrix protein M1, and HIV-gag. 45. The composition of claim 37, wherein the replication defective adenovirus vector further comprises a deletion in the E1 region. 46. The composition of claim 37, wherein the one or more nucleic acids are inserted in the E1 region. 47. The composition of claim 37, wherein the replication defective adenovirus vector further comprises a deletion in the E3 region. 48. The composition of claim 37, wherein the one or more nucleic acids are inserted in the E3 region. 49. The composition of claim 37, wherein the replication defective adenovirus vector is derived from Adenovirus serotype 5. 50. The composition of claim 37, wherein the composition comprises the replication defective adenovirus vector at a concentration of at least 1010 virus particles/ml. 51. The composition of claim 37, wherein the replication defective adenovirus vector is not helper dependent. 52. The composition of claim 37, further comprising an immunostimulant. 53. The composition of claim 37, further comprising a nucleic acid encoding an immunostimulant in a second vector, wherein the second vector comprises a deletion in the E2b region. 54. The composition of claim 53, wherein the second vector further comprises a deletion in the E 1 region. 55. The composition of claim 53, wherein the second vector further comprises a deletion in the E3 region. 56. The composition of claim 53, wherein the second vector is derived from Adenovirus serotype 5. 57. The composition of claim 52 or 53, wherein the immunostimulant is selected from the group consisting of granulocyte macrophage colony-stimulating factor (GM-CSF), granulocyte-colony stimulating factor (G-CSF), interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), interleukin-2 (IL-2), IL-7, IL-12, IL-4, IL-5, IL-6, IL-10, and IL-12. 58. The composition of claim 44, wherein the influenza virus selected from the group consisting of: H5N1, H3N2, H9N1, H1N1, H2N2, H7N7, H1N2, H9N2, H7N2, H7N3, and H10N7. 59. The composition of claim 37, wherein the composition is suitable for intramuscular, subcutaneous, or intradermal administration. 60. The composition of claim 37, wherein the one or more nucleic acids further encode an immunological fusion partner. 61. The composition of claim 60, wherein the immunological fusion partner comprises a Mycobacterium tuberculosis-derived Ra12 fragment, a protein D derivative, or LYTA. 62. A method of inducing an antigen-specific immune, immunoprotective, or immunotherapeutic response in a subject suffering from or at risk of a condition comprising: administering the composition of claim 37 to said subject.
1,600
347
15,485,026
1,627
A method for treating a subject afflicted with dementia is disclosed. The method comprises administering to the subject a composition comprising an effective amount of a benzoic acid, a salt, an ester, or a derivative thereof, and a pharmaceutically acceptable carrier or vehicle, wherein the subject is not administered any other neuropharmaceutical.
1. A method of treating dementia, comprising: administering to a patient with dementia, excluding a patient with a Clinical Dementia Rating (CDR) score of 0.5, a therapeutically effective amount of benzoic acid, a salt, an ester, or a derivative thereof. 2. The method of claim 1, further comprising administering an acetylcholine esterase inhibitor to the patient with dementia excluding the patient with the CDR score of 0.5. 3. The method of claim 1, wherein the administering step is performed daily for a period selected from the group consisting of more than 8 weeks, at least 16 weeks, at least 20 weeks, and at least 24 weeks. 4. The method of claim 1, wherein the therapeutically effective amount is 200-2000 mg per daily dosage unit. 5. The method of claim 1, wherein the therapeutically effective amount is 500-750 mg per daily dosage unit. 6. The method of claim 1, wherein the patient with dementia has a CDR score of 1. 7. A method for improvement in Clinician's Interview-Based Impression of Change plus Caregiver Input (CIBIC-plus) score in a dementia patient with a Clinical Dementia Rating (CDR) score of 1, comprising: administering to the patient with the CDR score of 1 a therapeutically effective amount of benzoic acid, a salt, an ester, or a derivative thereof for at least 24 weeks. 8. The method of claim 7, wherein the therapeutically effective amount is 200-2000 mg per daily dosage unit. 9. A method of treating a dementia patient with a Clinical Dementia Rating (CDR) score of 1, comprising: administering to the dementia patient with the CDR score of 1 a therapeutically effective amount of benzoic acid, a salt, an ester, or a derivative thereof, wherein the subject is optionally concurrently administered an acetylcholine esterase inhibitor hat not administered any other neuropharmaceutical. 10. The method of claim 9, wherein the administering step is performed daily for a period selected from the group consisting of more than 8 weeks, at least 16 weeks, at least 20 weeks, and at least 24 weeks. 11. The method of claim 9, wherein the salt thereof is sodium benzoate. 12. The method of claim 9, wherein the therapeutically effective amount is at least 250 mg per daily dosage unit. 13. The method of claim 12, wherein the therapeutically effective amount is at least 500 mg per daily dosage unit. 14. The method of claim 9, wherein the therapeutically effective amount is 200-2000 mg per daily dosage unit. 15. The method of claim 9, wherein the salt thereof is sodium benzoate and the therapeutically effective amount is 250-750 mg per daily dosage unit. 16. The method of claim 15, wherein the therapeutically effective amount is 500-750 mg per daily dosage unit. 17. The method of claim 9, wherein the administering step comprises: (a) administering to the patient sodium benzoate about 200-500 mg per daily dosage unit for the first treatment period; (b) administering to the patient sodium benzoate about 400-1000 per daily dosage unit fir the second treatment period; and (c) administering to the patient sodium benzoate about 500-1000 per daily dosage unit for the third treatment period. 18. The method of claim 9, wherein the patient is afflicted with Alzheimer disease or mild cognitive impairment. 19. The method of claim 9, wherein the patient is afflicted with an early phase Alzheimer disease or amnestic mild cognitive impairment. 20. The method of claim 9, wherein the therapeutically effective amount s greater than 1000 mg and less than or equal to 2000 mg per daily dosage unit.
A method for treating a subject afflicted with dementia is disclosed. The method comprises administering to the subject a composition comprising an effective amount of a benzoic acid, a salt, an ester, or a derivative thereof, and a pharmaceutically acceptable carrier or vehicle, wherein the subject is not administered any other neuropharmaceutical.1. A method of treating dementia, comprising: administering to a patient with dementia, excluding a patient with a Clinical Dementia Rating (CDR) score of 0.5, a therapeutically effective amount of benzoic acid, a salt, an ester, or a derivative thereof. 2. The method of claim 1, further comprising administering an acetylcholine esterase inhibitor to the patient with dementia excluding the patient with the CDR score of 0.5. 3. The method of claim 1, wherein the administering step is performed daily for a period selected from the group consisting of more than 8 weeks, at least 16 weeks, at least 20 weeks, and at least 24 weeks. 4. The method of claim 1, wherein the therapeutically effective amount is 200-2000 mg per daily dosage unit. 5. The method of claim 1, wherein the therapeutically effective amount is 500-750 mg per daily dosage unit. 6. The method of claim 1, wherein the patient with dementia has a CDR score of 1. 7. A method for improvement in Clinician's Interview-Based Impression of Change plus Caregiver Input (CIBIC-plus) score in a dementia patient with a Clinical Dementia Rating (CDR) score of 1, comprising: administering to the patient with the CDR score of 1 a therapeutically effective amount of benzoic acid, a salt, an ester, or a derivative thereof for at least 24 weeks. 8. The method of claim 7, wherein the therapeutically effective amount is 200-2000 mg per daily dosage unit. 9. A method of treating a dementia patient with a Clinical Dementia Rating (CDR) score of 1, comprising: administering to the dementia patient with the CDR score of 1 a therapeutically effective amount of benzoic acid, a salt, an ester, or a derivative thereof, wherein the subject is optionally concurrently administered an acetylcholine esterase inhibitor hat not administered any other neuropharmaceutical. 10. The method of claim 9, wherein the administering step is performed daily for a period selected from the group consisting of more than 8 weeks, at least 16 weeks, at least 20 weeks, and at least 24 weeks. 11. The method of claim 9, wherein the salt thereof is sodium benzoate. 12. The method of claim 9, wherein the therapeutically effective amount is at least 250 mg per daily dosage unit. 13. The method of claim 12, wherein the therapeutically effective amount is at least 500 mg per daily dosage unit. 14. The method of claim 9, wherein the therapeutically effective amount is 200-2000 mg per daily dosage unit. 15. The method of claim 9, wherein the salt thereof is sodium benzoate and the therapeutically effective amount is 250-750 mg per daily dosage unit. 16. The method of claim 15, wherein the therapeutically effective amount is 500-750 mg per daily dosage unit. 17. The method of claim 9, wherein the administering step comprises: (a) administering to the patient sodium benzoate about 200-500 mg per daily dosage unit for the first treatment period; (b) administering to the patient sodium benzoate about 400-1000 per daily dosage unit fir the second treatment period; and (c) administering to the patient sodium benzoate about 500-1000 per daily dosage unit for the third treatment period. 18. The method of claim 9, wherein the patient is afflicted with Alzheimer disease or mild cognitive impairment. 19. The method of claim 9, wherein the patient is afflicted with an early phase Alzheimer disease or amnestic mild cognitive impairment. 20. The method of claim 9, wherein the therapeutically effective amount s greater than 1000 mg and less than or equal to 2000 mg per daily dosage unit.
1,600
348
14,045,683
1,656
The present invention relates to a method of identifying a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) a yeast cell comprising: a) providing a mutated yeast cell comprising a deletion of at least one gene of the (PDH) by-pass, selected from the genes encoding the enzymes pyruvate decarboxylase (PDC), acetaldehyde dehydrogenase (ALD), and acetyl-CoA synthetase (ACS); b) transforming said mutated yeast cell with an expression vector comprising a heterologous nucleotide sequence encoding a candidate polypeptide having potential enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA; c) testing said recombinant mutated yeast cell for its ability to grow on minimal medium containing glucose as sole carbon source, and d) identifying said candidate polypeptide as a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) said yeast cell when growth of said cell is observed. The invention further relates to a method of producing a fermentation production such as butanol.
1. A method of identifying a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) a yeast cell comprising: providing a mutated yeast cell, wherein said mutation comprises an inactivation of at least one gene of the (PDH) by-pass, selected from the genes encoding the enzymes pyruvate decarboxylase (PDC), acetaldehyde dehydrogenase (ALD), and acetyl-CoA synthetase (ACS); transforming said mutated yeast cell with an expression vector comprising at least one heterologous nucleotide sequence operably linked to a promoter functional in yeast and said heterologous nucleotide sequence encoding a candidate polypeptide having potential enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl CoA; testing said recombinant mutated yeast cell for its ability to grow on minimal medium containing glucose as sole carbon source, and identifying said candidate polypeptide as a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl CoA in (the cytosol of) said yeast cell when growth of said cell is observed. 2. Method according to claim 1, wherein said yeast cell is a cell of Saccharomyces cerevisiae and wherein said heterologous nucleotide sequence is codon pair optimized for expression in Saccharomyces cerevisiae. 3. Method according to claim 2, wherein said mutation comprises an inactivation of the gene for acetyl-CoA synthetase isoform 2 (acs2). 4. Method according to claim 1, wherein said candidate polypeptide having enzymatic activity for converting acetaldehyde into acetyl-CoA is a (putative) acetylating acetaldehyde dehydrogenase (acdh). 5. A vector for the expression of heterologous polypeptides in yeast, said vector comprising a heterologous nucleotide sequence operably linked to a promoter functional in yeast and said heterologous nucleotide sequence encoding a polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) said yeast cell. 6. Vector according to claim 5, wherein said polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA. 7. Vector according to claim 5, wherein said polypeptide has more than 50%, preferably more than 60%, 70%, 80%, 90%, or 95% sequence identity with the amino acid sequence selected from SEQ ID NO: 19, 22, 25, 28 and 52. 8. Vector according to claim 5, for expression in Saccharomyces cerevisiae, wherein said heterologous nucleotide sequence is codon pair optimized for expression in Saccharomyces cerevisiae. 9. Expression vector according to claim 8, wherein said heterologous nucleotide sequence is selected from SEQ ID NO: 20, 23, 26, 29 and 51. 10. A recombinant yeast cell comprising a vector of claim 5. 11. A recombinant yeast cell comprising a heterologous nucleotide sequence encoding a polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) said yeast cell. 12. Yeast cell according to claim 10, further comprising an inactivation of at least one gene of the (PDH) by-pass, selected from the genes encoding the enzymes pyruvate decarboxylase (PDC), acetaldehyde dehydrogenase (ALD), and acetyl-CoA synthetase (ACS). 13. Yeast cell according to claim 10, wherein the yeast cell comprises an inactivation of a gene encoding an acetyl-CoA synthase. 14. Yeast cell according to claim 10, wherein said cell shows growth on minimal medium containing glucose as sole carbon source. 15. Yeast cell according to claim 10, further comprising an inactivation of a gene encoding an enzyme that catalyses the conversion of acetaldehyde into ethanol, preferably an alcohol dehydrogenase. 16. Yeast cell according to claim 10, further comprising one or more introduced genes encoding a recombinant pathway for the formation of 1-butanol from acetyl-CoA. 17. Yeast cell according to claim 16, wherein said one or more introduced genes encode enzymes that produce acetoacetyl-CoA, 3-hydroxybutyryl-CoA, crotonyl-CoA, butyryl-CoA, butylaldehyde and/or 1-butanol. 18. Yeast cell according to claim 10, wherein said yeast is Saccharomyces cerevisiae 19. A method of producing a fermentation product, comprising the steps of fermenting a suitable carbon substrate with a yeast cell according to claim 10 and recovering the fermentation product produced during said fermentation. 20. Method according to claim 19, wherein the fermentation product is butanol.
The present invention relates to a method of identifying a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) a yeast cell comprising: a) providing a mutated yeast cell comprising a deletion of at least one gene of the (PDH) by-pass, selected from the genes encoding the enzymes pyruvate decarboxylase (PDC), acetaldehyde dehydrogenase (ALD), and acetyl-CoA synthetase (ACS); b) transforming said mutated yeast cell with an expression vector comprising a heterologous nucleotide sequence encoding a candidate polypeptide having potential enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA; c) testing said recombinant mutated yeast cell for its ability to grow on minimal medium containing glucose as sole carbon source, and d) identifying said candidate polypeptide as a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) said yeast cell when growth of said cell is observed. The invention further relates to a method of producing a fermentation production such as butanol.1. A method of identifying a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) a yeast cell comprising: providing a mutated yeast cell, wherein said mutation comprises an inactivation of at least one gene of the (PDH) by-pass, selected from the genes encoding the enzymes pyruvate decarboxylase (PDC), acetaldehyde dehydrogenase (ALD), and acetyl-CoA synthetase (ACS); transforming said mutated yeast cell with an expression vector comprising at least one heterologous nucleotide sequence operably linked to a promoter functional in yeast and said heterologous nucleotide sequence encoding a candidate polypeptide having potential enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl CoA; testing said recombinant mutated yeast cell for its ability to grow on minimal medium containing glucose as sole carbon source, and identifying said candidate polypeptide as a heterologous polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl CoA in (the cytosol of) said yeast cell when growth of said cell is observed. 2. Method according to claim 1, wherein said yeast cell is a cell of Saccharomyces cerevisiae and wherein said heterologous nucleotide sequence is codon pair optimized for expression in Saccharomyces cerevisiae. 3. Method according to claim 2, wherein said mutation comprises an inactivation of the gene for acetyl-CoA synthetase isoform 2 (acs2). 4. Method according to claim 1, wherein said candidate polypeptide having enzymatic activity for converting acetaldehyde into acetyl-CoA is a (putative) acetylating acetaldehyde dehydrogenase (acdh). 5. A vector for the expression of heterologous polypeptides in yeast, said vector comprising a heterologous nucleotide sequence operably linked to a promoter functional in yeast and said heterologous nucleotide sequence encoding a polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) said yeast cell. 6. Vector according to claim 5, wherein said polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA. 7. Vector according to claim 5, wherein said polypeptide has more than 50%, preferably more than 60%, 70%, 80%, 90%, or 95% sequence identity with the amino acid sequence selected from SEQ ID NO: 19, 22, 25, 28 and 52. 8. Vector according to claim 5, for expression in Saccharomyces cerevisiae, wherein said heterologous nucleotide sequence is codon pair optimized for expression in Saccharomyces cerevisiae. 9. Expression vector according to claim 8, wherein said heterologous nucleotide sequence is selected from SEQ ID NO: 20, 23, 26, 29 and 51. 10. A recombinant yeast cell comprising a vector of claim 5. 11. A recombinant yeast cell comprising a heterologous nucleotide sequence encoding a polypeptide having enzymatic activity for converting pyruvate, acetaldehyde or acetate into acetyl-CoA in (the cytosol of) said yeast cell. 12. Yeast cell according to claim 10, further comprising an inactivation of at least one gene of the (PDH) by-pass, selected from the genes encoding the enzymes pyruvate decarboxylase (PDC), acetaldehyde dehydrogenase (ALD), and acetyl-CoA synthetase (ACS). 13. Yeast cell according to claim 10, wherein the yeast cell comprises an inactivation of a gene encoding an acetyl-CoA synthase. 14. Yeast cell according to claim 10, wherein said cell shows growth on minimal medium containing glucose as sole carbon source. 15. Yeast cell according to claim 10, further comprising an inactivation of a gene encoding an enzyme that catalyses the conversion of acetaldehyde into ethanol, preferably an alcohol dehydrogenase. 16. Yeast cell according to claim 10, further comprising one or more introduced genes encoding a recombinant pathway for the formation of 1-butanol from acetyl-CoA. 17. Yeast cell according to claim 16, wherein said one or more introduced genes encode enzymes that produce acetoacetyl-CoA, 3-hydroxybutyryl-CoA, crotonyl-CoA, butyryl-CoA, butylaldehyde and/or 1-butanol. 18. Yeast cell according to claim 10, wherein said yeast is Saccharomyces cerevisiae 19. A method of producing a fermentation product, comprising the steps of fermenting a suitable carbon substrate with a yeast cell according to claim 10 and recovering the fermentation product produced during said fermentation. 20. Method according to claim 19, wherein the fermentation product is butanol.
1,600
349
15,203,616
1,642
Methods for diagnosing pathology of the liver in a subject suspected of having such pathology are disclosed. The methods comprise quantifiably detecting lectin binding on proteins in biological fluids, and comparing the detected lectin binding with reference values for the binding of lectin of such proteins in healthy or disease states.
1. A method for diagnosing a liver pathology in a subject suspected of having the liver pathology comprising: obtaining biological fluid from the subject; contacting said biological fluid with a fucose-binding lectin and allowing said lectin to bind to a protein in said biological fluid, wherein said protein is selected from Hemopexin, HBsAg, hepatitis B viral particle, alpha-1-antichymotrypsin, alpha-1-antichymotrypsin His-Pro-less, Ceruloplasmin, alpha-2-macroglobulin, alpha-2-HS-glycoprotein, Haptoglobin, Fibrinogen gamma chain precursor, immunoglobulin, APO-D, Kininogen, Histidine rich glycoprotein, complement factor 1 precursor, complement factor I heavy chain, complement factor I light chain, Complement C1s, Complement factor B precursor, complement factor B Ba fragment, Complement factor B Bb fragment, Complement C3 precursor, Complement C3 beta chain, Complement C3 alpha chain, C3a anaphylatoxin, Complement, C3b alpha′ chain, Complement C3c fragment, Complement C3dg fragment, Complement C3g fragment, Complement C3d fragment, Complement C3f fragment, Complement C5, Complement C5 beta chain, Complement C5 alpha chain, C5a anaphylatoxin, Complement C5 alpha′ chain, Complement C7, alpha-1 B glycoprotein, B-2-glycoprotein, Vitamin D-binding protein, Inter-alpha-trypsin inhibitor heavy chain H2, Alpha-1B-glycoprotein, Angiotensinogen precursor, Angiotensin-1, Angiotensin-2, Angiotensin-3, GARP protein, beta-2-glycoprotein, Clusterin (Apo J), Integrin alpha-8 precursor glycoprotein, Integrin alpha-8 heavy chain, Integrin alpha-8 light chain, hepatitis C viral particle, and Leucine-rich repeat-containing protein 32 precursor; quantifiably detecting bound lectin in the biological fluid to obtain a detected bound lectin value; and comparing the detected bound lectin value with a reference value for lectin binding on said protein in a comparable biological fluid of subjects without said liver pathology, said detected bound lectin value relative to the reference value being indicative of the presence or absence of said liver pathology, wherein a detected bound lectin value that deviates from said reference value to a statistically significant degree is indicative of the presence of said liver pathology, or comparing the detected bound lectin value with a reference value for lectin binding on said protein in a comparable biological fluid of subjects in which said liver pathology is known to be present, said detected bound lectin value relative to the reference value being indicative of the presence or absence of said liver pathology, wherein a detected bound lectin value that deviates from said reference value to a statistically significant degree is indicative of the absence of said liver pathology. 2. The method according to claim 1 wherein said liver pathology is cirrhosis. 3. The method according to claim 1 wherein said liver pathology is fibrosis. 4. The method according to claim 1 wherein said liver pathology is hepatocellular carcinoma. 5. The method according to claim 1 wherein said liver pathology is hepatitis. 6. The method of claim 1 wherein the biological fluid is whole blood, serum, urine, saliva, tears, or mucous. 7. The method of claim 1, wherein the lectin is coupled to a detectable moiety.
Methods for diagnosing pathology of the liver in a subject suspected of having such pathology are disclosed. The methods comprise quantifiably detecting lectin binding on proteins in biological fluids, and comparing the detected lectin binding with reference values for the binding of lectin of such proteins in healthy or disease states.1. A method for diagnosing a liver pathology in a subject suspected of having the liver pathology comprising: obtaining biological fluid from the subject; contacting said biological fluid with a fucose-binding lectin and allowing said lectin to bind to a protein in said biological fluid, wherein said protein is selected from Hemopexin, HBsAg, hepatitis B viral particle, alpha-1-antichymotrypsin, alpha-1-antichymotrypsin His-Pro-less, Ceruloplasmin, alpha-2-macroglobulin, alpha-2-HS-glycoprotein, Haptoglobin, Fibrinogen gamma chain precursor, immunoglobulin, APO-D, Kininogen, Histidine rich glycoprotein, complement factor 1 precursor, complement factor I heavy chain, complement factor I light chain, Complement C1s, Complement factor B precursor, complement factor B Ba fragment, Complement factor B Bb fragment, Complement C3 precursor, Complement C3 beta chain, Complement C3 alpha chain, C3a anaphylatoxin, Complement, C3b alpha′ chain, Complement C3c fragment, Complement C3dg fragment, Complement C3g fragment, Complement C3d fragment, Complement C3f fragment, Complement C5, Complement C5 beta chain, Complement C5 alpha chain, C5a anaphylatoxin, Complement C5 alpha′ chain, Complement C7, alpha-1 B glycoprotein, B-2-glycoprotein, Vitamin D-binding protein, Inter-alpha-trypsin inhibitor heavy chain H2, Alpha-1B-glycoprotein, Angiotensinogen precursor, Angiotensin-1, Angiotensin-2, Angiotensin-3, GARP protein, beta-2-glycoprotein, Clusterin (Apo J), Integrin alpha-8 precursor glycoprotein, Integrin alpha-8 heavy chain, Integrin alpha-8 light chain, hepatitis C viral particle, and Leucine-rich repeat-containing protein 32 precursor; quantifiably detecting bound lectin in the biological fluid to obtain a detected bound lectin value; and comparing the detected bound lectin value with a reference value for lectin binding on said protein in a comparable biological fluid of subjects without said liver pathology, said detected bound lectin value relative to the reference value being indicative of the presence or absence of said liver pathology, wherein a detected bound lectin value that deviates from said reference value to a statistically significant degree is indicative of the presence of said liver pathology, or comparing the detected bound lectin value with a reference value for lectin binding on said protein in a comparable biological fluid of subjects in which said liver pathology is known to be present, said detected bound lectin value relative to the reference value being indicative of the presence or absence of said liver pathology, wherein a detected bound lectin value that deviates from said reference value to a statistically significant degree is indicative of the absence of said liver pathology. 2. The method according to claim 1 wherein said liver pathology is cirrhosis. 3. The method according to claim 1 wherein said liver pathology is fibrosis. 4. The method according to claim 1 wherein said liver pathology is hepatocellular carcinoma. 5. The method according to claim 1 wherein said liver pathology is hepatitis. 6. The method of claim 1 wherein the biological fluid is whole blood, serum, urine, saliva, tears, or mucous. 7. The method of claim 1, wherein the lectin is coupled to a detectable moiety.
1,600
350
15,206,623
1,619
A method of inhibiting PAR2 activation of keratinocytes by boosting the PAR2 inhibition ability of a vitamin B3 compound and an N-acyl amino acid compound with a Laminaria Saccharina extract. The method may be used to improve the appearance of hyperpigmented spots on skin. By determining the amount of Laminaria Saccharina extract needed to boost PAR2 inhibition ability of a vitamin B3 compound and an N-acyl amino acid compound, improved skin care composition can be formulated for the treatment of hyperpigmented skin.
1. A method of boosting the ability of a vitamin B3 compound and an N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes, comprising: a. identifying one or more keratinocytes in which inhibition of PAR2 activation is desired; b. applying an effective amount of a vitamin B3 compound and an N-acyl amino acid compound to the keratinocytes during a treatment period, wherein the amount of vitamin B3 compound and N-acyl amino acid compound is sufficient to inhibit PAR2 activation of the keratinocytes during the treatment period; and c. applying an effective amount of a Laminaria Saccharina extract to the keratinocytes, wherein the amount of Laminaria Saccharina extract is sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes relative to treatment without the Laminaria Saccharina extract. 2. The method of claim 1, wherein the vitamin B3 compound comprises niacinamide. 3. The method of claim 1, wherein the N-acyl amino acid compound comprises a N-acyl phenylalanine corresponding to the following formula: wherein R1 can be C1 to C30, saturated or unsaturated, straight or branched, substituted or unsubstituted alkyls; substituted or unsubstituted aromatic groups; or mixtures thereof. 4. The method of claim 3, wherein the N-acyl amino acid compound comprises N-undecylenoyl-L-phenylalanine. 5. The method of claim 1, wherein the N-acyl amino acid compound comprises an N-acyl tyrosine corresponds to the following formula: wherein R1 can be C1 to C30, saturated or unsaturated, straight or branched, substituted or unsubstituted alkyls; substituted or unsubstituted aromatic groups; or mixtures thereof. 6. The method of claim 1, wherein the effective amount of Laminaria Saccharina is sufficient to sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes by at least about 10%. 7. The method of claim 6, wherein the effective amount of Laminaria Saccharina is sufficient to sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes by at least about 50%. 8. A method of boosting the ability of a vitamin B3 compound and an N-acyl amino acid compound to improve the appearance of hyperpigmented skin, comprising: a. identifying a hperpigmented portion of skin; b. applying an effective amount of a vitamin B3 compound and an N-acyl amino acid compound to the hyperpigmented portion of skin during a treatment period, wherein the treatment period is sufficient for the vitamin B3 compound and N-acyl amino acid compound to improve the appearance of the hyperpigmentation; and c. applying an effective amount of a Laminaria Saccharina extract to the target portion of skin, wherein the amount of Laminaria Saccharina extract is sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to improve the appearance of the hyperpigmented portion of skin relative to treatment without the Laminaria Saccharina extract. 9. The method of claim 8, wherein the vitamin B3 compound, N-acyl amino acid compound, and Laminaria Saccharina extract are combined with a dermatologically acceptable carrier to provide a topical skin care composition. 10. The method of claim 9, wherein the skin care composition is applied to the hyperpigmented portion of skin at least once a day for at least four weeks. 11. The method of claim 8, wherein the composition comprises from about 0.001 to about 10% of the N-acyl amino acid. 12. The method of claim 8, wherein the composition further comprises a dermatologically acceptable carrier. 13. The method of claim 8, wherein the vitamin B3 compound and N-acyl amino acid compound are combined with a dermatologically acceptable carrier to provide a first topical skin care composition, and the Laminaria Saccharina extract is combined with a dermatologically acceptable carrier to provide a second topical skin care composition. 14. The method of claim 13, wherein the second topical skin care composition is applied after the first topical skin care composition. 15. A method of making a skin care composition that has improved ability to inhibit PAR2 activation of keratinocytes, comprising: a. applying a vitamin B3 compound and an N-acyl amino acid compound to keratinocytes during a treatment period; b. applying a Laminaria Saccharina extract to the keratinocytes during the treatment period; c. determining the effective amount of Laminaria Saccharina extract needed to boost PAR2 inhibition of the vitamin B3 compound and N-acyl amino acid compound during the treatment period; and d. combining the effective amount of Laminaria Saccharina extract with a dermatologically acceptable carrier to provide the skin care composition. 16. The method of claim 15, further comprising mixing a skin tone agent selected from arbutin, deoxyarbutin, sucrose dilaurante, bakuchoil, pyrenoine, panicum miliaceum seed extract, arlatone dioic acid, cinnamic acid, ferulic acid, achromaxyl, methyl nicotinamide, oil soluble licorice extract, folic acid, undecylenic acid, zinc undecylenate, thiamine, thiamine hydrochloride, L-tryptophan, hexylrescorcinol, helianthus annuus and vitis vinifera leaf extract, carnosine methyl gentisate, 1,2-hexandiol and 1,2-octandiol, inositol, koijic acid, hexamidine compounds, salicylic acid, retinoids, and combinations thereof into the skin care composition. 17. The method of claim 16, further comprising mixing an anti-inflammatory active into the skin care composition. 18. The method of claim 17, wherein the anti-inflammatory active is selected from glycyrrhizic acid, glycyrrhizic acid salts, licorice extract, bisabolol, and combinations thereof. 19. The method of claim 15, wherein the effective amount of Laminaria Saccharina extract is sufficient to boost the PAR2 inhibition ability of the vitamin B3 compound and N-acyl amino acid compound by at least about 10%.
A method of inhibiting PAR2 activation of keratinocytes by boosting the PAR2 inhibition ability of a vitamin B3 compound and an N-acyl amino acid compound with a Laminaria Saccharina extract. The method may be used to improve the appearance of hyperpigmented spots on skin. By determining the amount of Laminaria Saccharina extract needed to boost PAR2 inhibition ability of a vitamin B3 compound and an N-acyl amino acid compound, improved skin care composition can be formulated for the treatment of hyperpigmented skin.1. A method of boosting the ability of a vitamin B3 compound and an N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes, comprising: a. identifying one or more keratinocytes in which inhibition of PAR2 activation is desired; b. applying an effective amount of a vitamin B3 compound and an N-acyl amino acid compound to the keratinocytes during a treatment period, wherein the amount of vitamin B3 compound and N-acyl amino acid compound is sufficient to inhibit PAR2 activation of the keratinocytes during the treatment period; and c. applying an effective amount of a Laminaria Saccharina extract to the keratinocytes, wherein the amount of Laminaria Saccharina extract is sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes relative to treatment without the Laminaria Saccharina extract. 2. The method of claim 1, wherein the vitamin B3 compound comprises niacinamide. 3. The method of claim 1, wherein the N-acyl amino acid compound comprises a N-acyl phenylalanine corresponding to the following formula: wherein R1 can be C1 to C30, saturated or unsaturated, straight or branched, substituted or unsubstituted alkyls; substituted or unsubstituted aromatic groups; or mixtures thereof. 4. The method of claim 3, wherein the N-acyl amino acid compound comprises N-undecylenoyl-L-phenylalanine. 5. The method of claim 1, wherein the N-acyl amino acid compound comprises an N-acyl tyrosine corresponds to the following formula: wherein R1 can be C1 to C30, saturated or unsaturated, straight or branched, substituted or unsubstituted alkyls; substituted or unsubstituted aromatic groups; or mixtures thereof. 6. The method of claim 1, wherein the effective amount of Laminaria Saccharina is sufficient to sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes by at least about 10%. 7. The method of claim 6, wherein the effective amount of Laminaria Saccharina is sufficient to sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to inhibit PAR2 activation of keratinocytes by at least about 50%. 8. A method of boosting the ability of a vitamin B3 compound and an N-acyl amino acid compound to improve the appearance of hyperpigmented skin, comprising: a. identifying a hperpigmented portion of skin; b. applying an effective amount of a vitamin B3 compound and an N-acyl amino acid compound to the hyperpigmented portion of skin during a treatment period, wherein the treatment period is sufficient for the vitamin B3 compound and N-acyl amino acid compound to improve the appearance of the hyperpigmentation; and c. applying an effective amount of a Laminaria Saccharina extract to the target portion of skin, wherein the amount of Laminaria Saccharina extract is sufficient to boost the ability of the vitamin B3 compound and N-acyl amino acid compound to improve the appearance of the hyperpigmented portion of skin relative to treatment without the Laminaria Saccharina extract. 9. The method of claim 8, wherein the vitamin B3 compound, N-acyl amino acid compound, and Laminaria Saccharina extract are combined with a dermatologically acceptable carrier to provide a topical skin care composition. 10. The method of claim 9, wherein the skin care composition is applied to the hyperpigmented portion of skin at least once a day for at least four weeks. 11. The method of claim 8, wherein the composition comprises from about 0.001 to about 10% of the N-acyl amino acid. 12. The method of claim 8, wherein the composition further comprises a dermatologically acceptable carrier. 13. The method of claim 8, wherein the vitamin B3 compound and N-acyl amino acid compound are combined with a dermatologically acceptable carrier to provide a first topical skin care composition, and the Laminaria Saccharina extract is combined with a dermatologically acceptable carrier to provide a second topical skin care composition. 14. The method of claim 13, wherein the second topical skin care composition is applied after the first topical skin care composition. 15. A method of making a skin care composition that has improved ability to inhibit PAR2 activation of keratinocytes, comprising: a. applying a vitamin B3 compound and an N-acyl amino acid compound to keratinocytes during a treatment period; b. applying a Laminaria Saccharina extract to the keratinocytes during the treatment period; c. determining the effective amount of Laminaria Saccharina extract needed to boost PAR2 inhibition of the vitamin B3 compound and N-acyl amino acid compound during the treatment period; and d. combining the effective amount of Laminaria Saccharina extract with a dermatologically acceptable carrier to provide the skin care composition. 16. The method of claim 15, further comprising mixing a skin tone agent selected from arbutin, deoxyarbutin, sucrose dilaurante, bakuchoil, pyrenoine, panicum miliaceum seed extract, arlatone dioic acid, cinnamic acid, ferulic acid, achromaxyl, methyl nicotinamide, oil soluble licorice extract, folic acid, undecylenic acid, zinc undecylenate, thiamine, thiamine hydrochloride, L-tryptophan, hexylrescorcinol, helianthus annuus and vitis vinifera leaf extract, carnosine methyl gentisate, 1,2-hexandiol and 1,2-octandiol, inositol, koijic acid, hexamidine compounds, salicylic acid, retinoids, and combinations thereof into the skin care composition. 17. The method of claim 16, further comprising mixing an anti-inflammatory active into the skin care composition. 18. The method of claim 17, wherein the anti-inflammatory active is selected from glycyrrhizic acid, glycyrrhizic acid salts, licorice extract, bisabolol, and combinations thereof. 19. The method of claim 15, wherein the effective amount of Laminaria Saccharina extract is sufficient to boost the PAR2 inhibition ability of the vitamin B3 compound and N-acyl amino acid compound by at least about 10%.
1,600
351
15,100,780
1,663
The present invention relates to a method of producing sesquiterpene lactones of the thapsigargin family, the method comprising the steps of: (a) culturing plant cells of the genus Thapsia in a nutrient medium in a suspension cell culture, wherein the cells produce one or more sesquiterpene lactones of the thapsigargin family; and (b) recovering one or more sesquiterpene lactones of the thapsigargin family produced in (a). The present invention further relates to a suspension cell culture comprising plant cells of the genus Thapsia, wherein the plant cells are capable of producing one or more sesquiterpene lactones of the thapsigargin family and to a plant cell biomass comprising plant cells of the genus Thapsia obtained from said suspension cell culture.
1. A method of producing sesquiterpene lactones of the thapsigargin family, the method comprising the steps of: (a) culturing plant cells of the genus Thapsia in a nutrient medium in a suspension cell culture, wherein the cells produce one or more sesquiterpene lactones of the thapsigargin family; and (b) recovering one or more sesquiterpene lactone(s) of the thapsigargin family produced in (a). 2. The method of claim 1, wherein the one or more of the sesquiterpene lactones of the thapsigargin family recovered in step (b) comprises a 2β,3α,7β,8α,10β,11α-hexaoxygenated-6β,12-guaianolide nucleus. 3. The method of claim 2, wherein the sesquiterpene lactone of the thapsigargin family is thapsigargin. 4. The method of claim 1, wherein the one or more of the sesquiterpene lactone of the thapsigargin family recovered in step (b) comprises a 3α,7β,8α,10β,11α-pentaoxygenated-6β,12-guaianolide nucleus. 5. The method of claim 1, wherein the plant cells are selected from the group consisting of Thapsia garganica cells, Thapsia gymnesica cells and Thapsia villosa cells. 6. The method of claim 1, wherein the plant cells are non-embryogenic cells. 7. The method of claim 1, further comprising prior to step (a) an additional step of: (a-0) culturing Thapsia plant explants on medium, thereby obtaining friable callus material. 8. The method of claim 1, wherein the nutrient medium in step (a) is a growth medium capable of inducing a growth increase of at least 50% in one week. 9. The method of claim 1, wherein the nutrient medium is free of 2,4-Dichlorophenoxyacetic acid. 10. A suspension cell culture comprising plant cells of the genus Thapsia, wherein the plant cells are capable of producing one or more sesquiterpene lactones of the thapsigargin family. 11. A plant cell biomass comprising plant cells of the genus Thapsia obtained from the suspension cell culture of claim 10.
The present invention relates to a method of producing sesquiterpene lactones of the thapsigargin family, the method comprising the steps of: (a) culturing plant cells of the genus Thapsia in a nutrient medium in a suspension cell culture, wherein the cells produce one or more sesquiterpene lactones of the thapsigargin family; and (b) recovering one or more sesquiterpene lactones of the thapsigargin family produced in (a). The present invention further relates to a suspension cell culture comprising plant cells of the genus Thapsia, wherein the plant cells are capable of producing one or more sesquiterpene lactones of the thapsigargin family and to a plant cell biomass comprising plant cells of the genus Thapsia obtained from said suspension cell culture.1. A method of producing sesquiterpene lactones of the thapsigargin family, the method comprising the steps of: (a) culturing plant cells of the genus Thapsia in a nutrient medium in a suspension cell culture, wherein the cells produce one or more sesquiterpene lactones of the thapsigargin family; and (b) recovering one or more sesquiterpene lactone(s) of the thapsigargin family produced in (a). 2. The method of claim 1, wherein the one or more of the sesquiterpene lactones of the thapsigargin family recovered in step (b) comprises a 2β,3α,7β,8α,10β,11α-hexaoxygenated-6β,12-guaianolide nucleus. 3. The method of claim 2, wherein the sesquiterpene lactone of the thapsigargin family is thapsigargin. 4. The method of claim 1, wherein the one or more of the sesquiterpene lactone of the thapsigargin family recovered in step (b) comprises a 3α,7β,8α,10β,11α-pentaoxygenated-6β,12-guaianolide nucleus. 5. The method of claim 1, wherein the plant cells are selected from the group consisting of Thapsia garganica cells, Thapsia gymnesica cells and Thapsia villosa cells. 6. The method of claim 1, wherein the plant cells are non-embryogenic cells. 7. The method of claim 1, further comprising prior to step (a) an additional step of: (a-0) culturing Thapsia plant explants on medium, thereby obtaining friable callus material. 8. The method of claim 1, wherein the nutrient medium in step (a) is a growth medium capable of inducing a growth increase of at least 50% in one week. 9. The method of claim 1, wherein the nutrient medium is free of 2,4-Dichlorophenoxyacetic acid. 10. A suspension cell culture comprising plant cells of the genus Thapsia, wherein the plant cells are capable of producing one or more sesquiterpene lactones of the thapsigargin family. 11. A plant cell biomass comprising plant cells of the genus Thapsia obtained from the suspension cell culture of claim 10.
1,600
352
14,899,394
1,651
A cell culture article, including: a substrate comprising a polygalacturonic acid compound selected from at least one of: pectic acid; partially esterified pectic acid having a degree of esterification from 1 to 40 mol %, or salts thereof; and an adhesion polymer on the surface of the polygalacturonic acid compound. A method of making and using the article are also disclosed.
1. A cell culture article, comprising: a substrate comprising a polygalacturonic acid compound selected from at least one of: pectic acid; partially esterified pectic acid having a degree of esterification from 1 to 40 mol %, or salts thereof; and an adhesion polymer on the surface of the polygalacturonic acid compound. 2. The article of claim 1 wherein the polygalacturonic acid compound is covalently cross linked, ionically cross linked, or mixtures thereof. 3. The article of claim 1, wherein the partially esterified pectic acid comprises an alkyl carboxy ester having an alkyl group having from 1 to 10 carbon atoms. 4. The article of claim 1, wherein the adhesion polymer on the surface of the polygalacturonic acid compound comprises a polypeptide. 5. The article of claim 1, wherein the adhesion polymer on the surface of the polygalacturonic acid compound comprises a polymer having a conjugated polypeptide. 6. The article of claim 1, wherein the adhesion polymer on the surface of the polygalacturonic acid compound is a polymer having a conjugated polypeptide selected from at least one of: poly(MAA-PEG4-VN) homopolymer; poly(HEMA-co-MAA-PEG4-VN) copolymer; or mixtures thereof, where: MAA is methacrylic acid; HEMA is hydroxyethylmethacrylate; PEG4 is a polyethylene glycol tetra oligomer; and VN is a conjugated vitronectin polypeptide. 7. The article of claim 1, wherein the adhesion polymer is present in an amount of from 0.1 to 30 weight % based on the total weight of the article, or based on the total weight of the polygalacturonic acid compound or compounds selected. 8. The article of claim 1, wherein the adhesion polymer promotes the attachment of anchorage dependent live cells to the substrate. 9. The article of claim 1, wherein the substrate comprises a microcarrier particle. 10. A method for harvesting cultured cells, comprising: culturing cells on the surface of the article of claim 1; and contacting the cultured cells with a mixture of pectinase and a chelator to separate the cells from the article. 11. The method of claim 10, further comprising isolating the separated cells from the composition. 12. The method of claim 10, wherein the chelator is EDTA. 13. The method of claim 10, wherein the contacting is accomplished free of a protease. 14. A method of making the cell culture composition of claim 1 comprising: coating the surface of the substrate comprised of polygalacturonic acid compound with an adhesion polymer. 15. The method of claim 14 further comprising crosslinking the polygalacturonic acid compound. 16. The method of claim 15 wherein crosslinking the polygalacturonic acid compound is accomplished ionically, by internal gelation, or a combination thereof. 17. The method of claim 14 wherein the substrate is a microcarrier.
A cell culture article, including: a substrate comprising a polygalacturonic acid compound selected from at least one of: pectic acid; partially esterified pectic acid having a degree of esterification from 1 to 40 mol %, or salts thereof; and an adhesion polymer on the surface of the polygalacturonic acid compound. A method of making and using the article are also disclosed.1. A cell culture article, comprising: a substrate comprising a polygalacturonic acid compound selected from at least one of: pectic acid; partially esterified pectic acid having a degree of esterification from 1 to 40 mol %, or salts thereof; and an adhesion polymer on the surface of the polygalacturonic acid compound. 2. The article of claim 1 wherein the polygalacturonic acid compound is covalently cross linked, ionically cross linked, or mixtures thereof. 3. The article of claim 1, wherein the partially esterified pectic acid comprises an alkyl carboxy ester having an alkyl group having from 1 to 10 carbon atoms. 4. The article of claim 1, wherein the adhesion polymer on the surface of the polygalacturonic acid compound comprises a polypeptide. 5. The article of claim 1, wherein the adhesion polymer on the surface of the polygalacturonic acid compound comprises a polymer having a conjugated polypeptide. 6. The article of claim 1, wherein the adhesion polymer on the surface of the polygalacturonic acid compound is a polymer having a conjugated polypeptide selected from at least one of: poly(MAA-PEG4-VN) homopolymer; poly(HEMA-co-MAA-PEG4-VN) copolymer; or mixtures thereof, where: MAA is methacrylic acid; HEMA is hydroxyethylmethacrylate; PEG4 is a polyethylene glycol tetra oligomer; and VN is a conjugated vitronectin polypeptide. 7. The article of claim 1, wherein the adhesion polymer is present in an amount of from 0.1 to 30 weight % based on the total weight of the article, or based on the total weight of the polygalacturonic acid compound or compounds selected. 8. The article of claim 1, wherein the adhesion polymer promotes the attachment of anchorage dependent live cells to the substrate. 9. The article of claim 1, wherein the substrate comprises a microcarrier particle. 10. A method for harvesting cultured cells, comprising: culturing cells on the surface of the article of claim 1; and contacting the cultured cells with a mixture of pectinase and a chelator to separate the cells from the article. 11. The method of claim 10, further comprising isolating the separated cells from the composition. 12. The method of claim 10, wherein the chelator is EDTA. 13. The method of claim 10, wherein the contacting is accomplished free of a protease. 14. A method of making the cell culture composition of claim 1 comprising: coating the surface of the substrate comprised of polygalacturonic acid compound with an adhesion polymer. 15. The method of claim 14 further comprising crosslinking the polygalacturonic acid compound. 16. The method of claim 15 wherein crosslinking the polygalacturonic acid compound is accomplished ionically, by internal gelation, or a combination thereof. 17. The method of claim 14 wherein the substrate is a microcarrier.
1,600
353
14,834,914
1,663
The present invention relates to methods of producing lipids. In particular, the present invention relates to methods of increasing the level of one or more non-polar lipids and/or the total non-polar lipid content in a transgenic organism or part thereof. In one particular embodiment, the present invention relates to the use of an acyltransferase, for example, a monoacylglycerol acyltransferase (MGAT) to increase the level of one or more non-polar lipids and/or the total non-polar lipid content in plants, plant seed and/or leaves, algae and fungi.
1. A method of producing extracted lipid, the method comprising the steps of: i) obtaining a transgenic non-human organism or part thereof comprising one or more exogenous polynucleotides, wherein the transgenic non-human organism or part thereof has an increased level of one or more non-polar lipids when compared to a corresponding organism or part thereof lacking the one or more exogenous polynucleotides, and ii) extracting the lipid from the transgenic non-human organism or part thereof, thereby producing the extracted lipid. 2. The method of claim 1, wherein the level of the one or more non-polar lipids of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis than the corresponding non-human organism or part thereof. 3. The method of claim 1 or claim 2, wherein the level of the one or more non-polar lipids of the transgenic non-human organism or part thereof is at least 1% (w/w) greater on a relative basis than the corresponding non-human organism or part thereof. 4. The method of any one of claims 1 to 3, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is increased when compared to the corresponding organism or part thereof. 5. The method of claim 4, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis than the corresponding non-human organism or part thereof. 6. The method of claim 4 or claim 5, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is at least 1% (w/w) greater on a relative basis than the corresponding non-human organism or part thereof. 7. The method of any one of claims 1 to 6, wherein the transgenic non-human organism is a plant, alga, or an organism suitable for fermentation such as a yeast or fungus. 8. The method of claim 7, wherein the plant is Brassica sp., Gossypium hirsutum, Linum usitatissimum, Helianthus sp., Carthamus tinctorius, Glycine max, Zea mays, Arabidopsis thaliana, Sorghum bicolor, Sorghum vulgare, Avena sativa, Trifolium sp., Camelina sativa, Miscanthus x giganteus, or Miscanthus sinensis. 9. The method of any one of claims 1 to 8, wherein the part is a seed, fruit, or a vegetative part of a plant. 10. The method of claim 9, wherein the part is a plant seed and the extracted lipid is seedoil. 11. The method of claim 10, wherein the total oil content, or the total fatty acid content, of the seed is at least 0.5% (w/w) to 25% (w/w) greater on a weight basis than a corresponding seed lacking the one or more exogenous polynucleotides. 12. The method of claim 10 or claim 11, wherein the seed is from: i) a canola plant, ii) a corn plant, iii) a soybean plant, iv) a lupin plant, v) a peanut plant, vi) a sunflower plant, vii) a cotton plant, viii) a safflower plant, or ix) a flax plant. 13. The method of claim 12, wherein the canola seed comprises one or more of the following: i) at least 45% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 14. The method of claim 12, wherein the corn seed comprises one or more of the following: i) at least 5% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 15. The method of claim 12, wherein the soybean seed comprises one or more of the following: i) at least 20% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 16. The method of claim 12, wherein the lupin seed comprises one or more of the following: i) at least 10% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 17. The method of claim 12, wherein the peanut seed comprises one or more of the following: i) at least 50% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 18. The method of claim 9, wherein the vegetative part of the plant is an aerial plant part or a green part such as a leaf or stem. 19. The method of claim 18, wherein the TAG, DAG, TAG and DAG, or MAG content of the vegetative plant part is at least 10% (w/w) greater on a relative basis than the TAG, DAG, TAG and DAG, or MAG content of a corresponding vegetative plant part lacking the one or more exogenous polynucleotides. 20. The method of any one of claims 1 to 19, wherein at least 60% (mol %) of the fatty acid content of the extracted lipid is oleic acid. 21. The method of any one of claims 1 to 20, wherein the polyunsaturated fatty acid content of the extracted lipid is increased when compared to the lipid extracted from the corresponding organism or part thereof. 22. The method of any one of claims 1 to 21, wherein the level of a polyunsaturated fatty acid in the extracted lipid is increased when compared to the lipid extracted from the corresponding organism or part thereof, wherein the polyunsaturated fatty acid is eicosadienoic acid, arachidonic acid (ARA), alpha linolenic acid (ALA), stearidonic acid (SDA), eicosatrienoic acid (ETE), eicosatetraenoic acid (ETA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), or a combination of two of more thereof. 23. The method of any one of claims 1 to 22, wherein the level of the one or more lipids and/or the total non-polar lipid content is determinable by analysis by using gas chromatography of fatty acid methyl esters obtained from the extracted lipid. 24. The method of any one of claims 10 to 17, wherein the method further comprises harvesting the seed from the transgenic plant, pressing the seedoil from the seed, and/or purifying the seedoil. 25. The method of any one of claims 1 to 24, wherein the one or more exogenous polynucleotides encode: i) a monoacylglycerol acyltransferase (MGAT), ii) a diacylglycerol acyltransferase 2 (DGAT2), iii) a MGAT and a glycerol-3-phosphate acyltransferase (GPAT), iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT. 26. The method of claim 25, wherein the GPAT also has phosphatase activity to produce MAG, such as an Arabidopsis GPAT4 or GPAT6. 27. The method of claim 25 or claim 26, wherein the DGAT is a DGAT2. 28. The method of any one of claims 25 to 27, wherein the exogenous polynucleotide encoding MGAT comprises one or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:1 to 44, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:45 to 82, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 29. The method of any one of claims 25 to 28, wherein the exogenous polynucleotide encoding GPAT comprises one or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:84 to 141, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:144 to 201, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 30. The method of any one of claims 25 to 29, wherein the exogenous polynucleotide encoding DGAT2 comprises one or more of the following: i) a sequence of nucleotides of SEQ ID NO:204, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in SEQ ID NO:212, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 31. The method of any one of claims 25 to 30, wherein the level of the one or more non-polar lipids and/or the total non-polar lipid content of the transgenic organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% (w/w) greater on a relative basis than a corresponding organism or part thereof lacking the one or more exogenous polynucleotides but comprising an exogenous polynucleotide encoding an Arabidopsis thaliana DGAT1. 32. The method of any one of claims 1 to 31, wherein the transgenic non-human organism or part thereof further comprises one or more introduced mutations, and/or an exogenous polynucleotide which down-regulates the production and/or activity of an endogenous enzyme of the non-human transgenic organism or part thereof selected from DGAT, sn-1 glycerol-3-phosphate acyltransferase (sn-1 GPAT), 1-acyl-glycerol-3-phosphate acyltransferase (LPAAT), acyl-CoA:lysophosphatidylcholine acyltransferase (LPCAT), phosphatidic acid phosphatase (PAP), or a combination of two or more thereof. 33. The method of claim 32, wherein the exogenous polynucleotide is selected from: an antisense polynucleotide, a sense polynucleotide, a catalytic polynucleotide, a microRNA, a polynucleotide which encodes a polypeptide which binds the endogenous enzyme, and a double stranded RNA. 34. The method of any one of claims 1 to 33, wherein the non-polar lipid is TAG, DAG, TAG and DAG, MAG, total PUFA, or a specific PUFA which is eicosadienoic acid (EDA), arachidonic acid (ARA), alpha linolenic acid (ALA), stearidonic acid (SDA), eicosatrienoic acid (EYE), eicosatetraenoic acid (ETA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), or a combination of two of more thereof. 35. A transgenic non-human organism or part thereof comprising one or more exogenous polynucleotides, wherein the transgenic non-human organism or part thereof has an increased level of one or more non-polar lipids when compared to a corresponding organism or part thereof lacking the one or more exogenous polynucleotides. 36. The transgenic non-human organism or part thereof of claim 35, wherein the level of the one or more non-polar lipids of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% (w/w) greater on a relative basis than the corresponding organism or part thereof. 37. The transgenic non-human organism or part thereof of claim 35 or claim 36, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is increased compared to the corresponding organism or part thereof. 38. The transgenic non-human organism or part thereof of claim 37, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% greater on a relative basis than the corresponding organism or part thereof. 39. The transgenic non-human organism or part thereof of any one of claims 35 to 38, wherein the polyunsaturated fatty acid content in the extracted lipid of the transgenic non-human organism or part thereof is increased when compared to the extracted lipid of the corresponding organism or part thereof. 40. The transgenic non-human organism or part thereof of any one of claims 35 to 39, wherein the content of a polyunsaturated fatty acid in the extracted lipid is increased when compared to the lipid extracted from the corresponding organism or part thereof, wherein the polyunsaturated fatty acid comprises eicosadienoic acid (EDA), arachidonic acid (ARA), alpha linolenic acid (ALA), stearidonic acid (SDA), eicosatrienoic acid (ETE), eicosatetraenoic acid (ETA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), or a combination of two of more thereof. 41. The transgenic non-human organism or part thereof of any one of claims 35 to 40, wherein the one or more exogenous polynucleotides encode: i) a MGAT, ii) a DGAT2, iii) a MGAT and a GPAT, iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT. 42. The transgenic non-human organism of claim 41, wherein the GPAT also has phosphatase activity to produce MAG, such as an Arabidopsis GPAT4 or GPAT6. 43. The transgenic non-human organism of claim 41 or claim 42, wherein the DGAT is a DGAT2. 44. The transgenic non-human organism or part thereof of any one of claims 41 to 43, wherein the exogenous polynucleotide encoding MGAT comprises one or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:1 to 44, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:45 to 82, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 45. The transgenic non-human organism or part thereof of any one of claims 41 to 44, wherein the exogenous polynucleotide encoding GPAT comprises oen or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:84 to 141, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:144 to 201, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 46. The transgenic non-human organism or part thereof of any one of claims 41 to 45, wherein the exogenous polynucleotide encoding DGAT2 comprises one or more of the following: i) a sequence of nucleotides of SEQ ID NO:204, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in SEQ ID NO:212, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 47. The transgenic non-human organism or part thereof of any one of claims 44 to 46, wherein the level of the one or more non-polar lipids and/or the total lipid content of the transgenic organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% (w/w) greater on a relative basis than a corresponding organism or part thereof lacking the one or more exogenous polynucleotides but comprising an exogenous polynucleotide encoding an Arabidopsis thaliana DGAT1. 48. The transgenic non-human organism of any one of claims 35 to 47 which is a plant, alga, or an organism suitable for fermentation such as a yeast or fungus. 49. A transgenic non-human organism comprising one or more exogenous polynucleotides encoding: i) a MGAT, ii) a DGAT2, iii) a MGAT and a GPAT, iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT. 50. The transgenic non-human organism or part thereof of claim 49, further characterised by one or more features of any one of claims 35 to 48. 51. A method of obtaining a cell with enhanced ability to produce one or more non-polar lipids, the method comprising: i) introducing into a cell one or more exogenous polynucleotides encoding a) a MGAT, b) a DGAT2 c) a MGAT and a GPAT d) a MGAT and a DGAT, or e) a MGAT, a GPAT and a DGAT. wherein the one or more exogenous polynucleotides are operably linked to one or more promoters that are capable of directing expression of the one or more exogenous polynucleotides in the cell, ii) expressing the one or more exogenous polynucleotides in the cell, iii) analysing the lipid content of the cell, and iv) selecting a cell having an increased level of one or more non-polar lipids when compared to a corresponding cell lacking the exogenous polynucleotides. 52. The method of claim 51, wherein the selected cell has one or more of the features of the organism or part thereof defined in any one of claims 36 to 48. 53. The method of claim 51 or claim 52, wherein the first and second exogenous polynucleotides are stably integrated into the genome of the cell. 54. The method of claim 53, further comprising the step of regenerating a transgenic plant from the cell of step i). 55. A transgenic cell or plant obtained using the method of any one of claims 51 to 54, or a transgenic plant part obtained therefrom. 56. Use of one or more exogenous polynucleotides encoding: i) a MGAT, ii) a DGAT2, iii) a MGAT and a GPAT iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT, for producing a transgenic non-human organism or part thereof with enhanced ability to produce one or more non-polar lipids when compared to a corresponding organism or part thereof lacking the one or more exogenous polynucleotides. 57. A trans genic seed of a plant, the seed comprising one or more exogenous polynucleotides and having an increased level of one or more non-polar lipids when compared to a corresponding seed lacking the exogenous polynucleotides. 58. The transgenic seed of claim 57, which comprises one or more of the features defined in claims 36 to 48. 59. A transgenic plant which produces the seed of claim 57 or claim 58. 60. The transgenic plant of claim 59 which is Brassica sp., Gossypium hirsutum, Linum usitatissimum, Helianthus sp., Carthamus tinctorius, Glycine max, Zea mays, Arabidopsis thaliana, Sorghum bicolor, Sorghum vulgare, Avena sativa, Trifolium sp., Camelina sativa, Miscanthus x giganteus, or Miscanthus sinensis. 61. A method of producing seed, the method comprising: i) growing the transgenic plant of claim 59 or claim 60, and ii) harvesting the seed. 62. A fermentation process comprising the steps of: i) providing a vessel containing a liquid composition comprising the transgenic non-human organism of claim 48 which is suitable for fermentation, and constituents required for fermentation and fatty acid biosynthesis, and ii) providing conditions conducive to the fermentation of the liquid composition contained in said vessel. 63. Extracted lipid obtainable by the method of any one of claims 1 to 34, or obtainable from the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, or the transgenic plant of claim 59 or claim 60. 64. An extracted lipid obtainable from a transgenic non-human organism or a part thereof comprising a DAG content that is at least 1% (w/w) on a weight basis of the extracted lipid. 65. Use of the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, the transgenic plant of claim 59 or claim 60, or the extracted lipid of claim 63 or claim 64 for the manufacture of an industrial product. 66. The use of claim 65, wherein the industrial product is fuel. 67. A method of producing fuel, the method comprising: i) reacting the lipid of claim 63 or claim 64 with an alcohol, optionally, in the presence of a catalyst, to produce alkyl esters, and ii) optionally, blending the alkyl esters with petroleum based fuel. 68. The method of claim 67, wherein the alkyl esters are methyl esters. 69. A method of producing a feedstuff, the method comprising admixing the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, the transgenic plant of claim 59 or claim 60, or the extracted lipid of claim 63 or claim 64, or an extract or portion thereof, with at least one other food ingredient. 70. Feedstuffs, cosmetics or chemicals comprising the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, the transgenic plant of claim 59 or claim 60, or the extracted lipid of claim 63 or claim 64, or an extract or portion thereof. 71. A method for identifying a nucleic acid molecule encoding an acyltransferase having an increased ability to produce MAG, DAG and/or TAG in a cell, the method comprising: i) obtaining a cell comprising a nucleic acid molecule operably linked to a promoter which is active in the cell, wherein the nucleic acid molecule comprises a sequence of nucleotides as defined in any one or more of claim 27, 28, or 29 and/or a sequence of nucleotides encoding a polypeptide having one or more amino acid sequences as provided in SEQ ID NOs:220, 221, 222, 223, 224, 225, 226 and 227, or a sequence of amino acids which is at least 50%, preferably at least 60%, more preferably at least 65% identical thereto ii) determining if the level of MAG, DAG and/or TAG produced in the cell is increased when compared to a corresponding cell lacking the nucleic acid, and iii) identifying a nucleic acid molecule encoding a acyltransferase having an increased ability to produce MAG, DAG and/or TAG in the cell. 72. The method of claim 71, wherein the acyltransferase catalyzes an enzyme reaction in the MGAT pathway. 73. The method of claim 71 or claim 72, wherein the acyltransferase is a MGAT, GPAT and/or DGAT. 74. The method of claim 73, wherein the GPAT also has phosphatase activity to produce MAG, such as an Arabidopsis GPAT4 or GPAT6. 75. The method of claim 73 or claim 74, wherein the DGAT is DGAT2. 76. The method of any one of claims 71 to 75, further comprising introducing the nucleic acid molecule into a cell prior to step i). 77. The method of any one of claims 71 to 76, wherein the cell is a plant cell. 78. The method of any one of claims 71 to 77, wherein the acyltransferase increases TAG production in the cell by a greater amount than Arabidopsis thaliana DGAT1. 79. An isolated and/or recombinant polynucleotide comprising: i) a sequence of nucleotides selected from any one of SEQ ID NOs:1 to 6, or ii) a sequence of nucleotides which is at least 80% identical to i). 80. A vector comprising the polynucleotide of claim 79. 81. A transgenic cell comprising the polynucleotide of claim 79 or the vector of claim 80. 82. A transgenic non-human organism or part thereof comprising the polynucleotide of claim 79, the vector of claim 80, or the transgenic cell of claim 81.
The present invention relates to methods of producing lipids. In particular, the present invention relates to methods of increasing the level of one or more non-polar lipids and/or the total non-polar lipid content in a transgenic organism or part thereof. In one particular embodiment, the present invention relates to the use of an acyltransferase, for example, a monoacylglycerol acyltransferase (MGAT) to increase the level of one or more non-polar lipids and/or the total non-polar lipid content in plants, plant seed and/or leaves, algae and fungi.1. A method of producing extracted lipid, the method comprising the steps of: i) obtaining a transgenic non-human organism or part thereof comprising one or more exogenous polynucleotides, wherein the transgenic non-human organism or part thereof has an increased level of one or more non-polar lipids when compared to a corresponding organism or part thereof lacking the one or more exogenous polynucleotides, and ii) extracting the lipid from the transgenic non-human organism or part thereof, thereby producing the extracted lipid. 2. The method of claim 1, wherein the level of the one or more non-polar lipids of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis than the corresponding non-human organism or part thereof. 3. The method of claim 1 or claim 2, wherein the level of the one or more non-polar lipids of the transgenic non-human organism or part thereof is at least 1% (w/w) greater on a relative basis than the corresponding non-human organism or part thereof. 4. The method of any one of claims 1 to 3, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is increased when compared to the corresponding organism or part thereof. 5. The method of claim 4, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis than the corresponding non-human organism or part thereof. 6. The method of claim 4 or claim 5, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is at least 1% (w/w) greater on a relative basis than the corresponding non-human organism or part thereof. 7. The method of any one of claims 1 to 6, wherein the transgenic non-human organism is a plant, alga, or an organism suitable for fermentation such as a yeast or fungus. 8. The method of claim 7, wherein the plant is Brassica sp., Gossypium hirsutum, Linum usitatissimum, Helianthus sp., Carthamus tinctorius, Glycine max, Zea mays, Arabidopsis thaliana, Sorghum bicolor, Sorghum vulgare, Avena sativa, Trifolium sp., Camelina sativa, Miscanthus x giganteus, or Miscanthus sinensis. 9. The method of any one of claims 1 to 8, wherein the part is a seed, fruit, or a vegetative part of a plant. 10. The method of claim 9, wherein the part is a plant seed and the extracted lipid is seedoil. 11. The method of claim 10, wherein the total oil content, or the total fatty acid content, of the seed is at least 0.5% (w/w) to 25% (w/w) greater on a weight basis than a corresponding seed lacking the one or more exogenous polynucleotides. 12. The method of claim 10 or claim 11, wherein the seed is from: i) a canola plant, ii) a corn plant, iii) a soybean plant, iv) a lupin plant, v) a peanut plant, vi) a sunflower plant, vii) a cotton plant, viii) a safflower plant, or ix) a flax plant. 13. The method of claim 12, wherein the canola seed comprises one or more of the following: i) at least 45% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 14. The method of claim 12, wherein the corn seed comprises one or more of the following: i) at least 5% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 15. The method of claim 12, wherein the soybean seed comprises one or more of the following: i) at least 20% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 16. The method of claim 12, wherein the lupin seed comprises one or more of the following: i) at least 10% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 17. The method of claim 12, wherein the peanut seed comprises one or more of the following: i) at least 50% seedoil on a weight basis, ii) a relative seedoil content at least 5% (w/w) greater than a corresponding seed, iii) a relative DAG content at least 10% (w/w) greater than a corresponding seed, and iv) a relative TAG content at least 5% (w/w) greater than a corresponding seed. 18. The method of claim 9, wherein the vegetative part of the plant is an aerial plant part or a green part such as a leaf or stem. 19. The method of claim 18, wherein the TAG, DAG, TAG and DAG, or MAG content of the vegetative plant part is at least 10% (w/w) greater on a relative basis than the TAG, DAG, TAG and DAG, or MAG content of a corresponding vegetative plant part lacking the one or more exogenous polynucleotides. 20. The method of any one of claims 1 to 19, wherein at least 60% (mol %) of the fatty acid content of the extracted lipid is oleic acid. 21. The method of any one of claims 1 to 20, wherein the polyunsaturated fatty acid content of the extracted lipid is increased when compared to the lipid extracted from the corresponding organism or part thereof. 22. The method of any one of claims 1 to 21, wherein the level of a polyunsaturated fatty acid in the extracted lipid is increased when compared to the lipid extracted from the corresponding organism or part thereof, wherein the polyunsaturated fatty acid is eicosadienoic acid, arachidonic acid (ARA), alpha linolenic acid (ALA), stearidonic acid (SDA), eicosatrienoic acid (ETE), eicosatetraenoic acid (ETA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), or a combination of two of more thereof. 23. The method of any one of claims 1 to 22, wherein the level of the one or more lipids and/or the total non-polar lipid content is determinable by analysis by using gas chromatography of fatty acid methyl esters obtained from the extracted lipid. 24. The method of any one of claims 10 to 17, wherein the method further comprises harvesting the seed from the transgenic plant, pressing the seedoil from the seed, and/or purifying the seedoil. 25. The method of any one of claims 1 to 24, wherein the one or more exogenous polynucleotides encode: i) a monoacylglycerol acyltransferase (MGAT), ii) a diacylglycerol acyltransferase 2 (DGAT2), iii) a MGAT and a glycerol-3-phosphate acyltransferase (GPAT), iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT. 26. The method of claim 25, wherein the GPAT also has phosphatase activity to produce MAG, such as an Arabidopsis GPAT4 or GPAT6. 27. The method of claim 25 or claim 26, wherein the DGAT is a DGAT2. 28. The method of any one of claims 25 to 27, wherein the exogenous polynucleotide encoding MGAT comprises one or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:1 to 44, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:45 to 82, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 29. The method of any one of claims 25 to 28, wherein the exogenous polynucleotide encoding GPAT comprises one or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:84 to 141, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:144 to 201, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 30. The method of any one of claims 25 to 29, wherein the exogenous polynucleotide encoding DGAT2 comprises one or more of the following: i) a sequence of nucleotides of SEQ ID NO:204, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in SEQ ID NO:212, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 31. The method of any one of claims 25 to 30, wherein the level of the one or more non-polar lipids and/or the total non-polar lipid content of the transgenic organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% (w/w) greater on a relative basis than a corresponding organism or part thereof lacking the one or more exogenous polynucleotides but comprising an exogenous polynucleotide encoding an Arabidopsis thaliana DGAT1. 32. The method of any one of claims 1 to 31, wherein the transgenic non-human organism or part thereof further comprises one or more introduced mutations, and/or an exogenous polynucleotide which down-regulates the production and/or activity of an endogenous enzyme of the non-human transgenic organism or part thereof selected from DGAT, sn-1 glycerol-3-phosphate acyltransferase (sn-1 GPAT), 1-acyl-glycerol-3-phosphate acyltransferase (LPAAT), acyl-CoA:lysophosphatidylcholine acyltransferase (LPCAT), phosphatidic acid phosphatase (PAP), or a combination of two or more thereof. 33. The method of claim 32, wherein the exogenous polynucleotide is selected from: an antisense polynucleotide, a sense polynucleotide, a catalytic polynucleotide, a microRNA, a polynucleotide which encodes a polypeptide which binds the endogenous enzyme, and a double stranded RNA. 34. The method of any one of claims 1 to 33, wherein the non-polar lipid is TAG, DAG, TAG and DAG, MAG, total PUFA, or a specific PUFA which is eicosadienoic acid (EDA), arachidonic acid (ARA), alpha linolenic acid (ALA), stearidonic acid (SDA), eicosatrienoic acid (EYE), eicosatetraenoic acid (ETA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), or a combination of two of more thereof. 35. A transgenic non-human organism or part thereof comprising one or more exogenous polynucleotides, wherein the transgenic non-human organism or part thereof has an increased level of one or more non-polar lipids when compared to a corresponding organism or part thereof lacking the one or more exogenous polynucleotides. 36. The transgenic non-human organism or part thereof of claim 35, wherein the level of the one or more non-polar lipids of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% (w/w) greater on a relative basis than the corresponding organism or part thereof. 37. The transgenic non-human organism or part thereof of claim 35 or claim 36, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is increased compared to the corresponding organism or part thereof. 38. The transgenic non-human organism or part thereof of claim 37, wherein the total non-polar lipid content of the transgenic non-human organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% greater on a relative basis than the corresponding organism or part thereof. 39. The transgenic non-human organism or part thereof of any one of claims 35 to 38, wherein the polyunsaturated fatty acid content in the extracted lipid of the transgenic non-human organism or part thereof is increased when compared to the extracted lipid of the corresponding organism or part thereof. 40. The transgenic non-human organism or part thereof of any one of claims 35 to 39, wherein the content of a polyunsaturated fatty acid in the extracted lipid is increased when compared to the lipid extracted from the corresponding organism or part thereof, wherein the polyunsaturated fatty acid comprises eicosadienoic acid (EDA), arachidonic acid (ARA), alpha linolenic acid (ALA), stearidonic acid (SDA), eicosatrienoic acid (ETE), eicosatetraenoic acid (ETA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), or a combination of two of more thereof. 41. The transgenic non-human organism or part thereof of any one of claims 35 to 40, wherein the one or more exogenous polynucleotides encode: i) a MGAT, ii) a DGAT2, iii) a MGAT and a GPAT, iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT. 42. The transgenic non-human organism of claim 41, wherein the GPAT also has phosphatase activity to produce MAG, such as an Arabidopsis GPAT4 or GPAT6. 43. The transgenic non-human organism of claim 41 or claim 42, wherein the DGAT is a DGAT2. 44. The transgenic non-human organism or part thereof of any one of claims 41 to 43, wherein the exogenous polynucleotide encoding MGAT comprises one or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:1 to 44, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:45 to 82, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 45. The transgenic non-human organism or part thereof of any one of claims 41 to 44, wherein the exogenous polynucleotide encoding GPAT comprises oen or more of the following: i) a sequence of nucleotides selected from any one of SEQ ID NOs:84 to 141, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in any one of SEQ ID NOs:144 to 201, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 46. The transgenic non-human organism or part thereof of any one of claims 41 to 45, wherein the exogenous polynucleotide encoding DGAT2 comprises one or more of the following: i) a sequence of nucleotides of SEQ ID NO:204, ii) a sequence of nucleotides encoding a polypeptide comprising amino acids having a sequence as provided in SEQ ID NO:212, or a biologically active fragment thereof, iii) a sequence of nucleotides which is at least 50% identical to i) or ii), and iv) a sequence of nucleotides which hybridizes to any one of i) to iii) under stringent conditions. 47. The transgenic non-human organism or part thereof of any one of claims 44 to 46, wherein the level of the one or more non-polar lipids and/or the total lipid content of the transgenic organism or part thereof is at least 0.5% (w/w) greater on a weight basis and/or at least 1% (w/w) greater on a relative basis than a corresponding organism or part thereof lacking the one or more exogenous polynucleotides but comprising an exogenous polynucleotide encoding an Arabidopsis thaliana DGAT1. 48. The transgenic non-human organism of any one of claims 35 to 47 which is a plant, alga, or an organism suitable for fermentation such as a yeast or fungus. 49. A transgenic non-human organism comprising one or more exogenous polynucleotides encoding: i) a MGAT, ii) a DGAT2, iii) a MGAT and a GPAT, iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT. 50. The transgenic non-human organism or part thereof of claim 49, further characterised by one or more features of any one of claims 35 to 48. 51. A method of obtaining a cell with enhanced ability to produce one or more non-polar lipids, the method comprising: i) introducing into a cell one or more exogenous polynucleotides encoding a) a MGAT, b) a DGAT2 c) a MGAT and a GPAT d) a MGAT and a DGAT, or e) a MGAT, a GPAT and a DGAT. wherein the one or more exogenous polynucleotides are operably linked to one or more promoters that are capable of directing expression of the one or more exogenous polynucleotides in the cell, ii) expressing the one or more exogenous polynucleotides in the cell, iii) analysing the lipid content of the cell, and iv) selecting a cell having an increased level of one or more non-polar lipids when compared to a corresponding cell lacking the exogenous polynucleotides. 52. The method of claim 51, wherein the selected cell has one or more of the features of the organism or part thereof defined in any one of claims 36 to 48. 53. The method of claim 51 or claim 52, wherein the first and second exogenous polynucleotides are stably integrated into the genome of the cell. 54. The method of claim 53, further comprising the step of regenerating a transgenic plant from the cell of step i). 55. A transgenic cell or plant obtained using the method of any one of claims 51 to 54, or a transgenic plant part obtained therefrom. 56. Use of one or more exogenous polynucleotides encoding: i) a MGAT, ii) a DGAT2, iii) a MGAT and a GPAT iv) a MGAT and a DGAT, or v) a MGAT, a GPAT and a DGAT, for producing a transgenic non-human organism or part thereof with enhanced ability to produce one or more non-polar lipids when compared to a corresponding organism or part thereof lacking the one or more exogenous polynucleotides. 57. A trans genic seed of a plant, the seed comprising one or more exogenous polynucleotides and having an increased level of one or more non-polar lipids when compared to a corresponding seed lacking the exogenous polynucleotides. 58. The transgenic seed of claim 57, which comprises one or more of the features defined in claims 36 to 48. 59. A transgenic plant which produces the seed of claim 57 or claim 58. 60. The transgenic plant of claim 59 which is Brassica sp., Gossypium hirsutum, Linum usitatissimum, Helianthus sp., Carthamus tinctorius, Glycine max, Zea mays, Arabidopsis thaliana, Sorghum bicolor, Sorghum vulgare, Avena sativa, Trifolium sp., Camelina sativa, Miscanthus x giganteus, or Miscanthus sinensis. 61. A method of producing seed, the method comprising: i) growing the transgenic plant of claim 59 or claim 60, and ii) harvesting the seed. 62. A fermentation process comprising the steps of: i) providing a vessel containing a liquid composition comprising the transgenic non-human organism of claim 48 which is suitable for fermentation, and constituents required for fermentation and fatty acid biosynthesis, and ii) providing conditions conducive to the fermentation of the liquid composition contained in said vessel. 63. Extracted lipid obtainable by the method of any one of claims 1 to 34, or obtainable from the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, or the transgenic plant of claim 59 or claim 60. 64. An extracted lipid obtainable from a transgenic non-human organism or a part thereof comprising a DAG content that is at least 1% (w/w) on a weight basis of the extracted lipid. 65. Use of the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, the transgenic plant of claim 59 or claim 60, or the extracted lipid of claim 63 or claim 64 for the manufacture of an industrial product. 66. The use of claim 65, wherein the industrial product is fuel. 67. A method of producing fuel, the method comprising: i) reacting the lipid of claim 63 or claim 64 with an alcohol, optionally, in the presence of a catalyst, to produce alkyl esters, and ii) optionally, blending the alkyl esters with petroleum based fuel. 68. The method of claim 67, wherein the alkyl esters are methyl esters. 69. A method of producing a feedstuff, the method comprising admixing the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, the transgenic plant of claim 59 or claim 60, or the extracted lipid of claim 63 or claim 64, or an extract or portion thereof, with at least one other food ingredient. 70. Feedstuffs, cosmetics or chemicals comprising the transgenic non-human organism or part thereof of any one of claims 35 to 50, the transgenic cell or plant of claim 55, the transgenic seed of claim 57 or claim 58, the transgenic plant of claim 59 or claim 60, or the extracted lipid of claim 63 or claim 64, or an extract or portion thereof. 71. A method for identifying a nucleic acid molecule encoding an acyltransferase having an increased ability to produce MAG, DAG and/or TAG in a cell, the method comprising: i) obtaining a cell comprising a nucleic acid molecule operably linked to a promoter which is active in the cell, wherein the nucleic acid molecule comprises a sequence of nucleotides as defined in any one or more of claim 27, 28, or 29 and/or a sequence of nucleotides encoding a polypeptide having one or more amino acid sequences as provided in SEQ ID NOs:220, 221, 222, 223, 224, 225, 226 and 227, or a sequence of amino acids which is at least 50%, preferably at least 60%, more preferably at least 65% identical thereto ii) determining if the level of MAG, DAG and/or TAG produced in the cell is increased when compared to a corresponding cell lacking the nucleic acid, and iii) identifying a nucleic acid molecule encoding a acyltransferase having an increased ability to produce MAG, DAG and/or TAG in the cell. 72. The method of claim 71, wherein the acyltransferase catalyzes an enzyme reaction in the MGAT pathway. 73. The method of claim 71 or claim 72, wherein the acyltransferase is a MGAT, GPAT and/or DGAT. 74. The method of claim 73, wherein the GPAT also has phosphatase activity to produce MAG, such as an Arabidopsis GPAT4 or GPAT6. 75. The method of claim 73 or claim 74, wherein the DGAT is DGAT2. 76. The method of any one of claims 71 to 75, further comprising introducing the nucleic acid molecule into a cell prior to step i). 77. The method of any one of claims 71 to 76, wherein the cell is a plant cell. 78. The method of any one of claims 71 to 77, wherein the acyltransferase increases TAG production in the cell by a greater amount than Arabidopsis thaliana DGAT1. 79. An isolated and/or recombinant polynucleotide comprising: i) a sequence of nucleotides selected from any one of SEQ ID NOs:1 to 6, or ii) a sequence of nucleotides which is at least 80% identical to i). 80. A vector comprising the polynucleotide of claim 79. 81. A transgenic cell comprising the polynucleotide of claim 79 or the vector of claim 80. 82. A transgenic non-human organism or part thereof comprising the polynucleotide of claim 79, the vector of claim 80, or the transgenic cell of claim 81.
1,600
354
13,683,444
1,613
Disclosed are systems for use in administering inhaled nitric oxide gas as a therapeutic treatment, where the systems reduce the risk of inducing an increase in pulmonary capillary wedge pressure leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising a source of pharmaceutically acceptable nitric oxide gas for inhalation, a delivery device suitable for delivering nitric oxide gas from the source to a term or near-term neonate patient, and instructions for use in operating the system.
1. A system for use in administering inhaled nitric oxide gas, which system reduces the risk of inducing an increase in pulmonary capillary wedge pressure (PCWP) leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising: a source of pharmaceutically acceptable nitric oxide gas for inhalation; a delivery device suitable for delivering nitric oxide gas from the source to a term or near-term neonate patient; and instructions for use in operating the system, the instructions comprising: (i) a description of using the system to deliver 20 ppm inhaled nitric oxide gas to treat hypoxic respiratory failure in a term or near-term neonate; (ii) a contraindication for neonates who are dependent on right to left shunting of blood; and (iii) a warning that a patient with pre-existing left ventricular dysfunction may experience increased PCWP leading to pulmonary edema when treated with inhaled nitric oxide gas, wherein the warning: is effective to reduce use of inhaled nitric oxide gas to treat neonate patients who have hypoxic respiratory failure and pre-existing left ventricular dysfunction but who are not dependent on right to left shunting of blood, and thereby reduces the risk of occurrence of increased PCWP leading to pulmonary edema in neonates who have hypoxic respiratory failure. 2. The system of claim 1, wherein the warning reduces the risk of occurrence of increased PCWP leading to pulmonary edema resulting from treatment with 20 ppm nitric oxide during use of the system in neonates who have hypoxic respiratory failure. 3. The system of claim 1, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a compressed gas supplied in a cylinder. 4. The system of claim 1, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a mixture of nitric oxide gas and nitrogen gas. 5. The system of claim 1, wherein the warning also says that patients with pre-existing left ventricular dysfunction may experience other adverse events, besides increased PCWP leading to pulmonary edema, upon treatment with inhaled nitric oxide gas. 6. A system for use in administering inhaled nitric oxide gas, which system reduces the risk of inducing an increase in PCWP leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising: a source of pharmaceutically acceptable nitric oxide gas for inhalation; a delivery device suitable for delivering nitric oxide gas from the source to a term or near-term neonate patient; and instructions for use in operating the system, the instructions comprising (i) a description of using the system to deliver 20 ppm inhaled nitric oxide gas to treat hypoxic respiratory failure in a term or near-term neonate; and (ii) a warning that a patient with pre-existing left ventricular dysfunction may experience increased PCWP leading to pulmonary edema when treated with inhaled nitric oxide gas, wherein the warning: is effective to reduce use of inhaled nitric oxide gas to treat neonate patients who have hypoxic respiratory failure and pre-existing left ventricular dysfunction but who are not dependent on right to left shunting of blood, and thereby reduces the risk of occurrence of increased PCWP leading to pulmonary edema in neonates that have hypoxic respiratory failure. 7. The system of claim 6, wherein the warning reduces the risk of occurrence of increased PCWP leading to pulmonary edema resulting from treatment with 20 ppm nitric oxide during use of the system in neonates who have hypoxic respiratory failure. 8. The system of claim 6, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a compressed gas supplied in a cylinder. 9. The system of claim 6, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a mixture of nitric oxide gas and nitrogen gas. 10. The system of claim 6, wherein the warning also says that patients with pre-existing left ventricular dysfunction may experience other adverse events, besides increased PCWP leading to pulmonary edema, upon treatment with inhaled nitric oxide gas. 11. A system for use in administering inhaled nitric oxide gas, which system reduces the risk of inducing increased PCWP leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising: a source of pharmaceutically acceptable nitric oxide gas for inhalation; and instructions for administering nitric oxide gas from the source to treat hypoxic respiratory failure in a term or near-term neonate at a dosage of 20 ppm nitric oxide gas, the instructions comprising a warning that a patient with pre-existing left ventricular dysfunction may experience increased PCWP leading to pulmonary edema when treated with inhaled nitric oxide gas, wherein the warning: is effective to reduce use of inhaled nitric oxide gas to treat neonate patients who have hypoxic respiratory failure and pre-existing left ventricular dysfunction but who are not dependent on right to left shunting of blood, and thereby reduces the risk of occurrence of increased PCWP leading to pulmonary edema in neonates that have hypoxic respiratory failure. 12. The system of claim 11, wherein the warning reduces the risk of occurrence of increased PCWP leading to pulmonary edema resulting from treatment with 20 ppm nitric oxide during use of the system in neonates who have hypoxic respiratory failure. 13. The system of claim 11, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a compressed gas supplied in a cylinder. 14. The system of claim 11, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a mixture of nitric oxide gas and nitrogen gas. 15. The system of claim 11, wherein the warning also says that patients with pre-existing left ventricular dysfunction may experience other adverse events, besides increased PCWP leading to pulmonary edema, upon treatment with inhaled nitric oxide gas.
Disclosed are systems for use in administering inhaled nitric oxide gas as a therapeutic treatment, where the systems reduce the risk of inducing an increase in pulmonary capillary wedge pressure leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising a source of pharmaceutically acceptable nitric oxide gas for inhalation, a delivery device suitable for delivering nitric oxide gas from the source to a term or near-term neonate patient, and instructions for use in operating the system.1. A system for use in administering inhaled nitric oxide gas, which system reduces the risk of inducing an increase in pulmonary capillary wedge pressure (PCWP) leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising: a source of pharmaceutically acceptable nitric oxide gas for inhalation; a delivery device suitable for delivering nitric oxide gas from the source to a term or near-term neonate patient; and instructions for use in operating the system, the instructions comprising: (i) a description of using the system to deliver 20 ppm inhaled nitric oxide gas to treat hypoxic respiratory failure in a term or near-term neonate; (ii) a contraindication for neonates who are dependent on right to left shunting of blood; and (iii) a warning that a patient with pre-existing left ventricular dysfunction may experience increased PCWP leading to pulmonary edema when treated with inhaled nitric oxide gas, wherein the warning: is effective to reduce use of inhaled nitric oxide gas to treat neonate patients who have hypoxic respiratory failure and pre-existing left ventricular dysfunction but who are not dependent on right to left shunting of blood, and thereby reduces the risk of occurrence of increased PCWP leading to pulmonary edema in neonates who have hypoxic respiratory failure. 2. The system of claim 1, wherein the warning reduces the risk of occurrence of increased PCWP leading to pulmonary edema resulting from treatment with 20 ppm nitric oxide during use of the system in neonates who have hypoxic respiratory failure. 3. The system of claim 1, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a compressed gas supplied in a cylinder. 4. The system of claim 1, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a mixture of nitric oxide gas and nitrogen gas. 5. The system of claim 1, wherein the warning also says that patients with pre-existing left ventricular dysfunction may experience other adverse events, besides increased PCWP leading to pulmonary edema, upon treatment with inhaled nitric oxide gas. 6. A system for use in administering inhaled nitric oxide gas, which system reduces the risk of inducing an increase in PCWP leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising: a source of pharmaceutically acceptable nitric oxide gas for inhalation; a delivery device suitable for delivering nitric oxide gas from the source to a term or near-term neonate patient; and instructions for use in operating the system, the instructions comprising (i) a description of using the system to deliver 20 ppm inhaled nitric oxide gas to treat hypoxic respiratory failure in a term or near-term neonate; and (ii) a warning that a patient with pre-existing left ventricular dysfunction may experience increased PCWP leading to pulmonary edema when treated with inhaled nitric oxide gas, wherein the warning: is effective to reduce use of inhaled nitric oxide gas to treat neonate patients who have hypoxic respiratory failure and pre-existing left ventricular dysfunction but who are not dependent on right to left shunting of blood, and thereby reduces the risk of occurrence of increased PCWP leading to pulmonary edema in neonates that have hypoxic respiratory failure. 7. The system of claim 6, wherein the warning reduces the risk of occurrence of increased PCWP leading to pulmonary edema resulting from treatment with 20 ppm nitric oxide during use of the system in neonates who have hypoxic respiratory failure. 8. The system of claim 6, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a compressed gas supplied in a cylinder. 9. The system of claim 6, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a mixture of nitric oxide gas and nitrogen gas. 10. The system of claim 6, wherein the warning also says that patients with pre-existing left ventricular dysfunction may experience other adverse events, besides increased PCWP leading to pulmonary edema, upon treatment with inhaled nitric oxide gas. 11. A system for use in administering inhaled nitric oxide gas, which system reduces the risk of inducing increased PCWP leading to pulmonary edema in neonatal patients who have hypoxic respiratory failure and are candidates for inhaled nitric oxide treatment, the system comprising: a source of pharmaceutically acceptable nitric oxide gas for inhalation; and instructions for administering nitric oxide gas from the source to treat hypoxic respiratory failure in a term or near-term neonate at a dosage of 20 ppm nitric oxide gas, the instructions comprising a warning that a patient with pre-existing left ventricular dysfunction may experience increased PCWP leading to pulmonary edema when treated with inhaled nitric oxide gas, wherein the warning: is effective to reduce use of inhaled nitric oxide gas to treat neonate patients who have hypoxic respiratory failure and pre-existing left ventricular dysfunction but who are not dependent on right to left shunting of blood, and thereby reduces the risk of occurrence of increased PCWP leading to pulmonary edema in neonates that have hypoxic respiratory failure. 12. The system of claim 11, wherein the warning reduces the risk of occurrence of increased PCWP leading to pulmonary edema resulting from treatment with 20 ppm nitric oxide during use of the system in neonates who have hypoxic respiratory failure. 13. The system of claim 11, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a compressed gas supplied in a cylinder. 14. The system of claim 11, wherein the source of pharmaceutically acceptable nitric oxide gas for inhalation is a mixture of nitric oxide gas and nitrogen gas. 15. The system of claim 11, wherein the warning also says that patients with pre-existing left ventricular dysfunction may experience other adverse events, besides increased PCWP leading to pulmonary edema, upon treatment with inhaled nitric oxide gas.
1,600
355
15,519,559
1,627
Disclosed herein are methods and formulation for enhancing oral availability of CPT-11 during cancer therapy while at the same time, reducing its gastrointestinal (GI) toxicity, thus the methods and formulations as disclosed herein may augment the efficacy of cancer therapy.
1. A method of reducing CPT-11 induced gastrointestinal (GI) toxicity in a subject underwent a CPT-11 treatment, comprising, administering 1-50 mg/Kg of silychristin to the subject so as to ameliorate or alleviate symptoms of CPT-11 induced GI toxicity. 2. The method of claim 1, wherein the silychristin is administered to the subject at about 8 mg/kg. 3. The method of claim 1, wherein the silychristin is administered prior to, together with, or after the CPT-11 treatment for at least 5 days. 4. The method of claim 1, wherein the silychristin is administered prior to, together with, or after the CPT-11 treatment for at least 28 days. 5. The method of claim 1, wherein the subject has a cancer selected from the group consisting of, breast cancer, brain tumor, melanoma, lung cancer, lymphoma, neuroepithelioma, kidney cancer, prostate cancer, stomach cancer, colon cancer, rectal cancer, pancreatic cancer and uterus cancer. 6. The method of claim 5, wherein the cancer is colon cancer or rectal cancer. 7. The method of claim 5, wherein the cancer is metastatic. 8. The method of claim 1, wherein the subject is a human. 9. The method of claim 1, wherein the CPT-11 induced GI toxicity is diarrhea or bloody stool. 10. A method for improving oral availability of CPT-11 while reducing its gastrointestinal (GI) toxicity in a subject underwent a CPT-11 treatment, comprising administering to the subject in sequence, one dose of ursodeoxycholic acid (UDCA) that is about 0.1-10 mg/Kg; and at least one dose of silymarin, in which each dose of silymarin is about 1-50 mg/Kg; wherein each doses of silymarin is administered prior to, concurrently with, or after the CPT-11 treatment. 11. The method of claim 10, wherein the silymarin comprises silychristin. 12. The method of claim 11, wherein the UDCA is administered at about 2 mg/Kg. 13. The method of claim 11, wherein 5 doses of silymarin are administered to the subject, in which each doses of silymarin is about 8 mg/Kg. 14. The method of claim 11, wherein 28 doses of silymarin are administered to the subject, in which each doses of silymarin is about 8 mg/Kg. 15. The method of claim 11, wherein the CPT-11 treatment comprises administering one or more doses of CPT-11 to the subject after the administration of UDCA, in which each dose of CPT-11 is about 0.5-15 mg/Kg. 16. The method of claim 11, wherein the subject has a cancer selected from the group consisting of, breast cancer, brain tumor, melanoma, lung cancer, lymphoma, neuroepithelioma, kidney cancer, prostate cancer, stomach cancer, colon cancer, rectal cancer, pancreatic cancer and uterus cancer. 17. The method of claim 16, wherein the cancer is colon cancer or rectal cancer. 18. The method of claim 16, wherein the cancer is metastatic. 19. The method of claim 11, wherein the subject is a human 20. The method of claim 11, wherein the GI toxicity is CPT-11 induced diarrhea or bloody stool. 21-57. (canceled)
Disclosed herein are methods and formulation for enhancing oral availability of CPT-11 during cancer therapy while at the same time, reducing its gastrointestinal (GI) toxicity, thus the methods and formulations as disclosed herein may augment the efficacy of cancer therapy.1. A method of reducing CPT-11 induced gastrointestinal (GI) toxicity in a subject underwent a CPT-11 treatment, comprising, administering 1-50 mg/Kg of silychristin to the subject so as to ameliorate or alleviate symptoms of CPT-11 induced GI toxicity. 2. The method of claim 1, wherein the silychristin is administered to the subject at about 8 mg/kg. 3. The method of claim 1, wherein the silychristin is administered prior to, together with, or after the CPT-11 treatment for at least 5 days. 4. The method of claim 1, wherein the silychristin is administered prior to, together with, or after the CPT-11 treatment for at least 28 days. 5. The method of claim 1, wherein the subject has a cancer selected from the group consisting of, breast cancer, brain tumor, melanoma, lung cancer, lymphoma, neuroepithelioma, kidney cancer, prostate cancer, stomach cancer, colon cancer, rectal cancer, pancreatic cancer and uterus cancer. 6. The method of claim 5, wherein the cancer is colon cancer or rectal cancer. 7. The method of claim 5, wherein the cancer is metastatic. 8. The method of claim 1, wherein the subject is a human. 9. The method of claim 1, wherein the CPT-11 induced GI toxicity is diarrhea or bloody stool. 10. A method for improving oral availability of CPT-11 while reducing its gastrointestinal (GI) toxicity in a subject underwent a CPT-11 treatment, comprising administering to the subject in sequence, one dose of ursodeoxycholic acid (UDCA) that is about 0.1-10 mg/Kg; and at least one dose of silymarin, in which each dose of silymarin is about 1-50 mg/Kg; wherein each doses of silymarin is administered prior to, concurrently with, or after the CPT-11 treatment. 11. The method of claim 10, wherein the silymarin comprises silychristin. 12. The method of claim 11, wherein the UDCA is administered at about 2 mg/Kg. 13. The method of claim 11, wherein 5 doses of silymarin are administered to the subject, in which each doses of silymarin is about 8 mg/Kg. 14. The method of claim 11, wherein 28 doses of silymarin are administered to the subject, in which each doses of silymarin is about 8 mg/Kg. 15. The method of claim 11, wherein the CPT-11 treatment comprises administering one or more doses of CPT-11 to the subject after the administration of UDCA, in which each dose of CPT-11 is about 0.5-15 mg/Kg. 16. The method of claim 11, wherein the subject has a cancer selected from the group consisting of, breast cancer, brain tumor, melanoma, lung cancer, lymphoma, neuroepithelioma, kidney cancer, prostate cancer, stomach cancer, colon cancer, rectal cancer, pancreatic cancer and uterus cancer. 17. The method of claim 16, wherein the cancer is colon cancer or rectal cancer. 18. The method of claim 16, wherein the cancer is metastatic. 19. The method of claim 11, wherein the subject is a human 20. The method of claim 11, wherein the GI toxicity is CPT-11 induced diarrhea or bloody stool. 21-57. (canceled)
1,600
356
14,525,903
1,649
Methods are provided for treating and/or reducing the severity of multiple sclerosis in a human, by administering autologous mesenchymal stem cell-derived neural precursors. Also described is an in vitro method for differentiating mesenchymal stem-cell derived neural precursor oligodengroglial and neuronal cell types.
1-15. (canceled) 16. Isolated neural precursor cells, prepared by a process comprising the steps of: (a) expanding mesenchymal stem cells isolated from a human subject, (b) culturing a portion of the expanded mesenchymal stem cells obtained in step (a) in a neural progenitor basal medium (NPBM) supplemented with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and neural cell survival factor-1 (NSF-1), (c) collecting floating cell clusters from the culture of step (b), (d) measuring, in a test portion of the collected cells, expression of nestin, glial fibrillary acidic protein (GFAP), neurofilament-M (NF-M), and alpha smooth muscle (αSM) actin, relative to expression of nestin, GFAP, NF-M, and αSM actin in a test portion of the expanded mesenchymal stem cells obtained from step (a), and (e) selecting the remaining portion of the collected cells as neural precursors based on increased expression of nestin, GFAP, and NF-M, and decreased expression of αSM actin in said test portion of the collected cells, relative to the expression of nestin, GFAP, NF-M, and αSM actin in said test portion of the expanded mesenchymal stem cells. 17. The isolated neural precursor cells of claim 16, wherein said mesenchymal stem cells isolated from a human subject are isolated from the bone marrow of said human subject. 18. The isolated neural precursor cells of claim 16, wherein said mesenchymal stem cells isolated from a human subject are stored in cryopreservation media containing autologous serum and DMSO. 19. The isolated neural precursor cells of claim 16, wherein a portion of said isolated mesenchymal stem cells are tested for sterility, viability, and expression of one or more of: CD105, CD73, CD90, CD45, CD34, CD14, CD79, and HLA-DR, prior to expansion of said mesenchymal stem cells in step (a). 20. The isolated neural precursor cells of claim 16, wherein in step (a) said mesenchymal stem cells are expanded in mesenchymal stem cell basal medium comprising autologous serum. 21. The isolated neural precursor cells of claim 16, wherein said portion of the expanded mesenchymal stem cells are cultured in step (b) for 10-15 days. 22. The isolated neural precursor cells of claim 16, wherein said neural precursor cells are tested for ability to further differentiate by culturing the cells selected in step (e) in a medium comprising basic fibroblast growth factor. 23. The isolated neural precursor cells of claim 16, wherein said human subject suffers from multiple sclerosis. 24. A composition comprising the isolated neural precursor cells of claim 16 in a suitable carrier. 25. The composition of claim 24, wherein the suitable carrier is sterile sodium chloride.
Methods are provided for treating and/or reducing the severity of multiple sclerosis in a human, by administering autologous mesenchymal stem cell-derived neural precursors. Also described is an in vitro method for differentiating mesenchymal stem-cell derived neural precursor oligodengroglial and neuronal cell types.1-15. (canceled) 16. Isolated neural precursor cells, prepared by a process comprising the steps of: (a) expanding mesenchymal stem cells isolated from a human subject, (b) culturing a portion of the expanded mesenchymal stem cells obtained in step (a) in a neural progenitor basal medium (NPBM) supplemented with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and neural cell survival factor-1 (NSF-1), (c) collecting floating cell clusters from the culture of step (b), (d) measuring, in a test portion of the collected cells, expression of nestin, glial fibrillary acidic protein (GFAP), neurofilament-M (NF-M), and alpha smooth muscle (αSM) actin, relative to expression of nestin, GFAP, NF-M, and αSM actin in a test portion of the expanded mesenchymal stem cells obtained from step (a), and (e) selecting the remaining portion of the collected cells as neural precursors based on increased expression of nestin, GFAP, and NF-M, and decreased expression of αSM actin in said test portion of the collected cells, relative to the expression of nestin, GFAP, NF-M, and αSM actin in said test portion of the expanded mesenchymal stem cells. 17. The isolated neural precursor cells of claim 16, wherein said mesenchymal stem cells isolated from a human subject are isolated from the bone marrow of said human subject. 18. The isolated neural precursor cells of claim 16, wherein said mesenchymal stem cells isolated from a human subject are stored in cryopreservation media containing autologous serum and DMSO. 19. The isolated neural precursor cells of claim 16, wherein a portion of said isolated mesenchymal stem cells are tested for sterility, viability, and expression of one or more of: CD105, CD73, CD90, CD45, CD34, CD14, CD79, and HLA-DR, prior to expansion of said mesenchymal stem cells in step (a). 20. The isolated neural precursor cells of claim 16, wherein in step (a) said mesenchymal stem cells are expanded in mesenchymal stem cell basal medium comprising autologous serum. 21. The isolated neural precursor cells of claim 16, wherein said portion of the expanded mesenchymal stem cells are cultured in step (b) for 10-15 days. 22. The isolated neural precursor cells of claim 16, wherein said neural precursor cells are tested for ability to further differentiate by culturing the cells selected in step (e) in a medium comprising basic fibroblast growth factor. 23. The isolated neural precursor cells of claim 16, wherein said human subject suffers from multiple sclerosis. 24. A composition comprising the isolated neural precursor cells of claim 16 in a suitable carrier. 25. The composition of claim 24, wherein the suitable carrier is sterile sodium chloride.
1,600
357
15,609,253
1,611
The present invention relates to compositions and methods for the prevention and treatment of skin disorders and for the rejuvenation of the skin. In particular, the application describes topical compositions and methods of treatments comprising the combined use of one or more cannabinoids and one or more hydroxy acids in a suitable carrier.
1. A topical composition for the rejuvenation or treatment of skin in the form of an ointment, a cream, an emulsion, a lotion, a paste, an unguent, a gel or a sunscreen comprising therapeutically effective amounts of at least one cannabinoid and at least one hydroxy acid in a topically acceptable carrier, wherein the cannabinoid is present in a concentration between 0.1 and 30% by weight of the composition; wherein the hydroxy acid is present in a concentration between 0.1 and 10% by weight of the composition; and wherein the cannabinoid is one or more of a natural phytocannabinoid, an organic cannabinoid, an endocannabinoid, a cannabinoid analog, a cannabinoid derivative, a synthetic cannabinoid and a cannabinoid receptor agonist. 2. The topical composition of claim 1, wherein the organic cannabinoid is hemp oil or human breast milk. 3. The topical composition of claim 1, wherein the cannabinoid is one or more of cannabigerolic acid (CBGA), cannabigerolic acid monomethylether (CBGAM), cannabigerol (CBG), cannabigerol monomethylether (CBGM), cannabigerovarinic acid (CBGVA), cannabigerovarin (CBGV), cannabichromenic acid (CBCA), cannabichromene (CBC), cannabichromevarinic acid (CBCVA), cannabichromevarin (CBCV), cannabidiolic acid (CBDA), cannabidiol (CBD), cannabidiol monomethylether (CBDM), cannabidiol-C4 (CBD-C4), cannabidivarinic acid (CBDVA), cannabidivarin (CBDV), cannabidiorcol (CBD-C1), delta-9-tetrahydrocannabinolic acid A (THCA-A), delta-9-tetrahydrocannabinolic acid B (THCA-B), delta-9-tetrahydrocannabinol (THC), delta-9-tetrahydrocannabinolic acid-C4 (THCA-C4), delta-9-tetrahydrocannabinol-C4 (THC-C4), delta-9-tetrahydrocannabivarinic acid (THCVA), delta-9-tetrahydrocannabivarin (THCV), delta-9-tetrahydrocannabiorcolic acid (THCA-C1), delta-9-tetrahydrocannabiorcol (THC-C1), delta-7-cis-iso-tetrahydrocannabivarin, delta-8-tetrahydrocannabinolic acid (Δ8-THCA), delta-8-tetrahydrocannabinol (Δ8-THC), cannabicyclolic acid (CBLA), cannabicyclol (CBL), cannabicyclovarin (CBLV), cannnabielsoic acid A (CBEA-A), cannabielsoic acid B (CBEA-B), cannabielsoin (CBE), cannabinolic acid (CBNA), cannabinol (CBN), cannabinol methylether (CBNM), cannabinol-C4 (CBN-C4), cannabivarin (CBV), cannabinol-C2 (CBN-C2), cannabiorcol (CBN-C1), cannabinodiol (CBND), cannabinodivarin (CBVD), cannabitriol (CBT), 10-ethoxy-9-hydroxy-delta-6a-tetrahydrocannabinol, 8,9-dihydroxy-delta-6a-tetrahydrocannabinol, cannabitriolvarin (CBTV), ethoxy-cannabitriolvarin (CBTVE), dehydrocannabifuran (DCBF), cannabifuran (CBF), cannabichromanon (CBCN), cannabicitran (CBT), 10-oxo-delta-6a-tetrahydrocannabinol (OTHC), delta-9-cis-tetrahydrocannabinol (cis-THC), 3,4,5,6-tetrahydro-7-hydroxy-alpha-alpha-2-trimethyl-9-n-propyl-2,6-methano-2H-1-benzoxocin-5-methanol (OH-iso-HHCV), cannabiripsol (CBR) and trihydroxy-delta-9-tetrahydrocannabinol (triOH-THC). 4. The topical composition of claim 1, wherein the cannabinoid receptor agonist comprises one or more of a naphthoylindole, a naphthylmethylindole, a naphthoylpyrrole, a naphthylmethylindene, a phenylacetylindole and a cyclohexylphenol. 5. The topical composition of claim 1, wherein the hydroxy acid is an alpha hydroxy acid, a beta hydroxy acid or a combination thereof. 6. The topical composition of claim 5, wherein the alpha hydroxy acid is lactic acid, citric acid, glycolic acid, mandelic acid, benzylic acid, malic acid, tartaric acid, gluconolactone, galactonolactone, glucuronolactone, galacturonolactone, gulonolactone, ribonolactone, saccharic acid lactone, pantoyllactone, glucoheptonolactone, mannonolactone, or galactoheptonolactone. 7. The topical composition of claim 5, wherein the beta hydroxy acid is salicylic acid or lipohydroxy acid. 8. The topical composition of claim 1, further comprising a stabilizer selected from the group consisting of guar gum, xanthan gum cellulose hyaluronic acid, polyvinyl pyrrolidone (PVP), alginate, chondritin sulfate, poly gamma glutamic acid, gelatin, chitisin, corn starch and flour, in an amount from about 0.25% to about 30% (w/v). 9. The topical composition of claim 1, wherein the carrier comprises hemp oil and wherein the topical composition further comprises one or more of a thickening agent, an antibiotic, an antiseptic agent, an antifungal, an antibacterial agent, an analgesic, an antiviral agent or a UV absorbing agent in an amount between 0.1 and 5% by weight of the composition. 10. A method to treat a skin disorder or rejuvenate the skin in a subject in need thereof comprising topically administering to the subject a composition in the form of an ointment, a cream, an emulsion, a lotion, a paste, an unguent, a gel or a sunscreen comprising therapeutically effective amounts of at least one cannabinoid and at least one hydroxy acid in a pharmaceutically acceptable carrier, wherein the cannabinoid is present in a concentration between 0.1 and 30% by weight of the composition; wherein the hydroxy acid is present in a concentration between 0.1 and 10% by weight of the composition; and wherein the cannabinoid is one or more of a natural phytocannabinoid, an organic cannabinoid, an endocannabinoid, a cannabinoid analog, a cannabinoid derivative, a synthetic cannabinoid and a cannabinoid receptor agonist. 11. The method of claim 10, wherein the skin disorder is one or more of eczema, psoriasis, dermatitis, itching dermatosis, rosacea, perioral dermatitis, acne, non-melanoma cancer or melanoma. 12. The method of claim 10, wherein the subject presents a symptom which is one or more of pruritus, dryness, skin rash, redness, swelling of the skin, itching, crusting, flaking, blistering, cracking, oozing, or bleeding of the skin. 13. The method of claim 11, wherein the dermatitis is atopic dermatitis, contact dermatitis, xerotic eczema, or seborrheic dermatitis. 14. The method of claim 10, wherein the organic cannabinoid is hemp oil or human breast milk. 15. The method of claim 10, wherein the cannabinoid is one or more of cannabigerolic acid (CBGA), cannabigerolic acid monomethylether (CBGAM), cannabigerol (CBG), cannabigerol monomethylether (CBGM), cannabigerovarinic acid (CBGVA), cannabigerovarin (CBGV), cannabichromenic acid (CBCA), cannabichromene (CBC), cannabichromevarinic acid (CBCVA), cannabichromevarin (CBCV), cannabidiolic acid (CBDA), cannabidiol (CBD), cannabidiol monomethylether (CBDM), cannabidiol-C4 (CBD-C4), cannabidivarinic acid (CBDVA), cannabidivarin (CBDV), cannabidiorcol (CBD-C1), delta-9-tetrahydrocannabinolic acid A (THCA-A), delta-9-tetrahydrocannabinolic acid B (THCA-B), delta-9-tetrahydrocannabinol (THC), delta-9-tetrahydrocannabinolic acid-C4 (THCA-C4), delta-9-tetrahydrocannabinol-C4 (THC-C4), delta-9-tetrahydrocannabivarinic acid (THCVA), delta-9-tetrahydrocannabivarin (THCV), delta-9-tetrahydrocannabiorcolic acid (THCA-C1), delta-9-tetrahydrocannabiorcol (THC-C1), delta-7-cis-iso-tetrahydrocannabivarin, delta-8-tetrahydrocannabinolic acid (Δ8-THCA), delta-8-tetrahydrocannabinol (Δ8-THC), cannabicyclolic acid (CBLA), cannabicyclol (CBL), cannabicyclovarin (CBLV), cannnabielsoic acid A (CBEA-A), cannabielsoic acid B (CBEA-B), cannabielsoin (CBE), cannabinolic acid (CBNA), cannabinol (CBN), cannabinol methylether (CBNM), cannabinol-C4 (CBN-C4), cannabivarin (CBV), cannabinol-C2 (CBN-C2), cannabiorcol (CBN-C1), cannabinodiol (CBND), cannabinodivarin (CBVD), cannabitriol (CBT), 10-ethoxy-9-hydroxy-delta-6a-tetrahydrocannabinol, 8,9-dihydroxy-delta-6a-tetrahydrocannabinol, cannabitriolvarin (CBTV), ethoxy-cannabitriolvarin (CBTVE), dehydrocannabifuran (DCBF), cannabifuran (CBF), cannabichromanon (CBCN), cannabicitran (CBT), 10-oxo-delta-6a-tetrahydrocannabinol (OTHC), delta-9-cis-tetrahydrocannabinol (cis-THC), 3,4,5,6-tetrahydro-7-hydroxy-alpha-alpha-2-trimethyl-9-n-propyl-2,6-methano-2H-1-benzoxocin-5-methanol (OH-iso-HHCV), cannabiripsol (CBR) and trihydroxy-delta-9-tetrahydrocannabinol (triOH-THC). 16. The method of claim 10, wherein the cannabinoid receptor agonist comprises one or more of a naphthoylindole, a naphthylmethylindole, a naphthoylpyrrole, a naphthylmethylindene, a phenylacetylindole and a cyclohexylphenol. 17. The method of claim 10, wherein the hydroxy acid is an alpha hydroxy acid, a beta hydroxy acid or a combination thereof. 18. The method of claim 17, wherein the alpha hydroxy acid is lactic acid, citric acid, glycolic acid, mandelic acid, benzylic acid, malic acid, tartaric acid, gluconolactone, galactonolactone, glucuronolactone, galacturonolactone, gulonolactone, ribonolactone, saccharic acid lactone, pantoyllactone, glucoheptonolactone, mannonolactone, or galactoheptonolactone. 19. The method of claim 17, wherein the beta hydroxy acid is salicylic acid or lipohydroxy acid. 20. The method of claim 10, wherein the composition further comprises a stabilizer selected from the group consisting of guar gum, xanthan gum cellulose hyaluronic acid, polyvinyl pyrrolidone (PVP), alginate, chondritin sulfate, poly gamma glutamic acid, gelatin, chitisin, corn starch and flour, in an amount from about 0.25% to about 30% (w/v). 21. The method of claim 10, wherein the carrier comprises hemp oil and wherein the composition further comprises one or more of a thickening agent, an antibiotic, an antiseptic agent, an antifungal, an antibacterial agent, an analgesic, an antiviral agent or a UV absorbing agent in an amount between 0.1 and 5% by weight of the composition. 22. The method of claim 10, wherein the subject is a human and wherein the composition is topically administered to the subject in an amount between about 100 nmol to about 1 μmol/cm2.
The present invention relates to compositions and methods for the prevention and treatment of skin disorders and for the rejuvenation of the skin. In particular, the application describes topical compositions and methods of treatments comprising the combined use of one or more cannabinoids and one or more hydroxy acids in a suitable carrier.1. A topical composition for the rejuvenation or treatment of skin in the form of an ointment, a cream, an emulsion, a lotion, a paste, an unguent, a gel or a sunscreen comprising therapeutically effective amounts of at least one cannabinoid and at least one hydroxy acid in a topically acceptable carrier, wherein the cannabinoid is present in a concentration between 0.1 and 30% by weight of the composition; wherein the hydroxy acid is present in a concentration between 0.1 and 10% by weight of the composition; and wherein the cannabinoid is one or more of a natural phytocannabinoid, an organic cannabinoid, an endocannabinoid, a cannabinoid analog, a cannabinoid derivative, a synthetic cannabinoid and a cannabinoid receptor agonist. 2. The topical composition of claim 1, wherein the organic cannabinoid is hemp oil or human breast milk. 3. The topical composition of claim 1, wherein the cannabinoid is one or more of cannabigerolic acid (CBGA), cannabigerolic acid monomethylether (CBGAM), cannabigerol (CBG), cannabigerol monomethylether (CBGM), cannabigerovarinic acid (CBGVA), cannabigerovarin (CBGV), cannabichromenic acid (CBCA), cannabichromene (CBC), cannabichromevarinic acid (CBCVA), cannabichromevarin (CBCV), cannabidiolic acid (CBDA), cannabidiol (CBD), cannabidiol monomethylether (CBDM), cannabidiol-C4 (CBD-C4), cannabidivarinic acid (CBDVA), cannabidivarin (CBDV), cannabidiorcol (CBD-C1), delta-9-tetrahydrocannabinolic acid A (THCA-A), delta-9-tetrahydrocannabinolic acid B (THCA-B), delta-9-tetrahydrocannabinol (THC), delta-9-tetrahydrocannabinolic acid-C4 (THCA-C4), delta-9-tetrahydrocannabinol-C4 (THC-C4), delta-9-tetrahydrocannabivarinic acid (THCVA), delta-9-tetrahydrocannabivarin (THCV), delta-9-tetrahydrocannabiorcolic acid (THCA-C1), delta-9-tetrahydrocannabiorcol (THC-C1), delta-7-cis-iso-tetrahydrocannabivarin, delta-8-tetrahydrocannabinolic acid (Δ8-THCA), delta-8-tetrahydrocannabinol (Δ8-THC), cannabicyclolic acid (CBLA), cannabicyclol (CBL), cannabicyclovarin (CBLV), cannnabielsoic acid A (CBEA-A), cannabielsoic acid B (CBEA-B), cannabielsoin (CBE), cannabinolic acid (CBNA), cannabinol (CBN), cannabinol methylether (CBNM), cannabinol-C4 (CBN-C4), cannabivarin (CBV), cannabinol-C2 (CBN-C2), cannabiorcol (CBN-C1), cannabinodiol (CBND), cannabinodivarin (CBVD), cannabitriol (CBT), 10-ethoxy-9-hydroxy-delta-6a-tetrahydrocannabinol, 8,9-dihydroxy-delta-6a-tetrahydrocannabinol, cannabitriolvarin (CBTV), ethoxy-cannabitriolvarin (CBTVE), dehydrocannabifuran (DCBF), cannabifuran (CBF), cannabichromanon (CBCN), cannabicitran (CBT), 10-oxo-delta-6a-tetrahydrocannabinol (OTHC), delta-9-cis-tetrahydrocannabinol (cis-THC), 3,4,5,6-tetrahydro-7-hydroxy-alpha-alpha-2-trimethyl-9-n-propyl-2,6-methano-2H-1-benzoxocin-5-methanol (OH-iso-HHCV), cannabiripsol (CBR) and trihydroxy-delta-9-tetrahydrocannabinol (triOH-THC). 4. The topical composition of claim 1, wherein the cannabinoid receptor agonist comprises one or more of a naphthoylindole, a naphthylmethylindole, a naphthoylpyrrole, a naphthylmethylindene, a phenylacetylindole and a cyclohexylphenol. 5. The topical composition of claim 1, wherein the hydroxy acid is an alpha hydroxy acid, a beta hydroxy acid or a combination thereof. 6. The topical composition of claim 5, wherein the alpha hydroxy acid is lactic acid, citric acid, glycolic acid, mandelic acid, benzylic acid, malic acid, tartaric acid, gluconolactone, galactonolactone, glucuronolactone, galacturonolactone, gulonolactone, ribonolactone, saccharic acid lactone, pantoyllactone, glucoheptonolactone, mannonolactone, or galactoheptonolactone. 7. The topical composition of claim 5, wherein the beta hydroxy acid is salicylic acid or lipohydroxy acid. 8. The topical composition of claim 1, further comprising a stabilizer selected from the group consisting of guar gum, xanthan gum cellulose hyaluronic acid, polyvinyl pyrrolidone (PVP), alginate, chondritin sulfate, poly gamma glutamic acid, gelatin, chitisin, corn starch and flour, in an amount from about 0.25% to about 30% (w/v). 9. The topical composition of claim 1, wherein the carrier comprises hemp oil and wherein the topical composition further comprises one or more of a thickening agent, an antibiotic, an antiseptic agent, an antifungal, an antibacterial agent, an analgesic, an antiviral agent or a UV absorbing agent in an amount between 0.1 and 5% by weight of the composition. 10. A method to treat a skin disorder or rejuvenate the skin in a subject in need thereof comprising topically administering to the subject a composition in the form of an ointment, a cream, an emulsion, a lotion, a paste, an unguent, a gel or a sunscreen comprising therapeutically effective amounts of at least one cannabinoid and at least one hydroxy acid in a pharmaceutically acceptable carrier, wherein the cannabinoid is present in a concentration between 0.1 and 30% by weight of the composition; wherein the hydroxy acid is present in a concentration between 0.1 and 10% by weight of the composition; and wherein the cannabinoid is one or more of a natural phytocannabinoid, an organic cannabinoid, an endocannabinoid, a cannabinoid analog, a cannabinoid derivative, a synthetic cannabinoid and a cannabinoid receptor agonist. 11. The method of claim 10, wherein the skin disorder is one or more of eczema, psoriasis, dermatitis, itching dermatosis, rosacea, perioral dermatitis, acne, non-melanoma cancer or melanoma. 12. The method of claim 10, wherein the subject presents a symptom which is one or more of pruritus, dryness, skin rash, redness, swelling of the skin, itching, crusting, flaking, blistering, cracking, oozing, or bleeding of the skin. 13. The method of claim 11, wherein the dermatitis is atopic dermatitis, contact dermatitis, xerotic eczema, or seborrheic dermatitis. 14. The method of claim 10, wherein the organic cannabinoid is hemp oil or human breast milk. 15. The method of claim 10, wherein the cannabinoid is one or more of cannabigerolic acid (CBGA), cannabigerolic acid monomethylether (CBGAM), cannabigerol (CBG), cannabigerol monomethylether (CBGM), cannabigerovarinic acid (CBGVA), cannabigerovarin (CBGV), cannabichromenic acid (CBCA), cannabichromene (CBC), cannabichromevarinic acid (CBCVA), cannabichromevarin (CBCV), cannabidiolic acid (CBDA), cannabidiol (CBD), cannabidiol monomethylether (CBDM), cannabidiol-C4 (CBD-C4), cannabidivarinic acid (CBDVA), cannabidivarin (CBDV), cannabidiorcol (CBD-C1), delta-9-tetrahydrocannabinolic acid A (THCA-A), delta-9-tetrahydrocannabinolic acid B (THCA-B), delta-9-tetrahydrocannabinol (THC), delta-9-tetrahydrocannabinolic acid-C4 (THCA-C4), delta-9-tetrahydrocannabinol-C4 (THC-C4), delta-9-tetrahydrocannabivarinic acid (THCVA), delta-9-tetrahydrocannabivarin (THCV), delta-9-tetrahydrocannabiorcolic acid (THCA-C1), delta-9-tetrahydrocannabiorcol (THC-C1), delta-7-cis-iso-tetrahydrocannabivarin, delta-8-tetrahydrocannabinolic acid (Δ8-THCA), delta-8-tetrahydrocannabinol (Δ8-THC), cannabicyclolic acid (CBLA), cannabicyclol (CBL), cannabicyclovarin (CBLV), cannnabielsoic acid A (CBEA-A), cannabielsoic acid B (CBEA-B), cannabielsoin (CBE), cannabinolic acid (CBNA), cannabinol (CBN), cannabinol methylether (CBNM), cannabinol-C4 (CBN-C4), cannabivarin (CBV), cannabinol-C2 (CBN-C2), cannabiorcol (CBN-C1), cannabinodiol (CBND), cannabinodivarin (CBVD), cannabitriol (CBT), 10-ethoxy-9-hydroxy-delta-6a-tetrahydrocannabinol, 8,9-dihydroxy-delta-6a-tetrahydrocannabinol, cannabitriolvarin (CBTV), ethoxy-cannabitriolvarin (CBTVE), dehydrocannabifuran (DCBF), cannabifuran (CBF), cannabichromanon (CBCN), cannabicitran (CBT), 10-oxo-delta-6a-tetrahydrocannabinol (OTHC), delta-9-cis-tetrahydrocannabinol (cis-THC), 3,4,5,6-tetrahydro-7-hydroxy-alpha-alpha-2-trimethyl-9-n-propyl-2,6-methano-2H-1-benzoxocin-5-methanol (OH-iso-HHCV), cannabiripsol (CBR) and trihydroxy-delta-9-tetrahydrocannabinol (triOH-THC). 16. The method of claim 10, wherein the cannabinoid receptor agonist comprises one or more of a naphthoylindole, a naphthylmethylindole, a naphthoylpyrrole, a naphthylmethylindene, a phenylacetylindole and a cyclohexylphenol. 17. The method of claim 10, wherein the hydroxy acid is an alpha hydroxy acid, a beta hydroxy acid or a combination thereof. 18. The method of claim 17, wherein the alpha hydroxy acid is lactic acid, citric acid, glycolic acid, mandelic acid, benzylic acid, malic acid, tartaric acid, gluconolactone, galactonolactone, glucuronolactone, galacturonolactone, gulonolactone, ribonolactone, saccharic acid lactone, pantoyllactone, glucoheptonolactone, mannonolactone, or galactoheptonolactone. 19. The method of claim 17, wherein the beta hydroxy acid is salicylic acid or lipohydroxy acid. 20. The method of claim 10, wherein the composition further comprises a stabilizer selected from the group consisting of guar gum, xanthan gum cellulose hyaluronic acid, polyvinyl pyrrolidone (PVP), alginate, chondritin sulfate, poly gamma glutamic acid, gelatin, chitisin, corn starch and flour, in an amount from about 0.25% to about 30% (w/v). 21. The method of claim 10, wherein the carrier comprises hemp oil and wherein the composition further comprises one or more of a thickening agent, an antibiotic, an antiseptic agent, an antifungal, an antibacterial agent, an analgesic, an antiviral agent or a UV absorbing agent in an amount between 0.1 and 5% by weight of the composition. 22. The method of claim 10, wherein the subject is a human and wherein the composition is topically administered to the subject in an amount between about 100 nmol to about 1 μmol/cm2.
1,600
358
15,159,726
1,616
The present disclosure provides a method of adapting salmonidae fish for growth on soy protein-containing diets. The method comprises, within an effective period of time after the fish hatch, administering to the fish a fish feed composition. The fish feed composition comprises soy protein and an effective amount of an antioxidant. The antioxidant can be astaxanthin.
1. A method of adapting fish to be resistant to inflammatory enteritis induced by soy protein, the method comprising: a) providing to a fish population in water a feed composition containing at least about 1% (by weight) soy protein and an effective amount of an antioxidant after members of said fish population begin to feed by mouth; and b) continuing to provide said feed composition for a sufficient number of days after the fish begin to feed by mouth to thereby cause the fish to become resistant to inflammatory enteritis induced by soy protein. 2. The method of claim 1, wherein the antioxidant is astaxanthin. 3. The method of claim 2, wherein the feed further comprises animal by-product meal, nut-meal, and macrominerals, and wherein the fish population consists of salmonidae fish. 4. The method of claim 1, wherein the feed composition is provided to the fish for at least 100 days after the fish begin feeding by mouth. 5. The method of claim 4, wherein the feed composition is provided to the fish for 120 days after the fish begin feeding by mouth. 6. The method of claim 4, wherein the feed composition is provided to the fish for 190 days after the fish begin feeding by mouth. 7. The method of claim 4, wherein the feed composition is provided to the fish for 230 days after the fish begin feeding by mouth. 8. The method of claim 4, wherein the feed composition is provided to the fish for 365 days after the fish begin feeding by mouth. 9. The method of claim 1, wherein the feed composition is provided to the fish until the fish achieve a market size weight from about 1 to about 12 pounds. 10. The method of claim 1, wherein the feed composition is provided to the fish until the fish achieve a market size weight from about 1 to about 6 pounds. 11. The method of claim 1, wherein the feed composition is provided to the fish until the fish achieve a market size weight from about 1 to about 3 pounds. 12. The method of claim 2, wherein the astaxanthin is present in an amount from about 50 to about 2500 ppm of the feed composition. 13. The method of claim 2, wherein the astaxanthin is present in an amount from about 100 to about 1500 ppm of the feed composition. 14. The method of claim 2, wherein the astaxanthin is present in an amount from about 500 to about 1000 ppm of the feed composition. 15. The method of claim 1, wherein the soy protein is present in an amount from about 2% to about 80% (wt/wt) in the fish feed. 16. The method of claim 1, wherein the soy protein is present in an amount from about 10% to about 60% (wt/wt) in the fish feed. 17. The method of claim 1, wherein the soy protein is present in an amount from about 20% to about 50% (wt/wt) in the fish feed. 18. The method of claim 1, wherein the soy protein is present in an amount from about 25% to about 30% (wt/wt) in the fish feed. 19. The method of claim 2, wherein the astaxanthin is present in an amount from about 500 ppm to about 1000 ppm and the soy protein is present in an amount from about 25% to about 30% (wt/wt) in the fish feed. 20. A feed composition comprising at least about 20% (by weight) soy protein and an antioxidant, wherein said feed reduces the development of inflammatory enteritis induced by soy protein feeds in fish. 21. The feed composition of claim 20, wherein the antioxidant is astaxanthin. 22. The feed composition of claim 21, wherein the soy protein comprises a non-animal based protein concentrate, wherein the composition contains at least 55% protein content by weight, exopolysaccharides and contains oligosaccharides in an amount of between about 0.00 g/100 g to about 0.24 g/100 g on a dry matter basis. 23. The feed composition of claim 22, further comprising animal by-product meal, nut-meal, and macrominerals. 24. The feed composition of claim 23, wherein said feed comprises a composition containing up to 80% by weight of said non-animal based protein concentrate and up to 20% by weight of a mixture containing one or more compounds selected from the group consisting of lysine, methionine, lipids, biotin, choline, niacin, ascorbic acid, inositol, pantothenic acid, folic acid, pyridoxine, riboflavin, thiamin, vitamin A, vitamin B 12, vitamin D, vitamin E, vitamin K, calcium, phosphorus, potassium, sodium, magnesium, manganese, aluminum, iodine, cobalt, zinc, iron, selenium, and combinations thereof. 25. The feed composition of claim 21, wherein the protein is present in an amount of a least about 20% (by weight) and wherein astaxanthin is present in an amount of at least about 1 ppm.
The present disclosure provides a method of adapting salmonidae fish for growth on soy protein-containing diets. The method comprises, within an effective period of time after the fish hatch, administering to the fish a fish feed composition. The fish feed composition comprises soy protein and an effective amount of an antioxidant. The antioxidant can be astaxanthin.1. A method of adapting fish to be resistant to inflammatory enteritis induced by soy protein, the method comprising: a) providing to a fish population in water a feed composition containing at least about 1% (by weight) soy protein and an effective amount of an antioxidant after members of said fish population begin to feed by mouth; and b) continuing to provide said feed composition for a sufficient number of days after the fish begin to feed by mouth to thereby cause the fish to become resistant to inflammatory enteritis induced by soy protein. 2. The method of claim 1, wherein the antioxidant is astaxanthin. 3. The method of claim 2, wherein the feed further comprises animal by-product meal, nut-meal, and macrominerals, and wherein the fish population consists of salmonidae fish. 4. The method of claim 1, wherein the feed composition is provided to the fish for at least 100 days after the fish begin feeding by mouth. 5. The method of claim 4, wherein the feed composition is provided to the fish for 120 days after the fish begin feeding by mouth. 6. The method of claim 4, wherein the feed composition is provided to the fish for 190 days after the fish begin feeding by mouth. 7. The method of claim 4, wherein the feed composition is provided to the fish for 230 days after the fish begin feeding by mouth. 8. The method of claim 4, wherein the feed composition is provided to the fish for 365 days after the fish begin feeding by mouth. 9. The method of claim 1, wherein the feed composition is provided to the fish until the fish achieve a market size weight from about 1 to about 12 pounds. 10. The method of claim 1, wherein the feed composition is provided to the fish until the fish achieve a market size weight from about 1 to about 6 pounds. 11. The method of claim 1, wherein the feed composition is provided to the fish until the fish achieve a market size weight from about 1 to about 3 pounds. 12. The method of claim 2, wherein the astaxanthin is present in an amount from about 50 to about 2500 ppm of the feed composition. 13. The method of claim 2, wherein the astaxanthin is present in an amount from about 100 to about 1500 ppm of the feed composition. 14. The method of claim 2, wherein the astaxanthin is present in an amount from about 500 to about 1000 ppm of the feed composition. 15. The method of claim 1, wherein the soy protein is present in an amount from about 2% to about 80% (wt/wt) in the fish feed. 16. The method of claim 1, wherein the soy protein is present in an amount from about 10% to about 60% (wt/wt) in the fish feed. 17. The method of claim 1, wherein the soy protein is present in an amount from about 20% to about 50% (wt/wt) in the fish feed. 18. The method of claim 1, wherein the soy protein is present in an amount from about 25% to about 30% (wt/wt) in the fish feed. 19. The method of claim 2, wherein the astaxanthin is present in an amount from about 500 ppm to about 1000 ppm and the soy protein is present in an amount from about 25% to about 30% (wt/wt) in the fish feed. 20. A feed composition comprising at least about 20% (by weight) soy protein and an antioxidant, wherein said feed reduces the development of inflammatory enteritis induced by soy protein feeds in fish. 21. The feed composition of claim 20, wherein the antioxidant is astaxanthin. 22. The feed composition of claim 21, wherein the soy protein comprises a non-animal based protein concentrate, wherein the composition contains at least 55% protein content by weight, exopolysaccharides and contains oligosaccharides in an amount of between about 0.00 g/100 g to about 0.24 g/100 g on a dry matter basis. 23. The feed composition of claim 22, further comprising animal by-product meal, nut-meal, and macrominerals. 24. The feed composition of claim 23, wherein said feed comprises a composition containing up to 80% by weight of said non-animal based protein concentrate and up to 20% by weight of a mixture containing one or more compounds selected from the group consisting of lysine, methionine, lipids, biotin, choline, niacin, ascorbic acid, inositol, pantothenic acid, folic acid, pyridoxine, riboflavin, thiamin, vitamin A, vitamin B 12, vitamin D, vitamin E, vitamin K, calcium, phosphorus, potassium, sodium, magnesium, manganese, aluminum, iodine, cobalt, zinc, iron, selenium, and combinations thereof. 25. The feed composition of claim 21, wherein the protein is present in an amount of a least about 20% (by weight) and wherein astaxanthin is present in an amount of at least about 1 ppm.
1,600
359
15,430,815
1,652
The present invention comprises a method to diminish and/or eliminate atherosclerotic plaques, in mammals, through direct and indirect treatment of these plaques, in situ, using suitable substances which are capable of lipid removal, primarily through hydrolysis, either by a catalytic or stoichiometric process, wherein the substance targets receptors in and/or on the cell which lead to uptake into the lysosome. Such substances used to diminish and/or eliminate atherosclerotic plaques are generally comprised of lipid hydrolyzing proteins and/or polypeptides.
1. A pharmaceutical composition comprising a) human lysosomal acid lipase (hLAL) protein or peptide, or a protein or peptide having at least 85% sequence homology to human lysosomal acid lipase; and b) a pharmaceutically acceptable carrier; wherein the pharmaceutically acceptable carrier is an isotonic sterile injectable solution. 2. (canceled) 3. (canceled) 4. (canceled) 5. The composition of claim 1 wherein said lipid hydrolyzing protein or polypeptide is a protein having a Ser153 residue. 6. The composition of claim 1 wherein said lipid hydrolyzing protein or polypeptide is a polymorphic variant protein of lysosomal acid lipase with substitution of amino acid Pro(-6) to Thr and Gly2 to Arg. 7. The composition of claim 2 wherein the lysosomal acid lipase has fewer than six N-linked acetylglycosylation residues. 8. The composition of claim 2 wherein the lysosomal acid lipase has more than six N-linked acetylglycosylation residues. 9. The composition of claim 8 wherein the N-acetylglycosylation residue is oligosaccharide-terminated. 10. The composition of claim 9 wherein the oligosaccharide terminating residue is a mannose residue. 11. The composition of claim 10 wherein the N-acetylglycosylation residue is oligosaccharide-terminated. 12. The composition of claim 11 wherein the oligosaccharide terminating residue is a mannose residue. 13. (canceled) 14. (canceled) 15. A method for providing biologically active lysosomal acid lipase to mammalian cells, said method comprising administration into cells a vector comprising and expressing a DNA sequence encoding biologically active lysosomal acid lipase, and expressing the DNA sequence in said cells to produce biologically active lysosomal acid lipase capable of hydrolyzing lipids; wherein the expression level is in an amount sufficient to produce secretion of the biologically active lysosomal acid lipase from the cells in a therapeutic amount. 16. The method of claim 15 wherein the cells harboring the vector secrete the biologically active lysosomal acid lipase in an amount and form capable of being taken up by other cells deficient in lysosomal acid lipase. 17. The method of claim 15 wherein the cells are atheromatous plaque cells or cells of the liver. 18. The method of claim 17 wherein the vector is introduced to the cells ex vivo. 19. The method of claim 17 wherein the vector is introduced to the cells in vivo. 20. The method of claim 17, further comprising the administration of exogenously produced lysosomal acid lipase, contained in a pharmaceutically acceptable carrier. 21. The method of claim 15 wherein the cells harboring the vector secrete biologically active lysosomal acid lipase in an amount capable of reducing atherosclerotic plaque. 22. The method of claim 15 wherein the vector is a viral vector. 23. The method of claim 17 wherein the viral vector is selected from the group consisting of a lentivirus, adenovirus, adeno-associated virus and virus-like vectors. 24. The method of claim 15 wherein the vector is a plasmid. 25. The method of claim 17 wherein the biologically active lysosomal acid lipase is a polymorphic variant of lysosomal acid lipase with substitution of amino acid Pro(-6) to Thr and Gly2 to Arg. 26. A method for providing biologically active lysosomal acid lipase to cells of a mammal with atherosclerosis, comprising administration into the cells of said mammal an amount of a vector comprising and expressing a DNA sequence encoding lysosomal acid lipase and which is effective to transfect and sustain expression of biologically active lysosomal acid lipase in cells deficient therein. 27. The method of claim 26 wherein the expressed lysosomal acid lipase is secreted from the infected cells and is taken up by other cells deficient therein. 28. (canceled) 29. (canceled) 30. The pharmaceutical composition according to claim 1, wherein said protein or polypeptide comprises an oligosaccharide-terminated N-acetylglycosylated residue. 31. The pharmaceutical composition according to claim 1, wherein said protein or polypeptide is present in an amount sufficient to cause a decrease in a level of atherosclerotic plaques in a patient while minimizing undesired side effects. 32. The composition of claim 1, further comprising an anti-oxidant. 33. The composition of claim 1, further comprising a buffer. 34. The composition of claim 1, further comprising a bacteriostat. 35. The composition of claim 1, further comprising a preservative. 36. The composition of claim 1, further comprising a stabilizer. 37. The composition of claim 1, further comprising at least one of anti-oxidant, buffer, bacteriostat, preservative, or stabilizer. 38. The composition of claim 1, wherein the composition is stored in a unit-dose or a multi-dose container. 39. The composition of claim 38, wherein the container is sealed. 40. The composition of claim 38, wherein the container is a vial. 41. The composition of claim 1, wherein said human lysosomal acid lipase will target a receptor site selected from a mannose receptor, a mannose 6-phosphate receptor, a CD36 receptor, a LDL receptor, and combinations thereof for uptake into lysosomes, wherein said composition is for use in treating Wolman's Disease or Cholesteryl Ester Storage Disease in a mammal. 42. The composition of claim 38 or 39, wherein the container is an ampule. 43. A sealed vial containing a pharmaceutical composition in the form of a solution suitable for administration, said composition comprising: a. a protein or polypeptide comprising human lysosomal acid lipase or a protein or polypeptide having at least 85% sequence homology to human lysosomal acid lipase; and b. a pharmaceutically acceptable carrier, wherein the pharmaceutically acceptable carrier is an isotonic sterile injectable solution. 44. A sealed ampule containing a pharmaceutical composition in the form of a solution suitable for administration, said composition comprising: a. a protein or polypeptide comprising human lysosomal acid lipase or a protein or polypeptide having at least 85% sequence homology to human lysosomal acid lipase; and b. a pharmaceutically acceptable carrier, wherein the pharmaceutically acceptable carrier is an isotonic sterile injectable solution.
The present invention comprises a method to diminish and/or eliminate atherosclerotic plaques, in mammals, through direct and indirect treatment of these plaques, in situ, using suitable substances which are capable of lipid removal, primarily through hydrolysis, either by a catalytic or stoichiometric process, wherein the substance targets receptors in and/or on the cell which lead to uptake into the lysosome. Such substances used to diminish and/or eliminate atherosclerotic plaques are generally comprised of lipid hydrolyzing proteins and/or polypeptides.1. A pharmaceutical composition comprising a) human lysosomal acid lipase (hLAL) protein or peptide, or a protein or peptide having at least 85% sequence homology to human lysosomal acid lipase; and b) a pharmaceutically acceptable carrier; wherein the pharmaceutically acceptable carrier is an isotonic sterile injectable solution. 2. (canceled) 3. (canceled) 4. (canceled) 5. The composition of claim 1 wherein said lipid hydrolyzing protein or polypeptide is a protein having a Ser153 residue. 6. The composition of claim 1 wherein said lipid hydrolyzing protein or polypeptide is a polymorphic variant protein of lysosomal acid lipase with substitution of amino acid Pro(-6) to Thr and Gly2 to Arg. 7. The composition of claim 2 wherein the lysosomal acid lipase has fewer than six N-linked acetylglycosylation residues. 8. The composition of claim 2 wherein the lysosomal acid lipase has more than six N-linked acetylglycosylation residues. 9. The composition of claim 8 wherein the N-acetylglycosylation residue is oligosaccharide-terminated. 10. The composition of claim 9 wherein the oligosaccharide terminating residue is a mannose residue. 11. The composition of claim 10 wherein the N-acetylglycosylation residue is oligosaccharide-terminated. 12. The composition of claim 11 wherein the oligosaccharide terminating residue is a mannose residue. 13. (canceled) 14. (canceled) 15. A method for providing biologically active lysosomal acid lipase to mammalian cells, said method comprising administration into cells a vector comprising and expressing a DNA sequence encoding biologically active lysosomal acid lipase, and expressing the DNA sequence in said cells to produce biologically active lysosomal acid lipase capable of hydrolyzing lipids; wherein the expression level is in an amount sufficient to produce secretion of the biologically active lysosomal acid lipase from the cells in a therapeutic amount. 16. The method of claim 15 wherein the cells harboring the vector secrete the biologically active lysosomal acid lipase in an amount and form capable of being taken up by other cells deficient in lysosomal acid lipase. 17. The method of claim 15 wherein the cells are atheromatous plaque cells or cells of the liver. 18. The method of claim 17 wherein the vector is introduced to the cells ex vivo. 19. The method of claim 17 wherein the vector is introduced to the cells in vivo. 20. The method of claim 17, further comprising the administration of exogenously produced lysosomal acid lipase, contained in a pharmaceutically acceptable carrier. 21. The method of claim 15 wherein the cells harboring the vector secrete biologically active lysosomal acid lipase in an amount capable of reducing atherosclerotic plaque. 22. The method of claim 15 wherein the vector is a viral vector. 23. The method of claim 17 wherein the viral vector is selected from the group consisting of a lentivirus, adenovirus, adeno-associated virus and virus-like vectors. 24. The method of claim 15 wherein the vector is a plasmid. 25. The method of claim 17 wherein the biologically active lysosomal acid lipase is a polymorphic variant of lysosomal acid lipase with substitution of amino acid Pro(-6) to Thr and Gly2 to Arg. 26. A method for providing biologically active lysosomal acid lipase to cells of a mammal with atherosclerosis, comprising administration into the cells of said mammal an amount of a vector comprising and expressing a DNA sequence encoding lysosomal acid lipase and which is effective to transfect and sustain expression of biologically active lysosomal acid lipase in cells deficient therein. 27. The method of claim 26 wherein the expressed lysosomal acid lipase is secreted from the infected cells and is taken up by other cells deficient therein. 28. (canceled) 29. (canceled) 30. The pharmaceutical composition according to claim 1, wherein said protein or polypeptide comprises an oligosaccharide-terminated N-acetylglycosylated residue. 31. The pharmaceutical composition according to claim 1, wherein said protein or polypeptide is present in an amount sufficient to cause a decrease in a level of atherosclerotic plaques in a patient while minimizing undesired side effects. 32. The composition of claim 1, further comprising an anti-oxidant. 33. The composition of claim 1, further comprising a buffer. 34. The composition of claim 1, further comprising a bacteriostat. 35. The composition of claim 1, further comprising a preservative. 36. The composition of claim 1, further comprising a stabilizer. 37. The composition of claim 1, further comprising at least one of anti-oxidant, buffer, bacteriostat, preservative, or stabilizer. 38. The composition of claim 1, wherein the composition is stored in a unit-dose or a multi-dose container. 39. The composition of claim 38, wherein the container is sealed. 40. The composition of claim 38, wherein the container is a vial. 41. The composition of claim 1, wherein said human lysosomal acid lipase will target a receptor site selected from a mannose receptor, a mannose 6-phosphate receptor, a CD36 receptor, a LDL receptor, and combinations thereof for uptake into lysosomes, wherein said composition is for use in treating Wolman's Disease or Cholesteryl Ester Storage Disease in a mammal. 42. The composition of claim 38 or 39, wherein the container is an ampule. 43. A sealed vial containing a pharmaceutical composition in the form of a solution suitable for administration, said composition comprising: a. a protein or polypeptide comprising human lysosomal acid lipase or a protein or polypeptide having at least 85% sequence homology to human lysosomal acid lipase; and b. a pharmaceutically acceptable carrier, wherein the pharmaceutically acceptable carrier is an isotonic sterile injectable solution. 44. A sealed ampule containing a pharmaceutical composition in the form of a solution suitable for administration, said composition comprising: a. a protein or polypeptide comprising human lysosomal acid lipase or a protein or polypeptide having at least 85% sequence homology to human lysosomal acid lipase; and b. a pharmaceutically acceptable carrier, wherein the pharmaceutically acceptable carrier is an isotonic sterile injectable solution.
1,600
360
14,480,449
1,631
A method of characterising a sample from spectrometric data using calculation of spectral distance values is disclosed, for use in the field of mass spectrometry. Molecular formula assignment of peaks in mass spectral data is difficult and time-consuming, and the invention provides a computer implemented method of finding a most likely elemental composition of a measured spectral peak of interest. The method analyses isotopic peaks in a portion of the spectrum, using both their mass positions and intensities, to determine a spectral distance between those peaks and isotopic peaks of a candidate composition, finding peaks that match ( 140 ). A pattern spectral distance is determined ( 150 ) to provide a measure of the correspondence between a set of those peaks in the measured spectrum and peaks of each of a number of candidate compositions. The spectral fit is used to determine a most likely candidate composition.
1. A computer implemented method of characterizing a sample by identifying a most likely elemental composition of a measured peak of interest in a spectrum obtained from that sample, comprising the steps of: (a) identifying, for the peak of interest in the spectrum of the sample, a plurality of elemental compositions having a peak whose position lies within a predetermined tolerance of the position of the said measured peak of interest; (b) forming a set of candidate compositions from the identified plurality of elemental compositions; (c) generating a theoretical isotopic spectrum for each of the candidate compositions in the said formed set thereof; (d) selecting peaks within each theoretical isotopic spectrum thus generated, for further processing; (e) for each candidate composition of the set, in turn: (i) scaling the intensity of at least one of: all the selected peaks of the theoretical isotopic spectrum of the particular candidate composition under consideration, and/or all the peaks of the measured spectrum, to reduce the difference between the intensities of the peak of interest and the corresponding peak of the candidate composition; (ii) calculating a spectral distance, SD, between peaks in the theoretical isotopic spectrum and peaks in the measured spectrum based upon both the difference dM in the relative positions of a given peak in the theoretical isotopic spectrum and a given peak in the measured spectrum, and also the relative differences dI in scaled intensity thereof, wherein SD increases monotonically with dM and dI; (iii) either: for the peaks in the theoretical isotopic spectrum selected in step (d), identifying the peak in the measured spectrum which is considered most closely to correspond with a particular one of the peaks in the theoretical isotopic spectrum by determining the measured peak that has the lowest SD; or for the peaks in the measured spectrum, identifying the peak in the theoretical isotopic spectrum selected in step (d) which is considered most closely to correspond with a particular one of the peaks in the measured spectrum by determining the theoretical isotopic peak that has the lowest SD; (iv) calculating a pattern spectral distance (PSD) for a particular candidate composition, based upon a combination of both dM and dI between the measured peaks and the theoretical isotopic peaks, where the measured or theoretical isotopic peaks are those which have been identified in step (e) (iii), and wherein PSD increases monotonically with dM and dI; (v) normalizing the PSD for n, the number of selected peaks in the theoretical isotopic spectrum; (f) identifying which of the set of candidate elemental compositions of the measured peak of interest is the most likely elemental composition thereof, by determining which candidate composition has the lowest PSD; and (g) outputting data representative of the identified most likely elemental composition of the measured peak of interest; wherein steps (a)-(g) are performed by a computer executing computer software; wherein the measured spectrum is a mass spectrum generated by analyzing the sample in a mass spectrometer; and wherein the PSD is weighted according to the abundance of the peaks in either the theoretical isotopic mass spectrum or the measured mass spectrum such that a mass and/or intensity error of a less intense peak affects the PSD less than the same mass or intensity error of a more intense peak. 2. A method according to claim 1 wherein the weighted PSD is calculated according to one of: PSD=√[((dM 1 /M exp)2 *I 1f)+((dM 2 /M exp)2 *I 2f)+ . . . +((dI 1 /I exp)2 *I 1f)+((dI 2 /I exp)2 *I 2f)+ . . . ]  (A) PSD=√[((dM 1 /M exp)+((dM 2 /M exp)+ . . . +((dI 1 /I exp)2 *I 1f)+((dI 2 /I exp)2 *I 2f)+ . . . ]  (B) PSD=√[((dM 1 /M exp)2 *I 1f)+((dM 2 /M exp)2 *I 2f)+ . . . +((dI 1 /I exp)2)+((dI 2 /I exp)2)+ . . . ]  (C) and where: the intensity of the selected theoretical isotopic mass peak is Iip; and I1f, I2f etc. are fractional intensities. 3. A method according to claim 2, wherein the step of normalizing the PSD for n, the number of peaks in the theoretical isotopic spectrum, comprises multiplying the PSD by 1/√[2*I 1f+2*I 2f+ . . . ]  (a) 1/√[n+I 1f +I 2f+ . . . ]  (b) 1/√[n+I 1f +I 2f+ . . . ]  (c) for the cases (A), (B) and (C) of claim 2, respectively. 4. A computer implemented method of characterizing a sample by identifying a most likely elemental composition of a measured peak of interest in a spectrum obtained from that sample, comprising the steps of: (a) identifying, for the peak of interest in the spectrum of the sample, a plurality of elemental compositions having a peak whose position lies within a predetermined tolerance of the position of the said measured peak of interest; (b) forming a set of candidate compositions from the identified plurality of elemental compositions; (c) generating a theoretical isotopic spectrum for each of the candidate compositions in the said formed set thereof; (d) selecting peaks within each theoretical isotopic spectrum thus generated, for further processing; (e) for each candidate composition of the set, in turn: (i) scaling the intensity of at least one of: all the selected peaks of the theoretical isotopic spectrum of the particular candidate composition under consideration, and/or all the peaks of the measured spectrum, to reduce the difference between the intensities of the peak of interest and the corresponding peak of the candidate composition; (ii) calculating a spectral distance, SD, between peaks in the theoretical isotopic spectrum and peaks in the measured spectrum based upon both the difference dM in the relative positions of a given peak in the theoretical isotopic spectrum and a given peak in the measured spectrum, and also the relative differences dI in scaled intensity thereof, wherein SD increases monotonically with dM and dI; (iii) either: for the peaks in the theoretical isotopic spectrum selected in step (d), identifying the peak in the measured spectrum which is considered most closely to correspond with a particular one of the peaks in the theoretical isotopic spectrum by determining the measured peak that has the lowest SD; or for the peaks in the measured spectrum, identifying the peak in the theoretical isotopic spectrum selected in step (d) which is considered most closely to correspond with a particular one of the peaks in the measured spectrum by determining the theoretical isotopic peak that has the lowest SD; (iv) calculating a pattern spectral distance (PSD) for a particular candidate composition, based upon a combination of both dM and dI between the measured peaks and the theoretical isotopic peaks, where the measured or theoretical isotopic peaks are those which have been identified in step (e) (iii), and wherein PSD increases monotonically with dM and dI; (v) normalizing the PSD for n, the number of selected peaks in the theoretical isotopic spectrum; (f) identifying which of the set of candidate elemental compositions of the measured peak of interest is the most likely elemental composition thereof, by determining which candidate composition has the lowest PSD; and (g) outputting data representative of the identified most likely elemental composition of the measured peak of interest; wherein steps (a)-(g) are performed by a computer executing computer software; wherein the measured spectrum is a mass spectrum generated by analyzing the sample in a mass spectrometer; and wherein a criterion for selecting a theoretical isotopic peak comprises applying a threshold, such that only peaks of the theoretical isotopic spectrum having intensities greater than the threshold are selected for further processing. 5. A method according to claim 4, wherein the threshold is mathematically related to the noise in the measured spectrum. 6. A method according to claim 5, wherein the threshold in percent is set to 100*(4*Inoise/B), where B is the intensity of the identified peak of interest and the measured noise value of this peak is Inoise. 7. A method according to claim 4, wherein: SD is defined as any of: SD=√((dM/M exp)2+(dI/I exp)2)  (A) SD=|dM/M exp |+|dI/I exp|  (B) SD=√((dM/M exp)2)+|dI/I exp|  (C) SD=|dM/M exp|+√((dI/I exp)2)  (D) and wherein Mexp=expected positional error, Iexp=expected intensity error. 8. A method according to claim 4 wherein PSD is defined as any of: PSD=√(Σ(dM ip /M exp)2+Σ(dI ip /I exp)2)  (A) PSD=Σ|dM ip /M exp |+Σ|dI ip /I exp|  (B) PSD=√(Σ(dM ip /M exp)2)+Σ|dI ip /I exp|  (C) PSD=Σ|dM ip /M exp|+√(Σ(dI ip /I exp)2  (D) and wherein Σ is the sum over all selected theoretical isotopic peaks and their corresponding measured peaks, dMip is the positional difference between the identified corresponding measured peak and selected theoretical isotopic peak, and dIip is the intensity difference between the identified corresponding measured peak and selected theoretical isotopic peak. 9. A method according to claim 4 wherein between steps (e)(iii) and (e)(iv) the selected theoretical isotopic peaks and/or the peaks of the measured spectrum are scaled in intensity so as to minimize the sum of the difference in intensities, dI, between the intensity of each selected theoretical isotopic peak and the intensity of the corresponding measured peak. 10. A method according to claim 9 wherein the scaling is performed using a scaling factor N which is calculated and applied according to: I r =N*I 1 where: Ir is a scaled peak intensity, and I1 are the intensity of the peak in the spectrum to be scaled, either the selected theoretical isotopic peak intensity or the measured peak intensity; and where: I(1,abs) is the absolute intensity of the peak in the spectrum to be scaled, I(2,abs) is the absolute intensity of the corresponding peak in the other spectrum which is not to be scaled, and all summations are over all the peaks in the spectrum to be scaled; and in which the scaling factor is calculated as one of N=ΣI (2,abs) /ΣI (1,abs)  (a) N=Σ(I (2,abs) *I (1,abs))/Σ(I (1,abs) *I (1,abs))  (b) 11. A method according to claim 4 wherein the step of normalizing the PSD for n comprises multiplying the PSD by 1/√(2*n). 12. A computer implemented method of characterizing a sample by identifying a most likely elemental composition of a measured peak of interest in a spectrum obtained from that sample, comprising the steps of: (a) identifying, for the peak of interest in the spectrum of the sample, a plurality of elemental compositions having a peak whose position lies within a predetermined tolerance of the position of the said measured peak of interest; (b) forming a set of candidate compositions from the identified plurality of elemental compositions; (c) generating a theoretical isotopic spectrum for each of the candidate compositions in the said formed set thereof; (d) selecting peaks within each theoretical isotopic spectrum thus generated, for further processing; (e) for each candidate composition of the set, in turn: (i) scaling the intensity of at least one of: all the selected peaks of the theoretical isotopic spectrum of the particular candidate composition under consideration, and/or all the peaks of the measured spectrum, to reduce the difference between the intensities of the peak of interest and the corresponding peak of the candidate composition; (ii) calculating a spectral distance, SD, between peaks in the theoretical isotopic spectrum and peaks in the measured spectrum based upon both the difference dM in the relative positions of a given peak in the theoretical isotopic spectrum and a given peak in the measured spectrum, and also the relative differences dI in scaled intensity thereof, wherein SD increases monotonically with dM and dI; (iii) either: for the peaks in the theoretical isotopic spectrum selected in step (d), identifying the peak in the measured spectrum which is considered most closely to correspond with a particular one of the peaks in the theoretical isotopic spectrum by determining the measured peak that has the lowest SD; or for the peaks in the measured spectrum, identifying the peak in the theoretical isotopic spectrum selected in step (d) which is considered most closely to correspond with a particular one of the peaks in the measured spectrum by determining the theoretical isotopic peak that has the lowest SD; (iv) calculating a pattern spectral distance (PSD) for a particular candidate composition, based upon a combination of both dM and dI between the measured peaks and the theoretical isotopic peaks, where the measured or theoretical isotopic peaks are those which have been identified in step (e) (iii), and wherein PSD increases monotonically with dM and dI; (v) normalizing the PSD for n, the number of selected peaks in the theoretical isotopic spectrum; (f) identifying which of the set of candidate elemental compositions of the measured peak of interest is the most likely elemental composition thereof, by determining which candidate composition has the lowest PSD; and (g) outputting data representative of the identified most likely elemental composition of the measured peak of interest; wherein steps (a)-(g) are performed by a computer executing computer software; wherein the measured spectrum is a mass spectrum generated by analyzing the sample in a mass spectrometer; and wherein a peak is defined as missing if SD is larger than or equal to a given threshold, and wherein the calculation of the PSD involves applying a penalty for such missing peaks, the penalty further increasing the value of PSD. 13. A method according to claim 12 wherein the threshold is 1.0. 14. A method according to claim 12, wherein the penalty applied is calculated based on the signal to noise ratio, S/N, the peak would be expected to have, had it existed in the measured spectrum. 15. A method according to claim 12 wherein the penalty is dE, and dE is applied in one of the following ways: if dM ip >M exp the term dM ip /M exp is replaced with dE  (a) if dI ip >I exp the term dI ip /I exp is replaced with dE  (b) if both dM ip >M exp and dI ip >I exp both the term dM ip /M exp and dI ip /I exp are replaced with dE,  (c) and where dE is greater than or equal to 1.0. 16. A method according to claim 12, wherein: SD is defined as any of: SD=√((dM/M exp)2+(dI/I exp)2)  (A) SD=|dM/M exp |+|dI/I exp|  (B) SD=√((dM/M exp)2)+|dI/I exp|  (C) SD=|dM/M exp|+√((dI/I exp)2)  (D) and wherein Mexp=expected positional error, Iexp=expected intensity error. 17. A method according to claim 1 wherein PSD is defined as any of: PSD=√(Σ(dM ip /M exp)2+Σ(dI ip /I exp)2)  (A) PSD=Σ|dM ip /M exp |+Σ|dI ip /I exp|  (B) PSD=√(Σ(dM ip /M exp)2)+Σ|dI ip /I exp|  (C) PSD=Σ|dM ip /M exp|+√(Σ(dI ip /I exp)2  (D) and wherein Σ is the sum over all selected theoretical isotopic peaks and their corresponding measured peaks, dMip is the positional difference between the identified corresponding measured peak and selected theoretical isotopic peak, and dIip is the intensity difference between the identified corresponding measured peak and selected theoretical isotopic peak.
A method of characterising a sample from spectrometric data using calculation of spectral distance values is disclosed, for use in the field of mass spectrometry. Molecular formula assignment of peaks in mass spectral data is difficult and time-consuming, and the invention provides a computer implemented method of finding a most likely elemental composition of a measured spectral peak of interest. The method analyses isotopic peaks in a portion of the spectrum, using both their mass positions and intensities, to determine a spectral distance between those peaks and isotopic peaks of a candidate composition, finding peaks that match ( 140 ). A pattern spectral distance is determined ( 150 ) to provide a measure of the correspondence between a set of those peaks in the measured spectrum and peaks of each of a number of candidate compositions. The spectral fit is used to determine a most likely candidate composition.1. A computer implemented method of characterizing a sample by identifying a most likely elemental composition of a measured peak of interest in a spectrum obtained from that sample, comprising the steps of: (a) identifying, for the peak of interest in the spectrum of the sample, a plurality of elemental compositions having a peak whose position lies within a predetermined tolerance of the position of the said measured peak of interest; (b) forming a set of candidate compositions from the identified plurality of elemental compositions; (c) generating a theoretical isotopic spectrum for each of the candidate compositions in the said formed set thereof; (d) selecting peaks within each theoretical isotopic spectrum thus generated, for further processing; (e) for each candidate composition of the set, in turn: (i) scaling the intensity of at least one of: all the selected peaks of the theoretical isotopic spectrum of the particular candidate composition under consideration, and/or all the peaks of the measured spectrum, to reduce the difference between the intensities of the peak of interest and the corresponding peak of the candidate composition; (ii) calculating a spectral distance, SD, between peaks in the theoretical isotopic spectrum and peaks in the measured spectrum based upon both the difference dM in the relative positions of a given peak in the theoretical isotopic spectrum and a given peak in the measured spectrum, and also the relative differences dI in scaled intensity thereof, wherein SD increases monotonically with dM and dI; (iii) either: for the peaks in the theoretical isotopic spectrum selected in step (d), identifying the peak in the measured spectrum which is considered most closely to correspond with a particular one of the peaks in the theoretical isotopic spectrum by determining the measured peak that has the lowest SD; or for the peaks in the measured spectrum, identifying the peak in the theoretical isotopic spectrum selected in step (d) which is considered most closely to correspond with a particular one of the peaks in the measured spectrum by determining the theoretical isotopic peak that has the lowest SD; (iv) calculating a pattern spectral distance (PSD) for a particular candidate composition, based upon a combination of both dM and dI between the measured peaks and the theoretical isotopic peaks, where the measured or theoretical isotopic peaks are those which have been identified in step (e) (iii), and wherein PSD increases monotonically with dM and dI; (v) normalizing the PSD for n, the number of selected peaks in the theoretical isotopic spectrum; (f) identifying which of the set of candidate elemental compositions of the measured peak of interest is the most likely elemental composition thereof, by determining which candidate composition has the lowest PSD; and (g) outputting data representative of the identified most likely elemental composition of the measured peak of interest; wherein steps (a)-(g) are performed by a computer executing computer software; wherein the measured spectrum is a mass spectrum generated by analyzing the sample in a mass spectrometer; and wherein the PSD is weighted according to the abundance of the peaks in either the theoretical isotopic mass spectrum or the measured mass spectrum such that a mass and/or intensity error of a less intense peak affects the PSD less than the same mass or intensity error of a more intense peak. 2. A method according to claim 1 wherein the weighted PSD is calculated according to one of: PSD=√[((dM 1 /M exp)2 *I 1f)+((dM 2 /M exp)2 *I 2f)+ . . . +((dI 1 /I exp)2 *I 1f)+((dI 2 /I exp)2 *I 2f)+ . . . ]  (A) PSD=√[((dM 1 /M exp)+((dM 2 /M exp)+ . . . +((dI 1 /I exp)2 *I 1f)+((dI 2 /I exp)2 *I 2f)+ . . . ]  (B) PSD=√[((dM 1 /M exp)2 *I 1f)+((dM 2 /M exp)2 *I 2f)+ . . . +((dI 1 /I exp)2)+((dI 2 /I exp)2)+ . . . ]  (C) and where: the intensity of the selected theoretical isotopic mass peak is Iip; and I1f, I2f etc. are fractional intensities. 3. A method according to claim 2, wherein the step of normalizing the PSD for n, the number of peaks in the theoretical isotopic spectrum, comprises multiplying the PSD by 1/√[2*I 1f+2*I 2f+ . . . ]  (a) 1/√[n+I 1f +I 2f+ . . . ]  (b) 1/√[n+I 1f +I 2f+ . . . ]  (c) for the cases (A), (B) and (C) of claim 2, respectively. 4. A computer implemented method of characterizing a sample by identifying a most likely elemental composition of a measured peak of interest in a spectrum obtained from that sample, comprising the steps of: (a) identifying, for the peak of interest in the spectrum of the sample, a plurality of elemental compositions having a peak whose position lies within a predetermined tolerance of the position of the said measured peak of interest; (b) forming a set of candidate compositions from the identified plurality of elemental compositions; (c) generating a theoretical isotopic spectrum for each of the candidate compositions in the said formed set thereof; (d) selecting peaks within each theoretical isotopic spectrum thus generated, for further processing; (e) for each candidate composition of the set, in turn: (i) scaling the intensity of at least one of: all the selected peaks of the theoretical isotopic spectrum of the particular candidate composition under consideration, and/or all the peaks of the measured spectrum, to reduce the difference between the intensities of the peak of interest and the corresponding peak of the candidate composition; (ii) calculating a spectral distance, SD, between peaks in the theoretical isotopic spectrum and peaks in the measured spectrum based upon both the difference dM in the relative positions of a given peak in the theoretical isotopic spectrum and a given peak in the measured spectrum, and also the relative differences dI in scaled intensity thereof, wherein SD increases monotonically with dM and dI; (iii) either: for the peaks in the theoretical isotopic spectrum selected in step (d), identifying the peak in the measured spectrum which is considered most closely to correspond with a particular one of the peaks in the theoretical isotopic spectrum by determining the measured peak that has the lowest SD; or for the peaks in the measured spectrum, identifying the peak in the theoretical isotopic spectrum selected in step (d) which is considered most closely to correspond with a particular one of the peaks in the measured spectrum by determining the theoretical isotopic peak that has the lowest SD; (iv) calculating a pattern spectral distance (PSD) for a particular candidate composition, based upon a combination of both dM and dI between the measured peaks and the theoretical isotopic peaks, where the measured or theoretical isotopic peaks are those which have been identified in step (e) (iii), and wherein PSD increases monotonically with dM and dI; (v) normalizing the PSD for n, the number of selected peaks in the theoretical isotopic spectrum; (f) identifying which of the set of candidate elemental compositions of the measured peak of interest is the most likely elemental composition thereof, by determining which candidate composition has the lowest PSD; and (g) outputting data representative of the identified most likely elemental composition of the measured peak of interest; wherein steps (a)-(g) are performed by a computer executing computer software; wherein the measured spectrum is a mass spectrum generated by analyzing the sample in a mass spectrometer; and wherein a criterion for selecting a theoretical isotopic peak comprises applying a threshold, such that only peaks of the theoretical isotopic spectrum having intensities greater than the threshold are selected for further processing. 5. A method according to claim 4, wherein the threshold is mathematically related to the noise in the measured spectrum. 6. A method according to claim 5, wherein the threshold in percent is set to 100*(4*Inoise/B), where B is the intensity of the identified peak of interest and the measured noise value of this peak is Inoise. 7. A method according to claim 4, wherein: SD is defined as any of: SD=√((dM/M exp)2+(dI/I exp)2)  (A) SD=|dM/M exp |+|dI/I exp|  (B) SD=√((dM/M exp)2)+|dI/I exp|  (C) SD=|dM/M exp|+√((dI/I exp)2)  (D) and wherein Mexp=expected positional error, Iexp=expected intensity error. 8. A method according to claim 4 wherein PSD is defined as any of: PSD=√(Σ(dM ip /M exp)2+Σ(dI ip /I exp)2)  (A) PSD=Σ|dM ip /M exp |+Σ|dI ip /I exp|  (B) PSD=√(Σ(dM ip /M exp)2)+Σ|dI ip /I exp|  (C) PSD=Σ|dM ip /M exp|+√(Σ(dI ip /I exp)2  (D) and wherein Σ is the sum over all selected theoretical isotopic peaks and their corresponding measured peaks, dMip is the positional difference between the identified corresponding measured peak and selected theoretical isotopic peak, and dIip is the intensity difference between the identified corresponding measured peak and selected theoretical isotopic peak. 9. A method according to claim 4 wherein between steps (e)(iii) and (e)(iv) the selected theoretical isotopic peaks and/or the peaks of the measured spectrum are scaled in intensity so as to minimize the sum of the difference in intensities, dI, between the intensity of each selected theoretical isotopic peak and the intensity of the corresponding measured peak. 10. A method according to claim 9 wherein the scaling is performed using a scaling factor N which is calculated and applied according to: I r =N*I 1 where: Ir is a scaled peak intensity, and I1 are the intensity of the peak in the spectrum to be scaled, either the selected theoretical isotopic peak intensity or the measured peak intensity; and where: I(1,abs) is the absolute intensity of the peak in the spectrum to be scaled, I(2,abs) is the absolute intensity of the corresponding peak in the other spectrum which is not to be scaled, and all summations are over all the peaks in the spectrum to be scaled; and in which the scaling factor is calculated as one of N=ΣI (2,abs) /ΣI (1,abs)  (a) N=Σ(I (2,abs) *I (1,abs))/Σ(I (1,abs) *I (1,abs))  (b) 11. A method according to claim 4 wherein the step of normalizing the PSD for n comprises multiplying the PSD by 1/√(2*n). 12. A computer implemented method of characterizing a sample by identifying a most likely elemental composition of a measured peak of interest in a spectrum obtained from that sample, comprising the steps of: (a) identifying, for the peak of interest in the spectrum of the sample, a plurality of elemental compositions having a peak whose position lies within a predetermined tolerance of the position of the said measured peak of interest; (b) forming a set of candidate compositions from the identified plurality of elemental compositions; (c) generating a theoretical isotopic spectrum for each of the candidate compositions in the said formed set thereof; (d) selecting peaks within each theoretical isotopic spectrum thus generated, for further processing; (e) for each candidate composition of the set, in turn: (i) scaling the intensity of at least one of: all the selected peaks of the theoretical isotopic spectrum of the particular candidate composition under consideration, and/or all the peaks of the measured spectrum, to reduce the difference between the intensities of the peak of interest and the corresponding peak of the candidate composition; (ii) calculating a spectral distance, SD, between peaks in the theoretical isotopic spectrum and peaks in the measured spectrum based upon both the difference dM in the relative positions of a given peak in the theoretical isotopic spectrum and a given peak in the measured spectrum, and also the relative differences dI in scaled intensity thereof, wherein SD increases monotonically with dM and dI; (iii) either: for the peaks in the theoretical isotopic spectrum selected in step (d), identifying the peak in the measured spectrum which is considered most closely to correspond with a particular one of the peaks in the theoretical isotopic spectrum by determining the measured peak that has the lowest SD; or for the peaks in the measured spectrum, identifying the peak in the theoretical isotopic spectrum selected in step (d) which is considered most closely to correspond with a particular one of the peaks in the measured spectrum by determining the theoretical isotopic peak that has the lowest SD; (iv) calculating a pattern spectral distance (PSD) for a particular candidate composition, based upon a combination of both dM and dI between the measured peaks and the theoretical isotopic peaks, where the measured or theoretical isotopic peaks are those which have been identified in step (e) (iii), and wherein PSD increases monotonically with dM and dI; (v) normalizing the PSD for n, the number of selected peaks in the theoretical isotopic spectrum; (f) identifying which of the set of candidate elemental compositions of the measured peak of interest is the most likely elemental composition thereof, by determining which candidate composition has the lowest PSD; and (g) outputting data representative of the identified most likely elemental composition of the measured peak of interest; wherein steps (a)-(g) are performed by a computer executing computer software; wherein the measured spectrum is a mass spectrum generated by analyzing the sample in a mass spectrometer; and wherein a peak is defined as missing if SD is larger than or equal to a given threshold, and wherein the calculation of the PSD involves applying a penalty for such missing peaks, the penalty further increasing the value of PSD. 13. A method according to claim 12 wherein the threshold is 1.0. 14. A method according to claim 12, wherein the penalty applied is calculated based on the signal to noise ratio, S/N, the peak would be expected to have, had it existed in the measured spectrum. 15. A method according to claim 12 wherein the penalty is dE, and dE is applied in one of the following ways: if dM ip >M exp the term dM ip /M exp is replaced with dE  (a) if dI ip >I exp the term dI ip /I exp is replaced with dE  (b) if both dM ip >M exp and dI ip >I exp both the term dM ip /M exp and dI ip /I exp are replaced with dE,  (c) and where dE is greater than or equal to 1.0. 16. A method according to claim 12, wherein: SD is defined as any of: SD=√((dM/M exp)2+(dI/I exp)2)  (A) SD=|dM/M exp |+|dI/I exp|  (B) SD=√((dM/M exp)2)+|dI/I exp|  (C) SD=|dM/M exp|+√((dI/I exp)2)  (D) and wherein Mexp=expected positional error, Iexp=expected intensity error. 17. A method according to claim 1 wherein PSD is defined as any of: PSD=√(Σ(dM ip /M exp)2+Σ(dI ip /I exp)2)  (A) PSD=Σ|dM ip /M exp |+Σ|dI ip /I exp|  (B) PSD=√(Σ(dM ip /M exp)2)+Σ|dI ip /I exp|  (C) PSD=Σ|dM ip /M exp|+√(Σ(dI ip /I exp)2  (D) and wherein Σ is the sum over all selected theoretical isotopic peaks and their corresponding measured peaks, dMip is the positional difference between the identified corresponding measured peak and selected theoretical isotopic peak, and dIip is the intensity difference between the identified corresponding measured peak and selected theoretical isotopic peak.
1,600
361
14,776,323
1,613
Various adhesive compositions are described which may optionally comprise one or more active agents such as pharmaceutical agents. The incorporation of one or more absorbents in combination with one or more crystallization inhibitors improves adhesive characteristics of the compositions. Also described are related methods of improving adhesive characteristics of adhesive compositions with the use of a combination of absorbent and inhibitor. Also described are related methods of using the compositions and articles incorporating such compositions.
1. A method of enhancing the adhesive properties of an adhesive composition, wherein the adhesive composition comprises an adhesive component, an absorbent, and a vehicle, the method comprising: providing a crystallization inhibitor; incorporating the crystallization inhibitor in the adhesive composition, whereby the adhesive properties of the composition are enhanced. 2. The method of claim 1, wherein the absorbent is selected from the group consisting of insoluble swellable polymers, hydratable polymers, water soluble polymers, synthetic absorbents, super absorbent polymers, and combinations thereof. 3. The method of claim 2, wherein the absorbent is carboxymethyl cellulose. 4. The method of claim 3, wherein the carboxymethyl cellulose has a degree of substitution within a range from 0.2 to 1.5. 5. The method of any one of claims 3-4, wherein the carboxymethyl cellulose has a molecular weight in a range from 17,000 to 700,000. 6. The method of any one of claims 1-5 wherein the adhesive composition further comprises an active agent. 7. The method of claim 6, wherein the active agent is selected from the group consisting of analgesics, local anesthetics, anti-acne agents, anti-angina agents, antiarrhythmics, antibacterial, anti-convulsives, antidepressants, anti-rheumatics, sex hormones, anti-fungals, anti-hypertensives, anti-hypothyroid agents, anti-malarials, anti-migraine agents, anti-nausea agents, skin lighteners, dopamine receptor antagonists, muscle relaxants, sclerosing agents, vitamins and combinations thereof. 8. The method of claim 7, wherein the active agent includes an anti-rheumatic. 9. The method of claim 8, wherein the anti-rheumatic is ibuprofen. 10. The method of any one of claims 1-9, wherein the crystallization inhibitor is selected from the group consisting of polyvinylpyrrolidone, polyacrylamides, polyvinyl alcohols, polyacrylic acids, caseins, gelatins, polyamines/polyethyleneimines, polyethylene glycols, cellulose, cellulose derivatives, methylcellulose, hydroxypropyl cellulose, ethyl cellulose, carboxymethylcellulose, non-urethane associated thickeners, quaternary ammonium alginates, xanthan, pectin, guar gum, guar gum derivatives, carrageenan, carboxypolymethylene, agar, polyethoxylated sorbitols, butyl methacrylate, butyl methacrylate derivatives, 2-dimethylaminoethyl methacrylate, methyl methacrylate, polyaminoamides, polyaminoimidazolines, polyetherurethaneamines, polyethylene oxide, polyacrylic acid, silica, silicon dioxide, starch, starch derivatives, dextrin, cyclodextrins, dextran, rosin esters, sterols, bile acids, polyglucosamines, monoacylglycerols, glycerol monooleate, glycerol monolinoleate, glycerol monopalmitate, glycerol monostearate, glycerol monolaurate, glycerol monocaprylate, glycerol monocaprate, and combinations or mixtures thereof. 11. The method of claim 10, wherein the crystallization inhibitor includes polyvinylpyrrolidone. 12. The method of any one of claims 1-11, wherein the vehicle comprises a polyhydric alcohol. 13. The method of claim 12, wherein the polyhydric alcohol is selected from the group consisting of propylene glycol, glycerol, polyethylene glycol, and combinations thereof. 14. The method of claim 13, wherein the vehicle includes propylene glycol. 15. The method of any one of claims 1-14, wherein a weight ratio of the crystallization inhibitor to the vehicle is from about 0.1% to about 300%. 16. The method of any one of claims 1-15, wherein the adhesive component is a hot melt adhesive. 17. The method of any one of claims 1-16, wherein the incorporating includes bringing the adhesive composition to a temperature between about 60° C. and about 70° C. to soften the adhesive. 18. The method of any one of claims 1-17 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 0.5 N/in. 19. The method of claim 18 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 1.5 N/in. 20. The method of claim 19 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 2.5 N/in. 21. The method of any one of claims 1-20 wherein the adhesive composition exhibits a Tack value of at least 10 N/in. 22. The method of claim 21 wherein the adhesive composition exhibits a Tack value of at least 25 N/in. 23. The method of claim 22 wherein the adhesive composition exhibits a Tack value of at least 40 N/in. 24. The method of any one of claims 1-23 wherein the adhesive composition exhibits a static absorption of at least about 50 g/m2/24 hours. 25. The method of any one of claims 1-24 wherein the adhesive composition exhibits a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 26. The method of any one of claims 1-25 wherein the adhesive composition exhibits a static absorption of at least 500 g/m2/24 hours and a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 27. An adhesive composition comprising: an adhesive component; a vehicle; an absorbent; a crystallization inhibitor; wherein the adhesive composition exhibits a Peel on polyethylene value of at least 0.5 N/in and a Tack of at least 10 N/in. 28. The adhesive composition of claim 27 wherein the vehicle is a polyhydric alcohol. 29. The adhesive composition of any one of claims 27-28 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 1.5 N/in. 30. The adhesive composition of claim 29 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 2.5 N/in. 31. The adhesive composition of any one of claims 27-30 wherein the adhesive composition exhibits a Tack value of at least 25 N/in. 32. The adhesive composition of claim 31 wherein the adhesive composition exhibits a Tack value of at least 40 N/in. 33. The adhesive composition of any one of claims 27-32 further comprising: at least one active agent. 34. The adhesive composition of any one of claims 27-33, wherein the absorbent is selected from the group consisting of insoluble swellable polymers, hydratable polymers, water soluble polymers, synthetic absorbents, super absorbent polymers, and combinations thereof. 35. The adhesive composition of claim 34, wherein the absorbent is carboxymethyl cellulose. 36. The adhesive composition of claim 35, wherein the carboxymethyl cellulose has a degree of substitution within a range from 0.2 to 1.5. 37. The adhesive composition of any one of claims 35-36, wherein the carboxymethyl cellulose has a molecular weight in a range from 17,000 to 700,000. 38. The adhesive composition of any one of claims 33-37, wherein the active agent is selected from the group consisting of analgesics, local anesthetics, anti-acne agents, anti-angina agents, antiarrhythmics, antibacterial, anti-convulsives, antidepressants, anti-rheumatics, sex hormones, anti-fungals, anti-hypertensives, anti-hypothyroid agents, anti-malarials, anti-migraine agents, anti-nausea agents, skin lighteners, dopamine receptor antagonists, muscle relaxants, sclerosing agents, vitamins and combinations thereof. 39. The adhesive composition of claim 38, wherein the active agent includes an anti-rheumatic. 40. The adhesive composition of claim 39, wherein the anti-rheumatic is ibuprofen. 41. The adhesive composition of any one of claims 27-40, wherein the crystallization inhibitor is selected from the group consisting of polyvinylpyrrolidone, polyacrylamides, polyvinyl alcohols, polyacrylic acids, caseins, gelatins, polyamines/polyethyleneimines, polyethylene glycols, cellulose, cellulose derivatives, methylcellulose, hydroxypropyl cellulose, ethyl cellulose, carboxymethylcellulose, non-urethane associated thickeners, quaternary ammonium alginates, xanthan, pectin, guar gum, guar gum derivatives, carrageenan, carboxypolymethylene, agar, polyethoxylated sorbitols, butyl methacrylate, butyl methacrylate derivatives, 2-dimethylaminoethyl methacrylate, methyl methacrylate, polyaminoamides, polyaminoimidazolines, polyetherurethaneamines, polyethylene oxide, polyacrylic acid, silica, silicon dioxide, starch, starch derivatives, dextrin, cyclodextrins, dextran, rosin esters, sterols, bile acids, polyglucosamines, monoacylglycerols, glycerol monooleate, glycerol monolinoleate, glycerol monopalmitate, glycerol monostearate, glycerol monolaurate, glycerol monocaprylate, glycerol monocaprate, and combinations or mixtures thereof. 42. The adhesive composition of claim 41, wherein the crystallization inhibitor includes polyvinylpyrrolidone. 43. The adhesive composition of any one of claims 28-42, wherein the polyhydric alcohol is selected from the group consisting of propylene glycol, glycerol, polyethylene glycol, and combinations thereof. 44. The adhesive composition of any one of claims 27-43 wherein the adhesive composition exhibits a static absorption of at least about 50 g/m2/24 hours. 45. The adhesive composition of any one of claims 27-44 wherein the adhesive composition exhibits a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 46. The adhesive composition of any one of claims 27-45 wherein the adhesive composition exhibits a static absorption of at least 500 g/m2/24 hours and a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 47. An article for adhesive attachment to a surface of interest, the article comprising: a substrate defining at least one face; and a region of an adhesive composition disposed on at least a portion of the face of the substrate, the adhesive composition including (i) an adhesive component, (ii) a vehicle, (iii) an absorbent, and (iv) a crystallization inhibitor, wherein the adhesive composition exhibits a Peel on polyethylene value of at least 0.5 N/in and a Tack of at least 10 N/in. 48. The article of claim 47 wherein the article is a medical article. 49. The article of claim 48 wherein the medical article is selected from the group consisting of wound dressings, surgical dressings, medical tapes, athletic tapes, surgical tapes, sensors, electrodes, ostomy appliances, sealing rings, catheters, connector fittings, catheter hubs, catheter adapters, fluid delivery tubes, electrical wires and cables, negative pressure wound therapy (NPWT) components, surgical drains, wound draining components, IV site dressings, prostheses, stoma pouches, buccal patches, transdermal patches, dentures, hairpieces, bandages, diapers, medical padding, liposuction padding, hygiene pads, corn and callous pads, blister cushioning and protection pads, toe cushioning pads, and pads for protecting and cushioning tube sites such as tracheotomy tubes. 50. The article of any one of claims 47-49 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 1.5 N/in. 51. The article of claim 50 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 2.5 N/in. 52. The article of any one of claims 47-51 wherein the adhesive composition exhibits a Tack value of at least 25 N/in. 53. The article of claim 52 wherein the adhesive composition exhibits a Tack value of at least 40 N/in. 54. The article of any one of claims 47-53 wherein the vehicle is a polyhydric alcohol. 55. The article of any claim 54, wherein the polyhydric alcohol is selected from the group consisting of propylene glycol, glycerol, polyethylene glycol, and combinations thereof. 56. The article of claim 55 wherein the polyhydric alcohol is propylene glycol. 57. The article of any one of claims 47-56 wherein the adhesive composition further comprises: at least one active agent. 58. The article of any one of claims 47-57, wherein the absorbent is selected from the group consisting of insoluble swellable polymers, hydratable polymers, water soluble polymers, synthetic absorbents, super absorbent polymers, and combinations thereof. 59. The article of claim 58, wherein the absorbent is carboxymethyl cellulose. 60. The article of claim 59, wherein the carboxymethyl cellulose has a degree of substitution within a range from 0.2 to 1.5. 61. The article of any one of claims 59-60, wherein the carboxymethyl cellulose has a molecular weight in a range from 17,000 to 700,000. 62. The article of any one of claims 57-61, wherein the active agent is selected from the group consisting of analgesics, local anesthetics, anti-acne agents, anti-angina agents, antiarrhythmics, antibacterial, anti-convulsives, antidepressants, anti-rheumatics, sex hormones, anti-fungals, anti-hypertensives, anti-hypothyroid agents, anti-malarials, anti-migraine agents, anti-nausea agents, skin lighteners, dopamine receptor antagonists, muscle relaxants, sclerosing agents, vitamins and combinations thereof. 63. The article of claim 62, wherein the active agent includes an anti-rheumatic. 64. The article of claim 63, wherein the anti-rheumatic is ibuprofen. 65. The article of any one of claims 47-64, wherein the crystallization inhibitor is selected from the group consisting of polyvinylpyrrolidone, polyacrylamides, polyvinyl alcohols, polyacrylic acids, caseins, gelatins, polyamines/polyethyleneimines, polyethylene glycols, cellulose, cellulose derivatives, methylcellulose, hydroxypropyl cellulose, ethyl cellulose, carboxymethylcellulose, non-urethane associated thickeners, quaternary ammonium alginates, xanthan, pectin, guar gum, guar gum derivatives, carrageenan, carboxypolymethylene, agar, polyethoxylated sorbitols, butyl methacrylate, butyl methacrylate derivatives, 2-dimethylaminoethyl methacrylate, methyl methacrylate, polyaminoamides, polyaminoimidazolines, polyetherurethaneamines, polyethylene oxide, polyacrylic acid, silica, silicon dioxide, starch, starch derivatives, dextrin, cyclodextrins, dextran, rosin esters, sterols, bile acids, polyglucosamines, monoacylglycerols, glycerol monooleate, glycerol monolinoleate, glycerol monopalmitate, glycerol monostearate, glycerol monolaurate, glycerol monocaprylate, glycerol monocaprate, and combinations or mixtures thereof. 66. The article of claim 65, wherein the crystallization inhibitor includes polyvinylpyrrolidone. 67. The article of any one of claims 47-66 wherein the adhesive composition exhibits a static absorption of at least about 50 g/m2/24 hours. 68. The article of any one of claims 47-67 wherein the adhesive composition exhibits a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 69. The article of any one of claims 47-68 wherein the adhesive composition exhibits a static absorption of at least 500 g/m2/24 hours and a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours.
Various adhesive compositions are described which may optionally comprise one or more active agents such as pharmaceutical agents. The incorporation of one or more absorbents in combination with one or more crystallization inhibitors improves adhesive characteristics of the compositions. Also described are related methods of improving adhesive characteristics of adhesive compositions with the use of a combination of absorbent and inhibitor. Also described are related methods of using the compositions and articles incorporating such compositions.1. A method of enhancing the adhesive properties of an adhesive composition, wherein the adhesive composition comprises an adhesive component, an absorbent, and a vehicle, the method comprising: providing a crystallization inhibitor; incorporating the crystallization inhibitor in the adhesive composition, whereby the adhesive properties of the composition are enhanced. 2. The method of claim 1, wherein the absorbent is selected from the group consisting of insoluble swellable polymers, hydratable polymers, water soluble polymers, synthetic absorbents, super absorbent polymers, and combinations thereof. 3. The method of claim 2, wherein the absorbent is carboxymethyl cellulose. 4. The method of claim 3, wherein the carboxymethyl cellulose has a degree of substitution within a range from 0.2 to 1.5. 5. The method of any one of claims 3-4, wherein the carboxymethyl cellulose has a molecular weight in a range from 17,000 to 700,000. 6. The method of any one of claims 1-5 wherein the adhesive composition further comprises an active agent. 7. The method of claim 6, wherein the active agent is selected from the group consisting of analgesics, local anesthetics, anti-acne agents, anti-angina agents, antiarrhythmics, antibacterial, anti-convulsives, antidepressants, anti-rheumatics, sex hormones, anti-fungals, anti-hypertensives, anti-hypothyroid agents, anti-malarials, anti-migraine agents, anti-nausea agents, skin lighteners, dopamine receptor antagonists, muscle relaxants, sclerosing agents, vitamins and combinations thereof. 8. The method of claim 7, wherein the active agent includes an anti-rheumatic. 9. The method of claim 8, wherein the anti-rheumatic is ibuprofen. 10. The method of any one of claims 1-9, wherein the crystallization inhibitor is selected from the group consisting of polyvinylpyrrolidone, polyacrylamides, polyvinyl alcohols, polyacrylic acids, caseins, gelatins, polyamines/polyethyleneimines, polyethylene glycols, cellulose, cellulose derivatives, methylcellulose, hydroxypropyl cellulose, ethyl cellulose, carboxymethylcellulose, non-urethane associated thickeners, quaternary ammonium alginates, xanthan, pectin, guar gum, guar gum derivatives, carrageenan, carboxypolymethylene, agar, polyethoxylated sorbitols, butyl methacrylate, butyl methacrylate derivatives, 2-dimethylaminoethyl methacrylate, methyl methacrylate, polyaminoamides, polyaminoimidazolines, polyetherurethaneamines, polyethylene oxide, polyacrylic acid, silica, silicon dioxide, starch, starch derivatives, dextrin, cyclodextrins, dextran, rosin esters, sterols, bile acids, polyglucosamines, monoacylglycerols, glycerol monooleate, glycerol monolinoleate, glycerol monopalmitate, glycerol monostearate, glycerol monolaurate, glycerol monocaprylate, glycerol monocaprate, and combinations or mixtures thereof. 11. The method of claim 10, wherein the crystallization inhibitor includes polyvinylpyrrolidone. 12. The method of any one of claims 1-11, wherein the vehicle comprises a polyhydric alcohol. 13. The method of claim 12, wherein the polyhydric alcohol is selected from the group consisting of propylene glycol, glycerol, polyethylene glycol, and combinations thereof. 14. The method of claim 13, wherein the vehicle includes propylene glycol. 15. The method of any one of claims 1-14, wherein a weight ratio of the crystallization inhibitor to the vehicle is from about 0.1% to about 300%. 16. The method of any one of claims 1-15, wherein the adhesive component is a hot melt adhesive. 17. The method of any one of claims 1-16, wherein the incorporating includes bringing the adhesive composition to a temperature between about 60° C. and about 70° C. to soften the adhesive. 18. The method of any one of claims 1-17 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 0.5 N/in. 19. The method of claim 18 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 1.5 N/in. 20. The method of claim 19 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 2.5 N/in. 21. The method of any one of claims 1-20 wherein the adhesive composition exhibits a Tack value of at least 10 N/in. 22. The method of claim 21 wherein the adhesive composition exhibits a Tack value of at least 25 N/in. 23. The method of claim 22 wherein the adhesive composition exhibits a Tack value of at least 40 N/in. 24. The method of any one of claims 1-23 wherein the adhesive composition exhibits a static absorption of at least about 50 g/m2/24 hours. 25. The method of any one of claims 1-24 wherein the adhesive composition exhibits a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 26. The method of any one of claims 1-25 wherein the adhesive composition exhibits a static absorption of at least 500 g/m2/24 hours and a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 27. An adhesive composition comprising: an adhesive component; a vehicle; an absorbent; a crystallization inhibitor; wherein the adhesive composition exhibits a Peel on polyethylene value of at least 0.5 N/in and a Tack of at least 10 N/in. 28. The adhesive composition of claim 27 wherein the vehicle is a polyhydric alcohol. 29. The adhesive composition of any one of claims 27-28 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 1.5 N/in. 30. The adhesive composition of claim 29 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 2.5 N/in. 31. The adhesive composition of any one of claims 27-30 wherein the adhesive composition exhibits a Tack value of at least 25 N/in. 32. The adhesive composition of claim 31 wherein the adhesive composition exhibits a Tack value of at least 40 N/in. 33. The adhesive composition of any one of claims 27-32 further comprising: at least one active agent. 34. The adhesive composition of any one of claims 27-33, wherein the absorbent is selected from the group consisting of insoluble swellable polymers, hydratable polymers, water soluble polymers, synthetic absorbents, super absorbent polymers, and combinations thereof. 35. The adhesive composition of claim 34, wherein the absorbent is carboxymethyl cellulose. 36. The adhesive composition of claim 35, wherein the carboxymethyl cellulose has a degree of substitution within a range from 0.2 to 1.5. 37. The adhesive composition of any one of claims 35-36, wherein the carboxymethyl cellulose has a molecular weight in a range from 17,000 to 700,000. 38. The adhesive composition of any one of claims 33-37, wherein the active agent is selected from the group consisting of analgesics, local anesthetics, anti-acne agents, anti-angina agents, antiarrhythmics, antibacterial, anti-convulsives, antidepressants, anti-rheumatics, sex hormones, anti-fungals, anti-hypertensives, anti-hypothyroid agents, anti-malarials, anti-migraine agents, anti-nausea agents, skin lighteners, dopamine receptor antagonists, muscle relaxants, sclerosing agents, vitamins and combinations thereof. 39. The adhesive composition of claim 38, wherein the active agent includes an anti-rheumatic. 40. The adhesive composition of claim 39, wherein the anti-rheumatic is ibuprofen. 41. The adhesive composition of any one of claims 27-40, wherein the crystallization inhibitor is selected from the group consisting of polyvinylpyrrolidone, polyacrylamides, polyvinyl alcohols, polyacrylic acids, caseins, gelatins, polyamines/polyethyleneimines, polyethylene glycols, cellulose, cellulose derivatives, methylcellulose, hydroxypropyl cellulose, ethyl cellulose, carboxymethylcellulose, non-urethane associated thickeners, quaternary ammonium alginates, xanthan, pectin, guar gum, guar gum derivatives, carrageenan, carboxypolymethylene, agar, polyethoxylated sorbitols, butyl methacrylate, butyl methacrylate derivatives, 2-dimethylaminoethyl methacrylate, methyl methacrylate, polyaminoamides, polyaminoimidazolines, polyetherurethaneamines, polyethylene oxide, polyacrylic acid, silica, silicon dioxide, starch, starch derivatives, dextrin, cyclodextrins, dextran, rosin esters, sterols, bile acids, polyglucosamines, monoacylglycerols, glycerol monooleate, glycerol monolinoleate, glycerol monopalmitate, glycerol monostearate, glycerol monolaurate, glycerol monocaprylate, glycerol monocaprate, and combinations or mixtures thereof. 42. The adhesive composition of claim 41, wherein the crystallization inhibitor includes polyvinylpyrrolidone. 43. The adhesive composition of any one of claims 28-42, wherein the polyhydric alcohol is selected from the group consisting of propylene glycol, glycerol, polyethylene glycol, and combinations thereof. 44. The adhesive composition of any one of claims 27-43 wherein the adhesive composition exhibits a static absorption of at least about 50 g/m2/24 hours. 45. The adhesive composition of any one of claims 27-44 wherein the adhesive composition exhibits a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 46. The adhesive composition of any one of claims 27-45 wherein the adhesive composition exhibits a static absorption of at least 500 g/m2/24 hours and a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 47. An article for adhesive attachment to a surface of interest, the article comprising: a substrate defining at least one face; and a region of an adhesive composition disposed on at least a portion of the face of the substrate, the adhesive composition including (i) an adhesive component, (ii) a vehicle, (iii) an absorbent, and (iv) a crystallization inhibitor, wherein the adhesive composition exhibits a Peel on polyethylene value of at least 0.5 N/in and a Tack of at least 10 N/in. 48. The article of claim 47 wherein the article is a medical article. 49. The article of claim 48 wherein the medical article is selected from the group consisting of wound dressings, surgical dressings, medical tapes, athletic tapes, surgical tapes, sensors, electrodes, ostomy appliances, sealing rings, catheters, connector fittings, catheter hubs, catheter adapters, fluid delivery tubes, electrical wires and cables, negative pressure wound therapy (NPWT) components, surgical drains, wound draining components, IV site dressings, prostheses, stoma pouches, buccal patches, transdermal patches, dentures, hairpieces, bandages, diapers, medical padding, liposuction padding, hygiene pads, corn and callous pads, blister cushioning and protection pads, toe cushioning pads, and pads for protecting and cushioning tube sites such as tracheotomy tubes. 50. The article of any one of claims 47-49 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 1.5 N/in. 51. The article of claim 50 wherein the adhesive composition exhibits a Peel on polyethylene value of at least 2.5 N/in. 52. The article of any one of claims 47-51 wherein the adhesive composition exhibits a Tack value of at least 25 N/in. 53. The article of claim 52 wherein the adhesive composition exhibits a Tack value of at least 40 N/in. 54. The article of any one of claims 47-53 wherein the vehicle is a polyhydric alcohol. 55. The article of any claim 54, wherein the polyhydric alcohol is selected from the group consisting of propylene glycol, glycerol, polyethylene glycol, and combinations thereof. 56. The article of claim 55 wherein the polyhydric alcohol is propylene glycol. 57. The article of any one of claims 47-56 wherein the adhesive composition further comprises: at least one active agent. 58. The article of any one of claims 47-57, wherein the absorbent is selected from the group consisting of insoluble swellable polymers, hydratable polymers, water soluble polymers, synthetic absorbents, super absorbent polymers, and combinations thereof. 59. The article of claim 58, wherein the absorbent is carboxymethyl cellulose. 60. The article of claim 59, wherein the carboxymethyl cellulose has a degree of substitution within a range from 0.2 to 1.5. 61. The article of any one of claims 59-60, wherein the carboxymethyl cellulose has a molecular weight in a range from 17,000 to 700,000. 62. The article of any one of claims 57-61, wherein the active agent is selected from the group consisting of analgesics, local anesthetics, anti-acne agents, anti-angina agents, antiarrhythmics, antibacterial, anti-convulsives, antidepressants, anti-rheumatics, sex hormones, anti-fungals, anti-hypertensives, anti-hypothyroid agents, anti-malarials, anti-migraine agents, anti-nausea agents, skin lighteners, dopamine receptor antagonists, muscle relaxants, sclerosing agents, vitamins and combinations thereof. 63. The article of claim 62, wherein the active agent includes an anti-rheumatic. 64. The article of claim 63, wherein the anti-rheumatic is ibuprofen. 65. The article of any one of claims 47-64, wherein the crystallization inhibitor is selected from the group consisting of polyvinylpyrrolidone, polyacrylamides, polyvinyl alcohols, polyacrylic acids, caseins, gelatins, polyamines/polyethyleneimines, polyethylene glycols, cellulose, cellulose derivatives, methylcellulose, hydroxypropyl cellulose, ethyl cellulose, carboxymethylcellulose, non-urethane associated thickeners, quaternary ammonium alginates, xanthan, pectin, guar gum, guar gum derivatives, carrageenan, carboxypolymethylene, agar, polyethoxylated sorbitols, butyl methacrylate, butyl methacrylate derivatives, 2-dimethylaminoethyl methacrylate, methyl methacrylate, polyaminoamides, polyaminoimidazolines, polyetherurethaneamines, polyethylene oxide, polyacrylic acid, silica, silicon dioxide, starch, starch derivatives, dextrin, cyclodextrins, dextran, rosin esters, sterols, bile acids, polyglucosamines, monoacylglycerols, glycerol monooleate, glycerol monolinoleate, glycerol monopalmitate, glycerol monostearate, glycerol monolaurate, glycerol monocaprylate, glycerol monocaprate, and combinations or mixtures thereof. 66. The article of claim 65, wherein the crystallization inhibitor includes polyvinylpyrrolidone. 67. The article of any one of claims 47-66 wherein the adhesive composition exhibits a static absorption of at least about 50 g/m2/24 hours. 68. The article of any one of claims 47-67 wherein the adhesive composition exhibits a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours. 69. The article of any one of claims 47-68 wherein the adhesive composition exhibits a static absorption of at least 500 g/m2/24 hours and a moisture vapor transmission rate (MVTR) of at least 25 g/m2/24 hours.
1,600
362
15,533,710
1,612
Provided herein is an oral care composition comprising taurine or a salt thereof in an amount of from 0.001 weight % to 0.008 weight % by total weight of the composition, and an orally acceptable carrier. The composition is effective in accelerating HSP 27 production in damaged tissues of the oral cavity and enhances repair of the damaged tissue.
1. An oral care composition comprising taurine or a salt thereof in an amount of from 0.001 weight % to 0.05 weight %, e.g., 0.001 weight % to 0.008 weight % by total weight of the composition, and an orally acceptable carrier. 2. The composition of claim 1, wherein the taurine or salt thereof is present in the composition in an amount of from 0.003 weight % to 0.006 weight % by total weight of the composition. 3. The composition of claim 1 wherein the composition is selected from mouthwashes, sprays, dentifrices, oral strips, chewing gums and lozenges. 4. The composition of claim 1, wherein the oral care composition further comprises one or more agents selected from: surfactants, desensitizing agents, whitening agents, tartar control agents, binders, thickening agents, detergents, adhesion agents, foam modulators, pH modifying agents, mouth feel agents, sweeteners, flavorants, colorants, humectants, fluoride sources and combinations thereof. 5. The composition of claim 1, wherein the composition comprises free taurine and is substantially free of taurine salts. 6. The composition of claim 1 for use in preventing or repairing soft tissue damage in an oral cavity, or for use in healing a wound in a tissue of an oral cavity, or for use in preventing or treating inflammation in a tissue of an oral cavity, for use in reducing reactive oxygen species in a tissue of an oral cavity. 7. The composition for use according to claim 6, wherein the use comprises increasing or accelerating HSP 27 expression in the tissue. 8. The composition for use according to claim 6, wherein the tissue is gingival tissue. 9. The composition for use according to claim 6, wherein the use comprises contacting the oral cavity with the composition for a period of at least 30 seconds, at least 1 minute, at least 5 minutes, at least 10 minutes, at least 15 minutes or at least 1 hour. 10. A method of increasing or accelerating HSP 27 in a tissue of an oral cavity comprising administering to the tissue a composition according to claim 1. 11. The method of claim 10, wherein the method comprises preventing or repairing soft tissue damage in the oral cavity, or wherein the method comprises healing a wound in the tissue of the oral cavity, or wherein the method comprises preventing or treating inflammation in the tissue of the oral cavity, or wherein the method comprises reducing reactive oxygen species in the tissue of the oral cavity. 12. The method of claim 10, wherein the tissue is gingival tissue. 13. The method of claim 10, comprising contacting the oral cavity with the composition for a period of at least 30 seconds, at least 1 minute, at least 5 minutes, at least 10 minutes, at least 15 minutes or at least 1 hour. 14. (canceled) 15. (canceled) 16. A method of increasing or accelerating HSP 27 in a tissue of an oral cavity comprising administering to the tissue an effective amount of a taurine or a salt thereof. 17. The method of claim 16, wherein the effective amount of taurine or a salt thereof is from 10 to 1000 ppm, or from 10 to 500 ppm, or from 10 to 200 ppm, or from 10 to 100 ppm. 18. The method of claim 16, wherein the method comprises preventing or repairing soft tissue damage in the oral cavity, or comprises healing a wound in the tissue of the oral cavity, or comprises preventing or treating inflammation in the tissue of the oral cavity, or comprises reducing reactive oxygen species in the tissue of the oral cavity. 19. The method of claim 16, wherein the tissue is gingival tissue.
Provided herein is an oral care composition comprising taurine or a salt thereof in an amount of from 0.001 weight % to 0.008 weight % by total weight of the composition, and an orally acceptable carrier. The composition is effective in accelerating HSP 27 production in damaged tissues of the oral cavity and enhances repair of the damaged tissue.1. An oral care composition comprising taurine or a salt thereof in an amount of from 0.001 weight % to 0.05 weight %, e.g., 0.001 weight % to 0.008 weight % by total weight of the composition, and an orally acceptable carrier. 2. The composition of claim 1, wherein the taurine or salt thereof is present in the composition in an amount of from 0.003 weight % to 0.006 weight % by total weight of the composition. 3. The composition of claim 1 wherein the composition is selected from mouthwashes, sprays, dentifrices, oral strips, chewing gums and lozenges. 4. The composition of claim 1, wherein the oral care composition further comprises one or more agents selected from: surfactants, desensitizing agents, whitening agents, tartar control agents, binders, thickening agents, detergents, adhesion agents, foam modulators, pH modifying agents, mouth feel agents, sweeteners, flavorants, colorants, humectants, fluoride sources and combinations thereof. 5. The composition of claim 1, wherein the composition comprises free taurine and is substantially free of taurine salts. 6. The composition of claim 1 for use in preventing or repairing soft tissue damage in an oral cavity, or for use in healing a wound in a tissue of an oral cavity, or for use in preventing or treating inflammation in a tissue of an oral cavity, for use in reducing reactive oxygen species in a tissue of an oral cavity. 7. The composition for use according to claim 6, wherein the use comprises increasing or accelerating HSP 27 expression in the tissue. 8. The composition for use according to claim 6, wherein the tissue is gingival tissue. 9. The composition for use according to claim 6, wherein the use comprises contacting the oral cavity with the composition for a period of at least 30 seconds, at least 1 minute, at least 5 minutes, at least 10 minutes, at least 15 minutes or at least 1 hour. 10. A method of increasing or accelerating HSP 27 in a tissue of an oral cavity comprising administering to the tissue a composition according to claim 1. 11. The method of claim 10, wherein the method comprises preventing or repairing soft tissue damage in the oral cavity, or wherein the method comprises healing a wound in the tissue of the oral cavity, or wherein the method comprises preventing or treating inflammation in the tissue of the oral cavity, or wherein the method comprises reducing reactive oxygen species in the tissue of the oral cavity. 12. The method of claim 10, wherein the tissue is gingival tissue. 13. The method of claim 10, comprising contacting the oral cavity with the composition for a period of at least 30 seconds, at least 1 minute, at least 5 minutes, at least 10 minutes, at least 15 minutes or at least 1 hour. 14. (canceled) 15. (canceled) 16. A method of increasing or accelerating HSP 27 in a tissue of an oral cavity comprising administering to the tissue an effective amount of a taurine or a salt thereof. 17. The method of claim 16, wherein the effective amount of taurine or a salt thereof is from 10 to 1000 ppm, or from 10 to 500 ppm, or from 10 to 200 ppm, or from 10 to 100 ppm. 18. The method of claim 16, wherein the method comprises preventing or repairing soft tissue damage in the oral cavity, or comprises healing a wound in the tissue of the oral cavity, or comprises preventing or treating inflammation in the tissue of the oral cavity, or comprises reducing reactive oxygen species in the tissue of the oral cavity. 19. The method of claim 16, wherein the tissue is gingival tissue.
1,600
363
11,908,237
1,612
Methods of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject are provided. Methods of killing or inactivating microorganisms in at least a portion of the urethra of a subject are provided.
1. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, and a surfactant component distinct from the antimicrobial component, wherein the surfactant component is present in an amount of at least 0.5 wt-% and/or the surfactant component comprises an anionic surfactant, zwitterionic surfactant, poloxamer surfactant, amine oxide surfactant, or combinations thereof, with the proviso that when the internal cavity comprises a nasal passage, vagina, or oral cavity, the antimicrobial component does not include iodine or chlorhexidine. 2. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a surfactant component distinct from the antimicrobial component; and a vehicle comprising less than 1 wt-% water. 3. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a second active agent distinct from the antimicrobial component; and a surfactant component distinct from the antimicrobial component. 4. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, and at least 0.20 wt-% of an enhancer component comprising an alpha-hydroxy acid, a beta-hydroxy acid, a chelating agent, a (C1-C4)alkyl carboxylic acid, a (C6-C12)aryl carboxylic acid, a (C6-C12)aralkyl carboxylic acid, a (C6-C12)alkaryl carboxylic acid, a phenolic compound, a (C1-C10)alkyl alcohol, an ether glycol, or combinations thereof. 5. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinantions thereof, an effective amount of an enhancer component comprising a alpha-hydroxy acid, a beta-hydroxy acid, a chelating agent, a (C1-C4)alkyl carboxylic acid, a (C6-C12)aryl carboxylic acid, a (C6-C12)aralkyl carboxylic acid, a (C6-C12)alkaryl carboxylic acid, a phenolic compound, a (C1-C10)alkyl alcohol, an ether glycol, or combinations thereof, and a surfactant component distinct from the antimicrobial component. 6. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a hydrophilic vehicle other than water; and a surfactant component distinct from the antimicrobial component. 7. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof; a surfactant component distinct from the antimicrobial component; and wherein the composition has a viscosity of at least 1,000 cps at 23° C.; with the proviso that when the internal cavity comprises a nasal passage or vagina, the antimicrobial component does not include iodine. 8. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting the at least a portion of interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a hydrophilic component; and a hydrophobic component; wherein the hydrophobic component or the hydrophilic component forms the greatest portion of the composition by weight. 9. (canceled) 10. The method of any of claims 1-8 wherein the antiseptic comprises an antimicrobial lipid, a phenolic antiseptic, a cationic antiseptic, iodine and/or an iodophor, a peroxide antiseptic, an antimicrobial natural oil, or combinations thereof. 11-39. (canceled) 40. The method of any of claims 1-8 wherein the opening to the internal surface comprises at least a portion of the internal surface of the urethra, cervical opening, nasal passages, oral cavity, or a surgical incision/puncture site. 41. The method of any of claims 1-8 wherein the opening to the internal surface comprises at least a portion of the internal surface of the urethra. 42. The method of any of claims 1-8 wherein the instrument is selected from the group consisting of nasal gastric tubes, tracheotomy tubes, urinary catheters, peritoneal dialysis tubes, ventilator tubes, endotracheal tubes, and surgical instruments. 43-56. (canceled) 57. The method of any one of claims 1-8 wherein residual antimicrobial efficacy is provided to the surface to which the antimicrobial composition is applied. 58. The method of any one of claims 1-8 wherein the instrument is treated with the same or different antimicrobial composition prior to the inserting step. 59. (canceled) 60. The method of any one of claims 1-8 wherein the pH of the composition is less than 7. 61. The method of any one of claims 1-8 wherein the external tissue surrounding the opening is treated with the same or different antimicrobial composition prior to the inserting step.
Methods of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject are provided. Methods of killing or inactivating microorganisms in at least a portion of the urethra of a subject are provided.1. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, and a surfactant component distinct from the antimicrobial component, wherein the surfactant component is present in an amount of at least 0.5 wt-% and/or the surfactant component comprises an anionic surfactant, zwitterionic surfactant, poloxamer surfactant, amine oxide surfactant, or combinations thereof, with the proviso that when the internal cavity comprises a nasal passage, vagina, or oral cavity, the antimicrobial component does not include iodine or chlorhexidine. 2. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a surfactant component distinct from the antimicrobial component; and a vehicle comprising less than 1 wt-% water. 3. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a second active agent distinct from the antimicrobial component; and a surfactant component distinct from the antimicrobial component. 4. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, and at least 0.20 wt-% of an enhancer component comprising an alpha-hydroxy acid, a beta-hydroxy acid, a chelating agent, a (C1-C4)alkyl carboxylic acid, a (C6-C12)aryl carboxylic acid, a (C6-C12)aralkyl carboxylic acid, a (C6-C12)alkaryl carboxylic acid, a phenolic compound, a (C1-C10)alkyl alcohol, an ether glycol, or combinations thereof. 5. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinantions thereof, an effective amount of an enhancer component comprising a alpha-hydroxy acid, a beta-hydroxy acid, a chelating agent, a (C1-C4)alkyl carboxylic acid, a (C6-C12)aryl carboxylic acid, a (C6-C12)aralkyl carboxylic acid, a (C6-C12)alkaryl carboxylic acid, a phenolic compound, a (C1-C10)alkyl alcohol, an ether glycol, or combinations thereof, and a surfactant component distinct from the antimicrobial component. 6. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a hydrophilic vehicle other than water; and a surfactant component distinct from the antimicrobial component. 7. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting at least a portion of the interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof; a surfactant component distinct from the antimicrobial component; and wherein the composition has a viscosity of at least 1,000 cps at 23° C.; with the proviso that when the internal cavity comprises a nasal passage or vagina, the antimicrobial component does not include iodine. 8. A method of delaying the onset of an infection or preventing an infection caused by a microbial organism in an internal cavity of a subject, the method comprising: contacting the at least a portion of interior surface of an opening leading to the internal cavity with an antimicrobial composition; and subsequently at least partially inserting an instrument into the opening, wherein the antimicrobial composition comprises: an effective amount of an antimicrobial component comprising an antiseptic, an antibiotic, or combinations thereof, a hydrophilic component; and a hydrophobic component; wherein the hydrophobic component or the hydrophilic component forms the greatest portion of the composition by weight. 9. (canceled) 10. The method of any of claims 1-8 wherein the antiseptic comprises an antimicrobial lipid, a phenolic antiseptic, a cationic antiseptic, iodine and/or an iodophor, a peroxide antiseptic, an antimicrobial natural oil, or combinations thereof. 11-39. (canceled) 40. The method of any of claims 1-8 wherein the opening to the internal surface comprises at least a portion of the internal surface of the urethra, cervical opening, nasal passages, oral cavity, or a surgical incision/puncture site. 41. The method of any of claims 1-8 wherein the opening to the internal surface comprises at least a portion of the internal surface of the urethra. 42. The method of any of claims 1-8 wherein the instrument is selected from the group consisting of nasal gastric tubes, tracheotomy tubes, urinary catheters, peritoneal dialysis tubes, ventilator tubes, endotracheal tubes, and surgical instruments. 43-56. (canceled) 57. The method of any one of claims 1-8 wherein residual antimicrobial efficacy is provided to the surface to which the antimicrobial composition is applied. 58. The method of any one of claims 1-8 wherein the instrument is treated with the same or different antimicrobial composition prior to the inserting step. 59. (canceled) 60. The method of any one of claims 1-8 wherein the pH of the composition is less than 7. 61. The method of any one of claims 1-8 wherein the external tissue surrounding the opening is treated with the same or different antimicrobial composition prior to the inserting step.
1,600
364
14,966,843
1,649
CSF diagnostic in vitro method for the diagnosis of dementias and neuroinflammatory diseases, in which a determination of the procalcitonin immunoreactivity (PCT immunoreactivity) is carried out in a sample of cerebrospinal fluid (C SF) of a patient who is suffering from a dementia or neuroinflammatory disease or is suspected of suffering from such a disease. Conclusions about the presence, the course, the severity or the success of a treatment of the dementia or neuroinflammatory disease are drawn from a measured PCT immunoreactivity which is above a threshold value typical for healthy individuals.
1-13. (canceled) 14. A method for assisting in the detection and diagnosis of dementias selected from the group consisting of Alzheimer's dementia (AD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and various forms of vascular dementia (VAD), said method comprising: determining the level of procalcitonin (PCT) in a sample of cerebrospinal fluid (CSF) from a patient who is suffering from or, based on clinical manifestations, is suspected of having a dementia selected from the group consisting of Alzheimer's dementia (AD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and various forms of vascular dementia (VAD with the aid of a highly sensitive PCT immunoassay having a functional assay sensitivity (FAS) of 50 ng of PCT per liter (50 ng/l or 50 pg/ml) or better, said immunoassay comprising a pair of antibodies, wherein one of the pair of antibodies binds to calcitonin and the other of the pair of antibodies binds to katacalcin, and at least one of the pair of antibodies is an affinity-purified polyclonal antibody, wherein an increased level of procalcitonin in said sample when compared to levels of PCT in CSF from healthy individuals indicates dementia. 15. The method of claim 14, wherein said sensitive PCT immunoassay has a functional assay sensitivity (FAS) of 10 ng of PCT per liter (10 ng/l or 10 pg/ml) or better. 16. A method for measuring the level of procalcitonin (PCT) in a sample of cerebrospinal fluid (CSF) from a patient who is suffering from or is suspected of having a dementia, said method comprising: a) performing an immunoassay for PCT on a sample of CSF from a patient who is suffering from or, based on clinical manifestations, is suspected of having a dementia selected from the group consisting of Alzheimer's dementia (AD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and various forms of vascular dementia (VAD), wherein said immunoassay has a functional assay sensitivity (FAS) of 50 ng of PCT per liter (50ng/l or 50 pg/ml) or better, and said immunoassay comprises a pair of antibodies, wherein one of the pair of antibodies binds to calcitonin and the other of the pair of antibodies binds to katacalcin, and at least one of the pair of antibodies is an affinity-purified polyclonal antibody; b) determining the level of PCT in said sample, wherein an increased level of PCT in said sample when compared to levels of PCT in CSF from healthy individuals indicates dementia.
CSF diagnostic in vitro method for the diagnosis of dementias and neuroinflammatory diseases, in which a determination of the procalcitonin immunoreactivity (PCT immunoreactivity) is carried out in a sample of cerebrospinal fluid (C SF) of a patient who is suffering from a dementia or neuroinflammatory disease or is suspected of suffering from such a disease. Conclusions about the presence, the course, the severity or the success of a treatment of the dementia or neuroinflammatory disease are drawn from a measured PCT immunoreactivity which is above a threshold value typical for healthy individuals.1-13. (canceled) 14. A method for assisting in the detection and diagnosis of dementias selected from the group consisting of Alzheimer's dementia (AD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and various forms of vascular dementia (VAD), said method comprising: determining the level of procalcitonin (PCT) in a sample of cerebrospinal fluid (CSF) from a patient who is suffering from or, based on clinical manifestations, is suspected of having a dementia selected from the group consisting of Alzheimer's dementia (AD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and various forms of vascular dementia (VAD with the aid of a highly sensitive PCT immunoassay having a functional assay sensitivity (FAS) of 50 ng of PCT per liter (50 ng/l or 50 pg/ml) or better, said immunoassay comprising a pair of antibodies, wherein one of the pair of antibodies binds to calcitonin and the other of the pair of antibodies binds to katacalcin, and at least one of the pair of antibodies is an affinity-purified polyclonal antibody, wherein an increased level of procalcitonin in said sample when compared to levels of PCT in CSF from healthy individuals indicates dementia. 15. The method of claim 14, wherein said sensitive PCT immunoassay has a functional assay sensitivity (FAS) of 10 ng of PCT per liter (10 ng/l or 10 pg/ml) or better. 16. A method for measuring the level of procalcitonin (PCT) in a sample of cerebrospinal fluid (CSF) from a patient who is suffering from or is suspected of having a dementia, said method comprising: a) performing an immunoassay for PCT on a sample of CSF from a patient who is suffering from or, based on clinical manifestations, is suspected of having a dementia selected from the group consisting of Alzheimer's dementia (AD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and various forms of vascular dementia (VAD), wherein said immunoassay has a functional assay sensitivity (FAS) of 50 ng of PCT per liter (50ng/l or 50 pg/ml) or better, and said immunoassay comprises a pair of antibodies, wherein one of the pair of antibodies binds to calcitonin and the other of the pair of antibodies binds to katacalcin, and at least one of the pair of antibodies is an affinity-purified polyclonal antibody; b) determining the level of PCT in said sample, wherein an increased level of PCT in said sample when compared to levels of PCT in CSF from healthy individuals indicates dementia.
1,600
365
12,439,692
1,616
The invention provides aqueous pesticidal concentrates comprising at least one colloidal solid, a dispersed emulsion phase comprising at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase and at least one Ostwald ripening inhibitor.
1. A liquid pesticidal emulsion composition comprising (a) an aqueous continuous phase; (b) at least one colloidal solid; and (c) a dispersed oil emulsion phase comprising (i) at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase; and (ii) at least one Ostwald ripening inhibitor soluble or miscible in the oil phase or which itself serves as the oil phase. 2. The liquid pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises a liquid, substantially water-insoluble pesticidally active ingredient. 3. The liquid pesticidal composition of claim 2 wherein the liquid, substantially water-insoluble pesticidally active ingredient is dissolved in a solvent to form a low viscosity liquid. 4. The aqueous pesticidal composition of claim 1, wherein the water-insoluble pesticidally active ingredient is prepared by dissolving a solid, substantially water-insoluble pesticidally active ingredient in a solvent to form a low viscosity liquid. 5. The aqueous pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises a solid, substantially water-insoluble pesticidally active ingredient dispersed within the oil phase. 6. The aqueous pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises a substantially water-insoluble pesticidally active ingredient present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase. 7. The aqueous pesticidal composition of claim 1, wherein the aqueous phase contains at least one water-soluble agrochemical. 8. The aqueous pesticidal composition of claim 7, wherein the water-soluble agrochemical comprises at least one water-soluble agrochemical electrolyte. 9. The aqueous pesticidal composition of claim 8, wherein the at least one water-soluble agrochemical electrolyte comprises at least one member selected from the group consisting of ammonium sulfate, magnesium sulfate, dicamba, diquat, glyphosate, glufosinate, paraquat or mixtures thereof and agriculturally acceptable salts thereof. 10. The aqueous pesticidal composition of claim 9, wherein the water-soluble agrochemical electrolyte comprises at least one glyphosate salt selected from the group consisting of monosodium, monopotassium, diammonium, mono(dimethylammonium), mono(ethanolammonium), mono(isopropylammonium) and mono(trimethylsulfonium) salts. 11. The aqueous pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises an acetamide herbicide. 12. The aqueous pesticidal composition of claim 11, wherein the acetamide herbicide comprises at least one member selected from the group consisting of acetochlor, butachlor, metolachlor, S-metolachlor and pretilachlor, dimethenamid or dimethenamid-P. 13. The aqueous pesticidal composition of claim 12, wherein the acetamide comprises mixtures of metolachlor (S) and (R) isomers wherein the ratio of (S)-2-chloro-N-(2-ethyl-6-methylphenyl)-N-(2-methoxy-1-methylethyl)acetamide to (R)-2-chloro-N-(2-ethyl-6-methylphenyl)-N-(2-methoxy-1-methylethyl)acetamide is in the range of from 50-100% to 50-0%. 14. The aqueous pesticidal composition of claim 5, wherein the dispersed solid phase comprises at least one solid, substantially water-insoluble pesticidally active ingredient selected from the group consisting of chlorothalonil, isoxaflutole, mesotrione, including salts and chelates thereof, butafenacil, prodiamine, triazines, sulfonylurea herbicides, azoxystrobin, fludioxonil, tebuconazole and a compound of the formula (I): 15. The aqueous pesticidal composition of claim 14, wherein the dispersed solid phase comprises at least one solid, water-insoluble pesticidally active ingredient selected from the group consisting of mesotrione, atrazine, simazine, terbuthylazine, prodiamine, isoxaflutole, primisulfuron and prosulfuron. 16. The aqueous pesticidal composition of claim 1 wherein the composition comprises less than 0.5 weight % of an emulsifier formed of a low molecular weight or polymeric surfactant. 17. The aqueous pesticidal composition of claim 16 wherein the composition is entirely free of an emulsifier formed of a low molecular weight or polymeric surfactant. 18. A pesticidal composition obtained by diluting the pesticidal composition of claim 1 in a suitable carrier in an amount sufficient to obtain the desired final concentration of each of the active ingredients. 19. The pesticidal composition of claim 18 wherein the carrier is selected from water, liquid fertilizer or mixtures thereof. 20. A method for combating or preventing pests in crops of useful plants, said method comprising forming a liquid pesticidal emulsion composition comprising (a) an aqueous continuous phase; (b) at least one colloidal solid; and (c) a dispersed oil emulsion phase comprising (i) at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase; and (ii) at least one Ostwald ripening inhibitor soluble or miscible in the oil phase or which itself serves as the oil phase, diluting the emulsion composition, if necessary, in a suitable carrier in an amount sufficient to obtain the desired final concentration of each of the active ingredients and treating the desired area with said composition. 21. The method of combating or preventing pests in crops of useful plants according to claim 20, wherein the crops have been made tolerant to at least one of the pesticidally active ingredients as a result of conventional methods of breeding or genetic engineering. 22. The method of claim 21, wherein the crops are tolerant to ALS-, GS-, EPSPS-, PPO-, ACCase and/or HPPD-inhibitors and wherein the crops are treated post-emergence with said pesticidal compositions. 23. The method of claim 22, wherein the crops are tolerant to glyphosate and are selected from the group consisting of canola, cereals, cotton, maize, rice, soybeans and sugar beets. 24. A method of treating building materials or hides, said method comprising coating or impregnating a building material or treating said hides, with liquid, pesticidal emulsion compositions comprising (a) an aqueous continuous phase; (b) at least one colloidal solid; and (c) a dispersed oil emulsion phase comprising (i) at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase; and (ii) at least one Ostwald ripening inhibitor soluble or miscible in the oil phase or which itself serves as the oil phase. 25. The method of claim 24, wherein the composition is diluted in a suitable liquid carrier prior to coating or impregnating said building materials or hides.
The invention provides aqueous pesticidal concentrates comprising at least one colloidal solid, a dispersed emulsion phase comprising at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase and at least one Ostwald ripening inhibitor.1. A liquid pesticidal emulsion composition comprising (a) an aqueous continuous phase; (b) at least one colloidal solid; and (c) a dispersed oil emulsion phase comprising (i) at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase; and (ii) at least one Ostwald ripening inhibitor soluble or miscible in the oil phase or which itself serves as the oil phase. 2. The liquid pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises a liquid, substantially water-insoluble pesticidally active ingredient. 3. The liquid pesticidal composition of claim 2 wherein the liquid, substantially water-insoluble pesticidally active ingredient is dissolved in a solvent to form a low viscosity liquid. 4. The aqueous pesticidal composition of claim 1, wherein the water-insoluble pesticidally active ingredient is prepared by dissolving a solid, substantially water-insoluble pesticidally active ingredient in a solvent to form a low viscosity liquid. 5. The aqueous pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises a solid, substantially water-insoluble pesticidally active ingredient dispersed within the oil phase. 6. The aqueous pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises a substantially water-insoluble pesticidally active ingredient present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase. 7. The aqueous pesticidal composition of claim 1, wherein the aqueous phase contains at least one water-soluble agrochemical. 8. The aqueous pesticidal composition of claim 7, wherein the water-soluble agrochemical comprises at least one water-soluble agrochemical electrolyte. 9. The aqueous pesticidal composition of claim 8, wherein the at least one water-soluble agrochemical electrolyte comprises at least one member selected from the group consisting of ammonium sulfate, magnesium sulfate, dicamba, diquat, glyphosate, glufosinate, paraquat or mixtures thereof and agriculturally acceptable salts thereof. 10. The aqueous pesticidal composition of claim 9, wherein the water-soluble agrochemical electrolyte comprises at least one glyphosate salt selected from the group consisting of monosodium, monopotassium, diammonium, mono(dimethylammonium), mono(ethanolammonium), mono(isopropylammonium) and mono(trimethylsulfonium) salts. 11. The aqueous pesticidal composition of claim 1, wherein the substantially water-insoluble pesticidally active ingredient comprises an acetamide herbicide. 12. The aqueous pesticidal composition of claim 11, wherein the acetamide herbicide comprises at least one member selected from the group consisting of acetochlor, butachlor, metolachlor, S-metolachlor and pretilachlor, dimethenamid or dimethenamid-P. 13. The aqueous pesticidal composition of claim 12, wherein the acetamide comprises mixtures of metolachlor (S) and (R) isomers wherein the ratio of (S)-2-chloro-N-(2-ethyl-6-methylphenyl)-N-(2-methoxy-1-methylethyl)acetamide to (R)-2-chloro-N-(2-ethyl-6-methylphenyl)-N-(2-methoxy-1-methylethyl)acetamide is in the range of from 50-100% to 50-0%. 14. The aqueous pesticidal composition of claim 5, wherein the dispersed solid phase comprises at least one solid, substantially water-insoluble pesticidally active ingredient selected from the group consisting of chlorothalonil, isoxaflutole, mesotrione, including salts and chelates thereof, butafenacil, prodiamine, triazines, sulfonylurea herbicides, azoxystrobin, fludioxonil, tebuconazole and a compound of the formula (I): 15. The aqueous pesticidal composition of claim 14, wherein the dispersed solid phase comprises at least one solid, water-insoluble pesticidally active ingredient selected from the group consisting of mesotrione, atrazine, simazine, terbuthylazine, prodiamine, isoxaflutole, primisulfuron and prosulfuron. 16. The aqueous pesticidal composition of claim 1 wherein the composition comprises less than 0.5 weight % of an emulsifier formed of a low molecular weight or polymeric surfactant. 17. The aqueous pesticidal composition of claim 16 wherein the composition is entirely free of an emulsifier formed of a low molecular weight or polymeric surfactant. 18. A pesticidal composition obtained by diluting the pesticidal composition of claim 1 in a suitable carrier in an amount sufficient to obtain the desired final concentration of each of the active ingredients. 19. The pesticidal composition of claim 18 wherein the carrier is selected from water, liquid fertilizer or mixtures thereof. 20. A method for combating or preventing pests in crops of useful plants, said method comprising forming a liquid pesticidal emulsion composition comprising (a) an aqueous continuous phase; (b) at least one colloidal solid; and (c) a dispersed oil emulsion phase comprising (i) at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase; and (ii) at least one Ostwald ripening inhibitor soluble or miscible in the oil phase or which itself serves as the oil phase, diluting the emulsion composition, if necessary, in a suitable carrier in an amount sufficient to obtain the desired final concentration of each of the active ingredients and treating the desired area with said composition. 21. The method of combating or preventing pests in crops of useful plants according to claim 20, wherein the crops have been made tolerant to at least one of the pesticidally active ingredients as a result of conventional methods of breeding or genetic engineering. 22. The method of claim 21, wherein the crops are tolerant to ALS-, GS-, EPSPS-, PPO-, ACCase and/or HPPD-inhibitors and wherein the crops are treated post-emergence with said pesticidal compositions. 23. The method of claim 22, wherein the crops are tolerant to glyphosate and are selected from the group consisting of canola, cereals, cotton, maize, rice, soybeans and sugar beets. 24. A method of treating building materials or hides, said method comprising coating or impregnating a building material or treating said hides, with liquid, pesticidal emulsion compositions comprising (a) an aqueous continuous phase; (b) at least one colloidal solid; and (c) a dispersed oil emulsion phase comprising (i) at least one substantially water-insoluble pesticidally active ingredient, which is either itself an oily liquid comprising the oil phase, is a solid but is dissolved in an oily liquid present in the oil phase, is a solid and is dispersed within the oil phase or is present as a colloidal solid adsorbed to the liquid-liquid interface between the continuous aqueous phase and the dispersed oil phase; and (ii) at least one Ostwald ripening inhibitor soluble or miscible in the oil phase or which itself serves as the oil phase. 25. The method of claim 24, wherein the composition is diluted in a suitable liquid carrier prior to coating or impregnating said building materials or hides.
1,600
366
13,656,573
1,613
Vitamin E compositions, methods of making such compositions and therapeutic uses of vitamin E compositions are disclosed. Compositions of matter comprising a tocotrienol making up at least 15 dry basis weight percent of the composition of matter and a constituent selected from a triglyceride and a triglyceride ester, wherein upon mixing of the composition of matter with water the tocotrienol based composition is substantially emulsified; and wherein upon mixing of the composition of matter with water a resulting emulsion has an intensity-weighed mean droplet size of less than 700 nm are also disclosed.
1. A composition of matter comprising: a. a tocotrienol based composition making up at least 15 dry basis weight percent of the composition of matter; and b. a constituent selected from a triglyceride and a triglyceride ester; c. wherein upon mixing of the composition of matter with water the tocotrienol based composition is substantially emulsified; and d. wherein upon mixing of the composition of matter with water a resulting emulsion has an intensity-weighed mean droplet size of less than 700 nm. 2. The composition of matter of claim 1 further comprising a first emulsifier making up at least 0.25 dry basis weight percent of the composition of matter. 3. The composition of matter of claim 2 wherein the composition of matter is an emulsion. 4. The composition of matter of claim 2 wherein the first emulsifier is selected from Polysorbate 80 and phospholipid. 5. The composition of matter of claim 2 wherein the first emulsifier is Polysorbate 80 and wherein the emulsion further comprises a phospholipid. 6. The composition of matter of claim 1 wherein the constituent is selected from a triglyceride and a triglyceride ester is a medium chain triglyceride. 7. The composition of matter of claim 1 wherein the constituent is selected from a triglyceride and a triglyceride ester is a Caprylic/Capric triglyceride. 8. The composition of matter of claim 1 wherein the constituent is selected from a triglyceride and a triglyceride ester is a coconut oil. 9. The composition of matter of claim 1 further comprising cholesterol. 10. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester makes up at least five dry basis weight percent of the composition of matter. 11. The composition of matter of claim 1 further comprising a first nonionic copolymer. 12. The composition of matter of claim 11 wherein the first nonionic copolymer is a nonionic block copolymer. 13. The composition of matter of claim 11 wherein the first nonionic copolymer is poloxamer 188. 14. The composition of matter of claim 11 wherein the first nonionic copolymer makes up at least 0.5 weight percent of the composition of matter. 15. The composition of matter of claim 11 wherein the first nonionic copolymer makes up at least 1.0 weight percent of the composition of matter. 16. The composition of matter of claim 11 wherein the first non-ionic copolymer makes up at least 1.5 weight percent of the composition of matter. 17. The composition of matter of claim 11 wherein the first non-ionic copolymer makes up at least 2.5 weight percent of the composition of matter. 18. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the tocotrienol based composition makes up at least 10 weight percent of the oil phase. 19. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the tocotrienol based composition makes up at least 20 weight percent of the oil phase. 20. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the tocotrienol based composition makes up at least 30 weight percent of the oil phase. 21. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the oil phase is at least 5 weight percent of the composition of matter. 22. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the oil phase is at least 10 weight percent of the composition of matter. 23. The composition of matter of claim 1 further comprising a poloxamer. 24. The composition of matter of claim 1 wherein the composition of matter is not a hemolytic toxin. 25. The composition of matter of claim 1 wherein the composition of matter has growth inhibitory activity against a cell line selected from MCF-7 human mammary adenocarcinoma and SW-620 human colon adenocarcinoma. 26. The composition of matter of claim 1 wherein the composition of matter is a self-emulsifying drug delivery system. 27. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester is a triglyceride ester. 28. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester is polyoxyethylated castor oil. 29. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester makes up at least 10 dry basis weight percent of the composition of matter. 30. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester makes up at least 15 dry basis weight percent of the composition of matter. 31. The composition of matter of claim 1 further comprising a glycerol ester. 32. The composition of matter of claim 1 further comprising a coconut oil. 33. The composition of matter of claim 1 further comprising an alcohol. 34. The composition of matter of claim 34 wherein the alcohol is ethanol. 35. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 20 dry basis weight percent of the composition of matter. 36. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 30 dry basis weight percent of the composition of matter. 37. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 40 dry basis weight percent of the composition of matter. 38. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 50 dry basis weight percent of the composition of matter. 39. The composition of matter of claim 1 wherein upon mixing of the composition of matter with water the resulting emulsion has an intensity-weighed mean droplet size of less than 500 nm. 40. The composition of matter of claim 1 wherein upon mixing of the composition of matter with water the resulting emulsion has an intensity-weighed mean droplet size of less than 300 nm. 41. The composition of matter of claim 1 wherein upon mixing of the composition of matter with water the resulting emulsion has an intensity-weighed mean droplet size of less than 250 nm. 42. The composition of matter of claim 1 wherein the tocotrienol based composition is tocotrienol. 43. The composition of matter of claim 1 wherein the tocotrienol based composition is a tocotrienol derivative. 44. A self-emulsifying drug delivery system comprising: a. a tocotrienol based composition making up at least 15 dry basis weight percent of the composition of matter; b. a polyoxyethylated triglyceride making up at least 10 dry basis weight percent of the self-emulsifying drug delivery system; c. wherein upon mixing of the self-emulsifying drug delivery system with water the tocotrienol based composition is substantially emulsified; and d. wherein upon mixing of the self-emulsifying drug delivery system with water a resulting emulsion has an intensity-weighed mean droplet size of less than 700 nm. 45. The self-emulsifying drug delivery system of claim 44 further comprising: a. a glycerol ester; b. a coconut oil; and c. an alcohol; d. wherein the polyoxyethylated triglyceride is polyoxyethylated castor oil. 46. An emulsion comprising: a. a tocotrienol based composition making up at least 15 dry basis weight percent of the emulsion; and b. a medium chain triglyceride; c. wherein the tocotrienol based composition is substantially emulsified; and d. wherein the emulsion has an intensity-weighed mean droplet size of less than 500 nm. 47. The emulsion of claim 46 further comprising: a. a phospholipid; and b. a poloxamer; c. wherein the medium chain triglyceride makes up at least five dry basis weight percent of the emulsion; and d. wherein the emulsion has an oil phase and the tocotrienol based composition makes up at least 20 weight percent of the oil phase.
Vitamin E compositions, methods of making such compositions and therapeutic uses of vitamin E compositions are disclosed. Compositions of matter comprising a tocotrienol making up at least 15 dry basis weight percent of the composition of matter and a constituent selected from a triglyceride and a triglyceride ester, wherein upon mixing of the composition of matter with water the tocotrienol based composition is substantially emulsified; and wherein upon mixing of the composition of matter with water a resulting emulsion has an intensity-weighed mean droplet size of less than 700 nm are also disclosed.1. A composition of matter comprising: a. a tocotrienol based composition making up at least 15 dry basis weight percent of the composition of matter; and b. a constituent selected from a triglyceride and a triglyceride ester; c. wherein upon mixing of the composition of matter with water the tocotrienol based composition is substantially emulsified; and d. wherein upon mixing of the composition of matter with water a resulting emulsion has an intensity-weighed mean droplet size of less than 700 nm. 2. The composition of matter of claim 1 further comprising a first emulsifier making up at least 0.25 dry basis weight percent of the composition of matter. 3. The composition of matter of claim 2 wherein the composition of matter is an emulsion. 4. The composition of matter of claim 2 wherein the first emulsifier is selected from Polysorbate 80 and phospholipid. 5. The composition of matter of claim 2 wherein the first emulsifier is Polysorbate 80 and wherein the emulsion further comprises a phospholipid. 6. The composition of matter of claim 1 wherein the constituent is selected from a triglyceride and a triglyceride ester is a medium chain triglyceride. 7. The composition of matter of claim 1 wherein the constituent is selected from a triglyceride and a triglyceride ester is a Caprylic/Capric triglyceride. 8. The composition of matter of claim 1 wherein the constituent is selected from a triglyceride and a triglyceride ester is a coconut oil. 9. The composition of matter of claim 1 further comprising cholesterol. 10. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester makes up at least five dry basis weight percent of the composition of matter. 11. The composition of matter of claim 1 further comprising a first nonionic copolymer. 12. The composition of matter of claim 11 wherein the first nonionic copolymer is a nonionic block copolymer. 13. The composition of matter of claim 11 wherein the first nonionic copolymer is poloxamer 188. 14. The composition of matter of claim 11 wherein the first nonionic copolymer makes up at least 0.5 weight percent of the composition of matter. 15. The composition of matter of claim 11 wherein the first nonionic copolymer makes up at least 1.0 weight percent of the composition of matter. 16. The composition of matter of claim 11 wherein the first non-ionic copolymer makes up at least 1.5 weight percent of the composition of matter. 17. The composition of matter of claim 11 wherein the first non-ionic copolymer makes up at least 2.5 weight percent of the composition of matter. 18. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the tocotrienol based composition makes up at least 10 weight percent of the oil phase. 19. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the tocotrienol based composition makes up at least 20 weight percent of the oil phase. 20. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the tocotrienol based composition makes up at least 30 weight percent of the oil phase. 21. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the oil phase is at least 5 weight percent of the composition of matter. 22. The composition of matter of claim 1 wherein the composition of matter has an oil phase and the oil phase is at least 10 weight percent of the composition of matter. 23. The composition of matter of claim 1 further comprising a poloxamer. 24. The composition of matter of claim 1 wherein the composition of matter is not a hemolytic toxin. 25. The composition of matter of claim 1 wherein the composition of matter has growth inhibitory activity against a cell line selected from MCF-7 human mammary adenocarcinoma and SW-620 human colon adenocarcinoma. 26. The composition of matter of claim 1 wherein the composition of matter is a self-emulsifying drug delivery system. 27. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester is a triglyceride ester. 28. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester is polyoxyethylated castor oil. 29. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester makes up at least 10 dry basis weight percent of the composition of matter. 30. The composition of matter of claim 1 wherein the constituent selected from a triglyceride and a triglyceride ester makes up at least 15 dry basis weight percent of the composition of matter. 31. The composition of matter of claim 1 further comprising a glycerol ester. 32. The composition of matter of claim 1 further comprising a coconut oil. 33. The composition of matter of claim 1 further comprising an alcohol. 34. The composition of matter of claim 34 wherein the alcohol is ethanol. 35. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 20 dry basis weight percent of the composition of matter. 36. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 30 dry basis weight percent of the composition of matter. 37. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 40 dry basis weight percent of the composition of matter. 38. The composition of matter of claim 1 wherein the tocotrienol based composition makes up at least 50 dry basis weight percent of the composition of matter. 39. The composition of matter of claim 1 wherein upon mixing of the composition of matter with water the resulting emulsion has an intensity-weighed mean droplet size of less than 500 nm. 40. The composition of matter of claim 1 wherein upon mixing of the composition of matter with water the resulting emulsion has an intensity-weighed mean droplet size of less than 300 nm. 41. The composition of matter of claim 1 wherein upon mixing of the composition of matter with water the resulting emulsion has an intensity-weighed mean droplet size of less than 250 nm. 42. The composition of matter of claim 1 wherein the tocotrienol based composition is tocotrienol. 43. The composition of matter of claim 1 wherein the tocotrienol based composition is a tocotrienol derivative. 44. A self-emulsifying drug delivery system comprising: a. a tocotrienol based composition making up at least 15 dry basis weight percent of the composition of matter; b. a polyoxyethylated triglyceride making up at least 10 dry basis weight percent of the self-emulsifying drug delivery system; c. wherein upon mixing of the self-emulsifying drug delivery system with water the tocotrienol based composition is substantially emulsified; and d. wherein upon mixing of the self-emulsifying drug delivery system with water a resulting emulsion has an intensity-weighed mean droplet size of less than 700 nm. 45. The self-emulsifying drug delivery system of claim 44 further comprising: a. a glycerol ester; b. a coconut oil; and c. an alcohol; d. wherein the polyoxyethylated triglyceride is polyoxyethylated castor oil. 46. An emulsion comprising: a. a tocotrienol based composition making up at least 15 dry basis weight percent of the emulsion; and b. a medium chain triglyceride; c. wherein the tocotrienol based composition is substantially emulsified; and d. wherein the emulsion has an intensity-weighed mean droplet size of less than 500 nm. 47. The emulsion of claim 46 further comprising: a. a phospholipid; and b. a poloxamer; c. wherein the medium chain triglyceride makes up at least five dry basis weight percent of the emulsion; and d. wherein the emulsion has an oil phase and the tocotrienol based composition makes up at least 20 weight percent of the oil phase.
1,600
367
15,411,083
1,645
The present invention includes a composition including as one component a slurry matrix that is a liquid at room temperature and a gel at physiological pH, physiological salt concentrations and/or physiological temperatures and as a second component one or more antigens. Also include are methods of inducing an immune response in a subject and vaccinating a subject by administering such compositions.
1. A composition comprising as one component a slurry matrix that is a liquid at room temperature and a gel at physiological pH, physiological salt concentrations and/or physiological temperatures and as another component one or more antigens. 2. The composition of claim 1, wherein the slurry matrix comprises a peptide hydrogel, or a derivative thereof. 3. The composition of claim 1, wherein the slurry matrix comprises a peptide hydrogel of the peptide scaffold RADARADARADARADA, or a derivative thereof. 4. The composition of claim 3, wherein the peptide hydrogel comprises PURAMATRIX, or a derivative thereof. 5. The composition of claim 1, wherein the slurry matrix comprises MATRIGEL, or a derivative thereof. 6. The composition of claim 1 further comprising one of more adjuvants. 7. The composition of claim 1, further comprising a Toll-Like Receptor (TLR) agonist and/or a cytokine 8. The composition of claim 7, wherein the TLR agonist comprises a TLR4 and/or TLR9 agonist. 9. The composition of claim 8, wherein the TLR9 agonist comprises a CpG oligodeoxynucleotide (ODN). 10. The composition of claim 1, wherein the antigen comprises a hepatitis antigen, an influenza antigen, a schistosomiasis antigen, and/or a burkolderia antigen, or an antigenic fragment thereof. 11. A method of producing an immune response in a subject, the method comprising administering a composition of claim 1 to the subject. 12. A method of delivering one or more immunogenic antigens to a subject, the method comprising administering a composition of claim 1 to the subject. 13. A method of delivering one or more therapeutic antigens to a subject, the method comprising administering a composition of claim 1 to the subject. 14. The method of claim 11, wherein the subject is a domestic livestock or a companion animal. 15. The method of claim 11, wherein administration of the composition comprises subcutaneous (sc) delivery, intramuscular (im) delivery, intradermal delivery, transdermal delivery, mucosal delivery, intravaginal delivery, intrarectal delivery, intraperitoneal delivery, inhalation delivery, and/or aerosol delivery. 16. The method of claim 11, wherein administration of the composition comprises a primary and/or booster vaccination. 17. The method of claim 11, wherein administration of the composition comprises a booster vaccination after a primary vaccination with a polypeptide vaccine or a plasmid DNA vaccine. 18. A method of producing an anti-schistosome immune response in a bovoid, the method comprising administering a composition comprising as one component a slurry matrix that is a liquid at room temperature and is a gel at physiological conditions and as another component one or more schistosome antigens to the bovoid. 19. The method of claim 18, wherein the schistosome antigen comprises a SjCTPI polypeptide, a SjCTPI-Hsp70 polypeptide, a SjC23 polypeptide, and/or a SjC23-Hsp70 polypeptide, or an antigenic fragment thereof. 20. The method of claim 19, wherein administration of the composition comprises a booster administered after a primary vaccination with a SjCTPI-Hsp70 plasmid DNA vaccine.
The present invention includes a composition including as one component a slurry matrix that is a liquid at room temperature and a gel at physiological pH, physiological salt concentrations and/or physiological temperatures and as a second component one or more antigens. Also include are methods of inducing an immune response in a subject and vaccinating a subject by administering such compositions.1. A composition comprising as one component a slurry matrix that is a liquid at room temperature and a gel at physiological pH, physiological salt concentrations and/or physiological temperatures and as another component one or more antigens. 2. The composition of claim 1, wherein the slurry matrix comprises a peptide hydrogel, or a derivative thereof. 3. The composition of claim 1, wherein the slurry matrix comprises a peptide hydrogel of the peptide scaffold RADARADARADARADA, or a derivative thereof. 4. The composition of claim 3, wherein the peptide hydrogel comprises PURAMATRIX, or a derivative thereof. 5. The composition of claim 1, wherein the slurry matrix comprises MATRIGEL, or a derivative thereof. 6. The composition of claim 1 further comprising one of more adjuvants. 7. The composition of claim 1, further comprising a Toll-Like Receptor (TLR) agonist and/or a cytokine 8. The composition of claim 7, wherein the TLR agonist comprises a TLR4 and/or TLR9 agonist. 9. The composition of claim 8, wherein the TLR9 agonist comprises a CpG oligodeoxynucleotide (ODN). 10. The composition of claim 1, wherein the antigen comprises a hepatitis antigen, an influenza antigen, a schistosomiasis antigen, and/or a burkolderia antigen, or an antigenic fragment thereof. 11. A method of producing an immune response in a subject, the method comprising administering a composition of claim 1 to the subject. 12. A method of delivering one or more immunogenic antigens to a subject, the method comprising administering a composition of claim 1 to the subject. 13. A method of delivering one or more therapeutic antigens to a subject, the method comprising administering a composition of claim 1 to the subject. 14. The method of claim 11, wherein the subject is a domestic livestock or a companion animal. 15. The method of claim 11, wherein administration of the composition comprises subcutaneous (sc) delivery, intramuscular (im) delivery, intradermal delivery, transdermal delivery, mucosal delivery, intravaginal delivery, intrarectal delivery, intraperitoneal delivery, inhalation delivery, and/or aerosol delivery. 16. The method of claim 11, wherein administration of the composition comprises a primary and/or booster vaccination. 17. The method of claim 11, wherein administration of the composition comprises a booster vaccination after a primary vaccination with a polypeptide vaccine or a plasmid DNA vaccine. 18. A method of producing an anti-schistosome immune response in a bovoid, the method comprising administering a composition comprising as one component a slurry matrix that is a liquid at room temperature and is a gel at physiological conditions and as another component one or more schistosome antigens to the bovoid. 19. The method of claim 18, wherein the schistosome antigen comprises a SjCTPI polypeptide, a SjCTPI-Hsp70 polypeptide, a SjC23 polypeptide, and/or a SjC23-Hsp70 polypeptide, or an antigenic fragment thereof. 20. The method of claim 19, wherein administration of the composition comprises a booster administered after a primary vaccination with a SjCTPI-Hsp70 plasmid DNA vaccine.
1,600
368
13,997,344
1,613
Described herein are oral fluid compositions comprising one or more structuring agents, and methods of making and using the same.
1. A fluid composition comprising: one or more structuring agents selected from the group consisting of: a gum-type colloidal polymer; a cellulosic polymer; an acrylate polymer; and a clay or fine particulate; and an orally acceptable aqueous carrier; wherein the composition is a mouthwash or mouthrinse, and wherein the composition has a Flow Rate Index of less than 0.85. 2. The composition of claim 1, wherein the total concentration of said one or more structuring agents is less than about 5%, by weight, of the composition. 3. The composition of claim 2, wherein at least one of said one or more structuring agents is a gum-type colloidal polymer, a cellulosic polymer and an acrylate polymer. 4-5. (canceled) 6. The composition of claim 3, wherein structuring agents further comprises a clay or fine particulate. 7. The composition of claim 3, wherein at least one of said one or more structuring agents is a gum-type colloidal polymer selected from: agar, agarose, albumin, algae colloid, alginates, alginic acid and salts thereof, amber, ammoniac, amylopectins, arabinans, arabinogalactan, arabinoxylans, asafetida, bdellium, carageenans, casein, chicle, collagen, copal, curdlan, dermatin sulfate, dextrans, cross-linked dextrans, dextrin, emulsan, gelatin, fenugreek, frankincense, furcellarans, galactoglucomannas, galactomannans, gamboge, gellan, gellan gum, glucomannans, glycogens, guar, guar gum, hydroxypropylated guar gums, carboxymethyl guar gum, carboxymethyl(hydroxypropyl) guar gum, hydroxyethyl guar gum, gum arabic, gum elastic, gum ghatti, gum karaya, gum tragancanth (tragacanthin), heparin, hyaluronic acid, India rubber, inulin, karaya gum, keratin sulfate, konjac flour, konjac mannan, labdanum, laminarans, laurdimonium, laxseed saccharide (acidic), levan, locust bean gum, myrrh, okra gum, pectic acids, pectin, polydextrose, polyquaternium-4, polyquaternium-10, polyquaternium-28, protopectins, psyllium seed gum, pullulan, quince seed gum, sodium hyaluronate, raffinose, rhamsan, scleroglucan, sodium alginate, stachylose, starch from rice, corn, potato or wheat, tapioca starch, succinoglycan, tamarind seed gum, trant gum, water-soluble soybean polysaccharide, whelan, xanthan, xanthan gum, xylans, xyloglucans, and mixtures thereof; wherein at least one of said one or more structuring agents is a cellulosic polymer selected from: cellulose; methyl cellulose: ethyl cellulose; propyl cellulose; butyl cellulose: carboxymethyl cellulose; carboxyethyl cellulose; carboxymethyl methyl cellulose; carboxyethyl ethyl cellulose; carboxyethyl methyl cellulose; carboxymethyl ethyl cellulose; hydroxyalkyl cellulose; hydroxymethyl cellulose; hydroxyethyl cellulose; hydroxypropyl cellulose; hydroxybutyl cellulose; hydroxymethyl methyl cellulose; hydroxyethyl methyl cellulose: hydroxypropyl methyl cellulose; hydroxybutyl methyl cellulose; hydroxymethyl ethyl cellulose; hydroxyethyl ethyl cellulose; hydroxypropyl ethyl cellulose; hydroxybutyl ethyl cellulose; hydroxymethyl propyl cellulose; hydroxyethyl propyl cellulose; hydroxypropyl propyl cellulose; hydroxypropyl propyl cellulose; hydroxymethyl butyl cellulose; hydroxyethyl butyl cellulose; hydroxypropyl butyl cellulose: hydroxybutyl butyl cellulose; hydroxypropyl oxyethyl cellulose: steardimonium hydroxyethyl cellulose; cocodimonium hydroxypropyl oxyethyl cellulose; sodium carboxymethyl cellulose; nitrocellulose; sodium cellulose sulfate; chondroitin; chitin; chitosan; chitosan pyrrolidone carboxylate; chitosan glycolate chitosan lactate and mixtures thereof; wherein at least one of said one or more structuring agents is an acrylate polymer selected from: homopolymers of acrylic acid, crosslinked with an allyl ether pentaerythritol, allyl ether of sucrose or allyl ether of propylene, polyvinyl methylether, and carboxyvinyl polymer; polyoxyethylene polymers; polyoxyethylene/polyoxypropylene copolymers; acrylic polymers such as sodium polyacrylate, polyethyl acrylate, and polyacrylamide; synthetic water-soluble polymers such as polyethyleneimine and other kind of cationic polymers; semi-synthetic water-soluble polymers such as silicone-modified pulllan; and water-soluble inorganic polymers such as, bentonite, aluminum magnesium silicate, montmorillonite, beidellite, notronite, saponite, hectorite, and silicic anhydride. 8-9. (canceled) 10. The composition of claim 6, wherein at least one of said one or more structuring agents is a clay or fine particulate selected from: calcium magnesium silicate and amorphous silica. 11. The composition of claim 3, further comprising a humectant selected from: sorbitol, glycerin, propylene glycol, ethanol, and a combination of two or more thereof. 12. The composition of claim 3, wherein the orally acceptable aqueous carrier comprises greater than 50%, by weight, free water. 13. The composition of claim 3, further comprising one or more components selected from a fluoride ion source; a tartar control agent; a buffering agent; an abrasive; and a combination of two or more thereof. 14. The composition of claim 13, wherein at least one of said one or more components is a fluoride ion source selected from: stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate; ammonium fluorosilicate, amine fluoride, ammonium fluoride, and a combination of two or more thereof. 15. The composition of claim 3, comprising: from about 0.01 to about 0.5%, by weight, of a gum-type colloidal polymer; from about 0.01 to about 0.5%, by weight, of a cellulosic polymer; and from about 0.01 to about 0.5%, by weight, of an acrylate polymer. 16. The composition of claim 3, comprising: from about 0.01 to about 0.5%, by weight, of an additional gum-type colloidal polymer. 17. The composition of claim 6, wherein the clay or fine particulate is present in the composition at a concentration of 0.5%, by weight. 18. The composition of claim 3, further comprising a suspended material. 19. The composition of claim 18, wherein said suspended material is selected from: a colloidal metal; a film flake; a film strip; and a combination of two or more thereof. 20. A method of treating or preventing a disease or condition of the oral cavity comprising contacting an oral cavity surface of a subject in need thereof, with the composition of claim 1. 21. (canceled)
Described herein are oral fluid compositions comprising one or more structuring agents, and methods of making and using the same.1. A fluid composition comprising: one or more structuring agents selected from the group consisting of: a gum-type colloidal polymer; a cellulosic polymer; an acrylate polymer; and a clay or fine particulate; and an orally acceptable aqueous carrier; wherein the composition is a mouthwash or mouthrinse, and wherein the composition has a Flow Rate Index of less than 0.85. 2. The composition of claim 1, wherein the total concentration of said one or more structuring agents is less than about 5%, by weight, of the composition. 3. The composition of claim 2, wherein at least one of said one or more structuring agents is a gum-type colloidal polymer, a cellulosic polymer and an acrylate polymer. 4-5. (canceled) 6. The composition of claim 3, wherein structuring agents further comprises a clay or fine particulate. 7. The composition of claim 3, wherein at least one of said one or more structuring agents is a gum-type colloidal polymer selected from: agar, agarose, albumin, algae colloid, alginates, alginic acid and salts thereof, amber, ammoniac, amylopectins, arabinans, arabinogalactan, arabinoxylans, asafetida, bdellium, carageenans, casein, chicle, collagen, copal, curdlan, dermatin sulfate, dextrans, cross-linked dextrans, dextrin, emulsan, gelatin, fenugreek, frankincense, furcellarans, galactoglucomannas, galactomannans, gamboge, gellan, gellan gum, glucomannans, glycogens, guar, guar gum, hydroxypropylated guar gums, carboxymethyl guar gum, carboxymethyl(hydroxypropyl) guar gum, hydroxyethyl guar gum, gum arabic, gum elastic, gum ghatti, gum karaya, gum tragancanth (tragacanthin), heparin, hyaluronic acid, India rubber, inulin, karaya gum, keratin sulfate, konjac flour, konjac mannan, labdanum, laminarans, laurdimonium, laxseed saccharide (acidic), levan, locust bean gum, myrrh, okra gum, pectic acids, pectin, polydextrose, polyquaternium-4, polyquaternium-10, polyquaternium-28, protopectins, psyllium seed gum, pullulan, quince seed gum, sodium hyaluronate, raffinose, rhamsan, scleroglucan, sodium alginate, stachylose, starch from rice, corn, potato or wheat, tapioca starch, succinoglycan, tamarind seed gum, trant gum, water-soluble soybean polysaccharide, whelan, xanthan, xanthan gum, xylans, xyloglucans, and mixtures thereof; wherein at least one of said one or more structuring agents is a cellulosic polymer selected from: cellulose; methyl cellulose: ethyl cellulose; propyl cellulose; butyl cellulose: carboxymethyl cellulose; carboxyethyl cellulose; carboxymethyl methyl cellulose; carboxyethyl ethyl cellulose; carboxyethyl methyl cellulose; carboxymethyl ethyl cellulose; hydroxyalkyl cellulose; hydroxymethyl cellulose; hydroxyethyl cellulose; hydroxypropyl cellulose; hydroxybutyl cellulose; hydroxymethyl methyl cellulose; hydroxyethyl methyl cellulose: hydroxypropyl methyl cellulose; hydroxybutyl methyl cellulose; hydroxymethyl ethyl cellulose; hydroxyethyl ethyl cellulose; hydroxypropyl ethyl cellulose; hydroxybutyl ethyl cellulose; hydroxymethyl propyl cellulose; hydroxyethyl propyl cellulose; hydroxypropyl propyl cellulose; hydroxypropyl propyl cellulose; hydroxymethyl butyl cellulose; hydroxyethyl butyl cellulose; hydroxypropyl butyl cellulose: hydroxybutyl butyl cellulose; hydroxypropyl oxyethyl cellulose: steardimonium hydroxyethyl cellulose; cocodimonium hydroxypropyl oxyethyl cellulose; sodium carboxymethyl cellulose; nitrocellulose; sodium cellulose sulfate; chondroitin; chitin; chitosan; chitosan pyrrolidone carboxylate; chitosan glycolate chitosan lactate and mixtures thereof; wherein at least one of said one or more structuring agents is an acrylate polymer selected from: homopolymers of acrylic acid, crosslinked with an allyl ether pentaerythritol, allyl ether of sucrose or allyl ether of propylene, polyvinyl methylether, and carboxyvinyl polymer; polyoxyethylene polymers; polyoxyethylene/polyoxypropylene copolymers; acrylic polymers such as sodium polyacrylate, polyethyl acrylate, and polyacrylamide; synthetic water-soluble polymers such as polyethyleneimine and other kind of cationic polymers; semi-synthetic water-soluble polymers such as silicone-modified pulllan; and water-soluble inorganic polymers such as, bentonite, aluminum magnesium silicate, montmorillonite, beidellite, notronite, saponite, hectorite, and silicic anhydride. 8-9. (canceled) 10. The composition of claim 6, wherein at least one of said one or more structuring agents is a clay or fine particulate selected from: calcium magnesium silicate and amorphous silica. 11. The composition of claim 3, further comprising a humectant selected from: sorbitol, glycerin, propylene glycol, ethanol, and a combination of two or more thereof. 12. The composition of claim 3, wherein the orally acceptable aqueous carrier comprises greater than 50%, by weight, free water. 13. The composition of claim 3, further comprising one or more components selected from a fluoride ion source; a tartar control agent; a buffering agent; an abrasive; and a combination of two or more thereof. 14. The composition of claim 13, wherein at least one of said one or more components is a fluoride ion source selected from: stannous fluoride, sodium fluoride, potassium fluoride, sodium monofluorophosphate, sodium fluorosilicate; ammonium fluorosilicate, amine fluoride, ammonium fluoride, and a combination of two or more thereof. 15. The composition of claim 3, comprising: from about 0.01 to about 0.5%, by weight, of a gum-type colloidal polymer; from about 0.01 to about 0.5%, by weight, of a cellulosic polymer; and from about 0.01 to about 0.5%, by weight, of an acrylate polymer. 16. The composition of claim 3, comprising: from about 0.01 to about 0.5%, by weight, of an additional gum-type colloidal polymer. 17. The composition of claim 6, wherein the clay or fine particulate is present in the composition at a concentration of 0.5%, by weight. 18. The composition of claim 3, further comprising a suspended material. 19. The composition of claim 18, wherein said suspended material is selected from: a colloidal metal; a film flake; a film strip; and a combination of two or more thereof. 20. A method of treating or preventing a disease or condition of the oral cavity comprising contacting an oral cavity surface of a subject in need thereof, with the composition of claim 1. 21. (canceled)
1,600
369
15,528,604
1,612
The present disclosure is directed to a method for reducing bacteria in a precipitated calcium carbonate (CaCO 3 ) slurry, the method including: adding water to calcium oxide (CaO) to form Ca(OH) 2 ; treating the Ca(OH) 2 with CO 2 gas to form a slurry including precipitated CaCO 3 ; neutralizing the slurry; and exposing the slurry that was neutralized to ozone in an amount sufficient to reduce bacteria in the precipitated calcium carbonate slurry. Oral care compositions including the precipitated calcium carbonate of the disclosed process are also described.
1. A method for reducing bacteria in a precipitated calcium carbonate (CaCO3) slurry, the method comprising: adding water to calcium oxide (CaO) to form Ca(OH)2: treating the Ca(OH)2 with CO2 gas to form a slurry comprising precipitated CaCO3; neutralizing the slurry; and exposing the slurry that was neutralized to ozone in an amount sufficient to reduce bacteria in the precipitated calcium carbonate slurry. 2. The method of claim 1, wherein the exposing comprises injecting ozone gas into the slurry that was neutralized. 3. The method of claim 1, wherein the method further comprises: mixing the slurry that was neutralized and the ozone to create a mixture; and recirculating the mixture into a tank. 4. The method of claim 3, wherein the method further comprises sampling the slurry after the mixing and before the recirculating to determine a degree of reduction of the bacteria. 5. The method of claim 3, wherein the mixing comprises mixing the slurry and the ozone using a static mixer. 6. The method of claim 1, wherein a log reduction of the bacteria ranges from about a 1 log reduction to about a 5 log reduction. 7. The method of claim 1, wherein a log reduction of the bacteria ranges from about a 4 log reduction to about a 5 log reduction. 8. The method of claim 1, wherein a concentration of ozone in the slurry that was neutralized ranges from about 5 ppm to about 25 ppm. 9. The method of claim 1, wherein a concentration of ozone in the slurry that was neutralized is about 25 ppm. 10. The method of claim 1, wherein the bacteria comprise gram positive bacteria. 11. The method of claim 1, wherein the method further comprises: sampling the slurry that was neutralized before exposing the slurry to ozone; assessing a bioburden before exposing the slurry to ozone; sampling the slurry that was neutralized after exposing the slurry to ozone; and assessing a bioburden after exposing the slurry to ozone. 12. The method of claim 11, wherein the sampling the slurry that was neutralized after exposing the slurry to ozone occurs about 10 seconds to about 40 minutes after the slurry was exposed to zone. 13. The method of claim 11, wherein the sampling the slurry that was neutralized after exposing the slurry to ozone occurs about 3 seconds to about 10 seconds after the slurry was exposed to ozone. 14. The method of claim 1, wherein a pH of the slurry that was neutralized ranges from about 6.9 to about 8.5. 15. The method of claim 1, wherein a temperature of the slurry that was neutralized ranges from about 25° C. to about 47° C. 16. The method of claim 15, wherein a temperature of the slurry that was neutralized ranges from about 28° C. to about 32° C. 17. The method of claim 1, wherein a solid concentration of precipitated CaCO3 in the slurry ranges from about 12% w/w to about 40% w/w. 18. The method of claim 1, wherein the method further comprises drying the slurry to isolate the precipitated CaCO3 and form dried CaCO3. 19. The method of claim 18, wherein the method further comprises combining the dried CaCO3 with an oral care ingredient to form an oral care composition. 20. An oral care composition comprising the dried CaCO3 of claim 18.
The present disclosure is directed to a method for reducing bacteria in a precipitated calcium carbonate (CaCO 3 ) slurry, the method including: adding water to calcium oxide (CaO) to form Ca(OH) 2 ; treating the Ca(OH) 2 with CO 2 gas to form a slurry including precipitated CaCO 3 ; neutralizing the slurry; and exposing the slurry that was neutralized to ozone in an amount sufficient to reduce bacteria in the precipitated calcium carbonate slurry. Oral care compositions including the precipitated calcium carbonate of the disclosed process are also described.1. A method for reducing bacteria in a precipitated calcium carbonate (CaCO3) slurry, the method comprising: adding water to calcium oxide (CaO) to form Ca(OH)2: treating the Ca(OH)2 with CO2 gas to form a slurry comprising precipitated CaCO3; neutralizing the slurry; and exposing the slurry that was neutralized to ozone in an amount sufficient to reduce bacteria in the precipitated calcium carbonate slurry. 2. The method of claim 1, wherein the exposing comprises injecting ozone gas into the slurry that was neutralized. 3. The method of claim 1, wherein the method further comprises: mixing the slurry that was neutralized and the ozone to create a mixture; and recirculating the mixture into a tank. 4. The method of claim 3, wherein the method further comprises sampling the slurry after the mixing and before the recirculating to determine a degree of reduction of the bacteria. 5. The method of claim 3, wherein the mixing comprises mixing the slurry and the ozone using a static mixer. 6. The method of claim 1, wherein a log reduction of the bacteria ranges from about a 1 log reduction to about a 5 log reduction. 7. The method of claim 1, wherein a log reduction of the bacteria ranges from about a 4 log reduction to about a 5 log reduction. 8. The method of claim 1, wherein a concentration of ozone in the slurry that was neutralized ranges from about 5 ppm to about 25 ppm. 9. The method of claim 1, wherein a concentration of ozone in the slurry that was neutralized is about 25 ppm. 10. The method of claim 1, wherein the bacteria comprise gram positive bacteria. 11. The method of claim 1, wherein the method further comprises: sampling the slurry that was neutralized before exposing the slurry to ozone; assessing a bioburden before exposing the slurry to ozone; sampling the slurry that was neutralized after exposing the slurry to ozone; and assessing a bioburden after exposing the slurry to ozone. 12. The method of claim 11, wherein the sampling the slurry that was neutralized after exposing the slurry to ozone occurs about 10 seconds to about 40 minutes after the slurry was exposed to zone. 13. The method of claim 11, wherein the sampling the slurry that was neutralized after exposing the slurry to ozone occurs about 3 seconds to about 10 seconds after the slurry was exposed to ozone. 14. The method of claim 1, wherein a pH of the slurry that was neutralized ranges from about 6.9 to about 8.5. 15. The method of claim 1, wherein a temperature of the slurry that was neutralized ranges from about 25° C. to about 47° C. 16. The method of claim 15, wherein a temperature of the slurry that was neutralized ranges from about 28° C. to about 32° C. 17. The method of claim 1, wherein a solid concentration of precipitated CaCO3 in the slurry ranges from about 12% w/w to about 40% w/w. 18. The method of claim 1, wherein the method further comprises drying the slurry to isolate the precipitated CaCO3 and form dried CaCO3. 19. The method of claim 18, wherein the method further comprises combining the dried CaCO3 with an oral care ingredient to form an oral care composition. 20. An oral care composition comprising the dried CaCO3 of claim 18.
1,600
370
15,288,121
1,651
Biomass (e.g., plant biomass, animal biomass, and municipal waste biomass) is processed to produce useful products, such as fuels. For example, systems can use feedstock materials, such as cellulosic and/or lignocellulosic materials, to proceed ethanol and/or butanol, e.g., by fermentation.
1-18. (canceled) 19. A mixture comprising: an aqueous solution comprising a low molecular weight sugar, a fermenting microorganism, and a lignocellulosic material that has been irradiated with at least 10 Mrad of electron beam radiation, the lignocellulosic material having a BET surface area greater than 0.1 m2/g and a porosity greater than 50%. 20. The mixture of claim 19, wherein the low molecular weight sugar comprises xylose. 21. The mixture of claim 19, wherein the lignocellulosic material has a porosity greater than 70 percent. 22. The mixture of claim 19, wherein the lignocellulosic material has a BET surface area greater than 0.25 m2/g. 23. The mixture of claim 19, wherein the lignocellulosic material has a BET surface area greater than 1.0 m2/g. 24. The mixture of claim 19, wherein the lignocellulosic material is selected from the group consisting of wood, particle board, sawdust, agricultural waste, silage, grasses, rice hulls, bagasse, jute, hemp, flax, bamboo, sisal, abaca, straw, corn cobs, corn stover, switchgrass, alfalfa, hay, coconut hair, seaweed, and mixtures thereof. 25. The mixture of claim 24, wherein the lignocellulosic material comprises corn cobs and/or corn stover. 26. The mixture of claim 19, wherein the lignocellulosic material includes a plurality of saccharide units arranged in a molecular chain, wherein from about 1 out of every 2 to about 1 out of every 250 saccharide units includes a carboxylic acid group, or an ester or salt thereof.
Biomass (e.g., plant biomass, animal biomass, and municipal waste biomass) is processed to produce useful products, such as fuels. For example, systems can use feedstock materials, such as cellulosic and/or lignocellulosic materials, to proceed ethanol and/or butanol, e.g., by fermentation.1-18. (canceled) 19. A mixture comprising: an aqueous solution comprising a low molecular weight sugar, a fermenting microorganism, and a lignocellulosic material that has been irradiated with at least 10 Mrad of electron beam radiation, the lignocellulosic material having a BET surface area greater than 0.1 m2/g and a porosity greater than 50%. 20. The mixture of claim 19, wherein the low molecular weight sugar comprises xylose. 21. The mixture of claim 19, wherein the lignocellulosic material has a porosity greater than 70 percent. 22. The mixture of claim 19, wherein the lignocellulosic material has a BET surface area greater than 0.25 m2/g. 23. The mixture of claim 19, wherein the lignocellulosic material has a BET surface area greater than 1.0 m2/g. 24. The mixture of claim 19, wherein the lignocellulosic material is selected from the group consisting of wood, particle board, sawdust, agricultural waste, silage, grasses, rice hulls, bagasse, jute, hemp, flax, bamboo, sisal, abaca, straw, corn cobs, corn stover, switchgrass, alfalfa, hay, coconut hair, seaweed, and mixtures thereof. 25. The mixture of claim 24, wherein the lignocellulosic material comprises corn cobs and/or corn stover. 26. The mixture of claim 19, wherein the lignocellulosic material includes a plurality of saccharide units arranged in a molecular chain, wherein from about 1 out of every 2 to about 1 out of every 250 saccharide units includes a carboxylic acid group, or an ester or salt thereof.
1,600
371
13,392,596
1,653
The present invention relates to a production method for cryopreserved acellular dermal matrix and to cryopreserved acellular dermal matrix produced thereby, and more specifically it relates to a method in which a cryopreservation agent is made by adding sucrose to basic components consisting of glycerol and a basic solution and in which the resulting solution is used in the cryopreservation of skin tissue from which the cells in the epidermis and dermis have been removed, and relates to cryopreserved acellular dermal matrix produced thereby.
1. A method for preparing a cryopreserved acellular dermal matrix comprising: i) removing epidermis of allograft skin; ii) removing cells in dermis; iii) mixing glycerol, and a basic solution selected from a buffer solution and an animal cell culture medium; iv) dissolving sucrose in the solution to a final concentration of 20 to 40% by weight to obtain a cryoprotectant; v) penetrating the cryoprotectant into the skin from which epidermis and cells in dermis are removed; and vi) freezing the cryoprotectant-penetrated skin in a controlled rate freezer. 2. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the mixing ratio of glycerol and the basic solution is 0.5˜3:9 based on weight. 3. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the buffer solution is selected from PBS (phosphate buffered saline), TBS (Tris-buffered saline) and citric acid buffer. 4. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the animal cell culture medium is selected from MEM (Minimum Essential Media), DMEM (Dulbecco's Modified Eagle Media), RPMI 1640, IMDM (Iscove's Modified Dulbecco's Media), Defined Keratinocyte-SFM (without BPE), Keratinocyte-SFN (with BPE), KnockOut D-MEM, AmnioMAX-II Complete Medium and AmnioMAX-C100 Complete Medium. 5. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the final concentration of sucrose is 30% by weight. 6. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the cryoprotectant is penetrated into the separated skin in a 4° C. low-temperature bath for 6 to 24 hours. 7. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the cryoprotectant-penetrated skin is frozen in a controlled rate freezer at a freezing rate of −1° C. per minute. 8. An autograft substitute comprising the cryopreserved acellular dermal matrix which is prepared by the method according to any one of claims 1 to 7.
The present invention relates to a production method for cryopreserved acellular dermal matrix and to cryopreserved acellular dermal matrix produced thereby, and more specifically it relates to a method in which a cryopreservation agent is made by adding sucrose to basic components consisting of glycerol and a basic solution and in which the resulting solution is used in the cryopreservation of skin tissue from which the cells in the epidermis and dermis have been removed, and relates to cryopreserved acellular dermal matrix produced thereby.1. A method for preparing a cryopreserved acellular dermal matrix comprising: i) removing epidermis of allograft skin; ii) removing cells in dermis; iii) mixing glycerol, and a basic solution selected from a buffer solution and an animal cell culture medium; iv) dissolving sucrose in the solution to a final concentration of 20 to 40% by weight to obtain a cryoprotectant; v) penetrating the cryoprotectant into the skin from which epidermis and cells in dermis are removed; and vi) freezing the cryoprotectant-penetrated skin in a controlled rate freezer. 2. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the mixing ratio of glycerol and the basic solution is 0.5˜3:9 based on weight. 3. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the buffer solution is selected from PBS (phosphate buffered saline), TBS (Tris-buffered saline) and citric acid buffer. 4. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the animal cell culture medium is selected from MEM (Minimum Essential Media), DMEM (Dulbecco's Modified Eagle Media), RPMI 1640, IMDM (Iscove's Modified Dulbecco's Media), Defined Keratinocyte-SFM (without BPE), Keratinocyte-SFN (with BPE), KnockOut D-MEM, AmnioMAX-II Complete Medium and AmnioMAX-C100 Complete Medium. 5. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the final concentration of sucrose is 30% by weight. 6. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the cryoprotectant is penetrated into the separated skin in a 4° C. low-temperature bath for 6 to 24 hours. 7. The method for preparing a cryopreserved acellular dermal matrix according to claim 1, wherein the cryoprotectant-penetrated skin is frozen in a controlled rate freezer at a freezing rate of −1° C. per minute. 8. An autograft substitute comprising the cryopreserved acellular dermal matrix which is prepared by the method according to any one of claims 1 to 7.
1,600
372
12,922,432
1,663
A technique by which the production of plant biomass can be significantly increased is provided. A protein phosphatase 2C gene having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 from the N-terminal side in such order is over-expressed.
1. A plant, in which a gene encoding protein phosphatase 2C having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 in such order from the N-terminal side is over-expressed. 2. The plant according to claim 1, wherein the gene encoding protein phosphatase 2C is at least one gene selected from the group consisting of At1g03590-AtPP2C6-6, At1g16220, At1g79630, At5g01700, At3g02750, At5g36250, At5g26010, At4g32950, At3g16800, At3g05640, At5g27930-AtPP2C6-7, At2g20050, and At3g06270, or a gene functionally equivalent to the gene. 3. The plant according to claim 1, wherein the gene encoding protein phosphatase 2C encodes any one of the following proteins (a) to (c): (a) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48; (b) a protein comprising an amino acid sequence that has a deletion, a substitution, an addition, or an insertion of one or a plurality of amino acids with respect to an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48 and having protein phosphatase 2C activity; and (c) a protein that is encoded by a polynucleotide hybridizing under stringent conditions to a polynucleotide comprising a nucleotide sequence complementary to a nucleotide sequence selected from the group consisting of SEQ ID NOS: 4, 6, 35, 41, and 47 and has protein phosphatase 2C activity. 4. The plant according to claim 2, wherein the functionally equivalent gene is a protein phosphatase 2C gene from an organism other than Arabidopsis thaliana. 5. The plant according to claim 4, wherein an organism other than Arabidopsis thaliana is at least one type of organism selected from the group consisting of rice (Oryza sativa), black cottonwood (Populus trichocarpa), european grape (Vitis vinerfera), Medicago truncatula (Medicago truncatula), alfalfa (Medicago sativa), Physcomitrella patens (Physcomitrella patens), ice plant (Mesembryanthemum crystallinum), Chlamydomonas reinhardtii (Chlamydomonas reinhardtii), corn (Zea mays), rapeseed (Brassica rapa), tomato (Solanum lycopersicum), monkey flower (Mimulus guttatus), and monocellular red alga (Cyanidioschyzon merolae). 6-13. (canceled) 14. A method for increasing the production of biomass and/or seeds, by which a gene encoding protein phosphatase 2C having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 from the N-terminal side in such order is over-expressed by a plant. 15. The method according to claim 14, wherein the gene encoding protein phosphatase 2C is at least one gene selected from the group consisting of At1g03590-AtPP2C6-6, At1g16220, At1g79630, At5g01700, At3g02750, At5g36250, At5g26010, At4g32950, At3g16800, At3g05640, At5g27930-AtPP2C6-7, At2g20050, and At3g06270, or a gene functionally equivalent to the gene. 16. The method according to claim 14, wherein the gene encoding protein phosphatase 2C encodes any one of the following proteins (a) to (c): (a) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48; (b) a protein comprising an amino acid sequence that has a deletion, a substitution, an addition, or an insertion of one or a plurality of amino acids with respect to an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48 and having protein phosphatase 2C activity; and (c) a protein that is encoded by a polynucleotide hybridizing under stringent conditions to a polynucleotide comprising a nucleotide sequence complementary to a nucleotide sequence selected from the group consisting of SEQ ID NOS: 4, 6, 35, 41, and 47 and has protein phosphatase 2C activity. 17. The method according to claim 15, wherein the functionally equivalent gene is a protein phosphatase 2C gene from an organism other than Arabidopsis thaliana. 18. The method according to claim 17, wherein an organism other than Arabidopsis thaliana is at least one type of organism selected from the group consisting of rice (Oryza sativa), black cottonwood (Populus trichocarpa), european grape (Vitis vinerfera), Medicago truncatula (Medicago truncatula), alfalfa (Medicago sativa), Physcomitrella patens (Physcomitrella patens), ice plant (Mesembryanthemum crystallinum), Chlamydomonas reinhardtii (Chlamydomonas reinhardtii), corn (Zea mays), rapeseed (Brassica rapa), tomato (Solanum lycopersicum), monkey flower (Mimulus guttatus), and monocellular red alga (Cyanidioschyzon merolae). 19-26. (canceled) 27. A method for producing a plant, comprising the steps of: preparing a transformed plant in which a gene encoding protein phosphatase 2C having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 from the N-terminal side in such order is over-expressed; and measuring the amount of biomass and/or seeds of a progeny plant of the transformed plant and then selecting a line with significantly improved production of biomass and/or seeds. 28. The production method according to claim 27, wherein the gene encoding protein phosphatase 2C is at least one gene selected from the group consisting of At1g03590-AtPP2C6-6, At1g16220, At1g79630, At5g01700, At3g02750, At5g36250, At5g26010, At4g32950, At3g16800, At3g05640, At5g27930-AtPP2C6-7, At2g20050, and At3g06270, or a gene functionally equivalent to the gene. 29. The production method according to claim 27, wherein the gene encoding protein phosphatase 2C encodes any one of the following proteins (a) to (c): (a) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48; (b) a protein comprising an amino acid sequence that has a deletion, a substitution, an addition, or an insertion of one or a plurality of amino acids with respect to an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48 and having protein phosphatase 2C activity; and (c) a protein that is encoded by a polynucleotide hybridizing under stringent conditions to a polynucleotide comprising a nucleotide sequence complementary to a nucleotide sequence selected from the group consisting of SEQ ID NOS: 4, 6, 35, 41, and 47 and has protein phosphatase 2C activity. 30. The production method according to claim 28, wherein the functionally equivalent gene is a protein phosphatase 2C gene from an organism other than Arabidopsis thaliana. 31. The production method according to claim 30, wherein an organism other than Arabidopsis thaliana is at least one type of organism selected from the group consisting of rice (Oryza sativa), black cottonwood (Populus trichocarpa), european grape (Vitis vinerfera), Medicago truncatula (Medicago truncatula), alfalfa (Medicago sativa), Physcomitrella patens (Physcomitrella patens), ice plant (Mesembryanthemum crystallinum), Chlamydomonas reinhardtii (Chlamydomonas reinhardtii), corn (Zea mays), rapeseed (Brassica rapa), tomato (Solanum lycopersicum), monkey flower (Mimulus guttatus), and monocellular red alga (Cyanidioschyzon merolae). 32-39. (canceled)
A technique by which the production of plant biomass can be significantly increased is provided. A protein phosphatase 2C gene having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 from the N-terminal side in such order is over-expressed.1. A plant, in which a gene encoding protein phosphatase 2C having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 in such order from the N-terminal side is over-expressed. 2. The plant according to claim 1, wherein the gene encoding protein phosphatase 2C is at least one gene selected from the group consisting of At1g03590-AtPP2C6-6, At1g16220, At1g79630, At5g01700, At3g02750, At5g36250, At5g26010, At4g32950, At3g16800, At3g05640, At5g27930-AtPP2C6-7, At2g20050, and At3g06270, or a gene functionally equivalent to the gene. 3. The plant according to claim 1, wherein the gene encoding protein phosphatase 2C encodes any one of the following proteins (a) to (c): (a) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48; (b) a protein comprising an amino acid sequence that has a deletion, a substitution, an addition, or an insertion of one or a plurality of amino acids with respect to an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48 and having protein phosphatase 2C activity; and (c) a protein that is encoded by a polynucleotide hybridizing under stringent conditions to a polynucleotide comprising a nucleotide sequence complementary to a nucleotide sequence selected from the group consisting of SEQ ID NOS: 4, 6, 35, 41, and 47 and has protein phosphatase 2C activity. 4. The plant according to claim 2, wherein the functionally equivalent gene is a protein phosphatase 2C gene from an organism other than Arabidopsis thaliana. 5. The plant according to claim 4, wherein an organism other than Arabidopsis thaliana is at least one type of organism selected from the group consisting of rice (Oryza sativa), black cottonwood (Populus trichocarpa), european grape (Vitis vinerfera), Medicago truncatula (Medicago truncatula), alfalfa (Medicago sativa), Physcomitrella patens (Physcomitrella patens), ice plant (Mesembryanthemum crystallinum), Chlamydomonas reinhardtii (Chlamydomonas reinhardtii), corn (Zea mays), rapeseed (Brassica rapa), tomato (Solanum lycopersicum), monkey flower (Mimulus guttatus), and monocellular red alga (Cyanidioschyzon merolae). 6-13. (canceled) 14. A method for increasing the production of biomass and/or seeds, by which a gene encoding protein phosphatase 2C having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 from the N-terminal side in such order is over-expressed by a plant. 15. The method according to claim 14, wherein the gene encoding protein phosphatase 2C is at least one gene selected from the group consisting of At1g03590-AtPP2C6-6, At1g16220, At1g79630, At5g01700, At3g02750, At5g36250, At5g26010, At4g32950, At3g16800, At3g05640, At5g27930-AtPP2C6-7, At2g20050, and At3g06270, or a gene functionally equivalent to the gene. 16. The method according to claim 14, wherein the gene encoding protein phosphatase 2C encodes any one of the following proteins (a) to (c): (a) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48; (b) a protein comprising an amino acid sequence that has a deletion, a substitution, an addition, or an insertion of one or a plurality of amino acids with respect to an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48 and having protein phosphatase 2C activity; and (c) a protein that is encoded by a polynucleotide hybridizing under stringent conditions to a polynucleotide comprising a nucleotide sequence complementary to a nucleotide sequence selected from the group consisting of SEQ ID NOS: 4, 6, 35, 41, and 47 and has protein phosphatase 2C activity. 17. The method according to claim 15, wherein the functionally equivalent gene is a protein phosphatase 2C gene from an organism other than Arabidopsis thaliana. 18. The method according to claim 17, wherein an organism other than Arabidopsis thaliana is at least one type of organism selected from the group consisting of rice (Oryza sativa), black cottonwood (Populus trichocarpa), european grape (Vitis vinerfera), Medicago truncatula (Medicago truncatula), alfalfa (Medicago sativa), Physcomitrella patens (Physcomitrella patens), ice plant (Mesembryanthemum crystallinum), Chlamydomonas reinhardtii (Chlamydomonas reinhardtii), corn (Zea mays), rapeseed (Brassica rapa), tomato (Solanum lycopersicum), monkey flower (Mimulus guttatus), and monocellular red alga (Cyanidioschyzon merolae). 19-26. (canceled) 27. A method for producing a plant, comprising the steps of: preparing a transformed plant in which a gene encoding protein phosphatase 2C having 3 consensus sequences comprising the amino acid sequences shown in SEQ ID NOS: 1-3 from the N-terminal side in such order is over-expressed; and measuring the amount of biomass and/or seeds of a progeny plant of the transformed plant and then selecting a line with significantly improved production of biomass and/or seeds. 28. The production method according to claim 27, wherein the gene encoding protein phosphatase 2C is at least one gene selected from the group consisting of At1g03590-AtPP2C6-6, At1g16220, At1g79630, At5g01700, At3g02750, At5g36250, At5g26010, At4g32950, At3g16800, At3g05640, At5g27930-AtPP2C6-7, At2g20050, and At3g06270, or a gene functionally equivalent to the gene. 29. The production method according to claim 27, wherein the gene encoding protein phosphatase 2C encodes any one of the following proteins (a) to (c): (a) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48; (b) a protein comprising an amino acid sequence that has a deletion, a substitution, an addition, or an insertion of one or a plurality of amino acids with respect to an amino acid sequence selected from the group consisting of SEQ ID NOS: 5, 7, 36, 42, and 48 and having protein phosphatase 2C activity; and (c) a protein that is encoded by a polynucleotide hybridizing under stringent conditions to a polynucleotide comprising a nucleotide sequence complementary to a nucleotide sequence selected from the group consisting of SEQ ID NOS: 4, 6, 35, 41, and 47 and has protein phosphatase 2C activity. 30. The production method according to claim 28, wherein the functionally equivalent gene is a protein phosphatase 2C gene from an organism other than Arabidopsis thaliana. 31. The production method according to claim 30, wherein an organism other than Arabidopsis thaliana is at least one type of organism selected from the group consisting of rice (Oryza sativa), black cottonwood (Populus trichocarpa), european grape (Vitis vinerfera), Medicago truncatula (Medicago truncatula), alfalfa (Medicago sativa), Physcomitrella patens (Physcomitrella patens), ice plant (Mesembryanthemum crystallinum), Chlamydomonas reinhardtii (Chlamydomonas reinhardtii), corn (Zea mays), rapeseed (Brassica rapa), tomato (Solanum lycopersicum), monkey flower (Mimulus guttatus), and monocellular red alga (Cyanidioschyzon merolae). 32-39. (canceled)
1,600
373
14,057,243
1,644
The invention relates, in part, to antibodies with increased ADCC activity. Methods of producing such antibodies are also provided. The antibodies of the invention are produced in mammary epithelial cells, such as those in a non-human transgenic animal engineered to express and secrete the antibody in its milk. The antibodies or compositions comprising the antibodies can be used to treat disease in which ADCC activity provides a benefit. In one embodiment, therefore, the antibodies or compositions comprising the antibodies can be used to treat cancer, lymphoproliferative disease or autoimmune disease.
1. A method for enhancing the binding of the Fc region of an IgG antibody or IgG antibodies to an FcγRIII receptor, the method comprising: modifying the glycosylation of the IgG antibody or antibodies so that the binding of the Fc region of the antibody or antibodies to the FcγRIII receptor is enhanced, wherein the glycosylation is modified by producing the antibody or antibodies in mammalian mammary epithelial cells. 2. The method of claim 1, wherein the antibody or antibodies are of the isotype IgG1 or IgG2. 3. The method of claim 1, wherein the FcγRIII receptor is on monocytes, macrophages or natural killer cells. 4. The method of claim 1, wherein the mammalian mammary epithelial cells are of a non-human mammal engineered to express the antibody in its milk. 5. The method of claim 1, wherein the antibody is modified such that at least one chain of the antibody does not contain fucose. 6. The method of claim 1, wherein the antibody is modified such that the antibody contains an oligomannose or an additional oligomannose. 7. The method of claim 1, wherein the antibody is modified such that the carbohydrates of the antibody exhibit a high mannose glycosylation pattern. 8. The method of claim 1, wherein the antibody is modified such that at least one chain of the antibody is oligomannose-containing and non-fucosylated. 9. The method of claim 1, wherein the antibody is modified such that the major carbohydrate of the antibody is non-fucosylated. 10. The method of claim 1, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated oligomannose. 11. The method of claim 1, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated Man5. 12. The method of claim 1, wherein the antibody is modified such that less than 40% of the carbohydrates of the antibody contain fucose. 13. The method of claim 1, wherein the antibodies are modified such that at least 30% of the antibodies have at least one oligomannose. 14. The method of claim 1, wherein at least 60% of the carbohydrates of the antibodies are a non-fucosylated oligomannose and less than 40% of the carbohydrates of the antibodies are fucose-containing. 15. The method of claim 1, wherein the mammary epithelial cells are cells from a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 16. The method of claim 15, wherein the transgenic non-human mammal is a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 17. The method of claim 1, wherein the antibody is a chimeric antibody, humanized antibody or fully human antibody. 18. The method of claim 1, wherein the antibody is a full-length antibody. 19. The method of claim 1, wherein the full-length antibody comprises a heavy chain and a light chain. 20. The method of claim 1, wherein the antibody is an antibody fragment. 21. The method of claim 1, wherein the antibody is an anti CD137 antibody. 22. A composition comprising antibodies, wherein the antibodies have enhanced binding of the Fc region to an FcγRIII receptor, wherein the binding of the Fc region to an FcγRIII receptor is enhanced by producing the antibodies in mammalian mammary epithelial cells, wherein the binding of the Fc region to an FcγRIII receptor is enhanced compared to cell-culture derived antibodies, and wherein the antibody is of the isotype IgG. 23. The composition of claim 22, wherein the antibodies are of the isotype IgG1 or IgG2. 24. The composition of claim 22, wherein the FcγRIII receptor is on monocytes, macrophages or natural killer cells. 25. The composition of claim 22, wherein the mammalian mammary epithelial cells are of a non-human mammal engineered to express the antibodies in its milk. 26. The composition of claim 22, wherein the antibody is modified such that at least one chain of the antibody does not contain fucose. 27. The composition of claim 22, wherein the antibody is modified such that the antibody contains an oligomannose or an additional oligomannose. 28. The composition of claim 22, wherein the antibody is modified such that the carbohydrates of the antibody exhibit a high mannose glycosylation pattern. 29. The composition of claim 22, wherein the antibody is modified such that at least one chain of the antibody is oligomannose-containing and non-fucosylated. 30. The composition of claim 22, wherein the antibody is modified such that the major carbohydrate of the antibody is non-fucosylated. 31. The composition of claim 22, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated oligomannose. 32. The composition of claim 22, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated Man5. 33. The composition of claim 22, wherein the antibody is modified such that less than 40% of the carbohydrates of the antibody contain fucose. 34. The composition of claim 22, wherein the antibodies are modified such that at least 30% of the antibodies have at least one oligomannose. 35. The composition of claim 22, wherein at least 60% of the carbohydrates of the antibodies are a non-fucosylated oligomannose and less than 40% of the carbohydrates of the antibodies are fucose-containing. 36. The composition of claim 22, wherein the mammary epithelial cells are cells from a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 37. The composition of claim 36, wherein the transgenic non-human mammal is a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 38. The composition of claim 22, wherein the antibody is a chimeric antibody, humanized antibody or fully human antibody. 39. The composition of claim 22, wherein the antibody is a full-length antibody. 40. The composition of claim 22, wherein the full-length antibody comprises a heavy chain and a light chain. 41. The composition of claim 22, wherein the antibody is an antibody fragment. 42. The composition of claim 22, wherein the antibody is an anti CD137 antibody.
The invention relates, in part, to antibodies with increased ADCC activity. Methods of producing such antibodies are also provided. The antibodies of the invention are produced in mammary epithelial cells, such as those in a non-human transgenic animal engineered to express and secrete the antibody in its milk. The antibodies or compositions comprising the antibodies can be used to treat disease in which ADCC activity provides a benefit. In one embodiment, therefore, the antibodies or compositions comprising the antibodies can be used to treat cancer, lymphoproliferative disease or autoimmune disease.1. A method for enhancing the binding of the Fc region of an IgG antibody or IgG antibodies to an FcγRIII receptor, the method comprising: modifying the glycosylation of the IgG antibody or antibodies so that the binding of the Fc region of the antibody or antibodies to the FcγRIII receptor is enhanced, wherein the glycosylation is modified by producing the antibody or antibodies in mammalian mammary epithelial cells. 2. The method of claim 1, wherein the antibody or antibodies are of the isotype IgG1 or IgG2. 3. The method of claim 1, wherein the FcγRIII receptor is on monocytes, macrophages or natural killer cells. 4. The method of claim 1, wherein the mammalian mammary epithelial cells are of a non-human mammal engineered to express the antibody in its milk. 5. The method of claim 1, wherein the antibody is modified such that at least one chain of the antibody does not contain fucose. 6. The method of claim 1, wherein the antibody is modified such that the antibody contains an oligomannose or an additional oligomannose. 7. The method of claim 1, wherein the antibody is modified such that the carbohydrates of the antibody exhibit a high mannose glycosylation pattern. 8. The method of claim 1, wherein the antibody is modified such that at least one chain of the antibody is oligomannose-containing and non-fucosylated. 9. The method of claim 1, wherein the antibody is modified such that the major carbohydrate of the antibody is non-fucosylated. 10. The method of claim 1, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated oligomannose. 11. The method of claim 1, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated Man5. 12. The method of claim 1, wherein the antibody is modified such that less than 40% of the carbohydrates of the antibody contain fucose. 13. The method of claim 1, wherein the antibodies are modified such that at least 30% of the antibodies have at least one oligomannose. 14. The method of claim 1, wherein at least 60% of the carbohydrates of the antibodies are a non-fucosylated oligomannose and less than 40% of the carbohydrates of the antibodies are fucose-containing. 15. The method of claim 1, wherein the mammary epithelial cells are cells from a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 16. The method of claim 15, wherein the transgenic non-human mammal is a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 17. The method of claim 1, wherein the antibody is a chimeric antibody, humanized antibody or fully human antibody. 18. The method of claim 1, wherein the antibody is a full-length antibody. 19. The method of claim 1, wherein the full-length antibody comprises a heavy chain and a light chain. 20. The method of claim 1, wherein the antibody is an antibody fragment. 21. The method of claim 1, wherein the antibody is an anti CD137 antibody. 22. A composition comprising antibodies, wherein the antibodies have enhanced binding of the Fc region to an FcγRIII receptor, wherein the binding of the Fc region to an FcγRIII receptor is enhanced by producing the antibodies in mammalian mammary epithelial cells, wherein the binding of the Fc region to an FcγRIII receptor is enhanced compared to cell-culture derived antibodies, and wherein the antibody is of the isotype IgG. 23. The composition of claim 22, wherein the antibodies are of the isotype IgG1 or IgG2. 24. The composition of claim 22, wherein the FcγRIII receptor is on monocytes, macrophages or natural killer cells. 25. The composition of claim 22, wherein the mammalian mammary epithelial cells are of a non-human mammal engineered to express the antibodies in its milk. 26. The composition of claim 22, wherein the antibody is modified such that at least one chain of the antibody does not contain fucose. 27. The composition of claim 22, wherein the antibody is modified such that the antibody contains an oligomannose or an additional oligomannose. 28. The composition of claim 22, wherein the antibody is modified such that the carbohydrates of the antibody exhibit a high mannose glycosylation pattern. 29. The composition of claim 22, wherein the antibody is modified such that at least one chain of the antibody is oligomannose-containing and non-fucosylated. 30. The composition of claim 22, wherein the antibody is modified such that the major carbohydrate of the antibody is non-fucosylated. 31. The composition of claim 22, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated oligomannose. 32. The composition of claim 22, wherein the antibody is modified such that the major carbohydrate of the antibody is a non-fucosylated Man5. 33. The composition of claim 22, wherein the antibody is modified such that less than 40% of the carbohydrates of the antibody contain fucose. 34. The composition of claim 22, wherein the antibodies are modified such that at least 30% of the antibodies have at least one oligomannose. 35. The composition of claim 22, wherein at least 60% of the carbohydrates of the antibodies are a non-fucosylated oligomannose and less than 40% of the carbohydrates of the antibodies are fucose-containing. 36. The composition of claim 22, wherein the mammary epithelial cells are cells from a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 37. The composition of claim 36, wherein the transgenic non-human mammal is a goat, sheep, bison, camel, cow, pig, rabbit, buffalo, horse, rat, mouse or llama. 38. The composition of claim 22, wherein the antibody is a chimeric antibody, humanized antibody or fully human antibody. 39. The composition of claim 22, wherein the antibody is a full-length antibody. 40. The composition of claim 22, wherein the full-length antibody comprises a heavy chain and a light chain. 41. The composition of claim 22, wherein the antibody is an antibody fragment. 42. The composition of claim 22, wherein the antibody is an anti CD137 antibody.
1,600
374
13,825,601
1,654
The present invention relates to the GPR119 receptor agonists: 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N,N-dimethylbenzamide; -fluoro-4-(5-fluoro-6-(4-(3-(2-fluoro-propan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N-methylbenzamide; and 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)benzamide, and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful as a single pharmaceutical agent or in combination with one or more additional pharmaceutical agents, such as, a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, or an anti-diabetic peptide analogue, in the treatment of for example, a disorder selected from: a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; obesity; and complications related thereto.
1. A compound selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N,N-dimethylbenzamide; 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N-methylbenzamide; and 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)benzamide. 2. A compound according to claim 1 selected from 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N,N-dimethylbenzamide and pharmaceutically acceptable salts, solvates, and hydrates thereof. 3. A compound according to claim 1 selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N-methylbenzamide; and 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)benzamide. 4. A composition comprising a compound according to claim 2. 5. A composition comprising a compound according to claim 2 and a pharmaceutically acceptable carrier. 6. A method for preparing a composition comprising the step of admixing a compound according to claim 2 and a pharmaceutically acceptable carrier. 7. A composition comprising a compound according to claim 2 and a second pharmaceutical agent. 8. A method for preparing a composition comprising the step of admixing a compound according to claim 2 and a second pharmaceutical agent. 9. A pharmaceutical product selected from: a pharmaceutical composition, a formulation, a dosage form, a combined preparation, a twin pack, and a kit; wherein the product comprises a compound according to claim 2 and a second pharmaceutical agent. 10. A method for increasing the secretion of an incretin in an individual or increasing a blood incretin level in an individual, the method comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to claim 2. 11. A method for the treatment of a disorder selected from: a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; the method comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to claim 2. 12-24. (canceled) 25. A method for increasing the secretion of an incretin in an individual or increasing a blood incretin level in an individual, or for treating a disorder selected from a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; the method comprising administering to said individual in need thereof a compound according to claim 2 in combination with a second pharmaceutical agent. 26. (canceled) 27. A method for increasing the secretion of an incretin in an individual or increasing a blood incretin level in an individual, or for treating a disorder selected from a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; the method comprising prescribing to said individual in need thereof, a compound according to claim 2 in combination with a second pharmaceutical agent. 28-41. (canceled) 42. The method according to claim 58, wherein said compound and said second pharmaceutical agent are administered simultaneously. 43. The method according to claim 10, wherein said incretin is GLP-1. 44. The method according to claim 10, wherein said incretin is GIP. 45. The method according to claim 10, wherein said incretin is PYY. 46. A method of treating a condition characterized by low bone mass selected from: osteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget's disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine, and loss of height, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 47. A method of treating a neurological disorder selected from: stroke and Parkinson's disease, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 48. A method of treating a metabolic-related disorder selected from: diabetes, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glycosuria, metabolic acidosis, a cataract, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP-1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 49. A method of treating type 2 diabetes, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 50. (canceled) 51. The method according to claim 58 wherein said second pharmaceutical agent is selected from: a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, and an anti-diabetic peptide analogue. 52. The method according to claim 58, wherein said second pharmaceutical agent is a DPP-IV inhibitor selected from the following DPP-IV inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3(R)-amino-1-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl)butan-1-one; 1-[2-(3-hydroxyadamant-1-ylamino)acetyl]pyrrolidine-2(S)-carbonitrile; (1S,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-1-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopiperidin-1-yl]-3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidin-1-ylmethyl]benzonitrile; 8-[3(R)-aminopiperidin-1-yl]-7-(2-butynyl)-3-methyl-1-(4-methylquinazolin-2-ylmethyl)xanthine; 1-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid; 4(S)-fluoro-1-[2-[(1R,3S)-3-(1H-1,2,4-triazol-1-ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(S)-carbonitrile; 1-[(2S,3S,11bS)-2-amino-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one; (2S,4S)-2-cyano-4-fluoro-1-[(2-hydroxy-1,1-dimethyl) ethylamino]acetylpyrrolidine; 8-(cis-hexahydro-pyrrolo[3,2-b]pyrrol-1-yl)-3-methyl-7-(3-methyl-but-2-enyl)-1-(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione; 1-((3S,4S)-4-amino-1-(4-(3,3-difluoropyrrolidin-1-yl)-1,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one; (R)-2-((6-(3-aminopiperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)methyl)-4-fluorobenzonitrile; 5-{(S)-2-[2-((S)-2-cyano-pyrrolidin-1-yl)-2-oxo-ethylamino]-propyl}-5-(1H-tetrazol-5-yl)10,11-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide; ((2S,4S)-4-(4-(3-methyl-1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone; (2S,4S)-1-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-1-yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile; 6-[(3R)-3-amino-piperidin-1-yl]-5-(2-chloro-5-fluoro-benzyl)-1,3-dimethyl-1,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione; 2-({6-[(3R)-3-amino-3-methylpiperidin-1-yl]-1,3-dimethyl-2,4-dioxo-1,2,3,4-tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile; (2S)-1-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2-carbonitrile; (2S)-1-{[1,1-dimethyl-3-(4-pyridin-3-yl-imidazol-1-yl)-propylamino]-acetyl}-pyrrolidine-2-carbonitrile; (3,3-difluoropyrrolidin-1-yl)-((2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)methanone; (2S,4S)-1-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile; (2S,5R)-5-ethynyl-1-{N-(4-methyl-1-(4-carboxy-pyridin-2-yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile; and (1S,6R)-3-{[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}-6-(2,4,5-trifluorophenyl)cyclohex-3-en-1-amine. 53. The method according to claim 58, wherein said second pharmaceutical agent is a biguanide selected from the following biguanides and pharmaceutically acceptable salts, solvates, and hydrates thereof: (phenylethyl)biguanide, dimethylbiguanide, butylbiguanide, 1-(p-chlorophenyl)-5-isopropylbiguanide. 54. The method according to claim 58, wherein said second pharmaceutical agent is an alpha-glucosidase inhibitor selected from the following alpha-glucosidase inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof: (2R,3R,4R,5R)-4-((2R,3R,4R,5S,6R)-5-((2R,3R,4S,5S,6R)-3,4-dihydroxy-6-methyl-5-((1S,4R,5S,6S)-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2H-pyran-2-yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal; (2R,3R,4R,5S)-1-(2-hydroxyethyl)-2-(hydroxymethyl)piperidine-3,4,5-triol; (1S,2S,3R,4S,5S)-5-(1,3-dihydroxypropan-2-ylamino)-1-(hydroxymethyl)cyclohexane-1,2,3,4-tetraol. 55. The method according to claim 58, wherein-said or said second pharmaceutical agent is a sulfonylurea selected from the following sulfonylureas and pharmaceutically acceptable salts, solvates, and hydrates thereof: N-(4-(N-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2-carboxamide); 5-chloro-N-(4-(N-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide; 3-ethyl-4-methyl-N-(4-(N-((1r,4r)-4-methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo-2,5-dihydro-1H-pyrrole-1-carboxamide. 56. The method according to claim 58, wherein said second pharmaceutical agent is an SGLT2 inhibitor selected from the following SGLT2 inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof: (2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol; ethyl ((2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-1-isopropyl-5-methyl-1H-pyrazol-3-yloxy)tetrahydro-2H-pyran-2-yl)methyl carbonate; ethyl ((2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(2-(4-methoxybenzyl)phenoxy)tetrahydro-2H-pyran-2-yl)methyl carbonate. 57. The method according to claim 58, wherein said second pharmaceutical agent is a meglitinide selected from the following meglitinides and pharmaceutically acceptable salts, solvates, and hydrates thereof: (S)-2-ethoxy-4-(2-(3-methyl-1-(2-(piperidin-1-yl)phenyl)butylamino)-2-oxoethyl)benzoic acid; (R)-2-((1r,4R)-4-isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid; (S)-2-benzyl-4-((3aR,7aS)-1H-isoindol-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)-4-oxobutanoic acid. 58. A method for type 2 diabetes in an individual, comprising administering to said individual in need thereof, a compound according to claim 2 in combination with a second pharmaceutical agent. 59. The method according to claim 58, wherein said compound and said second pharmaceutical agent are administered separately or sequentially.
The present invention relates to the GPR119 receptor agonists: 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N,N-dimethylbenzamide; -fluoro-4-(5-fluoro-6-(4-(3-(2-fluoro-propan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N-methylbenzamide; and 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)benzamide, and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful as a single pharmaceutical agent or in combination with one or more additional pharmaceutical agents, such as, a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, or an anti-diabetic peptide analogue, in the treatment of for example, a disorder selected from: a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; obesity; and complications related thereto.1. A compound selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N,N-dimethylbenzamide; 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N-methylbenzamide; and 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)benzamide. 2. A compound according to claim 1 selected from 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N,N-dimethylbenzamide and pharmaceutically acceptable salts, solvates, and hydrates thereof. 3. A compound according to claim 1 selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)-N-methylbenzamide; and 3-fluoro-4-(5-fluoro-6-(4-(3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl)piperidin-1-yl)pyrimidin-4-ylamino)benzamide. 4. A composition comprising a compound according to claim 2. 5. A composition comprising a compound according to claim 2 and a pharmaceutically acceptable carrier. 6. A method for preparing a composition comprising the step of admixing a compound according to claim 2 and a pharmaceutically acceptable carrier. 7. A composition comprising a compound according to claim 2 and a second pharmaceutical agent. 8. A method for preparing a composition comprising the step of admixing a compound according to claim 2 and a second pharmaceutical agent. 9. A pharmaceutical product selected from: a pharmaceutical composition, a formulation, a dosage form, a combined preparation, a twin pack, and a kit; wherein the product comprises a compound according to claim 2 and a second pharmaceutical agent. 10. A method for increasing the secretion of an incretin in an individual or increasing a blood incretin level in an individual, the method comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to claim 2. 11. A method for the treatment of a disorder selected from: a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; the method comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to claim 2. 12-24. (canceled) 25. A method for increasing the secretion of an incretin in an individual or increasing a blood incretin level in an individual, or for treating a disorder selected from a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; the method comprising administering to said individual in need thereof a compound according to claim 2 in combination with a second pharmaceutical agent. 26. (canceled) 27. A method for increasing the secretion of an incretin in an individual or increasing a blood incretin level in an individual, or for treating a disorder selected from a GPR119-receptor-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; the method comprising prescribing to said individual in need thereof, a compound according to claim 2 in combination with a second pharmaceutical agent. 28-41. (canceled) 42. The method according to claim 58, wherein said compound and said second pharmaceutical agent are administered simultaneously. 43. The method according to claim 10, wherein said incretin is GLP-1. 44. The method according to claim 10, wherein said incretin is GIP. 45. The method according to claim 10, wherein said incretin is PYY. 46. A method of treating a condition characterized by low bone mass selected from: osteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget's disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine, and loss of height, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 47. A method of treating a neurological disorder selected from: stroke and Parkinson's disease, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 48. A method of treating a metabolic-related disorder selected from: diabetes, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glycosuria, metabolic acidosis, a cataract, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP-1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 49. A method of treating type 2 diabetes, wherein the method comprises administering to an individual in need of such treatment an effective amount of a compound according to claim 2. 50. (canceled) 51. The method according to claim 58 wherein said second pharmaceutical agent is selected from: a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, and an anti-diabetic peptide analogue. 52. The method according to claim 58, wherein said second pharmaceutical agent is a DPP-IV inhibitor selected from the following DPP-IV inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3(R)-amino-1-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl)butan-1-one; 1-[2-(3-hydroxyadamant-1-ylamino)acetyl]pyrrolidine-2(S)-carbonitrile; (1S,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-1-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopiperidin-1-yl]-3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidin-1-ylmethyl]benzonitrile; 8-[3(R)-aminopiperidin-1-yl]-7-(2-butynyl)-3-methyl-1-(4-methylquinazolin-2-ylmethyl)xanthine; 1-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid; 4(S)-fluoro-1-[2-[(1R,3S)-3-(1H-1,2,4-triazol-1-ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(S)-carbonitrile; 1-[(2S,3S,11bS)-2-amino-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one; (2S,4S)-2-cyano-4-fluoro-1-[(2-hydroxy-1,1-dimethyl) ethylamino]acetylpyrrolidine; 8-(cis-hexahydro-pyrrolo[3,2-b]pyrrol-1-yl)-3-methyl-7-(3-methyl-but-2-enyl)-1-(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione; 1-((3S,4S)-4-amino-1-(4-(3,3-difluoropyrrolidin-1-yl)-1,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one; (R)-2-((6-(3-aminopiperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)methyl)-4-fluorobenzonitrile; 5-{(S)-2-[2-((S)-2-cyano-pyrrolidin-1-yl)-2-oxo-ethylamino]-propyl}-5-(1H-tetrazol-5-yl)10,11-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide; ((2S,4S)-4-(4-(3-methyl-1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone; (2S,4S)-1-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-1-yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile; 6-[(3R)-3-amino-piperidin-1-yl]-5-(2-chloro-5-fluoro-benzyl)-1,3-dimethyl-1,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione; 2-({6-[(3R)-3-amino-3-methylpiperidin-1-yl]-1,3-dimethyl-2,4-dioxo-1,2,3,4-tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile; (2S)-1-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2-carbonitrile; (2S)-1-{[1,1-dimethyl-3-(4-pyridin-3-yl-imidazol-1-yl)-propylamino]-acetyl}-pyrrolidine-2-carbonitrile; (3,3-difluoropyrrolidin-1-yl)-((2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)methanone; (2S,4S)-1-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile; (2S,5R)-5-ethynyl-1-{N-(4-methyl-1-(4-carboxy-pyridin-2-yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile; and (1S,6R)-3-{[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}-6-(2,4,5-trifluorophenyl)cyclohex-3-en-1-amine. 53. The method according to claim 58, wherein said second pharmaceutical agent is a biguanide selected from the following biguanides and pharmaceutically acceptable salts, solvates, and hydrates thereof: (phenylethyl)biguanide, dimethylbiguanide, butylbiguanide, 1-(p-chlorophenyl)-5-isopropylbiguanide. 54. The method according to claim 58, wherein said second pharmaceutical agent is an alpha-glucosidase inhibitor selected from the following alpha-glucosidase inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof: (2R,3R,4R,5R)-4-((2R,3R,4R,5S,6R)-5-((2R,3R,4S,5S,6R)-3,4-dihydroxy-6-methyl-5-((1S,4R,5S,6S)-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2H-pyran-2-yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal; (2R,3R,4R,5S)-1-(2-hydroxyethyl)-2-(hydroxymethyl)piperidine-3,4,5-triol; (1S,2S,3R,4S,5S)-5-(1,3-dihydroxypropan-2-ylamino)-1-(hydroxymethyl)cyclohexane-1,2,3,4-tetraol. 55. The method according to claim 58, wherein-said or said second pharmaceutical agent is a sulfonylurea selected from the following sulfonylureas and pharmaceutically acceptable salts, solvates, and hydrates thereof: N-(4-(N-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2-carboxamide); 5-chloro-N-(4-(N-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide; 3-ethyl-4-methyl-N-(4-(N-((1r,4r)-4-methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo-2,5-dihydro-1H-pyrrole-1-carboxamide. 56. The method according to claim 58, wherein said second pharmaceutical agent is an SGLT2 inhibitor selected from the following SGLT2 inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof: (2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol; ethyl ((2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-1-isopropyl-5-methyl-1H-pyrazol-3-yloxy)tetrahydro-2H-pyran-2-yl)methyl carbonate; ethyl ((2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(2-(4-methoxybenzyl)phenoxy)tetrahydro-2H-pyran-2-yl)methyl carbonate. 57. The method according to claim 58, wherein said second pharmaceutical agent is a meglitinide selected from the following meglitinides and pharmaceutically acceptable salts, solvates, and hydrates thereof: (S)-2-ethoxy-4-(2-(3-methyl-1-(2-(piperidin-1-yl)phenyl)butylamino)-2-oxoethyl)benzoic acid; (R)-2-((1r,4R)-4-isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid; (S)-2-benzyl-4-((3aR,7aS)-1H-isoindol-2(3H,3aH,4H,5H,6H,7H,7aH)-yl)-4-oxobutanoic acid. 58. A method for type 2 diabetes in an individual, comprising administering to said individual in need thereof, a compound according to claim 2 in combination with a second pharmaceutical agent. 59. The method according to claim 58, wherein said compound and said second pharmaceutical agent are administered separately or sequentially.
1,600
375
14,424,198
1,613
Chlorhexidine gluconate solubilized in a hydrophobic monoacylglyceride is described. Compositions incorporating such materials, as well as methods of preparing such materials are also described.
1. A composition comprising chlorhexidine gluconate solubilized in a monoacylglyceride, wherein the monoacylglyceride is hydrophobic monoacylglyceride having a hydrophilic-lipophilic balance of no greater than 10 as determined using the HLB Method. 2. The composition of claim 1, comprising at least 5% by weight chlorhexidine gluconate dissolved in the hydrophobic monoacylglyceride based on the combined weight of the CHG and hydrophobic monoacylglyceride. 3. The composition of claim 1, wherein the monoacylglyceride has a solubility parameter of greater than 10 (cal/cm3)1/2. 4. The composition according to claim 1, wherein the monoacylglyceride has a binding energy to chlorhexidine gluconate of at least 25 kilocalories per mole. 5. The composition according to claim 1, wherein the monoacylglyceride comprises two vicinal hydrogen-bonding groups. 6. The composition according to claim 1, wherein the acyl group of the monoacylglyceride is a C8 to C18 acyl group. 7. The composition of claim 6, wherein the monoacylglyceride is selected from the group consisting of glycerol monocaprylate, glycerol monolaurate, glycerol monoisostearate, glycerol monooleate, and combinations thereof. 8. The composition according to claim 1, wherein the composition comprises no greater than 1 part by weight hydrophilic vehicle per 1 part by weight chlorhexidine gluconate. 9. The composition according to claim 1, wherein the composition comprises no greater than 0.1 parts by weight hydrophilic vehicle per 1 part by weight chlorhexidine gluconate. 10. The composition according to claim 1, wherein the composition comprises no greater than 0.1 parts by weight water per 1 part by weight chlorhexidine gluconate. 11. The composition according to claim 1, further comprising a carrier. 12. The composition of claim 11, wherein the carrier comprises a polymer. 13. The composition of claim 11, wherein the carrier comprises an organic solvent. 14. The composition according to claim 1, further comprising a thixotropic agent.
Chlorhexidine gluconate solubilized in a hydrophobic monoacylglyceride is described. Compositions incorporating such materials, as well as methods of preparing such materials are also described.1. A composition comprising chlorhexidine gluconate solubilized in a monoacylglyceride, wherein the monoacylglyceride is hydrophobic monoacylglyceride having a hydrophilic-lipophilic balance of no greater than 10 as determined using the HLB Method. 2. The composition of claim 1, comprising at least 5% by weight chlorhexidine gluconate dissolved in the hydrophobic monoacylglyceride based on the combined weight of the CHG and hydrophobic monoacylglyceride. 3. The composition of claim 1, wherein the monoacylglyceride has a solubility parameter of greater than 10 (cal/cm3)1/2. 4. The composition according to claim 1, wherein the monoacylglyceride has a binding energy to chlorhexidine gluconate of at least 25 kilocalories per mole. 5. The composition according to claim 1, wherein the monoacylglyceride comprises two vicinal hydrogen-bonding groups. 6. The composition according to claim 1, wherein the acyl group of the monoacylglyceride is a C8 to C18 acyl group. 7. The composition of claim 6, wherein the monoacylglyceride is selected from the group consisting of glycerol monocaprylate, glycerol monolaurate, glycerol monoisostearate, glycerol monooleate, and combinations thereof. 8. The composition according to claim 1, wherein the composition comprises no greater than 1 part by weight hydrophilic vehicle per 1 part by weight chlorhexidine gluconate. 9. The composition according to claim 1, wherein the composition comprises no greater than 0.1 parts by weight hydrophilic vehicle per 1 part by weight chlorhexidine gluconate. 10. The composition according to claim 1, wherein the composition comprises no greater than 0.1 parts by weight water per 1 part by weight chlorhexidine gluconate. 11. The composition according to claim 1, further comprising a carrier. 12. The composition of claim 11, wherein the carrier comprises a polymer. 13. The composition of claim 11, wherein the carrier comprises an organic solvent. 14. The composition according to claim 1, further comprising a thixotropic agent.
1,600
376
15,785,769
1,627
The present invention relates to a method for treating a patient suffering from a thermoregulatory dysfunction, especially hot flashes and flushes associated with hormonal changes due to naturally occurring menopause (whether male or female) or due to chemically or surgically induced menopause. The method is also applicable to treating the hot flashes, hot flushes, or night sweats associated with disease states that disrupt normal hormonal regulation of body temperature.
1. A method for treating a patient suffering from a thermoregulatory dysfunction comprising administering to said patient a compound selected from paroxetine, a pharmaceutically acceptable salt thereof, a hydrate or solvate of either, in any polymorphic form, and mixtures thereof; said compound being in an amount, based on the paroxctine moiety, which is at least about 0.1 mg/day up to less than a therapeutically effective antidepressant dosage of paroxetine. 2. The method of claim 1 wherein said compound is administered in an amount of at least 0.5 mg/day. 3. The method of claim 1 wherein said compound is administered in an amount of at least 1 mg/day. 4. The method of claim 1 wherein said compound is administered in an amount of at least 2 mg/day. 5. The method of claim 1 wherein said compound is administered in an amount of at least 4 mg/day. 6. The method of claim 1 wherein said compound is administered in an amount of not more than about 9.5 mg/day. 7. The method of claim 1 wherein said compound is administered in an amount of not more than about 9 mg/day. 8. The method of claim 1 wherein said compound is administered in an amount of not more than about 8.5 mg/day. 9. The method of claim 1 wherein said compound is administered in an amount of not more than about 8 mg/day. 10. The method of claim 1 wherein said compound is administered in an amount selected from 2 mg/day, 2.5 mg/day, 3 mg/day, 3.5 mg/day, 4 mg/day, 4.5 mg/day, 5 mg/day, 5.5 mg/day, 6 mg/day, 6.5 mg/day, 7 mg/day, 7.5 mg/day, 8 mg/day, and 8.5 mg/day. 11. The method of claim 1 wherein said thermoregulatory dysfunction is the result of a condition selected from female menopausal related hormonal changes, male menopausal related hormonal changes, chemically induced hormonal changes, surgically induced hormonal changes, hormonal disruption disease states, and any combination thereof. 12. The method of claim 1 wherein said therrnoregulatory dysfunction is a condition selected from the group consisting of hot flashes, hot flushes, night sweats and combinations thereof.
The present invention relates to a method for treating a patient suffering from a thermoregulatory dysfunction, especially hot flashes and flushes associated with hormonal changes due to naturally occurring menopause (whether male or female) or due to chemically or surgically induced menopause. The method is also applicable to treating the hot flashes, hot flushes, or night sweats associated with disease states that disrupt normal hormonal regulation of body temperature.1. A method for treating a patient suffering from a thermoregulatory dysfunction comprising administering to said patient a compound selected from paroxetine, a pharmaceutically acceptable salt thereof, a hydrate or solvate of either, in any polymorphic form, and mixtures thereof; said compound being in an amount, based on the paroxctine moiety, which is at least about 0.1 mg/day up to less than a therapeutically effective antidepressant dosage of paroxetine. 2. The method of claim 1 wherein said compound is administered in an amount of at least 0.5 mg/day. 3. The method of claim 1 wherein said compound is administered in an amount of at least 1 mg/day. 4. The method of claim 1 wherein said compound is administered in an amount of at least 2 mg/day. 5. The method of claim 1 wherein said compound is administered in an amount of at least 4 mg/day. 6. The method of claim 1 wherein said compound is administered in an amount of not more than about 9.5 mg/day. 7. The method of claim 1 wherein said compound is administered in an amount of not more than about 9 mg/day. 8. The method of claim 1 wherein said compound is administered in an amount of not more than about 8.5 mg/day. 9. The method of claim 1 wherein said compound is administered in an amount of not more than about 8 mg/day. 10. The method of claim 1 wherein said compound is administered in an amount selected from 2 mg/day, 2.5 mg/day, 3 mg/day, 3.5 mg/day, 4 mg/day, 4.5 mg/day, 5 mg/day, 5.5 mg/day, 6 mg/day, 6.5 mg/day, 7 mg/day, 7.5 mg/day, 8 mg/day, and 8.5 mg/day. 11. The method of claim 1 wherein said thermoregulatory dysfunction is the result of a condition selected from female menopausal related hormonal changes, male menopausal related hormonal changes, chemically induced hormonal changes, surgically induced hormonal changes, hormonal disruption disease states, and any combination thereof. 12. The method of claim 1 wherein said therrnoregulatory dysfunction is a condition selected from the group consisting of hot flashes, hot flushes, night sweats and combinations thereof.
1,600
377
15,108,098
1,612
The present invention provides an oral care composition comprising a carboxylic acid of the formula (I) wherein: R 1 is H, or is a substituted or unsubstituted alkyl group; and R 2 is H, OH, or NH 2 ; for use in: (a) selectively promoting, in an oral cavity: growth, metabolic activity or colonization of bacteria that have beneficial effects on oral health, relative to growth, metabolic activity or colonization of pathogenic oral bacteria; (b) selectively promoting, in an oral cavity, biofilm formation by bacteria that have beneficial effects on oral health, relative to biofilm formation by pathogenic oral bacteria; or (c) maintaining and/or re-establishing a healthy oral microflora.
1. An oral care composition comprising a carboxylic acid of the formula: wherein: R1 is H, or is a substituted or unsubstituted alkyl group; and R2 is H, OH, or NH2; for use in: (a) selectively promoting, in an oral cavity: growth, metabolic activity or colonization of bacteria that have beneficial effects on oral health, relative to growth, metabolic activity or colonization of pathogenic oral bacteria; (b) selectively promoting, in an oral cavity, biofilm formation by bacteria that have beneficial effects on oral health, relative to biofilm formation by pathogenic oral bacteria; or (c) maintaining and/or re-establishing a healthy oral microbiota. 2. The oral care composition of claim 1, wherein the carboxylic acid is present in the composition in an amount of from 0.01 weight % to 10 weight %, based on the weight of the oral care composition. 3. The oral care composition of claim 1, wherein the carboxylic acid is present in the composition in an amount of from 5 μmol/ml to 30 μmol/ml. 4. The oral care composition of claim 1, wherein R2 is H. 5. The oral care composition of claim 1, wherein R2 is OH. 6. The oral care composition of claim 1, wherein R2 is NH2. 7. The oral care composition of claim 6, wherein the carboxylic acid is an acidic amino acid. 8. The oral care composition of claim 7, wherein the acidic amino acid is an L-amino acid. 9. The oral care composition of claim 8, wherein the acidic amino acid is aspartic acid or glutamic acid. 10. The oral care composition of claim 1, wherein R1 is H. 11. The oral care composition of claim 1, wherein R1 is an unsubstituted alkyl group. 12. The oral care composition of claim 11, wherein R1 is an unsubstituted C1-C4 alkyl group. 13. The oral care composition of claim 12, wherein the carboxylic acid is L-lactic acid. 14. The oral care composition of claim 1, wherein R1 comprises a saccharide residue. 15. The oral care composition of claim 14, wherein the saccharide residue is a galactopyranose residue. 16. The oral care composition of claim 15, wherein the galactopyranose residue is an α-D-galactopyranose residue. 17. The oral care composition of claim 16, wherein the carboxylic acid is melibionic acid. 18. The oral care composition of claim 1, wherein R1 comprises at least one COOR3 group, wherein R3 is H or C1-4 alkyl. 19. The oral care composition of claim 18, wherein R1 comprises at least one C(O)COOH group. 20. The oral care composition of claim 18, wherein R1 is (CH2)nCOOR3, wherein n is from 1 to 5. 21. The oral care composition of claim 20, wherein n is 1. 22. The oral care composition of claim 21, wherein the carboxylic acid is succinic acid or L-malic acid. 23. The oral care composition of claim 21, wherein the carboxylic acid is monomethyl succinate. 24. The oral care composition of claim 19, wherein R1 is (CH2)mC(O)COOH, wherein m is from 1 to 4. 25. The oral care composition of claim 24, wherein the carboxylic acid is α-keto glutaric acid. 26. The oral care composition of claim 18, wherein R1 comprises two COOH groups. 27. The oral care composition of claim 26, wherein the carboxylic acid is citric acid. 28. The oral care composition of claim 1, wherein the composition is a dentifrice, a toothpaste, a gel, a tooth powder, a mouthwash, a mouthrinse, a lozenge, a tablet, a spray, a gum, or a film. 29. The oral care composition of claim 1, wherein the composition further comprises at least one species of bacteria that has beneficial effects on oral health. 30. The oral care composition of claim 29, wherein the species of bacteria that has beneficial effects on oral health is selected from Streptococcus mitis, Streptococcus salivarius, Streptococcus sanguinis, Actinomyces viscosus, Veillonella parvula, Streptococcus gordonii, Capnocytophaga sputigena, Actinomyces naeslundii and combinations thereof. 31. The oral care composition of claim 1, for use in preventing one or more of gingivitis, periodontitis, peri-implantitis, peri-implant mucositis, necrotizing gingivitis, necrotizing periodontitis and caries. 32. A method of selectively promoting, in an oral cavity of a subject: (a) growth, metabolic activity or colonization of bacteria that have beneficial effects on oral health, relative to growth, metabolic activity or colonization of pathogenic oral bacteria; or (b) biofilm formation by bacteria that have beneficial effects on oral health, relative to biofilm formation by pathogenic oral bacteria; the method comprising contacting an oral cavity of the subject with an oral care composition according to claim 1. 33. A method of maintaining and/or re-establishing a healthy oral microbiota in a subject, the method comprising contacting an oral cavity of the subject with an oral care composition according to claim 1. 34. A method of preventing one or more of gingivitis, periodontitis, peri-implantitis, peri-implant mucositis, necrotizing gingivitis, necrotizing periodontitis and caries in a subject, the method comprising contacting an oral cavity of the subject with an oral care composition according to claim 1. 35-37. (canceled)
The present invention provides an oral care composition comprising a carboxylic acid of the formula (I) wherein: R 1 is H, or is a substituted or unsubstituted alkyl group; and R 2 is H, OH, or NH 2 ; for use in: (a) selectively promoting, in an oral cavity: growth, metabolic activity or colonization of bacteria that have beneficial effects on oral health, relative to growth, metabolic activity or colonization of pathogenic oral bacteria; (b) selectively promoting, in an oral cavity, biofilm formation by bacteria that have beneficial effects on oral health, relative to biofilm formation by pathogenic oral bacteria; or (c) maintaining and/or re-establishing a healthy oral microflora.1. An oral care composition comprising a carboxylic acid of the formula: wherein: R1 is H, or is a substituted or unsubstituted alkyl group; and R2 is H, OH, or NH2; for use in: (a) selectively promoting, in an oral cavity: growth, metabolic activity or colonization of bacteria that have beneficial effects on oral health, relative to growth, metabolic activity or colonization of pathogenic oral bacteria; (b) selectively promoting, in an oral cavity, biofilm formation by bacteria that have beneficial effects on oral health, relative to biofilm formation by pathogenic oral bacteria; or (c) maintaining and/or re-establishing a healthy oral microbiota. 2. The oral care composition of claim 1, wherein the carboxylic acid is present in the composition in an amount of from 0.01 weight % to 10 weight %, based on the weight of the oral care composition. 3. The oral care composition of claim 1, wherein the carboxylic acid is present in the composition in an amount of from 5 μmol/ml to 30 μmol/ml. 4. The oral care composition of claim 1, wherein R2 is H. 5. The oral care composition of claim 1, wherein R2 is OH. 6. The oral care composition of claim 1, wherein R2 is NH2. 7. The oral care composition of claim 6, wherein the carboxylic acid is an acidic amino acid. 8. The oral care composition of claim 7, wherein the acidic amino acid is an L-amino acid. 9. The oral care composition of claim 8, wherein the acidic amino acid is aspartic acid or glutamic acid. 10. The oral care composition of claim 1, wherein R1 is H. 11. The oral care composition of claim 1, wherein R1 is an unsubstituted alkyl group. 12. The oral care composition of claim 11, wherein R1 is an unsubstituted C1-C4 alkyl group. 13. The oral care composition of claim 12, wherein the carboxylic acid is L-lactic acid. 14. The oral care composition of claim 1, wherein R1 comprises a saccharide residue. 15. The oral care composition of claim 14, wherein the saccharide residue is a galactopyranose residue. 16. The oral care composition of claim 15, wherein the galactopyranose residue is an α-D-galactopyranose residue. 17. The oral care composition of claim 16, wherein the carboxylic acid is melibionic acid. 18. The oral care composition of claim 1, wherein R1 comprises at least one COOR3 group, wherein R3 is H or C1-4 alkyl. 19. The oral care composition of claim 18, wherein R1 comprises at least one C(O)COOH group. 20. The oral care composition of claim 18, wherein R1 is (CH2)nCOOR3, wherein n is from 1 to 5. 21. The oral care composition of claim 20, wherein n is 1. 22. The oral care composition of claim 21, wherein the carboxylic acid is succinic acid or L-malic acid. 23. The oral care composition of claim 21, wherein the carboxylic acid is monomethyl succinate. 24. The oral care composition of claim 19, wherein R1 is (CH2)mC(O)COOH, wherein m is from 1 to 4. 25. The oral care composition of claim 24, wherein the carboxylic acid is α-keto glutaric acid. 26. The oral care composition of claim 18, wherein R1 comprises two COOH groups. 27. The oral care composition of claim 26, wherein the carboxylic acid is citric acid. 28. The oral care composition of claim 1, wherein the composition is a dentifrice, a toothpaste, a gel, a tooth powder, a mouthwash, a mouthrinse, a lozenge, a tablet, a spray, a gum, or a film. 29. The oral care composition of claim 1, wherein the composition further comprises at least one species of bacteria that has beneficial effects on oral health. 30. The oral care composition of claim 29, wherein the species of bacteria that has beneficial effects on oral health is selected from Streptococcus mitis, Streptococcus salivarius, Streptococcus sanguinis, Actinomyces viscosus, Veillonella parvula, Streptococcus gordonii, Capnocytophaga sputigena, Actinomyces naeslundii and combinations thereof. 31. The oral care composition of claim 1, for use in preventing one or more of gingivitis, periodontitis, peri-implantitis, peri-implant mucositis, necrotizing gingivitis, necrotizing periodontitis and caries. 32. A method of selectively promoting, in an oral cavity of a subject: (a) growth, metabolic activity or colonization of bacteria that have beneficial effects on oral health, relative to growth, metabolic activity or colonization of pathogenic oral bacteria; or (b) biofilm formation by bacteria that have beneficial effects on oral health, relative to biofilm formation by pathogenic oral bacteria; the method comprising contacting an oral cavity of the subject with an oral care composition according to claim 1. 33. A method of maintaining and/or re-establishing a healthy oral microbiota in a subject, the method comprising contacting an oral cavity of the subject with an oral care composition according to claim 1. 34. A method of preventing one or more of gingivitis, periodontitis, peri-implantitis, peri-implant mucositis, necrotizing gingivitis, necrotizing periodontitis and caries in a subject, the method comprising contacting an oral cavity of the subject with an oral care composition according to claim 1. 35-37. (canceled)
1,600
378
13,381,202
1,662
The invention relates to methods of producing a desired phenotype in a plant by manipulation of gene expression within the plant. The method relates to means to increase expression level of a bHLH subgroup 1b gene expression or activity, wherein a desired phenotype such as increased heat tolerance relative to a wild type control plant following heat stress results in reduced flower abortion and increased yield. Included are plants produced by said methods. The invention also relates to nucleic acid sequences and constructs useful such methods and methods of generating and isolating plants having increased expression of a bHLH subgroup 1b expression or activity.
1. A method of producing a transgenic plant, comprising transforming a plant, a plant tissue culture, or a plant cell with a vector comprising a nucleic acid construct that increases the expression or activity of a bHLH subgroup 1b gene to obtain a plant, tissue culture or a plant cell with increased bHLH expression or activity and growing said plant or regenerating a plant from said plant tissue culture or plant cell, wherein a plant having increased heat stress tolerance relative to a wild type control is produced. 2. The method of claim 1, wherein said nucleic acid construct comprises a sequence of a nucleic acid sequence encoding a bHLH subgroup 1b polypeptide. 3. The method of claim 1, wherein said nucleic acid construct comprises a constitutive promoter, an inducible promoter or a tissue specific promoter. 4. The method of claim 3, wherein said tissue specific promoter is a root promoter. 5. The plant produced by the method of claim 1. 6. A transgenic seed produced by the transgenic plant claim 5, wherein said seed produces a plant having increased heat stress tolerance relative to a wild type control. 7. A plant having a non-naturally occurring mutation in bHLH gene, wherein said plant has increased bHLH subgroup 1b gene expression or activity and said plant has increased heat stress tolerance relative to a wild type control. 8. A seed produced by the plant of claim 7, wherein said seed produces plant having increased heat stress tolerance relative to a wild type control.
The invention relates to methods of producing a desired phenotype in a plant by manipulation of gene expression within the plant. The method relates to means to increase expression level of a bHLH subgroup 1b gene expression or activity, wherein a desired phenotype such as increased heat tolerance relative to a wild type control plant following heat stress results in reduced flower abortion and increased yield. Included are plants produced by said methods. The invention also relates to nucleic acid sequences and constructs useful such methods and methods of generating and isolating plants having increased expression of a bHLH subgroup 1b expression or activity.1. A method of producing a transgenic plant, comprising transforming a plant, a plant tissue culture, or a plant cell with a vector comprising a nucleic acid construct that increases the expression or activity of a bHLH subgroup 1b gene to obtain a plant, tissue culture or a plant cell with increased bHLH expression or activity and growing said plant or regenerating a plant from said plant tissue culture or plant cell, wherein a plant having increased heat stress tolerance relative to a wild type control is produced. 2. The method of claim 1, wherein said nucleic acid construct comprises a sequence of a nucleic acid sequence encoding a bHLH subgroup 1b polypeptide. 3. The method of claim 1, wherein said nucleic acid construct comprises a constitutive promoter, an inducible promoter or a tissue specific promoter. 4. The method of claim 3, wherein said tissue specific promoter is a root promoter. 5. The plant produced by the method of claim 1. 6. A transgenic seed produced by the transgenic plant claim 5, wherein said seed produces a plant having increased heat stress tolerance relative to a wild type control. 7. A plant having a non-naturally occurring mutation in bHLH gene, wherein said plant has increased bHLH subgroup 1b gene expression or activity and said plant has increased heat stress tolerance relative to a wild type control. 8. A seed produced by the plant of claim 7, wherein said seed produces plant having increased heat stress tolerance relative to a wild type control.
1,600
379
14,127,606
1,655
The present invention generally relates to the prevention or treatment of disorders related to oxidative stress. For example, the present invention provides a composition for use in the prevention or treatment of oxidative stress related disorders under post-operative conditions. One embodiment of the present invention is a composition comprising DHA and EPA as active ingredients for use in the treatment or prevention of oxidative stress and/or related disorders.
1. A method for the treatment, prevention or alleviation of oxidative stress and/or related disorders induced by ischemia-reperfusion comprising administering a composition comprising DHA and EPA as active ingredients to a patient in need of same. 2. Method in accordance with claim wherein the composition comprises EPA and DHA in a weight ratio of about 2:1 to 1:1. 3. Method in accordance with claim 1, wherein DHA and EPA are provided from a lipid source comprising marine oils. 4. Method in accordance with claim al, wherein the composition comprises a lipid source selected from the group consisting of cocoa butter, soybean oil, fish oil and sunflower oil. 5. Method in accordance with claim 4, wherein the lipid source about 5-10 weight-% cocoa butter, about 45-55 weight-% soybean oil, about 15-25 weight-% fish oil and about 20-25 weight-% sunflower oil. 6. Method in accordance with claim 3, wherein the lipid source comprises about 18-22 weight-% saturated fatty acids, about 20-25 weight-% monounsaturated fatty acids and about 50-55 weight-% polyunsaturated fatty acids. 7. Method in accordance with claim 1, wherein the oxidative stress related disorder is linked to the presence of free radicals in a body. 8. Method in accordance with claim 1 for the treatment or prevention of oxidative damage to an organ selected from the group consisting of intestine, liver, lung, heart, kidney, and/or skin. 9. Method in accordance with claim 1, wherein the oxidative stress related disorder is selected from the group consisting of an inflammatory response to oxidative stress, retinal degeneration, mitohormesis, atherosclerosis, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), Friedreich' s ataxia, tardive dyskynesia, brain injuries, skin photoaging, reperfusion injury or stroke, myocardial infarction, hypertension, heart failure, epilepsy, hyperhomocysteinemia, physiological senescence, sepsis, stress following organ transplantation and combinations thereof. 10. Method in accordance with claim 1 to limit intestinal damages following ischemia-reperfusion (IR). 11. Method in accordance with claim 1, wherein EPA and DHA are provided in a daily dose of at least 400 mg. 12. Method in accordance with claim 1, wherein the composition is to be administered daily over a period of at least 3 days. 13. Method in accordance with claim 1, wherein the patient is a human or pet. 14. Method in accordance with claim 1, wherein the composition to be administered is a treatment selected from the group consisting of topically, orally, enterally and parenterally. 15. Method in accordance with claim 1, wherein the composition is selected from the group consisting of a food product, a pet food product, a nutraceutical, a drink, a food supplement, a powdered nutritional formula to be reconstituted in milk or water, and a medicament.
The present invention generally relates to the prevention or treatment of disorders related to oxidative stress. For example, the present invention provides a composition for use in the prevention or treatment of oxidative stress related disorders under post-operative conditions. One embodiment of the present invention is a composition comprising DHA and EPA as active ingredients for use in the treatment or prevention of oxidative stress and/or related disorders.1. A method for the treatment, prevention or alleviation of oxidative stress and/or related disorders induced by ischemia-reperfusion comprising administering a composition comprising DHA and EPA as active ingredients to a patient in need of same. 2. Method in accordance with claim wherein the composition comprises EPA and DHA in a weight ratio of about 2:1 to 1:1. 3. Method in accordance with claim 1, wherein DHA and EPA are provided from a lipid source comprising marine oils. 4. Method in accordance with claim al, wherein the composition comprises a lipid source selected from the group consisting of cocoa butter, soybean oil, fish oil and sunflower oil. 5. Method in accordance with claim 4, wherein the lipid source about 5-10 weight-% cocoa butter, about 45-55 weight-% soybean oil, about 15-25 weight-% fish oil and about 20-25 weight-% sunflower oil. 6. Method in accordance with claim 3, wherein the lipid source comprises about 18-22 weight-% saturated fatty acids, about 20-25 weight-% monounsaturated fatty acids and about 50-55 weight-% polyunsaturated fatty acids. 7. Method in accordance with claim 1, wherein the oxidative stress related disorder is linked to the presence of free radicals in a body. 8. Method in accordance with claim 1 for the treatment or prevention of oxidative damage to an organ selected from the group consisting of intestine, liver, lung, heart, kidney, and/or skin. 9. Method in accordance with claim 1, wherein the oxidative stress related disorder is selected from the group consisting of an inflammatory response to oxidative stress, retinal degeneration, mitohormesis, atherosclerosis, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), Friedreich' s ataxia, tardive dyskynesia, brain injuries, skin photoaging, reperfusion injury or stroke, myocardial infarction, hypertension, heart failure, epilepsy, hyperhomocysteinemia, physiological senescence, sepsis, stress following organ transplantation and combinations thereof. 10. Method in accordance with claim 1 to limit intestinal damages following ischemia-reperfusion (IR). 11. Method in accordance with claim 1, wherein EPA and DHA are provided in a daily dose of at least 400 mg. 12. Method in accordance with claim 1, wherein the composition is to be administered daily over a period of at least 3 days. 13. Method in accordance with claim 1, wherein the patient is a human or pet. 14. Method in accordance with claim 1, wherein the composition to be administered is a treatment selected from the group consisting of topically, orally, enterally and parenterally. 15. Method in accordance with claim 1, wherein the composition is selected from the group consisting of a food product, a pet food product, a nutraceutical, a drink, a food supplement, a powdered nutritional formula to be reconstituted in milk or water, and a medicament.
1,600
380
15,034,698
1,617
The present invention relates to a composition comprising a polymeric elastomeric phase and an odoriferous active compound. In particular the present invention relates to a composition comprising a polymeric elastomeric phase and an odoriferous active compound and releasing said odoriferous active compound over a given period of time. The present invention concerns also objects comprising a polymeric elastomeric phase and an odoriferous active compound and releasing said odoriferous active compound over a given period of time.
1. A composition comprising an elastomeric phase of macromolecular sequences having a flexible nature with a glass transition temperature of less than 20° C. and at least one active ingredient as odoriferous active compound which is an organic molecule having a molecular weight of at least 16 g/mol and an odour threshold value in air of at least 0.5 ppb. 2. A composition according to claim 1, wherein said elastomeric phase of macromolecular sequences is formed from blocks of a block copolymer. 3. A composition according to claim 2, wherein said block copolymer does not comprise any semicrystalline or crystalline blocks. 4. A composition according to claim 1, wherein said elastomeric phase of macromolecular sequences is formed from blocks of an acrylic block copolymer. 5. A composition according to claim 4, wherein said acrylic block copolymer is havinghas a general formula (A)nB in which: n is an integer of greater than or equal to 1, A is: an acrylic or methacrylic homo- or copolymer having a Tg of greater than 50° C., or polystyrene, or an acrylic/styrene or methacrylic/styrene copolymer. B is an acrylic or methacrylic homo- or copolymer having a Tg of less than 20° C. 6. A composition according to claim 1, wherein the macromolecular sequences of said elastomeric phase have a weight average molecular weight of between 10,000 g/mol and 500,000 g/mol. 7. A composition according to claim 1, wherein the at least one active ingredient as odoriferous active compound is present from 1 ppm to 10 wt %. 8. A composition according to claim 1, wherein the elastomeric phase of the composition makes up at least 1 wt % of the composition. 9. A composition according to claim 1, wherein the composition comprises a further polymer P. 10. A composition according to claim 9, wherein said further polymer P is a thermoplastic polymer. 11. A method of preparing a composition according to claim 1 comprising blending the elastomeric phase of macromolecular sequences and the at least one active ingredient as odoriferous active compound. 12. A method of preparing a composition according to claim 1, comprising the steps of mixing the at least one active ingredient as odoriferous active compound with at least one monomer and macromolecular sequences having a flexible nature followed by a polymerisation step. 13. An article comprising a composition according to claim 1. 14. An article according to claim 13 in form of a sheet, block, film, tube or profiled element. 15. for a method for making an article, comprising using a composition according to claim 1. 16. A composition according to claim 5, wherein A is a homo- or copolymer comprised of one or more monomers selected from the group consisting of methyl methacrylate (MMA), phenyl methacrylate, benzyl methacrylate and isobornyl methacrylate. 17. A composition according to claim 5, wherein A is selected from the group consisting of PMMA and PMMA modified with acrylic or methacrylic comonomers. 18. A composition according to claim 5, wherein B is an acrylic or methacrylic homo- or copolymer comprising monomers selected from the group consisting of methyl acrylate, ethyl acrylate, butyl acrylate (BuA), ethylhexyl acrylate and butyl methacrylate. 19. A composition according to claim 5, wherein B is an acrylic or methacrylic homo- or copolymer comprising butyl acrylate (BuA). 20. A composition according to claim 18, wherein said monomers make up at least 20 wt % of B.
The present invention relates to a composition comprising a polymeric elastomeric phase and an odoriferous active compound. In particular the present invention relates to a composition comprising a polymeric elastomeric phase and an odoriferous active compound and releasing said odoriferous active compound over a given period of time. The present invention concerns also objects comprising a polymeric elastomeric phase and an odoriferous active compound and releasing said odoriferous active compound over a given period of time.1. A composition comprising an elastomeric phase of macromolecular sequences having a flexible nature with a glass transition temperature of less than 20° C. and at least one active ingredient as odoriferous active compound which is an organic molecule having a molecular weight of at least 16 g/mol and an odour threshold value in air of at least 0.5 ppb. 2. A composition according to claim 1, wherein said elastomeric phase of macromolecular sequences is formed from blocks of a block copolymer. 3. A composition according to claim 2, wherein said block copolymer does not comprise any semicrystalline or crystalline blocks. 4. A composition according to claim 1, wherein said elastomeric phase of macromolecular sequences is formed from blocks of an acrylic block copolymer. 5. A composition according to claim 4, wherein said acrylic block copolymer is havinghas a general formula (A)nB in which: n is an integer of greater than or equal to 1, A is: an acrylic or methacrylic homo- or copolymer having a Tg of greater than 50° C., or polystyrene, or an acrylic/styrene or methacrylic/styrene copolymer. B is an acrylic or methacrylic homo- or copolymer having a Tg of less than 20° C. 6. A composition according to claim 1, wherein the macromolecular sequences of said elastomeric phase have a weight average molecular weight of between 10,000 g/mol and 500,000 g/mol. 7. A composition according to claim 1, wherein the at least one active ingredient as odoriferous active compound is present from 1 ppm to 10 wt %. 8. A composition according to claim 1, wherein the elastomeric phase of the composition makes up at least 1 wt % of the composition. 9. A composition according to claim 1, wherein the composition comprises a further polymer P. 10. A composition according to claim 9, wherein said further polymer P is a thermoplastic polymer. 11. A method of preparing a composition according to claim 1 comprising blending the elastomeric phase of macromolecular sequences and the at least one active ingredient as odoriferous active compound. 12. A method of preparing a composition according to claim 1, comprising the steps of mixing the at least one active ingredient as odoriferous active compound with at least one monomer and macromolecular sequences having a flexible nature followed by a polymerisation step. 13. An article comprising a composition according to claim 1. 14. An article according to claim 13 in form of a sheet, block, film, tube or profiled element. 15. for a method for making an article, comprising using a composition according to claim 1. 16. A composition according to claim 5, wherein A is a homo- or copolymer comprised of one or more monomers selected from the group consisting of methyl methacrylate (MMA), phenyl methacrylate, benzyl methacrylate and isobornyl methacrylate. 17. A composition according to claim 5, wherein A is selected from the group consisting of PMMA and PMMA modified with acrylic or methacrylic comonomers. 18. A composition according to claim 5, wherein B is an acrylic or methacrylic homo- or copolymer comprising monomers selected from the group consisting of methyl acrylate, ethyl acrylate, butyl acrylate (BuA), ethylhexyl acrylate and butyl methacrylate. 19. A composition according to claim 5, wherein B is an acrylic or methacrylic homo- or copolymer comprising butyl acrylate (BuA). 20. A composition according to claim 18, wherein said monomers make up at least 20 wt % of B.
1,600
381
14,512,071
1,611
There is provided herein an organofunctional polysiloxanes comprising hydroxyl polyester groups made by reaction of epoxy functional polyorganosiloxanes and oligmeric polyesters based on polyhydroxy carboxylic acids. There is also provided methods for making the organofunctional polysiloxanes and agricultural, coating, personal care and home care applications containing the organofunctional polysiloxanes.
1. A polysiloxane compound having the general formula (I): [MaDbD*cTdQe]f  (I) wherein M=R1R2R3SiO1/2; D=R4R5SiO2/2; D*=R6R7SiO2/2; T=R8SiO3/2; Q=SiO4/2; with a=1-10 b=0-1000 c=0-1000 d=0-1 e=0-1 f=1-10 wherein R1, R2, R3, R4, R5, R6 and R8 are each independently selected from the group consisting of monovalent hydrocarbon groups having from 1 to 8 carbon atoms, and an aryl or alkaryl hydrocarbon group of from 6 to 22 carbon atoms, or R7; R7 is selected from the group consisting of R9, R10 and R11, wherein R9 is selected from the group consisting of —Z-(A-E1)y, —Z-E2 and —Z—NH—C(O)—R12, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C20 hydrocarbon residue which can comprise one or more groups selected from —O—, —NH—,  and can be substituted by one or more OH groups, A is a bivalent residue selected from the group consisting of E1 is selected from the group consisting of E2 and E3 wherein E2=—O—C(O)—R12, wherein R12 is a dendrimer like branched hydrocarbon residue with up to 100 carbon atoms, which can comprise one or more groups selected from —O—, —C(O)—, and is substituted by one or more OH groups, wherein E2 is defined above, and x=1-4, y=1 or 2 R10 is selected from the group consisting of —Z-(A-E4)y′, —Z-E5 and —Z—NH—C(O)—R13, wherein Z and A are defined above, E4 is selected from the group consisting of E5 and E6 wherein E5=—O—C(O)—R13, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 9 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR14—, —C(O)—, and is substituted by one or more OH groups, wherein R14 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E5 is defined above, and x′=1-4, y′=1 or 2 R11 is selected from the group consisting of —Z-(A-E7)y, —Z-E8 and —Z—NH—C(O)—R15, wherein Z and A are defined above, E7 is selected from the group consisting of E8 and E9 wherein E8=—O—C(O)—R15, wherein R15 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with 10 to 50 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR16—, —C(O)—,)-, and is optionally substituted by one or more OH groups, wherein R16 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E8 is defined above, and x″=1-4, y″=1 or 2. 2. The polysiloxane compound according to claim 1, comprising structural elements selected from the following structures: wherein R17 is C1 to C22-alkyl, fluoro-substituted C1 to C22-alkyl or aryl, and g=0-600, wherein the groups R17 can be the same or different and are selected from C1 to C22-alkyl, fluoro-substituted C1 to C22-alkyl and aryl, and h=0-700, wherein R17 is as defined above, and i=0-10, wherein R7 is as defined above, and j=0-10, wherein R7 and R17 are as defined above, and k=0-30, wherein R17 is as defined above, and m=0-30, wherein n=0-10, g+h+i+j+k+m+n=12-1000. 3. The polysiloxane compound according to claim 2, comprising structural elements selected from the following structures: wherein R9 and R17 are defined above, and g1=0-300, wherein R10 and R17 are defined above, and g2=0-300, wherein R11 and R17 are defined above, and g3=0-300, wherein R9 is defined above, and j1=0-10, wherein R10 is defined above, and j2=0-10, wherein R11 is defined above, and j3=0-10, wherein R9 and R17 are defined above, and k1=0-15, wherein R10 and R17 are defined above, and k2=0-15, wherein R11 and R17 are defined above, and k3=0-15, and g1+g2+g3+h+i+j1+j2+j3+k1+k2+k3+m+n=12 to 1000. 4. The polysiloxane compound according to claim 2, wherein R17 is methyl or h=3-500 or i=0 or m=1-6 or n=0. 5. The polysiloxane compound according to claim 3, wherein R17 is methyl or g1=2-200 or g2=0-200 or g3=0-200 or j1=0 or j2=0 or j3=0 or k1=0-2 or k2=0-2 or k3=0-2 or g1+g2+g3+h+i+j1+j2+j3+k1+k2+k3+m+n=15 to 400 or h+k1=2-1000. 6. The polysiloxane compound according to claim 1, wherein the molar ratio of R9 to R17 is 10:1 to 1:10. 7. The polysiloxane compound according to claim 1, wherein the polysiloxane compound either do not contain hydrophilic residues R10 and/or lipophilic residues R11 or the molar ratio of the siloxy units comprising the dendrimer residue R9 to the siloxy units comprising hydrophilic residues R10 and lipophilic residues R11 in the polysiloxane compound is 1:0.01 to 1:100. 8. The polysiloxane compound according to claim 1, wherein the molar ratio of R9 to R10 and R11 is 1:0.1 to 1:10. 9. The polysiloxane compound according to claim 1, wherein R17 is C1 to C10-alkyl, optionally substituted with 1 to 13 fluoro atoms, and aryl. 10. The polysiloxane compound according to claim 1, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C10 hydrocarbon residue, which can comprise —O— groups and can be substituted by one or more OH groups. 11. The polysiloxane compound according to claim 1, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C6 hydrocarbon residue which can comprise one or more groups selected from —O—, —NH—, and can be substituted by one or more OH groups. 12. The polysiloxane compound according to claim 1, wherein R12 is dendrimer like branched hydrocarbon residue with up to 70 carbon atoms, which can comprise one or more groups selected from —O—, —C(O)—, and is substituted by one or more OH groups. 13. The polysiloxane compound according to claim 1, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 4 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR13—, —C(O)—, and is substituted by one or more OH groups, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms. 14. The polysiloxane compound according to claim 1, wherein R15 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with 10 to 30 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR13—, —C(O)—,)-, and is optionally substituted by one or more OH groups, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms. 15. The polysiloxane compound according to claim 1, wherein y=1 or x=1. 16. The polysiloxane compound according to claim 1 wherein R12 is wherein R19═R17 or H, w=1-3, R18═H or provided that the total number of carbon atoms in the dendrimer like residue R12 is 5 to 70 and at least one ester bond is present in the residue structure. 17. The polysiloxane compound according to claim 16 wherein R19 is CH3, and w=1. 18. The polysiloxane compound according to claim 16 wherein the number of ester bonds present in the residue structure is 1 to 14. 19. The polysiloxane compound according to claim 1 wherein Z═—CH2CH2CH2—O—CH2—, —CH2CH2CH2CH2—, —CH═CH2CH2—, —CH═CH2CH2CH2—, p=1 to 4, * marks a bond to the silicon atom in each case. 20. The polysiloxane compound according to claim 1 wherein —Z-A- is selected from the cyclic structures which are derived from cyclic epoxides. 21. The polysiloxane compound according to claim 1 wherein cyclic epoxides are selected from the group consisting of 22. A process for the production of the polysiloxane compound having the general formula (I): [MaDbD*cTdQe]f  (I) wherein M=R1R2R3SiO1/2; D=R4R5SiO2/2; D*=R6R7SiO2/2; T=R8SiO3/2; Q=SiO4/2; with a=1-10 b=0-1000 c=0-1000 d=0-1 e=0-1 f=1-10 wherein R1, R2, R3, R4, R5, R6 and R8 are each independently selected from the group consisting of monovalent hydrocarbon groups having from 1 to 8 carbon atoms, and an aryl or alkaryl hydrocarbon group of from 6 to 22 carbon atoms, or R7; R7 is selected from the group consisting of R9, R10 and R11, wherein R9 is selected from the group consisting of —Z-(A-E1)y, —Z-E2 and —Z—NH—C(O)—R12, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C20 hydrocarbon residue which can comprise one or more groups selected from —O—, —NH—,  and can be substituted by one or more OH groups, A is a bivalent residue selected from the group consisting of E1 is selected from the group consisting of E2 and E3 wherein E2=—O—C(O)—R12, wherein R12 is a dendrimer like branched hydrocarbon residue with up to 100 carbon atoms, which can comprise one or more groups selected from —O—, —C(O)—, and is substituted by one or more OH groups, wherein E2 is defined above, and x=1-4, y=1 or 2 R10 is selected from the group consisting of —Z-(A-E4)y′, —Z-E5 and —Z—NH—C(O)—R13, wherein Z and A are defined above, E4 is selected from the group consisting of E5 and E6 wherein E5=—O—C(O)—R13, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 9 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR14—, —C(O)—, and is substituted by one or more OH groups, wherein R14 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E5 is defined above, and x′=1-4, y′=1 or 2 R11 is selected from the group consisting of —Z-(A-E7)y, —Z-E8 and —Z—NH—C(O)—R15, wherein Z and A are defined above, E7 is selected from the group consisting of E8 and E9 wherein E8=—O—C(O)—R15, wherein R15 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with 10 to 50 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR16—, —C(O)—,)-, and is optionally substituted by one or more OH groups, wherein R16 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E8 is defined above, and x″=1-4, y″=1 or 2 is characterised in that an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, optionally at the same time or subsequently with other hydrophilic and/or hydrophobic carboxylic acids or partially esterified carboxylic anhydrides and, optionally, subsequently with primary or secondary amines. 23. The process of claim 22 wherein the process comprising the steps of: a) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, b) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, at the same time or subsequently with other hydrophilic and/or hydrophobic carboxylic acids or partially esterified carboxylic anhydrides, c) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, and subsequently with primary or secondary amines, d) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, at the same time or subsequently with other hydrophilic and/or hydrophobic carboxylic acids or partially esterified carboxylic anhydrides and subsequently with primary or secondary amines, and that for introducing the dendrimeric element R12 (a) preferably monocarboxy acid functionalized polyhydroxylated esters of the following structure are reacted with the epoxy functionalized silicones wherein R18 and R19 as defined above. 24. The process of claim 23 wherein R18 is H, w is 1 and R19 is CH3. 25. The process of claim 22 which is further characterized in that for introducing the dendrimeric element R12 (a), a copolymer comprising moieties derived from a monocarboxy acid functionalized polyhydroxylated acid is used, wherein the monocarboxy acid functionalized polyhydroxylated acid is selected from the group consisting 2,2-bis-(hydroxymethyl) propionic acid, hydrophilic C2 to C9 carboxylic acids, glycolic acid, lactic acid, γ-hydroxy butyric acid, 2,3-dihydroxy propionic acid, α,β-dihydroxy butyric acid, α,γ-dihydroxy butyric acid, gluconic acid, succinic acid, maleic acid, phthalic acid, terephthalic acid, citric acid, benzene 1,3,4 tricarboxylic acid, 1,3,5 tricarboxylic acid and mixtures thereof. 26. A cosmetic formulation comprising the polysiloxane compound according to claim 1 and at least one cosmetic ingredient. 27. The cosmetic formulation according to claim 26, wherein the polysiloxane compound according to claim 1 functions as an O/W emulsifier, an W/O emulsifier, viscosity regulators, antistatic agents, agents for a surface hydrophilization, mixture components for silicone rubbers which can be crosslinked to elastomers, modifiers of thermoplastics, components in coatings, for automobiles or medical equipment as well as additives for cleaning agents, detergents or preservative agents, an additive for toiletries, a coating agent for wood, paper and cardboard, a mould release agent, a biocompatible material for medicinal uses, a coating agent for textile fibres or textile fabrics, a coating agent for natural materials, defoaming agent for diesel oils and diesel fuels respectively, agents for the oil-water separation in crude oil and it's fractions, foam stabilizers in cold or warm hardening polyurethane hard or flexible foams, adjuvants in pesticides, agriculture, horticulture, turf, ornamental and forestry or emulsifier in compositions used therefore, a component in coating compositions, compatibilization of components, leveling, flow enhancement, deairing and the reduction of surface defects and drying accelerator. 28. The process for the production of carboxylic acid functions containing polyhydroxylated ester compounds according to claim 22, characterized in that an esterification is carried out in the presence of water and a strong acid as catalyst, the ratio of the monocarboxy acid functionalized polyhydroxylated acid or the acid mixture:water is 99.9:0.1 to 40:60, the reaction is carried out at 25 to 150° C., the catalyst concentration is in the range of 0.05 to 5%.
There is provided herein an organofunctional polysiloxanes comprising hydroxyl polyester groups made by reaction of epoxy functional polyorganosiloxanes and oligmeric polyesters based on polyhydroxy carboxylic acids. There is also provided methods for making the organofunctional polysiloxanes and agricultural, coating, personal care and home care applications containing the organofunctional polysiloxanes.1. A polysiloxane compound having the general formula (I): [MaDbD*cTdQe]f  (I) wherein M=R1R2R3SiO1/2; D=R4R5SiO2/2; D*=R6R7SiO2/2; T=R8SiO3/2; Q=SiO4/2; with a=1-10 b=0-1000 c=0-1000 d=0-1 e=0-1 f=1-10 wherein R1, R2, R3, R4, R5, R6 and R8 are each independently selected from the group consisting of monovalent hydrocarbon groups having from 1 to 8 carbon atoms, and an aryl or alkaryl hydrocarbon group of from 6 to 22 carbon atoms, or R7; R7 is selected from the group consisting of R9, R10 and R11, wherein R9 is selected from the group consisting of —Z-(A-E1)y, —Z-E2 and —Z—NH—C(O)—R12, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C20 hydrocarbon residue which can comprise one or more groups selected from —O—, —NH—,  and can be substituted by one or more OH groups, A is a bivalent residue selected from the group consisting of E1 is selected from the group consisting of E2 and E3 wherein E2=—O—C(O)—R12, wherein R12 is a dendrimer like branched hydrocarbon residue with up to 100 carbon atoms, which can comprise one or more groups selected from —O—, —C(O)—, and is substituted by one or more OH groups, wherein E2 is defined above, and x=1-4, y=1 or 2 R10 is selected from the group consisting of —Z-(A-E4)y′, —Z-E5 and —Z—NH—C(O)—R13, wherein Z and A are defined above, E4 is selected from the group consisting of E5 and E6 wherein E5=—O—C(O)—R13, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 9 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR14—, —C(O)—, and is substituted by one or more OH groups, wherein R14 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E5 is defined above, and x′=1-4, y′=1 or 2 R11 is selected from the group consisting of —Z-(A-E7)y, —Z-E8 and —Z—NH—C(O)—R15, wherein Z and A are defined above, E7 is selected from the group consisting of E8 and E9 wherein E8=—O—C(O)—R15, wherein R15 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with 10 to 50 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR16—, —C(O)—,)-, and is optionally substituted by one or more OH groups, wherein R16 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E8 is defined above, and x″=1-4, y″=1 or 2. 2. The polysiloxane compound according to claim 1, comprising structural elements selected from the following structures: wherein R17 is C1 to C22-alkyl, fluoro-substituted C1 to C22-alkyl or aryl, and g=0-600, wherein the groups R17 can be the same or different and are selected from C1 to C22-alkyl, fluoro-substituted C1 to C22-alkyl and aryl, and h=0-700, wherein R17 is as defined above, and i=0-10, wherein R7 is as defined above, and j=0-10, wherein R7 and R17 are as defined above, and k=0-30, wherein R17 is as defined above, and m=0-30, wherein n=0-10, g+h+i+j+k+m+n=12-1000. 3. The polysiloxane compound according to claim 2, comprising structural elements selected from the following structures: wherein R9 and R17 are defined above, and g1=0-300, wherein R10 and R17 are defined above, and g2=0-300, wherein R11 and R17 are defined above, and g3=0-300, wherein R9 is defined above, and j1=0-10, wherein R10 is defined above, and j2=0-10, wherein R11 is defined above, and j3=0-10, wherein R9 and R17 are defined above, and k1=0-15, wherein R10 and R17 are defined above, and k2=0-15, wherein R11 and R17 are defined above, and k3=0-15, and g1+g2+g3+h+i+j1+j2+j3+k1+k2+k3+m+n=12 to 1000. 4. The polysiloxane compound according to claim 2, wherein R17 is methyl or h=3-500 or i=0 or m=1-6 or n=0. 5. The polysiloxane compound according to claim 3, wherein R17 is methyl or g1=2-200 or g2=0-200 or g3=0-200 or j1=0 or j2=0 or j3=0 or k1=0-2 or k2=0-2 or k3=0-2 or g1+g2+g3+h+i+j1+j2+j3+k1+k2+k3+m+n=15 to 400 or h+k1=2-1000. 6. The polysiloxane compound according to claim 1, wherein the molar ratio of R9 to R17 is 10:1 to 1:10. 7. The polysiloxane compound according to claim 1, wherein the polysiloxane compound either do not contain hydrophilic residues R10 and/or lipophilic residues R11 or the molar ratio of the siloxy units comprising the dendrimer residue R9 to the siloxy units comprising hydrophilic residues R10 and lipophilic residues R11 in the polysiloxane compound is 1:0.01 to 1:100. 8. The polysiloxane compound according to claim 1, wherein the molar ratio of R9 to R10 and R11 is 1:0.1 to 1:10. 9. The polysiloxane compound according to claim 1, wherein R17 is C1 to C10-alkyl, optionally substituted with 1 to 13 fluoro atoms, and aryl. 10. The polysiloxane compound according to claim 1, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C10 hydrocarbon residue, which can comprise —O— groups and can be substituted by one or more OH groups. 11. The polysiloxane compound according to claim 1, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C6 hydrocarbon residue which can comprise one or more groups selected from —O—, —NH—, and can be substituted by one or more OH groups. 12. The polysiloxane compound according to claim 1, wherein R12 is dendrimer like branched hydrocarbon residue with up to 70 carbon atoms, which can comprise one or more groups selected from —O—, —C(O)—, and is substituted by one or more OH groups. 13. The polysiloxane compound according to claim 1, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 4 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR13—, —C(O)—, and is substituted by one or more OH groups, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms. 14. The polysiloxane compound according to claim 1, wherein R15 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with 10 to 30 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR13—, —C(O)—,)-, and is optionally substituted by one or more OH groups, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms. 15. The polysiloxane compound according to claim 1, wherein y=1 or x=1. 16. The polysiloxane compound according to claim 1 wherein R12 is wherein R19═R17 or H, w=1-3, R18═H or provided that the total number of carbon atoms in the dendrimer like residue R12 is 5 to 70 and at least one ester bond is present in the residue structure. 17. The polysiloxane compound according to claim 16 wherein R19 is CH3, and w=1. 18. The polysiloxane compound according to claim 16 wherein the number of ester bonds present in the residue structure is 1 to 14. 19. The polysiloxane compound according to claim 1 wherein Z═—CH2CH2CH2—O—CH2—, —CH2CH2CH2CH2—, —CH═CH2CH2—, —CH═CH2CH2CH2—, p=1 to 4, * marks a bond to the silicon atom in each case. 20. The polysiloxane compound according to claim 1 wherein —Z-A- is selected from the cyclic structures which are derived from cyclic epoxides. 21. The polysiloxane compound according to claim 1 wherein cyclic epoxides are selected from the group consisting of 22. A process for the production of the polysiloxane compound having the general formula (I): [MaDbD*cTdQe]f  (I) wherein M=R1R2R3SiO1/2; D=R4R5SiO2/2; D*=R6R7SiO2/2; T=R8SiO3/2; Q=SiO4/2; with a=1-10 b=0-1000 c=0-1000 d=0-1 e=0-1 f=1-10 wherein R1, R2, R3, R4, R5, R6 and R8 are each independently selected from the group consisting of monovalent hydrocarbon groups having from 1 to 8 carbon atoms, and an aryl or alkaryl hydrocarbon group of from 6 to 22 carbon atoms, or R7; R7 is selected from the group consisting of R9, R10 and R11, wherein R9 is selected from the group consisting of —Z-(A-E1)y, —Z-E2 and —Z—NH—C(O)—R12, wherein Z is a bivalent or trivalent straight-chained, cyclic or branched, saturated or unsaturated C2 to C20 hydrocarbon residue which can comprise one or more groups selected from —O—, —NH—,  and can be substituted by one or more OH groups, A is a bivalent residue selected from the group consisting of E1 is selected from the group consisting of E2 and E3 wherein E2=—O—C(O)—R12, wherein R12 is a dendrimer like branched hydrocarbon residue with up to 100 carbon atoms, which can comprise one or more groups selected from —O—, —C(O)—, and is substituted by one or more OH groups, wherein E2 is defined above, and x=1-4, y=1 or 2 R10 is selected from the group consisting of —Z-(A-E4)y′, —Z-E5 and —Z—NH—C(O)—R13, wherein Z and A are defined above, E4 is selected from the group consisting of E5 and E6 wherein E5=—O—C(O)—R13, wherein R13 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 9 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR14—, —C(O)—, and is substituted by one or more OH groups, wherein R14 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E5 is defined above, and x′=1-4, y′=1 or 2 R11 is selected from the group consisting of —Z-(A-E7)y, —Z-E8 and —Z—NH—C(O)—R15, wherein Z and A are defined above, E7 is selected from the group consisting of E8 and E9 wherein E8=—O—C(O)—R15, wherein R15 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with 10 to 50 carbon atoms, which can comprise one or more groups selected from —O—, —NH—, —NR16—, —C(O)—,)-, and is optionally substituted by one or more OH groups, wherein R16 is a straight-chained, cyclic or branched, saturated or unsaturated hydrocarbon residue with up to 6 carbon atoms, wherein E8 is defined above, and x″=1-4, y″=1 or 2 is characterised in that an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, optionally at the same time or subsequently with other hydrophilic and/or hydrophobic carboxylic acids or partially esterified carboxylic anhydrides and, optionally, subsequently with primary or secondary amines. 23. The process of claim 22 wherein the process comprising the steps of: a) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, b) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, at the same time or subsequently with other hydrophilic and/or hydrophobic carboxylic acids or partially esterified carboxylic anhydrides, c) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, and subsequently with primary or secondary amines, d) an epoxy functional polysiloxane is reacted with one or more than one monocarboxy functionalized dendrimer, at the same time or subsequently with other hydrophilic and/or hydrophobic carboxylic acids or partially esterified carboxylic anhydrides and subsequently with primary or secondary amines, and that for introducing the dendrimeric element R12 (a) preferably monocarboxy acid functionalized polyhydroxylated esters of the following structure are reacted with the epoxy functionalized silicones wherein R18 and R19 as defined above. 24. The process of claim 23 wherein R18 is H, w is 1 and R19 is CH3. 25. The process of claim 22 which is further characterized in that for introducing the dendrimeric element R12 (a), a copolymer comprising moieties derived from a monocarboxy acid functionalized polyhydroxylated acid is used, wherein the monocarboxy acid functionalized polyhydroxylated acid is selected from the group consisting 2,2-bis-(hydroxymethyl) propionic acid, hydrophilic C2 to C9 carboxylic acids, glycolic acid, lactic acid, γ-hydroxy butyric acid, 2,3-dihydroxy propionic acid, α,β-dihydroxy butyric acid, α,γ-dihydroxy butyric acid, gluconic acid, succinic acid, maleic acid, phthalic acid, terephthalic acid, citric acid, benzene 1,3,4 tricarboxylic acid, 1,3,5 tricarboxylic acid and mixtures thereof. 26. A cosmetic formulation comprising the polysiloxane compound according to claim 1 and at least one cosmetic ingredient. 27. The cosmetic formulation according to claim 26, wherein the polysiloxane compound according to claim 1 functions as an O/W emulsifier, an W/O emulsifier, viscosity regulators, antistatic agents, agents for a surface hydrophilization, mixture components for silicone rubbers which can be crosslinked to elastomers, modifiers of thermoplastics, components in coatings, for automobiles or medical equipment as well as additives for cleaning agents, detergents or preservative agents, an additive for toiletries, a coating agent for wood, paper and cardboard, a mould release agent, a biocompatible material for medicinal uses, a coating agent for textile fibres or textile fabrics, a coating agent for natural materials, defoaming agent for diesel oils and diesel fuels respectively, agents for the oil-water separation in crude oil and it's fractions, foam stabilizers in cold or warm hardening polyurethane hard or flexible foams, adjuvants in pesticides, agriculture, horticulture, turf, ornamental and forestry or emulsifier in compositions used therefore, a component in coating compositions, compatibilization of components, leveling, flow enhancement, deairing and the reduction of surface defects and drying accelerator. 28. The process for the production of carboxylic acid functions containing polyhydroxylated ester compounds according to claim 22, characterized in that an esterification is carried out in the presence of water and a strong acid as catalyst, the ratio of the monocarboxy acid functionalized polyhydroxylated acid or the acid mixture:water is 99.9:0.1 to 40:60, the reaction is carried out at 25 to 150° C., the catalyst concentration is in the range of 0.05 to 5%.
1,600
382
14,774,807
1,653
The invention relates to enzyme containing water-soluble films, and their use in detergents.
1. A method of making an enzyme containing water-soluble film, the method comprising use of a liquid enzyme formulation with an OD440 (10 mm path length) <0.15 per % AEP. 2. A method of making an enzyme containing water-soluble film, the method comprising use of a liquid enzyme formulation with an odor threshold of less than 20 ppm AEP. 3. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and an enzyme containing detergent, wherein the enzyme in the film is different from the enzyme in the detergent; preferably the enzyme in the film is protease. 4. A detergent pouch comprising at least one compartment formed by at least two separate enzymes containing water-soluble films, and a detergent, wherein the enzymes in the two films are different. In an embodiment, one film contains a protease. 5. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a detergent containing a builder with a Ca2+ logarithmic stability constant of above 4.5; preferably the detergent is a dishwash detergent. 6. The detergent pouch of claim 5, wherein the builder is a biodegradable chelating agent. 7. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a liquid detergent containing a viscosity modifier which is a substrate for the enzyme. 8. The detergent pouch of claim 7, wherein the enzyme containing water-soluble film is a lipase containing water-soluble film, and the viscosity modifier is hydrogenated castor oil. 9. A detergent pouch comprising a compartment formed by a lipase containing water-soluble film, a detergent, and a fragrance ester. 10. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a liquid detergent with an internal solvent system comprising at least one primary solvent having Hansen solubility (δ) of less than 30. 11. A detergent pouch comprising a compartment formed by at least two layers of film wherein at least one of the layers contain enzyme, and a detergent. 12. The detergent pouch of claim 11, wherein the layer of film containing enzyme is separated from the detergent by another layer of film. 13. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a detergent containing titanium dioxide. 14. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a detergent containing a reducing agent or antioxidant. 15. The detergent pouch of claim 5, wherein the detergent is a liquid detergent. 16. The detergent pouch of claim 9, wherein the detergent is a liquid detergent. 17. The detergent pouch of claim 11, wherein the detergent is a liquid detergent. 18. The detergent pouch of claim 13, wherein the detergent is a liquid detergent. 19. The detergent pouch of claim 14, wherein the detergent is a liquid detergent.
The invention relates to enzyme containing water-soluble films, and their use in detergents.1. A method of making an enzyme containing water-soluble film, the method comprising use of a liquid enzyme formulation with an OD440 (10 mm path length) <0.15 per % AEP. 2. A method of making an enzyme containing water-soluble film, the method comprising use of a liquid enzyme formulation with an odor threshold of less than 20 ppm AEP. 3. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and an enzyme containing detergent, wherein the enzyme in the film is different from the enzyme in the detergent; preferably the enzyme in the film is protease. 4. A detergent pouch comprising at least one compartment formed by at least two separate enzymes containing water-soluble films, and a detergent, wherein the enzymes in the two films are different. In an embodiment, one film contains a protease. 5. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a detergent containing a builder with a Ca2+ logarithmic stability constant of above 4.5; preferably the detergent is a dishwash detergent. 6. The detergent pouch of claim 5, wherein the builder is a biodegradable chelating agent. 7. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a liquid detergent containing a viscosity modifier which is a substrate for the enzyme. 8. The detergent pouch of claim 7, wherein the enzyme containing water-soluble film is a lipase containing water-soluble film, and the viscosity modifier is hydrogenated castor oil. 9. A detergent pouch comprising a compartment formed by a lipase containing water-soluble film, a detergent, and a fragrance ester. 10. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a liquid detergent with an internal solvent system comprising at least one primary solvent having Hansen solubility (δ) of less than 30. 11. A detergent pouch comprising a compartment formed by at least two layers of film wherein at least one of the layers contain enzyme, and a detergent. 12. The detergent pouch of claim 11, wherein the layer of film containing enzyme is separated from the detergent by another layer of film. 13. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a detergent containing titanium dioxide. 14. A detergent pouch comprising a compartment formed by an enzyme containing water-soluble film, and a detergent containing a reducing agent or antioxidant. 15. The detergent pouch of claim 5, wherein the detergent is a liquid detergent. 16. The detergent pouch of claim 9, wherein the detergent is a liquid detergent. 17. The detergent pouch of claim 11, wherein the detergent is a liquid detergent. 18. The detergent pouch of claim 13, wherein the detergent is a liquid detergent. 19. The detergent pouch of claim 14, wherein the detergent is a liquid detergent.
1,600
383
13,483,674
1,617
The present disclosure provides tissue products produced from adipose tissues, as well as methods for producing such tissue products. The tissue products can include acellular extracellular matrices. In addition, the present disclosure provides systems and methods for using such products.
1. A method for producing a tissue product, comprising: selecting an adipose tissue; mechanically processing the adipose tissue to reduce the tissue size; treating the mechanically processed tissue to remove substantially all cellular material from the tissue; suspending the tissue in a solution; and cross-linking the tissue to produce a stable three-dimensional structure. 2. The method of claim 1, further including drying the tissue. 3. The method of claim 2, wherein the tissue is dried after being mechanically processed and suspended but before being cross-linking. 4. The method of claim 2, wherein drying includes freeze-drying. 5. The method of claim 1, wherein cross-linking includes contacting the tissue with a cross-linking agent. 6. The method of claim 5, wherein the cross-linking agent includes at least one of glutaraldehyde, genepin, carbodiimides, and diisocyantes. 7. The method of claim 1, wherein cross-linking includes heating the tissue. 8. The method of claim 7, wherein the tissue is heated in a vacuum. 9. The method of claim 7, wherein the tissue is heated to 70° C. to 120° C. 10. The method of claim 1, further comprising placing the tissue in a mold to produce a desired shape prior to cross linking. 11. The method of claim 1, wherein the tissue is cross-linked such that the material maintains the stable three-dimensional structure when contacted with an aqueous environment. 12. The method of claim 11, wherein the material maintains the stable three-dimensional structure when implanted in a body. 13. The method of claim 1, wherein mechanically processing the tissue includes at least one of cutting, grinding, or blending to produce small particles. 14. The method of claim 1, further comprising contacting the tissue with a flat surface and applying force to compress the tissue to a thickness of about 2.0 mm or less and optionally cross-linking the tissue. 15. The method of claim 1, further comprising processing the tissue to form a drain or drain manifold in the form of a tube, column, or sheet, further processing the tissue to add grooves, channels, or holes, and cross-linking the tissue. 16. The method of claim 1, further comprising processing the tissue to form thin strips have a thickness of up to about 2.0 mm, rolling the thin strips to form a drain in the shape of a hollow tube, and cross-linking the tissue. 17. A tissue product made by a process comprising: selecting an adipose tissue; mechanically processing the adipose tissue to reduce the tissue size; treating the processed tissue to remove substantially all cellular material from the tissue; suspending the tissue in a solution; and cross-linking the tissue to produce a stable three-dimensional structure. 18. The product of claim 17, wherein the process further includes drying the tissue. 19. The product of claim 18, wherein the tissue is dried after being mechanically processed and suspended but before being cross-linking. 20. The product of claim 18, wherein drying includes freeze-drying. 21. The product of claim 17, wherein cross-linking includes contacting the tissue with a cross-linking agent. 22. The product of claim 21, wherein the cross-linking agent includes at least one of glutaraldehyde, genepin, carbodiimides, and diisocyantes. 23. The product of claim 17, wherein cross-linking includes heating the tissue. 24. The product of claim 23, wherein the tissue is heated in a vacuum. 25. The product of claim 23, wherein the tissue is heated to 70° C. to 120° C. 26. The product of claim 17, further comprising placing the tissue in a mold to produce a desired shape prior to cross linking. 27. The product of claim 17, wherein the tissue is cross-linked such that the material maintains the stable three-dimensional structure when contacted with an aqueous environment. 28. The product of claim 27, wherein the material maintains the stable three-dimensional structure when implanted in a body. 29. The product of claim 17, wherein the tissue contains hyaluronic acid and chondroitin sulfate. 30. The product of claim 17, wherein the tissue contains one or more growth factors selected from PECAM-1, HGF, VEGF, PDGF-BB, follistatin, IL-8, and FGF-basic. 31. The product of claim 17, wherein mechanically processing the tissue includes at least one of cutting, grinding, or blending to produce small particles. 32. The product of claim 17, further comprising contacting the tissue with a flat surface and applying force to compress the tissue to a thickness of about 2.0 mm or less and optionally cross-linking the tissue. 33. The product of claim 17, further comprising processing the tissue to form a drain or drain manifold in the form of a tube, column, or sheet, further processing the tissue to add grooves, channels, or holes, and cross-linking the tissue. 34. The product of claim 17, further comprising processing the tissue to form thin strips have a thickness of up to about 2.0 mm, rolling the thin strips to form a drain in the shape of a hollow tube, and cross-linking the tissue. 35-55. (canceled)
The present disclosure provides tissue products produced from adipose tissues, as well as methods for producing such tissue products. The tissue products can include acellular extracellular matrices. In addition, the present disclosure provides systems and methods for using such products.1. A method for producing a tissue product, comprising: selecting an adipose tissue; mechanically processing the adipose tissue to reduce the tissue size; treating the mechanically processed tissue to remove substantially all cellular material from the tissue; suspending the tissue in a solution; and cross-linking the tissue to produce a stable three-dimensional structure. 2. The method of claim 1, further including drying the tissue. 3. The method of claim 2, wherein the tissue is dried after being mechanically processed and suspended but before being cross-linking. 4. The method of claim 2, wherein drying includes freeze-drying. 5. The method of claim 1, wherein cross-linking includes contacting the tissue with a cross-linking agent. 6. The method of claim 5, wherein the cross-linking agent includes at least one of glutaraldehyde, genepin, carbodiimides, and diisocyantes. 7. The method of claim 1, wherein cross-linking includes heating the tissue. 8. The method of claim 7, wherein the tissue is heated in a vacuum. 9. The method of claim 7, wherein the tissue is heated to 70° C. to 120° C. 10. The method of claim 1, further comprising placing the tissue in a mold to produce a desired shape prior to cross linking. 11. The method of claim 1, wherein the tissue is cross-linked such that the material maintains the stable three-dimensional structure when contacted with an aqueous environment. 12. The method of claim 11, wherein the material maintains the stable three-dimensional structure when implanted in a body. 13. The method of claim 1, wherein mechanically processing the tissue includes at least one of cutting, grinding, or blending to produce small particles. 14. The method of claim 1, further comprising contacting the tissue with a flat surface and applying force to compress the tissue to a thickness of about 2.0 mm or less and optionally cross-linking the tissue. 15. The method of claim 1, further comprising processing the tissue to form a drain or drain manifold in the form of a tube, column, or sheet, further processing the tissue to add grooves, channels, or holes, and cross-linking the tissue. 16. The method of claim 1, further comprising processing the tissue to form thin strips have a thickness of up to about 2.0 mm, rolling the thin strips to form a drain in the shape of a hollow tube, and cross-linking the tissue. 17. A tissue product made by a process comprising: selecting an adipose tissue; mechanically processing the adipose tissue to reduce the tissue size; treating the processed tissue to remove substantially all cellular material from the tissue; suspending the tissue in a solution; and cross-linking the tissue to produce a stable three-dimensional structure. 18. The product of claim 17, wherein the process further includes drying the tissue. 19. The product of claim 18, wherein the tissue is dried after being mechanically processed and suspended but before being cross-linking. 20. The product of claim 18, wherein drying includes freeze-drying. 21. The product of claim 17, wherein cross-linking includes contacting the tissue with a cross-linking agent. 22. The product of claim 21, wherein the cross-linking agent includes at least one of glutaraldehyde, genepin, carbodiimides, and diisocyantes. 23. The product of claim 17, wherein cross-linking includes heating the tissue. 24. The product of claim 23, wherein the tissue is heated in a vacuum. 25. The product of claim 23, wherein the tissue is heated to 70° C. to 120° C. 26. The product of claim 17, further comprising placing the tissue in a mold to produce a desired shape prior to cross linking. 27. The product of claim 17, wherein the tissue is cross-linked such that the material maintains the stable three-dimensional structure when contacted with an aqueous environment. 28. The product of claim 27, wherein the material maintains the stable three-dimensional structure when implanted in a body. 29. The product of claim 17, wherein the tissue contains hyaluronic acid and chondroitin sulfate. 30. The product of claim 17, wherein the tissue contains one or more growth factors selected from PECAM-1, HGF, VEGF, PDGF-BB, follistatin, IL-8, and FGF-basic. 31. The product of claim 17, wherein mechanically processing the tissue includes at least one of cutting, grinding, or blending to produce small particles. 32. The product of claim 17, further comprising contacting the tissue with a flat surface and applying force to compress the tissue to a thickness of about 2.0 mm or less and optionally cross-linking the tissue. 33. The product of claim 17, further comprising processing the tissue to form a drain or drain manifold in the form of a tube, column, or sheet, further processing the tissue to add grooves, channels, or holes, and cross-linking the tissue. 34. The product of claim 17, further comprising processing the tissue to form thin strips have a thickness of up to about 2.0 mm, rolling the thin strips to form a drain in the shape of a hollow tube, and cross-linking the tissue. 35-55. (canceled)
1,600
384
15,084,441
1,627
The poly(ε-caprolactone)-ethoxylated fatty acid copolymers are block copolymers including ε-caprolactone units and ethoxylated fatty alcohol units, the block copolymer having the structural formula: where n and m are integers greater than 0 and R is an alkyl group. The block copolymer is prepared by polymerizing ε-caprolactone and an ethoxylated fatty alcohol in the presence of a catalyst, such as stannous octoate. The block copolymers have potential as delivery systems for various payloads, such as, but not limited to, lipid soluble drugs and diagnostic agents.
1. A poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer, comprising a block copolymer having at least one ε-caprolactone unit and ethoxylated fatty alcohol units, the block copolymer having the structural formula: where n and m are integers greater than 0 and R is an alkyl group, and wherein the poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer of claim 1, wherein n is 1 to 100, m is 10 to 100, and R is an alkyl group having 12-18 carbon atoms. 2-3. (canceled) 4. The poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer of claim 1, wherein the block copolymer has an average molecular weight of at least 3000 Daltons. 5. A method of making a poly(s-caprolactone)-ethoxylated fatty alcohol copolymer, comprising the steps of: mixing ε-caprolactone and an ethoxylated fatty alcohol in a reaction vessel to form a reaction mixture; adding a catalyst to the reaction mixture to catalyze polymerization; and heating the reaction vessel to 140° C. 6. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein said step of heating the reaction vessel to 140° C. comprises polymerizing the reaction mixture at a temperature of about 140° C. for between 4-5 hours. 7. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, further comprising the steps of: purging the reaction vessel of nitrogen; sealing the reaction vessel; and polymerizing the reaction mixture under vacuum. 8. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein said catalyst comprises stannous octoate. 9. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein said catalyst comprises bidentate sulfonamide zinc complex. 10. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein the ethoxylated fatty alcohol has a molecular weight between 1000 and 50,000 Daltons. 11. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, further comprising the step of cooling the reaction vessel to room temperature to terminate polymerization. 12. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein the ethoxylated fatty alcohol comprises polyoxyethylene stearyl ether. 13. (canceled) 14. A method of preparing self-assembled nanocarriers, comprising the steps of: dissolving a poly(ε-caprolactone)-ethoxylated fatty alcohol block copolymer in an organic solvent to form a solution; adding the solution drop-wise into distilled water; and evaporating the organic solvent to form self-assembled nanocarriers. 15. The method of preparing self-assembled nanocarriers according to claim 14, wherein the self-assembled nanocarriers have a mean diameter between 50 nm and 300 nm. 16. The method of preparing self-assembled nanocarriers according to claim 14, wherein the organic solvent comprises at least one solvent selected from the group consisting of acetone, tetrahydrofuran, acetonitrile, and dimethyl oxide.
The poly(ε-caprolactone)-ethoxylated fatty acid copolymers are block copolymers including ε-caprolactone units and ethoxylated fatty alcohol units, the block copolymer having the structural formula: where n and m are integers greater than 0 and R is an alkyl group. The block copolymer is prepared by polymerizing ε-caprolactone and an ethoxylated fatty alcohol in the presence of a catalyst, such as stannous octoate. The block copolymers have potential as delivery systems for various payloads, such as, but not limited to, lipid soluble drugs and diagnostic agents.1. A poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer, comprising a block copolymer having at least one ε-caprolactone unit and ethoxylated fatty alcohol units, the block copolymer having the structural formula: where n and m are integers greater than 0 and R is an alkyl group, and wherein the poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer of claim 1, wherein n is 1 to 100, m is 10 to 100, and R is an alkyl group having 12-18 carbon atoms. 2-3. (canceled) 4. The poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer of claim 1, wherein the block copolymer has an average molecular weight of at least 3000 Daltons. 5. A method of making a poly(s-caprolactone)-ethoxylated fatty alcohol copolymer, comprising the steps of: mixing ε-caprolactone and an ethoxylated fatty alcohol in a reaction vessel to form a reaction mixture; adding a catalyst to the reaction mixture to catalyze polymerization; and heating the reaction vessel to 140° C. 6. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein said step of heating the reaction vessel to 140° C. comprises polymerizing the reaction mixture at a temperature of about 140° C. for between 4-5 hours. 7. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, further comprising the steps of: purging the reaction vessel of nitrogen; sealing the reaction vessel; and polymerizing the reaction mixture under vacuum. 8. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein said catalyst comprises stannous octoate. 9. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein said catalyst comprises bidentate sulfonamide zinc complex. 10. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein the ethoxylated fatty alcohol has a molecular weight between 1000 and 50,000 Daltons. 11. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, further comprising the step of cooling the reaction vessel to room temperature to terminate polymerization. 12. The method of making a poly(ε-caprolactone)-ethoxylated fatty alcohol copolymer according to claim 5, wherein the ethoxylated fatty alcohol comprises polyoxyethylene stearyl ether. 13. (canceled) 14. A method of preparing self-assembled nanocarriers, comprising the steps of: dissolving a poly(ε-caprolactone)-ethoxylated fatty alcohol block copolymer in an organic solvent to form a solution; adding the solution drop-wise into distilled water; and evaporating the organic solvent to form self-assembled nanocarriers. 15. The method of preparing self-assembled nanocarriers according to claim 14, wherein the self-assembled nanocarriers have a mean diameter between 50 nm and 300 nm. 16. The method of preparing self-assembled nanocarriers according to claim 14, wherein the organic solvent comprises at least one solvent selected from the group consisting of acetone, tetrahydrofuran, acetonitrile, and dimethyl oxide.
1,600
385
13,043,021
1,611
There is provided a device comprising a body structure having one or more surfaces wherein at least one of the surfaces comprises a pH sensitive layer comprising a linear polymer, wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger. A method of forming a device, and a method of preventing or mitigating infection is also described.
1. A device comprising a body structure having one or more surfaces wherein at least one of the surfaces comprises a pH sensitive layer comprising a linear polymer, wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger and the linear polymer undergoes dissolution or erosion in an aqueous environment at the second water solubility. 2-13. (canceled) 14. The device of claim 1, wherein the device comprises more than three pH sensitive layers. 15. A method of forming a device comprising the steps of providing a structural layer and applying at least one pH sensitive layer thereto, said pH sensitive layer comprising a linear polymer wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger. 16-19. (canceled) 20. A method of preventing or mitigating infection associated with a device implanted or inserted into the human or animal body comprising the step of implanting or inserting a device into the human or animal body, wherein said device is a device according to claim 1. 21-27. (canceled) 28. A device comprising an extended body structure comprising: an extended structural layer having an inside surface and an outside surface, said inside surface defining an extended lumen, wherein the structural layer is substantially non-degradable or non-erodible at a pH of 2 to 10 and provides structural stability to the device; at least one extended pH sensitive degradable layer attached to, in centric arrangement with, and coextensive with the structural layer and being capable of controlled erosion, dissolution or degradation at a pH in the range of ≧5 to ≧7, each degradable layer comprising a pH sensitive linear polymer that ionizes and dissolves in an aqueous environment of use when the pH of the environment moves away from an initial pH to a pH that is indicative of bacterial contamination, wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger. 29. (canceled) 30. (canceled) 31. The device of claim 28 wherein plural pH sensitive degradable layers are present. 32-62. (canceled) 63. A multilayered catheter or stent comprising plural coextensive and centrically arranged layers, said layers defining a lumen, wherein: a first coextensive layer is a structural layer that is substantially non-degradable or non-erodible under physiological conditions; and at least one second coextensive layer is a pH sensitive layer comprising one or more pH sensitive linear polymers that ionize and dissolve, erode or degrade in an aqueous environment when the second coextensive layer is contaminated with bacteria. 64. The device according to claim 63 wherein the at least one second coextensive layer is in the interior of the first coextensive layer. 65. The device according to claim 63 wherein the at least one second coextensive layer is at the exterior of the first coextensive layer. 66. The device according to claim 63 wherein at least one second coextensive layer is in the interior of the first coextensive layer, and at least one second coextensive layer is at the exterior of the first coextensive layer. 67. The device according to claim 66 wherein two second coextensive layers are in the interior of the first coextensive layer, and one second coextensive layer is at the exterior of the first coextensive layer. 68. The device according to claim 67 wherein two interior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 69. The device according to claim 67 wherein all second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment 70. The device according to claim 66 wherein two second coextensive layers are at the exterior of the first coextensive layer, and one second coextensive layer is in the interior of the first coextensive layer. 71. The device according to claim 70 wherein two exterior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 72. The device according to claim 66 wherein two second coextensive layers are in the interior of the first coextensive layer, and two second coextensive layers are at the exterior of the first coextensive layer. 73. The device according to claim 72 wherein two interior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 74. The device according to claim 73 wherein two exterior second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment. 75. The device according to claim 72 wherein two exterior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 76. The device according to claim 75 wherein two interior second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment. 77. The device according to claim 72 wherein all second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment. 78. The device according to claim 63 wherein the bacterial contamination is caused by Escherichia coli, Lactobacillus bacterium, Proteus bacterium, other such bacteria known to be capable of contaminating the urinary tract. 79. The device according to claim 63 wherein the pH sensitive layer excludes a cross-linked pH sensitive polymer. 80. The device according to claim 63 wherein the pH sensitive layer comprises one or more linear pH sensitive polymers. 81. The device according to claim 63 wherein the structural layer comprises one or more of silicone, latex, (poly (vinyl chloride)), polyurethane, ethylene-vinylacetate copolymer, polyethylene, polypropylene, polyester, polystyrene, nylon. 82. The device according to claim 63 wherein the pH sensitive polymer is a cellulose-based linear polymer of the Formula I, wherein: a) at least one R is H, at least one R is —CH3, at least one R is —CH2CH(OH)CH3, at least one R is —COCH3, and at least one R is —COCH2CH2COOH, optionally wherein at least one R is —CH2CH(OCOCH2CH2COOH)CH3 or at least one R is —CH2CH(OCOCH3)CH3; b) at least one R is H, at least one R is —CH3, at least one R is —CH2CH(OH)CH3, and at least one R is —CO(C6H4)CO2H, optionally wherein at least one R is —CH2CH(OCO(C6H4)CO2H)CH3; c) at least one R is H, at least one R is —COCH3, and at least one R is —CO(C6H4)CO2H; d) at least one R is H, at least one R is —COCH3, and at least one R is —COCH2CH2CH3; or e) at least one R is H, at least one R is —COCH3, and at least one R is —CO(C6H3)(CO2H)2. 83. The device according to claim 63 wherein the pH trigger is ≧5, ≧5.5, ≧6, ≧6.2, ≧6.5, ≧6.8 or ≧7. 84. The device according to claim 63 wherein the linear pH sensitive polymer is selected from the group consisting of: a) hydroxypropyl methylcellulose acetate succinate having a pH trigger of ≧5.5, ≧6 or ≧6.8; b) hydroxypropyl methylcellulose phthalate having a pH trigger of ≧5 or ≧5.5; c) cellulose acetate trimellitate having a pH trigger of ≧5; d) cellulose acetate phthalate having a pH trigger of ≧6.2; or e) cellulose acetate butyrate having a pH trigger of ≧6. 85. The device according to claim 83 wherein the pH sensitive polymer having a pH trigger of about 7 is selected from the group consisting of copolymers of methacrylic acid and methyl methacrylate containing at least 2 methyl methacrylate units per methacrylic acid unit, partially esterified derivatives of hydroxypropyl methylcellulose containing at >/=12% acetyl content and </=7% succinoyl content. 86. The device according to claim 83 wherein the pH sensitive polymer having a pH trigger of about 6 is selected from the group consisting of partially esterified derivatives of hydroxypropyl methylcellulose containing at >/=9% acetyl content and </=11% succinoyl content, copolymers of methacrylic acid and methyl methacrylate containing at least 1 methyl methacrylate unit per methacrylic acid unit, cellulose acetate phthalate, cellulose acetate trimellitate, copolymers of ethylacrylate and methacrylic acid at a ratio of 1:1, wherein the layer further comprises one or more organic acids.
There is provided a device comprising a body structure having one or more surfaces wherein at least one of the surfaces comprises a pH sensitive layer comprising a linear polymer, wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger. A method of forming a device, and a method of preventing or mitigating infection is also described.1. A device comprising a body structure having one or more surfaces wherein at least one of the surfaces comprises a pH sensitive layer comprising a linear polymer, wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger and the linear polymer undergoes dissolution or erosion in an aqueous environment at the second water solubility. 2-13. (canceled) 14. The device of claim 1, wherein the device comprises more than three pH sensitive layers. 15. A method of forming a device comprising the steps of providing a structural layer and applying at least one pH sensitive layer thereto, said pH sensitive layer comprising a linear polymer wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger. 16-19. (canceled) 20. A method of preventing or mitigating infection associated with a device implanted or inserted into the human or animal body comprising the step of implanting or inserting a device into the human or animal body, wherein said device is a device according to claim 1. 21-27. (canceled) 28. A device comprising an extended body structure comprising: an extended structural layer having an inside surface and an outside surface, said inside surface defining an extended lumen, wherein the structural layer is substantially non-degradable or non-erodible at a pH of 2 to 10 and provides structural stability to the device; at least one extended pH sensitive degradable layer attached to, in centric arrangement with, and coextensive with the structural layer and being capable of controlled erosion, dissolution or degradation at a pH in the range of ≧5 to ≧7, each degradable layer comprising a pH sensitive linear polymer that ionizes and dissolves in an aqueous environment of use when the pH of the environment moves away from an initial pH to a pH that is indicative of bacterial contamination, wherein the water solubility of the linear polymer increases from a first water solubility to a second water solubility at a pH trigger. 29. (canceled) 30. (canceled) 31. The device of claim 28 wherein plural pH sensitive degradable layers are present. 32-62. (canceled) 63. A multilayered catheter or stent comprising plural coextensive and centrically arranged layers, said layers defining a lumen, wherein: a first coextensive layer is a structural layer that is substantially non-degradable or non-erodible under physiological conditions; and at least one second coextensive layer is a pH sensitive layer comprising one or more pH sensitive linear polymers that ionize and dissolve, erode or degrade in an aqueous environment when the second coextensive layer is contaminated with bacteria. 64. The device according to claim 63 wherein the at least one second coextensive layer is in the interior of the first coextensive layer. 65. The device according to claim 63 wherein the at least one second coextensive layer is at the exterior of the first coextensive layer. 66. The device according to claim 63 wherein at least one second coextensive layer is in the interior of the first coextensive layer, and at least one second coextensive layer is at the exterior of the first coextensive layer. 67. The device according to claim 66 wherein two second coextensive layers are in the interior of the first coextensive layer, and one second coextensive layer is at the exterior of the first coextensive layer. 68. The device according to claim 67 wherein two interior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 69. The device according to claim 67 wherein all second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment 70. The device according to claim 66 wherein two second coextensive layers are at the exterior of the first coextensive layer, and one second coextensive layer is in the interior of the first coextensive layer. 71. The device according to claim 70 wherein two exterior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 72. The device according to claim 66 wherein two second coextensive layers are in the interior of the first coextensive layer, and two second coextensive layers are at the exterior of the first coextensive layer. 73. The device according to claim 72 wherein two interior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 74. The device according to claim 73 wherein two exterior second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment. 75. The device according to claim 72 wherein two exterior second coextensive layers ionize and dissolve, erode or degrade at different pH values in an aqueous environment. 76. The device according to claim 75 wherein two interior second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment. 77. The device according to claim 72 wherein all second coextensive layers ionize and dissolve, erode or degrade at the same pH values in an aqueous environment. 78. The device according to claim 63 wherein the bacterial contamination is caused by Escherichia coli, Lactobacillus bacterium, Proteus bacterium, other such bacteria known to be capable of contaminating the urinary tract. 79. The device according to claim 63 wherein the pH sensitive layer excludes a cross-linked pH sensitive polymer. 80. The device according to claim 63 wherein the pH sensitive layer comprises one or more linear pH sensitive polymers. 81. The device according to claim 63 wherein the structural layer comprises one or more of silicone, latex, (poly (vinyl chloride)), polyurethane, ethylene-vinylacetate copolymer, polyethylene, polypropylene, polyester, polystyrene, nylon. 82. The device according to claim 63 wherein the pH sensitive polymer is a cellulose-based linear polymer of the Formula I, wherein: a) at least one R is H, at least one R is —CH3, at least one R is —CH2CH(OH)CH3, at least one R is —COCH3, and at least one R is —COCH2CH2COOH, optionally wherein at least one R is —CH2CH(OCOCH2CH2COOH)CH3 or at least one R is —CH2CH(OCOCH3)CH3; b) at least one R is H, at least one R is —CH3, at least one R is —CH2CH(OH)CH3, and at least one R is —CO(C6H4)CO2H, optionally wherein at least one R is —CH2CH(OCO(C6H4)CO2H)CH3; c) at least one R is H, at least one R is —COCH3, and at least one R is —CO(C6H4)CO2H; d) at least one R is H, at least one R is —COCH3, and at least one R is —COCH2CH2CH3; or e) at least one R is H, at least one R is —COCH3, and at least one R is —CO(C6H3)(CO2H)2. 83. The device according to claim 63 wherein the pH trigger is ≧5, ≧5.5, ≧6, ≧6.2, ≧6.5, ≧6.8 or ≧7. 84. The device according to claim 63 wherein the linear pH sensitive polymer is selected from the group consisting of: a) hydroxypropyl methylcellulose acetate succinate having a pH trigger of ≧5.5, ≧6 or ≧6.8; b) hydroxypropyl methylcellulose phthalate having a pH trigger of ≧5 or ≧5.5; c) cellulose acetate trimellitate having a pH trigger of ≧5; d) cellulose acetate phthalate having a pH trigger of ≧6.2; or e) cellulose acetate butyrate having a pH trigger of ≧6. 85. The device according to claim 83 wherein the pH sensitive polymer having a pH trigger of about 7 is selected from the group consisting of copolymers of methacrylic acid and methyl methacrylate containing at least 2 methyl methacrylate units per methacrylic acid unit, partially esterified derivatives of hydroxypropyl methylcellulose containing at >/=12% acetyl content and </=7% succinoyl content. 86. The device according to claim 83 wherein the pH sensitive polymer having a pH trigger of about 6 is selected from the group consisting of partially esterified derivatives of hydroxypropyl methylcellulose containing at >/=9% acetyl content and </=11% succinoyl content, copolymers of methacrylic acid and methyl methacrylate containing at least 1 methyl methacrylate unit per methacrylic acid unit, cellulose acetate phthalate, cellulose acetate trimellitate, copolymers of ethylacrylate and methacrylic acid at a ratio of 1:1, wherein the layer further comprises one or more organic acids.
1,600
386
15,330,732
1,661
A new cherry tree variety suitable for use as rootstock.
1. A new and distinct variety of cherry tree substantially as described and illustrated herein.
A new cherry tree variety suitable for use as rootstock.1. A new and distinct variety of cherry tree substantially as described and illustrated herein.
1,600
387
15,330,734
1,661
A new cherry tree variety suitable for use as rootstock.
1. A new and distinct variety of cherry tree substantially as described and illustrated herein.
A new cherry tree variety suitable for use as rootstock.1. A new and distinct variety of cherry tree substantially as described and illustrated herein.
1,600
388
14,356,650
1,628
A pharmaceutical, non-effervescent, solid composition comprising a mixture of a pharmaceutically effective amount of a ibuprofen salt and a pharmaceutically acceptable strong base in a molar ratio of from 1:0.01 to 1:0.8, said composition being such that, when dissolved in drinkable water for dilution, imparts a pH value ranging from 9 to 9.5 to the obtained solution, which do not cause sensory irritation to the oral cavity especially to the back of the mouth and throat, when swallowed.
1. A pharmaceutical, non-effervescent, solid composition comprising a mixture of a pharmaceutically effective amount of a ibuprofen salt and a pharmaceutically acceptable base in a molar ratio of from 1:0.01 to 1:0.8, said composition being such that, when dissolved in drinkable water for dilution, imparts a pH value ranging from 9.0 to 9.5 to the obtained solution. 2. The composition according to claim 1, wherein a mixture of a pharmaceutically effective amount of the ibuprofen salt and the pharmaceutically acceptable base is in a molar ratio of from 1:0.5 to 1:0.8. 3. The composition according to claim 1, wherein the pH value is 9.2. 4. The composition according to claim 1, wherein the ibuprofen salt is selected from L-arginine, L-lysine, sodium and potassium ibuprofen salt. 5. The composition according to claim 4, wherein the ibuprofen salt is ibuprofen L-arginine. 6. The composition according to claim 1, wherein the base is selected from an alkaline metal carbonate, an alkaline metal hydroxide and a tribasic metal phosphate. 7. The composition according to claim 6, wherein the alkaline metal carbonate is sodium carbonate. 8. The composition according to claim 6, wherein the alkaline metal hydroxide is sodium hydroxide. 9. The compositions according to claim 1, wherein the composition is in the form of powder suitably contained in sachets. 10. The composition according to claim 9, wherein the sachet contains an ibuprofen salt in an amount equivalent to a conventional oral unit dose of ibuprofen. 11. The composition according to claim 10 wherein to the conventional oral unit dose of ibuprofen is equal to 400 mg, 600 mg or 800 mg. 12. Method for preparing an aqueous solution having a pH value ranging from 9 to 9.5 with the composition of claim 1, said method comprising dissolving said composition in drinkable water for dilution in a volume of from 25 ml to 100 ml. 13. The method according to claim 12, wherein the volume of drinkable water is 50 ml. 14. A non-irritant liquid formulation obtainable by dissolving the composition according to claim 1 in drinkable water for dilution. 15. A liquid formulation according to claim 14 wherein the drinkable water for dilution has a volume of from 25 ml to 100 ml. 16. A method of avoiding sensory irritation to the oral cavity caused by a ibuprofen salt selected from the group consisting of L-arginine, L-lysine, sodium and potassium ibuprofen salts when swallowed in a non-effervescent liquid pharmaceutical oral solution thereof, said method comprising: mixing together a pharmaceutically effective amount of said ibuprofen salt with a pharmaceutically acceptable base selected from the group consisting of sodium carbonate, potassium carbonate; sodium hydroxide, potassium hydroxide; sodium tribasic phosphate and potassium tribasic phosphate in a molar ratio of from 1:0.01 to 1:0.8, with respect to ibuprofen salt before dissolving said ibuprofen salt into drinkable water for dilution. 17. A method according to claim 16 wherein the drinkable water for dilution has a volume of from 25 ml to 100 ml. 18. A method for eliminating unpleasant feeling of irritation of the oral cavity that usually follows the ingestion of a ibuprofen salt as an aqueous solution, said method comprising dissolving a composition according to claim 1 in drinkable water for dilution. 19. A method of making a pharmaceutical, non-effervescent, solid oral composition comprising a pharmaceutically effective amount of a ibuprofen salt and a pharmaceutically acceptable base to impart a pH value ranging from 9 to 9.5 to an aqueous solution of said composition, said method comprising: (i) mixing together a pharmaceutically acceptable amount of an ibuprofen salt and a base so that the molar ratio of the ibuprofen salt to a base may range from 1:0.01 to 1:0.8; (ii) preparing a solid oral composition; and (iii) filling said solid oral composition into a sachet. 20. The method according to claim 19, wherein said step (ii) comprises adding pharmaceutically acceptable excipients. 21. The method according to claim 19, wherein said method comprises a further step (iv) after step (iii), and wherein said further step (iv) comprises inserting one or more sachets into a package. 22. The composition according to claim 1 comprising a mixture of a pharmaceutically effective amount of ibuprofen L-arginine salt, and sodium carbonate or sodium hydroxide in a molar ratio of from 1:0.5 to 1:0.8; said composition being such that, when dissolved in drinkable water for dilution, imparts a pH value ranging from 9 to 9.5 to a said drinkable water solution.
A pharmaceutical, non-effervescent, solid composition comprising a mixture of a pharmaceutically effective amount of a ibuprofen salt and a pharmaceutically acceptable strong base in a molar ratio of from 1:0.01 to 1:0.8, said composition being such that, when dissolved in drinkable water for dilution, imparts a pH value ranging from 9 to 9.5 to the obtained solution, which do not cause sensory irritation to the oral cavity especially to the back of the mouth and throat, when swallowed.1. A pharmaceutical, non-effervescent, solid composition comprising a mixture of a pharmaceutically effective amount of a ibuprofen salt and a pharmaceutically acceptable base in a molar ratio of from 1:0.01 to 1:0.8, said composition being such that, when dissolved in drinkable water for dilution, imparts a pH value ranging from 9.0 to 9.5 to the obtained solution. 2. The composition according to claim 1, wherein a mixture of a pharmaceutically effective amount of the ibuprofen salt and the pharmaceutically acceptable base is in a molar ratio of from 1:0.5 to 1:0.8. 3. The composition according to claim 1, wherein the pH value is 9.2. 4. The composition according to claim 1, wherein the ibuprofen salt is selected from L-arginine, L-lysine, sodium and potassium ibuprofen salt. 5. The composition according to claim 4, wherein the ibuprofen salt is ibuprofen L-arginine. 6. The composition according to claim 1, wherein the base is selected from an alkaline metal carbonate, an alkaline metal hydroxide and a tribasic metal phosphate. 7. The composition according to claim 6, wherein the alkaline metal carbonate is sodium carbonate. 8. The composition according to claim 6, wherein the alkaline metal hydroxide is sodium hydroxide. 9. The compositions according to claim 1, wherein the composition is in the form of powder suitably contained in sachets. 10. The composition according to claim 9, wherein the sachet contains an ibuprofen salt in an amount equivalent to a conventional oral unit dose of ibuprofen. 11. The composition according to claim 10 wherein to the conventional oral unit dose of ibuprofen is equal to 400 mg, 600 mg or 800 mg. 12. Method for preparing an aqueous solution having a pH value ranging from 9 to 9.5 with the composition of claim 1, said method comprising dissolving said composition in drinkable water for dilution in a volume of from 25 ml to 100 ml. 13. The method according to claim 12, wherein the volume of drinkable water is 50 ml. 14. A non-irritant liquid formulation obtainable by dissolving the composition according to claim 1 in drinkable water for dilution. 15. A liquid formulation according to claim 14 wherein the drinkable water for dilution has a volume of from 25 ml to 100 ml. 16. A method of avoiding sensory irritation to the oral cavity caused by a ibuprofen salt selected from the group consisting of L-arginine, L-lysine, sodium and potassium ibuprofen salts when swallowed in a non-effervescent liquid pharmaceutical oral solution thereof, said method comprising: mixing together a pharmaceutically effective amount of said ibuprofen salt with a pharmaceutically acceptable base selected from the group consisting of sodium carbonate, potassium carbonate; sodium hydroxide, potassium hydroxide; sodium tribasic phosphate and potassium tribasic phosphate in a molar ratio of from 1:0.01 to 1:0.8, with respect to ibuprofen salt before dissolving said ibuprofen salt into drinkable water for dilution. 17. A method according to claim 16 wherein the drinkable water for dilution has a volume of from 25 ml to 100 ml. 18. A method for eliminating unpleasant feeling of irritation of the oral cavity that usually follows the ingestion of a ibuprofen salt as an aqueous solution, said method comprising dissolving a composition according to claim 1 in drinkable water for dilution. 19. A method of making a pharmaceutical, non-effervescent, solid oral composition comprising a pharmaceutically effective amount of a ibuprofen salt and a pharmaceutically acceptable base to impart a pH value ranging from 9 to 9.5 to an aqueous solution of said composition, said method comprising: (i) mixing together a pharmaceutically acceptable amount of an ibuprofen salt and a base so that the molar ratio of the ibuprofen salt to a base may range from 1:0.01 to 1:0.8; (ii) preparing a solid oral composition; and (iii) filling said solid oral composition into a sachet. 20. The method according to claim 19, wherein said step (ii) comprises adding pharmaceutically acceptable excipients. 21. The method according to claim 19, wherein said method comprises a further step (iv) after step (iii), and wherein said further step (iv) comprises inserting one or more sachets into a package. 22. The composition according to claim 1 comprising a mixture of a pharmaceutically effective amount of ibuprofen L-arginine salt, and sodium carbonate or sodium hydroxide in a molar ratio of from 1:0.5 to 1:0.8; said composition being such that, when dissolved in drinkable water for dilution, imparts a pH value ranging from 9 to 9.5 to a said drinkable water solution.
1,600
389
15,096,228
1,655
A method of treating hypertension can include administering to a patient in need thereof a therapeutically effective amount of an extract of Matricaria chamomilla L. The extract can be administered orally to the patient in an amount of about 100 mg/kg to about 200 mg/kg.
1. A method of treating hypertension, comprising administering orally to a patient in need thereof a therapeutically effective amount of an extract of Matricaria chamomilla only, wherein the Matricaria chamomilla is from the Al-Alnofood desert region of Saudi Arabia, further wherein the therapeutically effective amount of the extract of Matricaria chamomilla is about 100 mg/kg to about 200 mg/kg. 2. (canceled) 3. (canceled) 4. The method of treating hypertension according to claim 1, wherein the extract of Matricaria chamomilla is prepared by: providing a sample derived from aerial parts of Matricaria chamomilla, drying the sample, pulverizing the sample to provide a fine powder; and extracting the powder by alcohol extraction. 5. The method of treating hypertension according to claim 4, wherein the sample is obtained from flowers of Matricaria chamomilla. 6. The method of treating hypertension according to claim 4, wherein the alcohol extraction comprises: percolating the fine powder in alcohol to provide a crude alcoholic extract; filtering the extract to obtain a crude alcoholic extract; and concentrating the crude alcoholic extract to obtain a solid extract. 7. The method of treating hypertension according to claim 6, wherein the percolation is carried out for about 72 hours. 8. The method of treating hypertension according to claim 1, wherein the extract of Matricaria chamomilla is prepared by water distillation, the water distillation comprising boiling the Matricaria chamomilla to provide a heated mixture and separating essential oils from the heated mixture to provide an aqueous extract including essential oils. 9. The method of treating hypertension according to claim 8, wherein the water distillation method further comprises vaporizing water from the heated mixture using lyophilization to provide a dried extract. 10. The method of treating hypertension according to claim 8, wherein the extract is derived from aerial parts of Matricaria chamomilla. 11. A method of preparing an extract of Matricaria chamomilla, comprising: providing a sample derived from aerial parts of Matricaria chamomilla, drying the sample, pulverizing the sample to provide a fine powder; and extracting the powder by alcohol extraction. 12. The method of preparing an extract of Matricaria chamomilla according to claim 11, wherein the sample is obtained from flowers of Matricaria chamomilla. 13. The method of preparing an extract of Matricaria chamomilla according to claim 11, wherein the alcohol extraction comprises: percolating the fine powder in alcohol to provide a crude alcoholic extract; filtering the extract to obtain a crude alcoholic extract; and concentrating the crude alcoholic extract to obtain a solid extract. 14. The method of preparing an extract of Matricaria chamomilla according to claim 13, wherein the percolation is carried out for about 72 hours. 15. A method of preparing an extract of Matricaria chamomilla, comprising, water distillation of the sample, the water distillation including boiling the Matricaria chamomilla sample to provide a heated mixture and separating essential oils from the heated mixture to provide an aqueous extract including essential oils. 16. The method of preparing an extract of Matricaria chamomilla according to claim 15, wherein the water distillation method further comprises vaporizing water from the heated mixture using lyophilization to provide a dried extract.
A method of treating hypertension can include administering to a patient in need thereof a therapeutically effective amount of an extract of Matricaria chamomilla L. The extract can be administered orally to the patient in an amount of about 100 mg/kg to about 200 mg/kg.1. A method of treating hypertension, comprising administering orally to a patient in need thereof a therapeutically effective amount of an extract of Matricaria chamomilla only, wherein the Matricaria chamomilla is from the Al-Alnofood desert region of Saudi Arabia, further wherein the therapeutically effective amount of the extract of Matricaria chamomilla is about 100 mg/kg to about 200 mg/kg. 2. (canceled) 3. (canceled) 4. The method of treating hypertension according to claim 1, wherein the extract of Matricaria chamomilla is prepared by: providing a sample derived from aerial parts of Matricaria chamomilla, drying the sample, pulverizing the sample to provide a fine powder; and extracting the powder by alcohol extraction. 5. The method of treating hypertension according to claim 4, wherein the sample is obtained from flowers of Matricaria chamomilla. 6. The method of treating hypertension according to claim 4, wherein the alcohol extraction comprises: percolating the fine powder in alcohol to provide a crude alcoholic extract; filtering the extract to obtain a crude alcoholic extract; and concentrating the crude alcoholic extract to obtain a solid extract. 7. The method of treating hypertension according to claim 6, wherein the percolation is carried out for about 72 hours. 8. The method of treating hypertension according to claim 1, wherein the extract of Matricaria chamomilla is prepared by water distillation, the water distillation comprising boiling the Matricaria chamomilla to provide a heated mixture and separating essential oils from the heated mixture to provide an aqueous extract including essential oils. 9. The method of treating hypertension according to claim 8, wherein the water distillation method further comprises vaporizing water from the heated mixture using lyophilization to provide a dried extract. 10. The method of treating hypertension according to claim 8, wherein the extract is derived from aerial parts of Matricaria chamomilla. 11. A method of preparing an extract of Matricaria chamomilla, comprising: providing a sample derived from aerial parts of Matricaria chamomilla, drying the sample, pulverizing the sample to provide a fine powder; and extracting the powder by alcohol extraction. 12. The method of preparing an extract of Matricaria chamomilla according to claim 11, wherein the sample is obtained from flowers of Matricaria chamomilla. 13. The method of preparing an extract of Matricaria chamomilla according to claim 11, wherein the alcohol extraction comprises: percolating the fine powder in alcohol to provide a crude alcoholic extract; filtering the extract to obtain a crude alcoholic extract; and concentrating the crude alcoholic extract to obtain a solid extract. 14. The method of preparing an extract of Matricaria chamomilla according to claim 13, wherein the percolation is carried out for about 72 hours. 15. A method of preparing an extract of Matricaria chamomilla, comprising, water distillation of the sample, the water distillation including boiling the Matricaria chamomilla sample to provide a heated mixture and separating essential oils from the heated mixture to provide an aqueous extract including essential oils. 16. The method of preparing an extract of Matricaria chamomilla according to claim 15, wherein the water distillation method further comprises vaporizing water from the heated mixture using lyophilization to provide a dried extract.
1,600
390
11,432,226
1,611
A liquid or semi-solid matrix or magma or paste which is non-newtonian, thixotropic and pseudoplastic and composed of one or more controlled release agent, and/or one or more clays such as bentonite and/or one or more fillers in a non aqueous vehicle, and optionally materials selected from disintegrants, humectants, surfactants and stabilizers. The composition and physicochemical properties makes it harder or prevents dose dumping of narcotic analgesics in the presence of alcohol and harder to abuse opiod agonists or narcotic analgesics and discourages drug abuse via crushing, milling or grinding the dosage form to powder or heating the dosage form to vapour and snorting or inhalation by the nasal route or dissolving to abuse via the parenteral route.
1-117. (canceled) 118. The pharmaceutical composition of claim 140, wherein the paste is difficult to crush, dissolve, heat, evaporate, or extract. 119-128. (canceled) 129. A transdermal patch comprising the composition of claim 118. 130. A capsule comprising the composition of claim 118. 131. The capsule of claim 130, wherein the capsule is a hard gelatin capsule or a soft gelatin capsule. 132. The capsule of claim 130, which is coated with a film coat comprising cellulose esters or polymethacrylates or polyethylene glycol or cellulose ethers, esters or a combination thereof, and which in the presence of alcohol does not dose dump. 133. The capsule of claim 130, wherein the capsule comprises two different paste compositions, a first paste composition comprising the addictive substance and a second paste composition comprising an active substance that is the same or different than the addictive substance. 134. The capsule of claim 133, wherein the capsule further comprises a separation layer for separating the first paste composition and the second paste composition. a separation layer made of a wax such as carnuba wax or a high molecular weight polyethylene glycol. 135-138. (canceled) 139. The composition of claim 140, wherein the controlled release agent is selected from the group consisting of acacia, tragacanth, Xanthan gum, locust bean gum, guar-gum, karaya gum, pectin, arginic acid, polyethylene oxide, polyethylene glycol, polypropylene glycol arginate, hydroxypropyl methylcellulose, methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, carboxymethylcellulose sodium, polyvinylpyrrolidone, carboxyvinyl polymer, sodium polyacrylate, alpha starch, sodium carboxymethyl starch, albumin, dextrin, dextran sulfate, agar, gelatin, casein, sodium casein, pullulan, polyvinyl alcohol, deacetylated chitosan, polyethyoxazoline, poloxamers, ethylcellulose, chitin, chitosan, cellulose esters, aminoalkyl methacrylate polymer, anionic polymers of methacrylic acid and methacrylates, copolymers of acrylate and methacrylates with quaternary ammonium groups, ethylacrylate methylmethacrylate copolymers with a neutral ester group, polymethacrylates, surfactants, aliphatic polyesters, zein, polyvinyl acetate, polyvinyl chloride, and the like. 140. A pharmaceutical composition comprising: i) an addictive substance; ii) an oily, waxy, or fatty substance, or a combination thereof; iii) a clay selected from bentonite, montmorillonite, Pascalite, Smectite, illite, sepiolite, palygorskite, muscovite, allevardite, amesite, hectorite, fluorohectorite, saponite, beidellite, nontronite, stevensite, mica, vermiculite, fluorovermiculite, halloysite, fluorine-containing synthetic types of mica, phyllosilicates, beidellite, volkonskoite, hectorite, sauconite, sobockite, svinfordite, mixed illite/smectite minerals, and admixtures of illites with the clays named above; and iv) a controlled release agent; wherein the composition is dissolved, dispersed, emulsified, or suspended in the oily, waxy, or fatty substance to form a homogenous paste and is in a dosage form dispensing device and wherein the paste is on a structure. 141. The composition of claim 140, wherein the composition is non-newtonian, thixotropic and/or pseudoplastic. 142. The composition of claim 140, further comprising a disintegrant. 143. The composition of claim 140, further comprising an aqueous vehicle and an emulsifier. 144. The composition of claim 140, wherein the mixed illite/smectite minerals are rectorite, tarosovite, or ledikite. 145. The composition of claim 140, further comprising non-dissolved particles of a size less than 1000, 500, 200, or 100 microns. 146. The composition of claim 140, further comprising a nasal irritant. 147. The composition of claim 140, wherein the abuse potential of the addictive substance is reduced for at least one of the modes of crushing, milling or grinding the dosage form to powder and snorting or inhalation by the nasal route or dissolving for abuse via the parenteral route. 148. The composition of claim 140, wherein dissolution using a USP dissolution tester is not significantly affected by the rotation speed of the basket or paddle in the speed range from about 25 rpm to about 150 rpm. 149. The composition of claim 140, wherein there is no dose dumping during dissolution using a USP dissolution tester with basket or paddle assembly at 50 rpm in about 10% to about 20% ethanol. 150. The composition of claim 140, wherein there is no dose dumping during dissolution using a USP dissolution tester with basket or paddle assembly at 50 rpm in about 20% to about 30% ethanol. 151. The composition of claim 140, wherein the addictive substance is selected from opioid agonists, narcotic analgesics, and narcotic-like analgesics. 152. The composition of claim 140, wherein the addictive substance is selected from the group consisting of alfentanil, allylprodine, α prodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, etorphine, dihydroetorphine, fentanyl hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levorphanol, levophenacylmorphan, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, nalbuphene, normorphine, norpipanone, opium, oxycodone, oxymorphone, papaveretum, pentazocine, phenadoxone, phenomorphan, phenazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propoxyphene, sufentanil, tramadol, tilidine, α prodine, dextroporpoxyphene, propiram, profadol, phenampromide, thiambutene, pholcodeine, 3-transdimethylamino-4-phenyl-4-trans-carbethoxy-δ′-cyclohexene, 3-dimethylamino-O-(4-methoxyphenylcarbamoyl)-propiophenone oxime, (−)β-2′-hydroxy-2,9-dimethyl-5-phenyl-6,7-benzomorphan, (−)2′-hydroxy-2-(3-methyl-2-butenyl)-9-methyl-5-phenyl-6,7-benzomorphan, pirinitramide, (−)α-5,9-diethyl-2′-hydroxy-2-methyl-6,7-benzomorphan, ethyl 1-(2-dimethylaminoethyl)-4,5,6,7-tetrahydro-3-methyl-4-oxo-6-phenylindole-2-carboxylate, 1-benzoylmethyl-2,3-dimethyl-3-(m-hydroxyphenyl)-piperidine, N-allyl-7α-(1-(R)-hydroxy-1-methylbutyl)-6,14-endo-ethanotetrahydron ororipavine, (−)2′-hydroxy-2-methyl-6,7-benzomorphan, noracylmethadol, phenoperidine, α-dl-methadol, β-dl-methadol, α-1-methadol (215 mg.), β-dl-acetylmethadol, α-1-acetylmethadol, β-1-acetylmethadol, pharmaceutically acceptable salts thereof, stereoisomers thereof, ethers thereof, esters thereof, and mixtures thereof. 153. The composition of claim 140, wherein the controlled release agent is selected from the group consisting of naturally occurring or synthetic, anionic or nonionic, hydrophobic, hydrophilic rubbers, polymers, starch derivatives, cellulose derivatives, polysaccharides, carbomer, reseins, acrylics, proteins, vinyl-pyrrolidone-vinyl-acetate-copolymers, galactomannan and galactomannan derivatives, carrageenans and the like.
A liquid or semi-solid matrix or magma or paste which is non-newtonian, thixotropic and pseudoplastic and composed of one or more controlled release agent, and/or one or more clays such as bentonite and/or one or more fillers in a non aqueous vehicle, and optionally materials selected from disintegrants, humectants, surfactants and stabilizers. The composition and physicochemical properties makes it harder or prevents dose dumping of narcotic analgesics in the presence of alcohol and harder to abuse opiod agonists or narcotic analgesics and discourages drug abuse via crushing, milling or grinding the dosage form to powder or heating the dosage form to vapour and snorting or inhalation by the nasal route or dissolving to abuse via the parenteral route.1-117. (canceled) 118. The pharmaceutical composition of claim 140, wherein the paste is difficult to crush, dissolve, heat, evaporate, or extract. 119-128. (canceled) 129. A transdermal patch comprising the composition of claim 118. 130. A capsule comprising the composition of claim 118. 131. The capsule of claim 130, wherein the capsule is a hard gelatin capsule or a soft gelatin capsule. 132. The capsule of claim 130, which is coated with a film coat comprising cellulose esters or polymethacrylates or polyethylene glycol or cellulose ethers, esters or a combination thereof, and which in the presence of alcohol does not dose dump. 133. The capsule of claim 130, wherein the capsule comprises two different paste compositions, a first paste composition comprising the addictive substance and a second paste composition comprising an active substance that is the same or different than the addictive substance. 134. The capsule of claim 133, wherein the capsule further comprises a separation layer for separating the first paste composition and the second paste composition. a separation layer made of a wax such as carnuba wax or a high molecular weight polyethylene glycol. 135-138. (canceled) 139. The composition of claim 140, wherein the controlled release agent is selected from the group consisting of acacia, tragacanth, Xanthan gum, locust bean gum, guar-gum, karaya gum, pectin, arginic acid, polyethylene oxide, polyethylene glycol, polypropylene glycol arginate, hydroxypropyl methylcellulose, methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, carboxymethylcellulose sodium, polyvinylpyrrolidone, carboxyvinyl polymer, sodium polyacrylate, alpha starch, sodium carboxymethyl starch, albumin, dextrin, dextran sulfate, agar, gelatin, casein, sodium casein, pullulan, polyvinyl alcohol, deacetylated chitosan, polyethyoxazoline, poloxamers, ethylcellulose, chitin, chitosan, cellulose esters, aminoalkyl methacrylate polymer, anionic polymers of methacrylic acid and methacrylates, copolymers of acrylate and methacrylates with quaternary ammonium groups, ethylacrylate methylmethacrylate copolymers with a neutral ester group, polymethacrylates, surfactants, aliphatic polyesters, zein, polyvinyl acetate, polyvinyl chloride, and the like. 140. A pharmaceutical composition comprising: i) an addictive substance; ii) an oily, waxy, or fatty substance, or a combination thereof; iii) a clay selected from bentonite, montmorillonite, Pascalite, Smectite, illite, sepiolite, palygorskite, muscovite, allevardite, amesite, hectorite, fluorohectorite, saponite, beidellite, nontronite, stevensite, mica, vermiculite, fluorovermiculite, halloysite, fluorine-containing synthetic types of mica, phyllosilicates, beidellite, volkonskoite, hectorite, sauconite, sobockite, svinfordite, mixed illite/smectite minerals, and admixtures of illites with the clays named above; and iv) a controlled release agent; wherein the composition is dissolved, dispersed, emulsified, or suspended in the oily, waxy, or fatty substance to form a homogenous paste and is in a dosage form dispensing device and wherein the paste is on a structure. 141. The composition of claim 140, wherein the composition is non-newtonian, thixotropic and/or pseudoplastic. 142. The composition of claim 140, further comprising a disintegrant. 143. The composition of claim 140, further comprising an aqueous vehicle and an emulsifier. 144. The composition of claim 140, wherein the mixed illite/smectite minerals are rectorite, tarosovite, or ledikite. 145. The composition of claim 140, further comprising non-dissolved particles of a size less than 1000, 500, 200, or 100 microns. 146. The composition of claim 140, further comprising a nasal irritant. 147. The composition of claim 140, wherein the abuse potential of the addictive substance is reduced for at least one of the modes of crushing, milling or grinding the dosage form to powder and snorting or inhalation by the nasal route or dissolving for abuse via the parenteral route. 148. The composition of claim 140, wherein dissolution using a USP dissolution tester is not significantly affected by the rotation speed of the basket or paddle in the speed range from about 25 rpm to about 150 rpm. 149. The composition of claim 140, wherein there is no dose dumping during dissolution using a USP dissolution tester with basket or paddle assembly at 50 rpm in about 10% to about 20% ethanol. 150. The composition of claim 140, wherein there is no dose dumping during dissolution using a USP dissolution tester with basket or paddle assembly at 50 rpm in about 20% to about 30% ethanol. 151. The composition of claim 140, wherein the addictive substance is selected from opioid agonists, narcotic analgesics, and narcotic-like analgesics. 152. The composition of claim 140, wherein the addictive substance is selected from the group consisting of alfentanil, allylprodine, α prodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, etorphine, dihydroetorphine, fentanyl hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levorphanol, levophenacylmorphan, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, nalbuphene, normorphine, norpipanone, opium, oxycodone, oxymorphone, papaveretum, pentazocine, phenadoxone, phenomorphan, phenazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propoxyphene, sufentanil, tramadol, tilidine, α prodine, dextroporpoxyphene, propiram, profadol, phenampromide, thiambutene, pholcodeine, 3-transdimethylamino-4-phenyl-4-trans-carbethoxy-δ′-cyclohexene, 3-dimethylamino-O-(4-methoxyphenylcarbamoyl)-propiophenone oxime, (−)β-2′-hydroxy-2,9-dimethyl-5-phenyl-6,7-benzomorphan, (−)2′-hydroxy-2-(3-methyl-2-butenyl)-9-methyl-5-phenyl-6,7-benzomorphan, pirinitramide, (−)α-5,9-diethyl-2′-hydroxy-2-methyl-6,7-benzomorphan, ethyl 1-(2-dimethylaminoethyl)-4,5,6,7-tetrahydro-3-methyl-4-oxo-6-phenylindole-2-carboxylate, 1-benzoylmethyl-2,3-dimethyl-3-(m-hydroxyphenyl)-piperidine, N-allyl-7α-(1-(R)-hydroxy-1-methylbutyl)-6,14-endo-ethanotetrahydron ororipavine, (−)2′-hydroxy-2-methyl-6,7-benzomorphan, noracylmethadol, phenoperidine, α-dl-methadol, β-dl-methadol, α-1-methadol (215 mg.), β-dl-acetylmethadol, α-1-acetylmethadol, β-1-acetylmethadol, pharmaceutically acceptable salts thereof, stereoisomers thereof, ethers thereof, esters thereof, and mixtures thereof. 153. The composition of claim 140, wherein the controlled release agent is selected from the group consisting of naturally occurring or synthetic, anionic or nonionic, hydrophobic, hydrophilic rubbers, polymers, starch derivatives, cellulose derivatives, polysaccharides, carbomer, reseins, acrylics, proteins, vinyl-pyrrolidone-vinyl-acetate-copolymers, galactomannan and galactomannan derivatives, carrageenans and the like.
1,600
391
14,648,715
1,699
Disclosed herein are orally acceptable topical analgesic gels comprising a mixture of analgesic oils comprising (a) clove oil and/or eugenol, (b) a cooling agent, and (c) camphor, in an orally acceptable gel base, the gel base comprising an anionic polymer and a basic amino acid, and the analgesic gel providing controlled release of the mixture of analgesic oils following application to a tooth together with a methods of making and using the same.
1. An orally acceptable topical analgesic gel comprising: a mixture of analgesic oils comprising (a) clove oil and/or eugenol, (b) a cooling agent, and (c) camphor; and an orally acceptable gel base comprising: an anionic polymer and a basic amino acid; wherein the gel base provides controlled release of the mixture of analgesic oils following application to a tooth. 2. The analgesic gel of claim 1, wherein the gel base releases an effective amount of the analgesic oils to the tooth after 30 seconds. 3. The analgesic gel of claim 1, wherein the gel base releases an effective amount of the analgesic oils to the tooth after 60 seconds. 4. The analgesic gel of claim 1, wherein the gel base delivers an effective amount of the analgesic oils to the tooth for at least 5 minutes. 5. The analgesic gel of claim 1, wherein the gel base delivers an effective amount of the analgesic oils to the tooth for up to 120 minutes. 6. The analgesic gel of claim 1, wherein the mixture of analgesic oils comprises 1-10% by weight of the formulation. 7. The analgesic gel of claim 1, wherein the cooling agent is menthol. 8. The analgesic gel of claim 1, wherein the ratio of (a) to (b) to (c) is from about 8:1:1 to about 10:1:1. 9. The analgesic gel of claim 1, wherein the mixture of analgesic oils comprises clove oil:menthol:camphor in a ratio of 8:1:1 to about 10:1:1. 10. The analgesic gel of claim 1, wherein the basic amino acid is selected from arginine, lysine, citrullene, ornithine, creatine, histidine, diaminobutanoic acid, diaminoproprionic acid. 11. The analgesic gel of claim 1, wherein the basic amino acid is arginine. 12. The analgesic gel of claim 1, wherein the basic amino acid is present in an amount sufficient to partially or fully neutralize the anionic polymer. 13. The analgesic gel of claim 1, wherein anionic polymer comprises a crosslinked poly(acrylic acid). 14. The analgesic gel of claim 1, wherein the gel base comprises a methyl vinyl ether/maleic anhydride (PVM/MA) copolymer. 15. The analgesic gel of claim 1, wherein the gel base comprises one or more nonionic surfactants selected from poloxamers, polysorbates, and mixtures thereof. 16. The analgesic gel of claim 1, wherein the gel base has pH about 6 to about 8. 17. The analgesic gel of claim 1, wherein the gel base comprises one or more astringents. 18. The analgesic gel of claim 1, the gel base comprises a soluble potassium salt. 19. The analgesic gel of claim 1, wherein the gel base comprises a small particle occlusive agent. 20. The analgesic gel of claim 1, wherein the mixture of analgesic oils further comprises comprising a warming agent. 21. The gel of claim 1, wherein the gel base comprises: a) cross-linked poly(acrylic acid); b) propylene glycol; c) a nonionic surfactant selected from poloxamers, polysorbates, and mixtures thereof; and d) a neutralizing base. 22. A method of alleviating dental pain comprising administering an effective amount of an analgesic gel according to claim 1, to the affected area, wherein the composition is left on the affected area following application for at least sixty seconds. 23. (canceled) 24. A method of making an orally acceptable topical analgesic gel comprising a mixture of analgesic oils comprising (a) clove oil and/or eugenol, (b) a cooling agent, and (c) camphor; and an orally acceptable gel base comprising a cross-linked poly(acrylic acid) polymer, nonionic surfactants, a basic amino acid, and water, comprising: a) forming a water-in-oil emulsion wherein the oil phase comprises the mixture of analgesic oils, the water phase comprises the poly(acrylic acid) polymer and water, and the nonionic surfactants facilitate the emulsion formation, b) adding the basic amino acid to raise the pH of the emulsion thus formed to a level sufficient to ionize the carboxyl groups on the cross-linked poly(acrylic acid) polymer, thereby forming a stable gel.
Disclosed herein are orally acceptable topical analgesic gels comprising a mixture of analgesic oils comprising (a) clove oil and/or eugenol, (b) a cooling agent, and (c) camphor, in an orally acceptable gel base, the gel base comprising an anionic polymer and a basic amino acid, and the analgesic gel providing controlled release of the mixture of analgesic oils following application to a tooth together with a methods of making and using the same.1. An orally acceptable topical analgesic gel comprising: a mixture of analgesic oils comprising (a) clove oil and/or eugenol, (b) a cooling agent, and (c) camphor; and an orally acceptable gel base comprising: an anionic polymer and a basic amino acid; wherein the gel base provides controlled release of the mixture of analgesic oils following application to a tooth. 2. The analgesic gel of claim 1, wherein the gel base releases an effective amount of the analgesic oils to the tooth after 30 seconds. 3. The analgesic gel of claim 1, wherein the gel base releases an effective amount of the analgesic oils to the tooth after 60 seconds. 4. The analgesic gel of claim 1, wherein the gel base delivers an effective amount of the analgesic oils to the tooth for at least 5 minutes. 5. The analgesic gel of claim 1, wherein the gel base delivers an effective amount of the analgesic oils to the tooth for up to 120 minutes. 6. The analgesic gel of claim 1, wherein the mixture of analgesic oils comprises 1-10% by weight of the formulation. 7. The analgesic gel of claim 1, wherein the cooling agent is menthol. 8. The analgesic gel of claim 1, wherein the ratio of (a) to (b) to (c) is from about 8:1:1 to about 10:1:1. 9. The analgesic gel of claim 1, wherein the mixture of analgesic oils comprises clove oil:menthol:camphor in a ratio of 8:1:1 to about 10:1:1. 10. The analgesic gel of claim 1, wherein the basic amino acid is selected from arginine, lysine, citrullene, ornithine, creatine, histidine, diaminobutanoic acid, diaminoproprionic acid. 11. The analgesic gel of claim 1, wherein the basic amino acid is arginine. 12. The analgesic gel of claim 1, wherein the basic amino acid is present in an amount sufficient to partially or fully neutralize the anionic polymer. 13. The analgesic gel of claim 1, wherein anionic polymer comprises a crosslinked poly(acrylic acid). 14. The analgesic gel of claim 1, wherein the gel base comprises a methyl vinyl ether/maleic anhydride (PVM/MA) copolymer. 15. The analgesic gel of claim 1, wherein the gel base comprises one or more nonionic surfactants selected from poloxamers, polysorbates, and mixtures thereof. 16. The analgesic gel of claim 1, wherein the gel base has pH about 6 to about 8. 17. The analgesic gel of claim 1, wherein the gel base comprises one or more astringents. 18. The analgesic gel of claim 1, the gel base comprises a soluble potassium salt. 19. The analgesic gel of claim 1, wherein the gel base comprises a small particle occlusive agent. 20. The analgesic gel of claim 1, wherein the mixture of analgesic oils further comprises comprising a warming agent. 21. The gel of claim 1, wherein the gel base comprises: a) cross-linked poly(acrylic acid); b) propylene glycol; c) a nonionic surfactant selected from poloxamers, polysorbates, and mixtures thereof; and d) a neutralizing base. 22. A method of alleviating dental pain comprising administering an effective amount of an analgesic gel according to claim 1, to the affected area, wherein the composition is left on the affected area following application for at least sixty seconds. 23. (canceled) 24. A method of making an orally acceptable topical analgesic gel comprising a mixture of analgesic oils comprising (a) clove oil and/or eugenol, (b) a cooling agent, and (c) camphor; and an orally acceptable gel base comprising a cross-linked poly(acrylic acid) polymer, nonionic surfactants, a basic amino acid, and water, comprising: a) forming a water-in-oil emulsion wherein the oil phase comprises the mixture of analgesic oils, the water phase comprises the poly(acrylic acid) polymer and water, and the nonionic surfactants facilitate the emulsion formation, b) adding the basic amino acid to raise the pH of the emulsion thus formed to a level sufficient to ionize the carboxyl groups on the cross-linked poly(acrylic acid) polymer, thereby forming a stable gel.
1,600
392
10,605,669
1,653
The claimed subject matter comprises a device to collect and preserve cells comprising of: (1) a collection container comprised of a tube having an open end and a closed end, a closure in the open end of the tube, a vacuum drawn to a predetermined level inside the container; and (2) compounds including an anticoagulant agent and a fixative agent, wherein the compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, and thereby allowing said cells to be directly analyzed by a flow cytometer without further treatment. The claimed subject matter further comprises of a method of making a collection device for cells comprising of: (1) providing a tube having an open end and a closed end; (2) preloading compounds including: an anticoagulant agent, and a fixative agent into the tube, wherein the compounds are in a sufficient amount to preserve the cells' original morphology and antigenic sites without significant dilution of the cells, and thereby allowing the cells to be directly analyzed by a flow cytometer without further treatment; (3) inserting a closure into the open end of the tube; and (4) drawing a vacuum inside the tube to a predetermined level to form the collection device.
1. A method of making a collection device for cells comprising of: providing a tube having an open end and a closed end; preloading compounds including: an anticoagulant agent, and a fixative agent into said tube, said fixative agent selected from the group consisting of: diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-1,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1aza-3,7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1aza-3,7dioxabicyclo [3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1aza-3, 7dioxabi cyclo [3.3.0]octane, quaternary adamantine and combinations thereof; wherein said compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, and thereby allowing said cells to be directly analyzed by a flow cytometer without further treatment; inserting a closure into said open end of said tube; and drawing a vacuum inside said tube to a predetermined level to form said collection device. 2. The method of claim 1, wherein said anticoagulant agent is selected from the group consisting of ethylene diamine tetra acetic acid (EDTA), salts of EDTA, ethylene glycol tetra acetic acid (EGTA), salts of EGTA, hirudin, heparin, citric acid, salts of citric acid, oxalic acid, salts of oxalic acid, and a combination thereof. 3. The method of claim 1, wherein concentration of said fixative agent is less than about 1 g/ml. 4. The method of claim 1, wherein concentration of said anticoagulant agent is less than about 0.3 g/ml. 5. The method of claim 1, wherein said preloading step includes preloading a polyacylic acid into said tube. 6. The method of claim 1, wherein ratio of said compounds and a final composition comprising said cells and said compounds is less than about 2:100. 7. The method of claim 1, wherein said cells are selected from the group consisting of epithelial cells, bone marrow, spinal fluid, abnormal tissue sample in a cellular suspension, and a combination thereof. 8. The method of claim 1, further comprising of sterilizing said compounds before said compounds are preloaded into said tube. 9. The method of claim 1, further comprising of sterilizing at least all surface areas of said tube and said closure that can come into physical contact with said collected and preserved cells before said compounds are preloaded into said tube. 10. The method of claim 1, further comprising of providing at least one component selected from the group consisting of an alcohol swab, a gauze, a tube holder, a tourniquet, a glove, other cell collection tube, a needle, a lancet, adhesive strip, syringe, a test strip, a strip containing reagents for cell analysis, a packaging means for storing said at least one component and said collection device to form a kit, and a packaging means for transporting said collection device. 11. The method of claim 1, wherein said preloading step further comprises of freeze drying said compounds inside said tube. 12. The method of claim 1, further comprising of screening said collected and preserved cells using an instrument selected from the group consisting of: a flow cytometer, a hematology analyzer, H3 by Bayer Corporation, the Beckman Coulter STKS System, the Beckman Coulter Gen-S System, the Abbott Cell-Dyn 4000 Hematology System, Bayer ADVIA 120 System, the Sysmex XE2100 System, and other analyzers and a combination thereof. 13. The method of claim 1, further comprising of screening said collected and preserved cells for a purpose selected from the group consisting of: HIV, HPV, hepatitis, leukemia, cancer, and a combination thereof. 14. A collection device for cells comprising of: a collection container comprising of a tube having an open end and a closed end, a closure in said open end of said tube, a vacuum drawn to a predetermined level inside said container; and compounds including an anticoagulant agent and a fixative agent selected from the group consisting of: diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-1,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1aza-3, 7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1aza-3,7dioxabicyclo [3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1aza-3, 7dioxabi cyclo [3.3.0]octane, quaternary adamantine and combinations thereof, inside said tube, wherein said compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, and thereby allowing said cells to be directly analyzed by a flow cytometer without further treatment. 15. The device of claim 14, wherein said anticoagulant agent is selected from the group consisting of ethylene diamine tetra acetic acid (EDTA), salts of EDTA, ethylene glycol tetra acetic acid (EGTA), salts of EGTA, hirudin, heparin, citric acid, salts of citric acid, oxalic acid, salts of oxalic acid, and a combination thereof. 16. The device of claim 14, wherein concentration of said fixative agent is less than about 1 g/ml. 17. The device of claim 14, wherein concentration of said anticoagulant agent is less than about 0.3 g/ml. 18. The device of claim 14, wherein compounds further includes a polyacrylic acid. 19. The device of claim 14, wherein ratio of said compounds and a final composition comprising said cells and said compounds is less than about 2:100. 20. The device of claim 14, wherein said cells are selected from the group consisting of epithelial cells, bone marrow, spinal fluid, abnormal tissue sample in a cellular suspension, and a combination thereof. 21. The device of claim 14, wherein said compounds are sterile. 22. The device of claim 14, wherein at least all surface areas of said tube and said closure that can come into physical contact with said cells are sterile. 23. A kit comprising the device of claim 14 and further comprising of at least one component selected from the group consisting of an alcohol swab, a gauze, a tube holder, a tourniquet, a glove, other cell collection tube, a needle, a lancet, adhesive strip, syringe, a test strip, a strip containing reagents for cell analysis, a packaging means for storing said at least one component and said collection device to form a kit, and a packaging means for transporting said collection device. 24. The device of claim 14 wherein compounds contained in said tube are freeze dried. 25. The device of claim 14 wherein said device is used along with an instrument selected from the group consisting of: a flow cytometer, a hematology analyzer, H3 by Bayer Corporation, the Beckman Coulter STKS System, the Beckman Coulter Gen-S System, the Abbott Cell-Dyn 4000 Hematology System, Bayer ADVIA 120 System, the Sysmex XE2100 System, and a combination thereof to provide screening of said cells. 26. The device of claim 14 wherein said device is used in screening said cells for a purpose selected from the group consisting of: HIV, HPV, hepatitis, leukemia, cancer, and a combination thereof. 27. A method for transporting cells for analysis, said method comprising: providing a collection container under vacuum; and containing compounds including an anticoagulant agent and a fixative agent selected from the group consisting of: diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-1,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1aza-3, 7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1aza-3,7dioxabicyclo [3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1aza-3, 7dioxabi cyclo [3.3.0]octane, quaternary adamantine and combinations thereofinside said tube, wherein said compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, collecting said cells in said collection container and; transporting said cells for analysis.
The claimed subject matter comprises a device to collect and preserve cells comprising of: (1) a collection container comprised of a tube having an open end and a closed end, a closure in the open end of the tube, a vacuum drawn to a predetermined level inside the container; and (2) compounds including an anticoagulant agent and a fixative agent, wherein the compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, and thereby allowing said cells to be directly analyzed by a flow cytometer without further treatment. The claimed subject matter further comprises of a method of making a collection device for cells comprising of: (1) providing a tube having an open end and a closed end; (2) preloading compounds including: an anticoagulant agent, and a fixative agent into the tube, wherein the compounds are in a sufficient amount to preserve the cells' original morphology and antigenic sites without significant dilution of the cells, and thereby allowing the cells to be directly analyzed by a flow cytometer without further treatment; (3) inserting a closure into the open end of the tube; and (4) drawing a vacuum inside the tube to a predetermined level to form the collection device.1. A method of making a collection device for cells comprising of: providing a tube having an open end and a closed end; preloading compounds including: an anticoagulant agent, and a fixative agent into said tube, said fixative agent selected from the group consisting of: diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-1,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1aza-3,7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1aza-3,7dioxabicyclo [3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1aza-3, 7dioxabi cyclo [3.3.0]octane, quaternary adamantine and combinations thereof; wherein said compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, and thereby allowing said cells to be directly analyzed by a flow cytometer without further treatment; inserting a closure into said open end of said tube; and drawing a vacuum inside said tube to a predetermined level to form said collection device. 2. The method of claim 1, wherein said anticoagulant agent is selected from the group consisting of ethylene diamine tetra acetic acid (EDTA), salts of EDTA, ethylene glycol tetra acetic acid (EGTA), salts of EGTA, hirudin, heparin, citric acid, salts of citric acid, oxalic acid, salts of oxalic acid, and a combination thereof. 3. The method of claim 1, wherein concentration of said fixative agent is less than about 1 g/ml. 4. The method of claim 1, wherein concentration of said anticoagulant agent is less than about 0.3 g/ml. 5. The method of claim 1, wherein said preloading step includes preloading a polyacylic acid into said tube. 6. The method of claim 1, wherein ratio of said compounds and a final composition comprising said cells and said compounds is less than about 2:100. 7. The method of claim 1, wherein said cells are selected from the group consisting of epithelial cells, bone marrow, spinal fluid, abnormal tissue sample in a cellular suspension, and a combination thereof. 8. The method of claim 1, further comprising of sterilizing said compounds before said compounds are preloaded into said tube. 9. The method of claim 1, further comprising of sterilizing at least all surface areas of said tube and said closure that can come into physical contact with said collected and preserved cells before said compounds are preloaded into said tube. 10. The method of claim 1, further comprising of providing at least one component selected from the group consisting of an alcohol swab, a gauze, a tube holder, a tourniquet, a glove, other cell collection tube, a needle, a lancet, adhesive strip, syringe, a test strip, a strip containing reagents for cell analysis, a packaging means for storing said at least one component and said collection device to form a kit, and a packaging means for transporting said collection device. 11. The method of claim 1, wherein said preloading step further comprises of freeze drying said compounds inside said tube. 12. The method of claim 1, further comprising of screening said collected and preserved cells using an instrument selected from the group consisting of: a flow cytometer, a hematology analyzer, H3 by Bayer Corporation, the Beckman Coulter STKS System, the Beckman Coulter Gen-S System, the Abbott Cell-Dyn 4000 Hematology System, Bayer ADVIA 120 System, the Sysmex XE2100 System, and other analyzers and a combination thereof. 13. The method of claim 1, further comprising of screening said collected and preserved cells for a purpose selected from the group consisting of: HIV, HPV, hepatitis, leukemia, cancer, and a combination thereof. 14. A collection device for cells comprising of: a collection container comprising of a tube having an open end and a closed end, a closure in said open end of said tube, a vacuum drawn to a predetermined level inside said container; and compounds including an anticoagulant agent and a fixative agent selected from the group consisting of: diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-1,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1aza-3, 7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1aza-3,7dioxabicyclo [3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1aza-3, 7dioxabi cyclo [3.3.0]octane, quaternary adamantine and combinations thereof, inside said tube, wherein said compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, and thereby allowing said cells to be directly analyzed by a flow cytometer without further treatment. 15. The device of claim 14, wherein said anticoagulant agent is selected from the group consisting of ethylene diamine tetra acetic acid (EDTA), salts of EDTA, ethylene glycol tetra acetic acid (EGTA), salts of EGTA, hirudin, heparin, citric acid, salts of citric acid, oxalic acid, salts of oxalic acid, and a combination thereof. 16. The device of claim 14, wherein concentration of said fixative agent is less than about 1 g/ml. 17. The device of claim 14, wherein concentration of said anticoagulant agent is less than about 0.3 g/ml. 18. The device of claim 14, wherein compounds further includes a polyacrylic acid. 19. The device of claim 14, wherein ratio of said compounds and a final composition comprising said cells and said compounds is less than about 2:100. 20. The device of claim 14, wherein said cells are selected from the group consisting of epithelial cells, bone marrow, spinal fluid, abnormal tissue sample in a cellular suspension, and a combination thereof. 21. The device of claim 14, wherein said compounds are sterile. 22. The device of claim 14, wherein at least all surface areas of said tube and said closure that can come into physical contact with said cells are sterile. 23. A kit comprising the device of claim 14 and further comprising of at least one component selected from the group consisting of an alcohol swab, a gauze, a tube holder, a tourniquet, a glove, other cell collection tube, a needle, a lancet, adhesive strip, syringe, a test strip, a strip containing reagents for cell analysis, a packaging means for storing said at least one component and said collection device to form a kit, and a packaging means for transporting said collection device. 24. The device of claim 14 wherein compounds contained in said tube are freeze dried. 25. The device of claim 14 wherein said device is used along with an instrument selected from the group consisting of: a flow cytometer, a hematology analyzer, H3 by Bayer Corporation, the Beckman Coulter STKS System, the Beckman Coulter Gen-S System, the Abbott Cell-Dyn 4000 Hematology System, Bayer ADVIA 120 System, the Sysmex XE2100 System, and a combination thereof to provide screening of said cells. 26. The device of claim 14 wherein said device is used in screening said cells for a purpose selected from the group consisting of: HIV, HPV, hepatitis, leukemia, cancer, and a combination thereof. 27. A method for transporting cells for analysis, said method comprising: providing a collection container under vacuum; and containing compounds including an anticoagulant agent and a fixative agent selected from the group consisting of: diazolidinyl urea, imidazolidinyl urea, dimethoylol-5,5dimethylhydantoin, dimethylol urea, 2-bromo-2.-nitropropane-1,3-diol, oxazolidines, sodium hydroxymethyl glycinate, 5-hydroxymethoxymethyl-1-1aza-3, 7-dioxabicyclo [3.3.0]octane, 5-hydroxymethyl-1-1aza-3,7dioxabicyclo [3.3.0]octane, 5-hydroxypoly[methyleneoxy]methyl-1-1aza-3, 7dioxabi cyclo [3.3.0]octane, quaternary adamantine and combinations thereofinside said tube, wherein said compounds are in a sufficient amount to preserve said cells' original morphology and antigenic sites without significant dilution of said cells, collecting said cells in said collection container and; transporting said cells for analysis.
1,600
393
16,122,360
1,616
Abuse-resistant therapeutic pharmaceutical formulations include a cationic drug and at least one anionic polymer. Particular arrangements of the cationic drug and the at least one anionic polymer prevent the cationic drug from being extracted from the formulations in solvents and conditions commonly used by drug abusers attempting to isolate the cationic drug from its formulation.
1. An abuse-resistant therapeutic pharmaceutical formulation comprising: a cationic drug and at least one anionic polymer; wherein (i) the cationic drug is physically blended with but not ionically bound to the at least one anionic polymer, or (ii) more than 40% of the cationic drug is ionically bound (complexed) to the at least one anionic polymer; wherein the formulation prevents the cationic drug from being extracted from the formulation in a solvent selected from the group consisting of water, hydroalcohol solutions, pH 3 solutions, acetic acid solutions, and saline, at solution temperatures of 20-90° C. 2. The pharmaceutical formulation of claim 1, wherein the at least one anionic polymer comprises a poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer. 3. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with an allyl ether of pentaerythritol. 4. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with an allyl ether of sucrose. 5. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with an allyl ether of propylene. 6. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with a vinyl crosslinker. 7. The pharmaceutical formulation of claim 2, wherein the cationic drug is physically blended with but not ionically bound to the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer and the formulation further comprises an alkalinizing agent, wherein the weight ratio of the alkalizing agent to the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is between about 2% to 20%. 8. The pharmaceutical formulation of claim 7, wherein the weight ratio of the alkalizing agent to the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is between about 2% to 10%. 9. The pharmaceutical formulation of claim 7, wherein the weight ratio of the alkalizing agent to the poly(acrylic acid) homopolymer, copolymer or interpolymer is about 3.5%. 10. The pharmaceutical formulation of claim 7, wherein the alkalinizing agent is a bicarbonate salt. 11. The pharmaceutical formulation of claim 1, wherein the cationic drug is ionically bound (complexed) to the at least one anionic polymer. 12. The pharmaceutical formulation of claim 11, wherein the at least one anionic polymer comprises a poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer. 13. The pharmaceutical formulation of claim 12, wherein the cationic drug-poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer complex is made by reacting the cationic drug and the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer in an aqueous solution at a pH range of greater than the pKa−1 of the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer and lower than the pKa+1 of the cationic drug. 14. The pharmaceutical formulation of claim 13, wherein the aqueous solution comprises an alkalinizing agent that causes the pH range of the aqueous solution to be greater than the pKa−1 of the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer and lower than the pKa+1 of the cationic drug. 15. The pharmaceutical formulation of claim 14, wherein the alkalinizing agent is a bicarbonate salt.
Abuse-resistant therapeutic pharmaceutical formulations include a cationic drug and at least one anionic polymer. Particular arrangements of the cationic drug and the at least one anionic polymer prevent the cationic drug from being extracted from the formulations in solvents and conditions commonly used by drug abusers attempting to isolate the cationic drug from its formulation.1. An abuse-resistant therapeutic pharmaceutical formulation comprising: a cationic drug and at least one anionic polymer; wherein (i) the cationic drug is physically blended with but not ionically bound to the at least one anionic polymer, or (ii) more than 40% of the cationic drug is ionically bound (complexed) to the at least one anionic polymer; wherein the formulation prevents the cationic drug from being extracted from the formulation in a solvent selected from the group consisting of water, hydroalcohol solutions, pH 3 solutions, acetic acid solutions, and saline, at solution temperatures of 20-90° C. 2. The pharmaceutical formulation of claim 1, wherein the at least one anionic polymer comprises a poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer. 3. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with an allyl ether of pentaerythritol. 4. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with an allyl ether of sucrose. 5. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with an allyl ether of propylene. 6. The pharmaceutical formulation of claim 2, wherein the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is crosslinked with a vinyl crosslinker. 7. The pharmaceutical formulation of claim 2, wherein the cationic drug is physically blended with but not ionically bound to the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer and the formulation further comprises an alkalinizing agent, wherein the weight ratio of the alkalizing agent to the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is between about 2% to 20%. 8. The pharmaceutical formulation of claim 7, wherein the weight ratio of the alkalizing agent to the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer is between about 2% to 10%. 9. The pharmaceutical formulation of claim 7, wherein the weight ratio of the alkalizing agent to the poly(acrylic acid) homopolymer, copolymer or interpolymer is about 3.5%. 10. The pharmaceutical formulation of claim 7, wherein the alkalinizing agent is a bicarbonate salt. 11. The pharmaceutical formulation of claim 1, wherein the cationic drug is ionically bound (complexed) to the at least one anionic polymer. 12. The pharmaceutical formulation of claim 11, wherein the at least one anionic polymer comprises a poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer. 13. The pharmaceutical formulation of claim 12, wherein the cationic drug-poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer complex is made by reacting the cationic drug and the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer in an aqueous solution at a pH range of greater than the pKa−1 of the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer and lower than the pKa+1 of the cationic drug. 14. The pharmaceutical formulation of claim 13, wherein the aqueous solution comprises an alkalinizing agent that causes the pH range of the aqueous solution to be greater than the pKa−1 of the poly(acrylic acid) homopolymer, copolymer, terpolymer, or interpolymer and lower than the pKa+1 of the cationic drug. 15. The pharmaceutical formulation of claim 14, wherein the alkalinizing agent is a bicarbonate salt.
1,600
394
15,340,538
1,699
The present application provides for a compound of Formula IV, or a pharmaceutically acceptable salt, solvate, and/or ester thereof, compositions containing such compounds, therapeutic methods that include the administration of such compounds, and therapeutic methods and include the administration of such compounds with at least one additional therapeutic agent.
1-41. (canceled) 42. An in vivo metabolic product of Compound P: 43. The metabolic product of claim 42 which is substantially isolated. 44. A method of preparing the in vivo metabolic product of claim 42 comprising contacting Compound P with a mammal for a period of time sufficient to yield the in vivo metabolic product. 45. The method of claim 44 wherein the mammal is a rat, mouse, guinea pig, monkey, or human. 46. The method of claim 44 wherein Compound P is orally administered to the mammal. 47. The method of claim 44 wherein Compound P is radiolabeled. 48. The method of claim 47 wherein the radiolabel comprises C14 or H3. 49. A method of determining the optimal therapeutic dose of Compound P in a mammal comprising measuring the quantity of the in vivo metabolic product of claim 42 produced upon contacting the mammal with a radiolabeled Compound P and correlating the measured quantity of the in vivo metabolic product with therapeutic effectiveness of Compound P. 50. A prodrug of Compound P: 51. The prodrug of claim 50 which is an ester of Compound P. 52. A pharmaceutical composition comprising the prodrug of claim 50, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient. 53. A method of improving the pharmacokinetics of a drug, comprising administering to a patient treated with said drug, a therapeutically effective amount of the prodrug of claim 50, or a pharmaceutically acceptable salt thereof. 54. A solvate of Compound P: 55. A pharmaceutical composition comprising the solvate of claim 54, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient. 56. A method of improving the pharmacokinetics of a drug, comprising administering to a patient treated with said drug, a therapeutically effective amount of the solvate of claim 54, or a pharmaceutically acceptable salt thereof.
The present application provides for a compound of Formula IV, or a pharmaceutically acceptable salt, solvate, and/or ester thereof, compositions containing such compounds, therapeutic methods that include the administration of such compounds, and therapeutic methods and include the administration of such compounds with at least one additional therapeutic agent.1-41. (canceled) 42. An in vivo metabolic product of Compound P: 43. The metabolic product of claim 42 which is substantially isolated. 44. A method of preparing the in vivo metabolic product of claim 42 comprising contacting Compound P with a mammal for a period of time sufficient to yield the in vivo metabolic product. 45. The method of claim 44 wherein the mammal is a rat, mouse, guinea pig, monkey, or human. 46. The method of claim 44 wherein Compound P is orally administered to the mammal. 47. The method of claim 44 wherein Compound P is radiolabeled. 48. The method of claim 47 wherein the radiolabel comprises C14 or H3. 49. A method of determining the optimal therapeutic dose of Compound P in a mammal comprising measuring the quantity of the in vivo metabolic product of claim 42 produced upon contacting the mammal with a radiolabeled Compound P and correlating the measured quantity of the in vivo metabolic product with therapeutic effectiveness of Compound P. 50. A prodrug of Compound P: 51. The prodrug of claim 50 which is an ester of Compound P. 52. A pharmaceutical composition comprising the prodrug of claim 50, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient. 53. A method of improving the pharmacokinetics of a drug, comprising administering to a patient treated with said drug, a therapeutically effective amount of the prodrug of claim 50, or a pharmaceutically acceptable salt thereof. 54. A solvate of Compound P: 55. A pharmaceutical composition comprising the solvate of claim 54, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient. 56. A method of improving the pharmacokinetics of a drug, comprising administering to a patient treated with said drug, a therapeutically effective amount of the solvate of claim 54, or a pharmaceutically acceptable salt thereof.
1,600
395
13,905,015
1,618
Particulate constructs stabilized by amphiphilic copolymers and comprising at least one active coupled to a hydrophobic moiety provide sustained release of the active in both in vitro and in vivo environments.
1. A composition comprising particles which would result from forming said particle from a mixture of a) an amphiphilic stabilizer, b) a conjugate of the formula (active−linker)n−hydrophobic moiety  (1) wherein n is an integer of 1-100; and wherein “active” refers to at least a first therapeutic agent; “linker” is a covalent bond, a divalent residue of an organic molecule or a chelator which comprises a bond that is selectively cleavable to control release of said active; and “hydrophobic moiety” refers to the residue of an organic molecule that is insoluble in aqueous solution; and c) a second therapeutic agent different from the first therapeutic agent. 2. The composition of claim 1, wherein n is an integer of 2-100. 3. The composition of claim 1, wherein the hydrophobic moiety is vitamin E, vitamin A or vitamin K, or a retinol; or wherein the hydrophobic moiety is polycaprolactone, polylactic acid, polystyrene, polybutadiene, polycaproic acid, polymethylbenzylate, poly(D,L-lactide), poly(D,L-lactide-co-glycolide), poly(glycolide), poly(hydroxybutyrate), poly(alkylcarbonate) poly(orthoesters), polyesters, poly(hydroxyvaleric acid), or copolymers thereof. 4. The composition of claim 1, wherein the amphiphilic stabilizer is methoxypolyethylene glycol (mPEG)-polycaprolactone (PCL), mPEG-polystyrene, mPEG-polybutadiene, mPEG-polylactate, block copolymers of polyethylene oxide, and polypropylene oxide, or block copolymers of polyethylene oxide and polybutylene oxide. 5. The composition of claim 1, wherein the linker is the residue of a divalent organic molecule, and said residue of a divalent organic molecule comprises a site for hydrolytic cleavage and/or a site for enzymatic cleavage or a site for photolytic cleavage, or wherein the linker is a chelator. 6. The composition of claim 1, wherein said first and second therapeutic agents are maintained at non-antagonistic ratio when administered to a subject. 7. The composition of claim 1, wherein the particulate constructs comprise 103-107 conjugates of formula (1). 8. The composition of claim 1, wherein the particulate constructs have an average diameter less than 5μ. 9. The composition of claim 1, which is in the form of an emulsion. 10. The composition of claim 1 wherein said first and second therapeutic agents are coupled in conjugates of formula (1) to separate hydrophobic moieties. 11. The composition of claim 10 wherein the separate hydrophobic moieties are different. 12. The composition of claim 10 wherein said first and second therapeutic agent conjugates comprise a paclitaxel polylactic acid conjugate of formula (1) and a cis-diaminedichloroplatinum(II) polylactic acid diacid diethyl ester terminated polymer conjugate of formula (1). 13. The composition of claim 1 wherein said first therapeutic agent is present in the form of formula (1) and the second therapeutic agent is in unbound form. 14. The composition of claim 13 wherein the agent in the form of formula (1) is cis-diaminedichloroplatinum(II) and the agent in unbound form is paclitaxel. 15. The composition of claim 1 wherein said first and second therapeutic agents are coupled in a conjugate of formula (1) to the same hydrophobic moiety. 16. A method to administer a combination treatment to a subject, which method comprises administering a composition of claim 6 to a subject in need of such treatment.
Particulate constructs stabilized by amphiphilic copolymers and comprising at least one active coupled to a hydrophobic moiety provide sustained release of the active in both in vitro and in vivo environments.1. A composition comprising particles which would result from forming said particle from a mixture of a) an amphiphilic stabilizer, b) a conjugate of the formula (active−linker)n−hydrophobic moiety  (1) wherein n is an integer of 1-100; and wherein “active” refers to at least a first therapeutic agent; “linker” is a covalent bond, a divalent residue of an organic molecule or a chelator which comprises a bond that is selectively cleavable to control release of said active; and “hydrophobic moiety” refers to the residue of an organic molecule that is insoluble in aqueous solution; and c) a second therapeutic agent different from the first therapeutic agent. 2. The composition of claim 1, wherein n is an integer of 2-100. 3. The composition of claim 1, wherein the hydrophobic moiety is vitamin E, vitamin A or vitamin K, or a retinol; or wherein the hydrophobic moiety is polycaprolactone, polylactic acid, polystyrene, polybutadiene, polycaproic acid, polymethylbenzylate, poly(D,L-lactide), poly(D,L-lactide-co-glycolide), poly(glycolide), poly(hydroxybutyrate), poly(alkylcarbonate) poly(orthoesters), polyesters, poly(hydroxyvaleric acid), or copolymers thereof. 4. The composition of claim 1, wherein the amphiphilic stabilizer is methoxypolyethylene glycol (mPEG)-polycaprolactone (PCL), mPEG-polystyrene, mPEG-polybutadiene, mPEG-polylactate, block copolymers of polyethylene oxide, and polypropylene oxide, or block copolymers of polyethylene oxide and polybutylene oxide. 5. The composition of claim 1, wherein the linker is the residue of a divalent organic molecule, and said residue of a divalent organic molecule comprises a site for hydrolytic cleavage and/or a site for enzymatic cleavage or a site for photolytic cleavage, or wherein the linker is a chelator. 6. The composition of claim 1, wherein said first and second therapeutic agents are maintained at non-antagonistic ratio when administered to a subject. 7. The composition of claim 1, wherein the particulate constructs comprise 103-107 conjugates of formula (1). 8. The composition of claim 1, wherein the particulate constructs have an average diameter less than 5μ. 9. The composition of claim 1, which is in the form of an emulsion. 10. The composition of claim 1 wherein said first and second therapeutic agents are coupled in conjugates of formula (1) to separate hydrophobic moieties. 11. The composition of claim 10 wherein the separate hydrophobic moieties are different. 12. The composition of claim 10 wherein said first and second therapeutic agent conjugates comprise a paclitaxel polylactic acid conjugate of formula (1) and a cis-diaminedichloroplatinum(II) polylactic acid diacid diethyl ester terminated polymer conjugate of formula (1). 13. The composition of claim 1 wherein said first therapeutic agent is present in the form of formula (1) and the second therapeutic agent is in unbound form. 14. The composition of claim 13 wherein the agent in the form of formula (1) is cis-diaminedichloroplatinum(II) and the agent in unbound form is paclitaxel. 15. The composition of claim 1 wherein said first and second therapeutic agents are coupled in a conjugate of formula (1) to the same hydrophobic moiety. 16. A method to administer a combination treatment to a subject, which method comprises administering a composition of claim 6 to a subject in need of such treatment.
1,600
396
15,527,103
1,617
A pharmaceutical composition for aerosol administration and method of preventing or treating a pulmonary disease or condition in a subject with a SphK1 inhibitor are provided.
1. A method of preventing or treating a pulmonary disease or condition in a subject comprising administering to a subject in need of treatment an effective amount of a SphK1 inhibitor to prevent or treat the subject's pulmonary disease or condition, wherein the SphK1 inhibitor (a) exhibits at least a 20-fold greater selectivity for SphK1 than SphK2; and (b) is cell permeable, has an IC50 value of less than 10 μM, or has a Ki of less than 10 μM, or a combination thereof. 2. The method of claim 1, wherein the pulmonary disease or condition comprises pulmonary hypertension, adult respiratory distress syndrome, restrictive lung disease, chronic obstructive pulmonary disease, bronchiectasis, bronchiolectasis, bronchiolitis, bronchitis, emphysema, a diffuse interstitial or infiltrative lung disease, serofibrinous pleuritis, suppurative pleuritis, hemorrhagic pleuritis, a pleural effusion, idiopathic pulmonary fibrosis, hyperoxia or oxygen-induced lung injury, injury due to drug or chemotherapeutic toxicity, radiation-induced injury, or chemical injury. 3. The method of claim 1, wherein the SphK1 inhibitor comprises [(2R)-1-[[4-[[3-(benzenesulfonylmethyl)-5-methylphenoxy]methyl]phenyl]methyl] pyrrolidin-2-yl] methanol (PF-543) or an analog, derivative or pharmaceutically acceptable salt thereof. 4. The method of claim 3, wherein the pharmaceutically acceptable salt is a citric acid salt. 5. The method of claim 1, wherein said SphK1 inhibitor is administered to the lungs of the subject. 6. A method of treating oxygen-induced lung injury in a subject comprising administering to a subject in need of treatment an effective amount of [(2R)-1-[[4-[[3-(benzenesulfonylmethyl)-5-methylphenoxy] methyl]phenyl]methyl]pyrrolidin-2-yl]methanol (PF-543), or an analog, derivative or pharmaceutically acceptable salt thereof, to treat the subject's oxygen-induced lung injury. 7. The method of claim 6, wherein the pharmaceutically acceptable salt is a citric acid salt. 8. The method of claim 6, wherein the PF-543, or analog, derivative or pharmaceutically acceptable salt thereof, is administered to the lungs of the subject. 9. A pharmaceutical composition for aerosol administration comprising one or more propellants and [(2R)-1-[[4-[[3-(benzenesulfonylmethyl)-5-methylphenoxy] methyl]phenyl]methyl]pyrrolidin-2-yl]methanol (PF-543) or an analog, derivative or pharmaceutically acceptable salt thereof. 10. A metered dose inhaler for aerosol administration comprising an aerosol canister equipped with a metered dose valve, wherein the aerosol canister is filled with the pharmaceutical composition of claim 9.
A pharmaceutical composition for aerosol administration and method of preventing or treating a pulmonary disease or condition in a subject with a SphK1 inhibitor are provided.1. A method of preventing or treating a pulmonary disease or condition in a subject comprising administering to a subject in need of treatment an effective amount of a SphK1 inhibitor to prevent or treat the subject's pulmonary disease or condition, wherein the SphK1 inhibitor (a) exhibits at least a 20-fold greater selectivity for SphK1 than SphK2; and (b) is cell permeable, has an IC50 value of less than 10 μM, or has a Ki of less than 10 μM, or a combination thereof. 2. The method of claim 1, wherein the pulmonary disease or condition comprises pulmonary hypertension, adult respiratory distress syndrome, restrictive lung disease, chronic obstructive pulmonary disease, bronchiectasis, bronchiolectasis, bronchiolitis, bronchitis, emphysema, a diffuse interstitial or infiltrative lung disease, serofibrinous pleuritis, suppurative pleuritis, hemorrhagic pleuritis, a pleural effusion, idiopathic pulmonary fibrosis, hyperoxia or oxygen-induced lung injury, injury due to drug or chemotherapeutic toxicity, radiation-induced injury, or chemical injury. 3. The method of claim 1, wherein the SphK1 inhibitor comprises [(2R)-1-[[4-[[3-(benzenesulfonylmethyl)-5-methylphenoxy]methyl]phenyl]methyl] pyrrolidin-2-yl] methanol (PF-543) or an analog, derivative or pharmaceutically acceptable salt thereof. 4. The method of claim 3, wherein the pharmaceutically acceptable salt is a citric acid salt. 5. The method of claim 1, wherein said SphK1 inhibitor is administered to the lungs of the subject. 6. A method of treating oxygen-induced lung injury in a subject comprising administering to a subject in need of treatment an effective amount of [(2R)-1-[[4-[[3-(benzenesulfonylmethyl)-5-methylphenoxy] methyl]phenyl]methyl]pyrrolidin-2-yl]methanol (PF-543), or an analog, derivative or pharmaceutically acceptable salt thereof, to treat the subject's oxygen-induced lung injury. 7. The method of claim 6, wherein the pharmaceutically acceptable salt is a citric acid salt. 8. The method of claim 6, wherein the PF-543, or analog, derivative or pharmaceutically acceptable salt thereof, is administered to the lungs of the subject. 9. A pharmaceutical composition for aerosol administration comprising one or more propellants and [(2R)-1-[[4-[[3-(benzenesulfonylmethyl)-5-methylphenoxy] methyl]phenyl]methyl]pyrrolidin-2-yl]methanol (PF-543) or an analog, derivative or pharmaceutically acceptable salt thereof. 10. A metered dose inhaler for aerosol administration comprising an aerosol canister equipped with a metered dose valve, wherein the aerosol canister is filled with the pharmaceutical composition of claim 9.
1,600
397
14,651,356
1,611
Disclosed are extruded pesticide granules that when mixed with water form a near micro-emulsion that is highly stable. The extruded pesticide granules include at least one pesticide active ingredient, a non-ionic surfactant, and a carrier, which is desirably urea powder or urea pearl. Processes for making the extruded pesticide granules are also disclosed.
1. An extruded pesticide granular composition comprising urea, a non-ionic surfactant, a pesticide active ingredient, and water. 2. The extruded composition of claim 1 wherein the urea is in the form of urea pearls. 3. The extruded composition of claim 1 wherein the non-ionic surfactant is a block copolymer having a molecular weight of from about 10,000 to about 15,000 Daltons. 4. The extruded composition of claim 1 wherein the pesticide active ingredient is alpha cypermethrin. 5. The extruded composition of claim 1 wherein the pesticide active ingredient is dinotefuran. 6. The extruded composition of claim 1 wherein the pesticide active ingredient is water-soluble. 7. The extruded composition of claim 1 wherein the urea is present in the composition in an amount of from about 50 wt. % to about 95 wt. %. 8. The extruded composition of claim 1 wherein the urea is present in the composition in an amount of from about 70 wt. % to about 80 wt. %. 9. The extruded composition of claim 1 wherein the non-ionic surfactant is present in an amount of from about 5 wt. % to about 20 wt. %. 10. The extruded composition of claim 1 wherein the pesticide active ingredient is present in an amount of from about 5 wt. % to about 20 wt. %. 11. The extruded composition of claim 1 wherein the composition comprises less than 1% water. 12. The extruded composition of claim 1 wherein the weight ratio of non-ionic surfactant to pesticide active ingredient is from about 5:1 to about 1:1.25. 13. The extruded composition of claim 1 wherein the non-ionic surfactant is a block polymer comprising at least one polyethoxylate block and at least on poly-C3-C5-alkoxylate block. 14. The extruded composition of claim 1 wherein the non-ionic surfactant is a block polymer including a hydrophilic block and a hydrophobic block. 15. A method for controlling phytopathogenic fungi and/or undesired vegetation and/or undesired attack by insects or mites and/or for regulating the growth of plants, where the extruded pesticide granular composition of claim 1 is allowed to act on the respective pests, their environment or on the crop plants to be protected from the respective pests, on the soil and/or on undesired plants and/or on the crop plants and/or their environment. 16. A process for preparing an extruded pesticide granule comprising melting a non-ionic surfactant, dissolving a pesticide active into the molten non-ionic surfactant, introducing urea, adding water, and mixing to form a damp powder, extruding the damp powder to form an extrudate, and drying the extrudate to form the extruded pesticide granule.
Disclosed are extruded pesticide granules that when mixed with water form a near micro-emulsion that is highly stable. The extruded pesticide granules include at least one pesticide active ingredient, a non-ionic surfactant, and a carrier, which is desirably urea powder or urea pearl. Processes for making the extruded pesticide granules are also disclosed.1. An extruded pesticide granular composition comprising urea, a non-ionic surfactant, a pesticide active ingredient, and water. 2. The extruded composition of claim 1 wherein the urea is in the form of urea pearls. 3. The extruded composition of claim 1 wherein the non-ionic surfactant is a block copolymer having a molecular weight of from about 10,000 to about 15,000 Daltons. 4. The extruded composition of claim 1 wherein the pesticide active ingredient is alpha cypermethrin. 5. The extruded composition of claim 1 wherein the pesticide active ingredient is dinotefuran. 6. The extruded composition of claim 1 wherein the pesticide active ingredient is water-soluble. 7. The extruded composition of claim 1 wherein the urea is present in the composition in an amount of from about 50 wt. % to about 95 wt. %. 8. The extruded composition of claim 1 wherein the urea is present in the composition in an amount of from about 70 wt. % to about 80 wt. %. 9. The extruded composition of claim 1 wherein the non-ionic surfactant is present in an amount of from about 5 wt. % to about 20 wt. %. 10. The extruded composition of claim 1 wherein the pesticide active ingredient is present in an amount of from about 5 wt. % to about 20 wt. %. 11. The extruded composition of claim 1 wherein the composition comprises less than 1% water. 12. The extruded composition of claim 1 wherein the weight ratio of non-ionic surfactant to pesticide active ingredient is from about 5:1 to about 1:1.25. 13. The extruded composition of claim 1 wherein the non-ionic surfactant is a block polymer comprising at least one polyethoxylate block and at least on poly-C3-C5-alkoxylate block. 14. The extruded composition of claim 1 wherein the non-ionic surfactant is a block polymer including a hydrophilic block and a hydrophobic block. 15. A method for controlling phytopathogenic fungi and/or undesired vegetation and/or undesired attack by insects or mites and/or for regulating the growth of plants, where the extruded pesticide granular composition of claim 1 is allowed to act on the respective pests, their environment or on the crop plants to be protected from the respective pests, on the soil and/or on undesired plants and/or on the crop plants and/or their environment. 16. A process for preparing an extruded pesticide granule comprising melting a non-ionic surfactant, dissolving a pesticide active into the molten non-ionic surfactant, introducing urea, adding water, and mixing to form a damp powder, extruding the damp powder to form an extrudate, and drying the extrudate to form the extruded pesticide granule.
1,600
398
13,992,953
1,653
Variations in the differentiation and lineage potential of stem cells, including mesenchymal stem cells, currently limit their therapeutic use. The ability to identify, isolate, and specifically amplify stem cell populations with desired differentiation potential would contribute the use of stem cells in research and therapy. The present invention discloses a method of assessing differentiation potential of stem cells by measuring the differential expression of antigens CD146 and NG2 on the stem cells. The con elation between CD146 and NG2 expression and differentiation and trilineage potential is explored. The invention also discloses methods to specifically amplify or enrich stem cells with desired differentiation potential, monitor the differentiation potential of a heterogeneous stem cell population, quantify the heterogeneity in differentiation potential of a stem cell culture, and remove stem cells with specific differentiation potentials from a heterogeneous cell culture.
1-5. (canceled) 6. A method of identifying multipotent mesenchymal stem cells capable of high proliferation comprising the steps of collecting mesenchymal stem cells; measuring the expression of NG2; and selecting the mesenchymal stem cells with high expression of NG2. 7. The method identifying multipotent mesenchymal stem cells capable of high proliferation of claim 6 wherein the selected mesenchymal stem cells with high expression of NG2 have a colony forming efficiency of greater than 40%. 8. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 6 comprising the additional step of: minimizing the senescent cells in the mesenchymal stem cells selected in claim 6 by adding a marker for senescent cells to the mesenchymal stem cells; measuring the marker indicating senescent cells; and selecting the cells with high expression of NG2 and low indication of the marker for senescent cells. 9. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 6 comprising the additional step of: minimizing lineage-committed cells in the mesenchymal stem cells selected in claim 6 by adding a marker for lineage-committed cells to the mesenchymal stem cells; measuring the marker indicating lineage-committed cells; and selecting the cells with high expression of NG2 and low indication of the marker for lineage-committed cells. 10. A method of identifying multipotent mesenchymal stem cells capable of high proliferation comprising the steps of: collecting mesenchymal stem cells; coating a membrane with a substrate for NG2; placing the mesenchymal stem cells on one side of the membrane; placing a chemotactic agent that promotes cell migration on the other side of the membrane; allowing the mesenchymal stem cells to migrate through the membrane; and collecting the mesenchymal stem cells that migrate more quickly through the membrane. 11. A method of identifying multipotent mesenchymal stem cells capable of high proliferation comprising the steps of collecting mesenchymal stem cells; measuring the expression of CD146 and NG2; and selecting the mesenchymal stem cells with high expression of CD146 and NG2. 12. The method of claim 11 wherein the selected mesenchymal stem cells with high expression of CD146 and NG2 have a colony forming efficiency of greater than 40%. 13. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 11 comprising the additional step of minimizing the senescent cells in the mesenchymal stem cells selected in claim 11 by adding a marker for senescent cells to the mesenchymal stem cells; measuring the marker indicating senescent cells; and selecting the cells with high expression of CD146 and NG2 and low indication of the marker for senescent cells. 14. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 11 comprising the additional step of minimizing lineage-committed cells in the mesenchymal stem cells selected in claim 11 by adding a marker for lineage-committed cells to the mesenchymal stem cells; measuring the marker indicating lineage-committed cells; and selecting the cells with high expression of CD146 and NG2 and low indication of the marker for lineage-committed cells. 15. A method of identifying multipotent mesenchymal stem cells capable of high proliferation: collecting mesenchymal stem cells; coating a membrane with a substrate for NG2 and CD146; placing the mesenchymal stem cells on one side of the membrane; placing a chemotactic agent that promotes cell migration on the other side of the membrane; allowing the mesenchymal stem cells to migrate through the membrane; and collecting the mesenchymal stem cells that migrate more quickly through the membrane. 16-20. (canceled) 21. A method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential comprising the steps of: collecting mesenchymal stem cells from tissue; immunophenotyping the cells for high expression of NG2; and selecting the cells with high expression for NG2. 22. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 21 comprising the additional step of: culturing the cells selected in claim 21. 23. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 22, comprising the additional step of: cryopreserving the cultured cells. 24. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 21 comprising the additional steps of culturing the cells selected in claim 21; and monitoring the presence of NG2. 25. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 21 comprising the additional steps of: culturing the cells selected in claim 21; monitoring the presence of NG2; selecting the cells with high expression of NG2; and further culturing the cells selected in the prior step. 26. A method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential comprising the steps of collecting mesenchymal stem cells from tissue; immunophenotyping the cells for CD146 and NG2; and selecting the cells with high expression of CD146 and NG2. 27. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 26 comprising the additional step of: culturing the cells selected in claim 26. 28. The method for identification and culture of multipotent mesenchymal stem cells with high expression of CD146 and NG2 and with high proliferation potential of claim 27, comprising the additional step of: cryopreserving the cultured cells. 29. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 26 comprising the additional steps of: culturing the cells selected in claim 26; and monitoring the presence of CD146 and NG2. 30. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 26 comprising the additional steps of culturing the cells selected in claim 26; monitoring the presence of CD146 and NG2; selecting the cells with high expression of CD146 and NG2; and further culturing the cells selected in the prior step. 31-32. (canceled) 33. A composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential, at least 50% of said mesenchymal stem cells having high expression of NG2. 34. The composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential of claim 33, at least 75% of said mesenchymal stem cells having high expression of NG2. 35. A composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential, at least 50% of said mesenchymal stem cells having high expression of CD146 and NG2. 36. The composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential of claim 35, at least 75% of said mesenchymal stem cells having high expression of CD146 and NG2.
Variations in the differentiation and lineage potential of stem cells, including mesenchymal stem cells, currently limit their therapeutic use. The ability to identify, isolate, and specifically amplify stem cell populations with desired differentiation potential would contribute the use of stem cells in research and therapy. The present invention discloses a method of assessing differentiation potential of stem cells by measuring the differential expression of antigens CD146 and NG2 on the stem cells. The con elation between CD146 and NG2 expression and differentiation and trilineage potential is explored. The invention also discloses methods to specifically amplify or enrich stem cells with desired differentiation potential, monitor the differentiation potential of a heterogeneous stem cell population, quantify the heterogeneity in differentiation potential of a stem cell culture, and remove stem cells with specific differentiation potentials from a heterogeneous cell culture.1-5. (canceled) 6. A method of identifying multipotent mesenchymal stem cells capable of high proliferation comprising the steps of collecting mesenchymal stem cells; measuring the expression of NG2; and selecting the mesenchymal stem cells with high expression of NG2. 7. The method identifying multipotent mesenchymal stem cells capable of high proliferation of claim 6 wherein the selected mesenchymal stem cells with high expression of NG2 have a colony forming efficiency of greater than 40%. 8. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 6 comprising the additional step of: minimizing the senescent cells in the mesenchymal stem cells selected in claim 6 by adding a marker for senescent cells to the mesenchymal stem cells; measuring the marker indicating senescent cells; and selecting the cells with high expression of NG2 and low indication of the marker for senescent cells. 9. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 6 comprising the additional step of: minimizing lineage-committed cells in the mesenchymal stem cells selected in claim 6 by adding a marker for lineage-committed cells to the mesenchymal stem cells; measuring the marker indicating lineage-committed cells; and selecting the cells with high expression of NG2 and low indication of the marker for lineage-committed cells. 10. A method of identifying multipotent mesenchymal stem cells capable of high proliferation comprising the steps of: collecting mesenchymal stem cells; coating a membrane with a substrate for NG2; placing the mesenchymal stem cells on one side of the membrane; placing a chemotactic agent that promotes cell migration on the other side of the membrane; allowing the mesenchymal stem cells to migrate through the membrane; and collecting the mesenchymal stem cells that migrate more quickly through the membrane. 11. A method of identifying multipotent mesenchymal stem cells capable of high proliferation comprising the steps of collecting mesenchymal stem cells; measuring the expression of CD146 and NG2; and selecting the mesenchymal stem cells with high expression of CD146 and NG2. 12. The method of claim 11 wherein the selected mesenchymal stem cells with high expression of CD146 and NG2 have a colony forming efficiency of greater than 40%. 13. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 11 comprising the additional step of minimizing the senescent cells in the mesenchymal stem cells selected in claim 11 by adding a marker for senescent cells to the mesenchymal stem cells; measuring the marker indicating senescent cells; and selecting the cells with high expression of CD146 and NG2 and low indication of the marker for senescent cells. 14. The method of identifying multipotent mesenchymal stem cells capable of high proliferation of claim 11 comprising the additional step of minimizing lineage-committed cells in the mesenchymal stem cells selected in claim 11 by adding a marker for lineage-committed cells to the mesenchymal stem cells; measuring the marker indicating lineage-committed cells; and selecting the cells with high expression of CD146 and NG2 and low indication of the marker for lineage-committed cells. 15. A method of identifying multipotent mesenchymal stem cells capable of high proliferation: collecting mesenchymal stem cells; coating a membrane with a substrate for NG2 and CD146; placing the mesenchymal stem cells on one side of the membrane; placing a chemotactic agent that promotes cell migration on the other side of the membrane; allowing the mesenchymal stem cells to migrate through the membrane; and collecting the mesenchymal stem cells that migrate more quickly through the membrane. 16-20. (canceled) 21. A method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential comprising the steps of: collecting mesenchymal stem cells from tissue; immunophenotyping the cells for high expression of NG2; and selecting the cells with high expression for NG2. 22. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 21 comprising the additional step of: culturing the cells selected in claim 21. 23. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 22, comprising the additional step of: cryopreserving the cultured cells. 24. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 21 comprising the additional steps of culturing the cells selected in claim 21; and monitoring the presence of NG2. 25. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 21 comprising the additional steps of: culturing the cells selected in claim 21; monitoring the presence of NG2; selecting the cells with high expression of NG2; and further culturing the cells selected in the prior step. 26. A method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential comprising the steps of collecting mesenchymal stem cells from tissue; immunophenotyping the cells for CD146 and NG2; and selecting the cells with high expression of CD146 and NG2. 27. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 26 comprising the additional step of: culturing the cells selected in claim 26. 28. The method for identification and culture of multipotent mesenchymal stem cells with high expression of CD146 and NG2 and with high proliferation potential of claim 27, comprising the additional step of: cryopreserving the cultured cells. 29. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 26 comprising the additional steps of: culturing the cells selected in claim 26; and monitoring the presence of CD146 and NG2. 30. The method for identification and culture of multipotent mesenchymal stem cells with high proliferation potential of claim 26 comprising the additional steps of culturing the cells selected in claim 26; monitoring the presence of CD146 and NG2; selecting the cells with high expression of CD146 and NG2; and further culturing the cells selected in the prior step. 31-32. (canceled) 33. A composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential, at least 50% of said mesenchymal stem cells having high expression of NG2. 34. The composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential of claim 33, at least 75% of said mesenchymal stem cells having high expression of NG2. 35. A composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential, at least 50% of said mesenchymal stem cells having high expression of CD146 and NG2. 36. The composition comprising a population of mesenchymal stem cells having a colony forming efficiency of greater than 40% and trilineage potential of claim 35, at least 75% of said mesenchymal stem cells having high expression of CD146 and NG2.
1,600
399
15,697,723
1,626
The present invention provides a new process for the preparation of a substituted imidazothiazolone compound. The process of the present invention uses a fluoride-free Lewis acid which is cheap and friendly to environment, and provide high selectivity and yield.
1. A process for the preparation of a substituted imidazothiazolone compound of formula (I), or a stereoisomer thereof, or a stereoisomeric mixture thereof, which comprises: reacting a compound of formula (II), or a stereoisomer thereof, or a stereoisomeric mixture thereof, with a nucleophile in the presence of a fluoride-free Lewis acid to provide the compound of formula (I), or a stereoisomer thereof, or a stereoisomeric mixture thereof: wherein: R is H, alkyl or alkylcarbonyl; R1 is benzyl; and R2 is alkyl group, 2. The process of claim 1, wherein R2 is 1-phenyl-1-ethanonyl, 1-(4-chlorophenyl)-1-ethanonyl, 1-(4-methoxypheny)-1-ethanonyl, 2-oxocyclohexyl, 1-trimethylsilyloxy-2-oxocyclohexyl, 1-hydroxyl-2-oxocyclohexyl, or 2-methylpropanoate. 3. The process of claim 1, wherein the nucleophile is selected from enol ethers such as silyl enol ethers. 4. The process of claim 1, wherein the nucleophile is selected from the group consisting of the following structures (a)-(f): 5. The process of claim 1, wherein the fluoride-free Lewis acid is zinc halide such as ZnCl2, ZnBr2, and ZnI2. 6. The process of claim 1, wherein an organic solvent is used. 7. The process of claim 6, wherein the organic solvent used may be selected from the group consisting of ether such as tetrahydrofuran, hydrocarbons such as benzene and toluene, and chlorinated hydrocarbons such as chloroform, dichloromethane and dichloroethane. 8. The process of claim 1, wherein the reaction is carried out at a temperature of from −10° C. to 30° C., preferably, from 0° C. to room temperature.
The present invention provides a new process for the preparation of a substituted imidazothiazolone compound. The process of the present invention uses a fluoride-free Lewis acid which is cheap and friendly to environment, and provide high selectivity and yield.1. A process for the preparation of a substituted imidazothiazolone compound of formula (I), or a stereoisomer thereof, or a stereoisomeric mixture thereof, which comprises: reacting a compound of formula (II), or a stereoisomer thereof, or a stereoisomeric mixture thereof, with a nucleophile in the presence of a fluoride-free Lewis acid to provide the compound of formula (I), or a stereoisomer thereof, or a stereoisomeric mixture thereof: wherein: R is H, alkyl or alkylcarbonyl; R1 is benzyl; and R2 is alkyl group, 2. The process of claim 1, wherein R2 is 1-phenyl-1-ethanonyl, 1-(4-chlorophenyl)-1-ethanonyl, 1-(4-methoxypheny)-1-ethanonyl, 2-oxocyclohexyl, 1-trimethylsilyloxy-2-oxocyclohexyl, 1-hydroxyl-2-oxocyclohexyl, or 2-methylpropanoate. 3. The process of claim 1, wherein the nucleophile is selected from enol ethers such as silyl enol ethers. 4. The process of claim 1, wherein the nucleophile is selected from the group consisting of the following structures (a)-(f): 5. The process of claim 1, wherein the fluoride-free Lewis acid is zinc halide such as ZnCl2, ZnBr2, and ZnI2. 6. The process of claim 1, wherein an organic solvent is used. 7. The process of claim 6, wherein the organic solvent used may be selected from the group consisting of ether such as tetrahydrofuran, hydrocarbons such as benzene and toluene, and chlorinated hydrocarbons such as chloroform, dichloromethane and dichloroethane. 8. The process of claim 1, wherein the reaction is carried out at a temperature of from −10° C. to 30° C., preferably, from 0° C. to room temperature.
1,600