Unnamed: 0
int64
0
350k
ApplicationNumber
int64
9.75M
96.1M
ArtUnit
int64
1.6k
3.99k
Abstract
stringlengths
7
8.37k
Claims
stringlengths
3
292k
abstract-claims
stringlengths
68
293k
TechCenter
int64
1.6k
3.9k
500
14,594,757
1,642
Cancer antigens containing mutations in an expressed gene of cancer cells from a cancer patient are identified. Sequences from cancer cells obtained using a parallel sequencing platform are selected by comparing to the patient's normal genes or to normal genes from an HLA-matched individual. Sequences are further selected by identifying an HLA supertype of the cancer patient and selecting for that HLA supertype, sequences that have a particular amino acid at the mutant position and/or corresponding wild-type position in the effected gene. Peptides containing cancer antigens (i.e., mutations—once a mutation is defined, what makes it an immunogen is its ability to induce an immune response) are optionally tested for binding to HLA antigens of the cancer patient. Peptides containing the cancer antigens are evaluated for activating T cells (e.g., helper T lymphocytes and cytotoxic T lymphocytes (CTL)) cell lines from the cancer patient or from an HLA-matched donor. The cancer antigen(s) identified for a cancer patient are used to prepare a cancer vaccine and to treat the cancer patient.
1. A method of identifying cancer antigens for preparing a cancer vaccine, comprising a) obtaining a plurality of mutant sequences from the nucleic acid of cancer cells from a cancer patient, said mutant sequences coding for all or a portion of an expressed gene and wherein the mutant sequences each have a mutant position amino acid which substitutes for a wildtype position amino acid located at the same position in the wildtype sequence of the protein, wherein said mutant sequences are obtained using a parallel sequencing platform, said parallel sequencing platform employing parallel processing of said nucleic acid of cancer cells leading to sequence reads and mapping of the sequence reads to a database with reference gene sequences; and b) selecting mutant sequences from those identified in step a) by their ability to induce T cells that are specific for the cancer cells or by their ability to be recognized by patient cancer-specific T cells, wherein cancer antigens for preparing a cancer vaccine are identified. 2. The method of claim 1, further comprising, prior to step b), identifying an HLA class or supertype of the cancer patient and then selecting an amino acid for said HLA class or supertype as the mutant position amino acid and/or wildtype position amino acid using FIG. 7 wherein peptides are synthesized and evaluated for activation of T lymphocyte lines prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 3. The method of claim 1, wherein peptides comprising the selected sequences are evaluated for their ability to bind to HLA histocompatibility antigens prior to testing them in step b). 4. The method of claim 3, wherein the ability to bind to HLA histocompatibility antigens is carried out in silico using computer-based algorithm(s) for predicting HLA binding peptides. 5. The method of claim 4, wherein the peptides which bind to HLA histocompatibility antigens in silico are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 6. The method of claim 3, wherein the ability to bind to HLA histocompatibility antigens is carried out by synthesizing the peptides and testing them for binding to antigen-presenting cells that express HLA histocompatibility antigens. 7. The method of claim 6, wherein the peptides which bind to HLA histocompatibility antigens are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 8. The method of claim 1, wherein said parallel sequencing platform filters the sequencing results using a depth of coverage less than 20× and/or by not filtering with a base alignment quality (BAQ) algorithm. 9. The method of claim 2, wherein cancer antigens for preparing a cancer vaccine are identified, and wherein the HLA class or supertype is HLA-1 and the mutant amino acid is selected from the group consisting of phenylalanine, tyrosine, aspartic acid, glutamic acid, leucine, serine and threonine, and wherein the cancer patient expresses the HLA class or supertype HLA-A1 histocompatibility antigen. 10. The method of claim 2, wherein said mononuclear cells are enriched in CD8+ cells. 11. The method of claim 5, wherein said contacting further includes mononuclear cells that are enriched in CD8+ or the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or autologous CD4+ T cells and/or dendritic cells from the HLA-matched donor. 12. A method of identifying cancer antigens for preparing a cancer vaccine, comprising a) obtaining a plurality of mutant sequences from the nucleic acid of cancer cells from a cancer patient, said mutant sequences coding for all or a portion of an expressed gene and wherein the mutant sequences each have a mutant position amino acid which substitutes for a wildtype position amino acid located at the same position in the wildtype sequence of the protein, wherein said mutant sequences are obtained using a parallel sequencing platform, said parallel sequencing platform employing parallel processing of said nucleic acid of cancer cells leading to sequence reads and mapping of the sequence reads to a database with reference gene sequences; and b) identifying at least one mutant sequence for preparing a cancer vaccine from the plurality of mutant sequences obtained in step a) by determining that at least one peptide encoded by the at least one mutant sequence binds to an HLA class or supertype of the cancer patient. 13. The method of claim 12, wherein the peptides are synthesized comprising translating all or a portion of said mutant sequences from step b) and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 14. The method of claim 12, wherein peptides comprising the selected sequences from step b) are evaluated for their ability to bind to HLA histocompatibility antigens. 15. The method of claim 14, wherein the ability to bind to HLA histocompatibility antigens is carried out in silico using computer-based algorithm(s) for predicting HLA binding peptides. 16. The method of claim 15, wherein the peptides which bind to HLA histocompatibility antigens in silico are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 17. The method of claim 14, wherein the ability to bind to HLA histocompatibility antigens is carried out by synthesizing the peptides and testing them for binding to antigen-presenting cells that express HLA histocompatibility antigens. 18. The method of claim 17, wherein the peptides which bind to HLA histocompatibility antigens are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 19. The method of claim 12, wherein said parallel sequencing platform filters the sequencing results using a depth of coverage less than 20× and/or by not filtering with a base alignment quality (BAQ) algorithm. 20. The method of claim 12, wherein the HLA class or supertype is HLA-1 and the mutant amino acid is selected from the group consisting of phenylalanine, tyrosine, aspartic acid, glutamic acid, leucine, serine and threonine, and wherein the cancer patient expresses the HLA-A1 histocompatibility antigen. 21. The method of claim 13, wherein said mononuclear cells are enriched in CD8+ cells. 22. The method of claim 13, wherein said contacting further includes mononuclear cells that are enriched in CD8+ or the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or autologous CD4+ T cells and/or dendritic cells from the HLA-matched donor. 23. A cancer vaccine prepared using one or more of the cancer antigens identified using the method of claim 2. 24. The cancer vaccine of claim 23, which is a polypeptide that comprises one or more of the cancer antigens. 25. The cancer vaccine of claim 23, which is a nucleic acid that encodes for expression of one or more of the cancer antigens. 26. A method of treating a cancer patient, comprising: a) identifying cancer antigens from nucleic acid obtained from cancer cells of the cancer patient using the method of claim 1; b) preparing a vaccine with one or more said cancer antigens, and c) administering the vaccine to said cancer patient to generate T lymphocytes against the cancer cells; and/or d) administering the T lymphocytes prepared from the cancer patient or from an HLA-matched donor, wherein said T lymphocytes are i) prepared by contacting in vitro blood mononuclear cells from the cancer patient or from the HLA-matched donor with the vaccine; or ii) prepared by immunizing the donor with the vaccine and transferring immunized donor T lymphocytes to the cancer patient. 27. The method of claim 26, wherein said contacting further includes mononuclear cells that are enriched in CD8+ or the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or from the HLA-matched donor. 28. A method of treating a cancer patient, comprising: a) identifying cancer antigens from nucleic acid obtained from cancer cells of the cancer patient using the method of claim 1; b) contacting T cells from the patient or from an HLA-matched donor with the cancer antigens in vitro to stimulate cancer specific T lymphocytes; and c) administering the T lymphocytes to the cancer patient and treating the cancer. 29. The method of claim 28, wherein said contacting further includes the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or autologous CD4+ T cells and/or dendritic cells from the HLA-matched donor. 30. The method of claim 2, wherein the mutant position amino acid is selected from the group consisting of: phenylalanine, tyrosine, aspartic acid, glutamic acid, leucine, serine and threonine 31. The method of claim 2, wherein selecting mutant sequences identified in step a) by their ability to induce T cells that are specific for the cancer cells or by their ability to be recognized by cancer-specific T cells comprises using T cells from a donor that is HLA-matched at least one allele or immunizing HLA-transgenic animals with the mutated peptides.
Cancer antigens containing mutations in an expressed gene of cancer cells from a cancer patient are identified. Sequences from cancer cells obtained using a parallel sequencing platform are selected by comparing to the patient's normal genes or to normal genes from an HLA-matched individual. Sequences are further selected by identifying an HLA supertype of the cancer patient and selecting for that HLA supertype, sequences that have a particular amino acid at the mutant position and/or corresponding wild-type position in the effected gene. Peptides containing cancer antigens (i.e., mutations—once a mutation is defined, what makes it an immunogen is its ability to induce an immune response) are optionally tested for binding to HLA antigens of the cancer patient. Peptides containing the cancer antigens are evaluated for activating T cells (e.g., helper T lymphocytes and cytotoxic T lymphocytes (CTL)) cell lines from the cancer patient or from an HLA-matched donor. The cancer antigen(s) identified for a cancer patient are used to prepare a cancer vaccine and to treat the cancer patient.1. A method of identifying cancer antigens for preparing a cancer vaccine, comprising a) obtaining a plurality of mutant sequences from the nucleic acid of cancer cells from a cancer patient, said mutant sequences coding for all or a portion of an expressed gene and wherein the mutant sequences each have a mutant position amino acid which substitutes for a wildtype position amino acid located at the same position in the wildtype sequence of the protein, wherein said mutant sequences are obtained using a parallel sequencing platform, said parallel sequencing platform employing parallel processing of said nucleic acid of cancer cells leading to sequence reads and mapping of the sequence reads to a database with reference gene sequences; and b) selecting mutant sequences from those identified in step a) by their ability to induce T cells that are specific for the cancer cells or by their ability to be recognized by patient cancer-specific T cells, wherein cancer antigens for preparing a cancer vaccine are identified. 2. The method of claim 1, further comprising, prior to step b), identifying an HLA class or supertype of the cancer patient and then selecting an amino acid for said HLA class or supertype as the mutant position amino acid and/or wildtype position amino acid using FIG. 7 wherein peptides are synthesized and evaluated for activation of T lymphocyte lines prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 3. The method of claim 1, wherein peptides comprising the selected sequences are evaluated for their ability to bind to HLA histocompatibility antigens prior to testing them in step b). 4. The method of claim 3, wherein the ability to bind to HLA histocompatibility antigens is carried out in silico using computer-based algorithm(s) for predicting HLA binding peptides. 5. The method of claim 4, wherein the peptides which bind to HLA histocompatibility antigens in silico are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 6. The method of claim 3, wherein the ability to bind to HLA histocompatibility antigens is carried out by synthesizing the peptides and testing them for binding to antigen-presenting cells that express HLA histocompatibility antigens. 7. The method of claim 6, wherein the peptides which bind to HLA histocompatibility antigens are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 8. The method of claim 1, wherein said parallel sequencing platform filters the sequencing results using a depth of coverage less than 20× and/or by not filtering with a base alignment quality (BAQ) algorithm. 9. The method of claim 2, wherein cancer antigens for preparing a cancer vaccine are identified, and wherein the HLA class or supertype is HLA-1 and the mutant amino acid is selected from the group consisting of phenylalanine, tyrosine, aspartic acid, glutamic acid, leucine, serine and threonine, and wherein the cancer patient expresses the HLA class or supertype HLA-A1 histocompatibility antigen. 10. The method of claim 2, wherein said mononuclear cells are enriched in CD8+ cells. 11. The method of claim 5, wherein said contacting further includes mononuclear cells that are enriched in CD8+ or the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or autologous CD4+ T cells and/or dendritic cells from the HLA-matched donor. 12. A method of identifying cancer antigens for preparing a cancer vaccine, comprising a) obtaining a plurality of mutant sequences from the nucleic acid of cancer cells from a cancer patient, said mutant sequences coding for all or a portion of an expressed gene and wherein the mutant sequences each have a mutant position amino acid which substitutes for a wildtype position amino acid located at the same position in the wildtype sequence of the protein, wherein said mutant sequences are obtained using a parallel sequencing platform, said parallel sequencing platform employing parallel processing of said nucleic acid of cancer cells leading to sequence reads and mapping of the sequence reads to a database with reference gene sequences; and b) identifying at least one mutant sequence for preparing a cancer vaccine from the plurality of mutant sequences obtained in step a) by determining that at least one peptide encoded by the at least one mutant sequence binds to an HLA class or supertype of the cancer patient. 13. The method of claim 12, wherein the peptides are synthesized comprising translating all or a portion of said mutant sequences from step b) and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 14. The method of claim 12, wherein peptides comprising the selected sequences from step b) are evaluated for their ability to bind to HLA histocompatibility antigens. 15. The method of claim 14, wherein the ability to bind to HLA histocompatibility antigens is carried out in silico using computer-based algorithm(s) for predicting HLA binding peptides. 16. The method of claim 15, wherein the peptides which bind to HLA histocompatibility antigens in silico are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 17. The method of claim 14, wherein the ability to bind to HLA histocompatibility antigens is carried out by synthesizing the peptides and testing them for binding to antigen-presenting cells that express HLA histocompatibility antigens. 18. The method of claim 17, wherein the peptides which bind to HLA histocompatibility antigens are synthesized and evaluated for activating T lymphocytes prepared from the cancer patient or from an HLA-matched donor, said T lymphocytes obtained by contacting mononuclear cells from the cancer patient or from the HLA-matched donor with cancer cells from the cancer patient. 19. The method of claim 12, wherein said parallel sequencing platform filters the sequencing results using a depth of coverage less than 20× and/or by not filtering with a base alignment quality (BAQ) algorithm. 20. The method of claim 12, wherein the HLA class or supertype is HLA-1 and the mutant amino acid is selected from the group consisting of phenylalanine, tyrosine, aspartic acid, glutamic acid, leucine, serine and threonine, and wherein the cancer patient expresses the HLA-A1 histocompatibility antigen. 21. The method of claim 13, wherein said mononuclear cells are enriched in CD8+ cells. 22. The method of claim 13, wherein said contacting further includes mononuclear cells that are enriched in CD8+ or the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or autologous CD4+ T cells and/or dendritic cells from the HLA-matched donor. 23. A cancer vaccine prepared using one or more of the cancer antigens identified using the method of claim 2. 24. The cancer vaccine of claim 23, which is a polypeptide that comprises one or more of the cancer antigens. 25. The cancer vaccine of claim 23, which is a nucleic acid that encodes for expression of one or more of the cancer antigens. 26. A method of treating a cancer patient, comprising: a) identifying cancer antigens from nucleic acid obtained from cancer cells of the cancer patient using the method of claim 1; b) preparing a vaccine with one or more said cancer antigens, and c) administering the vaccine to said cancer patient to generate T lymphocytes against the cancer cells; and/or d) administering the T lymphocytes prepared from the cancer patient or from an HLA-matched donor, wherein said T lymphocytes are i) prepared by contacting in vitro blood mononuclear cells from the cancer patient or from the HLA-matched donor with the vaccine; or ii) prepared by immunizing the donor with the vaccine and transferring immunized donor T lymphocytes to the cancer patient. 27. The method of claim 26, wherein said contacting further includes mononuclear cells that are enriched in CD8+ or the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or from the HLA-matched donor. 28. A method of treating a cancer patient, comprising: a) identifying cancer antigens from nucleic acid obtained from cancer cells of the cancer patient using the method of claim 1; b) contacting T cells from the patient or from an HLA-matched donor with the cancer antigens in vitro to stimulate cancer specific T lymphocytes; and c) administering the T lymphocytes to the cancer patient and treating the cancer. 29. The method of claim 28, wherein said contacting further includes the addition of autologous CD4+ T cells and/or dendritic cells from the cancer patient or autologous CD4+ T cells and/or dendritic cells from the HLA-matched donor. 30. The method of claim 2, wherein the mutant position amino acid is selected from the group consisting of: phenylalanine, tyrosine, aspartic acid, glutamic acid, leucine, serine and threonine 31. The method of claim 2, wherein selecting mutant sequences identified in step a) by their ability to induce T cells that are specific for the cancer cells or by their ability to be recognized by cancer-specific T cells comprises using T cells from a donor that is HLA-matched at least one allele or immunizing HLA-transgenic animals with the mutated peptides.
1,600
501
15,255,263
1,623
A process for dissolving modified cellulose includes contacting modified cellulose solution with at least one non-solvent to form a plurality of modified cellulose particles.
1-24. (canceled) 25. A process for forming a gel comprising: forming an oxidized cellulose solution; forming a precipitating composition; and contacting the oxidized cellulose solution and the precipitating composition at a treatment site thereby precipitating oxidized cellulose from the oxidized cellulose solution and forming the gel. 26. The process according to claim 25, wherein the formation of the oxidized cellulose solution comprises: contacting an oxidized cellulose with a solvent under an inert atmosphere to form a swelled oxidized cellulose; adjusting the swelled oxidized cellulose mixture to a first temperature; contacting the swelled oxidized cellulose with a salt under the inert atmosphere to form the oxidized cellulose solution; and adjusting the oxidized cellulose solution to a second temperature that is lower than the first temperature. 27. The process according to claim 26, wherein the first temperature is from about 115° C. to about 145° C. and the second temperature is from about 90° C. to about 120° C. 28. The process according to claim 25, wherein the precipitating composition is selected from the group consisting of water, saline, phosphate buffered saline, and combinations thereof. 29. The process according to claim 25, wherein the precipitating composition is an aqueous solution of carboxymethylcellulose and the carboxymethylcellulose is present from about 0.5% by weight of the solution to about 5% by weight of the solution. 30. The process according to claim 25, wherein the precipitating composition is a solution of an acrylic polymer including at least one of methyl methacrylate, hydroxyethyl acrylate, hydroxyethyl methacrylate, glyceryl acrylate, glyceryl methacrylate, acrylic acid, methacrylic acid, acrylamide, or methacrylamide, and combinations thereof. 31. The process according to claim 30, wherein the precipitation composition solution comprises a solvent selected from the group consisting of acetone, ethyl acetate, dimethyl ether, and combinations thereof. 32. The process according to claim 25, further comprising convergently applying the oxidized cellulose solution and the precipitating composition onto a treatment site. 33. A process for forming a cross-linked gel comprising: forming an oxidized cellulose solution; forming a cross-linkable composition covalently cross-linkable with the oxidized cellulose solution; and contacting the oxidized cellulose solution and the cross-linkable composition at a treatment site thereby forming the cross-linked gel. 34. The process according to claim 33, wherein the cross-linkable composition comprises a Schiff-base compound selected from the group consisting of amoxicillin, cephalexin, and combinations thereof. 35. The process according to claim 33, wherein the cross-linkable composition comprises trilysine, albumin, polyethylene glycol amine, and combinations thereof. 36. The process according to claim 35, wherein the cross-linkable composition is an aqueous solution. 37. The process according to claim 34, further comprising convergently applying the oxidized cellulose solution and the cross-linkable composition onto a treatment site. 38. A process for forming a gel comprising: forming an oxidized cellulose solution; forming a gelation composition; and contacting the oxidized cellulose solution and the gelation composition at a treatment site thereby forming the gel. 39. The process according to claim 38, wherein the formation of the oxidized cellulose solution comprises: contacting an oxidized cellulose with a solvent under an inert atmosphere to form a swelled oxidized cellulose; adjusting the swelled oxidized cellulose to a first temperature; contacting the swelled oxidized cellulose with a salt under the inert atmosphere to form the oxidized cellulose solution; and adjusting the oxidized cellulose solution to a second temperature that is lower than the first temperature. 40. The process according to claim 39, wherein the first temperature is from about 115° C. to about 145° C. and the second temperature is from about 90° C. to about 120° C. 41. The process according to claim 38, wherein the gelation composition is an aqueous solution of chitosan having a pH from about 2.0 to about 6.0. 42. The process according to claim 38, wherein the oxidized cellulose solution has a pH from about 8.0 to about 9.5. 43. The process according to claim 38, wherein the gelation composition is an aqueous solution of at least one multivalent cation. 44. The process according to claim 38, wherein the at least one multivalent cation is selected from the group consisting of cations of calcium, barium, zinc, magnesium, chromium, platinum, and iron. 45. The process according to claim 38, wherein the gelation composition is selected from the group consisting of water, saline, phosphate buffered saline, and combinations thereof. 46. The process according to claim 38, wherein the gelation composition is an aqueous solution of carboxymethylcellulose and the carboxymethylcellulose is present from about 0.5% by weight of the solution to about 5% by weight of the solution. 47. The process according to claim 38, wherein the gelation composition is a solution of an acrylic polymer based on at least one of methyl methacrylate, hydroxyethyl acrylate, hydroxyethyl methacrylate, glyceryl acrylate, glyceryl methacrylate, acrylic acid, methacrylic acid, acrylamide, methacrylamide, and combinations thereof. 48. The process according to claim 47, wherein the oxidized cellulose solution comprises a solvent selected from the group consisting of acetone, ethyl acetate, dimethyl ether, and combinations thereof. 49. The process according to claim 38, wherein the gelation composition comprises a Schiff-base compound selected from the group consisting of amoxicillin, cephalexin, and combinations thereof. 50. The process according to claim 38, wherein the gelation composition comprises trilysine, albumin, polyethylene glycol amine, and combinations thereof. 51. The process according to claim 38, further comprising convergently applying the oxidized cellulose solution and the gelation composition onto a treatment site.
A process for dissolving modified cellulose includes contacting modified cellulose solution with at least one non-solvent to form a plurality of modified cellulose particles.1-24. (canceled) 25. A process for forming a gel comprising: forming an oxidized cellulose solution; forming a precipitating composition; and contacting the oxidized cellulose solution and the precipitating composition at a treatment site thereby precipitating oxidized cellulose from the oxidized cellulose solution and forming the gel. 26. The process according to claim 25, wherein the formation of the oxidized cellulose solution comprises: contacting an oxidized cellulose with a solvent under an inert atmosphere to form a swelled oxidized cellulose; adjusting the swelled oxidized cellulose mixture to a first temperature; contacting the swelled oxidized cellulose with a salt under the inert atmosphere to form the oxidized cellulose solution; and adjusting the oxidized cellulose solution to a second temperature that is lower than the first temperature. 27. The process according to claim 26, wherein the first temperature is from about 115° C. to about 145° C. and the second temperature is from about 90° C. to about 120° C. 28. The process according to claim 25, wherein the precipitating composition is selected from the group consisting of water, saline, phosphate buffered saline, and combinations thereof. 29. The process according to claim 25, wherein the precipitating composition is an aqueous solution of carboxymethylcellulose and the carboxymethylcellulose is present from about 0.5% by weight of the solution to about 5% by weight of the solution. 30. The process according to claim 25, wherein the precipitating composition is a solution of an acrylic polymer including at least one of methyl methacrylate, hydroxyethyl acrylate, hydroxyethyl methacrylate, glyceryl acrylate, glyceryl methacrylate, acrylic acid, methacrylic acid, acrylamide, or methacrylamide, and combinations thereof. 31. The process according to claim 30, wherein the precipitation composition solution comprises a solvent selected from the group consisting of acetone, ethyl acetate, dimethyl ether, and combinations thereof. 32. The process according to claim 25, further comprising convergently applying the oxidized cellulose solution and the precipitating composition onto a treatment site. 33. A process for forming a cross-linked gel comprising: forming an oxidized cellulose solution; forming a cross-linkable composition covalently cross-linkable with the oxidized cellulose solution; and contacting the oxidized cellulose solution and the cross-linkable composition at a treatment site thereby forming the cross-linked gel. 34. The process according to claim 33, wherein the cross-linkable composition comprises a Schiff-base compound selected from the group consisting of amoxicillin, cephalexin, and combinations thereof. 35. The process according to claim 33, wherein the cross-linkable composition comprises trilysine, albumin, polyethylene glycol amine, and combinations thereof. 36. The process according to claim 35, wherein the cross-linkable composition is an aqueous solution. 37. The process according to claim 34, further comprising convergently applying the oxidized cellulose solution and the cross-linkable composition onto a treatment site. 38. A process for forming a gel comprising: forming an oxidized cellulose solution; forming a gelation composition; and contacting the oxidized cellulose solution and the gelation composition at a treatment site thereby forming the gel. 39. The process according to claim 38, wherein the formation of the oxidized cellulose solution comprises: contacting an oxidized cellulose with a solvent under an inert atmosphere to form a swelled oxidized cellulose; adjusting the swelled oxidized cellulose to a first temperature; contacting the swelled oxidized cellulose with a salt under the inert atmosphere to form the oxidized cellulose solution; and adjusting the oxidized cellulose solution to a second temperature that is lower than the first temperature. 40. The process according to claim 39, wherein the first temperature is from about 115° C. to about 145° C. and the second temperature is from about 90° C. to about 120° C. 41. The process according to claim 38, wherein the gelation composition is an aqueous solution of chitosan having a pH from about 2.0 to about 6.0. 42. The process according to claim 38, wherein the oxidized cellulose solution has a pH from about 8.0 to about 9.5. 43. The process according to claim 38, wherein the gelation composition is an aqueous solution of at least one multivalent cation. 44. The process according to claim 38, wherein the at least one multivalent cation is selected from the group consisting of cations of calcium, barium, zinc, magnesium, chromium, platinum, and iron. 45. The process according to claim 38, wherein the gelation composition is selected from the group consisting of water, saline, phosphate buffered saline, and combinations thereof. 46. The process according to claim 38, wherein the gelation composition is an aqueous solution of carboxymethylcellulose and the carboxymethylcellulose is present from about 0.5% by weight of the solution to about 5% by weight of the solution. 47. The process according to claim 38, wherein the gelation composition is a solution of an acrylic polymer based on at least one of methyl methacrylate, hydroxyethyl acrylate, hydroxyethyl methacrylate, glyceryl acrylate, glyceryl methacrylate, acrylic acid, methacrylic acid, acrylamide, methacrylamide, and combinations thereof. 48. The process according to claim 47, wherein the oxidized cellulose solution comprises a solvent selected from the group consisting of acetone, ethyl acetate, dimethyl ether, and combinations thereof. 49. The process according to claim 38, wherein the gelation composition comprises a Schiff-base compound selected from the group consisting of amoxicillin, cephalexin, and combinations thereof. 50. The process according to claim 38, wherein the gelation composition comprises trilysine, albumin, polyethylene glycol amine, and combinations thereof. 51. The process according to claim 38, further comprising convergently applying the oxidized cellulose solution and the gelation composition onto a treatment site.
1,600
502
14,605,729
1,644
A method for inducing apoptosis of a neoplastic cell expressing C3aR or C5aR includes administering at least one complement antagonist to the cell so that the at least one complement antagonist substantially reduces or inhibits the activity of protein kinase B in the neoplastic cell.
1-43. (canceled) 44. A method of inducing apoptosis in a cancer cell expressing a C3a receptor (C3aR) and a C5a receptor (C5aR), the method comprising: administering amounts of a C3a antagonist and a C5a antagonist directly or locally to the cancer cell; wherein the amounts of the C3a antagonist and the C5a antagonist administered to the cancer are effective to induce apoptosis in the cancer cell, and wherein the C3a antagonist inhibits C3a mediated C3aR signaling of the cancer cell and the C5a antagonist inhibits C5a mediated C5aR signaling of the cancer cell. 45. The method of claim 44, the C3a antagonist and the C5a antagonist are each individually selected from the group consisting of a small molecule, a polypeptide, and a polynucleotide. 46. The method of claim 45, the C3a antagonist is an antibody directed against C3a and the C5a antagonist is an antibody directed against C5a. 47. The method of claim 44, the cancer cell being at least one of a melanoma cancer cell, fibrosarcoma cancer cell, breast cancer cell, or leukemia cancer cell. 48. A method of treating cancer in a subject in need thereof, the method comprising: administering directly or locally to cancer cells expressing C3aR and C5aR of the subject therapeutically effective amounts of a C3a antagonist and a C5a antagonist, wherein the amounts of C3a antagonist and C5a antagonist administered to the cancer cells induce apoptosis in the cancer cells, and the C3a antagonist inhibits C3a mediated C3aR signaling of the cancer cell and the C5a antagonist inhibits C5a mediated C5aR signaling of the cancer cell. 49. The method of claim 48, the C3a antagonist and the C5a antagonist being each being individually selected from the group consisting of a small molecule, a polypeptide, and a polynucleotide. 50. The method of claim 48, the C3a antagonist being an antibody directed against C3a and the C5a antagonist being an antibody directed against C5a. 51. The method of claim 48, the C3a antagonist and the C5a antagonist each being administered locally to the cancer in the subject. 52. The method of claim 48, the C3a antagonist and the C5a antagonist each being conjugated to targeting moieties that target the cancer being treated. 53. The method of claim 48, the C3a antagonist and the C5a antagonist each being administered systemically to the subject being treated. 54. The method of claim 48, the cancer being at least one of melanoma, fibrosarcoma, breast cancer cell, or leukemia. 55. A method of treating cancer in a subject, the method comprising: administering directly or locally to cancer cells expressing C3aR and C5aR of the subject therapeutically effective amounts of a C3a antibody and a C5a antibody to induce apoptosis in the cancer, wherein the C3a antagonist inhibits C3a mediated C3aR signaling of the cancer cell and the C5a antagonist inhibits C5a mediated C5aR signaling of the cancer cell. 56. The method of claim 55, the C3a antagonist and the C5a antagonist each being conjugated to targeting moieties that target the cancer being treated. 57. The method of claim 55, the C3a antagonist and the C5a antagonist reduce or substantially inhibit the activity of protein kinase B (PKB), induce Fas/FasL surface expression, down regulate Bcl-2/Bcl-xl transcripts, upregulate expression of Bax/Bim transcripts in the cancer cells. 58. The method of claim 55, the cancer being at least one of melanoma, fibrosarcoma, breast cancer cell, or leukemia.
A method for inducing apoptosis of a neoplastic cell expressing C3aR or C5aR includes administering at least one complement antagonist to the cell so that the at least one complement antagonist substantially reduces or inhibits the activity of protein kinase B in the neoplastic cell.1-43. (canceled) 44. A method of inducing apoptosis in a cancer cell expressing a C3a receptor (C3aR) and a C5a receptor (C5aR), the method comprising: administering amounts of a C3a antagonist and a C5a antagonist directly or locally to the cancer cell; wherein the amounts of the C3a antagonist and the C5a antagonist administered to the cancer are effective to induce apoptosis in the cancer cell, and wherein the C3a antagonist inhibits C3a mediated C3aR signaling of the cancer cell and the C5a antagonist inhibits C5a mediated C5aR signaling of the cancer cell. 45. The method of claim 44, the C3a antagonist and the C5a antagonist are each individually selected from the group consisting of a small molecule, a polypeptide, and a polynucleotide. 46. The method of claim 45, the C3a antagonist is an antibody directed against C3a and the C5a antagonist is an antibody directed against C5a. 47. The method of claim 44, the cancer cell being at least one of a melanoma cancer cell, fibrosarcoma cancer cell, breast cancer cell, or leukemia cancer cell. 48. A method of treating cancer in a subject in need thereof, the method comprising: administering directly or locally to cancer cells expressing C3aR and C5aR of the subject therapeutically effective amounts of a C3a antagonist and a C5a antagonist, wherein the amounts of C3a antagonist and C5a antagonist administered to the cancer cells induce apoptosis in the cancer cells, and the C3a antagonist inhibits C3a mediated C3aR signaling of the cancer cell and the C5a antagonist inhibits C5a mediated C5aR signaling of the cancer cell. 49. The method of claim 48, the C3a antagonist and the C5a antagonist being each being individually selected from the group consisting of a small molecule, a polypeptide, and a polynucleotide. 50. The method of claim 48, the C3a antagonist being an antibody directed against C3a and the C5a antagonist being an antibody directed against C5a. 51. The method of claim 48, the C3a antagonist and the C5a antagonist each being administered locally to the cancer in the subject. 52. The method of claim 48, the C3a antagonist and the C5a antagonist each being conjugated to targeting moieties that target the cancer being treated. 53. The method of claim 48, the C3a antagonist and the C5a antagonist each being administered systemically to the subject being treated. 54. The method of claim 48, the cancer being at least one of melanoma, fibrosarcoma, breast cancer cell, or leukemia. 55. A method of treating cancer in a subject, the method comprising: administering directly or locally to cancer cells expressing C3aR and C5aR of the subject therapeutically effective amounts of a C3a antibody and a C5a antibody to induce apoptosis in the cancer, wherein the C3a antagonist inhibits C3a mediated C3aR signaling of the cancer cell and the C5a antagonist inhibits C5a mediated C5aR signaling of the cancer cell. 56. The method of claim 55, the C3a antagonist and the C5a antagonist each being conjugated to targeting moieties that target the cancer being treated. 57. The method of claim 55, the C3a antagonist and the C5a antagonist reduce or substantially inhibit the activity of protein kinase B (PKB), induce Fas/FasL surface expression, down regulate Bcl-2/Bcl-xl transcripts, upregulate expression of Bax/Bim transcripts in the cancer cells. 58. The method of claim 55, the cancer being at least one of melanoma, fibrosarcoma, breast cancer cell, or leukemia.
1,600
503
14,937,142
1,658
The present invention relates to method for producing hypo-metallated redox-active metallothionein (MT) proteins, pharmaceutical compositions containing the proteins, and uses the pharmaceutical compositions for treatment of conditions originating from elevated intracellular oxidative stress and/or dis-balanced intracellular redox-potential and/or redox-potential-dependent imbalance of metal ions.
1. A method for producing a hypo-metallated redox-active metallothionein protein, the hypo-metallated redox-active metallothionein protein having 20 cysteine sulfhydryl groups and 7 binding pockets, 2 to 16 of the 20 cysteine sulfhydryl groups being free and reduced, and 1 to 6 of the 7 binding pockets being occupied by metal ions, comprising the following steps: providing a metallothioneinprotein; de-metallating the metallothioneinprotein; chemically reducing all the 20 cysteine sulfhydryl groups of the de-metallatedmetallothionein protein; and partiallymetallating the reduced de-metallatedmetallothionein protein by providing 1, 2, 3, 4, 5, or 6 metal ions to the 7 binding pockets. 2. The method of claim 1, wherein the metallothionein protein has an amino acid formula of [Xn1CXCXn2CXCXn3CXCXn4CXCXn5CXCXn6CCXCCXn7CXn8CXn9CXCXn10CXCCXn11], wherein X is any biogenic L-amino acid with the exception of cysteine, phenylalanine, tryptophan, and tyrosine; and n1 to n11 are integers of any value from 1 to 12. 3. The method of claim 1, further comprising a step of fully pre-metallating the metallothionein protein before the step of de-metallating the metallothionein protein. 4. The method of claim 1, wherein the step of de-metallating the metallothionein protein is performed by acidifying the metallothionein protein to a pH value below pH4. 5. The method of claim 1, further comprising a step of purifying the de-metallatedmetallothionein protein before the step of chemically reducing the de-metallatedmetallothionein protein. 6. The method of claim 5, wherein the step of purifying the de-metallatedmetallothionein protein is performed by one of dialysis, cation-exchange chromatography, reverse-phase chromatography, high-pressure-liquid chromatography, electrophoretic methods, gel filtration, and magnetic force. 7. The method of claim 1, wherein the step of chemically reducing the de-metallatedmetallothionein protein is performed by adding a reducing agent. 8. The method of claim 8, wherein the reducing agent is one of glutathione, reduced nicotinamide adenine dinucleotide (NADH), nicotinic acid, and dithiothreitol (DTT). 9. The method of claim 1, wherein the metal ions are selected from the group consisting of zinc, copper, iron, and selenium. 10. A pharmaceutical composition, comprising at least one hypo-metallated redox-active metallothionein protein produced by the method of claim 1. 11. Use of hypo-metallated redox-active metallothionein protein produced by the method of claim 1 for the manufacture of a medicament for treatment of a condition originating from elevated intracellular oxidative stress, dis-balanced intracellular redox-potential, or redox-potential-dependent imbalance of metal ions. 12. The use of claim 12, wherein the condition originating from elevated intracellular oxidative stress, dis-balanced intracellular redox-potential, or redox-potential-dependent imbalance of metal ions is one of cancers, neurodegenerative diseases, and metabolic disorders. 13. The use of claim 13, wherein the cancers are at least one of hepatocellular carcinoma, colon cancer, prostate cancer, and papillary thyroid cancer. 14. The use of claim 13, wherein the neurodegenerative diseases are one of Parkinson's disease, Alzheimer's disease, Wilson's Disease, Amyotrophic Lateral Sclerosis (ALS), and Huntington's Disease. 15. The use of claim 13, wherein the metabolic disorders are at least one of diabetes mellitus type I, diabetes mellitus type II, hyperlipidemia, obesity, and fatty liver syndrome.
The present invention relates to method for producing hypo-metallated redox-active metallothionein (MT) proteins, pharmaceutical compositions containing the proteins, and uses the pharmaceutical compositions for treatment of conditions originating from elevated intracellular oxidative stress and/or dis-balanced intracellular redox-potential and/or redox-potential-dependent imbalance of metal ions.1. A method for producing a hypo-metallated redox-active metallothionein protein, the hypo-metallated redox-active metallothionein protein having 20 cysteine sulfhydryl groups and 7 binding pockets, 2 to 16 of the 20 cysteine sulfhydryl groups being free and reduced, and 1 to 6 of the 7 binding pockets being occupied by metal ions, comprising the following steps: providing a metallothioneinprotein; de-metallating the metallothioneinprotein; chemically reducing all the 20 cysteine sulfhydryl groups of the de-metallatedmetallothionein protein; and partiallymetallating the reduced de-metallatedmetallothionein protein by providing 1, 2, 3, 4, 5, or 6 metal ions to the 7 binding pockets. 2. The method of claim 1, wherein the metallothionein protein has an amino acid formula of [Xn1CXCXn2CXCXn3CXCXn4CXCXn5CXCXn6CCXCCXn7CXn8CXn9CXCXn10CXCCXn11], wherein X is any biogenic L-amino acid with the exception of cysteine, phenylalanine, tryptophan, and tyrosine; and n1 to n11 are integers of any value from 1 to 12. 3. The method of claim 1, further comprising a step of fully pre-metallating the metallothionein protein before the step of de-metallating the metallothionein protein. 4. The method of claim 1, wherein the step of de-metallating the metallothionein protein is performed by acidifying the metallothionein protein to a pH value below pH4. 5. The method of claim 1, further comprising a step of purifying the de-metallatedmetallothionein protein before the step of chemically reducing the de-metallatedmetallothionein protein. 6. The method of claim 5, wherein the step of purifying the de-metallatedmetallothionein protein is performed by one of dialysis, cation-exchange chromatography, reverse-phase chromatography, high-pressure-liquid chromatography, electrophoretic methods, gel filtration, and magnetic force. 7. The method of claim 1, wherein the step of chemically reducing the de-metallatedmetallothionein protein is performed by adding a reducing agent. 8. The method of claim 8, wherein the reducing agent is one of glutathione, reduced nicotinamide adenine dinucleotide (NADH), nicotinic acid, and dithiothreitol (DTT). 9. The method of claim 1, wherein the metal ions are selected from the group consisting of zinc, copper, iron, and selenium. 10. A pharmaceutical composition, comprising at least one hypo-metallated redox-active metallothionein protein produced by the method of claim 1. 11. Use of hypo-metallated redox-active metallothionein protein produced by the method of claim 1 for the manufacture of a medicament for treatment of a condition originating from elevated intracellular oxidative stress, dis-balanced intracellular redox-potential, or redox-potential-dependent imbalance of metal ions. 12. The use of claim 12, wherein the condition originating from elevated intracellular oxidative stress, dis-balanced intracellular redox-potential, or redox-potential-dependent imbalance of metal ions is one of cancers, neurodegenerative diseases, and metabolic disorders. 13. The use of claim 13, wherein the cancers are at least one of hepatocellular carcinoma, colon cancer, prostate cancer, and papillary thyroid cancer. 14. The use of claim 13, wherein the neurodegenerative diseases are one of Parkinson's disease, Alzheimer's disease, Wilson's Disease, Amyotrophic Lateral Sclerosis (ALS), and Huntington's Disease. 15. The use of claim 13, wherein the metabolic disorders are at least one of diabetes mellitus type I, diabetes mellitus type II, hyperlipidemia, obesity, and fatty liver syndrome.
1,600
504
13,808,163
1,656
The present invention relates to methods of enhancing fermentation for the production of fermentation products. Specifically, the invention relates to enhancing fermentation in processes of producing ethanol from plant material using one or more fermenting organisms.
1. A method of fermenting sugars derived from plant material in a fermentation medium using a fermenting organism, wherein one or more GH61 polypeptides are added to the fermentation medium. 2. A process of producing a fermentation product from lignocellulose-containing material, comprising the steps of: (a) pre-treating lignocellulose-containing material; (b) hydrolyzing the material; (c) fermenting with a fermenting organism wherein one or more GH61 polypeptides are present in the fermentation medium. 3. The process of claim 1, wherein the lignocellulose-containing material originates from materials selected from the group consisting of corn stover, corn cobs, corn fiber, hardwood, softwood, cereal straw, wheat straw, switchgrass, rice hulls, Miscanthus, municipal solid waste, industrial organic waste, bagasse, and office paper, or mixtures thereof. 4. The process of claim 1, wherein the lignocellulose-containing material is chemically, mechanically or biologically pre-treated in step (a). 5. The process of claim 1, wherein the fermenting organism is a C6 or C5 fermenting organism. 6. The process of claim 1, wherein the fermentation product is ethanol. 7. The process of claim 1, wherein the fermentation product is recovered by distillation. 8-10. (canceled)
The present invention relates to methods of enhancing fermentation for the production of fermentation products. Specifically, the invention relates to enhancing fermentation in processes of producing ethanol from plant material using one or more fermenting organisms.1. A method of fermenting sugars derived from plant material in a fermentation medium using a fermenting organism, wherein one or more GH61 polypeptides are added to the fermentation medium. 2. A process of producing a fermentation product from lignocellulose-containing material, comprising the steps of: (a) pre-treating lignocellulose-containing material; (b) hydrolyzing the material; (c) fermenting with a fermenting organism wherein one or more GH61 polypeptides are present in the fermentation medium. 3. The process of claim 1, wherein the lignocellulose-containing material originates from materials selected from the group consisting of corn stover, corn cobs, corn fiber, hardwood, softwood, cereal straw, wheat straw, switchgrass, rice hulls, Miscanthus, municipal solid waste, industrial organic waste, bagasse, and office paper, or mixtures thereof. 4. The process of claim 1, wherein the lignocellulose-containing material is chemically, mechanically or biologically pre-treated in step (a). 5. The process of claim 1, wherein the fermenting organism is a C6 or C5 fermenting organism. 6. The process of claim 1, wherein the fermentation product is ethanol. 7. The process of claim 1, wherein the fermentation product is recovered by distillation. 8-10. (canceled)
1,600
505
15,131,586
1,658
A method for treating a hyperglycemia and hyperglycemia-dependent side effects in a subject undergoing chemotherapy includes a step of identifying a subject undergoing chemotherapy and being administered a hyperglycemia-inducing agent. Short-term starvation or a fasting mimicking diet is administered for a first time period to the subject to prevent hyperglycemia and sensitization to chemotherapy associated with increased glucose levels. The STS-mimicking drug Metformin is administrated between STS/FMD cycles to maintain STS-/FMD-like conditions during the re-feeding period.
1. A method for treating a hyperglycemia in a subject undergoing chemotherapy, the method comprising: a) identifying a subject undergoing chemotherapy and having hyperglycemia and/or being administered a hyperglycemia-inducing agent; and b) administering short-term starvation (STS), a fasting mimicking diet (FMD) or insulin to the subject for a first time period to prevent or reverse hyperglycemia and sensitization to chemotherapy associated with increased glucose levels. 2. The method of claim 1 wherein the hyperglycemia-inducing agent is selected form the group consisting of rapamycin, steroid medications including dexamethasone, and combinations thereof. 3. The method of claim 1 wherein short term starvation is administered for 48-140 hours prior to a round of chemotherapy and/or 4-56 hours following a round of chemotherapy. 4. The method of claim 1 wherein the FMD is administered for 48-140 hours prior to a round of chemotherapy and/or 4-56 hours following a round of chemotherapy. 5. The method of claim 1 wherein step b) is repeated a plurality of times at predetermined intervals. 6. The method of claim 5 wherein step b) is repeated at intervals from two weeks to 2 months. 7. The method of claim 6 wherein the subject is administered a normal diet in between repetition of step b). 8. The method of claim 1 wherein the first time period is from 3 to 10 days. 9. The method of claim 1 wherein the hyperglycemia-inducing agent is rapamycin. 10. The method of claim 1 wherein the hyperglycemia-inducing agent is a steroid medication. 11. The method of claim 1 wherein the hyperglycemia-inducing agent is dexamethasone. 12. A method for treating a hyperglycemia in a subject undergoing chemotherapy, the method comprising: a) identifying a subject undergoing chemotherapy and having hyperglycemia and/or being administered a hyperglycemia-inducing agent; and b) administering Metformin to subject to mimic effects of fasting. 13. The method of claim 12 wherein the hyperglycemia-inducing agent is selected form the group consisting of rapamycin, steroid medications including dexamethasone, and combinations thereof. 14. The method of claim 12 wherein the Metformin is administered at a dose of 1 to 2.5 mg/day. 15. A method for treating a hyperglycemia in a subject undergoing chemotherapy or other cancer therapy, the method comprising: a) identifying a subject undergoing chemotherapy and having hyperglycemia and/or being administered a hyperglycemia-inducing agent; b) administering short-term starvation (STS), a fasting mimicking diet (FMD) or insulin to the subject for a first time period to prevent or reverse hyperglycemia and sensitization to chemotherapy associated with increased glucose levels; and c) administering a normal diet to the subject after step b). 16. The method of claim 15 wherein the hyperglycemia-inducing agent is selected form the group consisting of rapamycin, steroid medications including dexamethasone, and combinations thereof. 17. The method of claim 15 wherein steps b) and c) is repeated a plurality of times at predetermined intervals. 18. The method of claim 17 wherein steps b) and c) is repeated at intervals from two weeks to 2 months. 19. The method of claim 15 wherein the first time period is from 3 to 10 days. 20. A method of replacing or enhancing an effect of a fasting mimicking diet on cancer cell sensitization, by administering metformin to patients receiving chemotherapy or another cancer therapy. 21. A method where metformin is administered in combination with breast cancer, ovarian cancer, colorectal cancer, melanoma, prostate cancer, cervical cancer, epidermoid carcinoma, neuroblastoma, or any additional cancer type in order to reduce glucose levels and promote differential stress sensitization to specifically kill cancer but not normal cells.
A method for treating a hyperglycemia and hyperglycemia-dependent side effects in a subject undergoing chemotherapy includes a step of identifying a subject undergoing chemotherapy and being administered a hyperglycemia-inducing agent. Short-term starvation or a fasting mimicking diet is administered for a first time period to the subject to prevent hyperglycemia and sensitization to chemotherapy associated with increased glucose levels. The STS-mimicking drug Metformin is administrated between STS/FMD cycles to maintain STS-/FMD-like conditions during the re-feeding period.1. A method for treating a hyperglycemia in a subject undergoing chemotherapy, the method comprising: a) identifying a subject undergoing chemotherapy and having hyperglycemia and/or being administered a hyperglycemia-inducing agent; and b) administering short-term starvation (STS), a fasting mimicking diet (FMD) or insulin to the subject for a first time period to prevent or reverse hyperglycemia and sensitization to chemotherapy associated with increased glucose levels. 2. The method of claim 1 wherein the hyperglycemia-inducing agent is selected form the group consisting of rapamycin, steroid medications including dexamethasone, and combinations thereof. 3. The method of claim 1 wherein short term starvation is administered for 48-140 hours prior to a round of chemotherapy and/or 4-56 hours following a round of chemotherapy. 4. The method of claim 1 wherein the FMD is administered for 48-140 hours prior to a round of chemotherapy and/or 4-56 hours following a round of chemotherapy. 5. The method of claim 1 wherein step b) is repeated a plurality of times at predetermined intervals. 6. The method of claim 5 wherein step b) is repeated at intervals from two weeks to 2 months. 7. The method of claim 6 wherein the subject is administered a normal diet in between repetition of step b). 8. The method of claim 1 wherein the first time period is from 3 to 10 days. 9. The method of claim 1 wherein the hyperglycemia-inducing agent is rapamycin. 10. The method of claim 1 wherein the hyperglycemia-inducing agent is a steroid medication. 11. The method of claim 1 wherein the hyperglycemia-inducing agent is dexamethasone. 12. A method for treating a hyperglycemia in a subject undergoing chemotherapy, the method comprising: a) identifying a subject undergoing chemotherapy and having hyperglycemia and/or being administered a hyperglycemia-inducing agent; and b) administering Metformin to subject to mimic effects of fasting. 13. The method of claim 12 wherein the hyperglycemia-inducing agent is selected form the group consisting of rapamycin, steroid medications including dexamethasone, and combinations thereof. 14. The method of claim 12 wherein the Metformin is administered at a dose of 1 to 2.5 mg/day. 15. A method for treating a hyperglycemia in a subject undergoing chemotherapy or other cancer therapy, the method comprising: a) identifying a subject undergoing chemotherapy and having hyperglycemia and/or being administered a hyperglycemia-inducing agent; b) administering short-term starvation (STS), a fasting mimicking diet (FMD) or insulin to the subject for a first time period to prevent or reverse hyperglycemia and sensitization to chemotherapy associated with increased glucose levels; and c) administering a normal diet to the subject after step b). 16. The method of claim 15 wherein the hyperglycemia-inducing agent is selected form the group consisting of rapamycin, steroid medications including dexamethasone, and combinations thereof. 17. The method of claim 15 wherein steps b) and c) is repeated a plurality of times at predetermined intervals. 18. The method of claim 17 wherein steps b) and c) is repeated at intervals from two weeks to 2 months. 19. The method of claim 15 wherein the first time period is from 3 to 10 days. 20. A method of replacing or enhancing an effect of a fasting mimicking diet on cancer cell sensitization, by administering metformin to patients receiving chemotherapy or another cancer therapy. 21. A method where metformin is administered in combination with breast cancer, ovarian cancer, colorectal cancer, melanoma, prostate cancer, cervical cancer, epidermoid carcinoma, neuroblastoma, or any additional cancer type in order to reduce glucose levels and promote differential stress sensitization to specifically kill cancer but not normal cells.
1,600
506
15,175,848
1,644
Humanized or chimeric anti-CD47 monoclonal antibodies are provided. The antibodies bind to and neutralize human CD47, and find use in various therapeutic methods. Preferred are non-activating antibodies. Embodiments of the invention include isolated antibodies and derivatives and fragments thereof, pharmaceutical formulations comprising one or more of the humanized or chimeric anti-CD47 monoclonal antibodies; and cell lines that produce these monoclonal antibodies. Also provided are amino acid sequences of the antibodies.
1. An isolated chimeric or humanized antibody that specifically binds to human CD47, and comprises at least one CDR sequence as set forth in SEQ ID NO:3-8, SEQ ID NO:20-25; SEQ ID NO:28-33 or SEQ ID NO:61-66.
Humanized or chimeric anti-CD47 monoclonal antibodies are provided. The antibodies bind to and neutralize human CD47, and find use in various therapeutic methods. Preferred are non-activating antibodies. Embodiments of the invention include isolated antibodies and derivatives and fragments thereof, pharmaceutical formulations comprising one or more of the humanized or chimeric anti-CD47 monoclonal antibodies; and cell lines that produce these monoclonal antibodies. Also provided are amino acid sequences of the antibodies.1. An isolated chimeric or humanized antibody that specifically binds to human CD47, and comprises at least one CDR sequence as set forth in SEQ ID NO:3-8, SEQ ID NO:20-25; SEQ ID NO:28-33 or SEQ ID NO:61-66.
1,600
507
15,135,677
1,619
Described herein is a hair care composition having from about 0.2% to about 10% of one or more surfactant soluble active agents, from about 1% to about 10% of one or more viscosity reducing agents having a partition dispersion coefficient of from about 0.05 to about 2.0, from about 16% to about 40% of one or more anionic surfactants, and from about 40% to about 83% of a carrier. The hair care composition has a liquid phase kinematic viscosity, measured at 40 degrees Celsius, of from about 10 cSt to about 1000 cSt.
1) A hair care composition comprising: a) from about 0.2% to about 10% of one or more surfactant soluble active agents, by weight of the hair care composition; b) from about 1% to about 10% of one or more viscosity reducing agents, by weight of the hair care composition, having a partition dispersion coefficient of from about 0.05 to about 2.0; c) from about 16% to about 40% of one or more anionic surfactants, by weight of the hair care composition; d) from about 40% to about 83% of a carrier, by weight of the hair care composition; wherein the one or more viscosity reducing agents has a molecular weight of from about 100 daltons to about 300 daltons; and wherein the hair care composition has a liquid phase kinematic viscosity, measured at 40 degrees Celsius, of from about 10 cSt to about 1,000 cSt. 2) The hair care composition according to claim 1, wherein the surfactant soluble active agent is selected from vitamins, perfume oils, UV absorbers, anti-dandruff actives, scalp benefit agents, skin actives, internal hair actives, antimicrobials, antibacterial actives, radical scavengers, skin lightening agents, chelators, antioxidants, proteins, amino acids, peptides, flavonoids, alpha-hydroxy acids, anti-acne actives, anti-inflammatories, and having a molecular weight ranging from about 100 g/mol to about 700 g/mol. 3) The hair care composition according to claim 2, wherein the surfactant soluble active agent is a perfume oil. 4) The hair care composition according to claim 1, wherein the surfactant soluble active agent is an anti-dandruff active selected from the group consisting of climbazole, ketoconazole, itraconazole, econazole, elubiol, octopirox (piroctone olamine), ciclopirox, rilopirox, MEA-Hydroxyoctyloxypyridinone, and combinations thereof. 5) The hair care composition according to claim 1, wherein the hair care composition further comprises from about 0.25% to about 14% of one or more co-surfactants, by weight of the hair care composition, selected from the group consisting of amphoteric, non-ionic, zwitterionic, and combinations thereof. 6) The hair care composition according to claim 1, wherein the partition dispersion coefficient of the one or more viscosity reducing agents is from about 0.08 to about 1.9. 7) The hair care composition according to claim 1, wherein the partition dispersion coefficient of the one or more viscosity reducing agents is from about 0.09 to about 1.8. 8) The hair care composition according to claim 1, wherein the partition dispersion coefficient of the one or more viscosity reducing agents is from about 0.095 to about 1.7. 9) The hair care composition according to claim 1, wherein the hair care composition comprises less than 4% of a counteracting additive having a partition dispersion coefficient of from about −3.1 to about −0.7, by weight of the hair care composition; and wherein the weight ratio of the one or more viscosity reducing agents to the counteracting additive is greater than 2:1. 10) The hair care composition according to claim 6, wherein the weight ratio of the one or more viscosity reducing agents to the counteracting additive is greater than 5:1. 11) The hair care composition according to claim 6, wherein the hair care composition is substantially free of a counteracting additive having a partition dispersion coefficient of from about −3.1 to about −0.7. 12) The hair care composition according to claim 1, wherein the liquid phase kinematic viscosity is from about 15 cSt to about 400 cSt. 13) The hair care composition according to claim 1, wherein the liquid phase kinematic viscosity is from about 20 cSt to about 300 cSt. 14) The hair care composition according to claim 1, wherein the liquid phase kinematic viscosity is from about 30 cSt to about 200 cSt. 15) The hair care composition according to claim 1, wherein the hair care composition comprises from about 20% to about 32% of the one or more anionic surfactants, by weight of the hair care composition. 16) The hair care composition according to claim 1, wherein the hair care composition comprises from about 4% to about 7% of the one or more amphoteric, non-ionic or zwitterionic co-surfactants, by weight of the hair care composition. 17) The hair care composition according to claim 1, wherein the hair care composition comprises from about 3.5% to about 8% of the one or more viscosity reducing agents, by weight of the hair care composition. 18) The hair care composition according to claim 1, wherein the one or more viscosity reducing agents is selected from the group consisting of veloutone, isoamyl salicylate, gamma-terpinene, linalyl iso butyrate, alpha-terpinene, limonene, dipentene, geranyl phenyl acetate, and combinations thereof. 19) The hair care composition according to claim 1, wherein the hair care composition further comprises a cationic polymer selected from the group consisting of guar polymers, non-guar galactomannan polymers, tapioca polymers, copolymers of acrylamide monomers and cationic monomers, cellulose polymers, and combinations thereof. 20) A method of treating the hair, the method comprising: a) providing the hair care composition according to claim 1 in a foam dispenser; b) dispensing the hair care composition from the foam dispenser as a foam; c) applying the foam to the hair; and d) rinsing the foam from the hair; wherein the foam has a density of from about 0.05 g/cm3 to about 0.30 g/cm3 when dispensed from the foam dispenser.
Described herein is a hair care composition having from about 0.2% to about 10% of one or more surfactant soluble active agents, from about 1% to about 10% of one or more viscosity reducing agents having a partition dispersion coefficient of from about 0.05 to about 2.0, from about 16% to about 40% of one or more anionic surfactants, and from about 40% to about 83% of a carrier. The hair care composition has a liquid phase kinematic viscosity, measured at 40 degrees Celsius, of from about 10 cSt to about 1000 cSt.1) A hair care composition comprising: a) from about 0.2% to about 10% of one or more surfactant soluble active agents, by weight of the hair care composition; b) from about 1% to about 10% of one or more viscosity reducing agents, by weight of the hair care composition, having a partition dispersion coefficient of from about 0.05 to about 2.0; c) from about 16% to about 40% of one or more anionic surfactants, by weight of the hair care composition; d) from about 40% to about 83% of a carrier, by weight of the hair care composition; wherein the one or more viscosity reducing agents has a molecular weight of from about 100 daltons to about 300 daltons; and wherein the hair care composition has a liquid phase kinematic viscosity, measured at 40 degrees Celsius, of from about 10 cSt to about 1,000 cSt. 2) The hair care composition according to claim 1, wherein the surfactant soluble active agent is selected from vitamins, perfume oils, UV absorbers, anti-dandruff actives, scalp benefit agents, skin actives, internal hair actives, antimicrobials, antibacterial actives, radical scavengers, skin lightening agents, chelators, antioxidants, proteins, amino acids, peptides, flavonoids, alpha-hydroxy acids, anti-acne actives, anti-inflammatories, and having a molecular weight ranging from about 100 g/mol to about 700 g/mol. 3) The hair care composition according to claim 2, wherein the surfactant soluble active agent is a perfume oil. 4) The hair care composition according to claim 1, wherein the surfactant soluble active agent is an anti-dandruff active selected from the group consisting of climbazole, ketoconazole, itraconazole, econazole, elubiol, octopirox (piroctone olamine), ciclopirox, rilopirox, MEA-Hydroxyoctyloxypyridinone, and combinations thereof. 5) The hair care composition according to claim 1, wherein the hair care composition further comprises from about 0.25% to about 14% of one or more co-surfactants, by weight of the hair care composition, selected from the group consisting of amphoteric, non-ionic, zwitterionic, and combinations thereof. 6) The hair care composition according to claim 1, wherein the partition dispersion coefficient of the one or more viscosity reducing agents is from about 0.08 to about 1.9. 7) The hair care composition according to claim 1, wherein the partition dispersion coefficient of the one or more viscosity reducing agents is from about 0.09 to about 1.8. 8) The hair care composition according to claim 1, wherein the partition dispersion coefficient of the one or more viscosity reducing agents is from about 0.095 to about 1.7. 9) The hair care composition according to claim 1, wherein the hair care composition comprises less than 4% of a counteracting additive having a partition dispersion coefficient of from about −3.1 to about −0.7, by weight of the hair care composition; and wherein the weight ratio of the one or more viscosity reducing agents to the counteracting additive is greater than 2:1. 10) The hair care composition according to claim 6, wherein the weight ratio of the one or more viscosity reducing agents to the counteracting additive is greater than 5:1. 11) The hair care composition according to claim 6, wherein the hair care composition is substantially free of a counteracting additive having a partition dispersion coefficient of from about −3.1 to about −0.7. 12) The hair care composition according to claim 1, wherein the liquid phase kinematic viscosity is from about 15 cSt to about 400 cSt. 13) The hair care composition according to claim 1, wherein the liquid phase kinematic viscosity is from about 20 cSt to about 300 cSt. 14) The hair care composition according to claim 1, wherein the liquid phase kinematic viscosity is from about 30 cSt to about 200 cSt. 15) The hair care composition according to claim 1, wherein the hair care composition comprises from about 20% to about 32% of the one or more anionic surfactants, by weight of the hair care composition. 16) The hair care composition according to claim 1, wherein the hair care composition comprises from about 4% to about 7% of the one or more amphoteric, non-ionic or zwitterionic co-surfactants, by weight of the hair care composition. 17) The hair care composition according to claim 1, wherein the hair care composition comprises from about 3.5% to about 8% of the one or more viscosity reducing agents, by weight of the hair care composition. 18) The hair care composition according to claim 1, wherein the one or more viscosity reducing agents is selected from the group consisting of veloutone, isoamyl salicylate, gamma-terpinene, linalyl iso butyrate, alpha-terpinene, limonene, dipentene, geranyl phenyl acetate, and combinations thereof. 19) The hair care composition according to claim 1, wherein the hair care composition further comprises a cationic polymer selected from the group consisting of guar polymers, non-guar galactomannan polymers, tapioca polymers, copolymers of acrylamide monomers and cationic monomers, cellulose polymers, and combinations thereof. 20) A method of treating the hair, the method comprising: a) providing the hair care composition according to claim 1 in a foam dispenser; b) dispensing the hair care composition from the foam dispenser as a foam; c) applying the foam to the hair; and d) rinsing the foam from the hair; wherein the foam has a density of from about 0.05 g/cm3 to about 0.30 g/cm3 when dispensed from the foam dispenser.
1,600
508
14,849,203
1,637
The invention provides methods for identifying modulators of ion channels without the use of recombinant cell lines over-expressing the ion channel proteins or the use of detection labels.
1. A method of determining ion channel modulating properties of a test reagent comprising: (a) applying cells to a surface of a colorimetric resonant reflectance optical biosensor; (b) substantially blocking the functional activity of one or more first types of ion channels, wherein the activity of one or more second types of ion channels is not blocked; (c) detecting a colorimetric resonant reflectance optical first peak wavelength value (PWV) for the cells; (d) applying a test reagent to the cells; (e) detecting a colorimetric resonant reflectance optical second PWV for the cells; and (f) determining ion channel modulation properties of the test reagent based on a comparison of the first PWV and second PWV. 2. The method of claim 1, wherein the functional activity of one or more types of ion channels is substantially blocked by an antibody, protein, or small organic molecule. 3. The method of claim 1, wherein the one or more first type of ion channels are voltage-gated sodium channels, voltage-gated calcium channels, mechano-sensitive channels, potassium channels, inward-rectifier potassium channels, calcium-activated potassium channels, voltage-gated potassium channels, two pore domain potassium channels, transient receptor potential channels, cation channels of sperm, cyclic nucleotide gated channels, hyperpolaraization activated cyclic nucleotide gated channels, two pore channels, ligand gated channels, and light-gated channels. 4. The method of claim 1, wherein the one or more second type of ion channels are voltage-gated sodium channels, voltage-gated calcium channels, mechano-sensitive channels, potassium channels, inward-rectifier potassium channels, calcium-activated potassium channels, voltage-gated potassium channels, two pore domain potassium channels, transient receptor potential channels, cation channels of sperm, cyclic nucleotide gated channels, hyperpolaraization activated cyclic nucleotide gated channels, two pore channels, ligand gated channels, and light-gated channels. 5. The method of claim 1, wherein the cells are incubated for a period of time after: (a) their application to the surface; (b) the blocking the functional activity of one or more first types of ion channels; (c) after the detecting the first PWV for the cells; (d) after the application of the test reagent to the cells; (e) after the detecting the first PWV for the cells, or (f) a combination thereof. 6. The method of claim 1, wherein the surface of the colorimetric resonant reflectance optical biosensor is an internal surface of a vessel selected from the group consisting of a microtiter well, microtiter plate, test tube, Petri dish, microfluidic channel, and microarray. 7. The method of claim 1, wherein the first and second PWVs are detected using a scanner with a lens having a lower limit pixel size of about 2 micrometers to about 200 micrometers. 8. The method of claim 1, wherein the cells and test reagent do not comprise detection labels. 9. The method of claim 1, wherein the method is performed at a temperature of about 2, 10, 15, 25, 30, or 37 degrees Celsius. 10. The method of claim 1, further comprising the steps of: (g) washing the cells; (h) equilibrating the cells; (i) optionally repeating steps (a)-(f). 11. A method of identifying a modulator of an ion channel comprising: (a) applying cells in a serum-free medium to a surface of a colorimetric resonant reflectance optical biosensor, wherein one or more extracellular matrix (ECM) ligands are immobilized to the surface of the biosensor; (b) detecting a colorimetric resonant reflectance optical first peak wavelength value (PWV) for the cells; (c) applying one or more test ion channel modulators to the surface of the biosensor; (d) effecting a change in the activity of one or more ion channels of the cells; (e) detecting a colorimetric resonant reflectance optical second PWV for the cells; and (f) determining if the one or more test ion channel modulators modulated the one or more ion channels of the cells. 12. The method of claim 11, further comprising the steps of: (g) washing the cells; (h) equilibrating the cells; (i) optionally repeating steps (a)-(f). 13. A method of identifying a modulator of an ion channel comprising: (a) applying cells in a serum-free medium to a surface of a colorimetric resonant reflectance optical biosensor, wherein one or more extracellular matrix (ECM) ligands are immobilized to the surface of the biosensor; (b) applying one or more test ion channel modulators to the surface of the biosensor; (c) effecting a change in one or more ion channels of the cells; (d) detecting a colorimetric resonant reflectance optical first PWV for the cells; (e) effecting a change in one or more ion channels of the cells; (f) detecting a colorimetric resonant reflectance optical second PWV for the cells; and (g) determining if the one or more test ion channel modulators modulated the one or more ion channels of the cells. 14. The method of claim 13, further comprising the steps of: (h) washing the cells; (j) equilibrating the cells; (k) optionally repeating steps (a)-(g).
The invention provides methods for identifying modulators of ion channels without the use of recombinant cell lines over-expressing the ion channel proteins or the use of detection labels.1. A method of determining ion channel modulating properties of a test reagent comprising: (a) applying cells to a surface of a colorimetric resonant reflectance optical biosensor; (b) substantially blocking the functional activity of one or more first types of ion channels, wherein the activity of one or more second types of ion channels is not blocked; (c) detecting a colorimetric resonant reflectance optical first peak wavelength value (PWV) for the cells; (d) applying a test reagent to the cells; (e) detecting a colorimetric resonant reflectance optical second PWV for the cells; and (f) determining ion channel modulation properties of the test reagent based on a comparison of the first PWV and second PWV. 2. The method of claim 1, wherein the functional activity of one or more types of ion channels is substantially blocked by an antibody, protein, or small organic molecule. 3. The method of claim 1, wherein the one or more first type of ion channels are voltage-gated sodium channels, voltage-gated calcium channels, mechano-sensitive channels, potassium channels, inward-rectifier potassium channels, calcium-activated potassium channels, voltage-gated potassium channels, two pore domain potassium channels, transient receptor potential channels, cation channels of sperm, cyclic nucleotide gated channels, hyperpolaraization activated cyclic nucleotide gated channels, two pore channels, ligand gated channels, and light-gated channels. 4. The method of claim 1, wherein the one or more second type of ion channels are voltage-gated sodium channels, voltage-gated calcium channels, mechano-sensitive channels, potassium channels, inward-rectifier potassium channels, calcium-activated potassium channels, voltage-gated potassium channels, two pore domain potassium channels, transient receptor potential channels, cation channels of sperm, cyclic nucleotide gated channels, hyperpolaraization activated cyclic nucleotide gated channels, two pore channels, ligand gated channels, and light-gated channels. 5. The method of claim 1, wherein the cells are incubated for a period of time after: (a) their application to the surface; (b) the blocking the functional activity of one or more first types of ion channels; (c) after the detecting the first PWV for the cells; (d) after the application of the test reagent to the cells; (e) after the detecting the first PWV for the cells, or (f) a combination thereof. 6. The method of claim 1, wherein the surface of the colorimetric resonant reflectance optical biosensor is an internal surface of a vessel selected from the group consisting of a microtiter well, microtiter plate, test tube, Petri dish, microfluidic channel, and microarray. 7. The method of claim 1, wherein the first and second PWVs are detected using a scanner with a lens having a lower limit pixel size of about 2 micrometers to about 200 micrometers. 8. The method of claim 1, wherein the cells and test reagent do not comprise detection labels. 9. The method of claim 1, wherein the method is performed at a temperature of about 2, 10, 15, 25, 30, or 37 degrees Celsius. 10. The method of claim 1, further comprising the steps of: (g) washing the cells; (h) equilibrating the cells; (i) optionally repeating steps (a)-(f). 11. A method of identifying a modulator of an ion channel comprising: (a) applying cells in a serum-free medium to a surface of a colorimetric resonant reflectance optical biosensor, wherein one or more extracellular matrix (ECM) ligands are immobilized to the surface of the biosensor; (b) detecting a colorimetric resonant reflectance optical first peak wavelength value (PWV) for the cells; (c) applying one or more test ion channel modulators to the surface of the biosensor; (d) effecting a change in the activity of one or more ion channels of the cells; (e) detecting a colorimetric resonant reflectance optical second PWV for the cells; and (f) determining if the one or more test ion channel modulators modulated the one or more ion channels of the cells. 12. The method of claim 11, further comprising the steps of: (g) washing the cells; (h) equilibrating the cells; (i) optionally repeating steps (a)-(f). 13. A method of identifying a modulator of an ion channel comprising: (a) applying cells in a serum-free medium to a surface of a colorimetric resonant reflectance optical biosensor, wherein one or more extracellular matrix (ECM) ligands are immobilized to the surface of the biosensor; (b) applying one or more test ion channel modulators to the surface of the biosensor; (c) effecting a change in one or more ion channels of the cells; (d) detecting a colorimetric resonant reflectance optical first PWV for the cells; (e) effecting a change in one or more ion channels of the cells; (f) detecting a colorimetric resonant reflectance optical second PWV for the cells; and (g) determining if the one or more test ion channel modulators modulated the one or more ion channels of the cells. 14. The method of claim 13, further comprising the steps of: (h) washing the cells; (j) equilibrating the cells; (k) optionally repeating steps (a)-(g).
1,600
509
14,355,903
1,627
The present invention relates to a method of treating a patient with a serine/threonine kinase inhibitor wherein resistance to the treatment with a serine/threonine kinase inhibitor is suppressed by administering the serine/threonine kinase inhibitor on an intermittent dosing schedule.
1. A method of treating a proliferative disease characterized by a mutation in BRAF kinase, which comprises suppressing resistance to treatment with a BRAF inhibitor of the Formula II by administering the BRAF inhibitor of Formula II on an intermittent dosing schedule. 2. A method of claim 1 wherein the BRAF mutation is a V600 mutation. 3. A method of claim 1 wherein the proliferative disease is melanoma which is characterized by a V600 mutation or colorectal cancer characterized by a V600 mutation. 4. A method of claim 1 wherein the proliferative disease is melanoma which is characterized by BRAFV600E or colorectal cancer characterized by BRAFV600E. 5. Use of a BRAF inhibitor of Formula II for the preparation of a medicament for the treatment of a proliferative disease whereby the BRAF inhibitor is administered on an intemittent dosing schedule.
The present invention relates to a method of treating a patient with a serine/threonine kinase inhibitor wherein resistance to the treatment with a serine/threonine kinase inhibitor is suppressed by administering the serine/threonine kinase inhibitor on an intermittent dosing schedule.1. A method of treating a proliferative disease characterized by a mutation in BRAF kinase, which comprises suppressing resistance to treatment with a BRAF inhibitor of the Formula II by administering the BRAF inhibitor of Formula II on an intermittent dosing schedule. 2. A method of claim 1 wherein the BRAF mutation is a V600 mutation. 3. A method of claim 1 wherein the proliferative disease is melanoma which is characterized by a V600 mutation or colorectal cancer characterized by a V600 mutation. 4. A method of claim 1 wherein the proliferative disease is melanoma which is characterized by BRAFV600E or colorectal cancer characterized by BRAFV600E. 5. Use of a BRAF inhibitor of Formula II for the preparation of a medicament for the treatment of a proliferative disease whereby the BRAF inhibitor is administered on an intemittent dosing schedule.
1,600
510
14,452,453
1,653
A tissue system includes: a support material; and a vascular network comprising a plurality of channels disposed in the support material. A method includes printing a bioink in a support structure to form a network of vascular precursor materials; and converting the vascular precursor materials into a physiologically relevant vascular network. Notably, the tissue systems, networks, etc. are physiologically-relevant, i.e. exhibiting one or more characteristics indicative of physiological relevance, such as a substantially fractal geometry, inter-vessel spacing, cellular composition, dermal structure, concentric multi-layered structure, etc.
1. A tissue system, comprising: a support material, and a vascular network comprising a plurality of channels disposed in the support material, wherein the vascular network is physiologically relevant. 2. The system as recited in claim 1, wherein the vascular network comprises a plurality of constituents selected from a group consisting of endothelial cells (EC), smooth muscle cells, growth factors, and adhesion proteins. 3. The system as recited in claim 2, further comprising a fugitive material configured to vacate an interior cavity of each channel in the vascular network in response to exposure to predetermined conditions. 4. The system as recited in claim 1, wherein the support material comprises one or more of: MATRIGEL™ Stock, MATRIGEL™/GM mixture, EXTRACELL™, PURAMATRIX™, Agarose, Sodium alginate/Calcium (II) chloride, Collagen (Types I-IV), lyophilized/reconstituted human cardiac ECM, gelatin, polyethylene glycol (PEG), polyethylene glycol diacrylate (PEGDA), and/or poly-L-lactic acid (PLLA), a buffer such as phosphate-buffered saline (PBS), and/or one or more cell-type specific culture growth media. 5. The system as recited in claim 1, wherein the vascular network has physical characteristics of being formed from omnidirectional printing of a bioink. 6. The system as recited in claim 1, wherein the vascular network comprises arterial pathways and venous pathways. 7. The system as recited in claim 1, each channel being characterized by an outer diameter in a range from approximately 0.5 microns to approximately 1 mm. 8. The system as recited in claim 1, wherein the channels comprise one or more of: large channels characterized by an outer large channel diameter between about 100 microns and about 20 mm; medium channels characterized by an outer medium channel diameter between about 7 microns and about 150 microns; and capillary channels characterized by an outer capillary diameter between about 5 microns and about 40 microns. 9. The system as recited in claim 1, wherein the vascular network is characterized by an inter-channel spacing between approximately 0.01 microns and approximately 200 microns. 10. The system as recited in claim 1, wherein the vascular network comprises a bifurcating network of the channels. 11. The system as recited in claim 1, wherein the vascular network has physical characteristics of being formed at least in part by vasculogenesis and/or angiogenesis. 12. A method, comprising: printing a bioink in a support structure to form a network of vascular precursor materials; and converting the vascular precursor materials into a physiologically relevant vascular network. 13. The method as recited in claim 12, wherein the printing comprises multiaxial extrusion of the bioink through a nozzle. 14. The method as recited in claim 12, wherein the printing comprises omnidirectional printing. 15. The method as recited in claim 12, wherein the printing forms the network in a geometric arrangement characterized by an inter-channel spacing between approximately 1 micron and approximately 175 microns. 16. The method as recited in claim 12, further comprising: incubating the support structure and the bioink under physiological conditions for a predetermined duration. 17. The method as recited in claim 16, further comprising: characterizing one or more tissues of the vascular network. 18. The method as recited in claim 17, wherein the characterizing comprises one or more of: optical imaging techniques, fluorescent imaging techniques, radiological imaging techniques, measuring tissue response to one or more compounds; and measuring tissue response to one or more stimuli. 19. The method as recited in claim 12, further comprising removing waste from one or more of: tissues and/or cells in the vascular network; and tissues and/or cells proximate to the vascular network. 20. The method as recited in claim 12, further comprising providing nutrients to one or more of: tissues and/or cells in the vascular network; and tissues and/or cells proximate to the vascular network.
A tissue system includes: a support material; and a vascular network comprising a plurality of channels disposed in the support material. A method includes printing a bioink in a support structure to form a network of vascular precursor materials; and converting the vascular precursor materials into a physiologically relevant vascular network. Notably, the tissue systems, networks, etc. are physiologically-relevant, i.e. exhibiting one or more characteristics indicative of physiological relevance, such as a substantially fractal geometry, inter-vessel spacing, cellular composition, dermal structure, concentric multi-layered structure, etc.1. A tissue system, comprising: a support material, and a vascular network comprising a plurality of channels disposed in the support material, wherein the vascular network is physiologically relevant. 2. The system as recited in claim 1, wherein the vascular network comprises a plurality of constituents selected from a group consisting of endothelial cells (EC), smooth muscle cells, growth factors, and adhesion proteins. 3. The system as recited in claim 2, further comprising a fugitive material configured to vacate an interior cavity of each channel in the vascular network in response to exposure to predetermined conditions. 4. The system as recited in claim 1, wherein the support material comprises one or more of: MATRIGEL™ Stock, MATRIGEL™/GM mixture, EXTRACELL™, PURAMATRIX™, Agarose, Sodium alginate/Calcium (II) chloride, Collagen (Types I-IV), lyophilized/reconstituted human cardiac ECM, gelatin, polyethylene glycol (PEG), polyethylene glycol diacrylate (PEGDA), and/or poly-L-lactic acid (PLLA), a buffer such as phosphate-buffered saline (PBS), and/or one or more cell-type specific culture growth media. 5. The system as recited in claim 1, wherein the vascular network has physical characteristics of being formed from omnidirectional printing of a bioink. 6. The system as recited in claim 1, wherein the vascular network comprises arterial pathways and venous pathways. 7. The system as recited in claim 1, each channel being characterized by an outer diameter in a range from approximately 0.5 microns to approximately 1 mm. 8. The system as recited in claim 1, wherein the channels comprise one or more of: large channels characterized by an outer large channel diameter between about 100 microns and about 20 mm; medium channels characterized by an outer medium channel diameter between about 7 microns and about 150 microns; and capillary channels characterized by an outer capillary diameter between about 5 microns and about 40 microns. 9. The system as recited in claim 1, wherein the vascular network is characterized by an inter-channel spacing between approximately 0.01 microns and approximately 200 microns. 10. The system as recited in claim 1, wherein the vascular network comprises a bifurcating network of the channels. 11. The system as recited in claim 1, wherein the vascular network has physical characteristics of being formed at least in part by vasculogenesis and/or angiogenesis. 12. A method, comprising: printing a bioink in a support structure to form a network of vascular precursor materials; and converting the vascular precursor materials into a physiologically relevant vascular network. 13. The method as recited in claim 12, wherein the printing comprises multiaxial extrusion of the bioink through a nozzle. 14. The method as recited in claim 12, wherein the printing comprises omnidirectional printing. 15. The method as recited in claim 12, wherein the printing forms the network in a geometric arrangement characterized by an inter-channel spacing between approximately 1 micron and approximately 175 microns. 16. The method as recited in claim 12, further comprising: incubating the support structure and the bioink under physiological conditions for a predetermined duration. 17. The method as recited in claim 16, further comprising: characterizing one or more tissues of the vascular network. 18. The method as recited in claim 17, wherein the characterizing comprises one or more of: optical imaging techniques, fluorescent imaging techniques, radiological imaging techniques, measuring tissue response to one or more compounds; and measuring tissue response to one or more stimuli. 19. The method as recited in claim 12, further comprising removing waste from one or more of: tissues and/or cells in the vascular network; and tissues and/or cells proximate to the vascular network. 20. The method as recited in claim 12, further comprising providing nutrients to one or more of: tissues and/or cells in the vascular network; and tissues and/or cells proximate to the vascular network.
1,600
511
15,346,926
1,627
The use of substances capable of inhibiting one or more enzymes of the histone deacetylase family (histone deacetylase inhibitors) for the therapeutic treatment of Philadelphia-negative myeloproliferative syndromes (polycythemia vera, essential thrombocythemia or idiopathic myelofibrosis) is described. The dosage of the above-mentioned substances is significantly lower than that normally used for the care of other tumour syndromes and may be from 10 to 150 mg/day/patient.
1-12. (canceled) 13. Method of treating Philadelphia-negative myeloproliferative syndromes in a patient suffering from said syndromes, said method comprising administering to said patient diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride, or other pharmaceutically acceptable salts and/or solvates, in a daily dosage amount of from 10 to 150 mg per patient. 14. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride is in monohydrate form. 15. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride is in monohydrate crystal form. 16. Method according to claim 13, wherein said daily dosage is from 30 to 120 mg per patient. 17. Method according to claim 13, wherein said daily dosage is from 50 to 100 mg per patient. 18. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is polycythemia vera. 19. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is essential thrombocythemia. 20. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is primary myeofibrosis. 21. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is secondary myelofibrosis. 22. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride, or other pharmaceutically acceptable salts and/or solvates, is not administered in combination with other active principles. 23. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride, or other pharmaceutically acceptable salts and/or solvates, is administered in combination with at least one other cytostatic active principle.
The use of substances capable of inhibiting one or more enzymes of the histone deacetylase family (histone deacetylase inhibitors) for the therapeutic treatment of Philadelphia-negative myeloproliferative syndromes (polycythemia vera, essential thrombocythemia or idiopathic myelofibrosis) is described. The dosage of the above-mentioned substances is significantly lower than that normally used for the care of other tumour syndromes and may be from 10 to 150 mg/day/patient.1-12. (canceled) 13. Method of treating Philadelphia-negative myeloproliferative syndromes in a patient suffering from said syndromes, said method comprising administering to said patient diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride, or other pharmaceutically acceptable salts and/or solvates, in a daily dosage amount of from 10 to 150 mg per patient. 14. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride is in monohydrate form. 15. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride is in monohydrate crystal form. 16. Method according to claim 13, wherein said daily dosage is from 30 to 120 mg per patient. 17. Method according to claim 13, wherein said daily dosage is from 50 to 100 mg per patient. 18. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is polycythemia vera. 19. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is essential thrombocythemia. 20. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is primary myeofibrosis. 21. Method according to claim 13, wherein said Philadelphia-negative myeloproliferative syndrome is secondary myelofibrosis. 22. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride, or other pharmaceutically acceptable salts and/or solvates, is not administered in combination with other active principles. 23. Method according to claim 13, wherein said diethyl-[6-(4-hydroxycarbamoyl-phenylcarbamoyloxymethyl)-naphthalen-2-yl-methyl]-ammonium chloride, or other pharmaceutically acceptable salts and/or solvates, is administered in combination with at least one other cytostatic active principle.
1,600
512
15,414,456
1,642
Disclosed herein are panels related to the diagnosis or recognition of advanced adenoma and colorectal cancer in a subject. The disclosed panels and related methods are used to predict or assess colon tumor status in a patient. They can be used to determine nature of tumor, recurrence, or patient response to treatments. Some embodiments of the methods include generating a report for clinical management.
1. A method of assessing colorectal health of an individual, comprising: obtaining a circulating blood sample from said individual; detecting protein levels for each member of a list of proteins in sample, said list of proteins comprising DPPIV, CO9, TFRC, ORM1, MIF, PKM, SAA, and CEA; and obtaining age information and gender information for said individual. 2. (canceled) 3. The method of claim 1, further comprising performing colonoscopy on said individual. 4. The method of claim 1, further comprising performing a treatment regimen upon said individual. 5. The method of claim 4, wherein said treatment regimen comprises polypectomy. 6. The method of claim 4, wherein said treatment regimen comprises radiation. 7. The method of claim 4, wherein said treatment regimen comprises chemotherapy. 8. (canceled) 9. (canceled) 10. (canceled) 11. (canceled) 12. (canceled) 13. (canceled) 14. (canceled) 15. (canceled) 16. (canceled) 17. (canceled) 18. The method of claim 1, further comprising transmitting a report to a health practitioner of results of said detecting. 19. The method of claim 18, wherein said report indicates recommendation for a colonoscopy for said individual. 20. The method of claim 18, wherein said report indicates recommendation for a polypectomy for said individual. 21. The method of claim 18, wherein said report indicates recommendation for radiation therapy for said individual. 22. The method of claim 18, wherein said report indicates recommendation for chemotherapy for said individual. 23. The method of claim 18, wherein said report indicates recommendation for undergoing an independent cancer assay. 24. The method claim 18, wherein said report indicates recommendation for undergoing a stool cancer assay. 25. The method of claim 1, wherein said list of proteins comprises no more than 15 proteins. 26. The method of claim 1, wherein said list of proteins comprises no more than 8 proteins.
Disclosed herein are panels related to the diagnosis or recognition of advanced adenoma and colorectal cancer in a subject. The disclosed panels and related methods are used to predict or assess colon tumor status in a patient. They can be used to determine nature of tumor, recurrence, or patient response to treatments. Some embodiments of the methods include generating a report for clinical management.1. A method of assessing colorectal health of an individual, comprising: obtaining a circulating blood sample from said individual; detecting protein levels for each member of a list of proteins in sample, said list of proteins comprising DPPIV, CO9, TFRC, ORM1, MIF, PKM, SAA, and CEA; and obtaining age information and gender information for said individual. 2. (canceled) 3. The method of claim 1, further comprising performing colonoscopy on said individual. 4. The method of claim 1, further comprising performing a treatment regimen upon said individual. 5. The method of claim 4, wherein said treatment regimen comprises polypectomy. 6. The method of claim 4, wherein said treatment regimen comprises radiation. 7. The method of claim 4, wherein said treatment regimen comprises chemotherapy. 8. (canceled) 9. (canceled) 10. (canceled) 11. (canceled) 12. (canceled) 13. (canceled) 14. (canceled) 15. (canceled) 16. (canceled) 17. (canceled) 18. The method of claim 1, further comprising transmitting a report to a health practitioner of results of said detecting. 19. The method of claim 18, wherein said report indicates recommendation for a colonoscopy for said individual. 20. The method of claim 18, wherein said report indicates recommendation for a polypectomy for said individual. 21. The method of claim 18, wherein said report indicates recommendation for radiation therapy for said individual. 22. The method of claim 18, wherein said report indicates recommendation for chemotherapy for said individual. 23. The method of claim 18, wherein said report indicates recommendation for undergoing an independent cancer assay. 24. The method claim 18, wherein said report indicates recommendation for undergoing a stool cancer assay. 25. The method of claim 1, wherein said list of proteins comprises no more than 15 proteins. 26. The method of claim 1, wherein said list of proteins comprises no more than 8 proteins.
1,600
513
14,432,011
1,611
The present invention relates to an emulsion-type cosmetic composition for coating keratin fibres, comprising: -an aqueous phase, -particles comprising: *at least one hard wax, preferentially in the form of an aqueous dispersion, having a melting point ranging from 65° C. to 120° C., the hard wax(es) being present at a total content greater than or equal to 10% by weight relative to the total weight of the composition, *at least one aqueous dispersion of particles of at least one film-forming polymer, the film forming polymer(s) being present at a dry matter content greater than or equal to 10% by weight relative to the total weight of the composition, -at least one emulsifying system capable of dispersing at least the hard wax(es), comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8, preferably greater than or equal to 10. The present invention also relates to a method for coating keratin fibres, to a method for preparing a cosmetic composition for coating keratin fibres, to a cosmetic composition for coating keratin fibres which is obtained by means of said production method, and also to a particular use.
1. An emulsion-type cosmetic composition, comprising: an aqueous phase; particles comprising: a hard wax having a melting point ranging from 65 to 120° C., the hard wax being present at a total content greater than or equal to 10% by weight relative to [[the]] a total weight of the composition, and an aqueous dispersion of particles of a film-forming polymer, the film-forming polymer being present at a dry matter content greater than or equal to 10% by weight relative to the total weight of the composition; and an emulsifying system capable of dispersing at least the hard wax, the emulsifying system comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8. 2. The composition according to claim 1, wherein the aqueous phase is from 30% to 80% by weight, relative to the total weight of the composition. 3. The composition according to claim 1, wherein the hard wax is polar. 4. The composition according to claim 1, wherein the particles of the hard wax are not introduced into the preparation of the composition in the form of a preprepared aqueous microdispersion of the hard wax. 5. The composition according to claim 1, wherein the particles of the film-forming polymer is introduced into the preparation of the composition in the form of a preprepared aqueous dispersion of film-forming polymer. 6. The composition according to claim 1, comprising a solids content greater than or equal to 42%. 7. The composition according to claim 1, wherein the particles of the hard wax and of the film-forming polymer have an average size expressed as volume-average “effective” diameter D[4,3] of less than or equal to 5 μm. 8. The composition according to claim 1, wherein the particles of the hard wax and of the film-forming polymer are present at a content greater than or equal to 30% by weight, relative to the total weight of the composition. 9. The composition according to claim 1, comprising a total content of particles of the hard wax, of greater than or equal to 12% by weight relative to the total weight of the composition. 10. The composition according to claim 1, comprising a total content of particles of the hard wax of at least 80% by weight, relative to the total weight of waxes. 11. The composition according to claim 1, comprising a total content of particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, of greater than or equal to 12% by weight, relative to the total weight of the composition. 12. The composition according to claim 1, comprising a total content of particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, of at least 80% by weight, relative to the total weight of the film-forming polymer(s) polymer. 13. The composition according to claim 1, wherein the particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, are chosen from synthetic polymers of free-radical type or of polycondensate type, polymers of natural origin, and mixtures thereof. 14. The composition according to claim 1, wherein the particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, are chosen from dispersions of acrylic polymers, dispersions of polyurethane, dispersions of sulfopolyesters, vinyl dispersions, dispersions of polyvinyl acetate, dispersions of vinylpyrrolidone/dimethylaminopropylmethacrylamide/lauryldimethylpropylmethacrylamidoammonium chloride terpolymer, dispersions of polyurethane/polyacrylic hybrid polymers, dispersions of particles of core-shell type, and mixtures thereof. 15. The composition according to claim 1, wherein the total content of particles of the hard wax and the total content of particles of the film-forming polymer, are such that the weight ratio of the particles of the hard wax to the particles of the film-forming polymer is greater than or equal to 1/2. 16. The composition according to claim 1, wherein the non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8, is chosen from: glycerol ethers, oxyalkylenated alcohols, fatty acid esters of polyethylene glycol, esters of fatty acids and of glycerol ethers, esters of fatty acids and of sorbitol ethers, and mixtures thereof. 17. The composition according to claim 1, wherein the emulsifying system comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8 is present at a content greater than or equal to 3% by weight relative to the total weight of the composition. 18. The composition according to claim 1, wherein the composition is free of an anionic surfactant. 19. The composition according to claim 1, comprising a colorant chosen from a pulverulent colorant. 20. The composition according to claim 1, having a viscosity at 25° C. ranging from 5 to 50 Pa·s. 21. A method for coating keratin fibres, comprising applying a cosmetic composition according to claim 1 to keratin fibres. 22. A method for preparing a cosmetic composition comprising: in a first phase, heating a hard wax and optionally an additional wax at a temperature above its melting point in order to melt the wax, adding an emulsifying system capable of dispersing at least the hard wax, adding water, wherein a water content added is greater than 25% by weight relative to the total weight of this first phase, and a total content of the emulsifying system is such that a weight ratio of the hard wax plus the additional wax/emulsifying system is inclusively between 2 and 6, adding a colorant, optionally adding a compound of thickening nature, wherein water is not present in the beginning of the first phase in order to avoid any early evaporation, and emulsifying the whole mixture with stirring at a temperature above the melting point of the wax: in a second phase, placing an aqueous phase dispersion of particles of film-forming polymers, in a vessel in which the temperature is inclusively between 0 and 45° C. the order in which the first phase and the second phase are prepared being of no importance; bringing the first and second phases together by pouring the first phase, still at a temperature above the melting point of the wax, into the vessel comprising the second phase having a temperature between 0 and 45° C.; leaving to stir until the temperature of the mixture stabilizes at the temperature, which is preferably regulated, between 0 and 45° C; and optionally adding a preserving system. 23. The method of preparation according to claim 22, wherein the method obtains a makeup compositions. 24. A cosmetic composition, comprising: an aqueous phase; particles comprising: a hard wax having a melting point ranging from 65 to 120° C., the hard wax being present at a total content greater than or equal to 10% by weight relative to a total weight of the composition, an aqueous dispersion of particles of a film-forming polymer, the film-forming polymer being present at a dry matter content greater than or equal to 10% by weight relative to the total weight of the composition; and an emulsifying system capable of dispersing at least the hard wax, the emulsifying system comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8, wherein the composition is obtained by the method of preparation according to claim 22.
The present invention relates to an emulsion-type cosmetic composition for coating keratin fibres, comprising: -an aqueous phase, -particles comprising: *at least one hard wax, preferentially in the form of an aqueous dispersion, having a melting point ranging from 65° C. to 120° C., the hard wax(es) being present at a total content greater than or equal to 10% by weight relative to the total weight of the composition, *at least one aqueous dispersion of particles of at least one film-forming polymer, the film forming polymer(s) being present at a dry matter content greater than or equal to 10% by weight relative to the total weight of the composition, -at least one emulsifying system capable of dispersing at least the hard wax(es), comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8, preferably greater than or equal to 10. The present invention also relates to a method for coating keratin fibres, to a method for preparing a cosmetic composition for coating keratin fibres, to a cosmetic composition for coating keratin fibres which is obtained by means of said production method, and also to a particular use.1. An emulsion-type cosmetic composition, comprising: an aqueous phase; particles comprising: a hard wax having a melting point ranging from 65 to 120° C., the hard wax being present at a total content greater than or equal to 10% by weight relative to [[the]] a total weight of the composition, and an aqueous dispersion of particles of a film-forming polymer, the film-forming polymer being present at a dry matter content greater than or equal to 10% by weight relative to the total weight of the composition; and an emulsifying system capable of dispersing at least the hard wax, the emulsifying system comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8. 2. The composition according to claim 1, wherein the aqueous phase is from 30% to 80% by weight, relative to the total weight of the composition. 3. The composition according to claim 1, wherein the hard wax is polar. 4. The composition according to claim 1, wherein the particles of the hard wax are not introduced into the preparation of the composition in the form of a preprepared aqueous microdispersion of the hard wax. 5. The composition according to claim 1, wherein the particles of the film-forming polymer is introduced into the preparation of the composition in the form of a preprepared aqueous dispersion of film-forming polymer. 6. The composition according to claim 1, comprising a solids content greater than or equal to 42%. 7. The composition according to claim 1, wherein the particles of the hard wax and of the film-forming polymer have an average size expressed as volume-average “effective” diameter D[4,3] of less than or equal to 5 μm. 8. The composition according to claim 1, wherein the particles of the hard wax and of the film-forming polymer are present at a content greater than or equal to 30% by weight, relative to the total weight of the composition. 9. The composition according to claim 1, comprising a total content of particles of the hard wax, of greater than or equal to 12% by weight relative to the total weight of the composition. 10. The composition according to claim 1, comprising a total content of particles of the hard wax of at least 80% by weight, relative to the total weight of waxes. 11. The composition according to claim 1, comprising a total content of particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, of greater than or equal to 12% by weight, relative to the total weight of the composition. 12. The composition according to claim 1, comprising a total content of particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, of at least 80% by weight, relative to the total weight of the film-forming polymer(s) polymer. 13. The composition according to claim 1, wherein the particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, are chosen from synthetic polymers of free-radical type or of polycondensate type, polymers of natural origin, and mixtures thereof. 14. The composition according to claim 1, wherein the particles of the film-forming polymer, as an aqueous dispersion or aqueous dispersions, are chosen from dispersions of acrylic polymers, dispersions of polyurethane, dispersions of sulfopolyesters, vinyl dispersions, dispersions of polyvinyl acetate, dispersions of vinylpyrrolidone/dimethylaminopropylmethacrylamide/lauryldimethylpropylmethacrylamidoammonium chloride terpolymer, dispersions of polyurethane/polyacrylic hybrid polymers, dispersions of particles of core-shell type, and mixtures thereof. 15. The composition according to claim 1, wherein the total content of particles of the hard wax and the total content of particles of the film-forming polymer, are such that the weight ratio of the particles of the hard wax to the particles of the film-forming polymer is greater than or equal to 1/2. 16. The composition according to claim 1, wherein the non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8, is chosen from: glycerol ethers, oxyalkylenated alcohols, fatty acid esters of polyethylene glycol, esters of fatty acids and of glycerol ethers, esters of fatty acids and of sorbitol ethers, and mixtures thereof. 17. The composition according to claim 1, wherein the emulsifying system comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8 is present at a content greater than or equal to 3% by weight relative to the total weight of the composition. 18. The composition according to claim 1, wherein the composition is free of an anionic surfactant. 19. The composition according to claim 1, comprising a colorant chosen from a pulverulent colorant. 20. The composition according to claim 1, having a viscosity at 25° C. ranging from 5 to 50 Pa·s. 21. A method for coating keratin fibres, comprising applying a cosmetic composition according to claim 1 to keratin fibres. 22. A method for preparing a cosmetic composition comprising: in a first phase, heating a hard wax and optionally an additional wax at a temperature above its melting point in order to melt the wax, adding an emulsifying system capable of dispersing at least the hard wax, adding water, wherein a water content added is greater than 25% by weight relative to the total weight of this first phase, and a total content of the emulsifying system is such that a weight ratio of the hard wax plus the additional wax/emulsifying system is inclusively between 2 and 6, adding a colorant, optionally adding a compound of thickening nature, wherein water is not present in the beginning of the first phase in order to avoid any early evaporation, and emulsifying the whole mixture with stirring at a temperature above the melting point of the wax: in a second phase, placing an aqueous phase dispersion of particles of film-forming polymers, in a vessel in which the temperature is inclusively between 0 and 45° C. the order in which the first phase and the second phase are prepared being of no importance; bringing the first and second phases together by pouring the first phase, still at a temperature above the melting point of the wax, into the vessel comprising the second phase having a temperature between 0 and 45° C.; leaving to stir until the temperature of the mixture stabilizes at the temperature, which is preferably regulated, between 0 and 45° C; and optionally adding a preserving system. 23. The method of preparation according to claim 22, wherein the method obtains a makeup compositions. 24. A cosmetic composition, comprising: an aqueous phase; particles comprising: a hard wax having a melting point ranging from 65 to 120° C., the hard wax being present at a total content greater than or equal to 10% by weight relative to a total weight of the composition, an aqueous dispersion of particles of a film-forming polymer, the film-forming polymer being present at a dry matter content greater than or equal to 10% by weight relative to the total weight of the composition; and an emulsifying system capable of dispersing at least the hard wax, the emulsifying system comprising a non-ionic surfactant with an HLB value at 25° C. greater than or equal to 8, wherein the composition is obtained by the method of preparation according to claim 22.
1,600
514
14,762,043
1,613
The present invention relates to a cosmetic or dermatological composition comprising, in a physiologically acceptable support: a) at least one merocyanine compound of formula (1) or (2) and b) at least one oily phase and c) at least one C 1 -C 4 monoalkanol. The present invention also relates to a non-therapeutic cosmetic process for caring for and/or making up a keratin material, comprising the application, to the surface of the said keratin material, of at least one composition as defined above. The invention also relates to a non-therapeutic cosmetic process for limiting the darkening of the skin and/or improving the colour and/or uniformity of the complexion, comprising the application, to the surface of the skin, of at least one composition as defined previously. The invention also relates to a non-therapeutic cosmetic process for preventing and/or treating the signs of ageing of a keratin material, comprising the application, to the surface of the keratin material, of at least one composition as defined previously.
1. Cosmetic or dermatological composition comprising, in a physiologically acceptable support: a) at least one merocyanine compound corresponding to one of the following formulae (1) and (2) or one of the E/E- or E/Z-geometrical isomer forms thereof: in which: R1 and R2 are, independently of each other, hydrogen; a C1-C22 alkyl group, a C2-C22 alkenyl group or a C2-C22 alkynyl group, it being possible for these groups to be substituted with at least one hydroxyl group or to be interrupted with at least one —O—; or alternatively R1 and R2 form, together with the nitrogen atom which connects them, a —(CH2)n— ring which may optionally be interrupted with —O— or —NH—; R3 is a group —(C═O)OR6 or a group —(CO)NHR6; R6 is a C1-C22 alkyl group, a C2-C22 alkenyl group, a C2-C22 alkynyl group, a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be substituted with one or more OH groups; R4 and R5 are hydrogens; or R4 and R5 form a —(CH2)n— ring which may be substituted with a C1-C4 alkyl group and/or interrupted with one or more —O— or with —NH—; n is a number between 2 and 7; R7 and R8 are, independently of each other, hydrogen; a C1-C22 alkyl group, a C2-C22 alkenyl group or a C2-C22 alkynyl group, it being possible for the said groups to be interrupted with one or more O and/or substituted with one or more OH groups; a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be interrupted with one or more —O—; or alternatively R7 and R8 form, together with the nitrogen which connects them, a —(CH2)n— ring which may be interrupted with one or more —O—; R9 and R10 are hydrogen; or R9 and R10 form a —(CH2)n— ring which may be substituted with a C1-C4 alkyl and/or interrupted with an —O— or —NH—; A is —O— or —NH; R11 is a C1-C22 alkyl group; a C2-C22 alkenyl group; a C2-C22 alkynyl group; a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be interrupted with one or more O; or a C1-C22 alkyl group or a C2-C22 alkenyl group which is substituted with a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said C3-C22 cycloalkyl group or C3-C22 cycloalkenyl group to be interrupted with one or more —O—; with the proviso that: (I) at least one of the groups R1, R2 or R6 is substituted with a hydroxyl; (II) if one of the groups R1 denotes a hydroxyethyl, R2 does not denote a hydrogen, a methyl or an ethyl, or a hydroxyethyl; and if R1 denotes hydrogen, R2 is not 1-hydroxy-3-methyl-2-butyl; (III) if R6 is substituted with one or more OH groups, one from among R1 and R2 is a C4-C22 alkyl group; or alternatively R1 and R2 form, together with the nitrogen to which they are attached, a piperidyl or morpholinyl radical; (IV) at least one among the radicals R7, R8 and R11 is interrupted with one or more —O—; and b) at least one oily phase, and c) at least one C1-C4 monoalkanol. 2. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R6 is a C1-C12 alkyl group which may be substituted with one or more hydroxyls. 3. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R6 is a C1-C12 alkyl group which may be substituted with one or more hydroxyls; one of the radicals R1 or R2 is a C4-C22 alkyl group; or alternatively R1 and R2 form, together with the nitrogen which connects them, a —(CH2)n— ring which may be interrupted with —O— and/or —NH. 4. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R11 is a radical —(CH2)m—O—R12, in which R12 is a C1-C12 alkyl group or a C1-C6-alkoxy-C1-C6-alkyl group; m is a number from 1 to 5. 5. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from those for which: R1 and R2, on the one hand, and R7 and R8, on the other hand, respectively form, together with the nitrogen atom to which they are respectively attached, a piperidyl radical or a morpholinyl radical. 6. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from those for which: R4 and R5 and R9 and R10 respectively form a carbon-based ring which contains 6 carbon atoms. 7. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R1 and R2 are, independently of each other, a hydrogen; or a C1-C22 alkyl group; or a C1-C22 hydroxyalkyl group; or R1 and R2 form, together with the nitrogen to which they are attached, a piperidyl or morpholinyl radical; R3 is a group —(C═O)OR6 or a group —(CO)NHR6; R6 is a C1-C22 alkyl group which may be substituted with one or more —OH groups; R4 and R5 are a hydrogen; or R4 and R5 are linked together to form a carbon-based ring which contains 6 carbon atoms. 8. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R1 and R2 are, independently of each other, a hydrogen; or a C1-C22 hydroxyalkyl group; in which at least one of the R1 and R2 radicals is a C1-C22 hydroxyalkyl group; R3 is a group —(C═O)OR6 or a group —(C═O)NHR6 ; R6 is a C1-C22 alkyl group; and R4 and R5 are hydrogens; or R4 and R5 are linked together to form a carbon-based ring which contains 6 carbon atoms. 9. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R7 and R8 are, independently of each other, a hydrogen or a C1-C8 alkyl group which may be interrupted with one or more —O—; A is —O— or —NH; R11 is a C1-C22 alkyl; and R9 and R10 are a hydrogen; or R9 and R10 are linked together to form a carbon-based ring which contains 6 carbon atoms. 10. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R7 and R8 form, together with the nitrogen atom to which they are attached, a morpholinyl or piperidyl radical; A is —O— or —NH; R11 is a C1-C22 alkyl group which may be interrupted with one or more —O—; and R9 and R10 are hydrogens; or R9 and R10 are linked together to form a carbon-based ring which contains 6 carbon atoms. 11. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R11 is a radical —(CH2)m—O—R12, in which R12 is a C1-C4 alkyl group or a C1-C4-alkoxy-C1-C4-alkyl group; m is a number from 1 to 3; R7 and R8 are, independently of each other, a hydrogen; a C1-C12 alkyl group which may be interrupted with one or more O; or R7 and R8 form, together with the nitrogen atom to which they are attached, a morpholinyl or piperidyl radical; R9 and R10 are hydrogens or together form a carbon-based ring which contains 6 carbon atoms; and A is —O— or —NH. 12. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from the following compounds and also the E/E- or E/Z-geometrical isomer forms thereof: TABLE A Compound Structure 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 13. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from those corresponding to the following formula (3) and also the E/E- or E/Z-geometrical isomer forms thereof: in which: A is —O— or —NH; R is a C1-C22 alkyl group, a C2-C22 alkenyl group, a C2-C22 alkynyl group, a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be interrupted with one or more O. 14. Composition according to claim 13, in which the merocyanines of formula (3) are chosen from the following compounds and also the E/E- or E/Z-geometrical isomer forms thereof: 15. Composition according to claim 14, in which the merocyanine of formula (3) is the compound 2-ethoxyethyl (2Z)-cyano{3-[(3-methoxypropyl)amino]cyclohex-2-en-1-ylidene}ethanoate (25) in its E/Z geometrical configuration having the following structure: and/or the E/E form having the following structure: 16. Composition according to claim 1, in which the monoalkanol is ethanol. 17. Composition according to claim 1, in which the merocyanine compound(s) of formula (I) are present in a concentration ranging from 0.1% to 10% by weight relative to the total weight of the composition. 18. Composition according to claim 1, in which the overall oily phase represents from 5% to 95% by weight relative to the total weight of the composition. 19. Composition according to claim 1, in which the C1-C4 monoalkanol(s) are present in concentrations ranging from 0.2% to 90% by weight relative to the total weight of the composition. 20. Non-therapeutic cosmetic process for caring for and/or making up a keratin material, comprising the application, to the surface of the said keratin material, of at least one composition as defined in claim 1. 21. Non-therapeutic cosmetic process for limiting the darkening of the skin and/or improving the colour and/or uniformity of the complexion, comprising the application, to the surface of the skin, of at least one composition as defined in claim 1. 22. Non-therapeutic cosmetic process for preventing and/or treating the signs of ageing of a keratin material, comprising the application, to the surface of the keratin material, of at least one composition as defined in claim 1.
The present invention relates to a cosmetic or dermatological composition comprising, in a physiologically acceptable support: a) at least one merocyanine compound of formula (1) or (2) and b) at least one oily phase and c) at least one C 1 -C 4 monoalkanol. The present invention also relates to a non-therapeutic cosmetic process for caring for and/or making up a keratin material, comprising the application, to the surface of the said keratin material, of at least one composition as defined above. The invention also relates to a non-therapeutic cosmetic process for limiting the darkening of the skin and/or improving the colour and/or uniformity of the complexion, comprising the application, to the surface of the skin, of at least one composition as defined previously. The invention also relates to a non-therapeutic cosmetic process for preventing and/or treating the signs of ageing of a keratin material, comprising the application, to the surface of the keratin material, of at least one composition as defined previously.1. Cosmetic or dermatological composition comprising, in a physiologically acceptable support: a) at least one merocyanine compound corresponding to one of the following formulae (1) and (2) or one of the E/E- or E/Z-geometrical isomer forms thereof: in which: R1 and R2 are, independently of each other, hydrogen; a C1-C22 alkyl group, a C2-C22 alkenyl group or a C2-C22 alkynyl group, it being possible for these groups to be substituted with at least one hydroxyl group or to be interrupted with at least one —O—; or alternatively R1 and R2 form, together with the nitrogen atom which connects them, a —(CH2)n— ring which may optionally be interrupted with —O— or —NH—; R3 is a group —(C═O)OR6 or a group —(CO)NHR6; R6 is a C1-C22 alkyl group, a C2-C22 alkenyl group, a C2-C22 alkynyl group, a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be substituted with one or more OH groups; R4 and R5 are hydrogens; or R4 and R5 form a —(CH2)n— ring which may be substituted with a C1-C4 alkyl group and/or interrupted with one or more —O— or with —NH—; n is a number between 2 and 7; R7 and R8 are, independently of each other, hydrogen; a C1-C22 alkyl group, a C2-C22 alkenyl group or a C2-C22 alkynyl group, it being possible for the said groups to be interrupted with one or more O and/or substituted with one or more OH groups; a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be interrupted with one or more —O—; or alternatively R7 and R8 form, together with the nitrogen which connects them, a —(CH2)n— ring which may be interrupted with one or more —O—; R9 and R10 are hydrogen; or R9 and R10 form a —(CH2)n— ring which may be substituted with a C1-C4 alkyl and/or interrupted with an —O— or —NH—; A is —O— or —NH; R11 is a C1-C22 alkyl group; a C2-C22 alkenyl group; a C2-C22 alkynyl group; a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be interrupted with one or more O; or a C1-C22 alkyl group or a C2-C22 alkenyl group which is substituted with a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said C3-C22 cycloalkyl group or C3-C22 cycloalkenyl group to be interrupted with one or more —O—; with the proviso that: (I) at least one of the groups R1, R2 or R6 is substituted with a hydroxyl; (II) if one of the groups R1 denotes a hydroxyethyl, R2 does not denote a hydrogen, a methyl or an ethyl, or a hydroxyethyl; and if R1 denotes hydrogen, R2 is not 1-hydroxy-3-methyl-2-butyl; (III) if R6 is substituted with one or more OH groups, one from among R1 and R2 is a C4-C22 alkyl group; or alternatively R1 and R2 form, together with the nitrogen to which they are attached, a piperidyl or morpholinyl radical; (IV) at least one among the radicals R7, R8 and R11 is interrupted with one or more —O—; and b) at least one oily phase, and c) at least one C1-C4 monoalkanol. 2. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R6 is a C1-C12 alkyl group which may be substituted with one or more hydroxyls. 3. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R6 is a C1-C12 alkyl group which may be substituted with one or more hydroxyls; one of the radicals R1 or R2 is a C4-C22 alkyl group; or alternatively R1 and R2 form, together with the nitrogen which connects them, a —(CH2)n— ring which may be interrupted with —O— and/or —NH. 4. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R11 is a radical —(CH2)m—O—R12, in which R12 is a C1-C12 alkyl group or a C1-C6-alkoxy-C1-C6-alkyl group; m is a number from 1 to 5. 5. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from those for which: R1 and R2, on the one hand, and R7 and R8, on the other hand, respectively form, together with the nitrogen atom to which they are respectively attached, a piperidyl radical or a morpholinyl radical. 6. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from those for which: R4 and R5 and R9 and R10 respectively form a carbon-based ring which contains 6 carbon atoms. 7. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R1 and R2 are, independently of each other, a hydrogen; or a C1-C22 alkyl group; or a C1-C22 hydroxyalkyl group; or R1 and R2 form, together with the nitrogen to which they are attached, a piperidyl or morpholinyl radical; R3 is a group —(C═O)OR6 or a group —(CO)NHR6; R6 is a C1-C22 alkyl group which may be substituted with one or more —OH groups; R4 and R5 are a hydrogen; or R4 and R5 are linked together to form a carbon-based ring which contains 6 carbon atoms. 8. Composition according to claim 1, in which the compounds of formula (1) are chosen from those for which: R1 and R2 are, independently of each other, a hydrogen; or a C1-C22 hydroxyalkyl group; in which at least one of the R1 and R2 radicals is a C1-C22 hydroxyalkyl group; R3 is a group —(C═O)OR6 or a group —(C═O)NHR6 ; R6 is a C1-C22 alkyl group; and R4 and R5 are hydrogens; or R4 and R5 are linked together to form a carbon-based ring which contains 6 carbon atoms. 9. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R7 and R8 are, independently of each other, a hydrogen or a C1-C8 alkyl group which may be interrupted with one or more —O—; A is —O— or —NH; R11 is a C1-C22 alkyl; and R9 and R10 are a hydrogen; or R9 and R10 are linked together to form a carbon-based ring which contains 6 carbon atoms. 10. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R7 and R8 form, together with the nitrogen atom to which they are attached, a morpholinyl or piperidyl radical; A is —O— or —NH; R11 is a C1-C22 alkyl group which may be interrupted with one or more —O—; and R9 and R10 are hydrogens; or R9 and R10 are linked together to form a carbon-based ring which contains 6 carbon atoms. 11. Composition according to claim 1, in which the compounds of formula (2) are chosen from those for which: R11 is a radical —(CH2)m—O—R12, in which R12 is a C1-C4 alkyl group or a C1-C4-alkoxy-C1-C4-alkyl group; m is a number from 1 to 3; R7 and R8 are, independently of each other, a hydrogen; a C1-C12 alkyl group which may be interrupted with one or more O; or R7 and R8 form, together with the nitrogen atom to which they are attached, a morpholinyl or piperidyl radical; R9 and R10 are hydrogens or together form a carbon-based ring which contains 6 carbon atoms; and A is —O— or —NH. 12. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from the following compounds and also the E/E- or E/Z-geometrical isomer forms thereof: TABLE A Compound Structure 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 13. Composition according to claim 1, in which the compounds of formula (1) or (2) are chosen from those corresponding to the following formula (3) and also the E/E- or E/Z-geometrical isomer forms thereof: in which: A is —O— or —NH; R is a C1-C22 alkyl group, a C2-C22 alkenyl group, a C2-C22 alkynyl group, a C3-C22 cycloalkyl group or a C3-C22 cycloalkenyl group, it being possible for the said groups to be interrupted with one or more O. 14. Composition according to claim 13, in which the merocyanines of formula (3) are chosen from the following compounds and also the E/E- or E/Z-geometrical isomer forms thereof: 15. Composition according to claim 14, in which the merocyanine of formula (3) is the compound 2-ethoxyethyl (2Z)-cyano{3-[(3-methoxypropyl)amino]cyclohex-2-en-1-ylidene}ethanoate (25) in its E/Z geometrical configuration having the following structure: and/or the E/E form having the following structure: 16. Composition according to claim 1, in which the monoalkanol is ethanol. 17. Composition according to claim 1, in which the merocyanine compound(s) of formula (I) are present in a concentration ranging from 0.1% to 10% by weight relative to the total weight of the composition. 18. Composition according to claim 1, in which the overall oily phase represents from 5% to 95% by weight relative to the total weight of the composition. 19. Composition according to claim 1, in which the C1-C4 monoalkanol(s) are present in concentrations ranging from 0.2% to 90% by weight relative to the total weight of the composition. 20. Non-therapeutic cosmetic process for caring for and/or making up a keratin material, comprising the application, to the surface of the said keratin material, of at least one composition as defined in claim 1. 21. Non-therapeutic cosmetic process for limiting the darkening of the skin and/or improving the colour and/or uniformity of the complexion, comprising the application, to the surface of the skin, of at least one composition as defined in claim 1. 22. Non-therapeutic cosmetic process for preventing and/or treating the signs of ageing of a keratin material, comprising the application, to the surface of the keratin material, of at least one composition as defined in claim 1.
1,600
515
14,356,109
1,653
A high yield method for fermenting carbohydrate to ethanol and prevention and/or disruption of biofilms, comprising: a) mixing a fermentation feedstock with a fermentation broth containing yeast and/or an enzyme, b) treating said mixture by adding a composition to the fermentor containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde, and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 0-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from Ci to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde in the fermentor is from about 0.25 to 3 kg/MT of fermentation feedstock, and c) isolating ethanol.
1. A high yield method of fermenting carbohydrate to ethanol in a fermentor, comprising: a) mixing a fermentation feedstock with a fermentation broth containing yeast and/or an enzyme, b) treating said mixture by adding a composition to the fermentor containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde, and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 0-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from C1 to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde in the fermentor is from about 0.25 to 3 kg/MT of fermentation feedstock, and c) isolating ethanol. 2. The method of claim 1, wherein the fermentation feedstock is corn, sorghum, wheat, triticale, rye, barley, rice or tubers. 3. The method of claim 1, wherein the fermentation feedstock is sugar cane or sugar beet. 4. The method of claim 1, wherein the carbohydrate to be fermented is derived from cellulose. 5. The method claim 1, wherein development of antibiotic-resistant strains of bacteria is inhibited. 6. A fermentation broth or slurry, comprising: a) carbohydrate feedstock to be fermented, yeast, and/or an enzyme, and b) a treatment composition containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 1-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from C1 to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde is from about 0.25 to 3 kg/MT of fermentation feedstock. 7. The fermentation broth of claim 6, wherein the carbohydrate feedstock is corn, sorghum, wheat, triticale, rye, barley, rice or tubers, and the aldehyde is formaldehyde with a concentration of 0.25 to 3.0 kg/MT. 8. The fermentation broth of claim 6, wherein the carbohydrate feedstock is sugar cane or sugar beet. 9. The fermentation broth of claim 6, wherein the carbohydrate feedstock is derived from cellulose. 10. The fermentation broth of claim 6, wherein development of antibiotic-resistant strains of bacteria is inhibited. 11. An improved method of fermenting carbohydrate to ethanol in a fermentor, comprising: a) mixing a fermentation feedstock with a fermentation broth containing yeast and/or an enzyme, b) treating said mixture by adding a composition to the fermentor containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde, and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 0-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from C1 to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde in the fermentor is from about 0.25 to 3 kg/MT of fermentation feedstock, and c) isolating ethanol, d) collecting material remaining after fermentation and adding it to animal feed. 12. The method of claim 11, wherein the organic acid is formic, acetic, propionic, or butyric. 13. The method of claim 11, comprising an antibiotic to control bacteria in an amount less than its MIC in fermentations without composition b). 14. The method of claim 11, which is free of antibiotic used to control bacteria in fermentation. 15. The method of claim 11, wherein bacteria comprise Lactobacillus spp. E. coli, Salmonella spp., Clostridium spp., Campylobacter spp., Shigella spp., Brachyspira spp., Listeria spp., Arcobacter spp, Pediococcus, Staphylococcus, Enterococcus, Acetobacter, Gluconobacter, A. pasterurianus, B. Subtilis, Leuconostoc mesenteroides, Weissella paramesenteroides and bacteria able to produce biofilms in solid surfaces. 16. The method of claim 11, which is free of virginiamycin or sulfuric acid. 17. The method of claim 11, wherein the carbohydrate feedstock is corn, sorghum, wheat, triticale, rye, barley, rice or tubers, and the aldehyde is formaldehyde with a concentration of 0.25 to 3.0 kg/MT. 18. The method of claim 11, wherein the carbohydrate feedstock is sugar cane or sugar beet. 19. The method of claim 11, wherein the carbohydrate feedstock is derived from cellulose. 20. The method of claim 11, wherein development of antibiotic-resistant strains of bacteria is inhibited.
A high yield method for fermenting carbohydrate to ethanol and prevention and/or disruption of biofilms, comprising: a) mixing a fermentation feedstock with a fermentation broth containing yeast and/or an enzyme, b) treating said mixture by adding a composition to the fermentor containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde, and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 0-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from Ci to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde in the fermentor is from about 0.25 to 3 kg/MT of fermentation feedstock, and c) isolating ethanol.1. A high yield method of fermenting carbohydrate to ethanol in a fermentor, comprising: a) mixing a fermentation feedstock with a fermentation broth containing yeast and/or an enzyme, b) treating said mixture by adding a composition to the fermentor containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde, and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 0-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from C1 to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde in the fermentor is from about 0.25 to 3 kg/MT of fermentation feedstock, and c) isolating ethanol. 2. The method of claim 1, wherein the fermentation feedstock is corn, sorghum, wheat, triticale, rye, barley, rice or tubers. 3. The method of claim 1, wherein the fermentation feedstock is sugar cane or sugar beet. 4. The method of claim 1, wherein the carbohydrate to be fermented is derived from cellulose. 5. The method claim 1, wherein development of antibiotic-resistant strains of bacteria is inhibited. 6. A fermentation broth or slurry, comprising: a) carbohydrate feedstock to be fermented, yeast, and/or an enzyme, and b) a treatment composition containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 1-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from C1 to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde is from about 0.25 to 3 kg/MT of fermentation feedstock. 7. The fermentation broth of claim 6, wherein the carbohydrate feedstock is corn, sorghum, wheat, triticale, rye, barley, rice or tubers, and the aldehyde is formaldehyde with a concentration of 0.25 to 3.0 kg/MT. 8. The fermentation broth of claim 6, wherein the carbohydrate feedstock is sugar cane or sugar beet. 9. The fermentation broth of claim 6, wherein the carbohydrate feedstock is derived from cellulose. 10. The fermentation broth of claim 6, wherein development of antibiotic-resistant strains of bacteria is inhibited. 11. An improved method of fermenting carbohydrate to ethanol in a fermentor, comprising: a) mixing a fermentation feedstock with a fermentation broth containing yeast and/or an enzyme, b) treating said mixture by adding a composition to the fermentor containing: 10-90 wt. % of an aldehyde selected from the group consisting of formaldehyde, para-formaldehyde, glutaraldehyde, another antimicrobial aldehyde, and mixtures thereof, 1-50 wt. % of a surfactant having an HLB from 4 to 18, 0-20 wt. % of an antimicrobial terpene, or essential oils, 1-50 wt. % of organic acids selected from C1 to C24 fatty acids, their salts, glycerides and esters thereof, and 1-50 wt. % water; wherein the concentration of aldehyde in the fermentor is from about 0.25 to 3 kg/MT of fermentation feedstock, and c) isolating ethanol, d) collecting material remaining after fermentation and adding it to animal feed. 12. The method of claim 11, wherein the organic acid is formic, acetic, propionic, or butyric. 13. The method of claim 11, comprising an antibiotic to control bacteria in an amount less than its MIC in fermentations without composition b). 14. The method of claim 11, which is free of antibiotic used to control bacteria in fermentation. 15. The method of claim 11, wherein bacteria comprise Lactobacillus spp. E. coli, Salmonella spp., Clostridium spp., Campylobacter spp., Shigella spp., Brachyspira spp., Listeria spp., Arcobacter spp, Pediococcus, Staphylococcus, Enterococcus, Acetobacter, Gluconobacter, A. pasterurianus, B. Subtilis, Leuconostoc mesenteroides, Weissella paramesenteroides and bacteria able to produce biofilms in solid surfaces. 16. The method of claim 11, which is free of virginiamycin or sulfuric acid. 17. The method of claim 11, wherein the carbohydrate feedstock is corn, sorghum, wheat, triticale, rye, barley, rice or tubers, and the aldehyde is formaldehyde with a concentration of 0.25 to 3.0 kg/MT. 18. The method of claim 11, wherein the carbohydrate feedstock is sugar cane or sugar beet. 19. The method of claim 11, wherein the carbohydrate feedstock is derived from cellulose. 20. The method of claim 11, wherein development of antibiotic-resistant strains of bacteria is inhibited.
1,600
516
14,397,060
1,644
Compositions and methods for inhibiting, treating, and/or preventing a B-cell neoplasm are provided.
1. A method of identifying a chemotherapeutic agent, said method comprising performing a binding assay between CD19 and phosphatidylinositide 3-kinase (PI3K) in the presence of at least one test compound, wherein a decrease in the amount of CD19-PI3K binding in the presence of the test compound compared to the binding in the absence of the test compound indicates that the compound is a chemotherapeutic agent. 2. The method of claim 1, wherein said binding assay is a cell-based screening assay. 3. The method of claim 1, wherein said CD19 is the cytoplasmic tail of CD19. 4. The method of claim 1, wherein said test compound is a small molecule, peptide, or antibody. 5. The method of claim 1, wherein said compound is a chemotherapeutic agent for the treatment of a B-cell neoplasm. 6. A method of inhibiting B-cell neoplasm in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one inhibitor of the CD19-PI3K interaction. 7. The method of claim 6, wherein said B-cell neoplasm is a lymphoma or B-cell acute lymphoblastic leukemia. 8. The method of claim 6, further comprising the administration of at least one other chemotherapeutic agent or radiation therapy to the subject. 9. The method of claim 6, wherein said inhibitor is a peptide mimic of the CD19 cytoplasmic tail or an antibody immunologically specific for the CD19 cytoplasmic tail.
Compositions and methods for inhibiting, treating, and/or preventing a B-cell neoplasm are provided.1. A method of identifying a chemotherapeutic agent, said method comprising performing a binding assay between CD19 and phosphatidylinositide 3-kinase (PI3K) in the presence of at least one test compound, wherein a decrease in the amount of CD19-PI3K binding in the presence of the test compound compared to the binding in the absence of the test compound indicates that the compound is a chemotherapeutic agent. 2. The method of claim 1, wherein said binding assay is a cell-based screening assay. 3. The method of claim 1, wherein said CD19 is the cytoplasmic tail of CD19. 4. The method of claim 1, wherein said test compound is a small molecule, peptide, or antibody. 5. The method of claim 1, wherein said compound is a chemotherapeutic agent for the treatment of a B-cell neoplasm. 6. A method of inhibiting B-cell neoplasm in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one inhibitor of the CD19-PI3K interaction. 7. The method of claim 6, wherein said B-cell neoplasm is a lymphoma or B-cell acute lymphoblastic leukemia. 8. The method of claim 6, further comprising the administration of at least one other chemotherapeutic agent or radiation therapy to the subject. 9. The method of claim 6, wherein said inhibitor is a peptide mimic of the CD19 cytoplasmic tail or an antibody immunologically specific for the CD19 cytoplasmic tail.
1,600
517
15,501,989
1,615
A starting (raw) material, for cosmetics, comprising a core corona type microgel dispersion obtained by radical polymerization of a specific polyethylene oxide macromonomer, a specific hydrophobic monomer, and a specific cross-linking monomer under specific conditions. The purpose of the present invention is to provide the raw material for cosmetics, with which the cosmetic having a high stability provides a good feeling in use and does not cause skin irritation, can be easily produced.
1.-8. (canceled) 9. A raw material, for cosmetics, comprising: a core-corona type microgel dispersion; wherein said core-corona type microgel dispersion is obtained by a radical polymerization of: (A) a polyethylene oxide macromonomer having chemical formula (1), Wherein R1 is an alkyl group having 1 to 3 carbon atoms, n is a number of 8 to 200 and X is H or CH3; (B) a hydrophobic monomer having chemical formula (2), Wherein R2 is an alkyl group having 1 to 3 carbon atoms, and R3 is an alkyl group having 1 to 12 carbon atoms; and (C) said hydrophobic monomer is at least one methacrylic acid selected from the group consisting of methacrylic acid derivatives that have alkyl groups having 1 to 8 carbon atoms; and (C) a cross-linking monomer having chemical formula (3), Wherein R4 and R5 are independent alkyl group having 1 to 3 carbon atoms, and m is a number of 0 to 2; and said radical polymerization further comprising a step of mixing; said (A) polyethylene oxide is in a mole ratio range of 1 to 25 of said (B) hydrophobic monomers, and said (C) cross-linking monomers that is in a range of 0.1 to 1.5% by mass of said hydrophobic monomers in a polymerization solvent of water and polyol that is at least one polyol selected from the group consisting of dipropylene glycol, 1,3-butylene glycol, and isoprene glycol, and said water is in a range of 90% to 10% by mass of said polyol; and a polymerization temperature is approximately 20° C. 10. The raw material for cosmetics according to claim 9, wherein: said raw material for cosmetics does not contain ethanol. 11. An emulsifier comprising said raw material for cosmetics according to claim 1. 12. A clouding agent comprising said raw material for cosmetics according to claim 1. 13. An oil-in-water emulsion cosmetic, comprising: (a) said emulsifier according to claim 11; (b) oil phase components; and (c) aqueous phase components; wherein (b) said oil phase components that are emulsified with (a) said emulsifier further comprising: at least one oil phase component selected from the group consisting of hydrocarbon oils, higher fatty acids, higher alcohols, synthetic ester oils, silicone oils, liquid oils and fats, solid oils and fats, waxes, and perfumes. 14. The oil-in-water emulsion cosmetic, according to claim 13, wherein: said oil-in-water emulsion cosmetic does not contain ethanol. 15. A white cloudy cosmetic comprising said clouding agent according to claim 12. 16. The white cloudy cosmetic, according to claim 15, wherein: wherein said white cloudy cosmetic does not contain ethanol.
A starting (raw) material, for cosmetics, comprising a core corona type microgel dispersion obtained by radical polymerization of a specific polyethylene oxide macromonomer, a specific hydrophobic monomer, and a specific cross-linking monomer under specific conditions. The purpose of the present invention is to provide the raw material for cosmetics, with which the cosmetic having a high stability provides a good feeling in use and does not cause skin irritation, can be easily produced.1.-8. (canceled) 9. A raw material, for cosmetics, comprising: a core-corona type microgel dispersion; wherein said core-corona type microgel dispersion is obtained by a radical polymerization of: (A) a polyethylene oxide macromonomer having chemical formula (1), Wherein R1 is an alkyl group having 1 to 3 carbon atoms, n is a number of 8 to 200 and X is H or CH3; (B) a hydrophobic monomer having chemical formula (2), Wherein R2 is an alkyl group having 1 to 3 carbon atoms, and R3 is an alkyl group having 1 to 12 carbon atoms; and (C) said hydrophobic monomer is at least one methacrylic acid selected from the group consisting of methacrylic acid derivatives that have alkyl groups having 1 to 8 carbon atoms; and (C) a cross-linking monomer having chemical formula (3), Wherein R4 and R5 are independent alkyl group having 1 to 3 carbon atoms, and m is a number of 0 to 2; and said radical polymerization further comprising a step of mixing; said (A) polyethylene oxide is in a mole ratio range of 1 to 25 of said (B) hydrophobic monomers, and said (C) cross-linking monomers that is in a range of 0.1 to 1.5% by mass of said hydrophobic monomers in a polymerization solvent of water and polyol that is at least one polyol selected from the group consisting of dipropylene glycol, 1,3-butylene glycol, and isoprene glycol, and said water is in a range of 90% to 10% by mass of said polyol; and a polymerization temperature is approximately 20° C. 10. The raw material for cosmetics according to claim 9, wherein: said raw material for cosmetics does not contain ethanol. 11. An emulsifier comprising said raw material for cosmetics according to claim 1. 12. A clouding agent comprising said raw material for cosmetics according to claim 1. 13. An oil-in-water emulsion cosmetic, comprising: (a) said emulsifier according to claim 11; (b) oil phase components; and (c) aqueous phase components; wherein (b) said oil phase components that are emulsified with (a) said emulsifier further comprising: at least one oil phase component selected from the group consisting of hydrocarbon oils, higher fatty acids, higher alcohols, synthetic ester oils, silicone oils, liquid oils and fats, solid oils and fats, waxes, and perfumes. 14. The oil-in-water emulsion cosmetic, according to claim 13, wherein: said oil-in-water emulsion cosmetic does not contain ethanol. 15. A white cloudy cosmetic comprising said clouding agent according to claim 12. 16. The white cloudy cosmetic, according to claim 15, wherein: wherein said white cloudy cosmetic does not contain ethanol.
1,600
518
14,409,419
1,631
Systems, methods and non-transitory computer readable media identify favored polymer conformations. One or more residues are identified and may be replaced in the polymer, or the original primary sequence of the polymer may be retained. The conformations of residues in a subset of residues in a region of the identified one or more residues are altered. This conformational adjustment is repeated for other subsets of residues in the region of the identified one or more residues, and for other conformations, thereby deriving a plurality of polymer structures. A set of clusters is generated for each residue of the polymer using the conformationally adjusted structures, thereby creating sets of clusters. Structures in the plurality of structures are grouped into subgroups when the structures fall into the same clusters across a threshold number of the sets of clusters. One or more physical properties are determined for structures in subgroups, thereby identifying one or more thermodynamically relevant polymer conformations for the polymer.
1. A method of identifying one or more thermodynamically relevant polymer conformations for a polymer comprising a plurality of atoms, wherein the polymer comprises at least one contiguous segment of main chain, the method comprising: at a computer system having one or more processors and memory storing one or more programs to be executed by the one of more processors: (A) obtaining an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, wherein each respective xi in {x1, . . . , xN} is a three dimensional coordinate for an atom in the plurality of atoms; (B) identifying, in silico, a residue of the polymer and optionally replacing the residue with a different residue; (C) identifying a region of the polymer comprising a plurality of residues; (D) altering a conformation, with respect to the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, for each respective subset of residues in the region of the polymer in a plurality of subsets of residues in the region of the polymer, thereby deriving a plurality of structures of the region of the polymer, wherein each respective subset of residues in the plurality of subsets of residues in the region of the polymer is selected from among all the residues in the region of the polymer using a deterministic, randomized or pseudo-randomized algorithm; (E) generating a plurality of sets of clusters, wherein each set of clusters in the plurality of sets of clusters is for (i) a side chain or a main chain of a different residue or (ii) a contiguous segment of the main chain in the at least one contiguous segment of main chain in the region of the polymer, a first structure in the plurality of structures is placed in a first cluster in a first set of clusters in the plurality of sets of clusters and a second structure in the plurality of structures is placed in a second cluster in the first set of clusters when a structural characteristic associated with the coordinates of the side chain or the main chain of the residue or the coordinates of the contiguous segment of the main chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain or the main chain of the residue in the second structure or the coordinates of the contiguous segment of the main chain by a threshold amount; (F) grouping respective structures in the plurality of structures into a plurality of subgroups, wherein each structure in a subgroup in the plurality of subgroups falls into the same cluster in a threshold number of the sets of clusters in the plurality of sets of clusters; and (G) determining a property of a plurality of structures in a subgroup in the plurality of subgroups, thereby identifying the one or more thermodynamically relevant polymer conformations for the polymer. 2. The method of claim 1, wherein the coordinates of the residue identified in (B) are altered without changing the residue type. 3. The method of claim 1, wherein a plurality of residues of the polymer are replaced in the identifying (B) with different residues. 4. The method of claim 1, wherein two residues of the polymer are replaced in the identifying (B) with different residues. 5. The method of claim 1, wherein the region of the polymer consists of the atoms in the plurality of atoms that are within a distance threshold of an atom of the residue identified in (B). 6. The method of any one of claims 1 through 4, wherein the region of the polymer consists of the atoms in the plurality of atoms that are within a distance threshold of a point proximate to the residue identified in (B). 7. The method of claim 6 wherein the distance threshold is 15 Angstroms. 8. The method of claim 1 wherein the conformation of each residue in a respective subset of residues in the plurality of subsets of residues is chosen in the altering (D) by a deterministic, randomized or pseudo-randomized algorithm. 9. The method of claim 1 wherein the identifying (B) comprises identifying a contiguous set of residues and wherein a coordinate of one or more main chain atoms in each residue in the contiguous set of residues is altered. 10. The method of claim 1 wherein the identifying (B) comprises identifying a contiguous set of residues and wherein a coordinate of one or more main chain atoms in each residue in the contiguous set of residues is altered. 11. The method of claim 1 wherein the altering of the conformation, for each respective subset of residues in the region of the polymer in the plurality of subsets of residues in the region of the polymer comprises: (i) setting a value t and defining an initial structure to be the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer; (ii) altering a conformation of each residue in a subset of residues in the polymer from that found in the initial structure thereby deriving a structure in the plurality of structures; (iii) accepting the structure derived in (ii) as the initial structure when a score of the structure derived in (ii) is less than a score of the initial structure; (iv) accepting, with a probability P(ΔE), the structure derived in (ii) as the initial structure when the score of the structure derived in (ii) is greater than the score of the initial structure, wherein P(ΔE) is a probability function that is dependent upon (1) a difference in score between the initial structure and the structure derived in (ii) and (2) the value t; (v) decreasing the value t by an amount; and (vi) executing (ii) through (v) until a first occurrence of an exit condition, thereby deriving the plurality of structures of the region of the polymer 12. The method of claim 11, wherein P(ΔE)=exp−[(ΔE)/(k*T)], wherein ΔE is a difference in score between the initial structure and the structure derived in (ii) and k is a constant. 13. The method of any one of claims 1-8, wherein the plurality of structures of the region of the polymer derived by the altering (C) are found using a genetic algorithm. 14. The method of any one of claims 1-13, wherein the polymer is a protein and the altering (D) alters a side chain of a residue in a subset of residues in the region of the polymer to a rotamer for the side chain found in a rotamer library. 15. The method of claim 14, wherein the rotamer for the side chain found in the rotamer library is selected from the rotamer library on a deterministic, random or pseudo-random basis. 16. The method of any one of claims 1-13, wherein the altering (D) alters a main chain of a residue in a subset of residues in the region of the polymer to a conformation found in a structural library. 17. The method of claim 16, wherein the conformation found in the structural library is selected from the structural library on a deterministic, random or pseudo-random basis. 18. The method of any one of claims 1-13, wherein the polymer is a protein and the altering (D) alters a side chain of a residue in a subset of residues in the region of the polymer to a conformation selected from a continuum of conformations for the side chain. 19. The method of claim 18, wherein the conformation is selected from the continuum of conformations for the side chain on a deterministic, random or pseudo-random basis. 20. The method of any one of claims 1-13, wherein the altering (D) alters a main chain of a residue in a subset of residues in the region of the polymer to a conformation selected from a continuum of conformations for the main chain. 21. The method of claim 20, wherein the conformation is selected from the continuum of conformations for the main chain on a deterministic, random or pseudo-random basis. 22. The method of any one of claims 1-21, wherein the generating (E) generates a set of clusters in the plurality of sets of clusters for a residue in the polymer using a clustering algorithm. 23. The method of claim 22, wherein the clustering algorithm comprises hierarchical clustering. 24. The method of claim 23, wherein the clustering algorithm comprises maximal linkage agglomerative clustering. 25. The method of claim 23, wherein the clustering algorithm comprises agglomerative clustering using (i) a nearest neighbor algorithm, (ii) a farthest-neighbor algorithm, (iii) an average linkage algorithm, (iv) a centroid algorithm, or (v) a sum-of-squares algorithm. 26. The method of claim 22, wherein the clustering algorithm comprises k-means clustering, fuzzy k-means clustering, Jarvis-Patrick or steepest-descent clustering. 27. The method of any one of claims 1-26, wherein the threshold number of the sets of clusters is all the sets of clusters in the plurality of sets of clusters. 28. The method of any one of claims 1-26, wherein the threshold number of the sets of clusters is all but one of the sets of clusters in the plurality of sets of clusters. 29. The method of any one of claims 1-26, wherein the threshold number of the sets of clusters is ninety percent of the sets of clusters in the plurality of sets of clusters. 30. The method of any one of claims 1-26, wherein sets of clusters in the plurality of clusters are selected on the basis of a physical property of the polymer. 31. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a mean distance between a first point in the plurality of structures and a second point in the plurality of structures. 32. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a mean distance between a first point in the plurality of structures and a second point in a molecule that binds to the polymer or is bound by the polymer. 33. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a mean energy of the plurality of structures. 34. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a measure of conformational variability within the subgroup 35. The method of claim 34 wherein the measure of conformational variability within the subgroup is configurational entropy. 36. The method of any one of claims 1-35, the method further comprising: (H) displaying a representation of the structures in each subgroup in the plurality of subgroups; (I) receiving a selection of a subgroup in the plurality of subgroups; (J) using a representation of the structures of the subgroup selected in (I) as an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer; and (K) repeating (D) through (G). 37. The method of any one of claims 1-35, the method further comprising: (H) displaying a representation of the structures in each subgroup in the plurality of subgroups; (I) receiving a selection of a subgroup in the plurality of subgroups; (J) using a representation of the structures of the subgroup selected in (I) as an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer; and (K) repeating (B) through (G). 38. The method of any one of claims 1-37, wherein the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer are obtained by x-ray crystallography, nuclear magnetic resonance, electron microscopy, or computer modeling. 39. The method of any one of claims 1-38 wherein the first set of clusters in the plurality of clusters is for a side chain of a residue in the region of the polymer, and the structural characteristic associated with the coordinates of the side chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain of the residue in the second structure by the threshold amount when the root-mean-square distance between corresponding non-hydrogen atoms in the side chain of the residue in the first structure and the side chain of the residue in the second structure deviate by more than 0.7 Angstroms. 40. The method of any one of claims 1-39, wherein the region of the polymer comprises ten residues. 41. The method of any one of claims 1-39, wherein the region of the polymer comprises one hundred residues. 42. A computer system for identifying a thermodynamically relevant conformation for a polymer comprising a plurality of atoms, the computer system comprising at least one processor and memory storing at least one program for execution by the at least one processor, the memory further comprising instructions for performing any of the methods of claims 1 through 41. 43. A computer system for identifying a thermodynamically relevant conformation for a polymer comprising a plurality of atoms, the computer system comprising at least one processor and memory storing at least one program for execution by the at least one processor, the memory further comprising instructions for performing any of the methods of claims 1 through 41. 44. A computer system for identifying one or more thermodynamically relevant polymer conformations for a polymer comprising a plurality of atoms, wherein the polymer comprises at least one contiguous segment of main chain, the computer system comprising at least one processor and memory storing at least one program for execution by the at least one processor, the memory further comprising instructions for: (A) obtaining an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, wherein each respective xi in {x1, . . . , xN} is a three dimensional coordinate for an atom in the plurality of atoms; (B) identifying, in silico, a residue of the polymer and optionally replacing the residue with a different residue; (C) identifying a region of the polymer comprising a plurality of residues; (D) altering a conformation, with respect to the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, for each respective subset of residues in the region of the polymer in a plurality of subsets of residues in the region of the polymer, thereby deriving a plurality of structures of the region of the polymer, wherein each respective subset of residues in the plurality of subsets of residues in the region of the polymer is selected from among all the residues in the region of the polymer using a deterministic, randomized or pseudo-randomized algorithm; (E) generating a plurality of sets of clusters, wherein each set of clusters in the plurality of sets of clusters is for (i) a side chain or a main chain of a different residue or (ii) a contiguous segment of the main chain in the at least one contiguous segment of main chain in the region of the polymer, a first structure in the plurality of structures is placed in a first cluster in a first set of clusters in the plurality of sets of clusters and a second structure in the plurality of structures is placed in a second cluster in the first set of clusters when a structural characteristic associated with the coordinates of the side chain or the main chain of the residue or the coordinates of the contiguous segment of the main chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain or the main chain of the residue in the second structure or the coordinates of the contiguous segment of the main chain by a threshold amount; (F) grouping respective structures in the plurality of structures into a plurality of subgroups, wherein each structure in a subgroup in the plurality of subgroups falls into the same cluster in a threshold number of the sets of clusters in the plurality of sets of clusters; and (G) determining a property of a plurality of structures in a subgroup in the plurality of subgroups, thereby identifying the one or more thermodynamically relevant polymer conformations for the polymer. 45. A non-transitory computer readable storage medium storing a computational module for identifying one or more thermodynamically relevant polymer conformations for a polymer comprising a plurality of atoms, wherein the polymer comprises at least one contiguous segment of main chain, the computational module comprising instructions for: (A) obtaining an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, wherein each respective xi in {x1, . . . , xN} is a three dimensional coordinate for an atom in the plurality of atoms; (B) identifying, in silico, a residue of the polymer and optionally replacing the residue with a different residue; (C) identifying a region of the polymer comprising a plurality of residues; (D) altering a conformation, with respect to the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, for each respective subset of residues in the region of the polymer in a plurality of subsets of residues in the region of the polymer, thereby deriving a plurality of structures of the region of the polymer, wherein each respective subset of residues in the plurality of subsets of residues in the region of the polymer is selected from among all the residues in the region of the polymer using a deterministic, randomized or pseudo-randomized algorithm; (E) generating a plurality of sets of clusters, wherein each set of clusters in the plurality of sets of clusters is for (i) a side chain or a main chain of a different residue or (ii) a contiguous segment of the main chain in the at least one contiguous segment of main chain in the region of the polymer, a first structure in the plurality of structures is placed in a first cluster in a first set of clusters in the plurality of sets of clusters and a second structure in the plurality of structures is placed in a second cluster in the first set of clusters when a structural characteristic associated with the coordinates of the side chain or the main chain of the residue or the coordinates of the contiguous segment of the main chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain or the main chain of the residue in the second structure or the coordinates of the contiguous segment of the main chain by a threshold amount; (F) grouping respective structures in the plurality of structures into a plurality of subgroups, wherein each structure in a subgroup in the plurality of subgroups falls into the same cluster in a threshold number of the sets of clusters in the plurality of sets of clusters; and (G) determining a property of a plurality of structures in a subgroup in the plurality of subgroups, thereby identifying the one or more thermodynamically relevant polymer conformations for the polymer.
Systems, methods and non-transitory computer readable media identify favored polymer conformations. One or more residues are identified and may be replaced in the polymer, or the original primary sequence of the polymer may be retained. The conformations of residues in a subset of residues in a region of the identified one or more residues are altered. This conformational adjustment is repeated for other subsets of residues in the region of the identified one or more residues, and for other conformations, thereby deriving a plurality of polymer structures. A set of clusters is generated for each residue of the polymer using the conformationally adjusted structures, thereby creating sets of clusters. Structures in the plurality of structures are grouped into subgroups when the structures fall into the same clusters across a threshold number of the sets of clusters. One or more physical properties are determined for structures in subgroups, thereby identifying one or more thermodynamically relevant polymer conformations for the polymer.1. A method of identifying one or more thermodynamically relevant polymer conformations for a polymer comprising a plurality of atoms, wherein the polymer comprises at least one contiguous segment of main chain, the method comprising: at a computer system having one or more processors and memory storing one or more programs to be executed by the one of more processors: (A) obtaining an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, wherein each respective xi in {x1, . . . , xN} is a three dimensional coordinate for an atom in the plurality of atoms; (B) identifying, in silico, a residue of the polymer and optionally replacing the residue with a different residue; (C) identifying a region of the polymer comprising a plurality of residues; (D) altering a conformation, with respect to the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, for each respective subset of residues in the region of the polymer in a plurality of subsets of residues in the region of the polymer, thereby deriving a plurality of structures of the region of the polymer, wherein each respective subset of residues in the plurality of subsets of residues in the region of the polymer is selected from among all the residues in the region of the polymer using a deterministic, randomized or pseudo-randomized algorithm; (E) generating a plurality of sets of clusters, wherein each set of clusters in the plurality of sets of clusters is for (i) a side chain or a main chain of a different residue or (ii) a contiguous segment of the main chain in the at least one contiguous segment of main chain in the region of the polymer, a first structure in the plurality of structures is placed in a first cluster in a first set of clusters in the plurality of sets of clusters and a second structure in the plurality of structures is placed in a second cluster in the first set of clusters when a structural characteristic associated with the coordinates of the side chain or the main chain of the residue or the coordinates of the contiguous segment of the main chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain or the main chain of the residue in the second structure or the coordinates of the contiguous segment of the main chain by a threshold amount; (F) grouping respective structures in the plurality of structures into a plurality of subgroups, wherein each structure in a subgroup in the plurality of subgroups falls into the same cluster in a threshold number of the sets of clusters in the plurality of sets of clusters; and (G) determining a property of a plurality of structures in a subgroup in the plurality of subgroups, thereby identifying the one or more thermodynamically relevant polymer conformations for the polymer. 2. The method of claim 1, wherein the coordinates of the residue identified in (B) are altered without changing the residue type. 3. The method of claim 1, wherein a plurality of residues of the polymer are replaced in the identifying (B) with different residues. 4. The method of claim 1, wherein two residues of the polymer are replaced in the identifying (B) with different residues. 5. The method of claim 1, wherein the region of the polymer consists of the atoms in the plurality of atoms that are within a distance threshold of an atom of the residue identified in (B). 6. The method of any one of claims 1 through 4, wherein the region of the polymer consists of the atoms in the plurality of atoms that are within a distance threshold of a point proximate to the residue identified in (B). 7. The method of claim 6 wherein the distance threshold is 15 Angstroms. 8. The method of claim 1 wherein the conformation of each residue in a respective subset of residues in the plurality of subsets of residues is chosen in the altering (D) by a deterministic, randomized or pseudo-randomized algorithm. 9. The method of claim 1 wherein the identifying (B) comprises identifying a contiguous set of residues and wherein a coordinate of one or more main chain atoms in each residue in the contiguous set of residues is altered. 10. The method of claim 1 wherein the identifying (B) comprises identifying a contiguous set of residues and wherein a coordinate of one or more main chain atoms in each residue in the contiguous set of residues is altered. 11. The method of claim 1 wherein the altering of the conformation, for each respective subset of residues in the region of the polymer in the plurality of subsets of residues in the region of the polymer comprises: (i) setting a value t and defining an initial structure to be the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer; (ii) altering a conformation of each residue in a subset of residues in the polymer from that found in the initial structure thereby deriving a structure in the plurality of structures; (iii) accepting the structure derived in (ii) as the initial structure when a score of the structure derived in (ii) is less than a score of the initial structure; (iv) accepting, with a probability P(ΔE), the structure derived in (ii) as the initial structure when the score of the structure derived in (ii) is greater than the score of the initial structure, wherein P(ΔE) is a probability function that is dependent upon (1) a difference in score between the initial structure and the structure derived in (ii) and (2) the value t; (v) decreasing the value t by an amount; and (vi) executing (ii) through (v) until a first occurrence of an exit condition, thereby deriving the plurality of structures of the region of the polymer 12. The method of claim 11, wherein P(ΔE)=exp−[(ΔE)/(k*T)], wherein ΔE is a difference in score between the initial structure and the structure derived in (ii) and k is a constant. 13. The method of any one of claims 1-8, wherein the plurality of structures of the region of the polymer derived by the altering (C) are found using a genetic algorithm. 14. The method of any one of claims 1-13, wherein the polymer is a protein and the altering (D) alters a side chain of a residue in a subset of residues in the region of the polymer to a rotamer for the side chain found in a rotamer library. 15. The method of claim 14, wherein the rotamer for the side chain found in the rotamer library is selected from the rotamer library on a deterministic, random or pseudo-random basis. 16. The method of any one of claims 1-13, wherein the altering (D) alters a main chain of a residue in a subset of residues in the region of the polymer to a conformation found in a structural library. 17. The method of claim 16, wherein the conformation found in the structural library is selected from the structural library on a deterministic, random or pseudo-random basis. 18. The method of any one of claims 1-13, wherein the polymer is a protein and the altering (D) alters a side chain of a residue in a subset of residues in the region of the polymer to a conformation selected from a continuum of conformations for the side chain. 19. The method of claim 18, wherein the conformation is selected from the continuum of conformations for the side chain on a deterministic, random or pseudo-random basis. 20. The method of any one of claims 1-13, wherein the altering (D) alters a main chain of a residue in a subset of residues in the region of the polymer to a conformation selected from a continuum of conformations for the main chain. 21. The method of claim 20, wherein the conformation is selected from the continuum of conformations for the main chain on a deterministic, random or pseudo-random basis. 22. The method of any one of claims 1-21, wherein the generating (E) generates a set of clusters in the plurality of sets of clusters for a residue in the polymer using a clustering algorithm. 23. The method of claim 22, wherein the clustering algorithm comprises hierarchical clustering. 24. The method of claim 23, wherein the clustering algorithm comprises maximal linkage agglomerative clustering. 25. The method of claim 23, wherein the clustering algorithm comprises agglomerative clustering using (i) a nearest neighbor algorithm, (ii) a farthest-neighbor algorithm, (iii) an average linkage algorithm, (iv) a centroid algorithm, or (v) a sum-of-squares algorithm. 26. The method of claim 22, wherein the clustering algorithm comprises k-means clustering, fuzzy k-means clustering, Jarvis-Patrick or steepest-descent clustering. 27. The method of any one of claims 1-26, wherein the threshold number of the sets of clusters is all the sets of clusters in the plurality of sets of clusters. 28. The method of any one of claims 1-26, wherein the threshold number of the sets of clusters is all but one of the sets of clusters in the plurality of sets of clusters. 29. The method of any one of claims 1-26, wherein the threshold number of the sets of clusters is ninety percent of the sets of clusters in the plurality of sets of clusters. 30. The method of any one of claims 1-26, wherein sets of clusters in the plurality of clusters are selected on the basis of a physical property of the polymer. 31. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a mean distance between a first point in the plurality of structures and a second point in the plurality of structures. 32. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a mean distance between a first point in the plurality of structures and a second point in a molecule that binds to the polymer or is bound by the polymer. 33. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a mean energy of the plurality of structures. 34. The method of any one of claims 1-29, wherein the property of the plurality of structures in the subgroup in the plurality of subgroups is a measure of conformational variability within the subgroup 35. The method of claim 34 wherein the measure of conformational variability within the subgroup is configurational entropy. 36. The method of any one of claims 1-35, the method further comprising: (H) displaying a representation of the structures in each subgroup in the plurality of subgroups; (I) receiving a selection of a subgroup in the plurality of subgroups; (J) using a representation of the structures of the subgroup selected in (I) as an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer; and (K) repeating (D) through (G). 37. The method of any one of claims 1-35, the method further comprising: (H) displaying a representation of the structures in each subgroup in the plurality of subgroups; (I) receiving a selection of a subgroup in the plurality of subgroups; (J) using a representation of the structures of the subgroup selected in (I) as an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer; and (K) repeating (B) through (G). 38. The method of any one of claims 1-37, wherein the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer are obtained by x-ray crystallography, nuclear magnetic resonance, electron microscopy, or computer modeling. 39. The method of any one of claims 1-38 wherein the first set of clusters in the plurality of clusters is for a side chain of a residue in the region of the polymer, and the structural characteristic associated with the coordinates of the side chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain of the residue in the second structure by the threshold amount when the root-mean-square distance between corresponding non-hydrogen atoms in the side chain of the residue in the first structure and the side chain of the residue in the second structure deviate by more than 0.7 Angstroms. 40. The method of any one of claims 1-39, wherein the region of the polymer comprises ten residues. 41. The method of any one of claims 1-39, wherein the region of the polymer comprises one hundred residues. 42. A computer system for identifying a thermodynamically relevant conformation for a polymer comprising a plurality of atoms, the computer system comprising at least one processor and memory storing at least one program for execution by the at least one processor, the memory further comprising instructions for performing any of the methods of claims 1 through 41. 43. A computer system for identifying a thermodynamically relevant conformation for a polymer comprising a plurality of atoms, the computer system comprising at least one processor and memory storing at least one program for execution by the at least one processor, the memory further comprising instructions for performing any of the methods of claims 1 through 41. 44. A computer system for identifying one or more thermodynamically relevant polymer conformations for a polymer comprising a plurality of atoms, wherein the polymer comprises at least one contiguous segment of main chain, the computer system comprising at least one processor and memory storing at least one program for execution by the at least one processor, the memory further comprising instructions for: (A) obtaining an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, wherein each respective xi in {x1, . . . , xN} is a three dimensional coordinate for an atom in the plurality of atoms; (B) identifying, in silico, a residue of the polymer and optionally replacing the residue with a different residue; (C) identifying a region of the polymer comprising a plurality of residues; (D) altering a conformation, with respect to the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, for each respective subset of residues in the region of the polymer in a plurality of subsets of residues in the region of the polymer, thereby deriving a plurality of structures of the region of the polymer, wherein each respective subset of residues in the plurality of subsets of residues in the region of the polymer is selected from among all the residues in the region of the polymer using a deterministic, randomized or pseudo-randomized algorithm; (E) generating a plurality of sets of clusters, wherein each set of clusters in the plurality of sets of clusters is for (i) a side chain or a main chain of a different residue or (ii) a contiguous segment of the main chain in the at least one contiguous segment of main chain in the region of the polymer, a first structure in the plurality of structures is placed in a first cluster in a first set of clusters in the plurality of sets of clusters and a second structure in the plurality of structures is placed in a second cluster in the first set of clusters when a structural characteristic associated with the coordinates of the side chain or the main chain of the residue or the coordinates of the contiguous segment of the main chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain or the main chain of the residue in the second structure or the coordinates of the contiguous segment of the main chain by a threshold amount; (F) grouping respective structures in the plurality of structures into a plurality of subgroups, wherein each structure in a subgroup in the plurality of subgroups falls into the same cluster in a threshold number of the sets of clusters in the plurality of sets of clusters; and (G) determining a property of a plurality of structures in a subgroup in the plurality of subgroups, thereby identifying the one or more thermodynamically relevant polymer conformations for the polymer. 45. A non-transitory computer readable storage medium storing a computational module for identifying one or more thermodynamically relevant polymer conformations for a polymer comprising a plurality of atoms, wherein the polymer comprises at least one contiguous segment of main chain, the computational module comprising instructions for: (A) obtaining an initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, wherein each respective xi in {x1, . . . , xN} is a three dimensional coordinate for an atom in the plurality of atoms; (B) identifying, in silico, a residue of the polymer and optionally replacing the residue with a different residue; (C) identifying a region of the polymer comprising a plurality of residues; (D) altering a conformation, with respect to the initial set of three-dimensional coordinates {x1, . . . , xN} for the polymer, for each respective subset of residues in the region of the polymer in a plurality of subsets of residues in the region of the polymer, thereby deriving a plurality of structures of the region of the polymer, wherein each respective subset of residues in the plurality of subsets of residues in the region of the polymer is selected from among all the residues in the region of the polymer using a deterministic, randomized or pseudo-randomized algorithm; (E) generating a plurality of sets of clusters, wherein each set of clusters in the plurality of sets of clusters is for (i) a side chain or a main chain of a different residue or (ii) a contiguous segment of the main chain in the at least one contiguous segment of main chain in the region of the polymer, a first structure in the plurality of structures is placed in a first cluster in a first set of clusters in the plurality of sets of clusters and a second structure in the plurality of structures is placed in a second cluster in the first set of clusters when a structural characteristic associated with the coordinates of the side chain or the main chain of the residue or the coordinates of the contiguous segment of the main chain, represented by the first set of clusters, in the first structure deviates from a structural characteristic associated with the coordinates of the side chain or the main chain of the residue in the second structure or the coordinates of the contiguous segment of the main chain by a threshold amount; (F) grouping respective structures in the plurality of structures into a plurality of subgroups, wherein each structure in a subgroup in the plurality of subgroups falls into the same cluster in a threshold number of the sets of clusters in the plurality of sets of clusters; and (G) determining a property of a plurality of structures in a subgroup in the plurality of subgroups, thereby identifying the one or more thermodynamically relevant polymer conformations for the polymer.
1,600
519
15,128,479
1,641
A chemiluminescent detection system, as well as kits and microfluidics devices containing same, are disclosed. Methods of using the system, kits, and devices are also disclosed. The first, second, and third antibodies or binding fragments thereof may be provided in any form that allows these antibodies/binding fragments to function in accordance with the presently disclosed and claimed inventive concept(s). For example, each of the first, second, and third antibodies/binding fragments may be a polyclonal antibody/binding fragment or a monoclonal antibody/binding fragment.
1. A kit containing a chemiluminescent detection system for a specific analyte, the kit comprising: (a) a first composition comprising a singlet-oxygen activatable chemiluminescent compound and a first antibody or binding fragment thereof associated therewith, wherein the first antibody or binding fragment thereof is a detection antibody that specifically binds to a first epitope of the analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the first antibody or binding fragment thereof; (b) a second composition comprising a singlet-oxygen activatable chemiluminescent compound and a second antibody or binding fragment thereof associated therewith, wherein the second antibody or binding fragment thereof is a detection antibody that specifically binds to a second epitope of the analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the second antibody or binding fragment thereof, and wherein the first and second epitopes at least partially overlap such that the first and second antibodies or binding fragments thereof cannot both bind to a single analyte molecule; and (c) a third composition comprising a third antibody or binding fragment thereof, the third antibody or binding fragment thereof being a capture antibody that specifically binds to a third epitope of the analyte that does not overlap with the first and second epitopes, whereby a single analyte molecule can bind the third antibody or binding fragment thereof and one of the first and second antibodies or binding fragments thereof, and wherein the third antibody or binding fragment thereof is capable of association with a sensitizer capable of generating singlet oxygen in its excited state, whereby association of the third antibody or binding fragment thereof with the sensitizer allows for the indirect binding of the sensitizer to the analyte. 2. The kit of claim 1, further comprising the sensitizer and wherein third composition further comprises the sensitizer associated with the third antibody or binding fragment thereof. 3. The kit of claim 1, wherein at least one of the first and second compositions further comprises at least one fluorescent molecule that is excited by the activated chemiluminescent compound. 4. The kit of any of claim 1, wherein at least one of (a)-(c) is further defined as being in the form of a lyophilized reagent. 5. The kit of claim 4, further comprising an excipient for the reconstitution of the lyophilized reagent. 6. A microfluidics device, comprising: at least one compartment containing: (i) a first composition comprising a singlet-oxygen activatable chemiluminescent compound and a first antibody or binding fragment thereof associated therewith, wherein the first antibody or binding fragment thereof is a detection antibody that specifically binds to a first epitope of a specific analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the first antibody or binding fragment thereof; (ii) a second composition comprising a singlet-oxygen activatable chemiluminescent compound and a second antibody or binding fragment thereof associated therewith, wherein the second antibody or binding fragment thereof is a detection antibody that specifically binds to a second epitope of the analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the second antibody or binding fragment thereof, and wherein the first and second epitopes at least partially overlap such that the first and second antibodies or binding fragments thereof cannot both bind to a single analyte molecule; and (iii) a third composition comprising a third antibody or binding fragment thereof, the third antibody or binding fragment thereof being a capture antibody that specifically binds to a third epitope of the analyte that does not overlap with the first and second epitopes, whereby a single analyte molecule can bind the third antibody or binding fragment thereof and one of the first and second antibodies or binding fragments thereof; and (iv) a sensitizer capable of association with the third antibody or binding fragment thereof, the sensitizer being capable of generating singlet oxygen in its excited state, and wherein association of the third antibody or binding fragment thereof with the sensitizer allows for the indirect binding of the sensitizer to the analyte. 7. The microfluidics device of claim 6, further comprising an inlet channel through which a sample may be disposed, wherein the at least one compartment is capable of being in fluidic communication with the inlet channel. 8. The microfluidics device of claim 7, further defined as comprising at least two compartments, wherein a first compartment contains (i), (ii), and (iii), and wherein a second compartment contains (iv). 9. The microfluidics device of claim 8, further comprising an inlet channel through which a sample may be disposed, wherein the first compartment is capable of being in fluidic communication with the inlet channel, and wherein the second compartment is capable of being in fluid communication with at least one of the inlet channel and the first compartment. 10. The microfluidics device of any of claims 6-9, further comprising at least one additional compartment capable of being in fluidic communication with at least one of the inlet channel and the at least one compartment, and wherein the at least one additional compartment contains a diluent. 11. A method for detecting the presence and/or concentration of a specific analyte in a sample, comprising the steps of: (a) combining, either simultaneously or wholly or partially sequentially: (i) a sample suspected of containing the specific analyte; (ii) a first composition comprising a singlet-oxygen activatable chemiluminescent compound and a first antibody or binding fragment thereof associated therewith, wherein the first antibody or binding fragment thereof is a detection antibody that specifically binds to a first epitope of a specific analyte; (iii) a second composition comprising a singlet-oxygen activatable chemiluminescent compound and a second antibody or binding fragment thereof associated therewith, wherein the second antibody or binding fragment thereof is a detection antibody that specifically binds to a second epitope of the analyte, and wherein the first and second epitopes at least partially overlap such that the first and second antibodies or binding fragments thereof cannot both bind to a single analyte molecule; (iv) a third composition comprising a third antibody or binding fragment thereof, the third antibody or binding fragment thereof being a capture antibody that specifically binds to a third epitope of the analyte that does not overlap with the first and second epitopes, whereby a single analyte molecule can bind the third antibody or binding fragment thereof and one of the first and second antibodies or binding fragments thereof; and (v) a sensitizer capable of association with the third antibody or binding fragment thereof, the sensitizer being capable of generating singlet oxygen in its excited state; (b) allowing the binding of (ii), (iii) and/or (iv) to analyte within the sample, wherein a first sandwich complex comprising an analyte molecule and (ii) and (iv) is formed, and a second sandwich complex comprising another analyte molecule and (iii) and (iv) is formed, and wherein (v) associates with (iv) in the first and second sandwich complexes, thus bringing the sensitizer into close proximity to the chemiluminescent compounds of (ii) and (iii); (c) activating the sensitizer to generate singlet oxygen, wherein activation of the sensitizer present in the first and second sandwich complexes causes the activation of the chemiluminescent compounds present in the first and second sandwich complexes; (d) determining the amount of chemiluminescence generated by the activated chemiluminescent compounds present in the first and second sandwich complexes; (e) optionally repeating steps (b)-(d); and (f) detecting the presence and/or concentration of the analyte by analyzing the amount of chemiluminescence so produced, wherein the amount of chemiluminescence is directly proportional to the amount of analyte in the sample. 12. The method of claim 11, wherein the sensitizer is a photosensitizer, and wherein step (c) is further defined as activating the photosensitizer via irradiation with light. 13. The method of claim 12, further comprising a step of exposing the sample to a separation step prior to combining with any of (ii)-(v). 14. The method of any of claim 11, wherein the singlet-oxygen activatable chemiluminescent compounds of the first and second compositions are the same. 15. The method of any of claim 11, wherein the singlet-oxygen activatable chemiluminescent compounds of the first and second compositions are different.
A chemiluminescent detection system, as well as kits and microfluidics devices containing same, are disclosed. Methods of using the system, kits, and devices are also disclosed. The first, second, and third antibodies or binding fragments thereof may be provided in any form that allows these antibodies/binding fragments to function in accordance with the presently disclosed and claimed inventive concept(s). For example, each of the first, second, and third antibodies/binding fragments may be a polyclonal antibody/binding fragment or a monoclonal antibody/binding fragment.1. A kit containing a chemiluminescent detection system for a specific analyte, the kit comprising: (a) a first composition comprising a singlet-oxygen activatable chemiluminescent compound and a first antibody or binding fragment thereof associated therewith, wherein the first antibody or binding fragment thereof is a detection antibody that specifically binds to a first epitope of the analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the first antibody or binding fragment thereof; (b) a second composition comprising a singlet-oxygen activatable chemiluminescent compound and a second antibody or binding fragment thereof associated therewith, wherein the second antibody or binding fragment thereof is a detection antibody that specifically binds to a second epitope of the analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the second antibody or binding fragment thereof, and wherein the first and second epitopes at least partially overlap such that the first and second antibodies or binding fragments thereof cannot both bind to a single analyte molecule; and (c) a third composition comprising a third antibody or binding fragment thereof, the third antibody or binding fragment thereof being a capture antibody that specifically binds to a third epitope of the analyte that does not overlap with the first and second epitopes, whereby a single analyte molecule can bind the third antibody or binding fragment thereof and one of the first and second antibodies or binding fragments thereof, and wherein the third antibody or binding fragment thereof is capable of association with a sensitizer capable of generating singlet oxygen in its excited state, whereby association of the third antibody or binding fragment thereof with the sensitizer allows for the indirect binding of the sensitizer to the analyte. 2. The kit of claim 1, further comprising the sensitizer and wherein third composition further comprises the sensitizer associated with the third antibody or binding fragment thereof. 3. The kit of claim 1, wherein at least one of the first and second compositions further comprises at least one fluorescent molecule that is excited by the activated chemiluminescent compound. 4. The kit of any of claim 1, wherein at least one of (a)-(c) is further defined as being in the form of a lyophilized reagent. 5. The kit of claim 4, further comprising an excipient for the reconstitution of the lyophilized reagent. 6. A microfluidics device, comprising: at least one compartment containing: (i) a first composition comprising a singlet-oxygen activatable chemiluminescent compound and a first antibody or binding fragment thereof associated therewith, wherein the first antibody or binding fragment thereof is a detection antibody that specifically binds to a first epitope of a specific analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the first antibody or binding fragment thereof; (ii) a second composition comprising a singlet-oxygen activatable chemiluminescent compound and a second antibody or binding fragment thereof associated therewith, wherein the second antibody or binding fragment thereof is a detection antibody that specifically binds to a second epitope of the analyte whereby the singlet-oxygen activatable chemiluminescent compound is capable of indirectly binding to the analyte via the second antibody or binding fragment thereof, and wherein the first and second epitopes at least partially overlap such that the first and second antibodies or binding fragments thereof cannot both bind to a single analyte molecule; and (iii) a third composition comprising a third antibody or binding fragment thereof, the third antibody or binding fragment thereof being a capture antibody that specifically binds to a third epitope of the analyte that does not overlap with the first and second epitopes, whereby a single analyte molecule can bind the third antibody or binding fragment thereof and one of the first and second antibodies or binding fragments thereof; and (iv) a sensitizer capable of association with the third antibody or binding fragment thereof, the sensitizer being capable of generating singlet oxygen in its excited state, and wherein association of the third antibody or binding fragment thereof with the sensitizer allows for the indirect binding of the sensitizer to the analyte. 7. The microfluidics device of claim 6, further comprising an inlet channel through which a sample may be disposed, wherein the at least one compartment is capable of being in fluidic communication with the inlet channel. 8. The microfluidics device of claim 7, further defined as comprising at least two compartments, wherein a first compartment contains (i), (ii), and (iii), and wherein a second compartment contains (iv). 9. The microfluidics device of claim 8, further comprising an inlet channel through which a sample may be disposed, wherein the first compartment is capable of being in fluidic communication with the inlet channel, and wherein the second compartment is capable of being in fluid communication with at least one of the inlet channel and the first compartment. 10. The microfluidics device of any of claims 6-9, further comprising at least one additional compartment capable of being in fluidic communication with at least one of the inlet channel and the at least one compartment, and wherein the at least one additional compartment contains a diluent. 11. A method for detecting the presence and/or concentration of a specific analyte in a sample, comprising the steps of: (a) combining, either simultaneously or wholly or partially sequentially: (i) a sample suspected of containing the specific analyte; (ii) a first composition comprising a singlet-oxygen activatable chemiluminescent compound and a first antibody or binding fragment thereof associated therewith, wherein the first antibody or binding fragment thereof is a detection antibody that specifically binds to a first epitope of a specific analyte; (iii) a second composition comprising a singlet-oxygen activatable chemiluminescent compound and a second antibody or binding fragment thereof associated therewith, wherein the second antibody or binding fragment thereof is a detection antibody that specifically binds to a second epitope of the analyte, and wherein the first and second epitopes at least partially overlap such that the first and second antibodies or binding fragments thereof cannot both bind to a single analyte molecule; (iv) a third composition comprising a third antibody or binding fragment thereof, the third antibody or binding fragment thereof being a capture antibody that specifically binds to a third epitope of the analyte that does not overlap with the first and second epitopes, whereby a single analyte molecule can bind the third antibody or binding fragment thereof and one of the first and second antibodies or binding fragments thereof; and (v) a sensitizer capable of association with the third antibody or binding fragment thereof, the sensitizer being capable of generating singlet oxygen in its excited state; (b) allowing the binding of (ii), (iii) and/or (iv) to analyte within the sample, wherein a first sandwich complex comprising an analyte molecule and (ii) and (iv) is formed, and a second sandwich complex comprising another analyte molecule and (iii) and (iv) is formed, and wherein (v) associates with (iv) in the first and second sandwich complexes, thus bringing the sensitizer into close proximity to the chemiluminescent compounds of (ii) and (iii); (c) activating the sensitizer to generate singlet oxygen, wherein activation of the sensitizer present in the first and second sandwich complexes causes the activation of the chemiluminescent compounds present in the first and second sandwich complexes; (d) determining the amount of chemiluminescence generated by the activated chemiluminescent compounds present in the first and second sandwich complexes; (e) optionally repeating steps (b)-(d); and (f) detecting the presence and/or concentration of the analyte by analyzing the amount of chemiluminescence so produced, wherein the amount of chemiluminescence is directly proportional to the amount of analyte in the sample. 12. The method of claim 11, wherein the sensitizer is a photosensitizer, and wherein step (c) is further defined as activating the photosensitizer via irradiation with light. 13. The method of claim 12, further comprising a step of exposing the sample to a separation step prior to combining with any of (ii)-(v). 14. The method of any of claim 11, wherein the singlet-oxygen activatable chemiluminescent compounds of the first and second compositions are the same. 15. The method of any of claim 11, wherein the singlet-oxygen activatable chemiluminescent compounds of the first and second compositions are different.
1,600
520
14,409,497
1,617
The invention provides high water content dentifrice compositions comprising a silica abrasive and a component containing microcrystalline cellulose and carboxymethylcellulose, as well as methods of making and using the same.
1. A dentifrice composition comprising by weight a. 30-50% of water; b. 0.5% to 1.5% of a component for enhancing flavor delivery and rheological profile comprising: b1. 80% to 90% of a microcrystalline cellulose; and b2. 10% to 20% of carboxymethylcellulose or a salt thereof; wherein the weight percentage of b1 and b2. is based on the total weight of component; and c. an effective amount of a silica abrasive, wherein the abrasive component comprises 15-25% of the composition. 2. The composition of claim 1 further comprising a synthetic anionic polymeric polycarboxylate. 3. The composition of claim 2 wherein the anionic polymer is a methyl vinyl ether/maleic anhydride (PVM/MA) copolymer having an average molecular weight (M.W.) of about 30,000 to about 1,000,000 and comprises 1-5% of the weight of the composition. 4. The composition according to claim 1 wherein the silica abrasive component comprises (a) a first population of silica abrasive particles, having a d50 of less than 5 microns, and (b) a second population of silica abrasive particles, having a d50 of greater than 8 microns, wherein the ratio of the first population to the second population by weight is between 1:2 and 1:4. 5. The composition according to claim 1 further comprising an effective amount of a fluoride ion source. 6. The composition according to claim 1 further comprising 25-35% of a humectant. 7. The composition according to claim 1 further comprising 1-2% sodium lauryl sulfate (SLS). 8. The composition according to claim further comprising a viscosity modifying amount of one or more of xanthan gum, carrageenan, silica thickener, or combinations thereof. 9. The composition according to claim 1 further comprising an antibacterially effective amount of a natural extract antibacterial, non-ionic bacterial agent, triclosan magnolol, tetrahydromagnolol, butyl magnolol, honokiol, tetrahydrohonokiol or mixtures thereof. 10. The composition according to claim 1 which is obtained or obtainable by premixing microcrystalline cellulose or a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose in water prior to combination with the other ingredients. 11. The composition according to claim 1 further comprising a) 25-35% glycerin as a humectant b) 1-3% PVM/MA copolymer c) 0.1-0.5% triclosan. 12. The composition according to claim 11 comprising: a) Glycerin 25-35% b) Water 30-50% c) PVM/MA copolymer 1-3% d) Sodium fluoride 0.1-0.5% e) Triclosan 0.1-1%   f) Sodium lauryl sulfate 1-2% g) Sodium carboxymethyl cellulose 0.5-2%   h) Carrageenan 0.1-1%   i) Silica abrasive 15-25% j) Mixture of 80-90%  0.7-1.2%. microcrystalline cellulose and 10-20% sodium carboxymethyl cellulose 13. A method of making a dentifrice composition according to claim 1 comprising dispersing a mixture of 80-90% microcrystalline cellulose and 10-20% sodium carboxymethyl cellulose in water, then admixing the remaining ingredients. 14. Use of the dentifrice compositions of claim 1 in the manufacture of a medicament for (i) reducing plaque accumulation, (ii) reducing or inhibiting demineralization and promote remineralization of the teeth, (iii) inhibiting microbial biofilm formation in the oral cavity, (iv) reducing or inhibiting gingivitis, (v) reducing or inhibiting formation of dental caries, (vi), reducing, repairing or inhibiting pre-carious lesions of the enamel, (vii) cleaning the teeth and oral cavity, (viii) reducing erosion, (ix) whitening teeth; and/or (x) promoting systemic health. 15. Use of microcrystalline cellulose in the manufacture of a dentifrice according to claim 1.
The invention provides high water content dentifrice compositions comprising a silica abrasive and a component containing microcrystalline cellulose and carboxymethylcellulose, as well as methods of making and using the same.1. A dentifrice composition comprising by weight a. 30-50% of water; b. 0.5% to 1.5% of a component for enhancing flavor delivery and rheological profile comprising: b1. 80% to 90% of a microcrystalline cellulose; and b2. 10% to 20% of carboxymethylcellulose or a salt thereof; wherein the weight percentage of b1 and b2. is based on the total weight of component; and c. an effective amount of a silica abrasive, wherein the abrasive component comprises 15-25% of the composition. 2. The composition of claim 1 further comprising a synthetic anionic polymeric polycarboxylate. 3. The composition of claim 2 wherein the anionic polymer is a methyl vinyl ether/maleic anhydride (PVM/MA) copolymer having an average molecular weight (M.W.) of about 30,000 to about 1,000,000 and comprises 1-5% of the weight of the composition. 4. The composition according to claim 1 wherein the silica abrasive component comprises (a) a first population of silica abrasive particles, having a d50 of less than 5 microns, and (b) a second population of silica abrasive particles, having a d50 of greater than 8 microns, wherein the ratio of the first population to the second population by weight is between 1:2 and 1:4. 5. The composition according to claim 1 further comprising an effective amount of a fluoride ion source. 6. The composition according to claim 1 further comprising 25-35% of a humectant. 7. The composition according to claim 1 further comprising 1-2% sodium lauryl sulfate (SLS). 8. The composition according to claim further comprising a viscosity modifying amount of one or more of xanthan gum, carrageenan, silica thickener, or combinations thereof. 9. The composition according to claim 1 further comprising an antibacterially effective amount of a natural extract antibacterial, non-ionic bacterial agent, triclosan magnolol, tetrahydromagnolol, butyl magnolol, honokiol, tetrahydrohonokiol or mixtures thereof. 10. The composition according to claim 1 which is obtained or obtainable by premixing microcrystalline cellulose or a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose in water prior to combination with the other ingredients. 11. The composition according to claim 1 further comprising a) 25-35% glycerin as a humectant b) 1-3% PVM/MA copolymer c) 0.1-0.5% triclosan. 12. The composition according to claim 11 comprising: a) Glycerin 25-35% b) Water 30-50% c) PVM/MA copolymer 1-3% d) Sodium fluoride 0.1-0.5% e) Triclosan 0.1-1%   f) Sodium lauryl sulfate 1-2% g) Sodium carboxymethyl cellulose 0.5-2%   h) Carrageenan 0.1-1%   i) Silica abrasive 15-25% j) Mixture of 80-90%  0.7-1.2%. microcrystalline cellulose and 10-20% sodium carboxymethyl cellulose 13. A method of making a dentifrice composition according to claim 1 comprising dispersing a mixture of 80-90% microcrystalline cellulose and 10-20% sodium carboxymethyl cellulose in water, then admixing the remaining ingredients. 14. Use of the dentifrice compositions of claim 1 in the manufacture of a medicament for (i) reducing plaque accumulation, (ii) reducing or inhibiting demineralization and promote remineralization of the teeth, (iii) inhibiting microbial biofilm formation in the oral cavity, (iv) reducing or inhibiting gingivitis, (v) reducing or inhibiting formation of dental caries, (vi), reducing, repairing or inhibiting pre-carious lesions of the enamel, (vii) cleaning the teeth and oral cavity, (viii) reducing erosion, (ix) whitening teeth; and/or (x) promoting systemic health. 15. Use of microcrystalline cellulose in the manufacture of a dentifrice according to claim 1.
1,600
521
14,917,045
1,663
This invention is intended to develop many DNA markers in plants of the genus Fragaria and to evaluate anthracnose resistance with high accuracy with the use of such many DNA markers. The markers associated with anthracnose resistance in plants of the genus Fragaria each comprise a continuous nucleic acid region sandwiched between the nucleotide sequence as shown in SEQ ID NO: 1 and the nucleotide sequence as shown in SEQ ID NO: 10 in the chromosome of a plant of the genus Fragaria.
1. A marker associated with anthracnose resistance in plants of the genus Fragaria comprising a continuous nucleic acid region sandwiched between the nucleotide sequence as shown in SEQ ID NO: 1 and the nucleotide sequence as shown in SEQ ID NO: 10 in the chromosome of the plant of the genus Fragaria. 2. The marker associated with anthracnose resistance in plants of the genus Fragaria according to claim 1, wherein the nucleic acid region comprises any nucleotide sequence selected from the group consisting of nucleotide sequences as shown in SEQ ID NOs: 1 to 10 or a part of the nucleotide sequence. 3. The marker associated with anthracnose resistance in plants of the genus Fragaria according to claim 1, wherein the nucleic acid region is located in a region sandwiched between the nucleotide sequence as shown in SEQ ID NO: 4 and the nucleotide sequence as shown in SEQ ID NO: 8 in the chromosome of the plant of the genus Fragaria. 4. A method for producing a plant line of the genus Fragaria with improved anthracnose resistance comprising: a step of extracting a chromosome of a progeny plant whose at least one parent is a plant of the genus Fragaria and/or a chromosome of the parent plant of the genus Fragaria; and a step of determining the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria according to claim 1 in the chromosome obtained above. 5. The method for producing a plant line of the genus Fragaria according to claim 4, wherein the step of determination comprises conducting a nucleic acid amplification reaction using a primer that specifically amplifies the marker associated with anthracnose resistance in the plant of the genus Fragaria to determine the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria. 6. The method for producing a plant line of the genus Fragaria according to claim 4, wherein the step of determination involves the use of a DNA chip comprising a probe corresponding to the marker associated with anthracnose resistance in the plant of the genus Fragaria. 7. The method for producing a plant line of the genus Fragaria according to claim 4, wherein the progeny plant is a seed or seedling and the chromosome is extracted from the seed or seedling. 8. A method for producing a plant line of the genus Fragaria with improved anthracnose resistance comprising: a step of extracting a chromosome of a progeny plant whose at least one parent is a plant of the genus Fragaria and/or a chromosome of the parent plant of the genus Fragaria; and a step of determining the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria according to claim 2 in the chromosome obtained above. 9. A method for producing a plant line of the genus Fragaria with improved anthracnose resistance comprising: a step of extracting a chromosome of a progeny plant whose at least one parent is a plant of the genus Fragaria and/or a chromosome of the parent plant of the genus Fragaria; and a step of determining the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria according to claim 3 in the chromosome obtained above.
This invention is intended to develop many DNA markers in plants of the genus Fragaria and to evaluate anthracnose resistance with high accuracy with the use of such many DNA markers. The markers associated with anthracnose resistance in plants of the genus Fragaria each comprise a continuous nucleic acid region sandwiched between the nucleotide sequence as shown in SEQ ID NO: 1 and the nucleotide sequence as shown in SEQ ID NO: 10 in the chromosome of a plant of the genus Fragaria.1. A marker associated with anthracnose resistance in plants of the genus Fragaria comprising a continuous nucleic acid region sandwiched between the nucleotide sequence as shown in SEQ ID NO: 1 and the nucleotide sequence as shown in SEQ ID NO: 10 in the chromosome of the plant of the genus Fragaria. 2. The marker associated with anthracnose resistance in plants of the genus Fragaria according to claim 1, wherein the nucleic acid region comprises any nucleotide sequence selected from the group consisting of nucleotide sequences as shown in SEQ ID NOs: 1 to 10 or a part of the nucleotide sequence. 3. The marker associated with anthracnose resistance in plants of the genus Fragaria according to claim 1, wherein the nucleic acid region is located in a region sandwiched between the nucleotide sequence as shown in SEQ ID NO: 4 and the nucleotide sequence as shown in SEQ ID NO: 8 in the chromosome of the plant of the genus Fragaria. 4. A method for producing a plant line of the genus Fragaria with improved anthracnose resistance comprising: a step of extracting a chromosome of a progeny plant whose at least one parent is a plant of the genus Fragaria and/or a chromosome of the parent plant of the genus Fragaria; and a step of determining the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria according to claim 1 in the chromosome obtained above. 5. The method for producing a plant line of the genus Fragaria according to claim 4, wherein the step of determination comprises conducting a nucleic acid amplification reaction using a primer that specifically amplifies the marker associated with anthracnose resistance in the plant of the genus Fragaria to determine the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria. 6. The method for producing a plant line of the genus Fragaria according to claim 4, wherein the step of determination involves the use of a DNA chip comprising a probe corresponding to the marker associated with anthracnose resistance in the plant of the genus Fragaria. 7. The method for producing a plant line of the genus Fragaria according to claim 4, wherein the progeny plant is a seed or seedling and the chromosome is extracted from the seed or seedling. 8. A method for producing a plant line of the genus Fragaria with improved anthracnose resistance comprising: a step of extracting a chromosome of a progeny plant whose at least one parent is a plant of the genus Fragaria and/or a chromosome of the parent plant of the genus Fragaria; and a step of determining the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria according to claim 2 in the chromosome obtained above. 9. A method for producing a plant line of the genus Fragaria with improved anthracnose resistance comprising: a step of extracting a chromosome of a progeny plant whose at least one parent is a plant of the genus Fragaria and/or a chromosome of the parent plant of the genus Fragaria; and a step of determining the presence or absence of the marker associated with anthracnose resistance in the plant of the genus Fragaria according to claim 3 in the chromosome obtained above.
1,600
522
14,141,946
1,616
Compositions for the transdermal delivery of NSAIDs in a flexible, finite form are described. The compositions comprise a polymer matrix that includes an NSAID and a polymer matrix comprising a blend of silicone and acrylic polymers. A flexible, occlusive backing material also is disclosed.
1. A composition for the transdermal delivery of an NSAID in the form of a flexible finite system for topical application, comprising a polymer matrix comprising (i) a therapeutically effective amount of an NSAID; (ii) a silicone polymer; and (iii) an acrylic polymer or an acrylic block copolymer. 2. The composition of claim 1, wherein the polymer matrix further comprises a styrene-isoprene-styrene block copolymer. 3. The composition of claim 1, wherein the polymer matrix further comprises a penetration enhancer. 4. The composition of claim 1, wherein the NSAID comprises flurbiprofen. 5. The composition of claim 1, wherein the polymer matrix comprises a non-functional acrylic polymer. 6. The composition of claim 1, wherein the polymer matrix comprises an acrylic block copolymer. 7. The composition of claim 1, wherein the polymer matrix comprises about 3% to 5% w/w flurbiprofen. 8. The composition of claim 1, wherein the polymer matrix comprises about 4 to 5% w/w acrylic polymer. 9. The composition of claim 1, wherein the polymer matrix comprises about 5% w/w acrylic block copolymer. 10. The composition of claim 1, wherein the polymer matrix comprises up to about 1% w/w styrene-isoprene-styrene block copolymer. 11. The composition of claim wherein the polymer matrix comprises at least about 80% w/w silicone polymer. 12. The composition of claim 1, wherein the system achieves delivery of the NSAID over a period of time of at least 8 hours. 13. The composition of claim 1, wherein the system achieves delivery of the NSAID over a period of time of at least 12 hours. 14. The composition of claim 1, wherein the system achieves delivery of the NSAID over a period of time of at least 24 hours. 15. The composition of claim 1, further comprising a backing layer. 16. The composition of claim 1, further comprising a flexible, occlusive backing layer. 17. The composition of claim 16, wherein the flexible, occlusive backing layer is stretchable. 18. The composition of any claim 16, wherein the flexible, occlusive backing layer is comprised of a fabric backing material coated with an occlusive coating. 19. The composition of claim 18, wherein the occlusive coating comprises polyisobutylene and/or styrene-isoprene-styrene (SIS) block copolymers. 20. A method for the transdermal delivery of an NSAID, comprising topically applying a composition as claimed in claim 1 to the skin or mucosa of a subject in need thereof. 21. A flexible, finite system for the transdermal delivery of an active agent comprising, (i) a polymer matrix comprising the active agent and one or more polymers and (ii) a flexible, occlusive backing layer comprised of a fabric backing material coated with an occlusive coating. 22. The flexible, finite system of claim 21, wherein the flexible, occlusive backing layer is stretchable. 23. The flexible, finite system of claim 22, wherein the flexible, finite system is stretchable. 24. A method for the transdermal delivery of an NSAID, comprising topically applying a flexible, finite system as claimed in claim 21 to the skin or mucosa of a subject in need thereof.
Compositions for the transdermal delivery of NSAIDs in a flexible, finite form are described. The compositions comprise a polymer matrix that includes an NSAID and a polymer matrix comprising a blend of silicone and acrylic polymers. A flexible, occlusive backing material also is disclosed.1. A composition for the transdermal delivery of an NSAID in the form of a flexible finite system for topical application, comprising a polymer matrix comprising (i) a therapeutically effective amount of an NSAID; (ii) a silicone polymer; and (iii) an acrylic polymer or an acrylic block copolymer. 2. The composition of claim 1, wherein the polymer matrix further comprises a styrene-isoprene-styrene block copolymer. 3. The composition of claim 1, wherein the polymer matrix further comprises a penetration enhancer. 4. The composition of claim 1, wherein the NSAID comprises flurbiprofen. 5. The composition of claim 1, wherein the polymer matrix comprises a non-functional acrylic polymer. 6. The composition of claim 1, wherein the polymer matrix comprises an acrylic block copolymer. 7. The composition of claim 1, wherein the polymer matrix comprises about 3% to 5% w/w flurbiprofen. 8. The composition of claim 1, wherein the polymer matrix comprises about 4 to 5% w/w acrylic polymer. 9. The composition of claim 1, wherein the polymer matrix comprises about 5% w/w acrylic block copolymer. 10. The composition of claim 1, wherein the polymer matrix comprises up to about 1% w/w styrene-isoprene-styrene block copolymer. 11. The composition of claim wherein the polymer matrix comprises at least about 80% w/w silicone polymer. 12. The composition of claim 1, wherein the system achieves delivery of the NSAID over a period of time of at least 8 hours. 13. The composition of claim 1, wherein the system achieves delivery of the NSAID over a period of time of at least 12 hours. 14. The composition of claim 1, wherein the system achieves delivery of the NSAID over a period of time of at least 24 hours. 15. The composition of claim 1, further comprising a backing layer. 16. The composition of claim 1, further comprising a flexible, occlusive backing layer. 17. The composition of claim 16, wherein the flexible, occlusive backing layer is stretchable. 18. The composition of any claim 16, wherein the flexible, occlusive backing layer is comprised of a fabric backing material coated with an occlusive coating. 19. The composition of claim 18, wherein the occlusive coating comprises polyisobutylene and/or styrene-isoprene-styrene (SIS) block copolymers. 20. A method for the transdermal delivery of an NSAID, comprising topically applying a composition as claimed in claim 1 to the skin or mucosa of a subject in need thereof. 21. A flexible, finite system for the transdermal delivery of an active agent comprising, (i) a polymer matrix comprising the active agent and one or more polymers and (ii) a flexible, occlusive backing layer comprised of a fabric backing material coated with an occlusive coating. 22. The flexible, finite system of claim 21, wherein the flexible, occlusive backing layer is stretchable. 23. The flexible, finite system of claim 22, wherein the flexible, finite system is stretchable. 24. A method for the transdermal delivery of an NSAID, comprising topically applying a flexible, finite system as claimed in claim 21 to the skin or mucosa of a subject in need thereof.
1,600
523
15,292,344
1,616
Disclosed is a cosmetic or dermatological preparation which comprises glycyrrhetic acid and/or glycyrrhizin and further comprises at least one substance selected from UV filter substances, self-tanning agents and antioxidants.
1. A cosmetic or dermatological preparation, wherein the preparation comprises at least one of glycyrrhetic acid and glycyrrhizin in an amount of at least 0.0001% by weight, based on a total weight of the preparation, and further comprises at least one substance which is selected from UV filter substances, self-tanning agents and antioxidants. 2. The preparation of claim 1, wherein the preparation comprises at least glycyrrhetic acid. 3. The preparation of claim 1, wherein the preparation comprises at least glycyrrhizin. 4. The preparation of claim 1, wherein the preparation comprises at least one UV filter substance. 5. The preparation of claim 1, wherein the preparation comprises not more than 20% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 6. The preparation of claim 5, wherein the preparation comprises at least 0.001% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 7. The preparation of claim 1, wherein the preparation comprises not more than 10% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 8. The preparation of claim 7, wherein the preparation comprises at least 0.01% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 9. The preparation of claim 1, wherein the preparation comprises not more than 1% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 10. The preparation of claim 1, wherein the preparation comprises at least one UV filter substance. 11. The preparation of claim 10, wherein the at least one UV filter substance comprises at least one of ethylhexyl methoxycinnamate, butyl methoxydibenzoylmethane, and ethylhexyl triazone. 12. The preparation of claim 1, wherein the preparation comprises at least one self-tanning substance. 13. The preparation of claim 12, wherein the at least one self-tanning substance comprises dihydroxyacetone. 14. The preparation of claim 12, wherein the at least one self-tanning substance comprises erythrulose. 15. The preparation of claim 12, wherein the at least one self-tanning substance is present in an amount of from 0.1% to 8% by weight, based on a total weight of the preparation. 16. The preparation of claim 1, wherein the preparation comprises at least one antioxidant. 17. The preparation of claim 16, wherein the at least one antioxidant comprises at least one of ascorbic acid, ascorbyl palmitate, Na or Mg ascorbyl phosphate, ascorbyl acetate, rutinic acid, alpha-glucosyl rutin, quercetin, isoquercetin, vitamin E and derivatives thereof, vitamin A and derivatives thereof, carnosine, butylated hydroxytoluene, butylated hydroxyanisole, beta-alanine, a carotenoide and phytoene. 18. The preparation of claim 16, wherein the at least one antioxidant comprises at least one of a carotenoide and phytoene. 19. The preparation of claim 16, wherein the at least one antioxidant comprises Na or Mg ascorbyl phosphate. 20. The preparation of claim 1, wherein the preparation further comprises ubiquinone (coenzyme Q10).
Disclosed is a cosmetic or dermatological preparation which comprises glycyrrhetic acid and/or glycyrrhizin and further comprises at least one substance selected from UV filter substances, self-tanning agents and antioxidants.1. A cosmetic or dermatological preparation, wherein the preparation comprises at least one of glycyrrhetic acid and glycyrrhizin in an amount of at least 0.0001% by weight, based on a total weight of the preparation, and further comprises at least one substance which is selected from UV filter substances, self-tanning agents and antioxidants. 2. The preparation of claim 1, wherein the preparation comprises at least glycyrrhetic acid. 3. The preparation of claim 1, wherein the preparation comprises at least glycyrrhizin. 4. The preparation of claim 1, wherein the preparation comprises at least one UV filter substance. 5. The preparation of claim 1, wherein the preparation comprises not more than 20% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 6. The preparation of claim 5, wherein the preparation comprises at least 0.001% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 7. The preparation of claim 1, wherein the preparation comprises not more than 10% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 8. The preparation of claim 7, wherein the preparation comprises at least 0.01% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 9. The preparation of claim 1, wherein the preparation comprises not more than 1% by weight of the at least one of glycyrrhetic acid and glycyrrhizin. 10. The preparation of claim 1, wherein the preparation comprises at least one UV filter substance. 11. The preparation of claim 10, wherein the at least one UV filter substance comprises at least one of ethylhexyl methoxycinnamate, butyl methoxydibenzoylmethane, and ethylhexyl triazone. 12. The preparation of claim 1, wherein the preparation comprises at least one self-tanning substance. 13. The preparation of claim 12, wherein the at least one self-tanning substance comprises dihydroxyacetone. 14. The preparation of claim 12, wherein the at least one self-tanning substance comprises erythrulose. 15. The preparation of claim 12, wherein the at least one self-tanning substance is present in an amount of from 0.1% to 8% by weight, based on a total weight of the preparation. 16. The preparation of claim 1, wherein the preparation comprises at least one antioxidant. 17. The preparation of claim 16, wherein the at least one antioxidant comprises at least one of ascorbic acid, ascorbyl palmitate, Na or Mg ascorbyl phosphate, ascorbyl acetate, rutinic acid, alpha-glucosyl rutin, quercetin, isoquercetin, vitamin E and derivatives thereof, vitamin A and derivatives thereof, carnosine, butylated hydroxytoluene, butylated hydroxyanisole, beta-alanine, a carotenoide and phytoene. 18. The preparation of claim 16, wherein the at least one antioxidant comprises at least one of a carotenoide and phytoene. 19. The preparation of claim 16, wherein the at least one antioxidant comprises Na or Mg ascorbyl phosphate. 20. The preparation of claim 1, wherein the preparation further comprises ubiquinone (coenzyme Q10).
1,600
524
14,416,438
1,612
The invention relates to nail composition sets comprising at least one primer and at least one color coat, wherein the primer comprises water and at least one adhesive compound and preferably further comprises at least one water-soluble film forming agent and/or at least one plasticizer.
1. A nail composition set comprising at least one primer and at least one color coat. 2. The nail composition set of claim 1, further comprising at least one basecoat and/or at least one topcoat. 3. The nail composition set of claim 1, wherein the color coat is a UV gel composition. 4. The nail composition set of claim 1, wherein the primer comprises (a) water and (b) at least one latex and/or at least one pseudolatex. 5. The nail composition set of claim 4, wherein the at least one latex and/or at least one pseudolatex is at least one aqueous polyurethane dispersion. 6. The nail composition set of claim 5, wherein the at least one aqueous polyurethane dispersion comprises a reaction product of a prepolymer comprising a dihydroxyl compound, a polyisocyanate, and a low molecular weight diol and at least two diamine compounds and wherein the composition is substantially free of triethanolamine stearate. 7. The composition of claim 5, wherein the at least one aqueous polyurethane dispersion comprises a reaction product of: a prepolymer according to the following formula: wherein R1 represents a dihydroxyl compound having a number average molecular weights of from about 700 to about 16,000, R2 represents a hydrocarbon radical of an aliphatic or cycloaliphatic polyisocyanate, and R3 represents a radical of a low molecular weight diol, n is from 0 to 5, and m is >1; at least one chain extender according to the formula: H2N—R4—NH2 wherein R4 represents an alkylene or alkylene oxide radical not substituted with ionic or potentially ionic groups; and at least one chain extender according to the formula: H2N—R5—NH2 wherein R5 represents an alkylene radical substituted with ionic or potentially ionic groups. 8. The nail composition set of claim 4, wherein the primer further comprises at least one water-soluble film forming agent. 9. The nail composition set of claim 4, wherein the primer further comprises at least one plasticizer. 10. The nail composition set of claim 1, wherein the color coat comprises at least one colorant. 11. The nail composition set of claim 1, wherein the primer is water-based. 12. A method of removing a nail composition set comprising at least one color coat and at least one primer, comprising removing the primer to effect removal of the primer as well as the color coat of the nail composition set. 13. The method of claim 12, wherein the color coat is a UV gel composition. 14. The method of claim 12, wherein the primer comprises (a) water and (b) at least one latex and/or at least one pseudolatex. 15. A kit for a nail composition set comprising (a) at least one primer composition comprising (i) water and (ii) latex and/or pseudolatex; and (b) one or more compositions selected from the group consisting of a topcoat composition, a basecoat composition, and a color coat composition. 16. The kit of claim 15, further comprising (c) instructions for removing a nail composition by removing the primer composition to effect removal of the nail composition set. 17. The kit of claim 15, wherein the kit comprises at least one color coat composition which is a UV gel composition.
The invention relates to nail composition sets comprising at least one primer and at least one color coat, wherein the primer comprises water and at least one adhesive compound and preferably further comprises at least one water-soluble film forming agent and/or at least one plasticizer.1. A nail composition set comprising at least one primer and at least one color coat. 2. The nail composition set of claim 1, further comprising at least one basecoat and/or at least one topcoat. 3. The nail composition set of claim 1, wherein the color coat is a UV gel composition. 4. The nail composition set of claim 1, wherein the primer comprises (a) water and (b) at least one latex and/or at least one pseudolatex. 5. The nail composition set of claim 4, wherein the at least one latex and/or at least one pseudolatex is at least one aqueous polyurethane dispersion. 6. The nail composition set of claim 5, wherein the at least one aqueous polyurethane dispersion comprises a reaction product of a prepolymer comprising a dihydroxyl compound, a polyisocyanate, and a low molecular weight diol and at least two diamine compounds and wherein the composition is substantially free of triethanolamine stearate. 7. The composition of claim 5, wherein the at least one aqueous polyurethane dispersion comprises a reaction product of: a prepolymer according to the following formula: wherein R1 represents a dihydroxyl compound having a number average molecular weights of from about 700 to about 16,000, R2 represents a hydrocarbon radical of an aliphatic or cycloaliphatic polyisocyanate, and R3 represents a radical of a low molecular weight diol, n is from 0 to 5, and m is >1; at least one chain extender according to the formula: H2N—R4—NH2 wherein R4 represents an alkylene or alkylene oxide radical not substituted with ionic or potentially ionic groups; and at least one chain extender according to the formula: H2N—R5—NH2 wherein R5 represents an alkylene radical substituted with ionic or potentially ionic groups. 8. The nail composition set of claim 4, wherein the primer further comprises at least one water-soluble film forming agent. 9. The nail composition set of claim 4, wherein the primer further comprises at least one plasticizer. 10. The nail composition set of claim 1, wherein the color coat comprises at least one colorant. 11. The nail composition set of claim 1, wherein the primer is water-based. 12. A method of removing a nail composition set comprising at least one color coat and at least one primer, comprising removing the primer to effect removal of the primer as well as the color coat of the nail composition set. 13. The method of claim 12, wherein the color coat is a UV gel composition. 14. The method of claim 12, wherein the primer comprises (a) water and (b) at least one latex and/or at least one pseudolatex. 15. A kit for a nail composition set comprising (a) at least one primer composition comprising (i) water and (ii) latex and/or pseudolatex; and (b) one or more compositions selected from the group consisting of a topcoat composition, a basecoat composition, and a color coat composition. 16. The kit of claim 15, further comprising (c) instructions for removing a nail composition by removing the primer composition to effect removal of the nail composition set. 17. The kit of claim 15, wherein the kit comprises at least one color coat composition which is a UV gel composition.
1,600
525
15,015,458
1,633
The present invention relates to vaccines comprising at least one mRNA encoding at least one antigen for use in the treatment of a disease in an elderly patient preferably exhibiting an age of at least 50 years, more preferably of at least 55 years, 60 years, 65 years, 70 years, or older, wherein the treatment comprises vaccination of the patient and eliciting an immune response in said patient. The present invention is furthermore directed to kits and kits of parts comprising such a vaccine and/or its components and to methods applying such a vaccine or kit.
1. A method for stimulating an immune response in a patient comprising selecting an elderly human patient exhibiting an age of at least 50 years and administering to the subject a composition comprising at least one mRNA encoding at least one antigen for the prophylaxis and/or treatment of a disease. 2. The method of claim 1, wherein the immune response is a Th1 immune response. 3. The method of claim 1, wherein the elderly patient is male or female and/or exhibits an age of at least 55 years, 60 years, 65 years, 70 years, or older. 4. The method of claim 1, wherein the disease is selected from infectious diseases, viral, bacterial or protozoological infectious diseases, autoimmune diseases, allergies or allergic diseases or cancer or tumour diseases. 5. The method of claim 1, wherein the antigen is selected from protein and peptide antigens, tumour antigens, self-antigens or auto-antigens, autoimmune self-antigens, pathogenic antigens, viral antigens, bacterial antigens, fungal antigens, protozoological antigens, animal antigens, allergy antigens. 6. The method of claim 1, wherein the vaccine is to be administered parenterally, orally, nasally, pulmonary, by inhalation, topically, rectally, buccally, vaginally, or via an implanted reservoir. 7. The method of claim 1, wherein the at least one mRNA encoding at least one antigen is to be administered in its naked form or is associated with or complexed with a cationic or polycationic compound. 8. The method of claim 1, wherein the at least one mRNA encoding at least one antigen is complexed with a polymeric carrier formed by disulfide-crosslinked cationic components selected from an oligopeptide having following sum formula (I): {(Arg)l;(Lys)m;(His)n;(Orn)0;(Xaa)x};  (formula (I) Wherein l+m+n+o+x=3-100, and l, m, n or o independently of each other is any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21-30, 31-40, 41-50, 51-60, 61-70, 71-80, 81-90 and 91-100 provided that the overall content of Arg (Arginine), Lys (Lysine), His (Histidine) and Orn (Ornithine) represents at least 10% of all amino acids of the oligopeptide of formula (V); and Xaa is any amino acid selected from native (=naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x is any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21-30, 31-40, 41-50, 51-60, 61-70, 71-80, 81-90, provided, that the overall content of Xaa does not exceed 90% of all amino acids of the oligopeptide of formula (I), or from a disulfide-crosslinked cationic component comprising as a repeat unit an oligopeptide having following subformula (Ia): {(Arg)l;(Lys)m;(His)n;(Orn)o;(Xaa′)x(Cys)y};  formula (Ia) wherein (Arg)l;(Lys)m;(His)n;(Orn)0; and x is preferably are as defined above for formula (I), Xaa′ is any amino acid selected from native (=naturally occurring) or non-native amino acids except of Arg, Lys, His, Orn or Cys and y is any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21-30, 31-40, 41-50, 51-60, 61-70, 71-80 and 81-90, provided that the overall content of Arg (Arginine), Lys (Lysine), His (Histidine) and Orn (Ornithine) represents at least 10% of all amino acids of the oligopeptide. or from a disulfide-crosslinked cationic component comprising as a repeat unit an oligopeptide having following subformula (Ib): Cys1{(Arg)l;(Lys)m;(His)n;(Orn)0;(Xaa)x}Cys2;  (formula (Ib)) wherein component {(Arg)l;(Lys)m;(His)n;(Orn)0;(Xaa)x} (formula (I)) within formula (Ib) is as defined herein and forms a core of subformula (Ib), and wherein Cys1 and Cys2 are Cysteines proximal to, or terminal to (Arg)l;(Lys)m;(His)n;(Orn)o;(Xaa)x. 9. The method of claim 1, wherein the at least one mRNA encoding at least one antigen is complexed with a polymeric carrier according to generic formula (VI): L-P1-S-[S-P2-S]n-S-P3-L wherein, P1 and P3 are different or identical to each other and represent a linear or branched hydrophilic polymer chain, each P1 and P3 exhibiting at least one —SH-moiety, capable to form a disulfide linkage upon condensation with component P2, the linear or branched hydrophilic polymer chain selected independent from each other from polyethylene glycol (PEG), poly-N-(2-hydroxypropyl)methacrylamide, poly-2-(methacryloyloxy)ethyl phosphorylcholines, poly(hydroxyalkyl L-asparagine), poly(2-(methacryloyloxy)ethyl phosphorylcholine), hydroxyethylstarch or poly(hydroxyalkyl L-glutamine), wherein the hydrophilic polymer chain exhibits a molecular weight of about 1 kDa to about 100 kDa, P2 is a cationic or polycationic peptide or protein, having a length of about 3 to about 100 amino acids, or is a cationic or polycationic polymer, having a molecular weight of about 0.5 kda to about 30 kda, each P2 exhibiting at least two —SH-moieties, capable to form a disulfide linkage upon condensation with further components P2 or component(s) P1 and/or P3; —S—S— is a (reversible) disulfide bond; L is an optional ligand, which may be present or not, and may be selected independent from the other from RGD, Transferrin, Folate, a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide (CPP), TAT, KALA, a ligand of a receptor, cytokines, hormones, growth factors, small molecules, carbohydrates, mannose, galactose, synthetic ligands, small molecule agonists, inhibitors or antagonists of receptors, or RGD peptidomimetic analogues; and n is an integer, selected from a range of about 1 to 50, preferably in a range of about 1, 2, 3, 4, or 5 to 10, more preferably in a range of about 1, 2, 3, or 4 to 9. 10. The method of claim 9, wherein the at least one mRNA encoding at least one antigen is complexed with a polymeric carrier molecule according to formula (VIa) L-P1-S-{[S-P2-S]a[S-(AA)x-S]b}-S-P3-L, wherein S, L, P1, P2 and P3 are preferably as defined above for formula (VI), a and b are integers, wherein a+b=n and n is an integer as defined above for formula (VI), x is an integer selected from a range of about 1 to 100, and (AA) is selected from an aromatic, a hydrophilic, a lipophilic, or a weak basic amino acid or peptide, or is a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide (CPPp), or is selected from therapeutically active proteins or peptides, from antigens, tumour antigens, pathogenic antigens, animal antigens, viral antigens, protozoan antigens, bacterial antigens, allergic antigens, autoimmune antigens, from allergens, from antibodies, from immunostimulatory proteins or peptides, or from antigen-specific T-cell receptors. 11. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 12. The method of claim 11, wherein the (m)RNA is an mRNA, an immunostimulatory nucleic acid, a CpG nucleic acid, a CpG-RNA, a CpG-DNA, or an immunostimulatory RNA (isRNA). 13. The method claim 1, wherein the vaccine furthermore comprises a pharmaceutically acceptable carrier and or vehicle. 14. The method of claim 1, wherein the vaccine furthermore comprises at least one adjuvant, an auxiliary substance selected from lipopolysaccharides, TNF-alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta, TNF-alpha, growth factors, and hGH, a ligand of human Toll-like receptor TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, a ligand of murine Toll-like receptor TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 OR TLR13, a ligand of a NOD-like receptor, a ligand of a RIG-I like receptor, an immunostimulatory nucleic acid, an immunostimulatory RNA (isRNA), a CpG-DNA, an antibacterial agent, or an anti-viral agent. 15. (canceled) 16. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 17. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 18. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 19. The method of claim 11, wherein the (m)RNA is an mRNA, an immunostimulatory nucleic acid, a CpG nucleic acid, a CpG-RNA, a CpG-DNA, or an immunostimulatory RNA (isRNA). 20. The method of claim 1, wherein the patient exhibits an age of at least 85 years. 21. The method of claim 1, wherein the vaccine is administered intradermally. 22. The method of claim 1, wherein the vaccine is administered at least a second time. 23. The method of claim 1, wherein the antigen is an infectious disease antigen and the wherein the method provides a protective immune response. 24. The method of claim 1, wherein the antigen is a prostate cancer antigen and the patient has prostate cancer.
The present invention relates to vaccines comprising at least one mRNA encoding at least one antigen for use in the treatment of a disease in an elderly patient preferably exhibiting an age of at least 50 years, more preferably of at least 55 years, 60 years, 65 years, 70 years, or older, wherein the treatment comprises vaccination of the patient and eliciting an immune response in said patient. The present invention is furthermore directed to kits and kits of parts comprising such a vaccine and/or its components and to methods applying such a vaccine or kit.1. A method for stimulating an immune response in a patient comprising selecting an elderly human patient exhibiting an age of at least 50 years and administering to the subject a composition comprising at least one mRNA encoding at least one antigen for the prophylaxis and/or treatment of a disease. 2. The method of claim 1, wherein the immune response is a Th1 immune response. 3. The method of claim 1, wherein the elderly patient is male or female and/or exhibits an age of at least 55 years, 60 years, 65 years, 70 years, or older. 4. The method of claim 1, wherein the disease is selected from infectious diseases, viral, bacterial or protozoological infectious diseases, autoimmune diseases, allergies or allergic diseases or cancer or tumour diseases. 5. The method of claim 1, wherein the antigen is selected from protein and peptide antigens, tumour antigens, self-antigens or auto-antigens, autoimmune self-antigens, pathogenic antigens, viral antigens, bacterial antigens, fungal antigens, protozoological antigens, animal antigens, allergy antigens. 6. The method of claim 1, wherein the vaccine is to be administered parenterally, orally, nasally, pulmonary, by inhalation, topically, rectally, buccally, vaginally, or via an implanted reservoir. 7. The method of claim 1, wherein the at least one mRNA encoding at least one antigen is to be administered in its naked form or is associated with or complexed with a cationic or polycationic compound. 8. The method of claim 1, wherein the at least one mRNA encoding at least one antigen is complexed with a polymeric carrier formed by disulfide-crosslinked cationic components selected from an oligopeptide having following sum formula (I): {(Arg)l;(Lys)m;(His)n;(Orn)0;(Xaa)x};  (formula (I) Wherein l+m+n+o+x=3-100, and l, m, n or o independently of each other is any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21-30, 31-40, 41-50, 51-60, 61-70, 71-80, 81-90 and 91-100 provided that the overall content of Arg (Arginine), Lys (Lysine), His (Histidine) and Orn (Ornithine) represents at least 10% of all amino acids of the oligopeptide of formula (V); and Xaa is any amino acid selected from native (=naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x is any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21-30, 31-40, 41-50, 51-60, 61-70, 71-80, 81-90, provided, that the overall content of Xaa does not exceed 90% of all amino acids of the oligopeptide of formula (I), or from a disulfide-crosslinked cationic component comprising as a repeat unit an oligopeptide having following subformula (Ia): {(Arg)l;(Lys)m;(His)n;(Orn)o;(Xaa′)x(Cys)y};  formula (Ia) wherein (Arg)l;(Lys)m;(His)n;(Orn)0; and x is preferably are as defined above for formula (I), Xaa′ is any amino acid selected from native (=naturally occurring) or non-native amino acids except of Arg, Lys, His, Orn or Cys and y is any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21-30, 31-40, 41-50, 51-60, 61-70, 71-80 and 81-90, provided that the overall content of Arg (Arginine), Lys (Lysine), His (Histidine) and Orn (Ornithine) represents at least 10% of all amino acids of the oligopeptide. or from a disulfide-crosslinked cationic component comprising as a repeat unit an oligopeptide having following subformula (Ib): Cys1{(Arg)l;(Lys)m;(His)n;(Orn)0;(Xaa)x}Cys2;  (formula (Ib)) wherein component {(Arg)l;(Lys)m;(His)n;(Orn)0;(Xaa)x} (formula (I)) within formula (Ib) is as defined herein and forms a core of subformula (Ib), and wherein Cys1 and Cys2 are Cysteines proximal to, or terminal to (Arg)l;(Lys)m;(His)n;(Orn)o;(Xaa)x. 9. The method of claim 1, wherein the at least one mRNA encoding at least one antigen is complexed with a polymeric carrier according to generic formula (VI): L-P1-S-[S-P2-S]n-S-P3-L wherein, P1 and P3 are different or identical to each other and represent a linear or branched hydrophilic polymer chain, each P1 and P3 exhibiting at least one —SH-moiety, capable to form a disulfide linkage upon condensation with component P2, the linear or branched hydrophilic polymer chain selected independent from each other from polyethylene glycol (PEG), poly-N-(2-hydroxypropyl)methacrylamide, poly-2-(methacryloyloxy)ethyl phosphorylcholines, poly(hydroxyalkyl L-asparagine), poly(2-(methacryloyloxy)ethyl phosphorylcholine), hydroxyethylstarch or poly(hydroxyalkyl L-glutamine), wherein the hydrophilic polymer chain exhibits a molecular weight of about 1 kDa to about 100 kDa, P2 is a cationic or polycationic peptide or protein, having a length of about 3 to about 100 amino acids, or is a cationic or polycationic polymer, having a molecular weight of about 0.5 kda to about 30 kda, each P2 exhibiting at least two —SH-moieties, capable to form a disulfide linkage upon condensation with further components P2 or component(s) P1 and/or P3; —S—S— is a (reversible) disulfide bond; L is an optional ligand, which may be present or not, and may be selected independent from the other from RGD, Transferrin, Folate, a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide (CPP), TAT, KALA, a ligand of a receptor, cytokines, hormones, growth factors, small molecules, carbohydrates, mannose, galactose, synthetic ligands, small molecule agonists, inhibitors or antagonists of receptors, or RGD peptidomimetic analogues; and n is an integer, selected from a range of about 1 to 50, preferably in a range of about 1, 2, 3, 4, or 5 to 10, more preferably in a range of about 1, 2, 3, or 4 to 9. 10. The method of claim 9, wherein the at least one mRNA encoding at least one antigen is complexed with a polymeric carrier molecule according to formula (VIa) L-P1-S-{[S-P2-S]a[S-(AA)x-S]b}-S-P3-L, wherein S, L, P1, P2 and P3 are preferably as defined above for formula (VI), a and b are integers, wherein a+b=n and n is an integer as defined above for formula (VI), x is an integer selected from a range of about 1 to 100, and (AA) is selected from an aromatic, a hydrophilic, a lipophilic, or a weak basic amino acid or peptide, or is a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide (CPPp), or is selected from therapeutically active proteins or peptides, from antigens, tumour antigens, pathogenic antigens, animal antigens, viral antigens, protozoan antigens, bacterial antigens, allergic antigens, autoimmune antigens, from allergens, from antibodies, from immunostimulatory proteins or peptides, or from antigen-specific T-cell receptors. 11. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 12. The method of claim 11, wherein the (m)RNA is an mRNA, an immunostimulatory nucleic acid, a CpG nucleic acid, a CpG-RNA, a CpG-DNA, or an immunostimulatory RNA (isRNA). 13. The method claim 1, wherein the vaccine furthermore comprises a pharmaceutically acceptable carrier and or vehicle. 14. The method of claim 1, wherein the vaccine furthermore comprises at least one adjuvant, an auxiliary substance selected from lipopolysaccharides, TNF-alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta, TNF-alpha, growth factors, and hGH, a ligand of human Toll-like receptor TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, a ligand of murine Toll-like receptor TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 OR TLR13, a ligand of a NOD-like receptor, a ligand of a RIG-I like receptor, an immunostimulatory nucleic acid, an immunostimulatory RNA (isRNA), a CpG-DNA, an antibacterial agent, or an anti-viral agent. 15. (canceled) 16. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 17. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 18. The method of claim 1, wherein the vaccine is formulated to comprise a) an (adjuvant) component, comprising or consisting of at least one (m)RNA, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and b) at least one free mRNA encoding an antigen. 19. The method of claim 11, wherein the (m)RNA is an mRNA, an immunostimulatory nucleic acid, a CpG nucleic acid, a CpG-RNA, a CpG-DNA, or an immunostimulatory RNA (isRNA). 20. The method of claim 1, wherein the patient exhibits an age of at least 85 years. 21. The method of claim 1, wherein the vaccine is administered intradermally. 22. The method of claim 1, wherein the vaccine is administered at least a second time. 23. The method of claim 1, wherein the antigen is an infectious disease antigen and the wherein the method provides a protective immune response. 24. The method of claim 1, wherein the antigen is a prostate cancer antigen and the patient has prostate cancer.
1,600
526
14,819,342
1,625
The present application relates to compounded compositions, methods of making compounded compositions, kits comprising compounded compositions, containers comprising compounded compositions, and methods of using compounded compositions. For example, disclosed herein are compounded compositions comprising an anti-fungal agent and methods of using a compounded composition to treat or prevent a fungal infection or a suspected fungal infection. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.
1. A method of treating or preventing a fungal infection in a subject, the method comprising: (i) adding to water contained within a foot bath a compounded composition comprising fluconazole and LoxaSperse™ excipient base powder; (ii) agitating the water using an agitator, thereby distributing the compounded composition throughout the water; and (iii) contacting the agitated water with at least a portion of one or both feet of a subject, wherein the subject has been diagnosed with, is suspected of having, or is at risk of developing a fungal infection. 2. A method of treating or preventing a fungal infection, the method comprising: (i) adding to water contained within a foot bath a compounded composition comprising an anti-fungal agent and an excipient base powder comprising a blend of micronized xylitol and poloxamers; (ii) agitating the water contained within the foot bath; and (iii) contacting the agitated water with at least a portion of one or both feet of a subject. 3. The method of claim 2, wherein the subject has been diagnosed with, is suspected of having, or is at risk of developing a fungal infection of one or both feet. 4. The method of claim 2, wherein the excipient base powder is LoxaSperse™ excipient base powder or XyliFos™ excipient base powder. 5. The method of claim 2, wherein the ratio of the anti-fungal agent to excipient base powder in the compounded composition is from about 1:60 to about 1:3. 6. The method of claim 2, wherein the ratio of the anti-fungal agent to excipient base powder in the compounded composition is about 1:15. 7. The method of claim 2, wherein the anti-fungal agent comprises fluconazole. 8. The method of claim 2, wherein the method comprises repeating steps (i)-(iii) daily. 9. The method of claim 2, wherein the method comprises heating the water contained within the foot bath. 10. The method of claim 2, wherein the compounded composition comprises one or more additional anti-infective agents. 11. The method of claim 10, wherein the additional anti-infective agent comprises an anti-bacterial agent. 12. The method of claim 2, wherein the method comprises administering to the subject an oral pharmaceutical composition comprising an anti-infective agent. 13. A compounded composition comprising fluconazole and an excipient base powder comprising a blend of micronized xylitol and poloxamers. 14. The compounded composition of claim 13, wherein the excipient base powder is LoxaSperse™ excipient base powder or XyliFos™ excipient base powder. 15. The compounded composition of claim 13, wherein the ratio of fluconazole to excipient base powder in the compounded composition is from about 1:60 to about 1:3. 16. The compounded composition of claim 13, wherein the ratio of fluconazole to excipient base powder in the compounded composition is about 1:15. 17. The compounded composition of claim 13, wherein the compounded composition comprises one or more additional anti-infective agents. 18. The compounded composition of claim 17, wherein the additional anti-infective agent comprises an anti-bacterial agent. 19. A method of making a compounded composition, the method comprising: obtaining an anti-fungal agent; obtaining an excipient base powder comprising a blend of micronized xylitol and poloxamers; and mixing a therapeutically effective amount of the anti-fungal agent with a sufficient amount of the excipient base powder to make a homogenous compounded composition. 20. The method of claim 19, wherein the excipient base is LoxaSperse™ excipient base powder or XyliFos™ excipient base powder. 21. The method of claim 19, wherein the anti-fungal agent comprises fluconazole. 22. The method of claim 19, wherein the ratio of the anti-fungal agent to excipient base powder in the compounded composition is from about 1:60 to about 1:3. 23. The method of claim 19, wherein the method comprises mixing a therapeutically effective amount of one or more additional anti-infective agents with the anti-fungal agent and the excipient base powder. 24. The method of claim 19, wherein the additional anti-infective agent comprises an anti-bacterial agent.
The present application relates to compounded compositions, methods of making compounded compositions, kits comprising compounded compositions, containers comprising compounded compositions, and methods of using compounded compositions. For example, disclosed herein are compounded compositions comprising an anti-fungal agent and methods of using a compounded composition to treat or prevent a fungal infection or a suspected fungal infection. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.1. A method of treating or preventing a fungal infection in a subject, the method comprising: (i) adding to water contained within a foot bath a compounded composition comprising fluconazole and LoxaSperse™ excipient base powder; (ii) agitating the water using an agitator, thereby distributing the compounded composition throughout the water; and (iii) contacting the agitated water with at least a portion of one or both feet of a subject, wherein the subject has been diagnosed with, is suspected of having, or is at risk of developing a fungal infection. 2. A method of treating or preventing a fungal infection, the method comprising: (i) adding to water contained within a foot bath a compounded composition comprising an anti-fungal agent and an excipient base powder comprising a blend of micronized xylitol and poloxamers; (ii) agitating the water contained within the foot bath; and (iii) contacting the agitated water with at least a portion of one or both feet of a subject. 3. The method of claim 2, wherein the subject has been diagnosed with, is suspected of having, or is at risk of developing a fungal infection of one or both feet. 4. The method of claim 2, wherein the excipient base powder is LoxaSperse™ excipient base powder or XyliFos™ excipient base powder. 5. The method of claim 2, wherein the ratio of the anti-fungal agent to excipient base powder in the compounded composition is from about 1:60 to about 1:3. 6. The method of claim 2, wherein the ratio of the anti-fungal agent to excipient base powder in the compounded composition is about 1:15. 7. The method of claim 2, wherein the anti-fungal agent comprises fluconazole. 8. The method of claim 2, wherein the method comprises repeating steps (i)-(iii) daily. 9. The method of claim 2, wherein the method comprises heating the water contained within the foot bath. 10. The method of claim 2, wherein the compounded composition comprises one or more additional anti-infective agents. 11. The method of claim 10, wherein the additional anti-infective agent comprises an anti-bacterial agent. 12. The method of claim 2, wherein the method comprises administering to the subject an oral pharmaceutical composition comprising an anti-infective agent. 13. A compounded composition comprising fluconazole and an excipient base powder comprising a blend of micronized xylitol and poloxamers. 14. The compounded composition of claim 13, wherein the excipient base powder is LoxaSperse™ excipient base powder or XyliFos™ excipient base powder. 15. The compounded composition of claim 13, wherein the ratio of fluconazole to excipient base powder in the compounded composition is from about 1:60 to about 1:3. 16. The compounded composition of claim 13, wherein the ratio of fluconazole to excipient base powder in the compounded composition is about 1:15. 17. The compounded composition of claim 13, wherein the compounded composition comprises one or more additional anti-infective agents. 18. The compounded composition of claim 17, wherein the additional anti-infective agent comprises an anti-bacterial agent. 19. A method of making a compounded composition, the method comprising: obtaining an anti-fungal agent; obtaining an excipient base powder comprising a blend of micronized xylitol and poloxamers; and mixing a therapeutically effective amount of the anti-fungal agent with a sufficient amount of the excipient base powder to make a homogenous compounded composition. 20. The method of claim 19, wherein the excipient base is LoxaSperse™ excipient base powder or XyliFos™ excipient base powder. 21. The method of claim 19, wherein the anti-fungal agent comprises fluconazole. 22. The method of claim 19, wherein the ratio of the anti-fungal agent to excipient base powder in the compounded composition is from about 1:60 to about 1:3. 23. The method of claim 19, wherein the method comprises mixing a therapeutically effective amount of one or more additional anti-infective agents with the anti-fungal agent and the excipient base powder. 24. The method of claim 19, wherein the additional anti-infective agent comprises an anti-bacterial agent.
1,600
527
15,154,499
1,631
Methods, system, and kits are provided for sample identification, and, more specifically, for designing, and/or making, and/or using sample discriminating codes or barcodes for identifying sample nucleic acids or other biomolecules or polymers. For example, a plurality of flowspace codewords may be generated, the codewords comprising a string of characters. A location for at least one padding character within the flowspace codewords may be determined. The padding character may be inserted into the flowspace codewords at the determined location. After the inserting, a plurality of the flowspace codewords may be selected based on satisfying a predetermined minimum distance criteria, wherein the selected codewords correspond to valid base space sequences according to a predetermined flow order. And the barcode sequences corresponding to the selected codewords may be manufactured.
1. A method for designing barcode sequences corresponding to flowspace codewords, comprising: generating a plurality of flowspace codewords, the codewords comprising a string of characters; determining a location for at least one padding character within the flowspace codewords; inserting the padding character into the flowspace codewords at the determined location; selecting, after the inserting, a plurality of the flowspace codewords based on satisfying a predetermined minimum distance criteria, wherein the selected codewords correspond to valid base space sequences according to a predetermined flow order; and manufacturing the barcode sequences corresponding to the selected codewords. 2. The method of claim 1, further comprising after the inserting, filtering at least one codeword that comprises an invalid base space translations according to the predetermined flow order. 3. The method of claim 1, wherein the selected codewords collectively comprise an error tolerant code that meets the predetermined minimum distance criteria. 4. The method of claim 3, wherein determining a location for the padding character within the flowspace codewords further comprises: iterating over a plurality of locations for the padding character within the codewords; for each iteration, calculating a number of codewords that correspond to a valid base space sequence according to the predetermined flow order; and selecting the location of the plurality of locations with the highest calculated number of codewords that correspond to a valid base space sequence. 5. The method of claim 4, wherein determining a location for the padding character within the flowspace codewords further comprises: determining, for each iteration, bases space sequences corresponding to the flowspace codewords that correspond to valid base space sequences after inserting the padding character at the iterated location into the codewords; filtering, for each iteration, the determined base space sequences based on at least a length criteria for the determined sequences; and calculating a number of valid base space sequences for the iterated location after the filtering. 6. The method of claim 5, wherein the filtering, for each iteration, further comprises filtering the determined base space sequences based on a percentage of nucleotide content criteria. 7. The method of claim 4, wherein the codewords of the error tolerant code are synchronized in flowspace after insertion of the at least one padding character. 8. The method of claim 3, wherein the generated flowspace codewords comprise a preliminary distance between the codewords such that the minimum distance between the selected codewords is greater than the minimum distance between the generated codewords. 9. The method of claim 8, wherein the preliminary distance between the codewords is maintained after insertion of the padding character. 10. The method of claim 9, wherein selecting the plurality of codewords further comprises: grouping the codewords such that an inner-group minimum distance between codewords within a group comprises a first value and an outer-group minimum distance for codewords between different groups comprises a second value, the first value being greater than the second value. 11. The method of claim 1, further comprising determining a subset of the selected codewords that comprise a terminating flow that does not indicate incorporation; and manufacturing a subset of barcode sequences corresponding to the subset of selected codewords such that an adaptor for the subset of barcode sequences is selected based on the terminating flow corresponding to the subset of codewords that does not indicate incorporation. 12. The method of claim 1, wherein manufacturing the barcode sequences further comprises appending to the barcode sequences a series of key bases, wherein, for a first portion of the barcode sequences, the appended key bases terminate with a repeated base. 13. The method of claim 12, wherein the first portion comprises half of the barcode sequences. 14. The method of claim 12, wherein, for a second portion of the barcode sequences, the appended key bases terminate with a non-repeating base. 15. The method of claim 14, wherein the selected codewords collectively comprise an error tolerant code that comprises a minimum distance between codewords such that a variance in terminating key bases appended to the selected codewords increases the minimum distance between codewords. 16. A method for sequencing polynucleotide samples comprising barcode sequences, the method comprising: incorporating at least some of a plurality of barcodes into a plurality of target nucleic acids to create polynucleotides, wherein the plurality of barcodes are designed such that the barcodes correspond with a flowspace codeword according to a predetermined flow order, the flowspace codewords comprise one or more error-tolerant codes, and the plurality of barcodes include at least 1000 barcodes; introducing, according to the predetermined flow ordering, a series of nucleotides to the polynucleotides; obtaining a series of signals resulting from the introducing of nucleotides to the target nucleic acids; and resolving the series of signals over the barcode sequences to render flowspace strings such that the rendered flowspace strings are matched to the codewords, wherein at least one rendered flowspace string is matched to at least one codeword in the presence of one or more errors. 17. The method of claim 15, wherein the at least one rendered flowspace string that is matched to at least one flowspace codeword in the presence of one or more errors is used to identify signals obtained over one of the target nucleic acid sequences associated with the barcode corresponding to the matched flowspace codeword. 18. A kit for use with a nucleic acid sequencing instrument, the kit comprising: a plurality of barcodes sequences meeting the following criteria: the barcode sequences correspond to flowspace codewords according to a predetermined flow order such that the corresponding codewords comprise an error-tolerant code of a minimum distance of at least three; the barcode sequences have a length within a predetermined length range; the barcode sequences are synchronized in flow space; and the plurality of barcode sequences is at least 500 different barcode sequences. 19. The kit of claim 16, wherein the plurality of barcode sequences are at least 1000 different barcode sequences.
Methods, system, and kits are provided for sample identification, and, more specifically, for designing, and/or making, and/or using sample discriminating codes or barcodes for identifying sample nucleic acids or other biomolecules or polymers. For example, a plurality of flowspace codewords may be generated, the codewords comprising a string of characters. A location for at least one padding character within the flowspace codewords may be determined. The padding character may be inserted into the flowspace codewords at the determined location. After the inserting, a plurality of the flowspace codewords may be selected based on satisfying a predetermined minimum distance criteria, wherein the selected codewords correspond to valid base space sequences according to a predetermined flow order. And the barcode sequences corresponding to the selected codewords may be manufactured.1. A method for designing barcode sequences corresponding to flowspace codewords, comprising: generating a plurality of flowspace codewords, the codewords comprising a string of characters; determining a location for at least one padding character within the flowspace codewords; inserting the padding character into the flowspace codewords at the determined location; selecting, after the inserting, a plurality of the flowspace codewords based on satisfying a predetermined minimum distance criteria, wherein the selected codewords correspond to valid base space sequences according to a predetermined flow order; and manufacturing the barcode sequences corresponding to the selected codewords. 2. The method of claim 1, further comprising after the inserting, filtering at least one codeword that comprises an invalid base space translations according to the predetermined flow order. 3. The method of claim 1, wherein the selected codewords collectively comprise an error tolerant code that meets the predetermined minimum distance criteria. 4. The method of claim 3, wherein determining a location for the padding character within the flowspace codewords further comprises: iterating over a plurality of locations for the padding character within the codewords; for each iteration, calculating a number of codewords that correspond to a valid base space sequence according to the predetermined flow order; and selecting the location of the plurality of locations with the highest calculated number of codewords that correspond to a valid base space sequence. 5. The method of claim 4, wherein determining a location for the padding character within the flowspace codewords further comprises: determining, for each iteration, bases space sequences corresponding to the flowspace codewords that correspond to valid base space sequences after inserting the padding character at the iterated location into the codewords; filtering, for each iteration, the determined base space sequences based on at least a length criteria for the determined sequences; and calculating a number of valid base space sequences for the iterated location after the filtering. 6. The method of claim 5, wherein the filtering, for each iteration, further comprises filtering the determined base space sequences based on a percentage of nucleotide content criteria. 7. The method of claim 4, wherein the codewords of the error tolerant code are synchronized in flowspace after insertion of the at least one padding character. 8. The method of claim 3, wherein the generated flowspace codewords comprise a preliminary distance between the codewords such that the minimum distance between the selected codewords is greater than the minimum distance between the generated codewords. 9. The method of claim 8, wherein the preliminary distance between the codewords is maintained after insertion of the padding character. 10. The method of claim 9, wherein selecting the plurality of codewords further comprises: grouping the codewords such that an inner-group minimum distance between codewords within a group comprises a first value and an outer-group minimum distance for codewords between different groups comprises a second value, the first value being greater than the second value. 11. The method of claim 1, further comprising determining a subset of the selected codewords that comprise a terminating flow that does not indicate incorporation; and manufacturing a subset of barcode sequences corresponding to the subset of selected codewords such that an adaptor for the subset of barcode sequences is selected based on the terminating flow corresponding to the subset of codewords that does not indicate incorporation. 12. The method of claim 1, wherein manufacturing the barcode sequences further comprises appending to the barcode sequences a series of key bases, wherein, for a first portion of the barcode sequences, the appended key bases terminate with a repeated base. 13. The method of claim 12, wherein the first portion comprises half of the barcode sequences. 14. The method of claim 12, wherein, for a second portion of the barcode sequences, the appended key bases terminate with a non-repeating base. 15. The method of claim 14, wherein the selected codewords collectively comprise an error tolerant code that comprises a minimum distance between codewords such that a variance in terminating key bases appended to the selected codewords increases the minimum distance between codewords. 16. A method for sequencing polynucleotide samples comprising barcode sequences, the method comprising: incorporating at least some of a plurality of barcodes into a plurality of target nucleic acids to create polynucleotides, wherein the plurality of barcodes are designed such that the barcodes correspond with a flowspace codeword according to a predetermined flow order, the flowspace codewords comprise one or more error-tolerant codes, and the plurality of barcodes include at least 1000 barcodes; introducing, according to the predetermined flow ordering, a series of nucleotides to the polynucleotides; obtaining a series of signals resulting from the introducing of nucleotides to the target nucleic acids; and resolving the series of signals over the barcode sequences to render flowspace strings such that the rendered flowspace strings are matched to the codewords, wherein at least one rendered flowspace string is matched to at least one codeword in the presence of one or more errors. 17. The method of claim 15, wherein the at least one rendered flowspace string that is matched to at least one flowspace codeword in the presence of one or more errors is used to identify signals obtained over one of the target nucleic acid sequences associated with the barcode corresponding to the matched flowspace codeword. 18. A kit for use with a nucleic acid sequencing instrument, the kit comprising: a plurality of barcodes sequences meeting the following criteria: the barcode sequences correspond to flowspace codewords according to a predetermined flow order such that the corresponding codewords comprise an error-tolerant code of a minimum distance of at least three; the barcode sequences have a length within a predetermined length range; the barcode sequences are synchronized in flow space; and the plurality of barcode sequences is at least 500 different barcode sequences. 19. The kit of claim 16, wherein the plurality of barcode sequences are at least 1000 different barcode sequences.
1,600
528
15,447,019
1,633
The invention provides polynucleotides and methods for expressing light-activated proteins in animal cells and altering an action potential of the cells by optical stimulation. The invention also provides animal cells and non-human animals comprising cells expressing the light-activated proteins.
1.-77. (canceled) 78. A light-activated protein comprising: a) a core polypeptide comprising an amino acid sequence at least 80% identical to the sequence shown in SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:4; b) an endoplasmic reticulum (ER) export signal; and c) a membrane trafficking signal. 79. The light-activated protein of claim 78, wherein the core polypeptide comprises an amino acid sequence at least 85% identical to the sequence shown in SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:4. 80. The light-activated protein of claim 78, wherein the core polypeptide comprises an amino acid sequence at least 90% identical to the sequence shown in SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:4. 81. The light-activated protein of claim 78, wherein the ER export signal comprises the amino acid sequence FCEYENEV (SEQ ID NO:12). 82. The light-activated protein of claim 78, wherein the membrane trafficking signal comprises the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:11). 83. The light-activated protein of claim 78, wherein the core amino acid sequence is at least 95% identical to amino acids 25-265 of SEQ ID NO:2. 84. The light-activated protein of claim 78, wherein the core amino acid sequence is at least 95% identical to SEQ ID NO:3. 85. The light-activated protein of claim 78, wherein the core amino acid sequence is at least 95% identical to SEQ ID NO:4. 86. An isolated polynucleotide comprising a nucleotide sequence encoding the light-activated protein of claim 78. 87. The polynucleotide of claim 86, wherein the light-activated protein-encoding nucleotide sequence is operably linked to a promoter. 88. The polynucleotide of claim 86, wherein the promoter is a CaMKIIa promoter. 89. The polynucleotide of claim 86, wherein the promoter is a synapsin I promoter. 90. The polynucleotide of claim 86, wherein the polynucleotide is in an expression vector. 91. The polynucleotide of claim 90, wherein the expression vector is a viral vector. 92. The polynucleotide of claim 91, wherein the viral vector is an adenoassociated virus vector, a retroviral vector, an adenoviral vector, a herpes simplex virus vector, or a lentiviral vector. 93. A system comprising: a) a delivery device comprising a polynucleotide that comprises a nucleotide sequence encoding the light-activated polypeptide of claim 78; b) a light source; and c) a control device that controls generation of light by the light source. 94. An animal cell comprising the light-activated polypeptide of claim 78 on its cell membrane. 95. A method of modulating an activity of a cell that expresses on its membrane the light-activated polypeptide of claim 78, the method comprising activating the light-activated polypeptide with light, wherein said activating modulates the activity of the cell.
The invention provides polynucleotides and methods for expressing light-activated proteins in animal cells and altering an action potential of the cells by optical stimulation. The invention also provides animal cells and non-human animals comprising cells expressing the light-activated proteins.1.-77. (canceled) 78. A light-activated protein comprising: a) a core polypeptide comprising an amino acid sequence at least 80% identical to the sequence shown in SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:4; b) an endoplasmic reticulum (ER) export signal; and c) a membrane trafficking signal. 79. The light-activated protein of claim 78, wherein the core polypeptide comprises an amino acid sequence at least 85% identical to the sequence shown in SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:4. 80. The light-activated protein of claim 78, wherein the core polypeptide comprises an amino acid sequence at least 90% identical to the sequence shown in SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:4. 81. The light-activated protein of claim 78, wherein the ER export signal comprises the amino acid sequence FCEYENEV (SEQ ID NO:12). 82. The light-activated protein of claim 78, wherein the membrane trafficking signal comprises the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:11). 83. The light-activated protein of claim 78, wherein the core amino acid sequence is at least 95% identical to amino acids 25-265 of SEQ ID NO:2. 84. The light-activated protein of claim 78, wherein the core amino acid sequence is at least 95% identical to SEQ ID NO:3. 85. The light-activated protein of claim 78, wherein the core amino acid sequence is at least 95% identical to SEQ ID NO:4. 86. An isolated polynucleotide comprising a nucleotide sequence encoding the light-activated protein of claim 78. 87. The polynucleotide of claim 86, wherein the light-activated protein-encoding nucleotide sequence is operably linked to a promoter. 88. The polynucleotide of claim 86, wherein the promoter is a CaMKIIa promoter. 89. The polynucleotide of claim 86, wherein the promoter is a synapsin I promoter. 90. The polynucleotide of claim 86, wherein the polynucleotide is in an expression vector. 91. The polynucleotide of claim 90, wherein the expression vector is a viral vector. 92. The polynucleotide of claim 91, wherein the viral vector is an adenoassociated virus vector, a retroviral vector, an adenoviral vector, a herpes simplex virus vector, or a lentiviral vector. 93. A system comprising: a) a delivery device comprising a polynucleotide that comprises a nucleotide sequence encoding the light-activated polypeptide of claim 78; b) a light source; and c) a control device that controls generation of light by the light source. 94. An animal cell comprising the light-activated polypeptide of claim 78 on its cell membrane. 95. A method of modulating an activity of a cell that expresses on its membrane the light-activated polypeptide of claim 78, the method comprising activating the light-activated polypeptide with light, wherein said activating modulates the activity of the cell.
1,600
529
15,597,676
1,612
Disclosed herein are oral care compositions comprising hemp seed oil and caprylyl glycol. In certain embodiments, the oral care composition optionally further comprises one or more ingredients selected from hyaluronic acid and aloe vera. Methods of making and using the compositions are also provided.
1. A mouthrinse comprising: hemp seed oil; caprylyl glycol; and an orally acceptable base. 2. The mouthrinse of claim 1, wherein said composition has a moisture retention capacity of at least 4.8, as measured in an n vitro moisture retention capacity assay. 3. The mouthrinse of claim 2, wherein said hemp seed oil is present at a concentration from 0.8 to 1.2%, by weight of the composition. 4. The mouthrinse of claim 3, wherein said hemp seed oil is present at a concentration of 1.0% by weight of the composition. 5. The mouthrinse of claim 3, wherein said caprylyl glycol is present at a concentration of 0.15 to 0.35%, by weight of the composition. 6. The mouthrinse of claim 5, wherein said caprylyl glycol is present at a concentration of 0.25%, by weight of the composition. 7. The mouthrinse of claim 5, further comprising aloe vera. 8. The mouthrinse of claim 7, wherein said aloe vera is present at a concentration of 0.15 to 0.35%, by weight of the composition. 9. The mouthrinse of claim 8, wherein said aloe vera is present at a concentration of 0.25%, by weight of the composition. 10. The mouthrinse of claim 5, further comprising hyaluronic acid. 11. The mouthrinse of claim 10, wherein said hyaluronic acid is present at a concentration of 0.01 to 0.1%, by weight of the composition. 12. The mouthrinse of claim 11, wherein said hyaluronic acid is present at a concentration of 0.05%, by weight of the composition. 13. A mouthrinse comprising a) hemp seed oil at a concentration from 0.8 to 1.2%, by weight of the composition; b) caprylyl glycol at a concentration of 0.15 to 0.35% by weight of the composition; c) optionally aloe vera at a concentration of 0.15 to 0.35%, by weight of the composition; and d) optionally hyaluronic acid at a concentration of 0.01 to 0.1%, by weight of the composition. 14. The mouthrinse of claim 13, having a moisture retention capacity of at least 4.8, as measured in an in vitro moisture retention capacity assay. 15. A method to improve oral health comprising applying an effective amount of the oral composition of any of the preceding claims set forth above to the oral cavity of a subject in need thereof. 16. The method of claim 15, wherein improving oral health may be selected from one or more of the following: a. reduce or inhibit formation of dental caries; b. reduce, repair or inhibit early enamel lesions; c. reduce or inhibit demineralization and promote remineralization of the teeth; d. reduce hypersensitivity of the teeth; e. reduce or inhibit gingivitis; f. promote healing of sores or cuts in the mouth; g. reduce levels of acid producing bacteria; h. increase relative levels of arginolytic bacteria; i. inhibit microbial biofilm formation in the oral cavity; j. raise and/or maintain plaque pH at levels of at least pH 5.5 following sugar challenge; k. reduce plaque accumulation; l. treat, relieve or reduce dry mouth; m. whiten teeth; n. enhance systemic health, including cardiovascular health; o. reduce erosion of the teeth; p. immunize the teeth against cariogenic bacteria and their effects; q. clean the teeth and oral cavity; r. reduce inflammation; s. increase anti-oxidant levels; t. reduce oral discomfort; u. increase lubrication; v. reduce tissue friction due to drying; and w. increase tissue hydration.
Disclosed herein are oral care compositions comprising hemp seed oil and caprylyl glycol. In certain embodiments, the oral care composition optionally further comprises one or more ingredients selected from hyaluronic acid and aloe vera. Methods of making and using the compositions are also provided.1. A mouthrinse comprising: hemp seed oil; caprylyl glycol; and an orally acceptable base. 2. The mouthrinse of claim 1, wherein said composition has a moisture retention capacity of at least 4.8, as measured in an n vitro moisture retention capacity assay. 3. The mouthrinse of claim 2, wherein said hemp seed oil is present at a concentration from 0.8 to 1.2%, by weight of the composition. 4. The mouthrinse of claim 3, wherein said hemp seed oil is present at a concentration of 1.0% by weight of the composition. 5. The mouthrinse of claim 3, wherein said caprylyl glycol is present at a concentration of 0.15 to 0.35%, by weight of the composition. 6. The mouthrinse of claim 5, wherein said caprylyl glycol is present at a concentration of 0.25%, by weight of the composition. 7. The mouthrinse of claim 5, further comprising aloe vera. 8. The mouthrinse of claim 7, wherein said aloe vera is present at a concentration of 0.15 to 0.35%, by weight of the composition. 9. The mouthrinse of claim 8, wherein said aloe vera is present at a concentration of 0.25%, by weight of the composition. 10. The mouthrinse of claim 5, further comprising hyaluronic acid. 11. The mouthrinse of claim 10, wherein said hyaluronic acid is present at a concentration of 0.01 to 0.1%, by weight of the composition. 12. The mouthrinse of claim 11, wherein said hyaluronic acid is present at a concentration of 0.05%, by weight of the composition. 13. A mouthrinse comprising a) hemp seed oil at a concentration from 0.8 to 1.2%, by weight of the composition; b) caprylyl glycol at a concentration of 0.15 to 0.35% by weight of the composition; c) optionally aloe vera at a concentration of 0.15 to 0.35%, by weight of the composition; and d) optionally hyaluronic acid at a concentration of 0.01 to 0.1%, by weight of the composition. 14. The mouthrinse of claim 13, having a moisture retention capacity of at least 4.8, as measured in an in vitro moisture retention capacity assay. 15. A method to improve oral health comprising applying an effective amount of the oral composition of any of the preceding claims set forth above to the oral cavity of a subject in need thereof. 16. The method of claim 15, wherein improving oral health may be selected from one or more of the following: a. reduce or inhibit formation of dental caries; b. reduce, repair or inhibit early enamel lesions; c. reduce or inhibit demineralization and promote remineralization of the teeth; d. reduce hypersensitivity of the teeth; e. reduce or inhibit gingivitis; f. promote healing of sores or cuts in the mouth; g. reduce levels of acid producing bacteria; h. increase relative levels of arginolytic bacteria; i. inhibit microbial biofilm formation in the oral cavity; j. raise and/or maintain plaque pH at levels of at least pH 5.5 following sugar challenge; k. reduce plaque accumulation; l. treat, relieve or reduce dry mouth; m. whiten teeth; n. enhance systemic health, including cardiovascular health; o. reduce erosion of the teeth; p. immunize the teeth against cariogenic bacteria and their effects; q. clean the teeth and oral cavity; r. reduce inflammation; s. increase anti-oxidant levels; t. reduce oral discomfort; u. increase lubrication; v. reduce tissue friction due to drying; and w. increase tissue hydration.
1,600
530
16,211,281
1,649
Methods and compositions are disclosed for enhancing neurogenesis, resolving neuropathy and improving neurological health and functioning using fungal extracts and their active ingredients, including species of mushrooms and mycelia containing psilocybin and psilocin, combined with erinacines and hericenones or fungal extracts containing those active ingredients, with the addition of nicotinic acid. The compositions may optionally be combined with nervine plants.
1. A composition comprising: psilocybin, psilocin, baeocystin, norbaeocystin, or salts thereof, psilocybin mushrooms or extracts thereof, or combinations thereof; and niacin. 2. The composition of claim 1, wherein the composition further comprises Bacopa species (Bacopa monnieri), Gotu kola (Centella asiatica), Gingko (Gingko biloba), Ginger (Zingiber officinale), Holy Basil (Ocimum sanctum), Hu Zhang (Polygonum cuspid atum), Oregano (Origanum vulgare, Origanum onites), Rosemary (Rosmarinus officinalis, Rosmarinus eriocalyx, Rosmarinus species), Turmeric (Curcuma longa), Green Tea (Camellia sinensis), lavender (Lavandula spica and Lavandula species), skullcap (Scutellaria lateriflora), oat straw (Avena sativa and Avena byzantine), Diviner's Sage (Salvia divinorum), ayahuasca (Banisteriopsis caapi and Psychotria species), Tabemanthe iboga, Voacanga africana, Tabemaemontana undulate, peyote (Lophophora williamsil), morning glory (Ipomoea tricolor, Argyreia nervosa), Cannabis sativa, Cannabis indica or Cannabis ruderalis, extracts thereof, or combinations thereof. 3. The composition of claim 1, wherein the composition further comprises: mycelia, fruitbodies, or extracts thereof of Antrodia, Beauveria, Copelandia, Cordyceps, Ganoderma, Grifola, Hericium, Inonotus, Isaria, Panaeolus, Phellinus or combinations thereof. 4. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 1 to a subject in need thereof. 5. The method of claim 4 where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof. 6. A composition comprising: psilocybin, psilocin, baeocystin, norbaeocystin, or salts thereof, psilocybin mushrooms or extracts thereof, or combinations thereof; Cannabis extracts comprising cannabidiol, tetrahydrocannabinol, or combinations thereof; and niacin. 7. The composition of claim 6, wherein the composition further comprises Bacopa species (Bacopa monnieri), Gotu kola (Centella asiatica), Gingko (Gingko biloba), Ginger (Zingiber officinale), Holy Basil (Ocimum sanctum), Hu Zhang (Polygonum cuspid atum), Oregano (Origanum vulgate, Origanum onites), Rosemary (Rosmarinus officinalis, Rosmarinus eriocalyx, Rosmarinus species), Turmeric (Curcuma longa), Green Tea (Camellia sinensis), lavender (Lavandula spica and Lavandula species), skullcap (Scutellaria lateriflora), oat straw (Avena sativa and Avena byzantine), Diviner's Sage (Salvia divinorum), ayahuasca (Banisteriopsis caapi and Psychotria species), Tabemanthe iboga, Voacanga africana, Tabemaemontana undulate, peyote (Lophophora williamsil), morning glory (Ipomoea tricolor, Argyreia nervosa), Cannabis sativa, Cannabis indica or Cannabis ruderalis, extracts thereof, or combinations thereof. 8. The composition of claim 6, wherein the composition further comprises: mycelia, fruitbodies, or extracts thereof of Antrodia, Beauveria, Copelandia, Cordyceps, Ganoderma, Grifola, Hericium, Inonotus, Isaria, Panaeolus, Phellinus or combinations thereof. 9. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 6 to a subject in need thereof. 10. The method of claim 9 where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof. 11. A composition comprising: psilocybin, psilocin, baeocystin, norbaeocystin, or salts thereof, psilocybin mushrooms or extracts thereof, or combinations thereof; and Cannabis extracts comprising cannabidiol, tetrahydrocannabinol, or combinations thereof. 12. The composition of claim 11, wherein the composition further comprises Bacopa species (Bacopa monnieri), Gotu kola (Centella asiatica), Gingko (Gingko biloba), Ginger (Zingiber officinale), Holy Basil (Ocimum sanctum), Hu Zhang (Polygonum cuspid atum), Oregano (Origanum vulgare, Origanum onites), Rosemary (Rosmarinus officinalis, Rosmarinus eriocalyx, Rosmarinus species), Turmeric (Curcuma longa), Green Tea (Camellia sinensis), lavender (Lavandula spica and Lavandula species), skullcap (Scutellaria lateriflora), oat straw (Avena sativa and Avena byzantine), Diviner's Sage (Salvia divinorum), ayahuasca (Banisteriopsis caapi and Psychotria species), Tabemanthe iboga, Voacanga africana, Tabemaemontana undulate, peyote (Lophophora williamsil), morning glory (Ipomoea tricolor, Argyreia nervosa), Cannabis sativa, Cannabis indica or Cannabis ruderalis, extracts thereof, or combinations thereof. 13. The composition of claim 11, wherein the composition further comprises: mycelia, fruitbodies, or extracts thereof of Antrodia, Beauveria, Copelandia, Cordyceps, Ganoderma, Grifola, Hericium, Inonotus, Isaria, Panaeolus, Phellinus or combinations thereof. 14. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 11 to a subject in need thereof. 15. The method of claim 14 where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof. 16. A composition comprising: mycelia, fruitbodies, or extracts thereof of Cordyceps, Ganoderma, Grifola, Hericium, or combinations thereof; and Cannabis extracts comprising cannabidiol, tetrahydrocannabinol, or combinations thereof. 17. The composition of claim 16, further comprising niacin. 18. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 16 to a subject in need thereof. 19. The method of claim 18, where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof.
Methods and compositions are disclosed for enhancing neurogenesis, resolving neuropathy and improving neurological health and functioning using fungal extracts and their active ingredients, including species of mushrooms and mycelia containing psilocybin and psilocin, combined with erinacines and hericenones or fungal extracts containing those active ingredients, with the addition of nicotinic acid. The compositions may optionally be combined with nervine plants.1. A composition comprising: psilocybin, psilocin, baeocystin, norbaeocystin, or salts thereof, psilocybin mushrooms or extracts thereof, or combinations thereof; and niacin. 2. The composition of claim 1, wherein the composition further comprises Bacopa species (Bacopa monnieri), Gotu kola (Centella asiatica), Gingko (Gingko biloba), Ginger (Zingiber officinale), Holy Basil (Ocimum sanctum), Hu Zhang (Polygonum cuspid atum), Oregano (Origanum vulgare, Origanum onites), Rosemary (Rosmarinus officinalis, Rosmarinus eriocalyx, Rosmarinus species), Turmeric (Curcuma longa), Green Tea (Camellia sinensis), lavender (Lavandula spica and Lavandula species), skullcap (Scutellaria lateriflora), oat straw (Avena sativa and Avena byzantine), Diviner's Sage (Salvia divinorum), ayahuasca (Banisteriopsis caapi and Psychotria species), Tabemanthe iboga, Voacanga africana, Tabemaemontana undulate, peyote (Lophophora williamsil), morning glory (Ipomoea tricolor, Argyreia nervosa), Cannabis sativa, Cannabis indica or Cannabis ruderalis, extracts thereof, or combinations thereof. 3. The composition of claim 1, wherein the composition further comprises: mycelia, fruitbodies, or extracts thereof of Antrodia, Beauveria, Copelandia, Cordyceps, Ganoderma, Grifola, Hericium, Inonotus, Isaria, Panaeolus, Phellinus or combinations thereof. 4. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 1 to a subject in need thereof. 5. The method of claim 4 where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof. 6. A composition comprising: psilocybin, psilocin, baeocystin, norbaeocystin, or salts thereof, psilocybin mushrooms or extracts thereof, or combinations thereof; Cannabis extracts comprising cannabidiol, tetrahydrocannabinol, or combinations thereof; and niacin. 7. The composition of claim 6, wherein the composition further comprises Bacopa species (Bacopa monnieri), Gotu kola (Centella asiatica), Gingko (Gingko biloba), Ginger (Zingiber officinale), Holy Basil (Ocimum sanctum), Hu Zhang (Polygonum cuspid atum), Oregano (Origanum vulgate, Origanum onites), Rosemary (Rosmarinus officinalis, Rosmarinus eriocalyx, Rosmarinus species), Turmeric (Curcuma longa), Green Tea (Camellia sinensis), lavender (Lavandula spica and Lavandula species), skullcap (Scutellaria lateriflora), oat straw (Avena sativa and Avena byzantine), Diviner's Sage (Salvia divinorum), ayahuasca (Banisteriopsis caapi and Psychotria species), Tabemanthe iboga, Voacanga africana, Tabemaemontana undulate, peyote (Lophophora williamsil), morning glory (Ipomoea tricolor, Argyreia nervosa), Cannabis sativa, Cannabis indica or Cannabis ruderalis, extracts thereof, or combinations thereof. 8. The composition of claim 6, wherein the composition further comprises: mycelia, fruitbodies, or extracts thereof of Antrodia, Beauveria, Copelandia, Cordyceps, Ganoderma, Grifola, Hericium, Inonotus, Isaria, Panaeolus, Phellinus or combinations thereof. 9. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 6 to a subject in need thereof. 10. The method of claim 9 where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof. 11. A composition comprising: psilocybin, psilocin, baeocystin, norbaeocystin, or salts thereof, psilocybin mushrooms or extracts thereof, or combinations thereof; and Cannabis extracts comprising cannabidiol, tetrahydrocannabinol, or combinations thereof. 12. The composition of claim 11, wherein the composition further comprises Bacopa species (Bacopa monnieri), Gotu kola (Centella asiatica), Gingko (Gingko biloba), Ginger (Zingiber officinale), Holy Basil (Ocimum sanctum), Hu Zhang (Polygonum cuspid atum), Oregano (Origanum vulgare, Origanum onites), Rosemary (Rosmarinus officinalis, Rosmarinus eriocalyx, Rosmarinus species), Turmeric (Curcuma longa), Green Tea (Camellia sinensis), lavender (Lavandula spica and Lavandula species), skullcap (Scutellaria lateriflora), oat straw (Avena sativa and Avena byzantine), Diviner's Sage (Salvia divinorum), ayahuasca (Banisteriopsis caapi and Psychotria species), Tabemanthe iboga, Voacanga africana, Tabemaemontana undulate, peyote (Lophophora williamsil), morning glory (Ipomoea tricolor, Argyreia nervosa), Cannabis sativa, Cannabis indica or Cannabis ruderalis, extracts thereof, or combinations thereof. 13. The composition of claim 11, wherein the composition further comprises: mycelia, fruitbodies, or extracts thereof of Antrodia, Beauveria, Copelandia, Cordyceps, Ganoderma, Grifola, Hericium, Inonotus, Isaria, Panaeolus, Phellinus or combinations thereof. 14. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 11 to a subject in need thereof. 15. The method of claim 14 where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof. 16. A composition comprising: mycelia, fruitbodies, or extracts thereof of Cordyceps, Ganoderma, Grifola, Hericium, or combinations thereof; and Cannabis extracts comprising cannabidiol, tetrahydrocannabinol, or combinations thereof. 17. The composition of claim 16, further comprising niacin. 18. A method for treating or improving neurological or mental health conditions comprising administration of an effective amount of the composition of claim 16 to a subject in need thereof. 19. The method of claim 18, where the neurological or mental health conditions comprise depression, memory loss, dementia, cognitive dysfunction, hearing loss, vision loss, neurologic pain, or combinations thereof.
1,600
531
12,758,315
1,645
A composition is provided comprising N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one antigenic component. The composition is suitable for use in vaccines and for treatment of infection, particularly meningococcal infection, demonstrating a broad spectrum of protection. A number of preferred antigenic components are described and include antigenic proteins and proteoglycans derived from N. meningitidis.
1. A composition comprising N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one antigenic component. 2. The composition of claim 1, wherein said antigenic component is an N. meningitidis antigenic protein. 3. The composition of claim 2, wherein said outer membrane vesicles are from a first strain of N. meningitidis and said antigenic component is from a second strain of N. meningitidis different from the first. 4. The composition of claim 1, wherein said outer membrane vesicles are enriched with a plurality of antigenic components from different strains of N. meningitidis. 5. The composition of claim 1, wherein said outer membrane vesicles comprise a mixture of outer membrane vesicles from different strains of N. meningitidis. 6. The composition of claim 1, wherein said antigenic component is an N. meningitidis antigenic proteoglycan. 7. The composition of claim 1, wherein said antigenic component is an N. meningitidis protein selected from the group consisting of a surface antigen, a periplasmic protein, a superoxide dismutase, and a glycoprotein. 8. The composition of claim 1, wherein said antigenic component is selected from the group consisting of Cu,Zn-superoxide dismutase; neisserial surface protein A (NspA); porA; OMP85; FrpB; PilQ; Hsf; HemK; sodC; mafA; N-acetyl glutamate synthetase; and macrophage infectivity potentiator-related protein. 9. The composition of claim 1, wherein said antigenic component is a peptide selected from the group consisting of SEQ ID NOS: 2; 4; 6; 8; 10; 12; 14; 16; 18; 20; 22; 24; 26; 28; 30; 32; 34; 36; 38; 40; 42; 44; 46; 48; 50; 52; 54; 56; 58; 60; 62; 64; 66; 68; 70; and 72. 10. The composition of claim 2, wherein said outer membrane vesicles are from a first strain of N. meningitidis and said antigenic component is from a second strain of N. meningitidis different from the first, further comprising a pharmaceutically acceptable carrier. 11. A vaccine composition comprising outer membrane vesicles from a first strain of N. meningitidis, as well as an antigenic component from a second strain of N. meningitidis different from the first, and a pharmaceutically acceptable carrier. 12. The vaccine composition of claim 11, wherein the antigenic component is selected from the group consisting of Cu,Zn-superoxide dismutase; neisserial surface protein A (NspA); porA; OMP85; FrpB; PilQ; Hsf; HemK; sodC; mafA; N-acetyl glutamate synthetase; and macrophage infectivity potentiator-related protein. 13. (canceled) 14. A method of manufacture of a composition, comprising: (a) extracting an antigenic component from an outer membrane of a bacteria, and preparing an aqueous solution of said antigenic component; (b) extracting outer membrane vesicles from a culture of N. meningitidis, and preparing an aqueous solution of said outer membrane vesicles; (c) obtaining a pharmaceutically acceptable carrier; and (d) admixing the solution prepared in (a), the solution prepared in (b) and the carrier obtained in (c). 15.-21. (canceled) 22. A method of manufacture of a composition, comprising: (a) recombinantly expressing a DNA that encodes an antigenic component in a bacteria; (b) extracting said antigenic component from the outer membrane of said bacteria, and preparing an aqueous solution of said antigenic component; (c) extracting outer membrane vesicles from a culture of N. meningitidis, and preparing an aqueous solution of said outer membrane vesicles; (d) obtaining a pharmaceutically acceptable carrier; and admixing the solution prepared in (b), the solution prepared in (c) and the carrier obtained in (d). 23. (canceled) 24. A method of manufacture of a composition, comprising: (a) recombinantly expressing a DNA that encodes an antigenic component in N. meningitidis; (b) extracting outer membrane vesicles from said N. meningitidis, and preparing an aqueous solution of said outer membrane vesicles, wherein said outer membrane vesicles comprise said antigenic component; (c) obtaining a pharmaceutically acceptable carrier; and (d) admixing the solution prepared in (b), with the carrier obtained in (c). 25.-27. (canceled) 28. A method of preventing N. meningitidis infection in an animal, comprising administering an effective dose of a composition comprising an outer membrane vesicle and an antigenic component selected from the group consisting of Cu,Zn-superoxide dismutase; neisserial surface protein A (NspA); porA; OMP85; FrpB; PiIQ; Hsf; HemK; sodC; mafA; N-acetyl glutamate synthetase; and macrophage infectivity potentiator-related protein, and a pharmaceutically acceptable carrier. 29.-31. (canceled) 32. A composition comprising isolated N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one isolated N. meningitidis antigenic protein, wherein the composition is obtained by a process comprising isolating said at least one N. meningitidis antigenic protein and combining the isolated outer membrane vesicles with the at least one isolated N. meningitidis antigenic protein, and said N. meningitidis antigenic protein is an N. meningitidis antigenic proteoglycan. 33. A composition comprising isolated N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one isolated N. meningitidis antigenic protein, wherein the composition is obtained by a process comprising isolating said at least one N. meningitidis antigenic protein and combining the isolated outer membrane vesicles with the at least one isolated N. meningitidis antigenic protein, and the composition further comprises Haemophilus surface fibril (Hsf). 34. A vaccine composition comprising isolated outer membrane vesicles from a first strain of N. meningitidis, an isolated N. meningitidis antigenic protein from a second strain of N. meningitidis different from the first, Haemophilus surface fibril (Hsf), and a pharmaceutically acceptable carrier. 35. A composition comprising N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with porA.
A composition is provided comprising N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one antigenic component. The composition is suitable for use in vaccines and for treatment of infection, particularly meningococcal infection, demonstrating a broad spectrum of protection. A number of preferred antigenic components are described and include antigenic proteins and proteoglycans derived from N. meningitidis.1. A composition comprising N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one antigenic component. 2. The composition of claim 1, wherein said antigenic component is an N. meningitidis antigenic protein. 3. The composition of claim 2, wherein said outer membrane vesicles are from a first strain of N. meningitidis and said antigenic component is from a second strain of N. meningitidis different from the first. 4. The composition of claim 1, wherein said outer membrane vesicles are enriched with a plurality of antigenic components from different strains of N. meningitidis. 5. The composition of claim 1, wherein said outer membrane vesicles comprise a mixture of outer membrane vesicles from different strains of N. meningitidis. 6. The composition of claim 1, wherein said antigenic component is an N. meningitidis antigenic proteoglycan. 7. The composition of claim 1, wherein said antigenic component is an N. meningitidis protein selected from the group consisting of a surface antigen, a periplasmic protein, a superoxide dismutase, and a glycoprotein. 8. The composition of claim 1, wherein said antigenic component is selected from the group consisting of Cu,Zn-superoxide dismutase; neisserial surface protein A (NspA); porA; OMP85; FrpB; PilQ; Hsf; HemK; sodC; mafA; N-acetyl glutamate synthetase; and macrophage infectivity potentiator-related protein. 9. The composition of claim 1, wherein said antigenic component is a peptide selected from the group consisting of SEQ ID NOS: 2; 4; 6; 8; 10; 12; 14; 16; 18; 20; 22; 24; 26; 28; 30; 32; 34; 36; 38; 40; 42; 44; 46; 48; 50; 52; 54; 56; 58; 60; 62; 64; 66; 68; 70; and 72. 10. The composition of claim 2, wherein said outer membrane vesicles are from a first strain of N. meningitidis and said antigenic component is from a second strain of N. meningitidis different from the first, further comprising a pharmaceutically acceptable carrier. 11. A vaccine composition comprising outer membrane vesicles from a first strain of N. meningitidis, as well as an antigenic component from a second strain of N. meningitidis different from the first, and a pharmaceutically acceptable carrier. 12. The vaccine composition of claim 11, wherein the antigenic component is selected from the group consisting of Cu,Zn-superoxide dismutase; neisserial surface protein A (NspA); porA; OMP85; FrpB; PilQ; Hsf; HemK; sodC; mafA; N-acetyl glutamate synthetase; and macrophage infectivity potentiator-related protein. 13. (canceled) 14. A method of manufacture of a composition, comprising: (a) extracting an antigenic component from an outer membrane of a bacteria, and preparing an aqueous solution of said antigenic component; (b) extracting outer membrane vesicles from a culture of N. meningitidis, and preparing an aqueous solution of said outer membrane vesicles; (c) obtaining a pharmaceutically acceptable carrier; and (d) admixing the solution prepared in (a), the solution prepared in (b) and the carrier obtained in (c). 15.-21. (canceled) 22. A method of manufacture of a composition, comprising: (a) recombinantly expressing a DNA that encodes an antigenic component in a bacteria; (b) extracting said antigenic component from the outer membrane of said bacteria, and preparing an aqueous solution of said antigenic component; (c) extracting outer membrane vesicles from a culture of N. meningitidis, and preparing an aqueous solution of said outer membrane vesicles; (d) obtaining a pharmaceutically acceptable carrier; and admixing the solution prepared in (b), the solution prepared in (c) and the carrier obtained in (d). 23. (canceled) 24. A method of manufacture of a composition, comprising: (a) recombinantly expressing a DNA that encodes an antigenic component in N. meningitidis; (b) extracting outer membrane vesicles from said N. meningitidis, and preparing an aqueous solution of said outer membrane vesicles, wherein said outer membrane vesicles comprise said antigenic component; (c) obtaining a pharmaceutically acceptable carrier; and (d) admixing the solution prepared in (b), with the carrier obtained in (c). 25.-27. (canceled) 28. A method of preventing N. meningitidis infection in an animal, comprising administering an effective dose of a composition comprising an outer membrane vesicle and an antigenic component selected from the group consisting of Cu,Zn-superoxide dismutase; neisserial surface protein A (NspA); porA; OMP85; FrpB; PiIQ; Hsf; HemK; sodC; mafA; N-acetyl glutamate synthetase; and macrophage infectivity potentiator-related protein, and a pharmaceutically acceptable carrier. 29.-31. (canceled) 32. A composition comprising isolated N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one isolated N. meningitidis antigenic protein, wherein the composition is obtained by a process comprising isolating said at least one N. meningitidis antigenic protein and combining the isolated outer membrane vesicles with the at least one isolated N. meningitidis antigenic protein, and said N. meningitidis antigenic protein is an N. meningitidis antigenic proteoglycan. 33. A composition comprising isolated N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with at least one isolated N. meningitidis antigenic protein, wherein the composition is obtained by a process comprising isolating said at least one N. meningitidis antigenic protein and combining the isolated outer membrane vesicles with the at least one isolated N. meningitidis antigenic protein, and the composition further comprises Haemophilus surface fibril (Hsf). 34. A vaccine composition comprising isolated outer membrane vesicles from a first strain of N. meningitidis, an isolated N. meningitidis antigenic protein from a second strain of N. meningitidis different from the first, Haemophilus surface fibril (Hsf), and a pharmaceutically acceptable carrier. 35. A composition comprising N. meningitidis outer membrane vesicles, wherein said outer membrane vesicles are enriched with porA.
1,600
532
12,675,614
1,634
The invention relates to the prediction of a sleep parameter (e.g., sleep efficiency (SE), latency to persistent sleep (LPS), wake after sleep onset (WASO), total sleep time (TST)) of an individual and the response of such an individual to a sleep inducing compound based on the individual's PER3 variable number tandem repeat (VNTR) genotype.
1. A method of predicting a sleep parameter of an individual, the method comprising: determining the individual's genotype at the PER3 variable number tandem repeat (VNTR) locus; and in the case that the individual's PER3 VNTR genotype is 5/5, predicting that the sleep parameter of the individual is indicative of better sleep than an individual whose PER3 VNTR genotype is 4/4 or 4/5. 2. The method of claim 1, wherein the sleep parameter is selected from a group consisting of: sleep efficiency (SE), total sleep time (TST), latency to persistent sleep (LPS), and wake after sleep onset (WASO). 3. The method of claim 1, wherein the individual's genotype is determined ex vivo. 4. The method of claim 1, wherein the individual is suffering from a sleep disorder. 5. The method of claim 4, wherein the sleep disorder is insomnia or insomnia is a symptom of another disorder. 6. The method of claim 5, wherein the insomnia is selected from a group consisting of: chronic insomnia and transient insomnia. 7. The method of claim 1, further comprising: in the case that the individual's PER3 VNTR genotype is 5/5, predicting that the individual will be less responsive to a sleep-inducing compound than would be an individual whose PER3 VNTR genotype is 4/4 or 4/5. 8. The method of claim 7, wherein the sleep-inducing compound includes a melatonin agonist. 9. The method of claim 8, wherein the melatonin agonist is (1R-trans)-N-[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide. 10. A method of treating a patient suffering from a sleep disorder or from insomnia as a symptom of another disorder, the method comprising: determining the patient's genotype at the PER3 variable number tandem repeat (VNTR) locus; and treating the patient based on the patient's PER3 VNTR genotype. 11. The method of claim 10, wherein treating includes administering to the patient an effective amount of a sleep-inducing compound. 12. The method of claim 11, wherein the effective amount is sufficient to improve at least one sleep parameter of the patient selected from a group consisting of: sleep efficiency (SE), total sleep time (TST), latency to persistent sleep (LPS), and wake after sleep onset (WASO). 13. The method of claim 11, wherein the sleep-inducing compound is a melatonin agonist. 14. The method of claim 13, wherein the melatonin agonist is (1R-trans)-N—[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide. 15. The method of claim 11, wherein the effective amount is based on the patient's PER3 VNTR genotype. 16. The method of claim 11, wherein the effective amount is between about 10 mg/day and about 300 mg/day. 17. The method of claim 16, wherein the effective amount is between about 10 mg/day and about 100 mg/day. 18. The method of claim 17, wherein the effective amount is between about 20 mg/day and about 50 mg/day. 19. The method of claim 15, wherein the effective amount is greater for a patient whose PER3 VNTR genotype is 5/5 than for a patient whose PER3 VNTR genotype is 4/4 or 4/5. 20. The method of claim 15, wherein, in the case that the patient's PER3 VNTR genotype is 5/5, treating the patient includes not treating the patient with (1R-trans)-N-[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide. 21. The method of claim 10, wherein the insomnia is selected from a group consisting of: chronic insomnia and transient insomnia. 22. A method of selecting individuals for inclusion in a sleep study, the method comprising: determining an individual's genotype for the PER3 VNTR locus; and in the case that the individual's PER3 VNTR genotype is 5/5, concluding that the individual will be less responsive to sleep-enhancing or sleep-inducing compositions than an individual having a PER3 VNTR genotype other than 5/5. 23. The method of claim 22, wherein the sleep study assesses at least one indicator selected from a group consisting of: latency to persistent sleep (LPS), wake after sleep onset (WASO), total sleep time (TST), and sleep efficiency (SE). 24. (1R-trans)-N-[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide for use in the treatment of a sleep disorder or one or more symptoms of insomnia associated with another disorder in a patient having a genotype associated with efficacious response to a sleep-inducing compound. 25. The use of claim 24, wherein the sleep disorder is selected from a group consisting of: chronic insomnia and transient insomnia.
The invention relates to the prediction of a sleep parameter (e.g., sleep efficiency (SE), latency to persistent sleep (LPS), wake after sleep onset (WASO), total sleep time (TST)) of an individual and the response of such an individual to a sleep inducing compound based on the individual's PER3 variable number tandem repeat (VNTR) genotype.1. A method of predicting a sleep parameter of an individual, the method comprising: determining the individual's genotype at the PER3 variable number tandem repeat (VNTR) locus; and in the case that the individual's PER3 VNTR genotype is 5/5, predicting that the sleep parameter of the individual is indicative of better sleep than an individual whose PER3 VNTR genotype is 4/4 or 4/5. 2. The method of claim 1, wherein the sleep parameter is selected from a group consisting of: sleep efficiency (SE), total sleep time (TST), latency to persistent sleep (LPS), and wake after sleep onset (WASO). 3. The method of claim 1, wherein the individual's genotype is determined ex vivo. 4. The method of claim 1, wherein the individual is suffering from a sleep disorder. 5. The method of claim 4, wherein the sleep disorder is insomnia or insomnia is a symptom of another disorder. 6. The method of claim 5, wherein the insomnia is selected from a group consisting of: chronic insomnia and transient insomnia. 7. The method of claim 1, further comprising: in the case that the individual's PER3 VNTR genotype is 5/5, predicting that the individual will be less responsive to a sleep-inducing compound than would be an individual whose PER3 VNTR genotype is 4/4 or 4/5. 8. The method of claim 7, wherein the sleep-inducing compound includes a melatonin agonist. 9. The method of claim 8, wherein the melatonin agonist is (1R-trans)-N-[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide. 10. A method of treating a patient suffering from a sleep disorder or from insomnia as a symptom of another disorder, the method comprising: determining the patient's genotype at the PER3 variable number tandem repeat (VNTR) locus; and treating the patient based on the patient's PER3 VNTR genotype. 11. The method of claim 10, wherein treating includes administering to the patient an effective amount of a sleep-inducing compound. 12. The method of claim 11, wherein the effective amount is sufficient to improve at least one sleep parameter of the patient selected from a group consisting of: sleep efficiency (SE), total sleep time (TST), latency to persistent sleep (LPS), and wake after sleep onset (WASO). 13. The method of claim 11, wherein the sleep-inducing compound is a melatonin agonist. 14. The method of claim 13, wherein the melatonin agonist is (1R-trans)-N—[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide. 15. The method of claim 11, wherein the effective amount is based on the patient's PER3 VNTR genotype. 16. The method of claim 11, wherein the effective amount is between about 10 mg/day and about 300 mg/day. 17. The method of claim 16, wherein the effective amount is between about 10 mg/day and about 100 mg/day. 18. The method of claim 17, wherein the effective amount is between about 20 mg/day and about 50 mg/day. 19. The method of claim 15, wherein the effective amount is greater for a patient whose PER3 VNTR genotype is 5/5 than for a patient whose PER3 VNTR genotype is 4/4 or 4/5. 20. The method of claim 15, wherein, in the case that the patient's PER3 VNTR genotype is 5/5, treating the patient includes not treating the patient with (1R-trans)-N-[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide. 21. The method of claim 10, wherein the insomnia is selected from a group consisting of: chronic insomnia and transient insomnia. 22. A method of selecting individuals for inclusion in a sleep study, the method comprising: determining an individual's genotype for the PER3 VNTR locus; and in the case that the individual's PER3 VNTR genotype is 5/5, concluding that the individual will be less responsive to sleep-enhancing or sleep-inducing compositions than an individual having a PER3 VNTR genotype other than 5/5. 23. The method of claim 22, wherein the sleep study assesses at least one indicator selected from a group consisting of: latency to persistent sleep (LPS), wake after sleep onset (WASO), total sleep time (TST), and sleep efficiency (SE). 24. (1R-trans)-N-[[2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]propanamide for use in the treatment of a sleep disorder or one or more symptoms of insomnia associated with another disorder in a patient having a genotype associated with efficacious response to a sleep-inducing compound. 25. The use of claim 24, wherein the sleep disorder is selected from a group consisting of: chronic insomnia and transient insomnia.
1,600
533
14,548,690
1,631
Devices and methods for detecting heart failure (HF) events or identifying patient at elevated risk of developing future HF events, such as events indicative of HF decompensation status, are described. The devices and methods can detect an HF event or predict HF risk using signal transfigurations on different portions of a physiologic signal. A system can comprise a physiologic signal analyzer circuit that can generate a signal trend of a signal feature calculated using one or more physiologic signals obtained from a patient. A signal transformation circuit can dynamically generates first and second transformations, apply the transformations to respective first and second portions of the signal trend, and generate respectively a first and second transformed signal trends. A target physiologic event detector circuit can detect a target physiologic event such as an event of worsening HF using a comparison of the first and second transformed signal trends.
1. A system, comprising: a physiologic signal analyzer circuit, including: a physiologic signal receiver circuit configured to receive one or more physiologic signals; and a signal trend generator configured to calculate a signal feature from the one or more physiologic signals and to generate a signal trend of the signal feature; a signal transformation circuit configured to dynamically generate first and second transformations using at least one characteristic measure of the signal trend, apply the first transformation to a first portion of the signal trend to generate a first transformed signal trend, and apply the second transformation to a second portion of the signal trend to generate a second transformed signal trend, the second portion of the signal trend different from the first portion of the signal trend; and a target physiologic event detector circuit configured to detect a target physiologic event using a comparison of the first and second transformed signal trends. 2. The system of claim 1, wherein the first portion of the signal trend does not overlap in time with the second portion of the signal trend. 3. The system of claim 1, wherein: the second portion of the signal trend includes data from the signal trend preceding the first portion of the signal trend in time, the second portion of the signal trend representing a baseline free of predicted target physiologic event; and the target physiologic event detector circuit is configured to detect the target physiologic event using a relative difference between the first and second transformed signal trends. 4. The system of claim 1, wherein the signal transformation circuit is configured to: generate the at least one characteristic measure including strength of the signal trend; and generate the first and second transformations each including a plurality of weight factors proportional to the strength of the signal trend. 5. The system of claim 1, wherein the signal transformation circuit is configured to generate the first and second transformations each including a plurality of time varying weight factors, the first transformation being different from the second transformation. 6. The system of claim 5, wherein the signal transformation circuit is configured to determine values of the plurality of time-varying weight factors as a linear or a non-linear function of relative time of the signal trend with respect to a reference time. 7. The system of claim 5, wherein the signal transformation circuit is configured to determine values of the plurality of time-varying weight factors as a monotonically increasing or monotonically decreasing function of relative time of the signal trend with respect to a reference time. 8. The system of claim 5, wherein the signal transformation circuit is configured to determine values of the plurality of time-varying weight factors as an exponential function of relative time of the signal trend with respect to a reference time. 9. The system of claim 5, wherein the first transformation includes a first plurality of time-varying weight factors and the second transformation includes a second plurality of time-varying weight factors, and wherein the signal transformation circuit is configured to: determine values of the first plurality of time-varying weight factors as a monotonically increasing function of relative time of the first portion of the signal trend with respect to a first reference time; and determine values of the second plurality of time-varying weight factors as a monotonically decreasing function of relative time of the second portion of the signal trend with respect to a second reference time. 10. The system of claim 1, comprising an auxiliary signal analyzer circuit configured to receive an auxiliary signal non-identical to the one or more physiologic signals, wherein the signal transformation circuit is configured to: generate the at least one characteristic measure including auxiliary signal strength; and dynamically generate the first and second transformations including a plurality of weight factors proportional to the auxiliary signal strength. 11. A system, comprising: a physiologic signal analyzer circuit, including: a physiologic signal receiver circuit configured to receive one or more physiologic signals; and a signal trend generator configured to calculate a signal feature from the one or more physiologic signals and to generate a signal trend of the signal feature; a signal transformation circuit configured to dynamically generate a transformation using strength of the signal trend, apply the transformation to the signal trend to generate a transformed signal trend using the transformation; and a target physiologic event detector circuit configured to calculate a representative value using the transformed signal trend, and to detect a target physiologic event in response to the representative value meeting a specified criterion. 12. The system of claim 11, wherein: the signal transformation circuit is configured to generate the transformation including a plurality of weight factors proportional to the strength of the signal trend; and the target physiologic event detector is configured to calculate the representative value including a central tendency of a selected portion of the transformed signal trend, and to detect the target physiologic event in response to the central tendency falling within a specified range. 13. A method, comprising: receiving one or more physiologic signals; generating a signal trend using a signal feature calculated from the one or more physiologic signals, the signal trend indicating the temporal variation of the signal feature; dynamically generating first and second transformations using at least one characteristic measure of the signal trend; transforming a first portion of the signal trend into a first transformed signal trend using the first transformation, and transforming a second portion of the signal trend into a second transformed signal trend using the second transformation, the second portion of the signal trend different from the first portion of the signal trend; and detecting a target physiologic event in response to the transformed signal trend meeting a specified criterion. 14. The method of claim 13, wherein transforming the first and second portions of the signal trends includes transforming the first portion of the signal trend non-overlapping in time with the second portion of the signal trend. 15. The method of claim 13, wherein: the second portion of the signal trend includes data from the signal trend preceding the first portion of the signal trend in time, the second portion of the signal trend representing a baseline free of predicted target physiologic event; and detecting a target physiologic event including determining whether a relative difference between the first and second transformed signal trends meets a specified criterion. 16. The method of claim 13, wherein dynamically generating the first and second transformation includes generating respectively first and second plurality of weight factors proportional to strength of the signal trend. 17. The method of claim 13, wherein dynamically generating the transformation includes generating respectively first and second plurality of timing-varying weight factors. 18. The method of claim 17, wherein generating the first and second plurality of time-varying weight factors includes determining values of the time-varying weight factors as one of a linear, a nonlinear, a monotonically increasing, or a monotonically decreasing function of relative time of the signal trend with respect to a reference time. 19. The method of claim 18, wherein generating the first and second plurality of time-varying weight factors includes determining the first plurality of time-varying weight factors as a monotonically increasing function of relative time of the first portion of the signal trend with respect to a first reference time, and determining the second plurality of time-varying weight factors as a monotonically decreasing function of relative time of the second portion of the signal trend with respect to a second reference time. 20. The method of claim 13, further comprising receiving an auxiliary signal non-identical to the one or more physiologic signals, wherein dynamically generating the transformation includes generating a plurality of weight factors proportional to strength of the auxiliary signal.
Devices and methods for detecting heart failure (HF) events or identifying patient at elevated risk of developing future HF events, such as events indicative of HF decompensation status, are described. The devices and methods can detect an HF event or predict HF risk using signal transfigurations on different portions of a physiologic signal. A system can comprise a physiologic signal analyzer circuit that can generate a signal trend of a signal feature calculated using one or more physiologic signals obtained from a patient. A signal transformation circuit can dynamically generates first and second transformations, apply the transformations to respective first and second portions of the signal trend, and generate respectively a first and second transformed signal trends. A target physiologic event detector circuit can detect a target physiologic event such as an event of worsening HF using a comparison of the first and second transformed signal trends.1. A system, comprising: a physiologic signal analyzer circuit, including: a physiologic signal receiver circuit configured to receive one or more physiologic signals; and a signal trend generator configured to calculate a signal feature from the one or more physiologic signals and to generate a signal trend of the signal feature; a signal transformation circuit configured to dynamically generate first and second transformations using at least one characteristic measure of the signal trend, apply the first transformation to a first portion of the signal trend to generate a first transformed signal trend, and apply the second transformation to a second portion of the signal trend to generate a second transformed signal trend, the second portion of the signal trend different from the first portion of the signal trend; and a target physiologic event detector circuit configured to detect a target physiologic event using a comparison of the first and second transformed signal trends. 2. The system of claim 1, wherein the first portion of the signal trend does not overlap in time with the second portion of the signal trend. 3. The system of claim 1, wherein: the second portion of the signal trend includes data from the signal trend preceding the first portion of the signal trend in time, the second portion of the signal trend representing a baseline free of predicted target physiologic event; and the target physiologic event detector circuit is configured to detect the target physiologic event using a relative difference between the first and second transformed signal trends. 4. The system of claim 1, wherein the signal transformation circuit is configured to: generate the at least one characteristic measure including strength of the signal trend; and generate the first and second transformations each including a plurality of weight factors proportional to the strength of the signal trend. 5. The system of claim 1, wherein the signal transformation circuit is configured to generate the first and second transformations each including a plurality of time varying weight factors, the first transformation being different from the second transformation. 6. The system of claim 5, wherein the signal transformation circuit is configured to determine values of the plurality of time-varying weight factors as a linear or a non-linear function of relative time of the signal trend with respect to a reference time. 7. The system of claim 5, wherein the signal transformation circuit is configured to determine values of the plurality of time-varying weight factors as a monotonically increasing or monotonically decreasing function of relative time of the signal trend with respect to a reference time. 8. The system of claim 5, wherein the signal transformation circuit is configured to determine values of the plurality of time-varying weight factors as an exponential function of relative time of the signal trend with respect to a reference time. 9. The system of claim 5, wherein the first transformation includes a first plurality of time-varying weight factors and the second transformation includes a second plurality of time-varying weight factors, and wherein the signal transformation circuit is configured to: determine values of the first plurality of time-varying weight factors as a monotonically increasing function of relative time of the first portion of the signal trend with respect to a first reference time; and determine values of the second plurality of time-varying weight factors as a monotonically decreasing function of relative time of the second portion of the signal trend with respect to a second reference time. 10. The system of claim 1, comprising an auxiliary signal analyzer circuit configured to receive an auxiliary signal non-identical to the one or more physiologic signals, wherein the signal transformation circuit is configured to: generate the at least one characteristic measure including auxiliary signal strength; and dynamically generate the first and second transformations including a plurality of weight factors proportional to the auxiliary signal strength. 11. A system, comprising: a physiologic signal analyzer circuit, including: a physiologic signal receiver circuit configured to receive one or more physiologic signals; and a signal trend generator configured to calculate a signal feature from the one or more physiologic signals and to generate a signal trend of the signal feature; a signal transformation circuit configured to dynamically generate a transformation using strength of the signal trend, apply the transformation to the signal trend to generate a transformed signal trend using the transformation; and a target physiologic event detector circuit configured to calculate a representative value using the transformed signal trend, and to detect a target physiologic event in response to the representative value meeting a specified criterion. 12. The system of claim 11, wherein: the signal transformation circuit is configured to generate the transformation including a plurality of weight factors proportional to the strength of the signal trend; and the target physiologic event detector is configured to calculate the representative value including a central tendency of a selected portion of the transformed signal trend, and to detect the target physiologic event in response to the central tendency falling within a specified range. 13. A method, comprising: receiving one or more physiologic signals; generating a signal trend using a signal feature calculated from the one or more physiologic signals, the signal trend indicating the temporal variation of the signal feature; dynamically generating first and second transformations using at least one characteristic measure of the signal trend; transforming a first portion of the signal trend into a first transformed signal trend using the first transformation, and transforming a second portion of the signal trend into a second transformed signal trend using the second transformation, the second portion of the signal trend different from the first portion of the signal trend; and detecting a target physiologic event in response to the transformed signal trend meeting a specified criterion. 14. The method of claim 13, wherein transforming the first and second portions of the signal trends includes transforming the first portion of the signal trend non-overlapping in time with the second portion of the signal trend. 15. The method of claim 13, wherein: the second portion of the signal trend includes data from the signal trend preceding the first portion of the signal trend in time, the second portion of the signal trend representing a baseline free of predicted target physiologic event; and detecting a target physiologic event including determining whether a relative difference between the first and second transformed signal trends meets a specified criterion. 16. The method of claim 13, wherein dynamically generating the first and second transformation includes generating respectively first and second plurality of weight factors proportional to strength of the signal trend. 17. The method of claim 13, wherein dynamically generating the transformation includes generating respectively first and second plurality of timing-varying weight factors. 18. The method of claim 17, wherein generating the first and second plurality of time-varying weight factors includes determining values of the time-varying weight factors as one of a linear, a nonlinear, a monotonically increasing, or a monotonically decreasing function of relative time of the signal trend with respect to a reference time. 19. The method of claim 18, wherein generating the first and second plurality of time-varying weight factors includes determining the first plurality of time-varying weight factors as a monotonically increasing function of relative time of the first portion of the signal trend with respect to a first reference time, and determining the second plurality of time-varying weight factors as a monotonically decreasing function of relative time of the second portion of the signal trend with respect to a second reference time. 20. The method of claim 13, further comprising receiving an auxiliary signal non-identical to the one or more physiologic signals, wherein dynamically generating the transformation includes generating a plurality of weight factors proportional to strength of the auxiliary signal.
1,600
534
14,170,373
1,631
A biological pathway is a series of actions that take place in an organism that lead to some resulting pathology or otherwise change the organism state. In the cell, these actions typically take place between molecules called proteins. Proteins within the cell interact in ways that are not fully understood, but evidence concerning these interactions is constantly being collected and published by microbiologists. The disclosed method automatically infers such biological pathways between proteins by looking at the overall system of published literature about those proteins.
1. A method for discovering a pathway among a set of biological and/or chemical entities, comprising: a) providing documents about each of the biological and/or chemical entities; b) creating a vector space representation of the documents based on words and/or phrases occurring in the documents; c) for each biological and/or chemical entity, creating a centroid in the vector space based on the vectors corresponding to documents mentioning that biological and/or chemical entity; d) creating a relative distance network of the biological and/or chemical entities, in view of the centroids, thereby identifying a particular pathway connecting the centroids; and e) finding at least one most connected centroid on said particular pathway, thereby identifying a particular biological and/or chemical entity for further investigation, wherein said particular biological and/or chemical entity corresponds to said at least one most connected centroid. 2. The method of claim 1, wherein, prior to step (b), documents matching more than one biological and/or chemical entity are removed. 3. The method of claim 1, wherein biological and/or chemical entities having less than a pre-defined threshold number of documents are removed. 4. The method of claim 1, wherein said biological and/or chemical entities are selected from the group consisting of human genes and proteins. 5. The method of claim 1, wherein said documents are provided in response to a query. 6. The method of claim 1, wherein said documents are provided over a network. 7. The method of claim 6, wherein said network is any of the following: local area network (LAN), wide area network (WAN), the Internet, or cellular network. 8. A method comprising: a. receiving a set of biological and/or chemical entities of interest, E; b. identifying a document set, R, mentioning any biological and/or chemical entity, and/or a variant thereof in E; c. creating a dictionary, D, from common terms and/or phrases in documents of document set R; d. assigning each document in document set R a numeric vector using a vector space model based on said dictionary D; e. computing a centroid for each biological and/or chemical entity in E by averaging numerical vectors of documents in R mentioning that biological and/or chemical entity; f. computing a distance matrix listing a distance between pairs of centroids; g. creating a relative neighborhood graph of biological and/or chemical entities in E based on said computed distance matrix, said relative neighborhood graph identifying a particular pathway connecting computed centroids; and h. identifying, from said relative neighborhood graph, at least one most connected centroid and outputting biological and/or chemical entity associated with said at least one most connected centroid. 9. The method of claim 8, wherein said step of creating said relative neighborhood graph comprises: g1. creating a candidate set, C, with biological and/or chemical entities in E; g2. selecting an initial biological and/or chemical entity in C as a new node, e, to add to a tree and removing said new node e from C; g3. comparing remaining biological and/or chemical entities in C to identify another biological and/or chemical entity to add to said tree with a shortest distance to existing nodes in said tree and adding said identified another biological and/or chemical entity to said tree and removing said another node from C, wherein step g3 is iteratively repeated for other entries in C until there are no more entries in C, with all entries in C being added to said tree; and resulting tree is output as part of said relative neighborhood graph. 10. The method of claim 8, wherein said distance between pairs of centroids is a cosine distance. 11. The method of claim 8, wherein, prior to step (c), documents in R matching more than one biological and/or chemical entity in E are removed from R. 12. The method of claim 8, wherein, prior to step (c), biological and/or chemical entities in E having less than a pre-defined threshold number of documents are removed from E. 13. The method of claim 8, wherein said method comprises displaying said computed centroids and documents in document set, R, via a scatter plot graph. 14. The method of claim 8, wherein said biological and/or chemical entities are selected from the group consisting of human genes and proteins. 15. The method of claim 8, wherein said document set R is identified in response to a query. 16. The method of claim 8, wherein said document set R is identified over a network. 17. The method of claim 16, wherein said network is any of the following: local area network (LAN), wide area network (WAN), the Internet, or cellular network. 18. A non-transitory, computer accessible memory medium storing program instructions for discovering a pathway among a set of biological and/or chemical entities, wherein the program instructions are executable by a processor to: a. receive a set of biological and/or chemical entities of interest, E; b. identify a document set, R, mentioning any biological and/or chemical entity, and/or a variant thereof in E; c. create a dictionary, D, from common terms and/or phrases in documents of document set R; d. assign each document in document set R a numeric vector using a vector space model based on said dictionary D; e. compute a centroid for each biological and/or chemical entity in E by averaging numerical vectors of documents in R mentioning that biological and/or chemical entity; f. compute a distance matrix listing a distance between pairs of centroids; g. create a relative neighborhood graph of biological and/or chemical entities in E based on said computed distance matrix, said relative neighborhood graph identifying a particular pathway connecting computed centroids; and h. identify, from said relative neighborhood graph, at least one most connected centroid and outputting biological and/or chemical entity associated with said at least one most connected centroid. 19. A system for discovering a pathway among a set of biological/chemical entities, the system comprising: one or more processors; and a memory comprising instructions which, when executed by the one or more processors, cause the one or more processors to: a. receive a set of biological and/or chemical entities of interest, E; b. identify a document set, R, mentioning any biological and/or chemical entity, and/or a variant thereof in E; c. create a dictionary, D, from common terms and/or phrases in documents of document set R; d. assign each document in document set R a numeric vector using a vector space model based on said dictionary D; e. compute a centroid for each biological and/or chemical entity in E by averaging numerical vectors of documents in R mentioning that biological and/or chemical entity; f. compute a distance matrix listing a distance between pairs of centroids; g. create a relative neighborhood graph of biological and/or chemical entities in E based on said computed distance matrix, said relative neighborhood graph identifying a particular pathway connecting computed centroids; and h. identify, from said relative neighborhood graph, at least one most connected centroid and outputting biological and/or chemical entity associated with said at least one most connected centroid.
A biological pathway is a series of actions that take place in an organism that lead to some resulting pathology or otherwise change the organism state. In the cell, these actions typically take place between molecules called proteins. Proteins within the cell interact in ways that are not fully understood, but evidence concerning these interactions is constantly being collected and published by microbiologists. The disclosed method automatically infers such biological pathways between proteins by looking at the overall system of published literature about those proteins.1. A method for discovering a pathway among a set of biological and/or chemical entities, comprising: a) providing documents about each of the biological and/or chemical entities; b) creating a vector space representation of the documents based on words and/or phrases occurring in the documents; c) for each biological and/or chemical entity, creating a centroid in the vector space based on the vectors corresponding to documents mentioning that biological and/or chemical entity; d) creating a relative distance network of the biological and/or chemical entities, in view of the centroids, thereby identifying a particular pathway connecting the centroids; and e) finding at least one most connected centroid on said particular pathway, thereby identifying a particular biological and/or chemical entity for further investigation, wherein said particular biological and/or chemical entity corresponds to said at least one most connected centroid. 2. The method of claim 1, wherein, prior to step (b), documents matching more than one biological and/or chemical entity are removed. 3. The method of claim 1, wherein biological and/or chemical entities having less than a pre-defined threshold number of documents are removed. 4. The method of claim 1, wherein said biological and/or chemical entities are selected from the group consisting of human genes and proteins. 5. The method of claim 1, wherein said documents are provided in response to a query. 6. The method of claim 1, wherein said documents are provided over a network. 7. The method of claim 6, wherein said network is any of the following: local area network (LAN), wide area network (WAN), the Internet, or cellular network. 8. A method comprising: a. receiving a set of biological and/or chemical entities of interest, E; b. identifying a document set, R, mentioning any biological and/or chemical entity, and/or a variant thereof in E; c. creating a dictionary, D, from common terms and/or phrases in documents of document set R; d. assigning each document in document set R a numeric vector using a vector space model based on said dictionary D; e. computing a centroid for each biological and/or chemical entity in E by averaging numerical vectors of documents in R mentioning that biological and/or chemical entity; f. computing a distance matrix listing a distance between pairs of centroids; g. creating a relative neighborhood graph of biological and/or chemical entities in E based on said computed distance matrix, said relative neighborhood graph identifying a particular pathway connecting computed centroids; and h. identifying, from said relative neighborhood graph, at least one most connected centroid and outputting biological and/or chemical entity associated with said at least one most connected centroid. 9. The method of claim 8, wherein said step of creating said relative neighborhood graph comprises: g1. creating a candidate set, C, with biological and/or chemical entities in E; g2. selecting an initial biological and/or chemical entity in C as a new node, e, to add to a tree and removing said new node e from C; g3. comparing remaining biological and/or chemical entities in C to identify another biological and/or chemical entity to add to said tree with a shortest distance to existing nodes in said tree and adding said identified another biological and/or chemical entity to said tree and removing said another node from C, wherein step g3 is iteratively repeated for other entries in C until there are no more entries in C, with all entries in C being added to said tree; and resulting tree is output as part of said relative neighborhood graph. 10. The method of claim 8, wherein said distance between pairs of centroids is a cosine distance. 11. The method of claim 8, wherein, prior to step (c), documents in R matching more than one biological and/or chemical entity in E are removed from R. 12. The method of claim 8, wherein, prior to step (c), biological and/or chemical entities in E having less than a pre-defined threshold number of documents are removed from E. 13. The method of claim 8, wherein said method comprises displaying said computed centroids and documents in document set, R, via a scatter plot graph. 14. The method of claim 8, wherein said biological and/or chemical entities are selected from the group consisting of human genes and proteins. 15. The method of claim 8, wherein said document set R is identified in response to a query. 16. The method of claim 8, wherein said document set R is identified over a network. 17. The method of claim 16, wherein said network is any of the following: local area network (LAN), wide area network (WAN), the Internet, or cellular network. 18. A non-transitory, computer accessible memory medium storing program instructions for discovering a pathway among a set of biological and/or chemical entities, wherein the program instructions are executable by a processor to: a. receive a set of biological and/or chemical entities of interest, E; b. identify a document set, R, mentioning any biological and/or chemical entity, and/or a variant thereof in E; c. create a dictionary, D, from common terms and/or phrases in documents of document set R; d. assign each document in document set R a numeric vector using a vector space model based on said dictionary D; e. compute a centroid for each biological and/or chemical entity in E by averaging numerical vectors of documents in R mentioning that biological and/or chemical entity; f. compute a distance matrix listing a distance between pairs of centroids; g. create a relative neighborhood graph of biological and/or chemical entities in E based on said computed distance matrix, said relative neighborhood graph identifying a particular pathway connecting computed centroids; and h. identify, from said relative neighborhood graph, at least one most connected centroid and outputting biological and/or chemical entity associated with said at least one most connected centroid. 19. A system for discovering a pathway among a set of biological/chemical entities, the system comprising: one or more processors; and a memory comprising instructions which, when executed by the one or more processors, cause the one or more processors to: a. receive a set of biological and/or chemical entities of interest, E; b. identify a document set, R, mentioning any biological and/or chemical entity, and/or a variant thereof in E; c. create a dictionary, D, from common terms and/or phrases in documents of document set R; d. assign each document in document set R a numeric vector using a vector space model based on said dictionary D; e. compute a centroid for each biological and/or chemical entity in E by averaging numerical vectors of documents in R mentioning that biological and/or chemical entity; f. compute a distance matrix listing a distance between pairs of centroids; g. create a relative neighborhood graph of biological and/or chemical entities in E based on said computed distance matrix, said relative neighborhood graph identifying a particular pathway connecting computed centroids; and h. identify, from said relative neighborhood graph, at least one most connected centroid and outputting biological and/or chemical entity associated with said at least one most connected centroid.
1,600
535
15,236,897
1,616
A herbicide and a method for killing targets plants is disclosed. The herbicide includes first and second amino acids applied to the leaves of the plant. The first and second amino acids, when combined interfere with at least one biological process.
1. A herbicide comprising: one of a first amino acid and a first and a second amino acid, the first and second amino acids, combined interfere with at least one biologic process within one of dandelions, and Canadian thistle. 2. The herbicide according to claim 1 wherein the one of a first amino acid and a first and a second amino acid inhibit aspartokinase in a target weed or plant. 3. The herbicide according to claim 1 wherein the one of a first amino acid include one of lysine, threonine, and methionine. 5. The herbicide according to claim 1 wherein the first and second amino acids include a mixture of lysine, threonine. 6. The herbicide according to claim 1 wherein the first and second amino acids include a mixture lysine, threonine having a concentration between 2 and 4 mM. 7. The herbicide according to claim 1 wherein the first and second amino acids comprises a mixture amino acids having a concentration of between 20-50% lysine and the balance of the amino acid mixture being the balance threonine. 8. The herbicide according to claim 1 wherein the herbicide contain first and second amino acids comprising a concentration of selected from the group of 20-40%, 20-35%, 20-30%, 20-25%, 25-50%, 30-50%, 35-50%, 40-50%, and 45-50%. 9. The herbicide according to claim 1 comprising a salt, which can have cations being one of potassium and ammonia ions. 10. The herbicide according to claim 9 having a concentration of between 0.2 and 0.5 M. 11. The herbicide according to claim 1 comprising a fertilizer. 12. The herbicide according to claim 1 consisting of an amino acid selected from the group of lysine, threonine, and methionine, valine, and tryptophan. 13. The herbicide according to according to claim 1 consisting of a pair of amino acids selected from the group of pairs of two of lysine, threonine, and methionine, valine, and tryptophan. 14. The herbicide according to according to claim 1 wherein the first amino acid oversupplies the first amino acid within at least one specific biological process so as to cause the plant to use up all of at least one amino acid, thus causing the plant to die from the deficit of the amino acid. 15. The herbicide according to claim 1 further comprising a surfactant to improve wetting of a target plant leaf. 16. A method of killing a target plant comprising: applying a herbicide comprising an amino acid to the target plant the first amino acid oversupplies the first amino acid within at least one specific biological process so as to cause the plant to use up all of at least one amino acid, thus causing the plant to die from the deficit of the amino acid . 17. The method according to claim 16, wherein applying a herbicide is spraying a liquid on the leaves of the plant. 18. A method of killing a target plant comprising: applying a herbicide comprising a pair amino acid to the target plant the pair of amino acids oversupplying at least a first amino acid within at least one specific biological process so as to cause the plant to use up all of at least a third amino acid, thus causing the plant to die from the deficit of the amino acid. 19. The method according to claim 18, wherein applying a herbicide is spraying a liquid on the leaves of the plant and wherein the pair of amino acids oversupplying at least a second amino acid within at least a second specific biological process so as to cause the plant to use up all of at least a fourth amino acid, thus causing the plant to die from the deficit of the third amino acid.
A herbicide and a method for killing targets plants is disclosed. The herbicide includes first and second amino acids applied to the leaves of the plant. The first and second amino acids, when combined interfere with at least one biological process.1. A herbicide comprising: one of a first amino acid and a first and a second amino acid, the first and second amino acids, combined interfere with at least one biologic process within one of dandelions, and Canadian thistle. 2. The herbicide according to claim 1 wherein the one of a first amino acid and a first and a second amino acid inhibit aspartokinase in a target weed or plant. 3. The herbicide according to claim 1 wherein the one of a first amino acid include one of lysine, threonine, and methionine. 5. The herbicide according to claim 1 wherein the first and second amino acids include a mixture of lysine, threonine. 6. The herbicide according to claim 1 wherein the first and second amino acids include a mixture lysine, threonine having a concentration between 2 and 4 mM. 7. The herbicide according to claim 1 wherein the first and second amino acids comprises a mixture amino acids having a concentration of between 20-50% lysine and the balance of the amino acid mixture being the balance threonine. 8. The herbicide according to claim 1 wherein the herbicide contain first and second amino acids comprising a concentration of selected from the group of 20-40%, 20-35%, 20-30%, 20-25%, 25-50%, 30-50%, 35-50%, 40-50%, and 45-50%. 9. The herbicide according to claim 1 comprising a salt, which can have cations being one of potassium and ammonia ions. 10. The herbicide according to claim 9 having a concentration of between 0.2 and 0.5 M. 11. The herbicide according to claim 1 comprising a fertilizer. 12. The herbicide according to claim 1 consisting of an amino acid selected from the group of lysine, threonine, and methionine, valine, and tryptophan. 13. The herbicide according to according to claim 1 consisting of a pair of amino acids selected from the group of pairs of two of lysine, threonine, and methionine, valine, and tryptophan. 14. The herbicide according to according to claim 1 wherein the first amino acid oversupplies the first amino acid within at least one specific biological process so as to cause the plant to use up all of at least one amino acid, thus causing the plant to die from the deficit of the amino acid. 15. The herbicide according to claim 1 further comprising a surfactant to improve wetting of a target plant leaf. 16. A method of killing a target plant comprising: applying a herbicide comprising an amino acid to the target plant the first amino acid oversupplies the first amino acid within at least one specific biological process so as to cause the plant to use up all of at least one amino acid, thus causing the plant to die from the deficit of the amino acid . 17. The method according to claim 16, wherein applying a herbicide is spraying a liquid on the leaves of the plant. 18. A method of killing a target plant comprising: applying a herbicide comprising a pair amino acid to the target plant the pair of amino acids oversupplying at least a first amino acid within at least one specific biological process so as to cause the plant to use up all of at least a third amino acid, thus causing the plant to die from the deficit of the amino acid. 19. The method according to claim 18, wherein applying a herbicide is spraying a liquid on the leaves of the plant and wherein the pair of amino acids oversupplying at least a second amino acid within at least a second specific biological process so as to cause the plant to use up all of at least a fourth amino acid, thus causing the plant to die from the deficit of the third amino acid.
1,600
536
14,007,577
1,628
The present application discloses 17β hydroxy steroid dehydrogenase (17β HSD) type 1, 3, 10 inhibitors and use thereof (alone and in combination) in the treatment of cancer and other afflictions. 17β HSD1 inhibitors include estradiol derivatives with a nieta-carbamoylbenzyl substituent at C 16. 17β HSD3/HSD10 inhibitors include androsterone derivatives substituted at the C3 position with a sulfonamide piperazine. Also disclosed are compounds that are inhibitors of both 17β HSD1 and 17β HSD3 that have a spiro-morpholine substituent at C20.
1. An inhibitor of 17β-HSD1. 2. The inhibitor of claim 1, wherein the inhibitor has the structure: wherein R is —OH, halo, —NH2, alkyl, alkoxy, aralkyl, carboxy, —CH2-heterocyclyl, heterocyclyloyl, —C(O)N(R′)(R″) or —B(OH)2, wherein R′ and R″ are independently or simultanesouly H or alkyl, or R′ and R″ are joined together, along with the nitrogen atom to which they are attached, to form a heterocyclic ring, or a pharaceutically acceptable salt or tautomer thereof. 3. The inhibitor of claim 2, wherein the inhibitor has the structure or a pharmaceutically acceptable salt thereof. 4. The inhibitor of claim 3, wherein the inhibitor has the structure 5. The inhibitor of claim 1, wherein the inhibitor has the structure: or a pharmaceutically acceptable salt thereof. 6. An inhibitor of 17β-HSD3. 7. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein R is heterocyclyl or —NRaRb, wherein Ra and Rb are each independently alkyl or aralkyl, or a pharmaceutically acceptable salt or tautomer thereof. 8. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein: X is or Y is —CH2—, —C(O)— or —S(O)2—; and Z is cycloalkyl, aryl or heterocyclyl, or a pharmaceutically acceptable salt or tautomer thereof. 9. The inhibitor of claim 8, wherein the inhibitor has the structure or a pharmaceutically acceptable salt thereof. 10. The inhibitor of claim 9, wherein the inhibitor has the structure 11. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein: R1 is alkyl, aralkyl, heterocyclyl, cycloalkyl, —CH2CH2S(O)2-aryl, heterocyclyl, —CH2CH2CH(O-acyl)CH2-heterocyclyl, or —CH2CH2NRaRb, wherein Ra and Rb are each independently alkyl, cycloalkyl, acyl, or aralkyl; and each R2 is independently hydrogen or alkyl, or a pharmaceutically acceptable salt or tautomer thereof. 12. The inhibitor of claim 11, wherein the inhibitor has the structure wherein, X1 is alkyl, R2 is independently hydrogen or alkyl, R3 is hydrogen, alkyl or aralkyl, or a pharmaceutically acceptable salt or tautomer thereof. 13. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein, R2 is independently hydrogen or alkyl, R4 is hydrogen or aralkyl, R5 is hydrogen, alkyl or aralkyl, or a pharmaceutically acceptable salt or tautomer thereof. 14. An inhibitor of 17β-HSD1 and 17β-HSD3. 15. The inhibitor of claim 14, wherein the inhibitor has the structure Xa or Xb: wherein: R6 and R7 are independently or simultaneously H, alkyl, cycloalkyl, aryl, aralkyl, or heterocyclyl, or a pharmaceutically acceptable salt or tautomer thereof. 16. An inhibitor of 17β-HSD10. 17. The inhibitor of claim 16, wherein the inhibitor has the structure: wherein: A is aryl or heteroaryl; W is —C(═O)—, —CH(—OH) or —CH(—COCH3); X is H, alkyl, thioalkyl, halo or alkoxy; Y is —CH2—, —C(═O)— or —S(═O)2; Z is H or alkyl, or a pharmaceutically acceptable salt or tautomer thereof. 18. A method of treating cancer in a subject comprising administering to said subject a compound of claim 1. 19. The method of claim 18, wherein said cancer is an estrogen-sensitive cancer. 20. The method of claim 19, wherein said cancer is breast cancer. 21. The method of claim 20, wherein said compound is a compound of claim 1. 22. The method of claim 18, wherein said cancer is an androgen-sensitive cancer. 23. The method of claim 18, wherein said cancer is prostate cancer. 24. The method of claim 23, wherein said compound is a compound of claim 1. 25. The method of claim 18, further comprising treating said subject with a secondary cancer therapy. 26. The method of claim 25, wherein said secondary cancer therapy is selected from the group consisting of chemotherapy, toxin therapy, radiation therapy, hormone or anti-hormone therapy, surgery, cryotherapy, or immunotherapy. 27. A method of treating a non-cancerous disease is a subject comprising administering to said subject a compound of claim 1. 28. The method of claim 27, wherein said non-cancerous disease is endometriosis. 29. The method of claim 28, wherein said compound is a compound of claim 1. 30. The method of claim 27, wherein said non-cancerous disease is benign prostatic hyperplasia. 31. The method of claim 30, wherein said compound is a compound of claim 6. 32. The method of claim 27, wherein said non-cancerous disease is Alzheimer's disease. 33. The method of claim 32, wherein said compound is a compound of claim 9. 34. The method of claim 27, further comprising treating said subject with a secondary therapy. 35. The method of claim 18, further comprising administering said compound at least a second time. 36. The method of any of claim 18, wherein said compound is administered intravenously, intra-arterially, subcutaneously, topically, or intramuscularly. 37. The method of claim 18, wherein said compound is administered systemically, regionally to a tumor/disease site, locally to a tumor/disease site, into tumor/tissue vasculature or intratumorally. 38. The method of claim 18, wherein said cancer is multi drug resistant. 39. The method of claim 18, wherein said cancer is metastatic. 40. The method of claim 18, wherein said cancer is recurrent. 41. The method of claim 18, wherein treating comprises inhibiting cancer growth, killing cancer cells, reducing tumor burden, reducing tumor size, improving said subject's quality of life or prolonging said subject's length of life. 42. The method of claim 18, wherein the cancer is lung cancer, brain cancer, head & neck cancer, skin cancer, liver cancer, pancreatic cancer, stomach cancer, colon cancer, rectal cancer, uterine cancer, cervical cancer, ovarian cancer, testicular cancer, skin cancer or esophageal cancer. 43. The method of claim 18, wherein said subject is a human. 44. The method of claim 18, wherein said subject is a non-human animal. 45. A radiolabelled compound comprising the structure wherein X is I123, I125, I131 or Br76, or a pharmaceutically acceptable salt or tautomer thereof. 46. A radiolabelled compound comprising the structure wherein X is I123, I125, I131 or Br76, or a pharmaceutically acceptable salt or tautomer thereof. 47. The radiolabelled compound of claim 45, for radioimaging or treating breast cancer or prostate cancer.
The present application discloses 17β hydroxy steroid dehydrogenase (17β HSD) type 1, 3, 10 inhibitors and use thereof (alone and in combination) in the treatment of cancer and other afflictions. 17β HSD1 inhibitors include estradiol derivatives with a nieta-carbamoylbenzyl substituent at C 16. 17β HSD3/HSD10 inhibitors include androsterone derivatives substituted at the C3 position with a sulfonamide piperazine. Also disclosed are compounds that are inhibitors of both 17β HSD1 and 17β HSD3 that have a spiro-morpholine substituent at C20.1. An inhibitor of 17β-HSD1. 2. The inhibitor of claim 1, wherein the inhibitor has the structure: wherein R is —OH, halo, —NH2, alkyl, alkoxy, aralkyl, carboxy, —CH2-heterocyclyl, heterocyclyloyl, —C(O)N(R′)(R″) or —B(OH)2, wherein R′ and R″ are independently or simultanesouly H or alkyl, or R′ and R″ are joined together, along with the nitrogen atom to which they are attached, to form a heterocyclic ring, or a pharaceutically acceptable salt or tautomer thereof. 3. The inhibitor of claim 2, wherein the inhibitor has the structure or a pharmaceutically acceptable salt thereof. 4. The inhibitor of claim 3, wherein the inhibitor has the structure 5. The inhibitor of claim 1, wherein the inhibitor has the structure: or a pharmaceutically acceptable salt thereof. 6. An inhibitor of 17β-HSD3. 7. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein R is heterocyclyl or —NRaRb, wherein Ra and Rb are each independently alkyl or aralkyl, or a pharmaceutically acceptable salt or tautomer thereof. 8. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein: X is or Y is —CH2—, —C(O)— or —S(O)2—; and Z is cycloalkyl, aryl or heterocyclyl, or a pharmaceutically acceptable salt or tautomer thereof. 9. The inhibitor of claim 8, wherein the inhibitor has the structure or a pharmaceutically acceptable salt thereof. 10. The inhibitor of claim 9, wherein the inhibitor has the structure 11. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein: R1 is alkyl, aralkyl, heterocyclyl, cycloalkyl, —CH2CH2S(O)2-aryl, heterocyclyl, —CH2CH2CH(O-acyl)CH2-heterocyclyl, or —CH2CH2NRaRb, wherein Ra and Rb are each independently alkyl, cycloalkyl, acyl, or aralkyl; and each R2 is independently hydrogen or alkyl, or a pharmaceutically acceptable salt or tautomer thereof. 12. The inhibitor of claim 11, wherein the inhibitor has the structure wherein, X1 is alkyl, R2 is independently hydrogen or alkyl, R3 is hydrogen, alkyl or aralkyl, or a pharmaceutically acceptable salt or tautomer thereof. 13. The inhibitor of claim 6, wherein the inhibitor has the structure: wherein, R2 is independently hydrogen or alkyl, R4 is hydrogen or aralkyl, R5 is hydrogen, alkyl or aralkyl, or a pharmaceutically acceptable salt or tautomer thereof. 14. An inhibitor of 17β-HSD1 and 17β-HSD3. 15. The inhibitor of claim 14, wherein the inhibitor has the structure Xa or Xb: wherein: R6 and R7 are independently or simultaneously H, alkyl, cycloalkyl, aryl, aralkyl, or heterocyclyl, or a pharmaceutically acceptable salt or tautomer thereof. 16. An inhibitor of 17β-HSD10. 17. The inhibitor of claim 16, wherein the inhibitor has the structure: wherein: A is aryl or heteroaryl; W is —C(═O)—, —CH(—OH) or —CH(—COCH3); X is H, alkyl, thioalkyl, halo or alkoxy; Y is —CH2—, —C(═O)— or —S(═O)2; Z is H or alkyl, or a pharmaceutically acceptable salt or tautomer thereof. 18. A method of treating cancer in a subject comprising administering to said subject a compound of claim 1. 19. The method of claim 18, wherein said cancer is an estrogen-sensitive cancer. 20. The method of claim 19, wherein said cancer is breast cancer. 21. The method of claim 20, wherein said compound is a compound of claim 1. 22. The method of claim 18, wherein said cancer is an androgen-sensitive cancer. 23. The method of claim 18, wherein said cancer is prostate cancer. 24. The method of claim 23, wherein said compound is a compound of claim 1. 25. The method of claim 18, further comprising treating said subject with a secondary cancer therapy. 26. The method of claim 25, wherein said secondary cancer therapy is selected from the group consisting of chemotherapy, toxin therapy, radiation therapy, hormone or anti-hormone therapy, surgery, cryotherapy, or immunotherapy. 27. A method of treating a non-cancerous disease is a subject comprising administering to said subject a compound of claim 1. 28. The method of claim 27, wherein said non-cancerous disease is endometriosis. 29. The method of claim 28, wherein said compound is a compound of claim 1. 30. The method of claim 27, wherein said non-cancerous disease is benign prostatic hyperplasia. 31. The method of claim 30, wherein said compound is a compound of claim 6. 32. The method of claim 27, wherein said non-cancerous disease is Alzheimer's disease. 33. The method of claim 32, wherein said compound is a compound of claim 9. 34. The method of claim 27, further comprising treating said subject with a secondary therapy. 35. The method of claim 18, further comprising administering said compound at least a second time. 36. The method of any of claim 18, wherein said compound is administered intravenously, intra-arterially, subcutaneously, topically, or intramuscularly. 37. The method of claim 18, wherein said compound is administered systemically, regionally to a tumor/disease site, locally to a tumor/disease site, into tumor/tissue vasculature or intratumorally. 38. The method of claim 18, wherein said cancer is multi drug resistant. 39. The method of claim 18, wherein said cancer is metastatic. 40. The method of claim 18, wherein said cancer is recurrent. 41. The method of claim 18, wherein treating comprises inhibiting cancer growth, killing cancer cells, reducing tumor burden, reducing tumor size, improving said subject's quality of life or prolonging said subject's length of life. 42. The method of claim 18, wherein the cancer is lung cancer, brain cancer, head & neck cancer, skin cancer, liver cancer, pancreatic cancer, stomach cancer, colon cancer, rectal cancer, uterine cancer, cervical cancer, ovarian cancer, testicular cancer, skin cancer or esophageal cancer. 43. The method of claim 18, wherein said subject is a human. 44. The method of claim 18, wherein said subject is a non-human animal. 45. A radiolabelled compound comprising the structure wherein X is I123, I125, I131 or Br76, or a pharmaceutically acceptable salt or tautomer thereof. 46. A radiolabelled compound comprising the structure wherein X is I123, I125, I131 or Br76, or a pharmaceutically acceptable salt or tautomer thereof. 47. The radiolabelled compound of claim 45, for radioimaging or treating breast cancer or prostate cancer.
1,600
537
14,904,407
1,629
The present invention relates to a pharmaceutical combination comprising a cyclin dependent kinase (CDK) inhibitor represented by a compound of formula I (as described herein) or a pharmaceutically acceptable salt thereof; and at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor, for use in the treatment of melanoma. The present invention also relates to a method for the treatment of melanoma comprising administering to a subject in need thereof, a therapeutically effective amount of a CDK inhibitor and a therapeutically effective amount of at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor.
1. A method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a CDK (cyclin dependent kinase) inhibitor selected from the compound of formula I or a pharmaceutically acceptable salt thereof; wherein Ar is phenyl, which is substituted by 1 or 2 different substituents selected from chlorine and trifluoromethyl; and a therapeutically effective amount of at least one anticancer agent selected from a BRAF (serine-threonine protein kinase B-raf) inhibitor or a MEK (mitogen activated protein kinase) inhibitor. 2. The method according to claim 1, wherein the CDK inhibitor is a compound of formula I or a pharmaceutically acceptable salt thereof; wherein the phenyl group is substituted by 2 different substituents selected from chlorine and trifluoromethyl. 3. The method according to claim 2, wherein the CDK inhibitor is (+)-trans-2-(2-chloro-4-trifluoromethylphenyl)-5,7-dihydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-chromen-4-one hydrochloride (compound A, also referred to as voruciclib). 4. The method according to claim 1, wherein the BRAF inhibitor or the MEK inhibitor is an inhibitor of V600 mutated form of BRAF. 5. The method according to claim 4, wherein the BRAF inhibitor is an inhibitor of V600E mutated form of BRAF. 6. The method according to claim 1, wherein the BRAF inhibitor is selected from sorafenib, vemurafenib, GDC-0879, dabrafenib, PLX4720, BMS-908662, LGX818, PLX3603, ARQ-736, DP-4978 or RAF265. 7. The method according to any one of the claims 4 to 6, wherein the BRAF inhibitor is vemurafenib. 8. The method according to any one of the claims 4 to 6, wherein the BRAF inhibitor is dabrafenib. 9. The method according to claim 1 or claim 4, wherein the MEK inhibitor is an inhibitor of V600E or V600K mutated form of BRAF. 10. The method according to claim 1, wherein the MEK inhibitor is selected from selumetinib, binimetinib, PD-0325901, trametinib, cobimetinib, refametinib, pimasertib, TAK-733 or WX-554. 11. The method according to claim 9 or claim 10, wherein the MEK inhibitor is trametinib. 12. The method according to any one of the claims 1 to 11, wherein the said CDK inhibitor; and the said at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor; are administered simultaneously to a subject in need thereof. 13. The method according to any one of the claims 1 to 11, wherein the said CDK inhibitor and the said at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor; are administered sequentially to a subject in need thereof. 14. The method according to claim 1, wherein the melanoma is non-refractory melanoma. 15. The method according to claim 14, wherein the melanoma is non-refractory BRAF mutant melanoma. 16. The method according to claim 15, wherein the melanoma is non-refractory BRAFV600 mutant melanoma 17. The method according to claim 16, wherein the melanoma is non-refractory BRAFV600E or BRAFV600K mutant melanoma. 18. The method according to claim 1, wherein the melanoma is recurrent or refractory melanoma. 19. The method according to claim 18, wherein the melanoma is recurrent or refractory BRAF mutant melanoma. 20. The method according to claim 19, wherein the melanoma is recurrent or refractory BRAFV600 mutant melanoma. 21. The method according to claim 20, wherein the melanoma is recurrent or refractory BRAFV600E melanoma or BRAFV600K mutant melanoma. 22. The method according to claim 1, wherein the melanoma is metastatic melanoma. 23. The method according to claim 22, wherein the melanoma is metastatic BRAF mutant melanoma. 24. The method according to claim 23, wherein the melanoma is metastatic BRAFV600 mutant melanoma. 25. The method according to claim 24, wherein the melanoma is metastatic BRAFV600E melanoma or BRAFV600K mutant melanoma.
The present invention relates to a pharmaceutical combination comprising a cyclin dependent kinase (CDK) inhibitor represented by a compound of formula I (as described herein) or a pharmaceutically acceptable salt thereof; and at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor, for use in the treatment of melanoma. The present invention also relates to a method for the treatment of melanoma comprising administering to a subject in need thereof, a therapeutically effective amount of a CDK inhibitor and a therapeutically effective amount of at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor.1. A method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a CDK (cyclin dependent kinase) inhibitor selected from the compound of formula I or a pharmaceutically acceptable salt thereof; wherein Ar is phenyl, which is substituted by 1 or 2 different substituents selected from chlorine and trifluoromethyl; and a therapeutically effective amount of at least one anticancer agent selected from a BRAF (serine-threonine protein kinase B-raf) inhibitor or a MEK (mitogen activated protein kinase) inhibitor. 2. The method according to claim 1, wherein the CDK inhibitor is a compound of formula I or a pharmaceutically acceptable salt thereof; wherein the phenyl group is substituted by 2 different substituents selected from chlorine and trifluoromethyl. 3. The method according to claim 2, wherein the CDK inhibitor is (+)-trans-2-(2-chloro-4-trifluoromethylphenyl)-5,7-dihydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-chromen-4-one hydrochloride (compound A, also referred to as voruciclib). 4. The method according to claim 1, wherein the BRAF inhibitor or the MEK inhibitor is an inhibitor of V600 mutated form of BRAF. 5. The method according to claim 4, wherein the BRAF inhibitor is an inhibitor of V600E mutated form of BRAF. 6. The method according to claim 1, wherein the BRAF inhibitor is selected from sorafenib, vemurafenib, GDC-0879, dabrafenib, PLX4720, BMS-908662, LGX818, PLX3603, ARQ-736, DP-4978 or RAF265. 7. The method according to any one of the claims 4 to 6, wherein the BRAF inhibitor is vemurafenib. 8. The method according to any one of the claims 4 to 6, wherein the BRAF inhibitor is dabrafenib. 9. The method according to claim 1 or claim 4, wherein the MEK inhibitor is an inhibitor of V600E or V600K mutated form of BRAF. 10. The method according to claim 1, wherein the MEK inhibitor is selected from selumetinib, binimetinib, PD-0325901, trametinib, cobimetinib, refametinib, pimasertib, TAK-733 or WX-554. 11. The method according to claim 9 or claim 10, wherein the MEK inhibitor is trametinib. 12. The method according to any one of the claims 1 to 11, wherein the said CDK inhibitor; and the said at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor; are administered simultaneously to a subject in need thereof. 13. The method according to any one of the claims 1 to 11, wherein the said CDK inhibitor and the said at least one anticancer agent selected from a BRAF inhibitor or a MEK inhibitor; are administered sequentially to a subject in need thereof. 14. The method according to claim 1, wherein the melanoma is non-refractory melanoma. 15. The method according to claim 14, wherein the melanoma is non-refractory BRAF mutant melanoma. 16. The method according to claim 15, wherein the melanoma is non-refractory BRAFV600 mutant melanoma 17. The method according to claim 16, wherein the melanoma is non-refractory BRAFV600E or BRAFV600K mutant melanoma. 18. The method according to claim 1, wherein the melanoma is recurrent or refractory melanoma. 19. The method according to claim 18, wherein the melanoma is recurrent or refractory BRAF mutant melanoma. 20. The method according to claim 19, wherein the melanoma is recurrent or refractory BRAFV600 mutant melanoma. 21. The method according to claim 20, wherein the melanoma is recurrent or refractory BRAFV600E melanoma or BRAFV600K mutant melanoma. 22. The method according to claim 1, wherein the melanoma is metastatic melanoma. 23. The method according to claim 22, wherein the melanoma is metastatic BRAF mutant melanoma. 24. The method according to claim 23, wherein the melanoma is metastatic BRAFV600 mutant melanoma. 25. The method according to claim 24, wherein the melanoma is metastatic BRAFV600E melanoma or BRAFV600K mutant melanoma.
1,600
538
15,019,694
1,627
A gel paste composition including (A) a crosslinking organopolysiloxane and (B) a liquid oil, the crosslinking organopolysiloxane (A) being obtained by reacting an organohydrogenpolysiloxane shown by the following general formula (I) with a polyoxyalkylene compound shown by the following general formula (II) in the presence of a catalyst for hydrosilylation reaction, the component (A) containing polyoxyethylene units in an amount of 20 wt % or more. As a result, the gel paste composition that contains a crosslinking organopolysiloxane having improved compatibility with ethylhexyl methoxycinnamate and water, and thus exhibits excellent dispersibility when blended to a cosmetic thereby providing good feeling and cosmetic sustainability. R 1 a H b SiO (4-a-b)/2   (I) C c H 2c-1 O(C 2 H 4 O) d (C 3 H 6 O) e C c H 2c-1   (II).
1. A gel paste composition comprising (A) a crosslinking organopolysiloxane; and (B) a liquid oil, the crosslinking organopolysiloxane (A) being obtained by reacting an organohydrogenpolysiloxane shown by the following general formula (I) with a polyoxyalkylene compound shown by the following general formula (II) in the presence of a catalyst for hydrosilylation reaction, the component (A) containing polyoxyethylene units in an amount of 20 wt % or more, R1 aHbSiO(4-a-b)/2  (I) wherein each R1 may be the same or different and represents a substituted or unsubstituted monovalent hydrocarbon group having 1 to 30 carbon atoms and not having an alkenyl group; and “a” and “b” each represent a positive number satisfying 1.0≦a≦2.5, 0.001≦b≦1.2, and 1.0≦a+b≦2.6; CcH2c-1O(C2H4O)d(C3H6O)eCcH2c-1  (II) wherein “c” represents an integer of 2 to 6; and “d” and “e” each represent an integer satisfying 5≦d≦200 and 0≦e≦200. 2. The gel paste composition according to claim 1, wherein the crosslinking organopolysiloxane (A) is obtained by reacting the organohydrogenpolysiloxane shown by the general formula (I) with the polyoxyalkylene compound shown by the general formula (II) and a polyoxyalkylene compound shown by the following general formula (III) in the presence of the catalyst for hydrosilylation reaction, CcH2c-1O(C2H4O)d(C3H6O)e—R2  (III) wherein R2 represents a hydrogen atom, a monovalent hydrocarbon group having 1 to 10 carbon atoms, or —(CO)R3 where R3 represents an alkyl group having 1 to 5 carbon atoms; and “c”, “d”, and “e” have the same meanings as defined above. 3. The gel paste composition according to claim 1, wherein the crosslinking organopolysiloxane (A) is obtained by reacting the organohydrogenpolysiloxane shown by the general formula (I) with the polyoxyalkylene compound shown by the general formula (II) and an organopolysiloxane shown by the following general formula (IV) in the presence of the catalyst for hydrosilylation reaction, R1 iR4 jSiO(4-i-j)/2  (IV) wherein R1 has the same meaning as defined above; R4 represents an alkenyl group having 2 to 10 carbon atoms; “i” and “j” each represent a positive number satisfying 1.0≦i≦2.999, 0.001≦j≦1.5, and 1.001≦i+j≦3. 4. The gel paste composition according to claim 1, wherein the organohydrogenpolysiloxane used to obtain the crosslinking organopolysiloxane (A) is shown by the following general formula (V), wherein R1 has the same meaning as defined above; “g” and “h” represent an integer satisfying 0≦g≦300 and 0≦h≦300; “x” is 0 to 2; and h+x≧2. 5. The gel paste composition according to claim 2, wherein the organohydrogenpolysiloxane used to obtain the crosslinking organopolysiloxane (A) is shown by the following general formula (V), wherein R1 has the same meaning as defined above; “g” and “h” represent an integer satisfying 0≦g≦300 and 0≦h≦300; “x” is 0 to 2; and h+x≧2. 6. The gel paste composition according to claim 3, wherein the organohydrogenpolysiloxane used to obtain the crosslinking organopolysiloxane (A) is shown by the following general formula (V), wherein R1 has the same meaning as defined above; “g” and “h” represent an integer satisfying 0≦g≦300 and 0≦h≦300; “x” is 0 to 2; and h+x≧2. 7. The gel paste composition according to claim 1, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 8. The gel paste composition according to claim 2, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 9. The gel paste composition according to claim 3, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 10. The gel paste composition according to claim 4, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 11. The gel paste composition according to claim 5, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 12. The gel paste composition according to claim 6, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 13. A cosmetic comprising a gel paste composition according to claim 1. 14. The cosmetic according to claim 13, further comprising an oil component (C) other than the liquid oil (B). 15. The cosmetic according to claim 13, wherein the cosmetic is an emulsified cosmetic and contains water and a water-soluble polymer in a continuous phase. 16. The cosmetic according to claim 15, wherein the water-soluble polymer is an alkali-thickened vinyl polymer or an acrylamidosulfonic acid polymer. 17. The cosmetic according to claim 15, wherein the emulsified cosmetic contains a surface-hydrophobized powder in an oil phase. 18. The cosmetic according to claim 17, wherein the surface-hydrophobized powder is a hydrophobized pigment.
A gel paste composition including (A) a crosslinking organopolysiloxane and (B) a liquid oil, the crosslinking organopolysiloxane (A) being obtained by reacting an organohydrogenpolysiloxane shown by the following general formula (I) with a polyoxyalkylene compound shown by the following general formula (II) in the presence of a catalyst for hydrosilylation reaction, the component (A) containing polyoxyethylene units in an amount of 20 wt % or more. As a result, the gel paste composition that contains a crosslinking organopolysiloxane having improved compatibility with ethylhexyl methoxycinnamate and water, and thus exhibits excellent dispersibility when blended to a cosmetic thereby providing good feeling and cosmetic sustainability. R 1 a H b SiO (4-a-b)/2   (I) C c H 2c-1 O(C 2 H 4 O) d (C 3 H 6 O) e C c H 2c-1   (II).1. A gel paste composition comprising (A) a crosslinking organopolysiloxane; and (B) a liquid oil, the crosslinking organopolysiloxane (A) being obtained by reacting an organohydrogenpolysiloxane shown by the following general formula (I) with a polyoxyalkylene compound shown by the following general formula (II) in the presence of a catalyst for hydrosilylation reaction, the component (A) containing polyoxyethylene units in an amount of 20 wt % or more, R1 aHbSiO(4-a-b)/2  (I) wherein each R1 may be the same or different and represents a substituted or unsubstituted monovalent hydrocarbon group having 1 to 30 carbon atoms and not having an alkenyl group; and “a” and “b” each represent a positive number satisfying 1.0≦a≦2.5, 0.001≦b≦1.2, and 1.0≦a+b≦2.6; CcH2c-1O(C2H4O)d(C3H6O)eCcH2c-1  (II) wherein “c” represents an integer of 2 to 6; and “d” and “e” each represent an integer satisfying 5≦d≦200 and 0≦e≦200. 2. The gel paste composition according to claim 1, wherein the crosslinking organopolysiloxane (A) is obtained by reacting the organohydrogenpolysiloxane shown by the general formula (I) with the polyoxyalkylene compound shown by the general formula (II) and a polyoxyalkylene compound shown by the following general formula (III) in the presence of the catalyst for hydrosilylation reaction, CcH2c-1O(C2H4O)d(C3H6O)e—R2  (III) wherein R2 represents a hydrogen atom, a monovalent hydrocarbon group having 1 to 10 carbon atoms, or —(CO)R3 where R3 represents an alkyl group having 1 to 5 carbon atoms; and “c”, “d”, and “e” have the same meanings as defined above. 3. The gel paste composition according to claim 1, wherein the crosslinking organopolysiloxane (A) is obtained by reacting the organohydrogenpolysiloxane shown by the general formula (I) with the polyoxyalkylene compound shown by the general formula (II) and an organopolysiloxane shown by the following general formula (IV) in the presence of the catalyst for hydrosilylation reaction, R1 iR4 jSiO(4-i-j)/2  (IV) wherein R1 has the same meaning as defined above; R4 represents an alkenyl group having 2 to 10 carbon atoms; “i” and “j” each represent a positive number satisfying 1.0≦i≦2.999, 0.001≦j≦1.5, and 1.001≦i+j≦3. 4. The gel paste composition according to claim 1, wherein the organohydrogenpolysiloxane used to obtain the crosslinking organopolysiloxane (A) is shown by the following general formula (V), wherein R1 has the same meaning as defined above; “g” and “h” represent an integer satisfying 0≦g≦300 and 0≦h≦300; “x” is 0 to 2; and h+x≧2. 5. The gel paste composition according to claim 2, wherein the organohydrogenpolysiloxane used to obtain the crosslinking organopolysiloxane (A) is shown by the following general formula (V), wherein R1 has the same meaning as defined above; “g” and “h” represent an integer satisfying 0≦g≦300 and 0≦h≦300; “x” is 0 to 2; and h+x≧2. 6. The gel paste composition according to claim 3, wherein the organohydrogenpolysiloxane used to obtain the crosslinking organopolysiloxane (A) is shown by the following general formula (V), wherein R1 has the same meaning as defined above; “g” and “h” represent an integer satisfying 0≦g≦300 and 0≦h≦300; “x” is 0 to 2; and h+x≧2. 7. The gel paste composition according to claim 1, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 8. The gel paste composition according to claim 2, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 9. The gel paste composition according to claim 3, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 10. The gel paste composition according to claim 4, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 11. The gel paste composition according to claim 5, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 12. The gel paste composition according to claim 6, wherein the liquid oil (B) is selected from a silicone oil and an ester oil. 13. A cosmetic comprising a gel paste composition according to claim 1. 14. The cosmetic according to claim 13, further comprising an oil component (C) other than the liquid oil (B). 15. The cosmetic according to claim 13, wherein the cosmetic is an emulsified cosmetic and contains water and a water-soluble polymer in a continuous phase. 16. The cosmetic according to claim 15, wherein the water-soluble polymer is an alkali-thickened vinyl polymer or an acrylamidosulfonic acid polymer. 17. The cosmetic according to claim 15, wherein the emulsified cosmetic contains a surface-hydrophobized powder in an oil phase. 18. The cosmetic according to claim 17, wherein the surface-hydrophobized powder is a hydrophobized pigment.
1,600
539
12,444,516
1,653
The invention relates to a process for the production of biogas from biomass, in particular from lignocellulose-containing biomass. According to the invention bio gas is produced from lignocellulose-containing biomass in a process, wherein the cellulose and hemicellulose in the lignocellulose are made accessible for bioconversion.
1. Process for the production of biogas from a feed stream of lignocellulose-containing biomass comprising the steps of: i) subjecting the biomass stream to a temperature of 150-250° C. at a pH of 3-7; and ii) subjecting the product of step i) to anaerobic digestion, thereby producing a stream of biogas. 2. Process according to claim 1, wherein a stream of biomass containing a low fraction of lignocellulose is first fed to a pre-treatment step wherein it is subjected to anaerobic digestion to produce said feed stream that is fed to step i). 3. Process according to claim 2, wherein said pre-treatment step is carried out in the same apparatus as step ii) and wherein the product of step i) is recycled to step ii). 4. Process according to claim 1, wherein the pH in step i) is 4-5. 5. Process according to claim 1, wherein the temperature in step i) is 160-220° C. 6. Process according to claim 1, wherein the amount of sulphide in step i) is at most 1000 mg/l. 7. Process according to claim 1, wherein the amount of sulphide in step i) is at most 500 mg/l. 8. Process according to claim 1, wherein step i) lasts from 2-15 minutes. 9. Process according to claim 1, wherein step i) is carried out at a pressure equal to or higher than the saturation vapour pressure at the used temperature.
The invention relates to a process for the production of biogas from biomass, in particular from lignocellulose-containing biomass. According to the invention bio gas is produced from lignocellulose-containing biomass in a process, wherein the cellulose and hemicellulose in the lignocellulose are made accessible for bioconversion.1. Process for the production of biogas from a feed stream of lignocellulose-containing biomass comprising the steps of: i) subjecting the biomass stream to a temperature of 150-250° C. at a pH of 3-7; and ii) subjecting the product of step i) to anaerobic digestion, thereby producing a stream of biogas. 2. Process according to claim 1, wherein a stream of biomass containing a low fraction of lignocellulose is first fed to a pre-treatment step wherein it is subjected to anaerobic digestion to produce said feed stream that is fed to step i). 3. Process according to claim 2, wherein said pre-treatment step is carried out in the same apparatus as step ii) and wherein the product of step i) is recycled to step ii). 4. Process according to claim 1, wherein the pH in step i) is 4-5. 5. Process according to claim 1, wherein the temperature in step i) is 160-220° C. 6. Process according to claim 1, wherein the amount of sulphide in step i) is at most 1000 mg/l. 7. Process according to claim 1, wherein the amount of sulphide in step i) is at most 500 mg/l. 8. Process according to claim 1, wherein step i) lasts from 2-15 minutes. 9. Process according to claim 1, wherein step i) is carried out at a pressure equal to or higher than the saturation vapour pressure at the used temperature.
1,600
540
14,346,262
1,699
An apparatus for detecting antibodies in a patient sample includes at least one incubation channel for accommodating a patient strip and for incubating the patient strip with a patient sample, a conjugate and a substrate and a control mechanism for visual inspection of incubation quality. A separate calibration strip has at least two control zones which are implemented in such a way that, owing to incubation with a reference sample, a conjugate and a substrate, a control band becomes visible in each case in the control zones in such a way that the color intensities of the control bands differ. An evaluation unit generates a calibration curve taking the different color intensities into account and ascertains, on the basis of the calibration curve, a quality value for an antibody-pathogen protein reaction which took place on the patient strip and information about antibody concentration in the patient sample examined.
1-8. (canceled) 9. An apparatus for detecting antibodies in a patient sample, comprising: at least one incubation channel (3) for accommodating a patient strip (1) and for incubating the patient strip (1) with a patient sample, a conjugate and a substrate; a control means for visual inspection of incubation quality; an evaluation unit and a separate calibration strip (4) as the control means, the calibration strip (4) having at least two control zones which are implemented in such a way that, owing to incubation with a reference sample, a conjugate and a substrate, a control band becomes visible in each case in the control zones in such a way that the color intensities of the control bands differ, and wherein the evaluation unit is configured to generate a calibration curve taking different color intensities into account and, wherein said evaluation unit is further configured to ascertain, on the basis of the calibration curve, a quality value for an antibody-pathogen protein reaction which took place on the patient strip (1) and information about antibody concentration in the patient sample examined. 10. The apparatus according to claim 9, wherein the control bands in the at least two control zones on the calibration strip (4) appear in varying color intensity at the same antibody concentration in the patient sample. 11. The apparatus according to claim 9, wherein the control bands in the at least two control zones on the calibration strip (4) each react specifically to antibodies provided in a reference sample. 12. The apparatus according to claim 9, wherein the calibration strip (4) has a positive control zone which appears as a band once the calibration strip (4) has been incubated with patient serum, conjugate and substrate. 13. The apparatus according to claim 9, wherein the calibration strip (4) has a negative control zone which is coated more thinly with an antigen than in a positive control zone or in which the antigen is directed to an antibody not suspected in the patient serum. 14. The apparatus according to claim 9, wherein the calibration strip (4) has at least one conjugate control zone in which a band appears once the calibration strip (4) has been incubated with the conjugate. 15. The apparatus according to claim 9, wherein the calibration strip (4) has an IgM conjugate control zone, an IgG conjugate control zone and an IgA conjugate control zone in which a band appears in each case once the calibration strip has been incubated with an IgM conjugate, IgG conjugate and/or IgA conjugate. 16. A method for detecting antibodies in a patient sample, comprising the steps of: inserting a patient strip (1) into an incubation channel (3); incubating said patient strip (1) with a patient sample, a conjugate and a substrate; capturing incubation quality by visual inspection of a control means, wherein as control means, a separate calibration strip (4) having at least two control zones is inserted into a further incubation channel (3) and incubated with a reference sample, a conjugate and a substrate under the same experimental conditions as the patient strip (1), a control band becoming visible in each case in the control zones after the calibration strip (4) has been incubated, the color intensities of the bands differing; generating a calibration curve (5) taking the different color intensities into account, wherein the curve is compared with a color intensity of at least one band of the at least one incubated patient strip (1); and on the basis of the comparison, ascertaining a quality value for an antibody-pathogen protein reaction which took place on the patient strip (1) and ascertaining information about antibody concentration in the patient sample examined.
An apparatus for detecting antibodies in a patient sample includes at least one incubation channel for accommodating a patient strip and for incubating the patient strip with a patient sample, a conjugate and a substrate and a control mechanism for visual inspection of incubation quality. A separate calibration strip has at least two control zones which are implemented in such a way that, owing to incubation with a reference sample, a conjugate and a substrate, a control band becomes visible in each case in the control zones in such a way that the color intensities of the control bands differ. An evaluation unit generates a calibration curve taking the different color intensities into account and ascertains, on the basis of the calibration curve, a quality value for an antibody-pathogen protein reaction which took place on the patient strip and information about antibody concentration in the patient sample examined.1-8. (canceled) 9. An apparatus for detecting antibodies in a patient sample, comprising: at least one incubation channel (3) for accommodating a patient strip (1) and for incubating the patient strip (1) with a patient sample, a conjugate and a substrate; a control means for visual inspection of incubation quality; an evaluation unit and a separate calibration strip (4) as the control means, the calibration strip (4) having at least two control zones which are implemented in such a way that, owing to incubation with a reference sample, a conjugate and a substrate, a control band becomes visible in each case in the control zones in such a way that the color intensities of the control bands differ, and wherein the evaluation unit is configured to generate a calibration curve taking different color intensities into account and, wherein said evaluation unit is further configured to ascertain, on the basis of the calibration curve, a quality value for an antibody-pathogen protein reaction which took place on the patient strip (1) and information about antibody concentration in the patient sample examined. 10. The apparatus according to claim 9, wherein the control bands in the at least two control zones on the calibration strip (4) appear in varying color intensity at the same antibody concentration in the patient sample. 11. The apparatus according to claim 9, wherein the control bands in the at least two control zones on the calibration strip (4) each react specifically to antibodies provided in a reference sample. 12. The apparatus according to claim 9, wherein the calibration strip (4) has a positive control zone which appears as a band once the calibration strip (4) has been incubated with patient serum, conjugate and substrate. 13. The apparatus according to claim 9, wherein the calibration strip (4) has a negative control zone which is coated more thinly with an antigen than in a positive control zone or in which the antigen is directed to an antibody not suspected in the patient serum. 14. The apparatus according to claim 9, wherein the calibration strip (4) has at least one conjugate control zone in which a band appears once the calibration strip (4) has been incubated with the conjugate. 15. The apparatus according to claim 9, wherein the calibration strip (4) has an IgM conjugate control zone, an IgG conjugate control zone and an IgA conjugate control zone in which a band appears in each case once the calibration strip has been incubated with an IgM conjugate, IgG conjugate and/or IgA conjugate. 16. A method for detecting antibodies in a patient sample, comprising the steps of: inserting a patient strip (1) into an incubation channel (3); incubating said patient strip (1) with a patient sample, a conjugate and a substrate; capturing incubation quality by visual inspection of a control means, wherein as control means, a separate calibration strip (4) having at least two control zones is inserted into a further incubation channel (3) and incubated with a reference sample, a conjugate and a substrate under the same experimental conditions as the patient strip (1), a control band becoming visible in each case in the control zones after the calibration strip (4) has been incubated, the color intensities of the bands differing; generating a calibration curve (5) taking the different color intensities into account, wherein the curve is compared with a color intensity of at least one band of the at least one incubated patient strip (1); and on the basis of the comparison, ascertaining a quality value for an antibody-pathogen protein reaction which took place on the patient strip (1) and ascertaining information about antibody concentration in the patient sample examined.
1,600
541
15,317,831
1,619
Disclosed herein is a hair care product is provided, which comprises an oil-in-water emulsion A and an oil-in-water emulsion C, wherein emulsion A has a pH-value from greater than 7.0 to 12.0 and contains: a1) at least one cationic surfactant A, and a2) at least one amino-functionalized silicone A; and emulsion C has a pH-value from 2.0 to lower than 7.0, and contains: c1) at least one cationic surfactant C, wherein surfactant A and surfactant C may be identical with or different from each other, and c2) at least one silicone C, wherein silicone C and the amino-functionalized silicone A may be identical with or different from each other, wherein emulsion A and emulsion C are separated from each other before use. Also disclosed herein is a process for hair caring by using the hair care product as contemplated herein.
1. A hair care product comprising: an oil-in-water emulsion A, where emulsion A has a pH-value from greater than about 7.0 to about 12.0 and comprises: a1) at least one cationic surfactant A, and a2) at least one amino-functionalized silicone A; and an oil-in-water emulsion C, wherein emulsion C has a pH-value from about 2.0 to lower than about 7.0 and comprises: c1) at least one cationic surfactant C, wherein surfactant A and surfactant C may be identical with or different from each other, and c2) at least one silicone C, wherein silicone C and the amino-functionalized silicone A may be identical with or different from each other, wherein emulsion A and emulsion C are separated from each other before use. 2. The hair care product according to claim 1, wherein component a1) and component c1), independent from each other, are selected from quaternary ammonium compounds. 3. The hair care product according to claim 1, wherein component a2) is selected from amodimethicones and their derivatives. 4. The hair care product according to claim 1, wherein component c2) is different from component a2), and component c2) is selected from dialkylpolysiloxanes; methylphenylpolysiloxanes; cyclic silicones; amino-, fatty acid-, alcohol-, polyether-, epoxy-, fluorine-, glycoside- and alkyl-modified silicone compounds; alkyloxylated silicones; their derivatives and any combinations thereof. 5. The hair care product according to claim 1, wherein emulsion A comprises: about 0.1 to about 10.0 wt.-% of component a1), and about 0.1 to about 20 wt.-% of component a2), based on the total amount of 100 wt % emulsion A. 6. The hair care product according to claim 1, wherein emulsion C comprises: about 0.1 to about 10 wt.-% of component c1), and about 0.1 to about 25 wt.-% of component c2), based on the total amount of 100 wt % emulsion C. 7. The hair care product according to claim 1, wherein emulsion A and emulsion C, independent from each other, further comprise at least one thickener and/or at least one emollient. 8. The hair care product according to claim 1, further comprising an additional component which is separated from emulsion A and emulsion C before use, wherein the additional component is a hair care substance in the form of oil-in-water emulsion, aqueous solution, spray, hair mousse, hair gel or hair wax. 9. A process for hair caring comprising the following steps in sequence: step a) applying an oil-in-water emulsion A to the hair, where emulsion A has a pH-value from greater than about 7.0 to about 12.0 and comprises: a1) at least one cationic surfactant A, and a2) at least one amino-functionalized silicone A; step b) washing the hair, and step c) applying an oil-in-water emulsion C to the hair, wherein emulsion C has a pH-value from about 2.0 to lower than about 7.0 and comprises: c1) at least one cationic surfactant C, wherein surfactant A and surfactant C may be identical with or different from each other, and c2) at least one silicone C, wherein silicone C and the amino-functionalized silicone A may be identical with or different from each other. 10. The process according to claim 9, further comprising, between step a) and step b), step i) applying an additional component to the hair. 11. The process according to claim 9, wherein the hair is washed after step c). 12. The hair care product according to claim 1, wherein component a1) and component c1), independent from each other, are selected from quaternary ammonium halides, ester-quats and any combinations thereof. 13. The hair care product according to claim 1, wherein component a1) and component c1), independent from each other, are selected from quaternary ammonium halides which have one to three C12-C22 alkyl. 14. The hair care product according to claim 3, wherein component a2) is selected from amodimethicones, Bis(C13-15 Alkoxy) PG Amodimethicones, Bis-Hydroxy/Methoxy Amodimethicones, Aminopropyl phenyl trimethicones, Aminopropyl dimethicones, Bisamino PEG/PPG-41/3 Aminoethyl PG-Propyl Dimethicones, Caprylyl Methicones, and any combinations thereof. 15. The hair care product according to claim 4, wherein component c2) is selected from dialkylpolysiloxanes 16. The hair care product according to claim 15, wherein component c2) is selected from dimethicones. 17. The hair care product according to claim 1, wherein: component a1) and component c1), independent from each other, are selected from quaternary ammonium compounds; component a2) is selected from amodimethicones and their derivatives, component c2) is different from component a2), and component c2) is selected from dialkylpolysiloxanes; methylphenylpolysiloxanes; cyclic silicones; amino-, fatty acid-, alcohol-, polyether-, epoxy-, fluorine-, glycoside- and alkyl-modified silicone compounds; alkyloxylated silicones; their derivatives and any combinations thereof. 18. The hair care product according to claim 17, wherein: emulsion A comprises: about 0.1 to about 10.0 wt.-% of component a1), and about 0.1 to about 20 wt.-% of component a2), based on the total amount of 100 wt % emulsion A; and emulsion C comprises: about 0.1 to about 10 wt.-% of component c1), and about 0.1 to about 25 wt.-% of component c2), based on the total amount of 100 wt % emulsion C.
Disclosed herein is a hair care product is provided, which comprises an oil-in-water emulsion A and an oil-in-water emulsion C, wherein emulsion A has a pH-value from greater than 7.0 to 12.0 and contains: a1) at least one cationic surfactant A, and a2) at least one amino-functionalized silicone A; and emulsion C has a pH-value from 2.0 to lower than 7.0, and contains: c1) at least one cationic surfactant C, wherein surfactant A and surfactant C may be identical with or different from each other, and c2) at least one silicone C, wherein silicone C and the amino-functionalized silicone A may be identical with or different from each other, wherein emulsion A and emulsion C are separated from each other before use. Also disclosed herein is a process for hair caring by using the hair care product as contemplated herein.1. A hair care product comprising: an oil-in-water emulsion A, where emulsion A has a pH-value from greater than about 7.0 to about 12.0 and comprises: a1) at least one cationic surfactant A, and a2) at least one amino-functionalized silicone A; and an oil-in-water emulsion C, wherein emulsion C has a pH-value from about 2.0 to lower than about 7.0 and comprises: c1) at least one cationic surfactant C, wherein surfactant A and surfactant C may be identical with or different from each other, and c2) at least one silicone C, wherein silicone C and the amino-functionalized silicone A may be identical with or different from each other, wherein emulsion A and emulsion C are separated from each other before use. 2. The hair care product according to claim 1, wherein component a1) and component c1), independent from each other, are selected from quaternary ammonium compounds. 3. The hair care product according to claim 1, wherein component a2) is selected from amodimethicones and their derivatives. 4. The hair care product according to claim 1, wherein component c2) is different from component a2), and component c2) is selected from dialkylpolysiloxanes; methylphenylpolysiloxanes; cyclic silicones; amino-, fatty acid-, alcohol-, polyether-, epoxy-, fluorine-, glycoside- and alkyl-modified silicone compounds; alkyloxylated silicones; their derivatives and any combinations thereof. 5. The hair care product according to claim 1, wherein emulsion A comprises: about 0.1 to about 10.0 wt.-% of component a1), and about 0.1 to about 20 wt.-% of component a2), based on the total amount of 100 wt % emulsion A. 6. The hair care product according to claim 1, wherein emulsion C comprises: about 0.1 to about 10 wt.-% of component c1), and about 0.1 to about 25 wt.-% of component c2), based on the total amount of 100 wt % emulsion C. 7. The hair care product according to claim 1, wherein emulsion A and emulsion C, independent from each other, further comprise at least one thickener and/or at least one emollient. 8. The hair care product according to claim 1, further comprising an additional component which is separated from emulsion A and emulsion C before use, wherein the additional component is a hair care substance in the form of oil-in-water emulsion, aqueous solution, spray, hair mousse, hair gel or hair wax. 9. A process for hair caring comprising the following steps in sequence: step a) applying an oil-in-water emulsion A to the hair, where emulsion A has a pH-value from greater than about 7.0 to about 12.0 and comprises: a1) at least one cationic surfactant A, and a2) at least one amino-functionalized silicone A; step b) washing the hair, and step c) applying an oil-in-water emulsion C to the hair, wherein emulsion C has a pH-value from about 2.0 to lower than about 7.0 and comprises: c1) at least one cationic surfactant C, wherein surfactant A and surfactant C may be identical with or different from each other, and c2) at least one silicone C, wherein silicone C and the amino-functionalized silicone A may be identical with or different from each other. 10. The process according to claim 9, further comprising, between step a) and step b), step i) applying an additional component to the hair. 11. The process according to claim 9, wherein the hair is washed after step c). 12. The hair care product according to claim 1, wherein component a1) and component c1), independent from each other, are selected from quaternary ammonium halides, ester-quats and any combinations thereof. 13. The hair care product according to claim 1, wherein component a1) and component c1), independent from each other, are selected from quaternary ammonium halides which have one to three C12-C22 alkyl. 14. The hair care product according to claim 3, wherein component a2) is selected from amodimethicones, Bis(C13-15 Alkoxy) PG Amodimethicones, Bis-Hydroxy/Methoxy Amodimethicones, Aminopropyl phenyl trimethicones, Aminopropyl dimethicones, Bisamino PEG/PPG-41/3 Aminoethyl PG-Propyl Dimethicones, Caprylyl Methicones, and any combinations thereof. 15. The hair care product according to claim 4, wherein component c2) is selected from dialkylpolysiloxanes 16. The hair care product according to claim 15, wherein component c2) is selected from dimethicones. 17. The hair care product according to claim 1, wherein: component a1) and component c1), independent from each other, are selected from quaternary ammonium compounds; component a2) is selected from amodimethicones and their derivatives, component c2) is different from component a2), and component c2) is selected from dialkylpolysiloxanes; methylphenylpolysiloxanes; cyclic silicones; amino-, fatty acid-, alcohol-, polyether-, epoxy-, fluorine-, glycoside- and alkyl-modified silicone compounds; alkyloxylated silicones; their derivatives and any combinations thereof. 18. The hair care product according to claim 17, wherein: emulsion A comprises: about 0.1 to about 10.0 wt.-% of component a1), and about 0.1 to about 20 wt.-% of component a2), based on the total amount of 100 wt % emulsion A; and emulsion C comprises: about 0.1 to about 10 wt.-% of component c1), and about 0.1 to about 25 wt.-% of component c2), based on the total amount of 100 wt % emulsion C.
1,600
542
14,382,624
1,653
Provided is a pesticidal Flavobacterium strain and bioactive compositions and metabolites derived therefrom as well as their methods of use for controlling pests.
1. An isolated strain of a Flavobacterium sp. that has the following characteristics: (A) a 16S rRNA gene sequence comprising the forward sequence having at least 99% identity to the sequence set forth in SEQ ID NO:3 and a reverse sequence having at least 99% identity to the sequence set forth in SEQ ID NO:4 and a consensus sequence having at least 99% identity to the sequence set forth in SEQ ID NO:5; (B) pesticidal activity; (C) growth modulating activity; (D) produces a pesticidal compound having the following properties: (1) has a molecular weight of about 150-195 as determined by Liquid Chromatography/Mass Spectroscopy (LC/MS); (2) has 1H NMR values of δ 7.60, 7.52, 6.83, 6.68, 2.74, 1.14 and has 13C NMR values of 203.96, 161.90, 145.11, 131.78, 131.78, 127.28, 123.83, 117.24, 117.24, 34.52, 8.89; (3) has an High Pressure Liquid Chromatography (HPLC) retention time of about 8-14 minutes, on a reversed phase C-18 HPLC (Phenomenex, Luna 5μ C18(2) 100 A, 100×4.60 mm) column using a water:acetonitrile (CH3CN) with a gradient solvent system (0-20 min; 90-0% aqueous CH3CN, 20-24 min; 100% CH3CN, 24-27 min; 0-90% aqueous CH3CN, 27-30 min; 90% aqueous CH3CN) at 0.5 mL/min flow rate and UV detection of 210 nm.; (E) is non-pathogenic to vertebrate animals; (F) is susceptible to tetracycline, erythromycin, streptomycin, penicillin, ampicillin, oxytetracycline, chloramphenicol, ciprofloxacin, gentamicin, piperacillin, imipenem and sulphamethoxazole-trimethoprim or substantially pure filtrate, supernatant, or extract derived therefrom. 2. The strain of claim 1, wherein the Flavobacterium species is a Flavobacterium strain having the identifying characteristics of Flavobacterium sp. H492 (NRRL Accession No. B-50584). 3. A substantially pure culture or whole cell broth comprising the strain of claim 1 or cell fraction, filtrate, supernatant, or extract derived therefrom. 4. A combination comprising (a) a first substance wherein said first substance is (1) a substantially pure culture or whole cell broth comprising the strain of claim 1 or (2) cell fraction, filtrate, supernatant, compound and/or extract derived from the strain of claim 1 and (b) at least one or more second substances, wherein said second substance is a chemical or biological pesticide or growth promoting agent. 5. The combination according to claim 4, wherein said second substance is a pesticide selected from the group consisting of nematicide, fungicide and insecticide. 6. The combination according to claim 4, wherein the combination is a composition. 7. A composition comprising (a) a first substance selected from the group consisting of a substantially pure culture, whole cell broth, cell fraction, filtrate, supernatant and/or extract derived from the strain of claim 1 and (b) a carrier, diluent, surfactant a carrier, surfactant or adjuvant. 8. A method for modulating pest infestation in a plant and/or modulating growth of a plant comprising applying to the plant and/or seeds thereof and/or substrate used for growing said plant an amount of at least one of: (A) a substantially pure culture or whole cell broth comprising or cell fraction, filtrate, supernatant, compound and/or extract derived from the strain of claim 1; (B) the combination of claim 4; (C) a compound having the following properties: (1) has a molecular weight of about 150-195 as determined by Liquid Chromatography/Mass Spectroscopy (LC/MS); (2) has 1H NMR values of δ 7.60, 7.52, 6.83, 6.68, 2.74, 1.14 and has 13C NMR values of 203.96, 161.90, 145.11, 131.78, 131.78, 127.28, 123.83, 117.24, 117.24, 34.52, 8.89 (3) has an High Pressure Liquid Chromatography (HPLC) retention time of about 8-14 minutes, on a reversed phase C-18 HPLC (Phenomenex, Luna 5μ C18(2) 100 A, 100×4.60 mm) column using a water:acetonitrile (CH3CN) with a gradient solvent system (0-20 min; 90-0% aqueous CH3CN, 20-24 min; 100% CH3CN, 24-27 min; 0-90% aqueous CH3CN, 27-30 min; 90% aqueous CH3CN) at 0.5 mL/min flow rate and UV detection of 210 nm; (4) is obtainable from a Flavobacterium species and (5) is a polyketide effective to modulate said pest infestation and/or plant growth. 9. The method according to claim 8, wherein said pest is a nematode pest. 10. The method according to claim 8, wherein said plant is selected from the group consisting of strawberry, squash, cucumber, tomato, rose, pepper, cucumber, eggplant, grapevine, cotton, onion, garlic, wheat, soy, corn and rice. 11. The method according to claim 8, wherein the compound is selected from the group consisting of (A) a compound having the structure ##STR001## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R wherein —R is lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (B) a compound having the structure ##STR001a## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R, wherein R is a lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (C) a compound having the structure ##STR001b## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R wherein —R is lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4, R5, R6, R, are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (D) a compound having the structure ##STR001c## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R, wherein R is lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4, R5, R6, R, are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (E) a compound having the structure ##STR001c## or a pesticidally acceptable salt or steriosomers thereof, wherein X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4 are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl. 12. The method according to claim 11, wherein said compound is (E)-4-phenylbut-3-en-2-one, Benzalacetophenone, (E)-4-(2-hydroxyphenyl)but-3-en-2-one, (3E,5E)-6-phenylhexa-3,5-dien-2-one, (2E,4E)-1,5-diphenylpenta-2,4-dien-1-one, (E)-1-(2-hydroxyphenyl)pent-1-en-3-one, (E)-3-(2-hydroxyphenyl)-1-phenylprop-2-en-1-one, (E)-1-(2-hydroxyphenyl)-5-methylhex-1-en-3-one, (E)-3-(4-hydroxyphenyl)-1-phenylprop-2-en-1-one, (E)-1-(4-hydroxyphenyl)-5-methylhex-1-en-3-one, (E)-4-(3-hydroxyphenyl)but-3-en-2-one, (E)-4-(2-hydroxyphenyl)but-3-en-2-one, 2-(2′-phenyl-2-oxoethoxy)benzaldehyde, (E)-4-(2-hydroxyphenyl)-3-methylbut-3-en-2-one, (E)-1-phenylpent-2-en-1-one, (3E,7Z)-deca-3,7-dien-2-one, (E)-4-(3-hydroxyphenyl)but-3-en-2-one, (1E,4E)-1,5-bis(3-hydroxyphenyl)penta-1,4-dien-3-one, (E)-1,4-diphenylbut-2-en-1-one, (E)-1-(4-hydroxyphenyl)-4-phenylbut-2-en-1-one, (1E,4E)-1,5-bis(3-hydroxyphenyl)-2-methylpenta-1,4-dien-3-one, (E)-3-(3-hydroxyphenyl)-1-phenylprop-2-en-1-one, (E)-1-phenylpent-1-en-3-one, (E)-4-(4-hydroxyphenyl)-3-methylbut-3-en-2-one, (E)-5-methyl-1-phenylhex-1-en-3-one, 2-bromo-1-phenylethanone, (E)-1-(3-hydroxyphenyl)-5-methylhex-1-en-3-one, (E)-4-(4-hydroxyphenyl)but-3-en-2-one, 3-hydroxybenzaldehyde, 4-hydroxybenzaldehyde, 2-hydroxybenzaldehyde, benzaldehyde, gamma-dodecalactone, gamma-octalactone, 2-octanone, 2-heptanone, 2-undecanone, cis-4-heptenal, 4-methyl-2-pentanone, isobutyraldehyde, (E)-4-phenylbut-2-enal, (E)-4-(4-hydroxyphenyl)but-2-enal, (E)-4-(3-hydroxyphenyl)but-2-enal, (E)-4-(2-hydroxyphenyl) but-2-enal, 3-phenylpropanal, 3-(4-hydroxyphenyl)propanal, 3-(3-hydroxyphenyl)propanal, 3-(2-hydroxyphenyl)propanal, 2-phenylacetaldehyde, 2-(4-hydroxyphenyl)acetaldehyde, 2-(3-hydroxyphenyl)acetaldehyde, 2-(2-hydroxyphenyl)acetaldehyde, cinnamaldehyde, (E)-3-(4-hydroxyphenyl)acrylaldehyde, (E)-3-(3-hydroxyphenyl)acrylaldehyde, (E)-3-(2-hydroxyphenyl)acrylaldehyde, (2E,4E)-5-phenylpenta-2,4-dienal, (2E,4E)-5-(4-hydroxyphenyl)penta-2,4-dienal, (2E,4E)-5-(3-hydroxyphenyl)penta-2,4-dienal, (2E,4E)-5-(2-hydroxyphenyl)penta-2,4-dienal, pentan-2-one, hexan-2-one, 3-methylheptan-2-one 3-methyloctan-2-one, 3-methylnonan-2-one and stereoisomers thereof. 13. A method for obtaining a polyketide compound from a strain of Flavobacterium comprising (i) culturing the strain of claim 1 for a time sufficient to obtain said compound and (ii) isolating the compound produced in step (a) from the supernatant of said culture. 14. A seed comprising the combination of claim 4. 15. Use of at least one of: (a) a substantially pure culture, whole cell broth, cell fraction, filtrate, supernatant and/or extract derived from the strain of claim 1 or metabolite derived from said pure culture, whole cell broth, cell fraction, filtrate, supernatant and/or extract; (b) the combination of claim 4; (c) a compound having the following properties: (1) has a molecular weight of about 150-195 as determined by Liquid Chromatography/Mass Spectroscopy (LC/MS); (2) has 1H NMR values of δ 7.60, 7.52, 6.83, 6.68, 2.74, 1.14 and has 13C NMR values of 203.96, 161.90, 145.11, 131, 78, 131.78, 127.28, 123.83, 117.24, 117.24, 34.52, 8.89 (3) has an High Pressure Liquid Chromatography (HPLC) retention time of about 8-14 minutes, on a reversed phase C-18 HPLC (Phenomenex, Luna 5μ C18(2) 100 A, 100×4.60 mm) column using a water:acetonitrile (CH3CN) with a gradient solvent system (0-20 min; 90-0% aqueous CH3CN, 20-24 min; 100% CH3CN, 24-27 min; 0-90% aqueous CH3CN, 27-30 min; 90% aqueous CH3CN) at 0.5 mL/min flow rate and UV detection of 210 nm; (4) is obtainable from a Flavobacterium species and (5) is a polyketide and optionally at least one of: (b) a second substance, wherein said second substance is a chemical or biological pesticide, growth promoting agent or (c) a carrier, diluent, surfactant a carrier, surfactant or adjuvant to modulate said pest infestation and/or plant growth.
Provided is a pesticidal Flavobacterium strain and bioactive compositions and metabolites derived therefrom as well as their methods of use for controlling pests.1. An isolated strain of a Flavobacterium sp. that has the following characteristics: (A) a 16S rRNA gene sequence comprising the forward sequence having at least 99% identity to the sequence set forth in SEQ ID NO:3 and a reverse sequence having at least 99% identity to the sequence set forth in SEQ ID NO:4 and a consensus sequence having at least 99% identity to the sequence set forth in SEQ ID NO:5; (B) pesticidal activity; (C) growth modulating activity; (D) produces a pesticidal compound having the following properties: (1) has a molecular weight of about 150-195 as determined by Liquid Chromatography/Mass Spectroscopy (LC/MS); (2) has 1H NMR values of δ 7.60, 7.52, 6.83, 6.68, 2.74, 1.14 and has 13C NMR values of 203.96, 161.90, 145.11, 131.78, 131.78, 127.28, 123.83, 117.24, 117.24, 34.52, 8.89; (3) has an High Pressure Liquid Chromatography (HPLC) retention time of about 8-14 minutes, on a reversed phase C-18 HPLC (Phenomenex, Luna 5μ C18(2) 100 A, 100×4.60 mm) column using a water:acetonitrile (CH3CN) with a gradient solvent system (0-20 min; 90-0% aqueous CH3CN, 20-24 min; 100% CH3CN, 24-27 min; 0-90% aqueous CH3CN, 27-30 min; 90% aqueous CH3CN) at 0.5 mL/min flow rate and UV detection of 210 nm.; (E) is non-pathogenic to vertebrate animals; (F) is susceptible to tetracycline, erythromycin, streptomycin, penicillin, ampicillin, oxytetracycline, chloramphenicol, ciprofloxacin, gentamicin, piperacillin, imipenem and sulphamethoxazole-trimethoprim or substantially pure filtrate, supernatant, or extract derived therefrom. 2. The strain of claim 1, wherein the Flavobacterium species is a Flavobacterium strain having the identifying characteristics of Flavobacterium sp. H492 (NRRL Accession No. B-50584). 3. A substantially pure culture or whole cell broth comprising the strain of claim 1 or cell fraction, filtrate, supernatant, or extract derived therefrom. 4. A combination comprising (a) a first substance wherein said first substance is (1) a substantially pure culture or whole cell broth comprising the strain of claim 1 or (2) cell fraction, filtrate, supernatant, compound and/or extract derived from the strain of claim 1 and (b) at least one or more second substances, wherein said second substance is a chemical or biological pesticide or growth promoting agent. 5. The combination according to claim 4, wherein said second substance is a pesticide selected from the group consisting of nematicide, fungicide and insecticide. 6. The combination according to claim 4, wherein the combination is a composition. 7. A composition comprising (a) a first substance selected from the group consisting of a substantially pure culture, whole cell broth, cell fraction, filtrate, supernatant and/or extract derived from the strain of claim 1 and (b) a carrier, diluent, surfactant a carrier, surfactant or adjuvant. 8. A method for modulating pest infestation in a plant and/or modulating growth of a plant comprising applying to the plant and/or seeds thereof and/or substrate used for growing said plant an amount of at least one of: (A) a substantially pure culture or whole cell broth comprising or cell fraction, filtrate, supernatant, compound and/or extract derived from the strain of claim 1; (B) the combination of claim 4; (C) a compound having the following properties: (1) has a molecular weight of about 150-195 as determined by Liquid Chromatography/Mass Spectroscopy (LC/MS); (2) has 1H NMR values of δ 7.60, 7.52, 6.83, 6.68, 2.74, 1.14 and has 13C NMR values of 203.96, 161.90, 145.11, 131.78, 131.78, 127.28, 123.83, 117.24, 117.24, 34.52, 8.89 (3) has an High Pressure Liquid Chromatography (HPLC) retention time of about 8-14 minutes, on a reversed phase C-18 HPLC (Phenomenex, Luna 5μ C18(2) 100 A, 100×4.60 mm) column using a water:acetonitrile (CH3CN) with a gradient solvent system (0-20 min; 90-0% aqueous CH3CN, 20-24 min; 100% CH3CN, 24-27 min; 0-90% aqueous CH3CN, 27-30 min; 90% aqueous CH3CN) at 0.5 mL/min flow rate and UV detection of 210 nm; (4) is obtainable from a Flavobacterium species and (5) is a polyketide effective to modulate said pest infestation and/or plant growth. 9. The method according to claim 8, wherein said pest is a nematode pest. 10. The method according to claim 8, wherein said plant is selected from the group consisting of strawberry, squash, cucumber, tomato, rose, pepper, cucumber, eggplant, grapevine, cotton, onion, garlic, wheat, soy, corn and rice. 11. The method according to claim 8, wherein the compound is selected from the group consisting of (A) a compound having the structure ##STR001## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R wherein —R is lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (B) a compound having the structure ##STR001a## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R, wherein R is a lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (C) a compound having the structure ##STR001b## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R wherein —R is lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4, R5, R6, R, are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (D) a compound having the structure ##STR001c## or a pesticidally acceptable salt or steriosomers thereof, wherein Y is OH, SH, NR, OR, SR, R, wherein R is lower chain alkyl containing 1, 2, 3, 4, 5, 6, 7, 8 or 9 alkyl moieties, aryl or arylalkyl moiety, substituted lower alkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4, R5, R6, R, are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl; (E) a compound having the structure ##STR001c## or a pesticidally acceptable salt or steriosomers thereof, wherein X is O, NH, NR or S; n is 0, 1, 2, 3, 4, 5, 6, 7, 8 or 9; R1, R2, R3, R4 are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, substituted cycloalkyl, alkoxy, substituted alkoxy, thioalkyl, substituted thioalkyl, hydroxy, halogen, amino, amido, carboxyl, —C(O)H, acyl, oxyacyl, carbamate, sulfonyl, sulfonamide, or sulfuryl. 12. The method according to claim 11, wherein said compound is (E)-4-phenylbut-3-en-2-one, Benzalacetophenone, (E)-4-(2-hydroxyphenyl)but-3-en-2-one, (3E,5E)-6-phenylhexa-3,5-dien-2-one, (2E,4E)-1,5-diphenylpenta-2,4-dien-1-one, (E)-1-(2-hydroxyphenyl)pent-1-en-3-one, (E)-3-(2-hydroxyphenyl)-1-phenylprop-2-en-1-one, (E)-1-(2-hydroxyphenyl)-5-methylhex-1-en-3-one, (E)-3-(4-hydroxyphenyl)-1-phenylprop-2-en-1-one, (E)-1-(4-hydroxyphenyl)-5-methylhex-1-en-3-one, (E)-4-(3-hydroxyphenyl)but-3-en-2-one, (E)-4-(2-hydroxyphenyl)but-3-en-2-one, 2-(2′-phenyl-2-oxoethoxy)benzaldehyde, (E)-4-(2-hydroxyphenyl)-3-methylbut-3-en-2-one, (E)-1-phenylpent-2-en-1-one, (3E,7Z)-deca-3,7-dien-2-one, (E)-4-(3-hydroxyphenyl)but-3-en-2-one, (1E,4E)-1,5-bis(3-hydroxyphenyl)penta-1,4-dien-3-one, (E)-1,4-diphenylbut-2-en-1-one, (E)-1-(4-hydroxyphenyl)-4-phenylbut-2-en-1-one, (1E,4E)-1,5-bis(3-hydroxyphenyl)-2-methylpenta-1,4-dien-3-one, (E)-3-(3-hydroxyphenyl)-1-phenylprop-2-en-1-one, (E)-1-phenylpent-1-en-3-one, (E)-4-(4-hydroxyphenyl)-3-methylbut-3-en-2-one, (E)-5-methyl-1-phenylhex-1-en-3-one, 2-bromo-1-phenylethanone, (E)-1-(3-hydroxyphenyl)-5-methylhex-1-en-3-one, (E)-4-(4-hydroxyphenyl)but-3-en-2-one, 3-hydroxybenzaldehyde, 4-hydroxybenzaldehyde, 2-hydroxybenzaldehyde, benzaldehyde, gamma-dodecalactone, gamma-octalactone, 2-octanone, 2-heptanone, 2-undecanone, cis-4-heptenal, 4-methyl-2-pentanone, isobutyraldehyde, (E)-4-phenylbut-2-enal, (E)-4-(4-hydroxyphenyl)but-2-enal, (E)-4-(3-hydroxyphenyl)but-2-enal, (E)-4-(2-hydroxyphenyl) but-2-enal, 3-phenylpropanal, 3-(4-hydroxyphenyl)propanal, 3-(3-hydroxyphenyl)propanal, 3-(2-hydroxyphenyl)propanal, 2-phenylacetaldehyde, 2-(4-hydroxyphenyl)acetaldehyde, 2-(3-hydroxyphenyl)acetaldehyde, 2-(2-hydroxyphenyl)acetaldehyde, cinnamaldehyde, (E)-3-(4-hydroxyphenyl)acrylaldehyde, (E)-3-(3-hydroxyphenyl)acrylaldehyde, (E)-3-(2-hydroxyphenyl)acrylaldehyde, (2E,4E)-5-phenylpenta-2,4-dienal, (2E,4E)-5-(4-hydroxyphenyl)penta-2,4-dienal, (2E,4E)-5-(3-hydroxyphenyl)penta-2,4-dienal, (2E,4E)-5-(2-hydroxyphenyl)penta-2,4-dienal, pentan-2-one, hexan-2-one, 3-methylheptan-2-one 3-methyloctan-2-one, 3-methylnonan-2-one and stereoisomers thereof. 13. A method for obtaining a polyketide compound from a strain of Flavobacterium comprising (i) culturing the strain of claim 1 for a time sufficient to obtain said compound and (ii) isolating the compound produced in step (a) from the supernatant of said culture. 14. A seed comprising the combination of claim 4. 15. Use of at least one of: (a) a substantially pure culture, whole cell broth, cell fraction, filtrate, supernatant and/or extract derived from the strain of claim 1 or metabolite derived from said pure culture, whole cell broth, cell fraction, filtrate, supernatant and/or extract; (b) the combination of claim 4; (c) a compound having the following properties: (1) has a molecular weight of about 150-195 as determined by Liquid Chromatography/Mass Spectroscopy (LC/MS); (2) has 1H NMR values of δ 7.60, 7.52, 6.83, 6.68, 2.74, 1.14 and has 13C NMR values of 203.96, 161.90, 145.11, 131, 78, 131.78, 127.28, 123.83, 117.24, 117.24, 34.52, 8.89 (3) has an High Pressure Liquid Chromatography (HPLC) retention time of about 8-14 minutes, on a reversed phase C-18 HPLC (Phenomenex, Luna 5μ C18(2) 100 A, 100×4.60 mm) column using a water:acetonitrile (CH3CN) with a gradient solvent system (0-20 min; 90-0% aqueous CH3CN, 20-24 min; 100% CH3CN, 24-27 min; 0-90% aqueous CH3CN, 27-30 min; 90% aqueous CH3CN) at 0.5 mL/min flow rate and UV detection of 210 nm; (4) is obtainable from a Flavobacterium species and (5) is a polyketide and optionally at least one of: (b) a second substance, wherein said second substance is a chemical or biological pesticide, growth promoting agent or (c) a carrier, diluent, surfactant a carrier, surfactant or adjuvant to modulate said pest infestation and/or plant growth.
1,600
543
14,775,621
1,627
A drink product having pharmaceutical compositions as an active ingredients of at least one phosphorylated inositol, optionally Genistein, optionally Ubiquinol, and optionally additional unphosphorylated inositol. Uses for prevention, treatment, and reduction in risk of developing or progression of a number of conditions are disclosed. This invention relates to certain drink products that generally are aqueous solutions containing Genistein (optionally), at least one phosphorylated myoinositol having 1 to 9 phosphate groups (and/or any of the optical isomers thereof) optionally enriched with any or all of myoinositol, optical isomers of myoinositol, electrolytes, flavors, vitamins, free radical scavengers, and sweeteners.
1. An aqueous liquid formulation comprising: (a) water and (b) at least one phosphorylated member selected from the group consisting of phosphorylated myoinositol having from one to nine phosphate groups inclusive of pyrophosphate groups in which each pyrophosphate is counted as two of the one to nine phosphate groups and/or an optical isomer of said phosphorylated myoinositol, and/or an orally acceptable salt thereof, (c) optionally Genistein (d) optionally Ubiquinol, in an orally absorbable form (e) further optionally comprising one or more components selected from the group consisting of: (1) an unphosphorylated member selected from myoinositol and/or an optical isomer thereof; (2) an orally acceptable free radical scavenger other than either of Genistein and Ubiquinol (3) a nutritionally acceptable orally administrable electrolyte; (4) a nutritionally acceptable orally administrable vitamin; (5) a flavor; (6) an orally administrable coloring agent; (7) an orally administrable sweetener; (8) an oral formulation acceptable thickener; (9) an orally administrable, liquid formulation processing aid; and (10) an orally administrable, liquid formulation auxiliary carrier other than water. 2. The formulation of claim 1 wherein the at least one phosphorylated member is selected from a tetramonophosphate, a pentamonophosphate, a hexamonophosphate, a pentamonophospho-monopyrophosphate, and a tetramonophospho-dipyryophosphate of (a) myoinositol or (b) an optical isomer of myoinositol. 3. The formulation of claim 1 wherein when said unphosphorylated inositol or optical isomer thereof is present, it is in the form of the inositol isomer that is present in at least one of the phosphorylated members when such phosphorylated members are viewed without consideration of the phosphorylations involved. 4. The formulation of claim 1 wherein when the unphosphorylated inositol or optical isomer thereof is present, it is selected from those members having a different inositol isomeric form from that in any of the phosphorylated members that are present when such phosphorylated members are viewed without consideration of the phosphorylations involved. 5. A method of reducing the risk of damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 6. A method of preventing damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 7. A method of treating damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 8. A method of reducing the risk of developing HIV and/or AIDS and secondary cancers related to HIV and AIDS comprising orally administering the formulation of claim 1. 9. The method of claim 5 wherein said cancers related to HIV and AIDS is selected from Kaposi sarcoma and non-Hodgkin lymphoma, Hodgkin disease and cancers of the lung, mouth, skin, cervix, and digestive system, liver, blood, soft tissue, and muscular tumors. 10. A method of preventing a HIV/AIDS related cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 11. A method of treating a cancer selected from Kaposi sarcoma and non-Hodgkin lymphoma, other AIDS-related cancers selected from Hodgkin disease and cancers of the lung, mouth, skin, cervix, and digestive system, liver, blood, soft tissue, and muscular tumors breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 12. A method of increasing T cells and or inhibiting HIV related CD4 reduction comprising administering the composition of claim 1 to a subject in need thereof. 13. A method for blocking the communication between a HIV cell surface to internal cellular compartment comprising administering the composition of claim 1 to a subject in need thereof. 14. A method for preventing an HIV particle from entering the internal cellular compartments thereby preventing the spread of the infection comprising administering the composition of claim 1 to a subject in need thereof. 15. A method of reducing the risk of adverse effects of common and current drug toxicities' associated with current anti-viral therapies comprising administering the composition of claim 1 to a subject in need thereof. 16. A method of reducing drug resistance known to occur with current drugs because it's a plant based product comprising administering the composition of claim 1 to a subject in need thereof. 17. A method for both prevention HIV infection of resting CD4 T-cells, viral DNA synthesis, and/or viral nuclear migration comprising administering the composition of claim 1 to a subject in need thereof. 18. A method for preventing Gag-NA chaperone, inositol does that interaction prior to membrane binding in HIV comprising administering the composition of claim 1 to a subject in need thereof. 19. A method for inhibiting the replication of HIV-1 comprising administering the composition of claim 1 to a subject in need thereof. 20. A method for AP3B1 regulation and HIV-1 Gag release comprising administering the composition of claim 1 to a subject in need thereof. 21. The formulation of claim 1 which is a liquid nutritional supplement. 22. The formulation of claim 1 which is an orally administered product. 23. A method of reducing the risk of damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 24. A method of preventing damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 25. A method of treating damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 26. A method of reducing the risk of developing a cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, wet tumors, multiple myeloma, brain, throat, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 27. A method of preventing cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, wet tumors, multiple myeloma, brain, throat, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 28. A method of treating a cancer selected from cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, wet tumors, multiple myeloma, brain, throat, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 29. A method of reducing adverse effects of diagnostic radiation treatments in a patient in need thereof comprising orally administering to such a patient, before, during, or after said diagnostic radiation treatment, the formulation of claim 1. 30. A method of reducing the risk of adverse effects of environmental radiation exposure to a human or animal in need thereof comprising administering to said human or said animal respectively, before, during, or after said environmental, radiation exposure, the composition of claim 1. 31. The method of claim 30 wherein said environmental radiation exposure is selected from the group consisting of: a) extreme elevation above sea level; b) flying; c) going into planetary orbit or further into space; and d) mining, purification, or handling of radioactive materials. 32. A method of reducing the risk of adverse effects of environmental exposure to at least one of air pollutants, cigarette smoke, and/or other free oxygen radical generating substances in a human or animal comprising administering to said human or said animal respectively, before, during, or after said exposure, the composition of claim 1. 33. A method of reducing the risk of or treating a neurodegenerative disease related to environmental condition exposure comprising administering the composition of claim 1. 34. The method of claim 33, wherein the neurodegenerative disease is Alzheimer's Disease and/or brain cancer. 35. A method of prophylactic treatment of a subject to prevent or slow the progression of an HIV infection wherein said subject is known to be or suspected to be at risk of an HIV infection due to actual or suspected exposure to HIV virus or has been determined to have a detectable titer of HIV/AID virus or surrogate markers therefor and has not yet presented with clinical symptoms of HIV/AIDS comprising administering a composition of claim 1 to said subject. 36. The aqueous liquid formulation of claim 1 wherein said Ubiquinol is present. 37. The aqueous liquid formulation of claim 36 wherein said Ubiquinol is in the form of Qunol Liquid Co Q10 or other form as set forth in U.S. Pat. No. 6,455,072. 38. The formulation of claim 1 wherein said phosphorylated member is myoinositol hexaphosphate or optical isomer thereof and said unphosphorylated myoinositol or optical isomer thereof is present and is unphosphorylated myoinositol. 39. The formulation of claim 38 or the composition of claim 38 wherein said phosphorylated member is myoinositol hexaphosphate and said unphosphorylated myoinositol or optical isomer thereof is myoinositol per se. 40. A composition comprising: (a) water and (b) at least one phosphorylated member selected from the group consisting of phosphorylated myoinositol having from one to nine phosphate groups inclusive of pyrophosphate groups in which each pyrophosphate is counted as two of the one to nine phosphate groups and/or an optical isomer of said phosphorylated myoinositol, and/or an orally acceptable salt thereof, (c) optionally Genistein (d) optionally Ubiquinol, in an orally absorbable form (e) further optionally comprising one or more components selected from the group consisting of: (1) an unphosphorylated member selected from myoinositol and/or an optical isomer thereof; (2) an orally acceptable free radical scavenger other than either of Genistein and Ubiquinol (3) a nutritionally acceptable orally administrable electrolyte; (4) a nutritionally acceptable orally administrable vitamin; (5) a flavor; (6) an orally administrable coloring agent; (7) an orally administrable sweetener; (8) an oral composition acceptable thickener; (9) an orally administrable, liquid formulation processing aid; and (10) an orally administrable, liquid formulation auxiliary carrier other than water. 41. The composition of claim 40 wherein the at least one phosphorylated member is selected from a tetramonophosphate, a pentamonophosphate, a hexamonophosphate, a pentamonophospho-monopyrophosphate, and a tetramonophospho-dipyryophosphate of (a) myoinositol or (b) an optical isomer of myoinositol. 42. The composition of claim 40 wherein when said unphosphorylated inositol or optical isomer thereof is present, it is in the form of the inositol isomer that is present in at least one of the phosphorylated members when such phosphorylated members are viewed without consideration of the phosphorylations involved. 43. The composition of claim 40 wherein when the unphosphorylated inositol or optical isomer thereof is present, it is selected from those members having a different inositol isomeric form from that in any of the phosphorylated members that are present when such phosphorylated members are viewed without consideration of the phosphorylations involved. 44. The composition of claim 40 wherein said Ubiquinol is present. 45. The composition of claim 44 wherein said Ubiquinol is in the form of Qunol Liquid Co Q10 or other form as set forth in U.S. Pat. No. 6,455,072. 46. The composition of claim 40 wherein said phosphorylated member is myoinositol hexaphosphate or optical isomer thereof and said unphosphorylated myoinositol or optical isomer thereof is present and is unphosphorylated myoinositol. 47. The composition of claim 49 wherein said phosphorylated member is myoinositol hexaphosphate and said unphosphorylated myoinositol or optical isomer thereof is myoinositol per se. 48. A method of making a formulation of claim 1 comprising dissolving the claim 1 components into an aqueous based carrier. 49. A method of making a composition of claim 40 comprising dissolving the claim 40 components into an aqueous based carrier.
A drink product having pharmaceutical compositions as an active ingredients of at least one phosphorylated inositol, optionally Genistein, optionally Ubiquinol, and optionally additional unphosphorylated inositol. Uses for prevention, treatment, and reduction in risk of developing or progression of a number of conditions are disclosed. This invention relates to certain drink products that generally are aqueous solutions containing Genistein (optionally), at least one phosphorylated myoinositol having 1 to 9 phosphate groups (and/or any of the optical isomers thereof) optionally enriched with any or all of myoinositol, optical isomers of myoinositol, electrolytes, flavors, vitamins, free radical scavengers, and sweeteners.1. An aqueous liquid formulation comprising: (a) water and (b) at least one phosphorylated member selected from the group consisting of phosphorylated myoinositol having from one to nine phosphate groups inclusive of pyrophosphate groups in which each pyrophosphate is counted as two of the one to nine phosphate groups and/or an optical isomer of said phosphorylated myoinositol, and/or an orally acceptable salt thereof, (c) optionally Genistein (d) optionally Ubiquinol, in an orally absorbable form (e) further optionally comprising one or more components selected from the group consisting of: (1) an unphosphorylated member selected from myoinositol and/or an optical isomer thereof; (2) an orally acceptable free radical scavenger other than either of Genistein and Ubiquinol (3) a nutritionally acceptable orally administrable electrolyte; (4) a nutritionally acceptable orally administrable vitamin; (5) a flavor; (6) an orally administrable coloring agent; (7) an orally administrable sweetener; (8) an oral formulation acceptable thickener; (9) an orally administrable, liquid formulation processing aid; and (10) an orally administrable, liquid formulation auxiliary carrier other than water. 2. The formulation of claim 1 wherein the at least one phosphorylated member is selected from a tetramonophosphate, a pentamonophosphate, a hexamonophosphate, a pentamonophospho-monopyrophosphate, and a tetramonophospho-dipyryophosphate of (a) myoinositol or (b) an optical isomer of myoinositol. 3. The formulation of claim 1 wherein when said unphosphorylated inositol or optical isomer thereof is present, it is in the form of the inositol isomer that is present in at least one of the phosphorylated members when such phosphorylated members are viewed without consideration of the phosphorylations involved. 4. The formulation of claim 1 wherein when the unphosphorylated inositol or optical isomer thereof is present, it is selected from those members having a different inositol isomeric form from that in any of the phosphorylated members that are present when such phosphorylated members are viewed without consideration of the phosphorylations involved. 5. A method of reducing the risk of damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 6. A method of preventing damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 7. A method of treating damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 8. A method of reducing the risk of developing HIV and/or AIDS and secondary cancers related to HIV and AIDS comprising orally administering the formulation of claim 1. 9. The method of claim 5 wherein said cancers related to HIV and AIDS is selected from Kaposi sarcoma and non-Hodgkin lymphoma, Hodgkin disease and cancers of the lung, mouth, skin, cervix, and digestive system, liver, blood, soft tissue, and muscular tumors. 10. A method of preventing a HIV/AIDS related cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 11. A method of treating a cancer selected from Kaposi sarcoma and non-Hodgkin lymphoma, other AIDS-related cancers selected from Hodgkin disease and cancers of the lung, mouth, skin, cervix, and digestive system, liver, blood, soft tissue, and muscular tumors breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 12. A method of increasing T cells and or inhibiting HIV related CD4 reduction comprising administering the composition of claim 1 to a subject in need thereof. 13. A method for blocking the communication between a HIV cell surface to internal cellular compartment comprising administering the composition of claim 1 to a subject in need thereof. 14. A method for preventing an HIV particle from entering the internal cellular compartments thereby preventing the spread of the infection comprising administering the composition of claim 1 to a subject in need thereof. 15. A method of reducing the risk of adverse effects of common and current drug toxicities' associated with current anti-viral therapies comprising administering the composition of claim 1 to a subject in need thereof. 16. A method of reducing drug resistance known to occur with current drugs because it's a plant based product comprising administering the composition of claim 1 to a subject in need thereof. 17. A method for both prevention HIV infection of resting CD4 T-cells, viral DNA synthesis, and/or viral nuclear migration comprising administering the composition of claim 1 to a subject in need thereof. 18. A method for preventing Gag-NA chaperone, inositol does that interaction prior to membrane binding in HIV comprising administering the composition of claim 1 to a subject in need thereof. 19. A method for inhibiting the replication of HIV-1 comprising administering the composition of claim 1 to a subject in need thereof. 20. A method for AP3B1 regulation and HIV-1 Gag release comprising administering the composition of claim 1 to a subject in need thereof. 21. The formulation of claim 1 which is a liquid nutritional supplement. 22. The formulation of claim 1 which is an orally administered product. 23. A method of reducing the risk of damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 24. A method of preventing damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 25. A method of treating damage to cells or tissues due to reactive oxygen species free radicals in a subject in need thereof comprising orally administering the formulation of claim 1. 26. A method of reducing the risk of developing a cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, wet tumors, multiple myeloma, brain, throat, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 27. A method of preventing cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, wet tumors, multiple myeloma, brain, throat, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 28. A method of treating a cancer selected from cancer selected from breast, pancreatic, ovarian, prostate, lung cancer, skin cancer, colon cancer liver, cervical, uterine, liver, blood, wet tumors, multiple myeloma, brain, throat, soft tissue, and muscular tumors in a subject in need thereof comprising orally administering the formulation of claim 1. 29. A method of reducing adverse effects of diagnostic radiation treatments in a patient in need thereof comprising orally administering to such a patient, before, during, or after said diagnostic radiation treatment, the formulation of claim 1. 30. A method of reducing the risk of adverse effects of environmental radiation exposure to a human or animal in need thereof comprising administering to said human or said animal respectively, before, during, or after said environmental, radiation exposure, the composition of claim 1. 31. The method of claim 30 wherein said environmental radiation exposure is selected from the group consisting of: a) extreme elevation above sea level; b) flying; c) going into planetary orbit or further into space; and d) mining, purification, or handling of radioactive materials. 32. A method of reducing the risk of adverse effects of environmental exposure to at least one of air pollutants, cigarette smoke, and/or other free oxygen radical generating substances in a human or animal comprising administering to said human or said animal respectively, before, during, or after said exposure, the composition of claim 1. 33. A method of reducing the risk of or treating a neurodegenerative disease related to environmental condition exposure comprising administering the composition of claim 1. 34. The method of claim 33, wherein the neurodegenerative disease is Alzheimer's Disease and/or brain cancer. 35. A method of prophylactic treatment of a subject to prevent or slow the progression of an HIV infection wherein said subject is known to be or suspected to be at risk of an HIV infection due to actual or suspected exposure to HIV virus or has been determined to have a detectable titer of HIV/AID virus or surrogate markers therefor and has not yet presented with clinical symptoms of HIV/AIDS comprising administering a composition of claim 1 to said subject. 36. The aqueous liquid formulation of claim 1 wherein said Ubiquinol is present. 37. The aqueous liquid formulation of claim 36 wherein said Ubiquinol is in the form of Qunol Liquid Co Q10 or other form as set forth in U.S. Pat. No. 6,455,072. 38. The formulation of claim 1 wherein said phosphorylated member is myoinositol hexaphosphate or optical isomer thereof and said unphosphorylated myoinositol or optical isomer thereof is present and is unphosphorylated myoinositol. 39. The formulation of claim 38 or the composition of claim 38 wherein said phosphorylated member is myoinositol hexaphosphate and said unphosphorylated myoinositol or optical isomer thereof is myoinositol per se. 40. A composition comprising: (a) water and (b) at least one phosphorylated member selected from the group consisting of phosphorylated myoinositol having from one to nine phosphate groups inclusive of pyrophosphate groups in which each pyrophosphate is counted as two of the one to nine phosphate groups and/or an optical isomer of said phosphorylated myoinositol, and/or an orally acceptable salt thereof, (c) optionally Genistein (d) optionally Ubiquinol, in an orally absorbable form (e) further optionally comprising one or more components selected from the group consisting of: (1) an unphosphorylated member selected from myoinositol and/or an optical isomer thereof; (2) an orally acceptable free radical scavenger other than either of Genistein and Ubiquinol (3) a nutritionally acceptable orally administrable electrolyte; (4) a nutritionally acceptable orally administrable vitamin; (5) a flavor; (6) an orally administrable coloring agent; (7) an orally administrable sweetener; (8) an oral composition acceptable thickener; (9) an orally administrable, liquid formulation processing aid; and (10) an orally administrable, liquid formulation auxiliary carrier other than water. 41. The composition of claim 40 wherein the at least one phosphorylated member is selected from a tetramonophosphate, a pentamonophosphate, a hexamonophosphate, a pentamonophospho-monopyrophosphate, and a tetramonophospho-dipyryophosphate of (a) myoinositol or (b) an optical isomer of myoinositol. 42. The composition of claim 40 wherein when said unphosphorylated inositol or optical isomer thereof is present, it is in the form of the inositol isomer that is present in at least one of the phosphorylated members when such phosphorylated members are viewed without consideration of the phosphorylations involved. 43. The composition of claim 40 wherein when the unphosphorylated inositol or optical isomer thereof is present, it is selected from those members having a different inositol isomeric form from that in any of the phosphorylated members that are present when such phosphorylated members are viewed without consideration of the phosphorylations involved. 44. The composition of claim 40 wherein said Ubiquinol is present. 45. The composition of claim 44 wherein said Ubiquinol is in the form of Qunol Liquid Co Q10 or other form as set forth in U.S. Pat. No. 6,455,072. 46. The composition of claim 40 wherein said phosphorylated member is myoinositol hexaphosphate or optical isomer thereof and said unphosphorylated myoinositol or optical isomer thereof is present and is unphosphorylated myoinositol. 47. The composition of claim 49 wherein said phosphorylated member is myoinositol hexaphosphate and said unphosphorylated myoinositol or optical isomer thereof is myoinositol per se. 48. A method of making a formulation of claim 1 comprising dissolving the claim 1 components into an aqueous based carrier. 49. A method of making a composition of claim 40 comprising dissolving the claim 40 components into an aqueous based carrier.
1,600
544
16,010,995
1,616
An oleogel comprising a non-polar liquid and a powder containing triterpene is provided as an oleogel that may be used for healing wounds.
1. A method for enhancing the rate of reepithelialization of a wound, comprising administering to the wound an oleogel comprising a triterpene fraction, wherein the triterpene fraction comprises betulin. 2. The method of claim 1, wherein the triterpene fraction comprises between 1 weight-percent and 20 weight-percent of the oleogel. 3. The method of claim 2, wherein the triterpene fraction comprises between 6 weight-percent and 12 weight-percent of the oleogel. 4. The method of claim 1, wherein the fraction of betulin in the triterpene fraction is greater than 60 weight-percent. 5. The method of claim 1, wherein the fraction of betulin in the triterpene fraction is greater than 80 weight-percent. 6. The method of claim 1, wherein the triterpene fraction further comprises lupeol. 7. The method of claim 6, wherein the joint fraction of betulin and lupeol in the triterpene fraction is greater than 80 weight-percent in the triterpene fraction. 8. The method of claim 1, wherein the joint fraction of betulin and lupeol in the triterpene fraction is greater than 85 weight-percent in the triterpene fraction. 9. The method of claim 1, wherein the oleogel further comprises a nonpolar liquid. 10. The method of claim 9, wherein the nonpolar liquid is a plant, animal, mineral, or synthetic oil. 11. The method of claim 10, wherein the plant oil is a vegetable oil which is selected from one of the following: sunflower oil, olive oil, avocado oil, and almond oil. 12. The method of claim 11, wherein the nonpolar liquid consists of sunflower oil. 13. The method of claim 9, wherein the nonpolar liquid fraction comprises between 80 weight-percent and 99 weight-percent in relation to the total weight of the oleogel, and wherein the triterpene fraction comprises between 1 weight-percent and 20 weight-percent in relation to the total weight of the oleogel. 14. The method of claim 1, wherein the oleogel further comprises betulinic acid, oleanolic acid, and erythrodiol. 15. The method of claim 1, wherein the method results in complete healing of the wound. 16. A method for healing a wound caused by a skin disease, comprising administering to the wound an oleogel comprising a triterpene fraction, wherein the triterpene fraction comprises betulin. 17. A method for healing a wound caused by a therapeutic intervention, comprising administering to the wound an oleogel comprising a triterpene fraction, wherein the triterpene fraction comprises betulin.
An oleogel comprising a non-polar liquid and a powder containing triterpene is provided as an oleogel that may be used for healing wounds.1. A method for enhancing the rate of reepithelialization of a wound, comprising administering to the wound an oleogel comprising a triterpene fraction, wherein the triterpene fraction comprises betulin. 2. The method of claim 1, wherein the triterpene fraction comprises between 1 weight-percent and 20 weight-percent of the oleogel. 3. The method of claim 2, wherein the triterpene fraction comprises between 6 weight-percent and 12 weight-percent of the oleogel. 4. The method of claim 1, wherein the fraction of betulin in the triterpene fraction is greater than 60 weight-percent. 5. The method of claim 1, wherein the fraction of betulin in the triterpene fraction is greater than 80 weight-percent. 6. The method of claim 1, wherein the triterpene fraction further comprises lupeol. 7. The method of claim 6, wherein the joint fraction of betulin and lupeol in the triterpene fraction is greater than 80 weight-percent in the triterpene fraction. 8. The method of claim 1, wherein the joint fraction of betulin and lupeol in the triterpene fraction is greater than 85 weight-percent in the triterpene fraction. 9. The method of claim 1, wherein the oleogel further comprises a nonpolar liquid. 10. The method of claim 9, wherein the nonpolar liquid is a plant, animal, mineral, or synthetic oil. 11. The method of claim 10, wherein the plant oil is a vegetable oil which is selected from one of the following: sunflower oil, olive oil, avocado oil, and almond oil. 12. The method of claim 11, wherein the nonpolar liquid consists of sunflower oil. 13. The method of claim 9, wherein the nonpolar liquid fraction comprises between 80 weight-percent and 99 weight-percent in relation to the total weight of the oleogel, and wherein the triterpene fraction comprises between 1 weight-percent and 20 weight-percent in relation to the total weight of the oleogel. 14. The method of claim 1, wherein the oleogel further comprises betulinic acid, oleanolic acid, and erythrodiol. 15. The method of claim 1, wherein the method results in complete healing of the wound. 16. A method for healing a wound caused by a skin disease, comprising administering to the wound an oleogel comprising a triterpene fraction, wherein the triterpene fraction comprises betulin. 17. A method for healing a wound caused by a therapeutic intervention, comprising administering to the wound an oleogel comprising a triterpene fraction, wherein the triterpene fraction comprises betulin.
1,600
545
14,396,249
1,652
The dairy industry today faces a problem of providing an alternative to adding sweeteners to fermented milk products in order to achieve the desired sweet taste without the added calories. Furthermore, it would be highly advantageous to establish a method for reducing lactose in fermented milk products to a level which is acceptable for lactose-intolerant consumers. The above problems have been solved by providing mutant Streptococcus thermophilus strains and mutant Lactobacillus delbrueckii subsp. bulgaricus strains that excrete glucose to the milk when the milk is inoculated and fermented with such Streptococcus thermophilus strains and Lactobacillus delbrueckii subsp, bulgaricus strains. Thus, the present invention relates to strains of Streptococcus thermophilus and Lactobacillus delbrueckii subsp, bulgaricus which secrete glucose to the milk substrate during fermentation, as well as to mixed cultures comprising the Streptococcus thermophilus strains and the Lactobacillus delbrueckii subsp, bulgaricus strains, starter cultures comprising the strains and dairy products manufactured with the cultures. The present method also relates to use of the strains for decreasing the lactose content of a fermented food product and for boosting growth of the probiotic BB-12®.
1. A galactose-fermenting Streptococcus thermophilus strain, wherein the strain carries a mutation in the DNA sequence of the glcK gene encoding a glucokinase protein, wherein the mutation inactivates the glucokinase protein or has a negative effect on expression of the gene. 2. The Streptococcus thermophilus strain according to claim 1, wherein the strain carries a mutation that reduces the transport of glucose into the cell. 3. The Streptococcus thermophilus strain according to claim 2, wherein the strain carries a mutation in a gene encoding a component of a glucose transporter, wherein the mutation inactivates the glucose transporter or has a negative effect on expression of the gene. 4. The Streptococcus thermophilus strain according to claim 3, wherein the strain carries a mutation in the DNA sequence of the manM gene encoding the IICMan protein of the glucose/mannose phosphotransferase system, wherein the mutation inactivates the IICMan protein or has a negative effect on expression of the gene. 5. The Streptococcus thermophilus strain according to any of the preceding claims, wherein the strain increases the amount of glucose in 9.5% B-milk to at least 5 mg/ml when inoculated into the 9.5% B-milk at a concentration of 106-107 CFU/ml and grown at 40° C. for 20 hours. 6. The Streptococcus thermophilus strain according to any of the preceding claims, wherein the strain increases the amount of glucose in 9.5% B-milk with 0.05% sucrose to at least 5 mg/ml when inoculated into the 9.5% B-milk with 0.05% sucrose at a concentration of 106-107 CFU/ml and grown at 40° C. for 20 hours. 7. A Streptococcus thermophilus strain, wherein the strain is selected from the group consisting of the Streptococcus thermophilus CHCC15757 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 25850, the Streptococcus thermophilus CHCC15887 strain that was deposited at Deutsche Sammlung von Mikroorganismen and Zellkulturen under accession No. DSM 25851, the Streptococcus thermophilus CHCC16404 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26722, and a mutant strain derived therefrom, wherein the mutant strain is obtained by using one of the deposited strains as starting material, and wherein the mutant has retained or further improved the lactose fermenting property and/or the glucose secreting property of said deposited strain. 8. A Lactobacillus delbrueckii subsp, bulgaricus strain, wherein the strain is resistant to 2-deoxyglucose and wherein the strain increases the amount of glucose in 9.5% B-milk to at least 5 mg/ml when inoculated into the 9.5% B-milk at a concentration of 106-107 CFU/ml and grown at 40° C. for at least 20 hours. 9. A Lactobacillus delbrueckii subsp, bulgaricus strain, wherein the strain is selected from the group consisting of the Lactobacillus delbrueckii subsp. bulgaricus strain CHCC16159 that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26420, the Lactobacillus delbrueckii subsp, bulgaricus strain CHCC16160 that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26421, and a mutant strain derived therefrom, wherein the mutant strain is obtained by using one of the deposited strains as starting material, and wherein the mutant has retained or further improved the lactose fermenting property and/or the glucose secreting property of said deposited strain. 10. A composition comprising from 104 to 1012 CFU/g of a Streptococcus thermophilus strain according to any of claims 1 to 7. 11. The composition of claim 10, wherein the Streptococcus thermophilus strain is unable to acidify the 9.5% B-milk, defined as resulting in a pH decrease of less than 1.0 when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C., and wherein the composition further comprises an amount of sucrose effective to trigger acidification of the 9.5% B-milk, defined as resulting in a pH decrease of 1.0 or more when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C. 12. The composition of claim 10 or 11 further comprising from 104 to 1012 CFU/g of a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 13. A method for producing a fermented milk product comprising inoculating and fermenting a milk substrate with at least one Streptococcus thermophilus strain according to any one of claims 1 to 7. 14. A method according to claim 13, wherein the Streptococcus thermophilus strain is unable to acidify 9.5% B-milk, defined as resulting in a pH decrease of less than 1.0 when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C., and wherein the milk substrate is added an amount of sucrose effective of triggering acidification of the B-milk, defined as resulting in a pH decrease of 1.0 or more when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C. 15. A method for producing a fermented milk product comprising inoculating and fermenting a milk substrate with at least one Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 16. A method according to any one of claims 13 to 15 comprising inoculating and fermenting a milk substrate with at least one Streptococcus thermophilus strain according to any one of claims 1 to 7 and at least one Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 17. A fermented milk product comprising at least one Streptococcus thermophilus strain according to any one of claims 1 to 7. 18. A fermented milk product comprising at least one Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 19. A fermented milk product according to claim 17 or 18 comprising at least one Streptococcus thermophilus strain according to any one of claims 1 to 7 and at least one Lactobacillus delbrueckii subsp bulgaricus strain according to claim 8 or 9. 20. Use of a Streptococcus thermophilus strain according to any one of claims 1 to 7 and a Lactobacillus delbrueckii subsp, bulgaricus strain according to claim 8 or 9 for the preparation of a fermented milk product. 21. Use of a Streptococcus thermophilus strain according to any one of claims 1 to 7 and a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9 for increasing the sweetness of a fermented milk product. 22. Use of a Streptococcus thermophilus strain according to any one of claims 1 to 7 and a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9 for decreasing the lactose content in a fermented milk product. 23. A fermented milk product according to any one of claims 17 to 19 for use in avoiding symptoms of lactose intolerance. 24. Use of a Streptococcus thermophilus strain according o any one of claims 1 to 7 for improving the growth of a Bifidobacterium strain. 25. Use of a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9 for improving the growth of a Bifidobacterium strain. 26. A method for screening and isolating a non-GMO Streptococcus thermophilus strain-according to any one of claims 1 to 7, wherein the method comprises the following steps: a) providing a galactose-fermenting Streptococcus thermophilus mother strain; b) selecting and isolating from a pool of Streptococcus thermophilus strains derived from the mother strain a pool of Streptococcus thermophilus strains which are resistant to 2-deoxyglucose; and c) selecting and isolating from the pool of Streptococcus thermophilus strains which are resistant to 2-deoxyglucose a Streptococcus thermophilus strain if the growth rate of the Streptococcus thermophilus strain is higher in M17 medium+2% galactose than in M17 medium+2% glucose. 27. A method according to claim 26 further comprising a step d) selecting and isolating from a pool of 2-deoxyglucose resistant Streptococcus thermophilus strains derived from the Streptococcus thermophilus strain selected in step c) a Streptococcus thermophilus strain if the growth rate of the Streptococcus thermophilus strain is high in M17 medium+2% sucrose but zero or at least 0-50% reduced compared to the growth rate of the mother strain in M17 medium+2% glucose. 28. The method according to claim 26 or 27, wherein the galactose-fermenting mother strain is selected from the group consisting of the Streptococcus thermophilus CHCC14994 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession no. DSM 25838, the Streptococcus thermophilus CHCC11976 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession no DSM 22934, and strains derived therefrom. 29. A method for screen ng and isolating a strain of Lactobacillus delbrueckii subsp. bulgaricus strain wherein the method comprises the following steps: a) providing a Lactobacillus delbrueckii subsp. bulgaricus mother strain; b) selecting and isolating from a pool of mutant Lactobacillus delbrueckii subsp. bulgaricus strains derived from the mother strain a pool of Lactobacillus delbrueckii subsp. bulgaricus strains which are resistant to 2-deoxyglucose; and c) selecting and isolating from the pool of Lactobacillus delbrueckii subsp. bulgaricus strains which are resistant to 2-deoxyglucose a Lactobacillus delbrueckii subsp. bulgaricus strain if the growth rate of the Lactobacillus delbrueckii subsp. bulgaricus strain is higher in MRS-IM medium+2% lactose than in MRS-IM medium+2% glucose. 30. A method according to claim 29, wherein the mother strain is selected from the group consisting of the Lactobacillus delbrueckii subsp. bulgaricus CHCC759 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26419, the Lactobacillus delbrueckii subsp. bulgaricus CHCC10019 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 19252, and strains derived therefrom.
The dairy industry today faces a problem of providing an alternative to adding sweeteners to fermented milk products in order to achieve the desired sweet taste without the added calories. Furthermore, it would be highly advantageous to establish a method for reducing lactose in fermented milk products to a level which is acceptable for lactose-intolerant consumers. The above problems have been solved by providing mutant Streptococcus thermophilus strains and mutant Lactobacillus delbrueckii subsp. bulgaricus strains that excrete glucose to the milk when the milk is inoculated and fermented with such Streptococcus thermophilus strains and Lactobacillus delbrueckii subsp, bulgaricus strains. Thus, the present invention relates to strains of Streptococcus thermophilus and Lactobacillus delbrueckii subsp, bulgaricus which secrete glucose to the milk substrate during fermentation, as well as to mixed cultures comprising the Streptococcus thermophilus strains and the Lactobacillus delbrueckii subsp, bulgaricus strains, starter cultures comprising the strains and dairy products manufactured with the cultures. The present method also relates to use of the strains for decreasing the lactose content of a fermented food product and for boosting growth of the probiotic BB-12®.1. A galactose-fermenting Streptococcus thermophilus strain, wherein the strain carries a mutation in the DNA sequence of the glcK gene encoding a glucokinase protein, wherein the mutation inactivates the glucokinase protein or has a negative effect on expression of the gene. 2. The Streptococcus thermophilus strain according to claim 1, wherein the strain carries a mutation that reduces the transport of glucose into the cell. 3. The Streptococcus thermophilus strain according to claim 2, wherein the strain carries a mutation in a gene encoding a component of a glucose transporter, wherein the mutation inactivates the glucose transporter or has a negative effect on expression of the gene. 4. The Streptococcus thermophilus strain according to claim 3, wherein the strain carries a mutation in the DNA sequence of the manM gene encoding the IICMan protein of the glucose/mannose phosphotransferase system, wherein the mutation inactivates the IICMan protein or has a negative effect on expression of the gene. 5. The Streptococcus thermophilus strain according to any of the preceding claims, wherein the strain increases the amount of glucose in 9.5% B-milk to at least 5 mg/ml when inoculated into the 9.5% B-milk at a concentration of 106-107 CFU/ml and grown at 40° C. for 20 hours. 6. The Streptococcus thermophilus strain according to any of the preceding claims, wherein the strain increases the amount of glucose in 9.5% B-milk with 0.05% sucrose to at least 5 mg/ml when inoculated into the 9.5% B-milk with 0.05% sucrose at a concentration of 106-107 CFU/ml and grown at 40° C. for 20 hours. 7. A Streptococcus thermophilus strain, wherein the strain is selected from the group consisting of the Streptococcus thermophilus CHCC15757 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 25850, the Streptococcus thermophilus CHCC15887 strain that was deposited at Deutsche Sammlung von Mikroorganismen and Zellkulturen under accession No. DSM 25851, the Streptococcus thermophilus CHCC16404 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26722, and a mutant strain derived therefrom, wherein the mutant strain is obtained by using one of the deposited strains as starting material, and wherein the mutant has retained or further improved the lactose fermenting property and/or the glucose secreting property of said deposited strain. 8. A Lactobacillus delbrueckii subsp, bulgaricus strain, wherein the strain is resistant to 2-deoxyglucose and wherein the strain increases the amount of glucose in 9.5% B-milk to at least 5 mg/ml when inoculated into the 9.5% B-milk at a concentration of 106-107 CFU/ml and grown at 40° C. for at least 20 hours. 9. A Lactobacillus delbrueckii subsp, bulgaricus strain, wherein the strain is selected from the group consisting of the Lactobacillus delbrueckii subsp. bulgaricus strain CHCC16159 that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26420, the Lactobacillus delbrueckii subsp, bulgaricus strain CHCC16160 that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26421, and a mutant strain derived therefrom, wherein the mutant strain is obtained by using one of the deposited strains as starting material, and wherein the mutant has retained or further improved the lactose fermenting property and/or the glucose secreting property of said deposited strain. 10. A composition comprising from 104 to 1012 CFU/g of a Streptococcus thermophilus strain according to any of claims 1 to 7. 11. The composition of claim 10, wherein the Streptococcus thermophilus strain is unable to acidify the 9.5% B-milk, defined as resulting in a pH decrease of less than 1.0 when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C., and wherein the composition further comprises an amount of sucrose effective to trigger acidification of the 9.5% B-milk, defined as resulting in a pH decrease of 1.0 or more when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C. 12. The composition of claim 10 or 11 further comprising from 104 to 1012 CFU/g of a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 13. A method for producing a fermented milk product comprising inoculating and fermenting a milk substrate with at least one Streptococcus thermophilus strain according to any one of claims 1 to 7. 14. A method according to claim 13, wherein the Streptococcus thermophilus strain is unable to acidify 9.5% B-milk, defined as resulting in a pH decrease of less than 1.0 when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C., and wherein the milk substrate is added an amount of sucrose effective of triggering acidification of the B-milk, defined as resulting in a pH decrease of 1.0 or more when 9.5% B-milk is inoculated with 106-107 CFU/ml of the Streptococcus thermophilus strain and incubated for 14 hours at 40° C. 15. A method for producing a fermented milk product comprising inoculating and fermenting a milk substrate with at least one Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 16. A method according to any one of claims 13 to 15 comprising inoculating and fermenting a milk substrate with at least one Streptococcus thermophilus strain according to any one of claims 1 to 7 and at least one Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 17. A fermented milk product comprising at least one Streptococcus thermophilus strain according to any one of claims 1 to 7. 18. A fermented milk product comprising at least one Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9. 19. A fermented milk product according to claim 17 or 18 comprising at least one Streptococcus thermophilus strain according to any one of claims 1 to 7 and at least one Lactobacillus delbrueckii subsp bulgaricus strain according to claim 8 or 9. 20. Use of a Streptococcus thermophilus strain according to any one of claims 1 to 7 and a Lactobacillus delbrueckii subsp, bulgaricus strain according to claim 8 or 9 for the preparation of a fermented milk product. 21. Use of a Streptococcus thermophilus strain according to any one of claims 1 to 7 and a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9 for increasing the sweetness of a fermented milk product. 22. Use of a Streptococcus thermophilus strain according to any one of claims 1 to 7 and a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9 for decreasing the lactose content in a fermented milk product. 23. A fermented milk product according to any one of claims 17 to 19 for use in avoiding symptoms of lactose intolerance. 24. Use of a Streptococcus thermophilus strain according o any one of claims 1 to 7 for improving the growth of a Bifidobacterium strain. 25. Use of a Lactobacillus delbrueckii subsp. bulgaricus strain according to claim 8 or 9 for improving the growth of a Bifidobacterium strain. 26. A method for screening and isolating a non-GMO Streptococcus thermophilus strain-according to any one of claims 1 to 7, wherein the method comprises the following steps: a) providing a galactose-fermenting Streptococcus thermophilus mother strain; b) selecting and isolating from a pool of Streptococcus thermophilus strains derived from the mother strain a pool of Streptococcus thermophilus strains which are resistant to 2-deoxyglucose; and c) selecting and isolating from the pool of Streptococcus thermophilus strains which are resistant to 2-deoxyglucose a Streptococcus thermophilus strain if the growth rate of the Streptococcus thermophilus strain is higher in M17 medium+2% galactose than in M17 medium+2% glucose. 27. A method according to claim 26 further comprising a step d) selecting and isolating from a pool of 2-deoxyglucose resistant Streptococcus thermophilus strains derived from the Streptococcus thermophilus strain selected in step c) a Streptococcus thermophilus strain if the growth rate of the Streptococcus thermophilus strain is high in M17 medium+2% sucrose but zero or at least 0-50% reduced compared to the growth rate of the mother strain in M17 medium+2% glucose. 28. The method according to claim 26 or 27, wherein the galactose-fermenting mother strain is selected from the group consisting of the Streptococcus thermophilus CHCC14994 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession no. DSM 25838, the Streptococcus thermophilus CHCC11976 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession no DSM 22934, and strains derived therefrom. 29. A method for screen ng and isolating a strain of Lactobacillus delbrueckii subsp. bulgaricus strain wherein the method comprises the following steps: a) providing a Lactobacillus delbrueckii subsp. bulgaricus mother strain; b) selecting and isolating from a pool of mutant Lactobacillus delbrueckii subsp. bulgaricus strains derived from the mother strain a pool of Lactobacillus delbrueckii subsp. bulgaricus strains which are resistant to 2-deoxyglucose; and c) selecting and isolating from the pool of Lactobacillus delbrueckii subsp. bulgaricus strains which are resistant to 2-deoxyglucose a Lactobacillus delbrueckii subsp. bulgaricus strain if the growth rate of the Lactobacillus delbrueckii subsp. bulgaricus strain is higher in MRS-IM medium+2% lactose than in MRS-IM medium+2% glucose. 30. A method according to claim 29, wherein the mother strain is selected from the group consisting of the Lactobacillus delbrueckii subsp. bulgaricus CHCC759 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 26419, the Lactobacillus delbrueckii subsp. bulgaricus CHCC10019 strain that was deposited at Deutsche Sammlung von Mikroorganismen und Zellkulturen under accession No. DSM 19252, and strains derived therefrom.
1,600
546
12,963,272
1,637
Reagents and methods for analysis of DNA hydroxymethylation are provided. Methods comprise modification of hydroxymethylated cytosine residues with a bulky moiety to protect hydroxymethylated positions from cleavage with a DNA endonuclease. For example, methods may comprise contacting DNA with a glucosyltransferase to glucosylate hydroxymethylated DNA positions and digesting the DNA with a DNA endonuclease to cleave DNA in positions lacking hydroxymethylation. Reagents and kits for hydroxymethylated DNA analysis are also provided.
1. A method for detecting DNA hydroxymethylation in a DNA sample comprising: (i) contacting a DNA sample comprising a glycosylated hydroxymethylcytosine with a DNA endonuclease to cleave the DNA; and (ii) detecting at least a first DNA sequence from a sample not cleaved by the DNA endonuclease to determine the presence of hydroxymethylation in the DNA sequence. 2. The method of claim 1, wherein contacting the DNA sample with a DNA endonuclease comprises contacting the DNA with two or more DNA endonucleases. 3. The method of claim 1 wherein the DNA endonuclease is a methylation dependent DNA endonuclease. 4. The method of claim 3, wherein contacting the DNA sample with a methylation dependent DNA endonuclease comprises contacting the DNA with two or more methylation dependent DNA endonucleases. 5. The method of claim 1, wherein detecting DNA sequences not cleaved by the DNA endonuclease comprises DNA sequencing or hybridization. 6. The method of claim 1, further comprising ligating the cleaved DNA to an oligonucleotide tag before said detecting of step (ii), wherein the oligonucleotide tag comprises a sequence for PCR primer binding. 7. The method of claim 6, wherein the oligonucleotide tag comprises a label. 8. The method of claim 7, wherein the label is a fluorescent, a colorimetric, an enzymatic, an antigen or a radioactive label. 9. The method of claim 6, wherein detecting DNA sequences not cleaved by the DNA endonuclease comprises sequencing the DNA using a primer that hybridizes to the oligonucleotide tag. 10. The method of claim 1, comprising determining the presence of DNA hydroxymethylation at a plurality of potential hydroxymethylation sites. 11. The method of claim 10, comprising determining the presence of DNA hydroxymethylation at least 5, 10, 15, 20, 50, 100, 500 or 1,000 potential hydroxymethylation sites. 12. The method of claim 10, wherein determining the presence of DNA hydroxymethylation at a potential methylation site comprises identifying a sequence corresponding detected DNA sequence on a genomic map. 13. The method of claim 1, further comprising detecting DNA hydroxymethylation in two or more DNA samples to determine differential DNA hydroxymethylation between two or more samples. 14. The method of claim 13, wherein said two or more samples comprise: samples from tissue culture cells grown under different conditions; samples from cells at different stages of differentiation; samples from healthy and diseases tissue; samples two or more different organisms or individuals; or samples from cells treated with a drug and placebo. 15. The method of claim 1, wherein the DNA sample comprises mammalian genomic DNA. 16. The method of claim 15, wherein the mammalian genomic DNA is human genomic DNA. 17. The method of claim 16, wherein the human genomic DNA is from a human subject. 18. The method of claim 15, wherein the human genomic DNA is from a cell line or tissue bank. 19. The method of claim 1, wherein the DNA sample is from cultured cells, a tissue biopsy blood, urine, saliva or skin. 20. The method of claim 19, wherein the cultured cells are neuronal cells or stem cells. 21. The method of claim 1, wherein the DNA endonuclease is MspI, BisI, GlaI, TaqαI or McrBC. 22. The method of claim 3, wherein the methylation dependent DNA endonuclease is BisI, GlaI or McrBC. 23. The method of claim 1, further comprising contacting the DNA sample with a methylation sensitive DNA endonuclease (MSE) before step (ii). 24. The method of claim 23, further comprising contacting the DNA sample with a methylation sensitive DNA endonuclease (MSE) before step (i) and detecting DNA sequences not cleaved by the methylation sensitive DNA endonuclease to determine the presence of DNA methylation. 25. The method of claim 1, further comprising treating a DNA sample to glycosylate hydroxymethylcytosine positions before step (i). 26. The method of claim 25, wherein treating the DNA sample to glycosylate hydroxymethylcytosine positions comprises contacting the DNA sample with a glucosyltransferase. 27. The method of claim 26, wherein the glucosyltransferase is recombinant. 28. The method of claim 26, wherein the glucosyltransferase is from a T-even bacteriophage. 29. The method of claim 26, wherein the glucosyltransferase is a β-glucosyltransferase. 30. The method of claim 1, further comprising contacting the DNA sample with a DNA methyltransferase prior to step (i). 31. The method of claim 30, wherein the DNA methyltransferase is M.SssI or M.CviPI. 32. A method for detecting DNA methylation and hydroxymethylation in a DNA sample comprising: (i) contacting a DNA sample with a methylation sensitive DNA endonuclease (MSE) to cleave the DNA; (ii) contacting the cleaved DNA sample comprising a glycosylated hydroxymethylcytosine with a methylation dependent DNA endonuclease to cleave the DNA; and (iii) detecting DNA sequences not cleaved by the methylation dependent DNA endonuclease to determine the presence of hydroxymethylation. 33-47. (canceled) 48. A substantially purified mammalian DNA sample comprising at least one glucosylated hydroxymethylcytosine.
Reagents and methods for analysis of DNA hydroxymethylation are provided. Methods comprise modification of hydroxymethylated cytosine residues with a bulky moiety to protect hydroxymethylated positions from cleavage with a DNA endonuclease. For example, methods may comprise contacting DNA with a glucosyltransferase to glucosylate hydroxymethylated DNA positions and digesting the DNA with a DNA endonuclease to cleave DNA in positions lacking hydroxymethylation. Reagents and kits for hydroxymethylated DNA analysis are also provided.1. A method for detecting DNA hydroxymethylation in a DNA sample comprising: (i) contacting a DNA sample comprising a glycosylated hydroxymethylcytosine with a DNA endonuclease to cleave the DNA; and (ii) detecting at least a first DNA sequence from a sample not cleaved by the DNA endonuclease to determine the presence of hydroxymethylation in the DNA sequence. 2. The method of claim 1, wherein contacting the DNA sample with a DNA endonuclease comprises contacting the DNA with two or more DNA endonucleases. 3. The method of claim 1 wherein the DNA endonuclease is a methylation dependent DNA endonuclease. 4. The method of claim 3, wherein contacting the DNA sample with a methylation dependent DNA endonuclease comprises contacting the DNA with two or more methylation dependent DNA endonucleases. 5. The method of claim 1, wherein detecting DNA sequences not cleaved by the DNA endonuclease comprises DNA sequencing or hybridization. 6. The method of claim 1, further comprising ligating the cleaved DNA to an oligonucleotide tag before said detecting of step (ii), wherein the oligonucleotide tag comprises a sequence for PCR primer binding. 7. The method of claim 6, wherein the oligonucleotide tag comprises a label. 8. The method of claim 7, wherein the label is a fluorescent, a colorimetric, an enzymatic, an antigen or a radioactive label. 9. The method of claim 6, wherein detecting DNA sequences not cleaved by the DNA endonuclease comprises sequencing the DNA using a primer that hybridizes to the oligonucleotide tag. 10. The method of claim 1, comprising determining the presence of DNA hydroxymethylation at a plurality of potential hydroxymethylation sites. 11. The method of claim 10, comprising determining the presence of DNA hydroxymethylation at least 5, 10, 15, 20, 50, 100, 500 or 1,000 potential hydroxymethylation sites. 12. The method of claim 10, wherein determining the presence of DNA hydroxymethylation at a potential methylation site comprises identifying a sequence corresponding detected DNA sequence on a genomic map. 13. The method of claim 1, further comprising detecting DNA hydroxymethylation in two or more DNA samples to determine differential DNA hydroxymethylation between two or more samples. 14. The method of claim 13, wherein said two or more samples comprise: samples from tissue culture cells grown under different conditions; samples from cells at different stages of differentiation; samples from healthy and diseases tissue; samples two or more different organisms or individuals; or samples from cells treated with a drug and placebo. 15. The method of claim 1, wherein the DNA sample comprises mammalian genomic DNA. 16. The method of claim 15, wherein the mammalian genomic DNA is human genomic DNA. 17. The method of claim 16, wherein the human genomic DNA is from a human subject. 18. The method of claim 15, wherein the human genomic DNA is from a cell line or tissue bank. 19. The method of claim 1, wherein the DNA sample is from cultured cells, a tissue biopsy blood, urine, saliva or skin. 20. The method of claim 19, wherein the cultured cells are neuronal cells or stem cells. 21. The method of claim 1, wherein the DNA endonuclease is MspI, BisI, GlaI, TaqαI or McrBC. 22. The method of claim 3, wherein the methylation dependent DNA endonuclease is BisI, GlaI or McrBC. 23. The method of claim 1, further comprising contacting the DNA sample with a methylation sensitive DNA endonuclease (MSE) before step (ii). 24. The method of claim 23, further comprising contacting the DNA sample with a methylation sensitive DNA endonuclease (MSE) before step (i) and detecting DNA sequences not cleaved by the methylation sensitive DNA endonuclease to determine the presence of DNA methylation. 25. The method of claim 1, further comprising treating a DNA sample to glycosylate hydroxymethylcytosine positions before step (i). 26. The method of claim 25, wherein treating the DNA sample to glycosylate hydroxymethylcytosine positions comprises contacting the DNA sample with a glucosyltransferase. 27. The method of claim 26, wherein the glucosyltransferase is recombinant. 28. The method of claim 26, wherein the glucosyltransferase is from a T-even bacteriophage. 29. The method of claim 26, wherein the glucosyltransferase is a β-glucosyltransferase. 30. The method of claim 1, further comprising contacting the DNA sample with a DNA methyltransferase prior to step (i). 31. The method of claim 30, wherein the DNA methyltransferase is M.SssI or M.CviPI. 32. A method for detecting DNA methylation and hydroxymethylation in a DNA sample comprising: (i) contacting a DNA sample with a methylation sensitive DNA endonuclease (MSE) to cleave the DNA; (ii) contacting the cleaved DNA sample comprising a glycosylated hydroxymethylcytosine with a methylation dependent DNA endonuclease to cleave the DNA; and (iii) detecting DNA sequences not cleaved by the methylation dependent DNA endonuclease to determine the presence of hydroxymethylation. 33-47. (canceled) 48. A substantially purified mammalian DNA sample comprising at least one glucosylated hydroxymethylcytosine.
1,600
547
14,081,917
1,642
An anti-c-Met antibody or antibody fragment and pharmaceutical composition comprising same, as well as a method for preventing and treating cancer by administering the antibody to a subject are provided.
1. A method of prevention or treatment of a cancer comprising administering an anti-c-Met or an antigen-binding fragment thereof to a subject in need of prevention or treatment of the cancer. 2. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 4, CDR-H2 having an amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 5, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 6; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 4 to 9 are respectively represented by following Formula I to VI: Xaa1-Xaa2-Tyr-Tyr-Met-Ser (SEQ ID NO: 4), wherein Xaa1 is Pro or Ser or is absent, and Xaa2 is Glu or Asp;  Formula I Arg-Asn-Xaa3-Xaa4-Asn-Gly-Xaa5-Thr (SEQ ID NO: 5), wherein Xaa3 is Asn or Lys, Xaa4 is Ala or Val, and Xaa5 is Asn or Thr;  Formula II Asp-Asn-Trp-Leu-Xaa6-Tyr (SEQ ID NO: 6); wherein Xaa6 is Ser or Thr;  Formula III Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13 (SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 3. The method of claim 2, wherein, the CDR-H1 a polypeptide having an amino acid sequence of SEQ ID NO: 22, 23, or 24; the CDR-H2 is a polypeptide having an amino acid sequence of SEQ ID NO: 25 or 26.; the CDR-H3 is a polypeptide having an amino acid sequence of SEQ ID NO: 27 or 28; the CDR-L1 is a polypeptide having an amino acid sequence of SEQ ID NO: 29, 30, 31, 32, 33, or 71; the CDR-L2 is a polypeptide having an amino acid sequence of SEQ ID NO: 34, 35, or 36; or the CDR-L3 is a polypeptide having an amino acid sequence of SEQ ID NO: 13, 14, 15, 16, or 37. 4. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1, CDR-H2 having an amino acid sequence of SEQ ID NO: 2, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 7 to 9 are respectively represented by Formula IV to VI below: Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13 (SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 5. The method of claim 4, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the light chain variable region comprises at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 10 or 71, CDR-L2 having an amino acid sequence of SEQ ID NO: 11, and CDR-L3 having an amino acid sequence of SEQ ID NO: 13, 14, 15, or 16. 6. The method of claim 4, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the heavy chain variable region has an amino acid sequence of SEQ ID NO: 17, and the light chain variable region has an amino acid sequence of SEQ ID NO: 18, 19, 20, 21, or 72. 7. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises: a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 68; a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 70; or a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence of SEQ ID NO: 73. 8. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 71, 72, or 73. 9. The method of claim 1, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, or a humanized antibody, 10. The method of claim 1, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 11. A method of prevention or inhibition of metastasis of a cancer comprising administering an anti-c-Met or an antigen-binding fragment thereof to a subject in need of prevention or treatment of the cancer. 12. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 4, CDR-H2 having an amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 5, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 6; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 4 to 9 are respectively represented by following Formula I to VI: Xaa1-Xaa2-Tyr-Tyr-Met-Ser (SEQ ID NO: 4), wherein Xaa1 is Pro or Ser or is absent, and Xaa2 is Glu or Asp;  Formula I Arg-Asn-Xaa3-Xaa4-Asn-Gly-Xaa5-Thr (SEQ ID NO: 5), wherein Xaa3 is Asn or Lys, Xaa4 is Ala or Val, and Xaa5 is Asn or Thr;  Formula II Asp-Asn-Trp-Leu-Xaa6-Tyr (SEQ ID NO: 6); wherein Xaa6 is Ser or Thr;  Formula III Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13(SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 13. The method of claim 12, wherein, the CDR-H1 a polypeptide having an amino acid sequence of SEQ ID NO: 22, 23, or 24; the CDR-H2 is a polypeptide having an amino acid sequence of SEQ ID NO: 25 or 26.; the CDR-H3 is a polypeptide having an amino acid sequence of SEQ ID NO: 27 or 28; the CDR-L1 is a polypeptide having an amino acid sequence of SEQ ID NO: 29, 30, 31, 32, 33, or 71; the CDR-L2 is a polypeptide having an amino acid sequence of SEQ ID NO: 34, 35, or 36; or the CDR-L3 is a polypeptide having an amino acid sequence of SEQ ID NO: 13, 14, 15, 16, or 37. 14. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1, CDR-H2 having an amino acid sequence of SEQ ID NO: 2, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 7 to 9 are respectively represented by Formula IV to VI below: Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13(SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 15. The method of claim 14, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the light chain variable region comprises at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 10 or 71, CDR-L2 having an amino acid sequence of SEQ ID NO: 11, and CDR-L3 having an amino acid sequence of SEQ ID NO: 13, 14, 15, or 16. 16. The method of claim 14, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the heavy chain variable region has an amino acid sequence of SEQ ID NO: 17, and the light chain variable region has an amino acid sequence of SEQ ID NO: 18, 19, 20, 21, or 72. 17. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises: a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 68; a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 70; or a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence of SEQ ID NO: 73. 18. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 71, 72, or 73. 19. The method of claim 11, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, or a humanized antibody, 20. The method of claim 11, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2.
An anti-c-Met antibody or antibody fragment and pharmaceutical composition comprising same, as well as a method for preventing and treating cancer by administering the antibody to a subject are provided.1. A method of prevention or treatment of a cancer comprising administering an anti-c-Met or an antigen-binding fragment thereof to a subject in need of prevention or treatment of the cancer. 2. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 4, CDR-H2 having an amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 5, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 6; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 4 to 9 are respectively represented by following Formula I to VI: Xaa1-Xaa2-Tyr-Tyr-Met-Ser (SEQ ID NO: 4), wherein Xaa1 is Pro or Ser or is absent, and Xaa2 is Glu or Asp;  Formula I Arg-Asn-Xaa3-Xaa4-Asn-Gly-Xaa5-Thr (SEQ ID NO: 5), wherein Xaa3 is Asn or Lys, Xaa4 is Ala or Val, and Xaa5 is Asn or Thr;  Formula II Asp-Asn-Trp-Leu-Xaa6-Tyr (SEQ ID NO: 6); wherein Xaa6 is Ser or Thr;  Formula III Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13 (SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 3. The method of claim 2, wherein, the CDR-H1 a polypeptide having an amino acid sequence of SEQ ID NO: 22, 23, or 24; the CDR-H2 is a polypeptide having an amino acid sequence of SEQ ID NO: 25 or 26.; the CDR-H3 is a polypeptide having an amino acid sequence of SEQ ID NO: 27 or 28; the CDR-L1 is a polypeptide having an amino acid sequence of SEQ ID NO: 29, 30, 31, 32, 33, or 71; the CDR-L2 is a polypeptide having an amino acid sequence of SEQ ID NO: 34, 35, or 36; or the CDR-L3 is a polypeptide having an amino acid sequence of SEQ ID NO: 13, 14, 15, 16, or 37. 4. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1, CDR-H2 having an amino acid sequence of SEQ ID NO: 2, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 7 to 9 are respectively represented by Formula IV to VI below: Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13 (SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 5. The method of claim 4, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the light chain variable region comprises at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 10 or 71, CDR-L2 having an amino acid sequence of SEQ ID NO: 11, and CDR-L3 having an amino acid sequence of SEQ ID NO: 13, 14, 15, or 16. 6. The method of claim 4, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the heavy chain variable region has an amino acid sequence of SEQ ID NO: 17, and the light chain variable region has an amino acid sequence of SEQ ID NO: 18, 19, 20, 21, or 72. 7. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises: a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 68; a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 70; or a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence of SEQ ID NO: 73. 8. The method of claim 1, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 71, 72, or 73. 9. The method of claim 1, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, or a humanized antibody, 10. The method of claim 1, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 11. A method of prevention or inhibition of metastasis of a cancer comprising administering an anti-c-Met or an antigen-binding fragment thereof to a subject in need of prevention or treatment of the cancer. 12. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 4, CDR-H2 having an amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 5, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 6; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 4 to 9 are respectively represented by following Formula I to VI: Xaa1-Xaa2-Tyr-Tyr-Met-Ser (SEQ ID NO: 4), wherein Xaa1 is Pro or Ser or is absent, and Xaa2 is Glu or Asp;  Formula I Arg-Asn-Xaa3-Xaa4-Asn-Gly-Xaa5-Thr (SEQ ID NO: 5), wherein Xaa3 is Asn or Lys, Xaa4 is Ala or Val, and Xaa5 is Asn or Thr;  Formula II Asp-Asn-Trp-Leu-Xaa6-Tyr (SEQ ID NO: 6); wherein Xaa6 is Ser or Thr;  Formula III Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13(SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 13. The method of claim 12, wherein, the CDR-H1 a polypeptide having an amino acid sequence of SEQ ID NO: 22, 23, or 24; the CDR-H2 is a polypeptide having an amino acid sequence of SEQ ID NO: 25 or 26.; the CDR-H3 is a polypeptide having an amino acid sequence of SEQ ID NO: 27 or 28; the CDR-L1 is a polypeptide having an amino acid sequence of SEQ ID NO: 29, 30, 31, 32, 33, or 71; the CDR-L2 is a polypeptide having an amino acid sequence of SEQ ID NO: 34, 35, or 36; or the CDR-L3 is a polypeptide having an amino acid sequence of SEQ ID NO: 13, 14, 15, 16, or 37. 14. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1, CDR-H2 having an amino acid sequence of SEQ ID NO: 2, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 7 to 9 are respectively represented by Formula IV to VI below: Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn;  Formula IV Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13(SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and  Formula V Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met.  Formula VI 15. The method of claim 14, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the light chain variable region comprises at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 10 or 71, CDR-L2 having an amino acid sequence of SEQ ID NO: 11, and CDR-L3 having an amino acid sequence of SEQ ID NO: 13, 14, 15, or 16. 16. The method of claim 14, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises the heavy chain variable region has an amino acid sequence of SEQ ID NO: 17, and the light chain variable region has an amino acid sequence of SEQ ID NO: 18, 19, 20, 21, or 72. 17. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises: a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 68; a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 70; or a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence of SEQ ID NO: 73. 18. The method of claim 11, wherein the anti-c-Met antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 71, 72, or 73. 19. The method of claim 11, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, or a humanized antibody, 20. The method of claim 11, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2.
1,600
548
13,646,589
1,642
An anti c-Met antibody or antibody fragment and pharmaceutical composition comprising same, as well as a method for preventing and treating cancer by administering the antibody to a subject.
1. An anti c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 4, CDR-H2 having an amino acid sequence of SEQ ID NO: 5, and CDR-H3 having an amino acid sequence of SEQ ID NO: 6; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 4 to 9 are respectively represented by following Formula I to VI: Formula I Xaa1-Xaa2-Tyr-Tyr-Met-Ser (SEQ ID NO: 4), wherein Xaa1 is Pro or Ser or is absent, and Xaa2 is Glu or Asp; Formula II Arg-Asn-Xaa3-Xaa4-Asn-Gly-Xaa5-Thr (SEQ ID NO: 5), wherein Xaa3 is Asn or Lys, Xaa4 is Ala or Val, and Xaa5 is Asn or Thr; Formula III Asp-Asn-Trp-Leu-Xaa6-Tyr (SEQ ID NO: 6); wherein Xaa6 is Ser or Thr; Formula IV Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn; Formula V Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13 (SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and Formula VI Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met. 2. The antibody or antigen-binding fragment of claim 1, wherein CDR-H1 a polypeptide having an amino acid sequence of SEQ ID NO: 22, 23, or 24. 3. The antibody or antigen-binding fragment of claim 1, wherein CDR-H2 is a polypeptide having an amino acid sequence of SEQ ID NO: 25 or 26. 4. The antibody or antigen-binding fragment of claim 1, wherein CDR-H3 is a polypeptide having an amino acid sequence of SEQ ID NO: 27 or 28. 5. The antibody or antigen-binding fragment of claim 1, wherein CDR-L1 is a polypeptide having an amino acid sequence of SEQ ID NO: 29, 30, 31, 32, 33, or 71. 6. The antibody or antigen-binding fragment of claim 1, wherein CDR-L2 is a polypeptide having an amino acid sequence of SEQ ID NO: 34, 35, or 36. 7. The antibody or antigen-binding fragment of claim 1, wherein CDR-L3 is a polypeptide having an amino acid sequence of SEQ ID NO: 13, 14, 15, 16, or 37. 8. An anti c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1, CDR-H2 having an amino acid sequence of SEQ ID NO: 2, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 7 to 9 are respectively represented by Formula IV to VI below: Formula IV Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn; Formula V Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13(SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and Formula VI Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met. 9. The antibody or antigen-binding fragment of claim 8, wherein the light chain variable region comprises at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 10 or 71, CDR-L2 having an amino acid sequence of SEQ ID NO: 11, and CDR-L3 having an amino acid sequence of SEQ ID NO: 13, 14, 15, or 16. 10. The antibody or antigen-binding fragment of claim 8, wherein the heavy chain variable region has an amino acid sequence of SEQ ID NO: 17, and the light chain variable region has an amino acid sequence of SEQ ID NO: 18, 19, 20, 21, or 72. 11. The antibody or antigen-binding fragment of claim 8, comprising a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 68. 12. The antibody or antigen-binding fragment of claim 8, comprising a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 70. 13. The antibody or antigen-binding fragment of claim 8, comprising a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence of SEQ ID NO: 73. 14. An anti c-Met antibody or antigen-binding fragment thereof comprising an amino acid sequence of SEQ ID NO: 71, 72, or 73. 15. The antibody or antigen-binding fragment of claim 1, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, a humanized antibody, 16. The antibody or antigen-binding fragment of claim 1, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 17. The antibody or antigen-binding fragment of claim 1, wherein c-Met is human c-Met, monkey c-Met, mouse c-Met, or rat c-Met. 18. The antibody or antigen-binding fragment of claim 8, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, a humanized antibody, 19. The antibody or antigen-binding fragment of claim 8, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 20. The antibody or antigen-binding fragment of claim 8, wherein c-Met is human c-Met, monkey c-Met, mouse c-Met, or rat c-Met. 21. The antibody or antigen-binding fragment of claim 14, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, a humanized antibody, 22. The antibody or antigen-binding fragment of claim 14, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 23. The antibody or antigen-binding fragment of claim 14, wherein c-Met is human c-Met, monkey c-Met, mouse c-Met, or rat c-Met. 24. A pharmaceutical composition comprising the anti c-Met antibody or antigen-binding fragment of claim 1 and a pharmaceutically acceptable carrier, a diluent, or an excipient. 25. A pharmaceutical composition comprising the anti c-Met antibody or antigen-binding fragment of claim 8 and a pharmaceutically acceptable carrier, a diluent, or an excipient. 26. A pharmaceutical composition comprising the anti c-Met antibody or antigen-binding fragment of claim 14 and a pharmaceutically acceptable carrier, a diluent, or an excipient. 27. A method of treating cancer comprising administering the antibody or antigen-binding fragment of claim 1 to a subject in need thereof. 28. The method of claim 27, wherein the cancer is squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal carcinoma, skin cancer, skin or intraocular melanoma, colorectal cancer, cancer near the anus, esophagus cancer, small intestinal tumor, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocytic lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, or a head or neck cancer. 29. A method of treating cancer comprising administering the antibody or antigen-binding fragment of claim 8 to a subject in need thereof. 30. The method of claim 29, wherein the cancer is squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal carcinoma, skin cancer, skin or intraocular melanoma, colorectal cancer, cancer near the anus, esophagus cancer, small intestinal tumor, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocytic lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, or a head or neck cancer. 31. A method of treating cancer comprising administering the antibody or antigen-binding fragment of claim 14 to a subject in need thereof. 32. The method of claim 31, wherein the cancer is squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal carcinoma, skin cancer, skin or intraocular melanoma, colorectal cancer, cancer near the anus, esophagus cancer, small intestinal tumor, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocytic lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, or a head or neck cancer. 33. A polypeptide having an amino acid sequence of SEQ ID NO: 71. 34. A polynucleotide encoding the antibody or antibody fragment of claim 1, optionally in a vector. 35. A cell comprising the polynucleotide of claim 34. 36. A polynucleotide encoding the antibody or antibody fragment of claim 8, optionally in a vector. 37. A cell comprising the polynucleotide of claim 36. 38. A polynucleotide encoding the antibody or antibody fragment of claim 14, optionally in a vector. 39. A cell comprising the polynucleotide of claim 38.
An anti c-Met antibody or antibody fragment and pharmaceutical composition comprising same, as well as a method for preventing and treating cancer by administering the antibody to a subject.1. An anti c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 4, CDR-H2 having an amino acid sequence of SEQ ID NO: 5, and CDR-H3 having an amino acid sequence of SEQ ID NO: 6; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 4 to 9 are respectively represented by following Formula I to VI: Formula I Xaa1-Xaa2-Tyr-Tyr-Met-Ser (SEQ ID NO: 4), wherein Xaa1 is Pro or Ser or is absent, and Xaa2 is Glu or Asp; Formula II Arg-Asn-Xaa3-Xaa4-Asn-Gly-Xaa5-Thr (SEQ ID NO: 5), wherein Xaa3 is Asn or Lys, Xaa4 is Ala or Val, and Xaa5 is Asn or Thr; Formula III Asp-Asn-Trp-Leu-Xaa6-Tyr (SEQ ID NO: 6); wherein Xaa6 is Ser or Thr; Formula IV Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn; Formula V Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13 (SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and Formula VI Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met. 2. The antibody or antigen-binding fragment of claim 1, wherein CDR-H1 a polypeptide having an amino acid sequence of SEQ ID NO: 22, 23, or 24. 3. The antibody or antigen-binding fragment of claim 1, wherein CDR-H2 is a polypeptide having an amino acid sequence of SEQ ID NO: 25 or 26. 4. The antibody or antigen-binding fragment of claim 1, wherein CDR-H3 is a polypeptide having an amino acid sequence of SEQ ID NO: 27 or 28. 5. The antibody or antigen-binding fragment of claim 1, wherein CDR-L1 is a polypeptide having an amino acid sequence of SEQ ID NO: 29, 30, 31, 32, 33, or 71. 6. The antibody or antigen-binding fragment of claim 1, wherein CDR-L2 is a polypeptide having an amino acid sequence of SEQ ID NO: 34, 35, or 36. 7. The antibody or antigen-binding fragment of claim 1, wherein CDR-L3 is a polypeptide having an amino acid sequence of SEQ ID NO: 13, 14, 15, 16, or 37. 8. An anti c-Met antibody or antigen-binding fragment thereof comprising: a heavy chain variable region comprising at least one heavy chain complementarity determining region (CDR) selected from the group consisting of CDR-H1 having an amino acid sequence of SEQ ID NO: 1, CDR-H2 having an amino acid sequence of SEQ ID NO: 2, and CDR-H3 having an amino acid sequence of SEQ ID NO: 3; and a light chain variable region comprising at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 7, CDR-L2 having an amino acid sequence of SEQ ID NO: 8, and CDR-L3 having an amino acid sequence of SEQ ID NO: 9, wherein SEQ ID NOS: 7 to 9 are respectively represented by Formula IV to VI below: Formula IV Lys-Ser-Ser-Xaa7-Ser-Leu-Leu-Ala-Xaa8-Gly-Asn-Xaa9-Xaa10-Asn-Tyr-Leu-Ala (SEQ ID NO: 7), wherein Xaa7 is His, Arg, Gln, or Lys, Xaa8 is Ser or Trp, Xaa9 is His or Gln, and Xaa10 is Lys or Asn; Formula V Trp-Xaa11-Ser-Xaa12-Arg-Val-Xaa13(SEQ ID NO: 8), wherein Xaa11 is Ala or Gly, Xaa12 is Thr or Lys, and Xaa13 is Ser or Pro; and Formula VI Xaa14-Gln-Ser-Tyr-Ser-Xaa15-Pro-Xaa16-Thr (SEQ ID NO: 9), wherein Xaa14 is Gly, Ala, or Gln, Xaa15 is Arg, His, Ser, Ala, Gly, or Lys, and Xaa16 is Leu, Tyr, Phe, or Met. 9. The antibody or antigen-binding fragment of claim 8, wherein the light chain variable region comprises at least one light chain CDR selected from the group consisting of CDR-L1 having an amino acid sequence of SEQ ID NO: 10 or 71, CDR-L2 having an amino acid sequence of SEQ ID NO: 11, and CDR-L3 having an amino acid sequence of SEQ ID NO: 13, 14, 15, or 16. 10. The antibody or antigen-binding fragment of claim 8, wherein the heavy chain variable region has an amino acid sequence of SEQ ID NO: 17, and the light chain variable region has an amino acid sequence of SEQ ID NO: 18, 19, 20, 21, or 72. 11. The antibody or antigen-binding fragment of claim 8, comprising a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 68. 12. The antibody or antigen-binding fragment of claim 8, comprising a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence from 21st to 220th of SEQ ID NO: 70. 13. The antibody or antigen-binding fragment of claim 8, comprising a heavy chain comprising an amino acid sequence from 18th to 462nd of SEQ ID NO: 62, an amino acid sequence from 18th to 461st of SEQ ID NO: 64, or amino acid sequence from 18th to 460th of SEQ ID NO: 66, and a light chain comprising an amino acid sequence of SEQ ID NO: 73. 14. An anti c-Met antibody or antigen-binding fragment thereof comprising an amino acid sequence of SEQ ID NO: 71, 72, or 73. 15. The antibody or antigen-binding fragment of claim 1, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, a humanized antibody, 16. The antibody or antigen-binding fragment of claim 1, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 17. The antibody or antigen-binding fragment of claim 1, wherein c-Met is human c-Met, monkey c-Met, mouse c-Met, or rat c-Met. 18. The antibody or antigen-binding fragment of claim 8, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, a humanized antibody, 19. The antibody or antigen-binding fragment of claim 8, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 20. The antibody or antigen-binding fragment of claim 8, wherein c-Met is human c-Met, monkey c-Met, mouse c-Met, or rat c-Met. 21. The antibody or antigen-binding fragment of claim 14, wherein the antibody or antigen-binding fragment is a monoclonal antibody, a mouse-derived antibody, a mouse-human chimeric antibody, a humanized antibody, 22. The antibody or antigen-binding fragment of claim 14, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2. 23. The antibody or antigen-binding fragment of claim 14, wherein c-Met is human c-Met, monkey c-Met, mouse c-Met, or rat c-Met. 24. A pharmaceutical composition comprising the anti c-Met antibody or antigen-binding fragment of claim 1 and a pharmaceutically acceptable carrier, a diluent, or an excipient. 25. A pharmaceutical composition comprising the anti c-Met antibody or antigen-binding fragment of claim 8 and a pharmaceutically acceptable carrier, a diluent, or an excipient. 26. A pharmaceutical composition comprising the anti c-Met antibody or antigen-binding fragment of claim 14 and a pharmaceutically acceptable carrier, a diluent, or an excipient. 27. A method of treating cancer comprising administering the antibody or antigen-binding fragment of claim 1 to a subject in need thereof. 28. The method of claim 27, wherein the cancer is squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal carcinoma, skin cancer, skin or intraocular melanoma, colorectal cancer, cancer near the anus, esophagus cancer, small intestinal tumor, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocytic lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, or a head or neck cancer. 29. A method of treating cancer comprising administering the antibody or antigen-binding fragment of claim 8 to a subject in need thereof. 30. The method of claim 29, wherein the cancer is squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal carcinoma, skin cancer, skin or intraocular melanoma, colorectal cancer, cancer near the anus, esophagus cancer, small intestinal tumor, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocytic lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, or a head or neck cancer. 31. A method of treating cancer comprising administering the antibody or antigen-binding fragment of claim 14 to a subject in need thereof. 32. The method of claim 31, wherein the cancer is squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal carcinoma, skin cancer, skin or intraocular melanoma, colorectal cancer, cancer near the anus, esophagus cancer, small intestinal tumor, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocytic lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, or a head or neck cancer. 33. A polypeptide having an amino acid sequence of SEQ ID NO: 71. 34. A polynucleotide encoding the antibody or antibody fragment of claim 1, optionally in a vector. 35. A cell comprising the polynucleotide of claim 34. 36. A polynucleotide encoding the antibody or antibody fragment of claim 8, optionally in a vector. 37. A cell comprising the polynucleotide of claim 36. 38. A polynucleotide encoding the antibody or antibody fragment of claim 14, optionally in a vector. 39. A cell comprising the polynucleotide of claim 38.
1,600
549
14,910,846
1,616
Spray drift reductants for agrochemical formulations and methods for reducing spray drift in agrochemical formulations are disclosed. An agrochemical formulation may comprise at least one spray drift reductant comprising a fatty acid ester of a C 3 to C 8 polyol, or an oligomer thereof having from 2 to 5 repeat units. The formulation also comprises at least one agrochemical active and/or nutrient, and optionally, a clathrate compound. A sprayable agrochemical formulation may comprise 0.01 wt. % to 1.0 wt. % of a spray drift reductant that is non-ionic, non-self-emulsifiable, has a hydrophilic-lipophilic balance of less than 7, and is capable of reducing spray drift by at least 10%, and also comprises at least on agrochemical active and/or nutrient, and optionally, a clathrate compound.
1. A sprayable agrochemical formulation comprising; i) at least one spray drift reductant comprising a fatty ester of a C3 to C8 polyol, or an oligomer thereof having from 2 to 5 repeat units; ii) at least one agrochemical active and/or nutrient; and iii) optionally a clathrate compound. 2. The formulation according to claim 1, wherein the fatty ester of a C3 to C8 polyol, or oligomer thereof, has a general structure (I): P.[R1]m  (I) wherein: P is the residue of a C3 to C8 polyol, or residue of an oligomer thereof having from 2 to 5 repeat units, each said polyol having m active hydrogen atoms, where m is an integer in the range from 2 to 7; each R1 independently represents hydrogen, a C1 to C28 hydrocarbyl, or an alkanoyl group represented by —C(O)R2 wherein R2 represents a C8 to C28 hydrocarbyl; and wherein at least one R1 group is or comprises an alkanoyl group represented by —C(O)R2. 3. The formulation according to claim 2, wherein the polyol residue is a C3 to C6 polyol residue. 4. The formulation according to claim 1, wherein said oligomer has from 2 to 4 repeat units. 5. The formulation according to claim 1, wherein the polyol oligomer is selected from diglycerol, triglycerol, tetraglycerol, dierythritol, trierythritol, tetraerythritol, di-1,3-propanediol, tri-1,3-propanediol, di(trimethylolpropane), or tri(trimethylolpropane). 6. The formulation according to claim 2, wherein the polyol has a value of m active hydrogen atoms in the range from 2 to 6. 7. The formulation according to claim 1, wherein the polyol is selected from ethylene glycol, isosorbide, 1,3-propanediol, propylene glycol, trimethyloipropane, trimethylolethane, glycerol, triglycerol, erythritol, threitol, pentaerythritol, sorbitan, arabitol, xylitol, ribitol, fucitol, mannitol, sorbitol, sucrose, maltose, galactitol, iditol, inositol, volemitol, isomalt, maltitol, or lactitol. 8. The formulation according to claim 2, wherein the C1 to C28 hydrocarbyl comprising group R′ is selected from a C1 to C28 alkyl or a C1 to C28 alkenyl. 9. The formulation according to claim 8, wherein the C1 to C28 alkyl or a C1 to C28 alkenyl is selected from dodecyl, tridecyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, or branched variants thereof, and the alkenyl radicals are selected from dodecenyl, tridecenyl, tetradecenyl, pentadecenyl, hexadecenyl, heptadecenyl, octadecenyl, nonadecenyl, eicosenyl, or branched variants thereof. 10. The formulation according to claim 2, wherein said alkanoyl group represented by —C(O)R2 is a residue of a fatty acid. 11. The formulation according to claim 10, wherein the fatty acid is selected from a C10 to C30 fatty acids. 12. The formulation according to claim 10, wherein the fatty acid is selected from caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, myristoleic acid, palmitoleic acid, sapienic acid, oleic acid, elaidic acid, vaccenic acid, linoleic acid, linoelaidic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, or docosahexaenoic acid. 13. The formulation according to claim 1, wherein the fatty ester of a C3 to C8 polyol or oligomer thereof is selected from glycerol, diglycerol, or sorbitan esters of oleic acid, elaidic acid, linoleic acid, or erucic acid. 14. The formulation according to claim 1, wherein the fatty ester of a C3 to C8 polyol or oligomer thereof has an HLB value of less than 7. 15. The formulation according to claim 1, wherein the clathrate compound is a urea clathrate or thiourea clathrate. 16. The formulation according to claim 1, wherein the molecular weight (weight average) of the spray drift reductant is in the range from 200 to 2,200. 17. The formulation according to claim 1, wherein the formulation comprises surfactants selected from non-ionic alkoxylates and alkoxylated fatty alcohol. 18. The formulation according to claim 17, wherein the non-ionic alkoxylate comprises an alkoxylated alcohol of the general formula: R3—O-(AO)x—H  (II) wherein R3 is a straight or branched chain, saturated or unsaturated, substituted or unsubstituted hydrocarbon group having from 4 to 30 carbon atoms; AO is an oxyalkylene group; and x represents an integer in the range from 1 to 30. 19. The formulation according to claim 18, wherein the oxyalkylene group AO is selected from oxyethylene (EO) and/or oxypropylene (PO). 20. The formulation according to either claim 18, wherein x is in the range from 2 to 25. 21. The formulation according to claim 18, wherein R3 is the residue of a fatty alcohol selected from capryl alcohol pelargonic alcohol, capric alcohol, undecyl alcohol, lauryl alcohol, tridecyl alcohol, myristyl alcohol, pentadecyl alcohol, cetyl alcohol, palmitoleyl alcohol, heptadecyl alcohol, stearyl alcohol, nonadecyl alcohol, arachidyl alcohol, heneicosyl alcohol, behenyl alcohol, oleyl alcohol, elaidyl alcohol, linoleyl alcohol, linolenyl alcohol, or erucyl alcohol. 22. The formulation according to claim 1, wherein the formulation is in the form of a suspension concentrate (SC), an oil-based suspension concentrate (OD), or a suspoemulsions (SE). 23. The formulation according to claim 1, wherein the spray drift reductant is present at a concentration of from 0.01 wt. % to 2 wt. %. 24. A sprayable agrochemical formulation comprising; i) in the range of from 0.01 wt. % to 1.0 wt. % spray drift reductant, wherein the reductant is non-ionic, non-self-emulsifiable, has a HLB of less than 7, and is capable of reducing spray drift by at least 10%; and ii) at least one agrochemical active and/or nutrient; and iii) optionally a clathrate compound. 25. A concentrate formulation for making a sprayable agrochemical formulation, said concentrate comprising a spray drift reductant of a fatty ester of a C3 to C8 polyol, or an oligomer thereof having from 2 to 5 repeat units. 26. A method of reducing spray drift in an agrochemical formulation comprising: applying the agrochemical formulation to vegetation, wherein the agrochemical formulation comprising a spray drift reductant comprising a fatty ester of a C3 to C8 polyol, or an oligomer thereof having from 2 to 5 repeat units, at least one agrochemical active and/or nutrient, and optionally a clathrate compound. 27. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction of spray droplets having a size of less than 150 μm of at least 10% at a spray pressure of 40 psi. 28. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction of spray droplets having a size of less than 105 μm of at least 10% at a spray pressure of 40 psi. 29. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction of spray droplets having a size of less than 500 μm of at least 5% at a spray pressure of 40 psi. 30. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction in relative span is at least 4% at a spray pressure of 40 psi 31. A method of reducing spray drift comprising applying a diluted concentrate formulation in accordance with claim 25 to vegetation. 32. A method of treating vegetation to control pests and/or to provide nutrients, the method comprising applying a formulation in accordance with claim 1 either to said vegetation or to the immediate environment of said vegetation.
Spray drift reductants for agrochemical formulations and methods for reducing spray drift in agrochemical formulations are disclosed. An agrochemical formulation may comprise at least one spray drift reductant comprising a fatty acid ester of a C 3 to C 8 polyol, or an oligomer thereof having from 2 to 5 repeat units. The formulation also comprises at least one agrochemical active and/or nutrient, and optionally, a clathrate compound. A sprayable agrochemical formulation may comprise 0.01 wt. % to 1.0 wt. % of a spray drift reductant that is non-ionic, non-self-emulsifiable, has a hydrophilic-lipophilic balance of less than 7, and is capable of reducing spray drift by at least 10%, and also comprises at least on agrochemical active and/or nutrient, and optionally, a clathrate compound.1. A sprayable agrochemical formulation comprising; i) at least one spray drift reductant comprising a fatty ester of a C3 to C8 polyol, or an oligomer thereof having from 2 to 5 repeat units; ii) at least one agrochemical active and/or nutrient; and iii) optionally a clathrate compound. 2. The formulation according to claim 1, wherein the fatty ester of a C3 to C8 polyol, or oligomer thereof, has a general structure (I): P.[R1]m  (I) wherein: P is the residue of a C3 to C8 polyol, or residue of an oligomer thereof having from 2 to 5 repeat units, each said polyol having m active hydrogen atoms, where m is an integer in the range from 2 to 7; each R1 independently represents hydrogen, a C1 to C28 hydrocarbyl, or an alkanoyl group represented by —C(O)R2 wherein R2 represents a C8 to C28 hydrocarbyl; and wherein at least one R1 group is or comprises an alkanoyl group represented by —C(O)R2. 3. The formulation according to claim 2, wherein the polyol residue is a C3 to C6 polyol residue. 4. The formulation according to claim 1, wherein said oligomer has from 2 to 4 repeat units. 5. The formulation according to claim 1, wherein the polyol oligomer is selected from diglycerol, triglycerol, tetraglycerol, dierythritol, trierythritol, tetraerythritol, di-1,3-propanediol, tri-1,3-propanediol, di(trimethylolpropane), or tri(trimethylolpropane). 6. The formulation according to claim 2, wherein the polyol has a value of m active hydrogen atoms in the range from 2 to 6. 7. The formulation according to claim 1, wherein the polyol is selected from ethylene glycol, isosorbide, 1,3-propanediol, propylene glycol, trimethyloipropane, trimethylolethane, glycerol, triglycerol, erythritol, threitol, pentaerythritol, sorbitan, arabitol, xylitol, ribitol, fucitol, mannitol, sorbitol, sucrose, maltose, galactitol, iditol, inositol, volemitol, isomalt, maltitol, or lactitol. 8. The formulation according to claim 2, wherein the C1 to C28 hydrocarbyl comprising group R′ is selected from a C1 to C28 alkyl or a C1 to C28 alkenyl. 9. The formulation according to claim 8, wherein the C1 to C28 alkyl or a C1 to C28 alkenyl is selected from dodecyl, tridecyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, or branched variants thereof, and the alkenyl radicals are selected from dodecenyl, tridecenyl, tetradecenyl, pentadecenyl, hexadecenyl, heptadecenyl, octadecenyl, nonadecenyl, eicosenyl, or branched variants thereof. 10. The formulation according to claim 2, wherein said alkanoyl group represented by —C(O)R2 is a residue of a fatty acid. 11. The formulation according to claim 10, wherein the fatty acid is selected from a C10 to C30 fatty acids. 12. The formulation according to claim 10, wherein the fatty acid is selected from caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, myristoleic acid, palmitoleic acid, sapienic acid, oleic acid, elaidic acid, vaccenic acid, linoleic acid, linoelaidic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, or docosahexaenoic acid. 13. The formulation according to claim 1, wherein the fatty ester of a C3 to C8 polyol or oligomer thereof is selected from glycerol, diglycerol, or sorbitan esters of oleic acid, elaidic acid, linoleic acid, or erucic acid. 14. The formulation according to claim 1, wherein the fatty ester of a C3 to C8 polyol or oligomer thereof has an HLB value of less than 7. 15. The formulation according to claim 1, wherein the clathrate compound is a urea clathrate or thiourea clathrate. 16. The formulation according to claim 1, wherein the molecular weight (weight average) of the spray drift reductant is in the range from 200 to 2,200. 17. The formulation according to claim 1, wherein the formulation comprises surfactants selected from non-ionic alkoxylates and alkoxylated fatty alcohol. 18. The formulation according to claim 17, wherein the non-ionic alkoxylate comprises an alkoxylated alcohol of the general formula: R3—O-(AO)x—H  (II) wherein R3 is a straight or branched chain, saturated or unsaturated, substituted or unsubstituted hydrocarbon group having from 4 to 30 carbon atoms; AO is an oxyalkylene group; and x represents an integer in the range from 1 to 30. 19. The formulation according to claim 18, wherein the oxyalkylene group AO is selected from oxyethylene (EO) and/or oxypropylene (PO). 20. The formulation according to either claim 18, wherein x is in the range from 2 to 25. 21. The formulation according to claim 18, wherein R3 is the residue of a fatty alcohol selected from capryl alcohol pelargonic alcohol, capric alcohol, undecyl alcohol, lauryl alcohol, tridecyl alcohol, myristyl alcohol, pentadecyl alcohol, cetyl alcohol, palmitoleyl alcohol, heptadecyl alcohol, stearyl alcohol, nonadecyl alcohol, arachidyl alcohol, heneicosyl alcohol, behenyl alcohol, oleyl alcohol, elaidyl alcohol, linoleyl alcohol, linolenyl alcohol, or erucyl alcohol. 22. The formulation according to claim 1, wherein the formulation is in the form of a suspension concentrate (SC), an oil-based suspension concentrate (OD), or a suspoemulsions (SE). 23. The formulation according to claim 1, wherein the spray drift reductant is present at a concentration of from 0.01 wt. % to 2 wt. %. 24. A sprayable agrochemical formulation comprising; i) in the range of from 0.01 wt. % to 1.0 wt. % spray drift reductant, wherein the reductant is non-ionic, non-self-emulsifiable, has a HLB of less than 7, and is capable of reducing spray drift by at least 10%; and ii) at least one agrochemical active and/or nutrient; and iii) optionally a clathrate compound. 25. A concentrate formulation for making a sprayable agrochemical formulation, said concentrate comprising a spray drift reductant of a fatty ester of a C3 to C8 polyol, or an oligomer thereof having from 2 to 5 repeat units. 26. A method of reducing spray drift in an agrochemical formulation comprising: applying the agrochemical formulation to vegetation, wherein the agrochemical formulation comprising a spray drift reductant comprising a fatty ester of a C3 to C8 polyol, or an oligomer thereof having from 2 to 5 repeat units, at least one agrochemical active and/or nutrient, and optionally a clathrate compound. 27. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction of spray droplets having a size of less than 150 μm of at least 10% at a spray pressure of 40 psi. 28. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction of spray droplets having a size of less than 105 μm of at least 10% at a spray pressure of 40 psi. 29. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction of spray droplets having a size of less than 500 μm of at least 5% at a spray pressure of 40 psi. 30. The method according to claim 26, wherein the spray drift reductant provides a percentage reduction in relative span is at least 4% at a spray pressure of 40 psi 31. A method of reducing spray drift comprising applying a diluted concentrate formulation in accordance with claim 25 to vegetation. 32. A method of treating vegetation to control pests and/or to provide nutrients, the method comprising applying a formulation in accordance with claim 1 either to said vegetation or to the immediate environment of said vegetation.
1,600
550
15,491,924
1,615
In various implementations, beta-hydroxybutyrate, related compounds, and/or one or more other compounds may be administered to an individual to cause weight loss, weight maintenance, elevate blood ketone levels, maintain blood ketone levels, reduce blood glucose levels, maintain blood glucose levels, improve energy, focus, mood, cognitive function, or aide with neurological or inflammatory disorders and/or combinations thereof. Other compounds may include short chain fatty acids, short chain triglycerides, medium chain fatty acids, medium chain triglycerides, long chain fatty acids, long chain triglycerides, berberine, metabolites of berberine (e.g., dihydroberberine), and/or combinations thereof.
1. A method for maintaining ketosis in an individual, the method comprising: orally administering a composition comprising approximately 0.5 g to approximately 10 g of R-beta-hydroxybutyrate; wherein administration induces or maintains ketosis in an individual. 2. The method of claim 1 wherein the amount of the composition administered comprises 0.5 to 3 g of R-beta-hydroxybutyrate. 3. The method of claim 1 wherein the composition further comprises at least one additional compound, and wherein at least one of the additional compounds comprises: a short chain fatty acid, an ester of short chain fatty acid; a medium chain fatty acid, an ester of medium chain fatty acid; a long chain fatty acid, or an ester of long chain fatty acid. 4. The method of claim 1 wherein the composition is administered up to 5 times daily. 5. The method of claim 1 wherein the composition further comprises at least one of: a flavoring, a vitamin, a mineral, or a binder. 6. The method of claim 1 wherein administration of the composition increases strength. 7. The method of claim 1 wherein administration of the composition increases mental acuity. 8. The method of claim 1 wherein administration of the composition increases at least one of metabolism, fat loss, fat oxidation, motor function, or muscle mass. 9. The method of claim 1 wherein the 0.5 to 10 g of R-beta-hydroxybutyrate comprises R-beta-hydroxybutyrate and at least one of a polymer of R-beta-hydroxybutyrate or R-beta-hydroxybutyrate-complex. 10. A composition comprising: approximately 0.5 g to approximately 10 g of R-beta-hydroxybutyrate; and one or more additional compounds capable of increasing ketone levels independently; wherein administration induces or maintains ketosis in an individual. 11. The composition of claim 10 wherein the R-beta-hydroxybutyrate comprises at least one of R-beta-hydroxybutyrate salt, R-beta-hydroxybutyrate-amino acid complex, or R-beta-hydroxybutyrate polymer. 12. The composition of claim 10 wherein at least one of the additional compounds comprises: a short chain fatty acid, an ester of short chain fatty acid; a medium chain fatty acid, an ester of medium chain fatty acid; a long chain fatty acid, or an ester of long chain fatty acid. 13. The composition of claim 10 wherein at least one of the additional compounds comprises at least one of a polymer of beta-hydroxybutyrate, D,L-beta-hydroxybutyrate, butyrate, butyric acid, or triglyceride tributyrin. 14. The composition of claim 10 wherein at least one of the additional compounds comprises at least one of berberine, dihydroberberine, or tetrahydroberberine. 15. A composition for inducing or maintaining ketosis, the composition comprising: pharmaceutically effective amounts of R-beta-hydroxybutyrate and amino acid. 16. The composition of claim 15 wherein the composition is approximately 1 g to approximately 5 grams of R-beta-hydroxybutyrate and approximately 0.5 to 2 g of amino acid. 17. The composition of claim 15 wherein the amino acid comprises Leucine. 18. The composition of claim 15 wherein the composition comprises a mixture of the R-beta-hydroxybutyrate and amino acid. 19. The composition of claim 15 wherein at least a portion of the R-beta-hydroxybutyrate is complexed with the amino acid. 20. The composition of claim 19 further comprising at least one R-beta-hydroxybutyrate salt.
In various implementations, beta-hydroxybutyrate, related compounds, and/or one or more other compounds may be administered to an individual to cause weight loss, weight maintenance, elevate blood ketone levels, maintain blood ketone levels, reduce blood glucose levels, maintain blood glucose levels, improve energy, focus, mood, cognitive function, or aide with neurological or inflammatory disorders and/or combinations thereof. Other compounds may include short chain fatty acids, short chain triglycerides, medium chain fatty acids, medium chain triglycerides, long chain fatty acids, long chain triglycerides, berberine, metabolites of berberine (e.g., dihydroberberine), and/or combinations thereof.1. A method for maintaining ketosis in an individual, the method comprising: orally administering a composition comprising approximately 0.5 g to approximately 10 g of R-beta-hydroxybutyrate; wherein administration induces or maintains ketosis in an individual. 2. The method of claim 1 wherein the amount of the composition administered comprises 0.5 to 3 g of R-beta-hydroxybutyrate. 3. The method of claim 1 wherein the composition further comprises at least one additional compound, and wherein at least one of the additional compounds comprises: a short chain fatty acid, an ester of short chain fatty acid; a medium chain fatty acid, an ester of medium chain fatty acid; a long chain fatty acid, or an ester of long chain fatty acid. 4. The method of claim 1 wherein the composition is administered up to 5 times daily. 5. The method of claim 1 wherein the composition further comprises at least one of: a flavoring, a vitamin, a mineral, or a binder. 6. The method of claim 1 wherein administration of the composition increases strength. 7. The method of claim 1 wherein administration of the composition increases mental acuity. 8. The method of claim 1 wherein administration of the composition increases at least one of metabolism, fat loss, fat oxidation, motor function, or muscle mass. 9. The method of claim 1 wherein the 0.5 to 10 g of R-beta-hydroxybutyrate comprises R-beta-hydroxybutyrate and at least one of a polymer of R-beta-hydroxybutyrate or R-beta-hydroxybutyrate-complex. 10. A composition comprising: approximately 0.5 g to approximately 10 g of R-beta-hydroxybutyrate; and one or more additional compounds capable of increasing ketone levels independently; wherein administration induces or maintains ketosis in an individual. 11. The composition of claim 10 wherein the R-beta-hydroxybutyrate comprises at least one of R-beta-hydroxybutyrate salt, R-beta-hydroxybutyrate-amino acid complex, or R-beta-hydroxybutyrate polymer. 12. The composition of claim 10 wherein at least one of the additional compounds comprises: a short chain fatty acid, an ester of short chain fatty acid; a medium chain fatty acid, an ester of medium chain fatty acid; a long chain fatty acid, or an ester of long chain fatty acid. 13. The composition of claim 10 wherein at least one of the additional compounds comprises at least one of a polymer of beta-hydroxybutyrate, D,L-beta-hydroxybutyrate, butyrate, butyric acid, or triglyceride tributyrin. 14. The composition of claim 10 wherein at least one of the additional compounds comprises at least one of berberine, dihydroberberine, or tetrahydroberberine. 15. A composition for inducing or maintaining ketosis, the composition comprising: pharmaceutically effective amounts of R-beta-hydroxybutyrate and amino acid. 16. The composition of claim 15 wherein the composition is approximately 1 g to approximately 5 grams of R-beta-hydroxybutyrate and approximately 0.5 to 2 g of amino acid. 17. The composition of claim 15 wherein the amino acid comprises Leucine. 18. The composition of claim 15 wherein the composition comprises a mixture of the R-beta-hydroxybutyrate and amino acid. 19. The composition of claim 15 wherein at least a portion of the R-beta-hydroxybutyrate is complexed with the amino acid. 20. The composition of claim 19 further comprising at least one R-beta-hydroxybutyrate salt.
1,600
551
14,701,779
1,644
The present invention relates to methods of treating immune disorders, particularly autoimmune and inflammatory disorders such as rheumatoid arthritis, and methods of producing antibodies for use in therapeutic strategies for treating such disorders. Generally, the present methods involve the use of antibodies that specifically bind to NKG2D receptors present on the surface of cells underlying the disorders.
1. A method of treating a patient with an inflammatory or autoimmune disorder, said method comprising administering to said patient a pharmaceutical composition comprising an inhibitor of an NKG2D receptor, and a pharmaceutically acceptable carrier. 2. The method of claim 1, wherein said inhibitor is a monoclonal antibody, or a fragment or derivative thereof. 3. The method of claim 2, wherein said antibody is humanized, chimeric, or human. 4. The method of claim 2, wherein said antibody is derived from the monoclonal antibody clone 1D11. 5. The method of claim 1, wherein said inhibitor interferes with the binding of an NKG2D ligand to the NKG2D receptor. 6. The method of claim 1, wherein said inflammatory or autoimmune disorder is selected from systemic lupus erythematosus, multiple sclerosis, Wegener's granulomatosus, temporal arteritis, Bechet's disease, or gastrointestinal inflammation. 7. (canceled) 8. The method of claim 1, wherein said method further comprises the administration of a pharmaceutical composition comprising a therapeutic compound selected from the group consisting of an inhibitor of TNF-alpha and an inhibitor of IL-15. 9. The method of claim 1, wherein said patient has an elevated level of NKG2D-expressing T cells. 10. The method of claim 9, wherein said T cells are CD4+. 11. The method of claim 10, wherein said T cells are CD28−. 12. The method of claim 1, wherein said method further comprises a diagnostic step in which, prior to the administration of said inhibitor, the prevalence of NKG2D-expressing CD4+CD28− T cells in said patient is assessed, wherein a detection of elevated levels of said cells in said patient indicates that the patient is suitable for the administration of said inhibitor. 13. The method of claim 12, wherein said diagnostic step comprises an immunoassay to detect the presence of CD4, CD28, or NKG2D on T cells obtained from said patient. 14. The method of claim 2, wherein said antibody is a cytotoxic antibody. 15. The method of claim 14, wherein said cytotoxic antibody comprises an element selected from the group consisting of a toxic peptide and a toxic small molecule. 16. A method of producing an antibody suitable for use in the treatment of an autoimmune or inflammatory disorder, said method comprising: i) providing a NKG2D receptor-expressing CD4+CD28− T cell and a plurality of monoclonal antibodies; ii) testing the ability of each of said antibodies to interfere with the binding of a ligand to said NKG2D receptor on said cells; iii) selecting an antibody from said plurality that reduces the binding of said ligand to said receptor; and iv) rendering said antibody suitable for human administration. 17-24. (canceled) 25. An antibody produced using the method of claim 16. 26. A pharmaceutical composition comprising an antibody produced using the method of claim 16, and a pharmaceutically acceptable carrier. 27. A kit comprising an antibody produced by the method of claim 16, and instructions for using said antibody in the treatment of rheumatoid arthritis. 28. A method for reducing CD4+ NKG2D+ autoreactive T cell stimulation in a patient in need thereof, said method comprising administering to said patient a pharmaceutical composition comprising an inhibitor of an NKG2D receptor, and a pharmaceutically acceptable carrier. 29. The method of claim 28, wherein the patient has an autoimmune disorder selected from rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, Wegener's granulomatosus, temporal arteritis, Bechet's disease or gastrointestinal inflammation.
The present invention relates to methods of treating immune disorders, particularly autoimmune and inflammatory disorders such as rheumatoid arthritis, and methods of producing antibodies for use in therapeutic strategies for treating such disorders. Generally, the present methods involve the use of antibodies that specifically bind to NKG2D receptors present on the surface of cells underlying the disorders.1. A method of treating a patient with an inflammatory or autoimmune disorder, said method comprising administering to said patient a pharmaceutical composition comprising an inhibitor of an NKG2D receptor, and a pharmaceutically acceptable carrier. 2. The method of claim 1, wherein said inhibitor is a monoclonal antibody, or a fragment or derivative thereof. 3. The method of claim 2, wherein said antibody is humanized, chimeric, or human. 4. The method of claim 2, wherein said antibody is derived from the monoclonal antibody clone 1D11. 5. The method of claim 1, wherein said inhibitor interferes with the binding of an NKG2D ligand to the NKG2D receptor. 6. The method of claim 1, wherein said inflammatory or autoimmune disorder is selected from systemic lupus erythematosus, multiple sclerosis, Wegener's granulomatosus, temporal arteritis, Bechet's disease, or gastrointestinal inflammation. 7. (canceled) 8. The method of claim 1, wherein said method further comprises the administration of a pharmaceutical composition comprising a therapeutic compound selected from the group consisting of an inhibitor of TNF-alpha and an inhibitor of IL-15. 9. The method of claim 1, wherein said patient has an elevated level of NKG2D-expressing T cells. 10. The method of claim 9, wherein said T cells are CD4+. 11. The method of claim 10, wherein said T cells are CD28−. 12. The method of claim 1, wherein said method further comprises a diagnostic step in which, prior to the administration of said inhibitor, the prevalence of NKG2D-expressing CD4+CD28− T cells in said patient is assessed, wherein a detection of elevated levels of said cells in said patient indicates that the patient is suitable for the administration of said inhibitor. 13. The method of claim 12, wherein said diagnostic step comprises an immunoassay to detect the presence of CD4, CD28, or NKG2D on T cells obtained from said patient. 14. The method of claim 2, wherein said antibody is a cytotoxic antibody. 15. The method of claim 14, wherein said cytotoxic antibody comprises an element selected from the group consisting of a toxic peptide and a toxic small molecule. 16. A method of producing an antibody suitable for use in the treatment of an autoimmune or inflammatory disorder, said method comprising: i) providing a NKG2D receptor-expressing CD4+CD28− T cell and a plurality of monoclonal antibodies; ii) testing the ability of each of said antibodies to interfere with the binding of a ligand to said NKG2D receptor on said cells; iii) selecting an antibody from said plurality that reduces the binding of said ligand to said receptor; and iv) rendering said antibody suitable for human administration. 17-24. (canceled) 25. An antibody produced using the method of claim 16. 26. A pharmaceutical composition comprising an antibody produced using the method of claim 16, and a pharmaceutically acceptable carrier. 27. A kit comprising an antibody produced by the method of claim 16, and instructions for using said antibody in the treatment of rheumatoid arthritis. 28. A method for reducing CD4+ NKG2D+ autoreactive T cell stimulation in a patient in need thereof, said method comprising administering to said patient a pharmaceutical composition comprising an inhibitor of an NKG2D receptor, and a pharmaceutically acceptable carrier. 29. The method of claim 28, wherein the patient has an autoimmune disorder selected from rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, Wegener's granulomatosus, temporal arteritis, Bechet's disease or gastrointestinal inflammation.
1,600
552
15,199,108
1,618
The present invention relates to an improved synthesis of N-monofluoroalkyl tropanes using fluoroalkyl iodide. The invention also provides the use of such method to prepare the non-radioactive tropane intermediate FP-CIT, and its subsequent conversion to the 123 I-labelled radiopharmaceutical DaTSCAN™ ( 123 I-ioflupane). Also provided is the use of fluoroalkyl iodide in the alkylation method of the invention.
1. A method of preparation of a non-radioactive N-monofluoroalkyl tropane compound of Formula (IIIA) which method comprises: (iii) provision of an amine of Formula (III): and (iii) alkylation of said amine with an alkylating agent of formula F—(CH2)mI, in the presence of a base in a suitable solvent, to give a reaction product comprising the N-monofluoroalkyl tropane of Formula (IIIA) and less than 1% of a transesterified impurity compound of Formula (V): wherein m is 2, 3, or 4. 2. The method of claim 1, wherein the amount of said alkylating agent is 1.0-1.1 equivalents, the amount of said base is 1.0-1.1 equivalents, and the amount of the amine of Formula (III) is 1 equivalent. 3. The method of claim 2, wherein the amount of said alkylating agent is 1.1 equivalents, and the amount of said base is 1.1 equivalents. 4. The method of claim 2, wherein said base is triethylamine 5. The method of claim 1, wherein said alkylation in step (ii) comprises heating in said suitable solvent. 6. The method of claim 5, wherein said heating is at 50-120° C. 7. The method of claim 1, wherein said suitable solvent is toluene. 8. The method of claim 1, wherein said alkylation in step (ii) comprises refluxing in said suitable solvent. 9. The method of claim 8, wherein said refluxing is done for a period of time that is about 2 hours. 10. The method of claim 1, wherein said reaction product comprises at least 90% of said compound of Formula (IIIA). 11. The method of claim 1, wherein the amount of said alkylating agent is 1.1 equivalents, the amount of said amine of Formula (III) is 1 equivalent, the amount of said base is 1.1 equivalents, and said alkylation in step (ii) comprises heating in a suitable solvent. 12. The method of claim 1, further comprising filtration and flash chromatographic purification of said reaction product from step (ii). 13. A method of preparation of a non-radioactive trialkyltin radioiodination precursor of Formula IV: wherein said method comprises: (i) provision of an amine of Formula (III): (i) alkylation of said amine with an alkylating agent of formula F—(CH2)mI in the presence of a base in a suitable solvent, to give a reaction product comprising the N-monofluoroalkyl tropane compound of Formula (IIIA): and less than 1% of a transesterified impurity compound of Formula (V): wherein m is 2, 3, or 4; and (ii) reaction of said reaction product from step (ii) with Sn2Rb 6 in the presence of a suitable catalyst to give the desired radioiodination precursor of Formula (IV) wherein each Rb is independently C1-4alkyl, and m is 2, 3, or 4. 14. A method of preparation of a radioiodinated tropane of Formula (IIIB): wherein m is 2, 3 or 4, and said method comprises: (i) provision of an amine of Formula (III): (ii) alkylation of said amine with an alkylating agent of formula F—(CH2)mI in the presence of a base in a suitable solvent, to give a reaction product comprising the N-monofluoroalkyl tropane of Formula (IIIA): and less than 1% of a transesterified impurity compound of Formula (V): wherein m is 2, 3, or 4; (iii) reaction of said reaction product from step (ii) with Sn2Rb 6 in the presence of a suitable catalyst to give a non-radioactive trialkyltin radioiodination precursor of Formula (IV): wherein m is 2, 3 or 4, and each Rb is independently C1-4 alkyl; and (iv) reacting the trialkyltin precursor of Formula IV from step (iii) with a supply of radioactive iodide [xI]-iodide, in the presence of a suitable oxidising agent to give the desired product of Formula (IIIB) wherein xI is 124I or 123I. 15. The method of claim 14, where xI is 123I. 16. The method of claim 15, wherein the radioiodinated tropane of Formula (IIIB) is provided as a pharmaceutical composition together with a biocompatible carrier medium. 17. The method of claim 14, wherein in step (ii), the amount of said alkylating agent is 1.0-1.1 equivalents, the amount of said base is 1.0-1.1 equivalents, and the amount of the amine of Formula (III) is 1 equivalent. 18. The method of claim 17, wherein said base is triethylamine 19. The method of claim 14, wherein said suitable solvent in step (ii) is toluene. 20. The method of claim 14, wherein said reaction product in step (ii) comprises at least 90% of said compound of Formula (IIIA). 21. The method of claim 14, wherein m is 3.
The present invention relates to an improved synthesis of N-monofluoroalkyl tropanes using fluoroalkyl iodide. The invention also provides the use of such method to prepare the non-radioactive tropane intermediate FP-CIT, and its subsequent conversion to the 123 I-labelled radiopharmaceutical DaTSCAN™ ( 123 I-ioflupane). Also provided is the use of fluoroalkyl iodide in the alkylation method of the invention.1. A method of preparation of a non-radioactive N-monofluoroalkyl tropane compound of Formula (IIIA) which method comprises: (iii) provision of an amine of Formula (III): and (iii) alkylation of said amine with an alkylating agent of formula F—(CH2)mI, in the presence of a base in a suitable solvent, to give a reaction product comprising the N-monofluoroalkyl tropane of Formula (IIIA) and less than 1% of a transesterified impurity compound of Formula (V): wherein m is 2, 3, or 4. 2. The method of claim 1, wherein the amount of said alkylating agent is 1.0-1.1 equivalents, the amount of said base is 1.0-1.1 equivalents, and the amount of the amine of Formula (III) is 1 equivalent. 3. The method of claim 2, wherein the amount of said alkylating agent is 1.1 equivalents, and the amount of said base is 1.1 equivalents. 4. The method of claim 2, wherein said base is triethylamine 5. The method of claim 1, wherein said alkylation in step (ii) comprises heating in said suitable solvent. 6. The method of claim 5, wherein said heating is at 50-120° C. 7. The method of claim 1, wherein said suitable solvent is toluene. 8. The method of claim 1, wherein said alkylation in step (ii) comprises refluxing in said suitable solvent. 9. The method of claim 8, wherein said refluxing is done for a period of time that is about 2 hours. 10. The method of claim 1, wherein said reaction product comprises at least 90% of said compound of Formula (IIIA). 11. The method of claim 1, wherein the amount of said alkylating agent is 1.1 equivalents, the amount of said amine of Formula (III) is 1 equivalent, the amount of said base is 1.1 equivalents, and said alkylation in step (ii) comprises heating in a suitable solvent. 12. The method of claim 1, further comprising filtration and flash chromatographic purification of said reaction product from step (ii). 13. A method of preparation of a non-radioactive trialkyltin radioiodination precursor of Formula IV: wherein said method comprises: (i) provision of an amine of Formula (III): (i) alkylation of said amine with an alkylating agent of formula F—(CH2)mI in the presence of a base in a suitable solvent, to give a reaction product comprising the N-monofluoroalkyl tropane compound of Formula (IIIA): and less than 1% of a transesterified impurity compound of Formula (V): wherein m is 2, 3, or 4; and (ii) reaction of said reaction product from step (ii) with Sn2Rb 6 in the presence of a suitable catalyst to give the desired radioiodination precursor of Formula (IV) wherein each Rb is independently C1-4alkyl, and m is 2, 3, or 4. 14. A method of preparation of a radioiodinated tropane of Formula (IIIB): wherein m is 2, 3 or 4, and said method comprises: (i) provision of an amine of Formula (III): (ii) alkylation of said amine with an alkylating agent of formula F—(CH2)mI in the presence of a base in a suitable solvent, to give a reaction product comprising the N-monofluoroalkyl tropane of Formula (IIIA): and less than 1% of a transesterified impurity compound of Formula (V): wherein m is 2, 3, or 4; (iii) reaction of said reaction product from step (ii) with Sn2Rb 6 in the presence of a suitable catalyst to give a non-radioactive trialkyltin radioiodination precursor of Formula (IV): wherein m is 2, 3 or 4, and each Rb is independently C1-4 alkyl; and (iv) reacting the trialkyltin precursor of Formula IV from step (iii) with a supply of radioactive iodide [xI]-iodide, in the presence of a suitable oxidising agent to give the desired product of Formula (IIIB) wherein xI is 124I or 123I. 15. The method of claim 14, where xI is 123I. 16. The method of claim 15, wherein the radioiodinated tropane of Formula (IIIB) is provided as a pharmaceutical composition together with a biocompatible carrier medium. 17. The method of claim 14, wherein in step (ii), the amount of said alkylating agent is 1.0-1.1 equivalents, the amount of said base is 1.0-1.1 equivalents, and the amount of the amine of Formula (III) is 1 equivalent. 18. The method of claim 17, wherein said base is triethylamine 19. The method of claim 14, wherein said suitable solvent in step (ii) is toluene. 20. The method of claim 14, wherein said reaction product in step (ii) comprises at least 90% of said compound of Formula (IIIA). 21. The method of claim 14, wherein m is 3.
1,600
553
14,310,201
1,618
Peptides having activity as protein binding agents are disclosed. The peptides have the following structure (I): including stereoisomers, pharmaceutically acceptable salts and prodrugs thereof, wherein R, R 1 , L 1 , L 2 , G, M, Y 1 Y 2 and SEQ are as defined herein. Methods associated with preparation and use of such peptides, as well as pharmaceutical compositions comprising such peptides, are also disclosed.
1. A cyclic peptide having the following structure (I): or a salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each individually optionally substituted linker moieties, each linker moiety optionally comprising a linkage to a solid support, a linkage to a reporter moiety, a linkage to a peptide ligand, a linkage to an azide or alkyne moiety or combinations thereof; G is a triazole, a carbon-carbon double bond or an amide; M is methionine; R is H, -L3-A or —C(═O)-L3-A, where L3 is a linker moiety and A is an alkyne, azide or a bond to a peptide ligand; R1 is H or C1-C6alkyl; Y1 and Y2 are each individually 0 or 1; and SEQ is an amino acid sequence comprising from 2 to 20 amino acids selected from natural and non-natural amino acids. 2. The cyclic peptide of claim 1, wherein G is a triazole. 3. The cyclic peptide of claim 1, wherein G is a carbon-carbon double bond. 4. The cyclic peptide of claim 1, wherein L1, L2, or both, comprise one or more substituents selected from alkyl, alkyne, azide and aminocarbonyl. 5. The cyclic peptide of claim 1, wherein L1, L2, or both, comprise a linkage selected from a linkage to a solid support, a linkage to a reporter moiety and a linkage to a peptide ligand. 6. The cyclic peptide of claim 1, wherein L1 and L2 are alkylene. 7. The cyclic peptide of claim 1, wherein the cyclic peptide has one of the following structures (Ia) or (Ib): wherein: R3 is H, a linkage to a solid support, a linkage to a reporter moiety, a linkage to a peptide ligand, a linkage to an azide or alkyne moiety or combinations thereof; and x and y are each independently an integer from 1 to 8. 8. The cyclic peptide of claim 1, wherein SEQ comprises from 2 to 9 amino acids. 9. The cyclic peptide of claim 8, wherein SEQ comprises from 5 to 7 amino acids. 10. The cyclic peptide of claim 1, wherein the amino acids are selected from D and L stereoisomers of Ala, Gly, Leu, Ile, Val, Phe, Tip, Arg, His, Lys, Asp, Glu, Asn, Gln, Ser, Thr, Tyr and Pro. 11. The cyclic peptide of claim 1, wherein R is H or —C(═O)-L3-A, where L3 is a linker moiety and A is a bond to a peptide ligand or an alkyne. 12. The cyclic peptide of claim 11, wherein A is an alkyne. 13. The cyclic peptide of claim 11, wherein A is a bond to a peptide ligand. 14. The cyclic peptide of claim 13, wherein the peptide ligand is a linear peptide. 15. The cyclic peptide of claim 13, wherein the peptide ligand is a cyclic peptide. 16. The cyclic peptide of claim 14, wherein the peptide ligand further comprises a second peptide ligand covalently bound thereto. 17. The cyclic peptide of claim 1, wherein SEQ has specific affinity for a binding region on Akt, pfLDH, pvLDH, pfHRP(II) or HIV-1 p24. 18. The cyclic peptide of claim 1, wherein SEQ has 90% sequence identity to any one of SEQ ID NOS: 1-33. 19. The cyclic peptide of claim 18, wherein SEQ comprises any one of SEQ ID NOS: 1-14. 20. The cyclic peptide of claim 1, wherein y1 and y2 are each 0. 21. A composition comprising the cyclic peptide of claim 1 and a pharmaceutically acceptable carrier. 22. A library comprising a plurality of cyclic peptides according to claim 1. 23. A method for identifying a target binding compound, the method comprising: A) providing a first peptide library comprising a plurality of first peptide library members, the first peptide library members optionally comprising an alkyne, azide or reporter moiety or combinations thereof; B) contacting the first peptide library with a target or a truncated analogue thereof, the target or truncated analogue thereof comprising a first binding site and optionally an alkyne, azide or reporter moiety or combinations thereof; C) identifying a first peptide library member with affinity for the first binding site and optionally modifying the first peptide library member to include an alkyne or azide moiety; and optionally: D) providing a second peptide library comprising a plurality of second peptide library members, the second peptide library members comprising an azide or alkyne or both; E) contacting the second peptide library with a composition comprising the target or truncated analogue thereof and the first peptide library member of step C; F) forming a triazole-linked conjugate between the first peptide library member of step C and a second peptide library member, the second peptide library member having affinity for a second binding site on the target or truncated analogue thereof, wherein the first peptide library, the second peptide library, or both, comprise cyclic peptides comprising: i. a sequence region comprising amino and carboxy termini and a variable peptide sequence of two to twenty amino acids selected from natural and non-natural amino acids; and ii. a linker region comprising a α-amino carbonyl, α-amido carbonyl, a methionine amino acid, or combinations thereof, and optionally comprising an alkyne, an azide, a linkage to a solid support or a linkage to a reporter moiety or a combination thereof, the linker region covalently linking the amino and carboxy termini of the sequence region. 24-52. (canceled) 53. A method of detecting Akt in a sample, the method comprising replacing an antibody or its equivalent in a cell-based or an immunoassay with the cyclic peptide of claim 1. 54. (canceled) 55. A method for inhibiting activity of a protein in a subject, the method comprising administering an effective amount of the cyclic peptide of claim 1 to a subject in need thereof. 56-58. (canceled) 59. A method of purifying a target, the method comprising immobilizing the cyclic peptide of claim 1 in a column based format, contacting the column with a matrix containing the target, washing the column, and eluting the target. 60. A method of imaging in vivo target expression, the method comprising: a) providing a cyclic peptide of claim 1, wherein SEQ is a peptide sequence having affinity for a location on or near a target expressing site in a subject, and modifying the cyclic peptide to include a small-molecule positron-emission-tomography ligand (PET ligand); b) administering the cyclic peptide of step a) to the subject; c) measuring the positron emission from the PET ligand at a first time; d) measuring the positron emission from the PET ligand at a second time; and e) comparing the positron emission from the PET ligand at the first and second times. 61. (canceled) 62. A method of imaging in vivo target expression, the method comprising: a) providing a cyclic peptide of claim 1, wherein SEQ is a peptide sequence having affinity for a location on or near a target expressing site in a subject, and modifying the cyclic peptide to include a small-molecule single-photon-emission-computed-tomography ligand (SPECT ligand); b) administering the cyclic peptide of step a) to the subject; c) measuring the photon emission from the SPECT ligand at a first time; d) measuring the photon emission from the SPECT ligand at a second time; and e) comparing the photon emission from the SPECT ligand at the first and second times. 63. (canceled) 64. A method of imaging in vivo target expression, the method comprising: a) providing a cyclic peptide of claim 1, wherein SEQ is a peptide sequence having affinity for a location on or near a target expressing site in a subject, and modifying the cyclic peptide to include a magnetic resonance ligand (MR ligand); b) administering the cyclic peptide of step a) to the subject; c) measuring the magnetic resonance from the MR ligand at a first time; d) measuring the magnetic resonance from the MR ligand at a second time; and e) comparing the magnetic resonance from the MR ligand at the first and second times. 65. (canceled) 66. The cyclic peptide of claim 1, wherein the cyclic peptide comprises a linkage to a reporter moiety, the reporter moiety selected from polyethylene glycol (PEG), biotin, thiol and fluorophores. 67. The cyclic peptide of claim 66, wherein the fluorophores are selected from carboxyfluorescein (FAM), fluorescein isothiocyanate (FITC), Cyanine-5 (Cy5), tetramethylrhodamine (TRITC) and Carboxytetramethylrhodamine (TAMRA).
Peptides having activity as protein binding agents are disclosed. The peptides have the following structure (I): including stereoisomers, pharmaceutically acceptable salts and prodrugs thereof, wherein R, R 1 , L 1 , L 2 , G, M, Y 1 Y 2 and SEQ are as defined herein. Methods associated with preparation and use of such peptides, as well as pharmaceutical compositions comprising such peptides, are also disclosed.1. A cyclic peptide having the following structure (I): or a salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each individually optionally substituted linker moieties, each linker moiety optionally comprising a linkage to a solid support, a linkage to a reporter moiety, a linkage to a peptide ligand, a linkage to an azide or alkyne moiety or combinations thereof; G is a triazole, a carbon-carbon double bond or an amide; M is methionine; R is H, -L3-A or —C(═O)-L3-A, where L3 is a linker moiety and A is an alkyne, azide or a bond to a peptide ligand; R1 is H or C1-C6alkyl; Y1 and Y2 are each individually 0 or 1; and SEQ is an amino acid sequence comprising from 2 to 20 amino acids selected from natural and non-natural amino acids. 2. The cyclic peptide of claim 1, wherein G is a triazole. 3. The cyclic peptide of claim 1, wherein G is a carbon-carbon double bond. 4. The cyclic peptide of claim 1, wherein L1, L2, or both, comprise one or more substituents selected from alkyl, alkyne, azide and aminocarbonyl. 5. The cyclic peptide of claim 1, wherein L1, L2, or both, comprise a linkage selected from a linkage to a solid support, a linkage to a reporter moiety and a linkage to a peptide ligand. 6. The cyclic peptide of claim 1, wherein L1 and L2 are alkylene. 7. The cyclic peptide of claim 1, wherein the cyclic peptide has one of the following structures (Ia) or (Ib): wherein: R3 is H, a linkage to a solid support, a linkage to a reporter moiety, a linkage to a peptide ligand, a linkage to an azide or alkyne moiety or combinations thereof; and x and y are each independently an integer from 1 to 8. 8. The cyclic peptide of claim 1, wherein SEQ comprises from 2 to 9 amino acids. 9. The cyclic peptide of claim 8, wherein SEQ comprises from 5 to 7 amino acids. 10. The cyclic peptide of claim 1, wherein the amino acids are selected from D and L stereoisomers of Ala, Gly, Leu, Ile, Val, Phe, Tip, Arg, His, Lys, Asp, Glu, Asn, Gln, Ser, Thr, Tyr and Pro. 11. The cyclic peptide of claim 1, wherein R is H or —C(═O)-L3-A, where L3 is a linker moiety and A is a bond to a peptide ligand or an alkyne. 12. The cyclic peptide of claim 11, wherein A is an alkyne. 13. The cyclic peptide of claim 11, wherein A is a bond to a peptide ligand. 14. The cyclic peptide of claim 13, wherein the peptide ligand is a linear peptide. 15. The cyclic peptide of claim 13, wherein the peptide ligand is a cyclic peptide. 16. The cyclic peptide of claim 14, wherein the peptide ligand further comprises a second peptide ligand covalently bound thereto. 17. The cyclic peptide of claim 1, wherein SEQ has specific affinity for a binding region on Akt, pfLDH, pvLDH, pfHRP(II) or HIV-1 p24. 18. The cyclic peptide of claim 1, wherein SEQ has 90% sequence identity to any one of SEQ ID NOS: 1-33. 19. The cyclic peptide of claim 18, wherein SEQ comprises any one of SEQ ID NOS: 1-14. 20. The cyclic peptide of claim 1, wherein y1 and y2 are each 0. 21. A composition comprising the cyclic peptide of claim 1 and a pharmaceutically acceptable carrier. 22. A library comprising a plurality of cyclic peptides according to claim 1. 23. A method for identifying a target binding compound, the method comprising: A) providing a first peptide library comprising a plurality of first peptide library members, the first peptide library members optionally comprising an alkyne, azide or reporter moiety or combinations thereof; B) contacting the first peptide library with a target or a truncated analogue thereof, the target or truncated analogue thereof comprising a first binding site and optionally an alkyne, azide or reporter moiety or combinations thereof; C) identifying a first peptide library member with affinity for the first binding site and optionally modifying the first peptide library member to include an alkyne or azide moiety; and optionally: D) providing a second peptide library comprising a plurality of second peptide library members, the second peptide library members comprising an azide or alkyne or both; E) contacting the second peptide library with a composition comprising the target or truncated analogue thereof and the first peptide library member of step C; F) forming a triazole-linked conjugate between the first peptide library member of step C and a second peptide library member, the second peptide library member having affinity for a second binding site on the target or truncated analogue thereof, wherein the first peptide library, the second peptide library, or both, comprise cyclic peptides comprising: i. a sequence region comprising amino and carboxy termini and a variable peptide sequence of two to twenty amino acids selected from natural and non-natural amino acids; and ii. a linker region comprising a α-amino carbonyl, α-amido carbonyl, a methionine amino acid, or combinations thereof, and optionally comprising an alkyne, an azide, a linkage to a solid support or a linkage to a reporter moiety or a combination thereof, the linker region covalently linking the amino and carboxy termini of the sequence region. 24-52. (canceled) 53. A method of detecting Akt in a sample, the method comprising replacing an antibody or its equivalent in a cell-based or an immunoassay with the cyclic peptide of claim 1. 54. (canceled) 55. A method for inhibiting activity of a protein in a subject, the method comprising administering an effective amount of the cyclic peptide of claim 1 to a subject in need thereof. 56-58. (canceled) 59. A method of purifying a target, the method comprising immobilizing the cyclic peptide of claim 1 in a column based format, contacting the column with a matrix containing the target, washing the column, and eluting the target. 60. A method of imaging in vivo target expression, the method comprising: a) providing a cyclic peptide of claim 1, wherein SEQ is a peptide sequence having affinity for a location on or near a target expressing site in a subject, and modifying the cyclic peptide to include a small-molecule positron-emission-tomography ligand (PET ligand); b) administering the cyclic peptide of step a) to the subject; c) measuring the positron emission from the PET ligand at a first time; d) measuring the positron emission from the PET ligand at a second time; and e) comparing the positron emission from the PET ligand at the first and second times. 61. (canceled) 62. A method of imaging in vivo target expression, the method comprising: a) providing a cyclic peptide of claim 1, wherein SEQ is a peptide sequence having affinity for a location on or near a target expressing site in a subject, and modifying the cyclic peptide to include a small-molecule single-photon-emission-computed-tomography ligand (SPECT ligand); b) administering the cyclic peptide of step a) to the subject; c) measuring the photon emission from the SPECT ligand at a first time; d) measuring the photon emission from the SPECT ligand at a second time; and e) comparing the photon emission from the SPECT ligand at the first and second times. 63. (canceled) 64. A method of imaging in vivo target expression, the method comprising: a) providing a cyclic peptide of claim 1, wherein SEQ is a peptide sequence having affinity for a location on or near a target expressing site in a subject, and modifying the cyclic peptide to include a magnetic resonance ligand (MR ligand); b) administering the cyclic peptide of step a) to the subject; c) measuring the magnetic resonance from the MR ligand at a first time; d) measuring the magnetic resonance from the MR ligand at a second time; and e) comparing the magnetic resonance from the MR ligand at the first and second times. 65. (canceled) 66. The cyclic peptide of claim 1, wherein the cyclic peptide comprises a linkage to a reporter moiety, the reporter moiety selected from polyethylene glycol (PEG), biotin, thiol and fluorophores. 67. The cyclic peptide of claim 66, wherein the fluorophores are selected from carboxyfluorescein (FAM), fluorescein isothiocyanate (FITC), Cyanine-5 (Cy5), tetramethylrhodamine (TRITC) and Carboxytetramethylrhodamine (TAMRA).
1,600
554
13,635,075
1,644
The present invention provides compositions and methods for generating and cryopreserving dendritic cells with superior functionality in producing stronger signals to T cells, resulting in a more potent DC-based anti-tumor vaccine. The present invention includes mature, antigen loaded DCs activated by Toll-like receptor agonists that induce clinically effective immune responses, preferably when used earlier in the disease process. The DCs of the present invention produce desirable levels of cytokines and chemokines, and further have the capacity to induce apoptosis of tumor cells. The cells can be cryopreserved and thawed for later use, thereby reducing the need for repeated pheresis and elutriation processes during vaccine production. These methods can also be utilized to directly target molecules involved in carcinogenetic signaling pathways and cancer stem cells.
1. A method of generating antigen loaded, activated dendritic cells (DC) for use in immunotherapy, comprising: loading at least one antigen into a DC; activating the DC with at least one TLR agonist; cryopreserving the DC; and thawing the DC; wherein the DC produces an effective amount of at least one cytokine to generate a T cell response. 2. The method of claim 1, wherein the antigen is a tumor antigen. 3. The method of claim 1, wherein the antigen is a microbial antigen. 4. The method of claim 1, wherein the TLR agonist is LPS. 5. The method of claim 1, wherein the cryopreserving comprises freezing the DC at a temperature of about −70° C. or lower. 6. The method of claim 1, wherein the recovery and viability of the DC after thawing is greater than or equal to about 70%. 7. The method of claim 1, wherein the recovery and viability of the DC after thawing is greater than or equal to about 80%. 8. The method of claim 1, wherein the DC are cryopreserved for at least about one week. 9. The method of claim 1, wherein the cytokine is IL12. 10. The method of claim 1, wherein the DC exhibits a killer function whereby the DC are capable of lysing targeted cancer cells. 11. A method of eliciting an immune response in a mammal, the method comprising administering a previously cryopreserved composition comprising an antigen loaded, activated DC into the mammal in need thereof, wherein the DC is antigen loaded and activated prior to being cryopreserved. 12. The method of claim 11, wherein the antigen is a tumor antigen. 13. The method of claim 11, wherein the antigen is a microbial antigen. 14. The method of claim 11, wherein the TLR agonist is LPS. 15. The method of claim 11, wherein the cryopreserving comprises freezing the DC at a temperature of about −70° C. or lower. 16. The method of claim 11, wherein the recovery and viability of the DC alter thawing is greater than or equal to about 70%. 17. The method of claim 11, wherein the recovery and viability of the DC after thawing is greater than or equal to about 80%. 18. The method of claim 11, wherein the DC are cryopreserved for at least about one week. 19. The method of claim 11, wherein the cytokine is IL12. 20. The method of claim 11, wherein the DC exhibits a killer function whereby the DC are capable of lysing targeted cancer cells. 21. A preservable composition for eliciting an immune response in a mammal, the composition comprising: a DC loaded with at least one antigen; wherein the DC has been activated by exposure to at least one TLR agonist; and wherein the DC produces an effective amount of at least one cytokine to generate a T cell response, irrespective of whether or not the composition has been cryopreserved. 22. The composition of claim 21, wherein the antigen is a tumor antigen. 23. The composition of claim 21, wherein the antigen is a microbial antigen. 24. The composition of claim 21, wherein the TLR agonist is LPS. 25. The composition of claim 21, wherein the composition has been cryopreserved at a temperature of about −70° C. or lower. 26. The composition of claim 25, wherein the recovery and viability of the DC after thawing is greater than or equal to about 70%. 27. The composition of claim 25, wherein the recovery and viability of the DC after thawing is greater than or equal to about 80%. 28. The composition of claim 25, wherein the composition is cryopreserved for at least about one week. 29. The composition of claim 21, wherein the cytokine is IL12. 30. The composition of claim 21, wherein the DC exhibits a killer function whereby the DC are capable of lysing targeted cancer cells.
The present invention provides compositions and methods for generating and cryopreserving dendritic cells with superior functionality in producing stronger signals to T cells, resulting in a more potent DC-based anti-tumor vaccine. The present invention includes mature, antigen loaded DCs activated by Toll-like receptor agonists that induce clinically effective immune responses, preferably when used earlier in the disease process. The DCs of the present invention produce desirable levels of cytokines and chemokines, and further have the capacity to induce apoptosis of tumor cells. The cells can be cryopreserved and thawed for later use, thereby reducing the need for repeated pheresis and elutriation processes during vaccine production. These methods can also be utilized to directly target molecules involved in carcinogenetic signaling pathways and cancer stem cells.1. A method of generating antigen loaded, activated dendritic cells (DC) for use in immunotherapy, comprising: loading at least one antigen into a DC; activating the DC with at least one TLR agonist; cryopreserving the DC; and thawing the DC; wherein the DC produces an effective amount of at least one cytokine to generate a T cell response. 2. The method of claim 1, wherein the antigen is a tumor antigen. 3. The method of claim 1, wherein the antigen is a microbial antigen. 4. The method of claim 1, wherein the TLR agonist is LPS. 5. The method of claim 1, wherein the cryopreserving comprises freezing the DC at a temperature of about −70° C. or lower. 6. The method of claim 1, wherein the recovery and viability of the DC after thawing is greater than or equal to about 70%. 7. The method of claim 1, wherein the recovery and viability of the DC after thawing is greater than or equal to about 80%. 8. The method of claim 1, wherein the DC are cryopreserved for at least about one week. 9. The method of claim 1, wherein the cytokine is IL12. 10. The method of claim 1, wherein the DC exhibits a killer function whereby the DC are capable of lysing targeted cancer cells. 11. A method of eliciting an immune response in a mammal, the method comprising administering a previously cryopreserved composition comprising an antigen loaded, activated DC into the mammal in need thereof, wherein the DC is antigen loaded and activated prior to being cryopreserved. 12. The method of claim 11, wherein the antigen is a tumor antigen. 13. The method of claim 11, wherein the antigen is a microbial antigen. 14. The method of claim 11, wherein the TLR agonist is LPS. 15. The method of claim 11, wherein the cryopreserving comprises freezing the DC at a temperature of about −70° C. or lower. 16. The method of claim 11, wherein the recovery and viability of the DC alter thawing is greater than or equal to about 70%. 17. The method of claim 11, wherein the recovery and viability of the DC after thawing is greater than or equal to about 80%. 18. The method of claim 11, wherein the DC are cryopreserved for at least about one week. 19. The method of claim 11, wherein the cytokine is IL12. 20. The method of claim 11, wherein the DC exhibits a killer function whereby the DC are capable of lysing targeted cancer cells. 21. A preservable composition for eliciting an immune response in a mammal, the composition comprising: a DC loaded with at least one antigen; wherein the DC has been activated by exposure to at least one TLR agonist; and wherein the DC produces an effective amount of at least one cytokine to generate a T cell response, irrespective of whether or not the composition has been cryopreserved. 22. The composition of claim 21, wherein the antigen is a tumor antigen. 23. The composition of claim 21, wherein the antigen is a microbial antigen. 24. The composition of claim 21, wherein the TLR agonist is LPS. 25. The composition of claim 21, wherein the composition has been cryopreserved at a temperature of about −70° C. or lower. 26. The composition of claim 25, wherein the recovery and viability of the DC after thawing is greater than or equal to about 70%. 27. The composition of claim 25, wherein the recovery and viability of the DC after thawing is greater than or equal to about 80%. 28. The composition of claim 25, wherein the composition is cryopreserved for at least about one week. 29. The composition of claim 21, wherein the cytokine is IL12. 30. The composition of claim 21, wherein the DC exhibits a killer function whereby the DC are capable of lysing targeted cancer cells.
1,600
555
15,475,519
1,619
A personal care composition for cosmetic treatment of skin that provides an improved moisturization signal to a user and exhibits good feel properties. The composition includes a dimethicone fluid or a blend of dimethicone fluids with a viscosity of greater than 1000 centistokes (cSt). The dimethicone fluid(s) are configured to provide the composition with a Time Weighted Force Area of between 5×104 and 12×104 and a Mean Break Time of between 0.30 and 0.75.
1. A personal care composition, comprising: a) about 5% to about 30%, by weight of the composition, of a blend of at least two dimethicone fluids, wherein the blend of dimethicone fluids exhibits a viscosity of greater than about 1000 centistokes (cSt) according to the Rheology Method; and b) a dermatologically acceptable carrier, wherein the composition exhibits a Time Weighted Force Area of between about 5×104 and about 12×104 and a Mean Break Time of between about 0.30 and about 0.75 according to the Tack Method. 2. The personal care composition of claim 1, wherein the blend of dimethicone fluids has a viscosity of less than about 10,000 Stokes. 3. The personal care composition of claim 1, wherein a first dimethicone fluid in the blend has a viscosity of about 1000 cSt and a second dimethicone in the blend has a viscosity of about 60,000 cSt. 4. The personal care composition of claim 3, wherein the first and second dimethicone fluids are present at a ratio of between about 20:1 and about 1:1. 5. The personal care composition of claim 1, further comprising about 1% to about 30%, by weight, of a silicone elastomer. 6. The personal care composition of claim 5, further comprising a weight ratio of silicone elastomer to dimethicone fluid of about 1:10 to about 1:1. 7. The personal care composition of claim 1, further comprising an initial break time of greater than about 1.0. 8. The personal care composition of claim 1, further comprising a tack force at 60 minutes, 80 minutes, or 100 minutes of greater than about 1000. 9. The personal care composition of claim 1, further comprising about 0.01% to about 5%, by weight, of a superabsorbent polymer. 10. The personal care composition of claim 1, further comprising an aqueous phase, wherein the superabsorbent polymer is present in the aqueous phase. 11. The personal care composition of claim 10, wherein the superabsorbent polymer has a non-swollen, number average particle size of between about 2 microns and 100 microns. 12. The personal care composition of claim 1, further comprising a skin care active selected from the group consisting of vitamins, minerals, peptides, oil control agents, anti-oxidants, anti-inflammatory agents, moisturizing agents, emollients, humectants, exfoliating agents, skin lightening agents, sunscreens, anti-acne actives, anti-wrinkle actives, antimicrobials, and combinations of these. 13. The personal care composition of claim 12, wherein the skin care active is a vitamin comprising a stabilized retinoid. 14. The personal care composition of claim 13, wherein the retinoid is stabilized by a fatty acid ester present at a ratio of fatty acid ester to retinoid of about 5:1 to about 50:1. 15. The personal care composition of claim 11, wherein the fatty acid ester comprises capric/caprylic triglyceride. 16. A method of cosmetically treating a skin condition with a composition that provides an improved moisture perception, comprising: a) identifying a target portion of skin where skin moisturization is needed or desired; and b) applying the skin care composition of claim 1 to the target portion of skin. 17. A personal care composition, comprising: a) about 10% to about 20%, by weight, of a dimethicone fluid, wherein the dimethicone fluid has a viscosity between about 1000 cSt and about 1,000,000 cSt according to the Rheology Method, and exhibits a Time Weighted Force Area of between about 5×104 and about 12×104 and a Mean Break Time of between about 0.30 and about 0.75 according to the Tack Method; and b) a dermatologically acceptable carrier. 18. The personal care composition of claim 17, further comprising an initial break time of greater than about 1.0. 19. The personal care composition of claim 1, further comprising a tack force of greater than about 1000 at 60 minutes, 80 minutes, and/or 100 minutes.
A personal care composition for cosmetic treatment of skin that provides an improved moisturization signal to a user and exhibits good feel properties. The composition includes a dimethicone fluid or a blend of dimethicone fluids with a viscosity of greater than 1000 centistokes (cSt). The dimethicone fluid(s) are configured to provide the composition with a Time Weighted Force Area of between 5×104 and 12×104 and a Mean Break Time of between 0.30 and 0.75.1. A personal care composition, comprising: a) about 5% to about 30%, by weight of the composition, of a blend of at least two dimethicone fluids, wherein the blend of dimethicone fluids exhibits a viscosity of greater than about 1000 centistokes (cSt) according to the Rheology Method; and b) a dermatologically acceptable carrier, wherein the composition exhibits a Time Weighted Force Area of between about 5×104 and about 12×104 and a Mean Break Time of between about 0.30 and about 0.75 according to the Tack Method. 2. The personal care composition of claim 1, wherein the blend of dimethicone fluids has a viscosity of less than about 10,000 Stokes. 3. The personal care composition of claim 1, wherein a first dimethicone fluid in the blend has a viscosity of about 1000 cSt and a second dimethicone in the blend has a viscosity of about 60,000 cSt. 4. The personal care composition of claim 3, wherein the first and second dimethicone fluids are present at a ratio of between about 20:1 and about 1:1. 5. The personal care composition of claim 1, further comprising about 1% to about 30%, by weight, of a silicone elastomer. 6. The personal care composition of claim 5, further comprising a weight ratio of silicone elastomer to dimethicone fluid of about 1:10 to about 1:1. 7. The personal care composition of claim 1, further comprising an initial break time of greater than about 1.0. 8. The personal care composition of claim 1, further comprising a tack force at 60 minutes, 80 minutes, or 100 minutes of greater than about 1000. 9. The personal care composition of claim 1, further comprising about 0.01% to about 5%, by weight, of a superabsorbent polymer. 10. The personal care composition of claim 1, further comprising an aqueous phase, wherein the superabsorbent polymer is present in the aqueous phase. 11. The personal care composition of claim 10, wherein the superabsorbent polymer has a non-swollen, number average particle size of between about 2 microns and 100 microns. 12. The personal care composition of claim 1, further comprising a skin care active selected from the group consisting of vitamins, minerals, peptides, oil control agents, anti-oxidants, anti-inflammatory agents, moisturizing agents, emollients, humectants, exfoliating agents, skin lightening agents, sunscreens, anti-acne actives, anti-wrinkle actives, antimicrobials, and combinations of these. 13. The personal care composition of claim 12, wherein the skin care active is a vitamin comprising a stabilized retinoid. 14. The personal care composition of claim 13, wherein the retinoid is stabilized by a fatty acid ester present at a ratio of fatty acid ester to retinoid of about 5:1 to about 50:1. 15. The personal care composition of claim 11, wherein the fatty acid ester comprises capric/caprylic triglyceride. 16. A method of cosmetically treating a skin condition with a composition that provides an improved moisture perception, comprising: a) identifying a target portion of skin where skin moisturization is needed or desired; and b) applying the skin care composition of claim 1 to the target portion of skin. 17. A personal care composition, comprising: a) about 10% to about 20%, by weight, of a dimethicone fluid, wherein the dimethicone fluid has a viscosity between about 1000 cSt and about 1,000,000 cSt according to the Rheology Method, and exhibits a Time Weighted Force Area of between about 5×104 and about 12×104 and a Mean Break Time of between about 0.30 and about 0.75 according to the Tack Method; and b) a dermatologically acceptable carrier. 18. The personal care composition of claim 17, further comprising an initial break time of greater than about 1.0. 19. The personal care composition of claim 1, further comprising a tack force of greater than about 1000 at 60 minutes, 80 minutes, and/or 100 minutes.
1,600
556
13,643,673
1,647
A method for alleviating a symptom of chemotherapy in a subject comprises identifying a subject undergoing chemotherapy and then administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject. Typically, the levels of IGF-1 and/or GH in the subject are monitored as well as chemotherapy related symptoms. A method of alleviating oxidative damage, cellular damage including mutations, and insulin resistance in a subject is also provided.
1. A method of alleviating a symptom of chemotherapy in a subject, the method comprising: identifying a subject undergoing chemotherapy; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject; and measuring the level of IGF-1 in the subject. 2. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone receptor antagonist. 3. The method of claim 1 wherein GH/IGF-1 Axis inhibitory composition comprises a human growth hormone variant includes at least one amino acid substitution selected from the group consisting of H18D, H21N, R167N, K168A, D171S, K172R, E174S, I179T, and G120R. 4. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises an IGF-I receptor antagonist. 5. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises Pegvisomant™. 6. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises a GH-releasing hormone (GHRH) receptor antagonist. 7. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises an antibody selected from the group consisting of growth hormone antibodies, IGF-1 receptor antibodies, and combinations thereof. 8. A method of alleviating a symptom of oxidative damage in a subject, the method comprising: identifying a subject predisposed to oxidative damage; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject; and measuring the level of IGF-1 in the subject. 9. The method of claim 8 wherein the subject is undergoing chemotherapy. 10. The method of claim 8 wherein the subject is predisposed to or exhibits symptoms of diabetes. 11. The method of claim 8 wherein the subject is predisposed to or exhibits symptoms of stroke. 12. The method of claim 8 wherein the subject is predisposed to cancer. 13. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone receptor antagonist. 14. The method of claim 8 wherein the subject has an IGF-I level in the upper half of the normal age- and sex-specific levels of IGF-I compared to an average level for general population. 15. The method of claim 8 wherein GH/IGF-1 Axis inhibitory composition comprises a human growth hormone variant includes at least one amino acid substitution selected from the group consisting of H18D, H21N, R167N, K168A, D171S, K172R, E174S, I179T, and G120R. 16. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a IGF-I receptor antagonist. 17. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises Pegvisomant™. 18. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 19. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 20. A method of inhibiting the development of a symptom of cancer in a subject, the method comprising: identifying a subject predisposed to developing cancer; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject; and measuring the level of IGF-1 in the subject. 21. The method of claim 20 wherein the subject expresses one or more genetic markers indicative of a predisposition of cancer. 22. The method of claim 20 wherein the subject has a family history of cancer. 23. The method of claim 20 wherein the subject is a smoker. 24. The method of claim 20 wherein the subject is exposed to cancer promoting environments or chemicals. 25. The method of claim 20 wherein the GH/IFG-1 Axis inhibitory composition comprises a GF-I receptor antagonist. 26. The method of claim 20 wherein the GH/IGF-1 Axis inhibitory composition comprises Pegvisomant™. 27. The method of claim 20 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 28. The method of claim 20 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 29. A method of alleviating a symptom oxidative damage in eukaryotic cells, the method comprising: identifying eukaryotic cells predisposed to oxidative damage; and administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the eukaryotic cells. 30. A method comprising: identifying a subject that does not suffer from acromegaly of less than 70 years of age with IGF-I levels in the highest quartile of the population; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject so that IGF-I levels are reduced to the median level for that population; and measuring the level of IGF-1 in the subject.
A method for alleviating a symptom of chemotherapy in a subject comprises identifying a subject undergoing chemotherapy and then administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject. Typically, the levels of IGF-1 and/or GH in the subject are monitored as well as chemotherapy related symptoms. A method of alleviating oxidative damage, cellular damage including mutations, and insulin resistance in a subject is also provided.1. A method of alleviating a symptom of chemotherapy in a subject, the method comprising: identifying a subject undergoing chemotherapy; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject; and measuring the level of IGF-1 in the subject. 2. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone receptor antagonist. 3. The method of claim 1 wherein GH/IGF-1 Axis inhibitory composition comprises a human growth hormone variant includes at least one amino acid substitution selected from the group consisting of H18D, H21N, R167N, K168A, D171S, K172R, E174S, I179T, and G120R. 4. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises an IGF-I receptor antagonist. 5. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises Pegvisomant™. 6. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises a GH-releasing hormone (GHRH) receptor antagonist. 7. The method of claim 1 wherein the GH/IGF-1 Axis inhibitory composition comprises an antibody selected from the group consisting of growth hormone antibodies, IGF-1 receptor antibodies, and combinations thereof. 8. A method of alleviating a symptom of oxidative damage in a subject, the method comprising: identifying a subject predisposed to oxidative damage; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject; and measuring the level of IGF-1 in the subject. 9. The method of claim 8 wherein the subject is undergoing chemotherapy. 10. The method of claim 8 wherein the subject is predisposed to or exhibits symptoms of diabetes. 11. The method of claim 8 wherein the subject is predisposed to or exhibits symptoms of stroke. 12. The method of claim 8 wherein the subject is predisposed to cancer. 13. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone receptor antagonist. 14. The method of claim 8 wherein the subject has an IGF-I level in the upper half of the normal age- and sex-specific levels of IGF-I compared to an average level for general population. 15. The method of claim 8 wherein GH/IGF-1 Axis inhibitory composition comprises a human growth hormone variant includes at least one amino acid substitution selected from the group consisting of H18D, H21N, R167N, K168A, D171S, K172R, E174S, I179T, and G120R. 16. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a IGF-I receptor antagonist. 17. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises Pegvisomant™. 18. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 19. The method of claim 8 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 20. A method of inhibiting the development of a symptom of cancer in a subject, the method comprising: identifying a subject predisposed to developing cancer; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject; and measuring the level of IGF-1 in the subject. 21. The method of claim 20 wherein the subject expresses one or more genetic markers indicative of a predisposition of cancer. 22. The method of claim 20 wherein the subject has a family history of cancer. 23. The method of claim 20 wherein the subject is a smoker. 24. The method of claim 20 wherein the subject is exposed to cancer promoting environments or chemicals. 25. The method of claim 20 wherein the GH/IFG-1 Axis inhibitory composition comprises a GF-I receptor antagonist. 26. The method of claim 20 wherein the GH/IGF-1 Axis inhibitory composition comprises Pegvisomant™. 27. The method of claim 20 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 28. The method of claim 20 wherein the GH/IGF-1 Axis inhibitory composition comprises a growth hormone antibody. 29. A method of alleviating a symptom oxidative damage in eukaryotic cells, the method comprising: identifying eukaryotic cells predisposed to oxidative damage; and administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the eukaryotic cells. 30. A method comprising: identifying a subject that does not suffer from acromegaly of less than 70 years of age with IGF-I levels in the highest quartile of the population; administering a therapeutically effective amount of a GH/IGF-1 Axis inhibitory composition to the subject so that IGF-I levels are reduced to the median level for that population; and measuring the level of IGF-1 in the subject.
1,600
557
14,858,181
1,611
The sunscreen compositions of the present invention include a phase-stable, oil-in-water emulsion that includes a continuous water phase containing a superhydrophilic amphiphilic copolymer and a suspension of styrene/acrylate copolymer particles; and a discontinuous oil phase homogeneously dispersed in the continuous water phase, where the discontinuous oil phase includes a UV absorbing compound and the sunscreen composition is essentially free of monomeric surfactant and has a viscosity of about 2000 cps or less.
1. A sunscreen composition, comprising a phase-stable, oil-in-water emulsion, comprising, a continuous water phase comprising a superhydrophilic amphiphilic copolymer and a suspension of styrene/acrylate copolymer particles; and a discontinuous oil phase homogeneously dispersed in the continuous water phase, the discontinuous oil phase comprising a UV absorbing compound, wherein the sunscreen composition is essentially free of monomeric surfactant and has a viscosity of about 2000 cps or less. 2. The sunscreen composition according to claim 1, comprising from about 0.75% to about 6% by weight of the superhydrophilic amphiphilic copolymer. 3. The sunscreen composition according to claim 1, wherein the superhydrophilic amphiphilic copolymer has a mole percent of amphiphilic unit that is about 5% or more up to about 10% or less and a weight average molecular weight that is less than about 200,000. 4. The sunscreen composition according to claim lcomprising from about 1% to about 10% by weight of the styrene/acrylate copolymer particles. 5. The sunscreen composition according to claim 1 comprising from about 1% to about 5% by weight of the styrene/acrylate copolymer particles. 6. The sunscreen composition according to claim 3, wherein the styrene/acrylate copolymer particles are suspended in a mixture of water and glycol. 7. The sunscreen composition according to claim 1, wherein the UV absorbing compound is selected from the group consisting of a UV-A absorbing moiety and a UV-B absorbing moiety. 8. The composition of claim 6, wherein the UV-A absorbing moiety is selected from the group consisting of tertrahydroxybenzophenones; dicarboxydihydroxybenzophenones and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxydibenzoylmethanes and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxystilbenes and alkane ester or acid halide derivatives thereof; bis(hydroxystyrenyl) benzenes; bis(carboxystyrenyl)benzenes and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy, and hydroxycarboxycarotenes and alkane ester or acid halide derivatives thereof; 2 cyano-3,3-diphenyl acrylic acid, 2-ethyl hexyl ester; benzotriazole; and bis-ethylhexyloxyphenol methoxyphenyl triazine. 9. The composition of claim 6, wherein the UV-B absorbing moiety is selected from the group consisting of a 4-aminobenzoic acid and alkane esters thereof; anthranilic acid and alkane esters thereof; salicylic acid and alkane esters thereof; hydroxycinnamic acid and alkane esters thereof; dihydroxy-, dicarboxy-, and hydroxycarboxybenzophenones and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxychalcones and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxycoumarins and alkane ester or acid halide derivatives thereof; and bis-ethylhexyloxyphenol methoxyphenyl triazine. 10. The foaming composition of claim 1 wherein the UV absorbing compound is selected from the group consisting of octyl salicylate, homosalate, oxybenzone, octocrylene, avobenzone, and Bis-ethyhexylophenol methoxyphenyl triazine. 11. The sunscreen composition according to claim 1, comprising from about 10% to about 55% by weight of the oil phase. 12. The sunscreen composition according to claim 1, comprising from about 20% to about 40% by weight of the oil phase. 13. The sunscreen composition according to claim 1, comprising from about 10% to about 40% by weight of the UV absorbing compound. 14. The sunscreen composition comprising a polyol. 15. The sunscreen composition according to claim 14, wherein the polyol is glycerin. 16. The sunscreen composition according to claim 15 comprising from about 1% to about 20% by weight of the glycerin. 17. The sunscreen composition according to claim 16, comprising from about 5% to about 12% by weight of the glycerin. 18. The foaming sunscreen composition according to claim 1 having a viscosity of about 2000 cps or less. 19. The sunscreen composition according to claim 1, wherein the viscosity of the sunscreen composition is about 1000 cps or less. 20. The sunscreen composition according to claim 16 further comprising Polyglyceryl-10 Laurate and bis-PEG-18 methyl ether dimethyl silane. 21. The sunscreen composition according to claim 20, comprising about 5% by weight of the glycerin, about 2.5% by weight of the styrene/acrylate copolymer particles, about 1% by weight of the Polyglyceryl-10 Laurate and about 1% by weight of the bis-PEG-18 methyl ether dimethyl silane. 22. The sunscreen composition according to claim 11, comprising from about 90% to about 45% by weight of the water phase. 23. The sunscreen composition according to claim 12, comprising from about 80% to about 60% by weight of the water phase. 24. The foaming sunscreen composition according to claim 1, wherein the oil phase consists essentially of the UV absorbing compound. 25. The foaming sunscreen composition according to claim 1, wherein the oil phase comprises about 80% or more by weight of the UV absorbing compound. 26. The foaming sunscreen composition according to claim 1, wherein the oil phase comprises about 95% or more by weight of the UV absorbing compound.
The sunscreen compositions of the present invention include a phase-stable, oil-in-water emulsion that includes a continuous water phase containing a superhydrophilic amphiphilic copolymer and a suspension of styrene/acrylate copolymer particles; and a discontinuous oil phase homogeneously dispersed in the continuous water phase, where the discontinuous oil phase includes a UV absorbing compound and the sunscreen composition is essentially free of monomeric surfactant and has a viscosity of about 2000 cps or less.1. A sunscreen composition, comprising a phase-stable, oil-in-water emulsion, comprising, a continuous water phase comprising a superhydrophilic amphiphilic copolymer and a suspension of styrene/acrylate copolymer particles; and a discontinuous oil phase homogeneously dispersed in the continuous water phase, the discontinuous oil phase comprising a UV absorbing compound, wherein the sunscreen composition is essentially free of monomeric surfactant and has a viscosity of about 2000 cps or less. 2. The sunscreen composition according to claim 1, comprising from about 0.75% to about 6% by weight of the superhydrophilic amphiphilic copolymer. 3. The sunscreen composition according to claim 1, wherein the superhydrophilic amphiphilic copolymer has a mole percent of amphiphilic unit that is about 5% or more up to about 10% or less and a weight average molecular weight that is less than about 200,000. 4. The sunscreen composition according to claim lcomprising from about 1% to about 10% by weight of the styrene/acrylate copolymer particles. 5. The sunscreen composition according to claim 1 comprising from about 1% to about 5% by weight of the styrene/acrylate copolymer particles. 6. The sunscreen composition according to claim 3, wherein the styrene/acrylate copolymer particles are suspended in a mixture of water and glycol. 7. The sunscreen composition according to claim 1, wherein the UV absorbing compound is selected from the group consisting of a UV-A absorbing moiety and a UV-B absorbing moiety. 8. The composition of claim 6, wherein the UV-A absorbing moiety is selected from the group consisting of tertrahydroxybenzophenones; dicarboxydihydroxybenzophenones and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxydibenzoylmethanes and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxystilbenes and alkane ester or acid halide derivatives thereof; bis(hydroxystyrenyl) benzenes; bis(carboxystyrenyl)benzenes and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy, and hydroxycarboxycarotenes and alkane ester or acid halide derivatives thereof; 2 cyano-3,3-diphenyl acrylic acid, 2-ethyl hexyl ester; benzotriazole; and bis-ethylhexyloxyphenol methoxyphenyl triazine. 9. The composition of claim 6, wherein the UV-B absorbing moiety is selected from the group consisting of a 4-aminobenzoic acid and alkane esters thereof; anthranilic acid and alkane esters thereof; salicylic acid and alkane esters thereof; hydroxycinnamic acid and alkane esters thereof; dihydroxy-, dicarboxy-, and hydroxycarboxybenzophenones and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxychalcones and alkane ester or acid halide derivatives thereof; dihydroxy-, dicarboxy-, and hydroxycarboxycoumarins and alkane ester or acid halide derivatives thereof; and bis-ethylhexyloxyphenol methoxyphenyl triazine. 10. The foaming composition of claim 1 wherein the UV absorbing compound is selected from the group consisting of octyl salicylate, homosalate, oxybenzone, octocrylene, avobenzone, and Bis-ethyhexylophenol methoxyphenyl triazine. 11. The sunscreen composition according to claim 1, comprising from about 10% to about 55% by weight of the oil phase. 12. The sunscreen composition according to claim 1, comprising from about 20% to about 40% by weight of the oil phase. 13. The sunscreen composition according to claim 1, comprising from about 10% to about 40% by weight of the UV absorbing compound. 14. The sunscreen composition comprising a polyol. 15. The sunscreen composition according to claim 14, wherein the polyol is glycerin. 16. The sunscreen composition according to claim 15 comprising from about 1% to about 20% by weight of the glycerin. 17. The sunscreen composition according to claim 16, comprising from about 5% to about 12% by weight of the glycerin. 18. The foaming sunscreen composition according to claim 1 having a viscosity of about 2000 cps or less. 19. The sunscreen composition according to claim 1, wherein the viscosity of the sunscreen composition is about 1000 cps or less. 20. The sunscreen composition according to claim 16 further comprising Polyglyceryl-10 Laurate and bis-PEG-18 methyl ether dimethyl silane. 21. The sunscreen composition according to claim 20, comprising about 5% by weight of the glycerin, about 2.5% by weight of the styrene/acrylate copolymer particles, about 1% by weight of the Polyglyceryl-10 Laurate and about 1% by weight of the bis-PEG-18 methyl ether dimethyl silane. 22. The sunscreen composition according to claim 11, comprising from about 90% to about 45% by weight of the water phase. 23. The sunscreen composition according to claim 12, comprising from about 80% to about 60% by weight of the water phase. 24. The foaming sunscreen composition according to claim 1, wherein the oil phase consists essentially of the UV absorbing compound. 25. The foaming sunscreen composition according to claim 1, wherein the oil phase comprises about 80% or more by weight of the UV absorbing compound. 26. The foaming sunscreen composition according to claim 1, wherein the oil phase comprises about 95% or more by weight of the UV absorbing compound.
1,600
558
14,053,012
1,644
Methods of assaying potency of a vaccine composition are provided. Said methods utilize a T cell receptor mimic that is reactive against a specific peptide/MHC complex. The potency of the vaccine is determined based upon the measured density of specific peptide/MHC complex present on the surface of the vaccine-treated antigen presenting cell.
1-20. (canceled) 21. A method of assaying the potency of a vaccine composition, the method comprising the steps of: delivering a vaccine composition to at least one antigen presenting cell so that the antigen presenting cell displays a peptide/MHC complex having an epitope derived from the vaccine composition; contacting the at least one antigen presenting cell with a T cell receptor mimic (TCRm) antibody or fragment thereof reactive against the peptide/MHC complex; quantitatively measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof; and determining the potency of the vaccine composition based on the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof. 22. The method of claim 21, wherein the at least one antigen presenting cell is a dendritic cell. 23. The method of claim 21, wherein the vaccine composition comprises a protein vaccine. 24. The method of claim 21, wherein the vaccine composition comprises a peptide vaccine. 25. The method of claim 21, wherein the TCRm antibody or fragment thereof is reactive to HLA-A2 MHC complexes. 26. The method of claim 21, wherein the step of quantitatively measuring the number of peptide/MHC complexes comprises measuring less than 60 complexes per cell. 27. The method of claim 21, wherein the step of determining the potency of the vaccine composition further comprises determining the level of antigen specific T cell stimulation. 28. The method of claim 27, wherein the step of determining the level of antigen specific T cell stimulation comprises determining the level of cytotoxic T cell (CTL) stimulation. 29. The method of claim 21, wherein the TCRm antibody or fragment thereof has a binding affinity for the specific peptide/MHC complex of about 10 nanomolar or greater. 30. The method of claim 21, wherein the step of quantitatively measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises detecting a label linked to the TCRm antibody or fragment thereof. 31. The method of claim 21, wherein the step of quantitatively measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises flow cytometry. 32. A method of determining the potency of a vaccine composition, the method comprising the steps of: contacting antigen presenting cells with a vaccine composition; contacting the antigen presenting cells with a T cell receptor mimic (TCRm) antibody or fragment thereof, wherein the antigen presenting cells display a peptide/MHC complex having an epitope derived from the vaccine composition and wherein the TCRm antibody or fragment thereof specifically binds the peptide/MHC complex; measuring the number of peptide/MHC complexes bound by the T cell receptor mimic antibody; and determining the potency of the vaccine composition based on the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof. 33. The method of claim 32, wherein the antigen presenting cells are dendritic cells. 34. The method of claim 32, wherein the vaccine composition comprises a protein vaccine. 35. The method of claim 32, wherein the vaccine composition comprises a peptide vaccine. 36. The method of claim 32, wherein the TCRm antibody or fragment thereof is reactive to HLA-A2 MHC complexes. 37. The method of claim 32, wherein the step of measuring the number of peptide/MHC complexes comprises measuring less than 60 complexes per cell. 38. The method of claim 32, wherein the step of determining the potency of the vaccine composition further comprises determining the level of antigen specific T cell stimulation. 39. The method of claim 38, wherein the step of determining the level of antigen specific T cell stimulation comprises determining the level of cytotoxic T cell (CTL) stimulation. 40. The method of claim 32, wherein the TCRm antibody or fragment thereof has a binding affinity for the specific peptide/MHC complex of about 10 nanomolar or greater. 41. The method of claim 32, wherein the step of measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises detecting a label linked to the TCRm antibody or fragment thereof. 42. The method of claim 32, wherein the step of measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises flow cytometry.
Methods of assaying potency of a vaccine composition are provided. Said methods utilize a T cell receptor mimic that is reactive against a specific peptide/MHC complex. The potency of the vaccine is determined based upon the measured density of specific peptide/MHC complex present on the surface of the vaccine-treated antigen presenting cell.1-20. (canceled) 21. A method of assaying the potency of a vaccine composition, the method comprising the steps of: delivering a vaccine composition to at least one antigen presenting cell so that the antigen presenting cell displays a peptide/MHC complex having an epitope derived from the vaccine composition; contacting the at least one antigen presenting cell with a T cell receptor mimic (TCRm) antibody or fragment thereof reactive against the peptide/MHC complex; quantitatively measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof; and determining the potency of the vaccine composition based on the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof. 22. The method of claim 21, wherein the at least one antigen presenting cell is a dendritic cell. 23. The method of claim 21, wherein the vaccine composition comprises a protein vaccine. 24. The method of claim 21, wherein the vaccine composition comprises a peptide vaccine. 25. The method of claim 21, wherein the TCRm antibody or fragment thereof is reactive to HLA-A2 MHC complexes. 26. The method of claim 21, wherein the step of quantitatively measuring the number of peptide/MHC complexes comprises measuring less than 60 complexes per cell. 27. The method of claim 21, wherein the step of determining the potency of the vaccine composition further comprises determining the level of antigen specific T cell stimulation. 28. The method of claim 27, wherein the step of determining the level of antigen specific T cell stimulation comprises determining the level of cytotoxic T cell (CTL) stimulation. 29. The method of claim 21, wherein the TCRm antibody or fragment thereof has a binding affinity for the specific peptide/MHC complex of about 10 nanomolar or greater. 30. The method of claim 21, wherein the step of quantitatively measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises detecting a label linked to the TCRm antibody or fragment thereof. 31. The method of claim 21, wherein the step of quantitatively measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises flow cytometry. 32. A method of determining the potency of a vaccine composition, the method comprising the steps of: contacting antigen presenting cells with a vaccine composition; contacting the antigen presenting cells with a T cell receptor mimic (TCRm) antibody or fragment thereof, wherein the antigen presenting cells display a peptide/MHC complex having an epitope derived from the vaccine composition and wherein the TCRm antibody or fragment thereof specifically binds the peptide/MHC complex; measuring the number of peptide/MHC complexes bound by the T cell receptor mimic antibody; and determining the potency of the vaccine composition based on the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof. 33. The method of claim 32, wherein the antigen presenting cells are dendritic cells. 34. The method of claim 32, wherein the vaccine composition comprises a protein vaccine. 35. The method of claim 32, wherein the vaccine composition comprises a peptide vaccine. 36. The method of claim 32, wherein the TCRm antibody or fragment thereof is reactive to HLA-A2 MHC complexes. 37. The method of claim 32, wherein the step of measuring the number of peptide/MHC complexes comprises measuring less than 60 complexes per cell. 38. The method of claim 32, wherein the step of determining the potency of the vaccine composition further comprises determining the level of antigen specific T cell stimulation. 39. The method of claim 38, wherein the step of determining the level of antigen specific T cell stimulation comprises determining the level of cytotoxic T cell (CTL) stimulation. 40. The method of claim 32, wherein the TCRm antibody or fragment thereof has a binding affinity for the specific peptide/MHC complex of about 10 nanomolar or greater. 41. The method of claim 32, wherein the step of measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises detecting a label linked to the TCRm antibody or fragment thereof. 42. The method of claim 32, wherein the step of measuring the number of peptide/MHC complexes bound by the TCRm antibody or fragment thereof comprises flow cytometry.
1,600
559
14,281,532
1,617
Foamable sanitizing compositions are disclosed. The compositions contain alcohol, water, a foaming agent, and a foam strengthening agent. The foaming agent which may comprise a derivatized dimethicone has been found to cause the alcohol solution to foam even though alcohol has various defoaming properties. The foam strengthening agent may comprise, for instance, a betaine that contains organic molecules wherein at least 90% of the organic molecules have a carbon chain length of 18 carbon atoms or greater. In one embodiment, the composition can be contained in a non-aerosol dispensing container that mixes the composition with air causing the composition to foam when dispensed.
1-23. (canceled) 24. A foamable composition comprising: (a) a linear dimethicone copolyol ester, (b) at least one C1-4 alcohol in an amount of from about 30% to about 90% by weight, (c) water in an amount of at least 10% by weight, and (d) betaine. 25. The foamable composition as defined in claim 24, wherein the dimethicone copolyol ester is present in an amount of from about 0.5% to about 15 by weight. 26. The foamable composition as defined in claim 24, wherein the dimethicone copolyol ester contains from about 2 moles to about 20 moles of polyoxyethylene glycol. 27. The foamable composition as defined in claim 24, wherein the alcohol is present in the composition in an amount of from about 40% to about 70% by weight. 28. The foamable composition as defined in claim 24, wherein the composition comprises at least two C1-4 alcohols. 29. The foamable composition as defined in claim 24, wherein the C1-4 alcohol comprises ethanol, 2-propanol, or n-propanol. 30. The foamable composition as defined in claim 24, wherein the water is present in the composition in an amount of from about 20% to about 70% by weight. 31. The foamable composition as defined in claim 24, wherein the composition further comprises from about 0.1% to about 5% by weight of a foam strengthening agent comprising an amphoteric or zwitterionic surfactant comprising surfactant molecules. 32. The foamable composition as defined in claim 24, wherein the composition further comprises a glycerin. 33. A foaming sanitizer product comprising: a dispensing container, the dispensing container including a non-aerosol pumping device, a foamable sanitizing composition contained within the dispensing container, the foamable sanitizing composition comprising (a) a linear dimethicone copolyol ester in an amount of from about 0.5% to about 15% by weight, (b) at least one C1-4 alcohol in an amount of from about 30% to about 90% by weight, (c) water in an amount of at east 10% by weight, and (d) betaine, wherein the pumping device of the dispensing container is configured to combine the sanitizing composition with air when dispensed from the container for producing a foam. 34. The foaming sanitizer product as defined in claim 33, wherein the linear dimethicone copolyol ester is present in an amount of from about 0.5% to about 15% by weight. 35. The foaming sanitizer product as defined in claim 33, wherein the dimethicone copolyol ester contains from about 2 moles to about 20 moles of polyoxyethylene glycol. 36. The foaming sanitizer product as defined in claim 33, wherein the alcohol is present in the composition in an amount of from about 40% to about 70% by weight. 37. The foaming sanitizer product as defined in claim 33, wherein the composition comprises at least two C1-4 alcohols. 38. The foaming sanitizer product as defined in claim 33, wherein the C1-4 alcohol comprises ethanol, 2-propanol, or n-propanol. 39. The foaming sanitizer product as defined in claim 33, wherein the water is present in the composition in an amount of from about 20% to about 70% by weight. 40. The foaming sanitizer product as defined in claim 33, wherein the composition further comprises from about 0.1% to about 5% by weight of a foam strengthening agent comprising an amphoteric or zwitterionic surfactant comprising surfactant molecules. 41. The foaming sanitizer product as defined in claim 33, wherein the composition further comprises a glycerin. 42. The foaming sanitizer product as defined in claim 33, wherein the pumping device comprises an air pump and a liquid pump in communication with an actuating head, and wherein displacing the actuating head manually causes the liquid pump to pump sanitizing composition and the air pump to pump air for mixing with the sanitizing composition and forming a foam that is dispensed from the container.
Foamable sanitizing compositions are disclosed. The compositions contain alcohol, water, a foaming agent, and a foam strengthening agent. The foaming agent which may comprise a derivatized dimethicone has been found to cause the alcohol solution to foam even though alcohol has various defoaming properties. The foam strengthening agent may comprise, for instance, a betaine that contains organic molecules wherein at least 90% of the organic molecules have a carbon chain length of 18 carbon atoms or greater. In one embodiment, the composition can be contained in a non-aerosol dispensing container that mixes the composition with air causing the composition to foam when dispensed.1-23. (canceled) 24. A foamable composition comprising: (a) a linear dimethicone copolyol ester, (b) at least one C1-4 alcohol in an amount of from about 30% to about 90% by weight, (c) water in an amount of at least 10% by weight, and (d) betaine. 25. The foamable composition as defined in claim 24, wherein the dimethicone copolyol ester is present in an amount of from about 0.5% to about 15 by weight. 26. The foamable composition as defined in claim 24, wherein the dimethicone copolyol ester contains from about 2 moles to about 20 moles of polyoxyethylene glycol. 27. The foamable composition as defined in claim 24, wherein the alcohol is present in the composition in an amount of from about 40% to about 70% by weight. 28. The foamable composition as defined in claim 24, wherein the composition comprises at least two C1-4 alcohols. 29. The foamable composition as defined in claim 24, wherein the C1-4 alcohol comprises ethanol, 2-propanol, or n-propanol. 30. The foamable composition as defined in claim 24, wherein the water is present in the composition in an amount of from about 20% to about 70% by weight. 31. The foamable composition as defined in claim 24, wherein the composition further comprises from about 0.1% to about 5% by weight of a foam strengthening agent comprising an amphoteric or zwitterionic surfactant comprising surfactant molecules. 32. The foamable composition as defined in claim 24, wherein the composition further comprises a glycerin. 33. A foaming sanitizer product comprising: a dispensing container, the dispensing container including a non-aerosol pumping device, a foamable sanitizing composition contained within the dispensing container, the foamable sanitizing composition comprising (a) a linear dimethicone copolyol ester in an amount of from about 0.5% to about 15% by weight, (b) at least one C1-4 alcohol in an amount of from about 30% to about 90% by weight, (c) water in an amount of at east 10% by weight, and (d) betaine, wherein the pumping device of the dispensing container is configured to combine the sanitizing composition with air when dispensed from the container for producing a foam. 34. The foaming sanitizer product as defined in claim 33, wherein the linear dimethicone copolyol ester is present in an amount of from about 0.5% to about 15% by weight. 35. The foaming sanitizer product as defined in claim 33, wherein the dimethicone copolyol ester contains from about 2 moles to about 20 moles of polyoxyethylene glycol. 36. The foaming sanitizer product as defined in claim 33, wherein the alcohol is present in the composition in an amount of from about 40% to about 70% by weight. 37. The foaming sanitizer product as defined in claim 33, wherein the composition comprises at least two C1-4 alcohols. 38. The foaming sanitizer product as defined in claim 33, wherein the C1-4 alcohol comprises ethanol, 2-propanol, or n-propanol. 39. The foaming sanitizer product as defined in claim 33, wherein the water is present in the composition in an amount of from about 20% to about 70% by weight. 40. The foaming sanitizer product as defined in claim 33, wherein the composition further comprises from about 0.1% to about 5% by weight of a foam strengthening agent comprising an amphoteric or zwitterionic surfactant comprising surfactant molecules. 41. The foaming sanitizer product as defined in claim 33, wherein the composition further comprises a glycerin. 42. The foaming sanitizer product as defined in claim 33, wherein the pumping device comprises an air pump and a liquid pump in communication with an actuating head, and wherein displacing the actuating head manually causes the liquid pump to pump sanitizing composition and the air pump to pump air for mixing with the sanitizing composition and forming a foam that is dispensed from the container.
1,600
560
15,513,132
1,646
The present invention relates to a therapeutic agent for pustular psoriasis or psoriatic erythroderma that is administered to a psoriasis patient that has been administered with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient; and to a therapeutic agent for psoriasis that is administered to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient. In addition, the present invention also relates to a method for the treatment of pustular psoriasis or psoriatic erythroderma, comprising administering an IL-17RA antagonist to a psoriasis patient that has been administered with an anti-TNF-alpha antibody; and to a method for the treatment of psoriasis, comprising administering an IL-17RA antagonist to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody.
1. A therapeutic agent for pustular psoriasis or psoriatic erythroderma that is administered to a psoriasis patient that has been administered with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient. 2. The therapeutic agent according to claim 1, wherein the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 3. The therapeutic agent according to claim 1 or 2, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 4. The therapeutic agent according to claim 3, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which a complementarity determining region (hereinafter, referred to as CDR) 1, CDR2, and CDR3 of a heavy chain variable region (hereinafter, referred to as VH) of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of a light chain variable region (hereinafter, referred to as VL) of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 5. The therapeutic agent according to any one of claims 1 to 4, wherein the clinical global impression (hereinafter, also referred to as CGI) of a patient after administration of the therapeutic agent becomes 2 or 1. 6. The therapeutic agent according to any one of claims 1 to 5, wherein the psoriasis area and severity index (hereinafter, referred to as PASI) score of a patient after administration of the therapeutic agent is lower than that before administration of the therapeutic agent. 7. A therapeutic agent for psoriasis that is administered to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient. 8. The therapeutic agent according to claim 7, wherein the patient that cannot be treated with the anti-TNF-alpha antibody is a patient that does not respond to the anti-TNF-alpha antibody or that is of insufficient tolerability to the anti-TNF-alpha antibody. 9. The therapeutic agent according to claim 7 or 8, the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 10. The therapeutic agent according to any one of claims 7 to 9, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 11. The therapeutic agent according to claim 10, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which CDR1, CDR2, and CDR3 of VH of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of VL of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 12. The therapeutic agent according to any one of claims 7 to 11, wherein the psoriasis is at least one selected from psoriasis vulgaris, psoriasis arthropica, pustular psoriasis, psoriatic erythroderma, and psoriasis guttata. 13. The therapeutic agent according to any one of claims 7 to 12, wherein the CGI of a patient after administration of the therapeutic agent becomes 2 or 1. 14. The therapeutic agent according to any one of claims 7 to 13, wherein the PASI score of a patient after administration of the therapeutic agent is lower than that before administration of the therapeutic agent. 15. A method for the treatment of pustular psoriasis or psoriatic erythroderma, comprising administering an IL-17RA antagonist to a psoriasis patient that has been administered with an anti-TNF-alpha antibody. 16. The method according to claim 15, wherein the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 17. The method according to claim 15 or 16, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 18. The method according to claim 17, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which CDR1, CDR2, and CDR3 of VH of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of VL of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 19. The method according to any one of claims 15 to 18, wherein the CGI of the patient after the administration of the IL-17RA antagonist becomes 2 or 1. 20. The method according to any one of claims 15 to 19, wherein the PASI score of a patient after the administration of the IL-17RA antagonist is lower than that before the administration of the antagonist. 21. The method according to any one of claims 15 to 20, wherein the IL-17RA antagonist is administered in a dose of 140 mg or more. 22. The method according to any one of claims 15 to 21, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg. 23. The method according to any one of claims 15 to 22, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg on the 1st day, the 1st week and the 2nd week, and thereafter once every two weeks. 24. A method for the treatment of psoriasis, comprising administering an IL-17RA antagonist to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody. 25. The method according to claim 24, wherein the patient that cannot be treated with the anti-TNF-alpha antibody is a patient that is ineffective to the anti-TNF-alpha antibody or that is of insufficient tolerability to the anti-TNF-alpha antibody. 26. The method according to claim 24 or 25, wherein the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 27. The method according to any one of claims 24 to 26, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 28. The method according to claim 27, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which CDR1, CDR2, and CDR3 of VH of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of VL of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 29. The method according to any one of claims 24 to 28, wherein the psoriasis is at least one selected from psoriasis vulgaris, psoriasis arthropica, pustular psoriasis, psoriatic erythroderma, and psoriasis guttata. 30. The method according to any one of claims 24 to 29, wherein the CGI of a patient after the administration of the IL-17RA antagonist becomes 2 or 1. 31. The method according to any one of claims 24 to 30, wherein the PASI score of a patient after the administration of the IL-17RA antagonist is lower than that before the administration of the antagonist. 32. The method according to any one of claims 24 to 31, wherein the IL-17RA antagonist is administered in a dose of 140 mg or more. 33. The method according to any one of claims 24 to 32, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg. 34. The method according to any one of claims 24 to 33, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg on the 1st day, the 1st week and the 2nd week, and thereafter once every two weeks.
The present invention relates to a therapeutic agent for pustular psoriasis or psoriatic erythroderma that is administered to a psoriasis patient that has been administered with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient; and to a therapeutic agent for psoriasis that is administered to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient. In addition, the present invention also relates to a method for the treatment of pustular psoriasis or psoriatic erythroderma, comprising administering an IL-17RA antagonist to a psoriasis patient that has been administered with an anti-TNF-alpha antibody; and to a method for the treatment of psoriasis, comprising administering an IL-17RA antagonist to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody.1. A therapeutic agent for pustular psoriasis or psoriatic erythroderma that is administered to a psoriasis patient that has been administered with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient. 2. The therapeutic agent according to claim 1, wherein the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 3. The therapeutic agent according to claim 1 or 2, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 4. The therapeutic agent according to claim 3, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which a complementarity determining region (hereinafter, referred to as CDR) 1, CDR2, and CDR3 of a heavy chain variable region (hereinafter, referred to as VH) of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of a light chain variable region (hereinafter, referred to as VL) of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 5. The therapeutic agent according to any one of claims 1 to 4, wherein the clinical global impression (hereinafter, also referred to as CGI) of a patient after administration of the therapeutic agent becomes 2 or 1. 6. The therapeutic agent according to any one of claims 1 to 5, wherein the psoriasis area and severity index (hereinafter, referred to as PASI) score of a patient after administration of the therapeutic agent is lower than that before administration of the therapeutic agent. 7. A therapeutic agent for psoriasis that is administered to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody, comprising an IL-17RA antagonist as an active ingredient. 8. The therapeutic agent according to claim 7, wherein the patient that cannot be treated with the anti-TNF-alpha antibody is a patient that does not respond to the anti-TNF-alpha antibody or that is of insufficient tolerability to the anti-TNF-alpha antibody. 9. The therapeutic agent according to claim 7 or 8, the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 10. The therapeutic agent according to any one of claims 7 to 9, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 11. The therapeutic agent according to claim 10, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which CDR1, CDR2, and CDR3 of VH of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of VL of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 12. The therapeutic agent according to any one of claims 7 to 11, wherein the psoriasis is at least one selected from psoriasis vulgaris, psoriasis arthropica, pustular psoriasis, psoriatic erythroderma, and psoriasis guttata. 13. The therapeutic agent according to any one of claims 7 to 12, wherein the CGI of a patient after administration of the therapeutic agent becomes 2 or 1. 14. The therapeutic agent according to any one of claims 7 to 13, wherein the PASI score of a patient after administration of the therapeutic agent is lower than that before administration of the therapeutic agent. 15. A method for the treatment of pustular psoriasis or psoriatic erythroderma, comprising administering an IL-17RA antagonist to a psoriasis patient that has been administered with an anti-TNF-alpha antibody. 16. The method according to claim 15, wherein the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 17. The method according to claim 15 or 16, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 18. The method according to claim 17, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which CDR1, CDR2, and CDR3 of VH of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of VL of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 19. The method according to any one of claims 15 to 18, wherein the CGI of the patient after the administration of the IL-17RA antagonist becomes 2 or 1. 20. The method according to any one of claims 15 to 19, wherein the PASI score of a patient after the administration of the IL-17RA antagonist is lower than that before the administration of the antagonist. 21. The method according to any one of claims 15 to 20, wherein the IL-17RA antagonist is administered in a dose of 140 mg or more. 22. The method according to any one of claims 15 to 21, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg. 23. The method according to any one of claims 15 to 22, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg on the 1st day, the 1st week and the 2nd week, and thereafter once every two weeks. 24. A method for the treatment of psoriasis, comprising administering an IL-17RA antagonist to a psoriasis patient that cannot be treated with an anti-TNF-alpha antibody. 25. The method according to claim 24, wherein the patient that cannot be treated with the anti-TNF-alpha antibody is a patient that is ineffective to the anti-TNF-alpha antibody or that is of insufficient tolerability to the anti-TNF-alpha antibody. 26. The method according to claim 24 or 25, wherein the anti-TNF-alpha antibody is at least one selected from adalimumab, infliximab, certolizumab pegol, certolizumab, and golimumab. 27. The method according to any one of claims 24 to 26, wherein the IL-17RA antagonist is an anti-IL-17RA antibody or an antibody fragment thereof. 28. The method according to claim 27, wherein the anti-IL-17RA antibody is selected from the following a) and b): a) a monoclonal antibody in which CDR1, CDR2, and CDR3 of VH of the antibody comprise the amino acid sequences shown in SEQ ID NOs:1, 2, and 3, respectively, and CDR1, CDR2, and CDR3 of VL of the antibody comprise the amino acid sequences shown in SEQ ID NOs:4, 5, and 6, respectively; and b) a monoclonal antibody in which VH of the antibody comprises the amino acid sequence shown in SEQ ID NO:7, and VL of the antibody comprises the amino acid sequence shown in SEQ ID NO:8. 29. The method according to any one of claims 24 to 28, wherein the psoriasis is at least one selected from psoriasis vulgaris, psoriasis arthropica, pustular psoriasis, psoriatic erythroderma, and psoriasis guttata. 30. The method according to any one of claims 24 to 29, wherein the CGI of a patient after the administration of the IL-17RA antagonist becomes 2 or 1. 31. The method according to any one of claims 24 to 30, wherein the PASI score of a patient after the administration of the IL-17RA antagonist is lower than that before the administration of the antagonist. 32. The method according to any one of claims 24 to 31, wherein the IL-17RA antagonist is administered in a dose of 140 mg or more. 33. The method according to any one of claims 24 to 32, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg. 34. The method according to any one of claims 24 to 33, wherein the IL-17RA antagonist is administered in a dose of 140 mg or 210 mg on the 1st day, the 1st week and the 2nd week, and thereafter once every two weeks.
1,600
561
14,174,317
1,617
Disclosed is a method of preparing a hair care composition comprising a step of controlling temperature when adding antidandruff agents. The method of the present invention provides reduced agglomeration of antidandruff agents selected from metal pyrithiones.
1. A method of preparing a antidandruff compositions, wherein the composition comprises: (a) a cationic surfactant system; (b) a high melting point fatty compound; (c) aqueous carrier; (d) an antidandruff agent selected from metal pyrithiones; and wherein the method comprises the steps of: (1) mixing the cationic surfactant system, high melting point fatty compound and aqueous carrier to form an emulsion; (2) adding the antidandruff agent to the emulsion having a temperature of from about 40° C. to about 66° C. 2. The method of claim 1, wherein the composition further contains a perfume, silicone, phenoxyethanol, benzyl alcohol, and/or parabens. 3. The method of claim 1, wherein the composition further contains perfumes and/or silicone compounds. 4. The method of claim 3, wherein the perfume has a Dielectric Constant (DC) of 15 or lower. 5. The method of claim 3, wherein the silicone compound has a hydrophilic group, 6. The method of claim 5, wherein the silicone compound is selected from the group consisting of aminosilicone, silicone copolyol, aminosilicone copolyol, quaternized silicone, quaternized silicone copolyol, and mixtures thereof. 7. The method of claim 1, wherein the cationic surfactant system comprises a mono-long alkyl quaternized ammonium salt and a di-long alkyl quaternized ammonium salt. 8. The method of claim 1, wherein the total level of the cationic surfactant system and high melting point fatty compound is 10.0% or less. 9. The method of claim 1, wherein the temperature is from about 45° C. to about 60° C., 10. The method of claim 1, wherein the temperature is from about 48° C. to about 55° C. 11. The method of claim 1, wherein the step (1) further comprising the steps of: (1-A1) mixing the cationic surfactant system, high melting point fatty compound and aqueous carrier wherein the temperature of the mixture is above the melting point of the high melting point fatty compounds; and (1-A2) the mixture is cooled down to a temperature gradually at a rate of from about 1° C. to 10° C./minute, to form an emulsion. 12. A composition made by the method of claim 1.
Disclosed is a method of preparing a hair care composition comprising a step of controlling temperature when adding antidandruff agents. The method of the present invention provides reduced agglomeration of antidandruff agents selected from metal pyrithiones.1. A method of preparing a antidandruff compositions, wherein the composition comprises: (a) a cationic surfactant system; (b) a high melting point fatty compound; (c) aqueous carrier; (d) an antidandruff agent selected from metal pyrithiones; and wherein the method comprises the steps of: (1) mixing the cationic surfactant system, high melting point fatty compound and aqueous carrier to form an emulsion; (2) adding the antidandruff agent to the emulsion having a temperature of from about 40° C. to about 66° C. 2. The method of claim 1, wherein the composition further contains a perfume, silicone, phenoxyethanol, benzyl alcohol, and/or parabens. 3. The method of claim 1, wherein the composition further contains perfumes and/or silicone compounds. 4. The method of claim 3, wherein the perfume has a Dielectric Constant (DC) of 15 or lower. 5. The method of claim 3, wherein the silicone compound has a hydrophilic group, 6. The method of claim 5, wherein the silicone compound is selected from the group consisting of aminosilicone, silicone copolyol, aminosilicone copolyol, quaternized silicone, quaternized silicone copolyol, and mixtures thereof. 7. The method of claim 1, wherein the cationic surfactant system comprises a mono-long alkyl quaternized ammonium salt and a di-long alkyl quaternized ammonium salt. 8. The method of claim 1, wherein the total level of the cationic surfactant system and high melting point fatty compound is 10.0% or less. 9. The method of claim 1, wherein the temperature is from about 45° C. to about 60° C., 10. The method of claim 1, wherein the temperature is from about 48° C. to about 55° C. 11. The method of claim 1, wherein the step (1) further comprising the steps of: (1-A1) mixing the cationic surfactant system, high melting point fatty compound and aqueous carrier wherein the temperature of the mixture is above the melting point of the high melting point fatty compounds; and (1-A2) the mixture is cooled down to a temperature gradually at a rate of from about 1° C. to 10° C./minute, to form an emulsion. 12. A composition made by the method of claim 1.
1,600
562
15,618,792
1,663
The present disclosure describes genetically-modified plants having enhanced tolerance to multiple abiotic stressors, such as extreme temperatures (heat or cold) and/or drought. Abiotic stress tolerance is enhanced by ectopic expression of a heterologous glutaredoxin. Abiotic stress tolerance (particularly drought) is also enhanced by inhibited function, activity, or expression of an endogenous glutaredoxin. Methods of producing such genetically-modified plants are also disclosed.
1. A method of producing a genetically-modified plant having enhanced tolerance to an abiotic stressor as compared to a control plant, said method comprising: transforming a plant with a heterologous, abiotic stress tolerance gene, wherein said gene encodes a glutaredoxin, thereby enhancing the tolerance of said genetically-modified plant to an abiotic stressor. 2. The method of claim 1, wherein said stress tolerance gene is Arabidopsis monothiol glutaredoxin AtGRXS17. 3. The method of claim 1, wherein said glutaredoxin is GRXS17. 4. The method of claim 1, wherein said abiotic stressor is heat stress. 5. The method of claim 1, wherein said transforming comprising: introducing into said plant a nucleic acid construct for said heterologous, abiotic stress tolerance gene encoding said glutaredoxin; and allowing said nucleic acid construct to be ectopically expressed in said plant. 6. The method of claim 5, wherein said nucleic acid construct comprises a nucleic acid coding sequence operably linked to a promoter that drives expression in said plant. 7. The method of claim 5, wherein the nucleic acid construct comprises a sequence selected from the group consisting of: (a) a nucleotide sequence comprising SEQ ID NO:1, 3, 5, or 30; (b) a nucleotide sequence comprising an antisense sequence corresponding to SEQ ID NO: 1, 3, 5, or 30; (c) a nucleotide sequence having at least about 70% sequence identity to SEQ ID NO: 1, 3, 5, or 30; (d) a nucleotide sequence encoding a glutaredoxin protein comprising SEQ ID NO:2, 4, 6, or 31; and (e) a nucleotide sequence encoding a glutaredoxin protein having at least about 50% amino acid identity to SEQ ID NO: 2, 4, 6, or 31 and retaining the functional characteristics thereof. 8. The method of claim 1, wherein said plant comprises a plant tissue, and said transforming comprising: culturing said tissue on media; and introducing said heterologous, abiotic stress tolerance gene into the cells of said tissue to yield transformed tissue. 9. The method of claim 8, further comprising: regenerating whole plants from said tissue, wherein said regenerated plants have enhanced tolerance of an abiotic stressor. 10. The method of claim 9, wherein said regenerating comprises: inducing callus formation from said transformed tissue; regenerating shoots; and rooting of said shoots in rooting media to regenerate said whole plant. 11. The method of claim 8, wherein said introducing is selected from the group consisting of: Agrobacterium-mediated transformation, PEG-mediated uptake, electroporation-mediated uptake, particle bombardment-mediated delivery, and microinjection. 12. The method of claim 1, wherein said genetically-modified plant is selected from the group consisting of wheat, oat, barley, rice, maize, millet, rye, sorghum, triticale, buckwheat, quinoa, soybeans, beans, peas, alfalfa, potatoes, sweet potatoes, cassava, yam, tomatoes, peppers, tobacco, and cotton. 13. The method of claim 1, wherein said genetically-modified plant comprises an identical phenotype as compared to said control plant when said plants are grown under non-stress conditions. 14. The method of claim 1, said method further comprising inhibiting the expression, activity, or function of an endogenous glutaredoxin gene of said genetically-modified plant. 15. A method of producing genetically-modified plants having enhanced tolerance to an abiotic stressor, said method comprising: crossing a first plant with a second plant to thereby produce progeny, wherein at least one of said first or second plants is a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene, wherein said gene encodes a glutaredoxin, said progeny having enhanced tolerance to an abiotic stressor. 16. The method of claim 15, wherein said progeny is homozygous for said abiotic stress tolerance gene. 17. The method of claim 15, wherein each of said first and second plants is a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene. 18. The method of claim 15, wherein said first plant is a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene, and said second plant has a characteristic selected from the group consisting of: pest resistance, herbicide resistance, geographic adaptation, and increased stalk strength. 19. A genetically-modified seed produced according to the method of claim 15. 20. A method of producing genetically-modified seed, said method comprising: providing a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene, wherein said gene encodes a glutaredoxin; and self-pollinating said plant to thereby produce genetically-modified seed, wherein said seed comprises said heterologous, abiotic stress tolerance gene.
The present disclosure describes genetically-modified plants having enhanced tolerance to multiple abiotic stressors, such as extreme temperatures (heat or cold) and/or drought. Abiotic stress tolerance is enhanced by ectopic expression of a heterologous glutaredoxin. Abiotic stress tolerance (particularly drought) is also enhanced by inhibited function, activity, or expression of an endogenous glutaredoxin. Methods of producing such genetically-modified plants are also disclosed.1. A method of producing a genetically-modified plant having enhanced tolerance to an abiotic stressor as compared to a control plant, said method comprising: transforming a plant with a heterologous, abiotic stress tolerance gene, wherein said gene encodes a glutaredoxin, thereby enhancing the tolerance of said genetically-modified plant to an abiotic stressor. 2. The method of claim 1, wherein said stress tolerance gene is Arabidopsis monothiol glutaredoxin AtGRXS17. 3. The method of claim 1, wherein said glutaredoxin is GRXS17. 4. The method of claim 1, wherein said abiotic stressor is heat stress. 5. The method of claim 1, wherein said transforming comprising: introducing into said plant a nucleic acid construct for said heterologous, abiotic stress tolerance gene encoding said glutaredoxin; and allowing said nucleic acid construct to be ectopically expressed in said plant. 6. The method of claim 5, wherein said nucleic acid construct comprises a nucleic acid coding sequence operably linked to a promoter that drives expression in said plant. 7. The method of claim 5, wherein the nucleic acid construct comprises a sequence selected from the group consisting of: (a) a nucleotide sequence comprising SEQ ID NO:1, 3, 5, or 30; (b) a nucleotide sequence comprising an antisense sequence corresponding to SEQ ID NO: 1, 3, 5, or 30; (c) a nucleotide sequence having at least about 70% sequence identity to SEQ ID NO: 1, 3, 5, or 30; (d) a nucleotide sequence encoding a glutaredoxin protein comprising SEQ ID NO:2, 4, 6, or 31; and (e) a nucleotide sequence encoding a glutaredoxin protein having at least about 50% amino acid identity to SEQ ID NO: 2, 4, 6, or 31 and retaining the functional characteristics thereof. 8. The method of claim 1, wherein said plant comprises a plant tissue, and said transforming comprising: culturing said tissue on media; and introducing said heterologous, abiotic stress tolerance gene into the cells of said tissue to yield transformed tissue. 9. The method of claim 8, further comprising: regenerating whole plants from said tissue, wherein said regenerated plants have enhanced tolerance of an abiotic stressor. 10. The method of claim 9, wherein said regenerating comprises: inducing callus formation from said transformed tissue; regenerating shoots; and rooting of said shoots in rooting media to regenerate said whole plant. 11. The method of claim 8, wherein said introducing is selected from the group consisting of: Agrobacterium-mediated transformation, PEG-mediated uptake, electroporation-mediated uptake, particle bombardment-mediated delivery, and microinjection. 12. The method of claim 1, wherein said genetically-modified plant is selected from the group consisting of wheat, oat, barley, rice, maize, millet, rye, sorghum, triticale, buckwheat, quinoa, soybeans, beans, peas, alfalfa, potatoes, sweet potatoes, cassava, yam, tomatoes, peppers, tobacco, and cotton. 13. The method of claim 1, wherein said genetically-modified plant comprises an identical phenotype as compared to said control plant when said plants are grown under non-stress conditions. 14. The method of claim 1, said method further comprising inhibiting the expression, activity, or function of an endogenous glutaredoxin gene of said genetically-modified plant. 15. A method of producing genetically-modified plants having enhanced tolerance to an abiotic stressor, said method comprising: crossing a first plant with a second plant to thereby produce progeny, wherein at least one of said first or second plants is a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene, wherein said gene encodes a glutaredoxin, said progeny having enhanced tolerance to an abiotic stressor. 16. The method of claim 15, wherein said progeny is homozygous for said abiotic stress tolerance gene. 17. The method of claim 15, wherein each of said first and second plants is a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene. 18. The method of claim 15, wherein said first plant is a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene, and said second plant has a characteristic selected from the group consisting of: pest resistance, herbicide resistance, geographic adaptation, and increased stalk strength. 19. A genetically-modified seed produced according to the method of claim 15. 20. A method of producing genetically-modified seed, said method comprising: providing a genetically-modified plant having ectopic expression of a heterologous, abiotic stress tolerance gene, wherein said gene encodes a glutaredoxin; and self-pollinating said plant to thereby produce genetically-modified seed, wherein said seed comprises said heterologous, abiotic stress tolerance gene.
1,600
563
15,305,114
1,627
The object of the present invention is to provide a formulation with the effect of effectively suppressing or inhibiting amyloid fibril formation by the dissolution, elimination (discharge), etc. of amyloid fibril formation in vivo. If an agent for suppressing or inhibiting an amyloid fibril formation comprising tranilast or a pharmacologically acceptable salt thereof as an active ingredient is administered by a method such as oral administration, amyloid fibril formation can be effectively suppressed or inhibited in vivo as a result of effects such as amyloid fibril dissolution or elimination (discharge). Therefore, it is possible to prevent or treat amyloid plaques, in which amyloid fibrils formed by the aggregation of amyloid protein have been deposited, and to prevent or treat diseases arising from amyloid fibril deposition, that is, diseases arising from the deposited amyloid fibrils themselves and diseases that cause dysfunction of organs or tissues as a result of amyloid fibril deposition.
1-11. (canceled) 12. A method for suppressing or inhibiting an amyloid fibril formation comprising administering an agent for suppressing or inhibiting an amyloid fibril formation to a patient in need of suppressing or inhibiting an amyloid fibril formation, wherein the agent comprises tranilast or a pharmacologically acceptable salt thereof as an active ingredient. 13. The method according to claim 12, wherein the agent is administered orally. 14. The method according to claim 12, wherein the agent has an effect of dissolving or discharging an amyloid fibril. 15. A method for preventing or treating an amyloid plaque comprising administering an agent for suppresing or inhibiting an amyloid fibril formation to a patient in need of preventing or treating an amyloid plaque, wherein the agent comprises tranilast or a pharmacologically acceptable salt thereof as an active ingredient. 16. The method according to claim 15, wherein the agent is administered orally. 17. The method according to claim 15, wherein the agent has an effect of dissolving or discharging an amyloid fibril. 18. The method according to claim 15, wherein the amyloid plaque is in the brain. 19. The method according to claim 16, wherein the amyloid plaque is in the brain. 20. The method according to claim 15, wherein the amyloid plaque is in the skin. 21. The method according to claim 16, wherein the amyloid plaque is in the skin. 22. A method for preventing or treating an amyloidosis comprising administering an agent for suppressing or inhibiting an amyloid fibril formation to a patient in need of preventing or treating an amyloidosis, wherein the agent comprises tranilast or a pharmacologically acceptable salt thereof as an active ingredient. 23. The method according to claim 22, wherein the agent is administered orally. 24. The method according to claim 22, wherein the agent has an effect of dissolving or discharging an amyloid fibril. 25. The method according to claim 22, wherein the amyloidosis is cerebral amyloidosis. 26. The method according to claim 25, wherein the cerebral amyloidosis is dementia arising from the deposition of an amyloid fibril in the brain. 27. The method according to claim 23, wherein the amyloidosis is cerebral amyloidosis. 28. The method according to claim 27, wherein the cerebral amyloidosis is dementia arising from the deposition of an amyloid fibril in the brain. 29. The method according to claim 22, wherein the amyloidosis is cutaneous amyloidosis. 30. The method according to claim 29, wherein the cutaneous amyloidosis is cutaneous lichen amyloidosis. 31. The method according to claim 23, wherein the amyloidosis is cutaneous amyloidosis.
The object of the present invention is to provide a formulation with the effect of effectively suppressing or inhibiting amyloid fibril formation by the dissolution, elimination (discharge), etc. of amyloid fibril formation in vivo. If an agent for suppressing or inhibiting an amyloid fibril formation comprising tranilast or a pharmacologically acceptable salt thereof as an active ingredient is administered by a method such as oral administration, amyloid fibril formation can be effectively suppressed or inhibited in vivo as a result of effects such as amyloid fibril dissolution or elimination (discharge). Therefore, it is possible to prevent or treat amyloid plaques, in which amyloid fibrils formed by the aggregation of amyloid protein have been deposited, and to prevent or treat diseases arising from amyloid fibril deposition, that is, diseases arising from the deposited amyloid fibrils themselves and diseases that cause dysfunction of organs or tissues as a result of amyloid fibril deposition.1-11. (canceled) 12. A method for suppressing or inhibiting an amyloid fibril formation comprising administering an agent for suppressing or inhibiting an amyloid fibril formation to a patient in need of suppressing or inhibiting an amyloid fibril formation, wherein the agent comprises tranilast or a pharmacologically acceptable salt thereof as an active ingredient. 13. The method according to claim 12, wherein the agent is administered orally. 14. The method according to claim 12, wherein the agent has an effect of dissolving or discharging an amyloid fibril. 15. A method for preventing or treating an amyloid plaque comprising administering an agent for suppresing or inhibiting an amyloid fibril formation to a patient in need of preventing or treating an amyloid plaque, wherein the agent comprises tranilast or a pharmacologically acceptable salt thereof as an active ingredient. 16. The method according to claim 15, wherein the agent is administered orally. 17. The method according to claim 15, wherein the agent has an effect of dissolving or discharging an amyloid fibril. 18. The method according to claim 15, wherein the amyloid plaque is in the brain. 19. The method according to claim 16, wherein the amyloid plaque is in the brain. 20. The method according to claim 15, wherein the amyloid plaque is in the skin. 21. The method according to claim 16, wherein the amyloid plaque is in the skin. 22. A method for preventing or treating an amyloidosis comprising administering an agent for suppressing or inhibiting an amyloid fibril formation to a patient in need of preventing or treating an amyloidosis, wherein the agent comprises tranilast or a pharmacologically acceptable salt thereof as an active ingredient. 23. The method according to claim 22, wherein the agent is administered orally. 24. The method according to claim 22, wherein the agent has an effect of dissolving or discharging an amyloid fibril. 25. The method according to claim 22, wherein the amyloidosis is cerebral amyloidosis. 26. The method according to claim 25, wherein the cerebral amyloidosis is dementia arising from the deposition of an amyloid fibril in the brain. 27. The method according to claim 23, wherein the amyloidosis is cerebral amyloidosis. 28. The method according to claim 27, wherein the cerebral amyloidosis is dementia arising from the deposition of an amyloid fibril in the brain. 29. The method according to claim 22, wherein the amyloidosis is cutaneous amyloidosis. 30. The method according to claim 29, wherein the cutaneous amyloidosis is cutaneous lichen amyloidosis. 31. The method according to claim 23, wherein the amyloidosis is cutaneous amyloidosis.
1,600
564
15,022,654
1,627
Provided herein are methods of treating or preventing a neurodegenerative by administering fenofibrate or an analog thereof.
1. A method of inducing PGC-1α expression in a neural cell comprising contacting a neural cell or population of neural cells with an effective amount of fenofibrate or an analog thereof. 2. The method of claim 1, wherein the contacting step is in vivo. 3. The method of claim 1 or 2, wherein the contacting step in vitro. 4. The method of any one of claims 1-3, wherein the induction of PGC-1α is PPARα independent. 5. The method of any one of claims 1-4, wherein the neural cell or population of neural cells comprises one or more neurons. 6. The method of claim 5, wherein the neuron or neurons are dopaminergic neuron. 7. The method of any of claims 1-4, wherein the neural cell or population of neural cells comprises one or more glial cells. 8. The method of any of claims 1-7, wherein the effective amount reduces one or more effects of oxidative stress. 9. The method of claim 1-7, wherein the effective amount increases a level of phosphorylated AMPK. 10. The method of claim 1-7, wherein the effective amount increases the number of mitochondria. 11. The method of claim 1-7, wherein the effective amount increases neural cell viability. 12. The method of claim 1-7, wherein the effective amount provides an anti-inflammatory effect. 13. A method of treating or preventing a neurodegenerative disease in a subject comprising selecting a subject with a neurodegenerative disease or at risk for a neurodegenerative disease; and administering to the subject an effective amount of fenofibrate or an analog thereof. 14. The method of claim 133, wherein the subject has an early stage neurodegenerative disease. 15. The method of claim 133 or 14, wherein the neurodegenerative disease is selected from the group consisting of Parkinson's Disease, Parkinson-plus syndrome, familial dementia, Alzheimer's Disease, Huntington's Disease, multiple sclerosis, dementia with Lewy bodies, Mild Cognitive Impairment, retinal neurodegeneration, and Amyotrophic Lateral Sclerosis. 16. The method of claim 155, wherein the Parkinson-plus syndrome is selected from the group consisting of multiple system atrophy (MSA), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD). 17. The method of any one of claims 133-166, wherein the fenofibrate or analog thereof is administered systemically. 18. The method of claim 177, wherein the fenofibrate or analog thereof is administered orally. 19. The method of any one of claims 143-188, wherein the effective amount of the fenofibrate or analog thereof induces PGC-1α expression in neural cells. 20. The method of any one of claims 143-199, wherein the fenofibrate or analog thereof is administered daily. 21. The method of any one of claims 143-20, further comprising administering a second therapeutic agent to the subject. 22. The method of claim 21, wherein the second therapeutic agent is selected from the group consisting of levadopa, a dopamine agonist, an anticholinergic agent, a monoamine oxidase inhibitor, a COMT inhibitor, amantadine, rivastigmine, an NMDA antagonist, a cholinesterase inhibitor, riluzole, an anti-psychotic agent, an antidepressant, and tetrabenazine. 23. The method of any one of claims 13-22, further comprising determining that the subject has a reduced level of PGC-1α expression as compared to a control subject. 24. A method of screening for an agent that promotes neuroprotection comprising contacting a cell with an agent to be screened and detecting PGC-1α level or activity in the cell, an increase in PGC-1α level or activity indicating the agent promotes neuroprotection. 25. The method of claim 24, wherein the cell is a neuron, glial cell or mononuclear blood cell.
Provided herein are methods of treating or preventing a neurodegenerative by administering fenofibrate or an analog thereof.1. A method of inducing PGC-1α expression in a neural cell comprising contacting a neural cell or population of neural cells with an effective amount of fenofibrate or an analog thereof. 2. The method of claim 1, wherein the contacting step is in vivo. 3. The method of claim 1 or 2, wherein the contacting step in vitro. 4. The method of any one of claims 1-3, wherein the induction of PGC-1α is PPARα independent. 5. The method of any one of claims 1-4, wherein the neural cell or population of neural cells comprises one or more neurons. 6. The method of claim 5, wherein the neuron or neurons are dopaminergic neuron. 7. The method of any of claims 1-4, wherein the neural cell or population of neural cells comprises one or more glial cells. 8. The method of any of claims 1-7, wherein the effective amount reduces one or more effects of oxidative stress. 9. The method of claim 1-7, wherein the effective amount increases a level of phosphorylated AMPK. 10. The method of claim 1-7, wherein the effective amount increases the number of mitochondria. 11. The method of claim 1-7, wherein the effective amount increases neural cell viability. 12. The method of claim 1-7, wherein the effective amount provides an anti-inflammatory effect. 13. A method of treating or preventing a neurodegenerative disease in a subject comprising selecting a subject with a neurodegenerative disease or at risk for a neurodegenerative disease; and administering to the subject an effective amount of fenofibrate or an analog thereof. 14. The method of claim 133, wherein the subject has an early stage neurodegenerative disease. 15. The method of claim 133 or 14, wherein the neurodegenerative disease is selected from the group consisting of Parkinson's Disease, Parkinson-plus syndrome, familial dementia, Alzheimer's Disease, Huntington's Disease, multiple sclerosis, dementia with Lewy bodies, Mild Cognitive Impairment, retinal neurodegeneration, and Amyotrophic Lateral Sclerosis. 16. The method of claim 155, wherein the Parkinson-plus syndrome is selected from the group consisting of multiple system atrophy (MSA), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD). 17. The method of any one of claims 133-166, wherein the fenofibrate or analog thereof is administered systemically. 18. The method of claim 177, wherein the fenofibrate or analog thereof is administered orally. 19. The method of any one of claims 143-188, wherein the effective amount of the fenofibrate or analog thereof induces PGC-1α expression in neural cells. 20. The method of any one of claims 143-199, wherein the fenofibrate or analog thereof is administered daily. 21. The method of any one of claims 143-20, further comprising administering a second therapeutic agent to the subject. 22. The method of claim 21, wherein the second therapeutic agent is selected from the group consisting of levadopa, a dopamine agonist, an anticholinergic agent, a monoamine oxidase inhibitor, a COMT inhibitor, amantadine, rivastigmine, an NMDA antagonist, a cholinesterase inhibitor, riluzole, an anti-psychotic agent, an antidepressant, and tetrabenazine. 23. The method of any one of claims 13-22, further comprising determining that the subject has a reduced level of PGC-1α expression as compared to a control subject. 24. A method of screening for an agent that promotes neuroprotection comprising contacting a cell with an agent to be screened and detecting PGC-1α level or activity in the cell, an increase in PGC-1α level or activity indicating the agent promotes neuroprotection. 25. The method of claim 24, wherein the cell is a neuron, glial cell or mononuclear blood cell.
1,600
565
15,698,846
1,613
The present disclosure is directed to an enhanced biocide composition containing an isothiazolinone and a pyrithione. In particular, the composition contains an isothiazolinone, a pyrithione, a zinc compound, and an organic amine. According to the present disclosure, the combination of components employed within the composition provides a synergistic effect against microorganisms, such as bacteria and fungi.
1. A biocide composition comprising: an isothiazolinone, a pyrithione, a zinc compound, and an organic amine. 2. The biocide composition of claim 1, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one, N-butyl-1,2-benzisothiazolin-3-one, N-methyl-1,2-benzisothiazolin-3-one, 2-methyl-2H-isothiazol-3-one, 5-chloro-2-methyl-2H-isothiazol-3-one, 2-octyl-3(2H)-isothiazolone, 4,5-dichloro-2n-octyl-3(2H)-isothiazolone, 2,2-dithiobis(N-methylbenzamide), or any combination thereof. 3. The biocide composition of claim 2, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one, N-butyl-1,2-benzisothiazolin-3-one, or any combination thereof. 4. The biocide composition of claim 3, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one. 5. The biocide composition of claim 1, wherein the pyrithione comprises sodium pyrithione, bismuth pyrithione, potassium pyrithione, lithium pyrithione, ammonium pyrithione, zinc pyrithione, copper pyrithione, calcium pyrithione, magnesium pyrithione, strontium pyrithione, silver pyrithione, gold pyrithione, manganese pyrithione, an organic amine pyrithione, or any combination thereof. 6. The biocide composition of claim 5, wherein the pyrithione comprises sodium pyrithione. 7. The biocide composition of claim 1, wherein the pyrithione comprises an alkali metal salt of pyrithione. 8. The biocide composition of claim 1, wherein the zinc compound comprises a zinc salt of an organic acid, a zinc salt of an inorganic acid, a zinc oxide, a zinc hydroxide, or any combination thereof. 9. The biocide composition of claim 8, wherein the zinc compound comprises a zinc salt of an inorganic acid. 10. The biocide composition of claim 9, wherein the zinc compound comprises zinc chloride. 11. The biocide composition of claim 1, wherein the zinc compound comprises a zinc salt selected from the group consisting of zinc acetate, zinc borate, zinc bromide, zinc carbonate, basic zinc carbonate, zinc chloride, zinc sulfate, zinc citrate, zinc fluoride, zinc iodide, zinc lactate, zinc oleate, zinc oxalate, zinc phosphate, zinc propionate, zinc salicylate, zinc selenate, zinc silicate, zinc stearate, zinc sulfide, zinc sulfate, zinc tannate, zinc tartrate, zinc valerate, or any combination thereof. 12. The biocide composition of claim 1, wherein the zinc compound comprises a binary zinc compound selected from the group consisting of zinc oxide, zinc sulfide, zinc fluoride, zinc iodide, zinc chloride, zinc bromide, zinc peroxide, zinc hydride, zinc carbide, zinc nitride, or any combination thereof. 13. The biocide composition of claim 1, wherein the organic amine comprises a compound of the following formula NR1R2R3 wherein R1, R2, and R3 are individually hydrogen or a C1-5 alkyl, optionally substituted, wherein at least one of R1, R2, and R3 is a C1-5 alkyl, optionally substituted. 14. The biocide composition of claim 13, wherein at least two of R1, R2, and R3 are hydrogen and at least one of R1, R2, and R3 is a C1-5 alkyl. 15. The biocide composition of claim 13, wherein the C1-5 alkyl is substituted with a hydroxyl group. 16. The biocide composition of claim 1, wherein the organic amine comprises an alkanolamine. 17. The biocide composition of claim 16, wherein the organic amine comprises ethanolamine. 18. The biocide composition of claim 1, wherein the composition further comprises a second organic amine. 19. The biocide composition of claim 18, wherein the second organic amine is selected from the group consisting of a monomer or a polymer of an alkyl diamine of the following formula R7R8N—[(CH2)p—CH2—NH—]q—H wherein R7 and R8 are individually hydrogen or a C1-5 alkyl, optionally substituted, p is 1 or 2, and q is from about 1 to about 2000. 20. The biocide composition of claim 1, wherein the composition comprises 1,2-benzisothiazolin-3-one, sodium pyrithione, zinc chloride, and ethanolamine. 21. The biocide composition of claim 1, wherein the weight ratio of the pyrithione to the isothiazolinone is from 0.1 to 10. 22. The biocide composition of claim 1, wherein the weight ratio of the isothiazolinone to the zinc compound is from 0.005 to 200. 23. An end-use formulation comprising the biocide composition of claim 1, 24. The end-use formulation of claim 23, wherein the end-use formulation comprising a coating composition. 25. A method of preserving a substrate, the method comprising applying the coating composition of claim 24 to the substrate. 26. A biocide composition concentrate comprising: an isothiazolinone, a pyrithione, a zinc compound, and an organic amine. 27. The biocide composition concentrate of claim 26, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one, the pyrithione comprises sodium pyrithione, the zinc compound comprises zinc chloride, and the organic amine comprises ethanolamine. 28. The biocide composition concentrate of claim 26, wherein the weight ratio of the pyrithione to the isothiazolinone is from 0.1 to 10. 29. The biocide composition concentrate of claim 26, wherein the weight ratio of the isothiazolinone to the zinc compound is from 0.005 to 200.
The present disclosure is directed to an enhanced biocide composition containing an isothiazolinone and a pyrithione. In particular, the composition contains an isothiazolinone, a pyrithione, a zinc compound, and an organic amine. According to the present disclosure, the combination of components employed within the composition provides a synergistic effect against microorganisms, such as bacteria and fungi.1. A biocide composition comprising: an isothiazolinone, a pyrithione, a zinc compound, and an organic amine. 2. The biocide composition of claim 1, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one, N-butyl-1,2-benzisothiazolin-3-one, N-methyl-1,2-benzisothiazolin-3-one, 2-methyl-2H-isothiazol-3-one, 5-chloro-2-methyl-2H-isothiazol-3-one, 2-octyl-3(2H)-isothiazolone, 4,5-dichloro-2n-octyl-3(2H)-isothiazolone, 2,2-dithiobis(N-methylbenzamide), or any combination thereof. 3. The biocide composition of claim 2, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one, N-butyl-1,2-benzisothiazolin-3-one, or any combination thereof. 4. The biocide composition of claim 3, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one. 5. The biocide composition of claim 1, wherein the pyrithione comprises sodium pyrithione, bismuth pyrithione, potassium pyrithione, lithium pyrithione, ammonium pyrithione, zinc pyrithione, copper pyrithione, calcium pyrithione, magnesium pyrithione, strontium pyrithione, silver pyrithione, gold pyrithione, manganese pyrithione, an organic amine pyrithione, or any combination thereof. 6. The biocide composition of claim 5, wherein the pyrithione comprises sodium pyrithione. 7. The biocide composition of claim 1, wherein the pyrithione comprises an alkali metal salt of pyrithione. 8. The biocide composition of claim 1, wherein the zinc compound comprises a zinc salt of an organic acid, a zinc salt of an inorganic acid, a zinc oxide, a zinc hydroxide, or any combination thereof. 9. The biocide composition of claim 8, wherein the zinc compound comprises a zinc salt of an inorganic acid. 10. The biocide composition of claim 9, wherein the zinc compound comprises zinc chloride. 11. The biocide composition of claim 1, wherein the zinc compound comprises a zinc salt selected from the group consisting of zinc acetate, zinc borate, zinc bromide, zinc carbonate, basic zinc carbonate, zinc chloride, zinc sulfate, zinc citrate, zinc fluoride, zinc iodide, zinc lactate, zinc oleate, zinc oxalate, zinc phosphate, zinc propionate, zinc salicylate, zinc selenate, zinc silicate, zinc stearate, zinc sulfide, zinc sulfate, zinc tannate, zinc tartrate, zinc valerate, or any combination thereof. 12. The biocide composition of claim 1, wherein the zinc compound comprises a binary zinc compound selected from the group consisting of zinc oxide, zinc sulfide, zinc fluoride, zinc iodide, zinc chloride, zinc bromide, zinc peroxide, zinc hydride, zinc carbide, zinc nitride, or any combination thereof. 13. The biocide composition of claim 1, wherein the organic amine comprises a compound of the following formula NR1R2R3 wherein R1, R2, and R3 are individually hydrogen or a C1-5 alkyl, optionally substituted, wherein at least one of R1, R2, and R3 is a C1-5 alkyl, optionally substituted. 14. The biocide composition of claim 13, wherein at least two of R1, R2, and R3 are hydrogen and at least one of R1, R2, and R3 is a C1-5 alkyl. 15. The biocide composition of claim 13, wherein the C1-5 alkyl is substituted with a hydroxyl group. 16. The biocide composition of claim 1, wherein the organic amine comprises an alkanolamine. 17. The biocide composition of claim 16, wherein the organic amine comprises ethanolamine. 18. The biocide composition of claim 1, wherein the composition further comprises a second organic amine. 19. The biocide composition of claim 18, wherein the second organic amine is selected from the group consisting of a monomer or a polymer of an alkyl diamine of the following formula R7R8N—[(CH2)p—CH2—NH—]q—H wherein R7 and R8 are individually hydrogen or a C1-5 alkyl, optionally substituted, p is 1 or 2, and q is from about 1 to about 2000. 20. The biocide composition of claim 1, wherein the composition comprises 1,2-benzisothiazolin-3-one, sodium pyrithione, zinc chloride, and ethanolamine. 21. The biocide composition of claim 1, wherein the weight ratio of the pyrithione to the isothiazolinone is from 0.1 to 10. 22. The biocide composition of claim 1, wherein the weight ratio of the isothiazolinone to the zinc compound is from 0.005 to 200. 23. An end-use formulation comprising the biocide composition of claim 1, 24. The end-use formulation of claim 23, wherein the end-use formulation comprising a coating composition. 25. A method of preserving a substrate, the method comprising applying the coating composition of claim 24 to the substrate. 26. A biocide composition concentrate comprising: an isothiazolinone, a pyrithione, a zinc compound, and an organic amine. 27. The biocide composition concentrate of claim 26, wherein the isothiazolinone comprises 1,2-benzisothiazolin-3-one, the pyrithione comprises sodium pyrithione, the zinc compound comprises zinc chloride, and the organic amine comprises ethanolamine. 28. The biocide composition concentrate of claim 26, wherein the weight ratio of the pyrithione to the isothiazolinone is from 0.1 to 10. 29. The biocide composition concentrate of claim 26, wherein the weight ratio of the isothiazolinone to the zinc compound is from 0.005 to 200.
1,600
566
15,394,485
1,662
The present invention relates to novel gene sequences encoding insecticidal proteins produced by Bacillus thuringiensis strains. Particularly, new chimeric genes encoding a CryIC, CryIB or CryID protein are provided which are useful to protect plants from insect damage. Also included herein are plant cells or plants comprising such genes and methods of making or using them, as well as plant cells or plants comprising one of such chimeric gene and at least one other such chimeric genes.
1. A chimeric gene comprising the following operably-linked sequences: a) a coding region encoding a Cry1C protein; comprising a DNA sequence with at least 98% sequence identity to the DNA of any one of SEQ ID Nos. 1, 3, 4 or 6, wherein said Cry1C protein is a protein comprising the amino acid sequence from the amino acid at position 29 to the amino acid at position 627 in SEQ ID No. 2, and b) a promoter region capable of directing expression in plant cells. 2. The chimeric gene of claim 1, wherein said promoter comprises the sequence of SEQ ID No. 18 or 19. 3. The chimeric gene of claim 1, further comprising a 3′ polyadenylation and transcript termination region. 4. The chimeric gene of claim 3, wherein said 3′ polyadenylation and transcript termination region is of the NADP-malic enzyme gene from Flaveria bidentis. 5. The chimeric gene of claim 1, further comprising the leader sequence of the tapetum specific E1 gene of Oryza sativa between the promoter and the coding region. 6. A DNA comprising the chimeric gene of claim 1, further comprising a second chimeric gene comprising the following operably-linked sequences: a) a second coding region encoding a Cry1B protein comprising a DNA sequence with at least 98% sequence identity to the DNA of SEQ ID No. 8 or 10, wherein said Cry1B protein is a protein comprising the amino acid sequence from the amino acid at position 31 to the amino acid at position 648 in SEQ ID No. 11, and b) a second promoter region capable of directing expression in plant cells. 7. A DNA comprising the chimeric gene of claim 1, further comprising a second chimeric gene comprising the following operably-linked sequences: a) a coding region encoding a Cry1D protein comprising a DNA sequence with at least 98% sequence identity to the DNA of SEQ ID No. 12 or 14, and wherein said Cry1D protein is a protein comprising the amino acid sequence from the amino acid at position 21 to the amino acid at position 604 in SEQ ID No. 15, and b) a promoter region capable of directing expression in plant cells. 8. The DNA of claim 6, wherein said second promoter region comprises the sequence of SEQ ID No. 18 or 19 and is different from said first promoter region. 9. The DNA of claim 6, wherein said second chimeric gene further comprises a 3′ polyadenylation and transcript termination region. 10. The DNA of claim 9, wherein said 3′ polyadenylation and transcript termination region is of the NADP-malic enzyme gene from Flaveria bidentis. 11. The DNA of claim 6, wherein said second chimeric gene further comprises the leader sequence of the tapetum specific E1 gene of Oryza sativa between the promoter and the coding region. 12. The DNA of claim 6, further comprising a third chimeric gene comprising the following operably-linked sequences: a) a coding region encoding a Cry1D protein comprising the DNA of SEQ ID No. 12 or 14, and b) a promoter region capable of directing expression in plants. 13. A transgenic plant cell comprising the gene of claim 1 stably incorporated in its genome. 14. A plant comprising the gene of claim 1 stably incorporated in its genome. 15. The plant of claim 14, which is a Brassica species plant or plant cell. 16. The plant of claim 15, wherein said plant is of the species Brassica oleraceae, Brassica napus, Brassica rapa, Brassica juncea or Brassica carinata. 17. The plant of claim 16, wherein said Brassica oleraceae is cabbage or cauliflower. 18. (canceled) 19. A method for controlling insects, comprising planting or sowing in a field, plants comprising the chimeric gene of claim 1. 20. (canceled) 21. A method of controlling insects in Brassica species plants, comprising expressing the chimeric gene of claim 1 in plants and planting or growing said plants. 22. A method of producing plants or seeds resistant to insects, comprising the steps of: a) obtaining a plant transformed with the, chimeric gene of claim 1, and b) selecting progeny of said plant or seeds thereof, containing said gene. 23. A microorganism comprising the chimeric gene of claim 1. 24. The microorganism of claim 23, wherein said microorganism is of the genus Escherichia, Bacillus or Agrobacterium. 25-33. (canceled) 34. A transgenic plant cell comprising the DNA of claim 6 stably incorporated in its genome. 35. A plant comprising the DNA of claim 6 stably incorporated in its genome. 36. The plant of claim 35, wherein said plant is of the species Brassica oleraceae, Brassica napus, Brassica rape, Brassica juncea, or Brassica carinata. 37. The plant of claim 36, wherein said plant is cabbage or cauliflower. 38. A method for controlling insects comprising planting or sowing in a field, plants comprising the DNA of claim 6. 39. A method of controlling insects in Brassica species plants, comprising expressing the DNA of claim 6 in said plants and planting or growing said plants. 40. A method of producing plants or seeds resistant to insects, comprising the steps of: (a) obtaining a plant transformed with the DNA of claim 6, and (b) selecting progeny of said plant or seeds thereof, containing said DNA. 41. A microorganism comprising the DNA of claim 6. 42. The microorganism of claim 41, wherein said microorganism is of the genus Escherichia, Bacillus, or Agrobacterium.
The present invention relates to novel gene sequences encoding insecticidal proteins produced by Bacillus thuringiensis strains. Particularly, new chimeric genes encoding a CryIC, CryIB or CryID protein are provided which are useful to protect plants from insect damage. Also included herein are plant cells or plants comprising such genes and methods of making or using them, as well as plant cells or plants comprising one of such chimeric gene and at least one other such chimeric genes.1. A chimeric gene comprising the following operably-linked sequences: a) a coding region encoding a Cry1C protein; comprising a DNA sequence with at least 98% sequence identity to the DNA of any one of SEQ ID Nos. 1, 3, 4 or 6, wherein said Cry1C protein is a protein comprising the amino acid sequence from the amino acid at position 29 to the amino acid at position 627 in SEQ ID No. 2, and b) a promoter region capable of directing expression in plant cells. 2. The chimeric gene of claim 1, wherein said promoter comprises the sequence of SEQ ID No. 18 or 19. 3. The chimeric gene of claim 1, further comprising a 3′ polyadenylation and transcript termination region. 4. The chimeric gene of claim 3, wherein said 3′ polyadenylation and transcript termination region is of the NADP-malic enzyme gene from Flaveria bidentis. 5. The chimeric gene of claim 1, further comprising the leader sequence of the tapetum specific E1 gene of Oryza sativa between the promoter and the coding region. 6. A DNA comprising the chimeric gene of claim 1, further comprising a second chimeric gene comprising the following operably-linked sequences: a) a second coding region encoding a Cry1B protein comprising a DNA sequence with at least 98% sequence identity to the DNA of SEQ ID No. 8 or 10, wherein said Cry1B protein is a protein comprising the amino acid sequence from the amino acid at position 31 to the amino acid at position 648 in SEQ ID No. 11, and b) a second promoter region capable of directing expression in plant cells. 7. A DNA comprising the chimeric gene of claim 1, further comprising a second chimeric gene comprising the following operably-linked sequences: a) a coding region encoding a Cry1D protein comprising a DNA sequence with at least 98% sequence identity to the DNA of SEQ ID No. 12 or 14, and wherein said Cry1D protein is a protein comprising the amino acid sequence from the amino acid at position 21 to the amino acid at position 604 in SEQ ID No. 15, and b) a promoter region capable of directing expression in plant cells. 8. The DNA of claim 6, wherein said second promoter region comprises the sequence of SEQ ID No. 18 or 19 and is different from said first promoter region. 9. The DNA of claim 6, wherein said second chimeric gene further comprises a 3′ polyadenylation and transcript termination region. 10. The DNA of claim 9, wherein said 3′ polyadenylation and transcript termination region is of the NADP-malic enzyme gene from Flaveria bidentis. 11. The DNA of claim 6, wherein said second chimeric gene further comprises the leader sequence of the tapetum specific E1 gene of Oryza sativa between the promoter and the coding region. 12. The DNA of claim 6, further comprising a third chimeric gene comprising the following operably-linked sequences: a) a coding region encoding a Cry1D protein comprising the DNA of SEQ ID No. 12 or 14, and b) a promoter region capable of directing expression in plants. 13. A transgenic plant cell comprising the gene of claim 1 stably incorporated in its genome. 14. A plant comprising the gene of claim 1 stably incorporated in its genome. 15. The plant of claim 14, which is a Brassica species plant or plant cell. 16. The plant of claim 15, wherein said plant is of the species Brassica oleraceae, Brassica napus, Brassica rapa, Brassica juncea or Brassica carinata. 17. The plant of claim 16, wherein said Brassica oleraceae is cabbage or cauliflower. 18. (canceled) 19. A method for controlling insects, comprising planting or sowing in a field, plants comprising the chimeric gene of claim 1. 20. (canceled) 21. A method of controlling insects in Brassica species plants, comprising expressing the chimeric gene of claim 1 in plants and planting or growing said plants. 22. A method of producing plants or seeds resistant to insects, comprising the steps of: a) obtaining a plant transformed with the, chimeric gene of claim 1, and b) selecting progeny of said plant or seeds thereof, containing said gene. 23. A microorganism comprising the chimeric gene of claim 1. 24. The microorganism of claim 23, wherein said microorganism is of the genus Escherichia, Bacillus or Agrobacterium. 25-33. (canceled) 34. A transgenic plant cell comprising the DNA of claim 6 stably incorporated in its genome. 35. A plant comprising the DNA of claim 6 stably incorporated in its genome. 36. The plant of claim 35, wherein said plant is of the species Brassica oleraceae, Brassica napus, Brassica rape, Brassica juncea, or Brassica carinata. 37. The plant of claim 36, wherein said plant is cabbage or cauliflower. 38. A method for controlling insects comprising planting or sowing in a field, plants comprising the DNA of claim 6. 39. A method of controlling insects in Brassica species plants, comprising expressing the DNA of claim 6 in said plants and planting or growing said plants. 40. A method of producing plants or seeds resistant to insects, comprising the steps of: (a) obtaining a plant transformed with the DNA of claim 6, and (b) selecting progeny of said plant or seeds thereof, containing said DNA. 41. A microorganism comprising the DNA of claim 6. 42. The microorganism of claim 41, wherein said microorganism is of the genus Escherichia, Bacillus, or Agrobacterium.
1,600
567
15,468,694
1,653
Provided are processes for facilitating the removal of a biological stain is provided wherein a substrate or coating including a protease is capable of enzymatically degrading of one or more components of the biological stain to facilitate biological stain removal from the substrate or said coating.
1. A method of facilitating the removal of a biological stain on a substrate or a coating comprising: providing a substrate or a coating; associating a protease with said substrate or said coating such that said substrate or said coating is capable of enzymatically degrading a component of a biological stain. 2. The method of claim 1 wherein said protease is a bacterial neutral thermolysin-like-protease from Bacillus stearothermophilus. 3. The method of claim 1 wherein the surface activity of the substrate or coating is 0.0075 units/cm2 or greater. 4. The method of claim 1 wherein said protease is covalently attached to said substrate or to said coating. 5. The method of claim 1 wherein said protease is non-covalently adhered to or admixed into said substrate or said coating. 6. The method of claim 1 wherein said substrate or said coating comprises an organic crosslinkable polymer resin. 7. The method of claim 6 wherein said organic crosslinkable polymer resin comprises a functional group of acetoacetate, acid, amine, carboxyl, epoxy, hydroxyl, isocyanate, silane, vinyl, or combinations thereof. 8. The method of claim 7 wherein said organic crosslinkable polymer resin is aminoplasts, melamine formaldehydes, carbamates, polyurethanes, polyacrylates, epoxies, polycarbonates, alkyds, vinyls, polyamides, polyolefins, phenolic resins, polyesters, polysiloxanes, or combinations thereof. 9. The method of claim 7 wherein said organic crosslinkable polymer is a hydroxyl-functionalized acrylate resin.
Provided are processes for facilitating the removal of a biological stain is provided wherein a substrate or coating including a protease is capable of enzymatically degrading of one or more components of the biological stain to facilitate biological stain removal from the substrate or said coating.1. A method of facilitating the removal of a biological stain on a substrate or a coating comprising: providing a substrate or a coating; associating a protease with said substrate or said coating such that said substrate or said coating is capable of enzymatically degrading a component of a biological stain. 2. The method of claim 1 wherein said protease is a bacterial neutral thermolysin-like-protease from Bacillus stearothermophilus. 3. The method of claim 1 wherein the surface activity of the substrate or coating is 0.0075 units/cm2 or greater. 4. The method of claim 1 wherein said protease is covalently attached to said substrate or to said coating. 5. The method of claim 1 wherein said protease is non-covalently adhered to or admixed into said substrate or said coating. 6. The method of claim 1 wherein said substrate or said coating comprises an organic crosslinkable polymer resin. 7. The method of claim 6 wherein said organic crosslinkable polymer resin comprises a functional group of acetoacetate, acid, amine, carboxyl, epoxy, hydroxyl, isocyanate, silane, vinyl, or combinations thereof. 8. The method of claim 7 wherein said organic crosslinkable polymer resin is aminoplasts, melamine formaldehydes, carbamates, polyurethanes, polyacrylates, epoxies, polycarbonates, alkyds, vinyls, polyamides, polyolefins, phenolic resins, polyesters, polysiloxanes, or combinations thereof. 9. The method of claim 7 wherein said organic crosslinkable polymer is a hydroxyl-functionalized acrylate resin.
1,600
568
14,414,218
1,613
Embodiments of various aspects described herein relates to compositions and methods for encapsulation and/or stabilization of oil, lipid, hydrophobic and/or lipophilic compounds in a silk-based material. The compositions described herein can be used in various applications, e.g., pharmaceutical, cosmetic, food, diagnostic, and tissue engineering applications.
1. A silk particle comprising at least two immiscible phases, a first immiscible phase comprising a silk-based material and a second immiscible phase comprising an active agent, wherein the first immiscible phase encapsulates the second immiscible phase and the second immiscible phase excludes a liposome. 2. The silk particle of claim 1, wherein the second immiscible phase comprises a lipid component. 3. The silk particle of claim 2, wherein the lipid component comprises oil. 4. The silk particle of any of claims 1-3, wherein the second immiscible phase forms a single compartment. 5. The silk particle of any of claims 1-3, wherein the second immiscible phase forms a plurality of compartments. 6. The silk particle of claim 4 or 5, wherein the size of the compartment or compartments ranges from about 1 nm to about 1000 μm, or from about 5 nm to about 500 μm. 7. The silk particle of any of claims 1-6, wherein the active agent present in the second immiscible phase comprises a hydrophobic or lipophilic molecule. 8. The silk particle of claim 7, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), a small molecule, or any combinations thereof. 9. The silk particle of any of claims 1-8, wherein the silk-based material comprises an additive. 10. The silk particle of claim 9, wherein the additive is selected from the group consisting of biocompatible polymers; plasticizers (e.g., glycerol); stimulus-responsive agents; active agents, small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; antibodies and antigen binding fragments thereof; nucleic acids; nucleic acid analogs and derivatives; glycogens or other sugars; immunogens; antigens; an extract made from biological materials such as bacteria, plants, fungi, or animal cells; animal tissues; naturally occurring or synthetic compositions; and any combinations thereof. 11. The silk particle of claim 9 or 10, wherein the additive is in a form of a particle (e.g., a nanoparticle or microparticle, including a plasmonic particle), a fiber, a tube, powder or any combinations thereof. 12. The silk particle of any of claims 9-11, wherein the additive comprises a silk material, e.g., silk particles, silk fibers, micro-sized silk fibers, unprocessed silk fibers, and any combinations thereof. 13. The silk particle of any of claims 1-12, wherein the second immiscible phase encapsulates a third immiscible phase. 14. The silk particle of any of claims 1-13, wherein the silk-based material is present in a form of a hydrogel. 15. The silk particle of any of claims 1-14, wherein the silk-based material is present in a dried state or lyophilized. 16. The silk particle of claim 15, wherein the lyophilized silk matrix is porous. 17. The silk particle of any of claims 1-16, wherein the silk-based material in the first immiscible phase is soluble in an aqueous solution. 18. The silk particle of any of claims 1-17, wherein beta-sheet content in the silk-based material is adjusted to an amount sufficient to enable the silk-based material to resist dissolution in an aqueous solution. 19. The silk particle of any of claims 1-18, wherein the size of the silk particle ranges from about 10 nm to about 10 mm, or from about 50 nm to about 5 mm. 20. A composition comprising a plurality of lipid compartments encapsulated in a silk-based material. 21. The composition of claim 20, wherein the size of the lipid compartments ranges from about 1 nm to about 1000 μm, or from about 5 nm to about 500 μm. 22. The composition of claim 20 or 21, wherein the volumetric ratio of the lipid compartments to the silk-based material ranges from about 1000:1 to about 1:1000, from about 500:1 to about 1:500, or from about 100:1 to about 1:100. 23. The composition of any of claims 20-22, wherein the silk-based material is in a form selected from the group consisting of a film, a sheet, a gel or hydrogel, a mesh, a mat, a non-woven mat, a fabric, a scaffold, a tube, a slab or block, a fiber, a particle, powder, a 3-dimensional construct, an implant, a foam or a sponge, a needle, a lyophilized material, a porous material, a non-porous material, and any combinations thereof. 24. The composition of any of claims 20-23, wherein the silk-based material comprises a film. 25. The composition of any of claims 20-24, wherein the silk-based material comprises a scaffold. 26. The composition of any of claims 20-25, wherein the silk-based material comprises an optical pattern. 27. The composition of claim 26, wherein the optical pattern comprises a hologram or an array of patterns that provides an optical functionality. 28. The composition of any of claims 20-27, wherein the lipid compartments further comprise an active agent. 29. The composition of claim 20-28, wherein the active agent comprises a hydrophobic or lipophilic molecule. 30. The composition of claim 29, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), a small molecule, or any combinations thereof. 31. The composition of any of claims 20-30, wherein the silk-based material comprises an additive. 32. The composition of claim 31, wherein the additive is selected from the group consisting of biocompatible polymers; plasticizers (e.g., glycerol); stimulus-responsive agents; small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; antibodies and antigen binding fragments thereof; nucleic acids; nucleic acid analogs and derivatives; glycogens or other sugars; immunogens; antigens; an extract made from biological materials such as bacteria, plants, fungi, or animal cells; animal tissues; naturally occurring or synthetic compositions; and any combinations thereof. 33. The composition of claim 31 or 32, wherein the additive is in a form selected from the group consisting of a particle, a fiber, a tube, a film, a gel, a mesh, a mat, a non-woven mat, a powder, and any combinations thereof. 34. The composition of any of claims 31-33, wherein the additive comprises a silk material, e.g., silk particles, silk fibers, micro-sized silk fibers, unprocessed silk fibers, and any combinations thereof. 35. A composition comprising a collection of silk particles of any of claims 1-19. 36. The composition of claim 35, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, a scaffold, or any combinations thereof. 37. The composition of claim 35 or 36, wherein the composition is formulated for use in a pharmaceutical product. 38. The composition of claim 35 or 36, wherein the composition is formulated for use in a cosmetic product. 39. The composition of claim 35 or 36, wherein the composition is formulated for use in a personal care product. 40. The composition of claim 35 or 36, wherein the composition is formulated for use in a food product. 41. A storage-stable composition comprising a silk particle of any of claims 1-19 or a composition of any of claims 20-40, wherein the active agent present in the second immiscible phase of the silk particle, or a hydrophobic or lipophilic molecule present in the lipid components retains at least about 30% of its original bioactivity after the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 42. The composition of claim 41, wherein the composition is maintained under exposure to light. 43. The composition of claim 41 or 42, wherein the composition is maintained at a relative humidity of at least about 10%. 44. The composition of any of claims 41-43, wherein the silk-based material of the silk particle or the composition is in a dried-state. 45. A method of producing a silk particle comprising: a. providing an emulsion of non-aqueous droplets dispersed in a silk solution undergoing a sol-gel transition (where the silk solution remains in a mixable state); and b. contacting a pre-determined volume of the emulsion with a non-aqueous phase, whereby the silk solution forms in the non-aqueous phase a silk particle entrapping at least one of the non-aqueous droplets therein. 46. The method of claim 45, wherein the sol-gel transition last for about at least 1 hour, or at least about 2 hours. 47. The method of claim 45 or 46, wherein the sol-gel transition of the silk solution is induced by sonication. 48. The method of claim 47, where the sonication is performed at an amplitude of about 5% to about 20%, or about 10% to about 15%. 49. The method of claim 47 or 48, wherein the sonication duration lasts for about 15 sec to about 60 sec, or from about 30 sec to about 45 sec. 50. The method of any of claims 45-49, wherein the silk solution has a concentration of about 1% (w/v) to about 15% (w/v), or about 2% (w/v) to about 7% (w/v). 51. The method of any of claims 45-50, further comprising adding an active agent into the silk fibroin solution undergoing a sol-gel transition. 52. The method of any of claims 45-51, wherein the non-aqueous droplets further comprise a hydrophobic or lipophilic molecule. 53. The method of claim 52, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), a small molecule, or any combinations thereof. 54. The method of any of claims 45-53, wherein the emulsion is produced by adding a non-aqueous, immiscible phase into the silk solution, thereby forming the non-aqueous droplets dispersed in the silk solution. 55. The method of any of claims 45-54, wherein the pre-determined volume of the emulsion substantially corresponds to a desirable size of the silk particle. 56. The method of any of claims 45-55, further comprising isolating the silk particle from the non-aqueous phase. 57. The method of any of claims 45-56, further comprising subjecting the silk particle to a post-treatment. 58. The method of claim 57, wherein the post-treatment further induces a conformational change in silk fibroin in the particle. 59. The method of claim 58, wherein said inducing conformational change comprises one or more of lyophilization or freeze-drying, water annealing, water vapor annealing, alcohol immersion, sonication, shear stress, electrogelation, pH reduction, salt addition, air-drying, electrospinning, stretching, or any combination thereof. 60. The method of any of claims 57-59, wherein the post-treatment comprises freeze-drying the silk particle. 61. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated in a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the active agent retains at least about 30% of its original bioactivity after the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 62. The method of claim 61, wherein the composition is maintained for at least about 1 month. 63. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated in a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the silk-based material is permeable to said at least one active agent such that the active agent is released through the silk-based material into an ambient surrounding at a pre-determined rate. 64. The method of claim 63, wherein the pre-determined rate is controlled by adjusting an amount of beta-sheet conformation of silk fibroin present in the silk-based material, porosity of the silk-based material, or a combination thereof. 65. The method of claim 63 or 64, wherein the composition is maintained at about room temperature. 66. The method of any of claims 61-65, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, or any combinations thereof. 67. The method of any of claims 61-66, wherein the composition is lyophilized. 68. The method of any of claims 61-67, wherein the composition is maintained at a temperature of about 37° C. or greater. 69. The method of any of claims 61-68, wherein the composition is maintained under exposure to light. 70. The method of any of claims 61-69, wherein the composition is maintained at a relative humidity of at least about 10%. 71. The method of any of claims 61-70, wherein the active agent comprises a hydrophobic or lipophilic active agent. 72. The method of claim 71, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 73. The method of any of claims 61-72, wherein the silk-based material comprises an additive. 74. The method of claim 73, wherein the additive is selected from the group consisting of biocompatible polymers; plasticizers (e.g., glycerol); stimulus-responsive agents; small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; antibodies and antigen binding fragments thereof; nucleic acids; nucleic acid analogs and derivatives; glycogens or other sugars; immunogens; antigens; an extract made from biological materials such as bacteria, plants, fungi, or animal cells; animal tissues; naturally occurring or synthetic compositions; and any combinations thereof. 75. The method of claim 73 or 74, wherein the additive is in a form selected from the group consisting of a particle, a fiber, a tube, a film, a gel, a mesh, a mat, a non-woven mat, a powder, and any combinations thereof. 76. The method of any of claims 73-75, wherein the additive comprises a silk material, e.g., silk particles, silk fibers, micro-sized silk fibers, unprocessed silk fibers, or any combinations thereof. 77. A method of delivering an active agent comprising applying or administering to a subject a composition comprising a silk-based material, the silk-based material encapsulating at least one lipid compartment with an active agent disposed therein, said silk-based material being permeable to the active agent such that the active agent is released through the silk-based material, at a pre-determined rate, upon application or administration of the composition to the subject. 78. The method of claim 77, wherein the active agent is released to an ambient surrounding. 79. The method of claim 77 or 78, wherein the active agent is released to at least one target cell of the subject. 80. The method of any of claims 77-79, wherein the active agent comprises a hydrophobic or lipophilic active agent. 81. The method of claim 80, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 82. The method of any of claims 77-81, wherein the silk-based material comprises an additive. 83. The method of any of claims 77-82, wherein the composition is applied or administered to the subject topically. 84. The method of claim 83, wherein the composition is applied on a skin of the subject. 85. The method of any of claims 77-82, wherein the composition is applied or administered to the subject orally. 86. A silk particle comprising at least two immiscible phases, a first immiscible phase comprising a silk-based material and a second immiscible phase comprising an active agent, wherein the first immiscible phase encapsulates the second immiscible phase and the second immiscible phase excludes a liposome. 87. The silk particle of claim 86, wherein the second immiscible phase comprises a lipid component. 88. The silk particle of claim 87, wherein the lipid component comprises oil. 89. The silk particle of any of claims 86-88, wherein the second immiscible phase forms a single compartment. 90. The silk particle of any of claims 86-89, wherein the second immiscible phase forms a plurality of compartments. 91. The silk particle of claim 89 or 90, wherein the size of the compartment or compartments ranges from about 1 μm to about 1000 μm, or from about 10 μm to about 500 μm. 92. The silk particle of any of claims 86-91, wherein the active agent present in the second immiscible phase comprises a hydrophobic or lipophilic molecule. 93. The silk particle of claim 92, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 94. The silk particle of any of claims 86-93, wherein the silk-based material comprises an additive. 95. The silk particle of claim 94, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 96. The silk particle of any of claims 86-95, wherein the second immiscible phase encapsulates a third immiscible phase. 97. The silk particle of any of claims 86-96, wherein the silk-based material is present in a form of a hydrogel. 98. The silk particle of any of claims 86-96, wherein the silk-based material is present in a dried state or lyophilized. 99. The silk particle of claim 98, wherein the lyophilized silk matrix is porous. 100. The silk particle of any of claims 86-99, wherein at least the silk-based material in the first immiscible phase is soluble in an aqueous solution. 101. The silk particle of any of claims 86-99, wherein beta-sheet content in the silk-based material is adjusted to an amount sufficient to enable the silk-based material to resist dissolution in an aqueous solution. 102. The silk particle of any of claims 86-101, wherein the size of the silk particle ranges from about 0.1 mm to about 10 mm, or from about 0.5 mm to about 5 mm. 103. A composition comprising a plurality of lipid compartments encapsulated in a silk-based material. 104. The composition of claim 103, wherein the size of the lipid compartments ranges from about 1 μm to about 1000 μm, or from about 10 μm to about 500 μm. 105. The composition of claim 103 or 104, wherein the volumetric ratio of the lipid compartments to the silk-based material ranges from about 1:1 to about 1:1000, from about 1:5 to about 1:500, or from about 1:10 to about 1:100. 106. The composition of any of claims 103-105, wherein the silk-based material comprises a film. 107. The composition of claim 106, wherein the silk-based material comprises an optical pattern. 108. The composition of claim 107, wherein the optical pattern comprises a hologram or an array of patterns that provides an optical functionality. 109. The composition of any of claims 103-108, wherein the silk-based material comprises a scaffold. 110. The composition of any of claims 103-109, wherein the lipid compartments further comprise an active agent. 111. The composition of claim 110, wherein the active agent comprises a hydrophobic or lipophilic molecule. 112. The composition of claim 111, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 113. The composition of any of claims 103-112, wherein the silk-based material comprises an additive. 114. The composition of claim 113, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 115. A composition comprising a collection of silk particles of any of claims 86-102. 116. The composition of claim 115, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, or any combinations thereof. 117. The composition of claim 115 or 116, wherein the composition is formulated for use in a pharmaceutical product. 118. The composition of claim 115 or 116, wherein the composition is formulated for use in a cosmetic product. 119. The composition of claim 115 or 116, wherein the composition is formulated for use in a food product. 120. A storage-stable composition comprising a silk particle of any of claims 86-102 or a composition of any of claims 103-119, where the active agent present in the second immiscible phase of the silk particle, or a hydrophobic or lipophilic molecule present in the lipid components retains at least about 30% of its original bioactivity when the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 121. The composition of claim 120, wherein the composition is maintained under exposure to light. 122. The composition of claim 120 or 121, wherein the composition is maintained at a relative humidity of at least about 10%. 123. The composition of any of claims 120-122, wherein the cross-linked silk matrix is in a dried-state. 124. A method of producing a silk particle comprising: a. providing or obtaining an emulsion of non-aqueous droplets dispersed in a silk solution undergoing a sol-gel transition (where the silk solution remains in a mixable state); and b. contacting a pre-determined volume of the emulsion with a non-aqueous phase, whereby the silk solution entraps at least one of the non-aqueous droplets and gels to form a silk particle dispersed in the non-aqueous phase. 125. The method of claim 124, wherein the sol-gel transition last for about at least 1 hour, or at least about 2 hours. 126. The method of claim 124 or 125, wherein the sol-gel transition of the silk solution is induced by sonication. 127. The method of claim 126, where the sonication is performed at an amplitude of about 5% to about 20%, or about 10% to about 15%. 128. The method of claim 126 or 127, wherein the sonication duration lasts for about 15 sec to about 60 sec, or from about 30 sec to about 45 sec. 129. The method of any of claims 124-128, wherein the silk solution has a concentration of about 1% (w/v) to about 15% (w/v), or about 2% (w/v) to about 7% (w/v). 130. The method of any of claims 124-129, further comprising adding an active agent into the silk fibroin solution undergoing a sol-gel transition. 131. The method of any of claims 124-130, wherein the non-aqueous droplets further comprise a hydrophobic or lipophilic molecule. 132. The method of claim 131, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 133. The method of any of claims 124-132, wherein the emulsion is produced by adding a non-aqueous, immiscible phase into the silk solution, thereby forming the non-aqueous droplets dispersed in the silk solution. 134. The method of any of claims 124-133, wherein the pre-determined volume of the emulsion is a volume corresponding to a desirable size of the silk particle. 135. The method of any of claims 124-134, further comprising isolating the silk particle from the non-aqueous phase. 136. The method of any of claims 124-135, further comprising freeze-drying the silk particle. 137. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the active agent retains at least about 30% of its original bioactivity when the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 138. The method of claim 137, wherein the composition is maintained for at least about 1 month. 139. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the silk-based material is permeable to said at least one active agent such that the active agent is released through the silk-based material into an ambient surrounding at a pre-determined rate. 140. The method of claim 139, wherein the pre-determined rate is controlled by adjusting an amount of beta-sheet conformation of silk fibroin present in the silk-based material, porosity of the silk-based material, or a combination thereof. 141. The method of claim 139 or 140, wherein the composition is maintained at about room temperature. 142. The method of any of claims 137-141, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, or any combinations thereof. 143. The method of any of claims 137-142, wherein the composition is lyophilized. 144. The method of any of claims 137-143, wherein the composition is maintained at a temperature of about 37° C. or greater. 145. The method of any of claims 137-144, wherein the composition is maintained under exposure to light. 146. The method of any of claims 137-145, wherein the composition is maintained at a relative humidity of at least about 10%. 147. The method of any of claims 137-146, wherein the active agent comprises a hydrophobic or lipophilic active agent. 148. The method of claim 147, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 149. The method of any of claims 137-148, wherein the silk-based material comprises an additive. 150. The method of claim 149, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 151. A method of delivering an active agent comprising applying or administering to a subject a composition comprising a silk-based material, the silk-based material encapsulating a lipid compartment with an active agent disposed therein, said silk-based material being permeable to the active agent such that the active agent is released through the silk-based material, at a pre-determined rate, upon application or administration of the composition to the subject. 152. The method of claim 151, wherein the active agent is released to an ambient surrounding. 153. The method of claim 151 or 152, wherein the active agent is released to at least one target cell of the subject. 154. The method of any of claims 151-153, wherein the active agent comprises a hydrophobic or lipophilic active agent. 155. The method of claim 154, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 156. The method of any of claims 151-155, wherein the silk-based material comprises an additive. 157. The method of claim 156, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 158. The method of any of claims 151-157, wherein the composition is applied or administered to the subject topically or orally. 159. The method of any of claims 151-158, wherein the composition is applied on skin of the subject.
Embodiments of various aspects described herein relates to compositions and methods for encapsulation and/or stabilization of oil, lipid, hydrophobic and/or lipophilic compounds in a silk-based material. The compositions described herein can be used in various applications, e.g., pharmaceutical, cosmetic, food, diagnostic, and tissue engineering applications.1. A silk particle comprising at least two immiscible phases, a first immiscible phase comprising a silk-based material and a second immiscible phase comprising an active agent, wherein the first immiscible phase encapsulates the second immiscible phase and the second immiscible phase excludes a liposome. 2. The silk particle of claim 1, wherein the second immiscible phase comprises a lipid component. 3. The silk particle of claim 2, wherein the lipid component comprises oil. 4. The silk particle of any of claims 1-3, wherein the second immiscible phase forms a single compartment. 5. The silk particle of any of claims 1-3, wherein the second immiscible phase forms a plurality of compartments. 6. The silk particle of claim 4 or 5, wherein the size of the compartment or compartments ranges from about 1 nm to about 1000 μm, or from about 5 nm to about 500 μm. 7. The silk particle of any of claims 1-6, wherein the active agent present in the second immiscible phase comprises a hydrophobic or lipophilic molecule. 8. The silk particle of claim 7, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), a small molecule, or any combinations thereof. 9. The silk particle of any of claims 1-8, wherein the silk-based material comprises an additive. 10. The silk particle of claim 9, wherein the additive is selected from the group consisting of biocompatible polymers; plasticizers (e.g., glycerol); stimulus-responsive agents; active agents, small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; antibodies and antigen binding fragments thereof; nucleic acids; nucleic acid analogs and derivatives; glycogens or other sugars; immunogens; antigens; an extract made from biological materials such as bacteria, plants, fungi, or animal cells; animal tissues; naturally occurring or synthetic compositions; and any combinations thereof. 11. The silk particle of claim 9 or 10, wherein the additive is in a form of a particle (e.g., a nanoparticle or microparticle, including a plasmonic particle), a fiber, a tube, powder or any combinations thereof. 12. The silk particle of any of claims 9-11, wherein the additive comprises a silk material, e.g., silk particles, silk fibers, micro-sized silk fibers, unprocessed silk fibers, and any combinations thereof. 13. The silk particle of any of claims 1-12, wherein the second immiscible phase encapsulates a third immiscible phase. 14. The silk particle of any of claims 1-13, wherein the silk-based material is present in a form of a hydrogel. 15. The silk particle of any of claims 1-14, wherein the silk-based material is present in a dried state or lyophilized. 16. The silk particle of claim 15, wherein the lyophilized silk matrix is porous. 17. The silk particle of any of claims 1-16, wherein the silk-based material in the first immiscible phase is soluble in an aqueous solution. 18. The silk particle of any of claims 1-17, wherein beta-sheet content in the silk-based material is adjusted to an amount sufficient to enable the silk-based material to resist dissolution in an aqueous solution. 19. The silk particle of any of claims 1-18, wherein the size of the silk particle ranges from about 10 nm to about 10 mm, or from about 50 nm to about 5 mm. 20. A composition comprising a plurality of lipid compartments encapsulated in a silk-based material. 21. The composition of claim 20, wherein the size of the lipid compartments ranges from about 1 nm to about 1000 μm, or from about 5 nm to about 500 μm. 22. The composition of claim 20 or 21, wherein the volumetric ratio of the lipid compartments to the silk-based material ranges from about 1000:1 to about 1:1000, from about 500:1 to about 1:500, or from about 100:1 to about 1:100. 23. The composition of any of claims 20-22, wherein the silk-based material is in a form selected from the group consisting of a film, a sheet, a gel or hydrogel, a mesh, a mat, a non-woven mat, a fabric, a scaffold, a tube, a slab or block, a fiber, a particle, powder, a 3-dimensional construct, an implant, a foam or a sponge, a needle, a lyophilized material, a porous material, a non-porous material, and any combinations thereof. 24. The composition of any of claims 20-23, wherein the silk-based material comprises a film. 25. The composition of any of claims 20-24, wherein the silk-based material comprises a scaffold. 26. The composition of any of claims 20-25, wherein the silk-based material comprises an optical pattern. 27. The composition of claim 26, wherein the optical pattern comprises a hologram or an array of patterns that provides an optical functionality. 28. The composition of any of claims 20-27, wherein the lipid compartments further comprise an active agent. 29. The composition of claim 20-28, wherein the active agent comprises a hydrophobic or lipophilic molecule. 30. The composition of claim 29, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), a small molecule, or any combinations thereof. 31. The composition of any of claims 20-30, wherein the silk-based material comprises an additive. 32. The composition of claim 31, wherein the additive is selected from the group consisting of biocompatible polymers; plasticizers (e.g., glycerol); stimulus-responsive agents; small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; antibodies and antigen binding fragments thereof; nucleic acids; nucleic acid analogs and derivatives; glycogens or other sugars; immunogens; antigens; an extract made from biological materials such as bacteria, plants, fungi, or animal cells; animal tissues; naturally occurring or synthetic compositions; and any combinations thereof. 33. The composition of claim 31 or 32, wherein the additive is in a form selected from the group consisting of a particle, a fiber, a tube, a film, a gel, a mesh, a mat, a non-woven mat, a powder, and any combinations thereof. 34. The composition of any of claims 31-33, wherein the additive comprises a silk material, e.g., silk particles, silk fibers, micro-sized silk fibers, unprocessed silk fibers, and any combinations thereof. 35. A composition comprising a collection of silk particles of any of claims 1-19. 36. The composition of claim 35, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, a scaffold, or any combinations thereof. 37. The composition of claim 35 or 36, wherein the composition is formulated for use in a pharmaceutical product. 38. The composition of claim 35 or 36, wherein the composition is formulated for use in a cosmetic product. 39. The composition of claim 35 or 36, wherein the composition is formulated for use in a personal care product. 40. The composition of claim 35 or 36, wherein the composition is formulated for use in a food product. 41. A storage-stable composition comprising a silk particle of any of claims 1-19 or a composition of any of claims 20-40, wherein the active agent present in the second immiscible phase of the silk particle, or a hydrophobic or lipophilic molecule present in the lipid components retains at least about 30% of its original bioactivity after the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 42. The composition of claim 41, wherein the composition is maintained under exposure to light. 43. The composition of claim 41 or 42, wherein the composition is maintained at a relative humidity of at least about 10%. 44. The composition of any of claims 41-43, wherein the silk-based material of the silk particle or the composition is in a dried-state. 45. A method of producing a silk particle comprising: a. providing an emulsion of non-aqueous droplets dispersed in a silk solution undergoing a sol-gel transition (where the silk solution remains in a mixable state); and b. contacting a pre-determined volume of the emulsion with a non-aqueous phase, whereby the silk solution forms in the non-aqueous phase a silk particle entrapping at least one of the non-aqueous droplets therein. 46. The method of claim 45, wherein the sol-gel transition last for about at least 1 hour, or at least about 2 hours. 47. The method of claim 45 or 46, wherein the sol-gel transition of the silk solution is induced by sonication. 48. The method of claim 47, where the sonication is performed at an amplitude of about 5% to about 20%, or about 10% to about 15%. 49. The method of claim 47 or 48, wherein the sonication duration lasts for about 15 sec to about 60 sec, or from about 30 sec to about 45 sec. 50. The method of any of claims 45-49, wherein the silk solution has a concentration of about 1% (w/v) to about 15% (w/v), or about 2% (w/v) to about 7% (w/v). 51. The method of any of claims 45-50, further comprising adding an active agent into the silk fibroin solution undergoing a sol-gel transition. 52. The method of any of claims 45-51, wherein the non-aqueous droplets further comprise a hydrophobic or lipophilic molecule. 53. The method of claim 52, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), a small molecule, or any combinations thereof. 54. The method of any of claims 45-53, wherein the emulsion is produced by adding a non-aqueous, immiscible phase into the silk solution, thereby forming the non-aqueous droplets dispersed in the silk solution. 55. The method of any of claims 45-54, wherein the pre-determined volume of the emulsion substantially corresponds to a desirable size of the silk particle. 56. The method of any of claims 45-55, further comprising isolating the silk particle from the non-aqueous phase. 57. The method of any of claims 45-56, further comprising subjecting the silk particle to a post-treatment. 58. The method of claim 57, wherein the post-treatment further induces a conformational change in silk fibroin in the particle. 59. The method of claim 58, wherein said inducing conformational change comprises one or more of lyophilization or freeze-drying, water annealing, water vapor annealing, alcohol immersion, sonication, shear stress, electrogelation, pH reduction, salt addition, air-drying, electrospinning, stretching, or any combination thereof. 60. The method of any of claims 57-59, wherein the post-treatment comprises freeze-drying the silk particle. 61. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated in a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the active agent retains at least about 30% of its original bioactivity after the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 62. The method of claim 61, wherein the composition is maintained for at least about 1 month. 63. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated in a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the silk-based material is permeable to said at least one active agent such that the active agent is released through the silk-based material into an ambient surrounding at a pre-determined rate. 64. The method of claim 63, wherein the pre-determined rate is controlled by adjusting an amount of beta-sheet conformation of silk fibroin present in the silk-based material, porosity of the silk-based material, or a combination thereof. 65. The method of claim 63 or 64, wherein the composition is maintained at about room temperature. 66. The method of any of claims 61-65, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, or any combinations thereof. 67. The method of any of claims 61-66, wherein the composition is lyophilized. 68. The method of any of claims 61-67, wherein the composition is maintained at a temperature of about 37° C. or greater. 69. The method of any of claims 61-68, wherein the composition is maintained under exposure to light. 70. The method of any of claims 61-69, wherein the composition is maintained at a relative humidity of at least about 10%. 71. The method of any of claims 61-70, wherein the active agent comprises a hydrophobic or lipophilic active agent. 72. The method of claim 71, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 73. The method of any of claims 61-72, wherein the silk-based material comprises an additive. 74. The method of claim 73, wherein the additive is selected from the group consisting of biocompatible polymers; plasticizers (e.g., glycerol); stimulus-responsive agents; small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; antibodies and antigen binding fragments thereof; nucleic acids; nucleic acid analogs and derivatives; glycogens or other sugars; immunogens; antigens; an extract made from biological materials such as bacteria, plants, fungi, or animal cells; animal tissues; naturally occurring or synthetic compositions; and any combinations thereof. 75. The method of claim 73 or 74, wherein the additive is in a form selected from the group consisting of a particle, a fiber, a tube, a film, a gel, a mesh, a mat, a non-woven mat, a powder, and any combinations thereof. 76. The method of any of claims 73-75, wherein the additive comprises a silk material, e.g., silk particles, silk fibers, micro-sized silk fibers, unprocessed silk fibers, or any combinations thereof. 77. A method of delivering an active agent comprising applying or administering to a subject a composition comprising a silk-based material, the silk-based material encapsulating at least one lipid compartment with an active agent disposed therein, said silk-based material being permeable to the active agent such that the active agent is released through the silk-based material, at a pre-determined rate, upon application or administration of the composition to the subject. 78. The method of claim 77, wherein the active agent is released to an ambient surrounding. 79. The method of claim 77 or 78, wherein the active agent is released to at least one target cell of the subject. 80. The method of any of claims 77-79, wherein the active agent comprises a hydrophobic or lipophilic active agent. 81. The method of claim 80, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 82. The method of any of claims 77-81, wherein the silk-based material comprises an additive. 83. The method of any of claims 77-82, wherein the composition is applied or administered to the subject topically. 84. The method of claim 83, wherein the composition is applied on a skin of the subject. 85. The method of any of claims 77-82, wherein the composition is applied or administered to the subject orally. 86. A silk particle comprising at least two immiscible phases, a first immiscible phase comprising a silk-based material and a second immiscible phase comprising an active agent, wherein the first immiscible phase encapsulates the second immiscible phase and the second immiscible phase excludes a liposome. 87. The silk particle of claim 86, wherein the second immiscible phase comprises a lipid component. 88. The silk particle of claim 87, wherein the lipid component comprises oil. 89. The silk particle of any of claims 86-88, wherein the second immiscible phase forms a single compartment. 90. The silk particle of any of claims 86-89, wherein the second immiscible phase forms a plurality of compartments. 91. The silk particle of claim 89 or 90, wherein the size of the compartment or compartments ranges from about 1 μm to about 1000 μm, or from about 10 μm to about 500 μm. 92. The silk particle of any of claims 86-91, wherein the active agent present in the second immiscible phase comprises a hydrophobic or lipophilic molecule. 93. The silk particle of claim 92, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 94. The silk particle of any of claims 86-93, wherein the silk-based material comprises an additive. 95. The silk particle of claim 94, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 96. The silk particle of any of claims 86-95, wherein the second immiscible phase encapsulates a third immiscible phase. 97. The silk particle of any of claims 86-96, wherein the silk-based material is present in a form of a hydrogel. 98. The silk particle of any of claims 86-96, wherein the silk-based material is present in a dried state or lyophilized. 99. The silk particle of claim 98, wherein the lyophilized silk matrix is porous. 100. The silk particle of any of claims 86-99, wherein at least the silk-based material in the first immiscible phase is soluble in an aqueous solution. 101. The silk particle of any of claims 86-99, wherein beta-sheet content in the silk-based material is adjusted to an amount sufficient to enable the silk-based material to resist dissolution in an aqueous solution. 102. The silk particle of any of claims 86-101, wherein the size of the silk particle ranges from about 0.1 mm to about 10 mm, or from about 0.5 mm to about 5 mm. 103. A composition comprising a plurality of lipid compartments encapsulated in a silk-based material. 104. The composition of claim 103, wherein the size of the lipid compartments ranges from about 1 μm to about 1000 μm, or from about 10 μm to about 500 μm. 105. The composition of claim 103 or 104, wherein the volumetric ratio of the lipid compartments to the silk-based material ranges from about 1:1 to about 1:1000, from about 1:5 to about 1:500, or from about 1:10 to about 1:100. 106. The composition of any of claims 103-105, wherein the silk-based material comprises a film. 107. The composition of claim 106, wherein the silk-based material comprises an optical pattern. 108. The composition of claim 107, wherein the optical pattern comprises a hologram or an array of patterns that provides an optical functionality. 109. The composition of any of claims 103-108, wherein the silk-based material comprises a scaffold. 110. The composition of any of claims 103-109, wherein the lipid compartments further comprise an active agent. 111. The composition of claim 110, wherein the active agent comprises a hydrophobic or lipophilic molecule. 112. The composition of claim 111, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 113. The composition of any of claims 103-112, wherein the silk-based material comprises an additive. 114. The composition of claim 113, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 115. A composition comprising a collection of silk particles of any of claims 86-102. 116. The composition of claim 115, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, or any combinations thereof. 117. The composition of claim 115 or 116, wherein the composition is formulated for use in a pharmaceutical product. 118. The composition of claim 115 or 116, wherein the composition is formulated for use in a cosmetic product. 119. The composition of claim 115 or 116, wherein the composition is formulated for use in a food product. 120. A storage-stable composition comprising a silk particle of any of claims 86-102 or a composition of any of claims 103-119, where the active agent present in the second immiscible phase of the silk particle, or a hydrophobic or lipophilic molecule present in the lipid components retains at least about 30% of its original bioactivity when the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 121. The composition of claim 120, wherein the composition is maintained under exposure to light. 122. The composition of claim 120 or 121, wherein the composition is maintained at a relative humidity of at least about 10%. 123. The composition of any of claims 120-122, wherein the cross-linked silk matrix is in a dried-state. 124. A method of producing a silk particle comprising: a. providing or obtaining an emulsion of non-aqueous droplets dispersed in a silk solution undergoing a sol-gel transition (where the silk solution remains in a mixable state); and b. contacting a pre-determined volume of the emulsion with a non-aqueous phase, whereby the silk solution entraps at least one of the non-aqueous droplets and gels to form a silk particle dispersed in the non-aqueous phase. 125. The method of claim 124, wherein the sol-gel transition last for about at least 1 hour, or at least about 2 hours. 126. The method of claim 124 or 125, wherein the sol-gel transition of the silk solution is induced by sonication. 127. The method of claim 126, where the sonication is performed at an amplitude of about 5% to about 20%, or about 10% to about 15%. 128. The method of claim 126 or 127, wherein the sonication duration lasts for about 15 sec to about 60 sec, or from about 30 sec to about 45 sec. 129. The method of any of claims 124-128, wherein the silk solution has a concentration of about 1% (w/v) to about 15% (w/v), or about 2% (w/v) to about 7% (w/v). 130. The method of any of claims 124-129, further comprising adding an active agent into the silk fibroin solution undergoing a sol-gel transition. 131. The method of any of claims 124-130, wherein the non-aqueous droplets further comprise a hydrophobic or lipophilic molecule. 132. The method of claim 131, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 133. The method of any of claims 124-132, wherein the emulsion is produced by adding a non-aqueous, immiscible phase into the silk solution, thereby forming the non-aqueous droplets dispersed in the silk solution. 134. The method of any of claims 124-133, wherein the pre-determined volume of the emulsion is a volume corresponding to a desirable size of the silk particle. 135. The method of any of claims 124-134, further comprising isolating the silk particle from the non-aqueous phase. 136. The method of any of claims 124-135, further comprising freeze-drying the silk particle. 137. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the active agent retains at least about 30% of its original bioactivity when the composition is (a) subjected to at least one freeze-thaw cycle, or (b) maintained for at least about 24 hours at a temperature of about room temperature or above, or (c) both (a) and (b). 138. The method of claim 137, wherein the composition is maintained for at least about 1 month. 139. A method comprising a step of: maintaining a composition, wherein the composition comprises at least one lipid compartment encapsulated a silk-based material and at least one active agent distributed in said at least one lipid compartment, and wherein the silk-based material is permeable to said at least one active agent such that the active agent is released through the silk-based material into an ambient surrounding at a pre-determined rate. 140. The method of claim 139, wherein the pre-determined rate is controlled by adjusting an amount of beta-sheet conformation of silk fibroin present in the silk-based material, porosity of the silk-based material, or a combination thereof. 141. The method of claim 139 or 140, wherein the composition is maintained at about room temperature. 142. The method of any of claims 137-141, wherein the composition is an emulsion, a colloid, a cream, a gel, a lotion, a paste, an ointment, a liniment, a balm, a liquid, a solid, a film, a sheet, a fabric, a mesh, a sponge, an aerosol, powder, or any combinations thereof. 143. The method of any of claims 137-142, wherein the composition is lyophilized. 144. The method of any of claims 137-143, wherein the composition is maintained at a temperature of about 37° C. or greater. 145. The method of any of claims 137-144, wherein the composition is maintained under exposure to light. 146. The method of any of claims 137-145, wherein the composition is maintained at a relative humidity of at least about 10%. 147. The method of any of claims 137-146, wherein the active agent comprises a hydrophobic or lipophilic active agent. 148. The method of claim 147, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 149. The method of any of claims 137-148, wherein the silk-based material comprises an additive. 150. The method of claim 149, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 151. A method of delivering an active agent comprising applying or administering to a subject a composition comprising a silk-based material, the silk-based material encapsulating a lipid compartment with an active agent disposed therein, said silk-based material being permeable to the active agent such that the active agent is released through the silk-based material, at a pre-determined rate, upon application or administration of the composition to the subject. 152. The method of claim 151, wherein the active agent is released to an ambient surrounding. 153. The method of claim 151 or 152, wherein the active agent is released to at least one target cell of the subject. 154. The method of any of claims 151-153, wherein the active agent comprises a hydrophobic or lipophilic active agent. 155. The method of claim 154, wherein the hydrophobic or lipophilic molecule comprises a therapeutic agent, a nutraceutical agent, a cosmetic agent, a coloring agent, a probiotic agent, a dye, an aromatic compound, an aliphatic compound (e.g., alkane, alkene, alkyne, cyclo-alkane, cyclo-alkene, and cyclo-alkyne), or any combinations thereof. 156. The method of any of claims 151-155, wherein the silk-based material comprises an additive. 157. The method of claim 156, wherein the additive comprises a biopolymer, an active agent, a plasmonic particle, glycerol, and any combinations thereof. 158. The method of any of claims 151-157, wherein the composition is applied or administered to the subject topically or orally. 159. The method of any of claims 151-158, wherein the composition is applied on skin of the subject.
1,600
569
15,064,792
1,641
Antibody/signal-generating moiety conjugates are disclosed that include an antibody covalently linked to a signal-generating moiety through a heterobifunctional polyalkyleneglycol linker. The disclosed conjugates show exceptional signal-generation in immunohistochemical and in situ hybridization assays on tissue sections and cytology samples. In one embodiment, enzyme-metallographic detection of nucleic acid sequences with hapten-labeled probes can be accomplished using the disclosed conjugates as a primary antibody without amplification.
1. An antibody-signal-generating moiety conjugate comprising an antibody covalently linked to a signal-generating moiety through a heterobifunctional PEG linker. 2. The conjugate of claim 1, wherein a thiol-reactive group of the linker is covalently linked to the antibody and an amine-reactive group of the heterobifunctional linker is covalently linked to the signal-generating moiety. 3. The conjugate of claim 2, wherein the thiol-reactive group of the linker is covalently linked to a cysteine residue of the antibody. 4. The conjugate of claim 2, wherein the thiol-reactive group of the linker is covalently linked to a thiol group that is introduced to the antibody. 5. The conjugate of claim 1, wherein an aldehyde-reactive group of the linker is covalently linked to the antibody and a thiol-reactive group of the linker is covalently linked to the signal-generating moiety. 6. The conjugate of claim 5, wherein the aldehyde-reactive group of the linker is covalently linked to an aldehyde formed on a glycosylated portion of the antibody. 7. The conjugate of claim 1, wherein the linker has the formula: wherein n=1 to 50; or, wherein m is from 1 to 50. 8. The conjugate of claim 7, wherein the conjugate has the formula: wherein Ab is an antibody, SM is a signal-generating moiety, n=1 to 50 and s=1 to 10. 9. The conjugate of claim 8, wherein the signal-generating moiety comprises an enzyme. 10. The conjugate of claim 9, wherein the enzyme comprises horseradish peroxidase or alkaline phosphatase. 11. The conjugate of claim 8, wherein s.=2 to 6. 12. The conjugate of claim 8, wherein n=4 to 12. 13. The conjugate of claim 7, wherein the conjugate has the formula: wherein Ab is an antibody, SM is a signal-generating moiety, m=1 to 50 and t=1 to 10. 14. The conjugate of claim 13, wherein the signal-generating moiety comprises an enzyme. 15. The conjugate of claim 14, wherein the enzyme comprises horseradish peroxidase or alkaline phosphatase. 16. The conjugate of claim 13, wherein t=2 to 6. 17. The conjugate of claim 13, wherein m=4 to 12.
Antibody/signal-generating moiety conjugates are disclosed that include an antibody covalently linked to a signal-generating moiety through a heterobifunctional polyalkyleneglycol linker. The disclosed conjugates show exceptional signal-generation in immunohistochemical and in situ hybridization assays on tissue sections and cytology samples. In one embodiment, enzyme-metallographic detection of nucleic acid sequences with hapten-labeled probes can be accomplished using the disclosed conjugates as a primary antibody without amplification.1. An antibody-signal-generating moiety conjugate comprising an antibody covalently linked to a signal-generating moiety through a heterobifunctional PEG linker. 2. The conjugate of claim 1, wherein a thiol-reactive group of the linker is covalently linked to the antibody and an amine-reactive group of the heterobifunctional linker is covalently linked to the signal-generating moiety. 3. The conjugate of claim 2, wherein the thiol-reactive group of the linker is covalently linked to a cysteine residue of the antibody. 4. The conjugate of claim 2, wherein the thiol-reactive group of the linker is covalently linked to a thiol group that is introduced to the antibody. 5. The conjugate of claim 1, wherein an aldehyde-reactive group of the linker is covalently linked to the antibody and a thiol-reactive group of the linker is covalently linked to the signal-generating moiety. 6. The conjugate of claim 5, wherein the aldehyde-reactive group of the linker is covalently linked to an aldehyde formed on a glycosylated portion of the antibody. 7. The conjugate of claim 1, wherein the linker has the formula: wherein n=1 to 50; or, wherein m is from 1 to 50. 8. The conjugate of claim 7, wherein the conjugate has the formula: wherein Ab is an antibody, SM is a signal-generating moiety, n=1 to 50 and s=1 to 10. 9. The conjugate of claim 8, wherein the signal-generating moiety comprises an enzyme. 10. The conjugate of claim 9, wherein the enzyme comprises horseradish peroxidase or alkaline phosphatase. 11. The conjugate of claim 8, wherein s.=2 to 6. 12. The conjugate of claim 8, wherein n=4 to 12. 13. The conjugate of claim 7, wherein the conjugate has the formula: wherein Ab is an antibody, SM is a signal-generating moiety, m=1 to 50 and t=1 to 10. 14. The conjugate of claim 13, wherein the signal-generating moiety comprises an enzyme. 15. The conjugate of claim 14, wherein the enzyme comprises horseradish peroxidase or alkaline phosphatase. 16. The conjugate of claim 13, wherein t=2 to 6. 17. The conjugate of claim 13, wherein m=4 to 12.
1,600
570
14,122,872
1,628
The present invention relates to a process for the preparation of a compound of formula (I) said process comprising the steps of: a) reacting a compound of formula (II), with an acylating or a silylating agent to produce a compound of formula (III), wherein P 1 and P 2 are each independently a protecting group selected from R 2− Si—R 3 R 4 , or R 1 CO—, wherein R 1 is a group selected from C 1-6 alkyl or C 3-6 cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; R 2 , R 3 and R 4 are each independently a group selected from C 1-6 alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; b) reacting the compound of formula (III) in the presence of palladium acetate or a derivative thereof to produce compound of formula (IV); and c) reacting the compound of formula (IV) with a reducing agent to produce compound of formula (I).
1. A process for the preparation of a compound of formula (I) said process comprising the steps of a) reacting a compound of formula (II), with an acylating or a silylating agent to produce a compound of formula (III), wherein P1 and P2 are each independently a protecting group selected from R2−Si—R3R4, or R1CO—, wherein R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) reacting the compound of formula (III) in the presence of palladium acetate or a derivative thereof to produce compound of formula (IV); and c) reacting the compound of formula (IV) with a reducing agent to produce compound of formula (I). 2. The process according to claim 1, wherein P1 is R1CO—. 3. The process according to claim 1, wherein P1 is R2−Si—R3R4. 4. The process according to claim 3, wherein P2 is R2−Si—R3R4. 5. The process according to claim 1, wherein P2 is R1CO—. 6. The process according to claim 5, wherein step (a) comprises the steps of (a1) protecting the hydroxyl of compound of formula (II) with a silylating agent to produce a compound of formula (IIa), wherein P1 is R2−Si—R3R4; and (a2) protecting the ketone of compound of formula (IIa) in the presence of an acylating agent to produce compound of formula (III). 7. The process according to claim 1, wherein the acylating agent is C2-6alkenylC1-6alkanoate or C2-6alkenylC3-6cycloalkanoate. 8. The process according to claim 1, wherein the silylating agent is selected from the group comprising C1-6alkylsilylchloride, C1-6alkylsilyltriflate, phenylsilyl chloride, phenylsilyltriflate, C1-6alkylphenylsilylchloride, C1-6alkylphenylsilyltriflate, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl. 9. The process according to claim 1, wherein step (b) is performed in the presence of a C1-6alkylene carbonate and an organotin compound. 10. The process according to claim 1, wherein said palladium acetate is present in stoichiometric amounts. 11. The process according to claim 1, wherein said reaction is performed with palladium acetate present in catalytic or sub-stoichiometric amounts, preferably the reaction is performed in an oxygen atmosphere. 12. The process according to claim 1, wherein the reducing agent in step (c) is selected from the group of metal hydride compounds. 13. The process according to claim 12, wherein the metal hydride compound is selected from the group comprising NaBH4/CeCl3, LiAlH4, NaBH4, NaBH(OAc)3, and ZnBH4. 14. A process for the preparation of estetrol, said process comprising preparing a compound of formula (I) by a process, said process comprising the steps of a) reacting a compound of formula (II), with an acylating or a silylating agent to produce a compound of formula (III), wherein P1 and P2 are each independently a protecting group selected from R2−Si—R3R4, or R1CO—, wherein R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) reacting the compound of formula (III) in the presence of palladium acetate or a derivative thereof to produce compound of formula (IV); and c) reacting the compound of formula (IV) with a reducing agent to produce compound of formula (I), and further reacting compound of formula (I) to produce the estetrol. 15. (canceled) 16. The process according to claim 14, wherein P1 is R1CO—. 17. The process according to claim 14, wherein P1 is R2−Si—R3R4. 18. The process according to claim 17, wherein P2 is R2−Si—R3R4. 19. The process according to claim 14, wherein P2 is R1CO—. 20. The process according to claim 19, wherein step (a) comprises the steps of (a1) protecting the hydroxyl of compound of formula (II) with a silylating agent to produce a compound of formula (IIa), wherein P1 is R2−Si—R3R4; and (a2) protecting the ketone of compound of formula (IIa) in the presence of an acylating agent to produce compound of formula (III). 21. The process according to claim 14, wherein the acylating agent is C2-6alkenylC1-6alkanoate or C2-6alkenylC3-6cycloalkanoate.
The present invention relates to a process for the preparation of a compound of formula (I) said process comprising the steps of: a) reacting a compound of formula (II), with an acylating or a silylating agent to produce a compound of formula (III), wherein P 1 and P 2 are each independently a protecting group selected from R 2− Si—R 3 R 4 , or R 1 CO—, wherein R 1 is a group selected from C 1-6 alkyl or C 3-6 cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; R 2 , R 3 and R 4 are each independently a group selected from C 1-6 alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; b) reacting the compound of formula (III) in the presence of palladium acetate or a derivative thereof to produce compound of formula (IV); and c) reacting the compound of formula (IV) with a reducing agent to produce compound of formula (I).1. A process for the preparation of a compound of formula (I) said process comprising the steps of a) reacting a compound of formula (II), with an acylating or a silylating agent to produce a compound of formula (III), wherein P1 and P2 are each independently a protecting group selected from R2−Si—R3R4, or R1CO—, wherein R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) reacting the compound of formula (III) in the presence of palladium acetate or a derivative thereof to produce compound of formula (IV); and c) reacting the compound of formula (IV) with a reducing agent to produce compound of formula (I). 2. The process according to claim 1, wherein P1 is R1CO—. 3. The process according to claim 1, wherein P1 is R2−Si—R3R4. 4. The process according to claim 3, wherein P2 is R2−Si—R3R4. 5. The process according to claim 1, wherein P2 is R1CO—. 6. The process according to claim 5, wherein step (a) comprises the steps of (a1) protecting the hydroxyl of compound of formula (II) with a silylating agent to produce a compound of formula (IIa), wherein P1 is R2−Si—R3R4; and (a2) protecting the ketone of compound of formula (IIa) in the presence of an acylating agent to produce compound of formula (III). 7. The process according to claim 1, wherein the acylating agent is C2-6alkenylC1-6alkanoate or C2-6alkenylC3-6cycloalkanoate. 8. The process according to claim 1, wherein the silylating agent is selected from the group comprising C1-6alkylsilylchloride, C1-6alkylsilyltriflate, phenylsilyl chloride, phenylsilyltriflate, C1-6alkylphenylsilylchloride, C1-6alkylphenylsilyltriflate, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl. 9. The process according to claim 1, wherein step (b) is performed in the presence of a C1-6alkylene carbonate and an organotin compound. 10. The process according to claim 1, wherein said palladium acetate is present in stoichiometric amounts. 11. The process according to claim 1, wherein said reaction is performed with palladium acetate present in catalytic or sub-stoichiometric amounts, preferably the reaction is performed in an oxygen atmosphere. 12. The process according to claim 1, wherein the reducing agent in step (c) is selected from the group of metal hydride compounds. 13. The process according to claim 12, wherein the metal hydride compound is selected from the group comprising NaBH4/CeCl3, LiAlH4, NaBH4, NaBH(OAc)3, and ZnBH4. 14. A process for the preparation of estetrol, said process comprising preparing a compound of formula (I) by a process, said process comprising the steps of a) reacting a compound of formula (II), with an acylating or a silylating agent to produce a compound of formula (III), wherein P1 and P2 are each independently a protecting group selected from R2−Si—R3R4, or R1CO—, wherein R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) reacting the compound of formula (III) in the presence of palladium acetate or a derivative thereof to produce compound of formula (IV); and c) reacting the compound of formula (IV) with a reducing agent to produce compound of formula (I), and further reacting compound of formula (I) to produce the estetrol. 15. (canceled) 16. The process according to claim 14, wherein P1 is R1CO—. 17. The process according to claim 14, wherein P1 is R2−Si—R3R4. 18. The process according to claim 17, wherein P2 is R2−Si—R3R4. 19. The process according to claim 14, wherein P2 is R1CO—. 20. The process according to claim 19, wherein step (a) comprises the steps of (a1) protecting the hydroxyl of compound of formula (II) with a silylating agent to produce a compound of formula (IIa), wherein P1 is R2−Si—R3R4; and (a2) protecting the ketone of compound of formula (IIa) in the presence of an acylating agent to produce compound of formula (III). 21. The process according to claim 14, wherein the acylating agent is C2-6alkenylC1-6alkanoate or C2-6alkenylC3-6cycloalkanoate.
1,600
571
15,008,646
1,623
The invention relates to a novel drug combination therapy useful in the treatment of dementia comprising administering a 1-aminocyclohexane derivative such as memantine and an acetylcholinesterase inhibitor (AChEI) such as donepezil.
1. A method for treating Alzheimer's disease in a subject in need thereof, comprising administration of a 1-aminocyclohexane derivative selected from memantine, and salts thereof, and an acetylcholinesterase inhibitor (AChEI) selected from donepezil, and salts thereof, wherein the combined treatment with the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) provides cognitive improvement. 2. The method of claim 1, wherein the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are administered conjointly. 3. The method of claim 2, wherein the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are administered in a single formulation. 4. The method of claim 1, wherein the dosages for each of the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are in the range of 1 to 200 mg per day. 5. The method of claim 4, wherein the dose for the 1-aminocyclohexane derivative is in the range of 10 to 40 mg per day and the dose for the acetylcholinesterase inhibitor (AChEI) is in the range of 5 to 24 mg per day. 6. The method of claim 1, wherein the cognitive improvement is measured by at least one clinical assessment selected from the group consisting of Severe Impairment Battery (SIB) Test, AD Cooperative Study-Activities of Daily Living (ADCS-ADL) Inventory and Clinician's Interview-Based Impression of Change Plus Version (CIBIC-plus). 7. A pharmaceutical composition comprising (i) a 1-aminocyclohexane derivative selected from memantine and salts thereof, (ii) an acetylcholinesterase inhibitor (AChEI) selected from donepezil, and salts thereof, and, optionally, (iii) a pharmaceutically acceptable carrier or excipient, wherein the 1-aminocyclohexane derivative and acetylcholinesterase inhibitor (AChEI) are present at therapeutically effective dosages. 8. The pharmaceutical composition of claim 7, wherein the dosages for each of the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are in the range of 1 to 200 mg. 9. The pharmaceutical composition of claim 8, wherein the dose for the 1-aminocyclohexane derivative is in the range of 10 to 40 mg and the dose for the acetylcholinesterase inhibitor (AChEI) is in the range of 5 to 24 mg. 10. The pharmaceutical composition of claim 7, which is a solid dosage form for oral administration. 11. The solid dosage form of claim 10, wherein the 1-aminocyclohexane derivative is present in an amount which is in the range of 10 to 40 mg and the acetylcholinesterase inhibitor (AChEI) is present in an amount which is in the range of 5 to 24 mg.
The invention relates to a novel drug combination therapy useful in the treatment of dementia comprising administering a 1-aminocyclohexane derivative such as memantine and an acetylcholinesterase inhibitor (AChEI) such as donepezil.1. A method for treating Alzheimer's disease in a subject in need thereof, comprising administration of a 1-aminocyclohexane derivative selected from memantine, and salts thereof, and an acetylcholinesterase inhibitor (AChEI) selected from donepezil, and salts thereof, wherein the combined treatment with the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) provides cognitive improvement. 2. The method of claim 1, wherein the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are administered conjointly. 3. The method of claim 2, wherein the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are administered in a single formulation. 4. The method of claim 1, wherein the dosages for each of the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are in the range of 1 to 200 mg per day. 5. The method of claim 4, wherein the dose for the 1-aminocyclohexane derivative is in the range of 10 to 40 mg per day and the dose for the acetylcholinesterase inhibitor (AChEI) is in the range of 5 to 24 mg per day. 6. The method of claim 1, wherein the cognitive improvement is measured by at least one clinical assessment selected from the group consisting of Severe Impairment Battery (SIB) Test, AD Cooperative Study-Activities of Daily Living (ADCS-ADL) Inventory and Clinician's Interview-Based Impression of Change Plus Version (CIBIC-plus). 7. A pharmaceutical composition comprising (i) a 1-aminocyclohexane derivative selected from memantine and salts thereof, (ii) an acetylcholinesterase inhibitor (AChEI) selected from donepezil, and salts thereof, and, optionally, (iii) a pharmaceutically acceptable carrier or excipient, wherein the 1-aminocyclohexane derivative and acetylcholinesterase inhibitor (AChEI) are present at therapeutically effective dosages. 8. The pharmaceutical composition of claim 7, wherein the dosages for each of the 1-aminocyclohexane derivative and the acetylcholinesterase inhibitor (AChEI) are in the range of 1 to 200 mg. 9. The pharmaceutical composition of claim 8, wherein the dose for the 1-aminocyclohexane derivative is in the range of 10 to 40 mg and the dose for the acetylcholinesterase inhibitor (AChEI) is in the range of 5 to 24 mg. 10. The pharmaceutical composition of claim 7, which is a solid dosage form for oral administration. 11. The solid dosage form of claim 10, wherein the 1-aminocyclohexane derivative is present in an amount which is in the range of 10 to 40 mg and the acetylcholinesterase inhibitor (AChEI) is present in an amount which is in the range of 5 to 24 mg.
1,600
572
15,129,811
1,646
The present invention relates to a therapeutic agent for nail and scalp psoriasis comprising an IL-17 Receptor A (IL-17RA or IL-17R) antigen binding proteins, such as monoclonal antibodies that bind IL-17RA, and method of using the same.
1. A method for treating nail or scalp psoriasis, comprising administering a composition comprising an antibody or fragment thereof to a patient wherein the antibody specifically binds to IL-17 Receptor A (IL-17RA) and has an antagonistic activity. 2. A method of according to claim 1, wherein the antibody or fragment thereof is selected from: a. an antibody, comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8; b. an antibody comprising the full length light chain amino acid sequence of SEQ ID NO: 9 and a full length heavy chain amino acid sequence of SEQ ID NO: 10 and c. an antibody, comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO:1, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO:2, a light chain CDR3 comprising the amino acid sequence of SEQ ID NO:3, a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO:4, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO:5, and a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO:6. 3. The method of claim 1 or 2, wherein the antibody or fragment thereof inhibits binding of IL-17A to said IL-17RA. 4. The method of any of the preceding claims wherein the patient is suffering from psoriasis. 5. The method of any of the preceding claims wherein the patient is suffering from plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 6. The method of any of the preceding claims wherein the patient has a NAPSI score of at least 6 or a modified NAPSI score of at least 2 on one or more nails. 7. The method of any of the preceding claims wherein the patient has a PSSI score of at least 15. 9. The method of any of the preceding claims where the patient has a SSA score of at least 30%. 10. The method of treating nail psoriasis according to any of preceding claims, wherein the composition is administered at a dose of antibody or fragment thereof effective to reduce or maintain a NAPSI score of 6 or less or a mNAPSI score of 3 or less on an affected nail. 11. The method of treating scalp psoriasis according to any of preceding claims wherein the composition is administered at a dose of antibody or fragment thereof effective to reduce and maintain a PSSI score of 15 or less. 12. The method of any of the preceding claims wherein the composition administered comprises a dose of antibody that is 70 mg, 140mg, 210 mg or 280 mg. 13. A method of treating nail or scalp psoriasis or any of the preceding claims wherein the patient has less than 10% body surface area affected by psoriasis and additionally having nail or scalp psoriasis. 14. The method of any of the preceding claims wherein the patient has less than 10% body surface area affected by moderate to severe psoriasis, plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 15. The method of claim 13 or 14 wherein the patient has about 7% or less body surface area affected by psoriasis. 16. The method of claim 13 or 14 wherein the patient has about 5% or less body surface area affected by psoriasis. 17. The method of claim 13 or 14 wherein the patient has about 3% or less body surface area affected by psoriasis. 18. The method of claim 13 or 14 wherein the patient has about 1% or less body surface area affected by psoriasis. 19. The method any one of the preceding claims, further comprising administering to said patient a second treatment. 20. The method of claim 19, wherein said second treatment is administered prior to, concurrent with, or subsequent to administration of said composition comprising said antibody. 21. The method of claim 19 or 20 wherein the second treatment is a topical treatment. 22. The method of claim 21 where in the topical treatment is selected from the group consisting fluorouracil, dithranol, tazarotene, cyclosporine, calcineneurin inhibitors, triamcinolone, fluocinonide, topical steroids, vitamin D3, vitamin D3 analogs, betamethasone dipropionate, betamethasone valerate, calcipotriol, clobetasol, XAMIOL, DAIVOBET, coal tar, urea, corticosteroids, retinoids, anthralin, topical methatrexate, keratolytics, salicylic acid, tofacitinab, apremilast, topical JAK inhibitors or a combination thereof. 23. The method of any one of claims 20-22, wherien the second treatment is selected from the group consisting of retinoids, acitretin cyclosporine, methotrexate, apremilast, tofacitinib, oral JAK inhibitors, oral PI3 kinase inhibitors, oral MAP kinase inhibitors, Fumaderm, fumarates, dimethyl fumarate, sulfasalazine, leflunomide, calcineurin inhibitors, azathioprine, thioguanine, hydroxyurea, hydroxychloroquine, sulfasalazine, antifungals or a combination thereof. 24. The method of any one of claims 20-23, wherein the second treatment an antibody or chimeric protein specific for TNF IL-17 IL-12/23 or IL-23. 25. The method of claim 24, wherein the antibody or chimeric protein is infliximab, adalimumab, etanercept, alefacept, ustekinumab, secukinumab, ixekizumab, guselkumab, antifungals or a combination thereof. 26. The method of any one of claims 20-25, wherein the second treatment is selected from the group consisting of is triamcinolone acetonide photochemotherapy, laser therapy, Excimer laser, oral/topical psorallen with UVA (PUVA), pulsed dye laser, radiation therapy, superficial radiotherapy, electron beam therapy, Grenz ray therapy, ermatome shaving, aloe vera extract, narrow band UV therapy UV therapy or a combination thereof. 27. The method of any one of the preceding claims, wherein said antibody is selected from the group consisting of: a. a humanized antibody; b. a chimeric antibody; c. a monoclonal antibody; d. an antigen-binding antibody fragment; e. a single chain antibody; f. a diabody; g. a triabody; h. a tetrabody; i. a Fab fragment; j. a F(ab')2 fragment; k. an IgD antibody; l. an IgE antibody; m. an IgM antibody; n. an IgG1 antibody; o. an IgG2 antibody; p. an IgG3 antibody; and q.an IgG4 antibody. 28. The method of any one of the preceding claims, wherein said composition is a pharmaceutical composition. 29. The method of any one of the preceding claims, wherein said pharmaceutical composition further comprises a pharmaceutically acceptable diluent. 30. The method of claim 28 or 29 wherein the pharmaceutical composition comprises an aqueous solution of a glutamic acid buffer comprising a) said glutamic acid buffer comprises a glutamic acid concentration of 5-30 mM.+-.0.2 mM; b) said glutamic acid buffer comprises a pH of 4.5-5.2.+-.0.2; c) said formulation further comprises 2-4% proline (w/v) and 0.005-0.02% (w/v) polysorbate 20 and d) the antibody or fragment thereof at a concentration of 100 to 150 mg/ml. 31. The method of claim 30 wherein the pharmaceutical composition has an osmolarity of 275 to 325 osm. 32. The method of claim 30 or 31 wherein the pharmaceutical composition has a viscosity of 5 to 7 cP at 25° C. 33. The method of any one of the preceding claims, wherein said antibody is a human IgG2 monoclonal antibody. 34. Use of an antibody or fragment thereof for the preparation of a medicament for the treatment of nail or scalp psoriasis, wherein the antibody or fragment thereof specifically binds to IL-17receptor RA (IL-17RA) and has an antagonistic activity. 35. A composition for use in the treatment of nail or scalp psoriasis, wherein the composition comprises an antibody or fragment thereof that specifically binds to IL-17receptor RA (IL-17RA) and has an antagonistic activity. 36. The use or composition of claim 34 or 35, wherein the antibody or fragment thereof is selected from: a. an antibody, comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8; b. an antibody comprising the full length light chain amino acid sequence of SEQ ID NO: 9 and a full length heavy chain amino acid sequence of SEQ ID NO: 10 and c. an antibody, comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO:1, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO:2, a light chain CDR3 comprising the amino acid sequence of SEQ ID NO:3, a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO:4, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO:5, and a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO:6. 37. The use or composition of any one of claims 34-36, wherein the antibody or fragment thereof inhibits binding of IL-17A to said IL-17RA. 38. The use or composition of any one of claims 34-37, wherein the medicament or composition is for administration to a patient suffering from psoriasis. 39. The use or composition of any one of claims 34-38, wherein the medicament or composition is for administration to a patient suffering from plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 40. The use or composition of claim 38 or 39, wherein the patient has a NAPSI score of at least 6 or a modified NAPSI score of at least 2 or 3 on one or more nails. 41. The use or composition of any one of claims 38-40, wherein the patient has a PSSI score of at least 15. 42. The use or composition of any one of claims 34-41, where the patient has a SSA score of at least 30%. 43. The use or composition of any one of claims 34-42, wherein the medicament or the composition comprises a dose of antibody or fragment thereof effective to reduce and maintain a NAPSI score of 6 or less or a mNAPSI score of 3 or less on an affected nail. 44. The use or composition of any one of claims 34-43, wherein the medicament or composition comprises a dose of antibody or fragment thereof effective to reduce and maintain a PSSI score of 15 or less. 45. The use or composition of any one of claims 34-44, wherein the medicament or composition comprises a dose of antibody that is 70 mg, 140mg, 210 mg or 280 mg. 46. The use or composition of any one of claims 34-45, wherein the medicament or composition is for administration to a patient having less than 10% body surface area affected by psoriasis and additionally having nail or scalp psoriasis. 47. The use or composition of any one of claims 34-46, wherein the medicament or composition is for administration to a patient having less than 10% body surface area affected by moderate to severe psoriasis, plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 48. The use or composition of any one of claims 34-46, wherein the medicament or composition is administered with a second treatment. 49. The use or composition of claim 48, wherein said second treatment is administered prior to, concurrent with, or subsequent to administration of said medicament or composition comprising said antibody. 50. The use or composition of claim 48 or 49, wherein the second treatment is a topical treatment. 51. The use or composition of claim 50, where in the topical treatment is selected from the group consisting fluorouracil, dithranol, tazarotene, cyclosporine, calcineneurin inhibitors, triamcinolone, fluocinonide, topical steroids, vitamin D3, vitamin D3 analogs, betamethasone dipropionate, betamethasone valerate, calcipotriol, clobetasol, XAMIOL, DAIVOBET, coal tar, urea, corticosteroids, retinoids, anthralin, topical methatrexate, keratolytics, salicylic acid, tofacitinab, apremilast, topical JAK inhibitors or a combination thereof. 52. The use or composition of any one of claims 48-51, wherein the second treatment is selected from the group consisting of retinoids, acitretin cyclosporine, methotrexate, apremilast, tofacitinib, oral JAK inhibitors, oral PI3 kinase inhibitors, oral MAP kinase inhibitors, Fumaderm, fumarates, dimethyl fumarate, sulfasalazine, leflunomide, calcineurin inhibitors, azathioprine, thioguanine, hydroxyurea, hydroxychloroquine, sulfasalazine, antifungals or a combination thereof. 53. The use or composition of any one of claims 48-52, wherein the second treatment an antibody or chimeric protein specific for TNF, IL-17, IL-12/23 or IL-23. 54. The use or composition of claim 53, wherein the antibody or chimeric protein is infliximab, adalimumab, etanercept, alefacept, ustekinumab, secukinumab, ixekizumab, guselkumab, antifungals or a combination thereof. 55. The use or composition of any one of claims 48-54, wherein the second treatment is selected from the group consisting of is triamcinolone acetonide photochemotherapy, laser therapy, Excimer laser, oral/topical psorallen with UVA (PUVA), pulsed dye laser, radiation therapy, superficial radiotherapy, electron beam therapy, Grenz ray therapy, ermatome shaving, aloe vera extract, narrow band UV therapy UV therapy or a combination thereof. 56. The use or composition of any one of claims 34-55, wherein said antibody is selected from the group consisting of: a. a humanized antibody; b. a chimeric antibody; c. a monoclonal antibody; d. an antigen-binding antibody fragment; e. a single chain antibody; f. a diabody; g. a triabody; h. a tetrabody; i. a Fab fragment; j. a F(ab')2 fragment; k. an IgD antibody; l. an IgE antibody; m. an IgM antibody; n. an IgG1 antibody; o. an IgG2 antibody; p. an IgG3 antibody; and q.an IgG4 antibody. 57. The use or composition of any one of claims 34-56, wherein said medicament or composition further comprises a pharmaceutically acceptable diluent. 58. The use or composition of any one of claims 34-57, wherein said medicament or composition comprises an aqueous solution of a glutamic acid buffer comprising a) said glutamic acid buffer comprises a glutamic acid concentration of 5-30 mM.+-.0.2 mM; b) said glutamic acid buffer comprises a pH of 4.5-5.2.+-.0.2; c) said formulation further comprises 2-4% proline (w/v) and 0.005-0.02% (w/v) polysorbate 20 and d) the antibody or fragment thereof at a concentration of 100 to 150 mg/ml. 59. The use or composition of claim 58, wherein the medicament or composition has an osmolarity of 275 to 325 osm. 60. The use or composition of claim 58 or 59, wherein the medicament or composition has a viscosity of 5 to 7 cP at 25° C. 61. The use or composition of any one of claims 34-60, wherein said antibody is a human IgG2 monoclonal antibody.
The present invention relates to a therapeutic agent for nail and scalp psoriasis comprising an IL-17 Receptor A (IL-17RA or IL-17R) antigen binding proteins, such as monoclonal antibodies that bind IL-17RA, and method of using the same.1. A method for treating nail or scalp psoriasis, comprising administering a composition comprising an antibody or fragment thereof to a patient wherein the antibody specifically binds to IL-17 Receptor A (IL-17RA) and has an antagonistic activity. 2. A method of according to claim 1, wherein the antibody or fragment thereof is selected from: a. an antibody, comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8; b. an antibody comprising the full length light chain amino acid sequence of SEQ ID NO: 9 and a full length heavy chain amino acid sequence of SEQ ID NO: 10 and c. an antibody, comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO:1, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO:2, a light chain CDR3 comprising the amino acid sequence of SEQ ID NO:3, a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO:4, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO:5, and a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO:6. 3. The method of claim 1 or 2, wherein the antibody or fragment thereof inhibits binding of IL-17A to said IL-17RA. 4. The method of any of the preceding claims wherein the patient is suffering from psoriasis. 5. The method of any of the preceding claims wherein the patient is suffering from plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 6. The method of any of the preceding claims wherein the patient has a NAPSI score of at least 6 or a modified NAPSI score of at least 2 on one or more nails. 7. The method of any of the preceding claims wherein the patient has a PSSI score of at least 15. 9. The method of any of the preceding claims where the patient has a SSA score of at least 30%. 10. The method of treating nail psoriasis according to any of preceding claims, wherein the composition is administered at a dose of antibody or fragment thereof effective to reduce or maintain a NAPSI score of 6 or less or a mNAPSI score of 3 or less on an affected nail. 11. The method of treating scalp psoriasis according to any of preceding claims wherein the composition is administered at a dose of antibody or fragment thereof effective to reduce and maintain a PSSI score of 15 or less. 12. The method of any of the preceding claims wherein the composition administered comprises a dose of antibody that is 70 mg, 140mg, 210 mg or 280 mg. 13. A method of treating nail or scalp psoriasis or any of the preceding claims wherein the patient has less than 10% body surface area affected by psoriasis and additionally having nail or scalp psoriasis. 14. The method of any of the preceding claims wherein the patient has less than 10% body surface area affected by moderate to severe psoriasis, plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 15. The method of claim 13 or 14 wherein the patient has about 7% or less body surface area affected by psoriasis. 16. The method of claim 13 or 14 wherein the patient has about 5% or less body surface area affected by psoriasis. 17. The method of claim 13 or 14 wherein the patient has about 3% or less body surface area affected by psoriasis. 18. The method of claim 13 or 14 wherein the patient has about 1% or less body surface area affected by psoriasis. 19. The method any one of the preceding claims, further comprising administering to said patient a second treatment. 20. The method of claim 19, wherein said second treatment is administered prior to, concurrent with, or subsequent to administration of said composition comprising said antibody. 21. The method of claim 19 or 20 wherein the second treatment is a topical treatment. 22. The method of claim 21 where in the topical treatment is selected from the group consisting fluorouracil, dithranol, tazarotene, cyclosporine, calcineneurin inhibitors, triamcinolone, fluocinonide, topical steroids, vitamin D3, vitamin D3 analogs, betamethasone dipropionate, betamethasone valerate, calcipotriol, clobetasol, XAMIOL, DAIVOBET, coal tar, urea, corticosteroids, retinoids, anthralin, topical methatrexate, keratolytics, salicylic acid, tofacitinab, apremilast, topical JAK inhibitors or a combination thereof. 23. The method of any one of claims 20-22, wherien the second treatment is selected from the group consisting of retinoids, acitretin cyclosporine, methotrexate, apremilast, tofacitinib, oral JAK inhibitors, oral PI3 kinase inhibitors, oral MAP kinase inhibitors, Fumaderm, fumarates, dimethyl fumarate, sulfasalazine, leflunomide, calcineurin inhibitors, azathioprine, thioguanine, hydroxyurea, hydroxychloroquine, sulfasalazine, antifungals or a combination thereof. 24. The method of any one of claims 20-23, wherein the second treatment an antibody or chimeric protein specific for TNF IL-17 IL-12/23 or IL-23. 25. The method of claim 24, wherein the antibody or chimeric protein is infliximab, adalimumab, etanercept, alefacept, ustekinumab, secukinumab, ixekizumab, guselkumab, antifungals or a combination thereof. 26. The method of any one of claims 20-25, wherein the second treatment is selected from the group consisting of is triamcinolone acetonide photochemotherapy, laser therapy, Excimer laser, oral/topical psorallen with UVA (PUVA), pulsed dye laser, radiation therapy, superficial radiotherapy, electron beam therapy, Grenz ray therapy, ermatome shaving, aloe vera extract, narrow band UV therapy UV therapy or a combination thereof. 27. The method of any one of the preceding claims, wherein said antibody is selected from the group consisting of: a. a humanized antibody; b. a chimeric antibody; c. a monoclonal antibody; d. an antigen-binding antibody fragment; e. a single chain antibody; f. a diabody; g. a triabody; h. a tetrabody; i. a Fab fragment; j. a F(ab')2 fragment; k. an IgD antibody; l. an IgE antibody; m. an IgM antibody; n. an IgG1 antibody; o. an IgG2 antibody; p. an IgG3 antibody; and q.an IgG4 antibody. 28. The method of any one of the preceding claims, wherein said composition is a pharmaceutical composition. 29. The method of any one of the preceding claims, wherein said pharmaceutical composition further comprises a pharmaceutically acceptable diluent. 30. The method of claim 28 or 29 wherein the pharmaceutical composition comprises an aqueous solution of a glutamic acid buffer comprising a) said glutamic acid buffer comprises a glutamic acid concentration of 5-30 mM.+-.0.2 mM; b) said glutamic acid buffer comprises a pH of 4.5-5.2.+-.0.2; c) said formulation further comprises 2-4% proline (w/v) and 0.005-0.02% (w/v) polysorbate 20 and d) the antibody or fragment thereof at a concentration of 100 to 150 mg/ml. 31. The method of claim 30 wherein the pharmaceutical composition has an osmolarity of 275 to 325 osm. 32. The method of claim 30 or 31 wherein the pharmaceutical composition has a viscosity of 5 to 7 cP at 25° C. 33. The method of any one of the preceding claims, wherein said antibody is a human IgG2 monoclonal antibody. 34. Use of an antibody or fragment thereof for the preparation of a medicament for the treatment of nail or scalp psoriasis, wherein the antibody or fragment thereof specifically binds to IL-17receptor RA (IL-17RA) and has an antagonistic activity. 35. A composition for use in the treatment of nail or scalp psoriasis, wherein the composition comprises an antibody or fragment thereof that specifically binds to IL-17receptor RA (IL-17RA) and has an antagonistic activity. 36. The use or composition of claim 34 or 35, wherein the antibody or fragment thereof is selected from: a. an antibody, comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8; b. an antibody comprising the full length light chain amino acid sequence of SEQ ID NO: 9 and a full length heavy chain amino acid sequence of SEQ ID NO: 10 and c. an antibody, comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO:1, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO:2, a light chain CDR3 comprising the amino acid sequence of SEQ ID NO:3, a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO:4, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO:5, and a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO:6. 37. The use or composition of any one of claims 34-36, wherein the antibody or fragment thereof inhibits binding of IL-17A to said IL-17RA. 38. The use or composition of any one of claims 34-37, wherein the medicament or composition is for administration to a patient suffering from psoriasis. 39. The use or composition of any one of claims 34-38, wherein the medicament or composition is for administration to a patient suffering from plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 40. The use or composition of claim 38 or 39, wherein the patient has a NAPSI score of at least 6 or a modified NAPSI score of at least 2 or 3 on one or more nails. 41. The use or composition of any one of claims 38-40, wherein the patient has a PSSI score of at least 15. 42. The use or composition of any one of claims 34-41, where the patient has a SSA score of at least 30%. 43. The use or composition of any one of claims 34-42, wherein the medicament or the composition comprises a dose of antibody or fragment thereof effective to reduce and maintain a NAPSI score of 6 or less or a mNAPSI score of 3 or less on an affected nail. 44. The use or composition of any one of claims 34-43, wherein the medicament or composition comprises a dose of antibody or fragment thereof effective to reduce and maintain a PSSI score of 15 or less. 45. The use or composition of any one of claims 34-44, wherein the medicament or composition comprises a dose of antibody that is 70 mg, 140mg, 210 mg or 280 mg. 46. The use or composition of any one of claims 34-45, wherein the medicament or composition is for administration to a patient having less than 10% body surface area affected by psoriasis and additionally having nail or scalp psoriasis. 47. The use or composition of any one of claims 34-46, wherein the medicament or composition is for administration to a patient having less than 10% body surface area affected by moderate to severe psoriasis, plaque psoriasis, pustular psoriasis or psoriatic erythroderma. 48. The use or composition of any one of claims 34-46, wherein the medicament or composition is administered with a second treatment. 49. The use or composition of claim 48, wherein said second treatment is administered prior to, concurrent with, or subsequent to administration of said medicament or composition comprising said antibody. 50. The use or composition of claim 48 or 49, wherein the second treatment is a topical treatment. 51. The use or composition of claim 50, where in the topical treatment is selected from the group consisting fluorouracil, dithranol, tazarotene, cyclosporine, calcineneurin inhibitors, triamcinolone, fluocinonide, topical steroids, vitamin D3, vitamin D3 analogs, betamethasone dipropionate, betamethasone valerate, calcipotriol, clobetasol, XAMIOL, DAIVOBET, coal tar, urea, corticosteroids, retinoids, anthralin, topical methatrexate, keratolytics, salicylic acid, tofacitinab, apremilast, topical JAK inhibitors or a combination thereof. 52. The use or composition of any one of claims 48-51, wherein the second treatment is selected from the group consisting of retinoids, acitretin cyclosporine, methotrexate, apremilast, tofacitinib, oral JAK inhibitors, oral PI3 kinase inhibitors, oral MAP kinase inhibitors, Fumaderm, fumarates, dimethyl fumarate, sulfasalazine, leflunomide, calcineurin inhibitors, azathioprine, thioguanine, hydroxyurea, hydroxychloroquine, sulfasalazine, antifungals or a combination thereof. 53. The use or composition of any one of claims 48-52, wherein the second treatment an antibody or chimeric protein specific for TNF, IL-17, IL-12/23 or IL-23. 54. The use or composition of claim 53, wherein the antibody or chimeric protein is infliximab, adalimumab, etanercept, alefacept, ustekinumab, secukinumab, ixekizumab, guselkumab, antifungals or a combination thereof. 55. The use or composition of any one of claims 48-54, wherein the second treatment is selected from the group consisting of is triamcinolone acetonide photochemotherapy, laser therapy, Excimer laser, oral/topical psorallen with UVA (PUVA), pulsed dye laser, radiation therapy, superficial radiotherapy, electron beam therapy, Grenz ray therapy, ermatome shaving, aloe vera extract, narrow band UV therapy UV therapy or a combination thereof. 56. The use or composition of any one of claims 34-55, wherein said antibody is selected from the group consisting of: a. a humanized antibody; b. a chimeric antibody; c. a monoclonal antibody; d. an antigen-binding antibody fragment; e. a single chain antibody; f. a diabody; g. a triabody; h. a tetrabody; i. a Fab fragment; j. a F(ab')2 fragment; k. an IgD antibody; l. an IgE antibody; m. an IgM antibody; n. an IgG1 antibody; o. an IgG2 antibody; p. an IgG3 antibody; and q.an IgG4 antibody. 57. The use or composition of any one of claims 34-56, wherein said medicament or composition further comprises a pharmaceutically acceptable diluent. 58. The use or composition of any one of claims 34-57, wherein said medicament or composition comprises an aqueous solution of a glutamic acid buffer comprising a) said glutamic acid buffer comprises a glutamic acid concentration of 5-30 mM.+-.0.2 mM; b) said glutamic acid buffer comprises a pH of 4.5-5.2.+-.0.2; c) said formulation further comprises 2-4% proline (w/v) and 0.005-0.02% (w/v) polysorbate 20 and d) the antibody or fragment thereof at a concentration of 100 to 150 mg/ml. 59. The use or composition of claim 58, wherein the medicament or composition has an osmolarity of 275 to 325 osm. 60. The use or composition of claim 58 or 59, wherein the medicament or composition has a viscosity of 5 to 7 cP at 25° C. 61. The use or composition of any one of claims 34-60, wherein said antibody is a human IgG2 monoclonal antibody.
1,600
573
14,122,892
1,628
The present invention relates to a process for the preparation of a compound of formula (I) comprising the steps of a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P 1 is a protecting group selected from R 2 Si—R 3 R 4 or R 1 CO—, R 1 is a group selected from C 1-6 alkyl or C 3-6 cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; R 2 , R 3 and R 4 are each independently a group selected from C 1-6 alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R 5 , and R 5 is a group selected from C 6-10 aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C 1-4 alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I).
1. A process for the preparation of a compound of formula (I) comprising the steps of a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P1 is a protecting group selected from R2—Si—R3R4 or R1CO—, R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R5, and R5 is a group selected from C6-10aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C1-4alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I). 2. The process according to claim 1, wherein step (b) is a sulfinylation and the sulfinylation is performed by reacting the compound of formula (III) with a base and with a sulfinylation reagent. 3. The process according to claim 1 wherein step (b) is a sulfinylation and a sulfinylation reagent is methyl 2-pyridinesulfinate, methyl benzenesulfinate, methyl 4-methyl-benzenesulfinate, methyl 4-chloro-benzene sulfinate. 4. The process according to claim 2, wherein the base used in the sulfinylation step is selected from the group comprising potassium hydride, potassium terbutylate, sodium hydride, sodium terbutylate and a mixture thereof. 5. The process according to claim 1, wherein step (b) is a halogenation and the halogenation is performed by reacting the compound of formula (III) with a halogenating reagent. 6. The process according to claim 1 wherein step (b) is a bromination and a brominating reagent is selected from the group comprising copper(II) bromide, bromine, and pyridine bromine perbromine. 7. The process according to claim 1, wherein the desulfinylation step is carried out with heat, or optionally in the presence of cupric sulfate. 8. The process according to claim 1, wherein the dehalogenation step is performed in the presence of a base. 9. The process according to claim 8, wherein the base is selected from the group comprising imidazole, collidine, 2,6-lutidine, triethylamine, or 1,8-diazabicyclo[5.4.0]undec-7-ene. 10. The process according to claim 1, wherein step (c) is performed using a reducing agent selected from the group of metal hydride compounds. 11. The process according to claim 1, wherein step (c) is performed using a reducing agent selected from the group comprising NaBH4/CeCl3, LiAlH4, NaBH4, NaBH(OAc)3, and ZnBH4. 12. The process according to claim 1, wherein the silylating agent is selected from the group comprising C1-6alkylsilylchloride, C1-6alkylsilyltriflate, phenylsilylchloride, phenylsilyltriflate, C1-6alkylphenylsilylchloride, C1-6alkylphenylsilyltriflate, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl. 13. The process according to claim 1, wherein the acylating agent is selected from the group comprising C2-6alkenylC1-6alkanoates, C2-6alkenylC3-6cycloalkanoate, acyl chlorides and anhydrides. 14. A process for the preparation of estetrol, said process comprising preparing a compound of formula (I) by a process, the process comprising the steps of: a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P1 is a protecting group selected from R2—Si—R3R4 or R1CO—, R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R5, and R5 is a group selected from C6-10aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C1-4alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I); and further reacting compound of formula (I) to produce estetrol. 15. A method of treating a disease or condition, the method comprising treating a subject with an estetrol of formula (I) wherein the estetrol is produced by a process comprising the steps of: a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P1 is a protecting group selected from R2—Si—R3R4 or R1CO—, R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R5, and R5 is a group selected from C6-10aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C1-4alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I), wherein the method is effective for hormone replacement therapy, vaginal dryness, contraception, a enhancing libido, treating skin, promoting wound healing, and treating or preventing a disorder selected from the group consisting of autoimmune diseases, breast tumors and colorectal tumors. 16. The process according to claim 14, wherein step (b) is a sulfinylation and the sulfinylation is performed by reacting the compound of formula (III) with a base and with a sulfinylation reagent. 17. The process according to claim 14 wherein step (b) is a sulfinylation and a sulfinylation reagent is methyl 2-pyridinesulfinate, methyl benzenesulfinate, methyl 4-methyl-benzenesulfinate, methyl 4-chloro-benzene sulfinate. 18. The process according to claim 16, wherein the base used in the sulfinylation step is selected from the group comprising potassium hydride, potassium terbutylate, sodium hydride, sodium terbutylate and a mixture thereof. 19. The process according to claim 14, wherein step (b) is a halogenation and the halogenation is performed by reacting the compound of formula (III) with a halogenating reagent. 20. The process according to claim 14 wherein step (b) is a bromination and a brominating reagent is selected from the group comprising copper(II) bromide, bromine, and pyridine bromine perbromine.
The present invention relates to a process for the preparation of a compound of formula (I) comprising the steps of a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P 1 is a protecting group selected from R 2 Si—R 3 R 4 or R 1 CO—, R 1 is a group selected from C 1-6 alkyl or C 3-6 cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; R 2 , R 3 and R 4 are each independently a group selected from C 1-6 alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C 1-4 alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R 5 , and R 5 is a group selected from C 6-10 aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C 1-4 alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I).1. A process for the preparation of a compound of formula (I) comprising the steps of a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P1 is a protecting group selected from R2—Si—R3R4 or R1CO—, R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R5, and R5 is a group selected from C6-10aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C1-4alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I). 2. The process according to claim 1, wherein step (b) is a sulfinylation and the sulfinylation is performed by reacting the compound of formula (III) with a base and with a sulfinylation reagent. 3. The process according to claim 1 wherein step (b) is a sulfinylation and a sulfinylation reagent is methyl 2-pyridinesulfinate, methyl benzenesulfinate, methyl 4-methyl-benzenesulfinate, methyl 4-chloro-benzene sulfinate. 4. The process according to claim 2, wherein the base used in the sulfinylation step is selected from the group comprising potassium hydride, potassium terbutylate, sodium hydride, sodium terbutylate and a mixture thereof. 5. The process according to claim 1, wherein step (b) is a halogenation and the halogenation is performed by reacting the compound of formula (III) with a halogenating reagent. 6. The process according to claim 1 wherein step (b) is a bromination and a brominating reagent is selected from the group comprising copper(II) bromide, bromine, and pyridine bromine perbromine. 7. The process according to claim 1, wherein the desulfinylation step is carried out with heat, or optionally in the presence of cupric sulfate. 8. The process according to claim 1, wherein the dehalogenation step is performed in the presence of a base. 9. The process according to claim 8, wherein the base is selected from the group comprising imidazole, collidine, 2,6-lutidine, triethylamine, or 1,8-diazabicyclo[5.4.0]undec-7-ene. 10. The process according to claim 1, wherein step (c) is performed using a reducing agent selected from the group of metal hydride compounds. 11. The process according to claim 1, wherein step (c) is performed using a reducing agent selected from the group comprising NaBH4/CeCl3, LiAlH4, NaBH4, NaBH(OAc)3, and ZnBH4. 12. The process according to claim 1, wherein the silylating agent is selected from the group comprising C1-6alkylsilylchloride, C1-6alkylsilyltriflate, phenylsilylchloride, phenylsilyltriflate, C1-6alkylphenylsilylchloride, C1-6alkylphenylsilyltriflate, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl. 13. The process according to claim 1, wherein the acylating agent is selected from the group comprising C2-6alkenylC1-6alkanoates, C2-6alkenylC3-6cycloalkanoate, acyl chlorides and anhydrides. 14. A process for the preparation of estetrol, said process comprising preparing a compound of formula (I) by a process, the process comprising the steps of: a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P1 is a protecting group selected from R2—Si—R3R4 or R1CO—, R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R5, and R5 is a group selected from C6-10aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C1-4alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I); and further reacting compound of formula (I) to produce estetrol. 15. A method of treating a disease or condition, the method comprising treating a subject with an estetrol of formula (I) wherein the estetrol is produced by a process comprising the steps of: a) reacting a compound of formula (II) with a silylating or an acylating agent to produce compound of formula (III), wherein P1 is a protecting group selected from R2—Si—R3R4 or R1CO—, R1 is a group selected from C1-6alkyl or C3-6cycloalkyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; R2, R3 and R4 are each independently a group selected from C1-6alkyl or phenyl, each group being optionally substituted by one or more substituents independently selected from fluoro or C1-4alkyl; b) halogenation or sulfinylation of the compound of formula (III) to produce a compound of formula (IV); wherein X is halo, or —O—SO—R5, and R5 is a group selected from C6-10aryl or heteroaryl, each group being optionally substituted by one or more substituents independently selected from chloro or C1-4alkyl; c) dehalogenation or desulfinylation of the compound of formula (IV) to produce compound of formula (V); and d) reacting the compound of formula (V) with a reducing agent to produce compound of formula (I), wherein the method is effective for hormone replacement therapy, vaginal dryness, contraception, a enhancing libido, treating skin, promoting wound healing, and treating or preventing a disorder selected from the group consisting of autoimmune diseases, breast tumors and colorectal tumors. 16. The process according to claim 14, wherein step (b) is a sulfinylation and the sulfinylation is performed by reacting the compound of formula (III) with a base and with a sulfinylation reagent. 17. The process according to claim 14 wherein step (b) is a sulfinylation and a sulfinylation reagent is methyl 2-pyridinesulfinate, methyl benzenesulfinate, methyl 4-methyl-benzenesulfinate, methyl 4-chloro-benzene sulfinate. 18. The process according to claim 16, wherein the base used in the sulfinylation step is selected from the group comprising potassium hydride, potassium terbutylate, sodium hydride, sodium terbutylate and a mixture thereof. 19. The process according to claim 14, wherein step (b) is a halogenation and the halogenation is performed by reacting the compound of formula (III) with a halogenating reagent. 20. The process according to claim 14 wherein step (b) is a bromination and a brominating reagent is selected from the group comprising copper(II) bromide, bromine, and pyridine bromine perbromine.
1,600
574
15,869,438
1,641
The invention relates to a method for classifying a patient at risk for heart failure, wherein said method comprises the steps of (i) measuring the concentration of IGFBP2 in a sample obtained from said patient and (ii) comparing the concentration of IGFBP2 measured in step (i) to a control value derived from the concentration of IGFBP2 in samples from patients who are at particular stages of heart failure and/or to a control value derived from the concentration of IGFBP2 in blood samples from healthy patients.
1-8. (canceled) 9. A method of treating heart failure in a patient in need thereof, comprising the steps of: a) obtaining a biological sample from said patient; b) measuring the concentration of IGFBP2 in the sample obtained in step a); c) determining the severity of the heart failure depending on the concentration of IGFBP2 measured in step b); and d) administering to said patient diagnosed as having heart failure an appropriate medical care for a subject suffering from heart failure depending on the severity of the heart failure as determined in step c). 10. The method according to claim 9, wherein the severity of the heart failure depending on the concentration of IGFBP2 measured in step b) is determined according to the NYHA heart failure classification. 11. The method according to claim 9, wherein step b) is performed by measuring the level of IGFBP2 protein in the sample obtained in step a). 12. The method according to claim 9, wherein the biological sample obtained in step a) is a urine sample. 13. The method according to claim 9, wherein the biological sample obtained in step a) is a plasma sample. 14. The method according to claim 9, wherein the biological sample obtained in step a) is a serum sample. 15. The method according to claim 9, wherein the measuring of the concentration of IGFBP2 is performed by using a set of antibodies direct against IGFBP2. 16. The method according to claim 13, wherein measuring the level of IGFBP2 protein is performed by ELISA.
The invention relates to a method for classifying a patient at risk for heart failure, wherein said method comprises the steps of (i) measuring the concentration of IGFBP2 in a sample obtained from said patient and (ii) comparing the concentration of IGFBP2 measured in step (i) to a control value derived from the concentration of IGFBP2 in samples from patients who are at particular stages of heart failure and/or to a control value derived from the concentration of IGFBP2 in blood samples from healthy patients.1-8. (canceled) 9. A method of treating heart failure in a patient in need thereof, comprising the steps of: a) obtaining a biological sample from said patient; b) measuring the concentration of IGFBP2 in the sample obtained in step a); c) determining the severity of the heart failure depending on the concentration of IGFBP2 measured in step b); and d) administering to said patient diagnosed as having heart failure an appropriate medical care for a subject suffering from heart failure depending on the severity of the heart failure as determined in step c). 10. The method according to claim 9, wherein the severity of the heart failure depending on the concentration of IGFBP2 measured in step b) is determined according to the NYHA heart failure classification. 11. The method according to claim 9, wherein step b) is performed by measuring the level of IGFBP2 protein in the sample obtained in step a). 12. The method according to claim 9, wherein the biological sample obtained in step a) is a urine sample. 13. The method according to claim 9, wherein the biological sample obtained in step a) is a plasma sample. 14. The method according to claim 9, wherein the biological sample obtained in step a) is a serum sample. 15. The method according to claim 9, wherein the measuring of the concentration of IGFBP2 is performed by using a set of antibodies direct against IGFBP2. 16. The method according to claim 13, wherein measuring the level of IGFBP2 protein is performed by ELISA.
1,600
575
15,520,756
1,628
The invention generally relates to methods of treating viral infections using known drugs and pharmaceutical compositions comprising same. More specifically, the disclosed methods are useful for the treatment of viral infections that are enveloped viruses. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.
1. A method for treating viral infection, the method comprising the step of administering to the subject an effective amount of at least one compound having a structure selected from: or a pharmaceutically acceptable salt thereof, thereby treating the subject for viral infection. 2. The method of claim 1, wherein the viral infection is selected from human immunodeficiency virus (HIV), human papillomavirus (HPV), influenza, chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, and viral pneumonia. 3. The method of claim 2, wherein the viral infection is influenza. 4. The method of claim 1, wherein the viral infection is an enveloped virus. 5. The method of claim 4, wherein the enveloped virus is a respiratory virus. 6. The method of claim 1, wherein the subject is a mammal. 7. The method of claim 6, wherein the mammal is a human. 8. The method of claim 1, wherein the subject has been diagnosed with a need for treatment of the viral infection prior to the administering step. 9. The method of claim 1, further comprising the step of identifying a subject in need of treatment of the viral infection. 10. The method of claim 1, wherein the effective amount is a therapeutically effective amount. 11. The method of claim 1, wherein the effective amount is a prophylactically effective amount. 12. The method off claim 1, wherein the compound has the structure: 13. A pharmaceutical composition comprising an effective amount of at least one compound having a structure selected from: or a pharmaceutically acceptable salt thereof; and an effective amount of: (a) at least one antiviral agent or a pharmaceutically acceptable salt thereof; or (b) at least one agent known to weaken the immune system or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier. 14. The composition of claim 13, wherein the effective amount is a therapeutically effective amount. 15. The composition of claim 13, wherein the effective amount is a prophylactically effective amount. 16. The composition of claim 13, wherein the compound has the structure: 17. A kit comprising an effective amount of at least one compound having a structure selected from: or a pharmaceutically acceptable salt thereof, and one or more of: (a) at least one antiviral agent; (b) at least one agent known to treat an autoimmune disorder; (c) instructions for treating a viral infection; (d) instructions for treating the autoimmune disorder; (e) instructions for administering the at least one compound in connection with treating a viral infection; or (f) instructions for administering the at least one compound in connection with reducing the risk of viral infection. 18. The kit of claim 17, wherein the at least one compound and the at least one agent are co-packaged. 19. The kit of claim 17, wherein the at least one compound and the at least one agent are co-formulated. 20. The kit of claim 17, wherein the at least one compound has the structure:
The invention generally relates to methods of treating viral infections using known drugs and pharmaceutical compositions comprising same. More specifically, the disclosed methods are useful for the treatment of viral infections that are enveloped viruses. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.1. A method for treating viral infection, the method comprising the step of administering to the subject an effective amount of at least one compound having a structure selected from: or a pharmaceutically acceptable salt thereof, thereby treating the subject for viral infection. 2. The method of claim 1, wherein the viral infection is selected from human immunodeficiency virus (HIV), human papillomavirus (HPV), influenza, chicken pox, infectious mononucleosis, mumps, measles, rubella, shingles, ebola, viral gastroenteritis, viral hepatitis, viral meningitis, human metapneumovirus, human parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, respiratory syncytial virus, and viral pneumonia. 3. The method of claim 2, wherein the viral infection is influenza. 4. The method of claim 1, wherein the viral infection is an enveloped virus. 5. The method of claim 4, wherein the enveloped virus is a respiratory virus. 6. The method of claim 1, wherein the subject is a mammal. 7. The method of claim 6, wherein the mammal is a human. 8. The method of claim 1, wherein the subject has been diagnosed with a need for treatment of the viral infection prior to the administering step. 9. The method of claim 1, further comprising the step of identifying a subject in need of treatment of the viral infection. 10. The method of claim 1, wherein the effective amount is a therapeutically effective amount. 11. The method of claim 1, wherein the effective amount is a prophylactically effective amount. 12. The method off claim 1, wherein the compound has the structure: 13. A pharmaceutical composition comprising an effective amount of at least one compound having a structure selected from: or a pharmaceutically acceptable salt thereof; and an effective amount of: (a) at least one antiviral agent or a pharmaceutically acceptable salt thereof; or (b) at least one agent known to weaken the immune system or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier. 14. The composition of claim 13, wherein the effective amount is a therapeutically effective amount. 15. The composition of claim 13, wherein the effective amount is a prophylactically effective amount. 16. The composition of claim 13, wherein the compound has the structure: 17. A kit comprising an effective amount of at least one compound having a structure selected from: or a pharmaceutically acceptable salt thereof, and one or more of: (a) at least one antiviral agent; (b) at least one agent known to treat an autoimmune disorder; (c) instructions for treating a viral infection; (d) instructions for treating the autoimmune disorder; (e) instructions for administering the at least one compound in connection with treating a viral infection; or (f) instructions for administering the at least one compound in connection with reducing the risk of viral infection. 18. The kit of claim 17, wherein the at least one compound and the at least one agent are co-packaged. 19. The kit of claim 17, wherein the at least one compound and the at least one agent are co-formulated. 20. The kit of claim 17, wherein the at least one compound has the structure:
1,600
576
15,599,420
1,646
Modulation of iron homeostasis by regulating BMP-6 activity is provided. Methods of using BMP-6 and BMP-6 protein-specific reagents, such as antibodies, for altering serum iron levels in humans are provided. Such antibodies are useful in pharmaceutical compositions for the prevention and treatment of hemochromatosis and anemia of inflammation.
1. A method of increasing serum transferrin saturation in a subject, the method comprising administering to said subject an effective amount of a pharmaceutical composition comprising a monoclonal antibody that specifically binds mature bone morphogenetic protein-6 (BMP-6) or fragments thereof. 2. The method of claim 1, wherein administration of said composition inhibits liver hepcidin mRNA production. 3. The method of claim 1, wherein administration of said composition inhibits hepatic hepcidin expression. 4. The method of claim 1, wherein the subject is a human and administration of said composition inhibits hepcidin expression in human hepatic cells. 5. The method of claim 1, wherein the antibody binds BMP-7. 6. The method of claim 5, wherein the antibody specifically binds BMP-6 with higher affinity than BMP-7. 7. The method of claim 6, wherein the antibody binds BMP-6 with at least 5-fold greater affinity than BMP-7. 8. The method of claim 6 wherein said BMP-6 is human BMP-6 and said BMP-7 is human BMP-7. 9. The method of claim 1, wherein the antibody binds to said BMP-6 with a Ka greater than or equal to about 104 M−1. 10. The method of claim 1, wherein administration of said composition reduces BMP-6 signaling. 11. The method of claim 1, wherein the mature BMP-6 comprises the amino acid sequence of SEQ ID NO: 2. 12. The method of claim 1, wherein the method increases serum iron. 13. The method of claim 1, wherein the composition is administered in an amount sufficient to inhibit an interaction between hemojuvelin and BMP-6. 14. The method of claim 1, wherein the subject is a human and the antibody preferably inhibits human BMP-6 over BMP-2, 4, 5, 7 or 9. 15. A method of inhibiting hepcidin expression in the liver of a subject, the method comprising administering to said subject an effective amount of a pharmaceutical composition comprising a monoclonal antibody that specifically binds bone morphogenetic protein-6 (BMP-6) or fragments thereof. 16. The method of claim 15, wherein administration of said composition inhibits liver hepcidin mRNA production. 17. The method of claim 15, wherein administration of said composition inhibits hepatic hepcidin expression. 18. The method of claim 15, wherein the subject is a human and administration of said composition inhibits hepcidin expression in human hepatic cells. 19. The method of claim 15, wherein administration of said composition increases serum transferrin saturation in the subject. 20. The method of claim 19, wherein the inhibition is in the presence of BMP-6 and BMP-2, BMP-4, BMP-5, BMP-7, or BMP-9. 21. The method of claim 15, wherein the antibody binds BMP-7. 22. The method of claim 21, wherein the antibody specifically binds BMP-6 with higher affinity than BMP-7. 23. The method of claim 22, wherein the antibody binds BMP-6 with at least 5-fold greater affinity than BMP-7. 24. The method of any one of claims 21, 22, or 23 wherein said BMP-6 is human BMP-6 and said BMP-7 is human BMP-7. 25. The method of claim 15, wherein the antibody binds to said BMP-6 with a Ka greater than or equal to about 104 M−1. 26. The method of claim 15, wherein administration of said composition reduces BMP-6 signaling. 27. The method of claim 15, wherein the mature BMP-6 comprises the amino acid sequence of SEQ ID NO: 2. 28. The method of claim 15, wherein the method increases serum iron. 29. The method of claim 15, wherein the composition is administered in an amount sufficient to inhibit an interaction between hemojuvelin and BMP-6. 30. The method of claim 15, wherein the subject is a human and the antibody preferably inhibits human BMP-6 over BMP-2, 4, 5, 7 or 9.
Modulation of iron homeostasis by regulating BMP-6 activity is provided. Methods of using BMP-6 and BMP-6 protein-specific reagents, such as antibodies, for altering serum iron levels in humans are provided. Such antibodies are useful in pharmaceutical compositions for the prevention and treatment of hemochromatosis and anemia of inflammation.1. A method of increasing serum transferrin saturation in a subject, the method comprising administering to said subject an effective amount of a pharmaceutical composition comprising a monoclonal antibody that specifically binds mature bone morphogenetic protein-6 (BMP-6) or fragments thereof. 2. The method of claim 1, wherein administration of said composition inhibits liver hepcidin mRNA production. 3. The method of claim 1, wherein administration of said composition inhibits hepatic hepcidin expression. 4. The method of claim 1, wherein the subject is a human and administration of said composition inhibits hepcidin expression in human hepatic cells. 5. The method of claim 1, wherein the antibody binds BMP-7. 6. The method of claim 5, wherein the antibody specifically binds BMP-6 with higher affinity than BMP-7. 7. The method of claim 6, wherein the antibody binds BMP-6 with at least 5-fold greater affinity than BMP-7. 8. The method of claim 6 wherein said BMP-6 is human BMP-6 and said BMP-7 is human BMP-7. 9. The method of claim 1, wherein the antibody binds to said BMP-6 with a Ka greater than or equal to about 104 M−1. 10. The method of claim 1, wherein administration of said composition reduces BMP-6 signaling. 11. The method of claim 1, wherein the mature BMP-6 comprises the amino acid sequence of SEQ ID NO: 2. 12. The method of claim 1, wherein the method increases serum iron. 13. The method of claim 1, wherein the composition is administered in an amount sufficient to inhibit an interaction between hemojuvelin and BMP-6. 14. The method of claim 1, wherein the subject is a human and the antibody preferably inhibits human BMP-6 over BMP-2, 4, 5, 7 or 9. 15. A method of inhibiting hepcidin expression in the liver of a subject, the method comprising administering to said subject an effective amount of a pharmaceutical composition comprising a monoclonal antibody that specifically binds bone morphogenetic protein-6 (BMP-6) or fragments thereof. 16. The method of claim 15, wherein administration of said composition inhibits liver hepcidin mRNA production. 17. The method of claim 15, wherein administration of said composition inhibits hepatic hepcidin expression. 18. The method of claim 15, wherein the subject is a human and administration of said composition inhibits hepcidin expression in human hepatic cells. 19. The method of claim 15, wherein administration of said composition increases serum transferrin saturation in the subject. 20. The method of claim 19, wherein the inhibition is in the presence of BMP-6 and BMP-2, BMP-4, BMP-5, BMP-7, or BMP-9. 21. The method of claim 15, wherein the antibody binds BMP-7. 22. The method of claim 21, wherein the antibody specifically binds BMP-6 with higher affinity than BMP-7. 23. The method of claim 22, wherein the antibody binds BMP-6 with at least 5-fold greater affinity than BMP-7. 24. The method of any one of claims 21, 22, or 23 wherein said BMP-6 is human BMP-6 and said BMP-7 is human BMP-7. 25. The method of claim 15, wherein the antibody binds to said BMP-6 with a Ka greater than or equal to about 104 M−1. 26. The method of claim 15, wherein administration of said composition reduces BMP-6 signaling. 27. The method of claim 15, wherein the mature BMP-6 comprises the amino acid sequence of SEQ ID NO: 2. 28. The method of claim 15, wherein the method increases serum iron. 29. The method of claim 15, wherein the composition is administered in an amount sufficient to inhibit an interaction between hemojuvelin and BMP-6. 30. The method of claim 15, wherein the subject is a human and the antibody preferably inhibits human BMP-6 over BMP-2, 4, 5, 7 or 9.
1,600
577
15,054,358
1,654
An apolipoprotein formulation is provided at a fixed dosage that is efficacious in the prophylactic and/or therapeutic treatment of diseases or conditions including, but not limited to cardiovascular disease, acute coronary syndrome, atherosclerosis, unstable angina pectoris, and myocardial infarction. More particularly, the fixed dosage apolipoprotein formulation displays relatively reduced inter-patient variability compared to weight-adjusted dosages. Typically, the apolipoprotein formulation is a reconstituted high density lipoprotein formulation comprising ApoA1, one or more lipids such as phosphatidylcholine, sphingomyelin and/or phophatidylglycerol and, optionally, a detergent such as cholate at a level that does not induce liver toxicity.
1. A method of prophylactically or therapeutically treating a disease, disorder or condition in a human in need thereof comprising administering to the human a fixed dosage apolipoprotein formulation in order to prophylactically or therapeutically treat the disease, disorder, or condition in the human, wherein the disease, disorder, or condition is selected from the group consisting of cardiovascular disease, hypercholesterolemia, and hypercholesterolemia. 2. (canceled) 3. (canceled) 4. The method according to claim 1, wherein the cardiovascular disease is selected from the group consisting of acute coronary syndrome (ACS), atherosclerosis, unstable angina pectoris, and myocardial infarction. 5. The method according to claim 1, wherein the apolipoprotein formulation is therapeutically effective upon administration to a human of any body weight, or of any body weight in a body weight range. 6. The method according to claim 5, wherein the body weight is in the body weight range 60-140 kg. 7. The method according to claim 1, wherein the apolipoprotein formulation displays relatively reduced inter-patient variability in exposure to the apolipoprotein of the formulation compared to that which would be observed or associated with a weight-adjusted dosage regime. 8. The method according to claim 7, wherein the inter-patient variability in exposure to apolipoprotein is less than 90% of that observed or associated with the weight-adjusted dosage regime. 9. The method according to claim 1, wherein the fixed dosage comprises an amount of apolipoprotein in a range selected from: 0.1 to 15 g; 1 to 10 g; 2 to 9 g; 3 to 8 g; 4 to 7 g; and 5 to 6 g of apolipoprotein. 10. The method according to claim 9, wherein the fixed dosage comprises 0.25 g, 0.5 g, 1 g, 1.7 g, 2 g, 3.4 g, 4 g, 5.1 g, kg, 6.8 g or 8 g apolipoprotein. 11. The method according to claim 1, wherein the apolipoprotein is Apo-A1 or a fragment thereof. 12. The method according to claim 1, wherein the apolipoprotein formulation is a reconstituted high density apolipoprotein (rHDL) formulation. 13. The method according to claim 12, wherein the rHDL formulation comprises the apolipoprotein and one or more lipids. 14. The method according to claim 13, further comprising a detergent. 15. The method according to claim 14, wherein the apolipoprotein is Apo A-1 purified from plasma, the lipid is phosphatidylcholine, and the detergent is sodium cholate. 16. The method according to claim 12, wherein the apolipoprotein is recombinant Apo A-I and the one or more lipids is a mixture of sphingomyelin and phosphatidylglycerol. 17. The method according to claim 16, wherein the ratio between the apolipoprotein and the lipid is between 1:80 and 1:120 (mol:mol) and the sphingomyelin and the phosphatidylglycerol are present in a ratio from 90:10 to 99:1 (w:w). 18-39. (canceled) 40. The method according to claim 15, wherein the ratio between the apolipoprotein and the lipid is from 1:40 to 1:75 (mol:mol), and the sodium cholate is present at a concentration of 0.5 to 0.9 g/L.
An apolipoprotein formulation is provided at a fixed dosage that is efficacious in the prophylactic and/or therapeutic treatment of diseases or conditions including, but not limited to cardiovascular disease, acute coronary syndrome, atherosclerosis, unstable angina pectoris, and myocardial infarction. More particularly, the fixed dosage apolipoprotein formulation displays relatively reduced inter-patient variability compared to weight-adjusted dosages. Typically, the apolipoprotein formulation is a reconstituted high density lipoprotein formulation comprising ApoA1, one or more lipids such as phosphatidylcholine, sphingomyelin and/or phophatidylglycerol and, optionally, a detergent such as cholate at a level that does not induce liver toxicity.1. A method of prophylactically or therapeutically treating a disease, disorder or condition in a human in need thereof comprising administering to the human a fixed dosage apolipoprotein formulation in order to prophylactically or therapeutically treat the disease, disorder, or condition in the human, wherein the disease, disorder, or condition is selected from the group consisting of cardiovascular disease, hypercholesterolemia, and hypercholesterolemia. 2. (canceled) 3. (canceled) 4. The method according to claim 1, wherein the cardiovascular disease is selected from the group consisting of acute coronary syndrome (ACS), atherosclerosis, unstable angina pectoris, and myocardial infarction. 5. The method according to claim 1, wherein the apolipoprotein formulation is therapeutically effective upon administration to a human of any body weight, or of any body weight in a body weight range. 6. The method according to claim 5, wherein the body weight is in the body weight range 60-140 kg. 7. The method according to claim 1, wherein the apolipoprotein formulation displays relatively reduced inter-patient variability in exposure to the apolipoprotein of the formulation compared to that which would be observed or associated with a weight-adjusted dosage regime. 8. The method according to claim 7, wherein the inter-patient variability in exposure to apolipoprotein is less than 90% of that observed or associated with the weight-adjusted dosage regime. 9. The method according to claim 1, wherein the fixed dosage comprises an amount of apolipoprotein in a range selected from: 0.1 to 15 g; 1 to 10 g; 2 to 9 g; 3 to 8 g; 4 to 7 g; and 5 to 6 g of apolipoprotein. 10. The method according to claim 9, wherein the fixed dosage comprises 0.25 g, 0.5 g, 1 g, 1.7 g, 2 g, 3.4 g, 4 g, 5.1 g, kg, 6.8 g or 8 g apolipoprotein. 11. The method according to claim 1, wherein the apolipoprotein is Apo-A1 or a fragment thereof. 12. The method according to claim 1, wherein the apolipoprotein formulation is a reconstituted high density apolipoprotein (rHDL) formulation. 13. The method according to claim 12, wherein the rHDL formulation comprises the apolipoprotein and one or more lipids. 14. The method according to claim 13, further comprising a detergent. 15. The method according to claim 14, wherein the apolipoprotein is Apo A-1 purified from plasma, the lipid is phosphatidylcholine, and the detergent is sodium cholate. 16. The method according to claim 12, wherein the apolipoprotein is recombinant Apo A-I and the one or more lipids is a mixture of sphingomyelin and phosphatidylglycerol. 17. The method according to claim 16, wherein the ratio between the apolipoprotein and the lipid is between 1:80 and 1:120 (mol:mol) and the sphingomyelin and the phosphatidylglycerol are present in a ratio from 90:10 to 99:1 (w:w). 18-39. (canceled) 40. The method according to claim 15, wherein the ratio between the apolipoprotein and the lipid is from 1:40 to 1:75 (mol:mol), and the sodium cholate is present at a concentration of 0.5 to 0.9 g/L.
1,600
578
13,962,241
1,651
Embodiments of the invention encompass the ex vivo production of cartilage from chondrocytes differentiated from fibroblasts or stem cells. In particular embodiments, fibroblasts are subjected to conditions to produce chondrocytes in the form of cartilage tissue, for example cartilage having a desired shape. In at least some embodiments, a mold for the desired shape of the cartilage is produced from imaging of a body region of an individual in need thereof, and the fibroblasts are seeded in the mold with particular conditions.
1. A method of generating cartilage ex vivo, comprising the step of subjecting fibroblasts or stem cells to conditions to differentiate said fibroblasts or stem cells into chondrocytes ex vivo to produce cartilage. 2. The method of claim 1, wherein the cartilage is configured in the form of a desired shape. 3. The method of claim 1, wherein said conditions comprise low oxygen, mechanical stress, or a combination thereof. 4. The method of claim 2, wherein said desired shape is at least part of an ear. 5. The method of claim 2, wherein said desired shape is at least part of a nose. 6. The method of claim 2, further comprising the step of generating a mold of the desired shape. 7. The method of claim 1, further comprising the step of providing the cartilage to an individual that is in need of cartilage repair. 8. The method of claim 2, wherein said desired shape is utilized to replace or repair cartilage in one or more regions of the body of an individual, wherein said region requires connective tissue. 9. The method of claim 1, further comprising the step of imaging a part of the body of an individual that is in need of cartilage repair or that is suspected of being in need of cartilage repair. 10. The method of claim 1, further comprising the step of imaging a part of the body of an individual that is in need of cartilage repair and generating therefrom a mold of a desired shape of cartilage. 11. The method of claim 1, further comprising the step of imaging a part of the body of an individual wherein that part is not in need of repair and using that image to generate a mold for growth of cartilage to replace or repair an area in need of repair. 12. The method of claim 7, wherein the cartilage is provided to the individual with one or more supports. 13. The method of claim 12, wherein the support is resorbable. 14. The method of claim 12, wherein the support is comprised of a material that would be resorbed by the body of the individual during and/or after its function of cartilage formation is completed. 15. The method of claim 12, wherein the support is non-resorbable. 16. The method of claim 15, wherein the support is comprised of metal or one or more other materials that may remain in the body and act as a scaffolding to maintain shape and function of the cartilage. 17. The method of claim 7, wherein the cartilage tissue is delivered to a nose, ear, knee, shoulder, elbow or other area of the body where connective tissue is required for the individual. 18. The method of claim 7, wherein the cartilage tissue is not delivered to a joint. 19. The method of claim 7, wherein the cartilage tissue is not delivered to a vertebral disc.
Embodiments of the invention encompass the ex vivo production of cartilage from chondrocytes differentiated from fibroblasts or stem cells. In particular embodiments, fibroblasts are subjected to conditions to produce chondrocytes in the form of cartilage tissue, for example cartilage having a desired shape. In at least some embodiments, a mold for the desired shape of the cartilage is produced from imaging of a body region of an individual in need thereof, and the fibroblasts are seeded in the mold with particular conditions.1. A method of generating cartilage ex vivo, comprising the step of subjecting fibroblasts or stem cells to conditions to differentiate said fibroblasts or stem cells into chondrocytes ex vivo to produce cartilage. 2. The method of claim 1, wherein the cartilage is configured in the form of a desired shape. 3. The method of claim 1, wherein said conditions comprise low oxygen, mechanical stress, or a combination thereof. 4. The method of claim 2, wherein said desired shape is at least part of an ear. 5. The method of claim 2, wherein said desired shape is at least part of a nose. 6. The method of claim 2, further comprising the step of generating a mold of the desired shape. 7. The method of claim 1, further comprising the step of providing the cartilage to an individual that is in need of cartilage repair. 8. The method of claim 2, wherein said desired shape is utilized to replace or repair cartilage in one or more regions of the body of an individual, wherein said region requires connective tissue. 9. The method of claim 1, further comprising the step of imaging a part of the body of an individual that is in need of cartilage repair or that is suspected of being in need of cartilage repair. 10. The method of claim 1, further comprising the step of imaging a part of the body of an individual that is in need of cartilage repair and generating therefrom a mold of a desired shape of cartilage. 11. The method of claim 1, further comprising the step of imaging a part of the body of an individual wherein that part is not in need of repair and using that image to generate a mold for growth of cartilage to replace or repair an area in need of repair. 12. The method of claim 7, wherein the cartilage is provided to the individual with one or more supports. 13. The method of claim 12, wherein the support is resorbable. 14. The method of claim 12, wherein the support is comprised of a material that would be resorbed by the body of the individual during and/or after its function of cartilage formation is completed. 15. The method of claim 12, wherein the support is non-resorbable. 16. The method of claim 15, wherein the support is comprised of metal or one or more other materials that may remain in the body and act as a scaffolding to maintain shape and function of the cartilage. 17. The method of claim 7, wherein the cartilage tissue is delivered to a nose, ear, knee, shoulder, elbow or other area of the body where connective tissue is required for the individual. 18. The method of claim 7, wherein the cartilage tissue is not delivered to a joint. 19. The method of claim 7, wherein the cartilage tissue is not delivered to a vertebral disc.
1,600
579
15,405,968
1,628
The present invention relates to a new use of tetrahydroxylated estrogens such as estetrol (1,3,5(10)-estratrien-3, 15α, 16α, 17β-tetrol), namely in a method of emergency contraception. The method of emergency contraception according to the invention comprises the oral administration of estetrol in a single dose within 120 hours of sexual intercourse.
1. A method of emergency contraception in a mammalian female comprising the administration of an emergency contraceptive comprising tetrahydroxylated estrogen represented by the general formula (I): wherein R1, R2, R3, R4 independently are a hydrogen atom; a hydroxyl group, an alkoxy group with 1-5 carbon atoms, or a keto group; wherein each of R5, R6, R7 is a hydroxyl group; and wherein no more than 3 of R1, R2, R3, R4 are hydrogen atoms, in an effective amount to inhibit ovulation and pregnancy in a female mammal. 2. The method according to claim 1, wherein said administration is performed through non-vaginal administration 3. The method according to any one of claim 1 or 2, wherein said administration is performed through oral, parenteral, rectal, transcutaneous or topical administration. 4. The method according to any one of claims 1 to 3, wherein said emergency contraceptive additionally comprises a pharmaceutically acceptable salt of said tetrahydroxylated estrogen or excipient. 5. The method according to any one of claims 1 to 4, wherein said emergency contraceptive additionally comprises one or more additional ingredients. 6. The method according to any one of claims 1 to 5, wherein said tetra-hydroxylated estrogen is estetrol (1,3,5(10)-estratrien-3, 15α, 16α, 17β-tetrol). 7. The method according to any one of claims 1 to 6, wherein said emergency contraceptive comprises the tetra-hydroxylated estrogen in a dose of between 0.5 and 7 mg/kg, between 1.0 to 5.0 mg/kg, between 1.5 to 2.5 mg/kg, or of about 2.0 mg/kg 8. The method according to any one of claims 1 to 6, wherein said emergency contraceptive comprises the tetra-hydroxylated estrogen in a dose of between 30 mg and 400 mg, between 40 mg and 250 mg, between 40 mg and 200 mg, between 40 mg and 150 mg, between 40 mg and 100 mg, between 50 mg and 100 mg of estetrol, or of about 100 mg. 9. The method according to any one of claims 1 to 8, wherein the administration is performed in a single dosage. 10. The method according to claim 1 to 8, wherein the administration is performed in a double dosage, e.g. with an interval of 6, 12, 18, or 24 hours. 11. The method according to any one of claims 1 to 10, wherein said administration is performed within 120 hours after sexual intercourse, preferably within 96, 72, 48, 24, 12 hours or less.
The present invention relates to a new use of tetrahydroxylated estrogens such as estetrol (1,3,5(10)-estratrien-3, 15α, 16α, 17β-tetrol), namely in a method of emergency contraception. The method of emergency contraception according to the invention comprises the oral administration of estetrol in a single dose within 120 hours of sexual intercourse.1. A method of emergency contraception in a mammalian female comprising the administration of an emergency contraceptive comprising tetrahydroxylated estrogen represented by the general formula (I): wherein R1, R2, R3, R4 independently are a hydrogen atom; a hydroxyl group, an alkoxy group with 1-5 carbon atoms, or a keto group; wherein each of R5, R6, R7 is a hydroxyl group; and wherein no more than 3 of R1, R2, R3, R4 are hydrogen atoms, in an effective amount to inhibit ovulation and pregnancy in a female mammal. 2. The method according to claim 1, wherein said administration is performed through non-vaginal administration 3. The method according to any one of claim 1 or 2, wherein said administration is performed through oral, parenteral, rectal, transcutaneous or topical administration. 4. The method according to any one of claims 1 to 3, wherein said emergency contraceptive additionally comprises a pharmaceutically acceptable salt of said tetrahydroxylated estrogen or excipient. 5. The method according to any one of claims 1 to 4, wherein said emergency contraceptive additionally comprises one or more additional ingredients. 6. The method according to any one of claims 1 to 5, wherein said tetra-hydroxylated estrogen is estetrol (1,3,5(10)-estratrien-3, 15α, 16α, 17β-tetrol). 7. The method according to any one of claims 1 to 6, wherein said emergency contraceptive comprises the tetra-hydroxylated estrogen in a dose of between 0.5 and 7 mg/kg, between 1.0 to 5.0 mg/kg, between 1.5 to 2.5 mg/kg, or of about 2.0 mg/kg 8. The method according to any one of claims 1 to 6, wherein said emergency contraceptive comprises the tetra-hydroxylated estrogen in a dose of between 30 mg and 400 mg, between 40 mg and 250 mg, between 40 mg and 200 mg, between 40 mg and 150 mg, between 40 mg and 100 mg, between 50 mg and 100 mg of estetrol, or of about 100 mg. 9. The method according to any one of claims 1 to 8, wherein the administration is performed in a single dosage. 10. The method according to claim 1 to 8, wherein the administration is performed in a double dosage, e.g. with an interval of 6, 12, 18, or 24 hours. 11. The method according to any one of claims 1 to 10, wherein said administration is performed within 120 hours after sexual intercourse, preferably within 96, 72, 48, 24, 12 hours or less.
1,600
580
15,414,722
1,642
The present invention concerns the use of VCAM-1, ICAM-1 and/or PIGF as biomarkers for predicting the outcome of the treatment with aflibercept, or ziv-aflibercept of a patient suspected to suffer from a cancer.
1-3. (canceled) 4. A method for treating a patient, wherein the patient has colon cancer, a colorectal cancer or a rectal cancer, comprising administering a therapeutically effective amount of aflibercept or ziv-aflibercept to the patient, wherein a level of a biomarker selected from the group consisting of VCAM-1, ICAM-1, and PIGFdetected in a biological sample obtained from the patient is lower than a reference level of expression of the biomarker. 5. A method for treating a patient, wherein the patient has colon cancer, a colorectal cancer or a rectal cancer, comprising administering a therapeutically effective amount of aflibercept or ziv-aflibercept to the patient, wherein levels of VCAM-1 and ICAM-1 detected in a biological sample obtained from the patient are lower than respective reference levels of expression of VCAM-1 and ICAM-1. 6. (canceled) 7. A method according to claim 4, wherein the reference level of expression of VCAM-1 is between 406 and 577 ng/mL. 8. A method according to claim 4, wherein the reference level of expression of VCAM-1 is 553 ng/mL. 9. A method according to claim 4, wherein the reference level of expression of ICAM-1 is between 92 and 145 ng/mL. 10. A method according to claim 4, wherein the reference level of on expression of ICAM-1 is 144 ng/mL. 11. A method according to claim 4, wherein the reference level of expression of PIGF is between 12 and 19 pg/mL. 12. A method according to claim 4, wherein the reference level of expression of PIGF is 17 pg/mL. 13. A method according to claim 4, wherein the biological sample is selected from the group consisting of blood, serum, and plasma. 14. (canceled) 15. A method according to claim 4 wherein the colorectal cancer is a metastatic colorectal cancer. 16. A method according to claim 4, wherein the VCAM-1, ICAM-1 and/or PIGF level which is determined is a circulating level. 17. A method according to claim 4, comprising administering to said patient therapeutically effective amounts of aflibercept, or ziv-aflibercept, folinic acid, 5-fluorouracil (5-FU) and irinotecan. 18. (canceled) 19. A kit for predicting whether a patient suspected to suffer from cancer is a candidate for aflibercept, or ziv-aflibercept therapy, which kit comprises: a) means for measuring the level of a biomarker selected from the group consisting of VCAM-1, ICAM-1 and PIGF; and b) optionally, a label giving instructions for the use of said kit in predicting whether a patient suspected to suffer from cancer is a candidate for aflibercept, or ziv-aflibercept therapy. 20. An article of manufacture comprising: a) a packaging material; b) means for measuring the level of a biomarker selected from the group consisting of VCAM-1, ICAM-1 and PIGF; and c) a label giving instructions for the use of said kit in predicting whether a a patient suspected to suffer from cancer is a candidate for aflibercept, or ziv-aflibercept therapy.
The present invention concerns the use of VCAM-1, ICAM-1 and/or PIGF as biomarkers for predicting the outcome of the treatment with aflibercept, or ziv-aflibercept of a patient suspected to suffer from a cancer.1-3. (canceled) 4. A method for treating a patient, wherein the patient has colon cancer, a colorectal cancer or a rectal cancer, comprising administering a therapeutically effective amount of aflibercept or ziv-aflibercept to the patient, wherein a level of a biomarker selected from the group consisting of VCAM-1, ICAM-1, and PIGFdetected in a biological sample obtained from the patient is lower than a reference level of expression of the biomarker. 5. A method for treating a patient, wherein the patient has colon cancer, a colorectal cancer or a rectal cancer, comprising administering a therapeutically effective amount of aflibercept or ziv-aflibercept to the patient, wherein levels of VCAM-1 and ICAM-1 detected in a biological sample obtained from the patient are lower than respective reference levels of expression of VCAM-1 and ICAM-1. 6. (canceled) 7. A method according to claim 4, wherein the reference level of expression of VCAM-1 is between 406 and 577 ng/mL. 8. A method according to claim 4, wherein the reference level of expression of VCAM-1 is 553 ng/mL. 9. A method according to claim 4, wherein the reference level of expression of ICAM-1 is between 92 and 145 ng/mL. 10. A method according to claim 4, wherein the reference level of on expression of ICAM-1 is 144 ng/mL. 11. A method according to claim 4, wherein the reference level of expression of PIGF is between 12 and 19 pg/mL. 12. A method according to claim 4, wherein the reference level of expression of PIGF is 17 pg/mL. 13. A method according to claim 4, wherein the biological sample is selected from the group consisting of blood, serum, and plasma. 14. (canceled) 15. A method according to claim 4 wherein the colorectal cancer is a metastatic colorectal cancer. 16. A method according to claim 4, wherein the VCAM-1, ICAM-1 and/or PIGF level which is determined is a circulating level. 17. A method according to claim 4, comprising administering to said patient therapeutically effective amounts of aflibercept, or ziv-aflibercept, folinic acid, 5-fluorouracil (5-FU) and irinotecan. 18. (canceled) 19. A kit for predicting whether a patient suspected to suffer from cancer is a candidate for aflibercept, or ziv-aflibercept therapy, which kit comprises: a) means for measuring the level of a biomarker selected from the group consisting of VCAM-1, ICAM-1 and PIGF; and b) optionally, a label giving instructions for the use of said kit in predicting whether a patient suspected to suffer from cancer is a candidate for aflibercept, or ziv-aflibercept therapy. 20. An article of manufacture comprising: a) a packaging material; b) means for measuring the level of a biomarker selected from the group consisting of VCAM-1, ICAM-1 and PIGF; and c) a label giving instructions for the use of said kit in predicting whether a a patient suspected to suffer from cancer is a candidate for aflibercept, or ziv-aflibercept therapy.
1,600
581
14,704,254
1,641
Management of the health status of an animal colony using a plurality of blood collection cards and the analysis of dried blood from members of the colony that has been collected on the cards. Members of the colony may be removed from the colony as a result of the analysis.
1. A method for managing a an animal colony, the method comprising: (a) collecting blood samples from a plurality of members of the colony on a plurality of collection cards; (b) allowing the blood samples to dry on the collection cards; (c) transporting the collection cards to a laboratory as a single unit; (d) extracting the samples from the cards; (e) analyzing the samples for the presence or absence of a biological marker for an infectious disease; and (f) removing one or more of the members from the colony based upon the presence or absence of the marker in the samples from the one or more members. 2. The method of claim 1, further comprising labeling each collection card in the plurality of collection cards to identify the member of the population associated with each sample on a card. 3. The method of claim 1, wherein the immunoassay comprises contacting the extracted blood with a fluorescently labeled binding partner for the antibody. 4. The method of claim 1, wherein the analyzing comprises conducting a multiplex fluorescence immunoassay on the samples extracted from the cards. 5. The method of claim 4, wherein the multiplex fluorescence immunoassay detects at least ten different analytes in the samples. 6. The method of claim 1, wherein the rodents are mice and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. MHV; b. MVN (MMV); c. NS1 (Generic Parvovirus); d. MPV (MPV1-5); e. MNV; f. TMEV; g. EDIM; h. Sendai virus; i. Mycoplasma pulmonis; j. PVM; k. REO3; l. LCMV; m. Ectromelia virus; n. MAD1; o. MAD2; p. Polyoma virus; q. Encephalitozoon cuniculi; r. CARB; s. Clostridium piliforme; t. MCMV; u. K virus; v. Hantaan virus; w. Lactate dehydrogenase-elevating virus; and x. MTV. 7. The method of claim 1, wherein the rodents are rats and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. RCV; b. NS1 (Generic Parvovirus); c. RPV; d. RMV; e. KRV; f. H-1; g. RTV (Rat theilovirus); h. Sendai virus; i. PVM; j. Mycoplasma pulmonis; k. REO3; l. LCMV; m. CARB; n. Hantaan virus; o. Clostridim piliforme; p. MAD1; q. MAD2; r. Encephalitozoom cuniculi; and s. IDIR. 8. The method of claim 4, wherein the rodents are mice and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. MHV; b. MVN (MMV); c. NS1 (Generic Parvovirus); d. MPV (MPV1-5); e. MNV; f. TMEV; g. EDIM; h. Sendai virus; i. Mycoplasma pulmonis; j. PVM; k. REO3; l. LCMV; m. Ectromelia virus; n. MAD1; o. MAD2; p. Polyoma virus; q. Encephalitozoon cuniculi; r. CARB; s. Clostridium piliforme; t. MCMV; u. K virus; v. Hantaan virus; w. Lactate dehydrogenase-elevating virus; and x. MTV. 9. The method of claim 4, wherein the rodents are rats and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. RCV; b. NS1 (Generic Parvovirus); c. RPV; d. RMV; e. KRV; f. H-1; g. RTV (Rat theilovirus); h. Sendai virus; i. PVM; j. Mycoplasma pulmonis; k. REO3; l. LCMV; m. CARB; n. Hantaan virus; o. Clostridim piliforme; p. MAD1; q. MAD2; r. Encephalitozoom cuniculi; and s. IDIR. 10. The method of claim 1, where in the extracting comprises extracting the blood with a buffer comprising ethylenediaminetetraacetic acid (EDTA). 11. The method of claim 4, where in the extracting comprises extracting the blood with a buffer comprising ethylenediaminetetraacetic acid (EDTA). 12. The method of claim 1, wherein each collection card contains segments for collecting up to five samples. 13. The method of claim 1, wherein each collection card contains segments for collecting up to fifty samples. 14. The method of claim 1, wherein each collection card contains segments for collecting up to 100 samples. 15. The method of claim 1, wherein the volume of blood of each sample collected on the card is 10-40 μL. 16. The method of claim 1, wherein the blood is collected from the animal's lateral saphenous vein, facial vein or temporal vein. 17. The method of claim 16, wherein the blood is collected on the card without a collection device. 18. The method of claim 1, wherein the samples are dried at room temperature. 19. The method of claim 1, wherein the collection cards are transported at room temperature. 20. The method of claim 12, wherein the segments are separated by a perforation that allows separation of the cards such that a single sample collection area is on each separated card. 21. The method of claim 20, wherein the identity of at least one of the rodent population and an individual member of the population is associated with each sample area of each separated card.
Management of the health status of an animal colony using a plurality of blood collection cards and the analysis of dried blood from members of the colony that has been collected on the cards. Members of the colony may be removed from the colony as a result of the analysis.1. A method for managing a an animal colony, the method comprising: (a) collecting blood samples from a plurality of members of the colony on a plurality of collection cards; (b) allowing the blood samples to dry on the collection cards; (c) transporting the collection cards to a laboratory as a single unit; (d) extracting the samples from the cards; (e) analyzing the samples for the presence or absence of a biological marker for an infectious disease; and (f) removing one or more of the members from the colony based upon the presence or absence of the marker in the samples from the one or more members. 2. The method of claim 1, further comprising labeling each collection card in the plurality of collection cards to identify the member of the population associated with each sample on a card. 3. The method of claim 1, wherein the immunoassay comprises contacting the extracted blood with a fluorescently labeled binding partner for the antibody. 4. The method of claim 1, wherein the analyzing comprises conducting a multiplex fluorescence immunoassay on the samples extracted from the cards. 5. The method of claim 4, wherein the multiplex fluorescence immunoassay detects at least ten different analytes in the samples. 6. The method of claim 1, wherein the rodents are mice and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. MHV; b. MVN (MMV); c. NS1 (Generic Parvovirus); d. MPV (MPV1-5); e. MNV; f. TMEV; g. EDIM; h. Sendai virus; i. Mycoplasma pulmonis; j. PVM; k. REO3; l. LCMV; m. Ectromelia virus; n. MAD1; o. MAD2; p. Polyoma virus; q. Encephalitozoon cuniculi; r. CARB; s. Clostridium piliforme; t. MCMV; u. K virus; v. Hantaan virus; w. Lactate dehydrogenase-elevating virus; and x. MTV. 7. The method of claim 1, wherein the rodents are rats and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. RCV; b. NS1 (Generic Parvovirus); c. RPV; d. RMV; e. KRV; f. H-1; g. RTV (Rat theilovirus); h. Sendai virus; i. PVM; j. Mycoplasma pulmonis; k. REO3; l. LCMV; m. CARB; n. Hantaan virus; o. Clostridim piliforme; p. MAD1; q. MAD2; r. Encephalitozoom cuniculi; and s. IDIR. 8. The method of claim 4, wherein the rodents are mice and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. MHV; b. MVN (MMV); c. NS1 (Generic Parvovirus); d. MPV (MPV1-5); e. MNV; f. TMEV; g. EDIM; h. Sendai virus; i. Mycoplasma pulmonis; j. PVM; k. REO3; l. LCMV; m. Ectromelia virus; n. MAD1; o. MAD2; p. Polyoma virus; q. Encephalitozoon cuniculi; r. CARB; s. Clostridium piliforme; t. MCMV; u. K virus; v. Hantaan virus; w. Lactate dehydrogenase-elevating virus; and x. MTV. 9. The method of claim 4, wherein the rodents are rats and the step of analyzing the extracted blood comprises analyzing for the presence or absence of seven or more diseases selected from the group consisting of: a. RCV; b. NS1 (Generic Parvovirus); c. RPV; d. RMV; e. KRV; f. H-1; g. RTV (Rat theilovirus); h. Sendai virus; i. PVM; j. Mycoplasma pulmonis; k. REO3; l. LCMV; m. CARB; n. Hantaan virus; o. Clostridim piliforme; p. MAD1; q. MAD2; r. Encephalitozoom cuniculi; and s. IDIR. 10. The method of claim 1, where in the extracting comprises extracting the blood with a buffer comprising ethylenediaminetetraacetic acid (EDTA). 11. The method of claim 4, where in the extracting comprises extracting the blood with a buffer comprising ethylenediaminetetraacetic acid (EDTA). 12. The method of claim 1, wherein each collection card contains segments for collecting up to five samples. 13. The method of claim 1, wherein each collection card contains segments for collecting up to fifty samples. 14. The method of claim 1, wherein each collection card contains segments for collecting up to 100 samples. 15. The method of claim 1, wherein the volume of blood of each sample collected on the card is 10-40 μL. 16. The method of claim 1, wherein the blood is collected from the animal's lateral saphenous vein, facial vein or temporal vein. 17. The method of claim 16, wherein the blood is collected on the card without a collection device. 18. The method of claim 1, wherein the samples are dried at room temperature. 19. The method of claim 1, wherein the collection cards are transported at room temperature. 20. The method of claim 12, wherein the segments are separated by a perforation that allows separation of the cards such that a single sample collection area is on each separated card. 21. The method of claim 20, wherein the identity of at least one of the rodent population and an individual member of the population is associated with each sample area of each separated card.
1,600
582
11,658,220
1,643
Early detection of tumors is a major determinant of survival of patients suffering from tumors, including bladder tumors. Members of the BTM or UBTM family can be highly and consistently accumulated in bladder tumor tissue and other tumor tissue, and/or can be accumulated in urine of patients, and thus are markers for bladder and other types of cancer. In certain embodiments, BTMs or UBTMs can accumulate in the urine, and detection of UBTM family members can be an effective diagnostic approach. In some embodiments, quantitative PCR methods have advantages over microarray methods. In other embodiments, detection and quantification of a plurality of BTMs or UBTMs can increase the sensitivity and specificity of detection of bladder cancer, and therefore provides methods for determining the stage and type of bladder cancer. Kits provide easy, convenient ways for carrying out the methods of this invention.
1. A method for detecting bladder cancer in a subject, comprising: detecting the accumulation of a urinary bladder tumor marker (“UBTM”) family member in the urine, said accumulation in said subject greater than about 1.2 times the accumulation of said UBTM in urine from a group of normal subjects not having malignant bladder cancer. 2. The method of claim 1, wherein said UBTM family member is not associated with blood to a substantial extent. 3. The method according to claim 1, wherein the UBTM is selected from the group consisting of GGH, SPP1, NRN1, SPARC, ADAMTS10, CNTN1, TLL2, PDIR, FBN1, KIAA0100 gene product, CALR, ITGBL1, ELA3B, SMOC2, HEXA, IGFBP7, MFAP2, CILP, OLFM1, LUM, SEM2, PRSS11, SULF1, SERPINH1, MGP, TIMP1, EGFL6, SPAG11, IGFBP5, SEMA3F, CDC2, TOP2A, UBE2C, STMN1, TUBA4, HIST1H1B, HMGB2, CCNA2, CDCA1, hypothetical protein MGC5576, DEK, MLF1IP, CDCA8, hypothetical protein FLJ20647, TYMS, SMC4L1, LYN, HMGB3, PTGIR, DONSON, HMMR, CLDN6, HIST1H1D, C10orf3, KNTC1, CKS1B, RRM2, HIST1H2BH, STK6, MPHOSPH1, CCNB2, GPR32, ENG, MFHAS1, HIST1H1C, AVPR2, CENPF, HOXA13, h4 histone family member g, MGC27121 gene, NP, ASPM, hypothetical protein FLJ11871, LBH, NUDT1, HELLS, ASB9, MCM5, IMP-2, DKFZP566M1046, TUBA2, GAS2L3, hypothetical protein FLJ12442, MCM6, DOK3, WDR18, CKAP2, KIF20A, putative fap protein, C6orf32, NEK2, CRY1, TGM2, DLG7, EIF2C2, DEPDC1, HIST2H4, MCM7, MTAP, KNTC2, HSPC150, SMC6L1, HIST1H2BC, ASF1B, ARH, LMNB1, hypothetical protein FLJ10719, hypothetical protein FLJ 10706, MAD2L1, SLC22A2, hypothetical protein MGC34923, SPAG5, ACVRL1, DSCR1, PRSS15, S100A9, MCM4, ST7L, PLEKHA4, EPHB1, CALD1, SMC1L1, Thy-1 co-transcribed, RAMP, FKBP11, C20orf129, HIST1H4H, CDKN3, MCAM, SNCAIP, NIPSNAP1, AP1M1, ANLN, C6orf69, TORC3, MAZ, TXNRD1, hypothetical protein xp 096695, C22orf4, VSNL1, similar to Carboxypeptidase N 83 kDa chain, KIAA1598, hypothetical protein FLJ13501, DKFZP4340047, hypothetical protein FLJ38716, similar to hypothetical protein (L1H3 region), hypothetical protein KIAA1875, PRIM1, hypothetical protein BC001096, MCM2, GJA3, C11orf30, similar to hypothetical protein FLJ30672, THY1, LRP3, LASS2, C18orf8, ZNF81, NARF, MTHFD2, D6T, SIAT7D, MMPL1, KLK11, KPNA2, FGFR1OP2, VIM, FLJ44108 protein, PAPOLG, FHOD1, RASL12, HMGN2, PITPNM2, DER1, EPHA4, VSIG1, RGS5, KIAA1639 protein, SH2B, PGLYRP4, CDC45L, MLSTD1, hypothetical protein MGC11266, TNFRSF13B, NET1, LHFPL5, MX2, SPHK1, ABCG4, SERPINB2, GALNT10, LEPR, MXD4, FAPP2, NUP210, CSK, NRP1, MGAT1, KIAA0100 gene product, LCN7, BMP7, ADAMTS10, PM5, NOMO3, CPA6, NPPC, hypothetical protein FLJ23221, ERP70, GALNT14, ITIH3, PAPPA2, LOXL1, TNFRSF6B, SPARC, MSMB, CLDN6, PTMA, AVPR2, similar to sodium- and chloride-dependent creatine transporter, TMEM19, hypothetical protein xp 047287, hypothetical protein FLJ11871, PROSC, MGC27121 gene, NQO1, CKAP4, hypothetical protein BC001096, PDPK1, regulator of mitotic spindle assembly 1, MIRAB13, PORCN, SIX6, GJB2, FLJ35784 protein, SLC37A3, SPRY4, LHX3, C7orf27, SLC39A1, ZNF307, MIF, BST2, PSTPIP1, SOX4, NCOA5, hypothetical protein FLJ31438, ODD, SLC23A2, SHFM1, SRPK2, RAMP2, BPGM, RGS5, CXADR, MEIS2, TENS1, SNAI2, CHST2, HCA127, Thy-1 co-transcribed (LOC94105), LRFN3, hypothetical protein FLJ22390, TRIB2, KRTHA3B, KIF21A, ANKRD17, RAG1, NUBP2, hypothetical protein FLJ20489, CASK, HIP1, PRKCDBP, TIE, C5orf15, CGI-72, ENTPD8, SH3BGRL3, NADH:ubiquinone oxidoreductase MLRQ subunit homolog, VG5Q, BG1, BCL2L11, ARK5, TLE3, ITIH5, RGS11, TM7SF3, SCRN3, PLXNA1, GJA4, hypothetical protein DKFZp434G1415, WSB2, CDA, GART, ZMPSTE24, TMEM33, GPI, hypothetical protein FLJ11000, CAMK1D, PTPN21, and TNS. 4. The method of claim 1, wherein said step of detecting is carried out by detecting accumulation of a bladder tumor marker (“BTM”) or UBTM mRNA. 5. The method of claim 1, wherein said detecting is carried out using a microarray. 6. The method of claim 1, wherein said detecting is carried out using quantitative polymerase chain reaction or hybridization methods. 7. The method of claim 1, wherein said step of detecting is carried out by detecting accumulation of a UBTM protein. 8. The method of claim 7, wherein said step of detecting is carried out by detecting accumulation of a UBTM peptide. 9. The method of claim 7, wherein said step of detecting is carried out using a UBTM antibody. 10. (canceled) 11. (canceled) 12. The method of claim 1, wherein the method includes detection of accumulation of two or more UBTM family members in said sample. 13. (canceled) 14. The method of claim 1, further comprising detecting one or more pairs of markers selected from the group consisting of TOP2A-HOXA13, TOP2A-IGFBP5, TOP2A-SEMA3F and CDC2-HOXA13. 15. A method for detecting bladder cancer, comprising: detecting the accumulation of a combination of two or more BTM family members selected from the group consisting of TOP2a and CDC2 (TOP2a-CDC2), SPAG5-TOP2A, SPAG5-CDC2, NRP1-UBE2c, SPAG5-MDK, TOP2a-THY1, SPAG5-CHGA, TOP2a-CHGA, SPAG5-THY1, TOP2a-MDK, CDC2-CHGA, TOP2a-ENG, TOP2a-MGP, SPAG5-NRP1, SPAG5-BIRC5, TOP2a-NRP1, SPAG5-MGP, SPAG5-EGFL6, TOP2a-HOXA13, TOP2a-EGFL6, TOP2a-BIRC5, SPAG5-ENG, SPAG5-HOXA13, CDC2-THY1, CDC2-MDK, SPAG5-SEM2, TOP2a-SMC4L1, TOP2a-SEM2, CDC2-NRP1, CDC2-MGP, CDC2-ENG, TOP2a-UBE2c, SPAG5-UBE2c, CDC2-EGFL6, CDC2-BIRC5, SPAG5-SMC4L1, CDC2-HOXA13, CDC2-SEM2, NRP1-MDK, NRP1-THY1, CDC2-SMC4L1, CDC2-UBE2c, CDC2-TOP2a, TOP2a-SPAG5, CDC2-SPAG5, SPAG5 and TOP2a and CDC2 (SPAG5-TOP2a-CDC5), SPAG5-TOP2a-NRP1, TOP2a-CDC2-NRP1, SPAG5-TOP2a-CHGA, SPAG5-CDC2-NRP1, TOP2a-CDC2-CHGA, SPAG5-TOP2a-SEM2, SPAG5-CDC2-CHGA, SPAG5-TOP2a-THY1, SPAG5-TOP2a-SMC4L1, SPAG5-TOP2a-MGP, SPAG5-TOP2a-MDK, SPAG5-TOP2a-ENG, SPAG5-TOP2a-UBE2c, SPAG5-TOP2a-EGFL6, TOP2a-CDC2-SEM2, SPAG5-TOP2a-BIRC5, TOP2a-CDC2-THY1, SPAG5-TOP2a-HOXA13, TOP2a-CDC2-SMC4L1, SPAG5-CDC2-SEM2, TOP2a-CDC2-MGP, TOP2a-CDC2-MDK, TOP2a-CDC2-ENG, SPAG5-CDC2-THY1, TOP2a-CDC2-UBE2c, SPAG5-CDC2-SMC4L1, SPAG5-CDC2-MGP, SPAG5-CDC2-MDK, SPAG5-CDC2-ENG, TOP2a-CDC2-EGFL6, SPAG5-CDC2-UBE2C TOP2a-CDC2-BIRC5, TOP2a-CDC2-HOXA13, SPAG5-CDC2-EGFL6, SPAG5-CDC2-BIRC5, SPAG5-CDC2-HOXA13, TOP2a-NRP1-CHGA, SPAG5-NRP1-CHGA, TOP2a-NRP1-SEM2, CDC2-NRP1-CHGA, TOP2a-NRP1-THY1, TOP2a-NRP1-SMC4L1, SPAG5-NRP1-SEM2, TOP2a-NRP1-MGP, TOP2a-NRP1-MDK, TOP2a-ENG-NRP1, SPAG5-NRP1-THY1, TOP2a-NRP1-UBE2c, SPAG5-NRP1-SMC4L1, SPAG5-NRP1-MGP, SPAG5-NRP1-MDK, SPAG5-ENG-NRP1, TOP2a-NRP1-EGFL6, SPAG5-NRP1-UBE2c, TOP2a-SEM2-CHGA, TOP2a-NRP1-BIRC5, TOP2a-CHGA-THY1, TOP2a-NRP1-HOXA13, SPAG5-NRP1-EGFL6, CDC2-NRP1-SEM2, TOP2a-CHGA-SMC4L1, SPAG5-SEM2-CHGA, SPAG5-NRP1-BIRC5, TOP2a-MGP-CHGA, CDC2-NRP1-THY1, TOP2a-CHGA-MDK, TOP2a-ENG-CHGA, SPAG5-CHGA-THY1, SPAG5-NRP1-HOXA13, CDC2-NRP1-SMC4L1, TOP2a-CHGA-UBE2c, CDC2-NRP1-MGP, SPAG5-CHGA-SMC4L1, CDC2-NRP1-MDK, SPAG5-MGP-CHGA, CDC2-ENG-NRP1, SPAG5-CHGA-MDK, SPAG5-ENG-CHGA, CDC2-NRP1-UBE2c, TOP2a-EGFL6-CHGA, SPAG5-CHGA-UBE2c, TOP2a-CHGA-BIRC5, TOP2a-CHGA-BIRC5, CDC2-NRP1-EGFL6, TOP2a-CHGA-HOXA13, SPAG5-EGFL6-CHGA, CDC2-SEM2-CHGA, CDC2-NRP1-BIRC5, SPAG5-CHGA-BIRC5, TOP2a-SEM2-THY1, CDC2-CHGA-THY1, CDC2-NRP1-HOXA13, SPAG5-CHGA-HOXA13, TOP2a-SEM2-SMC4L1, CDC2-CHGA-SMC4L1, TOP2a-MGP-SEM2, CDC2-MGP-CHGA, TOP2a-SEM2-MDK, CDC2-CHGA-MDK, TOP2a-THY1-SMC4L1, TOP2a-ENG-SEM2, SPAG5-SEM2-THY1, CDC2-ENG-CHGA, TOP2a-MGP-THY1, TOP2a-MDK-THY1, TOP2a-SEM2-UBE2c, TOP2a-ENG-THY1, CDC2-CHGA-UBE2c, SPAG5-SEM2-SMC4L1, TOP2a-MGP-SMC4L1, SPAG5-MGP-SEM2, TOP2a-MDK-SMC4L1, SPAG5-SEM2-MDK, TOP2a-UBE2c-THY1, TOP2a-MGP-MDK, TOP2a-ENG-SMC4L1, SPAG5-THY1-SMC4L1, SPAG5-ENG-SEM2, TOP2a-ENG-MGP, SPAG5-MGP-THY1, TOP2a-ENG-MDK, SPAG5-MDK-THY1, TOP2a-UBE2c-SMC4L1, TOP2a-EGFL6-SEM2, SPAG5-SEM2-UBE2c, SPAG5-ENG-THY1, CDC2-EGFL6-CHGA, TOP2a-MGP-UBE2c, SPAG5-MGP-SMC4L1, TOP2a-UBE2c-MDK, SPAG5-MDK-SMC4L1, TOP2a-EGFL6-THY1, TOP2a-ENG-UBE2c, TOP2a-SEM2-BIRC5, SPAG5-UBE2c-THY1, CDC2-CHGA-BIRC5, SPAG5-MGP-MDK, SPAG5-ENG-SMC4L1, SPAG5-ENG-MGP, SPAG5-ENG-MDK, TOP2a-BIRC5-THY1, TOP2a-SEM2-HOXA13, TOP2a-EGFL6-SMC4L1, SPAG5-UBE2c-SMC4L1, SPAG5-EGFL6-SEM2, CDC2-CHGA-HOXA13, TOP2a-EGFL6-MGP, SPAG5-MGP-UBE2c, TOP2a-EGFL6-MDK, SPAG5-UBE2c-MDK, TOP2a-HOXA13-THY1, TOP2a-BIRC5-SMC4L1, TOP2a-ENG-EGFL6, SPAG5-EGFL6-THY1, SPAG5-ENG-UBE2c, SPAG5-SEM2-BIRC5, TOP2a-MGP-BIRC5, CDC2-SEM2-THY1, TOP2a-BIRC5-MDK, TOP2a-EGFL6-UBE2c, TOP2a-ENG-BIRC5, TOP2a-HOXA13-SMC4L1, SPAG5-BIRC5-THY1, SPAG5-SEM2-HOXA13, SPAG5-EGFL6-SMC4L1, TOP2a-MGP-HOXA13, CDC2-SEM2-SMC4L1, SPAG5-EGFL6-MGP, TOP2a-HOXA13-MDK, SPAG5-EGFL6-MDK, CDC2-MGP-SEM2, TOP2a-BIRC5-UBE2c, TOP2a-ENG-HOXA13, SPAG5-HOXA13-THY1, CDC2-SEM2-MDK, SPAG5-BIRC5-SMC4L1, SPAG5-ENG-EGFL6, CDC2-THY1-SMC4L1, CDC2-ENG-SEM2, SPAG5-MGP-BIRC5, SPAG5-BIRC5-MDK, CDC2-MGP-THY1, TOP2a-UBE2c-HOXA13, CDC2-MDK-THY1, SPAG5-EGFL6-UBE2c, SPAG5-ENG-BIRC5, SPAG5-HOXA13-SMC4L1, CDC2-SEM2-UBE2c, CDC2-ENG-THY1, SPAG5-MGP-HOXA13, SPAG5-HOXA13-MDK, CDC2-MGP-SMC4L1, TOP2a-EGFL6-BIRC5, SPAG5-BIRC5-UBE2c, CDC2-MDK-SMC4L1, SPAG5-ENG-HOXA13, CDC2-UBE2c-THY1, CDC2-MGP-MDK, CDC2-ENG-SMC4L1, CDC2-ENG-MGP, TOP2a-EGFL6-HOXA13, SPAG5-UBE2c-HOXA13, CDC2-ENG-MDK, CDC2-UBE2C-SMC4L1, CDC2-EGFL6-SEM2, CDC2-MGP-UBE2c, TOP2a-BIRC5-HOXA13, CDC2-UBE2c-MDK, SPAG5-EGFL6-BIRC5, CDC2-EGFL6-THY1, CDC2-ENG-UBE2c, CDC2-SEM2-BIRC5, SPAG5-EGFL6-HOXA13, CDC2-BIRC5-THY1, CDC2-SEM2-HOXA13, CDC2-EGFL6-SMC4L1, CDC2-EGFL6-MGP, CDC2-EGFL6-MDK, SPAG5-BIRC5-HOXA13, CDC2-HOXA13-THY1, CDC2-BIRC5-SMC4L1, CDC2-ENG-EGFL6, CDC2-MGP-BIRC5, CDC2-BIRC5-MDK, CDC2-EGFL6-UBE2c, CDC2-ENG-BIRC5, CDC2-HOXA13-SMC4L1, CDC2-MGP-HOXA13, CDC2-HOXA13-MDK, CDC2-BIRC5-UBE2c, CDC2-ENG-HOXA13, CDC2-UBE2c-HOXA13, CDC2-EGFL6-BIRC5, CDC2-EGFL6-HOXA13, CDC2-BIRC5-HOXA13, NRP1-SEM2-CHGA, NRP1-CHGA-THY1, NRP1-CHGA-SMC4L1, NRP1-MGP-CHGA, NRP1-CHGA-MDK, ENG-NRP1-CHGA, NRP1-CHGA-UBE2c, NRP1-EGFL6-CHGA, NRP1-CHGA-BIRC5, NRP1-CHGA-HOXA13, NRP1-SEM2-THY1, NRP1-SEM2-SMC4L1, NRP1-MGP-SEM2, NRP1-SEM2-MDK, CDC2-TOP2a-CHGA, CDC2-TOP2a-NRP1, CDC2-TOP2a-SPAG5, CDC2-TOP2a-ENG, CDC2-TOP2a-SEM2, HOXA13-CDC2-TOP2a, CDC2-TOP2a-MDK, CDC2-TOP2a-IGFBP5, CDC2-TOP2a-SEMA3F, CDC2-TOP2a-EGFL6, TOP2a-SPAG5-CHGA, CDC2-SPAG5-CHGA, CDC2-TOP2a-SMC4L1, TOP2a-NRP1-SPAG5, TOP2a-NRP1-ENG, CDC2-NRP1-SPAG5, CDC2-NRP1-ENG, CDC2-TOP2a-NOV, UBE2c-CDC2-TOP2a, TOP2a-ENG-SPAG5, HOXA13-TOP2a-CHGA, CDC2-ENG-SPAG5, CDC2-TOP2a-THY1, HOXA13-CDC2-CHGA, BIRC5-CDC2-TOP2a, TOP2a-SPAG5-SEM2, TOP2a-MDK-CHGA, CDC2-SPAG5-SEM2, TOP2a-IGFBP5-CHGA, CDC2-MDK-CHGA, HOXA13-TOP2a-NRP1, CDC2-IGFBP5-CHGA, HOXA13-CDC2-NRP1, TOP2a-SEMA3F-CHGA, CDC2-SEMA3F-CHGA, TOP2a-NRP1-IGFBP5, HOXA13-TOP2a-SPAG5, CDC2-NRP1-IGFBP5, HOXA13-TOP2a-ENG, HOXA13-CDC2-SPAG5, TOP2a-NRP1-SEMA3F, HOXA13-CDC2-ENG, TOP2a-SPAG5-MDK, CDC2-NRP1-SEMA3F, HOXA13-TOP2a-SEM2, TOP2a-SPAG5-IGFBP5, TOP2a-SMC4L1-CHGA, CDC2-SPAG5-MDK, TOP2a-ENG-IGFBP5, HOXA13-CDC2-SEM2, CDC2-SPAG5-IGFBP5, CDC2-SMC4L1-CHGA, CDC2-ENG-IGFBP5, TOP2a-SPAG5-SEMA3F, NRP1-SPAG5-CHGA, TOP2a-ENG-SEMA3F, NRP1-ENG-CHGA, TOP2a-SEM2-IGFBP5, CDC2-SPAG5-SEMA3F, CDC2-ENG-SEMA3F, CDC2-SEM2-IGFBP5, TOP2a-NOV-CHGA, TOP2a-SPAG5-EGFL6, TOP2a-SEM2-SEMA3F, UBE2c-TOP2a-CHGA, CDC2-NOV-CHGA, CDC2-SPAG5-EGFL6, CDC2-SEM2-SEMA3F, UBE2c-CDC2-CHGA, ENG-SPAG5-CHGA, TOP2a-THY1-CHGA, BIRC5-THY1-CHGA, HOXA13-TOP2a-MDK, TOP2a-SPAG5-SMC4L1, CDC2-THY1-CHGA, TOP2a-NRP1-NOV, BIRC5-CDC2-CHGA, HOXA13-TOP2a-IGFBP5, HOXA13-CDC2-MDK, CDC2-SPAG5-SMC4L1, UBE2c-TOP2a-NRP1, CDC2-NRP1-NOV, ENG-SEM2-CHGA, HOXA13-CDC2-IGFBP5, UBE2c-CDC2-NRP1, NRP1-ENG-SPAG5, HOXA13-TOP2a-SEMA3F, HOXA13-NRP1-CHGA, TOP2a-MDK-IGFBP5, BIRC5-TOP2a-NRP1, HOXA13-CDC2-SEMA3F, CDC2-MDK-IGFBP5, TOP2a-NOV-SPAG5, BIRC5-CDC2-NRP1, TOP2a-ENG-NOV, NRP1-SPAG5-SEM2, UBE2c-TOP2a-SPAG5, TOP2a-MDK-SEMA3F, NRP1-MDK-CHGA, CDC2-NOV-SPAG5, NRP1-ENG-SEM2, HOXA13-TOP2a-EGFL6, UBE2c-TOP2a-ENG, and CDC2-ENG-NOV, in a biological sample from a patient suspected of having bladder cancer said accumulation of each of said markers greater than about 1.5 times the accumulation of each of said markers in a group of normal subjects not having malignant bladder cancer. 16. The method of claim 15, wherein the biological sample is from tissue or urine. 17. (canceled) 18. (canceled) 19. (canceled) 20. (canceled) 21. (canceled) 22. A device for detecting a BTM, comprising: a substrate having a combination of BTM or UBTM capture reagents thereon, the combination selected from the group consisting of TOP2a and CDC2 (TOP2a-CDC2), SPAG5-TOP2A, SPAG5-CDC2, NRP1-UBE2c, SPAG5-MDK, TOP2a-THY1, SPAG5-CHGA, TOP2a-CHGA, SPAG5-THY1, TOP2a-MDK, CDC2-CHGA, TOP2a-ENG, TOP2a-MGP, SPAG5-NRP1, SPAG5-BIRC5, TOP2a-NRP1, SPAG5-MGP, SPAG5-EGFL6, TOP2a-HOXA13, TOP2a-EGFL6, TOP2a-BIRC5, SPAG5-ENG, SPAG5-HOXA13, CDC2-THY1, CDC2-MDK, SPAG5-SEM2, TOP2a-SMC4L1, TOP2a-SEM2, CDC2-NRP1, CDC2-MGP, CDC2-ENG, TOP2a-UBE2c, SPAG5-UBE2c, CDC2-EGFL6, CDC2-BIRC5, SPAG5-SMC4L1, CDC2-HOXA13, CDC2-SEM2, NRP1-MDK, NRP1-THY1, CDC2-SMC4L1, CDC2-UBE2c, CDC2-TOP2a, TOP2a-SPAG5, CDC2-SPAG5, SPAG5 and TOP2a and CDC2 (SPAG5-TOP2a-CDC5), SPAG5-TOP2a-NRP1, TOP2a-CDC2-NRP1, SPAG5-TOP2a-CHGA, SPAG5-CDC2-NRP1, TOP2a-CDC2-CHGA, SPAG5-TOP2a-SEM2, SPAG5-CDC2-CHGA, SPAG5-TOP2a-THY1, SPAG5-TOP2a-SMC4L1, SPAG5-TOP2a-MGP, SPAG5-TOP2a-MDK, SPAG5-TOP2a-ENG, SPAG5-TOP2a-UBE2c, SPAG5-TOP2a-EGFL6, TOP2a-CDC2-SEM2, SPAG5-TOP2a-BIRC5, TOP2a-CDC2-THY1, SPAG5-TOP2a-HOXA13, TOP2a-CDC2-SMC4L1, SPAG5-CDC2-SEM2, TOP2a-CDC2-MGP, TOP2a-CDC2-MDK, TOP2a-CDC2-ENG, SPAG5-CDC2-THY1, TOP2a-CDC2-UBE2c, SPAG5-CDC2-SMC4L1, SPAG5-CDC2-MGP, SPAG5-CDC2-MDK, SPAG5-CDC2-ENG, TOP2a-CDC2-EGFL6, SPAG5-CDC2-UBE2c, TOP2a-CDC2-BIRC5, TOP2a-CDC2-HOXA13, SPAG5-CDC2-EGFL6, SPAG5-CDC2-BIRC5, SPAG5-CDC2-HOXA13, TOP2a-NRP1-CHGA, SPAG5-NRP1-CHGA, TOP2a-NRP1-SEM2, CDC2-NRP1-CHGA, TOP2a-NRP1-THY1, TOP2a-NRP1-SMC4L1, SPAG5-NRP1-SEM2, TOP2a-NRP1-MGP, TOP2a-NRP1-MDK, TOP2a-ENG-NRP1, SPAG5-NRP1-THY1, TOP2a-NRP1-UBE2c, SPAG5-NRP1-SMC4L1, SPAG5-NRP1-MGP, SPAG5-NRP1-MDK, SPAG5-ENG-NRP1, TOP2a-NRP1-EGFL6, SPAG5-NRP1-UBE2c, TOP2a-SEM2-CHGA, TOP2a-NRP1-BIRC5, TOP2a-CHGA-THY1, TOP2a-NRP1-HOXA13, SPAG5-NRP1-EGFL6, CDC2-NRP1-SEM2, TOP2a-CHGA-SMC4L1, SPAG5-SEM2-CHGA, SPAG5-NRP1-BIRC5, TOP2a-MGP-CHGA, CDC2-NRP1-THY1, TOP2a-CHGA-MDK, TOP2a-ENG-CHGA, SPAG5-CHGA-THY1, SPAG5-NRP1-HOXA13, CDC2-NRP1-SMC4L1, TOP2a-CHGA-UBE2c, CDC2-NRP1-MGP, SPAG5-CHGA-SMC4L1, CDC2-NRP1-MDK, SPAG5-MGP-CHGA, CDC2-ENG-NRP1, SPAG5-CHGA-MDK, SPAG5-ENG-CHGA, CDC2-NRP1-UBE2C, TOP2a-EGFL6-CHGA, SPAG5-CHGA-UBE2c, TOP2a-CHGA-BIRC5, TOP2a-CHGA-BIRC5, CDC2-NRP1-EGFL6, TOP2a-CHGA-HOXA13, SPAG5-EGFL6-CHGA, CDC2-SEM2-CHGA, CDC2-NRP1-BIRC5, SPAG5-CHGA-BIRC5, TOP2a-SEM2-THY1, CDC2-CHGA-THY1, CDC2-NRP1-HOXA13, SPAG5-CHGA-HOXA13, TOP2a-SEM2-SMC4L1, CDC2-CHGA-SMC4L1, TOP2a-MGP-SEM2, CDC2-MGP-CHGA, TOP2a-SEM2-MDK, CDC2-CHGA-MDK, TOP2a-THY1-SMC4L1, TOP2a-ENG-SEM2, SPAG5-SEM2-THY1, CDC2-ENG-CHGA, TOP2a-MGP-THY1, TOP2a-MDK-THY1, TOP2a-SEM2-UBE2c, TOP2a-ENG-THY1, CDC2-CHGA-UBE2c, SPAG5-SEM2-SMC4L1, TOP2a-MGP-SMC4L1, SPAG5-MGP-SEM2, TOP2a-MDK-SMC4L1, SPAG5-SEM2-MDK, TOP2a-UBE2c-THY1, TOP2a-MGP-MDK, TOP2a-ENG-SMC4L1, SPAG5-THY1-SMC4L1, SPAG5-ENG-SEM2, TOP2a-ENG-MGP, SPAG5-MGP-THY1, TOP2a-ENG-MDK, SPAG5-MDK-THY1, TOP2a-UBE2c-SMC4L1, TOP2a-EGFL6-SEM2, SPAG5-SEM2-UBE2c, SPAG5-ENG-THY1, CDC2-EGFL6-CHGA, TOP2a-MGP-UBE2c, SPAG5-MGP-SMC4L1, TOP2a-UBE2c-MDK, SPAG5-MDK-SMC4L1, TOP2a-EGFL6-THY1, TOP2a-ENG-UBE2c, TOP2a-SEM2-BIRC5, SPAG5-UBE2C-THY1, CDC2-CHGA-BIRC5, SPAG5-MGP-MDK, SPAG5-ENG-SMC4L1, SPAG5-ENG-MGP, SPAG5-ENG-MDK, TOP2a-BIRC5-THY1, TOP2a-SEM2-HOXA13, TOP2a-EGFL6-SMC4L1, SPAG5-UBE2c-SMC4L1, SPAG5-EGFL6-SEM2, CDC2-CHGA-HOXA13, TOP2a-EGFL6-MGP, SPAG5-MGP-UBE2c, TOP2a-EGFL6-MDK, SPAG5-UBE2c-MDK, TOP2a-HOXA13-THY1, TOP2a-BIRC5-SMC4L1, TOP2a-ENG-EGFL6, SPAG5-EGFL6-THY1, SPAG5-ENG-UBE2c, SPAG5-SEM2-BIRC5, TOP2a-MGP-BIRC5, CDC2-SEM2-THY1, TOP2a-BIRC5-MDK, TOP2a-EGFL6-UBE2c, TOP2a-ENG-BIRC5, TOP2a-HOXA13-SMC4L1, SPAG5-BIRC5-THY1, SPAG5-SEM2-HOXA13, SPAG5-EGFL6-SMC4L1, TOP2a-MGP-HOXA13, CDC2-SEM2-SMC4L1, SPAG5-EGFL6-MGP, TOP2a-HOXA13-MDK, SPAG5-EGFL6-MDK, CDC2-MGP-SEM2, TOP2a-BIRC5-UBE2c, TOP2a-ENG-HOXA13, SPAG5-HOXA13-THY1, CDC2-SEM2-MDK, SPAG5-BIRC5-SMC4L1, SPAG5-ENG-EGFL6, CDC2-THY1-SMC4L1, CDC2-ENG-SEM2, SPAG5-MGP-BIRC5, SPAG5-BIRC5-MDK, CDC2-MGP-THY1, TOP2a-UBE2c-HOXA13, CDC2-MDK-THY1, SPAG5-EGFL6-UBE2c, SPAG5-ENG-BIRC5, SPAG5-HOXA13-SMC4L1, CDC2-SEM2-UBE2c, CDC2-ENG-THY1, SPAG5-MGP-HOXA13, SPAG5-HOXA13-MDK CDC2-MGP-SMC4L1, TOP2a-EGFL6-BIRC5, SPAG5-BIRC5-UBE2c, CDC2-MDK-SMC4L1, SPAG5-ENG-HOXA13, CDC2-UBE2c-THY1, CDC2-MGP-MDK, CDC2-ENG-SMC4L1, CDC2-ENG-MGP, TOP2a-EGFL6-HOXA13, SPAG5-UBE2c-HOXA13, CDC2-ENG-MDK, CDC2-UBE2C-SMC4L1, CDC2-EGFL6-SEM2, CDC2-MGP-UBE2c, TOP2a-BIRC5-HOXA13, CDC2-UBE2c-MDK, SPAG5-EGFL6-BIRC5, CDC2-EGFL6-THY1, CDC2-ENG-UBE2c, CDC2-SEM2-BIRC5, SPAG5-EGFL6-HOXA13, CDC2-BIRC5-THY1, CDC2-SEM2-HOXA13, CDC2-EGFL6-SMC4L1, CDC2-EGFL6-MGP, CDC2-EGFL6-MDK, SPAG5-BIRC5-HOXA13, CDC2-HOXA13-THY1, CDC2-BIRC5-SMC4L1, CDC2-ENG-EGFL6, CDC2-MGP-BIRC5, CDC2-BIRC5-MDK, CDC2-EGFL6-UBE2c, CDC2-ENG-BIRC5, CDC2-HOXA13-SMC4L1, CDC2-MGP-HOXA13, CDC2-HOXA13-MDK, CDC2-BIRC5-UBE2c, CDC2-ENG-HOXA13, CDC2-UBE2c-HOXA13, CDC2-EGFL6-BIRC5, CDC2-EGFL6-HOXA13, CDC2-BIRC5-HOXA13, NRP1-SEM2-CHGA, NRP1-CHGA-THY1, NRP1-CHGA-SMC4L1, NRP1-MGP-CHGA, NRP1-CHGA-MDK, ENG-NRP1-CHGA, NRP1-CHGA-UBE2c, NRP1-EGFL6-CHGA, NRP1-CHGA-BIRC5, NRP1-CHGA-HOXA13, NRP1-SEM2-THY1, NRP1-SEM2-SMC4L1, NRP1-MGP-SEM2, NRP1-SEM2-MDK, CDC2-TOP2a-CHGA, CDC2-TOP2a-NRP1, CDC2-TOP2a-SPAG5, CDC2-TOP2a-ENG, CDC2-TOP2a-SEM2, HOXA13-CDC2-TOP2a, CDC2-TOP2a-MDK, CDC2-TOP2a-IGFBP5, CDC2-TOP2a-SEMA3F, CDC2-TOP2a-EGFL6, TOP2a-SPAG5-CHGA, CDC2-SPAG5-CHGA, CDC2-TOP2a-SMC4L1, TOP2a-NRP1-SPAG5, TOP2a-NRP1-ENG, CDC2-NRP1-SPAG5, CDC2-NRP1-ENG, CDC2-TOP2a-NOV, UBE2c-CDC2-TOP2a, TOP2a-ENG-SPAG5, HOXA13-TOP2a-CHGA, CDC2-ENG-SPAG5, CDC2-TOP2a-THY1, HOXA13-CDC2-CHGA, BIRC5-CDC2-TOP2a, TOP2a-SPAG5-SEM2, TOP2a-MDK-CHGA, CDC2-SPAG5-SEM2, TOP2a-IGFBP5-CHGA, CDC2-MDK-CHGA, HOXA13-TOP2a-NRP1, CDC2-IGFBP5-CHGA, HOXA13-CDC2-NRP1, TOP2a-SEMA3F-CHGA, CDC2-SEMA3F-CHGA, TOP2a-NRP1-IGFBP5, HOXA13-TOP2a-SPAG5, CDC2-NRP1-IGFBP5, HOXA13-TOP2a-ENG, HOXA13-CDC2-SPAG5, TOP2a-NRP1-SEMA3F, HOXA13-CDC2-ENG, TOP2a-SPAG5-MDK, CDC2-NRP1-SEMA3F, HOXA13-TOP2a-SEM2, TOP2a-SPAG5-IGFBP5, TOP2a-SMC4L1-CHGA, CDC2-SPAG5-MDK, TOP2a-ENG-IGFBP5, HOXA13-CDC2-SEM2, CDC2-SPAG5-IGFBP5, CDC2-SMC4L1-CHGA, CDC2-ENG-IGFBP5, TOP2a-SPAG5-SEMA3F, NRP1-SPAG5-CHGA, TOP2a-ENG-SEMA3F, NRP1-ENG-CHGA, TOP2a-SEM2-IGFBP5, CDC2-SPAG5-SEMA3F, CDC2-ENG-SEMA3F, CDC2-SEM2-IGFBP5, TOP2a-NOV-CHGA, TOP2a-SPAG5-EGFL6, TOP2a-SEM2-SEMA3F, UBE2c-TOP2a-CHGA, CDC2-NOV-CHGA, CDC2-SPAG5-EGFL6, CDC2-SEM2-SEMA3F, UBE2c-CDC2-CHGA, ENG-SPAG5-CHGA, TOP2a-THY1-CHGA, BIRC5-THY1-CHGA, HOXA13-TOP2a-MDK, TOP2a-SPAG5-SMC4L1, CDC2-THY1-CHGA, TOP2a-NRP1-NOV, BIRC5-CDC2-CHGA, HOXA13-TOP2a-IGFBP5, HOXA13-CDC2-MDK, CDC2-SPAG5-SMC4L1, UBE2c-TOP2a-NRP1, CDC2-NRP1-NOV, ENG-SEM2-CHGA, HOXA13-CDC2-IGFBP5, UBE2c-CDC2-NRP1, NRP1-ENG-SPAG5, HOXA13-TOP2a-SEMA3F, HOXA13-NRP1-CHGA, TOP2a-MDK-IGFBP5, BIRC5-TOP2a-NRP1, HOXA13-CDC2-SEMA3F, CDC2-MDK-IGFBP5, TOP2a-NOV-SPAG5, BIRC5-CDC2-NRP1, TOP2a-ENG-NOV, NRP1-SPAG5-SEM2, UBE2c-TOP2a-SPAG5, TOP2a-MDK-SEMA3F, NRP1-MDK-CHGA, CDC2-NOV-SPAG5, NRP1-ENG-SEM2, HOXA13-TOP2a-EGFL6, UBE2c-TOP2a-ENG, and CDC2-ENG-NOV; and a detector associated with said substrate, said detector capable of detecting said combination of BTM or UBTM associated with said capture reagents. 23. The device of claim 22, wherein at least one of said capture reagents comprises an oligonucleotide. 24. The device of claim 22, wherein at least one of said capture reagents comprises an antibody. 25. (canceled) 26. A kit for detecting cancer, comprising: a substrate; a combination of at least two BTM or UBTM capture reagents on said substrate, said combination selected from the group consisting of TOP2a and CDC2 (TOP2a-CDC2), SPAG5-TOP2A, SPAG5-CDC2, NRP1-UBE2C, SPAG5-MDK, TOP2a-THY1, SPAG5-CHGA, TOP2a-CHGA, SPAG5-THY1, TOP2a-MDK, CDC2-CHGA, TOP2a-ENG, TOP2a-MGP, SPAG5-NRP1, SPAG5-BIRC5, TOP2a-NRP1, SPAG5-MGP, SPAG5-EGFL6, TOP2a-HOXA13, TOP2a-EGFL6, TOP2a-BIRC5, SPAG5-ENG, SPAG5-HOXA13, CDC2-THY1, CDC2-MDK, SPAG5-SEM2, TOP2a-SMC4L1, TOP2a-SEM2, CDC2-NRP1, CDC2-MGP, CDC2-ENG, TOP2a-UBE2c, SPAG5-UBE2c, CDC2-EGFL6, CDC2-BIRC5, SPAG5-SMC4L1, CDC2-HOXA13, CDC2-SEM2, NRP1-MDK, NRP1-THY1, CDC2-SMC4L1, CDC2-UBE2c, CDC2-TOP2a, TOP2a-SPAG5, CDC2-SPAG5, SPAG5 and TOP2a and CDC2 (SPAG5-TOP2a-CDC5), SPAG5-TOP2a-NRP1, TOP2a-CDC2-NRP1, SPAG5-TOP2a-CHGA, SPAG5-CDC2-NRP1, TOP2a-CDC2-CHGA, SPAG5-TOP2a-SEM2, SPAG5-CDC2-CHGA, SPAG5-TOP2a-THY1, SPAG5-TOP2a-SMC4L1, SPAG5-TOP2a-MGP, SPAG5-TOP2a-MDK, SPAG5-TOP2a-ENG, SPAG5-TOP2a-UBE2c, SPAG5-TOP2a-EGFL6, TOP2a-CDC2-SEM2, SPAG5-TOP2a-BIRC5, TOP2a-CDC2-THY1, SPAG5-TOP2a-HOXA13, TOP2a-CDC2-SMC4L1, SPAG5-CDC2-SEM2, TOP2a-CDC2-MGP, TOP2a-CDC2-MDK, TOP2a-CDC2-ENG, SPAG5-CDC2-THY1, TOP2a-CDC2-UBE2c, SPAG5-CDC2-SMC4L1, SPAG5-CDC2-MGP, SPAG5-CDC2-MDK, SPAG5-CDC2-ENG, TOP2a-CDC2-EGFL6, SPAG5-CDC2-UBE2C, TOP2a-CDC2-BIRC5, TOP2a-CDC2-HOXA13, SPAG5-CDC2-EGFL6, SPAG5-CDC2-BIRC5, SPAG5-CDC2-HOXA13, TOP2a-NRP1-CHGA, SPAG5-NRP1-CHGA, TOP2a-NRP1-SEM2, CDC2-NRP1-CHGA, TOP2a-NRP1-THY1, TOP2a-NRP1-SMC4L1, SPAG5-NRP1-SEM2, TOP2a-NRP1-MGP, TOP2a-NRP1-MDK, TOP2a-ENG-NRP1, SPAG5-NRP1-THY1, TOP2a-NRP1-UBE2c, SPAG5-NRP1-SMC4L1, SPAG5-NRP1-MGP SPAG5-NRP1-MDK, SPAG5-ENG-NRP1, TOP2a-NRP1-EGFL6, SPAG5-NRP1-UBE2c, TOP2a-SEM2-CHGA, TOP2a-NRP1-BIRC5, TOP2a-CHGA-THY1, TOP2a-NRP1-HOXA13, SPAG5-NRP1-EGFL6, CDC2-NRP1-SEM2, TOP2a-CHGA-SMC4L1, SPAG5-SEM2-CHGA, SPAG5-NRP1-BIRC5, TOP2a-MGP-CHGA, CDC2-NRP1-THY1, TOP2a-CHGA-MDK, TOP2a-ENG-CHGA, SPAG5-CHGA-THY1, SPAG5-NRP1-HOXA13, CDC2-NRP1-SMC4L1, TOP2a-CHGA-UBE2c, CDC2-NRP1-MGP, SPAG5-CHGA-SMC4L1, CDC2-NRP1-MDK, SPAG5-MGP-CHGA, CDC2-ENG-NRP1, SPAG5-CHGA-MDK, SPAG5-ENG-CHGA, CDC2-NRP1-UBE2c, TOP2a-EGFL6-CHGA, SPAG5-CHGA-UBE2c, TOP2a-CHGA-BIRC5, TOP2a-CHGA-BIRC5, CDC2-NRP1-EGFL6, TOP2a-CHGA-HOXA13, SPAG5-EGFL6-CHGA, CDC2-SEM2-CHGA, CDC2-NRP1-BIRC5, SPAG5-CHGA-BIRC5, TOP2a-SEM2-THY1, CDC2-CHGA-THY1, CDC2-NRP1-HOXA13, SPAG5-CHGA-HOXA13, TOP2a-SEM2-SMC4L1, CDC2-CHGA-SMC4L1, TOP2a-MGP-SEM2, CDC2-MGP-CHGA, TOP2a-SEM2-MDK, CDC2-CHGA-MDK, TOP2a-THY1-SMC4L1, TOP2a-ENG-SEM2, SPAG5-SEM2-THY1, CDC2-ENG-CHGA, TOP2a-MGP-THY1, TOP2a-MDK-THY1, TOP2a-SEM2-UBE2c, TOP2a-ENG-THY1, CDC2-CHGA-UBE2c, SPAG5-SEM2-SMC4L1, TOP2a-MGP-SMC4L1, SPAG5-MGP-SEM2, TOP2a-MDK-SMC4L1, SPAG5-SEM2-MDK, TOP2a-UBE2c-THY1, TOP2a-MGP-MDK, TOP2a-ENG-SMC4L1, SPAG5-THY1-SMC4L1, SPAG5-ENG-SEM2, TOP2a-ENG-MGP, SPAG5-MGP-THY1, TOP2a-ENG-MDK, SPAG5-MDK-THY1, TOP2a-UBE2c-SMC4L1, TOP2a-EGFL6-SEM2, SPAG5-SEM2-UBE2c, SPAG5-ENG-THY1, CDC2-EGFL6-CHGA, TOP2a-MGP-UBE2c, SPAG5-MGP-SMC4L1, TOP2a-UBE2c-MDK, SPAG5-MDK-SMC4L1, TOP2a-EGFL6-THY1, TOP2a-ENG-UBE2c, TOP2a-SEM2-BIRC5, SPAG5-UBE2c-THY1, CDC2-CHGA-BIRC5, SPAG5-MGP-MDK, SPAG5-ENG-SMC4L1, SPAG5-ENG-MGP, SPAG5-ENG-MDK, TOP2a-BIRC5-THY1, TOP2a-SEM2-HOXA13, TOP2a-EGFL6-SMC4L1, SPAG5-UBE2c-SMC4L1, SPAG5-EGFL6-SEM2, CDC2-CHGA-HOXA13, TOP2a-EGFL6-MGP, SPAG5-MGP-UBE2c, TOP2a-EGFL6-MDK, SPAG5-UBE2c-MDK, TOP2a-HOXA13-THY1, TOP2a-BIRC5-SMC4L1, TOP2a-ENG-EGFL6, SPAG5-EGFL6-THY1, SPAG5-ENG-UBE2c, SPAG5-SEM2-BIRC5, TOP2a-MGP-BIRC5, CDC2-SEM2-THY1, TOP2a-BIRC5-MDK, TOP2a-EGFL6-UBE2c, TOP2a-ENG-BIRC5, TOP2a-HOXA13-SMC4L1, SPAG5-BIRC5-THY1, SPAG5-SEM2-HOXA13, SPAG5-EGFL6-SMC4L1, TOP2a-MGP-HOXA13, CDC2-SEM2-SMC4L1, SPAG5-EGFL6-MGP, TOP2a-HOXA13-MDK, SPAG5-EGFL6-MDK, CDC2-MGP-SEM2, TOP2a-BIRC5-UBE2c, TOP2a-ENG-HOXA13, SPAG5-HOXA13-THY1, CDC2-SEM2-MDK, SPAG5-BIRC5-SMC4L1, SPAG5-ENG-EGFL6, CDC2-THY1-SMC4L1, CDC2-ENG-SEM2, SPAG5-MGP-BIRC5, SPAG5-BIRC5-MDK, CDC2-MGP-THY1, TOP2a-UBE2c-HOXA13, CDC2-MDK-THY1, SPAG5-EGFL6-UBE2c, SPAG5-ENG-BIRC5, SPAG5-HOXA13-SMC4L1, CDC2-SEM2-UBE2c, CDC2-ENG-THY1, SPAG5-MGP-HOXA13, SPAG5-HOXA13-MDK, CDC2-MGP-SMC4L1, TOP2a-EGFL6-BIRC5, SPAG5-BIRC5-UBE2c, CDC2-MDK-SMC4L1, SPAG5-ENG-HOXA13, CDC2-UBE2c-THY1, CDC2-MGP-MDK, CDC2-ENG-SMC4L1, CDC2-ENG-MGP, TOP2a-EGFL6-HOXA13, SPAG5-UBE2c-HOXA13, CDC2-ENG-MDK, CDC2-UBE2c-SMC4L1, CDC2-EGFL6-SEM2, CDC2-MGP-UBE2c, TOP2a-BIRC5-HOXA13, CDC2-UBE2c-MDK, SPAG5-EGFL6-BIRC5, CDC2-EGFL6-THY1, CDC2-ENG-UBE2c, CDC2-SEM2-BIRC5, SPAG5-EGFL6-HOXA13, CDC2-BIRC5-THY1, CDC2-SEM2-HOXA13, CDC2-EGFL6-SMC4L1, CDC2-EGFL6-MGP, CDC2-EGFL6-MDK, SPAG5-BIRC5-HOXA13, CDC2-HOXA13-THY1, CDC2-BIRC5-SMC4L1, CDC2-ENG-EGFL6, CDC2-MGP-BIRC5, CDC2-BIRC5-MDK, CDC2-EGFL6-UBE2c, CDC2-ENG-BIRC5, CDC2-HOXA13-SMC4L1, CDC2-MGP-HOXA13, CDC2-HOXA13-MDK, CDC2-BIRC5-UBE2c, CDC2-ENG-HOXA13, CDC2-UBE2c-HOXA13, CDC2-EGFL6-BIRC5, CDC2-EGFL6-HOXA13, CDC2-BIRC5-HOXA13, NRP1-SEM2-CHGA, NRP1-CHGA-THY1, NRP1-CHGA-SMC4L1, NRP1-MGP-CHGA, NRP1-CHGA-MDK, ENG-NRP1-CHGA, NRP1-CHGA-UBE2c, NRP1-EGFL6-CHGA, NRP1-CHGA-BIRC5, NRP1-CHGA-HOXA13, NRP1-SEM2-THY1, NRP1-SEM2-SMC4L1, NRP1-MGP-SEM2, NRP1-SEM2-MDK, CDC2-TOP2a-CHGA, CDC2-TOP2a-NRP1, CDC2-TOP2a-SPAG5, CDC2-TOP2a-ENG, CDC2-TOP2a-SEM2, HOXA13-CDC2-TOP2a, CDC2-TOP2a-MDK, CDC2-TOP2a-IGFBP5, CDC2-TOP2a-SEMA3F, CDC2-TOP2a-EGFL6, TOP2a-SPAG5-CHGA, CDC2-SPAG5-CHGA, CDC2-TOP2a-SMC4L1, TOP2a-NRP1-SPAG5, TOP2a-NRP1-ENG, CDC2-NRP1-SPAG5, CDC2-NRP1-ENG, CDC2-TOP2a-NOV, UBE2c-CDC2-TOP2a, TOP2a-ENG-SPAG5, HOXA13-TOP2a-CHGA, CDC2-ENG-SPAG5, CDC2-TOP2a-THY1, HOXA13-CDC2-CHGA, BIRC5-CDC2-TOP2a, TOP2a-SPAG5-SEM2, TOP2a-MDK-CHGA, CDC2-SPAG5-SEM2, TOP2a-IGFBP5-CHGA, CDC2-MDK-CHGA, HOXA13-TOP2a-NRP1, CDC2-IGFBP5-CHGA, HOXA13-CDC2-NRP1, TOP2a-SEMA3F-CHGA, CDC2-SEMA3F-CHGA, TOP2a-NRP1-IGFBP5, HOXA13-TOP2a-SPAG5, CDC2-NRP1-IGFBP5, HOXA13-TOP2a-ENG, HOXA13-CDC2-SPAG5, TOP2a-NRP1-SEMA3F, HOXA13-CDC2-ENG, TOP2a-SPAG5-MDK, CDC2-NRP1-SEMA3F, HOXA13-TOP2a-SEM2, TOP2a-SPAG5-IGFBP5, TOP2a-SMC4L1-CHGA, CDC2-SPAG5-MDK, TOP2a-ENG-IGFBP5, HOXA13-CDC2-SEM2, CDC2-SPAG5-IGFBP5, CDC2-SMC4L1-CHGA, CDC2-ENG-IGFBP5, TOP2a-SPAG5-SEMA3F, NRP1-SPAG5-CHGA, TOP2a-ENG-SEMA3F, NRP1-ENG-CHGA, TOP2a-SEM2-IGFBP5, CDC2-SPAG5-SEMA3F, CDC2-ENG-SEMA3F, CDC2-SEM2-IGFBP5, TOP2a-NOV-CHGA, TOP2a-SPAG5-EGFL6, TOP2a-SEM2-SEMA3F, UBE2c-TOP2a-CHGA, CDC2-NOV-CHGA, CDC2-SPAG5-EGFL6, CDC2-SEM2-SEMA3F, UBE2c-CDC2-CHGA, ENG-SPAG5-CHGA, TOP2a-THY1-CHGA, BIRC5-THY1-CHGA, HOXA13-TOP2a-MDK, TOP2a-SPAG5-SMC4L1, CDC2-THY1-CHGA, TOP2a-NRP1-NOV, BIRC5-CDC2-CHGA, HOXA13-TOP2a-IGFBP5, HOXA13-CDC2-MDK, CDC2-SPAG5-SMC4L1, UBE2c-TOP2a-NRP1, CDC2-NRP1-NOV, ENG-SEM2-CHGA, HOXA13-CDC2-IGFBP5, UBE2c-CDC2-NRP1, NRP1-ENG-SPAG5, HOXA13-TOP2a-SEMA3F, HOXA13-NRP1-CHGA, TOP2a-MDK-IGFBP5, BIRC5-TOP2a-NRP1, HOXA13-CDC2-SEMA3F, CDC2-MDK-IGFBP5, TOP2a-NOV-SPAG5, BIRC5-CDC2-NRP1, TOP2a-ENG-NOV, NRP1-SPAG5-SEM2, UBE2c-TOP2a-SPAG5, TOP2a-MDK-SEMA3F, NRP1-MDK-CHGA, CDC2-NOV-SPAG5, NRP1-ENG-SEM2, HOXA13-TOP2a-EGFL6, UBE2c-TOP2a-ENG, and CDC2-ENG-NOV; and instructions for use. 27. (canceled) 28. (canceled) 29. (canceled) 30. A method for detecting the presence of bladder cancer in a subject, comprising: determining the amount in a urine sample of one or more first markers selected from the group consisting of BIRC2, HOXA13, IGFBP5, MGP, NOV, NRP1, SEMA3F, SPAG5, TOP2A, and wherein said first marker is not substantially present in blood of said subject. 31. The method of claim 30, further comprising an additional marker selected from the group consisting of IGFBP5, MGP, SEMA3F, CDC2, MDK, and HOXA13, wherein said additional marker is not the same as said first marker. 32. A method for distinguishing malignant bladder disease from non-malignant bladder disease, comprising: determining the accumulation in said patient's urine of one or more marker selected from the group consisting of HOXA13, IGFBP5, MDK, MGP, NRP1, CDC2, SEMA3F, SMC4L1, TOP2A and UBE2C; and determining the ratios of said one or more markers in said sample, a ratio of at least one of said markers being at least about 1.2 times that found in normal, non-malignant tissue. 33. (canceled) 34. (canceled) 35. (canceled) 36. A method for determining efficacy of therapy for bladder cancer, comprising: comparing the amount of one or more markers selected from the group consisting of GGH, SPP1, NRN1, SPARC, ADAMTS10, CNTN1, TLL2, PDIR, FBN1, KIAA0100 gene product, CALR, ITGBL1, ELA3B, SMOC2, HEXA, IGFBP7, MFAP2, CILP, OLFM1, LUM, SEM2, PRSS11, SULF1, SERPINH1, MGP, TIMP1, EGFL6, SPAG11, IGFBP5, SEMA3F, CDC2, TOP2A, UBE2C, STMN1, TUBA4, HIST1H1B, HMGB2, CCNA2, CDCA1, hypothetical protein MGC5576, DEK, MLF1IP, CDCA8, hypothetical protein FLJ20647, TYMS, SMC4L1, LYN, HMGB3, PTGIR, DONSON, HMMR, CLDN6, HIST1H1D, C10orf3, KNTC1, CKS1B, RRM2, HIST1H2BH, STK6, MPHOSPH1, CCNB2, GPR32, ENG, MFHAS1, HIST1H1C, AVPR2, CENPF, HOXA13, h4 histone family member g, MGC27121 gene, NP, ASPM, hypothetical protein FLJ11871, LBH, NUDT1, HELLS, ASB9, MCM5, IMP-2, DKFZP566M1046, TUBA2, GAS2L3, hypothetical protein FLJ12442, MCM6, DOK3, WDR18, CKAP2, KIF20A, putative fap protein, C6orf32, NEK2, CRY1, TGM2, DLG7, EIF2C2, DEPDC1, HIST2H4, MCM7, MTAP, KNTC2, HSPC150, SMC6L1, HIST1H2BC, ASF1B, ARH, LMNB1, hypothetical protein FLJ10719, hypothetical protein FLJ 10706, MAD2L1, SLC22A2, hypothetical protein MGC34923, SPAG5, ACVRL1, DSCR1, PRSS15, S100A9, MCM4, ST7L, PLEKHA4, EPHB1, CALD1, SMC1L1, Thy-1 co-transcribed, RAMP, FKBP11, C20orf129, HIST1H4H, CDKN3, MCAM, SNCAIP, NIPSNAP1, AP1M1, ANLN, C6orf69, TORC3, MAZ, TXNRD1, hypothetical protein xp 096695, C22orf4, VSNL1, similar to Carboxypeptidase N 83 kDa chain, KIAA1598, hypothetical protein FLJ13501, DKFZP4340047, hypothetical protein FLJ38716, similar to hypothetical protein (L1H3 region), hypothetical protein KIAA1875, PRIM1, hypothetical protein BC001096, MCM2, GJA3, C11orf30, similar to hypothetical protein FLJ30672, THY1, LRP3, LASS2, C18orf8, ZNF81, NARF, MTHFD2, D6T, SIAT7D, MMPL1, KLK11, KPNA2, FGFR1OP2, VIM, FLJ44108 protein, PAPOLG, FHOD1, RASL12, HMGN2, PITPNM2, DER1, EPHA4, VSIG1, RGS5, KIAA1639 protein, SH2B, PGLYRP4, CDC45L, MLSTD1, hypothetical protein MGC11266, TNFRSF13B, NET1, LHFPL5, MX2, SPHK1, ABCG4, SERPINB2, GALNT10, LEPR, MXD4, FAPP2, NUP210, CSK, NRP1, MGAT1, KIAA0100 gene product, LCN7, BMP7, ADAMTS10, PM5, NOMO3, CPA6, NPPC, hypothetical protein FLJ23221, ERP70, GALNT14, ITIH3, PAPPA2, LOXL1, TNFRSF6B, SPARC, MSMB, CLDN6, PTMA, AVPR2, similar to sodium- and chloride-dependent creatine transporter, TMEM19, hypothetical protein xp 047287, hypothetical protein FLJ11871, PROSC, MGC27121 gene, NQO1, CKAP4, hypothetical protein BC001096, PDPK1, regulator of mitotic spindle assembly 1, MIRAB13, PORCN, SOX6, GJB2, FLJ35784 protein, SLC37A3, SPRY4, LHX3, C7orf27, SLC39A1, ZNF307, MIF, BST2, PSTPIP1, SOX4, NCOA5, hypothetical protein FLJ31438, ODD, SLC23A2, SHFM1, SRPK2, RAMP2, BPGM, RGS5, CXADR, MEIS2, TENS1, SNAI2, CHST2, HCA127, Thy-1 co-transcribed (LOC94105), LRFN3, hypothetical protein FLJ22390, TRIB2, KRTHA3B, KIF21A, ANKRD17, RAG1, NUBP2, hypothetical protein FLJ20489, CASK, HIP1, PRKCDBP, TIE, C5orf15, CGI-72, ENTPD8, SH3BGRL3, NADH:ubiquinone oxidoreductase MLRQ subunit homolog, VG5Q, BG1, BCL2L11, ARK5, TLE3, ITIH5, RGS11, TM7SF3, SCRN3, PLXNA1, GJA4, hypothetical protein DKFZp434G1415, WSB2, CDA, GART, ZMPSTE24, TMEM33, GPI, hypothetical protein FLJ11000, CAMK1D, PTPN21, and TNS in a first sample from a patient with the amount of said one or more markers in a second sample from a patient after a period of treatment, the amount of said marker after said period of treatment is less than the amount of said marker before treatment. 37. A method for discriminating a stage of bladder cancer, comprising: measuring an amount of each of pairs of markers CDC2-HOXA13 or TOP2a-HOXA13 in the urine of a subject, said amounts of each of said pairs of markers are greater than the amounts of each of said pairs of markers in said subject's blood.
Early detection of tumors is a major determinant of survival of patients suffering from tumors, including bladder tumors. Members of the BTM or UBTM family can be highly and consistently accumulated in bladder tumor tissue and other tumor tissue, and/or can be accumulated in urine of patients, and thus are markers for bladder and other types of cancer. In certain embodiments, BTMs or UBTMs can accumulate in the urine, and detection of UBTM family members can be an effective diagnostic approach. In some embodiments, quantitative PCR methods have advantages over microarray methods. In other embodiments, detection and quantification of a plurality of BTMs or UBTMs can increase the sensitivity and specificity of detection of bladder cancer, and therefore provides methods for determining the stage and type of bladder cancer. Kits provide easy, convenient ways for carrying out the methods of this invention.1. A method for detecting bladder cancer in a subject, comprising: detecting the accumulation of a urinary bladder tumor marker (“UBTM”) family member in the urine, said accumulation in said subject greater than about 1.2 times the accumulation of said UBTM in urine from a group of normal subjects not having malignant bladder cancer. 2. The method of claim 1, wherein said UBTM family member is not associated with blood to a substantial extent. 3. The method according to claim 1, wherein the UBTM is selected from the group consisting of GGH, SPP1, NRN1, SPARC, ADAMTS10, CNTN1, TLL2, PDIR, FBN1, KIAA0100 gene product, CALR, ITGBL1, ELA3B, SMOC2, HEXA, IGFBP7, MFAP2, CILP, OLFM1, LUM, SEM2, PRSS11, SULF1, SERPINH1, MGP, TIMP1, EGFL6, SPAG11, IGFBP5, SEMA3F, CDC2, TOP2A, UBE2C, STMN1, TUBA4, HIST1H1B, HMGB2, CCNA2, CDCA1, hypothetical protein MGC5576, DEK, MLF1IP, CDCA8, hypothetical protein FLJ20647, TYMS, SMC4L1, LYN, HMGB3, PTGIR, DONSON, HMMR, CLDN6, HIST1H1D, C10orf3, KNTC1, CKS1B, RRM2, HIST1H2BH, STK6, MPHOSPH1, CCNB2, GPR32, ENG, MFHAS1, HIST1H1C, AVPR2, CENPF, HOXA13, h4 histone family member g, MGC27121 gene, NP, ASPM, hypothetical protein FLJ11871, LBH, NUDT1, HELLS, ASB9, MCM5, IMP-2, DKFZP566M1046, TUBA2, GAS2L3, hypothetical protein FLJ12442, MCM6, DOK3, WDR18, CKAP2, KIF20A, putative fap protein, C6orf32, NEK2, CRY1, TGM2, DLG7, EIF2C2, DEPDC1, HIST2H4, MCM7, MTAP, KNTC2, HSPC150, SMC6L1, HIST1H2BC, ASF1B, ARH, LMNB1, hypothetical protein FLJ10719, hypothetical protein FLJ 10706, MAD2L1, SLC22A2, hypothetical protein MGC34923, SPAG5, ACVRL1, DSCR1, PRSS15, S100A9, MCM4, ST7L, PLEKHA4, EPHB1, CALD1, SMC1L1, Thy-1 co-transcribed, RAMP, FKBP11, C20orf129, HIST1H4H, CDKN3, MCAM, SNCAIP, NIPSNAP1, AP1M1, ANLN, C6orf69, TORC3, MAZ, TXNRD1, hypothetical protein xp 096695, C22orf4, VSNL1, similar to Carboxypeptidase N 83 kDa chain, KIAA1598, hypothetical protein FLJ13501, DKFZP4340047, hypothetical protein FLJ38716, similar to hypothetical protein (L1H3 region), hypothetical protein KIAA1875, PRIM1, hypothetical protein BC001096, MCM2, GJA3, C11orf30, similar to hypothetical protein FLJ30672, THY1, LRP3, LASS2, C18orf8, ZNF81, NARF, MTHFD2, D6T, SIAT7D, MMPL1, KLK11, KPNA2, FGFR1OP2, VIM, FLJ44108 protein, PAPOLG, FHOD1, RASL12, HMGN2, PITPNM2, DER1, EPHA4, VSIG1, RGS5, KIAA1639 protein, SH2B, PGLYRP4, CDC45L, MLSTD1, hypothetical protein MGC11266, TNFRSF13B, NET1, LHFPL5, MX2, SPHK1, ABCG4, SERPINB2, GALNT10, LEPR, MXD4, FAPP2, NUP210, CSK, NRP1, MGAT1, KIAA0100 gene product, LCN7, BMP7, ADAMTS10, PM5, NOMO3, CPA6, NPPC, hypothetical protein FLJ23221, ERP70, GALNT14, ITIH3, PAPPA2, LOXL1, TNFRSF6B, SPARC, MSMB, CLDN6, PTMA, AVPR2, similar to sodium- and chloride-dependent creatine transporter, TMEM19, hypothetical protein xp 047287, hypothetical protein FLJ11871, PROSC, MGC27121 gene, NQO1, CKAP4, hypothetical protein BC001096, PDPK1, regulator of mitotic spindle assembly 1, MIRAB13, PORCN, SIX6, GJB2, FLJ35784 protein, SLC37A3, SPRY4, LHX3, C7orf27, SLC39A1, ZNF307, MIF, BST2, PSTPIP1, SOX4, NCOA5, hypothetical protein FLJ31438, ODD, SLC23A2, SHFM1, SRPK2, RAMP2, BPGM, RGS5, CXADR, MEIS2, TENS1, SNAI2, CHST2, HCA127, Thy-1 co-transcribed (LOC94105), LRFN3, hypothetical protein FLJ22390, TRIB2, KRTHA3B, KIF21A, ANKRD17, RAG1, NUBP2, hypothetical protein FLJ20489, CASK, HIP1, PRKCDBP, TIE, C5orf15, CGI-72, ENTPD8, SH3BGRL3, NADH:ubiquinone oxidoreductase MLRQ subunit homolog, VG5Q, BG1, BCL2L11, ARK5, TLE3, ITIH5, RGS11, TM7SF3, SCRN3, PLXNA1, GJA4, hypothetical protein DKFZp434G1415, WSB2, CDA, GART, ZMPSTE24, TMEM33, GPI, hypothetical protein FLJ11000, CAMK1D, PTPN21, and TNS. 4. The method of claim 1, wherein said step of detecting is carried out by detecting accumulation of a bladder tumor marker (“BTM”) or UBTM mRNA. 5. The method of claim 1, wherein said detecting is carried out using a microarray. 6. The method of claim 1, wherein said detecting is carried out using quantitative polymerase chain reaction or hybridization methods. 7. The method of claim 1, wherein said step of detecting is carried out by detecting accumulation of a UBTM protein. 8. The method of claim 7, wherein said step of detecting is carried out by detecting accumulation of a UBTM peptide. 9. The method of claim 7, wherein said step of detecting is carried out using a UBTM antibody. 10. (canceled) 11. (canceled) 12. The method of claim 1, wherein the method includes detection of accumulation of two or more UBTM family members in said sample. 13. (canceled) 14. The method of claim 1, further comprising detecting one or more pairs of markers selected from the group consisting of TOP2A-HOXA13, TOP2A-IGFBP5, TOP2A-SEMA3F and CDC2-HOXA13. 15. A method for detecting bladder cancer, comprising: detecting the accumulation of a combination of two or more BTM family members selected from the group consisting of TOP2a and CDC2 (TOP2a-CDC2), SPAG5-TOP2A, SPAG5-CDC2, NRP1-UBE2c, SPAG5-MDK, TOP2a-THY1, SPAG5-CHGA, TOP2a-CHGA, SPAG5-THY1, TOP2a-MDK, CDC2-CHGA, TOP2a-ENG, TOP2a-MGP, SPAG5-NRP1, SPAG5-BIRC5, TOP2a-NRP1, SPAG5-MGP, SPAG5-EGFL6, TOP2a-HOXA13, TOP2a-EGFL6, TOP2a-BIRC5, SPAG5-ENG, SPAG5-HOXA13, CDC2-THY1, CDC2-MDK, SPAG5-SEM2, TOP2a-SMC4L1, TOP2a-SEM2, CDC2-NRP1, CDC2-MGP, CDC2-ENG, TOP2a-UBE2c, SPAG5-UBE2c, CDC2-EGFL6, CDC2-BIRC5, SPAG5-SMC4L1, CDC2-HOXA13, CDC2-SEM2, NRP1-MDK, NRP1-THY1, CDC2-SMC4L1, CDC2-UBE2c, CDC2-TOP2a, TOP2a-SPAG5, CDC2-SPAG5, SPAG5 and TOP2a and CDC2 (SPAG5-TOP2a-CDC5), SPAG5-TOP2a-NRP1, TOP2a-CDC2-NRP1, SPAG5-TOP2a-CHGA, SPAG5-CDC2-NRP1, TOP2a-CDC2-CHGA, SPAG5-TOP2a-SEM2, SPAG5-CDC2-CHGA, SPAG5-TOP2a-THY1, SPAG5-TOP2a-SMC4L1, SPAG5-TOP2a-MGP, SPAG5-TOP2a-MDK, SPAG5-TOP2a-ENG, SPAG5-TOP2a-UBE2c, SPAG5-TOP2a-EGFL6, TOP2a-CDC2-SEM2, SPAG5-TOP2a-BIRC5, TOP2a-CDC2-THY1, SPAG5-TOP2a-HOXA13, TOP2a-CDC2-SMC4L1, SPAG5-CDC2-SEM2, TOP2a-CDC2-MGP, TOP2a-CDC2-MDK, TOP2a-CDC2-ENG, SPAG5-CDC2-THY1, TOP2a-CDC2-UBE2c, SPAG5-CDC2-SMC4L1, SPAG5-CDC2-MGP, SPAG5-CDC2-MDK, SPAG5-CDC2-ENG, TOP2a-CDC2-EGFL6, SPAG5-CDC2-UBE2C TOP2a-CDC2-BIRC5, TOP2a-CDC2-HOXA13, SPAG5-CDC2-EGFL6, SPAG5-CDC2-BIRC5, SPAG5-CDC2-HOXA13, TOP2a-NRP1-CHGA, SPAG5-NRP1-CHGA, TOP2a-NRP1-SEM2, CDC2-NRP1-CHGA, TOP2a-NRP1-THY1, TOP2a-NRP1-SMC4L1, SPAG5-NRP1-SEM2, TOP2a-NRP1-MGP, TOP2a-NRP1-MDK, TOP2a-ENG-NRP1, SPAG5-NRP1-THY1, TOP2a-NRP1-UBE2c, SPAG5-NRP1-SMC4L1, SPAG5-NRP1-MGP, SPAG5-NRP1-MDK, SPAG5-ENG-NRP1, TOP2a-NRP1-EGFL6, SPAG5-NRP1-UBE2c, TOP2a-SEM2-CHGA, TOP2a-NRP1-BIRC5, TOP2a-CHGA-THY1, TOP2a-NRP1-HOXA13, SPAG5-NRP1-EGFL6, CDC2-NRP1-SEM2, TOP2a-CHGA-SMC4L1, SPAG5-SEM2-CHGA, SPAG5-NRP1-BIRC5, TOP2a-MGP-CHGA, CDC2-NRP1-THY1, TOP2a-CHGA-MDK, TOP2a-ENG-CHGA, SPAG5-CHGA-THY1, SPAG5-NRP1-HOXA13, CDC2-NRP1-SMC4L1, TOP2a-CHGA-UBE2c, CDC2-NRP1-MGP, SPAG5-CHGA-SMC4L1, CDC2-NRP1-MDK, SPAG5-MGP-CHGA, CDC2-ENG-NRP1, SPAG5-CHGA-MDK, SPAG5-ENG-CHGA, CDC2-NRP1-UBE2c, TOP2a-EGFL6-CHGA, SPAG5-CHGA-UBE2c, TOP2a-CHGA-BIRC5, TOP2a-CHGA-BIRC5, CDC2-NRP1-EGFL6, TOP2a-CHGA-HOXA13, SPAG5-EGFL6-CHGA, CDC2-SEM2-CHGA, CDC2-NRP1-BIRC5, SPAG5-CHGA-BIRC5, TOP2a-SEM2-THY1, CDC2-CHGA-THY1, CDC2-NRP1-HOXA13, SPAG5-CHGA-HOXA13, TOP2a-SEM2-SMC4L1, CDC2-CHGA-SMC4L1, TOP2a-MGP-SEM2, CDC2-MGP-CHGA, TOP2a-SEM2-MDK, CDC2-CHGA-MDK, TOP2a-THY1-SMC4L1, TOP2a-ENG-SEM2, SPAG5-SEM2-THY1, CDC2-ENG-CHGA, TOP2a-MGP-THY1, TOP2a-MDK-THY1, TOP2a-SEM2-UBE2c, TOP2a-ENG-THY1, CDC2-CHGA-UBE2c, SPAG5-SEM2-SMC4L1, TOP2a-MGP-SMC4L1, SPAG5-MGP-SEM2, TOP2a-MDK-SMC4L1, SPAG5-SEM2-MDK, TOP2a-UBE2c-THY1, TOP2a-MGP-MDK, TOP2a-ENG-SMC4L1, SPAG5-THY1-SMC4L1, SPAG5-ENG-SEM2, TOP2a-ENG-MGP, SPAG5-MGP-THY1, TOP2a-ENG-MDK, SPAG5-MDK-THY1, TOP2a-UBE2c-SMC4L1, TOP2a-EGFL6-SEM2, SPAG5-SEM2-UBE2c, SPAG5-ENG-THY1, CDC2-EGFL6-CHGA, TOP2a-MGP-UBE2c, SPAG5-MGP-SMC4L1, TOP2a-UBE2c-MDK, SPAG5-MDK-SMC4L1, TOP2a-EGFL6-THY1, TOP2a-ENG-UBE2c, TOP2a-SEM2-BIRC5, SPAG5-UBE2c-THY1, CDC2-CHGA-BIRC5, SPAG5-MGP-MDK, SPAG5-ENG-SMC4L1, SPAG5-ENG-MGP, SPAG5-ENG-MDK, TOP2a-BIRC5-THY1, TOP2a-SEM2-HOXA13, TOP2a-EGFL6-SMC4L1, SPAG5-UBE2c-SMC4L1, SPAG5-EGFL6-SEM2, CDC2-CHGA-HOXA13, TOP2a-EGFL6-MGP, SPAG5-MGP-UBE2c, TOP2a-EGFL6-MDK, SPAG5-UBE2c-MDK, TOP2a-HOXA13-THY1, TOP2a-BIRC5-SMC4L1, TOP2a-ENG-EGFL6, SPAG5-EGFL6-THY1, SPAG5-ENG-UBE2c, SPAG5-SEM2-BIRC5, TOP2a-MGP-BIRC5, CDC2-SEM2-THY1, TOP2a-BIRC5-MDK, TOP2a-EGFL6-UBE2c, TOP2a-ENG-BIRC5, TOP2a-HOXA13-SMC4L1, SPAG5-BIRC5-THY1, SPAG5-SEM2-HOXA13, SPAG5-EGFL6-SMC4L1, TOP2a-MGP-HOXA13, CDC2-SEM2-SMC4L1, SPAG5-EGFL6-MGP, TOP2a-HOXA13-MDK, SPAG5-EGFL6-MDK, CDC2-MGP-SEM2, TOP2a-BIRC5-UBE2c, TOP2a-ENG-HOXA13, SPAG5-HOXA13-THY1, CDC2-SEM2-MDK, SPAG5-BIRC5-SMC4L1, SPAG5-ENG-EGFL6, CDC2-THY1-SMC4L1, CDC2-ENG-SEM2, SPAG5-MGP-BIRC5, SPAG5-BIRC5-MDK, CDC2-MGP-THY1, TOP2a-UBE2c-HOXA13, CDC2-MDK-THY1, SPAG5-EGFL6-UBE2c, SPAG5-ENG-BIRC5, SPAG5-HOXA13-SMC4L1, CDC2-SEM2-UBE2c, CDC2-ENG-THY1, SPAG5-MGP-HOXA13, SPAG5-HOXA13-MDK, CDC2-MGP-SMC4L1, TOP2a-EGFL6-BIRC5, SPAG5-BIRC5-UBE2c, CDC2-MDK-SMC4L1, SPAG5-ENG-HOXA13, CDC2-UBE2c-THY1, CDC2-MGP-MDK, CDC2-ENG-SMC4L1, CDC2-ENG-MGP, TOP2a-EGFL6-HOXA13, SPAG5-UBE2c-HOXA13, CDC2-ENG-MDK, CDC2-UBE2C-SMC4L1, CDC2-EGFL6-SEM2, CDC2-MGP-UBE2c, TOP2a-BIRC5-HOXA13, CDC2-UBE2c-MDK, SPAG5-EGFL6-BIRC5, CDC2-EGFL6-THY1, CDC2-ENG-UBE2c, CDC2-SEM2-BIRC5, SPAG5-EGFL6-HOXA13, CDC2-BIRC5-THY1, CDC2-SEM2-HOXA13, CDC2-EGFL6-SMC4L1, CDC2-EGFL6-MGP, CDC2-EGFL6-MDK, SPAG5-BIRC5-HOXA13, CDC2-HOXA13-THY1, CDC2-BIRC5-SMC4L1, CDC2-ENG-EGFL6, CDC2-MGP-BIRC5, CDC2-BIRC5-MDK, CDC2-EGFL6-UBE2c, CDC2-ENG-BIRC5, CDC2-HOXA13-SMC4L1, CDC2-MGP-HOXA13, CDC2-HOXA13-MDK, CDC2-BIRC5-UBE2c, CDC2-ENG-HOXA13, CDC2-UBE2c-HOXA13, CDC2-EGFL6-BIRC5, CDC2-EGFL6-HOXA13, CDC2-BIRC5-HOXA13, NRP1-SEM2-CHGA, NRP1-CHGA-THY1, NRP1-CHGA-SMC4L1, NRP1-MGP-CHGA, NRP1-CHGA-MDK, ENG-NRP1-CHGA, NRP1-CHGA-UBE2c, NRP1-EGFL6-CHGA, NRP1-CHGA-BIRC5, NRP1-CHGA-HOXA13, NRP1-SEM2-THY1, NRP1-SEM2-SMC4L1, NRP1-MGP-SEM2, NRP1-SEM2-MDK, CDC2-TOP2a-CHGA, CDC2-TOP2a-NRP1, CDC2-TOP2a-SPAG5, CDC2-TOP2a-ENG, CDC2-TOP2a-SEM2, HOXA13-CDC2-TOP2a, CDC2-TOP2a-MDK, CDC2-TOP2a-IGFBP5, CDC2-TOP2a-SEMA3F, CDC2-TOP2a-EGFL6, TOP2a-SPAG5-CHGA, CDC2-SPAG5-CHGA, CDC2-TOP2a-SMC4L1, TOP2a-NRP1-SPAG5, TOP2a-NRP1-ENG, CDC2-NRP1-SPAG5, CDC2-NRP1-ENG, CDC2-TOP2a-NOV, UBE2c-CDC2-TOP2a, TOP2a-ENG-SPAG5, HOXA13-TOP2a-CHGA, CDC2-ENG-SPAG5, CDC2-TOP2a-THY1, HOXA13-CDC2-CHGA, BIRC5-CDC2-TOP2a, TOP2a-SPAG5-SEM2, TOP2a-MDK-CHGA, CDC2-SPAG5-SEM2, TOP2a-IGFBP5-CHGA, CDC2-MDK-CHGA, HOXA13-TOP2a-NRP1, CDC2-IGFBP5-CHGA, HOXA13-CDC2-NRP1, TOP2a-SEMA3F-CHGA, CDC2-SEMA3F-CHGA, TOP2a-NRP1-IGFBP5, HOXA13-TOP2a-SPAG5, CDC2-NRP1-IGFBP5, HOXA13-TOP2a-ENG, HOXA13-CDC2-SPAG5, TOP2a-NRP1-SEMA3F, HOXA13-CDC2-ENG, TOP2a-SPAG5-MDK, CDC2-NRP1-SEMA3F, HOXA13-TOP2a-SEM2, TOP2a-SPAG5-IGFBP5, TOP2a-SMC4L1-CHGA, CDC2-SPAG5-MDK, TOP2a-ENG-IGFBP5, HOXA13-CDC2-SEM2, CDC2-SPAG5-IGFBP5, CDC2-SMC4L1-CHGA, CDC2-ENG-IGFBP5, TOP2a-SPAG5-SEMA3F, NRP1-SPAG5-CHGA, TOP2a-ENG-SEMA3F, NRP1-ENG-CHGA, TOP2a-SEM2-IGFBP5, CDC2-SPAG5-SEMA3F, CDC2-ENG-SEMA3F, CDC2-SEM2-IGFBP5, TOP2a-NOV-CHGA, TOP2a-SPAG5-EGFL6, TOP2a-SEM2-SEMA3F, UBE2c-TOP2a-CHGA, CDC2-NOV-CHGA, CDC2-SPAG5-EGFL6, CDC2-SEM2-SEMA3F, UBE2c-CDC2-CHGA, ENG-SPAG5-CHGA, TOP2a-THY1-CHGA, BIRC5-THY1-CHGA, HOXA13-TOP2a-MDK, TOP2a-SPAG5-SMC4L1, CDC2-THY1-CHGA, TOP2a-NRP1-NOV, BIRC5-CDC2-CHGA, HOXA13-TOP2a-IGFBP5, HOXA13-CDC2-MDK, CDC2-SPAG5-SMC4L1, UBE2c-TOP2a-NRP1, CDC2-NRP1-NOV, ENG-SEM2-CHGA, HOXA13-CDC2-IGFBP5, UBE2c-CDC2-NRP1, NRP1-ENG-SPAG5, HOXA13-TOP2a-SEMA3F, HOXA13-NRP1-CHGA, TOP2a-MDK-IGFBP5, BIRC5-TOP2a-NRP1, HOXA13-CDC2-SEMA3F, CDC2-MDK-IGFBP5, TOP2a-NOV-SPAG5, BIRC5-CDC2-NRP1, TOP2a-ENG-NOV, NRP1-SPAG5-SEM2, UBE2c-TOP2a-SPAG5, TOP2a-MDK-SEMA3F, NRP1-MDK-CHGA, CDC2-NOV-SPAG5, NRP1-ENG-SEM2, HOXA13-TOP2a-EGFL6, UBE2c-TOP2a-ENG, and CDC2-ENG-NOV, in a biological sample from a patient suspected of having bladder cancer said accumulation of each of said markers greater than about 1.5 times the accumulation of each of said markers in a group of normal subjects not having malignant bladder cancer. 16. The method of claim 15, wherein the biological sample is from tissue or urine. 17. (canceled) 18. (canceled) 19. (canceled) 20. (canceled) 21. (canceled) 22. A device for detecting a BTM, comprising: a substrate having a combination of BTM or UBTM capture reagents thereon, the combination selected from the group consisting of TOP2a and CDC2 (TOP2a-CDC2), SPAG5-TOP2A, SPAG5-CDC2, NRP1-UBE2c, SPAG5-MDK, TOP2a-THY1, SPAG5-CHGA, TOP2a-CHGA, SPAG5-THY1, TOP2a-MDK, CDC2-CHGA, TOP2a-ENG, TOP2a-MGP, SPAG5-NRP1, SPAG5-BIRC5, TOP2a-NRP1, SPAG5-MGP, SPAG5-EGFL6, TOP2a-HOXA13, TOP2a-EGFL6, TOP2a-BIRC5, SPAG5-ENG, SPAG5-HOXA13, CDC2-THY1, CDC2-MDK, SPAG5-SEM2, TOP2a-SMC4L1, TOP2a-SEM2, CDC2-NRP1, CDC2-MGP, CDC2-ENG, TOP2a-UBE2c, SPAG5-UBE2c, CDC2-EGFL6, CDC2-BIRC5, SPAG5-SMC4L1, CDC2-HOXA13, CDC2-SEM2, NRP1-MDK, NRP1-THY1, CDC2-SMC4L1, CDC2-UBE2c, CDC2-TOP2a, TOP2a-SPAG5, CDC2-SPAG5, SPAG5 and TOP2a and CDC2 (SPAG5-TOP2a-CDC5), SPAG5-TOP2a-NRP1, TOP2a-CDC2-NRP1, SPAG5-TOP2a-CHGA, SPAG5-CDC2-NRP1, TOP2a-CDC2-CHGA, SPAG5-TOP2a-SEM2, SPAG5-CDC2-CHGA, SPAG5-TOP2a-THY1, SPAG5-TOP2a-SMC4L1, SPAG5-TOP2a-MGP, SPAG5-TOP2a-MDK, SPAG5-TOP2a-ENG, SPAG5-TOP2a-UBE2c, SPAG5-TOP2a-EGFL6, TOP2a-CDC2-SEM2, SPAG5-TOP2a-BIRC5, TOP2a-CDC2-THY1, SPAG5-TOP2a-HOXA13, TOP2a-CDC2-SMC4L1, SPAG5-CDC2-SEM2, TOP2a-CDC2-MGP, TOP2a-CDC2-MDK, TOP2a-CDC2-ENG, SPAG5-CDC2-THY1, TOP2a-CDC2-UBE2c, SPAG5-CDC2-SMC4L1, SPAG5-CDC2-MGP, SPAG5-CDC2-MDK, SPAG5-CDC2-ENG, TOP2a-CDC2-EGFL6, SPAG5-CDC2-UBE2c, TOP2a-CDC2-BIRC5, TOP2a-CDC2-HOXA13, SPAG5-CDC2-EGFL6, SPAG5-CDC2-BIRC5, SPAG5-CDC2-HOXA13, TOP2a-NRP1-CHGA, SPAG5-NRP1-CHGA, TOP2a-NRP1-SEM2, CDC2-NRP1-CHGA, TOP2a-NRP1-THY1, TOP2a-NRP1-SMC4L1, SPAG5-NRP1-SEM2, TOP2a-NRP1-MGP, TOP2a-NRP1-MDK, TOP2a-ENG-NRP1, SPAG5-NRP1-THY1, TOP2a-NRP1-UBE2c, SPAG5-NRP1-SMC4L1, SPAG5-NRP1-MGP, SPAG5-NRP1-MDK, SPAG5-ENG-NRP1, TOP2a-NRP1-EGFL6, SPAG5-NRP1-UBE2c, TOP2a-SEM2-CHGA, TOP2a-NRP1-BIRC5, TOP2a-CHGA-THY1, TOP2a-NRP1-HOXA13, SPAG5-NRP1-EGFL6, CDC2-NRP1-SEM2, TOP2a-CHGA-SMC4L1, SPAG5-SEM2-CHGA, SPAG5-NRP1-BIRC5, TOP2a-MGP-CHGA, CDC2-NRP1-THY1, TOP2a-CHGA-MDK, TOP2a-ENG-CHGA, SPAG5-CHGA-THY1, SPAG5-NRP1-HOXA13, CDC2-NRP1-SMC4L1, TOP2a-CHGA-UBE2c, CDC2-NRP1-MGP, SPAG5-CHGA-SMC4L1, CDC2-NRP1-MDK, SPAG5-MGP-CHGA, CDC2-ENG-NRP1, SPAG5-CHGA-MDK, SPAG5-ENG-CHGA, CDC2-NRP1-UBE2C, TOP2a-EGFL6-CHGA, SPAG5-CHGA-UBE2c, TOP2a-CHGA-BIRC5, TOP2a-CHGA-BIRC5, CDC2-NRP1-EGFL6, TOP2a-CHGA-HOXA13, SPAG5-EGFL6-CHGA, CDC2-SEM2-CHGA, CDC2-NRP1-BIRC5, SPAG5-CHGA-BIRC5, TOP2a-SEM2-THY1, CDC2-CHGA-THY1, CDC2-NRP1-HOXA13, SPAG5-CHGA-HOXA13, TOP2a-SEM2-SMC4L1, CDC2-CHGA-SMC4L1, TOP2a-MGP-SEM2, CDC2-MGP-CHGA, TOP2a-SEM2-MDK, CDC2-CHGA-MDK, TOP2a-THY1-SMC4L1, TOP2a-ENG-SEM2, SPAG5-SEM2-THY1, CDC2-ENG-CHGA, TOP2a-MGP-THY1, TOP2a-MDK-THY1, TOP2a-SEM2-UBE2c, TOP2a-ENG-THY1, CDC2-CHGA-UBE2c, SPAG5-SEM2-SMC4L1, TOP2a-MGP-SMC4L1, SPAG5-MGP-SEM2, TOP2a-MDK-SMC4L1, SPAG5-SEM2-MDK, TOP2a-UBE2c-THY1, TOP2a-MGP-MDK, TOP2a-ENG-SMC4L1, SPAG5-THY1-SMC4L1, SPAG5-ENG-SEM2, TOP2a-ENG-MGP, SPAG5-MGP-THY1, TOP2a-ENG-MDK, SPAG5-MDK-THY1, TOP2a-UBE2c-SMC4L1, TOP2a-EGFL6-SEM2, SPAG5-SEM2-UBE2c, SPAG5-ENG-THY1, CDC2-EGFL6-CHGA, TOP2a-MGP-UBE2c, SPAG5-MGP-SMC4L1, TOP2a-UBE2c-MDK, SPAG5-MDK-SMC4L1, TOP2a-EGFL6-THY1, TOP2a-ENG-UBE2c, TOP2a-SEM2-BIRC5, SPAG5-UBE2C-THY1, CDC2-CHGA-BIRC5, SPAG5-MGP-MDK, SPAG5-ENG-SMC4L1, SPAG5-ENG-MGP, SPAG5-ENG-MDK, TOP2a-BIRC5-THY1, TOP2a-SEM2-HOXA13, TOP2a-EGFL6-SMC4L1, SPAG5-UBE2c-SMC4L1, SPAG5-EGFL6-SEM2, CDC2-CHGA-HOXA13, TOP2a-EGFL6-MGP, SPAG5-MGP-UBE2c, TOP2a-EGFL6-MDK, SPAG5-UBE2c-MDK, TOP2a-HOXA13-THY1, TOP2a-BIRC5-SMC4L1, TOP2a-ENG-EGFL6, SPAG5-EGFL6-THY1, SPAG5-ENG-UBE2c, SPAG5-SEM2-BIRC5, TOP2a-MGP-BIRC5, CDC2-SEM2-THY1, TOP2a-BIRC5-MDK, TOP2a-EGFL6-UBE2c, TOP2a-ENG-BIRC5, TOP2a-HOXA13-SMC4L1, SPAG5-BIRC5-THY1, SPAG5-SEM2-HOXA13, SPAG5-EGFL6-SMC4L1, TOP2a-MGP-HOXA13, CDC2-SEM2-SMC4L1, SPAG5-EGFL6-MGP, TOP2a-HOXA13-MDK, SPAG5-EGFL6-MDK, CDC2-MGP-SEM2, TOP2a-BIRC5-UBE2c, TOP2a-ENG-HOXA13, SPAG5-HOXA13-THY1, CDC2-SEM2-MDK, SPAG5-BIRC5-SMC4L1, SPAG5-ENG-EGFL6, CDC2-THY1-SMC4L1, CDC2-ENG-SEM2, SPAG5-MGP-BIRC5, SPAG5-BIRC5-MDK, CDC2-MGP-THY1, TOP2a-UBE2c-HOXA13, CDC2-MDK-THY1, SPAG5-EGFL6-UBE2c, SPAG5-ENG-BIRC5, SPAG5-HOXA13-SMC4L1, CDC2-SEM2-UBE2c, CDC2-ENG-THY1, SPAG5-MGP-HOXA13, SPAG5-HOXA13-MDK CDC2-MGP-SMC4L1, TOP2a-EGFL6-BIRC5, SPAG5-BIRC5-UBE2c, CDC2-MDK-SMC4L1, SPAG5-ENG-HOXA13, CDC2-UBE2c-THY1, CDC2-MGP-MDK, CDC2-ENG-SMC4L1, CDC2-ENG-MGP, TOP2a-EGFL6-HOXA13, SPAG5-UBE2c-HOXA13, CDC2-ENG-MDK, CDC2-UBE2C-SMC4L1, CDC2-EGFL6-SEM2, CDC2-MGP-UBE2c, TOP2a-BIRC5-HOXA13, CDC2-UBE2c-MDK, SPAG5-EGFL6-BIRC5, CDC2-EGFL6-THY1, CDC2-ENG-UBE2c, CDC2-SEM2-BIRC5, SPAG5-EGFL6-HOXA13, CDC2-BIRC5-THY1, CDC2-SEM2-HOXA13, CDC2-EGFL6-SMC4L1, CDC2-EGFL6-MGP, CDC2-EGFL6-MDK, SPAG5-BIRC5-HOXA13, CDC2-HOXA13-THY1, CDC2-BIRC5-SMC4L1, CDC2-ENG-EGFL6, CDC2-MGP-BIRC5, CDC2-BIRC5-MDK, CDC2-EGFL6-UBE2c, CDC2-ENG-BIRC5, CDC2-HOXA13-SMC4L1, CDC2-MGP-HOXA13, CDC2-HOXA13-MDK, CDC2-BIRC5-UBE2c, CDC2-ENG-HOXA13, CDC2-UBE2c-HOXA13, CDC2-EGFL6-BIRC5, CDC2-EGFL6-HOXA13, CDC2-BIRC5-HOXA13, NRP1-SEM2-CHGA, NRP1-CHGA-THY1, NRP1-CHGA-SMC4L1, NRP1-MGP-CHGA, NRP1-CHGA-MDK, ENG-NRP1-CHGA, NRP1-CHGA-UBE2c, NRP1-EGFL6-CHGA, NRP1-CHGA-BIRC5, NRP1-CHGA-HOXA13, NRP1-SEM2-THY1, NRP1-SEM2-SMC4L1, NRP1-MGP-SEM2, NRP1-SEM2-MDK, CDC2-TOP2a-CHGA, CDC2-TOP2a-NRP1, CDC2-TOP2a-SPAG5, CDC2-TOP2a-ENG, CDC2-TOP2a-SEM2, HOXA13-CDC2-TOP2a, CDC2-TOP2a-MDK, CDC2-TOP2a-IGFBP5, CDC2-TOP2a-SEMA3F, CDC2-TOP2a-EGFL6, TOP2a-SPAG5-CHGA, CDC2-SPAG5-CHGA, CDC2-TOP2a-SMC4L1, TOP2a-NRP1-SPAG5, TOP2a-NRP1-ENG, CDC2-NRP1-SPAG5, CDC2-NRP1-ENG, CDC2-TOP2a-NOV, UBE2c-CDC2-TOP2a, TOP2a-ENG-SPAG5, HOXA13-TOP2a-CHGA, CDC2-ENG-SPAG5, CDC2-TOP2a-THY1, HOXA13-CDC2-CHGA, BIRC5-CDC2-TOP2a, TOP2a-SPAG5-SEM2, TOP2a-MDK-CHGA, CDC2-SPAG5-SEM2, TOP2a-IGFBP5-CHGA, CDC2-MDK-CHGA, HOXA13-TOP2a-NRP1, CDC2-IGFBP5-CHGA, HOXA13-CDC2-NRP1, TOP2a-SEMA3F-CHGA, CDC2-SEMA3F-CHGA, TOP2a-NRP1-IGFBP5, HOXA13-TOP2a-SPAG5, CDC2-NRP1-IGFBP5, HOXA13-TOP2a-ENG, HOXA13-CDC2-SPAG5, TOP2a-NRP1-SEMA3F, HOXA13-CDC2-ENG, TOP2a-SPAG5-MDK, CDC2-NRP1-SEMA3F, HOXA13-TOP2a-SEM2, TOP2a-SPAG5-IGFBP5, TOP2a-SMC4L1-CHGA, CDC2-SPAG5-MDK, TOP2a-ENG-IGFBP5, HOXA13-CDC2-SEM2, CDC2-SPAG5-IGFBP5, CDC2-SMC4L1-CHGA, CDC2-ENG-IGFBP5, TOP2a-SPAG5-SEMA3F, NRP1-SPAG5-CHGA, TOP2a-ENG-SEMA3F, NRP1-ENG-CHGA, TOP2a-SEM2-IGFBP5, CDC2-SPAG5-SEMA3F, CDC2-ENG-SEMA3F, CDC2-SEM2-IGFBP5, TOP2a-NOV-CHGA, TOP2a-SPAG5-EGFL6, TOP2a-SEM2-SEMA3F, UBE2c-TOP2a-CHGA, CDC2-NOV-CHGA, CDC2-SPAG5-EGFL6, CDC2-SEM2-SEMA3F, UBE2c-CDC2-CHGA, ENG-SPAG5-CHGA, TOP2a-THY1-CHGA, BIRC5-THY1-CHGA, HOXA13-TOP2a-MDK, TOP2a-SPAG5-SMC4L1, CDC2-THY1-CHGA, TOP2a-NRP1-NOV, BIRC5-CDC2-CHGA, HOXA13-TOP2a-IGFBP5, HOXA13-CDC2-MDK, CDC2-SPAG5-SMC4L1, UBE2c-TOP2a-NRP1, CDC2-NRP1-NOV, ENG-SEM2-CHGA, HOXA13-CDC2-IGFBP5, UBE2c-CDC2-NRP1, NRP1-ENG-SPAG5, HOXA13-TOP2a-SEMA3F, HOXA13-NRP1-CHGA, TOP2a-MDK-IGFBP5, BIRC5-TOP2a-NRP1, HOXA13-CDC2-SEMA3F, CDC2-MDK-IGFBP5, TOP2a-NOV-SPAG5, BIRC5-CDC2-NRP1, TOP2a-ENG-NOV, NRP1-SPAG5-SEM2, UBE2c-TOP2a-SPAG5, TOP2a-MDK-SEMA3F, NRP1-MDK-CHGA, CDC2-NOV-SPAG5, NRP1-ENG-SEM2, HOXA13-TOP2a-EGFL6, UBE2c-TOP2a-ENG, and CDC2-ENG-NOV; and a detector associated with said substrate, said detector capable of detecting said combination of BTM or UBTM associated with said capture reagents. 23. The device of claim 22, wherein at least one of said capture reagents comprises an oligonucleotide. 24. The device of claim 22, wherein at least one of said capture reagents comprises an antibody. 25. (canceled) 26. A kit for detecting cancer, comprising: a substrate; a combination of at least two BTM or UBTM capture reagents on said substrate, said combination selected from the group consisting of TOP2a and CDC2 (TOP2a-CDC2), SPAG5-TOP2A, SPAG5-CDC2, NRP1-UBE2C, SPAG5-MDK, TOP2a-THY1, SPAG5-CHGA, TOP2a-CHGA, SPAG5-THY1, TOP2a-MDK, CDC2-CHGA, TOP2a-ENG, TOP2a-MGP, SPAG5-NRP1, SPAG5-BIRC5, TOP2a-NRP1, SPAG5-MGP, SPAG5-EGFL6, TOP2a-HOXA13, TOP2a-EGFL6, TOP2a-BIRC5, SPAG5-ENG, SPAG5-HOXA13, CDC2-THY1, CDC2-MDK, SPAG5-SEM2, TOP2a-SMC4L1, TOP2a-SEM2, CDC2-NRP1, CDC2-MGP, CDC2-ENG, TOP2a-UBE2c, SPAG5-UBE2c, CDC2-EGFL6, CDC2-BIRC5, SPAG5-SMC4L1, CDC2-HOXA13, CDC2-SEM2, NRP1-MDK, NRP1-THY1, CDC2-SMC4L1, CDC2-UBE2c, CDC2-TOP2a, TOP2a-SPAG5, CDC2-SPAG5, SPAG5 and TOP2a and CDC2 (SPAG5-TOP2a-CDC5), SPAG5-TOP2a-NRP1, TOP2a-CDC2-NRP1, SPAG5-TOP2a-CHGA, SPAG5-CDC2-NRP1, TOP2a-CDC2-CHGA, SPAG5-TOP2a-SEM2, SPAG5-CDC2-CHGA, SPAG5-TOP2a-THY1, SPAG5-TOP2a-SMC4L1, SPAG5-TOP2a-MGP, SPAG5-TOP2a-MDK, SPAG5-TOP2a-ENG, SPAG5-TOP2a-UBE2c, SPAG5-TOP2a-EGFL6, TOP2a-CDC2-SEM2, SPAG5-TOP2a-BIRC5, TOP2a-CDC2-THY1, SPAG5-TOP2a-HOXA13, TOP2a-CDC2-SMC4L1, SPAG5-CDC2-SEM2, TOP2a-CDC2-MGP, TOP2a-CDC2-MDK, TOP2a-CDC2-ENG, SPAG5-CDC2-THY1, TOP2a-CDC2-UBE2c, SPAG5-CDC2-SMC4L1, SPAG5-CDC2-MGP, SPAG5-CDC2-MDK, SPAG5-CDC2-ENG, TOP2a-CDC2-EGFL6, SPAG5-CDC2-UBE2C, TOP2a-CDC2-BIRC5, TOP2a-CDC2-HOXA13, SPAG5-CDC2-EGFL6, SPAG5-CDC2-BIRC5, SPAG5-CDC2-HOXA13, TOP2a-NRP1-CHGA, SPAG5-NRP1-CHGA, TOP2a-NRP1-SEM2, CDC2-NRP1-CHGA, TOP2a-NRP1-THY1, TOP2a-NRP1-SMC4L1, SPAG5-NRP1-SEM2, TOP2a-NRP1-MGP, TOP2a-NRP1-MDK, TOP2a-ENG-NRP1, SPAG5-NRP1-THY1, TOP2a-NRP1-UBE2c, SPAG5-NRP1-SMC4L1, SPAG5-NRP1-MGP SPAG5-NRP1-MDK, SPAG5-ENG-NRP1, TOP2a-NRP1-EGFL6, SPAG5-NRP1-UBE2c, TOP2a-SEM2-CHGA, TOP2a-NRP1-BIRC5, TOP2a-CHGA-THY1, TOP2a-NRP1-HOXA13, SPAG5-NRP1-EGFL6, CDC2-NRP1-SEM2, TOP2a-CHGA-SMC4L1, SPAG5-SEM2-CHGA, SPAG5-NRP1-BIRC5, TOP2a-MGP-CHGA, CDC2-NRP1-THY1, TOP2a-CHGA-MDK, TOP2a-ENG-CHGA, SPAG5-CHGA-THY1, SPAG5-NRP1-HOXA13, CDC2-NRP1-SMC4L1, TOP2a-CHGA-UBE2c, CDC2-NRP1-MGP, SPAG5-CHGA-SMC4L1, CDC2-NRP1-MDK, SPAG5-MGP-CHGA, CDC2-ENG-NRP1, SPAG5-CHGA-MDK, SPAG5-ENG-CHGA, CDC2-NRP1-UBE2c, TOP2a-EGFL6-CHGA, SPAG5-CHGA-UBE2c, TOP2a-CHGA-BIRC5, TOP2a-CHGA-BIRC5, CDC2-NRP1-EGFL6, TOP2a-CHGA-HOXA13, SPAG5-EGFL6-CHGA, CDC2-SEM2-CHGA, CDC2-NRP1-BIRC5, SPAG5-CHGA-BIRC5, TOP2a-SEM2-THY1, CDC2-CHGA-THY1, CDC2-NRP1-HOXA13, SPAG5-CHGA-HOXA13, TOP2a-SEM2-SMC4L1, CDC2-CHGA-SMC4L1, TOP2a-MGP-SEM2, CDC2-MGP-CHGA, TOP2a-SEM2-MDK, CDC2-CHGA-MDK, TOP2a-THY1-SMC4L1, TOP2a-ENG-SEM2, SPAG5-SEM2-THY1, CDC2-ENG-CHGA, TOP2a-MGP-THY1, TOP2a-MDK-THY1, TOP2a-SEM2-UBE2c, TOP2a-ENG-THY1, CDC2-CHGA-UBE2c, SPAG5-SEM2-SMC4L1, TOP2a-MGP-SMC4L1, SPAG5-MGP-SEM2, TOP2a-MDK-SMC4L1, SPAG5-SEM2-MDK, TOP2a-UBE2c-THY1, TOP2a-MGP-MDK, TOP2a-ENG-SMC4L1, SPAG5-THY1-SMC4L1, SPAG5-ENG-SEM2, TOP2a-ENG-MGP, SPAG5-MGP-THY1, TOP2a-ENG-MDK, SPAG5-MDK-THY1, TOP2a-UBE2c-SMC4L1, TOP2a-EGFL6-SEM2, SPAG5-SEM2-UBE2c, SPAG5-ENG-THY1, CDC2-EGFL6-CHGA, TOP2a-MGP-UBE2c, SPAG5-MGP-SMC4L1, TOP2a-UBE2c-MDK, SPAG5-MDK-SMC4L1, TOP2a-EGFL6-THY1, TOP2a-ENG-UBE2c, TOP2a-SEM2-BIRC5, SPAG5-UBE2c-THY1, CDC2-CHGA-BIRC5, SPAG5-MGP-MDK, SPAG5-ENG-SMC4L1, SPAG5-ENG-MGP, SPAG5-ENG-MDK, TOP2a-BIRC5-THY1, TOP2a-SEM2-HOXA13, TOP2a-EGFL6-SMC4L1, SPAG5-UBE2c-SMC4L1, SPAG5-EGFL6-SEM2, CDC2-CHGA-HOXA13, TOP2a-EGFL6-MGP, SPAG5-MGP-UBE2c, TOP2a-EGFL6-MDK, SPAG5-UBE2c-MDK, TOP2a-HOXA13-THY1, TOP2a-BIRC5-SMC4L1, TOP2a-ENG-EGFL6, SPAG5-EGFL6-THY1, SPAG5-ENG-UBE2c, SPAG5-SEM2-BIRC5, TOP2a-MGP-BIRC5, CDC2-SEM2-THY1, TOP2a-BIRC5-MDK, TOP2a-EGFL6-UBE2c, TOP2a-ENG-BIRC5, TOP2a-HOXA13-SMC4L1, SPAG5-BIRC5-THY1, SPAG5-SEM2-HOXA13, SPAG5-EGFL6-SMC4L1, TOP2a-MGP-HOXA13, CDC2-SEM2-SMC4L1, SPAG5-EGFL6-MGP, TOP2a-HOXA13-MDK, SPAG5-EGFL6-MDK, CDC2-MGP-SEM2, TOP2a-BIRC5-UBE2c, TOP2a-ENG-HOXA13, SPAG5-HOXA13-THY1, CDC2-SEM2-MDK, SPAG5-BIRC5-SMC4L1, SPAG5-ENG-EGFL6, CDC2-THY1-SMC4L1, CDC2-ENG-SEM2, SPAG5-MGP-BIRC5, SPAG5-BIRC5-MDK, CDC2-MGP-THY1, TOP2a-UBE2c-HOXA13, CDC2-MDK-THY1, SPAG5-EGFL6-UBE2c, SPAG5-ENG-BIRC5, SPAG5-HOXA13-SMC4L1, CDC2-SEM2-UBE2c, CDC2-ENG-THY1, SPAG5-MGP-HOXA13, SPAG5-HOXA13-MDK, CDC2-MGP-SMC4L1, TOP2a-EGFL6-BIRC5, SPAG5-BIRC5-UBE2c, CDC2-MDK-SMC4L1, SPAG5-ENG-HOXA13, CDC2-UBE2c-THY1, CDC2-MGP-MDK, CDC2-ENG-SMC4L1, CDC2-ENG-MGP, TOP2a-EGFL6-HOXA13, SPAG5-UBE2c-HOXA13, CDC2-ENG-MDK, CDC2-UBE2c-SMC4L1, CDC2-EGFL6-SEM2, CDC2-MGP-UBE2c, TOP2a-BIRC5-HOXA13, CDC2-UBE2c-MDK, SPAG5-EGFL6-BIRC5, CDC2-EGFL6-THY1, CDC2-ENG-UBE2c, CDC2-SEM2-BIRC5, SPAG5-EGFL6-HOXA13, CDC2-BIRC5-THY1, CDC2-SEM2-HOXA13, CDC2-EGFL6-SMC4L1, CDC2-EGFL6-MGP, CDC2-EGFL6-MDK, SPAG5-BIRC5-HOXA13, CDC2-HOXA13-THY1, CDC2-BIRC5-SMC4L1, CDC2-ENG-EGFL6, CDC2-MGP-BIRC5, CDC2-BIRC5-MDK, CDC2-EGFL6-UBE2c, CDC2-ENG-BIRC5, CDC2-HOXA13-SMC4L1, CDC2-MGP-HOXA13, CDC2-HOXA13-MDK, CDC2-BIRC5-UBE2c, CDC2-ENG-HOXA13, CDC2-UBE2c-HOXA13, CDC2-EGFL6-BIRC5, CDC2-EGFL6-HOXA13, CDC2-BIRC5-HOXA13, NRP1-SEM2-CHGA, NRP1-CHGA-THY1, NRP1-CHGA-SMC4L1, NRP1-MGP-CHGA, NRP1-CHGA-MDK, ENG-NRP1-CHGA, NRP1-CHGA-UBE2c, NRP1-EGFL6-CHGA, NRP1-CHGA-BIRC5, NRP1-CHGA-HOXA13, NRP1-SEM2-THY1, NRP1-SEM2-SMC4L1, NRP1-MGP-SEM2, NRP1-SEM2-MDK, CDC2-TOP2a-CHGA, CDC2-TOP2a-NRP1, CDC2-TOP2a-SPAG5, CDC2-TOP2a-ENG, CDC2-TOP2a-SEM2, HOXA13-CDC2-TOP2a, CDC2-TOP2a-MDK, CDC2-TOP2a-IGFBP5, CDC2-TOP2a-SEMA3F, CDC2-TOP2a-EGFL6, TOP2a-SPAG5-CHGA, CDC2-SPAG5-CHGA, CDC2-TOP2a-SMC4L1, TOP2a-NRP1-SPAG5, TOP2a-NRP1-ENG, CDC2-NRP1-SPAG5, CDC2-NRP1-ENG, CDC2-TOP2a-NOV, UBE2c-CDC2-TOP2a, TOP2a-ENG-SPAG5, HOXA13-TOP2a-CHGA, CDC2-ENG-SPAG5, CDC2-TOP2a-THY1, HOXA13-CDC2-CHGA, BIRC5-CDC2-TOP2a, TOP2a-SPAG5-SEM2, TOP2a-MDK-CHGA, CDC2-SPAG5-SEM2, TOP2a-IGFBP5-CHGA, CDC2-MDK-CHGA, HOXA13-TOP2a-NRP1, CDC2-IGFBP5-CHGA, HOXA13-CDC2-NRP1, TOP2a-SEMA3F-CHGA, CDC2-SEMA3F-CHGA, TOP2a-NRP1-IGFBP5, HOXA13-TOP2a-SPAG5, CDC2-NRP1-IGFBP5, HOXA13-TOP2a-ENG, HOXA13-CDC2-SPAG5, TOP2a-NRP1-SEMA3F, HOXA13-CDC2-ENG, TOP2a-SPAG5-MDK, CDC2-NRP1-SEMA3F, HOXA13-TOP2a-SEM2, TOP2a-SPAG5-IGFBP5, TOP2a-SMC4L1-CHGA, CDC2-SPAG5-MDK, TOP2a-ENG-IGFBP5, HOXA13-CDC2-SEM2, CDC2-SPAG5-IGFBP5, CDC2-SMC4L1-CHGA, CDC2-ENG-IGFBP5, TOP2a-SPAG5-SEMA3F, NRP1-SPAG5-CHGA, TOP2a-ENG-SEMA3F, NRP1-ENG-CHGA, TOP2a-SEM2-IGFBP5, CDC2-SPAG5-SEMA3F, CDC2-ENG-SEMA3F, CDC2-SEM2-IGFBP5, TOP2a-NOV-CHGA, TOP2a-SPAG5-EGFL6, TOP2a-SEM2-SEMA3F, UBE2c-TOP2a-CHGA, CDC2-NOV-CHGA, CDC2-SPAG5-EGFL6, CDC2-SEM2-SEMA3F, UBE2c-CDC2-CHGA, ENG-SPAG5-CHGA, TOP2a-THY1-CHGA, BIRC5-THY1-CHGA, HOXA13-TOP2a-MDK, TOP2a-SPAG5-SMC4L1, CDC2-THY1-CHGA, TOP2a-NRP1-NOV, BIRC5-CDC2-CHGA, HOXA13-TOP2a-IGFBP5, HOXA13-CDC2-MDK, CDC2-SPAG5-SMC4L1, UBE2c-TOP2a-NRP1, CDC2-NRP1-NOV, ENG-SEM2-CHGA, HOXA13-CDC2-IGFBP5, UBE2c-CDC2-NRP1, NRP1-ENG-SPAG5, HOXA13-TOP2a-SEMA3F, HOXA13-NRP1-CHGA, TOP2a-MDK-IGFBP5, BIRC5-TOP2a-NRP1, HOXA13-CDC2-SEMA3F, CDC2-MDK-IGFBP5, TOP2a-NOV-SPAG5, BIRC5-CDC2-NRP1, TOP2a-ENG-NOV, NRP1-SPAG5-SEM2, UBE2c-TOP2a-SPAG5, TOP2a-MDK-SEMA3F, NRP1-MDK-CHGA, CDC2-NOV-SPAG5, NRP1-ENG-SEM2, HOXA13-TOP2a-EGFL6, UBE2c-TOP2a-ENG, and CDC2-ENG-NOV; and instructions for use. 27. (canceled) 28. (canceled) 29. (canceled) 30. A method for detecting the presence of bladder cancer in a subject, comprising: determining the amount in a urine sample of one or more first markers selected from the group consisting of BIRC2, HOXA13, IGFBP5, MGP, NOV, NRP1, SEMA3F, SPAG5, TOP2A, and wherein said first marker is not substantially present in blood of said subject. 31. The method of claim 30, further comprising an additional marker selected from the group consisting of IGFBP5, MGP, SEMA3F, CDC2, MDK, and HOXA13, wherein said additional marker is not the same as said first marker. 32. A method for distinguishing malignant bladder disease from non-malignant bladder disease, comprising: determining the accumulation in said patient's urine of one or more marker selected from the group consisting of HOXA13, IGFBP5, MDK, MGP, NRP1, CDC2, SEMA3F, SMC4L1, TOP2A and UBE2C; and determining the ratios of said one or more markers in said sample, a ratio of at least one of said markers being at least about 1.2 times that found in normal, non-malignant tissue. 33. (canceled) 34. (canceled) 35. (canceled) 36. A method for determining efficacy of therapy for bladder cancer, comprising: comparing the amount of one or more markers selected from the group consisting of GGH, SPP1, NRN1, SPARC, ADAMTS10, CNTN1, TLL2, PDIR, FBN1, KIAA0100 gene product, CALR, ITGBL1, ELA3B, SMOC2, HEXA, IGFBP7, MFAP2, CILP, OLFM1, LUM, SEM2, PRSS11, SULF1, SERPINH1, MGP, TIMP1, EGFL6, SPAG11, IGFBP5, SEMA3F, CDC2, TOP2A, UBE2C, STMN1, TUBA4, HIST1H1B, HMGB2, CCNA2, CDCA1, hypothetical protein MGC5576, DEK, MLF1IP, CDCA8, hypothetical protein FLJ20647, TYMS, SMC4L1, LYN, HMGB3, PTGIR, DONSON, HMMR, CLDN6, HIST1H1D, C10orf3, KNTC1, CKS1B, RRM2, HIST1H2BH, STK6, MPHOSPH1, CCNB2, GPR32, ENG, MFHAS1, HIST1H1C, AVPR2, CENPF, HOXA13, h4 histone family member g, MGC27121 gene, NP, ASPM, hypothetical protein FLJ11871, LBH, NUDT1, HELLS, ASB9, MCM5, IMP-2, DKFZP566M1046, TUBA2, GAS2L3, hypothetical protein FLJ12442, MCM6, DOK3, WDR18, CKAP2, KIF20A, putative fap protein, C6orf32, NEK2, CRY1, TGM2, DLG7, EIF2C2, DEPDC1, HIST2H4, MCM7, MTAP, KNTC2, HSPC150, SMC6L1, HIST1H2BC, ASF1B, ARH, LMNB1, hypothetical protein FLJ10719, hypothetical protein FLJ 10706, MAD2L1, SLC22A2, hypothetical protein MGC34923, SPAG5, ACVRL1, DSCR1, PRSS15, S100A9, MCM4, ST7L, PLEKHA4, EPHB1, CALD1, SMC1L1, Thy-1 co-transcribed, RAMP, FKBP11, C20orf129, HIST1H4H, CDKN3, MCAM, SNCAIP, NIPSNAP1, AP1M1, ANLN, C6orf69, TORC3, MAZ, TXNRD1, hypothetical protein xp 096695, C22orf4, VSNL1, similar to Carboxypeptidase N 83 kDa chain, KIAA1598, hypothetical protein FLJ13501, DKFZP4340047, hypothetical protein FLJ38716, similar to hypothetical protein (L1H3 region), hypothetical protein KIAA1875, PRIM1, hypothetical protein BC001096, MCM2, GJA3, C11orf30, similar to hypothetical protein FLJ30672, THY1, LRP3, LASS2, C18orf8, ZNF81, NARF, MTHFD2, D6T, SIAT7D, MMPL1, KLK11, KPNA2, FGFR1OP2, VIM, FLJ44108 protein, PAPOLG, FHOD1, RASL12, HMGN2, PITPNM2, DER1, EPHA4, VSIG1, RGS5, KIAA1639 protein, SH2B, PGLYRP4, CDC45L, MLSTD1, hypothetical protein MGC11266, TNFRSF13B, NET1, LHFPL5, MX2, SPHK1, ABCG4, SERPINB2, GALNT10, LEPR, MXD4, FAPP2, NUP210, CSK, NRP1, MGAT1, KIAA0100 gene product, LCN7, BMP7, ADAMTS10, PM5, NOMO3, CPA6, NPPC, hypothetical protein FLJ23221, ERP70, GALNT14, ITIH3, PAPPA2, LOXL1, TNFRSF6B, SPARC, MSMB, CLDN6, PTMA, AVPR2, similar to sodium- and chloride-dependent creatine transporter, TMEM19, hypothetical protein xp 047287, hypothetical protein FLJ11871, PROSC, MGC27121 gene, NQO1, CKAP4, hypothetical protein BC001096, PDPK1, regulator of mitotic spindle assembly 1, MIRAB13, PORCN, SOX6, GJB2, FLJ35784 protein, SLC37A3, SPRY4, LHX3, C7orf27, SLC39A1, ZNF307, MIF, BST2, PSTPIP1, SOX4, NCOA5, hypothetical protein FLJ31438, ODD, SLC23A2, SHFM1, SRPK2, RAMP2, BPGM, RGS5, CXADR, MEIS2, TENS1, SNAI2, CHST2, HCA127, Thy-1 co-transcribed (LOC94105), LRFN3, hypothetical protein FLJ22390, TRIB2, KRTHA3B, KIF21A, ANKRD17, RAG1, NUBP2, hypothetical protein FLJ20489, CASK, HIP1, PRKCDBP, TIE, C5orf15, CGI-72, ENTPD8, SH3BGRL3, NADH:ubiquinone oxidoreductase MLRQ subunit homolog, VG5Q, BG1, BCL2L11, ARK5, TLE3, ITIH5, RGS11, TM7SF3, SCRN3, PLXNA1, GJA4, hypothetical protein DKFZp434G1415, WSB2, CDA, GART, ZMPSTE24, TMEM33, GPI, hypothetical protein FLJ11000, CAMK1D, PTPN21, and TNS in a first sample from a patient with the amount of said one or more markers in a second sample from a patient after a period of treatment, the amount of said marker after said period of treatment is less than the amount of said marker before treatment. 37. A method for discriminating a stage of bladder cancer, comprising: measuring an amount of each of pairs of markers CDC2-HOXA13 or TOP2a-HOXA13 in the urine of a subject, said amounts of each of said pairs of markers are greater than the amounts of each of said pairs of markers in said subject's blood.
1,600
583
15,476,849
1,635
Disclosed herein are compositions and pharmaceutical formulations that comprise a binding moiety conjugated to a polynucleic acid molecule and a polymer. Also described herein include methods for treating a cancer which utilize a composition or a pharmaceutical formulation comprising a binding moiety conjugated to a polynucleic acid molecule and a polymer.
1. A molecule of Formula (I): A-X-B-Y-C   Formula I wherein, A is an antibody or its binding fragments thereof; B is a polynucleotide; C is a polymer; X is a bond or first non-polymeric linker; and Y is a bond or second linker; wherein the polynucleotide comprises at least one 2′ modified nucleotide, at least one modified internucleotide linkage, or at least one inverted abasic moiety; and wherein A and C are not attached to B at the same terminus. 2. The molecule of claim 1, wherein the at least one 2′ modified nucleotide comprises 2′-O-methyl, 2′-O-methoxyethyl (2′-O-MOE), 2′-O-aminopropyl, 2′-deoxy, T-deoxy-2′-fluoro, 2′-O-aminopropyl (2′-O-AP), 2′-O-dimethylaminoethyl (2′-O-DMAOE), 2′-O-dimethylaminopropyl (2′-O-DMAP), T-O-dimethylaminoethyloxyethyl (2′-O-DMAEOE), or 2′-O-N-methylacetamido (2′-O-NMA) modified nucleotide. 3. The molecule of claim 1, wherein the at least one 2′ modified nucleotide comprises locked nucleic acid (LNA) or ethylene nucleic acid (ENA). 4. The molecule of claim 1, wherein the at least one modified internucleotide linkage comprises a phosphorothioate linkage or a phosphorodithioate linkage. 5. The molecule of claim 1, wherein the at least one inverted abasic moiety is at at least one terminus. 6. The molecule of claim 1, wherein the polynucleotide comprises a single strand. 7. The molecule of claim 1, wherein the polynucleotide comprises a first polynucleotide and a second polynucleotide hybridized to the first polynucleotide to form a double-stranded polynucleic acid molecule. 8. The molecule of claim 7, wherein the second polynucleotide comprises at least one modification. 9. The molecule of claim 7, wherein the first polynucleotide and the second polynucleotide are RNA molecules. 10. The molecule of claim 7, wherein the first polynucleotide comprises a sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 16-75, 452-1955, 1956-1962, 1967-2002, 2013-2032, 2082-2109, or 2117. 11. The molecule of claim 7, wherein the second polynucleotide comprises a sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 16-75, 452-1955, 1956-1962, 1967-2002, 2013-2032, 2082-2109, or 2117. 12. The molecule of claim 1, wherein X and Y are independently a bond. 13. The molecule of claim 1, wherein X and Y are independently a C1-C6 alkyl group. 14. The molecule of claim 1, wherein X is a homobifuctional linker or a heterobifunctional linker, optionally conjugated to a C1-C6 alkyl group. 15. The molecule of claim 1, wherein Y is a homobifuctional linker or a heterobifunctional linker. 16. The molecule of claim 1, wherein the antibody or binding fragment thereof comprises a humanized antibody or binding fragment thereof, chimeric antibody or binding fragment thereof, monoclonal antibody or binding fragment thereof, monovalent Fab′, divalent Fab2, single-chain variable fragment (scFv), diabody, minibody, nanobody, single-domain antibody (sdAb), or camelid antibody or binding fragment thereof. 17. The molecule of claim 1, wherein C is polyethylene glycol. 18. The molecule of claim 17, wherein C has a molecular weight of about 1000 Da, 2000 Da, or 5000 Da. 19. The molecule of claim 1, wherein A-X is conjugated to the 5′ end of B and Y-C is conjugated to the 3′ end of B, or Y-C is conjugated to the 5′ end of B and A-X is conjugated to the 3′ end of B. 20. The molecule of claim 1, further comprising D, wherein D is an endosomolytic moiety.
Disclosed herein are compositions and pharmaceutical formulations that comprise a binding moiety conjugated to a polynucleic acid molecule and a polymer. Also described herein include methods for treating a cancer which utilize a composition or a pharmaceutical formulation comprising a binding moiety conjugated to a polynucleic acid molecule and a polymer.1. A molecule of Formula (I): A-X-B-Y-C   Formula I wherein, A is an antibody or its binding fragments thereof; B is a polynucleotide; C is a polymer; X is a bond or first non-polymeric linker; and Y is a bond or second linker; wherein the polynucleotide comprises at least one 2′ modified nucleotide, at least one modified internucleotide linkage, or at least one inverted abasic moiety; and wherein A and C are not attached to B at the same terminus. 2. The molecule of claim 1, wherein the at least one 2′ modified nucleotide comprises 2′-O-methyl, 2′-O-methoxyethyl (2′-O-MOE), 2′-O-aminopropyl, 2′-deoxy, T-deoxy-2′-fluoro, 2′-O-aminopropyl (2′-O-AP), 2′-O-dimethylaminoethyl (2′-O-DMAOE), 2′-O-dimethylaminopropyl (2′-O-DMAP), T-O-dimethylaminoethyloxyethyl (2′-O-DMAEOE), or 2′-O-N-methylacetamido (2′-O-NMA) modified nucleotide. 3. The molecule of claim 1, wherein the at least one 2′ modified nucleotide comprises locked nucleic acid (LNA) or ethylene nucleic acid (ENA). 4. The molecule of claim 1, wherein the at least one modified internucleotide linkage comprises a phosphorothioate linkage or a phosphorodithioate linkage. 5. The molecule of claim 1, wherein the at least one inverted abasic moiety is at at least one terminus. 6. The molecule of claim 1, wherein the polynucleotide comprises a single strand. 7. The molecule of claim 1, wherein the polynucleotide comprises a first polynucleotide and a second polynucleotide hybridized to the first polynucleotide to form a double-stranded polynucleic acid molecule. 8. The molecule of claim 7, wherein the second polynucleotide comprises at least one modification. 9. The molecule of claim 7, wherein the first polynucleotide and the second polynucleotide are RNA molecules. 10. The molecule of claim 7, wherein the first polynucleotide comprises a sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 16-75, 452-1955, 1956-1962, 1967-2002, 2013-2032, 2082-2109, or 2117. 11. The molecule of claim 7, wherein the second polynucleotide comprises a sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs: 16-75, 452-1955, 1956-1962, 1967-2002, 2013-2032, 2082-2109, or 2117. 12. The molecule of claim 1, wherein X and Y are independently a bond. 13. The molecule of claim 1, wherein X and Y are independently a C1-C6 alkyl group. 14. The molecule of claim 1, wherein X is a homobifuctional linker or a heterobifunctional linker, optionally conjugated to a C1-C6 alkyl group. 15. The molecule of claim 1, wherein Y is a homobifuctional linker or a heterobifunctional linker. 16. The molecule of claim 1, wherein the antibody or binding fragment thereof comprises a humanized antibody or binding fragment thereof, chimeric antibody or binding fragment thereof, monoclonal antibody or binding fragment thereof, monovalent Fab′, divalent Fab2, single-chain variable fragment (scFv), diabody, minibody, nanobody, single-domain antibody (sdAb), or camelid antibody or binding fragment thereof. 17. The molecule of claim 1, wherein C is polyethylene glycol. 18. The molecule of claim 17, wherein C has a molecular weight of about 1000 Da, 2000 Da, or 5000 Da. 19. The molecule of claim 1, wherein A-X is conjugated to the 5′ end of B and Y-C is conjugated to the 3′ end of B, or Y-C is conjugated to the 5′ end of B and A-X is conjugated to the 3′ end of B. 20. The molecule of claim 1, further comprising D, wherein D is an endosomolytic moiety.
1,600
584
14,590,538
1,618
Embodiments of the present disclosure provide for compounds and methods of making compounds such as those shown in FIGS. 1.1A and 1.1B having formula 2, 3, 4, 5, 11, and 12 and formula 2′, 4′, and 11′,as well as uses for the compounds for imaging, and the like.
1. A method of imaging a T Cell comprising: administering to the subject a compound having formula 3; and imaging the subject, wherein detecting the presence of the compound corresponds to the presence of the T cell, wheiren formula 3 is represented by the following structure, wherein Ist is an isotope, wherein R′ is a compound having a formula selected from the group consisting of R′1, R′2, R′3, R′4, and R′5: 2. The method of claim 1, wherein the compound having formula 3 is a compound having formula 12, 3. The method of claim 1, wherein the isotope is selected from the group consisting of: 18F, 131I, 125I, 124I, 123I, 121I, 77Br, 77Br, 75Br, and 75Br. 4. A method of imaging T lymphoblasts comprising: administering to the subject a compound including formula 3; and imaging the subject, wherein detecting the presence of the compound corresponds to the presence of T lymphoblasts, wheiren formula 3 is represented by the following structure, wherein Ist is an isotope, wherein R′ is a compound having a formula selected from the group consisting of R′1, R′2, R′3, R′4, and R′5: 5. The method of claim 4, wherein the compound having formula 3 is a compound having formula 12, 6. The method of claim 4, wherein the isotope is selected from the group consisting of 18F, 131I, 125I, 124I, 123I, 121I, 77Br, 77Br, 75Br, and 75Br.
Embodiments of the present disclosure provide for compounds and methods of making compounds such as those shown in FIGS. 1.1A and 1.1B having formula 2, 3, 4, 5, 11, and 12 and formula 2′, 4′, and 11′,as well as uses for the compounds for imaging, and the like.1. A method of imaging a T Cell comprising: administering to the subject a compound having formula 3; and imaging the subject, wherein detecting the presence of the compound corresponds to the presence of the T cell, wheiren formula 3 is represented by the following structure, wherein Ist is an isotope, wherein R′ is a compound having a formula selected from the group consisting of R′1, R′2, R′3, R′4, and R′5: 2. The method of claim 1, wherein the compound having formula 3 is a compound having formula 12, 3. The method of claim 1, wherein the isotope is selected from the group consisting of: 18F, 131I, 125I, 124I, 123I, 121I, 77Br, 77Br, 75Br, and 75Br. 4. A method of imaging T lymphoblasts comprising: administering to the subject a compound including formula 3; and imaging the subject, wherein detecting the presence of the compound corresponds to the presence of T lymphoblasts, wheiren formula 3 is represented by the following structure, wherein Ist is an isotope, wherein R′ is a compound having a formula selected from the group consisting of R′1, R′2, R′3, R′4, and R′5: 5. The method of claim 4, wherein the compound having formula 3 is a compound having formula 12, 6. The method of claim 4, wherein the isotope is selected from the group consisting of 18F, 131I, 125I, 124I, 123I, 121I, 77Br, 77Br, 75Br, and 75Br.
1,600
585
15,976,346
1,617
A method for the treatment of facial Rosacea and skin erythema using pyrithione zinc aqueous solution of given concentration strength as the sole active ingredient in a topically applied fluid composition in which the pyrithione zinc aqueous solution comprises about 20% to about 30% of the fluid composition by volume, with an inactive moisturized carrier comprising the remainder of the fluid composition by volume.
1. A method of treating erythema resulting from facial Rosacea in a subject comprising the step of topically administering to the face of said subject a fluid composition consisting of an inactive moisturizer carrier and a single active ingredient, wherein the single active ingredient consists of a pyrithione zinc aqueous solution of about 20% to about 30% of the fluid composition by volume and the inactive moisturized carrier comprises the remainder of the fluid composition by volume, and wherein said pyrithione zinc aqueous solution included in the fluid composition is of a 1% to 4% concentration strength, and wherein said pyrithione zinc aqueous solution is the sole active ingredient of the fluid composition. 2. The method of claim 1 wherein a pharmaceutical form for administering the fluid composition includes an inactive moisturizer carrier selected from solutions, gels, lotions, creams, ointments, foams, emulsions, micro-emulsions, aerosols, sprays, and dispersions. 3. The method of claim 2 wherein a pharmaceutical form for administering said fluid composition includes about 20% to about 30% by volume of a pyrithione zinc aqueous solution of 1% to 4% concentration strength in a cocoa butter emulsion moisturizer cream carrier. 4. The method of claim 3 wherein said pharmaceutical form for administering said fluid composition consists of a mixture of one part pyrithione zinc aqueous solution by volume with 3 parts of a cocoa butter emulsion moisturizer cream carrier by volume. 5. A method of treating erythema resulting from facial Rosacea in a subject comprising the steps of: (a) topically administering to the facial area of said subject a fluid composition consisting of about 20% to about 30% of a pyrithione zinc aqueous solution of the fluid fluid composition by volume, wherein said 20% to about 30% of pyrithione zinc aqueous solution by volume is of a 1% to about 4% concentration strength, and is the sole active ingredient of the fluid composition; and (b) rinsing said fluid composition once administered from the face area of administration about 2-3 minutes thereafter. 6. The method of claim 5 wherein a pharmaceutical form for administering said fluid composition includes an inactive moisturizer carrier selected from solutions, gels, lotions, creams, ointments, foams, emulsions, micro-emulsions, aerosols sprays, and dispersions. 7. The method of claim 6 wherein a pharmaceutical form for administering said fluid composition in said treatment includes about 20% to about 30% by volume of a pyrithione zinc aqueous solution of 1% to 4% concentration strength in a cocoa butter emulsion moisturizer cream carrier. 8. The method of claim 7 wherein said pharmaceutical form for administering said fluid composition consists of a mixture of one part pyrithione zinc aqueous solution by volume
A method for the treatment of facial Rosacea and skin erythema using pyrithione zinc aqueous solution of given concentration strength as the sole active ingredient in a topically applied fluid composition in which the pyrithione zinc aqueous solution comprises about 20% to about 30% of the fluid composition by volume, with an inactive moisturized carrier comprising the remainder of the fluid composition by volume.1. A method of treating erythema resulting from facial Rosacea in a subject comprising the step of topically administering to the face of said subject a fluid composition consisting of an inactive moisturizer carrier and a single active ingredient, wherein the single active ingredient consists of a pyrithione zinc aqueous solution of about 20% to about 30% of the fluid composition by volume and the inactive moisturized carrier comprises the remainder of the fluid composition by volume, and wherein said pyrithione zinc aqueous solution included in the fluid composition is of a 1% to 4% concentration strength, and wherein said pyrithione zinc aqueous solution is the sole active ingredient of the fluid composition. 2. The method of claim 1 wherein a pharmaceutical form for administering the fluid composition includes an inactive moisturizer carrier selected from solutions, gels, lotions, creams, ointments, foams, emulsions, micro-emulsions, aerosols, sprays, and dispersions. 3. The method of claim 2 wherein a pharmaceutical form for administering said fluid composition includes about 20% to about 30% by volume of a pyrithione zinc aqueous solution of 1% to 4% concentration strength in a cocoa butter emulsion moisturizer cream carrier. 4. The method of claim 3 wherein said pharmaceutical form for administering said fluid composition consists of a mixture of one part pyrithione zinc aqueous solution by volume with 3 parts of a cocoa butter emulsion moisturizer cream carrier by volume. 5. A method of treating erythema resulting from facial Rosacea in a subject comprising the steps of: (a) topically administering to the facial area of said subject a fluid composition consisting of about 20% to about 30% of a pyrithione zinc aqueous solution of the fluid fluid composition by volume, wherein said 20% to about 30% of pyrithione zinc aqueous solution by volume is of a 1% to about 4% concentration strength, and is the sole active ingredient of the fluid composition; and (b) rinsing said fluid composition once administered from the face area of administration about 2-3 minutes thereafter. 6. The method of claim 5 wherein a pharmaceutical form for administering said fluid composition includes an inactive moisturizer carrier selected from solutions, gels, lotions, creams, ointments, foams, emulsions, micro-emulsions, aerosols sprays, and dispersions. 7. The method of claim 6 wherein a pharmaceutical form for administering said fluid composition in said treatment includes about 20% to about 30% by volume of a pyrithione zinc aqueous solution of 1% to 4% concentration strength in a cocoa butter emulsion moisturizer cream carrier. 8. The method of claim 7 wherein said pharmaceutical form for administering said fluid composition consists of a mixture of one part pyrithione zinc aqueous solution by volume
1,600
586
15,107,133
1,651
The invention provides a method for identifying methicillin resistant Staphylococcus aureus (MRSA) in a bacterial sample comprising the steps: classifying bacteria in the sample as Staphylococcus aureus (SA) and determining the presence or absence of the phenol soluble modulin peptide or a variant thereof wherein the presence of the PSM-mec peptide or variant thereof indicates methicillin resistant Staphylococcus aureus. The variant is preferably the formylated version of the PSM-mec peptide having a mass to charge ratio of 2415 in a singly protonated state.
1. A method for identifying methicillin resistant Staphylococcus aureus (MRSA) in a bacterial sample comprising the steps: classifying bacteria in the sample as Staphylococcus aureus (SA), directly depositing whole bacteria cells on a MALDI time of flight (MALDI-TOF) mass spectrometer target plate without prior extraction, acquiring a MALDI-TOF mass spectrum of the whole bacteria, determining the presence or absence of a variant of the phenol soluble modulin peptide (PSM-mec) wherein the presence of the variant of the PSM-mec peptide is determined by the presence of a mass signal in the MALDI-TOF mass spectrum centered at m/z 2415, and indicating the classified Staphylococcus aureus as a methicillin resistant Staphylococcus aureus when the variant of the PSM-mec peptide is present. 2. The method according to claim 1 wherein the bacteria are classified as Staphylococcus aureus by comparing the MALDI-TOF mass spectrum with reference mass spectra of a library comprising at least one reference mass spectrum of Staphylococcus aureus. 3-7. (canceled) 8. The method according to claim 1 wherein the presence of the variant of the PSM-mec peptide is determined by tandem mass spectrometry of the bacterial sample comprising the selection of parent ions centered at m/z 2415. 9. The method according to claim 1 wherein the variant of the PSM-mec peptide is the formylated version of the PSM-mec peptide. 10. The method according to claim 1 further comprising: determining a status of an agr system of Staphylococcus aureus of the bacterial sample, wherein the presence of the agr system and the presence of the variant of the PSM-mec peptide indicates a methicillin resistant Staphylococcus aureus. 11. The method according to claim 1 further comprising: determining a status of an agr system of Staphylococcus aureus of the bacterial sample, wherein the presence of the agr system and the absence of the variant of the PSM-mec peptide indicates a methicillin susceptible Staphylococcus aureus. 12. The method according to claim 11, wherein the presence or absence of delta-toxin is determined in the bacterial sample and the presence of the delta-toxin indicates the presence of the agr system. 13. The method according to claim 12, wherein the delta toxin is determined to be present in the bacterial sample when a mass signal at m/z 3007 or m/z 3037 is present in the MALDI TOF mass spectrum. 14. The method according to claim 1 wherein the bacterial sample stems from one of an agar plate, liquid nutrient broth, a smear, a body fluid and a blood culture. 15. The method according to claim 1 comprising the steps: (a) cultivating a sample to be analyzed on an agar plate; (b) selecting at least one bacterial colony of the agar plate as bacterial sample; (c) acquiring a MALDI TOF mass spectrum of whole cells of bacteria from the at least one colony; (d) classifying the bacteria by comparing the MALDI TOF mass spectrum with reference mass spectra of a library comprising at least one reference mass spectrum of Staphylococcus aureus; and (e) determining the presence or absence of a first and a second mass signal in the MALDI TOF mass spectrum, wherein the first mass signal is centered at m/z 3007 or m z 3037 and the second mass signal is centered at m z 2415; wherein the bacteria of the at least one colony are determined to be methicillin resistant if the first and the second mass signal are present and wherein the bacteria of the at least one colony are determined to be methicillin susceptible if the first mass signal is present and the second mass signal is absent. 16. The method according to claim 1 wherein an additional antibiotic susceptibility test is performed after determining the presence of the PSM-mec peptide or variant thereof in order to confirm the identification of methicillin resistant Staphylococcus aureus. 17. The method according to claim 10, wherein the presence or absence of delta-toxin is determined in the bacterial sample and the presence of the delta-toxin indicates the presence of the agr system. 18. The method according to claim 17, wherein the delta toxin is determined to be present in the bacterial sample when a mass signal at m/z 3007 or m/z 3037 is present in a the MALDI TOF mass spectrum. 19. The method according to claim 1 further comprising: determining a status of an agr system of Staphylococcus aureus of the bacterial sample and assigning the Staphylococcus aureus to be undetermined with regard to methicillin resistant and methicillin susceptible in case of an agr negative status. 20. The method according to claim 19, wherein the presence or absence of delta-toxin is determined in the bacterial sample and the presence of the delta-toxin indicates the presence of the agr system. 21. The method according to claim 20, wherein the delta toxin is determined to be present in the bacterial sample when a mass signal at m/z 3007 or m/z 3037 is present in a the MALDI TOF mass spectrum. 22. The method according to claim 15 wherein the bacteria are assigned to be undetermined with regard to methicillin resistant and methicillin susceptible if the first mass signal is absent. 23. A method for identifying methicillin resistant Staphylococcus aureus (MRSA) in a bacterial sample comprising the steps: classifying bacteria in the sample as Staphylococcus aureus (SA) and determining the presence or absence of a variant of the phenol soluble modulin peptide (PSM-mec) wherein a solvent is applied to the bacterial sample, a mass spectrum of a resulting supernatant is acquired and the presence of the variant of the PSM-mec peptide is determined by the presence of a mass signal in the mass spectrum centered at m/z 2415.
The invention provides a method for identifying methicillin resistant Staphylococcus aureus (MRSA) in a bacterial sample comprising the steps: classifying bacteria in the sample as Staphylococcus aureus (SA) and determining the presence or absence of the phenol soluble modulin peptide or a variant thereof wherein the presence of the PSM-mec peptide or variant thereof indicates methicillin resistant Staphylococcus aureus. The variant is preferably the formylated version of the PSM-mec peptide having a mass to charge ratio of 2415 in a singly protonated state.1. A method for identifying methicillin resistant Staphylococcus aureus (MRSA) in a bacterial sample comprising the steps: classifying bacteria in the sample as Staphylococcus aureus (SA), directly depositing whole bacteria cells on a MALDI time of flight (MALDI-TOF) mass spectrometer target plate without prior extraction, acquiring a MALDI-TOF mass spectrum of the whole bacteria, determining the presence or absence of a variant of the phenol soluble modulin peptide (PSM-mec) wherein the presence of the variant of the PSM-mec peptide is determined by the presence of a mass signal in the MALDI-TOF mass spectrum centered at m/z 2415, and indicating the classified Staphylococcus aureus as a methicillin resistant Staphylococcus aureus when the variant of the PSM-mec peptide is present. 2. The method according to claim 1 wherein the bacteria are classified as Staphylococcus aureus by comparing the MALDI-TOF mass spectrum with reference mass spectra of a library comprising at least one reference mass spectrum of Staphylococcus aureus. 3-7. (canceled) 8. The method according to claim 1 wherein the presence of the variant of the PSM-mec peptide is determined by tandem mass spectrometry of the bacterial sample comprising the selection of parent ions centered at m/z 2415. 9. The method according to claim 1 wherein the variant of the PSM-mec peptide is the formylated version of the PSM-mec peptide. 10. The method according to claim 1 further comprising: determining a status of an agr system of Staphylococcus aureus of the bacterial sample, wherein the presence of the agr system and the presence of the variant of the PSM-mec peptide indicates a methicillin resistant Staphylococcus aureus. 11. The method according to claim 1 further comprising: determining a status of an agr system of Staphylococcus aureus of the bacterial sample, wherein the presence of the agr system and the absence of the variant of the PSM-mec peptide indicates a methicillin susceptible Staphylococcus aureus. 12. The method according to claim 11, wherein the presence or absence of delta-toxin is determined in the bacterial sample and the presence of the delta-toxin indicates the presence of the agr system. 13. The method according to claim 12, wherein the delta toxin is determined to be present in the bacterial sample when a mass signal at m/z 3007 or m/z 3037 is present in the MALDI TOF mass spectrum. 14. The method according to claim 1 wherein the bacterial sample stems from one of an agar plate, liquid nutrient broth, a smear, a body fluid and a blood culture. 15. The method according to claim 1 comprising the steps: (a) cultivating a sample to be analyzed on an agar plate; (b) selecting at least one bacterial colony of the agar plate as bacterial sample; (c) acquiring a MALDI TOF mass spectrum of whole cells of bacteria from the at least one colony; (d) classifying the bacteria by comparing the MALDI TOF mass spectrum with reference mass spectra of a library comprising at least one reference mass spectrum of Staphylococcus aureus; and (e) determining the presence or absence of a first and a second mass signal in the MALDI TOF mass spectrum, wherein the first mass signal is centered at m/z 3007 or m z 3037 and the second mass signal is centered at m z 2415; wherein the bacteria of the at least one colony are determined to be methicillin resistant if the first and the second mass signal are present and wherein the bacteria of the at least one colony are determined to be methicillin susceptible if the first mass signal is present and the second mass signal is absent. 16. The method according to claim 1 wherein an additional antibiotic susceptibility test is performed after determining the presence of the PSM-mec peptide or variant thereof in order to confirm the identification of methicillin resistant Staphylococcus aureus. 17. The method according to claim 10, wherein the presence or absence of delta-toxin is determined in the bacterial sample and the presence of the delta-toxin indicates the presence of the agr system. 18. The method according to claim 17, wherein the delta toxin is determined to be present in the bacterial sample when a mass signal at m/z 3007 or m/z 3037 is present in a the MALDI TOF mass spectrum. 19. The method according to claim 1 further comprising: determining a status of an agr system of Staphylococcus aureus of the bacterial sample and assigning the Staphylococcus aureus to be undetermined with regard to methicillin resistant and methicillin susceptible in case of an agr negative status. 20. The method according to claim 19, wherein the presence or absence of delta-toxin is determined in the bacterial sample and the presence of the delta-toxin indicates the presence of the agr system. 21. The method according to claim 20, wherein the delta toxin is determined to be present in the bacterial sample when a mass signal at m/z 3007 or m/z 3037 is present in a the MALDI TOF mass spectrum. 22. The method according to claim 15 wherein the bacteria are assigned to be undetermined with regard to methicillin resistant and methicillin susceptible if the first mass signal is absent. 23. A method for identifying methicillin resistant Staphylococcus aureus (MRSA) in a bacterial sample comprising the steps: classifying bacteria in the sample as Staphylococcus aureus (SA) and determining the presence or absence of a variant of the phenol soluble modulin peptide (PSM-mec) wherein a solvent is applied to the bacterial sample, a mass spectrum of a resulting supernatant is acquired and the presence of the variant of the PSM-mec peptide is determined by the presence of a mass signal in the mass spectrum centered at m/z 2415.
1,600
587
15,291,504
1,613
Process for thickening a composition, through the use of a direct emulsion in water, which is alkali-swellable, of a polymer of the ASE or HASE type, which is both rich in acrylic acid and which has a certain quantity of AMPS. The use of such emulsions simultaneously allows there to be no obligation to use surfactants and organic solvents other than water, and allows the thickening phenomenon to be activated for pHs of less than 7: this latter characteristic is particularly advantageous for formulations intended to he used in contact with skin.
1. A method for thickening a formulation, the method comprising: contacting the formulation with a direct aqueous emulsion of a polymer followed by regulating a pH of the formulation to a value between 5 and 7, thereby forming a thickened formulation, wherein the emulsion is free from surfactants and organic solvents other than water and the polymer consists, expressed as a content by weight of monomers therein, of: a) from 20% to 60% by weight of acrylic acid and, optionally, methacrylic acid, wherein a content by weight of acrylic acid compared to a total weight of acrylic and methacrylic acid is at least 50%, b) from 40% to 80% by weight of ethyl acrylate, butyl acrylate, methyl methacrylate, or any combination thereof, c) from 0.05% to 22% by weight of 2-acrylamido-2-methylpropane sulfonic acid, and d) from 0 to 1% by weight of at least one cross-linked monomer. 2-3. (canceled) 4. The method according to claim 1, wherein the cross-linked monomer is ethylene glycol dimethacrylate, trimethylolpropanetriacrylate, diallyl phtalate, allyle acrylate, an allyl maleate, methylene-bis-acrylamide, methylene-bis-methacrylamide, tetrallyloxyethane, a triallylcyanurate, an allylic ether, an acrylic ester obtained from a polyol, a methacrylic ester obtained from a polyol, a methacrylurethane ester obtained from a polyol or any combination thereof. 5. (canceled) 6. The method according to one of the claim 1, wherein the aqueous emulsion has a dry extract of between 10% and 50% by dry weight of polymer, compared to its total weight. 7. The method according to claim 1, wherein the emulsion has a particle size of between 50 nm and 500 nm. 8. The method according to claim 1, wherein the polymer has an average molar mass by weight of between 20,000 g/mole and 1,000,000 g/mole. 9. The method according to claim 1, wherein the content by weight of acrylic acid compared to the total weight of the acrylic acid and methacrylic acid is from 80% to 100%. 10. The method according to claim 1, wherein where the content by weight of acrylic acid compared to the total weight of the acrylic acid and methacrylic acid is from 98% to 100%. 11. The method according to claim 1, wherein the polymer does not comprise methacrylic acid. 12. (canceled) 13. The method according to claim 1, wherein the formulation is a cosmetic formulation. 14. The method according to claim 13, wherein the cosmetic formulation comprises a surfactant and is suitable for application to human skin. 15. (canceled) 16. The method according to claim 1, wherein the formulation has a pH of from 5 to 6.5 after thickening. 17. The method according to claim 11, wherein the formulation has a pH of from 5.5 to 6 after thickening. 18. The method according to claim 1, wherein the polymer does not comprise a monomer having a hydrophobic group. 19. The method according to claim 1, wherein the 40% to 80% by weight of (b) is a combination of two monomers selected from the group consisting of ethyl acrylate, butyl acrylate, and methyl methacrylate, or else is a combination of ethyl acrylate, butyl acrylate, and methyl methacrylate. 20. The method according to claim 1, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 1200 mPa·s at a pH of 7. 21. The method according to claim 1, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 400 mPa·s greater at a pH of 6 than at a of 5. 22. The method according to claim 1, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 1200 mPa·s greater at a pH of 7 than at a pH of 5. 23. The method according to claim 21, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 1200 mPa·s greater at a pH. of 7 than at a pH of 5.
Process for thickening a composition, through the use of a direct emulsion in water, which is alkali-swellable, of a polymer of the ASE or HASE type, which is both rich in acrylic acid and which has a certain quantity of AMPS. The use of such emulsions simultaneously allows there to be no obligation to use surfactants and organic solvents other than water, and allows the thickening phenomenon to be activated for pHs of less than 7: this latter characteristic is particularly advantageous for formulations intended to he used in contact with skin.1. A method for thickening a formulation, the method comprising: contacting the formulation with a direct aqueous emulsion of a polymer followed by regulating a pH of the formulation to a value between 5 and 7, thereby forming a thickened formulation, wherein the emulsion is free from surfactants and organic solvents other than water and the polymer consists, expressed as a content by weight of monomers therein, of: a) from 20% to 60% by weight of acrylic acid and, optionally, methacrylic acid, wherein a content by weight of acrylic acid compared to a total weight of acrylic and methacrylic acid is at least 50%, b) from 40% to 80% by weight of ethyl acrylate, butyl acrylate, methyl methacrylate, or any combination thereof, c) from 0.05% to 22% by weight of 2-acrylamido-2-methylpropane sulfonic acid, and d) from 0 to 1% by weight of at least one cross-linked monomer. 2-3. (canceled) 4. The method according to claim 1, wherein the cross-linked monomer is ethylene glycol dimethacrylate, trimethylolpropanetriacrylate, diallyl phtalate, allyle acrylate, an allyl maleate, methylene-bis-acrylamide, methylene-bis-methacrylamide, tetrallyloxyethane, a triallylcyanurate, an allylic ether, an acrylic ester obtained from a polyol, a methacrylic ester obtained from a polyol, a methacrylurethane ester obtained from a polyol or any combination thereof. 5. (canceled) 6. The method according to one of the claim 1, wherein the aqueous emulsion has a dry extract of between 10% and 50% by dry weight of polymer, compared to its total weight. 7. The method according to claim 1, wherein the emulsion has a particle size of between 50 nm and 500 nm. 8. The method according to claim 1, wherein the polymer has an average molar mass by weight of between 20,000 g/mole and 1,000,000 g/mole. 9. The method according to claim 1, wherein the content by weight of acrylic acid compared to the total weight of the acrylic acid and methacrylic acid is from 80% to 100%. 10. The method according to claim 1, wherein where the content by weight of acrylic acid compared to the total weight of the acrylic acid and methacrylic acid is from 98% to 100%. 11. The method according to claim 1, wherein the polymer does not comprise methacrylic acid. 12. (canceled) 13. The method according to claim 1, wherein the formulation is a cosmetic formulation. 14. The method according to claim 13, wherein the cosmetic formulation comprises a surfactant and is suitable for application to human skin. 15. (canceled) 16. The method according to claim 1, wherein the formulation has a pH of from 5 to 6.5 after thickening. 17. The method according to claim 11, wherein the formulation has a pH of from 5.5 to 6 after thickening. 18. The method according to claim 1, wherein the polymer does not comprise a monomer having a hydrophobic group. 19. The method according to claim 1, wherein the 40% to 80% by weight of (b) is a combination of two monomers selected from the group consisting of ethyl acrylate, butyl acrylate, and methyl methacrylate, or else is a combination of ethyl acrylate, butyl acrylate, and methyl methacrylate. 20. The method according to claim 1, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 1200 mPa·s at a pH of 7. 21. The method according to claim 1, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 400 mPa·s greater at a pH of 6 than at a of 5. 22. The method according to claim 1, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 1200 mPa·s greater at a pH of 7 than at a pH of 5. 23. The method according to claim 21, wherein, for a 1% dry weight content of the polymer in the thickened formulation, a viscosity of the thickened formulation is at least 1200 mPa·s greater at a pH. of 7 than at a pH of 5.
1,600
588
13,630,577
1,632
The present invention relates to the field of degradation with hyperthermophilic organisms, and in particular to the use of hyperthermophilic degradation to produce heat and energy rich components including hydrogen and ethanol from a biomass. In some embodiments, a biomass is fermented in the presence of hyperthermophilic organisms to produce heat. The heat is used to heat a liquid which is used directly in a heat pump or radiant heat or to produce electricity or drive a steam turbine. In some embodiments, acetate is utilized as a substrate to produce energy by methanogenesis.
1. A process comprising: providing a biomass that contains a pathogen and a population of at least one genus of a hyperthermophilic organism; culturing said population of at least one genus of a hyperthermophilic organism under anaerobic conditions on said biomass at a temperature of 80° C. or higher; maintaining said culture at temperature of 80° C. or higher so that said pathogens are destroyed to provide a decontaminated biomass residue; and processing said decontaminated biomass residue. 2. The process of claim 1, further comprising the step of separating at least one degradation product from said biomass. 3. The process of claim 1, further comprising the step of converting said degradation product into heat or electricity. 4. The process of claim 1, wherein said biomass is selected from the group consisting of sewage, agricultural waste products, food waste and manure. 5. The process of claim 4, wherein said biomass selected from the group consisting of sewage, agricultural waste products, food waste and manure is mixed with an algae biomass. 6. The process of claim 1, wherein at least one degradation product is selected from the group consisting of hydrogen, acetate, methane and ethanol and combinations thereof. 7. The process of claim 1, wherein said degradation product is acetate and further comprising the step of feeding said acetate to methanogenic bacteria under conditions such that said acetate is converted to methane. 8. The process of claim 1, further comprising the step of feeding at least one degradation product to a culture system comprising algae under conditions such that the growth of said algae is enhanced. 9. The process of claim 8, wherein said degradation product is acetate. 10. The process of claim 1, wherein said conditions comprising maintaining said microorganism in a stationary phase.
The present invention relates to the field of degradation with hyperthermophilic organisms, and in particular to the use of hyperthermophilic degradation to produce heat and energy rich components including hydrogen and ethanol from a biomass. In some embodiments, a biomass is fermented in the presence of hyperthermophilic organisms to produce heat. The heat is used to heat a liquid which is used directly in a heat pump or radiant heat or to produce electricity or drive a steam turbine. In some embodiments, acetate is utilized as a substrate to produce energy by methanogenesis.1. A process comprising: providing a biomass that contains a pathogen and a population of at least one genus of a hyperthermophilic organism; culturing said population of at least one genus of a hyperthermophilic organism under anaerobic conditions on said biomass at a temperature of 80° C. or higher; maintaining said culture at temperature of 80° C. or higher so that said pathogens are destroyed to provide a decontaminated biomass residue; and processing said decontaminated biomass residue. 2. The process of claim 1, further comprising the step of separating at least one degradation product from said biomass. 3. The process of claim 1, further comprising the step of converting said degradation product into heat or electricity. 4. The process of claim 1, wherein said biomass is selected from the group consisting of sewage, agricultural waste products, food waste and manure. 5. The process of claim 4, wherein said biomass selected from the group consisting of sewage, agricultural waste products, food waste and manure is mixed with an algae biomass. 6. The process of claim 1, wherein at least one degradation product is selected from the group consisting of hydrogen, acetate, methane and ethanol and combinations thereof. 7. The process of claim 1, wherein said degradation product is acetate and further comprising the step of feeding said acetate to methanogenic bacteria under conditions such that said acetate is converted to methane. 8. The process of claim 1, further comprising the step of feeding at least one degradation product to a culture system comprising algae under conditions such that the growth of said algae is enhanced. 9. The process of claim 8, wherein said degradation product is acetate. 10. The process of claim 1, wherein said conditions comprising maintaining said microorganism in a stationary phase.
1,600
589
15,307,948
1,651
In the present invention, for test cells which are either stem cells whose state of differentiation is unknown or cells obtained from stem cells by differentiation induction, an LC-MS or GC-MS analysis is performed on culture supernatants collected from a culture dish of the test cells and a culture dish of control cells whose state of differentiation is known, and the state of differentiation of the test cells is assessed based on the amount, determined as a result of the aforementioned analysis, of at least one compound selected from the group of putrescine, kynurenine, cystathionine, ascorbic acid, riboflavin, pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid in both the culture supernatant of the test cells and the culture supernatant of the control cells.
1. A cell differentiation state assessment method, comprising: assessing a state of differentiation of test cells based on an amount of a specified substance in a culture supernatant of the test cells, the test cells being stem cells whose state of differentiation is unknown or cells obtained from stem cells by differentiation induction, wherein the specified substance is at least one compound selected from the group consisting of putrescine, kynurenine, cystathionine, ascorbic acid, riboflavin, pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid. 2. The cell differentiation state assessment method according to claim 1, wherein the state of differentiation of the test cells is assessed by comparing the amount of the specified substance in a culture supernatant of the test cells and the amount of the specified substance in a culture supernatant of control cells whose state of differentiation is known. 3. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably differentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of putrescine, kynurenine, cystathionine, ascorbic acid and riboflavin in the supernatant of the test cells to the amount of the same compound in the supernatant of the control cells is equal to or higher than a predetermined threshold, it is concluded that the stem cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 4. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably differentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid in the supernatant of the control cells to the amount of the same compound in the supernatant of the test cells is equal to or higher than a threshold, it is concluded that the stem cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 5. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably undifferentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of putrescine, kynurenine, cystathionine, ascorbic acid and riboflavin in the supernatant of the test cells to the amount of the same compound in the supernatant of the control cells is equal to or higher than a predetermined threshold, it is concluded that the stem cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 6. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably undifferentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid in the supernatant of the control cells to the amount of the same compound in the supernatant of the test cells is equal to or higher than a threshold, it is concluded that the stern cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 7. The cell differentiation state assessment method according to claim 1, wherein the stem cells are pluripotent stem cells. 8. The cell differentiation state assessment method according to claim 1, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 9. The cell differentiation state assessment method according to claim 2, wherein the stem cells are pluripotent stem cells. 10. The cell differentiation state assessment method according to claim 2, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 11. The cell differentiation state assessment method according to claim 3, wherein the stem cells are pluripotent stem cells. 12. The cell differentiation state assessment method according to claim 3, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 13. The cell differentiation state assessment method according to claim 4, wherein the stem cells are pluripotent stem cells. 14. The cell differentiation state assessment method according to claim 4, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 15. The cell differentiation state assessment method according to claim 5, wherein the stem cells are pluripotent stem cells. 16. The cell differentiation state assessment method according to claim 5, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 17. The cell differentiation state assessment method according to claim 6, wherein the stem cells are pluripotent stem cells. 18. The cell differentiation state assessment method according to claim 6, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 19. The cell differentiation state assessment method according to claim 7, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 20. The cell differentiation state assessment method according to claim 9, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry.
In the present invention, for test cells which are either stem cells whose state of differentiation is unknown or cells obtained from stem cells by differentiation induction, an LC-MS or GC-MS analysis is performed on culture supernatants collected from a culture dish of the test cells and a culture dish of control cells whose state of differentiation is known, and the state of differentiation of the test cells is assessed based on the amount, determined as a result of the aforementioned analysis, of at least one compound selected from the group of putrescine, kynurenine, cystathionine, ascorbic acid, riboflavin, pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid in both the culture supernatant of the test cells and the culture supernatant of the control cells.1. A cell differentiation state assessment method, comprising: assessing a state of differentiation of test cells based on an amount of a specified substance in a culture supernatant of the test cells, the test cells being stem cells whose state of differentiation is unknown or cells obtained from stem cells by differentiation induction, wherein the specified substance is at least one compound selected from the group consisting of putrescine, kynurenine, cystathionine, ascorbic acid, riboflavin, pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid. 2. The cell differentiation state assessment method according to claim 1, wherein the state of differentiation of the test cells is assessed by comparing the amount of the specified substance in a culture supernatant of the test cells and the amount of the specified substance in a culture supernatant of control cells whose state of differentiation is known. 3. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably differentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of putrescine, kynurenine, cystathionine, ascorbic acid and riboflavin in the supernatant of the test cells to the amount of the same compound in the supernatant of the control cells is equal to or higher than a predetermined threshold, it is concluded that the stem cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 4. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably differentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid in the supernatant of the control cells to the amount of the same compound in the supernatant of the test cells is equal to or higher than a threshold, it is concluded that the stem cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 5. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably undifferentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of putrescine, kynurenine, cystathionine, ascorbic acid and riboflavin in the supernatant of the test cells to the amount of the same compound in the supernatant of the control cells is equal to or higher than a predetermined threshold, it is concluded that the stem cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 6. The cell differentiation state assessment method according to claim 2, wherein cells which are unmistakably undifferentiated are used as the control cells, and if a ratio of the amount of at least one compound selected from a group of pyruvic acid, serine, cysteine, threonic acid, citric acid, and orotic acid in the supernatant of the control cells to the amount of the same compound in the supernatant of the test cells is equal to or higher than a threshold, it is concluded that the stern cells whose state of differentiation is unknown are in an undifferentiated state or that undifferentiated cells are mixed in the cells obtained from the stem cells by differentiation induction. 7. The cell differentiation state assessment method according to claim 1, wherein the stem cells are pluripotent stem cells. 8. The cell differentiation state assessment method according to claim 1, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 9. The cell differentiation state assessment method according to claim 2, wherein the stem cells are pluripotent stem cells. 10. The cell differentiation state assessment method according to claim 2, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 11. The cell differentiation state assessment method according to claim 3, wherein the stem cells are pluripotent stem cells. 12. The cell differentiation state assessment method according to claim 3, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 13. The cell differentiation state assessment method according to claim 4, wherein the stem cells are pluripotent stem cells. 14. The cell differentiation state assessment method according to claim 4, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 15. The cell differentiation state assessment method according to claim 5, wherein the stem cells are pluripotent stem cells. 16. The cell differentiation state assessment method according to claim 5, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 17. The cell differentiation state assessment method according to claim 6, wherein the stem cells are pluripotent stem cells. 18. The cell differentiation state assessment method according to claim 6, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 19. The cell differentiation state assessment method according to claim 7, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry. 20. The cell differentiation state assessment method according to claim 9, wherein the amount of the specified substance in the culture supernatant is quantitatively determined by mass spectrometry.
1,600
590
15,094,113
1,618
Described herein is the androgen receptor modulator of formula (I) in the treatment of prostate cancer in combination with other therapeutic options and in the treatment of diseases or conditions that are amenable to treatment with the androgen receptor modulator, as well as pharmaceutical compositions and medicaments that include such compound.
1. A method of treating metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with: (a) abiraterone acetate (Zytiga); (b) Ipilimumab; (c) everolimus; (d) leuprolide or leuprolide acetate (Lupron or Eligard); or (e) goserelin acetate (Zoladex). 2. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with abiraterone acetate (Zytiga) at a dose in a range of from about 500 mg per day to about 1000 mg per day together with prednisone at a dose of about 5 mg twice per day. 3. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination Ipilimumab, administered by intravenous infusion at a dose of about 1.5 mg/Kg to about 3.0 mg/kg IV every 3 weeks for a total of 4 doses. 4. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with everolimus, administered at a dose of about 5 mg per day to about 20 mg per day. 5. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with leuprolide or leuprolide acetate (Lupron or Eligard), administered as a depot injection at a dose of about 7.5 mg every 4 weeks, or 22.5 mg every 3 months, or about 30 mg every 4 months, or about 45 mg every 6 months. 6. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with goserelin acetate (Zoladex), administered as a subcutaneous implant at a dose of about 3.6 mg every 4 weeks or about 10.8 mg every 12 weeks. 7. A method of treating metastatic castration-resistant prostate cancer, or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with a taxane or tubulin inhibitor, comprising Docetaxel or Cabazitaxel. 8. The method of claim 7, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with Docetaxel, administered by intravenous infusion at a dose of about 35 mg/m2 to about 75 mg/m2 every 3 weeks 9. The method of claim 7, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with Cabazitaxel administered by intravenous infusion at a dose of about 13 mg/m2 to about 25 mg/m2 every 3 weeks 10. A method of treating metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with: (a) a CYP17 inhibitor, wherein the CYP17 inhibitor is: (i) abiraterone acetate (Zytiga) administered at a dose in a range of from about 500 mg per day to about 1000 mg per day together with prednisone at a dose of about 5 mg twice per day; or (ii) TAK-700 (orteronel) administered at a dose in a range of from about 300 mg twice per day to about 600 mg twice per day together with prednisone at about 5 mg twice per day; or (iii) TOK-001 (galeterone); or (iv) VT-464; or (b) an osteoprotective agent, that is: (i) Denosumab, administered by subcutaneous injection at a dose of about 60 mg to about 120 mg every 4 weeks to every 6 months; or (ii) Zoledronic acid, it is administered by intravenous infusion at a dose of about 4 mg 5 every 4 weeks to every 12 weeks; or (iii) AMG-0007, CEP-37251, ALX-0141, Alendronate sodium (Fosamax), Pamidronate disodium (Aredia), Neridronic acid (Nerixia), Minodronic acid (Recalbon) or Risedronate sodium (Actonel); or (c) a radiation therapy that comprises: (i) Alpharadin, administered by intravenous infusion at a dose of about 25 to about 50 kBq/kg every 4 weeks; (ii)177Lu-J591, administered by intravenous infusion at a dose of about 30 mCi/m2 to about 70 mCi/m2; or (iii) external beam radiation therapy (including Proton beam), or brachytherapy; or (d) Provenge administered in 3 doses at approximately 2 weeks interval; or (e) Prostvac administered by subcutaneous injection Ipilimumab; or (f) BMS-936558 administered by intravenous infusion at a dose of about 1.0 mg/kg to about 10 mg/kg on days 1, 15 and 29 of 6-week cycles; or (g) Ipilimumab administered by intravenous infusion at a dose of about 1.5 mg/Kg to about 3.0 mg/kg IV every 3 weeks for a total of 4 doses; or (h) an anti-STEAP-1 antibody drug conjugate, comprising RG7450 (DSTP3086S); or (i) a HSP90 or HSP27 pathway modulator, comprising: (i) OGX-011 (Custirsen), administered by intravenous infusion at a dose of about 320 mg to about 640 mg every week; or (ii) OGX-427, administered by intravenous infusion at a loading dose of about 300 mg to about 600 mg followed by about 500 mg to about 1000 mg every week, (iii) AUY922, HSP990, PF-04928473, PF-04929113 (SNX-5422), Retaspimycin or AT13387. 11. The method of claim 10, further comprising administering a therapeutically effective amount of a gonadotropin-releasing hormone agonist or antagonist to the male human patient. 12. A method of treating post-abiraterone acetate treated metastatic castration-resistant prostate cancer, metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof: to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day in combination with: (a) a phosphoinositide 3-kinase (PI3K) inhibitor, TORC inhibitor, or dual PI3K/TORC inhibitor, that comprises everolimus, BEZ-235, BKM120, BGT226, BYL-719, GDC0068, GDC-0980, GDC0941, GDC0032, MK-2206, OSI-027, CC-223, AZD8055, SAR245408, SAR245409, PF04691502, WYE125132, GSK2126458, GSK-2636771, BAY806946, PF-05212384, SF1126, PX866, AMG319, ZSTK474, Cal101, PWT33597, LY-317615 (enzastaurin hydrochloride), CU-906, or CUDC-907, and if the TORC inhibitor is everolimus, it is administered at a dose of about 5 mg per day to about 20 mg per day; or (b) a kinase inhibitor, comprising: (i) a MET or VEGFR kinase inhibitor, comprising Cabozantinib (XL184), PF-2341066 (Crizotinib), ARQ-197 (Tivantinib), MK-2461, JNJ-38877605, MK-8033, INCB-28060, BMS-777607, AMG-208, LY-2801653, EMD-1214063, EMD-1204831, AMG-337, HMPL-504 (Volitinib), SAR-125844, LY2875358, ABR-215050 (Tasquinimod), CHIR-258 (Dovitinib), EXEL-7647, OSI-930, BIBF-1120, BAY-73-4506 (Regorafenib), BMS-582664 (Brivanib), JNJ-26483327, AZD-2171 (Cediranib), Sorafenib, Aflibercept, Enzastaurin, AG-013736 (Axitinib), OSI-632, or GSK-786034 (Pazopanib), and if Cabozantinib, it is administered orally at a dose of about 40 mg per day to about 100 mg per day; or (ii) an EGFR, MEK, or SRC kinase inhibitor, comprising Erlotinib, Cetuximab, Gefitinib, Canertinib, Panitumumab, Nimotuzumab, Lapatinib, Vandetanib, Afatinib, MP-412, AEE-788, Neratinib, XL-647, AC-480, Dacomitinib, AZD-8931, CUDC-101, AP-26113, CO-1686, Trametinib, Selumetinib, MEK-162, Refametinib, TAK-733, RO-5126766, BI-847325, AZD6244, GSK1120212, PF-5208763 (Bosutinib), or AZD-0530 (Saracatinib), and if Erlotinib, it is administered orally at a dose in a range of from about 100 mg to about 150 mg; and if Gefitinib, it is administered orally at a dose of about 250 mg; and if Trametinib, it is administered orally at a dose of about 1 mg to about 2 mg; or (iii) an AKT, RAF, FGFR, or CDK4/6 kinase inhibitor, comprising GDC0068, MK-2206, AT7867, GSK2110183, GSK2141795, GSK690693, Vemurafenib (PLX4032/RG7204), GSK2118436, Dabrafenib (GSK208436), LGX818, RAF265, LY2780301, Dovitinib (TKI258), BGJ398, AZD4547, PD-0332991 or LEE011. 13. The method of claim 12, further comprising administering a therapeutically effective amount of a gonadotropin-releasing hormone agonist or antagonist to the male human. 14. A method of treating metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with a gonadotropin-releasing hormone agonist or antagonist, comprising Lupron, Zoladex (Goserelin), Degarelix, Ozarelix, ABT-620 (Elagolix), TAK-385 (Relugolix), EP-100 or KLH-2109, and if Lupron, it is administered as a depot injection at a dose of about 7.5 mg every 4 weeks, or 22.5 mg every 3 months, or about 30 mg every 4 months, or about 45 mg every 6 months, and if Zoladex (Goserelin), it is administered as a subcutaneous implant at a dose of about 3.6 mg every 4 weeks or about 10.8 mg every 12 weeks, and if Degarelix, it is administered as a subcutaneous injection at a dose of about 240 mg followed by about 80 mg administered every 4 weeks. 15. A method of treating breast cancer, androgen dependent hirsutism, androgenic alopecia, uterine fibroids, leiomyoma, endometrial carcinoma or endometriosis in a human patient, said method comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human patient in need of such treatment at a dose in a range of from about 30 mg per day to about 480 mg per day on a continuous daily dosing schedule 16. The method of claim 15, further comprising administering a therapeutically effective amount of a gonadotropin-releasing hormone agonist or antagonist to the male human patient. 17. A method of treating a male human patient having advanced castration-sensitive prostate cancer, castration-resistant prostate cancer, or high-risk localized prostate cancer comprising administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, at a dose in a range of from about 30 mg per day to about 480 mg per day to a male human patient in need of such treatment. 18. The method of claim 17, wherein the treating results in a decrease in the patient's prostate-specific antigen levels of at least 50% from baseline after 3 months of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, on a continuous daily dosing schedule.
Described herein is the androgen receptor modulator of formula (I) in the treatment of prostate cancer in combination with other therapeutic options and in the treatment of diseases or conditions that are amenable to treatment with the androgen receptor modulator, as well as pharmaceutical compositions and medicaments that include such compound.1. A method of treating metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with: (a) abiraterone acetate (Zytiga); (b) Ipilimumab; (c) everolimus; (d) leuprolide or leuprolide acetate (Lupron or Eligard); or (e) goserelin acetate (Zoladex). 2. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with abiraterone acetate (Zytiga) at a dose in a range of from about 500 mg per day to about 1000 mg per day together with prednisone at a dose of about 5 mg twice per day. 3. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination Ipilimumab, administered by intravenous infusion at a dose of about 1.5 mg/Kg to about 3.0 mg/kg IV every 3 weeks for a total of 4 doses. 4. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with everolimus, administered at a dose of about 5 mg per day to about 20 mg per day. 5. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with leuprolide or leuprolide acetate (Lupron or Eligard), administered as a depot injection at a dose of about 7.5 mg every 4 weeks, or 22.5 mg every 3 months, or about 30 mg every 4 months, or about 45 mg every 6 months. 6. The method of claim 1, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with goserelin acetate (Zoladex), administered as a subcutaneous implant at a dose of about 3.6 mg every 4 weeks or about 10.8 mg every 12 weeks. 7. A method of treating metastatic castration-resistant prostate cancer, or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with a taxane or tubulin inhibitor, comprising Docetaxel or Cabazitaxel. 8. The method of claim 7, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with Docetaxel, administered by intravenous infusion at a dose of about 35 mg/m2 to about 75 mg/m2 every 3 weeks 9. The method of claim 7, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with Cabazitaxel administered by intravenous infusion at a dose of about 13 mg/m2 to about 25 mg/m2 every 3 weeks 10. A method of treating metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with: (a) a CYP17 inhibitor, wherein the CYP17 inhibitor is: (i) abiraterone acetate (Zytiga) administered at a dose in a range of from about 500 mg per day to about 1000 mg per day together with prednisone at a dose of about 5 mg twice per day; or (ii) TAK-700 (orteronel) administered at a dose in a range of from about 300 mg twice per day to about 600 mg twice per day together with prednisone at about 5 mg twice per day; or (iii) TOK-001 (galeterone); or (iv) VT-464; or (b) an osteoprotective agent, that is: (i) Denosumab, administered by subcutaneous injection at a dose of about 60 mg to about 120 mg every 4 weeks to every 6 months; or (ii) Zoledronic acid, it is administered by intravenous infusion at a dose of about 4 mg 5 every 4 weeks to every 12 weeks; or (iii) AMG-0007, CEP-37251, ALX-0141, Alendronate sodium (Fosamax), Pamidronate disodium (Aredia), Neridronic acid (Nerixia), Minodronic acid (Recalbon) or Risedronate sodium (Actonel); or (c) a radiation therapy that comprises: (i) Alpharadin, administered by intravenous infusion at a dose of about 25 to about 50 kBq/kg every 4 weeks; (ii)177Lu-J591, administered by intravenous infusion at a dose of about 30 mCi/m2 to about 70 mCi/m2; or (iii) external beam radiation therapy (including Proton beam), or brachytherapy; or (d) Provenge administered in 3 doses at approximately 2 weeks interval; or (e) Prostvac administered by subcutaneous injection Ipilimumab; or (f) BMS-936558 administered by intravenous infusion at a dose of about 1.0 mg/kg to about 10 mg/kg on days 1, 15 and 29 of 6-week cycles; or (g) Ipilimumab administered by intravenous infusion at a dose of about 1.5 mg/Kg to about 3.0 mg/kg IV every 3 weeks for a total of 4 doses; or (h) an anti-STEAP-1 antibody drug conjugate, comprising RG7450 (DSTP3086S); or (i) a HSP90 or HSP27 pathway modulator, comprising: (i) OGX-011 (Custirsen), administered by intravenous infusion at a dose of about 320 mg to about 640 mg every week; or (ii) OGX-427, administered by intravenous infusion at a loading dose of about 300 mg to about 600 mg followed by about 500 mg to about 1000 mg every week, (iii) AUY922, HSP990, PF-04928473, PF-04929113 (SNX-5422), Retaspimycin or AT13387. 11. The method of claim 10, further comprising administering a therapeutically effective amount of a gonadotropin-releasing hormone agonist or antagonist to the male human patient. 12. A method of treating post-abiraterone acetate treated metastatic castration-resistant prostate cancer, metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof: to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day in combination with: (a) a phosphoinositide 3-kinase (PI3K) inhibitor, TORC inhibitor, or dual PI3K/TORC inhibitor, that comprises everolimus, BEZ-235, BKM120, BGT226, BYL-719, GDC0068, GDC-0980, GDC0941, GDC0032, MK-2206, OSI-027, CC-223, AZD8055, SAR245408, SAR245409, PF04691502, WYE125132, GSK2126458, GSK-2636771, BAY806946, PF-05212384, SF1126, PX866, AMG319, ZSTK474, Cal101, PWT33597, LY-317615 (enzastaurin hydrochloride), CU-906, or CUDC-907, and if the TORC inhibitor is everolimus, it is administered at a dose of about 5 mg per day to about 20 mg per day; or (b) a kinase inhibitor, comprising: (i) a MET or VEGFR kinase inhibitor, comprising Cabozantinib (XL184), PF-2341066 (Crizotinib), ARQ-197 (Tivantinib), MK-2461, JNJ-38877605, MK-8033, INCB-28060, BMS-777607, AMG-208, LY-2801653, EMD-1214063, EMD-1204831, AMG-337, HMPL-504 (Volitinib), SAR-125844, LY2875358, ABR-215050 (Tasquinimod), CHIR-258 (Dovitinib), EXEL-7647, OSI-930, BIBF-1120, BAY-73-4506 (Regorafenib), BMS-582664 (Brivanib), JNJ-26483327, AZD-2171 (Cediranib), Sorafenib, Aflibercept, Enzastaurin, AG-013736 (Axitinib), OSI-632, or GSK-786034 (Pazopanib), and if Cabozantinib, it is administered orally at a dose of about 40 mg per day to about 100 mg per day; or (ii) an EGFR, MEK, or SRC kinase inhibitor, comprising Erlotinib, Cetuximab, Gefitinib, Canertinib, Panitumumab, Nimotuzumab, Lapatinib, Vandetanib, Afatinib, MP-412, AEE-788, Neratinib, XL-647, AC-480, Dacomitinib, AZD-8931, CUDC-101, AP-26113, CO-1686, Trametinib, Selumetinib, MEK-162, Refametinib, TAK-733, RO-5126766, BI-847325, AZD6244, GSK1120212, PF-5208763 (Bosutinib), or AZD-0530 (Saracatinib), and if Erlotinib, it is administered orally at a dose in a range of from about 100 mg to about 150 mg; and if Gefitinib, it is administered orally at a dose of about 250 mg; and if Trametinib, it is administered orally at a dose of about 1 mg to about 2 mg; or (iii) an AKT, RAF, FGFR, or CDK4/6 kinase inhibitor, comprising GDC0068, MK-2206, AT7867, GSK2110183, GSK2141795, GSK690693, Vemurafenib (PLX4032/RG7204), GSK2118436, Dabrafenib (GSK208436), LGX818, RAF265, LY2780301, Dovitinib (TKI258), BGJ398, AZD4547, PD-0332991 or LEE011. 13. The method of claim 12, further comprising administering a therapeutically effective amount of a gonadotropin-releasing hormone agonist or antagonist to the male human. 14. A method of treating metastatic castration-resistant prostate cancer, non-metastatic castration-resistant prostate cancer, metastatic castration-sensitive prostate cancer, non-metastatic castration-sensitive prostate cancer or high-risk localized prostate cancer in a male human patient comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human male patient in need of such treatment at a dose of about 30 mg per day to about 480 mg per day, in combination with a gonadotropin-releasing hormone agonist or antagonist, comprising Lupron, Zoladex (Goserelin), Degarelix, Ozarelix, ABT-620 (Elagolix), TAK-385 (Relugolix), EP-100 or KLH-2109, and if Lupron, it is administered as a depot injection at a dose of about 7.5 mg every 4 weeks, or 22.5 mg every 3 months, or about 30 mg every 4 months, or about 45 mg every 6 months, and if Zoladex (Goserelin), it is administered as a subcutaneous implant at a dose of about 3.6 mg every 4 weeks or about 10.8 mg every 12 weeks, and if Degarelix, it is administered as a subcutaneous injection at a dose of about 240 mg followed by about 80 mg administered every 4 weeks. 15. A method of treating breast cancer, androgen dependent hirsutism, androgenic alopecia, uterine fibroids, leiomyoma, endometrial carcinoma or endometriosis in a human patient, said method comprising orally administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a human patient in need of such treatment at a dose in a range of from about 30 mg per day to about 480 mg per day on a continuous daily dosing schedule 16. The method of claim 15, further comprising administering a therapeutically effective amount of a gonadotropin-releasing hormone agonist or antagonist to the male human patient. 17. A method of treating a male human patient having advanced castration-sensitive prostate cancer, castration-resistant prostate cancer, or high-risk localized prostate cancer comprising administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, at a dose in a range of from about 30 mg per day to about 480 mg per day to a male human patient in need of such treatment. 18. The method of claim 17, wherein the treating results in a decrease in the patient's prostate-specific antigen levels of at least 50% from baseline after 3 months of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, on a continuous daily dosing schedule.
1,600
591
14,987,256
1,699
Provided in this disclosure are methods and compositions that find use in a variety of multiplex cellular/tissue section analyses. In certain aspects, a tissue section (or planar cellular slide) is stained with a combination of “visible” labels and “invisible” labels for specific targets of interest. The visible labels are observed to obtain a result and then, based on the result, one or more of the invisible labels are detected, e.g., using digital microscopy.
1. A method for multiplex analysis of a tissue section, comprising: staining a tissue section for a first target and a second target, wherein the first target is stained with a detectable label in the visible spectrum and the second target is stained with a detectable label in the non-visible spectrum; detecting the first stain on the tissue section to obtain a result; and detecting the second stain on the tissue section based on the obtained result, wherein the second stain is not detected by fluorescence microscopy. 2. The method of claim 1, wherein detecting the first stain on the tissue section comprises visual inspection under bright field microscopy. 3. The method of claim 1, wherein detecting the second stain on the tissue section comprises digitally scanning the slide. 4. The method of claim 1, wherein the second stain is detected only on a sub-region of the tissue section, wherein the sub-region is selected based on the obtained result. 5. The method of claim 1, wherein staining for the first target is selected from the group consisting of: immunohistochemistry (IHC) staining, in-situ hybridization (ISH), histological stain, and combinations thereof. 6. The method of claim 1, wherein staining for the second target is selected from the group consisting of: immunohistochemistry (IHC) staining, in-situ hybridization (ISH), and combinations thereof. 7. The method of claim 1, wherein the tissue section is a formalin fixed and embedded in paraffin wax (FFPE) tissue section. 8. The method of claim 1, further comprising comparing the relative location of the detected first and second stains on the tissue section. 9. The method of claim 1, wherein the non-visible stain absorbs light in the range of from about 700 nm to about 1000 nm wavelength. 10. The method of claim 9, wherein the non-visible stain is a near infra-red absorbing (NIR) organic material. 11. The method of claim 9, wherein the NIR organic material that comprises one or more of the following: a cyanine group, a squarine group, a crocanaine group, a phthalocyanine group, a naphthalocyanine group, a dithiolene group, a dithiolene metal complex, or combinations thereof; or wherein the NIR organic material is 2,5-bis[(4-carboxylic-piperidylamino)thiophenyl]-croconium. 12. The method of claim 1, wherein the first or second stain is produced by an enzymatic reaction or by an organometallic catalyst. 13. The method of claim 1, wherein the tissue section is a section of a biopsy obtained from a patient. 14. The method of claim 13, wherein the first target and/or the second target are disease biomarkers. 15. The method of claim 14, wherein the disease biomarkers are selected from the group consisting of: infections disease biomarkers, cancer biomarkers, immune or autoimmune response biomarkers, genetic biomarkers, and combinations thereof. 16. The method of claim 1, further comprising staining the tissue section for at least one additional target, wherein the at least one additional target is stained with a detectable label in the visible spectrum that is distinguishable from the detectable label for the first target. 17. The method of claim 1, further comprising staining the tissue section for at least one additional target, wherein the at least one additional target is stained with a detectable label in the non-visible spectrum that is distinguishable from the detectable label for the second target. 18. A kit for staining a tissue section, comprising: one or more first labeling reagents for detecting a first target on a tissue section, wherein the one or more first labeling reagents stain the first target with a detectable label in the visible spectrum; and one or more second labeling reagents for detecting a second target on a tissue section, wherein the one or more second labeling reagents stain the second target with a detectable label in the non-visible spectrum. 19. The kit of claim 18, wherein the non-visible stain absorbs light in the range of from about 700 nm to about 1000 nm wavelength. 20. The kit of claim 19, wherein the non-visible stain is a NIR organic material that comprises one or more of the following: a cyanine group, a squarine group, a crocanaine group, a phthalocyanine group, a naphthalocyanine group, a dithiolene group, a dithiolene metal complex, or combinations thereof; or wherein the NIR organic material is 2,5-bis[(4-carboxylic-piperidylamino)thiophenyl]-croconium.
Provided in this disclosure are methods and compositions that find use in a variety of multiplex cellular/tissue section analyses. In certain aspects, a tissue section (or planar cellular slide) is stained with a combination of “visible” labels and “invisible” labels for specific targets of interest. The visible labels are observed to obtain a result and then, based on the result, one or more of the invisible labels are detected, e.g., using digital microscopy.1. A method for multiplex analysis of a tissue section, comprising: staining a tissue section for a first target and a second target, wherein the first target is stained with a detectable label in the visible spectrum and the second target is stained with a detectable label in the non-visible spectrum; detecting the first stain on the tissue section to obtain a result; and detecting the second stain on the tissue section based on the obtained result, wherein the second stain is not detected by fluorescence microscopy. 2. The method of claim 1, wherein detecting the first stain on the tissue section comprises visual inspection under bright field microscopy. 3. The method of claim 1, wherein detecting the second stain on the tissue section comprises digitally scanning the slide. 4. The method of claim 1, wherein the second stain is detected only on a sub-region of the tissue section, wherein the sub-region is selected based on the obtained result. 5. The method of claim 1, wherein staining for the first target is selected from the group consisting of: immunohistochemistry (IHC) staining, in-situ hybridization (ISH), histological stain, and combinations thereof. 6. The method of claim 1, wherein staining for the second target is selected from the group consisting of: immunohistochemistry (IHC) staining, in-situ hybridization (ISH), and combinations thereof. 7. The method of claim 1, wherein the tissue section is a formalin fixed and embedded in paraffin wax (FFPE) tissue section. 8. The method of claim 1, further comprising comparing the relative location of the detected first and second stains on the tissue section. 9. The method of claim 1, wherein the non-visible stain absorbs light in the range of from about 700 nm to about 1000 nm wavelength. 10. The method of claim 9, wherein the non-visible stain is a near infra-red absorbing (NIR) organic material. 11. The method of claim 9, wherein the NIR organic material that comprises one or more of the following: a cyanine group, a squarine group, a crocanaine group, a phthalocyanine group, a naphthalocyanine group, a dithiolene group, a dithiolene metal complex, or combinations thereof; or wherein the NIR organic material is 2,5-bis[(4-carboxylic-piperidylamino)thiophenyl]-croconium. 12. The method of claim 1, wherein the first or second stain is produced by an enzymatic reaction or by an organometallic catalyst. 13. The method of claim 1, wherein the tissue section is a section of a biopsy obtained from a patient. 14. The method of claim 13, wherein the first target and/or the second target are disease biomarkers. 15. The method of claim 14, wherein the disease biomarkers are selected from the group consisting of: infections disease biomarkers, cancer biomarkers, immune or autoimmune response biomarkers, genetic biomarkers, and combinations thereof. 16. The method of claim 1, further comprising staining the tissue section for at least one additional target, wherein the at least one additional target is stained with a detectable label in the visible spectrum that is distinguishable from the detectable label for the first target. 17. The method of claim 1, further comprising staining the tissue section for at least one additional target, wherein the at least one additional target is stained with a detectable label in the non-visible spectrum that is distinguishable from the detectable label for the second target. 18. A kit for staining a tissue section, comprising: one or more first labeling reagents for detecting a first target on a tissue section, wherein the one or more first labeling reagents stain the first target with a detectable label in the visible spectrum; and one or more second labeling reagents for detecting a second target on a tissue section, wherein the one or more second labeling reagents stain the second target with a detectable label in the non-visible spectrum. 19. The kit of claim 18, wherein the non-visible stain absorbs light in the range of from about 700 nm to about 1000 nm wavelength. 20. The kit of claim 19, wherein the non-visible stain is a NIR organic material that comprises one or more of the following: a cyanine group, a squarine group, a crocanaine group, a phthalocyanine group, a naphthalocyanine group, a dithiolene group, a dithiolene metal complex, or combinations thereof; or wherein the NIR organic material is 2,5-bis[(4-carboxylic-piperidylamino)thiophenyl]-croconium.
1,600
592
15,590,598
1,634
Methods of identifying potential skin moisturizing actives for the treatment of dry skin and method of formulating a moisturizing skin care composition using actives identified by the method. Moisturizing agents can be identified by comparing the transcriptional profile of a skin tissue sample contacted by a test agent to a negative or positive control to determine if the regulation of certain genes corresponds to the appropriate direction of regulation indicated by the control. Agents identified as skin moisturizing agents can then be incorporated into a skin moisturizing composition.
1. A screening method for identifying a cosmetic test agent as a skin moisturizing agent, the method comprising: a. contacting a skin tissue sample with a cosmetic test agent; b. generating a transcriptional profile for the skin tissue sample, wherein the transcriptional profile comprises data related to the transcription of at least two genes selected from v-rel avian reticuloendotheliosis viral oncogene homolog gene (REL); integrin beta-1 gene (ITGB1); transforming growth factor beta receptor II gene (TGFBR2); YY1-associated protein 1 gene (YY1AP1); mitogen-activated protein kinase 3 gene (MAP3K); caspase 3, apoptosis-related cysteine peptidase gene (CASP3); telomerase reverse transcriptase gene (TERT); mdm2 p53 binding protein homolog gene (MDM2); and laminin, gamma 2 gene (LAMC2); c. comparing the transcriptional profile for the skin tissue sample to a positive control transcriptional profile; and d. identifying the cosmetic test agent as a skin moisturizing agent for human skin when the transcriptional profile for the skin tissue sample and the positive control transcriptional profile are concordant. 2. The method of claim 1, wherein the skin tissue sample comprises keratinocytes and fibroblasts. 3. The method of claim 1, wherein the tissue sample is subjected to laser capture microdissection. 4. The method of claim 1, wherein generating the gene expression profile comprises: i. isolating RNA from the tissue sample, ii. using the isolated RNA to create cRNA, labeling the cRNA with a fluorescent dye, and iv. hybridizing the labeled cRNA to a microarray. 5. The method of claim 1, wherein the transcriptional profile comprises data related to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, YY1AP1, MAP3K, CASP3. 6. The method of claim 1, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, TERT, MAP3K, LAMC2. 7. The method of claim 6, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from TERT, MAP3K, LAMC2. 8. The method of claim 6, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, ITGB1, TGFBR2. 9. The method of claim 1, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, TERT, MDM2, MAP3K, LAMC2. 10. The method of claim 9, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from TERT, MAP3K, LAMC2, MDM2. 11. The method of claim 10, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from TERT, MDM2, LAMC2. 12. The method of claim 1, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, YY1AP1, MAP3K, CASP3. 13. The method of claim 12, wherein the transcriptional profile comprises data related to the transcription of at least REL and MAP3K. 14. The method of claim 13, wherein the transcriptional profile comprises data related to the transcription of at least CASP3. 15. The method of claim 1, wherein the transcriptional profile comprises data related to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, YY1AP1, MAP3K, CASP3, TERT, MDM2, and LAMC2, the positive control transcriptional profile is replaced with a negative control transcriptional profile, and identifying the cosmetic test agent as a skin moisturizing agent for human skin when the transcriptional profile for the skin tissue sample and the negative control transcriptional profile are discordant. 16. A method of formulating a moisturizing skin care composition for human skin, comprising: a. identifying a cosmetic test agent as skin moisturizing agent according to the screening method of claim 1; and b. combining an effective amount of the cosmetic test agent with a dermatologically acceptable carrier to produce the skin care composition.
Methods of identifying potential skin moisturizing actives for the treatment of dry skin and method of formulating a moisturizing skin care composition using actives identified by the method. Moisturizing agents can be identified by comparing the transcriptional profile of a skin tissue sample contacted by a test agent to a negative or positive control to determine if the regulation of certain genes corresponds to the appropriate direction of regulation indicated by the control. Agents identified as skin moisturizing agents can then be incorporated into a skin moisturizing composition.1. A screening method for identifying a cosmetic test agent as a skin moisturizing agent, the method comprising: a. contacting a skin tissue sample with a cosmetic test agent; b. generating a transcriptional profile for the skin tissue sample, wherein the transcriptional profile comprises data related to the transcription of at least two genes selected from v-rel avian reticuloendotheliosis viral oncogene homolog gene (REL); integrin beta-1 gene (ITGB1); transforming growth factor beta receptor II gene (TGFBR2); YY1-associated protein 1 gene (YY1AP1); mitogen-activated protein kinase 3 gene (MAP3K); caspase 3, apoptosis-related cysteine peptidase gene (CASP3); telomerase reverse transcriptase gene (TERT); mdm2 p53 binding protein homolog gene (MDM2); and laminin, gamma 2 gene (LAMC2); c. comparing the transcriptional profile for the skin tissue sample to a positive control transcriptional profile; and d. identifying the cosmetic test agent as a skin moisturizing agent for human skin when the transcriptional profile for the skin tissue sample and the positive control transcriptional profile are concordant. 2. The method of claim 1, wherein the skin tissue sample comprises keratinocytes and fibroblasts. 3. The method of claim 1, wherein the tissue sample is subjected to laser capture microdissection. 4. The method of claim 1, wherein generating the gene expression profile comprises: i. isolating RNA from the tissue sample, ii. using the isolated RNA to create cRNA, labeling the cRNA with a fluorescent dye, and iv. hybridizing the labeled cRNA to a microarray. 5. The method of claim 1, wherein the transcriptional profile comprises data related to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, YY1AP1, MAP3K, CASP3. 6. The method of claim 1, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, TERT, MAP3K, LAMC2. 7. The method of claim 6, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from TERT, MAP3K, LAMC2. 8. The method of claim 6, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, ITGB1, TGFBR2. 9. The method of claim 1, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, TERT, MDM2, MAP3K, LAMC2. 10. The method of claim 9, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from TERT, MAP3K, LAMC2, MDM2. 11. The method of claim 10, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from TERT, MDM2, LAMC2. 12. The method of claim 1, wherein the transcriptional profile comprises data corresponding to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, YY1AP1, MAP3K, CASP3. 13. The method of claim 12, wherein the transcriptional profile comprises data related to the transcription of at least REL and MAP3K. 14. The method of claim 13, wherein the transcriptional profile comprises data related to the transcription of at least CASP3. 15. The method of claim 1, wherein the transcriptional profile comprises data related to the transcription of at least two genes selected from REL, ITGB1, TGFBR2, YY1AP1, MAP3K, CASP3, TERT, MDM2, and LAMC2, the positive control transcriptional profile is replaced with a negative control transcriptional profile, and identifying the cosmetic test agent as a skin moisturizing agent for human skin when the transcriptional profile for the skin tissue sample and the negative control transcriptional profile are discordant. 16. A method of formulating a moisturizing skin care composition for human skin, comprising: a. identifying a cosmetic test agent as skin moisturizing agent according to the screening method of claim 1; and b. combining an effective amount of the cosmetic test agent with a dermatologically acceptable carrier to produce the skin care composition.
1,600
593
15,300,984
1,628
Embodiments of the disclosure include methods and compositions related to treatment of one or more medical conditions with one or more negative modulators of GABA A receptors. In specific embodiments, depression and/or suicidability is treated or ameliorated or prevented with one or more negative modulators of GABA A receptors, such as a partial inverse agonist of a GABA A receptor comprising an alpha5 subunit.
1. A method of treating or preventing or ameliorating at least one symptom of a medical condition in an individual, comprising the step of providing to the individual a therapeutically effective amount of one or more negative modulators of GABAA receptors, wherein the medical condition is selected from the group consisting of a depression-related disorder; an anxiety-related disorder; an attention-related disorder; a psychosis-related disorder; an eating disorder; a personality disorder; cognitive impairment following traumatic brain injury; neuropathic pain; chronic muscle or bone pain; diabetic complications resulting in nerve injury; generalized attack of muscular weakness; recurring sleep episodes during the day; migraine; addiction; suicidality; and a combination thereof. 2. The method of claim 1, wherein the onset of amelioration of one or more depression-related symptoms occurs within hours, days, or weeks. 3. The method of claim 1, wherein the GABAA receptor comprises an alpha5 subunit. 4. The method of claim 1, wherein the negative modulator is selected from the group consisting of a negative allosteric modulators acting at the benzodiazepine binding site; negative allosteric modulators acting at the barbiturate steroid binding site; negative allosteric modulators acting at the neuroactive steroid binding site; competitive antagonists of the GABAAR; negative modulators acting in the pore of the GABAAR channel; nicotinamide and related compounds; inverse agonists and antagonists of the propofol binding site of the GABAAR; and a combination thereof. 5. The method of claim 1, wherein the depression-related disorder is major depressive disorder (MDD); dysthymia; cyclothymic disorder; seasonal affective disorder/seasonal depression; depression after traumatic brain injury; postpartum depression; premenstrual dysphoric disorder; depressive symptoms associated with menopause; depression following substance abuse/withdrawal; bipolar disorder; bipolar disorder in remission; or depressive episodes of bipolar disorder. 6. The method of claim 1, wherein the anxiety-related disorder is general anxiety disorder; obsessive compulsive disorder; Impulse control disorder; anxiousness associated with depression; repeated episodes of anxiety, extreme apprehension or fear of social interaction (social phobia); panic disorders; posttraumatic stress syndrome or posttraumatic stress disorder; or separation anxiety disorder. 7. The method of claim 1, wherein the attention-related disorder is attention deficit hyperactive disorder; or adult attention deficit hyperactive disorder. 8. The method of claim 1, wherein the psychosis-related disorder is schizophrenia, schizophrenia-spectrum disorder or psychotic depressive illness. 9. The method of claim 1, wherein the eating disorder is anorexia nervosa; bulimia; or obesity. 10. The method of claim 1, wherein the individual has avoidant personality disorder; antisocial personality disorder; borderline personality disorder; conduct disorder; dependent personality disorder; depressive personality disorder; histrionic personality disorder; narcissistic personality disorder; negativistic personality disorder; obsessive-compulsive personality disorder; paranoid personality disorder; schizoid personality disorder; or schizotypal personality disorder. 11. The method of claim 1, wherein the individual has been diagnosed with the medical condition. 12. The method of claim 1, wherein the individual has not been diagnosed with the medical condition. 13. (canceled) 14. (canceled) 15. (canceled) 16. (canceled) 17. (canceled) 18. The method of claim 1, wherein the negative modulator of GABAA receptor is a partial inverse agonist of a GABAA receptor comprising an α5 subunit. 19. The method of claim 18, wherein the partial inverse agonist of a GABAA receptor comprising an α5 subunit is L-655,708, R04938581, CP-457,920, MRK-016, or a combination thereof. 20. The method of claim 19, wherein the partial inverse agonist is L-655,708. 21. (canceled) 22. (canceled) 23. (canceled) 24. The method of claim 1, further comprising the step of providing to the individual a therapeutically effective amount of another therapy. 25. The method of claim 24, wherein the other therapy is selected from the group consisting of monoamine oxidase inhibitors (MAOis), selective serotonin reuptake inhibitors(SSRis), serotonin-norepinephrine reuptake inhibitors (SNRis), norepinephrine reuptake inhibitors (NRIs), triple reuptake inhibitors, modulators of CNS acetylcholine function, stimulants, anti-glucocorticoids, antagonists of NMDA-type glutamate receptors, tricylic antidepressants (TCAs), and a combination thereof. 26. The method of claim 24, wherein the other therapy is an anti-depressant.
Embodiments of the disclosure include methods and compositions related to treatment of one or more medical conditions with one or more negative modulators of GABA A receptors. In specific embodiments, depression and/or suicidability is treated or ameliorated or prevented with one or more negative modulators of GABA A receptors, such as a partial inverse agonist of a GABA A receptor comprising an alpha5 subunit.1. A method of treating or preventing or ameliorating at least one symptom of a medical condition in an individual, comprising the step of providing to the individual a therapeutically effective amount of one or more negative modulators of GABAA receptors, wherein the medical condition is selected from the group consisting of a depression-related disorder; an anxiety-related disorder; an attention-related disorder; a psychosis-related disorder; an eating disorder; a personality disorder; cognitive impairment following traumatic brain injury; neuropathic pain; chronic muscle or bone pain; diabetic complications resulting in nerve injury; generalized attack of muscular weakness; recurring sleep episodes during the day; migraine; addiction; suicidality; and a combination thereof. 2. The method of claim 1, wherein the onset of amelioration of one or more depression-related symptoms occurs within hours, days, or weeks. 3. The method of claim 1, wherein the GABAA receptor comprises an alpha5 subunit. 4. The method of claim 1, wherein the negative modulator is selected from the group consisting of a negative allosteric modulators acting at the benzodiazepine binding site; negative allosteric modulators acting at the barbiturate steroid binding site; negative allosteric modulators acting at the neuroactive steroid binding site; competitive antagonists of the GABAAR; negative modulators acting in the pore of the GABAAR channel; nicotinamide and related compounds; inverse agonists and antagonists of the propofol binding site of the GABAAR; and a combination thereof. 5. The method of claim 1, wherein the depression-related disorder is major depressive disorder (MDD); dysthymia; cyclothymic disorder; seasonal affective disorder/seasonal depression; depression after traumatic brain injury; postpartum depression; premenstrual dysphoric disorder; depressive symptoms associated with menopause; depression following substance abuse/withdrawal; bipolar disorder; bipolar disorder in remission; or depressive episodes of bipolar disorder. 6. The method of claim 1, wherein the anxiety-related disorder is general anxiety disorder; obsessive compulsive disorder; Impulse control disorder; anxiousness associated with depression; repeated episodes of anxiety, extreme apprehension or fear of social interaction (social phobia); panic disorders; posttraumatic stress syndrome or posttraumatic stress disorder; or separation anxiety disorder. 7. The method of claim 1, wherein the attention-related disorder is attention deficit hyperactive disorder; or adult attention deficit hyperactive disorder. 8. The method of claim 1, wherein the psychosis-related disorder is schizophrenia, schizophrenia-spectrum disorder or psychotic depressive illness. 9. The method of claim 1, wherein the eating disorder is anorexia nervosa; bulimia; or obesity. 10. The method of claim 1, wherein the individual has avoidant personality disorder; antisocial personality disorder; borderline personality disorder; conduct disorder; dependent personality disorder; depressive personality disorder; histrionic personality disorder; narcissistic personality disorder; negativistic personality disorder; obsessive-compulsive personality disorder; paranoid personality disorder; schizoid personality disorder; or schizotypal personality disorder. 11. The method of claim 1, wherein the individual has been diagnosed with the medical condition. 12. The method of claim 1, wherein the individual has not been diagnosed with the medical condition. 13. (canceled) 14. (canceled) 15. (canceled) 16. (canceled) 17. (canceled) 18. The method of claim 1, wherein the negative modulator of GABAA receptor is a partial inverse agonist of a GABAA receptor comprising an α5 subunit. 19. The method of claim 18, wherein the partial inverse agonist of a GABAA receptor comprising an α5 subunit is L-655,708, R04938581, CP-457,920, MRK-016, or a combination thereof. 20. The method of claim 19, wherein the partial inverse agonist is L-655,708. 21. (canceled) 22. (canceled) 23. (canceled) 24. The method of claim 1, further comprising the step of providing to the individual a therapeutically effective amount of another therapy. 25. The method of claim 24, wherein the other therapy is selected from the group consisting of monoamine oxidase inhibitors (MAOis), selective serotonin reuptake inhibitors(SSRis), serotonin-norepinephrine reuptake inhibitors (SNRis), norepinephrine reuptake inhibitors (NRIs), triple reuptake inhibitors, modulators of CNS acetylcholine function, stimulants, anti-glucocorticoids, antagonists of NMDA-type glutamate receptors, tricylic antidepressants (TCAs), and a combination thereof. 26. The method of claim 24, wherein the other therapy is an anti-depressant.
1,600
594
13,929,252
1,633
The present disclosure relates generally to methods of treatment of tissue prior to implantation. In one aspect, the methods of treatment include washing adipose tissue with detergents to improve the viability of adipose cells for implantation and/or to increase the amount of viable adipose cells per volume of tissue for implantation.
1. A method for treating tissue comprising: selecting a tissue comprising adipocytes for implantation; contacting the tissue with a detergent at a concentration and time sufficient to remove non-viable materials from the tissue; and rinsing the tissue to remove the detergent. 2. The method according to claim 1, wherein the tissue comprises adipose tissue. 3. The method according to claim 1 or 2, wherein the method is performed prior to implantation of the tissue during autologous fat transfer. 4. The method according to claim 1, wherein the detergent is a nonionic detergent. 5. The method according to claim 1, wherein the detergent is a biocompatible detergent. 6. The method according to claim 1, wherein a poloxamer is not used in the contacting step. 7. The method according to claim 1, wherein the detergent comprises TWEEN. 8. The method according to claim 1, wherein the detergent comprises TRITON-X. 9. The method according to claim 1, wherein the non-viable materials comprise at least one of extracellular matrix proteins, blood, water, pharmaceutical agents, and epinephrine. 10. The method according to claim 1, wherein the tissue is contacted with a detergent at a time and temperature sufficient to reduce the water content of the tissue. 11. The method according to claim 1, further comprising agitating the tissue while the tissue is in contact with the detergent. 12. The method of claim 11, wherein agitating the tissue comprises at least one of stirring, shaking, and causing the detergent to flow in contact with the tissue. 13. The method according to claim 1, further comprising repeating the steps of contacting the tissue with the detergent and rinsing the tissue. 14. The method according to claim 1, wherein the washed tissue is rinsed with a rinsing fluid comprising a saline solution. 15. The method according to claim 1, further comprising implanting the tissue in a patient. 16. The method according to claim 1, further comprising removing water from the tissue. 17. A tissue composition, comprising an adipocyte-containing tissue prepared according to claim 1. 18. The tissue composition of claim 17, wherein the tissue comprises adipose tissue, and wherein the tissue is prepared such that the number of viable adipocytes per volume of tissue is greater than the number of viable adipocytes per volume of tissue in unprocessed human adipose tissue. 19. The tissue composition of claim 18, wherein the tissue is prepared such that the number of viable adipocytes per volume of tissue is at least two times the number of viable adipocytes per volume of tissue in unprocessed human adipose tissue.
The present disclosure relates generally to methods of treatment of tissue prior to implantation. In one aspect, the methods of treatment include washing adipose tissue with detergents to improve the viability of adipose cells for implantation and/or to increase the amount of viable adipose cells per volume of tissue for implantation.1. A method for treating tissue comprising: selecting a tissue comprising adipocytes for implantation; contacting the tissue with a detergent at a concentration and time sufficient to remove non-viable materials from the tissue; and rinsing the tissue to remove the detergent. 2. The method according to claim 1, wherein the tissue comprises adipose tissue. 3. The method according to claim 1 or 2, wherein the method is performed prior to implantation of the tissue during autologous fat transfer. 4. The method according to claim 1, wherein the detergent is a nonionic detergent. 5. The method according to claim 1, wherein the detergent is a biocompatible detergent. 6. The method according to claim 1, wherein a poloxamer is not used in the contacting step. 7. The method according to claim 1, wherein the detergent comprises TWEEN. 8. The method according to claim 1, wherein the detergent comprises TRITON-X. 9. The method according to claim 1, wherein the non-viable materials comprise at least one of extracellular matrix proteins, blood, water, pharmaceutical agents, and epinephrine. 10. The method according to claim 1, wherein the tissue is contacted with a detergent at a time and temperature sufficient to reduce the water content of the tissue. 11. The method according to claim 1, further comprising agitating the tissue while the tissue is in contact with the detergent. 12. The method of claim 11, wherein agitating the tissue comprises at least one of stirring, shaking, and causing the detergent to flow in contact with the tissue. 13. The method according to claim 1, further comprising repeating the steps of contacting the tissue with the detergent and rinsing the tissue. 14. The method according to claim 1, wherein the washed tissue is rinsed with a rinsing fluid comprising a saline solution. 15. The method according to claim 1, further comprising implanting the tissue in a patient. 16. The method according to claim 1, further comprising removing water from the tissue. 17. A tissue composition, comprising an adipocyte-containing tissue prepared according to claim 1. 18. The tissue composition of claim 17, wherein the tissue comprises adipose tissue, and wherein the tissue is prepared such that the number of viable adipocytes per volume of tissue is greater than the number of viable adipocytes per volume of tissue in unprocessed human adipose tissue. 19. The tissue composition of claim 18, wherein the tissue is prepared such that the number of viable adipocytes per volume of tissue is at least two times the number of viable adipocytes per volume of tissue in unprocessed human adipose tissue.
1,600
595
14,441,901
1,611
The present invention relates to an effervescent tablet, which upon dissolution in water provides a solution useful as a mouth wash or oral rinse for the prevention or treatment of inflammatory processes of the soft tissues of the mouth, throat and oral cavity.
1. An effervescent tablet comprising: a) a pharmaceutically effective amount of one or more phosphate salts; b) a pharmaceutically effective amount of a calcium salt; c) one or more effervescing organic acid component; and d) one or more effervescing base component; wherein said tablet upon dissolution in water provides a supersaturated solution of calcium and phosphate ions. 2. The effervescent tablet according to claim 1, wherein the phosphate salt is a sodium phosphate. 3. The effervescent tablet according to claim 2, wherein the tablet comprises a pharmaceutically effective amount of dibasic, sodium phosphate and monobasic, sodium phosphate. 4. The effervescent tablet according to claim 1, wherein the calcium salt is calcium chloride. 5. The effervescent tablet according to claim 1, wherein the effervescing organic acid component is selected from the group consisting of organic mono-, di- and tricarboxylic acids, and oxyacids, salts of organic acids and salts of inorganic acids, and combinations thereof. 6. The effervescent tablet according to claim 5, wherein the effervescing organic acid component is citric acid. 7. The effervescent tablet according to claim 1, wherein the one or more effervescing base components is a carbonate salt. 8. The effervescent tablet according to claim 7, wherein the one or more effervescing bases is selected from the group consisting of sodium carbonate, magnesium carbonate, sodium hydrogen carbonate, potassium carbonate, potassium hydrogen carbonate, calcium carbonate and magnesium carbonate. 9. The effervescent tablet according to claim 8, wherein composition comprises sodium carbonate and sodium hydrogen carbonate. 10. The effervescent tablet according to claim 1, wherein the tablet upon dissolution in water results in a solution of about pH 6.5. 11. The effervescent tablet according to claim 1, wherein the tablet upon dissolution results in a solution remaining clear for at least about 2 hr. 12. The effervescent tablet according to claim 1, wherein the tablet comprises one or more additional excipients. 13. The effervescent tablet according to claim 12, wherein the additional excipient is one or more carbohydrates selected from the group consisting of polyalcohols, dextrines and saccharides. 14. The effervescent tablet according to claim 12, wherein the additional excipient is one or more polyalcohol selected from the group consisting of sorbitol, mannitol, xylitol, and inositol or the mixture thereof. 15. The effervescent tablet according to claim 12, wherein the additional excipient is one or more saccharides selected from the group consisting of glucose, fructose and sucrose, or the mixture thereof. 16. The effervescent tablet according to claim 12, wherein the excipient is a flavour and/or sweetener. 17. The effervescent tablet according to claim 12, wherein the flavour and/or sweetener is selected from the group consisting of saccharin, aspartame, and acesulfame. 18. The effervescent tablet according to claim 1, wherein the amount of calcium per tablet is in the range of about 14-24 mg. 19. The effervescent tablet according to claim 1, wherein the amount of phosphate per tablet is in the range of about 28-36 mg. 20. The effervescent tablet according to claim 1, wherein the tablet comprises a molar ratio of NaH2PO4:Na2HPO4:CaCl2×2H2O of about 1:3:6. 21. The effervescent tablet according to claim 1, comprising: about 69.1 mg calcium chloride dihydrate; about 9.2 mg monobasic sodium phosphate; about 31.1 mg dibasic sodium phosphate; about 450.0 mg sodium hydrogen carbonate; about 50.0 mg sodium carbonate; about 238.5 mg citric acid; about 225.5 mg maltodextrine; and about 26.5 mg sorbitol. 22. The effervescent tablet according to claim 1, comprising: about 69.1 mg calcium chloride dihydrate; about 9.2 mg monobasic sodium phosphate; about 31.1 mg dibasic sodium phosphate; about 512.3 mg sodium hydrogen carbonate; about 60.6 mg sodium carbonate; about 272.7 mg citric acid; about 214.6 mg maltodextrine; and about 30.3 mg sorbitol. 23. The effervescent tablet according to claim 1, comprising: about 69.1 mg calcium chloride dihydrate; about 9.2 mg monobasic sodium phosphate; about 31.1 mg dibasic sodium phosphate; about 1690.5 mg sodium hydrogen carbonate; about 200.0 mg sodium carbonate; about 900.0 mg citric acid; and about 100.0 mg sorbitol. 24. A process for the preparation of effervescent tablets according to claim 1, comprising the steps of: a) mixing the one or more effervescing organic acid components and a dilutent tp provide a mixture; b) subjecting the mixture of step a) to granulation to provide a granulate; c) drying the granulate obtained in step b) to provide a dried granulate; d) mixing the dried granulate of step c) with the one or more effervescing base component, a pharmaceutically effective amount of one or more phosphate salt(s), a pharmaceutically effective amount of a calcium salt, and optionally one or more pharmaceutically acceptable excipients; and e) compressing the mixture of step d) to form tablets. 25. A process for preventing or treating inflammatory processes of the soft tissues of the mouth, throat and oral cavity, comprising the steps of diluting an effervescent tablet according to claim 1 in a suitable amount of water in order to dissolve the tablet, and administering the obtained solution to the patient as an oral rinse.
The present invention relates to an effervescent tablet, which upon dissolution in water provides a solution useful as a mouth wash or oral rinse for the prevention or treatment of inflammatory processes of the soft tissues of the mouth, throat and oral cavity.1. An effervescent tablet comprising: a) a pharmaceutically effective amount of one or more phosphate salts; b) a pharmaceutically effective amount of a calcium salt; c) one or more effervescing organic acid component; and d) one or more effervescing base component; wherein said tablet upon dissolution in water provides a supersaturated solution of calcium and phosphate ions. 2. The effervescent tablet according to claim 1, wherein the phosphate salt is a sodium phosphate. 3. The effervescent tablet according to claim 2, wherein the tablet comprises a pharmaceutically effective amount of dibasic, sodium phosphate and monobasic, sodium phosphate. 4. The effervescent tablet according to claim 1, wherein the calcium salt is calcium chloride. 5. The effervescent tablet according to claim 1, wherein the effervescing organic acid component is selected from the group consisting of organic mono-, di- and tricarboxylic acids, and oxyacids, salts of organic acids and salts of inorganic acids, and combinations thereof. 6. The effervescent tablet according to claim 5, wherein the effervescing organic acid component is citric acid. 7. The effervescent tablet according to claim 1, wherein the one or more effervescing base components is a carbonate salt. 8. The effervescent tablet according to claim 7, wherein the one or more effervescing bases is selected from the group consisting of sodium carbonate, magnesium carbonate, sodium hydrogen carbonate, potassium carbonate, potassium hydrogen carbonate, calcium carbonate and magnesium carbonate. 9. The effervescent tablet according to claim 8, wherein composition comprises sodium carbonate and sodium hydrogen carbonate. 10. The effervescent tablet according to claim 1, wherein the tablet upon dissolution in water results in a solution of about pH 6.5. 11. The effervescent tablet according to claim 1, wherein the tablet upon dissolution results in a solution remaining clear for at least about 2 hr. 12. The effervescent tablet according to claim 1, wherein the tablet comprises one or more additional excipients. 13. The effervescent tablet according to claim 12, wherein the additional excipient is one or more carbohydrates selected from the group consisting of polyalcohols, dextrines and saccharides. 14. The effervescent tablet according to claim 12, wherein the additional excipient is one or more polyalcohol selected from the group consisting of sorbitol, mannitol, xylitol, and inositol or the mixture thereof. 15. The effervescent tablet according to claim 12, wherein the additional excipient is one or more saccharides selected from the group consisting of glucose, fructose and sucrose, or the mixture thereof. 16. The effervescent tablet according to claim 12, wherein the excipient is a flavour and/or sweetener. 17. The effervescent tablet according to claim 12, wherein the flavour and/or sweetener is selected from the group consisting of saccharin, aspartame, and acesulfame. 18. The effervescent tablet according to claim 1, wherein the amount of calcium per tablet is in the range of about 14-24 mg. 19. The effervescent tablet according to claim 1, wherein the amount of phosphate per tablet is in the range of about 28-36 mg. 20. The effervescent tablet according to claim 1, wherein the tablet comprises a molar ratio of NaH2PO4:Na2HPO4:CaCl2×2H2O of about 1:3:6. 21. The effervescent tablet according to claim 1, comprising: about 69.1 mg calcium chloride dihydrate; about 9.2 mg monobasic sodium phosphate; about 31.1 mg dibasic sodium phosphate; about 450.0 mg sodium hydrogen carbonate; about 50.0 mg sodium carbonate; about 238.5 mg citric acid; about 225.5 mg maltodextrine; and about 26.5 mg sorbitol. 22. The effervescent tablet according to claim 1, comprising: about 69.1 mg calcium chloride dihydrate; about 9.2 mg monobasic sodium phosphate; about 31.1 mg dibasic sodium phosphate; about 512.3 mg sodium hydrogen carbonate; about 60.6 mg sodium carbonate; about 272.7 mg citric acid; about 214.6 mg maltodextrine; and about 30.3 mg sorbitol. 23. The effervescent tablet according to claim 1, comprising: about 69.1 mg calcium chloride dihydrate; about 9.2 mg monobasic sodium phosphate; about 31.1 mg dibasic sodium phosphate; about 1690.5 mg sodium hydrogen carbonate; about 200.0 mg sodium carbonate; about 900.0 mg citric acid; and about 100.0 mg sorbitol. 24. A process for the preparation of effervescent tablets according to claim 1, comprising the steps of: a) mixing the one or more effervescing organic acid components and a dilutent tp provide a mixture; b) subjecting the mixture of step a) to granulation to provide a granulate; c) drying the granulate obtained in step b) to provide a dried granulate; d) mixing the dried granulate of step c) with the one or more effervescing base component, a pharmaceutically effective amount of one or more phosphate salt(s), a pharmaceutically effective amount of a calcium salt, and optionally one or more pharmaceutically acceptable excipients; and e) compressing the mixture of step d) to form tablets. 25. A process for preventing or treating inflammatory processes of the soft tissues of the mouth, throat and oral cavity, comprising the steps of diluting an effervescent tablet according to claim 1 in a suitable amount of water in order to dissolve the tablet, and administering the obtained solution to the patient as an oral rinse.
1,600
596
15,700,446
1,613
Antimicrobial formulations and coatings for medical devices and processes therefor are disclosed. The formulations include at least one water permeable polymer with at least one antimicrobial agent in a liquid medium and are prepared by wet milling the components and can form antimicrobial coatings having uniformly dispersed particles having an average size of no greater than 50 microns.
1. A medical device comprising an antimicrobial coating, wherein the coating comprises at least one water permeable polymer and uniformly dispersed particles therein of at least one antimicrobial agent, and wherein the particles and any agglomerations of the antimicrobial agent have an average size of no greater than 50 microns. 2. The medical device of claim 1, wherein the average size is no greater than 20 microns. 3. The medical device of claim 2, wherein the average size is no greater than 5 microns. 4. The medical device of claim 1, wherein the at least one water permeable polymer encapsulates the at least one antimicrobial agent. 5. The medical device of claim 1, wherein the at least one water permeable polymer includes at least one of a polyurethane or a thermoplastic polyurethane elastomer. 6. The medical device of claim 1, wherein the at least one polymer includes at least one of a polyurethane, thermoplastic polyurethane elastomer, polyester, polylactic acid, polyglycolic acid, polytetramethylene glycol, polyacrylamide, polyacrylic acid, polyacrylate, poly(2-hydroxyethyl methacrylate), polyethylene-imine, poly-sulfonate and copolymers thereof. 7. The medical device of claim 1, wherein the at least one polymer has a weight average molecular weight of from about 70,000 to about 120,000 Daltons. 8. The medical device of claim 1, wherein the at least one antimicrobial agent comprises silver sulfadiazine, and wherein the coating has a release profile wherein at least 0.50 micrograms per centimeter (μg/cm) of silver is continuously released after 72 hours. 9. The medical device of claim 1, wherein the coating further comprises chlorhexidine diacetate, and at least 10 μg/cm of chlorhexidine diacetate is continuously released after 72 hours. 10. The medical device of claim 1, wherein the at least one antimicrobial agent comprises a silver-based salt and a polybiguanide salt. 11. The medical device of claim 10, wherein the silver-based salt is silver sulfadiazine, and the polybiguanide salt is chlorhexidine diacetate. 12. The medical device of claim 11, wherein the coating comprises from about 2 weight percent (wt. %) to about 10 wt. % of silver sulfadiazine, wherein the coating comprises at least 9 wt. % of chlorhexidine diacetate, and wherein the coating comprises from about 70 wt. % to about 90 wt. % of the water permeable polymer. 13. The medical device of claim 12, wherein the coating comprises from about 3.5 wt. % to about 7 wt. % silver sulfadiazine. 14. The medical device of claim 13, wherein the coating has a release profile wherein at least 0.50 μg/cm of silver is continuously released after 150 hours. 15. The medical device of claim 12, wherein the coating comprises at least 11 wt. % of chlorhexidine diacetate. 16. The medical device of claim 15, wherein the coating has a release profile wherein at least 10 μg/cm of chlorhexidine diacetate is continuously released after about 150 hours. 17. The medical device of claim 1, wherein the coating has a thickness of between about 20 microns to about 80 microns. 18. An antimicrobial formulation for coating a medical device prepared according to a process comprising: milling at least one water permeable polymer which is dissolved in a liquid medium with at least one antimicrobial agent which is insoluble in the liquid medium to form the antimicrobial formulation in which the at least one water permeable polymer encapsulates the at least one antimicrobial agent; and adding a primary C1-6 alcohol to the antimicrobial formulation, wherein the at least one antimicrobial agent includes a silver-based antimicrobial agent, and wherein the at least one water permeable polymer includes at least one of a polyurethane or a thermoplastic polyurethane elastomer. 19. The antimicrobial formulation of claim 18, wherein the at least one antimicrobial agent includes a combination of the silver-based antimicrobial agent and a polybiguanide or salt thereof. 20. The antimicrobial formulation of claim 18, wherein the at least one antimicrobial agent includes a combination of silver sulfadiazine and chlorhexidine diacetate.
Antimicrobial formulations and coatings for medical devices and processes therefor are disclosed. The formulations include at least one water permeable polymer with at least one antimicrobial agent in a liquid medium and are prepared by wet milling the components and can form antimicrobial coatings having uniformly dispersed particles having an average size of no greater than 50 microns.1. A medical device comprising an antimicrobial coating, wherein the coating comprises at least one water permeable polymer and uniformly dispersed particles therein of at least one antimicrobial agent, and wherein the particles and any agglomerations of the antimicrobial agent have an average size of no greater than 50 microns. 2. The medical device of claim 1, wherein the average size is no greater than 20 microns. 3. The medical device of claim 2, wherein the average size is no greater than 5 microns. 4. The medical device of claim 1, wherein the at least one water permeable polymer encapsulates the at least one antimicrobial agent. 5. The medical device of claim 1, wherein the at least one water permeable polymer includes at least one of a polyurethane or a thermoplastic polyurethane elastomer. 6. The medical device of claim 1, wherein the at least one polymer includes at least one of a polyurethane, thermoplastic polyurethane elastomer, polyester, polylactic acid, polyglycolic acid, polytetramethylene glycol, polyacrylamide, polyacrylic acid, polyacrylate, poly(2-hydroxyethyl methacrylate), polyethylene-imine, poly-sulfonate and copolymers thereof. 7. The medical device of claim 1, wherein the at least one polymer has a weight average molecular weight of from about 70,000 to about 120,000 Daltons. 8. The medical device of claim 1, wherein the at least one antimicrobial agent comprises silver sulfadiazine, and wherein the coating has a release profile wherein at least 0.50 micrograms per centimeter (μg/cm) of silver is continuously released after 72 hours. 9. The medical device of claim 1, wherein the coating further comprises chlorhexidine diacetate, and at least 10 μg/cm of chlorhexidine diacetate is continuously released after 72 hours. 10. The medical device of claim 1, wherein the at least one antimicrobial agent comprises a silver-based salt and a polybiguanide salt. 11. The medical device of claim 10, wherein the silver-based salt is silver sulfadiazine, and the polybiguanide salt is chlorhexidine diacetate. 12. The medical device of claim 11, wherein the coating comprises from about 2 weight percent (wt. %) to about 10 wt. % of silver sulfadiazine, wherein the coating comprises at least 9 wt. % of chlorhexidine diacetate, and wherein the coating comprises from about 70 wt. % to about 90 wt. % of the water permeable polymer. 13. The medical device of claim 12, wherein the coating comprises from about 3.5 wt. % to about 7 wt. % silver sulfadiazine. 14. The medical device of claim 13, wherein the coating has a release profile wherein at least 0.50 μg/cm of silver is continuously released after 150 hours. 15. The medical device of claim 12, wherein the coating comprises at least 11 wt. % of chlorhexidine diacetate. 16. The medical device of claim 15, wherein the coating has a release profile wherein at least 10 μg/cm of chlorhexidine diacetate is continuously released after about 150 hours. 17. The medical device of claim 1, wherein the coating has a thickness of between about 20 microns to about 80 microns. 18. An antimicrobial formulation for coating a medical device prepared according to a process comprising: milling at least one water permeable polymer which is dissolved in a liquid medium with at least one antimicrobial agent which is insoluble in the liquid medium to form the antimicrobial formulation in which the at least one water permeable polymer encapsulates the at least one antimicrobial agent; and adding a primary C1-6 alcohol to the antimicrobial formulation, wherein the at least one antimicrobial agent includes a silver-based antimicrobial agent, and wherein the at least one water permeable polymer includes at least one of a polyurethane or a thermoplastic polyurethane elastomer. 19. The antimicrobial formulation of claim 18, wherein the at least one antimicrobial agent includes a combination of the silver-based antimicrobial agent and a polybiguanide or salt thereof. 20. The antimicrobial formulation of claim 18, wherein the at least one antimicrobial agent includes a combination of silver sulfadiazine and chlorhexidine diacetate.
1,600
597
14,725,714
1,619
According to the subject invention, there is disclosed, a dosage and packaging configuration which includes the use of color-coded pre-filled syringes and vials to fill and refill infusion systems with existing and new dosage forms of intrathecal baclofen.
1. A drug delivery system comprising a labeled pre-filled syringe containing a sterile aqueous solution of baclofen that is suitable for intrathecal delivery, wherein the sterile aqueous solution of baclofen was sterilized by heating at 121° C. for a F0 value of 7 minutes. 2. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen contains no more than 0.5% of 4-(4-chlorophenyl)-2-pyrrolidone by weight of baclofen. 3. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen further comprises sodium chloride. 4. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen is substantially free of any particulates. 5. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen has a baclofen concentration from about 0.5 mg/mL to about 4.0 mg/mL. 6. The drug delivery system of claim 5, wherein the labeled pre-filled syringe contains from 5 mL to 40 mL of the sterile aqueous solution of baclofen. 7. The drug delivery system of claim 5, further comprising a color-coding system indicative of the concentration of baclofen in the labeled pre-filled syringe, size of the pre-filled syringe, or both. 8. The drug delivery system of claim 7, further comprising a plunger, wherein the color-coding system is on the plunger, on a label of the labeled pre-filled syringe, or both. 9. The drug delivery system of claim 1, wherein the labeled pre-filled syringe further comprises a leur-lock tip. 10. A drug delivery system comprising a labeled pre-filled syringe containing a sterile aqueous solution of baclofen that is suitable for intrathecal delivery, wherein the sterile aqueous solution of baclofen contains no more than 0.5% of 4-(4-chlorophenyl)-2-pyrrolidone by weight of baclofen. 11. The drug delivery system of claim 10, wherein the sterile aqueous solution of baclofen further comprises sodium chloride. 12. The drug delivery system of claim 10, wherein the sterile aqueous solution of baclofen is substantially free of any particulates. 13. The drug delivery system of claim 10, wherein the sterile aqueous solution of baclofen has a baclofen concentration from about 0.5 mg/mL to about 4.0 mg/mL. 14. The drug delivery system of claim 13, wherein the labeled pre-filled syringe contains from 5 mL to 40 mL of the sterile aqueous solution of baclofen. 15. The drug delivery system of claim 13, further comprising a color-coding system indicative of the concentration of baclofen in the labeled pre-filled syringe, size of the pre-filled syringe, or both. 16. The drug delivery system of claim 15, further comprising a plunger, wherein the color-coding system is on the plunger, on a label of the labeled pre-filled syringe, or both. 17. The drug delivery system of claim 10, wherein the labeled pre-filled syringe further comprises a leur-lock tip.
According to the subject invention, there is disclosed, a dosage and packaging configuration which includes the use of color-coded pre-filled syringes and vials to fill and refill infusion systems with existing and new dosage forms of intrathecal baclofen.1. A drug delivery system comprising a labeled pre-filled syringe containing a sterile aqueous solution of baclofen that is suitable for intrathecal delivery, wherein the sterile aqueous solution of baclofen was sterilized by heating at 121° C. for a F0 value of 7 minutes. 2. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen contains no more than 0.5% of 4-(4-chlorophenyl)-2-pyrrolidone by weight of baclofen. 3. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen further comprises sodium chloride. 4. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen is substantially free of any particulates. 5. The drug delivery system of claim 1, wherein the sterile aqueous solution of baclofen has a baclofen concentration from about 0.5 mg/mL to about 4.0 mg/mL. 6. The drug delivery system of claim 5, wherein the labeled pre-filled syringe contains from 5 mL to 40 mL of the sterile aqueous solution of baclofen. 7. The drug delivery system of claim 5, further comprising a color-coding system indicative of the concentration of baclofen in the labeled pre-filled syringe, size of the pre-filled syringe, or both. 8. The drug delivery system of claim 7, further comprising a plunger, wherein the color-coding system is on the plunger, on a label of the labeled pre-filled syringe, or both. 9. The drug delivery system of claim 1, wherein the labeled pre-filled syringe further comprises a leur-lock tip. 10. A drug delivery system comprising a labeled pre-filled syringe containing a sterile aqueous solution of baclofen that is suitable for intrathecal delivery, wherein the sterile aqueous solution of baclofen contains no more than 0.5% of 4-(4-chlorophenyl)-2-pyrrolidone by weight of baclofen. 11. The drug delivery system of claim 10, wherein the sterile aqueous solution of baclofen further comprises sodium chloride. 12. The drug delivery system of claim 10, wherein the sterile aqueous solution of baclofen is substantially free of any particulates. 13. The drug delivery system of claim 10, wherein the sterile aqueous solution of baclofen has a baclofen concentration from about 0.5 mg/mL to about 4.0 mg/mL. 14. The drug delivery system of claim 13, wherein the labeled pre-filled syringe contains from 5 mL to 40 mL of the sterile aqueous solution of baclofen. 15. The drug delivery system of claim 13, further comprising a color-coding system indicative of the concentration of baclofen in the labeled pre-filled syringe, size of the pre-filled syringe, or both. 16. The drug delivery system of claim 15, further comprising a plunger, wherein the color-coding system is on the plunger, on a label of the labeled pre-filled syringe, or both. 17. The drug delivery system of claim 10, wherein the labeled pre-filled syringe further comprises a leur-lock tip.
1,600
598
14,233,546
1,631
The present application mainly relates to specific methods for inferring activity of one or more cellular signaling pathway(s) in tissue of a medical subject based at least on the expression level(s) of one or more target gene(s) of the cellular signaling pathway(s) measured in an extracted sample of the tissue of the medical subject, an apparatus comprising a digital compressor configured to perform such methods and a non-transitory storage medium storing instructions that are executable by a digital processing device to perform such methods.
1. A method comprising: inferring activity of one or more cellular signaling pathway(s) in tissue of a medical subject based at least on the expression level(s) (20) of one or more target gene(s) of the cellular signaling pathway(s) measured in an extracted sample of the tissue of the medical subject, wherein the inferring comprises: inferring activity of the cellular signaling pathway(s) in the tissue of the medical subject by evaluating at least a portion of a probabilistic model (40-1, . . . , 40-7), preferably a Bayesian network (40-1, . . . , 40-7), representing the cellular signaling pathway(s) for a set of inputs including at least the expression level(s) (20) of the one or more target genes of the cellular signaling pathway(s) measured in the extracted sample of the tissue of the medical subject; estimating a level (46) in the tissue of the medical subject of at least one transcription factor (TF) element, the at least one TF element controlling transcription of the one or more target gene(s) of the cellular signaling pathway(s), the estimating being based at least in part on conditional probabilities relating the at least one TF element and the expression level(s) (20) of the one or more target gene(s) of the cellular signaling pathway(s) measured in the extracted sample of the tissue of the medical subject; inferring activity of the cellular signaling pathway(s) based on the estimated level in the tissue sample of the transcription factor; and determining whether the cellular signaling pathway(s) is/are operating abnormally in the tissue of the medical subject based on the inferred activity of the cellular signaling pathway(s) in the tissue of the medical subject; wherein the inferring is performed by a digital processing device (12) using the probabilistic model (40-1, . . . , 40-7) of the cellular signaling pathway(s). 2. A method according to claim 1, wherein the inferring comprises: estimating a level (46) in the tissue of the medical subject of at least one transcription factor (TF) element represented by a TF node of the probabilistic model, the TF element controlling transcription of the one or more target gene(s) of the cellular signaling pathway(s), the estimating being based at least in part on conditional probabilities of the probabilistic model (40-1, . . . , 40-7) relating the TF node and nodes in the probabilistic model representing the one or more target gene(s) of the cellular signaling pathway(s) measured in the extracted sample of the tissue of the medical subject, and wherein the inferring is preferably performed by using a Bayesian network (40-1, . . . , 40-7) comprising nodes representing information about the signaling pathway(s) and conditional probability relationships between connected nodes of the Bayesian network. 3. The method of claim 1, wherein the cellular signaling pathway(s) comprise(s) a Wnt pathway, an ER pathway, an AR pathway and/or a Hedgehog pathway. 4. The method of claim 3, wherein the inferring comprises: inferring activity of a Wnt pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the Wnt pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6 and FZD7. 5. The method of claim 3, wherein the inferring (also) comprises: inferring activity of an ER pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the ER pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: CDH26, SGK3, PGR, GREB1, CA12, XBP1, CELSR2, WISP2, DSCAM, ERBB2, CTSD, TFF1 and NRIP1. 6. The method of claim 3, wherein the inferring (also) comprises: inferring activity of a Hedgehog pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the Hedgehog pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN and CTSL1. 7. The method of claim 3, wherein the inferring (also) comprises: inferring activity of an AR pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the AR pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: KLK2, PMEPA1, TMPRSS2, NKX3—1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR and EAF2. 8. The method of claim 4, wherein the inferring is further based on expression levels (20) of at least one target gene of the Wnt pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A and LECT2. 9. The method of claim 5, wherein the inferring is further based on expression levels (20) of at least one target gene of the ER pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: AP1B1, ATP5J, COL18A1, COX7A2L, EBAG9, ESR1, HSPB1, IGFBP4, KRT19, MYC, NDUFV3, PISD, PREDM15, PTMA, RARA, SOD1 and TRIM25. 10. The method of claim 6, wherein the inferring is further based on expression levels (20) of at least one target gene of the Hedgehog pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: BCL2, FOXA2, FOXF1, H19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1 and TOM1. 11. The method of claim 7, wherein the inferring is further based on expression levels (20) of at least one target gene of the AR pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1 and TACC2. 12. (canceled) 13. (canceled) 14. The method of claim 1 comprising: inferring activity of a Wnt pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the Wnt pathway measured in an extracted sample of the tissue of the medical subject and/or inferring activity of an ER pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the ER pathway measured in an extracted sample of the tissue of the medical subject and/or inferring activity of a Hedgehog pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the Hedgehog pathway measured in an extracted sample of the tissue of the medical subject, and/or inferring activity of an AR pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the AR pathway measured in an extracted sample of the tissue of the medical subject. 15. The method of claim 14, wherein the set of target genes of the Wnt pathway includes at least nine, preferably all target genes selected from the group comprising: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6 and FZD7, and/or the set of target genes of the ER pathway includes at least nine, preferably all target genes selected from the group comprising: CDH26, SGK3, PGR, GREB1, CA12, XBP1, CELSR2, WISP2, DSCAM, ERBB2, CTSD, TFF1 and NRIP1, and/or the set of target genes of the Hedgehog pathway includes at least nine, preferably all target genes selected from the group comprising: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN and CTSL1, and/or the set of target genes of the AR pathway includes at least nine, preferably all target genes selected from the group comprising: KLK2, PMEPA1, TMPRSS2, NKX3—1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR and EAF2. 16. The method of claim 15, wherein the set of target genes of the Wnt pathway further includes at least one target gene selected from the group comprising: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A, and LECT2, and/or the set of target genes of the ER pathway further includes at least one target gene selected from the group comprising: AP1B1, ATP5J, COL18A1, COX7A2L, EBAG9, ESR1, HSPB1, IGFBP4, KRT19, MYC, NDUFV3, PISD, PREDM15, PTMA, RARA, SOD1 and TRIM25, and/or the set of target genes of the Hedgehog pathway further includes at least one target gene selected from the group comprising: BCL2, FOXA2, FOXF1, G19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1 and TOM1, and/or the set of target genes of the AR pathway further includes at least one target gene selected from the group comprising: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1 and TACC2. 17. An apparatus comprising a digital processor (12) configured to perform a method as set forth in claim 1. 18. A non-transitory storage medium storing instructions that are executable by a digital processing device (12) to perform a method as set forth in claim 1. 19. A computer program comprising program code means for causing a digital processing device (12) to perform a method as set forth in claim 1. 20. (canceled)
The present application mainly relates to specific methods for inferring activity of one or more cellular signaling pathway(s) in tissue of a medical subject based at least on the expression level(s) of one or more target gene(s) of the cellular signaling pathway(s) measured in an extracted sample of the tissue of the medical subject, an apparatus comprising a digital compressor configured to perform such methods and a non-transitory storage medium storing instructions that are executable by a digital processing device to perform such methods.1. A method comprising: inferring activity of one or more cellular signaling pathway(s) in tissue of a medical subject based at least on the expression level(s) (20) of one or more target gene(s) of the cellular signaling pathway(s) measured in an extracted sample of the tissue of the medical subject, wherein the inferring comprises: inferring activity of the cellular signaling pathway(s) in the tissue of the medical subject by evaluating at least a portion of a probabilistic model (40-1, . . . , 40-7), preferably a Bayesian network (40-1, . . . , 40-7), representing the cellular signaling pathway(s) for a set of inputs including at least the expression level(s) (20) of the one or more target genes of the cellular signaling pathway(s) measured in the extracted sample of the tissue of the medical subject; estimating a level (46) in the tissue of the medical subject of at least one transcription factor (TF) element, the at least one TF element controlling transcription of the one or more target gene(s) of the cellular signaling pathway(s), the estimating being based at least in part on conditional probabilities relating the at least one TF element and the expression level(s) (20) of the one or more target gene(s) of the cellular signaling pathway(s) measured in the extracted sample of the tissue of the medical subject; inferring activity of the cellular signaling pathway(s) based on the estimated level in the tissue sample of the transcription factor; and determining whether the cellular signaling pathway(s) is/are operating abnormally in the tissue of the medical subject based on the inferred activity of the cellular signaling pathway(s) in the tissue of the medical subject; wherein the inferring is performed by a digital processing device (12) using the probabilistic model (40-1, . . . , 40-7) of the cellular signaling pathway(s). 2. A method according to claim 1, wherein the inferring comprises: estimating a level (46) in the tissue of the medical subject of at least one transcription factor (TF) element represented by a TF node of the probabilistic model, the TF element controlling transcription of the one or more target gene(s) of the cellular signaling pathway(s), the estimating being based at least in part on conditional probabilities of the probabilistic model (40-1, . . . , 40-7) relating the TF node and nodes in the probabilistic model representing the one or more target gene(s) of the cellular signaling pathway(s) measured in the extracted sample of the tissue of the medical subject, and wherein the inferring is preferably performed by using a Bayesian network (40-1, . . . , 40-7) comprising nodes representing information about the signaling pathway(s) and conditional probability relationships between connected nodes of the Bayesian network. 3. The method of claim 1, wherein the cellular signaling pathway(s) comprise(s) a Wnt pathway, an ER pathway, an AR pathway and/or a Hedgehog pathway. 4. The method of claim 3, wherein the inferring comprises: inferring activity of a Wnt pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the Wnt pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6 and FZD7. 5. The method of claim 3, wherein the inferring (also) comprises: inferring activity of an ER pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the ER pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: CDH26, SGK3, PGR, GREB1, CA12, XBP1, CELSR2, WISP2, DSCAM, ERBB2, CTSD, TFF1 and NRIP1. 6. The method of claim 3, wherein the inferring (also) comprises: inferring activity of a Hedgehog pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the Hedgehog pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN and CTSL1. 7. The method of claim 3, wherein the inferring (also) comprises: inferring activity of an AR pathway in the tissue of the medical subject based at least on expression levels (20) of one or more, preferably at least three, target gene(s) of the AR pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: KLK2, PMEPA1, TMPRSS2, NKX3—1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR and EAF2. 8. The method of claim 4, wherein the inferring is further based on expression levels (20) of at least one target gene of the Wnt pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A and LECT2. 9. The method of claim 5, wherein the inferring is further based on expression levels (20) of at least one target gene of the ER pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: AP1B1, ATP5J, COL18A1, COX7A2L, EBAG9, ESR1, HSPB1, IGFBP4, KRT19, MYC, NDUFV3, PISD, PREDM15, PTMA, RARA, SOD1 and TRIM25. 10. The method of claim 6, wherein the inferring is further based on expression levels (20) of at least one target gene of the Hedgehog pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: BCL2, FOXA2, FOXF1, H19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1 and TOM1. 11. The method of claim 7, wherein the inferring is further based on expression levels (20) of at least one target gene of the AR pathway measured in the extracted sample of the tissue of the medical subject selected from the group comprising: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1 and TACC2. 12. (canceled) 13. (canceled) 14. The method of claim 1 comprising: inferring activity of a Wnt pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the Wnt pathway measured in an extracted sample of the tissue of the medical subject and/or inferring activity of an ER pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the ER pathway measured in an extracted sample of the tissue of the medical subject and/or inferring activity of a Hedgehog pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the Hedgehog pathway measured in an extracted sample of the tissue of the medical subject, and/or inferring activity of an AR pathway in tissue of a medical subject based at least on expression levels (20) of two, three or more target genes of a set of target genes of the AR pathway measured in an extracted sample of the tissue of the medical subject. 15. The method of claim 14, wherein the set of target genes of the Wnt pathway includes at least nine, preferably all target genes selected from the group comprising: KIAA1199, AXIN2, RNF43, TBX3, TDGF1, SOX9, ASCL2, IL8, SP5, ZNRF3, KLF6, CCND1, DEFA6 and FZD7, and/or the set of target genes of the ER pathway includes at least nine, preferably all target genes selected from the group comprising: CDH26, SGK3, PGR, GREB1, CA12, XBP1, CELSR2, WISP2, DSCAM, ERBB2, CTSD, TFF1 and NRIP1, and/or the set of target genes of the Hedgehog pathway includes at least nine, preferably all target genes selected from the group comprising: GLI1, PTCH1, PTCH2, IGFBP6, SPP1, CCND2, FST, FOXL1, CFLAR, TSC22D1, RAB34, S100A9, S100A7, MYCN, FOXM1, GLI3, TCEA2, FYN and CTSL1, and/or the set of target genes of the AR pathway includes at least nine, preferably all target genes selected from the group comprising: KLK2, PMEPA1, TMPRSS2, NKX3—1, ABCC4, KLK3, FKBP5, ELL2, UGT2B15, DHCR24, PPAP2A, NDRG1, LRIG1, CREB3L4, LCP1, GUCY1A3, AR and EAF2. 16. The method of claim 15, wherein the set of target genes of the Wnt pathway further includes at least one target gene selected from the group comprising: NKD1, OAT, FAT1, LEF1, GLUL, REG1B, TCF7L2, COL18A1, BMP7, SLC1A2, ADRA2C, PPARG, DKK1, HNF1A, and LECT2, and/or the set of target genes of the ER pathway further includes at least one target gene selected from the group comprising: AP1B1, ATP5J, COL18A1, COX7A2L, EBAG9, ESR1, HSPB1, IGFBP4, KRT19, MYC, NDUFV3, PISD, PREDM15, PTMA, RARA, SOD1 and TRIM25, and/or the set of target genes of the Hedgehog pathway further includes at least one target gene selected from the group comprising: BCL2, FOXA2, FOXF1, G19, HHIP, IL1R2, JAG2, JUP, MIF, MYLK, NKX2.2, NKX2.8, PITRM1 and TOM1, and/or the set of target genes of the AR pathway further includes at least one target gene selected from the group comprising: APP, NTS, PLAU, CDKN1A, DRG1, FGF8, IGF1, PRKACB, PTPN1, SGK1 and TACC2. 17. An apparatus comprising a digital processor (12) configured to perform a method as set forth in claim 1. 18. A non-transitory storage medium storing instructions that are executable by a digital processing device (12) to perform a method as set forth in claim 1. 19. A computer program comprising program code means for causing a digital processing device (12) to perform a method as set forth in claim 1. 20. (canceled)
1,600
599
14,150,488
1,611
A method for sterilizing an antiseptic solution includes providing a container containing the antiseptic solution, the antiseptic solution having an initial purity, selecting a sterilization temperature from about 85° C. to about 135° C. and an sterilization time from about 1 minute to about 19 hours, heating the antiseptic solution to the selected sterilization temperature, maintaining the temperature for the selected sterilization time, and terminating the heating of the antiseptic solution when the sterilization time expires. After terminating the heating, the antiseptic solution has a post-sterilization purity. The sterilization temperature and the sterilization time are selected such that after terminating the heating, the antiseptic solution is sterile and has a post-sterilization purity of at least about 92% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 5%.
1. The sterile antiseptic solution of claim 23, prepared by a method comprising the steps of: providing a container containing a pre-sterilized antiseptic solution, the pre-sterilized antiseptic solution having an initial purity; selecting a sterilization temperature from about 85° C. to about 135° C. and a sterilization time from about 1 minute to about 19 hours; heating the pre-sterilized antiseptic solution to the selected sterilization temperature; maintaining the pre-sterilized antiseptic solution at the sterilization temperature for the selected sterilization time; and terminating the heating of the pre-sterilized antiseptic solution when the selected sterilization time expires, thereby providing the sterile antiseptic solution, wherein after terminating the heating, the sterile antiseptic solution has a post-sterilization purity, and wherein the sterilization temperature and the sterilization time are selected such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 92% and a percentage point change in purity from the initial purity to the post-sterilization purity of at most about 5%. 2. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 85° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.38·ln x+156.9 for 21.5≦x≦1123; or b) 125° C.≦y≦135° C., x≧1 and y≦−10.38·ln x+156.9 for 9.1≦x≦21.5, wherein y is the sterilization temperature and x is the sterilization time in minutes. 3. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 85° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.37·ln x+154.6 for 17.5≦x≦900; or b) 125° C.≦y≦135° C., x≧1 and y≦−10.37·ln x+154.6 for 7.3≦x≦17.5, wherein y is the sterilization temperature and x is the sterilization time in minutes. 4. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 85° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.4·ln x+151.7 for 13≦x≦670; or b) 125° C.≦y≦135° C., x≧1 and y≦−10.4·ln x+151.7 for 5.5≦x≦13, wherein y is the sterilization temperature and x is the sterilization time in minutes. 5. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 90° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.6·ln x+148.3 for 9≦x≦260, or b) 125° C.≦y≦135° C., x≧1, and y≦−10.6·ln x+148.3 for 3.7≦x≦9 wherein y is the sterilization temperature and x is the sterilization time in minutes. 6. The sterile antiseptic solution of claim 1, wherein the selected sterilization temperature and the selected sterilization time are chosen such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 94% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 4%. 7. The sterile antiseptic solution of claim 1, wherein the selected sterilization temperature and the selected sterilization time are chosen such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 96% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 3%. 8. The sterile antiseptic solution of claim 1, wherein the selected sterilization temperature and the selected sterilization time are chosen such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 98% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 2%. 9. The sterile antiseptic solution of claim 23, wherein the sterile antiseptic solution is completely free of microbes. 10. The sterile antiseptic solution of claim 23, wherein the antimicrobial molecule is selected from the group consisting of chlorhexidine gluconate and octenidine dihydrochloride; and wherein the solvent is selected from the group consisting of ethanol, isopropanol, and n-propanol. 11. The sterile antiseptic solution of claim 23, comprising from about 40% to about 90% v/v of the solvent and from about 0.1% to about 2.5% w/v of the antimicrobial molecule, wherein the solvent comprises isopropanol and the antimicrobial molecule comprises chlorhexidine gluconate. 12. The sterile antiseptic solution of claim 23, comprising about 70% v/v of the solvent and about 2.0% w/v of the antimicrobial molecule, wherein the solvent comprises isopropanol and the antimicrobial molecule comprises chlorhexidine gluconate. 13. The sterile antiseptic solution of claim 1, wherein the sterilization time is from about 6 minutes to about one hour. 14. The sterile antiseptic solution of claim 1, wherein the sterilization temperature is about 95° C. and the sterilization time is from about 1.5 hours to about 6.5 hours. 15. The sterile antiseptic solution of claim 1, wherein the sterilization temperature is about 110° C. and the sterilization time is from about 6 minutes to about 90 minutes. 16. The sterile antiseptic solution of claim 1, wherein the sterilization temperature is about 120° C. and the sterilization time is from about 2 minutes to about 35 minutes. 17. The sterile antiseptic solution of claim 1, wherein the container comprises glass or plastic. 18. The sterile antiseptic solution of claim 1, wherein the method further comprises the steps of: providing a cascading water sterilizer that produces a constant, cascading waterfall, wherein heating the antiseptic solution to the selected sterilization temperature and maintaining the selected sterilization temperature comprises contacting the container with the cascading waterfall for the duration of the sterilization time. 19. The sterile antiseptic solution of claim 1, wherein the method further comprises a step of cooling the sterile antiseptic solution after terminating the heating. 20. The sterile antiseptic solution of claim 1, wherein prior to heating the pre-sterilized antiseptic solution to the selected sterilization temperature, the container and the pre-sterilized antiseptic solution have a temperature from about 20° C. to about 35° C. 21. The sterile antiseptic solution of claim 1, wherein the container is sealed prior to heating the pre-sterlized antiseptic solution to the selected sterilization temperature. 22. The sterile antiseptic solution of claim 10, further comprising water. 23. A sterile antiseptic solution comprising: an antimicrobial molecule selected from the group consisting of a bis-(dihydropyridinyl)-decane derivative and a biguanide; and a solvent, wherein the sterile antiseptic solution is sterile and at least 92% pure. 24. The sterile antiseptic solution of claim 23, wherein the antiseptic solution is at least 94% pure. 25. The sterile antiseptic solution of claim 23, wherein the antiseptic solution is at least 96% pure. 26. The sterile antiseptic solution of claim 23, wherein the antiseptic solution is at least 98% pure. 27. The sterile antiseptic solution of claim 23, comprising from about 40% to about 90% v/v of the solvent, from about 0.1% to about 2.5% w/v of the antimicrobial molecule, and from about 10% to about 60% v/v water, wherein the solvent comprises isopropanol and the antimicrobial molecule comprises chlorhexidine gluconate. 28. The sterile antiseptic solution of claim 23, wherein the sterile antiseptic solution maintains 7-day sterility in accordance with U.S. Pharmacopeia Convention (USP) 36, Chapter 55, Biological Indicators—Resistance Performance Tests.
A method for sterilizing an antiseptic solution includes providing a container containing the antiseptic solution, the antiseptic solution having an initial purity, selecting a sterilization temperature from about 85° C. to about 135° C. and an sterilization time from about 1 minute to about 19 hours, heating the antiseptic solution to the selected sterilization temperature, maintaining the temperature for the selected sterilization time, and terminating the heating of the antiseptic solution when the sterilization time expires. After terminating the heating, the antiseptic solution has a post-sterilization purity. The sterilization temperature and the sterilization time are selected such that after terminating the heating, the antiseptic solution is sterile and has a post-sterilization purity of at least about 92% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 5%.1. The sterile antiseptic solution of claim 23, prepared by a method comprising the steps of: providing a container containing a pre-sterilized antiseptic solution, the pre-sterilized antiseptic solution having an initial purity; selecting a sterilization temperature from about 85° C. to about 135° C. and a sterilization time from about 1 minute to about 19 hours; heating the pre-sterilized antiseptic solution to the selected sterilization temperature; maintaining the pre-sterilized antiseptic solution at the sterilization temperature for the selected sterilization time; and terminating the heating of the pre-sterilized antiseptic solution when the selected sterilization time expires, thereby providing the sterile antiseptic solution, wherein after terminating the heating, the sterile antiseptic solution has a post-sterilization purity, and wherein the sterilization temperature and the sterilization time are selected such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 92% and a percentage point change in purity from the initial purity to the post-sterilization purity of at most about 5%. 2. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 85° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.38·ln x+156.9 for 21.5≦x≦1123; or b) 125° C.≦y≦135° C., x≧1 and y≦−10.38·ln x+156.9 for 9.1≦x≦21.5, wherein y is the sterilization temperature and x is the sterilization time in minutes. 3. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 85° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.37·ln x+154.6 for 17.5≦x≦900; or b) 125° C.≦y≦135° C., x≧1 and y≦−10.37·ln x+154.6 for 7.3≦x≦17.5, wherein y is the sterilization temperature and x is the sterilization time in minutes. 4. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 85° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.4·ln x+151.7 for 13≦x≦670; or b) 125° C.≦y≦135° C., x≧1 and y≦−10.4·ln x+151.7 for 5.5≦x≦13, wherein y is the sterilization temperature and x is the sterilization time in minutes. 5. The sterile antiseptic solution of claim 1, wherein the selected sterilization time and selected sterilization temperature satisfy the following relationship: a) 90° C.≦y<125° C., y≧−6.14·ln x+123.2 for 1≦x≦552, and y≦−10.6·ln x+148.3 for 9≦x≦260, or b) 125° C.≦y≦135° C., x≧1, and y≦−10.6·ln x+148.3 for 3.7≦x≦9 wherein y is the sterilization temperature and x is the sterilization time in minutes. 6. The sterile antiseptic solution of claim 1, wherein the selected sterilization temperature and the selected sterilization time are chosen such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 94% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 4%. 7. The sterile antiseptic solution of claim 1, wherein the selected sterilization temperature and the selected sterilization time are chosen such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 96% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 3%. 8. The sterile antiseptic solution of claim 1, wherein the selected sterilization temperature and the selected sterilization time are chosen such that after terminating the heating, the sterile antiseptic solution has a post-sterilization purity of at least about 98% and the percentage point change in purity from the initial purity to the post-sterilization purity is at most about 2%. 9. The sterile antiseptic solution of claim 23, wherein the sterile antiseptic solution is completely free of microbes. 10. The sterile antiseptic solution of claim 23, wherein the antimicrobial molecule is selected from the group consisting of chlorhexidine gluconate and octenidine dihydrochloride; and wherein the solvent is selected from the group consisting of ethanol, isopropanol, and n-propanol. 11. The sterile antiseptic solution of claim 23, comprising from about 40% to about 90% v/v of the solvent and from about 0.1% to about 2.5% w/v of the antimicrobial molecule, wherein the solvent comprises isopropanol and the antimicrobial molecule comprises chlorhexidine gluconate. 12. The sterile antiseptic solution of claim 23, comprising about 70% v/v of the solvent and about 2.0% w/v of the antimicrobial molecule, wherein the solvent comprises isopropanol and the antimicrobial molecule comprises chlorhexidine gluconate. 13. The sterile antiseptic solution of claim 1, wherein the sterilization time is from about 6 minutes to about one hour. 14. The sterile antiseptic solution of claim 1, wherein the sterilization temperature is about 95° C. and the sterilization time is from about 1.5 hours to about 6.5 hours. 15. The sterile antiseptic solution of claim 1, wherein the sterilization temperature is about 110° C. and the sterilization time is from about 6 minutes to about 90 minutes. 16. The sterile antiseptic solution of claim 1, wherein the sterilization temperature is about 120° C. and the sterilization time is from about 2 minutes to about 35 minutes. 17. The sterile antiseptic solution of claim 1, wherein the container comprises glass or plastic. 18. The sterile antiseptic solution of claim 1, wherein the method further comprises the steps of: providing a cascading water sterilizer that produces a constant, cascading waterfall, wherein heating the antiseptic solution to the selected sterilization temperature and maintaining the selected sterilization temperature comprises contacting the container with the cascading waterfall for the duration of the sterilization time. 19. The sterile antiseptic solution of claim 1, wherein the method further comprises a step of cooling the sterile antiseptic solution after terminating the heating. 20. The sterile antiseptic solution of claim 1, wherein prior to heating the pre-sterilized antiseptic solution to the selected sterilization temperature, the container and the pre-sterilized antiseptic solution have a temperature from about 20° C. to about 35° C. 21. The sterile antiseptic solution of claim 1, wherein the container is sealed prior to heating the pre-sterlized antiseptic solution to the selected sterilization temperature. 22. The sterile antiseptic solution of claim 10, further comprising water. 23. A sterile antiseptic solution comprising: an antimicrobial molecule selected from the group consisting of a bis-(dihydropyridinyl)-decane derivative and a biguanide; and a solvent, wherein the sterile antiseptic solution is sterile and at least 92% pure. 24. The sterile antiseptic solution of claim 23, wherein the antiseptic solution is at least 94% pure. 25. The sterile antiseptic solution of claim 23, wherein the antiseptic solution is at least 96% pure. 26. The sterile antiseptic solution of claim 23, wherein the antiseptic solution is at least 98% pure. 27. The sterile antiseptic solution of claim 23, comprising from about 40% to about 90% v/v of the solvent, from about 0.1% to about 2.5% w/v of the antimicrobial molecule, and from about 10% to about 60% v/v water, wherein the solvent comprises isopropanol and the antimicrobial molecule comprises chlorhexidine gluconate. 28. The sterile antiseptic solution of claim 23, wherein the sterile antiseptic solution maintains 7-day sterility in accordance with U.S. Pharmacopeia Convention (USP) 36, Chapter 55, Biological Indicators—Resistance Performance Tests.
1,600