![]() USES OF DA-DKP, COMPOSITION AND METHOD FOR THE SUPPLY OF STEM CELLS TO UMINDIVIDUAL
专利摘要:
Patent Specification: "Trunk cell mobilization, migration, expansion and differentiation compositions and methods of using them". The present invention relates to compositions that enhance stem cell mobilization, migration, expansion and / or differentiation and methods of using them for treating mammals. 25745783v1 1/1 25745783v1 公开号:BR112015020469A2 申请号:R112015020469 申请日:2014-03-17 公开日:2020-01-28 发明作者:Bar- Or David;Thomas Greg 申请人:Ampio Pharmaceuticals Inc; IPC主号:
专利说明:
Invention Patent Descriptive Report for USES OF DA-DKP, COMPOSITION AND METHOD FOR SUPPLYING STEM CELLS TO AN INDIVIDUAL. CROSS REFERENCE TO RELATED APPLICATIONS [001] This application claims priority benefit under 35 U.S.C. § 119 (e) United States Provisional Patent Application Serial No. 61 / 791,623, filed on March 15, 2013; United States Provisional Patent Application Serial No. 61 / 832,713, filed on June 7, 2013; and United States Provisional Patent Application Serial No. 61 / 897,449, filed on October 30, 2013; the description of each of which is incorporated by reference. TECHNICAL FIELD [002] The present invention relates to the field of stem cell technology. More particularly, the invention describes compositions for the mobilization, migration, expansion and / or differentiation of stem cells and methods of using them. BACKGROUND [003] Stem cells have the ability to divide into indefinite periods in culture and become a wide variety of types of cells and specialized tissues which can then be used for basic research, drug discovery and treatment ( or prevention) of many diseases. Stem cells are typically divided into two main groups: adult stem cells and embryonic stem cells. [004] Adult stem cells are undifferentiated, but are present in differentiated tissues and are capable of differentiating in the cell types from which the adult stem cell originates. Adult stem cells have been derived from several sources, Petition 870170016572, of 03/14/2017, p. 5/13 2/115 such as the nervous system, bone marrow, adipose tissue, dermis, pancreas and liver. Stem cells were also isolated from the umbilical cord and the placenta. Adult-type stem cells are also believed to be found in smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, spongy bone tissue, cartilaginous tissue, pancreatic dutch tissue, spleen tissue, thymus, tonsil tissue, Peyer plaque tissue, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, cardiac tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, renal tissue , digestive tract tissue, esophageal tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterine tissue, eye tissue (including retinal tissue), testicular tissue, ovarian tissue, prostate tissue, connective tissue , endocrine tissue and mesentery tissue. [005] Embryonic stem cells are undifferentiated cells derived from the embryo. Typically, these cells are extracted from the internal cell mass of blastocytes and, when grown under unique conditions, either alone or in combination with a variety of feeder cells, embryonic stem cells maintain an euploid karyotype, do not undergo senescence and retain senescence. ability to differentiate in cells of the endodermal, ectodermal and mesodermal lines. [006] Mesenchymal stem cells (Mesenchymal Stem Cells MSCs) refers to cells that have the potential for self-renewal and are capable of differentiation into multiple mesenchymal strains. These cells may have the ability to give rise to other types of cells in the germ layer, such as neuronal cells. The main source of MSCs is isolation from bone marrow and other tissues, such as adipose tissue. Due to its scarcity in fabrics 3/115 adults, MSCs are normally isolated only in small amounts, however, they have a high proliferation capacity and can be readily expanded in culture to generate clinically relevant numbers of cells through multiple passages. These cells have been shown to support tissue repair and regeneration, making them a promising tool for various therapeutic applications. [007] One of the first clinical uses of stem cells was for bone marrow transplants in patients with hematological malignancies, in which hematopoietic stem cells derived from the bone marrow donor were administered to the recipient after treating the recipient with a dose of radiation and / or chemotherapy in order to perform ablation not only of hematological malignancy, but also non-malignant hematopoiesis. The administration of non-malignant hematopoietic stem cells results in donor-specific hematopoiesis and, in some patients, cure of the malignancy. This was first described in 1957 in patients with acute leukemia after myeloablation. Stem cells have also been used as an autologous bone marrow transplant in patients given high doses of chemotherapy and / or radiation for the treatment of solid tumors, in order to restore bone marrow. The use of autologous hematopoietic cell transplants combined with high doses of chemotherapy / radiotherapy for solid tumors has been extensively investigated for cancers of the breast, colon, lung, nasopharynx and other types. [008] Clinical use of autologous stem cells has also been carried out for a variety of autoimmune indications, including rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus and systemic sclerosis. [009] The importance of technologies used to expand speed 4/115 las-stem, both adult and / or embryonic bypass, is illustrated by the clinical uses of these cells in the treatment of a wide variety of diseases. The cell culture systems used in these treatments (for example, liquid static culture, semi-solid culture and long-term bone marrow culture) appear to have their own requirements, which must be met before human stem cells can be cultured or other species of mammals. To date, however, a common requirement and disadvantage of stem cell culture systems has been the requirement for undefined components contained in animal sera (eg fetal bovine serum or horse serum) for optimal stem cell growth or expansion. . [0010] The use of serum in the culture of hematopoietic cells is disadvantageous for several reasons. Serum is a major source of undefined differentiating factors and thus tends to promote hematopoietic cell differentiation, rather than expansion. The serum efficiency varies between serum lots. Some batches of serum were found to be toxic to cells. In addition, the serum may be contaminated with infectious agents, such as mycoplasma, bacteriophages and viruses. These problems cause inconsistencies in the growth support properties of the medium, making it difficult to standardize stem cell production processes and making it difficult to interpret experiments performed on media containing serum. Thus, the use of serum represents an important obstacle to the clinical implementation of therapies related to stem cells. [0011] As a result, researchers have tried to replace animal serums or conditioned media with serum-free culture of varying degrees of chemical definition. These attempts have had varying degrees of success, depending on the identity of the type of cell you are trying to expand. The development of exempt means 5/115 serum was reviewed (Sandstrom, EE etal., Biotech. & Bioengin. 43: 706733 (1994); Collins, PC et al., Curr. Opin. Biotech. 7: 223-230 (1996); McAdams, TA etal., TIBTECH 14: 341-349 (1996)). A more attractive alternative would be a defined serum-free medium. Therefore, it is desirable to develop defined serum-free methods and media for (i) isolation of stem cells and (ii) expansion of these cells over an extended period of time through multiple passages, while maintaining their potential for differentiation into multiple strains. The creation of defined serum-free media and methods would provide a robust platform that would help enable the clinical implementation of stem cell-based therapies. SUMMARY OF THE INVENTION [0012] One embodiment of the present invention is a method of causing an effect selected from the group consisting of stem cell mobilization, stem cell migration, stem cell expansion and stem cell differentiation in an individual. The method includes administering DA-DKP to an individual who needs it. In this embodiment and in all other embodiments described here, DA-DKP can be administered in various formulations, be administered through various routes of administration and be produced by various methods. For example, DA-DKP can be administered as a pharmaceutical composition that includes DA-DKP. Such pharmaceutical compositions can also include N-acetyl tryptophan, caprylate and / or caprylic acid. Such pharmaceutical compositions may include a fraction of human serum albumin, in which substantially all of the albumin has been removed from the fraction, or may include a low molecular weight fraction of human serum albumin, such as a fraction of molecular weight below 5,000. Such fractions of human serum albumin can be produced by filtration. In addition, the pharmaceutical composition can be prepared by 6/115 heating of human serum albumin under conditions effective to cause the formation of DA-DKP or through a process that includes contact of human serum albumin with an enzyme that cleaves the two N-terminal amino acids of human albumin under conditions effective to produce DA-DKP. DA-DKP can be a synthetic DA-DKP. [0013] In all embodiments of the present invention, DA-DKP can be administered locally to the individual. For example, the site of local administration can be selected from a joint, a surgical site, a site of a segmented skeletal opening or unbroken fracture, a wound, an ulcer and an inflammatory rash. In addition, DA-DKP can be administered as a parenteral formulation, it can be administered systemically to the individual, or it can be administered as an oral dosage formulation. In addition, DA-DKP can be formulated as part of or an implantable device, such as one selected from a sponge, biocompatible polymer, bioerodible polymer, mass, gel, bone matrix, artificial bone matrix, screw, pin, tube endotracheal, stent, contact lens, pacemaker, central IV tube, Foley catheter and intracranial device. [0014] In all embodiments of the present invention, administration of DA-DKP can increase the production of CXCR4, decrease the production of CXCL12, increase the production of MMP14 or MMP13, increase the production of agrecana, increase the production of SDF1, increase the production of collagen 2A1 or any combination of the foregoing. Also, administration of DA-DKP may decrease the production of a protein selected from the group consisting of MAPK-activated protein kinase 3, beta-adrenergic receptor kinase 1, ADAM metallopeptidase with type I thrombopondin motif, activated protein kinase 2 by MAPK, C-Src kinase, Macrophage Removing Receptor, Nogina, Bruton tyrosine kinase, 7/115 glycogen synthase kinase-3 alpha-beta, HSP 90 alpha / beta, phosphoinositide kinase-3, alpha catalytic subunit and eukaryotic translation initiation factor 4A, Fibroblast Growth Factor 17 and combinations thereof. In addition, administration of DA-DKP can decrease the production of a protein selected from MAPK-activated protein 3 kinase, Nogine, phosphatidylinositol 3-kinase, alpha catalytic subunit and combinations thereof. Administration of DA-DKP can also increase the production of a protein selected from clusterin (Apolipoprotein J), prothrombin, C1QBP (hyaluronan binding protein 1), TNFSF 15 (VEGF inhibitor), mamaglobin 2, MIP3b (CCL 19), MCP 1 (CCL 2), PTHrP, spondin 1, elafin (elastase inhibitor), IL 11, NPS-PLA2, CFC 1 (cryptic protein), Testicana 1 (SPOCK 1), angiogenin, URB, MMP-3 , IP10 (CXCL 10), BSSP 4, IL 8 (CXCL 8), Rspo2, cystatin C, bFGF, Factor H, Coagulation Factor IX, SDF-1 (cxcl 12), CATC (dipeptidylpeptidase 1), PIGR, Ck- b-8-1 (variant with MPIF 1 splicing), C1s, EMR2, ART, DPP 2, SAA, TIMP-1, Semaphorin 3A and combinations thereof. Administration of DA-DKP may also increase the production of a protein selected from clusterin (Apolipoprotein J), C1QBP (hyaluronan-binding protein 1), MCP 1 (CCL 2), PTHrP, Elafine (elastase inhibitor), IL 11, MMP-3, bFGF, AEA, TIMP-1, Semaphorin 3A and combinations thereof. Administration of DA-DKP can also decrease the production of a protein selected from MAPK-activated protein kinase 3, Nogina, phosphatidylinositol 3-kinase, alpha catalytic subunit and combinations thereof and increase the production of a protein selected from the group consisting of Clusterin (Apolipoprotein J), C1QBP (hyaluronan binding protein 1), MCP 1 (CCL 2), PTHrP, Elafin (elastase inhibitor), IL 11, MMP-3, bFGF, AEA, TIMP-1 , Semaphorin 3A and combinations thereof. In addition, the administration of DA8 / 115 DKP can down-regulate Akt pathways in the individual. [0015] Another embodiment of the present invention is a method of stimulating chondrogenesis in an individual by administering a pharmaceutical composition that includes DA-DKP and optionally a component selected from N-acetyl tryptophan, caprylate, caprylic acid and combinations of them to an individual who needs it. In this modality, chondrogenesis can be stimulated in a stem cell, such as a stem cell selected from a parent cell and mesenchymal stem cells (Mesenchymal Stem Cells - MSCs). In this modality, stimulation of chondrogenesis can promote the repair of cartilage, bone and / or ligament or induce the repair or regeneration of chondral tissue in the individual. In addition, chondrogenesis can treat or ameliorate a chondrogenic disease in the individual, and chondrogenic disease can be a congenital, degenerative or fibrotic joint cartilage, rheumatoid arthritis or osteoarthritis. In this modality, chondrogenesis can treat or repair a condition selected from a cartilage defect, a skeletal defect or a fracture resulting from trauma or surgery. In addition, administration of DA-DKP can stimulate the formation of new bone tissue or cartilaginous tissue. [0016] In the chondrogenesis stimulation method, administration may include ex vivo stimulating stem cells and then administration of stimulated stem cells to the individual. For example, stem cells can be stimulated ex vivo by culturing a population of chondrocyte stem cells with DADKP, or a composition comprising a DA-DKP, for a time sufficient to stimulate chondrogenesis and the administration includes implantation of stimulated cells in a desired location in the individual. In this modality, chondrogenesis can result in an increase in the production of collagen, type 2A1 collagen, collagen of 9/115 type 1A1 or a 2-fold, 4-fold, 10-fold, 20-fold or 20-fold increase in collagen production. [0017] Other embodiments of the present invention include the use of DA-DKP or a composition including DA-DKP in the preparation of a medicament for stimulating chondrogenesis in mammals and the use of DA-DKP or a composition including DA-DKP for the chondrogenesis stimulation in mammals. [0018] Another embodiment of the invention is a method of stimulating the development of nerve tissue in an individual by administering a pharmaceutical composition that includes DA-DKP and optionally a component selected from N-acetyl tryptophan, caprylate, caprylic acid and combinations thereof to an individual who needs it. In this modality, the development of nervous tissue can be stimulated in a stem cell, such as a progenitor cell or mesenchymal stem cells (Mesenchymal Stem Cells - MSCs). Stimulation of nerve tissue development can promote the repair of brain nerves, spinal cord and / or peripheral nerves or induce the repair or regeneration of neurons, neuroglia and / or astrocytes in the individual. The development of nervous tissue can treat or ameliorate a disease of the central nervous system and / or a disease of the peripheral nervous system in the individual, such as a neurodegenerative disease. In addition, the development of nerve tissue can treat or repair a condition selected from an injury resulting from trauma or surgery. [0019] In the nerve tissue development stimulation method, administration may include ex vivo stimulating stem cells and then administration of stimulated stem cells to the individual. For example, stem cells can be stimulated ex vivo by culturing a population of stem cells from the line of neurons, neuroglia and / or astrocytes with DA-DKP, or composition 10/115 which includes DA-DKP, for a time sufficient to stimulate the development of nervous tissue and the administration stage includes implantation of the stimulated cells in a desired location in the individual. [0020] Other embodiments of the present invention include the use of DA-DKP or a composition that includes DA-DKP in the preparation of a medicament for stimulating nerve tissue development in an individual and the use of DA-DKP or a composition that includes DA-DKP for the stimulation of nerve tissue development in an individual. [0021] Another embodiment of the invention is a method of stimulating tissue development in an individual by administering DA-DKP to an individual who needs it. In this embodiment, the tissue can be selected from nervous tissue, adipose tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, spongy bone tissue, cartilaginous tissue, pancreatic dutch tissue, spleen tissue, thymus tissue, tonsil tissue, Peyer plaque tissue, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, cardiac tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophageal tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterine tissue, eye tissue, testicular tissue, ovarian tissue, prostate tissue, tissue connective tissue, endocrine tissue and mesentery tissue. [0022] Other embodiments of the invention are a supplement for serum-free eukaryotic cell culture media that includes DADKP, wherein a cell culture medium supplemented with said supplement is capable of supporting the expansion of stem cells and a composition which includes stem cells in a serum-free medium su11 / 115 supplemented with the media supplement. [0023] Another embodiment of the invention is a stem cell expansion method that includes contact of the stem cells with DA-DKP and culture of the stem cells under suitable conditions to facilitate the expansion of the stem cells. [0024] Another embodiment of the invention is a method of providing stem cells to a mammal through contact of stem cells with DA-DKP, culture of stem cells under suitable conditions to facilitate cell expansion; and introducing expanded cells into an individual. [0025] Another embodiment of the invention is a method for causing stem cells to differentiate into a particular type of cells by contacting stem cells with DA-DKP, culture of stem cells under conditions suitable to facilitate expansion of stem cells and addition of one or more differentiating factors or modification of culture conditions to induce stem cell differentiation to form a different type of cell. [0026] Another embodiment of the invention is a method of providing differentiated stem cells to an individual. The method includes contacting the stem cells with DA-DKP, culturing the stem cells under suitable conditions to facilitate the expansion of the stem cells and adding one or more differentiating factors or changing the culture conditions to induce cell differentiation to form a different type of cell. The method also includes the introduction of differentiated cells in the individual. [0027] This Summary of the Invention is not intended, nor should it be, interpreted as being representative of the full extent and scope of the present invention. In addition, references made herein to the present invention, or aspects thereof, should be understood to mean certain embodiments of the present invention and not to 12/115 must necessarily be understood as limiting all modalities to a particular description. The present invention is presented at various levels in detail in the Summary of the Invention, as well as in the attached drawings and Detailed Description and no limitation as to the scope of the present invention is intended by including or not including elements, components, etc. in this Summary of the Invention. Additional aspects of the present invention will become more easily apparent from the Detailed Description, particularly when taken in conjunction with the drawings. BRIEF DESCRIPTION OF THE DRAWINGS [0028] Figure 1 illustrates the average variation in pain scores for patients with osteoarthritis who received Ampion ™ compared to patients who received saline. [0029] Figure 2 are photographs of cell aggregates from stem cells treated with saline, dexamethasone, DADKP and Ampion ™. [0030] Figure 3 illustrates the increase in collagen and agrecan transcription by stem cells treated with DA-DKP when compared to saline. [0031] Figure 4 illustrates the increase in CXCR4 production by mesenchymal stem cells grown in 3D culture by Ampion ™ compared to saline. [0032] Figure 5 illustrates the decrease in CXCL12 production by mesenchymal stem cells grown in 3D culture by Ampion ™ compared to saline. [0033] Figure 6 illustrates the effect of Ampion ™ on stem cell migration in vitro. [0034] Figure 7 shows the effect of Ampion ™ on the transcription of collagen 2A1 by bone marrow derived from human mesenchymal stem cells. 13/115 [0035] Figure 8 demonstrates the effect of Ampion ™ on bone marrow agrecana transcription derived from human mesenchymal stem cells. [0036] Figure 9 demonstrates the effect of Ampion ™ on the transcription of GAPDH by bone marrow derived from human mesenchymal stem cells. [0037] Figure 10 illustrates the patient's layout of the clinical trial described in Example 8. [0038] Figure 11 illustrates the reduction in pain for patients with osteoarthritis who received injections of LMWF-5A when compared to vehicle control. DETAILED DESCRIPTION [0039] The present invention relates to methods for causing the mobilization, migration and / or differentiation of stem cells in an individual who needs it. The present invention is also directed to compositions that promote, in a safe and effective way, the expansion of stem cells and methods of using such compositions and expanded cells in the treatment of pathological conditions susceptible to treatment with stem cells. Definitions [0040] The term albumin substitute refers to any compound that can be used in place of human serum albumin in compositions of the invention to provide substantially similar or better results than human serum albumin. Preferably, albumin is of human origin. Most preferably, albumin is human serum albumin. [0041] The term expand or expand refers to the growth and division, not differentiation, of stem cells in culture. The term differentiation refers to the development of a cell of a particular type into a cell of another type. The development of a city 14/115 pluripotent hematopoietic stem squid in a myeloid precursor is an example of differentiation. Likewise, the development of precursor cells in another cell type is an example of differentiation. [0042] The term stem cell refers, in general, to all cells that have the ability to divide for indefinite periods of time and give rise to specialized cells. Within the definition of stem cells the following is included: a) totipotent cells, such as an embryonic stem cell, an extraembryonic stem cell, a cloned stem cell, a cell derived from parthenogenesis, a cell reprogrammed to have totipotent properties or a primordial germ cell; b) pluripotent cells, such as hematopoietic stem cells, a stem cell derived from adipose tissue, a mesenchymal stem cell, a stem cell from umbilical cord blood, a stem cell derived from placenta, a stem cell derived exfoliated tooth, a hair follicle stem cell or a neuronal stem cell; and c) a tissue-specific stem cell, such as a precursor cell for the neuronal, hepatic, nephrogenic, adipogenic, osteoblastic, osteoclastic, alveolar, cardiac, intestinal or endothelial lineage. Cells can be derived, for example, from tissues such as pancreatic tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone marrow tissue, spongy bone tissue, tissue cartilaginous, pancreatic tissue, pancreatic duct tissue, spleen tissue, thymus tissue, Peyer plaque tissue, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, cardiac tissue, lung tissue, vascular tissue, tissue endothelial, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophageal tissue, stomach tissue, small intestine tissue, intestine tissue 15/115 thick, adipose tissue, uterine tissue, ocular tissue (including retinal tissue), testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue and mesentery tissue. Specific stem cells are described below in the section entitled Ex Vivo Treatment Methods. [0043] The term totipotent cells refers to mammalian cells that have the potential to become any cell type in the adult body and any cell type in the extraembryonic membranes (eg placenta). Totipotent cells are the fertilized egg and approximately the first 4 cells produced through its divage. [0044] The term pluripotent stem cells refers to true stem cells with the potential to become any differentiated cell in the body, but cannot contribute to producing the components of extraembryonic membranes which are derived from the trophoblast. The amnion develops from the epiblast, not from the trophoblast. Three types of pluripotent stem cells have been confirmed: embryonic stem cells (Embryonic Stem - ES) (may also be totipotent in primates), embryonic lineage cells (Embryonic Germ - EG) and embryonic carcinoma cells (Embryonic Carcinoma - EC) . These EC cells can be isolated from teratocarcinomas, a tumor that occasionally occurs in a fetus' gonad. Unlike the other two, EC cells are usually aneuploid. [0045] The term multipotent stem cells refers to true stem cells that can differ only in a limited number of types. For example, the bone marrow contains multipotent stem cells that give rise to all blood cells, but may not be able to differentiate into other types of cells. [0046] The term mesenchymal stem cells refers to cells 16/115 multipotent stroma that can differentiate into a variety of cell types, including osteoblasts, chondrocytes, adipocytes and adipocytes (fat cells). [0047] The term hematopoietic stem cells or HSC refers to a stem cell that is capable of differentiating both in myeloid strains (ie, monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes / platelets and some cells dendritic) and lymphoid strains (ie, T cells, B cells, NK cells and some dendritic cells). [0048] The term ingredient refers to any compound, whether of biological or chemical origin, which can be used in cell culture medium to maintain or promote the growth or proliferation of cells. The terms component, nutrient and ingredient can be used interchangeably and all are meant to refer to these compounds. Typical ingredients that are used in cell culture media include amino acids, salts, metals, sugars, lipids, nucleic acids, hormones, vitamins, fatty acids, proteins and the like. Other ingredients that promote or maintain cell growth ex vivo can be selected by those skilled in the art according to the particular need. [0049] Cell culture means cells or tissues that are maintained, cultured or grown in an artificial environment in vitro. [0050] The term culture vessel refers to glass containers, plastic containers or other containers of various sizes that can provide an aseptic environment for growing cells. For example, flasks, plates with a single or multiple wells, discs with a single or multiple wells or microplates with multiple wells can be used. In addition, a bioreactor can be used for cell culture. [0051] The terms cell culture medium, culture medium and 17/115 medium formulation refers to a nutrient solution for cell culture or growth. [0052] The term contact refers to the mixing, addition, culture or agitation of one or more cells with one or more compounds, solutions, media, etc. [0053] A serum-free medium is a medium that does not contain complete serum (for example, human serum, fetal bovine serum, horse serum, goat serum, etc.). [0054] The term buffering agent refers to an agent that acts to stabilize the concentration of hydrogen ions and, therefore, the pH of a solution by neutralizing, within limits, both acids and bases. Suitable buffering agents which can be used in the supplement and in the medium of the present invention include N- [2-hydroxyethyl] piperazine-N '- [2-ethanesulfonic] (HEPES), β-glycerol-phosphate and bicarbonate buffer. [0055] The term individual refers to any animal, including mammals, birds, reptiles and amphibians and, in preferred embodiments, mammals, including humans, companion animals, farm animals and wild animals. Compositions [0056] The present invention provides compositions that are useful in methods of the present invention, such methods including in vivo methods for causing mobilization, migration and / or differentiation of stem cells in an individual by administering such compositions and methods of Expansion of stem cells ex vivo by providing stem cells to an individual to make stem cells differentiate. Thus, such compositions are useful in administration to an individual and indirect contact with stem cells or as an additive or supplement to the cell culture medium. The compositions of the present invention are particularly suitable for their use. 18/115 port the expansion of mesenchymal stem cells (Mesenchymal Stem Cells - MSCs). Such cells include, but are not limited to, chondrocytes, osteoblasts, osteoclasts and epithelial cells of the skin and vascular tissue. The compositions of the present invention are also suitable to support the expansion of both primary and immortalized cells from most or all embryonic origins (for example, ectoderm-derived, mesoderm-derived and endoderm-derived). These cells include cells of the central and peripheral nervous system (neurons, glial cells and astrocytes), epithelial cells (sensory epithelial cells, epidermal cells (skin, breast, hair, nails, pituitary gland, sebaceous gland)), connective tissue cells ( cartilage, bone, striated and smooth muscle, hematopoietic cells, lymphoid cells, kidney cells, adrenal cells, gonads) and parenchymal cells of the liver, pancreas, thyroid, thymus, as well as epithelial linings of the urinary bladder, urethra, tympanic cavity and fallopian tube Eustachian. These cells can be of human origin or another mammal or eukaryote. [0057] The compositions of the invention include aspartylalanyl diketopiperazine (i.e., Asp-Ala DKP or DA-DKP). Diketopiperazines (DKP) are a class of cyclic organic compounds that result from peptide bonds between two amino acids to form a lactam. They are the smallest possible cyclic peptides. The invention also provides a pharmaceutical product comprising a composition of DA-DKP. [0058] Methods of producing diketopiperazines, such as DA-DKP, are well known in the art and these methods can be employed to synthesize the diketopiperazines of the invention. See, for example, United States Patents Nos. 4,694,081, 5,817,751, 5,990,112, 5,932,579 and 6,555,543, United States Patent Application Publication No. 2004/0024180, PCT Applications WO 96/00391 and WO 97/48685 19/115 and Smith et al., Bioorg. Med. Chem. Letters, 8, 2369-2374 (1998), the full descriptions of which are incorporated herein by reference. [0059] For example, diketopiperazines, such as DA-DKP, can be prepared first through dipeptide synthesis. Dipeptides can be synthesized by methods well known in the art using L-amino acids, D-amino acids or a combination of D- and L-amino acids. For example, solid phase peptide synthesis methods can be used. Of course, dipeptides are also commercially available from numerous sources, including DMI Synthesis Ltd., Cardiff, United Kingdom (synthesis to order), Sigma-Aldrich, St. Louis, MO (mainly synthesis to order), Phoenix Pharmaceuticals, Inc. , Belmont, CA (synthesis to order), Fisher Scientific (synthesis to order) and Advanced ChemTech, Louisville, KY. [0060] Once the dipeptide is synthesized or acquired, it is cyclized to form a diketopiperazine. This can be accomplished through a variety of techniques. For example, United States Patent Application Publication No. 2004/0024180 describes a method of cyclizing dipeptides. Briefly, the dipeptide is heated in an organic solvent while distilling water. Preferably, the organic solvent is a low boiling azeotrope with water, such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n-butanol, 2-butanol, t-butanol, acetic acid butyl ester, carbon tetrachloride, chlorobenzene , chloroform, cyclohexane, 1,2-dichloroethane, diethyl acetal, dimethyl acetal, ethyl ester of acetic acid, heptane, methyl isobutyl ketone, 3-pentanol, toluene and xylene. The temperature depends on the reaction speed at which the cyclization occurs and the type of azeotropic agent used. The reaction is preferably carried out at 50-200Ό, more preferably 80-150Ό. The pH range in which cyclization occurs can be easily determined by 20/115 those versed in the technique. Advantageously, it will be 2-9, preferably 3-7. [0061] When one of the dipeptide amino acids has, or is derived to have, a carboxyl group on its side chain (e.g., aspartic acid), the dipeptide is preferably cyclized as described in United States Patent No. 6,555. 543. Briefly, the dipeptide, with the carboxyl group on the side chain still protected, is heated under neutral conditions. Typically, the dipeptide will be heated from about 80 ° to about 180 °, preferably about 120 °. The solvent will be a neutral solvent. For example, the solvent can comprise an alcohol (such as butanol, methanol, ethanol and higher alcohols, but not phenol) and an azeotropic co-solvent (such as toluene, benzene or xylene). Preferably, the alcohol is butan-2-ol and the azeotropic co-solvent is toluene. Heating is continued until the reaction is complete and such times can be determined empirically. Typically, the dipeptide will be cyclized by refluxing it for about 8-24 hours, preferably about 18 hours. Finally, the protecting group is removed from diketopiperazine. In doing so, the use of strong acids (mineral acids, such as sulfuric or hydrochloric acids), strong bases (alkaline bases, such as potassium hydroxide or sodium hydroxide) and strong reducing agents (for example, aluminum and lithium hydride ) should be avoided in order to maintain the chirality of the final compound. [0062] Dipeptides made on solid phase resins can be cyclized and released from the resin in a single step. See, for example, United States Patent No. 5,817,751. For example, the resin having a trapped N-alkylated dipeptide is suspended in toluene or toluene / ethanol in the presence of acetic acid (for example, 1%) or triethylamine (for example, 4%). Typically, basic cycling conditions are preferred for their faster cycling times. 21/115 [0063] Other methods of cyclizing dipeptides and producing diketopiperazines are known in the art and can be used in the preparation of diketopiperazines useful in the practice of the invention. See, for example, those references listed above. In addition, many diketopiperazines suitable for use in the present invention can be produced, as described below, from proteins and peptides. In addition, diketopiperazines for use in the practice of the invention can be obtained commercially, for example, from DMI Synthesis Ltd., Cardiff, United Kingdom (synthesis to order). [0064] The DA-DKP composition and / or products of the present invention can be prepared from solutions containing DA-DKP, including from commercially available pharmaceutical compositions comprising albumin, such as human serum albumin. DA-DKP has been found to be present in some commercially available intravenous pharmaceutical compositions containing albumin. The DA-DKP present in these pharmaceutical preparations is formed by means of heating steps frequently used in the manufacture of these pharmaceutical compositions. The heating results in the dividing and cyclization of the two N-terminal amino acids of the proteins to form DA-DKP. [0065] Consequently, DA-DKP can be prepared by heating solutions of albumin, as well as other proteins and peptides. For example, a solution of albumin in phosphate buffer at neutral pH is prepared. Preferably, the solution is a concentrated solution (for example, about 100-500 mM) to obtain protonation of the N-terminal amino acid. The solution is heated to 60Ό for between about 2 hours to several days, preferably about 4 days, to cause the formation of DA-DKP. Protein denaturation should preferably be avoided. This can be accomplished using shorter times and / or by adding caprylic acid or N22 / 115 acetyl tryptophan at about 0.02 M for each. [0066] DA-DKP can also be prepared by contacting a solution of albumin with an enzyme that can cleave the two N-terminal amino acids of the protein or peptide (eg, dipeptidyl peptidases, particularly DPP IV). Suitable dipeptidyl peptidases are commercially available, for example, from Sigma. The reaction should be carried out at a pH of 6-8, preferably in a buffer, such as phosphate buffer, at a temperature high enough to accelerate the reaction, but not so high that the protein is denatured (for example, 37Ό ). [0067] The preparation of DA-DKP compositions can be through well-known methods, such as ultrafiltration, chromatography (for example, size exclusion chromatography), affinity chromatography (for example, using a column of beads having, stuck an antibody or antibodies to the desired diketopiperazine (s) or an antibody or antibodies to the truncated peptide or protein), anion exchange or cation exchange, sucrose gradient centrifugation, chromatography, salt precipitation or sonication, which will remove some or all of the albumin in the solution. The resulting composition and / or product containing DA-DKP can be used and incorporated into pharmaceutical compositions as described above. [0068] In a specific embodiment, a composition of the present invention is prepared by treating a feed stream containing albumin through tangential flow filtration. As used here, the term tangential flow refers to the flow direction of the feed stream containing albumin in relation to the filtration medium. This flow direction can be tangential (also commonly referred to as cross flow) or normal flow or a combination of both. Tangential flow refers to a current 23/115 feed containing albumin characterized by the fact that most of the current flows through the surface of the filtration medium, whereas normal flow refers to a current characterized by the fact that most of the current flows through the filtration media in an angle of 90 ° to the surface of the filtration means. [0069] In another embodiment, a composition of the invention is produced by treating a feed stream containing albumin via chromatography. Reference here to chromatography is the mechanical and / or physical operation of separating a fraction of the feed stream containing albumin from the remaining fraction using a pressure drop across a stationary phase. The term mechanical chromatography, as used here, refers, without limitation, to size exclusion chromatography. The term physical chromatography, as used here, refers to, but is not limited to, affinity chromatography, ion exchange chromatography, rapid protein liquid chromatography and immunoaffinity chromatography. [0070] The stationary phase of a chromatography step may include, but is not limited to, resins (i.e. polystyrene, polystyrene divinylbenzene and polyacrylamide), ion exchange resins (i.e., sulfonated functional groups, quaternary ammonium, carboxylate and diethyl ammonium ), crosslinked agarose, dextrans, phosphocellulose, porous glass and silica, alumina and zirconia matrices. In addition, the stationary phase can be immobilized on a solid support particle or on the inside wall of a cylinder through physical attraction, chemical bonding and / or in situ polymerization after coating. The immobilized stationary phase can coat the outer surfaces of the particles and cylinder and / or fill any available pores within the solid particles. The stationary phase can be functionalized with biospecific ligands which include, but are not limited to, antibodies, receptors 24/115 protein tors, steroid hormones, proteins, vitamins and enzyme inhibitors. [0071] Using an ultrafiltration separation method, a human serum albumin composition can be passed through an ultrafiltration membrane having a molecular weight cut that retains albumin, while DA-DKP passes to the resulting filtrate or fraction . This filtrate can comprise components that have molecular weights of less than about 50 kDa, less than about 40 kDa, less than 30 kDa, less than about 20 kDa, less than about 10 kDa, less than about 5 kDa , less than about 3 kDa. Preferably, the filtrate comprises components with molecular weights of less than about 5 Da (also referred to as <5000MW). This fraction or filtrate of <5000MW contains DA-DKP which is formed after the aspartate-alanine dipeptide is cleaved from albumin and subsequently cyclized to diketopiperazine. [0072] Physiologically acceptable salts of the DA-DKP of the invention can also be used in the practice of the invention. Physiologically acceptable salts include conventional non-toxic salts, such as salts derived from inorganic acids (such as hydrochloric, hydrobromic, sulfuric, phosphoric, nitric and so on), organic acids (such as acetic, propionic, succinic, glycolic, stearic acids , lactic, malic, tartaric, citric, glutamic, aspartic, benzoic, salicylic, oxalic, ascorbic acid and so on) or bases (such as hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or N-derived organic cations, Ndibenzylethylenediamine, D-glucosamine or ethylenediamine). The salts are prepared in a conventional manner, for example, by neutralizing the free base form of the compound with an acid. [0073] The composition of the present invention can be a pharmaceutical solution having a concentration range of DA-DKP with a 25/115 minimum threshold of about 10 μΜ, about 20 μΜ, about 30 μΜ, about 40 μΜ, about 50 μΜ, about 60 μΜ, about 70 μΜ, about 80 μΜ, about 90 μΜ about 100 μΜ, about 110 μΜ, about 120 μΜ, about 130 μΜ, about 140 μΜ, about 150 μΜ, about 160 μΜ, about 170 μΜ, about 180 μΜ, about 180 μΜ, about 190 μΜ , about 200 μΜ, about 210 μΜ, about 220 μΜ, about 230 μΜ, about 240 μΜ, about 240, about 250 μΜ, about 260 μΜ, about 270 μΜ, about 280 μΜ, about 290 μΜ, about 300 μΜ, about 310, about 320 μΜ, about 330 μΜ, about 340 μΜ, about 350 μΜ, about 360 μΜ, about 370 μΜ, about 380 μΜ, about 390 μΜ or about 400 μΜ. The composition of the present invention can be a pharmaceutical solution having a DA-DKP concentration range with an upper limit of about 600 μΜ, about 580 μΜ, about 570 μΜ, about 560 μΜ, about 550 μΜ, about about 540 μΜ, about 530 μΜ, about 520 μΜ, about 510 μΜ, about 500 μΜ, about 490 μΜ, about 480 μΜ, about 470 μΜ, about 460 μΜ, about 450 μΜ, about 450 440 μΜ, about 430 μΜ, about 420 μΜ, about 410 μΜ, about 400 μΜ, about 390 μΜ, about 380 μΜ, about 370 μΜ, about 360 μΜ, about 350, about 350 340 μΜ, about 330 μΜ, about 320 μΜ, about 310 μΜ, about 300 μΜ, about 290 μΜ, about 280, about 270 μΜ, about 260 μΜ, about 250 μΜ, about 240 μΜ, about 230 μΜ, about 220 μΜ, about 210 μΜ or about 200 μΜ. [0074] An effective amount of DA-DKP in the composition of the present invention for treating the diseases described here can be a range with a minimum threshold of about 10 pg, about 15 pg, about 20 pg, about 25 pg, about 30 pg, about 35 pg, about 40 pg, about 45 pg, about 50 pg, about 55 pg, about 60 pg, about 65 pg, about 70 pg, about 75 pg, about 80 pg, 26/115 about 85 pg, about 90 pg, about 95 pg, about 100 pg, about 110 pg, about 120 pg, about 130 pg, about 140 pg, about 150 pg, about 160 pg, about 170 pg, about 180 pg, about 190 pg, about 200 pg, about 210 pg, about 220 pg, about 230 pg, about 240 pg, about 250 pg, about 260 pg, about 270 pg, about 280 pg, about 290 pg, about 300 pg, about 310 pg, about 320 pg, about 330 pg, about 340 pg, about 350 pg , about 360 pg, about 370 pg, about 380 pg, about 390 pg, about 400 pg, about 425 pg, about 450 pg, about 475 pg, or about 500 pg. In addition, an effective amount of DA-DKP in the composition of the present invention for the treatment of conditions described herein can be a range with a maximum limit of about 500 pg, about 490 pg, about 480 ng. about 470 ng. about 460 pg, about 450 pg, about 440 ng. about 430 ng. about 420 pg, about 410 pg, about 400 ng. about 390 ng. about 380 pg, about 370 pg, about 360 ng. about 350 ng. about 340 pg, about 330 pg, about 320 ng. about 310 ng. about 300 pg, about 290 pg, about 280 ng. about 270 ng. about 260 pg, about 250 pg, about 240 ng. about 230 ng. about 220 pg, about 210 pg, about 200 ng. about 190 ng. about 180 pg, about 170 pg, about 160 ng. about 150 ng. about 140 pg, about 130 pg, about 120 ng. about 110 ng. about 100 pg, about 90 pg, about 80 pg, about 70 pg, about 60 pg, about 50 pg, about 40 pg, about 30 pg, or about 20 pg. [0075] Dosage forms for topical or transdermal administration of compounds of the invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, plasters and drops. The active ingredient can be mixed under sterile conditions with a pharmaceutically acceptable carrier and any buffers or propellants. 27/115 that may be required. [0076] Ointments, pastes, creams and gels may contain, in addition to the active ingredient, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid , talc and zinc oxide or mixtures thereof. [0077] Powders and sprays may contain, in addition to the active ingredient, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder or mixtures of these substances. Sprays may additionally contain usual propellants, such as chlorofluorocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. [0078] Transdermal patches have the additional advantage of allowing controlled administration of the compounds of the invention to the body. Such dosage forms can be prepared by dissolving, dispersing or otherwise incorporating one or more compounds of the invention in a suitable medium, such as an elastomeric matrix material. Absorption enhancers can also be used to increase the flow of the compound through the skin. The rate of such flow can be controlled by providing a rate control membrane or dispersing the compound in a polymeric matrix or gel. [0079] Pharmaceutical compositions of the present invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions or emulsions or sterile powders that can be reconstituted into solutions or sterile injectable dispersions immediately before use, which may contain antioxidants, buffers, solutes which make the formulation isotonic with the intended recipient's blood or suspending or thickening agents. 28/115 [0080] Examples of suitable aqueous and non-aqueous vehicles that can be used in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and so on) and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Adequate fluidity can be maintained, for example, by using coating materials, such as lecithin, maintaining the required particle size in the case of dispersions and using surfactants. These compositions can also contain adjuvants such as wetting agents, emulsifying agents and dispersing agents. It may also be desirable to include isotonic agents, such as sugars, sodium chloride and so on in the compositions. In addition, prolonged absorption of the injectable pharmaceutical form can be achieved by including agents which delay absorption, such as aluminum monostearate and gelatin. [0082] In some cases, in order to prolong the effect of a drug, it is desirable to delay the absorption of the drug from subcutaneous or intramuscular injection. This can be achieved by using a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of drug absorption then depends on its rate of dissolution which, in turn, may depend on the crystal size and crystalline shape. Alternatively, delayed absorption of a parenterally administered drug is achieved by dissolving or suspending the drug in an oily vehicle. [0083] Injectable deposit forms are made by forming microencapsulation matrices of the drug in biodegradable polymers, such as polylactide-polyglycolide. Depending on the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples 29/115 of other biodegradable polymers include (poly) orthoesters and (poly) anhydrides. Injectable depot formulations are also prepared by enclosing the drug in liposomes or microemulsions that are compatible with body tissue. Injectable materials can be sterilized, for example, by means of filtration through a bacterial retention filter. [0084] The formulations can be presented in sealed containers with unit doses or multiple doses, for example, ampoules and vials, and can be stored in a lyophilized condition that requires only the addition of the sterile liquid vehicle, for example, water for injection, immediately before use. Extemporaneous injectable solutions and suspensions can be prepared from sterile powders, granules and tablets of the type described above. [0085] Kits comprising the pharmaceutical products of the present invention are also provided. The kits may comprise a DA-DKP composition formulated for administration by injection. DA-DKP can be prepared as described here, such as by removing albumin from a solution of a human albumin composition. The kits can contain sealed containers with unit doses or multiple doses, for example, ampoules and vials, and can be stored in a lyophilized condition that requires only the addition of the sterile liquid vehicle, for example, water for injection, immediately before use. Kits can also be stored in a state where the content is ready for direct use or injection. [0086] Although it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition). The pharmaceutical compositions of the present invention comprise a compound or compounds of the invention as an active ingredient in admixture with one or more pharmaceutically acceptable carriers and, optionally, with a 30/115 or more of other compounds, drugs or other materials. Each vehicle must be acceptable in the sense of being compatible with the other ingredients of the formulation and not harmful to the animal. Pharmaceutically acceptable vehicles are well known in the art. Regardless of the route of administration selected, the compounds of the present invention are formulated in pharmaceutically acceptable dosage forms by conventional methods known to those skilled in the art. See, for example, Remington's Pharmaceutical Sciences. [0087] The composition of the present invention may further comprise N-acetyl tryptophan (NAT), caprylic acid, caprylate or combinations thereof. Preferably, the composition may comprise NAT. The compositions of the present invention having NAT, caprylic acid, caprylate or combinations thereof can be a pharmaceutical composition having NAT, caprylic acid, caprylate or combinations thereof in a concentration range with a minimum limit of about 1 mM, about 2 mM, about 3 mM, about 4 mm, about 5 mM, about 6 mM, about 7 mM, about 8 mm, about 9 mm, about 10 mM, about 11 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 16 mM, about 17 mM, about 18 mM, about 19 mM or about 20 mM. In addition, the compositions of the present invention having NAT, caprylic acid, caprylate or combinations thereof can be a pharmaceutical composition having NAT, caprylic acid, caprylate or combinations thereof in a concentration range with a maximum limit of about 40 mM, about 39 mM, about 38 mM, about 37 mM, about 36 mM, about 35 mM, about 34 mM, about 33 mM, about 32 mM, about 32 mM, about 31 mM, about 30 mM, about 29 mM, about 28 mM, about 27 mM, about 26 mM, about 25 mM, about 24 mM, about 23 mM, about 22, or about 21 mM. Preferably, the concentration range is 31/115 about 4 mm to about 20 mM. [0088] In addition, the composition of the present invention may also comprise a second drug, such as an analgesic (such as lidocaine or paracetamol), an anti-inflammatory (such as betamethasone, non-steroidal anti-inflammatory drugs (Non-Steroid Anti -Inflammatory Drugs - NSAIDs), acetaminophen, ibuprofen, naproxen) and / or other suitable drugs. [0089] Instead of purifying the DA-DKP, pharmaceutical compositions comprising albumin found in the mammalian receptor of the treatments of the present invention can be administered to stimulate the expansion of stem cells in the mammal. Although compositions comprising these proteins and / or peptides which are commercially available can be used if they contain diketopiperazines, especially DA-DKP, it is preferable to treat albumin as described above to increase the DA-DKP content prior to administration of the improved compositions. The mammal is preferably a human being and the proteins and / or peptides are preferably human proteins and / or peptides. Parenteral routes of administration are preferred. [0090] Using the compositions or cell culture media supplemented with the compositions of the present invention, the growth, expansion and differentiation of stem cells can be regulated by the addition of defined growth factors or other cytokines. Such an influence on stem cells in culture containing normal serum is not possible, since the undefined components in the serum obscure cellular responses to defined factors. Thus, using the compositions of the present invention, stem cells can be expanded and differentiated in suspension culture and in the absence of stromal cells, collagen or support matrices. [0091] The supplement and compositions or culture of suple cells 32/115 combined with the compositions of the present invention allow the growth and expansion of both pluripotent stem cell populations and differentiated offspring. [0092] Compositions or cell culture media supplemented with the compositions of the present invention can be used for culture of stem cells derived from a number of animals, including humans, monkeys, murines, rats, hamsters, rabbits, guinea pigs horse, cows, pigs, dogs, horses, cats, goats and sheep. Preferably, human stem cells are cultured. Yet another specific modality of aspects one or two is that in which stem cells are totipotent, pluripotent or multipotent stem cells. In a particular embodiment, stem cells are embryonic stem cells, fetal stem cells, extraembryonic stem cells or adult stem cells. [0093] Compositions or cell culture media supplemented with the compositions of the present invention may also include one or more ingredients selected from the group consisting of one or more antioxidants, one or more albumin or albumin substitutes, one or more lipid agents , one or more insulins or insulin substitutes, one or more transferrins or transferrin substitutes, one or more trace elements, one or more glucocorticoids, N-acetyl-L-cysteine, Human Ex-Cite®, ethanolamine, human zinc insulin , human transferrin saturated with iron, selenium, hydrocortisone, D, tocopherol L-acetate and 2-mercaptoethanol. For example, N-acetyl-L-cysteine can be solubilized in doubly distilled water. The pH of N-acetyl-L-cysteine at this point is approximately 2.0. To avoid protein denaturation, the pH of N-acetyl-Lcysteine is adjusted to 7.0 with 5N sodium hydroxide and is added to the mixture. The entire mixture is then filtered through a 0.2 micron low-protein filter. 33/115 [0094] The supplement or medium of the present invention can be in liquid form or can be kept in dry form. The type of liquid carrier and the method used to dissolve the ingredients in solution can vary and can be determined by those skilled in the art with no more than routine experimentation. In general, the liquid vehicle is water. [0095] The supplement or concentrated medium or formulation of the present invention (both aqueous and dry forms) are typically sterilized to prevent unwanted contamination. Sterilization can be achieved, for example, by ultraviolet light, filtration or heat. [0096] All the supplement ingredients of the present invention which are of human origin (e.g., human serum albumin, transferrin) are heat treated before use by heating at 600 ° C for 10 hours. [0097] Those skilled in the art are familiar with methods for culturing hematopoietic cells. For example, guidelines for culturing hematopoietic cells are described in Freshney, R. I. et a!., Eds., Culture of Hematopoietic Cells, Wiley-Liss, New York, 1994, pages 81-98. [0098] The present invention also provides a kit comprising a carrier medium, such as a compartmentalized cardboard box or package to receive, in narrow confinement therein, one or more container means, such as vials, tubes, ampoules, vials and so on. onwards, wherein a first container contains the cell culture compositions or media supplemented with the compositions of the present invention. Optionally, a second container medium contains a basal medium. Preferably, the container containing the supplement of the present invention can be stored from about -135 to about 40, preferably from about -5 to about 800, more preferably from about -5 to about -200 and, still 34/115 more preferably, about -200. A container containing the medium of the invention is preferably stored at about 2 to about 80 and, more preferably, about 40. [0099] The present invention also provides a composition comprising stem cells in a serum-free medium, wherein the serum-free medium, which is supplemented with compositions of the present invention, is capable of supporting the growth of stem cells in serum-free culture. Aliquots of this composition can be frozen at about -800 and below. Aliquots of this composition can be stored indefinitely at less than or equal to about -135 O. After an aliquot of the composition is thawed and opened, using the sterile cell culture technique, stem cells can be grown in serum-free culture. . [00100] Another modality is the use of a composition of the invention in the storage and transport of stem cells in order to maintain the viability, mobility and function of stem cells. A composition of the invention can be used individually or added to a variety of agents known in the art to allow transport of stem cells. This is particularly important in situations of bone marrow stem cell transport in which freezing and thawing cells is not carried out in various situations. The ability to properly store stem cells during transport would allow tissue extraction in physical locations separate from the stem cell processing unit. [00101] Another embodiment of the invention is a pharmaceutical preparation comprising a composition of the invention generated in a Good Manufacturing Practices / Good Tissue Practices environment, so that it is suitable for clinical use. Subsequent to the concentration and quantification of units of activity, the composition of the invention can be diluted in an excipient or vehicle. Can be 35/115 it is advantageous to formulate the compositions in unit dosage form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units suitable as unitary dosages for the subject to be treated, each unit containing a predetermined amount of active compounds of the compositions of the present invention calculated to produce the desired therapeutic effect in combination with the required pharmaceutical carrier. The unit dosage forms of the invention are dependent on the necessary amount of a composition to stimulate the proliferation of the respective stem cells whose proliferation and / or differentiation is being sought. The amount of composition necessary to stimulate the proliferation and / or differentiation of the desired stem cells can be formulated in a single dose or can be formulated in several dosage units. Treatment may require one dose at once or it may require repeated doses. [00102] The actual formulation of the compositions of the invention will be carried out according to conventional practices that are known to those skilled in the art. These are well known in the art and the one chosen is based on the route of administration that will be used, as well as specific pharmacokinetic properties that are desired. For example, the preferred form of therapy using a composition of the invention is an injectable dosage and, more preferably, an injection formulated for administration in the specific area that requires stem cell regeneration. However, several modalities are possible. For example, parenteral routes of administration may include, for example, intra-articular, intravenous, intradermal, intraspinal, intraperitoneal, subcutaneous or intramuscular injection, oral (for example, ingestion or inhalation), transdermal (topical), transmucosal and rectal . Solutions or suspensions for formulating Therapeutic compositions of the present invention may include: a sterile diluent, such as water for injection, saline (eg, phosphate buffered saline (PBS, UPS)), fixed oils, glycerin or other synthetic solvents; antibacterial and antifungal agents, such as parabens, a polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol and so on), chlorobutanol, phenol, ascorbic acid, thimerosal and so on; antioxidants, such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylene diaminetetraacetic acid; buffers, such as acetates, citrates or phosphates, and agents for adjusting tonicity, such as sodium chloride or dextrose. The desired fluidity can be maintained, for example, by using a coating, such as lecithin, by maintaining the required particle size in the case of dispersion and by using surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, sodium chloride, polyalcohols, such as mannitol or sorbitol, in the composition. Prolonged administration of the injectable compositions can be obtained including an agent that delays absorption. Such agents include, for example, aluminum monostearate and gelatin. The parenteral preparation can be enclosed in ampoules, disposable syringes or multi-dose vials made of glass or plastic. It is known in the art, and common practice for oral compositions, to generally include an inert diluent or an edible carrier. Oral compositions may be liquid or may be enclosed in gelatin capsules or compressed into tablets. Tablets, pills, capsules, troches and so on can contain any of the following ingredients or compounds of a similar nature: a binder, such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient, such as starch or lactose; a disintegrating agent, such as alginic acid, Primogel or corn starch; a lubricant, such as magnesium stearate or 37/115 Sterotes; colloidal silicon dioxide. Transmucosal administration can be achieved using nasal sprays or suppositories. For transdermal administration, the active compounds are formulated in ointments, ointments, gels or creams, as generally known in the art. In Vivo Treatment Methods [00103] Modalities of the invention include the use of the compositions of the invention to obtain the mobilization, migration, differentiation and / or expansion of stem cells within a living organism. Clinical situations where administration of the compositions of the invention is desirable include conditions where an increase in the number of stem cells is required due to a disease or senescence of endogenous stem cells or conditions that benefit from the repair function on the surrounding tissues or elsewhere of the organism. Such stem cells can be present in pancreatic tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone marrow tissue, spongy bone tissue, cartilaginous tissue, pancreatic tissue, pancreatic duct tissue , spleen tissue, thymus tissue, Peyer plaque tissue, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, cardiac tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue , kidney tissue, digestive tract tissue, esophageal tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterine tissue, eye tissue (including retinal tissue), testicular tissue, ovarian tissue, prostate tissue connective tissue, endocrine tissue and mesentery tissue. [00104] The administration of the compositions of the invention can be carried out systemically or in an environment located in the individual. For example, a local administration area can be any location 38/115 of the body in which tissue development is desired or beneficial, such as a joint, a surgical site, a site of a segmented skeletal opening or unbroken fracture, a wound, an ulcer or an inflammatory skin rash. The compositions of the invention can also be administered systemically, such as via an oral dosage formulation. In addition, the compositions of the invention can be administered to an individual in, on or as part of an implantable device. For example, such a device may be a sponge, biocompatible polymer, bioerodible polymer, mass, gel, bone matrix, artificial bone matrix, screw, pin, endotracheal tube, stent, contact lenses, pacemaker, central IV tube, catheter Foley or intracranial device. [00105] The invention includes a method for causing an effect selected from stem cell mobilization, stem cell migration, stem cell expansion and stem cell differentiation in an individual by administering DA-DKP to a individual who needs them. In this method, the DA-DKP administration step can have an effect of increasing the production of CXCR4, decreasing the production of CXCL12, increasing the production of MMP14 or MMP13, increasing the production of agrecana, increasing the production of SDF1, increasing the collagen production 2A1 or any combination of the foregoing. Also, administration of DADKP may decrease the production of a protein selected from the group consisting of MAPK-activated protein kinase 3, beta-adrenergic receptor kinase 1, ADAM metallopeptidase with type I thrombopondin motif, activated protein kinase 2 by MAPK, C-Src kinase, Macrophage Remover Receptor, Nogine, Bruton tyrosine kinase, glycogen synthase alpha-beta kinase-3, HSP 90 alpha / beta, phosphoinositide kinase-3, alpha catalytic subunit and factor 4A of eukaryotic translation start, Fi Growth Factor 17 39/115 broblasts and combinations thereof. In addition, administration of DA-DKP can decrease the production of a protein selected from MAPK-activated protein 3 kinase, Nogine, phosphatidylinositol 3-kinase, alpha catalytic subunit and combinations thereof. Administration of DA-DKP can also increase the production of a protein selected from clusterin (Apolipoprotein J), prothrombin, C1QBP (hyaluronan binding protein 1), TNFSF 15 (VEGF inhibitor), mamaglobin 2, MIP3b (CCL 19), MCP 1 (CCL 2), PTHrP, spondin 1, elafin (elastase inhibitor), IL 11, NPS-PLA2, CFG 1 (cryptic protein), Testicana 1 (SPOCK 1), angiogenin, URB, MMP-3 , IP10 (CXCL 10), BSSP 4, IL 8 (CXCL 8), Rspo2, cystatin C, bFGF, Factor H, Coagulation Factor IX, SDF-1 (cxcl 12), CATC (dipeptidyl-peptidase 1), PIGR, Ck-b-8-1 (variant with MPIF 1 splicing), C1s, EMR2, ART, DPP 2, SAA, TIMP-1, Semaphorin 3A and combinations thereof. Administration of DA-DKP may also increase the production of a protein selected from clusterin (Apolipoprotein J), C1QBP (hyaluronan-binding protein 1), MCP 1 (CCL 2), PTHrP, Elafine (elastase inhibitor), IL 11, MMP-3, bFGF, AEA, TIMP-1, Semaphorin 3A and combinations thereof. Administration of DA-DKP can also decrease the production of a protein selected from MAPK-activated protein kinase 3, Nogina, phosphatidylinositol 3-kinase, alpha catalytic subunit and combinations thereof and increase the production of a protein selected from the group consisting of Clusterin (Apolipoprotein J), C1QBP (hyaluronan binding protein 1), MCP 1 (CCL 2), PTHrP, Elafin (elastase inhibitor), IL 11, MMP-3, bFGF, AEA, TIMP-1 , Semaphorin 3A and combinations thereof. In addition, administration of DA-DKP can down-regulate Akt pathways in the individual. Methods of the invention include methods of stimulating the development of tissues in an individual through administration 40/115 of DA-DKP to an individual. These methods are suitable for the development of any tissue in the individual, including one or more nervous tissue, adipose tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, spongy bone tissue , cartilaginous tissue, pancreatic dutch tissue, spleen tissue, thymus tissue, tonsil tissue, Peyer's plaque tissue, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, cardiac tissue, lung tissue, tissue vascular, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophageal tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterine tissue, eye tissue, tissue lung, testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue and mesentery tissue. [00106] A specific method of in vivo treatment includes conditions in which a greater number and / or faster recovery of stem cells is required in a patient after a medical procedure. For example, after a bone marrow transplant, hematopoietic cell expansion is desirable so that the patient does not succumb to bacterial or viral infections. Such an expansion of granulocyte and monocyte precursors would be useful to reinforce the immune defenses following a bone marrow transplant. Thus, the present invention provides methods and compositions that can be administered to a patient who has undergone a bone marrow transplant. [00107] Another modality of an in vivo method supports the expansion of endogenous stem cells after an injury has occurred and where endogenous stem cells are mobilized or begin to differentiate, but do not do so at levels high enough to stimulate a response beneficial. Thus, another modality of information 41/115 is the administration of compositions of the invention to patients having an injury in a selected tissue of pancreatic tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone marrow tissue, spongy bone tissue, cartilage tissue, liver tissue, pancreatic tissue, pancreatic duct tissue, spleen tissue, thymus tissue, Peyer's plaque tissue, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, tissue cardiac, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophageal tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterine tissue , ocular tissue (including retinal tissue), testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue and bone tissue esentério. [00108] Another embodiment of the invention is the administration of a composition of the present invention individually or in combination with growth factors to promote healing. [00109] Another embodiment of the invention is a method of treating diabetes by administering compositions of the invention to an individual with diabetes to induce islet regeneration. This method may also include the administration of other factors capable of inducing islet regeneration. Such factors can be, for example, soluble proteins, membrane-bound proteins or transcription factors that act intracellularly. For example, it is known that the administration, in mice, of combinations of GLP-1, EGF and gastrin leads to the regeneration of islets or islet-like cells that are functionally effective in models such as NOD or streptozocin-induced diabetes. The compositions of the invention can be added to islet cultures in differentiation in vitro to expand the numbers of stem cells, but can also be administered 42/115 directly to a patient's pancreas in vivo in order to restore islet cell function or amplify the effect of administered hormones. [00110] Another embodiment of an in vivo method of the invention is a treatment for multiple sclerosis by administering a composition of the invention to a patient having multiple sclerosis for expansion of neuronal progenitor cells. Alternatively, a composition of the invention can be administered to patients suffering from multiple sclerosis individually or in combination with agents capable of inhibiting the autoimmune process. The synergy in therapeutic effects is anticipated through the concomitant induction of tissue healing and repair of the immune system. [00111] Another embodiment of the invention is the treatment of an immune disorder, such as an autoimmune disease, by extracting stem cells from an autologous patient, treating the cells with a composition of the invention and / or other combinations of expansion compounds stem cells, ablation of the patient's immune system and subsequent reintroduction of stem cells into the host for reconstitution. A composition of the invention can subsequently be supplied directly to the host in order to accelerate the reconstitution of hematopoiesis. Autoimmune diseases that can be treated through these processes include, but are not limited to, type 1 diabetes, multiple sclerosis, rheumatoid arthritis, systemic sclerosis, Hashimoto's thyroiditis, myasthenia gravis, scleroderma, systemic lupus erythematosus, graft versus host disease and so on against. [00112] Another embodiment of the invention is a treatment for an autoimmune disease, such as type 1 diabetes, multiple sclerosis, rheumatoid arthritis, Hashimoto's thyroiditis, myasthenia gravis, scleroderma and so on comprising administering a make up 43/115 location of the invention to a relevant location within the organism that needs such treatment to increase the production of stem cells, such as administration to a joint or pancreas of an organism to increase chondrogenesis or production of islet cells. [00113] Another embodiment of the invention includes the use of a composition of the invention for expansion of antigen-specific and / or non-specific immune regulatory cells within an organism. The expansion of such cells is used to control pathological immune responses. For example, it is known that Th2 cells, TR1 cells, CD4 + CD25 + FoxP3 + cells and doubly negative CD3 + cells are able to suppress immune responses in an antigen-specific manner, whereas NKT cells, myeloid suppressor cells, M2 cells and cells Immature dendritic cells are able to suppress immune responses in a non-antigen-specific manner. A composition of the invention can be used for culture and ex vivo expansion of immune regulatory cells derived from a patient who needs it. A composition of the invention can be used individually or in combination with factors known to be involved in the development of said cells for administration to an organism to increase the proliferation of these cells without loss of function. [00114] Another embodiment of the invention is a stroke treatment by administering a composition of the present invention to a patient with a stroke to cause the in vivo mobilization, migration, expansion and / or differentiation of endogenous neural cells. This method may also include administering a composition of the invention in combination with polypeptides and proteins known in the art for expanding neural cells. [00115] Another embodiment of the invention is the use of a composition of the invention as an adjuvant for the treatment of a disease 44/115 degenerative by applying a combination of a composition of the invention with known therapies in order to enhance the beneficial effects of known therapies. [00116] Another embodiment of the invention relates to the use of a composition of the invention described here for the preparation of a medicament to increase the expansion of stem cells in vivo or ex vivo. [00117] Another embodiment of the invention is an in vivo method for the treatment of a degenerative joint disease by administering a composition of the present invention to cause mobilization, migration, expansion and / or differentiation of stem cells in the individual. A degenerative joint disease is a progressive deterioration of the articular cartilage that covers joints. A degenerative joint disease (osteoarthritis) is a progressive non-infectious load-bearing disorder in the joints. The cartilage of the normal joint is smooth, white and translucent. It is composed of cartilaginous cells (chondrocytes) integrated in a sponge-like matrix made of collagen, protein polysaccharides and water. With early primary arthritis, the cartilage becomes yellow and opaque, with irregular localized areas and softening of the surfaces. As degeneration progresses, the soft areas become cracked and worn, exposing the bone under the cartilage. The bone then begins to remodel and increase in density, while the remaining cartilage begins to wear away. Eventually, osteophytes (spurs of new bone) covered by cartilage form at the edge of the joint. As the mechanical effort increases, the cartilage requires repair. Cartilage cells are unable to produce enough spongy-type matrix and, therefore, damaged cartilage cannot be repaired. Cartilage has no blood supply to increase healing. Most degenerative joint diseases are the result of instability or changes in 45/115 articulation by aging. This includes degenerative arthritis in the elderly and, in younger individuals, may be the result of injuries, bruises, abnormal joint configuration (ie, hip dysplasia) or mechanical wear due to rupture of the anterior cruciate ligament, patellar dislocation or dissecting osteochondritis, for example. example. Degenerative joint disease can occur in any joint in the body including, without limitation, knee, hip, shoulder, hand and spine. [00118] Conventional pharmaceutical therapies for degenerative joint disease include administration of paracetamol, non-steroidal anti-inflammatory drugs (Non-Steroid Anti-Inflammatory Drugs NSAIDs), narcotics and corticosteroids. [00119] A particular embodiment of the invention is a method of stimulating chondrogenesis in an individual by administering a pharmaceutical composition that includes DA-DKP. The pharmaceutical composition can include any pharmaceutical composition described herein and, therefore, can also include a selected component of N-acetyl tryptophan, caprylate, caprylic acid and combinations. In this method, chondrogenesis can be stimulated in a stem cell and the stem cell can be a progenitor cell or a mesenchymal stem cell (Mesenchymal Stem Cell - MSC). The stimulation of chondrogenesis in the present method can promote the repair of cartilage, bone and / or ligament or induce the repair or regeneration of chondral tissue in the individual. Thus, the method is useful for treating or ameliorating a chondrogenic disease, such as a congenital, degenerative or fibrotic joint, cartilaginous disease, rheumatoid arthritis or osteoarthritis in the individual. Also, chondrogenesis can be useful to treat or repair a selected condition of a cartilage defect, a skeletal defect and a fracture due to trauma or surgery. For example, a skeletal defect can be a large segmental skeletal opening and the cartilage defect can be a 46/115 tearing of articular cartilage, a congenital cartilage defect or damage to cartilage induced by bone fractures. [00120] This method can be performed by stimulating stem cells ex vivo and then administering the stimulated stem cells to the individual. For example, stem cells can be stimulated ex vivo by culturing a stem cell population of the chondrocyte lineage with DA-DKP, or a composition comprising a DA-DKP, for a time sufficient to stimulate chondrogenesis and then be implanted in a desired location in the individual. [00121] This method can result in an increase in collagen production, including type 2A1 collagen and / or type 1A1 collagen. Specifically, chondrogenesis can result in at least a 2-fold, 4-fold, 10-fold, 20-fold, 20-fold, 25-fold increase in collagen production. Another embodiment of the present invention includes a method of stimulating nerve tissue development in an individual by administering a pharmaceutical composition that includes DA-DKP. The pharmaceutical composition can include any pharmaceutical composition described herein and, therefore, can also include a selected component of N-acetyl tryptophan, caprylate, caprylic acid and combinations thereof, to an individual. In this method, the development of nerve tissue can be stimulated in a stem cell, which can be selected from a progenitor cell and a mesenchymal stem cell (Mesenchymal Stem Cell MSC). Stimulation of nerve tissue development can promote brain, spinal cord and / or peripheral nerve repair or induce the repair or regeneration of neurons, neuroglia and / or astrocytes in the individual. In addition, the development of nervous tissue can treat or ameliorate a disease of the central nervous system and / or a disease of the peripheral nervous system in the individual. Such of 47/115 children can be selected from neurodegenerative diseases. The development of nerve tissue can also treat or repair a selected condition of an injury resulting from trauma or surgery. [00122] This method can be performed through stimulation of stem cells ex vivo and then administration of stimulated stem cells to the individual. For example, stem cells can be stimulated ex vivo by culturing a population of stem cells from the line of neurons, neuroglia and / or astrocytes with the pharmaceutical composition for a time sufficient to stimulate the development of nerve tissue and then be implanted in a desired location in the individual. Ex Vivo Treatment Methods [00123] The ex vivo expansion of stem cells for systemic administration represents an effective treatment modality for disorders or diseases susceptible to treatment with stem cells. For example, mesenchymal stem cells expanded in vitro reduced graft versus host disease both acute and chronic in a patient suffering from grade IV severe graft versus host disease in the liver and intestine after bone marrow transplantation. The administration of 2 million cells / kg on day 73 after bone marrow transplantation led to a long-term remission of graft versus host disease (Le Blanc et al., 2004, Lancet 363: 14391441), demonstrating that [00124] A the present invention further provides methods for increasing the expansion of stem cells ex vivo. The method involves contacting the stem cells with a composition of the invention or mixing or incubating the stem cells with a growth medium that includes a composition of the present invention to expand the population of stem cells by subsequent administration, either locally to 48/115 a necessary location or systemically. Such methods make it possible to use populations of stem cells, such as mesenchymal stem cells, which expand relatively slowly and, therefore, are often not practical for generalized clinical use. [00125] The compositions of the invention, in addition to increasing the rate of proliferation of stem cells, can also maintain stem cells in an undifferentiated state. Stem cells reside in unique physiological niches, and although cell growth within mimics from such niches has been accomplished, mimics from the stem cell niche are often unusable in clinical situations. An example of this is the fact that early hematopoietic stem cells require feeder cell lines to be expanded in high amounts or the fact that optimal growth of embryonic stem cells is still achieved primarily using murine feeders. The present invention provides compositions for increasing stem cell expansion that can recreate conditions similar to stem cell niches using approaches that are viable in the clinical setting. [00126] As discussed above, the methods of extraction, expansion and identification of specific stem cell phenotypes is important for clinical implementation. For example, bone marrow is commonly used as a source of therapeutic stem cells for myocardial disease, angina and hematopoietic cell transplantation. However, bone marrow in general contains a large number of different stem cells, in addition to the well-known hematopoietic CD34 + stem cells. CD34- hematopoietic stem cells, mesenchymal stem cells and myogenic precursor cells were all found in the bone marrow, in addition to T cells, B cells and relatively high levels of CD4 + CD25 + regulatory T cells. Given the heterogeneity of bone marrow as a starting material for 49/115 stem cell therapy, it is evident that the understanding of particular cell populations, as well as the ability to isolate and expand them, would substantially advance the field of stem cell therapeutic products. Consequently, if a population of stem cells is derived from adult or embryonic sources, stem cells can be grown in a culture medium containing a composition of the invention to increase the population of a heterogeneous mixture of cells, or a purified population of stem cells in order to increase the rate of expansion or growth of stem cells when grown in culture. [00128] Various methods of growing stem cells outside the body have been developed and are known in the art. Originally, most of the work in the area of growth and expansion of stem cells was carried out in the hematopoietic system using bone marrow cells. The ability of newly isolated or cultured bone marrow cells to form colonies on methyl cellulose or agar was used as an option. [00129] The development of stem cell expansion techniques began with work aimed at increasing the number of colonies formed in semi-solid media. The first experiments used a variety of uncharacterized sera and conditioned media. For example, conditioned media from the trophoblast cell line, conditioned media from xenogenic stromal cells, tumor cell supernatants and healthy lymphocytes were used. The work was also carried out to design serum-free systems using ingredients such as human transferrin and bovine insulin. The specific advantage of such systems is that hematopoietic stem cells could be expanded without concurrent differentiation. Early long-term culture systems required the 50/115 use of murine feeder cells (stromal), since human strains had certain disadvantages in terms of hematopoietic promoting activity. There were numerous drawbacks in using murine feeder cell lines to maintain the viability and proliferative potential of stem cells. As a result, an effort was made to overcome the difficulties in growing human feeder cells and a variety of such cells were developed. [00130] The stem cells to be expanded by the methods of the present invention can be isolated from any organ of any mammalian organism by any means known to those skilled in the art. Stem cells can be derived from embryonic or adult tissue. Those skilled in the art can determine how to isolate stem cells from the organ or tissue of particular interest using methods known in the art. Stem cell populations can also be enriched using antibodies to other stem cell markers on the surface. Such markers include, but are not limited to, FLK-1, AC133, CD34, ckit, CXCR-4, Oct-4, Rex-1, CD9, CD13, CD29, CD44, CD166, CD90, CD105, SH-3, SH- 4, TRA-1-60, TRA-1-81, SSEA-4, Sox-2 and so on. Those skilled in the art will be able to determine the specific cell marker useful for isolating stem cells from the desired tissue. [00131] Those skilled in the art will be able to determine a suitable growth medium for the initial preparation of stem cells. Growth media commonly used for stem cells include, but are not limited to, Iscove modified Dulbecco media (Iscove's Modified Dulbecco's Media - IMDM), DMEM, KODMEM, DMEM / F12, RPMI 1640 medium, McCoy medium 5A, essential alpha medium (a-MEM), F-12K nutrient mixing medium (mo 51/115 Kaighn specification, F-12K), X-vivo 20, Stemline, CC100, H2000, Stemspan, MCDB 131 medium, Eagle basal medium (Basal Media Eagle BME), Glasgow essential minimum medium, modified Eagle medium (Modified Eagle Medium - MEM), Opti-MEM I reduced serum medium, Waymouth MB 752/1 medium, Williams medium E, NCTC-109 medium, neuroplasm medium, BGJb medium, Brinster BMOC-3 medium, medium CMRL, CO2-independent medium, Leibovitz L-15 medium and so on. [00132] If desired, other components, such as growth factors, can be added as desired. Examples of growth factors and other components that can be added include, but are not limited to, thrombopoietin (ThromboPOietin - TPO), stem cell factor (Stem Cell Factor - SCF), IL-1, IL-3, IL-7, flt-3 ligand (Flt-3L), G-CSF, GM-CSF, Epo, FGF-1, FGF-2, FGF-4, FGF-20, IGF, EGF, NGF, LIF, PDGF, bone morphogenic protein (Bone Morphogenic Protein- BMP), activin-A, VEGF, forskolin, glucocorticoids and so on. In addition, the initial isolation medium may contain serum such as fetal calf serum, horse serum or human serum or, more preferably, serum substitute components. Numerous agents have been introduced in means to alleviate the need for serum. For example, serum substitutes included bovine serum albumin (Bovine Serum Albumin - BSA), insulin, 2mercaptoethanol and transferrin (TransFerrin - TF). [00133] Stem cells can then be stored for a desired period of time, if necessary. Stem cell storage methods are known to those skilled in the art. Typically, stem cells are treated with a cryoprotection process, then stored frozen until needed. [00134] Stem cells can be purified prior to contact with a composition of the present invention by methods co 52/115 known in the art using, for example, antibody technology, such as cell panning, through the use of fluorescence activated cell sorting (FACS) methods or activated cell separation methods magnets, such as MACS apparatus, to isolate cells having the desired stem cell markers or to remove unwanted contaminating cell types having unwanted cell markers prior to contact with the compositions of the invention. Other methods of purification or concentration of stem cells may include the use of techniques such as counterflow centrifugation elutriation, density balance centrifugation, unitary gravity velocity sedimentation, formation of immune rosettes and immune adhesion, depletion of T lymphocytes. Examples Stem cell markers that may be useful in purification include, but are not limited to, FLK-1, AC133, CD34, c-kit, CXCR-4, Oct-4, Rex-1, CD9, CD13, CD29, CD44, CD166, CD90, CD105, SH-3, SH-4, TRA-1-60, TRA-1-81, SSEA-4, Sox-2 and so on. Examples of markers on the cell surface that can be used as markers of unwanted contaminating cell types depend on the stem cell phenotype sought. For example, if collecting pluripotent hematopoietic cells is desired, the contaminating cells will have markers dedicated to differentiated hematopoietic cells, such as CD38 or CD33. In addition, contamination of non-hematopoietic cells would be detected by the lack of CD45 expression. If selection of stromal mesenchymal cells is desired, then the contaminating cells would be detected by the expression of hematopoietic markers, such as CD45. In addition, stem cells can be purified based on properties, such as size, density, adhesion to certain substrates, or the efflux of certain dyes, such as Hoechst 33342 or Rhodamine 123. 53/115 [00135] Stem cells can be genetically modified at any stage of preparation. For example, a gene that encodes a selectable marker or another gene of interest can be introduced into the prepared stem cells. [00136] To increase the growth of stem cells, stem cells are contacted with a composition of the invention or mixed or incubated with media containing a composition of the invention. [00137] Antibiotics, antifungals or other compounds that prevent contamination can be added to the incubation medium, if desired. Exemplary compounds include, but are not limited to, penicillin, streptomycin, gentamicin, fungizone or others known in the art. [00138] Stem cells in contact with the compositions of the invention can be expanded through multiple passages using methods known to those skilled in the art. For example, stem cells isolated from an individual are counted and the density of viable cells determined using trypan blue dye exclusion methods. For subculture (or passage) of cells, they are again plated at a density of 5000 cells / cm2 in culture medium containing a composition of the invention in new tissue culture flasks coated with protein, such as gelatin or fibronectin, and left grow in a humidified incubator at 37Ό and 5% CO2. If necessary, the culture medium is replenished. Upon reaching subconfluence (about 90%), the culture medium is removed and the adherent cells trypsinized with 0.25% trypsin and 1 mM EDTA for 3-5 min at 37Ό. Adherent cells are washed twice with PBS before trypsinization. Trypsin is neutralized and the cells are pelleted by centrifugation at 300 x g for 10 min and resuspended in culture medium. 54/115 which may also contain a composition of the invention. The cells are washed by centrifugation at 300 x g for 10 min and resuspended in hot culture medium also containing a composition of the invention. Stem cells can be subcultured through multiple passages that use the same passage protocol. [00139] Stem cells can be contacted with the compositions of the invention, for example, simply by mixing the composition (s) with the stem cell culture. The mixing can be carried out in a multiplicity of suitable containers capable of maintaining the viability of stem cells. Said containers may include, but are not limited to, tissue culture flasks, conical tubes, culture bags, bioreactors or cultures that are continuously mixed. The stem cells can then be allowed to grow as desired. In some situations, it will be desirable to use a combined culture system, in which the cells are cultured with a first type of culture condition, then, subsequently, another culture condition is used. For example, when rapid expansion of hematopoietic stem cells is required without differentiation, the cells can be cultured initially in a high concentration of a composition of the invention for 48 hours or a period of time necessary to induce the cycling of the stem cells. Subsequently, media containing cytokines can be added to the culture for passages after the first 48 hours. Those skilled in the art will understand that, depending on the type of stem cells and level of differentiation desired, different concentrations of the compositions of the invention can be added at different time points in the culture. For example, in a particular culture situation, adding compositions of the invention at the beginning of the culture may not be ideal. 55/115 [00140] The desired ratio of stem cells to the compositions of the invention can be determined by those skilled in the art. For example, a ratio of less than about 1: 1000 to 1000: 1 or more (stem cell preparation for composing the invention) can be used. For example, a ratio of stem cells to composition preparation of the invention from about 1: 750, 1: 500, 1: 250 or 1: 100 to about 100: 1,250: 1,500: 1 or 750: 1 can be used . This proportion can vary, for example, depending on the temperature, incubation time, number of stem cells, desired activity sought in the stem cells, the type of stem cells, the purity of the stem cells, the amount of placental tissue used as a starting point and so on. Stem cells can be isolated from their growth medium prior to contact with a composition of the invention or stem cells can remain in their growth medium with an added composition of the invention. [00141] The period of time during which the stem cell is in contact with a composition of the invention can be determined by those skilled in the art. In general, the contact step can range from less than about 1 second, 30 seconds or 60 seconds to about 2 or 3 weeks or more. Preferably, the contact step is between about 2, 5, 10, 30 or 45 minutes to about 12, 14, 16, 18 or 20 days. More preferably, the contact step is between about 1.3, 5, 8 or 24 hours to about 3, 5, 7 or 10 days. [00142] In addition, conditions that promote the proliferation or differentiation of certain types of cells can be used during culture. These conditions include, but are not limited to, variation in temperature, variation in the content of carbon dioxide / oxygen, variation in the turbidity of said medium or exposure to small molecules that modify cell cultures, such as nutrients, inhibitors of certain enzymes, stimulators of certain enzymes, 56/115 inhibitors of histone deacetylase activity, such as valproic acid, trichostatin A, trapoxin A or depsipeptide, inhibitors of DNA methyltransferase activity, such as 5-azacytidine, inhibitors of the GSK-3 enzyme and so on. [00143] The role of oxygen tension in self-renewal and viability of stem cells is also an important issue that is considered in the present invention. It is known that hematopoietic stem cells tend to reside in bone marrow hypoxia niches and that, as cells differentiate into more mature offspring, they progressively migrate to areas of bone marrow with higher oxygen tension (Ivanovic et al., 2002, Exp Hematol 30: 67-73, which is incorporated herein by reference in its entirety). This important variable in tissue culture has been explored in studies that show that maximum expansion of human CD34 stem cells capable of complete hematopoietic reconstitution of NOD-SCID mice was obtained under hypoxia conditions using an oxygen tension as low as 1.5%. In addition, other stem cells, such as neuronal stem cells, also appear to be located in hypoxic niches and have preferential expansion at low oxygen content under in vitro conditions, as opposed to normal oxygen tension. In addition, embryonic stem cells, which grow at similar proliferative rates between normoxia and hypoxia, retain a superior ability to form teratomas in vivo and embryo bodies in vitro when grown under hypoxic conditions. Consequently, one embodiment of the invention is the use of hypoxic conditions during part or all of the incubation of stem cells in specialized incubators with an oxygen tension ranging from 0.1% to 7.5%, preferably 0.5% to 5%, more preferably 3% -5%. In addition, another embodiment of the invention is the use of hypoxic conditions in combination with the compositions of the invention in order to increase 57/115 the proliferation without differentiation of stem cells that are being grown in culture. [00144] In terms of improving the ability of a composition of the invention to stimulate stem cell proliferation without differentiation, an adjuvant approach that is considered to be a modality of the invention is the use of enzyme inhibitors in conjunction with a composition of the invention. For example, histone deacetylases are a class of enzymes involved in epigenetically opening parts of chromatin to transcription factors, thus allowing the expression of genes that, under normal conditions in adults, would not be expressed. For example, the telomerase gene (hTERT catalytic subunit) is involved in the process of cellular immortalization and is expressed under physiological conditions only in embryonic stem cells, as well as some hematopoietic cells in the bone marrow, in an abnormal way. The functional role of the telomerase enzyme is to repair the ends of the shortened telomeric chromosomes so that the cells can escape replicative senescence. Pathologically, telomerase is the enzyme responsible for the ability of cancer cells to proliferate indefinitely in cell culture. Under normal physiological conditions, fibroblasts do not express telomerase and undergo replicative senescence. A variety of reports have been published describing that treatment of fibroblasts with histone deacetylase inhibitors, such as tricostatin A, again induces the expression of functional telomerase (Mukhopadhyay et al., 2005, J Cell Mol Med 9: 662-669; Hou et al ., 2002, Exp Cell Res 274: 25-34; Cong et al., 2000, J Biol Chem 275: 35665-35668, each of which is incorporated herein by reference in its entirety). This suggests that manipulation of the histone deacetylase pathway can be used as a method of de-differentiating cells or offers the possibility of rejuvenating parents who are almost in replicative exhaustion. In 58/115 In fact, it has been demonstrated that the observed effect of prolonging life due to caloric restriction is related to the histone deacetylase pathway (Howitz et al., 2003, Nature 425: 191-196, which is incorporated by reference in the in full). The clinical relevance of manipulating this pathway is illustrated in experiments with valproic acid, an antidepressant that is in clinical use and is a histone deacetylase inhibitor with a potency similar to tricostatin A in some models. Treatment of hematopoietic stem cells derived from bone marrow with valproic acid has been shown to increase both proliferation and self-renewal by accelerating the progression of the cell cycle (Bug, supra). Said acceleration was accompanied by a negative regulation of the inhibitory factor p21 (cip-1 / waf-1). In addition, treatment with valproic acid suppressed GSK3 activity and activated the Wnt signaling pathway, both of which are associated with self-renewal in both hematopoietic stem cells (Gotoh et al., 1997, Cell Growth Differ 8: 721-729 ; Baba et al., 2005, Immunity 23: 599-609, each of which is incorporated herein by reference in its entirety), as well as embryonic (Sato et al., 2004, Nat Med 10: 55-63; He et al. , 2005, Clin Lung Cancer 7 ·. 54-60, each of which is incorporated herein by reference in its entirety). The power of valproic acid to synergize with known hematopoietic stem cell stimulating cytokines, such as Flt3L, TPO, SCF and IL-3, has been demonstrated (De Felice et al., 2005, Cancer Res 65: 1505-1513, which is incorporated by reference here in full). [00145] Through contact with a composition of the invention, stem cells are able to increase their growth rate and expand rapidly. When desired, culture conditions are used which allow the compositions of the invention to increase stem cell proliferation without inducing differentiation. Any suitable method for determining the rate of growth and 59/115 stem cell differentiation can be used to determine the growth rate and count of the stem cells thus produced. For example, flow cytometry analysis of markers associated with stem cell retention, assays in semi-solid media for quantification of early and dedicated parents and NOD-SCID Repopulation Activity Assays in vivo to quantify the number of stem cells in vivo with reconstitution activity. These assays can be modified and altered to allow detection of specific stem cell subtypes. The assays can also be developed in immunocompromised mice, such as the NOD-SCID genus, by inducing a pathology for which human stem cells are expected to be therapeutic. For example, it has been shown that human stem cells have therapeutic activity in a variety of non-hematopoietic configurations in NOD-SCID mice, as well as nude mice. The rate of cell growth can also depend on other factors such as, for example, temperature, type of stem cells, media content and the time allowed for the placenta incubation stage and contact stage. Those skilled in the art will be able to alter these variables to adjust the growth rate as needed. [00146] Another embodiment of the invention is the use of compositions, or cell culture media supplemented with the compositions of the present invention, to stimulate stem cell proliferation including, for example: [00147] - human embryonic stem cells characterized by the expression of markers, such as SSEA-4, GC ™ -2 antigen, TRA 1-60, Cripto, gastrin releasing peptide receptor (GRP), podocalixin-like protein ( PODXL) or human telomerase reverse transcriptase (hTERT); 60/115 [00148] - stem cells that produce human oocytes characterized by the expression of markers, such as Vasa, Oct-4, Dazl, Stella, Fragilis, Nobox, c-Kit and Sca-1; [00149] - parthenogenetically generated stem cells characterized by the expression of markers, such as Oct-4, alkaline phosphatase, telomerase, SSEA-4, TRA 1-60 and TRA 1-81; [00150] - sperm stem cells reprogrammed for pluripotent germ stem cells characterized by the expression of markers, such as Oct-4, Nanog, Dppa5 and Rex1; [00151] - hematopoietic stem cells characterized by markers, such as Stem Cell Antigen (SCA +), negative lineage (lin-), c c-kit +, CD34 +, CD38-, CD33-; [00152] - mesenchymal stem cells characterized by markers, such as LFA-3, ICAM-1, PECAM-1, P-selectin, L-selectin, CD49b / CD29, CD49c / CD29, CD49d / CD29, CD61, CD18, CD29, 6-19, thrombomodulin, telomerase, CD10, CD13, STRO-1, STRO-2, VCAM1, CD146, THY-1; [00153] - placenta-derived multipotent cells characterized by markers, such as Oct-4, Rex-1, CD9, CD13, CD29, CD44, CD166, CD90, CD105, SH-3, SH-4, TRA-1-60 , TRA-1-81, SSEA-4 and Sox-2; [00154] - stem cells derived from adipose tissue characterized by markers, such as CD13, CD29, CD44, CD63, CD73, CD90, CD166, aldehyde dehydrogenase (ALDH) and ABCG2; [00155] - umbilical cord blood stem cells characterized by markers, such as CD34, c-kit and CXCR-4; [00156] - deciduous tooth stem cells characterized by markers, such as STRO-1, CD146 (MUC18), alkaline phosphatase, EFEM and bFGF; [00157] - neural stem cells characterized by markers, such as 61/115 as RC-2, 3CB2, BLB, Sox-2hh, GLAST, Pax 6, Nesting, Muashi-1 and prominine; [00158] - epithelial stem cells of the stomach characterized by markers, such as Musashi-1, c-Hairy-1 and HES-5; [00159] - skeletal muscle stem cells characterized by markers, such as positive desmin, SCA-1 +, CD45- and having a lateral population profile in flow cytometry by dye exclusion; [00160] - mammary gland stem cells characterized by markers, such as SCA-1 positive, CD45- and keratin-6; [00161] - dermal stem cells characterized by markers, such as SCA-1 positive, CD34 +, CD45- and positive for alpha6integrin, beta-integrin, keratin 14 and keratin 19; [00162] - myocardial stem cells characterized by markers, such as SCA-1 positive, c-kit positive and having a lateral population profile in flow cytometry by dye exclusion; [00163] - mesangial stem cells characterized by markers, such as SCA-1 positive, c-kit positive and having a profile of the lateral population in flow cytometry by dye exclusion; [00164] - hepatic oval stem cells characterized by markers, such as SCA-1 positive, c-kit positive and CD34 positive; or [00165] - pancreatic stem cells characterized by markers, such as nestin, CK-8, CK-18, lsl-1, Pdx-1, Pax-4 and Ngn-3. [00166] The compositions of the invention can be used as a proliferation stimulator itself or as an additive to media known to be useful for culturing said cells. An example of such a tissue culture medium is Dulbecco's modified Eagle's medium (Dulbecco's Modified Eagle's Medium - DMEM). [00167] Another embodiment of the invention is the use of cell culture compositions or media supplemented with the compositions of 62/115 present invention to stimulate the proliferation of totipotent stem cells generated through cloning using nuclear transfer technologies. [00168] Methods of administering stem cells to a mammal include contacting stem cells with a composition of the invention, culturing the stem cells under suitable conditions to facilitate the expansion of the stem cells and introducing the expanded cells into a mammal . [00169] Thus, the serum-free supplement of the present invention can also be used to prepare a type of stem cell of interest for exploding in a mammal. In this embodiment, cells which have been made to differentiate ex vivo are introduced into a mammal. For example, hematopoietic cells that have been made to differentiate into a hematopoietic stem cell, precursor or progenitor cell can be introduced into the bone marrow or blood stream of the mammal. Differentiated cells can be introduced, for example, into the bone marrow or blood of the mammal by well-known techniques. [00170] In some embodiments of the invention, a method for expanding or growing stem cells is provided which includes contact of a stem cell with the growth medium containing a composition of the invention. The stem cell can be: [00171] a) a totipotent cell, such as an embryonic stem cell, an extraembryonic stem cell, a cloned stem cell, a cell derived from parthenogenesis; [00172] b) a pluripotent cell, such as hematopoietic stem cells, a stem cell derived from adipose tissue, a mesenchymal stem cell, an umbilical cord blood stem cell, a placenta derived stem cell, a stem cell stem derived from exfoliated tooth, a hair follicle stem cell or a cell 63/115 neural trunk; or [00173] c) a tissue specific progenitor cell, such as a precursor cell for the neuronal, hepatic, nephrogenic, adipogenic, osteoblastic, osteoclastic, alveolar, cardiac, intestinal or endothelial lineage. [00174] The incubation step can take place, for example, at a suitable temperature, such as from about 320 to about 400. [00175] Within these methods, stem cells can be manipulated during their isolation or production or during their expansion or immediately before their administration to a mammal. Such manipulation can adapt stem cells to perform a therapeutic function after administration to the mammal. For example, it has been shown that bone marrow mesenchymal cells treated with the DNA inhibitor methyltransferase 5-aza-cytidine transdifferentiate in myocardial tissue and improve the left ventricular ejection fraction and inhibit cardiac remodeling. Other types of stem cells have been used to improve myocardial activity, perfusion and decrease ventricular remodeling, including mesenchymal stem cells, endothelial stem cells and skeletal myoblasts. A limiting factor in these studies is the lack of reproducible methods to expand a sufficient number of semi-differentiated progenitor stem cells that have a high propensity to repair the heart. This drawback is, in part, due to the lack of suitable culture media for the expansion of such single cell populations. The compositions and methods of the present invention overcome these drawbacks by providing means of reproducible expansion and maintenance of progenitor cell populations for subsequent therapeutic administration to a mammal. [00176] Each publication or patent cited here is incorporated by reference in its entirety. 64/115 Mechanism of Action [00177] DA-DKP and, in particular, low molecular weight fractions of commercial human serum albumin, such as Ampion ™ (Ampio Pharmaceuticals, Greenwood Village, CO) have several activities that can be classified as anti-inflammatory and remodeling / healing. The anti-inflammatory properties of Ampion ™ involve inhibiting vascular permeability by rearranging the cytoskeleton in endothelial cells (forming a cortical actin ring), inhibiting activation of memory T cells (non-virgin) by antiCD3 / CD28 or APC and reduction in the amount of pro-inflammatory cytokines (TNFa) produced by PBMCs in response to a strong pro-inflammatory stimulus (LPS). The remodeling / healing effects are through mobilization and differentiation of stem cells into tissue-specific cells (such as differentiation into chondrocytes in the case of the knee). [00178] Ampion ™ activates the transcription factor activated by ligand (Transcription Factor - TF) Peroxisome Proliferator Activator Receptor (Peroxisome Proliferator Activator Receptor - PPAR), possibly by binding to the molecule's co-activator site in both stem cells mesenchymal and PBMCs. TF arrays demonstrated activation of PPAR and its binding partner, Retinoid X Receptor (Retinoid X Receptor - RXR). PPAR belongs to the nuclear hormone receptor superfamily, expressed in inflammatory and immune cells. PPAR heterodimerizes with the Retinoid X Receptor (Retinoid X Receptor - RXR) in the nucleus and, together, the dimer binds to the peroxisome proliferation response element (Peroxisome Proliferation Response Element - PPRE) in the target gene promoter. The genes involved are anti-inflammatory, involved in the protection and differentiation of stem cells. Four functional domains were identified in PPAR: A / B, C, D and E / F. The N-terminal A / B domain contains the ligand independent activation function (AF-1), which is responsible for 65/115 stable by phosphorylation of PPAR. The C domain or DNA binding domain (DNA Binding Domain - DBD) promotes the binding of PPAR to PPRE in the promoter region of the target genes. Domain D is the cofactor binding site. The E / F domain is the ligand binding domain (LBD) responsible for the specificity of ligand and activation of PPAR binding to PPRE that increases the expression of target genes. [00179] The invention having now been described in general will be more easily understood by reference to the following examples, which are included only for the purpose of illustrating certain aspects of the modalities of the present invention. The examples are not intended to limit the invention, since those skilled in the art will recognize, from the above teachings and the examples below, that other techniques and methods can satisfy the claims and can be employed without departing from the scope of the invention claimed. EXAMPLES Example 1 [00180] A clinical trial was conducted to investigate the effect of intra-articular injection into the knee of the fraction <5000 MW (also referred to here as Ampion ™) to improve joint function and reduce knee osteoarthritis pain in adults with symptomatic primary osteoarthritis in the knees. A randomized, double-blind, placebo-controlled parallel study with 43 evaluable subjects was chosen as the appropriate design to estimate the treatment and safety effect of the <5000 MW fraction when it was injected into the knees in the study. Study Drug [00181] 2 arms; each subject received a single injection of 4 ml in one knee with either Amipon ™ or saline. 66/115 Study Population [00182] The study population consisted of 43 male or female patients aged 40-83 years (mean 63.0, standard deviation (SD) 9.6) 28 were male and 15 were male feminine. All subjects were Caucasian. The individuals' height ranged from 162-192 cm (mean 175.3, SD 8.1), with screening weight ranging from 56117 kg (mean 88.8, SD 13.89). The subjects were fully ambulatory, with symptomatic primary osteoarthritis of the knee for more than 6 months before screening with Grade II or III Kellgren Lawrence (indicating mild to moderate osteoarthritis). Grade II for 6 individuals and Grade III for 36 individuals. One individual was Grade IV. If both knees of an individual were osteroarthritic, one knee was chosen for the study, while the other knee received a standard of care. Exclusion Criteria [00183] The following are the exclusion criteria for the study population: [00184] 1. Unable as a result of medical evaluation and screening investigation. [00185] 2. History of allergic reactions to albumin. [00186] 3. history of allergic reactions to excipients in 5% human albumin. [00187] 4. Any local intra-articular or periarticular injection, injection or surgery in the indicator knee (last 6 months). [00188] 5. Operative arthroscopy (last 3 months). [00189] 6. Other surgical procedure on the indicator knee other than arthroscopy (last 12 months). [00190] 7. Any product under investigation for the knee (last months). [00191] 8. Kellgren Lawrence knee osteoarthritis of grade I or 67/115 IV (doubtful or serious). [00192] 9. Inflammatory or crystal arthropathies, acute fractures, severe loss of bone density, bone necrosis. [00193] 10. Isolated patellofemoral syndrome or chondromalacia. [00194] 11. Any other disease or condition that interferes with the free use and evaluation of the indicator knee. [00195] 12. Main injury to the index knee (previous 12 months). [00196] 13. Severe osteoarthritis of the hip ipsilateral to the index knee. [00197] 14. Any pain that could interfere with the assessment of pain in the indicator knee. [00198] 15. Any pharmacological or non-pharmacological treatment started or exchanged (previous 4 weeks). [00199] 16. Use of one of a. Any topical treatment (latest 48h), b. All painkillers and NSAIDs, with the exception of paracetamol (last 48 h), c. Anticoagulant therapy (last 48 h), d. Any treatments with systemic steroids (last 14 days), e. All immunosuppressants within a period of 5 times the half-life of the drug before randomization, f. Corticosteroids> 10 mg equivalent prednisolone per day (last 30 days), g. Any treatment with albumin (last 3 months). [00200] 17. Women who are pregnant or breastfeeding. [00201] 18. Women of childbearing potential who have a positive pregnancy test on day 1 before treatment. Study Evaluation [00202] The study consisted of a three-week screening period and an 84-day study participation period. Follow-up evaluations were performed 6 hours, 24 hours and 72 hours after the injection. Individuals were contacted by phone on Day 8, 68/115 Day 30 and Day 84 to assess general pain and mobility and monitor adverse events. Individuals were offered the option of intra-articular injection of betamethasone to the knee under investigation for pain relief after Day 8, if deemed necessary after an assessment by the investigator. Primary Result [00203] The numerical pain rating scale (NRS) in the knee study was completed at the pre-dose (pre-injection baseline), 6 hours after the dose on Day 1, 24 hours post-dose on Day 2, 72 hours post-dose on Day 4 and post-dose on Day 8, Day 30 and Day 84 (EOS or End-of-Study). Pain NRS is a numerical rating of 0-10, with 0 being no pain, 5 being moderate pain and 10 being the worst possible pain. Safety Endpoints [00204] The safety parameters of the study were incidence of adverse events, vital signs at the pre-dose and Day 4 of the study, twelve ECG readings on the screening and 24 hours post-dose and clinical safety blood tests ( biochemistry and hematology) assessed at screening and 24 hours post-dose. Secondary Endpoints [00205] The secondary endpoints of the study were percentage of responders on Day 30 and Day 84, defined as an improvement in pain NRS of two or more points, the change from the pre-injection baseline in the WOMAC 3.1 Osteoarthritis index (scale complete, subtotal of pain, subtotal of stiffness and subtotal of function) in 24 and 72 hours after intra-articular injection, the change from pre-injection baseline to emergency medication requirement (paracetamol) up to 24 hours and 72 hours after injection and intra-articular changes over time in mobility on Day 8, Day 30 and Day 84 post-dose compared to before the dose and the immediate post-dose period. 69/115 Intent to Treat (ITT) and Safety Population [00206] Study participants who were randomized and received at least one dose of study medication. ITT refers to individuals who met the inclusion / exclusion criteria. Population By protocol [00207] Study participants in the ITT set whose pre-dose pain score did not violate the inclusion / exclusion criteria. Efficacy Population [00208] Study participants in the pre-protocol population who did not receive emergency medication between 8 and 30 days. Statistical Analysis [00209] Primary: Analysis model of covariance (ANCOVA) to examine the mean difference (SD) between the treatment groups regarding the mean change in pain on day 30 and day 84 (EOS), adjusted for NRS of pain of baseline. [00210] Additional: Test X 2 for differences in the percentage of respondents. Cochran-Armitage trend test for differences in clinically significant improvements. Student's t-test: mean difference (SD) in pain NRS in 30 days. Security Analysis [00211] Adverse events and serious adverse events were listed by individual. Abstracts were presented for the treatment of adverse events classified by the MedDRA System Organ Class and Preferred Term regarding the global incidence and by severity and relationship with the study medication. The incidence of adverse events arising from treatment was compared between treatment groups. All clinical safety and tolerability data were listed for each individual and summarized by treatment. Vital signs and ECG parameters were noted and summarized by treatment. Laboratory values were listed, along with observations regarding the 70/115 clinical relevance, regarding values outside the normal range of the laboratory. Changes in screening were assessed for clinical significance. Results Table 4. Population Analysis set Study Size (n) Ampion ™ (n) Saline Solution (n) Security set 43 22 21 ITT Set 43 22 21 Per-protocol set a 41 20 21 Assessed by effectiveness 0 32 17 15 [00212] a: 2 subjects in the Ampion ™ group had baseline pain NRS <4 points [00213] b : 5 subjects in the Ampion ™ group and 6 subjects in the saline group required emergency medication Emergency [00214] Betamethasone injection: there was no apparent difference between the use of betamethasone injections between individuals who received Ampion ™ (5 of 22 individuals, 23%) compared with individuals who received saline solution (6 of 21 individuals, 29% ). [00215] Emergency medications (paracetamol): emergency pain relief medication in the study period with the knees within 24 hours after the injection occurred in a similar number of subjects who received Ampion ™ (6 out of 22 subjects) compared with subjects who received saline solution (6 of 21 subjects), with similar average doses of paracetamol used in each treatment group. Efficiency Results 71/115 Table 5. NRS of pain per treatment, mean (SD) of the population per protocol Treatment Prose 6 hPostdose 24 hPostdose 72 hPostdose Day 8Postdose 30thPostdose Day 84Postdose Ampion ™ 4.70 (0.7) 2.00(1.3) 3.20(1.5) 2.60(2.1) 2.90(2.1) 2.90(1.8) 3.21(1.8) Saline solution 5.29(1.4) 2.67(1.9) 3.00(17) 2.86(2.1) 3.33(1.9) 3.86(2.2) 4.81(2.3) Table 6. Mean change in least squares (LS) in pain NRS: population per protocol Treatment 6 hPostdose 24 h Postdose 72 hPostdose Day 8Postdose 30thPostdose Day 84Postdose D (Ampion ™) -3.06 -1.69 -2.31 -2.00 -2.16 -1.60 E (Saline solution) -2.28 -2.11 -2.22 -1.76 -1.09 -0.36 P value 0.15 0.42 0.89 0.71 0.12 0.07 [00216] Scale: -10 = greatest possible improvement in pain from baseline, 10 = lowest possible improvement (greatest increase) in pain from baseline. [00217] Day 1: 6 hours post-dose [00218] * adjusted for baseline pain NRS Table 7. Mean LS change in pain NRS: population evaluable for effectiveness Treatment 6 hPostdose 24 h Postdose 72 hPostdose Day 8Postdose 30thPostdose Day 84Postdose D (Ampion ™) -2.91 -1.99 -2.94 -2.45 -2.29 -2.22 E (Saline solution) -2.62 -2.61 -2.79 -2.22 -1.17 -0.46 P value 0.62 0.19 0.79 0.71 0.19 0.04 [00219] Scale: -10 = greatest possible improvement in pain from the li 72/115 baseline, 10 = lowest possible improvement (greatest increase) in pain from baseline. [00220] Day 1: 6 hours post-dose [00221] * adjusted for baseline pain NRS Percentage of responders on Day 84 (EPS): population by protocol (see Table 8) [00222] Respondent: decrease in pain NRS on Day 84 from -2 to -10 points (with -10 being the greatest possible improvement in pain). [00223] Non-responder: decrease in pain NRS on Day 84 from 1 to 10 (10 being the largest possible increase in pain). Table 8. Pain trends in 30 days from baseline, by treatment group Treatment Non-responder Answerer P value Ampion ™ 47.4% 52.6% 0.06 Saline solution 76.2% 23.8% Percentage of responders on Day 84 (EPS): population assessable for effectiveness (see Table 9) [00224] Respondent: decrease in pain NRS on Day 84 from -2 to -10 points (with -10 being the greatest possible improvement in pain) . [00225] Non-responder: decrease in pain NRS on Day 84 from 1 to 10 points (10 being the greatest possible increase in pain). [00226] Table 9. Trends in pain in 30 days from baseline, by treatment group Treatment Non-responder Answerer P value Ampion ™ 35.7% 64.3% 0.10 Saline solution 66.7% 33.3% Summary of achad os: Effectiveness: [00227] Overall pain (as assessed by the numerical pain score score) and WPMAC scores were reduced postdose for each of the treatment groups throughout the study period (p <0.05), except placebo on Day 84. In addition addition, there was a 73/115 trend in a significant difference between changes from baseline on Day 30 and Day 84 for subjects who received Ampion ™ compared to subjects who received the placebo saline solution (Day 30: p = 0.12; Day 84: p = 0.07). This trend became statistically significant in individuals who did not receive emergency medication (p = 0.04). There was a trend towards a higher percentage of responders at the end of the study (Day 84) for individuals who received Ampion ™ vs. placebo (p = 0.06). Use of paracetamol emergency medication up to 72 hours post-dose was higher in treatment group E (saline). See Figure 1. Adverse events (AEs) [00228] Emerging treatment AEs were reported by 20 of the 43 subjects (47%) after dose administration, with a total of 27 adverse events. Commonly occurring AEs were headache and joint swelling and knee stiffness. Most individuals reported adverse events classified as mild only (16 of 43 patients, 37%). Only 4 individuals (9%) reported adverse events of moderate severity: [00229] - Ampion ™: joint damage and hypertension [00230] - Saline: Back pain and hematoma at the blood vessel puncture site [00231] There was no apparent difference in the incidence of moderate AEs between individuals who received Ampion ™ (2 individuals, 9%) compared to individuals who received saline solution (2 individuals, 10%). These AEs were all considered to be probably or definitely unrelated to the study drug. [00232] There were no AEs classified as serious adverse events. [00233] AEs considered to be (possibly) related to the administration of the study drug were reported in 3 74/115 of 43 individuals (7%). There was no apparent difference in the incidence of related AEs between individuals who received Ampion ™ (1 subject, 5%), compared with individuals who received saline (2 subjects, 10%): [00234] a. Mild severity headache that started five minutes after administration of the treatment and resolved 1.8 hours later (Ampion ™). [00235] b. Mild severity headache that started 5 hours after treatment and resolved 0.5 hours later (saline). [00236] c. Swelling of the joints of the right knee (study knee) of mild severity that started 2.4 days after the administration of the treatment and resolved 21 hours later (saline). [00237] In general, a greater proportion of adverse events arising from treatment were reported in subjects who received saline (12 subjects, 57%) compared to subjects who received Ampion ™ (8 subjects, 36%). AEs considered to be (possibly) related to the administration of the study drug were reported in 3 of 43 individuals (7%) and included headache and swelling of the knee joints. There were no deaths or other serious adverse events. There was no evident difference in terms of safety, assessed through clinical laboratory biochemical tests, vital signs and ECG assessments between treatments. Study Findings [00238] Pain (as assessed by numerical pain score) and WOMAC score was reduced post-dose for each treatment group during the study period, except placebo on Day 84, with no significant differences between groups of treatment. Despite a higher baseline pain NRS for the saline group compared to the Ampion ™ group, there was a 75/115 trend of a long-term effect of the study drug, with a higher percentage of subjects who responded to Ampion ™ on Day 84 compared to saline. In subjects who received Ampion ™, overall pain was reduced post-dose during the study period, whereas subjects who received saline did not experience a reduction in post-dose pain on Day 84. Use of paracetamol emergency medication up to 72 hours post-dose was higher in treatment group E (saline). Ampion ™ was considered safe and well tolerated at the dose used in the study. The improvement in pain in this study on Day 84 is consistent with tissue regeneration caused by the administration of Ampion ™. Example 2 [00239] This example demonstrates the increase in chondrogenesis in stem cells by DA-DKP and other drug treatments. [00240] The inventors investigated the possibility that DA-DKP directly increases chondrogenesis or has an additive or synergistic effect on the transcription and / or translation of genes important to the chondrocyte lineage. [00241] Materials: [00242] - Stem cells: mesenchymal stem cells derived from human bone marrow: mesenchymal stem cells derived from human bone marrow from passage 5 (HUXMA - 01001, Cyagen Biosciences, Sunnyvale CA) [00243] - Mesenchymal Stem Cell Chondrogenic Medium Differentiation (GUXMX-90041, Cyagen Biosciences, Sunnyvale CA) [00244] - Chemically defined stem cell medium TheraPEAK MSCGM (190.632 Lonza) [00245] - 1 mM dexamethasone acetate and mifepristone in absolute ethanol (Sigma) [00246] - Saline solution or 0.9% sodium chloride for ZR injection 76/115 Flush (Excelsior Medical, Neptune NJ) [00247] - 0.6 mM sodium caprylate in sterile filtered saline (Sigma) [00248] - 3 mM NAT in sterile filtered saline (Sigma); to prepare, heat to 60Ό for 30 minutes, then sonicate for 5 minutes. [00249] - 10 mM DADKP in sterile filtered saline [00250] - 0.2 μ syringe filters [00251] - HEPES buffered saline, trypsin / EDTA, Trypsin neutralization solution (Lonza reagent pack ) [00252] - 182 cm 2 tissue culture flasks [00253] - Sterile pipettes and tips [00254] Tissue culture hood, humidified CO 2 incubator, water bath [00255] - 24-well tissue culture plates [ 00256] - Qiagen RNeasy plus centrifuge columns (Qiagen 74134) [00257] - Qiagen RT2 qPCR primer pairs for Collagen 2A1, MMP13, TIMP1, agrecana, GAPDH and actin B [00258] Roche Sybr Green I master mix and Invitrogen Superscript VILO master mix [00259] - Roche LightCycler 480 [00260] The following drug treatment solutions were prepared in saline for injection and heated to 37 ° C in a bath: dexamethasone at 4 μΜ mifepristone at 4 μΜ NAT at 3 mM o Caprilate at 0.6 mM o DADKP at 80 μΜ 77/115 o 3 mM NAT mix, 0.6 mM caprylate, 80 μΜ DADKP (Ampion mix [00261] Cyagen Chondrogenic Differentiation medium was prepared using the manufacturer's protocols, but excluding dexamethasone and TGF beta 3 supplements and heated to 37Ό in a bath [00262] The treatment protocol included expansion of HUXMA 5 stem cells in 182 cm2 flasks containing 40 ml of TheraPEAK ™ MSCGM at 80-90% confluence. The cells were trypsinized from the flasks and a suspension 1.0 x 107 HUXMA stem cells were heated in Ciagen Chondrogenic Differentiation medium 20 μΙ of heated cell suspension were placed in the middle of each well of a tissue culture plate with 24 wells (200,000 cells per point) and incubated at 37Ό and 5% CO2 for one hour. [00263] The plates were removed from the incubator and an additional 720 μΙ of chondrogenic medium was added to each well. 250 μΙ of saline or drug solutions were added to the appropriate wells in triplicate (bringing the final drug dilution to a quarter of the concentrations of the drug treatment solution). 10 μΙ of TGF beta 3 solution (supplied by Cyagen) was added to each well and the plates were returned to the incubator. [00264] Media changes were performed every 3-4 days by aspirating the medium from the cavities and replacing with fresh chondrogenic medium, diluted stocks and TGF beta 3, as described above. [00265] RNA isolation and analysis were performed for all plates as follows on days 7, 14 and 22 post-treatment (with the described medium changes). Half of the wells were removed and saved for protein analysis. Qiagen RNeasy plus lysis buffer (with 2-mercaptoethanol) was added to each well and gently stirred for 10 minutes. The cell suspensions subjected to lysis in each well were transferred to Qiashredder columns and 78/115 centrifuged at 14,000 rpm for 2 minutes. RNA isolation was performed by RNeasy ™ plus according to the manufacturer's protocol. The RNA was eluted from the Qiagen columns using 25 μΙ of water-free RNase. [00266] The synthesis of cDNA and PCR in real time were performed as follows. The first cDNA strand synthesis of all samples was performed in a total volume of 20 μΙ using 10 μΙ of isolated RNA. All cDNA reactions were then diluted with 30 μΙ of nuclease-free water. Real-time PCR was then performed using 5 μΙ of diluted cDNA, Roche Syber Green master mix and Qiagen RT2 ™ qPCR primer pairs in a total reaction volume of 20 μΙ. The relative expression of the gene was determined using the delta Ct method. Data / Results Table 10. Results for Day 7 130515 gene expression by RTPCR O OAverage the << 2AACpAverage Standard deviation Sample õ O Actin the < the < vs saline vs saline Regulation Regulation Regulation Collagen 2A1 saline 35.5 28.71 6.79 6,733 35.82 28.35 7.47 34.62 28.68 5.94 Collagen 2A1 Dexamethasone 32.42 25.67 6.75 5,797 0.017 0.9885 1.01 1.44 2.122932.42 27.07 5.35-1.38 2.6087 2.61 33.41 28.12 5.29-1.44 2.7195 2.72 Collagen 2A1 Mifepristone 33.67 26.77 6.9 7.433 0.167 0.8909 1.12 1.82 1.117932.73 24.36 8.371,637 0.3216 3.11 34.82 27.79 7.030.297 0.8141 1.23 Collagen 2A1 Ampion 31.34 27.08 4.26 4.15 -2.47 5.5533 5.55 6.46 3.121933.5 28.73 4.77-1.96 3.8996 3.90 79/115 130515 gene expression by RTPCR O OAverage the << 2AACpAverage Standard deviation Sample õ O Actin the < the < vs saline vs saline Regulation Regulation Regulation31.94 28.52 3.42-3.31 9,9406 9.94 Collagen 2A1 NAT 34.84 27.62 7.22 7.063 0.477 0.7137 1.40 0.61 1.400333.83 27.11 6.72-0.01 1.0093 1.01 34.09 26.84 7.250.517 0.699 1.43 Collagen 2A1 Caprylate 33.77 27.74 6.03 6.573 -0.7 1.6283 1.63 0.18 1.564333.95 27.14 6.810.077 0.9482 1.05 33.94 27.06 6.880.147 0.9033 1.11 Collagen 2A1 DADKP 32.82 26.81 6.01 6,993 -0.72 1,651 1.65 0.62 2.036134.44 26.52 7.921,187 0.4393 2.28 32.87 25.82 7.050.317 0.8029 1.25 Mixture of collagen 2A1 32.57 26.67 5.9 6.6 -0.83 1.7818 1.78 0.18 1.705633.8 26.98 6.820.087 0.9417 1.06 33.45 26.37 7.08 7.08 0.347 0.7864 1.27 MMP13 saline 32.64 28.71 3.93 4.637 33.19 28.35 4.84 33.82 28.68 5.14 MMP13 Dexamethasone 31 25.67 5.33 4,493 0.693 0.6184 1.62 0.45 1.797730.99 27.07 3.92-0.72 1.6434 1.64 32.35 28.12 4.23-0.41 1.3256 1.33 MMP13 Mifepristone 31.98 26.77 5.21 5.7 0.573 0.6721 1.49 2.47 1.825431.19 24.36 6.832,193 0.2186 4.57 32.85 27.79 5.060.423 0.7457 1.34 MMP13 Ampion 30.53 27.08 3.45 3.36 -1.19 2.2763 2.28 2.59 1.187532.69 28.73 3.96-0.68 1.5984 1.60 31.19 28.52 2.67-1.97 3.9086 3.91 80/115 130515 gene expression by RTPCR O OAverage the << 2AACpAverage Standard deviation Sample õ O Actin O< O< vs saline vs saline Regulation Regulation Regulation MMP13NAT 32.77 27.62 5.15 5.523 0.513 0.7006 1.43 1.89 0.517532.6 27.11 5.490.853 0.5535 1.81 32.77 26.84 5.931.293 0.408 2.45 MMP13 Caprilate 33.77 27.74 6.03 5.623 1.393 0.3807 2.63 1.55 2.248331.73 27.14 4.59-0.05 1.0329 1.03 33.31 27.06 6.251,613 0.3268 3.06 MMP13DADKP 31.81 26.81 5 5,017 0.363 0.7774 1.29 0.67 1.489631.12 26.52 4.6-0.04 1.0257 1.03 31.27 25.82 5.450.813 0.5691 1.76 mix MMP13 31.21 26.67 4.54 5.323 -0.1 1.0693 1.07 1.10 1.964732.31 26.98 5.330.693 0.6184 1.62 32.47 26.37 6.11,463 0.3627 2.76 [00267] On day seven, the inventors observed high expression of type 2A1 and MMP13 collagen. All cultures were disk-shaped, except for DADKP-treated wells, which are presented as loose granules. Cell aggregate images were taken on day 8 post-treatment (see Figure 2). 81/115 Table 11. Results for Day 14 130521 RTPCR gene expression O OMean the << 2AACpAverage Standard deviation Sample õ O Actin the < the < vs saline vs saline regulation regulation regulation Collagen 2A1 Saline 29.3 16.51 12.79 11.06 27.99 17.59 10.4 27.35 17.37 9.98 Collagen 2A1 Dexamethasone 22.58 14.39 8.19 8.07 -2.87 7.2938 7.29 8.08 2.025922.58 14.9 7.68-3.38 10,387 10.39 22.85 14.51 8.34-2.72 6.5735 6.57 Collagen 2A1 Mifepristone 27.52 16.74 10.78 10.72 -0.28 1.2114 1.21 1.28 0.255626.77 15.81 10.96-0.1 1.0693 1.07 27.31 16.9 10.41-0.65 1.5655 1.57 Collagen 2A1 Ampion 26.87 16.58 10.29 10.91 -0.77 1.7013 1.70 0.40 2.441626.7 16.58 10.12-0.94 1.9141 1.91 28.7 16.37 12.331,273 0.4137 -2.42 Collagen 2A1 NAT 29.41 16.58 12.83 9.72 1,773 0.2925 -3.42 3.81 6.271625.69 17.46 8.23-2.83 7.0943 7.09 24.51 16.41 8.1-2.96 7.7633 7.76 Collagen 2A1 Caprylate 25.88 17.67 8.21 8,477 -2.85 7.1934 7.19 6.14 1.617624.6 16.34 8.26-2.8 6.9483 6.95 25.75 16.79 8.962.1 4.2772 4.28 Collagen 2A1 DADKP 28.86 18.37 10.49 10.43 -0.57 1.4811 1.48 1.01 1.838727.3 17.6 9.7-1.36 2.5609 2.56 27.64 16.55 11.090.033 0.9772 -1.02 Mixture of collagen 2A1 28.5 16.47 12.03 10.05 0.973 0.5093 -1.96 2.00 3.430724.97 15.93 9.04-2.02 4.0465 4.05 27.33 18.24 9.09 9.09 -1.97 3.9086 3.91 Pellet cellsCells with nodules 82/115 130521 RTPCR gene expression O OMean the << 2AACpAverage Standard deviation Sample õ O Actin the < the < vs saline vs saline regulation regulation regulation S0X9 Saline solution 24.94 16.51 8.43 7.18 24.31 17.59 6.72 23.76 17.37 6.39 S0X9Dexamethasone 22.6 14.39 8.21 7.91 1.03 0.4889 -2.04 1.69 0.411922.39 14.9 7.490.31 0.8066 -1.24 22.54 14.51 8.030.85 0.5548 -1.80 S0X9 Mifepristone 23.61 16.74 6.87 6,927 -0.31 1.2397 1.24 1.19 0.041122.76 15.81 6.95-0.23 1.1728 1.17 23.86 16.9 6.96-0.22 1.1647 1.16 S0X9 Ampion 24.87 16.58 8.29 8.357 1.11 0.4633 -2.16 2.30 0.531625.29 16.58 8.711.53 0.3463 -2.89 24.44 16.37 8.070.89 0.5396 -1.85 S0X9 NAT 24.96 16.58 8.38 6,877 1.2 0.4353 -2.30 0.62 2.526423.56 17.46 6.1-1.08 2,114 2.11 22.56 16.41 6.15-1.03 2,042 2.04 S0X9 Caprilato 24.64 17.67 6.97 6.383 -0.21 1.1567 1.16 1.81 0.583222.53 16.34 6.19-0.99 1.9862 1.99 22.78 16.79 5.99-1.19 2.2815 2.28 S0X9 DADKP 26.22 18.37 7.85 7.323 0.67 0.6285 -1.59 0.19 1.546224.68 17.6 7.08-0.1 1.0718 1.07 23.59 16.55 7.04-0.14 1.1019 1.10 Mixture S0X9 24.4 16.47 7.93 7.053 0.75 0.5946 -1.68 0.43 1.825122.54 15.93 6.61-0.57 1.4845 1.48 24.86 18.24 6.62-0.56 1.4743 1.47 [00268] On the fourteenth day, the inventors observed some cultures that exhibited different phenotypes. Comparison of the wells treated with DADKP with the control in a pellet indicated that the transcription was still high. Additional drug treatments appear to have an effect on collagen transcription. 83/115 Table 12. Results for Day 22 130529 RTPCR gene expression OAverage the << O<<CMAverage Standard deviation Sample O O GAPDH the < the < vs saline vs saline regulation regulation regulation Collagen 2A1 Saline 31.83 18.36 13.47 13.15 32.21 18.54 13.67 30.25 17.95 12.3 Collagen 2A1 Dexamethasone 22.42 16.42 6 5.76 -7.15 141.7 141.70 168.82 27,28621.94 16.41 5.53-7.62 196.27 196.27 21.42 15.67 5.757.4 168.51 168.51 Collagen 2A1 Mifepristone 25.99 17.47 8.52 9.22 -4.63 24,704 24.70 16.82 8.369327.6 17.46 10.14-3.01 8.0371 8.04 26.84 17.84 9-4.15 17,712 17.71 Collagen 2A1 Ampion 21.42 18.45 2.97 2,233 -10.2 1157.4 1157.40 2077.99 938.4321.3 19.15 2.15-11 2043.3 2043.27 20.57 18.99 1.58-11.6 3033.3 3033.29 Collagen 2A1 NAT 24.2 19.45 4.75 5.207 -8.4 337.01 337.01 262.94 105.8623.63 18.76 4.87-8.28 310.12 310.12 24.67 18.67 6-7.15 141.7 141.70 Collagen 2A1 Caprylate 22.93 16.85 6.08 6.557 -7.07 134.05 134.05 101.18 36,37325.6 18.41 7.19-5.96 62,106 62.11 24.26 17.86 6.4-6.75 107.39 107.39 Collagen 2A1 DADKP 27.41 17.52 9.89 7.223 -3.26 9.5577 9.56 136.83 170.1425.44 18.44 7-6.15 70,849 70.85 22.94 18.16 4.78-8.37 330.08 330.08 Mixture of collagen 2A1 20.54 17.93 2.61 4.03 -10.5 1485.4 1485.43 723.78 661.4722.61 17.66 4.95-8.2 293.39 293.39 21.87 17.34 4.53 4.53 -8.62 392.53 392.53 Pellet cellsCells with nodules 84/115 O OAverage the << the <<CMAverage Standard deviation Sample õ O GAPDH the < the < vs saline vs saline regulation regulation regulation MMP13 Saline solution 21.77 18.36 3.41 2.993 21.37 18.54 2.83 20.69 17.95 2.74 MMP13Dexamethasone 20.77 16.42 4.35 4,433 1.357 0.3905 -2.56 -2.73 0.425520.68 16.41 4.271,277 0.4127 -2.42 20.35 15.67 4.681,687 0.3106 -3.22 MMP13 Mifepristone 20.98 17.47 3.51 3,633 0.517 0.699 -1.43 -1.57 0.221221.32 17.46 3.860.867 0.5484 -1.82 21.37 17.84 3.530.537 0.6889 -1.45 MMP13Ampion 21.18 18.45 2.73 2.96 -0.26 1.2002 1.20 0.35 1.516921.82 19.15 2.67-0.32 1.2512 1.25 22.47 18.99 3.480.477 0.7137 -1.40 MMP13NAT 22.68 19.45 3.23 3,053 0.237 0.8487 -1.18 0.29 1.269121.71 18.76 2.95-0.04 1.0305 1.03 21.65 18.67 2.98-0.01 1.0093 1.01 MMP13 Caprilate 20.28 16.85 3.43 2,733 0.437 0.7388 -1.35 0.62 1.789720.31 18.41 1.9-1.09 2.1337 2.13 20.73 17.86 2.87-0.12 1.0892 1.09 MMP13DADKP 20.34 17.52 2.82 2.96 -0.17 1.1277 1.13 0.36 1.237521.41 18.44 2.97-0.02 1.0163 1.02 21.25 18.16 3.090.097 0.9352 -1.07 Mixture MMP13 20.78 17.93 2.85 3,287 -0.14 1.1045 1.10 -0.58 1,46321.23 17.66 3.570.577 0.6705 -1.49 20.78 17.34 3.440.447 0.7337 -1.36 O OAverage the << 2AACpAverage Standard deviation SampleActin the < the < vs saline vs saline regulation regulation regulation Agrecana Saline solution 27.57 16.69 10.88 10.22 26.34 16.4 9.94 25.55 15.72 9.83 Agrecana Dexamethasone 20.48 14.2 6.28 6.523 -3.94 15,313 15.31 13.03 2.007420.68 13.99 6.69-3.53 11,525 11.52 85/115 20.44 13.84 6.6-3.62 12,267 12.27 Agrecana Mifepristona 25.97 15.68 10.29 9,247 0.073 0.9504 -1.05 1.55 2.311124.4 15.42 8.98-1.24 2.3565 2.36 23.94 15.47 8.47-1.75 3.358 3.36 Agrecana Ampion 23.54 16.68 6.86 6.067 -3.36 10,244 10.24 19.16 8.443423.28 17.4 5.88-4.34 20.205 20.21 22.66 17.2 5.46-4.76 27,033 27.03 Agrecana NAT 25.43 17.19 8.24 7.1 -1.98 3.9358 3.94 11.66 10,95223.3 17.68 5.62-4.6 24,195 24.20 24.4 16.96 7.44-2.78 6.8527 6.85 Agrecana Caprilato 21.64 15.53 6.11 7.42 -4.11 17,228 17.23 10.81 8.320925.43 15.71 9.72-0.5 1.4109 1.41 21.95 15.52 6.43-3.79 13.801 13.80 Agrecana DADKP 24.7 15.64 9.06 7.28 -1.16 2.2294 2.23 10.65 8.51123.56 16.73 6.83-3.39 10,459 10.46 22.36 16.41 5.95-4.27 19,248 19.25 Agrecana mix 21.47 16.62 4.85 5,023 -5.37 41.26 41.26 37.33 8.64921.36 15.92 5.44-4.78 27,411 27.41 20.34 15.56 4.78-5.44 43.311 43.31 Pellet cellsCells with nodules O OAverage the << O<<CMAverage Standard deviation Sample TIMP1 Actin the < the < vs saline vs saline regulation regulation regulation TIMP1 Saline solution 21.32 16.69 4.63 3.767 19.91 16.4 3.51 18.88 15.72 3.16 TIMP1 Dexamethasone 17.35 14.2 3.15 3.16 -0.62 1.5333 1.53 1.53 0.205717.35 13.99 3.36-0.41 1.3256 1.33 16.81 13.84 2.97-0.8 1.7371 1.74 TIMP1 Mifepristone 19.69 15.68 4.01 4,227 0.243 0.8448 -1.18 -1.38 0.179219.8 15.42 4.380.613 0.6537 -1.53 86/115 19.76 15.47 4.290.523 0.6958 -1.44 TIMP1 Ampion 19.44 16.68 2.76 2.993 -1.01 2.0093 2.01 1.72 0.266920.42 17.4 3.02-0.75 1.6779 1.68 20.4 17.2 3.2-0.57 1.4811 1.48 TIMP1 NAT 20.59 17.19 3.4 3,537 -0.37 1.2894 1.29 1.18 0.098721.28 17.68 3.6-0.17 1.1225 1.12 20.57 16.96 3.61-0.16 1,1147 1.11 TIMP1 Caprilato 17.65 15.53 2.12 3,143 -1.65 3.1311 3.13 1.12 2.307318.77 15.71 3.06-0.71 1,632 1.63 19.77 15.52 4.250.483 0.7153 -1.40 TIMP1 DADKP 18.64 15.64 3 3.04 -0.77 1.7013 1.70 1.66 0.1319.9 16.73 3.17-0.6 1.5122 1.51 19.36 16.41 2.95-0.82 1.7613 1.76 Mixture TIMP1 19.61 16.62 2.99 3.56 -0.78 1.7132 1.71 0.53 1.575419.51 15.92 3.59-0.18 1.1303 1.13 19.66 15.56 4.10.333 0.7937 -1.26 [00269] On day 22, the inventors observed significantly increased collagen transcription after treatment with DADKP. In addition, drug treatments also showed some activity, but not as pronounced as DADKP. The transcription of collagen 2a1 and agrecan showed an increase of 2000 times and 20 times, respectively, on day 22 after treatment with DADKP and, similarly, 22 days after treatment with Ampion (see Figure 3). Discussion / Conclusion [00270] Treatment with DADKP exhibited an increased transcription of collagen 2A1 and possibly agrecan. Visual observations indicate that the culture architecture can also affect collagen transcription. Example 3 [00271] Following the experiments described in Example 2, a standard procedure for testing the mesenchymal chondrogenesis protocol is developed. This example establishes the standardized chondrogenic testing protocol and expected results. Materials / Equipment: Materials 87/115 [00272] - Human mesenchymal stem cells derived from passage 5 bone marrow (HUXMA - 01001, Cyagen Biosciences, Sunnyvale CA) [00273] - Mesenchymal Stem Cell Chondrogenic Differentiation Medium (GUXMX-90041, Cyagen Biosciences, Sunnyvale CA ) [00274] - TheraPEAK MSCGM chemically defined stem cell medium (190,632 Lonza) [00275] - 1 mM dexamethasone acetate and mifepristone in absolute ethanol [00276] - Saline solution or 0.9% sodium chloride for injection ZR Flush [00277] - 0.6 mM sodium caprylate in sterile filtered saline [00278] - 3 mM NAT in sterile filtered saline; to prepare, heat to 60Ό for 30 minutes, then sleep for 5 minutes. [00279] - 10 mM DADKP in sterile filtered saline solution [00280] - 0.2 μΜ syringe filters [00281] - HEPES buffered saline solution, trypsin / EDTA, Trypsin neutralization solution (Lonza reagent pack ) [00282] - Tissue culture flasks of 75 and 182 cm 2 [00283] - Pipettes and sterile tips [00284] - Hood for tissue culture, humidified incubator with CO2, water bath [00285] - Tissue culture plates with 24 wells [00286] - Centrifuge columns Qiagen RNeasy plus (Qiagen 74134) [00287] - Qiagen RT2 qPCR primer pairs for Collagen 2A1, MMP13, TIMP1, agrecana, GAPDH and actin B 88/115 [00288] - Roche Sybr Green I master mix and Invitrogen Superscript VILO master mix [00289] - Roche LightCycler 480 Procedure / Methodologies Expansion of Cells from Frozen Stocks [00290] - HUXMA cells from passage 4 were cryopreserved using conventional procedures and stored in liquid nitrogen before use. [00291] - Remove a liquid nitrogen storage flask and place in three 75 cm 2 tissue culture flasks containing 15ml of TheraPEAK MSCGM each. [00292] - Incubate the cells at 37Ό and 5% CO2 in a humidified incubator until 60-70% confluence is reached (about 3 days). [00293] - Trypsinize the cells of the flasks using conventional protocol and place the cells from a 75 cm 2 flask to a 182 cm 2 flask containing 40 ml of TheraPEAK MSCGM. [00294] - Incubate the cells until 80-90% of confluence is reached (approximately 4 days). [00295] - Trypsinize the cells again and proceed to the next step. Cell Coating and Treatment [00296] - Prepare the following working dilutions in saline for injection from the stocks listed above and heat to 37Ό in a bath (controls and solutions designed to mimic the final concentrations of some known components of Ampion). dexamethasone at 4 μΜ and mifepristone at NAT at 3 mM caprilate at 0.6 mM o DADKP at 80 μΜ 89/115 o Mixture of NAT at 3 mM, Caprylate at 0.6 mM, DADKP at 80 μΜ [00297] - In addition, heat the solution and stock of Ampion to 37Ό in bath. [00298] - Mix Cyagen Chondrogenic Differentiation medium following the instructions, but EXCLUDE the dexamethasone and TGF beta 3 supplements. Heat to 37Ό in the bath. [00299] - Prepare a cell suspension to 1.0 x 107 HUXMA stem cells in warm chondrogenic medium, then place 20 μΙ in the middle of each well to be used in tissue culture plates with 24 wells (200,000 cells per Score). [00300] - Incubate at 37Ό and 5% CO2 for one hour. [00301] - Remove the plates from the incubator and gently add 720 μΐ of chondrogenic medium to each well. [00302] Add 250 μΐ of saline, Ampion or control solutions to the appropriate wells in triplicate. [00303] - Then, add 10 μΐ of the TGF Beta 3 solution provided by Cyagen to each well. [00304] - Place the plates back in the incubator. [00305] - Media changes were performed every 3-4 days by aspirating the medium from the cavities and replacing it with fresh chondrogenic medium, diluted stocks and TGF beta 3, as described above. Isolation and Analysis of RNA [00306] - The plates were processed as follows on days 7, 14 and 22 post-treatment (with the media changes described). [00307] - Remove the medium from the cavities and save for later analysis of proteins. [00308] - Add Qiagen RNeasy plus lysis buffer (with 2ME) to each well and shake gently for 10 minutes. [00309] - transfer the solution to Qiashredder and centrifuge columns 90/115 gar at 14,000 rpm for 2 minutes. [00310] - Proceed with RNeasy plus protocol according to the manufacturer's recommendations. [00311] - Elute the RNA from columns using 25 μΙ of RNase-free water. Synthesis of cDNA and Real-Time PCR [00312] - The synthesis of the first cDNA strand from all samples was then performed according to the protocol in a total volume of 20 μΐ recommended using 10 μΙ of isolated RNA. [00313] - All cDNA reactions were then diluted with 30 ml of nuclease-free water. [00314] - Real-time PCR, then, was performed using 5 μΙ of diluted cDNA, Roche Syber Green master mix and Qiagen RT2 qPCR primer pairs (total volume 20 μΙ). Expected Results [00315] Treatment of bone marrow mesenchymal / stromal stem cells (MSCs) with DADKP significantly increases chondrogenesis, at least as measured by collagen 2a1 and agrecan transcription. After treating these cells with a composition containing N-acetyl tryptophan (NAT), caprylate and DADKP, an increase in collagen 2a1 transcription of more than 1000 times can be observed and an increase of more than 2000 times can be observed. [00316] Treatment of the same cells with dexamethasone and mifepristone shows a slight increase in chondrogenesis markers, substantially below the 1000-fold increase in collagen 2a1 transcription seen after treatment with DADKP or the composition containing N-acetyl tryptophan, caprylate and DADKP . Similarly, treatment with N-acetyl tryptophan or caprylate also results in a moderate increase in chondrogenesis markers. 91/115 Example 4 [00317] This example demonstrates the effect that Ampion ™ has on the transcription of CXCR4 or CXCL12 by mesenchymal stem cells (MSC) grown in 3D culture. Several groups have shown that CXCR4 transcription and expression is lost when stem cells are cultured. This effect is more pronounced if the cells are grown directly on plastic in a 2D conformation. In this example, the MSCs are grown in suspended droplet cultures in the presence of Ampion ™, and then the mRNA is evaluated using RT-PCR. Materials [00318] - Human mesenchymal stem cells derived from passage 5 bone marrow (HUXMA - 01001, Cyagen Biosciences, Sunnyvale CA) [00319] - Mesenchymal Stem Cell Chondrogenic Differentiation Medium (GUXMX-90041, Cyagen Biosciences, Sunnyvale CA) [ 00320] - TheraPEAK chemically defined stem cell medium MSCGM (190,632 Lonza) [00321] - Lonza MSGM (contains serum) [00322] - 10 mM cobalt chloride in saline, sterile filtrate (Sigma) [00323] - Saline or 0.9% sodium chloride for ZR injection Flush (Excelsior Medical, Neptune NJ) [00324] - 0.2 μ syringe filters [00325] - HEPES buffered saline, trypsin / EDTA, trypsin neutralization solution (Lonza reagent pack) [00326] - 182 cm2 tissue culture flasks [00327] - Pipettes and sterile tips [00328] - Hood for tissue culture, humidified incubator with CO2, water bath [00329] - Petri dishes of 10 cm 2 92/115 [00330] - Qiagen RNeasy plus centrifuge columns (Qiagen 74134) [00331] - Qiagen RT2 qPCR primer pairs for CXCR4, CXCL12, Collagen 2A1, MMP14, MMP13, agrecana, IGA4, GAPDH and actin B [00332] - Master mix Roche Sybr I Green and Master mix Invitrogen Superscript VILO [00333] - Roche LightCycler 480 Cell Expansion [00334] - HUXMA cells from passage 5 were removed from storage in liquid nitrogen and expanded in 182 cm 2 flasks containing 40 ml of TheraPEAK MSCGM up to 80-90% confluence. [00335] - Trypsinize the cells using the conventional protocol and proceed to the next step. Preparation of Droplet Cultures in Suspension [00336] - Prepare the following working dilutions in saline solution for injection from the stocks listed above. [00337] - o Saline solution [00338] - o Cobalt chloride at 800 μΜ in saline solution [00339] - the pure Ampion [00340] - Heat dilutions and Lonza MSCGM to 37Ό in a bath. [00341] - Prepare a suspension of 1.0 X 106 cells of HUXMA stem cells in heated MSCGM medium. [00342] - In a sterile tube, mix 250 μΐ of appropriate working dilution, 250 μΐ of cell suspension (250,000 cells per reaction), 500 μΐ of heated MSCGM. [00343] - Carefully place 40 μΐ points of the resulting solution on the bottom surface of a Petri dish lid (this should result in a total of 40 points). 93/115 [00344] - Place 20 ml of sterile PBS in the lower reservoir of the Petri dish, then invert the cap over the PBS. [00345] - Incubate at 37Ό and 5% CO2. Isolations and RNA Analysis [00346] - After 4 days, remove the plates from the incubator. [00347] - Wash the spheres of the lid with 5 ml of hot PBS at the bottom of each plate and transfer to conical tubes for a 15 ml centrifuge. [00348] - Centrifuge the cells at 1000 RPM for 5 minutes and aspirate the solution. [00349] - Add 350 μΐ of Qiagen Lysis Buffer to each tube RNeasy plus (with 2ME). [00350] - Transfer the solution to Qiashredder columns and centrifuge at 14,000 rpm for 2 minutes. [00351] - Proceed with RNeasy plus protocol according to the manufacturer's recommendations. [00352] - Elute the RNA from columns with 30 μΐ of RNase free of water. Synthesis of cDNA and Real-Time PCR [00353] - The synthesis of the first cDNA strand from all samples was then performed according to the protocol in a total volume of 20 μΐ recommended using 10 μΐ of isolated RNA. [00354] - All cDNA reactions were then diluted with 30 μΐ of nuclease-free water. [00355] - Real-time PCR, then, was performed using 5 μΐ of diluted cDNA, Roche Syber Green master mix and Qiagen RT2 qPCR primer pairs (total volume 20 I). [00356] - Relative gene expression of determined by means of the delta Ct method. [00357] Cobalt treatment seemed to greatly affect the 94/115 GAPDH expression of these cells. The cp calls were shown very early and showed several bands. As a result, actin B was also used to evaluate relative expression using the delta method. The results are shown below in Tables 13 and 14. Table 13. Effect on CXCR4 Normalization for actinCp CpAACp 2-AACpSample cxc Actin ACp vs sun, saline vs sun, saline regulation CXCR4 Saline solution 32.93 18.43 14.25 32.47 18.47 Average 32.7 18.45 CXCR4 Cobalt 29.88 18.6 11.19 -3.065 8.369 8.3729.6 18.51 Average 29.74 18,555 CXCR4 Ampion 007 31.61 20.31 11.43 -2.825 7.086 7.0931.83 20.28 Average 31.72 20,295Standardization for GAPDHCp CpAACp 2-AACpSample cxc GAPDH ACp vs saline Saline Vs regulation CXCR4 Saline solution 32.93 19.27 13.47 32.47 19.19 Average 32.7 19.23 CXCR4 Ampion 007 31.61 20.13 11.59 -1.88 3,681 3.6831.83 20.13 Average 31.72 20.13Average normalization for actin / GAPDHAverage CP Average CPAACp 2-AACpSample CXC Act / Gap ACp vs saline vs saline regulation CXCR4 Saline solution 32.7 18.84 13.86CXCR4 A mpion 007 31.72 20.2125 11.51 -2.353 5.107 5.11 95/115 Table 14. Effect on CXCR12 Normalization for actinCp CpAACp 2-AACpSample CXC Actin ACp vs saline vs saline regulation CXCL12 Saline solution 25.57 18.43 7.13 25.59 18.47 Average 25.58 18.45 CXCL12 Cobalt 33.29 18.6 14.91 7.78 0.005 -219.7933.64 18.51 Average 33.47 18,555 CXCL12 Ampion 007 31.61 20.31 11.43 4,295 0.051 -19.6331.83 20.28 Average 31.72 20,295Standardization for GAPDHCp CpAACp 2-AACpSample CXC GAPDH ACp vs saline vs saline regulation CXCL12 Saline solution 25.57 19.27 6.35 25.59 19.19 Average 25.58 19.23 CXCL12 Ampion 007 31.61 20.13 11.59 5.24 0.026 -37.7931.83 20.13 Average 31.72 20.13Average normalization for actin / GA PDHAverage CP Average CPAACp 2-AACpSample CXC Act / Gap ACp Vs saline solution vs saline regulation CXCL12 saline 25.58 18.84 6.74CXCL12 Ampion 007 31.72 20.2125 11.51 4,768 0.037 -27.24 [00358] These results are also shown in Figures 4 and 5. [00359] The results of this experiment demonstrate that Ampion affects CXCR4 and CXCL12. More particularly, Ampion caused a 3-7-fold increase in CXCR4, while reducing 96/115 CXCL12 in 19-37 times. Example 5 [00360] This example will demonstrate the effect that Ampion ™ has on stem cell migration in vitro. Previous data suggested that the transcription of CXCR4 or CXCL12 was both affected by treatment with Ampion. This experiment will be designed to establish whether the migration of MSCs is altered as well. Materials [00361] - Human mesenchymal stem cells derived from passage 5 bone marrow (HUXMA - 01001, Cyagen Biosciences, Sunnyvale CA) [00362] - TheraPEAK MSCGM chemically defined stem cell medium (190,632 Lonza) [00363] - Lonza MSGM, Gibco RPMI, Gibco defined fetal bovine serum [00364] - 10 mM cobalt chloride in saline, sterile filtrate (Sigma) [00365] Saline or 0.9% sodium chloride for ZR Flush injection (Excelsior Medical , Neptune NJ) [00366] - Filters for 0.2 μΜ syringe. [00367] - HEPES buffered saline, trypsin / EDTA, trypsin neutralization solution (Lonza reagent pack) [00368] - Tissue culture flasks of 25 and 175 cm 2 [00369] - Pipettes and sterile tips [00370 ] - Hood for tissue culture, humidified incubator with CO 2 , water bath [00371] - Thincert tissue culture inserts and 24 well tissue culture plates (Greiner) [00372] - 50 μg Calceina AM bottles (BD Biosciences) 97/115 [00373] - DMSO (Sigma) Cell Expansion [00374] - HUXMA cells from passage 5 were removed from storage in liquid nitrogen and expanded in 175 cm 2 flasks containing 40 ml of TheraPEAK MSCGM up to 80-90% confluence. [00375] - Trypsinize the cells using the conventional protocol and proceed to the next step. Cell Treatment [00376] - Prepare the following working dilutions in saline solution for injection from the stocks listed above: [00377] o Saline solution [00378] o Cobalt chloride at 800 μΜ in saline solution [00379] o Pure Ampion [00380] - Heat dilutions and Lonza MSCGM to 37Ό in a bath. [00381] - Prepare a suspension of stem cell cells HUXMA in heated MSCGM medium. [00382] - Count and then take 250,000 from the cell suspension in 4 ml of heated MSCGM. [00383] - Add the diluted cell solution to the tissue culture flasks of 25 cm 2 . [00384] - Add 1 ml of the appropriate working dilution to the bottle. [00385] - Incubate at 37Ό and 5% CO2 for 72 hours. Invasion Assay [00386] - Remove cells from the flask with trypsin and place in 600 μι of RPMI + 0.5% FBS. [00387] - Add 200 μΐ of the resulting cell suspension to the upper chamber (3 Thinserts inserts per treatment group) over a tissue culture plate with 24 wells. 98/115 [00388] - Add 600 μΐ of RPMI + 0.5% FBS to the lower chamber of the Transwell system. [00389] - Add 66 μΐ of one of the following solutions to the lower chamber for each treatment group. [00390] o 550 ng / ml SDF (final 50 ng / ml) [00391] o 110 ng / ml TGF beta3 (final 10 ng / ml) [00392] serum (final 10%) [00393] Incubate the plate for 24 hours at 37Ό and 5% CO 2 . Cell Marking and Detection [00394] - Remove the two vials of AM calcein and add 20 μΙ of DMSO. [00395] - Transfer the contents of both vials to 12.5 ml of RPMI containing 0.2% BSA. [00396] - Place 450 ml of the AM calcein solution (8 μΜ) in each well of a 24 well tissue culture plate. [00397] - Transfer the inserts from the above to cavities containing AM calcein and incubate 45 minutes at 37Ό and 5% CO 2 . [00398] - After loading the cells with AM calcein, transfer the inserts to tissue culture plates with 24 wells containing 450 μΐ I of EDTA / trypsin and incubate for another 10 minutes. [00399] Loosen the inserts, then add 450 μΐ of trypsin neutralization solution to each well. [00400] - Transfer 250 μΐ of the resulting solution to a flat bottom plate with 96 black wells (each reaction in triplicate). [00401] - Read fluorescence at 485 nm excitation and 530 nm emission. [00402] The results of this experiment are shown below in Table 15 and in Figure 6. 99/115 Table 15. Migration test Assay using pre-treated cells P valueSample Average FU FU Standard deviation FU 1 FU 2 FU 3 vs saline % Increase 10% FBS + saline 265.79 17.95 284.62 248.87 263.89 FBS + Cobalt at 200 μΜ 343.27 12.69 346.49 329.28 354.03 0.005 29.1% FBS + Ampion 324.99 28.29 292.35 342.54 340.07 0.047 22.3% SDF-1 + saline 252.74 36.85 210.41 270.2 277.61 SDF + Cobalt at 200 μΜ 255.86 36.34 215.24 267.07 285.27 0.922 1.2% SDF + Ampion 329.89 40.14 310.92 302.75 376 0.071 30.5% TGF B3 + saline 307.86 21.33 295.48 295.6 332.49 TGF + Cobalt at 200 μΜ 286.77 11.06 274.97 288.44 296.9 0.226 -6.8% TGF + Ampion 354.67 24.38 360.25 327.99 375.78 0.068 16.3% [00403] For all chemotactic signals tested, pretreatment of MSCs with Ampion ™ increased the detectable amount of cells in the lower chamber after 24 hours. Ampion increased the migration of FBS by 22%, SDF-1 by 31% and TGF beta3 by 15%. Example 6 [00404] This example demonstrates that Ampion ™ accelerates chondrogenesis. In addition, Ampion ™ has an additive or synergistic effect on the transcription or translation of genes important to the chondrocyte lineage. Materials [00405] - Human mesenchymal stem cells derived from passage 5 bone marrow (HUXMA - 01001, Cyagen Biosciences, Sunnyvale CA) [00406] - Mesenchymal Stem Cell Chondrogenic Differentiation Medium (GUXMX-90041, Cyagen Biosciences, Sunnyvale CA) [ 00407] - Chemically defined stem cell medium TheraPEAK MSCGM (190,632 Lonza) [00408] - 1 mM dexamethasone and mifepristone in absolute ethanol (Sigma) [00409] - 0.9% saline or sodium chloride for ZR injection 100/115 Flush (Excelsior Medical, Neptune NJ) [00410] - 0.6 mM sodium caprylate in sterile filtered saline (Sigma) [00411] - 3 mM NAT in sterile filtered saline (Sigma); to prepare, heat to 60Ό for 30 minutes, then sonicate for 5 minutes. [00412] - 10 mM DADKP in sterile filtered saline solution [00413] - Filters for 0.2 μΜ syringe HEPES-buffered saline solution, trypsin / EDTA, trypsin neutralization solution (Lonza reagent pack) [00414] - 182 cm 2 tissue culture flasks [00415] - Sterile pipettes and tips [00416] - Hood for tissue culture, humidified incubator with CO2, water bath [00417] - Tissue culture plates with 24 wells [00418] - Centrifuge columns Qiagen RNeasy plus (Qiagen 74134) [00419] - Qiagen RT2 qPCR primer pairs for Collagen 2A1, MMP13, TIMP1, agrecana, GAPDH and actin B [00420] - Master mix Roche Sybr I Green and Master mix Invitrogen Superscript VILO [00421] - Roche LightCycler 480 Cell Expansion [00422] - HUXMA cells from passage 5 were expanded in 175 cm2 flasks containing 40 ml of TheraPEAK MSCGM to 8090% confluence. [00423] - Trypsinize the cells in the flasks using the conventional protocol and proceed to the next step. Cell Coating and Treatment [00424] - Prepare the following working dilutions in solution 101/115 saline for injection from the stocks listed above and heat to 37Ό in a bath (controls and solutions designed to mimic the final concentrations of some known components of Ampion) [00425] 4 μΜ dexamethasone and mifepristone [00426] NAT 3 mM [00427] 0,6 mM caprylate [00428] o 80 μΜ DADKP [00429] o 3 mM NAT mix, 0.6 mM caprylate, 80μΜ DADKP [00430] - In addition, heat the saline solution and 37ion Ampion stock in bath. [00431] - Mix Cyagen Chondrogenic Differentiation medium following the instructions, but EXCLUDE the dexamethasone and TGF beta 3 supplements. Heat to 37Ό in the bath. [00432] - Prepare a cell suspension of 1.0 x 107 HUXMA stem cells in warm chondrogenic medium, then place 20 μΙ in the middle of each well to be used in tissue culture plates with 24 wells (200,000 cells per Score). [00433] - Incubate at 37Ό and 5% CO 2 for one hour. [00434] - Remove the plates from the incubator and gently add 720 μΙ of chondrogenic medium to each well. [00435] - Add 250 μΙ of saline, Ampion or control solutions to the appropriate wells in triplicate. [00436] - Then, add 10 μΙ of the TGF Beta 3 solution provided by Cyagen to each well. [00437] - Place the plates back in the incubator. [00438] - Media changes were carried out every 3-4 days by aspirating the medium from the cavities and replacing it with fresh chondrogenic medium, diluted stocks and TGF beta 3, as described above. 102/115 Isolations and RNA Analysis [00439] - The plates were processed as follows on days 7, 14 and 22 post-treatment (with the media changes described). [00440] - Remove the medium from the cavities and save for later analysis of proteins. [00441] - Add Qiagen RNeasy plus Lysis Buffer (with 2ME) to each well and shake gently for 10 minutes. [00442] - Transfer the solution to Qiashredder columns and centrifuge at 14,000 rpm for 2 minutes. [00443] - Proceed with RNeasy plus protocol according to the manufacturer's recommendations. [00444] - Elute the RNA from columns using 25 μΙ of RNase-free water. Synthesis of cDNA and Real-Time PCR [00445] - The synthesis of the first cDNA strand from all samples was then performed according to the protocol in a total volume of 20 μΙ recommended using 10 μΙ of isolated RNA. [00446] - All cDNA reactions were then diluted with 30μΙ of nuclease-free water. [00447] - Real-time PCR, then, was performed using 5 μΙ of diluted cDNA, Roche Syber Green master mix and Qiagen RT2 qPCR primer pairs (total volume 20 I). [00448] - Relative gene expression determined using the delta Ct method. Results [00449] The results of this experiment on days 7 and 22 are shown below in Tables 15-16 and in Figures 7-9. 103/115 Table 15. Results for Day 7 130515 RTPCR gene expression O OAverage the << O<<CMAverage Standard deviation Sample õ O Actin the < the < vs saline vs saline regulation regulation regulation Collagen 2A1 Saline 35.5 28.71 6.79 6,733 35.82 28.35 7.47 34.62 28.68 5.94 Collagen 2A1 Dexamethasone 32.42 25.67 6.75 5,797 0.017 0.9885 1.01 1.44 2.122932.42 27.07 5.35-1.38 2.6087 2.61 33.41 28.12 5.29-1.44 2.7195 2.72 Collagen 2A1 Mifepristone 33.67 26.77 6.9 7.433 0.167 0.8909 1.12 -1.82 1.117932.73 24.36 8.371,637 0.3216 3.11 34.82 27.79 7.030.297 0.8141 1.23 Collagen 2A1 Ampion 31.34 27.08 4.26 4.15 -2.47 5.5533 5.55 6.46 3.121933.5 28.73 4.77-1.96 3.8996 3.90 31.94 28.52 3.42-3.31 9,9406 9.94 Collagen 2A1 NAT 34.84 27.62 7.22 7.063 0.477 0.7137 1.40 -0.61 1.400333.83 27.11 6.72-0.01 1.0093 1.01 34.09 26.84 7.250.517 0.699 1.43 Collagen 2A1 Caprylate 33.77 27.74 6.03 6.573 -0.7 1.6283 1.63 -0.18 1.564333.95 27.14 6.810.077 0.9482 1.05 33.94 27.06 6.880.147 0.9033 1.11 Collagen 2A1 DADKP 32.82 26.81 6.01 6,993 -0.72 1,651 1.65 -0.62 2.036134.44 26.52 7.921,187 0.4393 2.28 32.87 25.82 7.050.317 0.8029 1.25 Mixture of collagen 2A1 32.57 26.67 5.9 6.6 -0.83 1.7818 1.78 -0.18 1.705633.8 26.98 6.820.087 0.9417 1.06 104/115 33.45 26.37 7.08 7.08 0.347 0.7864 1.27 Cp OAverage the << the <<CMAverage Standard deviation Sample Col Actin the < the < vs saline vs saline regulation regulation regulation MMP13 Saline solution 32.64 28.71 3.93 4.637 33.19 28.35 4.84 33.82 28.68 5.14 MMP13Dexamethasone 31 25.67 5.33 4,493 0.693 0.6184 1.62 0.45 1.797730.99 27.07 3.92-0.72 1.6434 1.64 32.35 28.12 4.23-0.41 1.3256 1.33 MMP13 Mifepristone 31.98 26.77 5.21 5.7 0.573 0.6721 1.49 -2.47 1.825431.19 24.36 6.832,193 0.2186 4.57 32.85 27.79 5.060.423 0.7457 1.34 MMP13 Ampion 30.53 27.08 3.45 3.36 -1.19 2.2763 2.28 2.59 1.187532.69 28.73 3.96-0.68 1.5984 1.60 31.19 28.52 2.67-1.97 3.9086 3.91 MMP13NAT 32.77 27.62 5.15 5.523 0.513 0.7006 1.43 -1.89 0.517532.6 27.11 5.490.853 0.5535 1.81 32.77 26.84 5.931.293 0.408 2.45 MMP13 Caprilate 33.77 27.74 6.03 5.623 1.393 0.3807 2.63 -1.55 2.248331.73 27.14 4.59-0.05 1.0329 1.03 33.31 27.06 6.251,613 0.3268 3.06 MMP13DADKP 31.81 26.81 5 5,017 0.363 0.7774 1.29 -0.67 1.489631.12 26.52 4.6-0.04 1.0257 1.03 31.27 25.82 5.450.813 0.5691 1.76 MMP13MX 31.21 26.67 4.54 5.323 -0.1 1.0693 1.07 -1.10 1.964732.31 26.98 5.330.693 0.6184 1.62 32.47 26.37 6.11,463 0.3627 2.76 [00450] On day seven, high expression of type 2A1 collagen and MMP13 was observed. All cultures were discoid shaped, 105/115 except Ampion ™ treated wells that were loose granules. Table 16. Results for Day 22 130529RTPCR gene expression O OAverage the << O<<CMAverage Standard deviation Sample õ O GAPDH the < the < vs saline vs saline regulation regulation regulation Collagen 2A1 Saline 31.83 18.36 13.47 13.15 32.21 18.54 13.67 30.25 17.95 12.3 Collagen 2A1 Dexamethasone 22.42 16.42 6 5.76 -7.15 141.7 141.70 168.82 27.28621.94 16.41 5.53-7.62 196.27 196.27 21.42 15.67 5.757.4 168.51 168.51 Collagen 2A1 Mifepristone 25.99 17.47 8.52 9.22 -4.63 24,704 24.70 16.82 8.369327.6 17.46 10.14-3.01 8.0371 8.04 26.84 17.84 9-4.15 17,712 17.71 Collagen 2A1 Ampion 21.42 18.45 2.97 2,233 -10.2 1157.4 1157.40 2077.99 938.4321.3 19.15 2.15-11 2043.3 2043.27 20.57 18.99 1.58-11.6 3033.3 3033.29 Collagen 2A1 NAT 24.2 19.45 4.75 5.207 -8.4 337.01 337.01 262.94 105.8623.63 18.76 4.87-8.28 310.12 310.12 24.67 18.67 6-7.15 141.7 141.70 Collagen 2A1 Caprylate 22.93 16.85 6.08 6.557 -7.07 134.05 134.05 101.18 36.37325.6 18.41 7.19-5.96 62,106 62.11 24.26 17.86 6.4-6.75 107.39 107.39 Collagen 2A1 DADKP 27.41 17.52 9.89 7.223 -3.26 9.5577 9.56 136.83 170.1425.44 18.44 7-6.15 70,849 70.85 22.94 18.16 4.78-8.37 330.08 330.08 Mixture of collagen 2A1 20.54 17.93 2.61 4.03 -10.5 1485.4 1485.43 723.78 661.4722.61 17.66 4.95-8.2 293.39 293.39 21.87 17.34 4.53 4.53 -8.62 392.53 392.53 106/115 cells in pelletscells with nodules O OAverage the << the <<CMAverage Standard deviation Sample õ O GAPDH the < the < vs saline vs saline regulation regulation regulation MMP13 Saline solution 21.77 18.36 3.41 2.993 21.37 18.54 2.83 20.69 17.95 2.74 MMP13 Dexamethasone 20.77 16.42 4.35 4,433 1.357 0.3905 -2.56 -2.73 0.425520.68 16.41 4.271,277 0.4127 -2.42 20.35 15.67 4.681,687 0.3106 -3.22 MMP13 Mifepristone 20.98 17.47 3.51 3,633 0.517 0.699 -1.43 -1.57 0.221221.32 17.46 3.860.867 0.5484 -1.82 21.37 17.84 3.530.537 0.6889 -1.45 MMP13 Ampion 21.18 18.45 2.73 2.96 -0.26 1.2002 1.20 0.35 1.516921.82 19.15 2.67-0.32 1.2512 1.25 22.47 18.99 3.480.477 0.7137 -1.40 MMP13NAT 22.68 19.45 3.23 3,053 0.237 0.8487 -1.18 0.29 1.269121.71 18.76 2.95-0.04 1.0305 1.03 21.65 18.67 2.98-0.01 1.0093 1.01 MMP13 Caprilate 20.28 16.85 3.43 2,733 0.437 0.7388 -1.35 0.62 1.789720.31 18.41 1.9-1.09 2.1337 2.13 20.73 17.86 2.87-0.12 1.0892 1.09 MMP13DADKP 20.34 17.52 2.82 2.96 -0.17 1.1277 1.13 0.36 1.237521.41 18.44 2.97-0.02 1.0163 1.02 21.25 18.16 3.090.097 0.9352 -1.07 MMP13MX 20.78 17.93 2.85 3,287 -0.14 1.1045 1.10 -0.58 1,46321.23 17.66 3.570.577 0.6705 -1.49 20.78 17.34 3.440.447 0.7337 -1.36 107/115 O OAverage the << O<<CMAverage Standard deviation SampleActin the < the < vs saline vs saline regulation regulation regulation Agrecana Saline solution 27.57 16.69 10.88 10.22 26.34 16.4 9.94 25.55 15.72 9.83 Agrecana Dexamethasone 20.48 14.2 6.28 6.523 -3.94 15,313 15.31 13.03 2.007420.68 13.99 6.69-3.53 11,525 11.52 20.44 13.84 6.6-3.62 12,267 12.27 Agrecana Mifepristona 25.97 15.68 10.29 9,247 0.073 0.9504 -1.05 1.55 2.311124.4 15.42 8.98-1.24 2.3565 2.36 23.94 15.47 8.47-1.75 3.358 3.36 Agrecana Ampion 23.54 16.68 6.86 6.067 -3.36 10,244 10.24 19.16 8.443423.28 17.4 5.88-4.34 20.205 20.21 22.66 17.2 5.46-4.76 27,033 27.03 Agrecana NAT 25.43 17.19 8.24 7.1 -1.98 3.9358 3.94 11.66 10,95223.3 17.68 5.62-4.6 24,195 24.20 24.4 16.96 7.44-2.78 6.8527 6.85 Agrecana Caprilato 21.64 15.53 6.11 7.42 -4.11 17,228 17.23 10.81 8.320925.43 15.71 9.72-0.5 1.4109 1.41 21.95 15.52 6.43-3.79 13.801 13.80 Agrecana DADKP 24.7 15.64 9.06 7.28 -1.16 2.2294 2.23 10.65 8.51123.56 16.73 6.83-3.39 10,459 10.46 22.36 16.41 5.95-4.27 19,248 19.25 Agrecana mix 21.47 16.62 4.85 5,023 -5.37 41.26 41.26 37.33 8.64921.36 15.92 5.44-4.78 27,411 27.41 20.34 15.56 4.78-5.44 43.311 43.31 cells in pelletscells with nodules 108/115 O OAverage the << 2AACpAverage Standard deviation Sample TIMP1 Actin the < the < vs saline vs saline regulation Regulation regulation TIMP1 Saline solution 21.32 16.69 4.63 3.767 19.91 16.4 3.51 18.88 15.72 3.16 TIMP1 Dexamethasone 17.35 14.2 3.15 3.16 -0.62 1.5333 1.53 1.53 0.205717.35 13.99 3.36-0.41 1.3256 1.33 16.81 13.84 2.97-0.8 1.7371 1.74 TIMP1 Mifepristone 19.69 15.68 4.01 4,227 0.243 0.8448 -1.18 1.38 0.179219.8 15.42 4.380.613 0.6537 -1.53 19.76 15.47 4.290.523 0.6958 -1.44 TIMP1 Ampion 19.44 16.68 2.76 2.993 -1.01 2.0093 2.01 1.72 0.266920.42 17.4 3.02-0.75 1.6779 1.68 20.4 17.2 3.2-0.57 1.4811 1.48 TIMP1 NAT 20.59 17.19 3.4 3,537 -0.37 1.2894 1.29 1.18 0.098721.28 17.68 3.6-0.17 1.1225 1.12 20.57 16.96 3.61-0.16 1,1147 1.11 TIMP1 Caprilato 17.65 15.53 2.12 3,143 -1.65 3.1311 3.13 1.12 2.307318.77 15.71 3.06-0.71 1,632 1.63 19.77 15.52 4.250.483 0.7153 -1.40 TIMP1 DADKP 18.64 15.64 3 3.04 -0.77 1.7013 1.70 1.66 0.1319.9 16.73 3.17-0.6 1.5122 1.51 19.36 16.41 2.95-0.82 1.7613 1.76 Mixture TIMP1 19.61 16.62 2.99 3.56 -0.78 1.7132 1.71 0.53 1.575419.51 15.92 3.59-0.18 1.1303 1.13 19.66 15.56 4.10.333 0.7937 -1.26 [00451] On Day 22, collagen transcription is greatly increased by Ampion ™. The components also showed some activity, but not as pronounced as Ampion ™. Agrecana is also high. [00452] The results of this example show that Ampion has the ability to increase the transcription of collagen 2A1 and agrecana. Example 7 [00453] The following example is a proteomic analysis of synovial fluid from patients treated with Ampion ™ compared to the fluid 109/115 synovial of patients who received saline. [00454] Synovial fluid was taken from patients' knees in a clinical trial for the use of Ampion ™ to treat knee osteoarthritis at the beginning of the study and 12 weeks after treatment with 10cc Ampion ™ or saline. Baseline up to 12 weeks were compared for both saline and Ampion. [00455] Synovial fluid samples were analyzed by SomaLogic, Inc., of Boulder, Colorado, USA using their patented SOMAscan ™ technology. [00456] The proteins shown below in Table 17 were low in Ampion ™ treated synovial fluid compared to saline treated synovial fluid. [00457] Table 17. Low content proteins in the sample treated with Ampion ™ MAPK-activated protein kinase 3 Beta-adrenergic receptor kinase 1 ADAM metallopeptidase with type I thrombospondin motif MAPK-activated protein kinase 2 C-Src kinase Macrophage remover receptor Nogina Bruton's tyrosine kinase Kinase-3 alpha / beta glycogen synthase Kinase-3 alpha / beta glycogen synthase HSP 90 alpha / beta HSP 90 alpha / beta Phosphoinositide 3-kinase, alpha catalytic subunit Phosphoinositide 3-kinase, alpha catalytic subunit Eukaryotic translation start factor 4A Fibroblast growth factor 17 [00458] The proteins shown below in Table 18 were elevated in the synovial fluid treated with Ampion ™ compared to the synovial fluid treated with saline. 110/115 Table 18. Proteins in high content in the sample treated with Ampion ™ Clusterin (Apolipoprotein J) C1QBP (hyaluronan-binding protein 1) Mamaglobin 2 MCP 1 (CCL 2) Spondin 1 IL 11 CFC 1 (cryptic protein) Angiogenin MMP-3 BSSP4 RSPO2 bFGF Coagulation factor IX CATC (Dipeptidyl peptidase 1) Ck-b-8-1 (variant with MPIF 1 splicing) C1s EMR2 ART DPP 2 SAA TIMP-1 Semaphorin 3A Prothrombin TNFSF 15 (VEGF inhibitor) MIP3b (CCL 19) PTHrP Elafin (elastase inhibitor) NPS-PLA2 Testicana 1 (SPOCK 1) URB IP10 (cxcl 10) IL 8 (cxcl 8) Cystatin C H factor SDF-1 (cxcl 12) PIGR [00459] The proteins shown below in Table 19 were meant 111/115 differently (up or down) in Ampion ™ treated synovial fluid compared to saline treated synovial fluid and are known to influence cartilage and synovial fluid production. Table 19. High proteins in the sample treated with Ampion ™ NameProtein Steering compared to saline Description of Protein Clusterina elevated Known for stimulating the proliferation and stability of different stem cells C1QBP elevated a hyaluronic binding protein and inhibits C1, thereby inhibiting complement-induced apoptosis MAPKAPK3 decreased Modulates polycomb-mediated repression of gene expression (via the Akt pathway), which is necessary for the maintenance of hematopoietic stem cells MCP-1 elevated Recruits macrophages in inflammatory conditions IL-11 elevated Stimulates the production of hematopoietic stem cells and megakaryocyte precursor cells MMP3 elevated A metalloproteinase which also breaks down proteoglycans bFGF elevated Stimulates the growth of mesenchymal cells Nogina decreased Essential for the formation of cartilage in embryos, inhibitor of bone morphogenetic proteins PIK3CA decreased Akt Regulations SAA elevated Plays an important role in HDL metabolism and is positively regulated in many inflammatory diseases TIMP 1 elevated A major inhibitor of MMPs and extracellular tissue matrix PTHrP elevated Regulates epithelial-mesenchymal interactions Elafina elevated Inhibits elastin (protease) [00460] The results of this example suggest that the administration of Ampion ™ negatively regulates the Akt pathways. Akt, also known as protein kinase Β (PKB), is a serine / threonine-specific protein kinase that plays a key role in several cellular processes, such as cell proliferation, transcription and cell migration. Example 8 [00461] This example demonstrates the effect of intra injections 112/115 joints of a low molecular weight fraction of 5% human serum albumin (LMWF-5A) for the treatment of knee pain due to osteoarthritis. [00462] This was a double-blind, parallel, vehicle-controlled, randomized, multicenter study designed to evaluate the safety and efficacy of two doses of an intra-articular injection of LMWF-5A. Patients with symptomatic knee osteoarthritis were randomized 1: 1: 1: 1 to receive a single intra-articular injection of 4 mL or 10 mL into the knee of LMWF-5A or vehicle control (saline). The primary efficacy endpoint was the difference between treatment groups in pain variation by Western Ontario and McMaster Universities (WOMAC) from baseline over 12 weeks. Safety was examined as the incidence and severity of adverse events (AEs). [00463] A total of 329 patients with knee pain due to OA were randomized 1: 1: 1: 1 through four study arms: 4 mL of LMWF-5A, 4 mL of vehicle control with saline, 10 mL LMWF-5A or 10 mL of vehicle control with saline. The disposition of the patients is shown in Figure 10. [00464] The LMWF-5A initiation material, HSA purchased from Octapharma (Lachen, Switzerland), was subjected to centrifugation / ultrafiltration under sterile conditions and the ultrafiltrate, containing species with a molecular weight below 5000 Da, was separated. The ultrafiltrate contained DA-DKP (about 50-200 mM) and excipients (i.e., sodium caprylate and sodium acetyltryptophanate). The ultrafiltrate was transferred to aseptic filling to produce the sterile medicated product. [00465] The clinical effects of OA treatment were assessed during clinical visits at 6 and 12 weeks and telephone contacts at 2, 4, 8 and 10 weeks, using the Likert 5-point osteomaarthritis Womach 3.1 score, Assessment Global Patient 113/115 (PGA) of disease severity using a Likert score of 5 points and the amount of acetaminophen after intra-articular injection. Acetaminophen was supplied in 500 mg tablets at the start of the study as an emergency medicine and taken as a tablet every 4 hours, as needed. Safety was assessed by recording adverse events (through 24 hours post-dose and in all follow-up contacts), vital signs and physical examination results (baseline, weeks 6 and 12). [00466] LMWF-5A resulted in a statistically significant improvement in pain compared to the control vehicle (20.93 vs 20.72, respectively). An injection volume effect was not observed (p = 0.64). The difference calculated from the control was 20.25 (Cl 95%: 20.08-20.41), p = 0.004. The reduction in pain with LMWF-5A compared to vehicle control was observed as early as 4 weeks (p = 0.03) and persisted until week 12 (p = 0.004). The percentage of pain reduction over time was significantly higher for LMWF5A compared to vehicle control (12 weeks: 42.3% and 31.7%, respectively), as shown in Figure 11. [00467] Patients treated with LMWF-5A demonstrated significant improvements in the following secondary parameters compared to vehicle control: PGA (20.87 vs 20.65, p = 0.01), physical function, (20.78 vs 20 , 64, p = 0.04); resting pain (20.91 vs 20.70, p = 0.004); pain with movement (20.96 vs 20.75, p = 0.01), Table 3. There were no differences in reduced stiffness between treatment groups. There was a downward trend in the number of paracetamol tablets used over the study period for LMWF-5A compared to vehicle control (median (IQR)): 24.0 (0.62) vs 34.0 (5, 85.5), p = 0.09. Improvement in pain in this study is consistent with tissue regeneration caused by the administration of Ampion ™. 114/115 Example 9 [00468] This example is a 20-week extension of the clinical study described in Example 8, demonstrating the effects of LMWF-5A for the treatment of knee pain due to osteoarthritis. [00469] This analysis is a 20-week extension of a multicenter, vehicle-controlled, double-blind randomized study (NCT01839331) that evaluated the efficacy and safety of the low molecular weight fraction of 5% human serum albumin (LMWF-5A ) for the treatment of pain associated with inflammation in symptomatic knee osteoarthritis (OAK). [00470] Ninety-seven patients who received an intraarticular injection of 4 mL of LMWF-5A or control vehicle were followed up for an additional 8 weeks beyond the end of the initial 12-week study. Efficacy measures included changes from baseline in Western Ontario and McMaster Universities Osteoarthritis (WOMAC) pain scores and function. Patients were considered responders if they achieved> 40% improvement in pain and WOMAC function. Differences between treatment groups were assessed using the chi-square test or ANCOVA, adjusted for baseline values. [00471] In a subgroup of patients with moderate to severe OAK (Kellgren-Lawrence grades 3-4; n = 64), there were statistically significant improvements in WOMAC pain scores (mean change from baseline -0.99 vs -0.65) and function (-0.85 vs -0.58) for 20 weeks for patients who received LMWF-5A compared to vehicle control, respectively. Differences associated with treatment-versus-placebo in pain (-0.95 vs -0.77) and function (-0.79 vs 0.63), respectively, were also observed in the population per protocol. At 20 weeks, the percentage of responders to pain in the moderate to severe subgroup was significantly higher for pa 115/115 patients who received LMWF-5A (50%) compared to those who received vehicle control (25%). Similar rates and severity of adverse events were observed in the LMWF-5A and control groups. [00472] This example demonstrates that a single injection of LMWF5A was associated with sustained improvements in knee pain and may be a therapeutic option for patients with moderate to severe OAK. These results demonstrate a significant benefit of LMWF5A for patients with objective evidence of true OAK and high therapeutic need. [00473] The preceding examples of the present invention have been presented for purposes of illustration and description. Furthermore, these examples are not intended to limit the invention to the form described here. Consequently, variations and modifications commensurate with the teachings of the description of the invention and the skill or knowledge of the relevant technique are within the scope of the present invention. The specific modalities described in the examples provided here are intended to explain the best known method for practicing the invention and to allow those skilled in the art to use the invention in these or other modalities and with various modifications required by the particular applications or uses of the present invention. The attached claims are intended to be interpreted as including alternative modalities to the extent permitted by the state of the art.
权利要求:
Claims (16) [1] 1. Use of DA-DKP, characterized by the fact that it is for the preparation of a pharmaceutical composition for use in a method to cause an effect selected from the group consisting of stem cell mobilization, stem cell migration, cell expansion -stem and stem cell differentiation in an individual. [2] 2. Use according to claim 1, characterized by the fact that the pharmaceutical composition further comprises N-acetyl tryptophan, caprylate and / or caprylic acid. [3] 3. Use according to claim 1 or 2, characterized by the fact that the pharmaceutical composition comprises a low molecular weight fraction of human serum albumin, with substantially all of the albumin removed from the fraction. [4] 4. Use, according to claim 3, characterized by the fact that the low molecular weight fraction of human serum albumin is produced by means of filtration. [5] 5. Use according to any one of claims 1 to 4, characterized by the fact that the composition is administered locally to the individual, the administration site being selected from the group consisting of a joint, a surgical site, a site of a segmented skeletal opening or unbroken fracture, a wound, an ulcer and an inflammatory rash. [6] Use according to any one of claims 1 to 4, characterized in that the composition is formulated as part of or an implantable device selected from a sponge, biocompatible polymer, bioerodible polymer, mass, gel, bone matrix, matrix artificial bone, screw, pin, endotracheal tube, stent, contact lens, pacemaker, central IV tube, Foley catheter and intracranial device. Petition 870170016572, of 03/14/2017, p. 6/13 2/4 [7] 7. Use according to any one of claims 1 to 6, characterized by the fact that the administration of the composition increases the production of a compound selected from the group consisting of CXCR4, MMP14, MMP13, agrecana, SDF1, collagen 2A1 and combinations thereof. [8] 8. Use according to any one of claims 1 to 7, characterized by the fact that the administration of the composition decreases the production of a protein selected from the group consisting of CXCL12, MAPK-activated protein kinase 3, beta-adrenergic receptor kinase 1, ADAM metallopeptidase with type I thrombopondin motif , MAPK-activated protein kinase 2, C-Src kinase, Macrophage Remover Receptor, Nogina, Bruton tyrosine kinase, glycogen synthase alpha-beta kinase-3, HSP 90 alpha / beta, phosphoinositide kinase-3, subunit alpha catalytic and eukaryotic translation initiation factor 4A, Fibroblast Growth Factor 17 and combinations thereof. [9] 9. Use according to any one of claims 1 to 8, characterized by the fact that the administration of the composition increases the production of a protein selected from the group consisting of clusterin (Apolipoprotein J), prothrombin, C1QBP (hyaluronan-binding protein 1), TNFSF 15 (VEGF inhibitor), mamaglobin 2, MIP3b (CCL 19), MCP 1 (CCL 2), PTHrP, spondin 1, elafin (elastase inhibitor), IL 11, NPS-PLA2, CFG 1 (cryptic protein), Testicana 1 (SPOCK 1), angiogenin, URB, MMP-3, IP10 (CXCL 10), BSSP 4, IL 8 (CXCL 8), Rspo2, cystatin C, bFGF, Factor H, Coagulation Factor IX, SDF-1 (cxcl 12), CATC (dipeptidyl-peptidase 1), PIGR, Ck-b-8-1 (variant with MPIF splicing 1), C1s, EMR2, ART, DPP 2, SAA, TIMP-1, Semaphorin 3A and combinations thereof. [10] 10. Use according to any one of claims 1 Petition 870170016572, of 03/14/2017, p. 7/13 3/4 to 9, characterized by the fact that the administration of the composition negatively regulates the Akt pathways in the individual. [11] 11. Use of DA-DKP, characterized by the fact that it is for the preparation of a pharmaceutical composition for use in a method of stimulation of chondrogenesis in an individual, in which the pharmaceutical composition comprises DA-DKP and a component selected from the group consisting of in N-acetyl tryptophan, caprylate, caprylic acid and combinations thereof. [12] 12. Use according to claim 11, characterized by the fact that chondrogenesis treats or improves a chondrogenic disease in the mammal. [13] 13. Use according to any one of claims 1 to 10, characterized by the fact that the effect comprises the stimulation of tissue in a subject in need of it, the tissue being selected from the group consisting of tissue from the nervous system , adipose tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, spongy bone tissue, cartilaginous tissue, dutch pancreatic tissue, spleen tissue, thymus tissue, tonsil tissue, tissue of Peyer's plaque, lymph node tissue, thyroid tissue, epidermal tissue, dermal tissue, subcutaneous tissue, cardiac tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, renal tissue, digestive tract tissue, tissue esophagus, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterine tissue, eye tissue, lung tissue, testicular tissue, tissue ovarian, prostate tissue, connective tissue, endocrine tissue, mesentery tissue and combinations thereof. [14] 14. Composition, characterized by the fact that it comprises stem cells in a medium free from supplemented serum Petition 870170016572, of 03/14/2017, p. 8/13 4/4 with the supplement for medium comprising DA-DKP, and the cell culture medium supplemented with said supplement is capable of supporting the expansion of stem cells. [15] 15. Method for providing stem cells to an individual, characterized by the fact that it comprises: (a) stem cell contact with DA-DKP; (b) culture of stem cells under suitable conditions to facilitate the expansion of stem cells; (c) optionally, adding one or more differentiating factors or changing the culture conditions to induce differentiation of cells to form a different type of cell; and (d) introducing the cells into the individual. [16] 16. Invention, characterized by being in any form of its embodiments or in any applicable category of claim, for example, product or process or use encompassed by the matter initially described, revealed or illustrated in the patent application.
类似技术:
公开号 | 公开日 | 专利标题 US11026940B2|2021-06-08|Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same ES2604581T3|2017-03-07|Modulation of the differentiation of cytoblasts and progenitor cells, assays and uses thereof MXPA04011851A|2005-03-31|Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes. KR20210107144A|2021-08-31|Treatment of spinal cord injury and traumatic brain injury using placental stem cells JP2018141027A|2018-09-13|Use of stem cells to prevent neuronal dieback Shimamura et al.2017|Neuro-regeneration therapy using human Muse cells is highly effective in a mouse intracerebral hemorrhage model JP6960120B2|2021-11-05|Liver disease therapeutic agents and methods for treating liver disease US10870830B2|2020-12-22|Method for culturing differentiation-promoting and -sustaining spheroid form of tonsil-derived stem cells US20190314423A1|2019-10-17|Placental mesenchymal stem cells, methods of production, and placental mesenchymal stem cell based therapeutics WO2019092939A1|2019-05-16|Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease Kawashima et al.2020|Engraftment potential of maternal adipose-derived stem cells for fetal transplantation KR101590722B1|2016-02-03|Pharmaceutical composition for treating nervous system disease comprising human blood derived hematosphere AU2012200787B2|2014-12-04|Modulation of stem and progenitor cell differentiation, assays, and uses thereof US20180085405A1|2018-03-29|Treatment of stroke by amniotic fluid derived stem cell conditioned media and products derived thereof KR20140021156A|2014-02-20|Pharmaceutical composition for treating nervous system disease comprising human blood derived hematosphere AU2013295793A1|2015-02-19|Extracts isolated from electroporated ambhibian oocytes and use thereof in treating diseases and disorders
同族专利:
公开号 | 公开日 WO2014145729A2|2014-09-18| CN105101965B|2021-03-09| US9808454B2|2017-11-07| JP2016515533A|2016-05-30| EP2968315A2|2016-01-20| US20140286913A1|2014-09-25| EP2968315B1|2020-06-03| US11026940B2|2021-06-08| CA2906864A1|2014-09-18| NZ712630A|2021-07-30| US20180153880A1|2018-06-07| MX2015010937A|2015-10-29| AU2014232728A1|2015-10-29| SG10201707619RA|2017-10-30| KR20150132508A|2015-11-25| IL240473D0|2015-09-24| AU2014232728B2|2019-02-21| ZA201507291B|2019-04-24| WO2014145729A3|2014-12-24| US20220016110A1|2022-01-20| PH12015502048A1|2016-01-18| EP2968315A4|2016-08-31| EA201500943A1|2016-08-31| HK1214143A1|2016-07-22| SG11201506267XA|2015-09-29| JP6588005B2|2019-10-09| CN105101965A|2015-11-25|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US3941790A|1967-07-03|1976-03-02|National Research Development Corporation|Bis diketopiperazines| FR2092556A5|1970-04-02|1972-01-21|Snam Progetti| GB1353304A|1971-03-30|1974-05-15|Sagami Chem Res|Process for production of diketopiperazine dihydroxamates and intermediate therefor| US4006261A|1973-09-28|1977-02-01|Firmenich S.A.|Flavoring with mixtures of theobromine and cyclic dipeptides| US3928330A|1973-12-19|1975-12-23|Ciba Geigy Corp|Substituted piperazinedione carboxylic acids and metal salts thereof| GB1459488A|1974-03-19|1976-12-22|Wyeth John & Brother Ltd|Piperazinedione derivatives| US4088649A|1975-07-02|1978-05-09|Firmenich S.A.|Process for preparing a diketo-piperazine| JPS5225019A|1975-08-19|1977-02-24|New Zealand Inventions Dev|Production of serum albumin| US4205057A|1978-01-17|1980-05-27|Government Of The United States|Cerebrospinal fluid protein fragments| DE2940654A1|1979-10-06|1981-04-16|Bayer Ag, 5090 Leverkusen|DIMERES KETEN OF 1,2 ,, - TRIAZOL-3-CARBONIC ACID| US4289759A|1980-06-23|1981-09-15|Ortho Pharmaceutical Corporation|Immunoregulatory diketopiperazine compounds| US4331595A|1980-06-23|1982-05-25|Ortho Pharmaceutical Corporation|Immunoregulatory diketopiperazine compounds| JPH0113075Y2|1981-08-31|1989-04-17| JPS5973574A|1982-10-21|1984-04-25|Grelan Pharmaceut Co Ltd|Cyclic dipeptide| US4694061A|1983-10-12|1987-09-15|Ciba-Geigy Corporation|Radiation-sensitive polycondensates, processes for their preparation coated material and its use| US4727018A|1984-05-18|1988-02-23|Eichner Ronald D|Immunoregulation of transplantable tissue| US4806538A|1984-11-02|1989-02-21|Fujisawa Pharmaceutical Co., Ltd.|Piperazine compound as PAF-antagonist| CS254868B1|1985-06-11|1988-02-15|Jiri Vanzura|Method of cyclic dipeptides production| JPS6236331A|1985-08-12|1987-02-17|Kenji Suzuki|Anti-infective agent and immunological activator| US4771056A|1985-08-29|1988-09-13|Roman Rozencwaig|Method of medical treatment with serotonin antagonists| US4661500A|1985-08-29|1987-04-28|Roman Rozencwaig|Method of medical treatment with serotonin antagonists| US5047401A|1985-09-09|1991-09-10|Board Of Regents, The University Of Texas System|Use of dipeptide alkyl esters to treat GVHD| US4694081A|1985-09-23|1987-09-15|Monsanto Company|Process to prepare 2,5-diketopiperazines| PT83613B|1985-10-28|1988-11-21|Lilly Co Eli|Process for the selective chemical removal of a protein amino-terminal residue| JPS61112060A|1985-10-30|1986-05-30|Fujisawa Pharmaceut Co Ltd|Piperazine compound| JPS63290868A|1987-05-22|1988-11-28|Fujisawa Pharmaceut Co Ltd|Diketopiperazine derivative and salts thereof| JPS6413075A|1987-07-03|1989-01-17|Nippon Chemiphar Co|Production of 2, 5-piperazinedione derivative| US5512544A|1987-09-13|1996-04-30|Yeda Research And Development Co. Ltd.|Pharmaceutical compositions comprising an anticytokine| US4992552A|1988-08-31|1991-02-12|Eastman Kodak Company|Process for preparation of amino acids| US5144073A|1988-08-31|1992-09-01|Hubbs John C|Process for preparation of dipeptides| JP2001055340A|1988-10-31|2001-02-27|Welfide Corp|Production of albumin preparation| US5238938A|1989-02-10|1993-08-24|Otsuka Pharmaceutical Co., Ltd.|Indole derivatives| DE3906952A1|1989-03-04|1990-09-06|Boehringer Mannheim Gmbh|) 6 -C 1 8 ) ALKANSULFINYL AND 2 SULPHONE -METHOXYMETHYL-PROPYL) - PHOSPHATES, METHOD FOR PRODUCING THE MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS| ES2070878T3|1989-10-13|1995-06-16|Phobos Nv|PROCEDURE FOR THE CONTINUOUS PRODUCTION OF HIGH MOLECULAR POLYESTER RESIN.| JPH03176478A|1989-12-05|1991-07-31|Snow Brand Milk Prod Co Ltd|New diketopiperazine derivative and immunosuppressive agent containing the same as active ingredient| DK0478729T3|1990-03-15|1996-09-16|Nutrasweet Co|Process for the preparation of aspartame from a diketopiperazine and novel intermediates and derivatives thereof| US6180616B1|1990-05-10|2001-01-30|Atsuo F. Fukunaga|Use of purine receptor agonists to alleviate or normalize physiopathologically excited sensory nerve function| GB9022543D0|1990-10-17|1990-11-28|Wellcome Found|Antibody production| EP0557388A4|1990-11-14|1994-07-06|Univ Company Pty Ltd|Diagnosis and treatment of multiple sclerosis| US5776892A|1990-12-21|1998-07-07|Curative Health Services, Inc.|Anti-inflammatory peptides| JPH04234374A|1990-12-27|1992-08-24|Ajinomoto Co Inc|Production of diketopiperazine derivative| GB9102997D0|1991-02-13|1991-03-27|Pfizer Ltd|Therapeutic agents| US5543503A|1991-03-29|1996-08-06|Genentech Inc.|Antibodies to human IL-8 type A receptor| US5538993A|1991-09-12|1996-07-23|Yissum Research Development Company|Certain tetrahydrocannabinol-7-oic acid derivatives| AU2905092A|1991-10-28|1993-06-07|Cytoven|Pharmaceutical dipeptide compositions and methods of use thereof| JPH05244982A|1991-12-06|1993-09-24|Sumitomo Chem Co Ltd|Humanized b-b10| JPH05163148A|1991-12-18|1993-06-29|Kanebo Ltd|Anti-neoplastic agent| GB9200210D0|1992-01-07|1992-02-26|British Bio Technology|Compounds| US5352461A|1992-03-11|1994-10-04|Pharmaceutical Discovery Corporation|Self assembling diketopiperazine drug delivery system| US5578323A|1992-06-15|1996-11-26|Emisphere Technologies, Inc.|Proteinoid carriers and methods for preparation and use thereof| US6099856A|1992-06-15|2000-08-08|Emisphere Technologies, Inc.|Active agent transport systems| US5418218A|1992-07-10|1995-05-23|The University Of Maryland At Baltimore|Histidyl-proline diketopiperazine a cns-active pharmacologic agent| US5648486A|1992-07-13|1997-07-15|Cytomed, Inc.|Compounds and methods for the treatment of inflammatory and immune disorders| US5358938A|1992-07-13|1994-10-25|Cytomed, Inc.|Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase| US5463083A|1992-07-13|1995-10-31|Cytomed, Inc.|Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders| GB9217331D0|1992-08-14|1992-09-30|Xenova Ltd|Pharmaceutical compounds| WO1994004537A2|1992-08-20|1994-03-03|Cytomed, Inc.|Dual functional anti-inflammatory and immunosuppressive agents| US5434151A|1992-08-24|1995-07-18|Cytomed, Inc.|Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase| US5728553A|1992-09-23|1998-03-17|Delta Biotechnology Limited|High purity albumin and method of producing| EP0674661B1|1992-12-18|2003-07-02|Molecular Rx., Inc.|Assay and treatment for demyelinating diseases such as multiple sclerosis| ES2068742B1|1993-02-11|1995-11-16|Uriach & Cia Sa J|NEW DERIVATIVES OF PIRIDINIO.| WO1994020063A2|1993-03-04|1994-09-15|Cytoven International N.V.|Pharmaceutical tryptophan containing dipeptide compositions and methods of use thereof| US6107050A|1993-05-03|2000-08-22|The United States Of America As Represented By The Department Of Health And Human Services|Diagnostic test for alzheimers disease| US5523226A|1993-05-14|1996-06-04|Biotechnology Research And Development Corp.|Transgenic swine compositions and methods| EP0711168A4|1993-07-23|1998-04-01|Lxr Biotechnology Inc|Methods of treating apoptosis and associated conditions| GB9324872D0|1993-12-03|1994-01-19|Univ Pasteur|Pharmaceutical compounds| IL112627D0|1994-02-14|1995-05-26|Xenova Ltd|Diketopiperazines, their preparation and pharmaceutical or veterinary compositions containing them| JP2921731B2|1994-03-10|1999-07-19|日清製油株式会社|Gelling agents for organic solvents or fats and oils| FR2717484A1|1994-03-16|1995-09-22|Pf Medicament|New pseudo-bis-peptide cpds.| GB9410387D0|1994-05-24|1994-07-13|Xenova Ltd|Pharmaceutical compounds| WO1996000391A1|1994-06-23|1996-01-04|Affymax Technologies N.V.|Methods for the synthesis of diketopiperazines| US5550132A|1994-06-22|1996-08-27|University Of North Carolina|Hydroxyalkylammonium-pyrimidines or purines and nucleoside derivatives, useful as inhibitors of inflammatory cytokines| US5817751A|1994-06-23|1998-10-06|Affymax Technologies N.V.|Method for synthesis of diketopiperazine and diketomorpholine derivatives| US5792776A|1994-06-27|1998-08-11|Cytomed, Inc.,|Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders| EE9600184A|1994-06-27|1997-06-16|Cytomed, Inc.|Compounds and methods for the treatment of cardiovascular, inflammatory and immune diseases| EP0801065A4|1994-07-29|1998-02-25|Nikken Chemicals Co Ltd|1,4-dihydropyridine compound and medicinal composition containing the same| US5561115A|1994-08-10|1996-10-01|Bayer Corporation|Low temperature albumin fractionation using sodium caprylate as a partitioning agent| US5693338A|1994-09-29|1997-12-02|Emisphere Technologies, Inc.|Diketopiperazine-based delivery systems| US6331318B1|1994-09-30|2001-12-18|Emisphere Technologies Inc.|Carbon-substituted diketopiperazine delivery systems| ES2184808T3|1994-10-05|2003-04-16|Cari Loder|TREATMENT OF MULTIPLE SCLEROSIS AND OTHER DEMELINIZING STATES USING LOFEPRAMINE IN COMBINATION WITH L-PHENYLALANINE, THYROSINE OR TRIPTOPHAN AND OPTIONALLY A VITAMIN B12 COMPOSITE.| ES2087038B1|1994-11-07|1997-03-16|Uriach & Cia Sa J|NEW PIPERIDINES WITH ANTAGONIST ACTIVITY OF THE PAF.| CZ282794A3|1994-11-16|1996-04-17|Galena|Pentapeptidic precursors of biologically active cyclic dipeptides| AU691361B2|1994-12-01|1998-05-14|Toyama Chemical Co. Ltd.|Novel 2,3-diketopiperazine derivative or salt thereof| JP3634891B2|1995-04-06|2005-03-30|株式会社海洋バイオテクノロジー研究所|Chitinase inhibitor| US6096871A|1995-04-14|2000-08-01|Genentech, Inc.|Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life| US5750565A|1995-05-25|1998-05-12|Cytomed, Inc.|Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders| US5703093A|1995-05-31|1997-12-30|Cytomed, Inc.|Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders| AU6311996A|1995-07-06|1997-02-05|Zeneca Limited|Peptide inhibitors of fibronectine| US5811241A|1995-09-13|1998-09-22|Cortech, Inc.|Method for preparing and identifying N-substitued 1,4-piperazines and N-substituted 1,4-piperazinediones| US5902790A|1995-10-03|1999-05-11|Cytran, Inc.|Pharmaceutical angiostatic dipeptide compositions and method of use thereof| US6060452A|1996-03-13|2000-05-09|Cytran, Inc.|Analogs of L-Glu-L-Trp having pharmacological activity| US5665714A|1995-12-07|1997-09-09|Clarion Pharmaceuticals Inc.|N-substituted glycerophosphoethanolamines| US6541224B2|1996-03-14|2003-04-01|Human Genome Sciences, Inc.|Tumor necrosis factor delta polypeptides| WO1997038011A1|1996-04-03|1997-10-16|Pepresearch A/S|Non-dendritic backbone peptide carrier| WO1997036888A1|1996-04-03|1997-10-09|Merck & Co., Inc.|Inhibitors of farnesyl-protein transferase| US5919785A|1996-04-03|1999-07-06|Merck & Co., Inc.|Inhibitors of farnesyl-protein transferase| EP0904072A4|1996-04-12|2003-07-30|Peptide Technology Pty Ltd|Methods of treating immunopathologies using polyunsaturated fattyacids| US5990112A|1996-06-18|1999-11-23|Affymax Technologies N.V.|Inhibitors of metalloproteases pharmaceutical compositions comprising same and methods of their use| US5932579A|1996-06-18|1999-08-03|Affymax Technologies N.V.|Collagenase-1 and stromelysin-1 inhibitors, pharmaceutical compositions comprising same and methods of their use| US5985581A|1996-07-25|1999-11-16|The Mclean Hospital Corporation|Use of presenilin-1 for diagnosis of alzheimers disease| AU4720397A|1996-09-09|1998-03-26|Microbial Screening Technologies Pty. Limited|Terpenylated diketopiperazines, | NZ335544A|1996-10-04|2001-08-31|Neuronz Ltd|Use of GPE in form of Gly-Pro-Glu or Gly-Pro or Pro-Glu as a neuromodulator| RU2125728C1|1996-10-10|1999-01-27|Пермская государственная медицинская академия|Method of immunodiagnostics of multiple sclerosis| EP0835660A1|1996-10-14|1998-04-15|Gaston Edmond Filomena Merckx|Products containing methylcobalamin for the treatment of multiple sclerosis or other demyelinating conditions| US6441172B1|1996-11-07|2002-08-27|Torrey Pines Institute For Molecular Studies|Diketodiazacyclic compounds, diazacyclic compounds and combinatorial libraries thereof| US5932112A|1996-11-27|1999-08-03|Browning Transport Management, Inc.|Method and apparatus for killing microorganisms in ship ballast water| RU2112242C1|1996-12-09|1998-05-27|Пермская государственная медицинская академия|Method for diagnosing the cases of multiple sclerosis| JPH10226615A|1997-02-18|1998-08-25|Pola Chem Ind Inc|Composition containing aspartic acid-phenylalanine cyclic dipeptide derivative| JPH10245315A|1997-03-05|1998-09-14|Pola Chem Ind Inc|Composition containing cyclodipeptide derivative| US6306909B1|1997-03-12|2001-10-23|Queen's University At Kingston|Anti-epileptogenic agents| WO1998040748A1|1997-03-14|1998-09-17|Neuromark|Diagnosing neurologic disorders| US20070208087A1|2001-11-02|2007-09-06|Sanders Virginia J|Compounds, compositions and methods for the treatment of inflammatory diseases| US6265535B1|1997-05-30|2001-07-24|The Trustees Of The University Of Pennsylvania|Peptides and peptide analogues designed from binding sites of tumor necrosis factor receptor superfamily and their uses| RU2128840C1|1997-06-16|1999-04-10|Пермская государственная медицинская академия|Method for diagnosing the cases of multiple sclerosis| US6222029B1|1997-08-01|2001-04-24|Genset|5′ ESTs for secreted proteins expressed in brain| US5834032A|1997-08-11|1998-11-10|Song; Moon K.|Compositions and methods for treating diabetes| US7202279B1|1998-02-11|2007-04-10|Georgetown University|Cyclic dipeptides and azetidinone compounds and their use in treating CNS injury and neurodegenerative disorders| EP0939124A3|1998-02-24|2001-03-21|Smithkline Beecham Plc|MGBP1 sequences| AU3370099A|1998-03-31|1999-10-18|Mayo Foundation For Medical Education And Research|Use of platelet activating factor inhibitors to inhibit il-5 induced eosinophil activation or degranulation| AU3467099A|1998-04-03|1999-10-25|Cytran Ltd.|Methods for production of therapeutic cytokines| EP1067951A2|1998-04-03|2001-01-17|Cytran Ltd.|Use of l-glu-l-trp in the treatment of hiv infection| AU767241B2|1998-09-14|2003-11-06|Qiang Xu|Immunosuppressive agents| WO2000020840A1|1998-10-02|2000-04-13|Ischemia Technologies, Inc.|Tests for the rapid evaluation of ischemic states and kits| US6492179B1|1998-10-02|2002-12-10|Ischemia Techologies, Inc.|Test for rapid evaluation of ischemic states and kit| US6461875B1|1998-10-02|2002-10-08|Ischemia Technologies, Inc.|Test for rapid evaluation of ischemic states and kit| AU6279399A|1998-10-02|2000-04-26|Diagnostic Markers, Inc.|Methods and materials for detection and measurement of free radical damage| US6475743B1|1998-10-02|2002-11-05|Ischemia Technologies, Inc.|Marker useful for detection and measurement of free radical damage and method| DE19847690A1|1998-10-15|2000-04-20|Brahms Diagnostica Gmbh|Diagnosing sepsis and severe infections, useful for assessing severity and progress of treatment, by measuring content of peptide prohormone or their derived fragments| US7026322B2|1998-11-12|2006-04-11|Nereus Pharmaceuticals, Inc.|Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds| US6358957B1|1998-11-12|2002-03-19|Nereus Pharmaceuticals, Inc.|Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds| CA2359561A1|1999-01-20|2000-07-27|Shiro Akinaga|Proteasome inhibitors| AU3760400A|1999-03-19|2000-10-09|Vanderbilt University|Methods and reagents for the diagnosis and treatment of multiple sclerosis| US6090780A|1999-04-07|2000-07-18|Chandon Prasad|Histidyl-proline diketopiperazine and method of use| US6689765B2|1999-05-04|2004-02-10|Schering Corporation|Piperazine derivatives useful as CCR5 antagonists| JP2000327575A|1999-05-26|2000-11-28|Teika Seiyaku Kk|Remedy for inflammatiory disease containing diketopiperazine derivative and new diketopiperazine derivative| DE19937721A1|1999-08-10|2001-02-15|Max Planck Gesellschaft|New diketopiperazines| JP2003517468A|1999-11-12|2003-05-27|ワイス|Branched adamantyl and noradamatyl aryl- and aralkylpiperazines having serotonin 5-HT1A activity| US6677473B1|1999-11-19|2004-01-13|Corvas International Inc|Plasminogen activator inhibitor antagonists| US6248363B1|1999-11-23|2001-06-19|Lipocine, Inc.|Solid carriers for improved delivery of active ingredients in pharmaceutical compositions| US20050096323A1|1999-11-30|2005-05-05|Novoscience Pharma Inc.|Diketopiperazine derivatives to inhibit thrombin| EP1260230A4|2000-02-29|2008-08-06|Chugai Pharmaceutical Co Ltd|Preparations stabilized over long time| DE10019879A1|2000-04-20|2001-10-25|Degussa|Production of known and new 2,5-diketopiperazine derivatives useful for the synthesis of bioactive compounds, e.g. cyclo| HU0302358A3|2000-05-09|2007-09-28|Adpharma|Piperazinedione compounds and use of them for producing pharmaceutical compositions| US7288545B2|2000-05-09|2007-10-30|Angiorx Corporation|Piperazinedione compounds| DE10026998A1|2000-05-31|2001-12-13|Fresenius Kabi De Gmbh|Process for the preparation of a cosmetic composition comprising human serum albumin obtained from transgenic non-human mammals| EP1289990A1|2000-06-08|2003-03-12|Lilly Icos LLC|Tetracyclic diketopierazine compounds as pdev inhibitors| AU7693401A|2000-07-18|2002-02-05|Joslin Diabetes Center Inc|Methods of modulating fibrosis| CZ20002680A3|2000-07-21|2002-03-13|Ev®En Ing. Csc. Kasafírek|Cyclic alkylthiopeptides| CZ20002681A3|2000-07-21|2002-03-13|Ev®En Ing. Csc. Kasafírek|Cyclic tyrosine dipeptides| US6967202B2|2000-08-04|2005-11-22|Dmi Biosciences, Inc.|Method of synthesizing diketopiperazines| CA2417960C|2000-08-04|2012-07-10|Dmi Biosciences, Inc.|Method of using diketopiperazines and composition containing them| AU2001297611A1|2000-12-29|2002-08-19|Celltech R And D, Inc.|Pharmaceutical uses and synthesis of diketopiperazines| GB2372740A|2001-01-17|2002-09-04|Xenova Ltd|Diketopiperazines| AU2002225219A1|2001-01-26|2002-08-06|Oxford Glycosciences Ltd|Diagnosis and treatment of multiple sclerosis| EP1389206B1|2001-04-13|2006-09-13|Vertex Pharmaceuticals Incorporated|Inhibitors of c-jun n-terminal kinases and other protein kinases| EP1408986B1|2001-05-08|2008-09-24|Yale University|Proteomimetic compounds and methods| US7368421B2|2001-06-27|2008-05-06|Probiodrug Ag|Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis| AU2002332122A1|2001-10-15|2003-04-28|The Medstar Research Institute|Modulation of akt-dependent response to prevent restenosis| EP2325193A3|2001-11-02|2012-05-02|Insert Therapeutics, Inc.|Methods and compositions for therapeutic use of RNAinterference| US20040063654A1|2001-11-02|2004-04-01|Davis Mark E.|Methods and compositions for therapeutic use of RNA interference| GB0128108D0|2001-11-23|2002-01-16|Astrazeneca Ab|Therapeutic use| WO2003056026A1|2001-12-27|2003-07-10|Ajinomoto Co., Inc.|Process for production of glutamic acid derivatives| AU2002358163A1|2002-01-18|2003-07-30|Unilever Plc|Cosmetic compositions comprising a cyclodipeptide compound| AU2003223491A1|2002-04-05|2003-10-27|Nitromed, Inc.|Nitric oxide donors, compositions and methods of use| ES2204294B2|2002-07-02|2005-02-01|Universidade De Santiago De Compostela|NEW ACTIVE ANTIBIOTICS AGAINST THE ANGUILLARUM VIBRIO AND ITS APPLICATIONS IN CROPS OF FISH, CRUSTACEANS, MOLLUSCS AND OTHER AQUACULTURE ACTIVITIES.| US20040038865A1|2002-08-01|2004-02-26|Mannkind Corporation|Cell transport compositions and uses thereof| US20080260838A1|2003-08-01|2008-10-23|Mannkind Corporation|Glucagon-like peptide 1 pharmaceutical formulations| BRPI0313363A8|2002-08-02|2019-01-02|Beyondspring Pharmaceuticals Inc|dehydrophenylhistines and their analogues, and their synthesis| US7919497B2|2002-08-02|2011-04-05|Nereus Pharmaceuticals, Inc.|Analogs of dehydrophenylahistins and their therapeutic use| KR20050042146A|2002-08-06|2005-05-04|아플라겐 게엠베하|Binding molecules| DE10238144A1|2002-08-15|2004-02-26|Basf Ag|Use of aryl-substituted 3,6-dialkylidene-2,5-piperazinedione derivatives as photostable UV filters in cosmetic or pharmaceutical compositions for protecting human skin or hair against solar radiation| JP4641796B2|2002-09-03|2011-03-02|ジョージタウンユニヴァーシティ|AKT inhibitor, pharmaceutical composition, and use thereof| AT520981T|2002-10-02|2011-09-15|Dmi Biosciences Inc|DIAGNOSIS AND MONITORING OF DISEASES| US7196169B2|2002-10-11|2007-03-27|Queen's University At Kingston|Isolated post-translationally modified mammalian proteins for monitoring and diagnosing muscle damage| JP2006515574A|2002-11-22|2006-06-01|ノボ・ノルデイスク・エー/エス|Compounds used in the treatment of obesity| KR20120101164A|2003-05-15|2012-09-12|디엠아이 바이오사이언시스, 인크|Treatment of t-cell mediated diseases| CN1791420A|2003-05-15|2006-06-21|Dmi生物科学公司|Treatment of T-cell mediated diseases| PL1651620T3|2003-07-30|2012-04-30|Xenon Pharmaceuticals Inc|Piperazine derivatives and their use as therapeutic agents| US8067425B2|2003-09-03|2011-11-29|Neuren Pharmaceuticals Limited|Neuroprotective bicyclic compounds and methods for their use| EP1768986A2|2004-06-29|2007-04-04|Amgen Inc.|Furanopyrimidines| ES2710025T3|2004-08-20|2019-04-22|Prometic Biosciences Ltd|Schemes of isolation and sequential protein purification by affinity chromatography| BRPI0514293A|2004-08-23|2008-06-10|Mannkind Corp|diketopiperazine salts, diketomorpholine salts or diketodioxane salts for drug administration| AU2006216998B2|2005-02-14|2010-12-23|Merck Sharp & Dohme Corp.|Inhibitors of Akt activity| CA2621595A1|2005-09-21|2007-04-12|Surmodics, Inc.|Coatings and articles including natural biodegradable polysaccharides| MX2008010721A|2006-02-22|2008-09-01|Mannkind Corp|A method for improving the pharmaceutic properties of microparticles comprising diketopiperazine and an active agent.| KR101438839B1|2006-04-14|2014-10-02|맨카인드 코포레이션|Glucagon-like peptide 1 (glp-1)pharmaceutical formulations| US20080017576A1|2006-06-15|2008-01-24|Rensselaer Polytechnic Institute|Global model for optimizing crossflow microfiltration and ultrafiltration processes| AR061240A1|2006-06-20|2008-08-13|Lilly Co Eli|COMPOUNDS OF 4-ISOQUINOLIN-PHENOL, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND THEIR USE AS ANTINEOPLASIC AND / OR ANTIVIRAL AGENTS.| CA2657578A1|2006-07-11|2008-01-17|Harkness Pharmaceuticals, Inc.|Methods of treating obesity using satiety factors| US8231929B2|2006-11-09|2012-07-31|Cook Medical Technologies Llc|Medical device coating process| US20100042206A1|2008-03-04|2010-02-18|Icon Medical Corp.|Bioabsorbable coatings for medical devices| KR101618037B1|2007-07-12|2016-05-04|유니버시티 오브 사우스 플로리다|Inhibitors of Akt/PKB with anti-tumor activity| US20090038416A1|2007-08-07|2009-02-12|Aleta Behrman Bonner|System and method for biological sample collection and analyte detection| WO2009032651A1|2007-08-31|2009-03-12|Smithkline Beecham Corporation|Inhibitors of akt activity| NZ583576A|2007-09-25|2012-06-29|Abbott Lab|Octahydropentalene compounds as chemokine receptor antagonists| US20090163936A1|2007-12-21|2009-06-25|Chunlin Yang|Coated Tissue Engineering Scaffold| WO2009114725A2|2008-03-12|2009-09-17|Children's Hospital Medical Center|Mobilization of hematopoietic stem cells| JP5856843B2|2008-05-27|2016-02-10|アンピオ ファーマシューティカルズ,インコーポレイテッド|Pharmaceutical composition using diketopiperazine| US8314106B2|2008-12-29|2012-11-20|Mannkind Corporation|Substituted diketopiperazine analogs for use as drug delivery agents| TWI528982B|2009-03-04|2016-04-11|曼凱公司|An improved dry powder drug delivery system| US8748380B2|2009-10-30|2014-06-10|Novozymes Biopharma Dk A/S|Albumin variants| CN101914490B|2010-08-13|2012-10-10|中国医科大学|Human amnion mesenchymal stem cell serum-free culture medium and culture method thereof| JP5941047B2|2010-09-07|2016-06-29|アンピオ ファーマシューティカルズ,インコーポレイテッド|Drug for treating disease and kit containing the same| US8507496B2|2010-09-07|2013-08-13|Dmi Acquisition Corp.|Treatment of diseases| WO2012045094A1|2010-10-01|2012-04-05|The Trustees Of Columbia University In The City Of New York|Derivation of fibrochondrocytes from progenitor cells| WO2012174472A1|2011-06-17|2012-12-20|Mannkind Corporation|High capacity diketopiperazine microparticles| SG10201608087WA|2011-10-10|2016-11-29|Ampio Pharmaceuticals Inc|Implantable medical devices with increased immune tolerance, and methods for making and implanting| JP6203734B2|2011-10-10|2017-09-27|アンピオ ファーマシューティカルズ,インコーポレイテッド|Treatment of osteoarthritis| KR20140095479A|2011-10-28|2014-08-01|앰피오 파마슈티컬스 인코퍼레이티드|Treatment of Rhinitis| JP3176478U|2012-04-11|2012-06-21|ネイス株式会社|Exercise air trampoline| US20150366932A1|2013-02-01|2015-12-24|Ampio Pharmaceuticals, Inc.|Methods for producing diketopiperazines and compositions containing diketopiperazines| NZ712630A|2013-03-15|2021-07-30|Ampio Pharmaceuticals Inc|Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same| CA2958080A1|2014-08-18|2016-02-25|Ampio Pharmaceuticals, Inc.|Treatment of joint conditions| WO2016209969A1|2015-06-22|2016-12-29|Ampio Pharmaceuticals, Inc.|Use of low molecular weight fractions of human serum albumin in treating diseases|CA2417960C|2000-08-04|2012-07-10|Dmi Biosciences, Inc.|Method of using diketopiperazines and composition containing them| KR20120101164A|2003-05-15|2012-09-12|디엠아이 바이오사이언시스, 인크|Treatment of t-cell mediated diseases| JP5856843B2|2008-05-27|2016-02-10|アンピオ ファーマシューティカルズ,インコーポレイテッド|Pharmaceutical composition using diketopiperazine| US8507496B2|2010-09-07|2013-08-13|Dmi Acquisition Corp.|Treatment of diseases| JP6203734B2|2011-10-10|2017-09-27|アンピオ ファーマシューティカルズ,インコーポレイテッド|Treatment of osteoarthritis| SG10201608087WA|2011-10-10|2016-11-29|Ampio Pharmaceuticals Inc|Implantable medical devices with increased immune tolerance, and methods for making and implanting| KR20140095479A|2011-10-28|2014-08-01|앰피오 파마슈티컬스 인코퍼레이티드|Treatment of Rhinitis| NZ712630A|2013-03-15|2021-07-30|Ampio Pharmaceuticals Inc|Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same| CA2958080A1|2014-08-18|2016-02-25|Ampio Pharmaceuticals, Inc.|Treatment of joint conditions| KR101960592B1|2016-03-09|2019-03-21|울산대학교 산학협력단|Composition for promoting proliferation, differentiation or anti-aging of stem cell using Janus kinase 1 inhibitor| AU2017235446A1|2016-03-16|2018-11-08|Cell Medicine, Inc.|Mesenchymal stem cells with enhanced efficacy| EP3432895B1|2016-03-23|2021-12-29|Centre Hospitalier Régional et Universitaire de Lille|Platelet pellet lysate for use in treating neurological disorders| CN107418934B|2016-05-24|2021-01-01|南开大学|Application of TNFSF15 protein in-vitro amplification of human cord blood hematopoietic stem cells| US10426796B2|2016-06-13|2019-10-01|SMART SURGICAL, Inc.|Compositions for biological systems and methods for preparing and using the same| US10456423B2|2016-06-13|2019-10-29|SMART SURGICAL, Inc.|Compositions for biological systems and methods for preparing and using the same| CN106834223B|2017-04-05|2020-03-27|上海逍鹏生物科技有限公司|Method for inducing differentiation of umbilical cord mesenchymal stem cells into chondrocytes| CN111073857A|2019-12-30|2020-04-28|深圳爱生再生医学科技有限公司|Stem cell biological product for treating arthritis and preparation method and application thereof|
法律状态:
2018-01-23| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]| 2018-02-27| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]| 2019-08-20| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]|Free format text: NOTIFICACAO DE ANUENCIA RELACIONADA COM O ART 229 DA LPI | 2020-02-04| B15I| Others concerning applications: loss of priority|Free format text: PERDA DAS PRIORIDADES US 61/791,623, DE 15.03.2013; US 61/832,713, DE 07.06.2013; US 61/897,449, DE 30.10.2013; US 61/923,314, DE 03.01.2014; US 61/939,625, DE 13.02.2014, POR AUSENCIA DE CUMPRIMENTO DA EXIGENCIA PUBLICADA NA RPI NO 19/11/2019. | 2020-02-11| B08F| Application dismissed because of non-payment of annual fees [chapter 8.6 patent gazette]|Free format text: REFERENTE A 6A ANUIDADE. | 2020-08-04| B08K| Patent lapsed as no evidence of payment of the annual fee has been furnished to inpi [chapter 8.11 patent gazette]|Free format text: REFERENTE AO DESPACHO 8.6 PUBLICADO NA RPI 2562 DE 11/02/2020. | 2021-10-13| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US201361791623P| true| 2013-03-15|2013-03-15| US201361832713P| true| 2013-06-07|2013-06-07| US201361897449P| true| 2013-10-30|2013-10-30| US201461923314P| true| 2014-01-03|2014-01-03| US201461939625P| true| 2014-02-13|2014-02-13| PCT/US2014/030538|WO2014145729A2|2013-03-15|2014-03-17|Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|