WO1996039487A1 - Chemically defined medium for human mesenchymal stem cells - Google Patents

Chemically defined medium for human mesenchymal stem cells Download PDF

Info

Publication number
WO1996039487A1
WO1996039487A1 PCT/US1996/008405 US9608405W WO9639487A1 WO 1996039487 A1 WO1996039487 A1 WO 1996039487A1 US 9608405 W US9608405 W US 9608405W WO 9639487 A1 WO9639487 A1 WO 9639487A1
Authority
WO
WIPO (PCT)
Prior art keywords
insulin
composition
serum
cells
growth factor
Prior art date
Application number
PCT/US1996/008405
Other languages
French (fr)
Inventor
Daniel R. Marshak
James J. Holecek
Original Assignee
Osiris Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Osiris Therapeutics, Inc. filed Critical Osiris Therapeutics, Inc.
Priority to AU59692/96A priority Critical patent/AU5969296A/en
Priority to EP96916987A priority patent/EP0832188A4/en
Priority to JP9501017A priority patent/JPH11506610A/en
Publication of WO1996039487A1 publication Critical patent/WO1996039487A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/36Lipids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/80Neurotransmitters; Neurohormones
    • C12N2501/825Serotonine (5-HT); Melatonine

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

A composition and method for maintaining the viability of human mesenchymal precursor cells in a serum-free environment which composition includes: (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; and (5) at least one amino acid selected from the group consisting of glutamine, arginine and cysteine, and is free of serum. Also, a composition and method for culture expanding human mesenchymal precursor cells in a serum-free environment. This composition further includes a mitogen, particularly a serotonergic agonist. The cells are preferably isolated human mesenchymal stem cells.

Description

CHEMICALLY DEFINED MEDIUM FOR HUMAN MESENCHYMAL STEM CELLS
This invention relates to the field of cell and tissue culture media, particularly to such media which can support viability and/or growth of cells in the absence of serum.
Bone marrow is a complex tissue composed of hematopoietic, endothelial and stromal elements. Marrow stroma consists of a network of cells and matrix which provide the physical scaffold and a suitable chemical environment for hematopoiesis. Included in the stromal cell population are fibroblasts, reticulocytes, adipocytes and oεteogenic cells. It has been postulated that, within the marrow stromal cell population, stem cells exist which have fibroblastic, adipogenic, reticular, osteogenic or chondrogenic potential. These cells are referred to as mesenchymal stem cells (MSCs) . The development of bone and cartilage in diffusion chamber assays following the introduction of whole marrow, suspensions of marrow cells or cultured marrow-derived mesenchymal cells supports the notion that MSCs are osteochondral progenitor cells that can be isolated from, inter alia, bone marrow. Whole marrow and cultured marrow-derived fibroblastic cells have also given rise to bone and cartilage when combined with porous tricalcium phosphate-hydroxyapatite ceramic cubes which were then implanted subcutaneously into syngeneic or immuno- compromised hosts.
Bone formation has been observed when cultured marrow-derived MSCs from human, rat, murine, canine, rabbit and avian donors were used in the ceramic cube assay. Cells from all of these sources were maintained in vi tro in serum-- supplemented medium. Serum plays a critical role in the growth of cells in vi tro by providing components such as amino acids, lipids, growth factors, vitamins, hormones and attachment factors, by acting as a pH buffer and by providing protease inhibitors (Barnes and Sato, Anal . Biochem. , 102 : 270 , 1980; Iscove, In Cell Cul ture Methods for Molecules and Cell Biology (ed. D.W. Barnes et al . ) , pp. 169-185, 1984; and Barnes, BioTechniques , 5:534-541, 1987) . Nevertheless, the high degree of variability of different lots of serum, the relatively poor degree of characterization of serum components and the necessity of laboriously testing serum prior to its purchase makes the replacement of serum by a chemically defined medium highly desirable.
The growth and expansion of these MSC is accomplished using a nutrient medium fortified with fetal bovine serum. Although serum supplementation is useful in that it provides the cells with nutrients, attachment factors, and growth factors necessary for their in vi tro survival (Barnes supra, 1980 S 1987; Iscove, supra, 1984) , the use of serum is problematic in that there is high cost, lot to lot variation, and a costly screen process is required. In addition to the time and cost problems, the use of serum also adds complexity to any studies which investigate the effect peptide growth factors and small molecules have on the differentiation and growth of cells in vi tro. At present there is a chemically defined medium for the growth and expansion of rat marrow derived mesenchymal stem cells (Lennon et al. 1995, submitted) and several for human marrow hematopoietic stem cells (Iscove supra, 1984; Drouet et al . , Br. J. Hematol . , 73:143-147, 1989; and Sandstrom et al . , Biotechnology and Bioengineering , 43:706-733, 1994 but not one that would support the growth of human MSCs.
The invention is based on the discovery by the inventors of a composition of chemically defined components which support viability and/or growth of mesenchymal progenitor cells in the absence of serum.
Thus, in one aspect the invention provides a composition for maintaining the viability of human mesenchymal precursor cells in a serum-free environment which composition comprises
(1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; and (5) glutamine, and which is free of serum. This aspect also contemplates such a composition in aqueous liquid form in combination with at least one human mesenchymal progenitor cell, particularly an isolated human mesenchymal stem cell.
The invention also provides a method for maintaining the viability of human mesenchymal precursor cells in a serum- free environment which method comprises maintaining viabile human mesenchymal precursor cells in a medium which is free of serum and which comprises (1) a minimum essential medium;
(2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; and (5) glutamine, and which is free of serum.
The invention also provides a composition for culture expanding human mesenchymal precursor cells in a serum-free environment which composition comprises (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; (5) glutamine; and (6) a mitogen, and which is free of serum. This aspect also contemplates such a composition in aqueous liquid form in combination with at least one human mesenchymal progenitor cell, particularly an isolated human mesenchymal stem cell.
The invention also provides a method for culture expanding human mesenchymal precursor cells in a serum-free environment which method comprises culturing viabile human mesenchymal precursor cells in a medium which is free of serum and which comprises (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin¬ like growth factor; (5) glutamine; and (6) a mitogen, and which is free of serum.
The invention will now be further described by reference to a brief description of each of the Figures, but in no way are a limitation of the scope of the invention.
Figures 1A and IB each graphically illustrate comparisons of human MSC proliferation when cultured in the Chemically Defined Medium (containing PDGF-/3/8) of the invention or in Complete Medium (containing serum) which were made by a crystal violet dye binding assay.
Figures 2A and 2B graphically illustrate the MSC proliferative response observed when human MSCs were grown for 6 days in the Chemically Defined Medium of the invention with the presence of a range of serotonin concentrations and in the presence or absence of PDGF, respectively.
Figures 3A and 3B are photomicrographs of human MSCs cultured in Complete Medium and the Chemically Defined Medium (CDM) of the invention, respectively. Figures 4A and 4B graphically illustrate the level of osteogenic differentiation, as measured by alkaline phosphatase of hMSCs grown in Chemically Defined Medium or in Complete Medium with and without Osteogenic Supplements (OS) , as follows:
Figure 4A shows that cells grown in Chemically Defined Medium with OS exhibited an increase in alkaline phosphatase activity that was higher than that of cells in CDM without OS and that was comparable to that of cells grown in Complete Medium with OS; and
Figure 4B shows that this observed increase was still apparent when the alkaline phosphatase was normalized with respect to cell number.
The invention will now be described in more detail with respect to numerous embodiments and examples in support thereof.
As used herein the term "minimum essential medium" refers to any serum-free animal cell culture preparation or medium of known composition which will support the viability of human mesenchymal stem cells in vi tro. Examples are any of the Eagle's based media, i.e., Dulbecco's Modified Eagle's Medium (DMEM) ; Iscove'ε Modified Eagle's Medium, alpha Modified Eagle's Medium, and alεo McCoy's 5A and BGJb (Fitton- Jackson Modification) .
As used herein the term "albumin" refers to serum albumin of any mammalian species, but not to other forms of albumin, such as ovalbumin, lactalbumin, or grain or soybean albumins .
As used herein the term "iron source" refers to any species that will release the reduced, ferric, form of iron to the medium, including but not limited to transferrin, FeS04 or ferritin.
As used herein the term "insulin" refers to any of the various insulins that are known. Insulins are divided into three categories according to promptness, duration and intensity of action following subcutaneous administration, i.e., as mentioned above, rapid, intermediate or long-acting. Crystalline regular insulin is prepared by precipitation in the presence of zinc chloride and modified forms have been developed to alter the pattern of activity. Protamine zinc insulin (PZI) is the result of the reaction of insulin and zinc with the basic protein, protamine, to form a protein complex which disεolves and is absorbed more slowly than crystalline regular insulin but is highly reliable for absorption at a steady rate. Isophane is a modified crystalline protamine zinc insulin whose effects are comparable to a mixture of predominantly regular insulin with a lesser portion of protamine zinc insulin. The extended and prompt insulin-zinc suspensions are also contemplated for use in the invention. The insulin can be, for example, of human bovine, ovine or other animal origin or can be a recombinant product.
Human insulin is now widely available as a result of its production by recombinant DNA techniques; in theory it should be slightly less immunogenic than purified porcine insulin, which in turn should be less immunogenic than bovine insulin. Bovine insulin differs from human insulin by three amino acid residues, whereas porcine differs from human insulin by only one amino acid at the carboxyl-terminus of the B chain. However, when highly purified, all three inεulins have a relatively low, but measurable, capacity to stimulate the immune response. Short- or rapid-acting insulins are simply solutions of regular, crystalline zinc insulin (insulin injection) dissolved in a buffer at neutral pH. These have the most rapid onset of action but the shortest duration, i.e., glucose levels reach a low point within 20-30 minutes and return to baseline in about 2-3 hours.
Intermediate-acting insulins are formulated so that they dissolve more gradually when administered subcutaneously; their durations of action are thus longer. The two preparations most frequently used are neutral protamine Hagedorn (NPH) insulin (isophane insulin suspension) and Lente insulin (insulin zinc suspension) . NPH insulin is a suspension of insulin in a complex with zinc and protamine in a phosphate buffer. Lente insulin is a mixture of crystallized (Ultralente) and amorphous (Semilente) insulins in an acetate buffer, which minimizes the solubility of insulin. The preparations have similar pharmacokinetic profiles.
Ultralente insulin (extended insulin zinc suspension) and protamine zinc insulin suεpension are long-acting insulins; they have a very slow onset and a prolonged ("flat") peak of action. These insulins are advocated to provide a low basal concentration of insulin throughout the day.
As used herein the term insulin is also contemplated to encompass insulin analogs. Recent development of insulin that have altered rates of absorption have raised interest. Inεulin with aspartate and glutamate substituted at positions B9 and B27, respectively, crystallizes poorly and has been termed "monomeric insulin" . This insulin is absorbed more rapidly from subcutaneous depots and thus may be useful in meeting postprandial demands. By contrast, other insulin analogs tend to crystallize at the site of injection and are absorbed more slowly. Insulins with enhanced potency have been produced by substitution of aspartate for histidine at position BIO and by modification of the carboxyl-terminal residues of the B chain.
As used herein the terms "somatomedin(s) " or "insulin¬ like growth factor(s) " refer to peptide hormones structurally related to human proinsulin. Insulin-like growth factor-I or SM-C is a basic, 7649-molecular-weight, 70-amino acid, growth hormone (GH) -dependent, single-chain peptide. Insulin-like growth factor-II is a 67-amino acid. 7471-molecular-weight neutral peptide structurally similar to IGF-I, but much less GH dependent. Both peptides demonstrate stimulation of sulfate incorporation into cartilage, insulin-like activity in muscle and adipose tisεue, and mitogenic activity, particularly in fibroblasts. Circulatory SM-C is primarily synthesized by the liver. Receptors for IGF peptides have been detected in most cells. Two types of receptors have been described. Type I receptor shows a high affinity for IGF-I, a somewhat lower affinity for IGF-II, and a low affinity for insulin. Type II receptor has a high affinity for IGF-II, a low to moderate affinity for IGF-I, and no affinity for insulin. Binding of the growth factor to its receptor causes autophosphorylation of receptor tyrosine residues. IGFs regulate the synthetic activity of cells. In cartilage, IGFs stimulate sulfate and leucine uptake into glycosaminoglycans and proline conversion to hydroxyproline. In adipocytes, IGFs show insulin-like activity including stimulation of glycolysis, inhibition of lipolysis, and inhibition of glycogenolysis.
As used herein the term "growth factor" refers to proteins that cause resting cells to undergo cell division and, in some cases, differentiation. Some growth factors are cell type-specific, stimulating division of only those cells with appropriate receptors; other growth factors are more general in their effects. Among the well-studied growth factors are epidermal growth factor (EGF) , nerve growth factor (NGF) , fibroblast growth factor (FGF) , platelet- derived growth factor (PDGF) , erythropoietin, and a family of proteins called lymphokines, which includes interleukins (IL- 1, IL-2, etc.) and interferon γ. There are also extracellular factors that antagonize the effects of growth factors, slowing or preventing cell division; transforming growth factor β (TGF/3) and tumor necrosis factor (TNF) are such factors.
Platelets are the main source of mitogenic activity found in whole blood serum and missing in plasma. The growth factor with this mitogenic activity is platelet-derived grwoth factor (PDGF) . PDGF has a molecular weight of 28,000 to 35,000. It is made up of two chains, which share 60% homology. The A chain has a molecular weight of 17,000. The B chain is sufficient for the mitogenic effect of PDGF.
Platelet-derived growth factor is a mitogen for cells bearing its receptor and acts synergistically with other growth factors as a competence factor. Platelet-derived growth factor nonreciprocally inhibits EGF binding to the EGF receptor, and stimulates the production and release of somatomedin-C (SM-C) -like factor by cultured human fibroblastε and porcine aorta smooth-muscle cells. Therefore, PDGF modulates growth factor synthesis, secretion, and effects at a tisεue level.
A particularly preferred growth factor for use in the hCDM is platelet-derived growth factor, particularly the ββ homodimer of human recombinant platelet-derived growth factor(hrPDGF3/3) . As used herein the term "antioxidant" refers to any substance that slows down the oxidation of hydrocarbons, oils, fats or the like and thus helps to delay or prevent deterioration. Examples include ascorbic acid and its analogues, particularly ascorbic acid-2-phosphate. Other categories of antioxidants include monothiogycerol, pyruvic acid, citric acid, and retinol acetate.
As used herein the term "antibiotic/antimycotic" refers to preparations of antibacterial and antifungal agents that are typically used in human and animal tissue culture.
As used herein the term "essential amino acids" refers to glutamine or commercially available stabilized analogs, such as GlutaMax-I supplement (GIBCO) which can be used in lieu of glutamine.
As used herein the term "lipid" refers to biological lipids. Biological lipids are a chemically diverse group of compounds which are insoluble or only very slightly soluble in water. Their biological functions are equally diverse. Fats and oils are the principal stored forms of energy in many organisms, and phospho-lipids and sterols make up about half the mass of biological membranes. Other lipids, although present in relatively small quantities, play crucial roles as enzyme cofactors, electron carriers, light-absorbing pigments, hydrophobic anchors, emulsifying agents, hormones, and intracellular mesεengers.
As used herein the term "fatty acid" refers to long chain carboxylic acids which generally contain 4-36 carbon atoms and preferably contain at least 12 carbon atoms, most preferably 12 to 22. In some cases this carbon chain is fully saturated and unbranched, while others contain one or more double bonds. They can have saturated, unsaturated, branched or straight chain hydrocarbon chains. A few contain 3-carbon rings or hydroxy1 groups. The compounds are generally not surface active. They are poorly soluble in water and the longer the acid chain, the fewer the double bonds, the lower the solubility in water. The carboxylic acid group is polar and ionized at neutral pH. This accounts for the slight solubility of short-chain acids in water.
Examples of such acids are those ranging from C16 to C22 with up to three unsaturated bonds (also branching) . Examples of saturated straight chain acids are n-dodecanoic acid, n-tetradecanoic acid, n-hexadecanoic acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, montanic acid and melisεic acid. Alεo useful are unsaturated monoolefinic straight chain monocarboxylic acids. Examples of these are oleic acid, palmitoleic, acid gadoleic acid and erucic acid. Also useful are unsaturated (polyolefinic) straight chain monocarboxylic acids. Examples of these are linoleic acid, ricinoleic acid, linolenic acid, arachidonic acid and behenolic acid. Useful branched acids include, for example, diacetyl tartaric acid.
The physical properties of the fatty acids, and of compounds that contain them, are largely determined by the length and degree of unsaturation of the hydrocarbon chain. The nonpolar hydrocarbon chain accounts for the poor solubility of fatty acids in water. The longer the fatty acyl chain and the fewer the double bonds, the lower the solubility in water. The carboxylic acid group is polar (and ionized at neutral pH) , and accounts for the slight solubility of short-chain fatty acids in water.
In mammals, free fatty acids (having a free carboxylate group) circulate in the blood bound to a protein carrier, serum albumin. However, fatty acids are present mostly as carboxylic acid derivatives such as esters or amides. Lacking the charged carboxylate group," these fatty acid derivatives are generally even less soluble in water than are the free carboxylic acids.
The simplest lipids constructed from fatty acids are the triacylglycerols, also referred to as triglycerideε, fats, or neutral fats . Triacylglycerols are composed of three fatty acids each in ester linkage with a single glycerol (Fig. 9- 2) . Those containing the same kind of fatty acid in all three positions are called simply triacylglycerols, and are named after the fatty acid they contain. In most eukaryotic cells, triacylglycerols form a separate phase of microscopic, oily droplets in the aqueous cytosol, serving as depots of metabolic fuel.
As used herein the term "lipoprotein" refers to a conjugated protein formed from a lipid prosthetic group bound to an apolipoprotein. Apolipoproteins are lipid-binding proteins in the blood, responεible for the transport of triacylglycerol , phospholipids, cholesterol, and cholesteryl esterε between organs. Apolipoproteins ("apo" designates the protein in its lipid-free form) combine with various lipids to form several classes of lipoprotein particles, spherical aggregates with hydrophobic lipids at the core and hydrophilic protein side chains and lipid head groups at the surface. Various combinations of lipid and protein produce particles of different densities, ranging from chylomicrons and very low-density lipoproteins (VLDL) to very high-density lipoproteins (VHDL) , which may be separated by ultracentrifugation.
As used herein the term "non-essential amino acids" refers to the remainder of the known naturally-occuring amino acids that participate in physiological processes and are used in protein synthesis.
As used herein the term "mitogen" refers to a substance that stimulates mitosis and/or lymphocyte transformation.
Phospholipids such as lecithin, myoinositol, phosphatidylethanolamine, sphingomyelin and phosphatidyl choline can be added to supplement their availability for cell membrane synthesis. A suitable source is the the product "Ex-Cyte" (Miles, Inc., Naperville, IL) which contains a mixture of fatty acids, phospholipidε, lipoproteins and cholesterol.
Serotonin (5-hydroxytryptamine) is found predominantly in enterochromaffin cells throughout the gastrointestinal tract, in platelets and in parts of the brain. It is known to be a potent neurotransmitter. It stimulates and inhibits nerves and smooth muscles in the cardiovascular, respiratory and gastrointestinal systems. Serotonergic agonists, which mimic the effect of serotonin when interacting with one of its receptor, have been identified and include for example, ipεapirone, gepirone, buεpirone, 1- [2- (4-aminophenyl) -ethyl] - 4- (3-bbyfluromethyl phenyl) piperazine (PADD) and N, N- dipropyl-5-carboxamidotryptamine (DP-5CT) . Furhter, see Hamon et al, in the Neuropharmacology of Seratonin, Whitabker-Azmitia and Perouthka (eds) , Ann. N.Y. Acad. Sci., 600:114, 1990. It has been discovered in accordance with the invention that serotonin and its agonists exert a mitogenic activity on hMSC when included in the Chemically Defined Medium.
Sodium pyruvate can also be added to the formulation to supplement its availability for the Kreb's cycle. Biotin can also be added to the formulation to supplement its availability as an coenzyme important in carboxyl group transfer reactions. Vitamin B12 (cyanocobalamin) or its coenzyme precursor (cobalamin) can also be added to the formulation to supplement its availability as a coenzyme for group transfer and methylation reactions. Trace element mix can also be added to the formulation to provide a supplemental source of metals necessary for electron transport and many metalloenzymes and proteins. Nucleoside mix can alεo be added to the formulation as a supplemental source of purine and pyrimidine bases necessary for DNA and RNA synthesis.
Example 1 Culture Expansion of Human MSCs in a Serum-Free Medium
Isolation and Preparation of Human MSCs
Human bone marrow cells were from marrow aspirated from the iliac crest of normal donors . The donors included both female and male ranging in age from 19 to 52 years .
Human mesenchymal stem cells (MSC) were purified and cultured using a modification of a previously reported method (Haynesworth et al . , 1992a) . Iliac crest marrow (20 to 40 ml) was vortexed to disperse the cells. The marrow was equally divided between three 50 ml centrifuge tubes each containing 20 ml of Dulbecco'ε Modified Eagles Medium (DMEM) supplemented with fetal bovine serum (10% v/v) from selected lots (BioCell) (Complete Medium) . The cells were pelleted by a 5 minute spin at 1200 rpm in a Beckman GS-34 table top centrifuge fitted with a GH 3.8 horizontal rotor (Beckman) . The cell pellets (5.0 ml) were layered over preformed gradients of 70% Percoll. The cells were fractionated by centrifuging the gradients at 460 x g for 15 minutes. The MSCs were isolated from the top 25% of the gradient, transferred to a 50 ml centrifuge tube and rinsed with 30 ml of complete medium. The cells were counted 'using a hemacytometer, after lysing the red blood cells. The cells were plated onto 100 mm tissue culture plates at 1 x 107 cells per dish. The cultures were maintained at 37°C in a humidified atmosphere of 95% air and 5% C02 and the culture medium was changed every 3 to 4 days.
Culturing and Passaging of Human MSC
When the colonies on the plates became large and near confluent the cells were lifted off the plates by incubating the cells with 0.25% trypsin containing 1 mM EDTA (GIBCO) for 5 minutes at 37°C. The trypsin reaction waε quenched by adding bovine calf serum (Hyclone) at half the volume of the cell sample. The cells were replated at a ratio of 1:3. The first passage cultures were subcultured again when the cultures reached 80% to 90% confluence.
The screening of components to be added to the serum free Chemically Defined Medium (CDM) was done using second or third passage cells seeded onto 35 mm dishes at a density of 2.5 x 104 cells in Complete Medium. A seeding density of 3.- x 10* was used for the in vitro osteogenesis assay. The cells were allowed to attach for 24 hours. After the cells had attached, the medium was removed, the cell layer was rinsed with serum free DMEM and the serum free formulation containing the test components was added. Each component was tested in triplicate and repeated a second time with a different donor sample. The medium was changed as usual and the cells examined daily under an Olympus microscope.
Serum-Free Human Chemically Defined Medium (hCDM)
The minimum essential medium for the serum free formulation of this example was Iscove' s modified Dulbecco's Medium (IMDM) (Gibco) . IMDM was supplemented with human serum albumin (Miles) 5 mg/ml, human Ex-Cyte lipoprotein (Miles) 100 μg/ml, saturated human transferrin (Boehringer Mannheim) 2 μg/ml, human recombinant insulin (Boehringer Mannheim) 10 μg/ml, 100 x MEM vitamins' (Sigma) 1.0% MEM essential amino acids (Gibco) 0.89% MEM nonessential amino acids (Gibco) 0.4% sodium pyruvate (Gibco) 1 mM, GlutaMAX-I supplement (Gibco) 1 mM, folic acid (Gibco) 10 μg/ml, Ascorbic acid 2-phosphate (Whako BioProducts) 10 μM, Biotin (Gibco) 1.0 μg/ml, vitamin B12 mix (Gibco) 1.36 μg/ml, trace element mix (Gibco) diluted 500 fold, FeS04 (Sigma) 4 x 10"8M, nucleoside mix (Sigma ribonucleosides , 2 ' - deoxyribonucleosides, uridine, and thymidine) 10 μg/ml, antibiotic/antimycotic (Gibco) 1.0%, and either human recombinant PDGF ββ homodimer (Boehringer Mannheim) 10-20 ng/ml or 5-hydroxytryptamine (Sigma) 10"5 to 10"6M.
Cell Proliferation Assay
The rate of cell proliferation was measured by a colorimetric dye binding asεay uεing crystal violet dye. The method used was a modification of the methods described by Westergen-Thorsson et al . (1991) and Lennon et al . (1995). Since crystal violet is specific for the nucleus of the cell the intensity of staining is proportional to cell number and can be quantified by measuring the absorbance of the extracted dye at 595 nm. For each batch of crystal violet a standard curve was generated using a fixed number of MSCs (5 x 103 to 1 x 105 cells/35 mm dish) . The slope of the line (correlation coefficient = 0.99) was used to calculate the cell number in each the test well. For the proliferation assay, human MSCs were plated into 35 mm plates at 2.5 to 3 x 104 cells per plate. The plates were removed on day 7 from 37° C02 incubator. The cell layer was rinsed 2 times with 1.0 ml of Tyrode's balanced salt solution, and then was fixed for 15 minutes with 1.0 ml of 196 (v/v) glutaraldehyde in Tyrode's. The cell layer was rinsed 2 times with 1.0 ml H20 and allowed to air dry. The cells were stained with 0.1% (v/v) Crystal Violet in H20 for 30 minutes at ambient temperature. The plates were rinsed 3 times with H20 and the bound dye was extracted by adding 2.0 ml of 1.096 (v/v) Triton XlOO in H20 to each plate and gently agitating on a rotating shaker for 4 hourε at room temperature.
The extracted dye (200 μl) was transferred to a well of a 96 well plate and the absorbance measured at 595 nm on an ELISA plate reader (BioRad) using 1.0% Triton XlOO as a blank.
Alkaline Phosphatase Histochemistry
The level of alkaline phosphatase activity of human MSCs was measured using an alkaline phosphatase kit (Sigma) . The plates were removed from a 37° C02 incubator at set time points, rinsed 2 times with 1.0 ml of PBS and fixed for 1 minute with a citrate/acetone solution. The cells were rinsed 2 times with H20 (1.0 ml) before adding 2.0 ml of Fast Violet Solution (0.5 ml of Napthol AS-MX Phosphate alkaline solution in 12 ml of Fast violet solution) to each plate. The plates incubated in the dark at room temperature for 1 hour, the cells were then rinsed 2 times with H20 and air dried before microscopic examination.
Quantification of Alkaline Phosphatase Activity
The cell layer were washed 2 times with Tyrode's (2.0 ml) . Subεtrate, p-nitrophenol phosphate (Sigma 104 tablet 5.0 mg) , dissolved in substrate buffer (50 mM glycine pH 11, 1.0 mM MgCl) (1.0 ml), was added to each plate. The plates were incubated at 37°C for 5 to 20 minutes. The amount of released p-nitrophenol (PNP) was measured at 405 nm in an ELISA plate reader. Enzyme activity was expressed both as PNP released min"1 and PNP min^.lO6 cell"1. Von Kossa Staining of Mineralized Nodules
Plates were rinsed 2 times with Ca and Mg free Phosphate buffered saline (2.0 ml) . The cells were fixed for 1 hour with 10% buffered formalin and then the plates were rinsed 2 times with H20 (2.0 ml) . A 2% (w/v) AgNO in H20 (1.0 ml) and incubate at in the dark for 10 minutes. The plates were rinsed with H20 (3x 2.0 ml) . With the last rinse left on the plates, they were exposed to a bright light for 15 minutes. The plates were rinsed again with H20 and air dried.
Alkaline Phosphatase Activity and Mineralized Nodule Formation by Cultures Grown in Serum Free Medium Supplemented With Dexamethasone
Alkaline phosphatase activity and mineralized nodule formation was determined using medium supplemented with dexamethasone (10"7M) , ascorbic acid-2-phosphate (0.05 mM) , and β-glycerophosphate (10 mM) osteogenic supplements (OS) . Confluent first or εecond passage cells were subcultured into 35 mm dishes at a density of 3.0 x 104 cells per dish. Cells were allowed to attach as usual. The medium was removed and replaced with either serum free medium or serum free medium containing OS. Complete Medium was used as a positive control. On days 4, 8, 12, and 16 the cultures were removed from a 37° C02 incubator and cell proliferation, alkaline phosphatase activity (histochemical and biochemical) and mineralized nodule formation were measured.
Preparation of Human MSC for Ceramic Cube Assay
Cultured MSCs were trypsinized from the plate as described above. The cells were rinssd with serum free medium (2x 10 ml) and counted. The cell density was adjusted to 5 x 106 cell per ml with serum free medium. Porous 3 mm3 ceramic cubes composed of a mixture of 60% tricalcium phosphate and 40% hydroxyapatite (Zimmer Corporation, Warsaw, Indiana) , and prepared as described by Dennis et al . (1992) and Lennon et al . (1995), were added to the cell suspension. A slight negative atmospheric pressure was generated by evacuating 10 ml of air to ensure that the cells would infiltrate into the pores of the cubes . The cubes with cells were incubated at 37°C for 1 to 2 hours prior to implanting into severe combined immunodeficiency (SCID) mice.
Immunocytochemical Staining
Human MSCs (1st to 3rd passage) in Complete Medium were seeded into 35 mm dishes at 30,000 cells per dish and the cells were allowed to attach for 16 hours before switching to the serum free medium. The cells were grown in the test medium for 3 to 4 days and control plates were left in Complete Medium. The cells were rinsed three times with PBS. the cultures were incubated for 1 hour with 100 μl of either SH2 or SH4 hybridoma culture supernatant (Haynesworth et al . , 1992b) . Chick specific SB1 Bruder et al . hybridoma supernatant was used as a negative control. Plates were rinsed with a 0.1% BSA in PBS solution (3x 2.0 ml) and incubated for 45 minuteε with an FITC-conjugated goat antimouεe IgG diluted in 0.1% BSA-PBS. The diεhes were again rinsed with 0.1% BSA-PBS (3x 2.0 ml) . The cell layer was covered with a coverslip after a drop of PPD immunofluorescence mounting medium was added. Immunofluorescence was observed on an Olympus BX50 epi- fluorescence microscope.
RESULTS
Human MSC Proliferation in Serum Free Medium
The growth kinetics of human MSCs in serum free medium was measured using the crystal violet dye binding assay with MSCs grown in Complete Medium as a positive control. When human MSCs were cultured in CDM containing 10 mg/ml of PDGF- ββ , they exhibited growth kinetics sitv.ilar to those cells grown in Complete Medium (Figures 1A and IB) . The proliferation of cells maintained in CDM was generally between 80 to 90% of that observed in the cultures maintained in Complete Medium. The cell proliferation, however, declined slightly at days 10 to 12. Since these cultures were generally between 80 to 90% confluent by day 8 the decrease in cell proliferation observed beyond day 8 was not considered problematic. Passage of cells grown in CDM generally returned the proliferative capacity of these cells in CDM.
Lauder, TINS, 16 : 233 -239, 1993, has suggested that 5- hydroxytrypamine (5HT) may be involved in regulation of morphogenic activities such as cell proliferation and differentiation. To determine if 5HT was able to simulate the proliferation of human MSCs, a dose response evaluation was made using the crystal violet cell proliferation assay. Human MSC were grown in either CDM, with or without PDGF in the presence of increasing concentrations of 5HT for six days. (Figures 2A and 2B respectively) . As illustrated in the figures there is a dose reεponse effect of 5HT on cell number. The optimum concentration for MSC proliferation appears to be between 10"5M to 10"6M of 5HT.
Cell Morphology
In addition to the measuring proliferation of human MSC grown in CDM it was also important tha the cells maintain the correct morphology, one that is similar to that seen in cultures maintained in Complete Medium. Human MSC from first, second and third passages maintained in Complete Medium have the same morphology, most cells having a fibroblastic morphology, with a few polygonal, or rounded cells.
Human MSCs maintained in CDM had a morphology that was similar to that of cells grown in Complete Medium (Figure 3) . The majority of the cells had the same spindle shaped fibroblastic morphology as the cells grown in Complete Medium. There were also some larger rounded cells in the CDM cultures, but these cells were a small minority.
In Vi tro Osteogenesis of Human MSC in SFhDM
Dexamethasone, 3-glycerophosphate and ascorbate 2- phoεphate was added to the CDM (OS) to measure the in vi tro osteogenic potential of human MSCs grown in CDM. Human MSC were grown in CDM + PDGFJ/3 with and without OS for 16 days'. The level of oεteogenic differentiation alkaline phoεphatase activity (histochemically and biochemically) and the degree of mineralized nodule formation were measured at days 4, 8, 12, and 16. The positive control for these asεays was human MSC grown in Complete Medium with and without OS. The cells grown in CDM + OS exhibited an increase in alkaline phosphatase activity that was higher than that of cells in CDM and the same as that of cells grown in complete medium + OS (Figure 4A) . This observed increase was true even when alkaline phoεphaεe activity waε normalized to cell number (Figure 4B) . The hiεtochemical staining mirrored that of the biochemical activity. There was an increase in the number of alkaline phosphatase positive cells in the culture maintained in CDM + OS. This staining pattern was similar to that of cells grown in Complete Medium + OS. On day 16 the alkaline phosphataεe positive cells in both the cultures maintained in CDM + OS and Complete Medium + OS had a rounded and polygonal morphology. The cells maintained in either CDM or Complete Medium on day 16 did not lose their fibroblastic morphology. This was true for both alkaline phosphatase positive and negative cells.
The appearance of Von Kossa positive staining nodules were apparent by day 16 in both the CDM + OS and Complete Medium + OS cultures. These cells had Von Kossa positive nodules beginning on day 12 and increasing in number through day 16. Cells maintained in either CDK or Complete Medium never developed Von Kossa positive nodules.
In Vivo Bone Formation in Ceramic Cubes
The osteochondrogenic potential of human MSC has been demonstrated by their ability to form bone when loaded into ceramic cubes and implanted subcutaneously into Nude mice
(Haynesworth et al . , 1992a) . To determine the osteochondrogenic potential of human MSCs grown in CDM these cells were grown in CDM, loaded into ceramic cubes and implanted into SCID mice. SCID mice were used because there is no immunological responεe to human cells. Human MSCs, 1st or 2nd passage, were subcultured in completed medium onto 100 mm tissue culture plates at a density of 3 x 10s cells per dish. After the cells attached (16 hours; , the cultures were rinsed 2 times with sterile Tyrode's before switching to CDM + PDGF. The MSCs were grown in this medium until the cultures became near confluent. The MSCs were then harvested, loaded into ceramic cubes and implanted subcutaneously into SCID mice. The cubes were harvested at 3 weeks and 6 weeks.
Ceramic cubes loaded with control MSCs or MSCs grown in CDM were negative for bone 3 weeks post implantation. Six weeks poεt implantation cubeε containing either cellε grown in CDM or Complete Medium were positive for bone. After 6 weeks of incubation in SCID mice, bone was observed in 90 to 100% of the ceramic cubes containing control MSC as compared to 85% to 90% of the cubes which contai ed MSCs grown in CDM + PDGF contained bone.
Immunohistochemical Staining of MSC
To further characterize cells grown in CDM these cells were probed for their expression of the SH antigens characteristic of human MSCs (Haynesworth et al. , 1992b) . The immunoreactivity of antibodies SH4 and SH2 to human MSCs from 2nd passage was evident in both cells grown in CDM and Complete Medium. There was no observed difference in reactivity of the SH antibodies to human MSC when maintained in either Complete Medium or CDM. The antibodies and the reactivity was much greater with SH4 than SH2. This was also true for cells maintained in complete medium.

Claims

What Is Claimed Is:
1. A composition for maintaining the viability of human mesenchymal precursor cells in a serum-free environment which composition comprises (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; and (5) glutamine, and which is free of serum.
2. The composition of claim 1 wherein the iron source is transferrin.
3. The composition of claim 1 wherein the insulin-like growth factor is IGF-1 or IGF-2.
4. The composition of claim 1 which further includes a lipid.
5. The composition of claim 1 which further comprises a human mesenchymal progenitor cell.
6. A method for maintaining the viability of human mesenchymal precursor cells in a serum-free environment which method comprises maintaining viabile human mesenchymal precursor cells in a medium which is free of serum and which comprises (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; and (5) glutamine, and which is free of serum.
7. The method of claim 6 wherein the iron source is transferrin.
8. The method of claim 6 wherein the insulin-like growth factor is IGF-1 or IGF-2.
9. The method of claim 6 wherein the composition further includes a lipid.
10. The method of claim 6 which further comprises an antioxidant.
11. The method of claim 10 wherein the antioxidant is ascorbic acid or an analog or derivative thereof.
12. A composition for culture expanding human mesenchymal precursor cells in a serum-free environment which composition comprises (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin¬ like growth factor; (5) glutamine; and (6) a mitogen, and which is free of serum.
13. The composition of claim 12 wherein the iron source is transferrin.
14. The composition of claim 12 wherein the insulin¬ like growth factor is IGF-1 or IGF-2.
15. The composition of claim 12 which further includes a lipid.
16. The composition of claim 12 wherein the mitogen is a growth factor.
17. The composition of claim 16 wherein the growth factor is a platelet-derived growth factor.
18. The composition of claim 12 wherein the mitogen is a serotonergic agonist.
19. The composition of claim 18 wherein the serotonergic agonist is serotonin.
20. The composition of claim 12 which further comprises a human mesenchymal progenitor cell.
21. The composition of claim 20 wherein the human mesenchymal progenitor cell is a human mesenchymal stem cell.
22. A method for culture expanding human mesenchymal precursor cells in a serum-free environment which method compriseε culturing viabile human mesenchymal precursor cells in a medium which is free of serum and which comprises (1) a minimum essential medium; (2) serum albumin; (3) an iron source; (4) insulin or an insulin-like growth factor; (5) glutamine; and (6) a mitogen, and which is free of serum.
23. The method of claim 22 wherein the iron source is transferrin.
24. The method of claim 22 wherein the insulin-like growth factor is IGF-1 or IGF-2.
25. The method of claim 22 wherein the composition further includes a lipid.
26. The method of claim 22 wherein the composition further compriseε an antioxidant.
27. The method of claim 26 wherein the antioxidant is ascorbic acid or an analog or derivative thereof.
28. The method of claim 22 wherein the mitogen is a growth factor.
29. The method of claim 28 wherein the growth factor is a platelet-derived growth factor.
30. The method of claim 22 wherein the mitogen is a serotonergic agonist.
31. The method of claim 30 wherein the serotonergic agonist is serotonin.
PCT/US1996/008405 1995-06-05 1996-06-03 Chemically defined medium for human mesenchymal stem cells WO1996039487A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU59692/96A AU5969296A (en) 1995-06-05 1996-06-03 Chemically defined medium for human mesenchymal stem cells
EP96916987A EP0832188A4 (en) 1995-06-05 1996-06-03 Chemically defined medium for human mesenchymal stem cells
JP9501017A JPH11506610A (en) 1995-06-05 1996-06-03 Synthetic medium for human mesenchymal stem cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/464,599 US5908782A (en) 1995-06-05 1995-06-05 Chemically defined medium for human mesenchymal stem cells
US08/464,599 1995-06-05

Publications (1)

Publication Number Publication Date
WO1996039487A1 true WO1996039487A1 (en) 1996-12-12

Family

ID=23844553

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/008405 WO1996039487A1 (en) 1995-06-05 1996-06-03 Chemically defined medium for human mesenchymal stem cells

Country Status (6)

Country Link
US (1) US5908782A (en)
EP (1) EP0832188A4 (en)
JP (1) JPH11506610A (en)
AU (1) AU5969296A (en)
CA (1) CA2223582A1 (en)
WO (1) WO1996039487A1 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0948255A1 (en) * 1996-12-06 1999-10-13 Osiris Therapeutics, Inc. Improved chondrogenic differentiation of human mesenchymal stem cells
WO2000027996A1 (en) * 1998-11-09 2000-05-18 Consorzio Per La Gestione Del Centro Di Biotecnologie Avanzate Serum free medium for chondrocyte-like cells
US6733746B2 (en) 1996-03-12 2004-05-11 Invitrogen Corporation Hematopoietic cell culture nutrient supplement
WO2004052177A2 (en) 2002-12-05 2004-06-24 Case Western Reserve University Cell-based therapies for ischemia
EP1478734A2 (en) * 2002-01-25 2004-11-24 Genzyme Corporation Serum-free media for chondrocytes and methods of use thereof
EP1629089A1 (en) * 2003-06-03 2006-03-01 Organ Recovery Systems, Inc. Selection and propagation of progenitor cells
WO2008002914A3 (en) * 2006-06-26 2008-04-10 Gambro Bct Inc Method of culturing mesenchymal stem cells
WO2009084662A1 (en) * 2007-12-28 2009-07-09 Fujirebio Inc. Medium for mammalian somatic cells and additive therefor
WO2011069091A1 (en) 2009-12-04 2011-06-09 Boston Biomedical Research Institute, Inc. Method for cloning pluripotent stem cells
WO2011159797A2 (en) 2010-06-15 2011-12-22 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
WO2012006440A2 (en) 2010-07-07 2012-01-12 Cellular Dynamics International, Inc. Endothelial cell production by programming
US8198084B2 (en) 1996-08-30 2012-06-12 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
WO2012109208A2 (en) 2011-02-08 2012-08-16 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
WO2014130770A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
WO2014132137A2 (en) 2013-03-01 2014-09-04 Université De Genève Transgenic cell selection
WO2015164228A1 (en) 2014-04-21 2015-10-29 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
US9623048B2 (en) 2013-02-08 2017-04-18 Shanghai Institutes For Biological Sciences, Chinese Academy Of Sciences Human hepatocyte-like cells and uses thereof
WO2017070337A1 (en) 2015-10-20 2017-04-27 Cellular Dynamics International, Inc. Methods for directed differentiation of pluripotent stem cells to immune cells
WO2017070145A1 (en) 2015-10-19 2017-04-27 Cellular Dynamics International, Inc. Production of virus-receptive pluripotent stem cell (psc)-derived hepatocytes
WO2017079215A1 (en) 2015-11-03 2017-05-11 Glycomimetics, Inc. Methods and compositions for the production of monoclonal antibodies, hematopoietic stem cells, and methods of using the same
WO2018067836A1 (en) 2016-10-05 2018-04-12 Cellular Dynamics International, Inc. Methods for directed differentiation of pluripotent stem cells to hla homozygous immune cells
WO2018067826A1 (en) 2016-10-05 2018-04-12 Cellular Dynamics International, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
WO2018195175A1 (en) 2017-04-18 2018-10-25 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
WO2019186274A2 (en) 2018-03-30 2019-10-03 University Of Geneva Micro rna expression constructs and uses thereof
US10472605B2 (en) 2014-02-14 2019-11-12 National University Of Ireland, Galway Serum-free medium
US10669519B2 (en) 2010-10-08 2020-06-02 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
WO2021240240A1 (en) 2020-05-27 2021-12-02 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
WO2022235586A1 (en) 2021-05-03 2022-11-10 Astellas Institute For Regenerative Medicine Methods of generating mature corneal endothelial cells
WO2022235869A1 (en) 2021-05-07 2022-11-10 Astellas Institute For Regenerative Medicine Methods of generating mature hepatocytes
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
WO2023213983A2 (en) 2022-05-04 2023-11-09 Antion Biosciences Sa Expression construct

Families Citing this family (134)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110091428A1 (en) * 2000-07-31 2011-04-21 New York Medical College Compositions of adult organ stem cells and uses thereof
US7862810B2 (en) * 2000-07-31 2011-01-04 New York Medical College Methods and compositions for the repair and/or regeneration of damaged myocardium
US20020098167A1 (en) * 2000-07-31 2002-07-25 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium
US20050013804A1 (en) * 2000-09-12 2005-01-20 Yukio Kato Method of culturing mesenchymal stem cells
EP1349918B1 (en) 2000-12-06 2014-08-06 Anthrogenesis Corporation Method of collecting placental stem cells
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
CA2856986C (en) 2001-02-14 2019-08-13 Anthrogenesis Corporation Post-partum mammalian placental stem cells for use in the treatment of neurological or renal diseases and disorders
US6506576B2 (en) 2001-03-14 2003-01-14 Board Of Trustees Of The University Of Arkansas Serum-and steroid-free culture media for cerebellar granule neurons
US20070141036A1 (en) * 2002-01-09 2007-06-21 Alberto Gorrochategui Barrueta Composition and procedure for tissue creation, regeneration and repair by a cell-bearing biological implant enriched with platelet concentrate and supplements
WO2003057865A1 (en) * 2002-01-09 2003-07-17 Derbiotek, S.L. Composition and method of creating, regenerating and repairing tissues using a cell-carrying biological implant which is enriched with a platelet concentrate and supplements
WO2003073998A2 (en) * 2002-03-02 2003-09-12 Board Of Regents, The University Of Texas System Local production and/or delivery of anti-cancer agents by stromal cell precursors
AU2003235403A1 (en) * 2002-05-22 2003-12-02 Fushimi Pharmaceutical Co., Ltd. Method of utilizing physiological activity of rare saccharide and compositions containing rare saccharide
CN1717177A (en) 2002-11-26 2006-01-04 人类起源公司 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
DE10311620A1 (en) * 2003-03-17 2004-10-07 Biotissue Technologies Gmbh Cartilage cell culture medium and use thereof
CA2521217C (en) * 2003-04-01 2013-05-14 United States Of America Department Of Veteran's Affairs Stem-cell, precursor cell, or target cell-based treatment of multi-organ failure and renal dysfunction
US20050260748A1 (en) * 2004-02-27 2005-11-24 Michigan State University Adult stem cells and uses thereof
EP1747265B1 (en) * 2004-04-23 2011-04-20 BioE LLC Multi-lineage progenitor cells
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
US20050265980A1 (en) * 2004-05-14 2005-12-01 Becton, Dickinson And Company Cell culture environments for the serum-free expansion of mesenchymal stem cells
WO2006053031A2 (en) * 2004-11-12 2006-05-18 Mayo Foundation For Medical Education And Research Photocrosslinkable poly(caprolactone fumarate)
US20060165667A1 (en) * 2004-12-03 2006-07-27 Case Western Reserve University Novel methods, compositions and devices for inducing neovascularization
US8017395B2 (en) 2004-12-17 2011-09-13 Lifescan, Inc. Seeding cells on porous supports
CA2512667A1 (en) * 2005-01-07 2006-07-07 Takahiro Ochiya Human hepatocyte-like cells and uses thereof
JP5199866B2 (en) * 2005-03-23 2013-05-15 メイヨ フオンデーシヨン フオー メデイカル エジユケーシヨン アンド リサーチ Photocrosslinkable oligo (poly (ethylene glycol) fumarate) hydrogels for cell and drug delivery
WO2006108218A1 (en) * 2005-04-15 2006-10-19 Regenertech Pty Limited Use of neuropeptide y (npy) and agonists and antagonists thereof for tissue regeneration
JP5527968B2 (en) * 2005-04-29 2014-06-25 メイヨ フオンデーシヨン フオー メデイカル エジユケーシヨン アンド リサーチ Hydrophilic / hydrophobic polymer networks based on poly (caprolactone fumarate), poly (ethylene glycol fumarate) and copolymers thereof
JP2008544957A (en) * 2005-05-10 2008-12-11 ユナイテッド ステイツ オブ アメリカ デパートメント オブ ヴェテランズ アフェアーズ Treatment of renal diseases and multiple organ failure by mesenchymal stem cells and mesenchymal stem cell conditioned medium
US20090304650A1 (en) * 2005-05-11 2009-12-10 Scott Edward W Repair of the Bone Marrow Vasculature
JP2008545703A (en) 2005-05-27 2008-12-18 バイアセル インコーポレーティッド Treatment of ischemia using stem cells
AU2006202209B2 (en) * 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
CN101484575B (en) * 2005-06-08 2013-10-02 森托科尔公司 Cellular therapy for ocular degeneration
US20090169523A1 (en) 2005-06-13 2009-07-02 Catherine Verfaillie Hsc self-renewal
WO2007044418A2 (en) * 2005-10-06 2007-04-19 Moscatello David K Cell culture media, kits and methods of use
PE20070771A1 (en) 2005-10-13 2007-08-11 Anthrogenesis Corp IMMUNOMODULATION THROUGH THE USE OF PLACENTA STEM CELLS
EP2471904B1 (en) 2005-12-29 2018-10-24 Celularity, Inc. Placental stem cell populations
CA2633775A1 (en) 2005-12-29 2007-07-12 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
CZ301148B6 (en) * 2006-01-25 2009-11-18 Univerzita Karlova V Praze Method of culturing human mezenchymal stem cells, particularly for facilitating fracture healing process, and bioreactor for carrying out the method
DE602007008627D1 (en) * 2006-02-14 2010-10-07 Genetix Ltd Cell culture medium
US8119123B2 (en) * 2006-02-16 2012-02-21 New York Medical College Compositions comprising vascular and myocyte progenitor cells and methods of their use
DK1993575T3 (en) 2006-03-07 2019-10-14 Geeta Shroff COMPOSITIONS INCLUDING HUMAN EMBYO STAM CELLS AND DERIVATIVES THEREOF, METHODS OF USE AND PROCEDURES FOR MANUFACTURING
CA2646491A1 (en) * 2006-04-17 2007-10-25 Bioe, Inc. Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
CN101490245B (en) * 2006-06-20 2013-01-16 建新公司 Serum-free media and their uses for chondrocyte expansion
NZ597779A (en) 2007-02-12 2013-07-26 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
WO2008129563A2 (en) * 2007-04-23 2008-10-30 Stempeutics Research Private Limited, Human mesenchymal stem cells and preparation thereof
TWM322542U (en) * 2007-05-23 2007-11-21 Universal Scient Ind Co Ltd Testing machine
US9080145B2 (en) * 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
CN101835479A (en) * 2007-07-25 2010-09-15 佰欧益有限公司 Differentiation of multi-lineage progenitor cells to chondrocytes
CN101952415B (en) 2007-07-31 2017-06-27 生命扫描有限公司 The differentiation of human embryo stem cell
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
SI2203176T1 (en) 2007-09-28 2015-04-30 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
AU2008323719B2 (en) * 2007-11-09 2013-04-04 New York Medical College Methods for the repair and/or regeneration of damaged myocardium using variants of hepatocyte growth factor
CN107574142B (en) 2007-11-27 2021-07-06 生命扫描有限公司 Differentiation of human embryonic stem cells
WO2009073518A1 (en) * 2007-11-30 2009-06-11 New York Medical College Methods of isolating non-senescent cardiac stem cells and uses thereof
US8124071B2 (en) * 2007-11-30 2012-02-28 New York Medical College Methods of reducing transplant rejection and cardiac allograft vasculopathy by implanting autologous stem cells
EP2245140A2 (en) * 2007-11-30 2010-11-03 New York Medical College Compositions comprising hdac inhibitors and methods of their use in restoring stem cell function and preventing heart failure
US8512696B2 (en) 2007-11-30 2013-08-20 Autologous, Llc Methods of isolating non-senescent cardiac stem cells and uses thereof
AU2009205886B2 (en) 2008-01-18 2015-08-27 Katholieke Universiteit Leuven Stem cell aggregates and methods for making and using
WO2009105570A2 (en) 2008-02-21 2009-08-27 Centocor Ortho Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US20100015710A1 (en) * 2008-04-25 2010-01-21 Sunghoon Jung Methods and Compositions for Isolating, Maintaining and Serially Expanding Human Mesenchymal Stem Cells
EP2279246A4 (en) * 2008-05-02 2012-03-28 Massachusetts Inst Technology Methods and compositions for modulating immunological tolerance
WO2009143241A2 (en) * 2008-05-21 2009-11-26 Bioe, Inc. Differentiation of multi-lineage progenitor cells to pancreatic cells
BRPI0913925A2 (en) * 2008-06-30 2015-08-04 Centocor Ortho Biotech Inc Differentiation of pluripotent stem cells
JP5734183B2 (en) 2008-06-30 2015-06-17 ヤンセン バイオテツク,インコーポレーテツド Differentiation of pluripotent stem cells
US20100028307A1 (en) * 2008-07-31 2010-02-04 O'neil John J Pluripotent stem cell differentiation
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
NZ591292A (en) 2008-08-20 2012-10-26 Anthrogenesis Corp Improved cell composition and methods of making the same
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
CN102186970A (en) 2008-09-12 2011-09-14 克里奥普拉斯低温生物有限公司 Ischemic tissue cell therapy
EP2182055A1 (en) 2008-10-14 2010-05-05 Heinrich-Heine-Universität Düsseldorf Human cord blood derived unrestricted somatic stem cells (USSC)
CA2742267C (en) * 2008-10-31 2019-06-04 Centocor Ortho Biotech Inc. Differentiation of human embryonic stem cells to the pancreatic endocrine lineage
BRPI0919885A2 (en) * 2008-10-31 2015-08-11 Centocor Ortho Biotech Inc Differentiation of human embryonic stem cells into pancreatic endocrine lineage
RU2015130665A (en) 2008-11-19 2018-12-24 Антродженезис Корпорейшн AMNIOTIC ADHESIVE CELLS
US9192695B2 (en) 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
RU2555538C2 (en) 2008-11-20 2015-07-10 Сентокор Орто Байотек Инк. Culture of pluripotent stem cells on microcarriers
AU2009316583B2 (en) * 2008-11-20 2016-04-21 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
MX353489B (en) * 2009-07-02 2018-01-16 Anthrogenesis Corp Method of producing erythrocytes without feeder cells.
RU2540021C2 (en) * 2009-07-20 2015-01-27 Янссен Байотек, Инк. Differentiating human embryonic stem cells
EP2456862A4 (en) 2009-07-20 2013-02-27 Janssen Biotech Inc Differentiation of human embryonic stem cells
GB2485113B (en) * 2009-07-20 2016-12-28 Janssen Biotech Inc Differentiation of human embryonic stem cells into cells of the pancreatic endoderm lineage
CN102625829B (en) 2009-07-21 2015-05-06 Abt控股公司 Use of stem cells to reduce leukocyte extravasation
US20110020292A1 (en) 2009-07-21 2011-01-27 Abt Holding Company Use of Stem Cells to Reduce Leukocyte Extravasation
PL2494035T3 (en) * 2009-10-29 2018-07-31 Janssen Biotech, Inc. Pluripotent stem cells
RU2586506C2 (en) * 2009-12-23 2016-06-10 Янссен Байотек, Инк. Differentiation of human embryonic stem cells
CN102741395B (en) 2009-12-23 2016-03-16 詹森生物科技公司 The differentiation of human embryo stem cell
WO2011094181A1 (en) 2010-01-26 2011-08-04 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
EP3940060A1 (en) 2010-02-25 2022-01-19 ABT Holding Company Modulation of macrophage activation
SG183498A1 (en) 2010-02-25 2012-09-27 Abt Holding Co Modulation of angiogenesis
AU2011223900A1 (en) 2010-03-01 2012-09-13 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
PT2556145T (en) 2010-04-07 2016-10-25 Anthrogenesis Corp Angiogenesis using placental stem cells
WO2011127113A1 (en) 2010-04-08 2011-10-13 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
MX351515B (en) 2010-05-12 2017-10-17 Janssen Biotech Inc Differentiation of human embryonic stem cells.
CA3128483A1 (en) 2010-05-12 2011-11-17 Abt Holding Company Modulation of splenocytes in cell therapy
US9090878B2 (en) 2010-06-17 2015-07-28 Katholieke Universiteit Leuven Methods for differentiating cells into hepatic stellate cells and hepatic sinusoidal endothelial cells, cells produced by the methods, and methods for using the cells
EP2582788A4 (en) 2010-06-17 2014-03-19 Stemrd Inc Serum-free chemically defined cell culture medium
JP5996533B2 (en) 2010-07-13 2016-09-21 アントフロゲネシス コーポレーション How to generate natural killer cells
AU2011293440B2 (en) 2010-08-24 2016-05-05 Katholieke Universiteit Leuven Non-static suspension culture of cell aggregates
CA2809305C (en) 2010-08-31 2019-06-11 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
AU2011296381B2 (en) 2010-08-31 2016-03-31 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9528090B2 (en) 2010-08-31 2016-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
WO2012092485A1 (en) 2010-12-31 2012-07-05 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
US9388388B2 (en) 2011-01-31 2016-07-12 Katholieke Universiteit Leuven Methods for making cells with an extra-embryonic endodermal precursor phenotype
AU2012262273B2 (en) 2011-06-01 2017-09-14 Celularity Inc. Treatment of pain using placental stem cells
SG10201604543PA (en) 2011-06-06 2016-07-28 ReGenesys BVBA Expansion of stem cells in hollow fiber bioreactors
US20140328811A1 (en) 2011-08-01 2014-11-06 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
GB201119335D0 (en) 2011-11-09 2011-12-21 Univ Leuven Kath Hepatitis virus infectable stem cells
AU2012355698B2 (en) 2011-12-22 2018-11-29 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
KR20140131999A (en) 2012-03-07 2014-11-14 얀센 바이오테크 인코포레이티드 Defined Media for Expansion and Maintenance of Pluripotent Stem Cells
CN108103006A (en) 2012-06-08 2018-06-01 詹森生物科技公司 Differentiation of the human embryo stem cell to pancreatic endocrine cell
US9321995B2 (en) * 2012-12-20 2016-04-26 Suzhou Biowisetech Co., Ltd. Stem cell culture medium and its applications as well as a stem cell culture method
JP6529440B2 (en) 2012-12-31 2019-06-12 ヤンセン バイオテツク,インコーポレーテツド Suspension and clustering of human pluripotent cells for differentiation to pancreatic endocrine cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
KR102036780B1 (en) 2012-12-31 2019-10-25 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
KR102084561B1 (en) 2012-12-31 2020-03-04 얀센 바이오테크 인코포레이티드 Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
EP2953635A4 (en) 2013-02-05 2016-10-26 Anthrogenesis Corp Natural killer cells from placenta
KR102312720B1 (en) 2013-03-15 2021-10-13 알로소스 Cell repopulated collagen matrix for soft tissue repair and regeneration
KR102161726B1 (en) 2013-04-12 2020-10-06 사베리오 라프란체스카 Improving organs for transplantation
DK2992088T3 (en) 2013-04-30 2019-11-11 Univ Leuven Kath CELL THERAPY FOR MYELODYSPLASTIC SYNDROMES
JP6617231B2 (en) 2013-08-28 2019-12-11 国立大学法人岐阜大学 Method for producing induced pluripotent stem cells
JP5889856B2 (en) * 2013-10-15 2016-03-22 佛教慈濟醫療財團法人 Method for proliferating human mesenchymal stem cells ex vivo, adjuvant used therefor, method for extracting growth factor of human mesenchymal stem cells ex vivo, and use of the growth factor
WO2015148704A1 (en) 2014-03-25 2015-10-01 Terumo Bct, Inc. Passive replacement of media
CA2949056A1 (en) 2014-05-16 2015-11-19 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
JP6830059B2 (en) 2014-09-26 2021-02-17 テルモ ビーシーティー、インコーポレーテッド Scheduled cell feeding
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
SG11201806245TA (en) 2016-01-21 2018-08-30 Abt Holding Co Stem cells for wound healing
MA45479A (en) 2016-04-14 2019-02-20 Janssen Biotech Inc DIFFERENTIATION OF PLURIPOTENT STEM CELLS IN ENDODERMAL CELLS OF MIDDLE INTESTINE
WO2017205667A1 (en) 2016-05-25 2017-11-30 Terumo Bct, Inc. Cell expansion
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US20200246386A1 (en) 2017-04-24 2020-08-06 Pluristem Ltd. Methods and compositions for treating neurological disorders

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444778A (en) * 1981-08-27 1984-04-24 Coughlin Shaun R Method and composition for treating atherosclerosis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NO162160C (en) * 1987-01-09 1989-11-15 Medi Cult As SERUM-FREE GROWTH MEDIUM AND USE THEREOF.
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444778A (en) * 1981-08-27 1984-04-24 Coughlin Shaun R Method and composition for treating atherosclerosis

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BROWER M, ET AL.: "GROWTH OF CELL LINES AND CLINICAL SPECIMENS OF HUMAN NON-SMALL CELL LUNG CANCER IN A SERUM-FREE DEFINED MEDIUM", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 46, no. 01, 1 February 1986 (1986-02-01), US, pages 798 - 806, XP002948928, ISSN: 0008-5472 *
JESSOP H L, NOBLE B S, CRYER A: "THE DIFFERENTITATION OF A POTENTIAL MESENCHYMAL STEM CELL POPULATION WITHIN OVINE BONE MARROW", BIOCHEMICAL SOCIETY TRANSACTIONS, PORTLAND PRESS LTD, GB, vol. 22, no. 03, 1 August 1994 (1994-08-01), GB, pages 248S, XP002948929, ISSN: 0300-5127 *
MAURER H. R.: "TOWARDS CHEMICALLY-DEFINED, SERUM-FREE MEDIA FOR MAMMALIAN CELL CULTURE.", ANIMAL CELL CULTURE, XX, XX, 1 January 1986 (1986-01-01), XX, pages 13 - 31., XP002912242 *
See also references of EP0832188A4 *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6733746B2 (en) 1996-03-12 2004-05-11 Invitrogen Corporation Hematopoietic cell culture nutrient supplement
US8198084B2 (en) 1996-08-30 2012-06-12 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
US9321996B2 (en) 1996-08-30 2016-04-26 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
US8815573B2 (en) 1996-08-30 2014-08-26 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
US8785194B2 (en) 1996-08-30 2014-07-22 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
US8455246B2 (en) 1996-08-30 2013-06-04 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
EP0948255A4 (en) * 1996-12-06 2002-10-23 Osiris Therapeutics Inc Improved chondrogenic differentiation of human mesenchymal stem cells
EP0948255A1 (en) * 1996-12-06 1999-10-13 Osiris Therapeutics, Inc. Improved chondrogenic differentiation of human mesenchymal stem cells
WO2000027996A1 (en) * 1998-11-09 2000-05-18 Consorzio Per La Gestione Del Centro Di Biotecnologie Avanzate Serum free medium for chondrocyte-like cells
US6617159B1 (en) 1998-11-09 2003-09-09 Consorzio Per La Gestione Del Centro Di Biotechnologie Avanzate Serum free medium for chondrocyte cells
US7109032B2 (en) 1998-11-09 2006-09-19 Consorzio Per La Gestione Del Centro Di Biotecnologie Avanzate Serum-free medium for mesenchymal stem cells
EP1478734A2 (en) * 2002-01-25 2004-11-24 Genzyme Corporation Serum-free media for chondrocytes and methods of use thereof
EP1478734A4 (en) * 2002-01-25 2006-04-05 Genzyme Corp Serum-free media for chondrocytes and methods of use thereof
WO2004052177A2 (en) 2002-12-05 2004-06-24 Case Western Reserve University Cell-based therapies for ischemia
EP1629089A1 (en) * 2003-06-03 2006-03-01 Organ Recovery Systems, Inc. Selection and propagation of progenitor cells
WO2008002914A3 (en) * 2006-06-26 2008-04-10 Gambro Bct Inc Method of culturing mesenchymal stem cells
WO2009084662A1 (en) * 2007-12-28 2009-07-09 Fujirebio Inc. Medium for mammalian somatic cells and additive therefor
WO2011069091A1 (en) 2009-12-04 2011-06-09 Boston Biomedical Research Institute, Inc. Method for cloning pluripotent stem cells
WO2011159797A2 (en) 2010-06-15 2011-12-22 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
WO2012006440A2 (en) 2010-07-07 2012-01-12 Cellular Dynamics International, Inc. Endothelial cell production by programming
US11773363B2 (en) 2010-10-08 2023-10-03 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11746319B2 (en) 2010-10-08 2023-09-05 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US10669519B2 (en) 2010-10-08 2020-06-02 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US10870827B2 (en) 2010-10-08 2020-12-22 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
WO2012109208A2 (en) 2011-02-08 2012-08-16 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
US9623048B2 (en) 2013-02-08 2017-04-18 Shanghai Institutes For Biological Sciences, Chinese Academy Of Sciences Human hepatocyte-like cells and uses thereof
WO2014130770A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
WO2014132137A2 (en) 2013-03-01 2014-09-04 Université De Genève Transgenic cell selection
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11708554B2 (en) 2013-11-16 2023-07-25 Terumo Bct, Inc. Expanding cells in a bioreactor
US10472605B2 (en) 2014-02-14 2019-11-12 National University Of Ireland, Galway Serum-free medium
WO2015164228A1 (en) 2014-04-21 2015-10-29 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
WO2017070145A1 (en) 2015-10-19 2017-04-27 Cellular Dynamics International, Inc. Production of virus-receptive pluripotent stem cell (psc)-derived hepatocytes
US10947502B2 (en) 2015-10-20 2021-03-16 FUJIFILM Cellular Dynamics, Inc. Methods for directed differentiation of pluripotent stem cells to immune cells
WO2017070337A1 (en) 2015-10-20 2017-04-27 Cellular Dynamics International, Inc. Methods for directed differentiation of pluripotent stem cells to immune cells
WO2017070333A1 (en) 2015-10-20 2017-04-27 Cellular Dynamics International, Inc. Multi-lineage hematopoietic precursor cell production by genetic programming
WO2017079215A1 (en) 2015-11-03 2017-05-11 Glycomimetics, Inc. Methods and compositions for the production of monoclonal antibodies, hematopoietic stem cells, and methods of using the same
WO2018067826A1 (en) 2016-10-05 2018-04-12 Cellular Dynamics International, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
WO2018067836A1 (en) 2016-10-05 2018-04-12 Cellular Dynamics International, Inc. Methods for directed differentiation of pluripotent stem cells to hla homozygous immune cells
WO2018195175A1 (en) 2017-04-18 2018-10-25 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
EP4083063A2 (en) 2017-04-18 2022-11-02 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
US11649455B2 (en) 2018-03-30 2023-05-16 University Of Geneva Micro RNA expression constructs and uses thereof
WO2019186274A2 (en) 2018-03-30 2019-10-03 University Of Geneva Micro rna expression constructs and uses thereof
WO2021240240A1 (en) 2020-05-27 2021-12-02 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
WO2022235586A1 (en) 2021-05-03 2022-11-10 Astellas Institute For Regenerative Medicine Methods of generating mature corneal endothelial cells
WO2022235869A1 (en) 2021-05-07 2022-11-10 Astellas Institute For Regenerative Medicine Methods of generating mature hepatocytes
WO2023213983A2 (en) 2022-05-04 2023-11-09 Antion Biosciences Sa Expression construct

Also Published As

Publication number Publication date
JPH11506610A (en) 1999-06-15
CA2223582A1 (en) 1996-12-12
AU5969296A (en) 1996-12-24
EP0832188A1 (en) 1998-04-01
EP0832188A4 (en) 2001-07-25
US5908782A (en) 1999-06-01

Similar Documents

Publication Publication Date Title
US5908782A (en) Chemically defined medium for human mesenchymal stem cells
US9321995B2 (en) Stem cell culture medium and its applications as well as a stem cell culture method
Kato et al. Growth requirements of low‐density rabbit costal chondrocyte cultures maintained in serum‐free medium
EP1988159B1 (en) Additive for culture medium for use in serum-free culture of animal cell, kit, and use of the additive or kit
RU2272839C2 (en) Serum-free medium for cultured cells used for reconstruction of osseous and cartilage segments (variants)
US5324656A (en) Media for normal human muscle satellite cells
Kato et al. Fibroblast growth factor stimulates colony formation of differentiated chondrocytes in soft agar
AU742638B2 (en) Improved chondrogenic differentiation of human mesenchymal stem cells
Dollenmeier et al. Proliferation and differentiation of chick skeletal muscle cells cultured in a chemically defined medium
KR20060076781A (en) Cell culture media
WO2014094386A1 (en) Stem cell culture medium and use thereof and stem cell culture method
Ernst et al. Osteoblastlike cells in a serum-free methylcellulose medium form colonies: effects of insulin and insulinlike growth factor I
US20120213748A1 (en) Cardiomyocytes and methods of producing and purifying cardiomyocytes
Rockwell et al. The growth requirements of SV40 virus transformed Balb/c‐3T3 cells in serum‐free monolayer culture
TW200927927A (en) Stem cell medium
Loo et al. Serum‐free mouse embryo cells: growth responses in vitro
CN112708591B (en) Culture medium with definite chemical components for in-vitro differentiation of muscle stem cells
Webber et al. Serum‐free culture of rabbit meniscal fibrochondrocytes: Proliferative response
US20230117670A1 (en) Bioactive substance composition, serum-free medium comprising the composition, and uses thereof
CLARK et al. Islet cell culture in defined serum-free medium
Kumegawa et al. In vitro effects of thyroxine and insulin on myoblasts from chick embryo skeletal muscle
AU734174B2 (en) Chemically defined medium for human mesenchymal stem cells
CN111621470B (en) High-efficiency low-toxicity myocardial purification culture medium and method
Suryawan et al. The primary cell culture system for preadipocytes
CN117343898A (en) Serum-free culture medium and application thereof in preparation of large yellow croaker cell culture meat

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref country code: JP

Ref document number: 1997 501017

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref document number: 2223582

Country of ref document: CA

Ref country code: CA

Ref document number: 2223582

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996916987

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996916987

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1996916987

Country of ref document: EP