US20050265980A1 - Cell culture environments for the serum-free expansion of mesenchymal stem cells - Google Patents

Cell culture environments for the serum-free expansion of mesenchymal stem cells Download PDF

Info

Publication number
US20050265980A1
US20050265980A1 US11/129,612 US12961205A US2005265980A1 US 20050265980 A1 US20050265980 A1 US 20050265980A1 US 12961205 A US12961205 A US 12961205A US 2005265980 A1 US2005265980 A1 US 2005265980A1
Authority
US
United States
Prior art keywords
cell culture
serum
medium
bfgf
collagen
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/129,612
Inventor
Chang Chen
Andrea Liebmann-Vinson
Ruiling Xu
Jonathan Rowley
Perry Haaland
Bryce Chaney
Matthew Mitchell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Becton Dickinson and Co
Original Assignee
Becton Dickinson and Co
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 Becton Dickinson and Co filed Critical Becton Dickinson and Co
Priority to US11/129,612 priority Critical patent/US20050265980A1/en
Assigned to BECTON, DICKINSON AND COMPANY reassignment BECTON, DICKINSON AND COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANEY, BRYCE N., HAALAND, PERRY D., CHEN, CHANG, LIEBMAN-VINSON, ANDREA, MITCHELL, MATTHEW, ROWLEY, JONATHAN ALLEN, XU, RUILING
Publication of US20050265980A1 publication Critical patent/US20050265980A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C12N2500/36Lipids
    • 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
    • C12N2500/42Organic phosphate, e.g. beta glycerophosphate
    • 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
    • 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
    • C12N2501/11Epidermal growth factor [EGF]
    • 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
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • 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
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • 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
    • C12N2501/10Growth factors
    • C12N2501/14Erythropoietin [EPO]
    • 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
    • C12N2501/145Thrombopoietin [TPO]
    • 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
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • 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
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • C12N2501/165Vascular endothelial growth factor [VEGF]
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/21Chemokines, e.g. MIP-1, MIP-2, RANTES, MCP, PF-4
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2303Interleukin-3 (IL-3)
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2305Interleukin-5 (IL-5)
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2306Interleukin-6 (IL-6)
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2311Interleukin-11 (IL-11)
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/26Flt-3 ligand (CD135L, flk-2 ligand)
    • 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
    • C12N2501/30Hormones
    • C12N2501/32Angiotensins [AT], angiotensinogen
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • 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
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • 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
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/58Adhesion molecules, e.g. ICAM, VCAM, CD18 (ligand), CD11 (ligand), CD49 (ligand)
    • 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
    • C12N2501/60Transcription factors
    • 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
    • C12N2501/70Enzymes
    • 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
    • C12N2501/999Small molecules not provided for elsewhere
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/52Fibronectin; Laminin
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides

Definitions

  • the present invention relates to serum-free cell culture systems that provide for mesenchymal stem cell expansion while maintaining a pluripotent phenotype, and methods of use for the expanded mesenchymal stem cell populations.
  • MSCs Mesenchymal stem cells
  • MSCs Mesenchymal stem cells
  • MSCs have generated a high level of experimental and clinical interest due to their potential for a range of therapeutic uses including repair of damaged or diseased tissues (Baksh et al. (2004) J. Cell. Mol. Med. 8:301-316; Barry and Murphy (2004) Int. J. Biochem. Cell Bio. 36:568-584).
  • MSCs do very poorly in serum-free environments because they detach and die in culture. These MSCs can be maintained in an attached state in vitro with minimal serum (e.g., ⁇ 1%), although such an environment provides little stimulation for MSCs to proliferate and grow.
  • serum-free cell culture environments have been described for MSC expansion (Lennon et al. (1995) Exp. Cell Res. 219:211-222; U.S. Pat. No. 5,908,782), the current industry standard still contains a large amount of serum.
  • compositions and methods for promoting mesenchymal stem cell (MSC) expansion while maintaining a pluripotent phenotype are provided.
  • the compositions include serum-free cell culture systems for MSC expansion that comprise a serum-free cell culture medium and a two-dimensional or three-dimensional cell culture surface.
  • at least one insoluble substrate protein is presented from the cell culture surface.
  • the cell culture surface comprises a cell culture support bound to a cell adhesion resistant (CAR) material, which in turn is bound to at least one insoluble substrate protein.
  • CAR cell adhesion resistant
  • Insoluble substrate proteins for use in the present invention include extracellular matrix (ECM) proteins such as fibronectin, laminin, hyaluronic acid (HA), vitronectin, collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof.
  • ECM extracellular matrix
  • the cell culture surface comprises a cell culture support bound to a cell adhesion resistant (CAR) material, which in turn is bound to at least one insoluble substrate protein, for example, at least one ECM protein.
  • the serum-free cell culture medium is a solution that comprises a mixture of soluble MSC growth-promoting factors.
  • Compositions further include kits comprising serum-free cell culture media and a two-dimensional or three-dimensional cell culture surface suitable for MSC expansion.
  • Methods of the present invention comprise the use of these serum-free cell culture systems to promote the expansion of MSCs.
  • Further methods comprise the use of these serum-free cell culture systems and expanded MSCs for cell transplantation or to engineer tissues to treat various disorders or diseases, including those of the cardiovascular system, muscle, ligament, bone, tendon, cartilage, nervous system, blood, immune system, liver, or pancreas.
  • Further methods comprise the use of these serum-free cell culture systems to promote the expansion of MSCs within primary aspirates from whole bone marrow such that the MSCs are co-cultured with non-MSCs present in the primary aspirates.
  • FIG. 1 shows expansion of total mesenchymal stem cells (MSCs) over time in the G2 serum-free culture system and collagen 1+fibronectin surface of the present invention (BDT Environment) versus expansion in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic.
  • MSCs mesenchymal stem cells
  • FIG. 2 shows the adipocyte differentiation capacity of MSCs expanded in the G2 serum-free medium and collagen 1+fibronectin surface (BDT Environment) or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic.
  • FIG. 3 shows the bone differentiation capacity of MSCs expanded in the G2 serum-free medium collagen 1+fibronectin surfaces (BDT Environment) or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic.
  • FIG. 4 shows a comparison of the number of hMSCs after culture in a serum-free base medium (BDTM) with or without growth factors on either a tissue culture polystyrene (TCPS) or cell adhesion resistant (CAR) surface.
  • BDTM serum-free base medium
  • TCPS tissue culture polystyrene
  • CAR cell adhesion resistant
  • FIG. 5 shows a comparison of various serum-free compositions and collagen 1+fibronectin surfaces for MSC expansion that are labeled C6, D3, C2, and G5, compared to G2 medium and a serum containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 6A shows the adipocyte differentiation capacity of MSCs expanded in D3, C2, and G5 serum-free culture media and collagen 1/fibronectin surfaces or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 6B shows the bone differentiation capacity of MSCs expanded in D3, C2, and G5 serum-free culture media and collagen 1/fibronectin surfaces or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 7 shows a comparison of further refined serum-free composition for MSC expansion labeled G7, G4, and C8 and cultured on collagen 1/fibronectin surfaces compared to expansion in Cambrex Biosciences (Baltimore, Md.) serum-containing medium (10% FBS CM).
  • FIG. 8 shows a comparison of MSC expansion in various media conditions and collagen 1/fibronectin surfaces, including G4 serum-free medium, G4+TGF ⁇ , FGF only, EGF only, and TGF ⁇ only, as compared to basal medium and Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 9 shows a comparison of MSC expansion in various media conditions where selected growth factor combinations were added to basal medium, as compared to basal medium alone and Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (TCPS).
  • Growth factor combinations were bFGF+TGF- ⁇ , WNT-3a+bFGF, bFGF+EGF, WNT-3a+TGF- ⁇ , WNT-3a+EGF, EGF+TGF- ⁇ , WNT-3a alone, bFGF+EGF+TGF- ⁇ , WNT-3a+bFGF+EGF+TGF- ⁇ , and bFGF+EGF+TGF- ⁇ +BIO.
  • the present invention relates to compositions and methods for promoting mesenchymal stem cell (MSC) expansion while maintaining the pluripotent phenotype of these cells.
  • Serum-free cell culture systems for MSC expansion are provided. These cell culture systems comprise serum-free cell culture medium and a two-dimensional or three-dimensional cell culture surface.
  • the serum-free cell culture medium is a solution that comprises a mixture of soluble MSC growth-promoting and self-renewal factors.
  • at least one insoluble substrate protein is presented from the cell culture surface.
  • the cell culture surface comprises a cell culture support bound to a cell adhesion resistant (CAR) material, which in turn is bound to at least one insoluble substrate protein.
  • CAR cell adhesion resistant
  • Insoluble substrate proteins may include but are not limited to extracellular matrix (ECM) proteins such as fibronectin, laminin, hyaluronic acid (HA), vitronectin, or collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof.
  • ECM extracellular matrix
  • HA hyaluronic acid
  • collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof.
  • kits comprising the serum-free cell culture systems of the invention, which are suitable for MSC expansion.
  • Methods of the present invention are directed to the use of these serum-free cell culture systems to promote the expansion of MSCs and to engineer tissues.
  • the expanded MSCs of the present invention can be used to treat various disorders or diseases, particularly those of the cardiovascular system, muscle, ligament, bone, tendon, cartilage, nervous system, blood, immune system, liver, or pancreas.
  • meenchymal is intended to mean such cells as bone marrow, endothelial cells, epithelial cell progenitors, cardiomyocytes, astrocytes, neurons, chondrocytes, osteoblasts, pancreatic cells, hepatocytes, and other cells of mesenchymal origin.
  • meenchymal stem cell or “MSC” refers to a cell that gives rise to a cell of mesenchymal lineage.
  • expanded is intended to mean that the resultant cell population is derived from ex vivo culture of stem cells in media compositions comprising mixtures of cytokines, where the outgoing (cultured) number of cells exceeds the ingoing (non-cultured) number of cells.
  • the term “expanded” is not to be construed or limited by any mechanism or theory of cellular origin and may comprise cells that originate de novo in culture.
  • the serum-free cell culture system of the present invention comprises a serum-free cell culture medium and a cell culture surface.
  • at least one insoluble substrate protein is presented from the cell culture surface, meaning that the insoluble substrate protein is bound, adsorbed, linked, attached or in some way associated with the cell culture surface.
  • Suitable insoluble substrate proteins include the ECM proteins disclosed herein.
  • the insoluble substrate protein presented from the cell culture surface is a combination of collagen I and fibronectin.
  • the cell culture surface comprises a cell culture support bound to a cell adhesion resistant material, which in turn is bound to at least one insoluble substrate protein.
  • the cell culture support may be solid or porous, polymer, metal, glass, ceramic, or combinations thereof. Culturing of MSCs in the presence of this cell culture surface and the serum-free cell culture medium unexpectedly provides for expansion of these cells while maintaining their pluripotent phenotype.
  • CAR cell adhesion resistant
  • a material that, when present on a surface, prevents, inhibits, or reduces the non-specific binding (adhesion) of cells, proteins, or polypeptides found on cell surfaces.
  • CAR materials are resistant to adhesion of mammalian cells and also to microorganisms.
  • CAR materials are sometimes referred to as non-fouling substrates, inert coatings, low affinity reagents, or non-adhesive coatings.
  • the cell culture support to which is bound the CAR material may be two- or three-dimensional, solid or porous, and may be constructed of any of a variety of materials, including natural polymers, synthetic polymers, hydrogels, metals, ceramics, and inorganic or organic-inorganic composites.
  • the cell culture support may be shaped using methods such as, for example, solvent casting, compression molding, filament drawing, meshing, leaching, weaving, and coating.
  • suitable polymers and hydrogels include, but are not limited to, collagen, glycosaminoglycan (GAG)-based materials, alginate, hyaluronate poly(alpha esters) such as poly(lactate acid), poly(glycolic acid), polyorthoesters and polyanhydrides and their copolymers, cellulose ether, cellulose, cellulosic ester, fluorinated polyethylene, phenolic, poly-4-methylpentene, polyacrylonitrile, polyamide, polyamideimide, polyacrylate, polybenzoxazole, polycarbonate, polycyanoarylether, polyestercarbonate, polyether, polyetheretherketone, polyetherimide, polyetherketone, polyethersulfone, polyethylene, polyfluoroolefin, polylmide, polyolefin, polyoxadiazole, polyphenylene oxide, polyphenylene sulfide, polypropylene, polystyrene, polysulfide, polysulf
  • polysebacic acid polymers such as polylactic acid (PLA), polyglycolic acid (PGA), poly(orthoester), polysebacic acid polymers (PSA), poly(lactic-co-glycolic) acid copolymers (PLGA), poly(lactic-co-sebacic) acid copolymers (PLSA), poly(glycolic-co-sebacic) acid copolymers (PGSA), hydrogels such as polyhydroxyethylmethacrylate (poly-HEMA) or polyethylene oxide-polypropylene oxide copolymer, polyethylegylcols (PEG), polyvinylalchols (PVA), polyvinylpyrrolidone (PVP) and polyhydroxyalkanoate (PHA).
  • PLA polylactic acid
  • PGA polyglycolic acid
  • PVA poly(orthoester)
  • PSA polysebacic acid polymers
  • PLA poly(lactic-co-glycolic) acid copolymers
  • Hybrid materials containing naturally derived and synthetic polymer materials e.g., PGA and PLGA may also be used. Non-limiting examples of such materials are disclosed in Chen et al. (2000) Advanced Materials 12:455-457.
  • polymers useful in the present invention include polymers or copolymers of caprolactones, carbonates, amides, amino acids, orthoesters, acetals, cyanoacrylates and degradable urethanes, as well as copolymers of these with straight chain or branched, substituted or unsubstituted, alkanyl, haloalkyl, thioalkyl, aminoalkyl, alkenyl, or aromatic hydroxy- or di-carboxylic acids.
  • the biologically important amino acids with reactive side-chain groups such as lysine, arginine, aspartic acid, glutamic acid, serine, threonine, tyrosine and cysteine, or their enantiomers, may be included in copolymers with any of the aforementioned materials.
  • polymer surfaces are selected from the group consisting of polystyrene, polyethylene, polypropylene, polyethylene terephthalate, polytetrafluoroethylene, polylactide, and cellulose. Silicone polymers such as polydimethylsiloxane (PDMS) are also used.
  • PDMS polydimethylsiloxane
  • Inorganic composites include, e.g., calcium phosphate ceramics, bioglasses and bioactive glass-ceramics, in particular composites combining calcium hydroxyapatite and silicon stabilized tricalcium phosphate.
  • preferred cell culture supports are polystyrene (PS), polypropylene, polyethylene, polyethylene terephthalate, polytri- or tetra-fluoroethylene, polyhexafluoropropylene, polyvinyl chloride, polyvinylidine fluoride, polyactide, cellulose, glass, or a ceramic.
  • CAR material any suitable CAR material, many of which are known to those skilled in the art, may be bound to the cell culture support.
  • Typical CAR materials include hyaluronic acid (HA) or a derivative thereof, alginic acid (AA) or a derivative thereof, poly-HEMA, polyethylene glycol (PEG), glyme or a derivative thereof, polypropylacrylamide, polyisopropylacrylamide, or a combination of these compounds.
  • the CAR material is HA.
  • one or more of a proteoglycan, a biglycan, a glycosaminoglycan, or MatrigelTM may be bound to the CAR material.
  • proteins or other substances may be bound to the CAR material either covalently or non-covalently, but are preferably covalently bound.
  • the proteins or other substances comprise, for example, ECM proteins or polycationic polymers.
  • ECM proteins for use in the present invention may include fibronectin, laminin, hyaluronic acid (HA), vitronectin, or collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof.
  • Various types of covalent bonds can form, some of which are discussed in more detail in co-pending, commonly assigned U.S. Patent Application Publication Nos. 20050036980, 20040062882, 20030113813, and 20030113812, all of which are incorporated herein by reference.
  • one or more ECM proteins as disclosed herein and/or one or more polycationic polymer are bound to the CAR material.
  • a mixture of collagen I and fibronectin is bound to the CAR material.
  • the ECM protein(s) can be in the form of a naturally occurring polypeptide (protein), a recombinant polypeptide, or a synthetic or semi-synthetic polypeptide, including any fragment of the peptide or protein, or combination thereof.
  • polypeptide and “protein” are used interchangeably herein.
  • ECM proteins Methods of cloning, expressing, and purifying polypeptides, such as ECM proteins, are conventional, as are methods of generating synthetic or semi-synthetic polypeptides. ECM proteins can also be obtained from commercial sources.
  • ECM proteins or polycationic polymers can be bound to the CAR material either covalently or non-covalently (for example, passively adsorbed, such as by electrostatic forces, ionic or hydrogen bonds, hydrophilic or hydrophobic interactions, Van der Waals forces, etc.).
  • the binding is covalent.
  • U.S. Patent Application Publication Nos. 20040062882, 20030113813, and 20030113812 describe such covalent binding of molecules to CAR surfaces.
  • one method of attaching a CAR material to a cell culture support comprises treating the cell culture support with an oxidizing plasma, and binding the CAR material to the treated cell culture support.
  • Another method of attaching a CAR material to a cell culture support comprises treating the cell culture support with an oxidizing plasma; exposing the treated cell culture support to a polycationic polymer with amino groups (such as PEI, PDL, poly-L-lysine (PLL), poly-L-ornithine (PLO), poly-D-ornithine (PDO), poly(vinylamine) (PVA) or poly(allylamine) (PAA), in one embodiment, PEI or PDL to form an intermediate layer, and binding the CAR material to the intermediate layer.
  • a polycationic polymer with amino groups such as PEI, PDL, poly-L-lysine (PLL), poly-L-ornithine (PLO), poly-D-ornithine (PDO), poly(vinylamine) (PVA) or poly(allylamine) (PAA)
  • PEI or PDL to form an intermediate layer, and binding the CAR material to the intermediate layer.
  • This invention also speaks to the use of flexible substrates in culture.
  • Flexercell culture systems from Flexcell International Corporation are able to apply tensile, compressive, or shear stresses to cultured cells. See, for example, U.S. Pat. Nos. 4,789,601, 4,822,741, 4,839,280, 6,037,141, 6,048,723, and 6,218,178.
  • U.S. Pat. No. 6,057,150 discloses the application of a biaxial strain to an elastic membrane that may be coated with extracellular matrix proteins and covered with cultured cells.
  • U.S. Pat. No. 6,107,081 discloses another system in which a unidirectional cell stretching device comprising an elastic strip is coated with an extracellular matrix on which cells are cultured and stretched.
  • a flexible substrate can be deformed easily and in a controlled manner, and also supports cell adhesion and growth comparable to conventional cell culture substrates.
  • Silicones such as poly(dimethyl siloxane) (PDMS), are particularly suitable for this application because they are not only highly flexible but also provide optical clarity that allows microscopic observation of the cell cultures.
  • Another aspect of the invention is a method of making the above cell culture surface of the invention, where the method comprises (a) attaching a CAR material to a cell culture support, and (b) attaching to the CAR material collagen I and fibronectin, or biologically active fragment or variant thereof, and, optionally, one or more other ECM proteins (or a biologically active fragment or variant of the ECM protein) and/or one or more polycationic polymers. Any of the ECM proteins or polycationic polymers disclosed herein, or others, may be used.
  • the CAR material is attached to the cell culture support by treating the cell culture support with an oxidizing plasma, and binding the CAR material to the treated cell culture support.
  • the CAR material is attached to the cell culture support by treating the cell culture support with an oxidizing plasma; exposing the treated cell culture support to a polycationic polymer with amino groups to form an intermediate layer; and binding the CAR material to the intermediate layer.
  • the polycationic polymer is polyethylene imine (PEI) or poly-D-lysine (PDL).
  • a CAR material such as HA
  • a CAR material is bonded directly to polymeric nitrogen-containing surfaces.
  • examples of such surfaces are ammonia plasma-treated polymers and Primaria TM-treated polystyrene (PS) surfaces, and poly D-lysine coated surfaces.
  • Polymeric substrates suitable for use in the invention include polystyrene, polypropylene, polyethylene terephthalate, polylactide, cellulose, and the like, though polystyrene is preferred.
  • HA is immobilized directly on an intermediate polyamine layer, e.g., polyethyleneimine, poly-D-Lysine, or poly-L-lysine, or directly bound to the polymer with the HA bound to that intermediate layer).
  • an intermediate polyamine layer e.g., polyethyleneimine, poly-D-Lysine, or poly-L-lysine, or directly bound to the polymer with the HA bound to that intermediate layer).
  • the plasma treatment process may be any process that is capable of causing nitrogen to be incorporated onto the surface of the polymer article resulting in reactive amine or other nitrogen-containing groups, including direct as well as remote plasma treatment methods.
  • suitable plasma treatments are ones using reactive gases such as nitrogen, nitrogen oxide, nitrogen dioxide or ammonia in the gas phase, alone or in mixture with air, argon, or other inert gases, and may be preceded or followed by treatments employing argon or other inert gases.
  • the plasma maybe sustained over the full treatment time or maybe administered in pulses.
  • the plasma gas is ammonia, and treatment is performed with a power charge of between 1 and 400 W, preferably between 10 and 150 W, a pressure between 10 mtorr and 10 ton, and a treatment time between 1 second and 1 hour, preferably between 10 seconds and 30 minutes.
  • Plasma-treated polystyrene can be prepared, for example by pumping the treatment chamber to a 0.3 mTorr base pressure, establishing a 200 mTorr argon atmosphere, and applying a 60 sec argon plasma treatment, followed by a 120 sec, 375 mTorr NH 3 plasma treatment at 95 W.
  • Other suitable treatments will be known to those of skill in the art.
  • the plasma-treated surface may be exposed to an aqueous solution containing HA or a derivative thereof, or alginic acid (alginate; AA) in the presence of a condensing agent such as a carbodiimide, for example, ethyldimethylaminopropyl carbodiimide (EDC), in aqueous solution, or dicyclohexylcarbodiimide (DCC), in organic solvents.
  • a condensing agent such as a carbodiimide, for example, ethyldimethylaminopropyl carbodiimide (EDC), in aqueous solution, or dicyclohexylcarbodiimide (DCC), in organic solvents.
  • a condensing agent such as a carbodiimide, for example, ethyldimethylaminopropyl carbodiimide (EDC), in aqueous solution, or dicyclohexylcarbodiimide
  • exposure or “exposing” as used herein is intended to include any type of contact made between a liquid and a solid, for example by pipetting, pouring, spraying, dripping, immersing, pouring, dipping, injecting, etc., without limitation.
  • HA is an anionic polysaccharide composed of repeating units of beta-1,4-glucuronatebeta-1,3-N-acetylglucosamine.
  • a reactive —COO ⁇ group is present on every repeat unit of HA that can be utilized to covalently couple HA to an amine containing surface using methods described herein.
  • a condensing agent such as EDC activates the —COO ⁇ groups present in HA, creating a reactive ester intermediate (ester (o-acylisourea) intermediate).
  • This intermediate is highly unstable and subject to hydrolysis, leading to the cleaving off of the activated ester intermediate, forming an isourea, and regenerating the —COO ⁇ group.
  • N-hydroxysuccinimides include, but are not limited to, N-hydroxy-succinimide (NHS), hydroxy-sulfosuccinimide (sulfo-NHS) or hydroxybenzotriazole hydrate.
  • HA covalently bonded to plasma-treated polystyrene in this way prevents attachment of any of a number of types of cells, including NIH3T3 and osteoblast MC3T3 cells as well as MSC's and a variety of primary and stem cells.
  • Suitable derivatives of HA that may be used in the invention will be known to the skilled artisan, and are described, for example, in U.S. Pat. No. 4,851,521. These include partial esters of HA with alcohols of the aliphatic, araliphatic, cycloaliphatic and heterocyclic series, and salts of such partial esters with inorganic or organic bases. Similar derivatives of alginic acid should also be useful. Further, other plasma treatment methods for producing surfaces with amine and other nitrogen-containing groups are also suitable, and are known to those of skill in the art.
  • the cell culture surfaces for use in the serum-free cell culture systems include but are not limited to standard tissue culture vessels and two-dimensional surfaces, including sheets, slides, culture dishes, culture flasks, bags, culture bottles, or multiwell dishes.
  • three-dimensional cell culture surfaces such as microcarriers or three dimensional (3-D) scaffolds, including but not limited to foams, hydrogels, or fiber meshes may be used for generating a three-dimensional cell culture, tissue, or organ.
  • Three-dimensional cell culture surfaces such as microcarriers or three dimensional (3-D) scaffolds, including but not limited to foams, hydrogels, or fiber meshes may be used for generating a three-dimensional cell culture, tissue, or organ.
  • Three-dimensional scaffold refers herein to a 3-D porous template that provides a very high surface area to volume ratio for cell culture. These scaffolds may be used for initial cell attachment and expansion, or subsequently for tissue formation either in vitro or in vivo.
  • a 3-D scaffold according to this invention comprises base materials (described below), a CAR layer and bound thereto one or more insoluble substrate proteins, such as the ECMs disclosed herein, and, optionally, other substances, which promote or enhance cell attachment, growth, migration, and/or differentiation.
  • the scaffold is seeded with MSCs and contacted with the serum-free cell culture medium described herein below in order to permit cell growth and differentiation in a structural environment that more closely mimics the in vivo setting.
  • Cells derived therefrom can be isolated from the scaffold or can be implanted with or without the scaffold into a suitable location in the body of a mammal, preferably a human patient. However, the use of this technology could easily be translated to non-human mammals such as cats, dogs, horses, and the like.
  • the shape and dimensions of the 3-D scaffold are determined based on the organ being replaced or supplemented, and the type of scaffold material being used to create the construct. For example, if a polymeric scaffold is used for heart tissue replacement or supplementation, the dimension of the polymeric scaffold can vary in terms of width and length of the polymeric scaffold. One of skill in the art recognizes that the size and dimensions of the polymeric scaffold will be determined based on the area of the organ being replaced or supplemented.
  • other suitable articles may constitute a cell culture support surface of the present invention, including medical devices, extracorporeal devices and artificial joints, tubes, sutures, stents, orthopedic devices, vascular grafts, membranes, films, biosensors, or microparticles.
  • One embodiment includes a method of forming tissue-engineered constructs using a 3-D scaffold material or other suitable article that comprises the CAR surface onto which one or more insoluble substrate proteins, for example, the ECMs disclosed herein, are attached, either covalently or non-covalently.
  • a scaffold in combination with the serum-free cell culture medium disclosed herein supports the maturation, development and differentiation, of additional cultured MSCs in vitro to form components of adult tissues analogous to their in vivo counterparts.
  • the tissue-engineered constructs are created using scaffold materials disclosed herein as the substrate onto which cells are deposited and cultured in the presence of the serum-free cell culture medium described herein below, and on which cells are grown and adhere.
  • the scaffold allows optimum cell-cell interactions, thereby allowing a more natural formation of cellular phenotypes and a tissue microenvironment.
  • the scaffold also allows MSCs to continue to grow actively, proliferate, and differentiate to produce a tissue-engineered construct that is also capable of supporting the growth, proliferation, and differentiation of additional cultured cell populations, if needed.
  • the scaffold is biocompatible and conducive to cell attachment and subsequent tissue growth.
  • Other surface properties can be modified to suit the intended application without altering other properties of the scaffold such as its mechanical strength or thermal properties.
  • Useful surface modifications could include, for example, changes in chemical group functionality, surface charge, hydrophobicity, hydrophilicity, and wettability.
  • the CAR surface technology can easily be translated to the surfaces of 3-D scaffolds. Such surface modifications are well known in the art.
  • Sterilization is performed prior to seeding the scaffold with cells. Heat sterilization is often impractical since the heat treatment could deform the device, especially if the materials have a melting temperature below that required for the heat sterilization treatment. For example, cold ethylene oxide gas, vapor hydrogen peroxide treatments can be used for sterilization.
  • the 3-D scaffolds of the present invention comprise any suitable base material for construction, including the polymeric materials disclosed herein above.
  • the polymeric matrix can be fabricated to have a controlled pore structure that allows nutrients from the serum-free cell culture medium described herein below to reach the deposited cell population but prevent cultured cells from migrating through the pores. In vitro cell attachment and cell viability can be assessed using scanning electron microscopy, histology and quantitative assessment with radioisotopes.
  • the polymeric matrix can be shaped into any number of desirable configurations to satisfy any number of overall system, geometry, or space restrictions.
  • the polymeric matrix can be shaped to different sizes to conform to the organs of different sized patients.
  • the tissue-engineered construct can be flat, tubular, or of complex geometry. The shape of the construct will be decided by its intended use.
  • the construct can be implanted to repair, supplement, or replace diseased or damaged parts of organs.
  • the scaffold base material is a hydrogel composed of crosslinked polymer networks that are typically insoluble or poorly soluble in water, but can swell to an equilibrium size in the presence of excess water.
  • the MSCs can be placed in a hydrogel and the hydrogel injected into desired locations within the organ.
  • the hydrogel compositions can include, without limitation, for example, poly(esters), poly(hydroxy acids), poly(lactones), poly(amides), poly(ester-amides), poly(amino acids), poly(anhydrides), poly(ortho-esters), poly(carbonates), poly(phosphazines), poly(thioesters), polysaccharides and mixtures thereof.
  • compositions can also include, for example, a poly(hydroxy) acid including poly(alpha-hydroxy) acids and poly(betahydroxy) acids.
  • poly(hydroxy) acids include, for example, polylactic acid, polyglycolic acid, polycaproic acid, polybutyric acid, polyvaleric acid, and copolymers and mixtures thereof. Due to the unique properties of hydrogels and their potential applications in such areas as controlled drug delivery, various types of hydrogels have been synthesized and characterized.
  • the matrix materials of the present invention encompass both conventional foam or sponge materials and the so-called hydrogel sponges (see, e.g., U.S. Pat. No. 5,451,613).
  • Mesenchymal stem cells grown on the scaffold materials in the presence of the serum-free cell culture medium disclosed herein in accordance with the present invention may grow in multiple layers, forming a cellular structure that resembles physiologic conditions found in vivo.
  • the scaffold can support the proliferation of different types of cells and the formation of a number of different tissues. Examples include, but are not limited to, kidney, heart, skin, liver, pancreas, adrenal and neurological tissue, as well as tissues of the gastrointestinal and genitourinary tracts, and the circulatory system.
  • the cells grown or expanded on the aforementioned scaffold can be used, alone or in combination with the scaffold, in a variety of applications.
  • the scaffold and cells can be implanted into a subject.
  • Implants can be used to replace or supplement existing tissue, for example, by treating a subject with a cardiovascular disorder by replacing or supplementing natural cardiovascular tissue.
  • the subject can be monitored after implantation for amelioration of the cardiovascular disorder.
  • a three-dimensional biocompatible scaffold may be brought into contact with vasculature-promoting expanded MSCs of the invention and then brought into contact with a host tissue at a target site (e.g., within the organ) or where the organ tissue is grown on the scaffold prior to implantation.
  • the graft is then able to grow and proliferate within the target site and replace or supplement the depleted activity of the organ.
  • the construct can be added at a single location in the host or, alternatively, a plurality of constructs can be created and added to multiple sites in the host.
  • target site refers to a region in the host or organ that requires replacement or supplementation.
  • the target site can be a single region in the organ or host, or can be multiple regions in the organ or host.
  • the supplementation or replacement results in the same physiological response as a normal organ.
  • the scaffold is created using parts of a natural decellularized organ.
  • Parts of organs can be decellularized by removing the entire cellular and tissue content from the organ (see, e.g., U.S. Pat. No. 6,479,064).
  • the term “decellularized” or “decellularization” as used herein refers to a biostructure (e.g., an organ, or part of an organ) from which the cellular and tissue content has been removed leaving behind an intact acellular infrastructure.
  • the process of decellularization removes the specialized tissue, leaving behind the complex three-dimensional network of connective tissue.
  • the connective tissue infrastructure is primarily composed of collagen.
  • the decellularized structure provides a matrix material onto which different cell populations can be infused.
  • Decellularized biostructures can be rigid, or semi-rigid, having an ability to alter their shapes. Culture and construction of decellularized biostructures can be performed, for example, as describe in U.S. Pat. No. 6,479,064, which is herein incorporated by reference in its entirety.
  • the two-dimensional and three-dimensional cell culture surfaces serve as a substrate onto which MSCs can be seeded and cultured in the presence of a serum-free cell culture medium disclosed herein.
  • seeded MSCs can be expanded ex vivo by contacting them with a serum-free cell culture medium that comprises a defined cytokine cocktail.
  • the defined combinations comprise at least 2, at least 3, at least, 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, or at least 15 cytokines from the group comprising brain-derived neurotrophic factor (BDNF), basic fibroblast growth factor (bFGF), bone morphogenic protein 2 (BMP-2), bone morphogenic protein 4 (BMP-4), dickkopf 1 (DKK-1), epidermal growth factor (EGF), erythropoietin (EPO), fibronectin, Flt-3/Flk-2 ligand, granulocyte colony stimulating factor (G-CSF), insulin-like growth factor (IGF-1), interleukin-11 (IL-11), interleukin-3 (IL-3), interleukin-5 (IL-5), interleukin-6 (IL-6), leukemia inhibitory factor (LIF), platelet-derived endothelial cell growth factor (PD-ECGF), stem cell factor (SCF), stromal cell derived
  • WNT signaling agonists include, but are not limited to (2′Z,3′E)-6-bromoindirubin-3′-oxime (BIO) and WNT proteins including WNT-1, WNT-2, WNT-2b, WNT-3, WNT-3a, WNT-4, WNT-5a, WNT-5b, WNT-6, WNT-7a, WNT-7b, WNT-8a, WNT-8b, WNT-9a, WNT-9b, WNT-10a, WNT-10b, WNT-11, and WNT-16.
  • the WNT protein is WNT-3a.
  • the defined cytokine cocktail is termed “G2” and comprises a combination of fibronectin, SDF-11, IL-6, SCF, IL-5, BDNF, PD-ECGF, IL-11, IL-3, EPO, Flt-3/Flk-2 ligand, BMP-4, thrombospondin, IGF-1, and bFGF.
  • the defined cytokine cocktail is termed “C6” and comprises a combination of BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1 ⁇ , and WNT-3a.
  • C6 comprises a combination of BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1 ⁇ , and WNT-3a.
  • the defined cytokine cocktail is termed “D3” and comprises a combination of bFGF, BMP-2, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a.
  • the defined cytokine cocktail is termed “C2” and comprises a combination of bFGF, BIO, BMP-2, BMP-4, EGF, EPO, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a.
  • the defined cytokine cocktail is termed “G5” and comprises a combination of bFGF, BMP-4, DKK-1, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-6, PD-ECGF, SDF-1 ⁇ , and WNT-3a.
  • the defined cytokine cocktail is termed “G7” and comprises a combination of bFGF, BMP-2, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, LIF, and WNT-3a.
  • the defined cytokine cocktail is termed “G4” and comprises a combination of bFGF, BMP-2, EGF, IL-11, PD-ECGF, and WNT-3a.
  • the defined cytokine cocktail is termed “C8” and comprises a combination of bFGF, BMP-2, EGF, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, IL-5, LIF, PD-ECGF, and WNT-3a.
  • the cytokine cocktail comprises bFGF in combination with at least growth factor selected from the group consisting of a WNT signaling agonist, TGF- ⁇ , and EGF.
  • the cytokine cocktail comprises a WNT signaling agonist in combination with at least one growth factor selected from the group consisting of bFGF, TGF- ⁇ , and EGF.
  • the cytokine cocktail comprises TGF- ⁇ in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and EGF.
  • the cytokine cocktail comprises EGF in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and TGF- ⁇ .
  • the cytokine cocktail comprises at least two growth factors selected from the group consisting of bFGF, TGF- ⁇ , EGF, and a WNT signaling agonist.
  • the WNT signaling agonist is WNT-3a; in other of these embodiments, the WNT signaling agonist is BIO.
  • adding complexity to a media composition comprising at least two growth factors selected from the group consisting of bFGF, TGF- ⁇ , EGF, and a WNT signaling agonist with the addition of other factors enhances MSC expansion.
  • bFGF bFGF
  • TGF- ⁇ TGF- ⁇
  • EGF EGF
  • WNT signaling agonist a WNT signaling agonist
  • media containing combinations of bFGF, TGF- ⁇ , EGF, and a WNT signaling agonist produce MSC expansion
  • the addition of other signaling molecules such as BMPs, soluble ECMs, and other cytokines and factors can further enhance MSC expansion.
  • the G2 combination of growth factors allows for rapid expansion of MSCs, at similar or even better than commercially available serum containing media.
  • this expansion when compared to serum containing media that is greater than 10 days old, will outperform the serum containing media.
  • the MSCs expanded in G2 media are still pluripotent and have been demonstrated to differentiate into both bone and fat.
  • the ability of these MSCs to maintain a pluripotent phenotype is significant as this is evidence that serum can be removed from the culture during expansion and the cells have not changed considerably nor have they been pushed towards specific differentiated phenotypes that would prevent the use of the expanded cells from being used in different therapeutic applications.
  • the defined cytokine combinations are added to a serum-free base medium to provide a serum-free culture medium suitable for expanding MSCs.
  • a serum-free nutritive medium suitable for the culture of MSCs may be used.
  • the base medium is as described in Lennon et al. (1995) Exp. Cell Res., 219:211-222, comprising Dulbecco's Modified Eagle's Medium (DMEM), MCDB-201 (Sigma-Aldrich (St. Louis, Mo.)), ascorbic acid 2-phosphate, dexamethasone, linoleic acid-bovine serum albumin, insulin, transferrin, and sodium selenite.
  • DMEM Dulbecco's Modified Eagle's Medium
  • MCDB-201 Sigma-Aldrich (St. Louis, Mo.)
  • ascorbic acid 2-phosphate dexamethasone
  • dexamethasone linoleic acid-bovine serum albumin
  • the base medium comprises a ratio of DMEM to MCDB-201 of 60:40, the concentration of ascorbic acid 2-phosphate is 1 ⁇ 10 4 M, the concentration of dexamethasone is 1 ⁇ 10 ⁇ 9 M, linoleic acid-bovine serum albumin is in an amount of about 0.1%, insulin is in an amount of about 5 ⁇ g/mL, transferrin is in an amount of about 5 ⁇ g/mL, and sodium selenite is in an amount of about 5 ng/mL (see Table 2, herein below).
  • Many different base media could be used, and human or other animal-derived components may be substituted such as human albumin for bovine albumin, or human insulin.
  • the final concentration of cytokines in the serum-free cell culture medium can range from about 1 femtogram/ml to about 1 picogram/ml to about 1 nanogram/ml to about 1 microgram/ml to about 1 milligram/ml.
  • the concentration of any one of the cytokines can be about 1 pg/ml, about 5 pg/ml, about 10 pg/ml, about 15 pg/ml, about 20 pg/ml, about 25 pg/ml, about 30 pg/ml, about 35 pg/ml, about 40 pg/ml, about 45 pg/ml, about 50 pg/ml, about 55 pg/ml, about 60 pg/ml, about 65 pg/ml, about 70 pg/ml, about 75 pg/ml, about 80 pg/ml, about 85 pg/ml, about 90 pg/ml, about 95 pg/ml, about 100 pg/ml, about 110 pg/ml, about 120 pg/ml, about 130 pg/ml, about 140 pg/ml, about 150 pg/ml, about 160
  • the serum-free cell culture medium comprises a concentration of BDNF from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of bFGF from about 1 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of BIO of about 1 pM to about 1 pM. In another embodiment, the serum-free cell culture medium comprises a concentration of BMP-2 from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of BMP-4 from about 10 pg/ml to about 500 ng/ml.
  • the serum-free cell culture medium comprises a concentration of DKK-1 from about 10 pg/ml to about 10 ⁇ g/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of EGF from about 10 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of erythropoietin from about 0.0001 units/ml to about 50 units/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of fibronectin from about 10 pg/ml to about 100 ⁇ g/ml.
  • the serum-free cell culture medium comprises a concentration of Flt-3/Flk-2 ligand from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of G-CSF from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IGF-1 from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-11 from about 1 pg/ml to about 100 ng/ml.
  • the serum-free cell culture medium comprises a concentration of IL-3 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-5 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-6 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of LIF from about 10 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of PD-ECGF from about 10 pg/ml to about 500 ng/ml.
  • the serum-free cell culture medium comprises a concentration of SCF from about 10 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of SDF-1 ⁇ from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of TGF- ⁇ from about 10 fg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of thrombospondin from about 10 pg/ml to about 1 ⁇ g/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of WNT-3a from about 10 pg/ml to about 500 ng/ml.
  • the serum-free cell culture medium comprises BDNF in an amount of about 1 ng/ml; bFGF in an amount of about 2.5 ng/ml; BMP-4 in an amount of about 0.5 ng/ml; erythropoietin in an amount of about 0.05 U/ml; fibronectin in an amount of about 10 ng/ml; Flt-3/Flk-2 ligand in an amount of about 5 ng/ml; IGF-1 in an amount of about 2.5 ng/ml; IL-11 in an amount of about 0.1 ng/ml; IL-3 in an amount of about 1 ng/ml; IL-5 in an amount of about 0.1 ng/ml; IL-6 in an amount of about 0.2 ng/ml; PD-ECGF in an amount of about 2 ng/ml; SCF in an amount of about 2 ng/ml; SDF-1 ⁇ in an amount of about 3 ng/ml; and thrombospondin in an amount of
  • cytokines for use in the present invention may be concentrated and, in some instances, lyophilized before addition to the base medium to obtain the examples of final concentrations listed above.
  • mass weight added to culture will depend on the specific biological activity of the cytokine preparation. Bioassays to determine the biological potency of cytokines are well known in the art. Therefore, where the biological activity is correlated to a mass weight, then biological “units” as defined by the assay are used.
  • Populations of MSCs that have been expanded using the serum-free cell culture system of the present invention comprise cells expressing cell surface markers of interest.
  • the term “cell surface marker” or “marker” is intended to mean a protein that is expressed on the surface of a cell, which can be detected using specific antibodies.
  • the expanded MSCs of the present invention express markers reported for commercial MSCs, including CD166, CD44, CD105, and CD29.
  • the expanded MSCs of the invention may also express combinations of these cell markers, and may express CD73, CD90, CD 106, CD 146, or any combination thereof.
  • the expression of a cell surface marker defines non-mesenchymal cell populations such as mature blood cells and hematopoietic stem cells, for example: CD3; CD14; CD19; CD34; CD42a; CD45; and any combinations of these markers.
  • the term “substantially free” is intended to mean that less than 5%, less than 4%, less than 3%, less than 2%, or less than 1% of the cells within the population express the marker of interest.
  • the expanded cell population is substantially free of T-cells (expressing the CD3 antigen), B-cells (expressing the CD 19 antigen), or mature granulocytes, NK lymphocytes, or macrophages (expressing the CD16 antigen).
  • substantially proportion is intended to mean that at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or up to 100% of the cells express the marker of interest.
  • substantially diminished is intended to mean that at least less than 10%, at least less than 5%, or at least less than 1% of cells express the marker of interest.
  • cells with increased cell surface marker expression can be detected and isolated by any means including flow cytometric sorting, antibody panning, and the like.
  • flow cytometric analysis one of skill in the art must first set a detection threshold for fluorescence. In setting the threshold, a negative control sample population will be recorded and a gate will be set around the population of interest according to the desired forward scatter (FSC) and side scatter (SSC). The detection threshold is then adjusted so that 97% or more of the cells do not fluoresce. Once the detection threshold is set, the fluorescence of cell population of interest is recorded. A cell is considered “positive for expression” when it expresses the marker of interest, whether a protein or a gene.
  • any method may be used to determine expression such as gene expression profiles, FACS, and the like.
  • the term “ + ” indicates that the cell is positive for expression of the marker of interest.
  • the term “ ⁇ ” indicates that the cell does not have detectable levels of expression of the marker of interest.
  • the antibodies used to detect various lineages may be conjugated to different fluorochromes. These include phycobiliproteins, e.g., phycoerythrin and allophycocyanins; fluorescein; and Texas red. Dead cells can also be detected using dyes that selectively accumulate in dead cells (e.g., propidium iodide and 7-amino actinomycin D).
  • MSCs are characterized by a capacity for self-renewal. Further, the cells are characterized by an ability to commit to multilineage development.
  • multilineage development is intended that the cell is capable of differentiating into cell types of mesenchymal origin. These cells have a capability of limited self renewal and are constrained to committed development to various mesenchymal tissues such as bone, cartilage, fat, tendon, muscle, and marrow stroma (Pittenger et al. (1999) Science. 284:5411 143).
  • Stem cell function can be assayed using both in vitro and in vivo methods.
  • In vitro testing comprises culturing MSCs in semi-solid medium similar to hematopoetic stem cells. Along with colony forming assays, MSCs may be tested for multilineage potential by culturing these cells in the presence of factors that send them down specific pathways such as bone, fat, and cartilage.
  • Media for differentiating expanded MSCs are well known in the art (Pittenger and Martin (2004) Circ. Res. 95:9-20), and are commercially available through several sources such as Cambrex (Baltimore, Md.), Stem Cell Technologies (Vancouver, B. C., Canada), and R&D Systems (Minneapolis, Minn.).
  • Expanded cells are cultured either on surfaces such as culture dishes with these media for 7-30 days for bone, fat, or muscle phenotypes, or in pellets in the bottom or tubes or flasks for differentiating down cartilage phenotypes.
  • tissue specific tests are performed such as alkaline phosphatase enzyme, osteocalcin or osteopontin for bone, oil O red staining for lipid accumulation in MSCs that differentiate towards an adipocyte lineage.
  • an “expression profile” comprises one or more values corresponding to a measurement of the relative abundance of a gene expression product. Such values may include measurements of RNA levels or protein abundance.
  • the expression profile can comprise values representing the measurement of the transcriptional state or the translational state of the gene (see U.S. Pat. Nos. 6,040,138, 5,800,992, 6,020135, 6,344,316, and 6,033,860).
  • the transcriptional state of a sample includes the identities and relative abundance of the RNA species, especially mRNAs present in the sample. Preferably, a substantial fraction of all constituent RNA species in the sample are measured, but at least a sufficient fraction to characterize the transcriptional state of the sample is measured.
  • the transcriptional state can be conveniently determined by measuring transcript abundance by any of several existing gene expression technologies.
  • Translational state includes the identities and relative abundance of the constituent protein species in the sample. As is known to those of skill in the art, the transcriptional state and translational state are related.
  • microarrays can be used to measure the values to be included in the expression profiles. Microarrays are particularly well suited for this purpose because of the reproducibility between different experiments.
  • DNA microarrays provide one method for the simultaneous measurement of the expression levels of large numbers of genes. Each array consists of a reproducible pattern of capture probes attached to a cell culture support. Labeled RNA or DNA is hybridized to complementary probes on the array and then detected by laser scanning. Hybridization intensities for each probe on the array are determined and converted to a quantitative value representing relative gene expression levels (see U.S. Pat. Nos. 6,040,138, 5,800,992 and 6,020,135, 6,033,860, and 6,344,316). High-density oligonucleotide arrays are particularly useful for determining the gene expression profile for a large number of RNAs in a sample.
  • Array is intended to mean a cell culture support or substrate with peptide or nucleic acid probes attached to the cell culture support or substrate. Arrays typically comprise a plurality of different nucleic acid or peptide capture probes that are coupled to a surface of a substrate in different, known locations. These arrays, also described as “microarrays” or colloquially “chips,” have been generally described in the art, for example, in U.S. Pat. Nos. 5,143,854, 5,445,934, 5,744,305, 5,677,195, 6,040,193, 5,424,186, 6,329,143, and 6,309,831 and Fodor et al. (1991) Science 251:767-77.
  • the methods of the invention comprise culturing or expanding MSCs from a stem cell source in a serum-free cell culture system that comprises a serum-free cell culture medium disclosed herein above, which comprises a cytokine cocktail and base medium.
  • a serum-free cell culture medium disclosed herein above, which comprises a cytokine cocktail and base medium.
  • Methods of cell culture are well known in the art (see, e.g., Waymouth (1984) “Preparation and Use of Serum-free Culture Media,” in Cell Culture Methods for Molecular and Cell Biology, Vol. 1, Methods for Preparation of Media, Supplements, and Substrata for Serum - Free Animal Cell Culture , eds. Barnes et al. (Alan R. Liss), pp. 23-68).
  • cell-culture is intended to mean the expansion or maintenance of cells in an artificial in vitro environment while maintaining a pluripotent phenotype. It is to be understood that the term “cell culture” is a generic term and may be used to encompass the cultivation not only of individual cells but also tissues, organ systems, or whole organisms.
  • cells derived from a stem cell source are seeded onto a two-dimensional or three-dimensional cell culture surface described herein and contacted with the serum-free cell culture medium of the invention.
  • the medium may initially contain the cytokine cocktail as well as the base medium, or the cytokine cocktail can be added later.
  • Cells are then incubated with the serum-free cell culture medium comprising the cytokine cocktail and cell culture support at a temperature suitable for cell growth (for some embodiments about 37° C.), for at least about 24 hours, at least about 48 hours, at least about 72 hours, at least about 96 hours, at least about 120 hours, at least about 144 hours, at least about 168 hours, at least about 192 hours, or longer.
  • Cells may be harvested in any manner known in the art including but not limited to centrifugation after decanting non-adherent cells, trypsinizing adherent cells, or scraping cells from the surface of the cell culture support.
  • the MSCs used in culture can include cells derived from any stem cell source, such as umbilical cord, umbilical cord blood, placenta, embryonic stem cells, adipose tissue, bone marrow, or other tissue-specific mesenchyme. These samples may be fresh, frozen, or refrigerated. Methods of freezing cells are well known in the art (see, e.g., Doyle et al. (1995) Cell & Tissue Culture: Laboratory Procedures (John Wiley & Sons, Chichester)).
  • Cryopreservation of stem cells prior to culture or cryopreservation of expanded cells disclosed herein may be carried out according to known methods.
  • cells may be suspended in a “freezing medium” such as, for example, culture medium further comprising 10% dimethylsulfoxide (DMSO), with or without 5-10% glycerol, at a density, for example, of about 1-2 ⁇ 10 6 cells/ml.
  • the cells may be dispensed into glass or plastic vials, which are then sealed and transferred to a freezing chamber of a programmable or passive freezer.
  • the optimal rate of freezing may be determined empirically. For example, a freezing program that gives a change in temperature of about ⁇ 1° C./min through the heat of fusion may be used.
  • vials containing the cells Once vials containing the cells have reached ⁇ 80° C., they may be transferred to a liquid nitrogen storage area.
  • Cryopreserved cells may be stored for a period of years.
  • freshly isolated cells from any stem cell source may be cryopreserved to constitute a bank of cells, portions of which may be withdrawn by thawing and then used to produce the expanded cells of the invention as needed.
  • Thawing may be carried out rapidly, for example, by transferring a vial from liquid nitrogen to a 37° C. water bath. The thawed contents of the vial may be immediately transferred under sterile conditions to a culture vessel containing an appropriate medium such as nutritive medium.
  • the cells Once in culture, the cells may be examined daily, for example, with an inverted microscope to detect cell proliferation, and subcultured as soon as they reach an appropriate density.
  • Cells may be withdrawn from a cell bank as needed, and used for the production of new stem cells or tissue either in vitro, for example, as a three-dimensional scaffold culture, as described below, or in vivo, for example, by direct administration of cells to the site where tissue reconstitution or repair is needed.
  • the expanded MSCs of the invention may be used to reconstitute or repair tissue in a subject where the cells were originally isolated from that subject's own bone marrow or other tissue (i.e., autologous cells).
  • the expanded MSCs disclosed herein may be used as ubiquitous donor cells to reconstitute or repair tissue in any subject (i.e., heterologous cells).
  • Non-adherent cells may be removed after 1 to 3 days. Adherent, spindle shaped fibroblast-like MSCs are kept and expanded. After several hours to days, non-adherent cells are washed away and the MSCs remain. It has also been reported that MSCs may be obtained from cord blood, placenta and adipose tissues.
  • Cultured MSCs can be further isolated using any method known in the art. Generally, the MSCs are contacted with monoclonal antibodies directed to cell surface antigens and either positively or negatively selected. Such techniques for selection are well known in the art and include sorting by immunomagnetic beads, by complement mediated lysis, by “panning” with antibody attached to a solid matrix, agglutination methods, magnetic activated cell sorting (MACS), or fluorescence activated cell sorting (FACS).
  • monoclonal antibodies directed to cell surface antigens either positively or negatively selected.
  • Such techniques for selection are well known in the art and include sorting by immunomagnetic beads, by complement mediated lysis, by “panning” with antibody attached to a solid matrix, agglutination methods, magnetic activated cell sorting (MACS), or fluorescence activated cell sorting (FACS).
  • the expanded MSCs of the invention have broad application in treating and ameliorating disease and injury.
  • the expanded MSCs of the invention are useful in many therapeutic applications including repairing, reconstituting, and regenerating tissue as well as gene delivery.
  • the MSCs of the invention can comprise both lineage-committed and uncommitted cells; thus, both cell types can be used together to accomplish multiple therapeutic goals, even simultaneously in some embodiments.
  • the expanded MSCs of the invention can be used directly as stem cell transplants or be used in stem cell grafts either in suspension or on a cell culture support scaffold as noted herein above.
  • expanded MSCs of the invention can be placed in a carrier medium before administration.
  • expanded MSCs of the invention can be administered in any physiologically acceptable medium, intravascularly, including intravenously, although they may also be introduced into other convenient sites such as into the bone marrow, where the cells may find an appropriate site for regeneration and differentiation.
  • the MSCs may be introduced by any method including injection, catheterization, or the like.
  • Drugs of interest include 5-fluorouracil and growth factors including cytokines such as IL-2, IL-3, G-CSF, M-CSF, GM-CSF, IFN ⁇ , and erythropoietin.
  • the MSCs can be injected with collagen, Matrigel, alone or with other hydrogels.
  • Administered MSCs may also comprise a mixture of cells herein described and additional cells of interest. Additional cells of interest include, without limitation, differentiated liver cells, differentiated cardiac muscle, differentiated pancreatic cells, and the like. These combinations are particularly useful when the expanded MSCs of the invention are seeded on a three-dimensional scaffold, hydrogel, or without carrier, as disclosed herein above.
  • the expanded MSC population of the invention can be used to repair or reconstitute damaged or diseased mesenchymal tissues, such as the heart, the pancreas, the liver, fat tissue, bone, cartilage, endothelium, nerves, astrocytes, dermis, and the like.
  • the expanded MSCs migrate to or are placed at the site of injury, they can differentiate to form new tissues and supplement organ function.
  • the cells are used to promote vascularization and, therefore, improve oxygenation and waste removal from tissues.
  • the expanded MSCs of the invention can be used to increase function of differentiated tissues and organs such as the ischemic heart as in cardiac failure or ischemic nerves as in stroke. Therefore, the expanded MSCs of the invention are useful in any disease where cellular function or organ function has been decremented.
  • “Supplementing a damaged organ” or “supplementing organ function” is intended to mean increasing, enhancing, or improving the function of an organ that is operating at less than optimum capacity.
  • the term is used to refer to a gain in function so that the organ is operating at a physiologically acceptable capacity for that subject.
  • the physiological acceptable capacity for an organ from a child e.g., a kidney or heart
  • the entire organ or part of the organ can be supplemented.
  • the supplementation results in an organ with the same physiological response as a non-damaged or non-diseased organ.
  • an organ is supplemented in capacity when it is functioning to at least at about 10% of its natural capacity.
  • the expanded MSCs of the present invention can be used for implantation by contacting the cells with a tissue-engineered construct prior to grafting as noted herein above. The construct containing these cells is then implanted into a host in need of such a graft.
  • the cells of the invention are particularly useful for promoting bone and cartilage generation, thereby facilitating tissue regeneration and repair. These cells may also be used in applications relating to graft versus host disease.
  • the expanded MSCs of the invention may also be useful as vasculature-promoting stem cells. “Vascularization promoting” or “vasculature promoting” is intended to mean promoting the growth of new vessels (vasculogenesis) or inducing outgrowth from existing vessels (angiogenesis), or any combination thereof.
  • the expanded cells of the invention may comprise both lineage-committed and uncommitted MSCs.
  • the MSCs may give rise to both the differentiated tissue and act as the source for the vasculature-promoting stem cells.
  • the MSCs may give rise to both the differentiated tissue and act as the source for the cardiovascular-promoting stem cells.
  • the MSCs may give rise to both the differentiated tissue and act as the source for the bone and cartilage-promoting stem cells.
  • the source of differentiated tissues may comprise cells or tissue from the intended graft recipient or another donor.
  • the cell or tissue source may be differentiated prior to implantation.
  • pancreatic beta cells can be differentiated using conditions described for embryoid body formation as detailed in Itskovich-Eldor et al. (2000) Mol. Med. 6:88.
  • the vasculature-promoting stem cells may be contacted with angiogenic growth factors such as VEGF and bFGF. These stem cells may be contacted with angiogenic growth factors prior to or after seeding onto the scaffold before engrafting the tissue-engineered construct into a damaged organ.
  • angiogenic growth factors such as VEGF and bFGF.
  • cytokine-impregnated polymers can release the angiogenic growth factor, such as VEGF and bFGF, over time.
  • the scaffold may be programmed to drive expansion and proliferation of seeded cells (see, e.g., U.S. Patent Application Publication No. 20040063206).
  • microspheres or microcarriers may be contacted with the vasculature-promoting stem cells and placed at a target site.
  • Microsphere-based scaffolds are well known in the art (see, e.g., Mahoney and Saltzman (2001) Nature Biotech. 19:934).
  • the cardiovascular- or bone and cartilage-promoting stem cells may be contacted with growth factors such as BDNF, bFGF, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF 1- ⁇ , TGF- ⁇ , thrombospondin, and WNT signaling agonists, including but not limited to BIO and WNT proteins, WNT-1, WNT-2, WNT-2b, WNT-3, WNT-3a, WNT-4, WNT-5a, WNT-5b, WNT-6, WNT-7a, WNT-7b, WNT-8a, WNT-8b, WNT-9a, WNT-9b, WNT-10a, WNT-10b, WNT-11, and WNT-16.
  • the WNT protein is WNT-3a.
  • the expanded MSCs of the invention can also be used for gene therapy in patients in need thereof.
  • more mature lineage-committed cells will be useful, especially where transient gene expression is needed or where gene transduction is facilitated by the maturation and division of the cells.
  • some retroviral vectors require that the cell be cycling for the gene to be integrated. Methods for transducing stem and progenitor cells to deliver new and therapeutic genes are known in the art.
  • kits useful for promoting the attachment, survival, and/or proliferation of MSCs comprising a serum-free cell culture system of the invention.
  • a kit comprises a serum-free cell culture medium and a cell culture surface described herein, and can comprise one or more other components or reagents suitable for culturing the cell.
  • the serum-free cell culture medium comprises any of the cytokine cocktails described herein, for example, the cocktail shown in Table 1 below, and a base medium, for example, the base medium shown in Table 2 below, and a cell culture surface comprising a cell culture support bound to a CAR, which in turn is bound to an ECM protein.
  • the ECM is a mixture of collagen I and fibronectin.
  • the kits comprise these components and also comprise one or more reagents for measuring cell proliferation in the culture.
  • Such kits have many uses, which will be evident to one of skill in the art. For example, they can be used to propagate MSCs to be used in methods of cell therapy. Such kits could be of commercial use, e.g., in high-throughput drug studies.
  • Base Medium for Serum-Free Cell Culture Medium Base Medium Component Amount DMEM/MCDB-201 Media 60:40 Ascorbic acid 2-phosphate 1 ⁇ 10 ⁇ 4 M Dexamethasone 1 ⁇ 10 ⁇ 9 M Linoleic Acid-Bovine Serum 0.1% Albumin Insulin 5 ⁇ g/mL Transferrin 5 ⁇ g/mL Sodium selenite 5 ng/mL
  • MSCs along with complete growth medium were purchased from Cambrex Biosciences (Baltimore, Md.). Frozen cells were thawed and cultured following manufacturer's instruction. After reaching ⁇ 90% confluency, MSCs were washed once with PBS, removed from the culture surface using Trypsin/EDTA and replated at a density of 1600 cells/well (for 96 well plates) or at a density of 50,000 cells/well (for 6-well plates) which corresponds to the manufacturers recommended seeding density of 5000 cells/cm 2 .
  • MSCs induced to form adipocytes were seeded at 7000 cells/well and cultured in Stem Cell Technologies (Vancouver, B. C., Canada) adipogenic medium supplement.
  • MSCs induced to form osteoblasts were seeded at 1000 cells/well using Cambrex Biosciences (Baltimore, Md.) osteogenic media supplements.
  • MSCs expanded in serum-free media of the present invention on CAR Col 1/FN surface for 7 days remained multipotent.
  • MSCs expanded in culture environments of the present invention were removed from the serum-free media and then cultured in either adipogenic (fat) or osteogenic (bone) induction media (per manufacturer's suggestions).
  • MSCs expanded in the serum-free environments of the present invention were able to differentiate towards both adipogenic (see FIG. 2 ) and osteogenic (see FIG. 3 ) lineages.
  • MSCs cultured in serum-free environments for 7 days retain cell surface markers showing equivalency to Cambrex cell populations expanded in serum-containing media.
  • MSCs cultured in serum-free environments lacked CD45 marker expression, indicating that these cells do not differentiate towards hematopoietic lineages.
  • MSCs cultured in three different media all expand better on a Col 1/FN CAR surface as compared to TCPS surfaces (see FIG. 4 ). Also, MSCs cultured in G2 serum-free medium expand to equivalent levels as MSCs cultured in serum-containing medium.
  • the G2 serum-free medium served as a control for this optimization experiment. Screening plates were prepared using 96-well plates containing 30 factors in 60 different compositions. One thousand six hundred (1600) MSCs were incubated in each well for 7 days. Best media compositions were selected based on the average number of live cells obtained from the repeats from each screen.
  • the C6 well contained cytokines BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1 ⁇ , and WNT-3a.
  • the D3 well contained cytokines bFGF, BMP-2, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a
  • the C2 well contained cytokines bFGF, BIO, BMP-2, BMP-4, EGF, EPO, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a.
  • the G5 well contained cytokines bFGF, BMP-4, DKK-1, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-6, PD-ECGF, SDF-1 ⁇ , and WNT-3a.
  • Cytokine compositions of wells C6, D3, C2, and G5 are referred to hereinafter as “C6,” “D3,” “C2,” and “G5,” respectively.
  • G2 serum free medium controls and 10% serum-containing complete medium (see FIG. 5 ).
  • serum free media compositions contain 22, 11, 11, and 10 growth factor/cytokine additives, respectively.
  • serum free media G2 and D3 both performed as well or better than serum-containing complete medium controls and significantly outperform a previously published serum free MSC expansion medium (Lennon et al. (1995) Exp. Cell Res. 219:211-222; U.S. Pat. No. 5,908,782).
  • MSCs Three of these serum-free media compositions (D3, C2, and G5) were used to confirm MSCs retained multipotentiality. MSCs were cultured in one of the three serum-free media for 7 days. Expanded MSCs were removed from the serum-free media and then cultured in either adipogenic (fat) or osteogenic (bone) induction media (per manufacturer's suggestions). MSCs remained multipotential and differentiated towards both adipogenic and osteogenic lineages in all three serum-free media, each comparable to the manufacturer's recommended serum-containing complete medium controls (see FIGS. 6A and 6B ).
  • the G7 well contained cytokines bFGF, BMP-2, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, LIF, and WNT-3a.
  • the G4 well contained cytokines bFGF, BMP-2, EGF, IL-11, PD-ECGF, and WNT-3a.
  • the C8 well contained cytokines bFGF, BMP-2, EGF, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, IL-5, LIF, PD-ECGF, and WNT-3a.
  • Cytokine compositions of wells G7, G4, and C8 are referred to hereinafter as “G7,” “G4,” and “C8,” respectively.
  • Media compositions G7, G4, and C8 all expanded MSCs as well as or better than serum-containing controls (see FIG. 7 ).
  • MSCs were cultured in: 1) the G4 serum-free medium composition; 2) G4+TGF- ⁇ ; 3) bFGF in base medium only; 4) EGF in base medium only; 5) TGF- ⁇ in base medium only; 6) base medium alone; and 7) serum-containing medium.
  • MSCs cultured in G4 expanded as well or better than serum-containing medium
  • MSCs cultured in G4+TGF- ⁇ far outperformed any other media composition, nearly triple the serum-containing medium controls (see FIG. 8 ).
  • the single factors, while promoting MSC expansion did not perform as well as serum in expanding MSCs.
  • a serum-free medium comprising base medium and WNT-3a, bFGF, and TGF- ⁇ was compared to media compositions containing the same components but adding one, two, three, four, five, six, or seven additional cytokines that include EGF, fibronectin, thrombospondin, BMP-2, BMP-4, IL-11, and PD-ECGF in their ability to expand MSCs in culture. Adding one, two, three, four, five, six, or seven growth factors to the serum free medium comprising base medium and WNT-3a, bFGF, and TGF- ⁇ had a positive effect on MSC expansion.
  • EGF EGF-induced fibroblast proliferation
  • the addition of EGF to the three growth factors already present in the serum-free medium resulted in approximately a 20% increase in MSC expansion over compositions that did not contain EGF, and both BMPs on top of EGF further enhanced expansion.
  • the addition of all seven cytokines to WNT-3a, bFGF, and TGF- ⁇ gave the best expansion, demonstrating that building complexity into the serum free environments may further enhance MSC expansion.
  • the synergies between WNT-3a, bFGF, EGF, and TGF- ⁇ were found to have the greatest effects on MSC serum-free expansion on collagen 1+fibronectin surfaces.
  • a serum-free medium comprising base medium was compared to media comprising the base medium plus various combinations of WNT-3a, BIO, bFGF, EGF, and TGF- ⁇ in their ability to expand MSCs in culture (see FIG. 9 ).
  • the combinations tested were: base medium alone (no growth factors), Cambrex Biosciences (Baltimore, Md.) serum-containing medium on TCPS, base medium+bFGF+TGF- ⁇ , base medium+WNT-3a+bFGF, base medium+bFGF+EGF, base medium+WNT-3a+TGF- ⁇ , base medium+WNT-3a+EGF, base medium+EGF+TGF- ⁇ , base medium+WNT-3a, base medium+bFGF+EGF+TGF- ⁇ , base medium+WNT-3a+bFGF+EGF+TGF- ⁇ , and base medium+bFGF+EGF+TGF- ⁇ +BIO.
  • the growth factor combinations that produced the greatest MSC expansion were base medium+EGF+TGF- ⁇ , base medium+bFGF+TGF- ⁇ , base medium+bFGF+EGF+TGF- ⁇ , base medium+WNT-3a+bFGF+EGF+TGF- ⁇ , and base medium+bFGF+EGF+TGF- ⁇ +BIO.

Abstract

Compositions and methods for promoting mesenchymal stem cell expansion while maintaining a pluripotent phenotype are disclosed. Serum-free cell culture systems and kits and methods of use for mesenchymal stem cell expansion are provided. Methods also comprise the use of the expanded mesenchymal stem cells to treat various disorders or diseases, particularly those of the cardiovascular system, bone, or cartilage.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 60/571,212, filed May 14, 2004, the content of which is herein incorporated by reference it its entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to serum-free cell culture systems that provide for mesenchymal stem cell expansion while maintaining a pluripotent phenotype, and methods of use for the expanded mesenchymal stem cell populations.
  • BACKGROUND OF THE INVENTION
  • Mesenchymal stem cells (MSCs) are present in adult tissues and constitute a population of cells that can be isolated, expanded in culture, and characterized in vitro and in vivo (Pittenger and Martin (2004) Circ. Res. 95:9-20). MSCs are able to differentiate into multiple cell lineages, including osteoblasts, chondrocytes, endothelial cells, and neuronal cells, and can express phenotypic characteristics of endothelial, neural, smooth muscle, skeletal myoblast, and cardiac myocyte cells (Kassem et al. (2004) Basic Clin. Pharmacol. Toxicol. 95:209-214; Pittenger and Martin (2004) Circ. Res. 95:9-20). In recent years, MSCs have generated a high level of experimental and clinical interest due to their potential for a range of therapeutic uses including repair of damaged or diseased tissues (Baksh et al. (2004) J. Cell. Mol. Med. 8:301-316; Barry and Murphy (2004) Int. J. Biochem. Cell Bio. 36:568-584).
  • Typically, MSCs do very poorly in serum-free environments because they detach and die in culture. These MSCs can be maintained in an attached state in vitro with minimal serum (e.g., <1%), although such an environment provides little stimulation for MSCs to proliferate and grow. Although serum-free cell culture environments have been described for MSC expansion (Lennon et al. (1995) Exp. Cell Res. 219:211-222; U.S. Pat. No. 5,908,782), the current industry standard still contains a large amount of serum.
  • In addition to the desirability of serum-free environments for the expansion of MSCs, serum-free media and culture systems have great utility in the field of cellular therapy. The creation of highly defined environments for cell expansion is of great importance for quality purposes, and serum levels are typically very ill-defined (see, e.g., U.S. Pat. No. 5,908,782). In addition, there is a risk of Bovine Spongiform Encephalopathy (BSE) contamination in patients receiving cells cultured in the presence of serum. Such a risk raises the possibility that the FDA will not allow therapies involving cells cultured in the presence of animal sera.
  • For the aforementioned reasons, the development of new serum-free cell culture systems for the expansion of MSCs is therefore desirable.
  • SUMMARY OF THE INVENTION
  • Compositions and methods for promoting mesenchymal stem cell (MSC) expansion while maintaining a pluripotent phenotype are provided. The compositions include serum-free cell culture systems for MSC expansion that comprise a serum-free cell culture medium and a two-dimensional or three-dimensional cell culture surface. In the serum-free cell culture system of the present invention, at least one insoluble substrate protein is presented from the cell culture surface. In one embodiment, the cell culture surface comprises a cell culture support bound to a cell adhesion resistant (CAR) material, which in turn is bound to at least one insoluble substrate protein. Insoluble substrate proteins for use in the present invention include extracellular matrix (ECM) proteins such as fibronectin, laminin, hyaluronic acid (HA), vitronectin, collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof. In one embodiment, the cell culture surface comprises a cell culture support bound to a cell adhesion resistant (CAR) material, which in turn is bound to at least one insoluble substrate protein, for example, at least one ECM protein. The serum-free cell culture medium is a solution that comprises a mixture of soluble MSC growth-promoting factors. Compositions further include kits comprising serum-free cell culture media and a two-dimensional or three-dimensional cell culture surface suitable for MSC expansion.
  • Methods of the present invention comprise the use of these serum-free cell culture systems to promote the expansion of MSCs. Further methods comprise the use of these serum-free cell culture systems and expanded MSCs for cell transplantation or to engineer tissues to treat various disorders or diseases, including those of the cardiovascular system, muscle, ligament, bone, tendon, cartilage, nervous system, blood, immune system, liver, or pancreas. Further methods comprise the use of these serum-free cell culture systems to promote the expansion of MSCs within primary aspirates from whole bone marrow such that the MSCs are co-cultured with non-MSCs present in the primary aspirates.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows expansion of total mesenchymal stem cells (MSCs) over time in the G2 serum-free culture system and collagen 1+fibronectin surface of the present invention (BDT Environment) versus expansion in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic.
  • FIG. 2 shows the adipocyte differentiation capacity of MSCs expanded in the G2 serum-free medium and collagen 1+fibronectin surface (BDT Environment) or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic.
  • FIG. 3 shows the bone differentiation capacity of MSCs expanded in the G2 serum-free medium collagen 1+fibronectin surfaces (BDT Environment) or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic.
  • FIG. 4 shows a comparison of the number of hMSCs after culture in a serum-free base medium (BDTM) with or without growth factors on either a tissue culture polystyrene (TCPS) or cell adhesion resistant (CAR) surface.
  • FIG. 5 shows a comparison of various serum-free compositions and collagen 1+fibronectin surfaces for MSC expansion that are labeled C6, D3, C2, and G5, compared to G2 medium and a serum containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 6A shows the adipocyte differentiation capacity of MSCs expanded in D3, C2, and G5 serum-free culture media and collagen 1/fibronectin surfaces or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (10% FBS CM). FIG. 6B shows the bone differentiation capacity of MSCs expanded in D3, C2, and G5 serum-free culture media and collagen 1/fibronectin surfaces or expanded in Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 7 shows a comparison of further refined serum-free composition for MSC expansion labeled G7, G4, and C8 and cultured on collagen 1/fibronectin surfaces compared to expansion in Cambrex Biosciences (Baltimore, Md.) serum-containing medium (10% FBS CM).
  • FIG. 8 shows a comparison of MSC expansion in various media conditions and collagen 1/fibronectin surfaces, including G4 serum-free medium, G4+TGFβ, FGF only, EGF only, and TGFβ only, as compared to basal medium and Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (10% FBS CM).
  • FIG. 9 shows a comparison of MSC expansion in various media conditions where selected growth factor combinations were added to basal medium, as compared to basal medium alone and Cambrex Biosciences (Baltimore, Md.) serum-containing medium on tissue culture plastic (TCPS). Growth factor combinations were bFGF+TGF-β, WNT-3a+bFGF, bFGF+EGF, WNT-3a+TGF-β, WNT-3a+EGF, EGF+TGF-β, WNT-3a alone, bFGF+EGF+TGF-β, WNT-3a+bFGF+EGF+TGF-β, and bFGF+EGF+TGF-β+BIO.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to compositions and methods for promoting mesenchymal stem cell (MSC) expansion while maintaining the pluripotent phenotype of these cells. Serum-free cell culture systems for MSC expansion are provided. These cell culture systems comprise serum-free cell culture medium and a two-dimensional or three-dimensional cell culture surface. The serum-free cell culture medium is a solution that comprises a mixture of soluble MSC growth-promoting and self-renewal factors. In the serum-free cell culture system of the present invention, at least one insoluble substrate protein is presented from the cell culture surface. In one embodiment, the cell culture surface comprises a cell culture support bound to a cell adhesion resistant (CAR) material, which in turn is bound to at least one insoluble substrate protein. Insoluble substrate proteins may include but are not limited to extracellular matrix (ECM) proteins such as fibronectin, laminin, hyaluronic acid (HA), vitronectin, or collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof. Other compositions of the present invention include kits comprising the serum-free cell culture systems of the invention, which are suitable for MSC expansion.
  • Methods of the present invention are directed to the use of these serum-free cell culture systems to promote the expansion of MSCs and to engineer tissues. The expanded MSCs of the present invention can be used to treat various disorders or diseases, particularly those of the cardiovascular system, muscle, ligament, bone, tendon, cartilage, nervous system, blood, immune system, liver, or pancreas.
  • The term “mesenchymal” is intended to mean such cells as bone marrow, endothelial cells, epithelial cell progenitors, cardiomyocytes, astrocytes, neurons, chondrocytes, osteoblasts, pancreatic cells, hepatocytes, and other cells of mesenchymal origin. As used herein, the term “mesenchymal stem cell” or “MSC” refers to a cell that gives rise to a cell of mesenchymal lineage.
  • The term “expanded” is intended to mean that the resultant cell population is derived from ex vivo culture of stem cells in media compositions comprising mixtures of cytokines, where the outgoing (cultured) number of cells exceeds the ingoing (non-cultured) number of cells. The term “expanded” is not to be construed or limited by any mechanism or theory of cellular origin and may comprise cells that originate de novo in culture.
  • The serum-free cell culture system of the present invention comprises a serum-free cell culture medium and a cell culture surface. In one embodiment, at least one insoluble substrate protein is presented from the cell culture surface, meaning that the insoluble substrate protein is bound, adsorbed, linked, attached or in some way associated with the cell culture surface. Suitable insoluble substrate proteins include the ECM proteins disclosed herein. In one such embodiment, the insoluble substrate protein presented from the cell culture surface is a combination of collagen I and fibronectin. In another embodiment, the cell culture surface comprises a cell culture support bound to a cell adhesion resistant material, which in turn is bound to at least one insoluble substrate protein. The cell culture support may be solid or porous, polymer, metal, glass, ceramic, or combinations thereof. Culturing of MSCs in the presence of this cell culture surface and the serum-free cell culture medium unexpectedly provides for expansion of these cells while maintaining their pluripotent phenotype.
  • As used herein, the term cell adhesion resistant (CAR) refers to a material that, when present on a surface, prevents, inhibits, or reduces the non-specific binding (adhesion) of cells, proteins, or polypeptides found on cell surfaces. CAR materials are resistant to adhesion of mammalian cells and also to microorganisms. CAR materials are sometimes referred to as non-fouling substrates, inert coatings, low affinity reagents, or non-adhesive coatings.
  • The cell culture support to which is bound the CAR material may be two- or three-dimensional, solid or porous, and may be constructed of any of a variety of materials, including natural polymers, synthetic polymers, hydrogels, metals, ceramics, and inorganic or organic-inorganic composites. The cell culture support may be shaped using methods such as, for example, solvent casting, compression molding, filament drawing, meshing, leaching, weaving, and coating.
  • Examples of suitable polymers and hydrogels include, but are not limited to, collagen, glycosaminoglycan (GAG)-based materials, alginate, hyaluronate poly(alpha esters) such as poly(lactate acid), poly(glycolic acid), polyorthoesters and polyanhydrides and their copolymers, cellulose ether, cellulose, cellulosic ester, fluorinated polyethylene, phenolic, poly-4-methylpentene, polyacrylonitrile, polyamide, polyamideimide, polyacrylate, polybenzoxazole, polycarbonate, polycyanoarylether, polyestercarbonate, polyether, polyetheretherketone, polyetherimide, polyetherketone, polyethersulfone, polyethylene, polyfluoroolefin, polylmide, polyolefin, polyoxadiazole, polyphenylene oxide, polyphenylene sulfide, polypropylene, polystyrene, polysulfide, polysulfone, polytetrafluoroethylene, polythioether, polytriazole, polyurethane, polyvinylidene fluoride, regenerated cellulose, urea-formaldehyde, or copolymers or physical blends of these materials.
  • Further examples include synthetic polymers, such as polylactic acid (PLA), polyglycolic acid (PGA), poly(orthoester), polysebacic acid polymers (PSA), poly(lactic-co-glycolic) acid copolymers (PLGA), poly(lactic-co-sebacic) acid copolymers (PLSA), poly(glycolic-co-sebacic) acid copolymers (PGSA), hydrogels such as polyhydroxyethylmethacrylate (poly-HEMA) or polyethylene oxide-polypropylene oxide copolymer, polyethylegylcols (PEG), polyvinylalchols (PVA), polyvinylpyrrolidone (PVP) and polyhydroxyalkanoate (PHA). PHAs and their production are described in, for example, PCT Publication Nos. WO 99/14313, WO 99/32536 and WO 00/56376. Hybrid materials containing naturally derived and synthetic polymer materials (e.g., PGA and PLGA) may also be used. Non-limiting examples of such materials are disclosed in Chen et al. (2000) Advanced Materials 12:455-457.
  • Other polymers useful in the present invention include polymers or copolymers of caprolactones, carbonates, amides, amino acids, orthoesters, acetals, cyanoacrylates and degradable urethanes, as well as copolymers of these with straight chain or branched, substituted or unsubstituted, alkanyl, haloalkyl, thioalkyl, aminoalkyl, alkenyl, or aromatic hydroxy- or di-carboxylic acids. In addition, the biologically important amino acids with reactive side-chain groups, such as lysine, arginine, aspartic acid, glutamic acid, serine, threonine, tyrosine and cysteine, or their enantiomers, may be included in copolymers with any of the aforementioned materials.
  • In one embodiment, polymer surfaces are selected from the group consisting of polystyrene, polyethylene, polypropylene, polyethylene terephthalate, polytetrafluoroethylene, polylactide, and cellulose. Silicone polymers such as polydimethylsiloxane (PDMS) are also used.
  • Inorganic composites include, e.g., calcium phosphate ceramics, bioglasses and bioactive glass-ceramics, in particular composites combining calcium hydroxyapatite and silicon stabilized tricalcium phosphate. Among preferred cell culture supports are polystyrene (PS), polypropylene, polyethylene, polyethylene terephthalate, polytri- or tetra-fluoroethylene, polyhexafluoropropylene, polyvinyl chloride, polyvinylidine fluoride, polyactide, cellulose, glass, or a ceramic.
  • Any suitable CAR material, many of which are known to those skilled in the art, may be bound to the cell culture support. Typical CAR materials include hyaluronic acid (HA) or a derivative thereof, alginic acid (AA) or a derivative thereof, poly-HEMA, polyethylene glycol (PEG), glyme or a derivative thereof, polypropylacrylamide, polyisopropylacrylamide, or a combination of these compounds. In one embodiment, the CAR material is HA.
  • In some embodiments, one or more of a proteoglycan, a biglycan, a glycosaminoglycan, or Matrigel™ may be bound to the CAR material.
  • In other embodiments, proteins or other substances may be bound to the CAR material either covalently or non-covalently, but are preferably covalently bound. The proteins or other substances comprise, for example, ECM proteins or polycationic polymers. ECM proteins for use in the present invention may include fibronectin, laminin, hyaluronic acid (HA), vitronectin, or collagen proteins such as collagen I, collagen II, collagen III, collagen IV, collagen V, and collagen VI, or any combination thereof. Various types of covalent bonds can form, some of which are discussed in more detail in co-pending, commonly assigned U.S. Patent Application Publication Nos. 20050036980, 20040062882, 20030113813, and 20030113812, all of which are incorporated herein by reference. These applications also disclose other aspects of making and using surfaces that include cell culture supports with bound CAR materials and ECM proteins.
  • In one embodiment, one or more ECM proteins as disclosed herein and/or one or more polycationic polymer are bound to the CAR material. In one embodiment, a mixture of collagen I and fibronectin is bound to the CAR material.
  • The ECM protein(s) can be in the form of a naturally occurring polypeptide (protein), a recombinant polypeptide, or a synthetic or semi-synthetic polypeptide, including any fragment of the peptide or protein, or combination thereof. The terms “polypeptide” and “protein” are used interchangeably herein.
  • Methods of cloning, expressing, and purifying polypeptides, such as ECM proteins, are conventional, as are methods of generating synthetic or semi-synthetic polypeptides. ECM proteins can also be obtained from commercial sources.
  • ECM proteins or polycationic polymers can be bound to the CAR material either covalently or non-covalently (for example, passively adsorbed, such as by electrostatic forces, ionic or hydrogen bonds, hydrophilic or hydrophobic interactions, Van der Waals forces, etc.). In a preferred embodiment, the binding is covalent. U.S. Patent Application Publication Nos. 20040062882, 20030113813, and 20030113812 describe such covalent binding of molecules to CAR surfaces.
  • Methods of making surfaces in which a CAR material is bound to a cell culture support, and in which ECM proteins, polycationic polymers, or the like, are bound to the CAR material, are described in detail in U.S. Patent Application Publication Nos. 20040062882, 20030113813, and 20030113812. In brief, one method of attaching a CAR material to a cell culture support comprises treating the cell culture support with an oxidizing plasma, and binding the CAR material to the treated cell culture support. Another method of attaching a CAR material to a cell culture support comprises treating the cell culture support with an oxidizing plasma; exposing the treated cell culture support to a polycationic polymer with amino groups (such as PEI, PDL, poly-L-lysine (PLL), poly-L-ornithine (PLO), poly-D-ornithine (PDO), poly(vinylamine) (PVA) or poly(allylamine) (PAA), in one embodiment, PEI or PDL to form an intermediate layer, and binding the CAR material to the intermediate layer. Methods of binding an ECM or a polycationic polyaminoacid to a CAR material are conventional. These include, for example, sodium periodate oxidation and reductive amination, EDC/NHS carbodiimide coupling, or the like.
  • This invention also speaks to the use of flexible substrates in culture. For example, Flexercell culture systems from Flexcell International Corporation are able to apply tensile, compressive, or shear stresses to cultured cells. See, for example, U.S. Pat. Nos. 4,789,601, 4,822,741, 4,839,280, 6,037,141, 6,048,723, and 6,218,178. U.S. Pat. No. 6,057,150 discloses the application of a biaxial strain to an elastic membrane that may be coated with extracellular matrix proteins and covered with cultured cells. U.S. Pat. No. 6,107,081 discloses another system in which a unidirectional cell stretching device comprising an elastic strip is coated with an extracellular matrix on which cells are cultured and stretched. A flexible substrate can be deformed easily and in a controlled manner, and also supports cell adhesion and growth comparable to conventional cell culture substrates. Silicones, such as poly(dimethyl siloxane) (PDMS), are particularly suitable for this application because they are not only highly flexible but also provide optical clarity that allows microscopic observation of the cell cultures.
  • Another aspect of the invention is a method of making the above cell culture surface of the invention, where the method comprises (a) attaching a CAR material to a cell culture support, and (b) attaching to the CAR material collagen I and fibronectin, or biologically active fragment or variant thereof, and, optionally, one or more other ECM proteins (or a biologically active fragment or variant of the ECM protein) and/or one or more polycationic polymers. Any of the ECM proteins or polycationic polymers disclosed herein, or others, may be used.
  • In one embodiment, the CAR material is attached to the cell culture support by treating the cell culture support with an oxidizing plasma, and binding the CAR material to the treated cell culture support. In another embodiment, the CAR material is attached to the cell culture support by treating the cell culture support with an oxidizing plasma; exposing the treated cell culture support to a polycationic polymer with amino groups to form an intermediate layer; and binding the CAR material to the intermediate layer. Preferably, the polycationic polymer is polyethylene imine (PEI) or poly-D-lysine (PDL).
  • In a one embodiment, a CAR material, such as HA, is bonded directly to polymeric nitrogen-containing surfaces. Examples of such surfaces are ammonia plasma-treated polymers and Primaria ™-treated polystyrene (PS) surfaces, and poly D-lysine coated surfaces. Polymeric substrates suitable for use in the invention include polystyrene, polypropylene, polyethylene terephthalate, polylactide, cellulose, and the like, though polystyrene is preferred.
  • HA is immobilized directly on an intermediate polyamine layer, e.g., polyethyleneimine, poly-D-Lysine, or poly-L-lysine, or directly bound to the polymer with the HA bound to that intermediate layer).
  • The use of plasma techniques are familiar to those of skill in the art (see, for example, Garbassi et al., (1994) Polymer Surfaces, from Physics to Technology (Wiley, Chichester), and Inagaki (1996) Plasma Surface Modification and Plasma Polymerization (Technomic Publishing Company, Lancaster). In the present invention, the plasma treatment process may be any process that is capable of causing nitrogen to be incorporated onto the surface of the polymer article resulting in reactive amine or other nitrogen-containing groups, including direct as well as remote plasma treatment methods. Examples of suitable plasma treatments are ones using reactive gases such as nitrogen, nitrogen oxide, nitrogen dioxide or ammonia in the gas phase, alone or in mixture with air, argon, or other inert gases, and may be preceded or followed by treatments employing argon or other inert gases. The plasma maybe sustained over the full treatment time or maybe administered in pulses. Preferably, the plasma gas is ammonia, and treatment is performed with a power charge of between 1 and 400 W, preferably between 10 and 150 W, a pressure between 10 mtorr and 10 ton, and a treatment time between 1 second and 1 hour, preferably between 10 seconds and 30 minutes.
  • Plasma-treated polystyrene can be prepared, for example by pumping the treatment chamber to a 0.3 mTorr base pressure, establishing a 200 mTorr argon atmosphere, and applying a 60 sec argon plasma treatment, followed by a 120 sec, 375 mTorr NH3 plasma treatment at 95 W. Other suitable treatments will be known to those of skill in the art. Following plasma treatment of the surface to be coated, the plasma-treated surface may be exposed to an aqueous solution containing HA or a derivative thereof, or alginic acid (alginate; AA) in the presence of a condensing agent such as a carbodiimide, for example, ethyldimethylaminopropyl carbodiimide (EDC), in aqueous solution, or dicyclohexylcarbodiimide (DCC), in organic solvents. The term “expose” or “exposing” as used herein is intended to include any type of contact made between a liquid and a solid, for example by pipetting, pouring, spraying, dripping, immersing, pouring, dipping, injecting, etc., without limitation.
  • HA is an anionic polysaccharide composed of repeating units of beta-1,4-glucuronatebeta-1,3-N-acetylglucosamine. A reactive —COO group is present on every repeat unit of HA that can be utilized to covalently couple HA to an amine containing surface using methods described herein. In this manner, a condensing agent such as EDC activates the —COO groups present in HA, creating a reactive ester intermediate (ester (o-acylisourea) intermediate). This intermediate is highly unstable and subject to hydrolysis, leading to the cleaving off of the activated ester intermediate, forming an isourea, and regenerating the —COO group. To stabilize this reactive ester intermediate, and increase reaction yield, a molecule able to enhance the reaction promoted by EDC should also be present. Such stabilizing compounds are generally selected from the class of N-hydroxysuccinimides and aryl or heterocyclic derivatives thereof. Preferred N-hydroxysuccinimides include, but are not limited to, N-hydroxy-succinimide (NHS), hydroxy-sulfosuccinimide (sulfo-NHS) or hydroxybenzotriazole hydrate. Although not intended to be bound to a particular theory, it is believed that attachment of HA to the amine containing polymer surface occurs through a mechanism wherein (for example) EDC and NHS combine to create an active ester polysaccharide with a carboxyl group capable of coupling to an amine. When coupling occurs, NHS is released. Other compounds known in the art that are able to react with EDC in this manner and which serve as reactive intermediate ester stabilizing compounds should also be effective in the invention. HA covalently bonded to plasma-treated polystyrene in this way prevents attachment of any of a number of types of cells, including NIH3T3 and osteoblast MC3T3 cells as well as MSC's and a variety of primary and stem cells.
  • Suitable derivatives of HA that may be used in the invention will be known to the skilled artisan, and are described, for example, in U.S. Pat. No. 4,851,521. These include partial esters of HA with alcohols of the aliphatic, araliphatic, cycloaliphatic and heterocyclic series, and salts of such partial esters with inorganic or organic bases. Similar derivatives of alginic acid should also be useful. Further, other plasma treatment methods for producing surfaces with amine and other nitrogen-containing groups are also suitable, and are known to those of skill in the art.
  • The cell culture surfaces for use in the serum-free cell culture systems include but are not limited to standard tissue culture vessels and two-dimensional surfaces, including sheets, slides, culture dishes, culture flasks, bags, culture bottles, or multiwell dishes. Alternatively, three-dimensional cell culture surfaces, such as microcarriers or three dimensional (3-D) scaffolds, including but not limited to foams, hydrogels, or fiber meshes may be used for generating a three-dimensional cell culture, tissue, or organ. “Three-dimensional scaffold” refers herein to a 3-D porous template that provides a very high surface area to volume ratio for cell culture. These scaffolds may be used for initial cell attachment and expansion, or subsequently for tissue formation either in vitro or in vivo. A 3-D scaffold according to this invention comprises base materials (described below), a CAR layer and bound thereto one or more insoluble substrate proteins, such as the ECMs disclosed herein, and, optionally, other substances, which promote or enhance cell attachment, growth, migration, and/or differentiation. In one embodiment, the scaffold is seeded with MSCs and contacted with the serum-free cell culture medium described herein below in order to permit cell growth and differentiation in a structural environment that more closely mimics the in vivo setting. Cells derived therefrom can be isolated from the scaffold or can be implanted with or without the scaffold into a suitable location in the body of a mammal, preferably a human patient. However, the use of this technology could easily be translated to non-human mammals such as cats, dogs, horses, and the like.
  • The shape and dimensions of the 3-D scaffold are determined based on the organ being replaced or supplemented, and the type of scaffold material being used to create the construct. For example, if a polymeric scaffold is used for heart tissue replacement or supplementation, the dimension of the polymeric scaffold can vary in terms of width and length of the polymeric scaffold. One of skill in the art recognizes that the size and dimensions of the polymeric scaffold will be determined based on the area of the organ being replaced or supplemented. Furthermore, other suitable articles may constitute a cell culture support surface of the present invention, including medical devices, extracorporeal devices and artificial joints, tubes, sutures, stents, orthopedic devices, vascular grafts, membranes, films, biosensors, or microparticles.
  • One embodiment includes a method of forming tissue-engineered constructs using a 3-D scaffold material or other suitable article that comprises the CAR surface onto which one or more insoluble substrate proteins, for example, the ECMs disclosed herein, are attached, either covalently or non-covalently. Such a scaffold in combination with the serum-free cell culture medium disclosed herein supports the maturation, development and differentiation, of additional cultured MSCs in vitro to form components of adult tissues analogous to their in vivo counterparts.
  • The tissue-engineered constructs, in one embodiment, are created using scaffold materials disclosed herein as the substrate onto which cells are deposited and cultured in the presence of the serum-free cell culture medium described herein below, and on which cells are grown and adhere. The scaffold allows optimum cell-cell interactions, thereby allowing a more natural formation of cellular phenotypes and a tissue microenvironment. The scaffold also allows MSCs to continue to grow actively, proliferate, and differentiate to produce a tissue-engineered construct that is also capable of supporting the growth, proliferation, and differentiation of additional cultured cell populations, if needed.
  • In one embodiment, the scaffold is biocompatible and conducive to cell attachment and subsequent tissue growth. Other surface properties can be modified to suit the intended application without altering other properties of the scaffold such as its mechanical strength or thermal properties. Useful surface modifications could include, for example, changes in chemical group functionality, surface charge, hydrophobicity, hydrophilicity, and wettability. The CAR surface technology can easily be translated to the surfaces of 3-D scaffolds. Such surface modifications are well known in the art. Sterilization is performed prior to seeding the scaffold with cells. Heat sterilization is often impractical since the heat treatment could deform the device, especially if the materials have a melting temperature below that required for the heat sterilization treatment. For example, cold ethylene oxide gas, vapor hydrogen peroxide treatments can be used for sterilization.
  • The 3-D scaffolds of the present invention comprise any suitable base material for construction, including the polymeric materials disclosed herein above. The polymeric matrix can be fabricated to have a controlled pore structure that allows nutrients from the serum-free cell culture medium described herein below to reach the deposited cell population but prevent cultured cells from migrating through the pores. In vitro cell attachment and cell viability can be assessed using scanning electron microscopy, histology and quantitative assessment with radioisotopes.
  • The polymeric matrix can be shaped into any number of desirable configurations to satisfy any number of overall system, geometry, or space restrictions. The polymeric matrix can be shaped to different sizes to conform to the organs of different sized patients. Thus, the tissue-engineered construct can be flat, tubular, or of complex geometry. The shape of the construct will be decided by its intended use. The construct can be implanted to repair, supplement, or replace diseased or damaged parts of organs.
  • In one embodiment, the scaffold base material is a hydrogel composed of crosslinked polymer networks that are typically insoluble or poorly soluble in water, but can swell to an equilibrium size in the presence of excess water. For example, the MSCs can be placed in a hydrogel and the hydrogel injected into desired locations within the organ. The hydrogel compositions can include, without limitation, for example, poly(esters), poly(hydroxy acids), poly(lactones), poly(amides), poly(ester-amides), poly(amino acids), poly(anhydrides), poly(ortho-esters), poly(carbonates), poly(phosphazines), poly(thioesters), polysaccharides and mixtures thereof. Furthermore, the compositions can also include, for example, a poly(hydroxy) acid including poly(alpha-hydroxy) acids and poly(betahydroxy) acids. Such poly(hydroxy) acids include, for example, polylactic acid, polyglycolic acid, polycaproic acid, polybutyric acid, polyvaleric acid, and copolymers and mixtures thereof. Due to the unique properties of hydrogels and their potential applications in such areas as controlled drug delivery, various types of hydrogels have been synthesized and characterized. The matrix materials of the present invention encompass both conventional foam or sponge materials and the so-called hydrogel sponges (see, e.g., U.S. Pat. No. 5,451,613).
  • Mesenchymal stem cells grown on the scaffold materials in the presence of the serum-free cell culture medium disclosed herein in accordance with the present invention may grow in multiple layers, forming a cellular structure that resembles physiologic conditions found in vivo. The scaffold can support the proliferation of different types of cells and the formation of a number of different tissues. Examples include, but are not limited to, kidney, heart, skin, liver, pancreas, adrenal and neurological tissue, as well as tissues of the gastrointestinal and genitourinary tracts, and the circulatory system.
  • The cells grown or expanded on the aforementioned scaffold can be used, alone or in combination with the scaffold, in a variety of applications. For example, the scaffold and cells can be implanted into a subject. Implants can be used to replace or supplement existing tissue, for example, by treating a subject with a cardiovascular disorder by replacing or supplementing natural cardiovascular tissue. The subject can be monitored after implantation for amelioration of the cardiovascular disorder. A three-dimensional biocompatible scaffold may be brought into contact with vasculature-promoting expanded MSCs of the invention and then brought into contact with a host tissue at a target site (e.g., within the organ) or where the organ tissue is grown on the scaffold prior to implantation. The graft is then able to grow and proliferate within the target site and replace or supplement the depleted activity of the organ. The construct can be added at a single location in the host or, alternatively, a plurality of constructs can be created and added to multiple sites in the host.
  • The term “target site” as used herein refers to a region in the host or organ that requires replacement or supplementation. The target site can be a single region in the organ or host, or can be multiple regions in the organ or host. In some embodiments, the supplementation or replacement results in the same physiological response as a normal organ.
  • In another embodiment, the scaffold is created using parts of a natural decellularized organ. Parts of organs can be decellularized by removing the entire cellular and tissue content from the organ (see, e.g., U.S. Pat. No. 6,479,064). The term “decellularized” or “decellularization” as used herein refers to a biostructure (e.g., an organ, or part of an organ) from which the cellular and tissue content has been removed leaving behind an intact acellular infrastructure. The process of decellularization removes the specialized tissue, leaving behind the complex three-dimensional network of connective tissue. The connective tissue infrastructure is primarily composed of collagen. The decellularized structure provides a matrix material onto which different cell populations can be infused. Decellularized biostructures can be rigid, or semi-rigid, having an ability to alter their shapes. Culture and construction of decellularized biostructures can be performed, for example, as describe in U.S. Pat. No. 6,479,064, which is herein incorporated by reference in its entirety.
  • The two-dimensional and three-dimensional cell culture surfaces serve as a substrate onto which MSCs can be seeded and cultured in the presence of a serum-free cell culture medium disclosed herein. In this manner, seeded MSCs can be expanded ex vivo by contacting them with a serum-free cell culture medium that comprises a defined cytokine cocktail. In one embodiment, the defined combinations comprise at least 2, at least 3, at least, 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, or at least 15 cytokines from the group comprising brain-derived neurotrophic factor (BDNF), basic fibroblast growth factor (bFGF), bone morphogenic protein 2 (BMP-2), bone morphogenic protein 4 (BMP-4), dickkopf 1 (DKK-1), epidermal growth factor (EGF), erythropoietin (EPO), fibronectin, Flt-3/Flk-2 ligand, granulocyte colony stimulating factor (G-CSF), insulin-like growth factor (IGF-1), interleukin-11 (IL-11), interleukin-3 (IL-3), interleukin-5 (IL-5), interleukin-6 (IL-6), leukemia inhibitory factor (LIF), platelet-derived endothelial cell growth factor (PD-ECGF), stem cell factor (SCF), stromal cell derived factor 1-α (SDF 1-α), transforming growth factor β (TGF-β), thrombospondin, and a WNT signaling agonist. WNT signaling agonists include, but are not limited to (2′Z,3′E)-6-bromoindirubin-3′-oxime (BIO) and WNT proteins including WNT-1, WNT-2, WNT-2b, WNT-3, WNT-3a, WNT-4, WNT-5a, WNT-5b, WNT-6, WNT-7a, WNT-7b, WNT-8a, WNT-8b, WNT-9a, WNT-9b, WNT-10a, WNT-10b, WNT-11, and WNT-16. In one embodiment, the WNT protein is WNT-3a.
  • In one embodiment, the defined cytokine cocktail is termed “G2” and comprises a combination of fibronectin, SDF-11, IL-6, SCF, IL-5, BDNF, PD-ECGF, IL-11, IL-3, EPO, Flt-3/Flk-2 ligand, BMP-4, thrombospondin, IGF-1, and bFGF.
  • In another embodiment, the defined cytokine cocktail is termed “C6” and comprises a combination of BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1α, and WNT-3a.
  • In another embodiment, the defined cytokine cocktail is termed “D3” and comprises a combination of bFGF, BMP-2, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a.
  • In another embodiment, the defined cytokine cocktail is termed “C2” and comprises a combination of bFGF, BIO, BMP-2, BMP-4, EGF, EPO, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a.
  • In another embodiment, the defined cytokine cocktail is termed “G5” and comprises a combination of bFGF, BMP-4, DKK-1, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-6, PD-ECGF, SDF-1α, and WNT-3a.
  • In another embodiment, the defined cytokine cocktail is termed “G7” and comprises a combination of bFGF, BMP-2, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, LIF, and WNT-3a.
  • In another embodiment, the defined cytokine cocktail is termed “G4” and comprises a combination of bFGF, BMP-2, EGF, IL-11, PD-ECGF, and WNT-3a.
  • In another embodiment, the defined cytokine cocktail is termed “C8” and comprises a combination of bFGF, BMP-2, EGF, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, IL-5, LIF, PD-ECGF, and WNT-3a.
  • In another embodiment, the cytokine cocktail comprises bFGF in combination with at least growth factor selected from the group consisting of a WNT signaling agonist, TGF-β, and EGF. In another embodiment, the cytokine cocktail comprises a WNT signaling agonist in combination with at least one growth factor selected from the group consisting of bFGF, TGF-β, and EGF. In another embodiment, the cytokine cocktail comprises TGF-β in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and EGF. In another embodiment, the cytokine cocktail comprises EGF in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and TGF-β. In another embodiment, the cytokine cocktail comprises at least two growth factors selected from the group consisting of bFGF, TGF-β, EGF, and a WNT signaling agonist. In some of these embodiments, the WNT signaling agonist is WNT-3a; in other of these embodiments, the WNT signaling agonist is BIO.
  • In one embodiment, adding complexity to a media composition comprising at least two growth factors selected from the group consisting of bFGF, TGF-β, EGF, and a WNT signaling agonist with the addition of other factors enhances MSC expansion. For example, although media containing combinations of bFGF, TGF-β, EGF, and a WNT signaling agonist produce MSC expansion, the addition of other signaling molecules such as BMPs, soluble ECMs, and other cytokines and factors can further enhance MSC expansion.
  • The G2 combination of growth factors allows for rapid expansion of MSCs, at similar or even better than commercially available serum containing media. In particular, this expansion, when compared to serum containing media that is greater than 10 days old, will outperform the serum containing media. Furthermore, the MSCs expanded in G2 media are still pluripotent and have been demonstrated to differentiate into both bone and fat. The ability of these MSCs to maintain a pluripotent phenotype is significant as this is evidence that serum can be removed from the culture during expansion and the cells have not changed considerably nor have they been pushed towards specific differentiated phenotypes that would prevent the use of the expanded cells from being used in different therapeutic applications.
  • The defined cytokine combinations are added to a serum-free base medium to provide a serum-free culture medium suitable for expanding MSCs. Any serum-free nutritive medium suitable for the culture of MSCs may be used. In one embodiment, the base medium is as described in Lennon et al. (1995) Exp. Cell Res., 219:211-222, comprising Dulbecco's Modified Eagle's Medium (DMEM), MCDB-201 (Sigma-Aldrich (St. Louis, Mo.)), ascorbic acid 2-phosphate, dexamethasone, linoleic acid-bovine serum albumin, insulin, transferrin, and sodium selenite. In a further embodiment, the base medium comprises a ratio of DMEM to MCDB-201 of 60:40, the concentration of ascorbic acid 2-phosphate is 1×104 M, the concentration of dexamethasone is 1×10−9 M, linoleic acid-bovine serum albumin is in an amount of about 0.1%, insulin is in an amount of about 5 μg/mL, transferrin is in an amount of about 5 μg/mL, and sodium selenite is in an amount of about 5 ng/mL (see Table 2, herein below). Many different base media could be used, and human or other animal-derived components may be substituted such as human albumin for bovine albumin, or human insulin. The ability to substitute non-animal derived components for animal derived is known in the field, and several of these components are commercially available, for example from Cambrex Biosciences (Baltimore, Md.) or Sigma-Aldrich (St. Louis, Mo.).
  • The final concentration of cytokines in the serum-free cell culture medium can range from about 1 femtogram/ml to about 1 picogram/ml to about 1 nanogram/ml to about 1 microgram/ml to about 1 milligram/ml. In some embodiments, the concentration of any one of the cytokines can be about 1 pg/ml, about 5 pg/ml, about 10 pg/ml, about 15 pg/ml, about 20 pg/ml, about 25 pg/ml, about 30 pg/ml, about 35 pg/ml, about 40 pg/ml, about 45 pg/ml, about 50 pg/ml, about 55 pg/ml, about 60 pg/ml, about 65 pg/ml, about 70 pg/ml, about 75 pg/ml, about 80 pg/ml, about 85 pg/ml, about 90 pg/ml, about 95 pg/ml, about 100 pg/ml, about 110 pg/ml, about 120 pg/ml, about 130 pg/ml, about 140 pg/ml, about 150 pg/ml, about 160 pg/ml, about 170 pg/ml, about 180 pg/ml, about 190 pg/ml, about 200 pg/ml, about 210 pg/ml, about 220 pg/ml, about 230 pg/ml, about 240 pg/ml, about 250 pg/ml, about 260 pg/ml, about 270 pg/ml, about 280 pg/ml, about 290 pg/ml, about 300 pg/ml, about 310 pg/ml, about 320 pg/ml, about 330 pg/ml, about 340 pg/ml, about 350 pg/ml, about 360 pg/ml, about 370 pg/ml, about 380 pg/ml, about 390 pg/ml, about 400 pg/ml, about 410 pg/ml, about 420 pg/ml, about 430 pg/ml, about 440 pg/ml, about 450 pg/ml, about 460 pg/ml, about 470 pg/ml, about 480 pg/ml, about 490 pg/ml, about 500 pg/ml, about 510 pg/ml, about 520 pg/ml, about 530 pg/ml, about 540 pg/ml, about 550 pg/ml, about 560 pg/ml, about 570 pg/ml, about 580 pg/ml, about 590 pg/ml, about 600 pg/ml, about 610 pg/ml, about 620 pg/ml, about 630 pg/ml, about 640 pg/ml, about 650 pg/ml, about 660 pg/ml, about 670 pg/ml, about 680 pg/ml, about 690 pg/ml, about 700 pg/ml, about 710 pg/ml, about 720 pg/ml, about 730 pg/ml, about 740 pg/ml, about 750 pg/ml, about 760 pg/ml, about 770 pg/ml, about 780 pg/ml, about 790 pg/ml, about 800 pg/ml, about 810 pg/ml, about 820 pg/ml, about 830 pg/ml, about 840 pg/ml, about 850 pg/ml, about 860 pg/ml, about 870 pg/ml, about 880 pg/ml, about 890 pg/ml, about 900 pg/ml, about 910 pg/ml, about 920 pg/ml, about 930 pg/ml, about 940 pg/ml, about 950 pg/ml, about 960 pg/ml, about 970 pg/ml, about 980 pg/ml, about 990 pg/ml, about 1 ng/ml, about 1.5 ng/ml, about 2 ng/ml, about 2.5 ng/ml, about 3 ng/ml, about 3.5 ng/ml, about 4 ng/ml, about 4.5 ng/ml, about 5 ng/ml, about 6 ng/ml, about 7 ng/ml, about 8 ng/ml, about 9 ng/ml, about 10 ng/ml, about 15 ng/ml, about 20 ng/ml, about 25 ng/ml, about 30 ng/ml, about 35 ng/ml, about 40 ng/ml, about 45 ng/ml, about 50 ng/ml, about 55 ng/ml, about 60 ng/ml, about 65 ng/ml, about 70 ng/ml, about 75 ng/ml, about 80 ng/ml, about 85 ng/ml, about 90 ng/ml, about 95 ng/ml, about 100 ng/ml, about 110 ng/ml, about 120 ng/ml, about 130 ng/ml, about 140 ng/ml, about 150 ng/ml, about 160 ng/ml, about 170 ng/ml, about 180 ng/ml, about 190 ng/ml, about 200 ng/ml, about 210 ng/ml, about 220 ng/ml, about 230 ng/ml, about 240 ng/ml, about 250 ng/ml, about 260 ng/ml, about 270 ng/ml, about 280 ng/ml, about 290 ng/ml, about 300 ng/ml, about 310 ng/ml, about 320 ng/ml, about 330 ng/ml, about 340 ng/ml, about 350 ng/ml, about 360 ng/ml, about 370 ng/ml, about 380 ng/ml, about 390 ng/ml, about 400 ng/ml, about 410 ng/ml, about 420 ng/ml, about 430 ng/ml, about 440 ng/ml, about 450 ng/ml, about 460 ng/ml, about 470 ng/ml, about 480 ng/ml, about 490 ng/ml, about 500 ng/ml, about 510 ng/ml, about 520 ng/ml, about 530 ng/ml, about 540 ng/ml, about 550 ng/ml, about 560 ng/ml, about 570 ng/ml, about 580 ng/ml, about 590 ng/ml, about 600 ng/ml, about 610 ng/ml, about 620 ng/ml, about 630 ng/ml, about 640 ng/ml, about 650 ng/ml, about 660 ng/ml, about 670 ng/ml, about 680 ng/ml, about 690 ng/ml, about 700 ng/ml, about 710 ng/ml, about 720 ng/ml, about 730 ng/ml, about 740 ng/ml, about 750 ng/ml, about 760 ng/ml, about 770 ng/ml, about 780 ng/ml, about 790 ng/ml, about 800 ng/ml, about 810 ng/ml, about 820 ng/ml, about 830 ng/ml, about 840 ng/ml, about 850 ng/ml, about 860 ng/ml, about 870 ng/ml, about 880 ng/ml, about 890 ng/ml, about 900 ng/ml, about 910 ng/ml, about 920 ng/ml, about 930 ng/ml, about 940 ng/ml, about 950 ng/ml, about 960 ng/ml, about 970 ng/ml, about 980 ng/ml, about 990 ng/ml, or about 1000 ng/ml.
  • In one embodiment, the serum-free cell culture medium comprises a concentration of BDNF from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of bFGF from about 1 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of BIO of about 1 pM to about 1 pM. In another embodiment, the serum-free cell culture medium comprises a concentration of BMP-2 from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of BMP-4 from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of DKK-1 from about 10 pg/ml to about 10 μg/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of EGF from about 10 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of erythropoietin from about 0.0001 units/ml to about 50 units/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of fibronectin from about 10 pg/ml to about 100 μg/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of Flt-3/Flk-2 ligand from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of G-CSF from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IGF-1 from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-11 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-3 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-5 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of IL-6 from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of LIF from about 10 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of PD-ECGF from about 10 pg/ml to about 500 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of SCF from about 10 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of SDF-1α from about 1 pg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of TGF-β from about 10 fg/ml to about 100 ng/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of thrombospondin from about 10 pg/ml to about 1 μg/ml. In another embodiment, the serum-free cell culture medium comprises a concentration of WNT-3a from about 10 pg/ml to about 500 ng/ml.
  • In one embodiment, the serum-free cell culture medium comprises BDNF in an amount of about 1 ng/ml; bFGF in an amount of about 2.5 ng/ml; BMP-4 in an amount of about 0.5 ng/ml; erythropoietin in an amount of about 0.05 U/ml; fibronectin in an amount of about 10 ng/ml; Flt-3/Flk-2 ligand in an amount of about 5 ng/ml; IGF-1 in an amount of about 2.5 ng/ml; IL-11 in an amount of about 0.1 ng/ml; IL-3 in an amount of about 1 ng/ml; IL-5 in an amount of about 0.1 ng/ml; IL-6 in an amount of about 0.2 ng/ml; PD-ECGF in an amount of about 2 ng/ml; SCF in an amount of about 2 ng/ml; SDF-1α in an amount of about 3 ng/ml; and thrombospondin in an amount of about 10 ng/ml.
  • Those skilled in the art will recognize that cytokines for use in the present invention may be concentrated and, in some instances, lyophilized before addition to the base medium to obtain the examples of final concentrations listed above. Those skilled in the art will also recognize that the mass weight added to culture will depend on the specific biological activity of the cytokine preparation. Bioassays to determine the biological potency of cytokines are well known in the art. Therefore, where the biological activity is correlated to a mass weight, then biological “units” as defined by the assay are used.
  • Populations of MSCs that have been expanded using the serum-free cell culture system of the present invention comprise cells expressing cell surface markers of interest. The term “cell surface marker” or “marker” is intended to mean a protein that is expressed on the surface of a cell, which can be detected using specific antibodies. For example, the expanded MSCs of the present invention express markers reported for commercial MSCs, including CD166, CD44, CD105, and CD29. The expanded MSCs of the invention may also express combinations of these cell markers, and may express CD73, CD90, CD 106, CD 146, or any combination thereof. In some instances, the expression of a cell surface marker defines non-mesenchymal cell populations such as mature blood cells and hematopoietic stem cells, for example: CD3; CD14; CD19; CD34; CD42a; CD45; and any combinations of these markers.
  • The term “substantially free” is intended to mean that less than 5%, less than 4%, less than 3%, less than 2%, or less than 1% of the cells within the population express the marker of interest. For example, in one embodiment, the expanded cell population is substantially free of T-cells (expressing the CD3 antigen), B-cells (expressing the CD 19 antigen), or mature granulocytes, NK lymphocytes, or macrophages (expressing the CD16 antigen).
  • The term “substantial proportion” is intended to mean that at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or up to 100% of the cells express the marker of interest. Conversely, the term “substantially diminished” is intended to mean that at least less than 10%, at least less than 5%, or at least less than 1% of cells express the marker of interest.
  • Those skilled in the art recognize that cells with increased cell surface marker expression can be detected and isolated by any means including flow cytometric sorting, antibody panning, and the like. Generally in flow cytometric analysis, one of skill in the art must first set a detection threshold for fluorescence. In setting the threshold, a negative control sample population will be recorded and a gate will be set around the population of interest according to the desired forward scatter (FSC) and side scatter (SSC). The detection threshold is then adjusted so that 97% or more of the cells do not fluoresce. Once the detection threshold is set, the fluorescence of cell population of interest is recorded. A cell is considered “positive for expression” when it expresses the marker of interest, whether a protein or a gene. Any method may be used to determine expression such as gene expression profiles, FACS, and the like. The term “+” indicates that the cell is positive for expression of the marker of interest. The term “” indicates that the cell does not have detectable levels of expression of the marker of interest.
  • The antibodies used to detect various lineages may be conjugated to different fluorochromes. These include phycobiliproteins, e.g., phycoerythrin and allophycocyanins; fluorescein; and Texas red. Dead cells can also be detected using dyes that selectively accumulate in dead cells (e.g., propidium iodide and 7-amino actinomycin D).
  • These expanded MSCs are characterized by a capacity for self-renewal. Further, the cells are characterized by an ability to commit to multilineage development. By “multilineage development” is intended that the cell is capable of differentiating into cell types of mesenchymal origin. These cells have a capability of limited self renewal and are constrained to committed development to various mesenchymal tissues such as bone, cartilage, fat, tendon, muscle, and marrow stroma (Pittenger et al. (1999) Science. 284:5411 143).
  • Stem cell function can be assayed using both in vitro and in vivo methods. In vitro testing comprises culturing MSCs in semi-solid medium similar to hematopoetic stem cells. Along with colony forming assays, MSCs may be tested for multilineage potential by culturing these cells in the presence of factors that send them down specific pathways such as bone, fat, and cartilage. Media for differentiating expanded MSCs are well known in the art (Pittenger and Martin (2004) Circ. Res. 95:9-20), and are commercially available through several sources such as Cambrex (Baltimore, Md.), Stem Cell Technologies (Vancouver, B. C., Canada), and R&D Systems (Minneapolis, Minn.). Expanded cells are cultured either on surfaces such as culture dishes with these media for 7-30 days for bone, fat, or muscle phenotypes, or in pellets in the bottom or tubes or flasks for differentiating down cartilage phenotypes. The test for function, tissue specific tests are performed such as alkaline phosphatase enzyme, osteocalcin or osteopontin for bone, oil O red staining for lipid accumulation in MSCs that differentiate towards an adipocyte lineage.
  • The expanded MSCs of the invention can be analyzed based on gene expression profiles. In this manner, the multilineage commitment potential can be determined. As used herein, an “expression profile” comprises one or more values corresponding to a measurement of the relative abundance of a gene expression product. Such values may include measurements of RNA levels or protein abundance. Thus, the expression profile can comprise values representing the measurement of the transcriptional state or the translational state of the gene (see U.S. Pat. Nos. 6,040,138, 5,800,992, 6,020135, 6,344,316, and 6,033,860).
  • The transcriptional state of a sample includes the identities and relative abundance of the RNA species, especially mRNAs present in the sample. Preferably, a substantial fraction of all constituent RNA species in the sample are measured, but at least a sufficient fraction to characterize the transcriptional state of the sample is measured. The transcriptional state can be conveniently determined by measuring transcript abundance by any of several existing gene expression technologies. Translational state includes the identities and relative abundance of the constituent protein species in the sample. As is known to those of skill in the art, the transcriptional state and translational state are related.
  • In one embodiment of the invention, microarrays can be used to measure the values to be included in the expression profiles. Microarrays are particularly well suited for this purpose because of the reproducibility between different experiments. DNA microarrays provide one method for the simultaneous measurement of the expression levels of large numbers of genes. Each array consists of a reproducible pattern of capture probes attached to a cell culture support. Labeled RNA or DNA is hybridized to complementary probes on the array and then detected by laser scanning. Hybridization intensities for each probe on the array are determined and converted to a quantitative value representing relative gene expression levels (see U.S. Pat. Nos. 6,040,138, 5,800,992 and 6,020,135, 6,033,860, and 6,344,316). High-density oligonucleotide arrays are particularly useful for determining the gene expression profile for a large number of RNAs in a sample.
  • “Array” is intended to mean a cell culture support or substrate with peptide or nucleic acid probes attached to the cell culture support or substrate. Arrays typically comprise a plurality of different nucleic acid or peptide capture probes that are coupled to a surface of a substrate in different, known locations. These arrays, also described as “microarrays” or colloquially “chips,” have been generally described in the art, for example, in U.S. Pat. Nos. 5,143,854, 5,445,934, 5,744,305, 5,677,195, 6,040,193, 5,424,186, 6,329,143, and 6,309,831 and Fodor et al. (1991) Science 251:767-77.
  • The methods of the invention comprise culturing or expanding MSCs from a stem cell source in a serum-free cell culture system that comprises a serum-free cell culture medium disclosed herein above, which comprises a cytokine cocktail and base medium. Methods of cell culture are well known in the art (see, e.g., Waymouth (1984) “Preparation and Use of Serum-free Culture Media,” in Cell Culture Methods for Molecular and Cell Biology, Vol. 1, Methods for Preparation of Media, Supplements, and Substrata for Serum-Free Animal Cell Culture, eds. Barnes et al. (Alan R. Liss), pp. 23-68). The term “cell-culture” is intended to mean the expansion or maintenance of cells in an artificial in vitro environment while maintaining a pluripotent phenotype. It is to be understood that the term “cell culture” is a generic term and may be used to encompass the cultivation not only of individual cells but also tissues, organ systems, or whole organisms.
  • In some embodiments, cells derived from a stem cell source are seeded onto a two-dimensional or three-dimensional cell culture surface described herein and contacted with the serum-free cell culture medium of the invention. The medium may initially contain the cytokine cocktail as well as the base medium, or the cytokine cocktail can be added later. Cells are then incubated with the serum-free cell culture medium comprising the cytokine cocktail and cell culture support at a temperature suitable for cell growth (for some embodiments about 37° C.), for at least about 24 hours, at least about 48 hours, at least about 72 hours, at least about 96 hours, at least about 120 hours, at least about 144 hours, at least about 168 hours, at least about 192 hours, or longer. Cells may be harvested in any manner known in the art including but not limited to centrifugation after decanting non-adherent cells, trypsinizing adherent cells, or scraping cells from the surface of the cell culture support.
  • The MSCs used in culture can include cells derived from any stem cell source, such as umbilical cord, umbilical cord blood, placenta, embryonic stem cells, adipose tissue, bone marrow, or other tissue-specific mesenchyme. These samples may be fresh, frozen, or refrigerated. Methods of freezing cells are well known in the art (see, e.g., Doyle et al. (1995) Cell & Tissue Culture: Laboratory Procedures (John Wiley & Sons, Chichester)).
  • Cryopreservation of stem cells prior to culture or cryopreservation of expanded cells disclosed herein may be carried out according to known methods. For example, cells may be suspended in a “freezing medium” such as, for example, culture medium further comprising 10% dimethylsulfoxide (DMSO), with or without 5-10% glycerol, at a density, for example, of about 1-2×106 cells/ml. The cells may be dispensed into glass or plastic vials, which are then sealed and transferred to a freezing chamber of a programmable or passive freezer. The optimal rate of freezing may be determined empirically. For example, a freezing program that gives a change in temperature of about −1° C./min through the heat of fusion may be used. Once vials containing the cells have reached −80° C., they may be transferred to a liquid nitrogen storage area. Cryopreserved cells may be stored for a period of years.
  • In some embodiments, freshly isolated cells from any stem cell source may be cryopreserved to constitute a bank of cells, portions of which may be withdrawn by thawing and then used to produce the expanded cells of the invention as needed. Thawing may be carried out rapidly, for example, by transferring a vial from liquid nitrogen to a 37° C. water bath. The thawed contents of the vial may be immediately transferred under sterile conditions to a culture vessel containing an appropriate medium such as nutritive medium. Once in culture, the cells may be examined daily, for example, with an inverted microscope to detect cell proliferation, and subcultured as soon as they reach an appropriate density.
  • Cells may be withdrawn from a cell bank as needed, and used for the production of new stem cells or tissue either in vitro, for example, as a three-dimensional scaffold culture, as described below, or in vivo, for example, by direct administration of cells to the site where tissue reconstitution or repair is needed. As described herein, the expanded MSCs of the invention may be used to reconstitute or repair tissue in a subject where the cells were originally isolated from that subject's own bone marrow or other tissue (i.e., autologous cells). Alternatively, the expanded MSCs disclosed herein may be used as ubiquitous donor cells to reconstitute or repair tissue in any subject (i.e., heterologous cells).
  • Methods of MSC isolation from bone marrow are well established. Prior to culture, a large proportion of non-mesenchymal linage cells may be removed from a stem cell source by negative or positive selection. For example, large numbers of lineage-committed cells can be removed by selective magnetic bead separations. In some embodiments, at least about 80%, usually at least about 70% of the differentiated cells will be removed prior to culture. Mononuclear cells from these tissues may be collected by density gradient centrifugation and cultured in tissue culture containers. Furthermore, bone marrow aspirates may be seeded onto tissue culture plates. Non-adherent cells may be removed after 1 to 3 days. Adherent, spindle shaped fibroblast-like MSCs are kept and expanded. After several hours to days, non-adherent cells are washed away and the MSCs remain. It has also been reported that MSCs may be obtained from cord blood, placenta and adipose tissues.
  • Cultured MSCs can be further isolated using any method known in the art. Generally, the MSCs are contacted with monoclonal antibodies directed to cell surface antigens and either positively or negatively selected. Such techniques for selection are well known in the art and include sorting by immunomagnetic beads, by complement mediated lysis, by “panning” with antibody attached to a solid matrix, agglutination methods, magnetic activated cell sorting (MACS), or fluorescence activated cell sorting (FACS).
  • The expanded MSCs of the invention have broad application in treating and ameliorating disease and injury. The expanded MSCs of the invention are useful in many therapeutic applications including repairing, reconstituting, and regenerating tissue as well as gene delivery. The MSCs of the invention can comprise both lineage-committed and uncommitted cells; thus, both cell types can be used together to accomplish multiple therapeutic goals, even simultaneously in some embodiments. For example, in some embodiments, the expanded MSCs of the invention can be used directly as stem cell transplants or be used in stem cell grafts either in suspension or on a cell culture support scaffold as noted herein above.
  • The expanded MSCs of the invention can be placed in a carrier medium before administration. For infusion, expanded MSCs of the invention can be administered in any physiologically acceptable medium, intravascularly, including intravenously, although they may also be introduced into other convenient sites such as into the bone marrow, where the cells may find an appropriate site for regeneration and differentiation. Usually, at least about 1×105 cells/kg, at least about 5×105 cells/kg, at least about 1×106 cells/kg, at least about 2×106 cells/kg, at least about 3×106 cells/kg, at least about 4×106 cells/kg, at least about 5×106 cells/kg, at least about 6×106 cells/kg, at least about 7×106 cells/kg, at least about 8×106 cells/kg, at least about 9×106 cells/kg, at least about 10×106 cells/kg, or more will be administered. See, for example, Ballen et al. (2001) Transplantation 7:635-645. The MSCs may be introduced by any method including injection, catheterization, or the like. If desired, additional drugs or growth factors can be co-administered. Drugs of interest include 5-fluorouracil and growth factors including cytokines such as IL-2, IL-3, G-CSF, M-CSF, GM-CSF, IFNγ, and erythropoietin. In addition, the MSCs can be injected with collagen, Matrigel, alone or with other hydrogels.
  • Administered MSCs may also comprise a mixture of cells herein described and additional cells of interest. Additional cells of interest include, without limitation, differentiated liver cells, differentiated cardiac muscle, differentiated pancreatic cells, and the like. These combinations are particularly useful when the expanded MSCs of the invention are seeded on a three-dimensional scaffold, hydrogel, or without carrier, as disclosed herein above.
  • In one embodiment, the expanded MSC population of the invention can be used to repair or reconstitute damaged or diseased mesenchymal tissues, such as the heart, the pancreas, the liver, fat tissue, bone, cartilage, endothelium, nerves, astrocytes, dermis, and the like. Once the expanded MSCs migrate to or are placed at the site of injury, they can differentiate to form new tissues and supplement organ function. In some embodiments, the cells are used to promote vascularization and, therefore, improve oxygenation and waste removal from tissues. In these embodiments, the expanded MSCs of the invention can be used to increase function of differentiated tissues and organs such as the ischemic heart as in cardiac failure or ischemic nerves as in stroke. Therefore, the expanded MSCs of the invention are useful in any disease where cellular function or organ function has been decremented.
  • “Supplementing a damaged organ” or “supplementing organ function” is intended to mean increasing, enhancing, or improving the function of an organ that is operating at less than optimum capacity. The term is used to refer to a gain in function so that the organ is operating at a physiologically acceptable capacity for that subject. For example, the physiological acceptable capacity for an organ from a child, e.g., a kidney or heart, would be different from the physiological acceptable capacity of an adult, or an elderly patient. The entire organ or part of the organ can be supplemented. Preferably the supplementation results in an organ with the same physiological response as a non-damaged or non-diseased organ. In one embodiment, an organ is supplemented in capacity when it is functioning to at least at about 10% of its natural capacity.
  • The expanded MSCs of the present invention can be used for implantation by contacting the cells with a tissue-engineered construct prior to grafting as noted herein above. The construct containing these cells is then implanted into a host in need of such a graft. The cells of the invention are particularly useful for promoting bone and cartilage generation, thereby facilitating tissue regeneration and repair. These cells may also be used in applications relating to graft versus host disease. The expanded MSCs of the invention may also be useful as vasculature-promoting stem cells. “Vascularization promoting” or “vasculature promoting” is intended to mean promoting the growth of new vessels (vasculogenesis) or inducing outgrowth from existing vessels (angiogenesis), or any combination thereof.
  • “Differentiated cells” is intended to mean cells that are committed to restricted tissue development. In some embodiments, the expanded cells of the invention may comprise both lineage-committed and uncommitted MSCs. Thus, in some tissue-engineered constructs, the MSCs may give rise to both the differentiated tissue and act as the source for the vasculature-promoting stem cells. In addition, in some tissue-engineered constructs, the MSCs may give rise to both the differentiated tissue and act as the source for the cardiovascular-promoting stem cells. In addition, in some tissue-engineered constructs, the MSCs may give rise to both the differentiated tissue and act as the source for the bone and cartilage-promoting stem cells. In other embodiments, the source of differentiated tissues may comprise cells or tissue from the intended graft recipient or another donor. The cell or tissue source may be differentiated prior to implantation. For example, pancreatic beta cells can be differentiated using conditions described for embryoid body formation as detailed in Itskovich-Eldor et al. (2000) Mol. Med. 6:88.
  • In one specific embodiment, the vasculature-promoting stem cells may be contacted with angiogenic growth factors such as VEGF and bFGF. These stem cells may be contacted with angiogenic growth factors prior to or after seeding onto the scaffold before engrafting the tissue-engineered construct into a damaged organ. In some embodiments, cytokine-impregnated polymers can release the angiogenic growth factor, such as VEGF and bFGF, over time. In other embodiments, the scaffold may be programmed to drive expansion and proliferation of seeded cells (see, e.g., U.S. Patent Application Publication No. 20040063206). In other embodiments, microspheres or microcarriers may be contacted with the vasculature-promoting stem cells and placed at a target site. Microsphere-based scaffolds are well known in the art (see, e.g., Mahoney and Saltzman (2001) Nature Biotech. 19:934).
  • In one specific embodiment, the cardiovascular- or bone and cartilage-promoting stem cells may be contacted with growth factors such as BDNF, bFGF, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF 1-α, TGF-β, thrombospondin, and WNT signaling agonists, including but not limited to BIO and WNT proteins, WNT-1, WNT-2, WNT-2b, WNT-3, WNT-3a, WNT-4, WNT-5a, WNT-5b, WNT-6, WNT-7a, WNT-7b, WNT-8a, WNT-8b, WNT-9a, WNT-9b, WNT-10a, WNT-10b, WNT-11, and WNT-16. In one embodiment, the WNT protein is WNT-3a.
  • The expanded MSCs of the invention can also be used for gene therapy in patients in need thereof. In some embodiments, more mature lineage-committed cells will be useful, especially where transient gene expression is needed or where gene transduction is facilitated by the maturation and division of the cells. For example, some retroviral vectors require that the cell be cycling for the gene to be integrated. Methods for transducing stem and progenitor cells to deliver new and therapeutic genes are known in the art.
  • Another aspect of the invention is a kit useful for promoting the attachment, survival, and/or proliferation of MSCs, comprising a serum-free cell culture system of the invention is provided. Such a kit comprises a serum-free cell culture medium and a cell culture surface described herein, and can comprise one or more other components or reagents suitable for culturing the cell. In one embodiment, the serum-free cell culture medium comprises any of the cytokine cocktails described herein, for example, the cocktail shown in Table 1 below, and a base medium, for example, the base medium shown in Table 2 below, and a cell culture surface comprising a cell culture support bound to a CAR, which in turn is bound to an ECM protein. In some embodiments, the ECM is a mixture of collagen I and fibronectin. In other embodiments, the kits comprise these components and also comprise one or more reagents for measuring cell proliferation in the culture. Such kits have many uses, which will be evident to one of skill in the art. For example, they can be used to propagate MSCs to be used in methods of cell therapy. Such kits could be of commercial use, e.g., in high-throughput drug studies.
  • The following examples are offered by way of illustration and not by way of limitation.
  • EXPERIMENTAL
  • For these examples, the concentrations of various cytokines in the culture media are described in Table 1, while the base medium composition is described in Table 2.
    TABLE 1
    Cytokine Cocktail for Serum-Free Cell Culture Medium
    Concentration
    Growth Factor Abbrev. Factor Full Name Vendor (ng/ml)
    BDNF Brain-derived Neurotrophic Factor R&D Systems, MN 1
    bFGF Basic Fibroblast Growth Factor BD Biosciences, MA 2.5
    BIO (2′Z,3′E)-6-bromoindirubin-3′-oxime Calbiochem 50 nM
    BMP-2 Bone Morphogenic Protein 2 R&D Systems, MN 0.5
    BMP-4 Bone Morphogenic Protein 4 R&D Systems, MN 0.5
    DKK-1 Dickkopf 1 R&D Systems, MN 10
    EGF Epidermal Growth Factor BD Biosciences, MA 10
    Erythropoietin Erythropoietin R&D Systems, MN .05 units/ml
    Fibronectin Fibronectin BD Biosciences, MA 10
    Flt-3/Flk-2 ligand Flt-3/Flk-2 ligand R&D Systems, MN 5
    G-CSF Granulocytes colony stimulating factor R&D Systems, MN 1
    IGF-1 Insulin-like Growth Factor 1 R&D Systems, MN 2.5
    IL-11 Interleukin-11 R&D Systems, MN 0.1
    IL-3 Interleukin-3 R&D Systems, MN 1
    IL-5 Interleukin-5 R&D Systems, MN 0.1
    IL-6 Interleukin-6 R&D Systems, MN 0.2
    LIF Leukemia Inhibitory Factor Chemicon 10
    PD-ECGF Platelet-derived Endothelial Cell Growth Factor R&D Systems, MN 2
    SCF Stem Cell Factor BD Biosciences, MA 2
    SDF-1 α Stromal Cell Derived Factor 1 α R&D Systems, MN 3
    TGF-β Transforming Growth Factor-β BD Biosciences 0.1-1
    Thrombospondin Thrombospondin Sigma-Aldrich, MO 10
    WNT-3a Wnt protein 3a R&D Systems, MN 10
  • TABLE 2
    Base Medium for Serum-Free Cell Culture Medium
    Base Medium Component Amount
    DMEM/MCDB-201 Media 60:40
    Ascorbic acid 2-phosphate 1 × 10−4 M
    Dexamethasone
    1 × 10−9 M
    Linoleic Acid-Bovine Serum 0.1%
    Albumin
    Insulin
    5 μg/mL
    Transferrin
    5 μg/mL
    Sodium selenite
    5 ng/mL
  • Example 1 MSC Expansion in Various Serum-Free Media
  • MSCs along with complete growth medium were purchased from Cambrex Biosciences (Baltimore, Md.). Frozen cells were thawed and cultured following manufacturer's instruction. After reaching ˜90% confluency, MSCs were washed once with PBS, removed from the culture surface using Trypsin/EDTA and replated at a density of 1600 cells/well (for 96 well plates) or at a density of 50,000 cells/well (for 6-well plates) which corresponds to the manufacturers recommended seeding density of 5000 cells/cm2.
  • At day 6 or 7, cells were washed once with PBS, fixed using 4% paraformaldehyde for 15 minutes and then stained using DAPI according to manufacturer's suggestions (Molecular Probes, Eugene, Oreg.). For 96-well plate experiments, one image per well was taken at 4× magnification using Molecular Devices' Discovery-1 high content screening system. Cell nuclei enumeration was determined using Metamorph Image Analysis Software (Molecular Devices, Sunnyvale, Calif.). The number of DAPI stained cell nuclei corresponds directly with the total number of cells per well. For 6-well plates, cell enumeration was determined using Trypan Blue exclusion and manual counting using a hemocytometer. Multiple donors (different preps of MSCs) were tested in serum-free conditions while the cells were also cultured on tissue culture polystyrene (TCPS) plates using the manufacturer's standard serum-containing medium as the control.
  • Several serum-free expansion conditions were scaled up into the 6-well plate format (˜10 cm2) and cultured for 7 days. Cells were then washed with PBS and then removed from the 6-well plates using Trypsin/EDTA. Cells were then reseeded into 96-well plates at two different densities according to manufacturer's specifications. MSCs induced to form adipocytes were seeded at 7000 cells/well and cultured in Stem Cell Technologies (Vancouver, B. C., Canada) adipogenic medium supplement. MSCs induced to form osteoblasts were seeded at 1000 cells/well using Cambrex Biosciences (Baltimore, Md.) osteogenic media supplements. Two to three weeks after induction, cells were washed once with PBS and fixed using 4% paraformaldehyde. Fixed cells were then stained with Oil Red O (Sigma-Aldrich (St. Louis, Mo.)) for adipocytes and BCIP/NBT (Sigma-Aldrich (St. Louis, Mo.)) for alkaline phosphatase/osteoblasts. Bright field images at multiple sites per well were taken of stained cells at 10× magnification. Percentage of differentiation was determined using Metamorph Image Analysis Software (Molecular Devices, Sunnyvale, Calif.) to calculate total stained area by threshold image analysis.
  • Initial experiments compared serum-free medium expansion of MSCs in G2 medium on Collagen I (Col 1, Sigma-Aldrich)/Fibronectin (FN, BD Biosciences) cell adhesion resistant (CAR) surface modified 96-well plates. MSCs were expanded for 7 days in G2 serum-free medium on the CAR surface and cell nuclei counts were compared to MSCs expanded in the manufacturer's recommended serum-containing medium on tissue culture polystyrene (TCPS) 96-well plates. MSCs expanded in the G2-CAR surface serum free environment were comparable to the manufacturer's recommended serum-containing expansion conditions up to 7 days (see FIG. 1).
  • Example 2 Differentiation Capacity and Surface Marker Characterization of G2 Expanded MSCs
  • MSCs expanded in serum-free media of the present invention on CAR Col 1/FN surface for 7 days remained multipotent. Briefly, MSCs expanded in culture environments of the present invention were removed from the serum-free media and then cultured in either adipogenic (fat) or osteogenic (bone) induction media (per manufacturer's suggestions). MSCs expanded in the serum-free environments of the present invention were able to differentiate towards both adipogenic (see FIG. 2) and osteogenic (see FIG. 3) lineages. These results demonstrate that the serum-free environments of the present invention help to maintain stem cell pluripotency in expanded MSCs at least as well as the industry standard serum-containing media (Cambrex). This demonstrates that these cells are not committed towards specific lineages, and that therefore these MSCs may still be used for a variety or research or clinical applications.
  • MSCs cultured in serum-free environments for 7 days retain cell surface markers showing equivalency to Cambrex cell populations expanded in serum-containing media. In addition, MSCs cultured in serum-free environments lacked CD45 marker expression, indicating that these cells do not differentiate towards hematopoietic lineages.
  • Example 3 Comparison of Media and Cell Culture Surface Conditions
  • MSCs cultured in three different media (G2, base medium without G2 growth factors/cytokines, and in serum-containing complete medium) all expand better on a Col 1/FN CAR surface as compared to TCPS surfaces (see FIG. 4). Also, MSCs cultured in G2 serum-free medium expand to equivalent levels as MSCs cultured in serum-containing medium.
  • Example 4 Media Optimization
  • Media compositions that support the expansion of MSCs were identified. The G2 serum-free medium served as a control for this optimization experiment. Screening plates were prepared using 96-well plates containing 30 factors in 60 different compositions. One thousand six hundred (1600) MSCs were incubated in each well for 7 days. Best media compositions were selected based on the average number of live cells obtained from the repeats from each screen.
  • Best well “hits” of the cytokine screen were the C6, D3, C2, and G5 wells, respectively.
  • The C6 well contained cytokines BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1α, and WNT-3a.
  • The D3 well contained cytokines bFGF, BMP-2, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a
  • The C2 well contained cytokines bFGF, BIO, BMP-2, BMP-4, EGF, EPO, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and WNT-3a.
  • The G5 well contained cytokines bFGF, BMP-4, DKK-1, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-6, PD-ECGF, SDF-1α, and WNT-3a.
  • Cytokine compositions of wells C6, D3, C2, and G5 are referred to hereinafter as “C6,” “D3,” “C2,” and “G5,” respectively.
  • Media compositions C6, D3, C2, and G5 all outperformed G2 serum free medium controls and 10% serum-containing complete medium (see FIG. 5). These serum free media compositions contain 22, 11, 11, and 10 growth factor/cytokine additives, respectively. In addition, serum free media G2 and D3 both performed as well or better than serum-containing complete medium controls and significantly outperform a previously published serum free MSC expansion medium (Lennon et al. (1995) Exp. Cell Res. 219:211-222; U.S. Pat. No. 5,908,782).
  • Three of these serum-free media compositions (D3, C2, and G5) were used to confirm MSCs retained multipotentiality. MSCs were cultured in one of the three serum-free media for 7 days. Expanded MSCs were removed from the serum-free media and then cultured in either adipogenic (fat) or osteogenic (bone) induction media (per manufacturer's suggestions). MSCs remained multipotential and differentiated towards both adipogenic and osteogenic lineages in all three serum-free media, each comparable to the manufacturer's recommended serum-containing complete medium controls (see FIGS. 6A and 6B).
  • Further media optimization was performed and the best well “hits” from this cytokine screen were the G7, G4 and C8 wells, respectively.
  • The G7 well contained cytokines bFGF, BMP-2, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, LIF, and WNT-3a.
  • The G4 well contained cytokines bFGF, BMP-2, EGF, IL-11, PD-ECGF, and WNT-3a.
  • The C8 well contained cytokines bFGF, BMP-2, EGF, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, IL-5, LIF, PD-ECGF, and WNT-3a.
  • Cytokine compositions of wells G7, G4, and C8 are referred to hereinafter as “G7,” “G4,” and “C8,” respectively. Media compositions G7, G4, and C8 all expanded MSCs as well as or better than serum-containing controls (see FIG. 7).
  • Additional optimization data was collected employing a variety of media compositions. In one experiment, MSCs were cultured in: 1) the G4 serum-free medium composition; 2) G4+TGF-β; 3) bFGF in base medium only; 4) EGF in base medium only; 5) TGF-β in base medium only; 6) base medium alone; and 7) serum-containing medium. Although MSCs cultured in G4 expanded as well or better than serum-containing medium, MSCs cultured in G4+TGF-β far outperformed any other media composition, nearly triple the serum-containing medium controls (see FIG. 8). Furthermore, the single factors, while promoting MSC expansion, did not perform as well as serum in expanding MSCs. However, synergistic combinations of these factors (e.g., G4) expand MSCs more than any single factor, and the addition of TGF-β substantially enhanced expansion. These factor synergies, in combination with the collagen 1+fibronectin surface, comprise serum-free environments that substantially outperform anything that has been established to date. G4+TGF-β serum-free medium was scaled up and used to expand MSCs in 6-well Col 1/FN CAR plates. MSCs were seeded on 6-well plates at 50,000 per well (n=3). Cells in serum containing complete media were seeded onto a 6-well TCPS plate. At confluency, all cells were removed and cell number was determined using Trypan Blue exclusion and hemocytometer counting. Cells were replated at 50,000 per well under the same condition for a second round of expansion. Cell numbers were again determined as above. After the first round of expansion (approximately 6 days), MSCs expanded in serum-free conditions outperformed the manufacturer's recommended serum-containing medium controls. After the second round of expansion (approximately 6 days), the MSCs continued to expand in the serum-free medium at an increased rate as compared to the serum-containing medium controls. G4+TGFb provided 5.4 and 6.5 fold expansion in the first and second rounds, respectively, and regular medium gave 3.5 and 3.1 fold expansion in the first and second rounds, respectively.
  • In another experiment, a serum-free medium comprising base medium and WNT-3a, bFGF, and TGF-β was compared to media compositions containing the same components but adding one, two, three, four, five, six, or seven additional cytokines that include EGF, fibronectin, thrombospondin, BMP-2, BMP-4, IL-11, and PD-ECGF in their ability to expand MSCs in culture. Adding one, two, three, four, five, six, or seven growth factors to the serum free medium comprising base medium and WNT-3a, bFGF, and TGF-β had a positive effect on MSC expansion. In particular, the addition of EGF to the three growth factors already present in the serum-free medium resulted in approximately a 20% increase in MSC expansion over compositions that did not contain EGF, and both BMPs on top of EGF further enhanced expansion. Furthermore, the addition of all seven cytokines to WNT-3a, bFGF, and TGF-β gave the best expansion, demonstrating that building complexity into the serum free environments may further enhance MSC expansion. However, the synergies between WNT-3a, bFGF, EGF, and TGF-β were found to have the greatest effects on MSC serum-free expansion on collagen 1+fibronectin surfaces.
  • In another experiment, a serum-free medium comprising base medium was compared to media comprising the base medium plus various combinations of WNT-3a, BIO, bFGF, EGF, and TGF-β in their ability to expand MSCs in culture (see FIG. 9). Specifically, the combinations tested were: base medium alone (no growth factors), Cambrex Biosciences (Baltimore, Md.) serum-containing medium on TCPS, base medium+bFGF+TGF-β, base medium+WNT-3a+bFGF, base medium+bFGF+EGF, base medium+WNT-3a+TGF-β, base medium+WNT-3a+EGF, base medium+EGF+TGF-β, base medium+WNT-3a, base medium+bFGF+EGF+TGF-β, base medium+WNT-3a+bFGF+EGF+TGF-β, and base medium+bFGF+EGF+TGF-β+BIO. Although all conditions outperformed base medium alone, the growth factor combinations that produced the greatest MSC expansion were base medium+EGF+TGF-β, base medium+bFGF+TGF-β, base medium+bFGF+EGF+TGF-β, base medium+WNT-3a+bFGF+EGF+TGF-β, and base medium+bFGF+EGF+TGF-β+BIO.
  • All publications and patent applications mentioned in the specification are indicative of the level of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually to be incorporated by reference.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims.

Claims (26)

1. A serum-free cell culture system comprising a serum-free cell culture medium and a cell culture surface that promotes the adhesion and expansion of mesenchymal stem cells, wherein at least one insoluble substrate protein is presented from said cell culture surface, and wherein said serum-free cell culture medium is selected from the group consisting of:
a) a medium comprising bFGF in combination with at least one growth factor selected from the group consisting of a WNT signaling agonist, TGF-β, and EGF;
b) a medium comprising a WNT signaling agonist in combination with at least one growth factor selected from the group consisting of bFGF, TGF-β, and EGF;
c) a medium comprising TGF-β in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and EGF;
d) a medium comprising EGF in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and TGF-β;
e) a medium comprising fibronectin, SDF-1 α, IL-6, SCF, IL-5, BDNF, PD-ECGF, IL-11, IL-3, EPO, Flt-3/Flk-2 ligand, BMP-4, thrombospondin, IGF-1, and bFGF;
f) a medium comprising BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1α, and a WNT signaling agonist;
g) a medium comprising bFGF, BMP-2, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and a WNT signaling agonist;
h) a medium comprising bFGF, BIO, BMP-2, BMP-4, EGF, EPO, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and a WNT signaling agonist;
i) a medium comprising bFGF, BMP-4, DKK-1, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-6, PD-ECGF, SDF-1α, and a WNT signaling agonist;
j) a medium comprising bFGF, BMP-2, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, LIF, and a WNT signaling agonist;
k) a medium comprising bFGF, BMP-2, EGF, IL-11, PD-ECGF, and a WNT signaling agonist; and
l) a medium comprising bFGF, BMP-2, EGF, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, IL-5, LIF, PD-ECGF, and a WNT signaling agonist.
2. The serum-free cell culture system of claim 1, wherein said WNT signaling agonist is BIO or WNT-3a.
3. The serum-free cell culture system of claim 1, wherein the insoluble substrate protein is an extracellular matrix protein.
4. The serum-free cell culture system of claim 3, wherein the extracellular matrix protein is fibronectin, laminin, HA, vitronectin, collagen I, collagen II, collagen III, collagen IV, collagen V, collagen VI, or any combination thereof.
5. The serum-free cell culture system of claim 4, wherein the extracellular matrix protein is a combination of collagen I and fibronectin.
6. The serum-free cell culture system of claim 5, wherein the WNT signaling agonist is BIO or WNT-3a, and wherein the cell culture surface comprises a cell culture support bound to a cell adhesion resistant material.
7. A method of expanding a population of mesenchymal stem cells using the cell culture system of claim 1.
8. The method of claim 7, wherein said WNT signaling agonist is BIO or WNT-3a, wherein the cell culture surface comprises a cell culture support bound to a cell adhesion resistant material, and wherein the insoluble substrate protein is a combination of collagen I and fibronectin.
9. An expanded population of mesenchymal stem cells that has never been exposed to serum, wherein said population is produced by the method of claim 7.
10. A method of transplanting an expanded population of mesenchymal stem cells, comprising administering the population of claim 9 to a patient in need thereof, wherein the population is administered in an amount effective to reconstitute cardiovascular, bone, or cartilage tissue.
11. A kit comprising the serum-free cell culture system of claim 1.
12. The kit of claim 11, wherein said WNT signaling agonist is BIO or WNT-3a, wherein the cell culture surface comprises a cell culture support bound to a cell adhesion resistant material, and wherein the insoluble substrate protein is a combination of collagen I and fibronectin.
13. A serum-free cell culture system comprising a serum-free cell culture medium and a cell culture surface, wherein said cell culture surface promotes the adhesion and expansion of mesenchymal stem cells and comprises a cell culture support bound to a cell adhesion resistant material, and wherein at least one insoluble substrate protein is presented from the cell culture surface.
14. The serum-free cell culture system of claim 13, wherein the insoluble substrate protein is an extracellular matrix protein.
15. The serum-free cell culture system of claim 14, wherein the extracellular matrix protein is fibronectin, laminin, HA, vitronectin, collagen I, collagen II, collagen III, collagen IV, collagen V, collagen VI, or any combination thereof.
16. The serum-free cell culture system of claim 15, wherein the extracellular matrix protein is a combination of collagen I and fibronectin.
17. The serum-free cell culture system of claim 13, wherein said serum-free cell culture medium is selected from the group consisting of:
a) a medium comprising bFGF in combination with at least one growth factor selected from the group consisting of a WNT signaling agonist, TGF-β, and EGF;
b) a medium comprising a WNT signaling agonist in combination with at least one growth factor selected from the group consisting of bFGF, TGF-β, and EGF;
c) a medium comprising TGF-β in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and EGF;
d) a medium comprising EGF in combination with at least one growth factor selected from the group consisting of bFGF, a WNT signaling agonist, and TGF-β;
e) a medium comprising fibronectin, SDF-1 α, IL-6, SCF, IL-5, BDNF, PD-ECGF, IL-11, IL-3, EPO, Flt-3/Flk-2 ligand, BMP-4, thrombospondin, IGF-1, and bFGF;
f) a medium comprising BDNF, bFGF, BIO, BMP-2, BMP-4, DKK-1, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, G-CSF, IGF-1, IL-11, IL-3, IL-5, IL-6, LIF, PD-ECGF, SCF, SDF-1α, and a WNT signaling agonist;
g) a medium comprising bFGF, BMP-2, EGF, EPO, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and a WNT signaling agonist;
h) a medium comprising bFGF, BIO, BMP-2, BMP-4, EGF, EPO, Flt-3/Flk-2 ligand, IGF-1, IL-11, IL-5, and a WNT signaling agonist;
i) a medium comprising bFGF, BMP-4, DKK-1, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-6, PD-ECGF, SDF-1α, and a WNT signaling agonist;
j) a medium comprising bFGF, BMP-2, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, LIF, and a WNT signaling agonist;
k) a medium comprising bFGF, BMP-2, EGF, IL-11, PD-ECGF, and a WNT signaling agonist; and
l) a medium comprising bFGF, BMP-2, EGF, fibronectin, thrombospondin, Flt-3/Flk-2 ligand, IL-11, IL-5, LIF, PD-ECGF, and a WNT signaling agonist.
18. The serum-free cell culture system of claim 17, wherein said WNT signaling agonist is BIO or WNT-3a.
19. The serum-free cell culture system of claim 17, wherein the insoluble substrate protein is an extracellular matrix protein and wherein the extracellular matrix protein is fibronectin, laminin, HA, vitronectin, collagen I, collagen II, collagen III, collagen IV, collagen V, collagen VI, or any combination thereof.
20. The serum-free cell culture system of claim 19, wherein the extracellular matrix protein is a combination of collagen I and fibronectin.
21. A method of expanding a population of mesenchymal stem cells using the cell culture system of claim 13.
22. The method of claim 21, wherein the insoluble substrate protein is a combination of collagen I and fibronectin.
23. An expanded population of mesenchymal stem cells, said population produced by the method of claim 21, wherein said cells were unexposed to serum.
24. A method of transplanting an expanded population of mesenchymal stem cells, comprising administering the population of claim 23 to a patient in need thereof, wherein the population is administered in an amount effective to reconstitute cardiovascular, bone, or cartilage tissue.
25. A kit comprising the serum-free cell culture system of claim 13.
26. The kit of claim 25, wherein the insoluble substrate protein is a combination of collagen I and fibronectin.
US11/129,612 2004-05-14 2005-05-13 Cell culture environments for the serum-free expansion of mesenchymal stem cells Abandoned US20050265980A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/129,612 US20050265980A1 (en) 2004-05-14 2005-05-13 Cell culture environments for the serum-free expansion of mesenchymal stem cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57121204P 2004-05-14 2004-05-14
US11/129,612 US20050265980A1 (en) 2004-05-14 2005-05-13 Cell culture environments for the serum-free expansion of mesenchymal stem cells

Publications (1)

Publication Number Publication Date
US20050265980A1 true US20050265980A1 (en) 2005-12-01

Family

ID=34978975

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/128,950 Expired - Fee Related US7790458B2 (en) 2004-05-14 2005-05-13 Material and methods for the growth of hematopoietic stem cells
US11/129,612 Abandoned US20050265980A1 (en) 2004-05-14 2005-05-13 Cell culture environments for the serum-free expansion of mesenchymal stem cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/128,950 Expired - Fee Related US7790458B2 (en) 2004-05-14 2005-05-13 Material and methods for the growth of hematopoietic stem cells

Country Status (4)

Country Link
US (2) US7790458B2 (en)
EP (2) EP1756267A2 (en)
JP (2) JP2007536935A (en)
WO (2) WO2005113751A1 (en)

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050261189A1 (en) * 2004-04-16 2005-11-24 Hydra Biosciences, Inc. Methods of promoting cardiac cell proliferation
WO2007136760A2 (en) * 2006-05-19 2007-11-29 The Johns Hopkins University Method of growth of mesenchymal cells under non-adherent conditions for clinical applications
US20070292949A1 (en) * 2006-06-20 2007-12-20 Genzyme Corporation Serum-free media and their uses for chondrocyte expansion
US20080078000A1 (en) * 2004-10-19 2008-03-27 Regeneron Pharmaceuticals, Inc. Conditioned culture media
US20080269895A1 (en) * 2005-09-20 2008-10-30 Steinwachs Matthias R Implant for the Repair of a Cartilage Defect and Method for Manufacturing the Implant
WO2008130650A1 (en) * 2007-04-19 2008-10-30 University Of Massachusetts Medical School Thermal responsive polymer siloxanes, compositions, and methods and applications related thereto
US20090061517A1 (en) * 2007-05-31 2009-03-05 Kisaalita William S Cell culture apparatus and methods of making and using same
US20090191634A1 (en) * 2008-01-30 2009-07-30 Arthur W Martin (meth)acrylate surfaces for cell culture, methods of making and using the surfaces
US20090191632A1 (en) * 2008-01-30 2009-07-30 Fadeev Andrei G Swellable (meth)acrylate surfaces for culturing cells in chemically defined media
US20090191627A1 (en) * 2008-01-30 2009-07-30 Andrei Gennadyevich Fadeev Synthetic surfaces for culturing cells in chemically defined media
US20090191633A1 (en) * 2008-01-30 2009-07-30 Christopher Bankole Shogbon Synthetic Surfaces for Culturing Stem Cell Derived Cardiomyocytes
US20090191626A1 (en) * 2008-01-30 2009-07-30 Christopher Bankole Shogbon Synthetic Surfaces for Culturing Stem Cell Derived Oligodendrocyte Progenitor Cells
EP2098244A1 (en) * 2008-03-04 2009-09-09 Medizinische Hochschule Hannover Pharmaceutical composition for the treatment of myocardial infarction
WO2009120043A2 (en) * 2008-03-28 2009-10-01 경희대학교 산학협력단 Use for cd45+ cells or a cd45+ cell culture fluid for promoting the growth of mesenchymal stem cells
US20100015710A1 (en) * 2008-04-25 2010-01-21 Sunghoon Jung Methods and Compositions for Isolating, Maintaining and Serially Expanding Human Mesenchymal Stem Cells
US20100035344A1 (en) * 2008-06-11 2010-02-11 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
US20100040582A1 (en) * 2006-05-31 2010-02-18 Nadir Askenasy Methods of selecting stem cells and uses thereof
US20100278783A1 (en) * 2009-05-01 2010-11-04 Biocardia, Inc. Method of Preparing Autologous Cells and Method of Use for Therapy
US20110065809A1 (en) * 2009-09-15 2011-03-17 Medtronic, Inc. Polymerization of Multifunctional Azides, and Polymers Therefrom
US20110111498A1 (en) * 2008-03-17 2011-05-12 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
US20110129919A1 (en) * 2008-03-17 2011-06-02 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
US20110129447A1 (en) * 2006-03-23 2011-06-02 Pluristem Ltd. Methods for Cell Expansion and Uses of Cells and Conditioned Media Produced Thereby for Therapy
US20110171182A1 (en) * 2006-03-23 2011-07-14 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US20110207166A1 (en) * 2009-11-06 2011-08-25 Sarah Rivkah Vaiselbuh Human bone marrow microenvironments and uses thereof
US20110212523A1 (en) * 2008-11-11 2011-09-01 Yukio Kato Differentiation-inducing culture medium additive and use thereof
US20110293583A1 (en) * 2006-03-23 2011-12-01 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
CN102449141A (en) * 2009-03-31 2012-05-09 德克萨斯大学系统董事会 Isolation of human umbilical cord blood-derived mesenchymal stem cells
US20120219531A1 (en) * 2008-03-17 2012-08-30 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
WO2012135813A1 (en) * 2011-03-31 2012-10-04 University Of Rochester Methods and compositions for mesenchymal stem cell proliferation
US8480757B2 (en) 2005-08-26 2013-07-09 Zimmer, Inc. Implants and methods for repair, replacement and treatment of disease
US8497121B2 (en) 2006-12-20 2013-07-30 Zimmer Orthobiologics, Inc. Method of obtaining viable small tissue particles and use for tissue repair
US8518433B2 (en) 2003-12-11 2013-08-27 Zimmer, Inc. Method of treating an osteochondral defect
WO2013165252A1 (en) * 2012-05-03 2013-11-07 Erasmus University Medical Center Rotterdam Culturing of mesenchymal stem cells
US8637309B2 (en) 2008-03-17 2014-01-28 Agency For Science, Technology And Research Microcarriers for stem cell culture
NL2010225C2 (en) * 2013-02-01 2014-08-04 Conradus Ghosal Gho Composition and method for preserving, transporting and storing living biological materials.
NL2010222C2 (en) * 2013-02-01 2014-08-04 Conradus Ghosal Gho Composition and method for generating a desired cell type and/or tissue type from hair follicular stem cells.
US8829166B2 (en) 2002-06-26 2014-09-09 Zimmer Orthobiologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
US8932858B2 (en) 2008-03-07 2015-01-13 Corning Incorporated Modified polysaccharide for cell culture and release
NL2011170C2 (en) * 2013-07-15 2015-01-21 Univ Erasmus Medical Ct Method and culture medium for in vitro culturing of stem cells.
US9074176B2 (en) 2006-01-13 2015-07-07 Two Cells Co., Ltd. Culture medium additive for use in serum-free culturing of animal cell, kit and use thereof
US9138318B2 (en) 2007-04-12 2015-09-22 Zimmer, Inc. Apparatus for forming an implant
US20160032248A1 (en) * 2013-03-15 2016-02-04 Stemcell Technologies, Inc. Compositions and methods for obtaining enriched mesenchymal stem cell cultures
WO2016053758A1 (en) * 2014-10-01 2016-04-07 Commence Bio, Inc. Induction medium and methods for stem cell culture and therapy
US9394521B2 (en) 2010-03-10 2016-07-19 Two Cells Co., Ltd. Cell preparation containing mesenchymal stem cells, and method for producing same
US9458431B2 (en) 2008-03-17 2016-10-04 Agency For Science, Technology And Research Microcarriers for stem cell culture
US9541480B2 (en) 2011-06-29 2017-01-10 Academia Sinica Capture, purification, and release of biological substances using a surface coating
US9617511B2 (en) 2010-09-07 2017-04-11 The Board Of Regents Of The University Of Texas System Tissue-specific differentiation matrices and uses thereof
CN107028981A (en) * 2007-09-19 2017-08-11 普拉里斯坦有限公司 Adherent cell from fat or placenta tissue and its purposes in the treatment
US9758762B2 (en) 2008-09-02 2017-09-12 Pluristem Ltd. Perfusion bioreactor for culturing CD200—placenta adherent cells
US9873862B2 (en) 2007-01-22 2018-01-23 The Board Of Trustees Of The University Of Arkansas Maintenance and propagation of mesenchymal stem cells
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane
US10112198B2 (en) 2014-08-26 2018-10-30 Academia Sinica Collector architecture layout design
US10150950B2 (en) * 2013-09-05 2018-12-11 Medipost Co., Ltd Method for culturing mesenchymal stem cells according to cell size
WO2018226435A1 (en) * 2017-06-10 2018-12-13 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10167447B2 (en) 2012-12-21 2019-01-01 Zimmer, Inc. Supports and methods for promoting integration of cartilage tissue explants
US20190025286A1 (en) * 2017-07-20 2019-01-24 Seqvera Ltd. Oy In vitro method for measurement and model-free evaluation of time-invariant biomaterials functions
EP3441458A1 (en) * 2009-02-03 2019-02-13 Koninklijke Nederlandse Akademie van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
US10213463B2 (en) 2016-06-13 2019-02-26 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10246679B2 (en) 2015-03-02 2019-04-02 The University Of Tokyo Method for producing pluripotent stem cell having improved chimera forming ability from primed pluripotent stem cell, and composition and combination therefor
CN109609443A (en) * 2019-01-14 2019-04-12 上海银海圣生物科技有限公司 A kind of mouse liver primitive cell culture base, cultural method and incubator
CN109715785A (en) * 2016-07-25 2019-05-03 宇部兴产株式会社 Inhibit the method for the method of the differentiation of stem cell, the method and induction stem cell of modulating stem cell
US20190224244A1 (en) * 2016-07-11 2019-07-25 Bonus Therapeutics Ltd. Cell compositions for tissue regeneration
US10443037B2 (en) * 2015-01-26 2019-10-15 Ube Industries, Ltd. Long-term cell-cultivation using polyimide porous membrane and cell-cryopreservation method using polyimide porous membrane
US10456423B2 (en) 2016-06-13 2019-10-29 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10495644B2 (en) 2014-04-01 2019-12-03 Academia Sinica Methods and systems for cancer diagnosis and prognosis
CN110662833A (en) * 2017-04-25 2020-01-07 北海道公立大学法人札幌医科大学 Method for producing mesenchymal stem cell, marker for treatment effect of mesenchymal stem cell, method for determining treatment effect, and cell preparation containing mesenchymal stem cell
US10722541B2 (en) 2011-03-22 2020-07-28 Pluristem Ltd. Methods for treating radiation or chemical injury
US11034935B2 (en) 2010-07-29 2021-06-15 Koninklijke Nederlandse Akademie Van Wetenschappen Liver organoid, uses thereof and culture method for obtaining them
US11278573B2 (en) * 2008-12-05 2022-03-22 Regenexx, LLC Methods and compositions to facilitate repair of avascular tissue

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7723106B2 (en) * 2004-07-29 2010-05-25 Food Industry Research & Development Institute Stroma-free, serum-free, and chemically defined medium and method for ex vivo mononuclear cell expansion using the same
DK1759536T3 (en) 2004-06-01 2011-09-05 Kwalata Trading Ltd In vitro techniques for use with stem cells
EP1776453B1 (en) * 2004-08-13 2009-02-25 Medtronic, Inc. Isolation of endothelial progenitor cell subsets and methods for their use
US7273756B2 (en) * 2004-10-01 2007-09-25 Isto Technologies, Inc. Method for chondrocyte expansion with phenotype retention
GB0505970D0 (en) * 2005-03-23 2005-04-27 Univ Edinburgh Culture medium containing kinase inhibitor, and uses thereof
EP1891437B1 (en) * 2005-06-06 2014-04-02 Glykos Finland Oy Method for purification of a cell population
US20070207544A1 (en) * 2005-07-29 2007-09-06 Food Industry Research & Development Institute Stroma-free, serum-free, and chemically defined medium and method for ex vivo mononuclear cell expansion using the same
WO2007095069A2 (en) * 2006-02-10 2007-08-23 The Trustees Of The University Of Pennsylvania Nf-ya activates multiple hematopoietic stem cell (hsc) regulatory genes and promotes hsc self-renewal
TW200734462A (en) * 2006-03-08 2007-09-16 In Motion Invest Ltd Regulating stem cells
JP5523825B2 (en) * 2006-03-20 2014-06-18 ジ・オハイオ・ステイト・ユニバーシティ・リサーチ・ファウンデイション MicroRNA fingerprints during human megakaryocyte formation
US8734827B2 (en) * 2006-04-28 2014-05-27 University Of Hong Kong Bioengineered intervertebral discs and methods for their preparation
EP2039759A1 (en) * 2007-09-24 2009-03-25 Franca Fagioli Ex-vivo expansion method under clinical grade conditions of haematopoietic stem cells isolated from bone marow to treat ischaemic diseases such as acute myocardial infartcion
RU2359030C1 (en) * 2008-03-19 2009-06-20 Общество С Ограниченной Ответственностью "Лаборатория Клеточных Технологий" Method for obtaining endotheliocytes from human embryonic stem cells (versions)
US8288513B2 (en) * 2008-07-25 2012-10-16 Becton, Dickinson And Company Defined cell culturing surfaces and methods of use
WO2010056336A1 (en) * 2008-11-14 2010-05-20 Howmedica Osteonics Corp. Cells for joint fluid
CN101580816B (en) * 2009-04-23 2012-02-29 中国科学院广州生物医药与健康研究院 Novel serum-free culture medium for inducing fast and efficient production of pluripotent stem cells and use method thereof
EP2488187A4 (en) * 2009-10-13 2014-03-26 Allocure Inc Assay for the prediction of therapeutic effectiveness of mesenchymal stromal cells, and methods of using same
KR20140001900A (en) * 2010-10-18 2014-01-07 선샤인 바이오테크 인크 Human multipotent embryonic stem cell-like progenitor cells
EP2638159B1 (en) 2010-11-11 2019-04-24 University of Miami Compositions, kits and methods for treatment of cardiovascular, immunological, and inflammatory diseases
JP5993113B2 (en) * 2011-01-31 2016-09-14 株式会社 バイオミメティクスシンパシーズ Human adipose tissue-derived mesenchymal stem cells for the treatment of Alzheimer's disease
US8709401B2 (en) 2011-02-25 2014-04-29 Howmedica Osteonics Corp. Primed stem cells and uses thereof to treat inflammatory conditions in joints
CN102191215B (en) * 2011-03-25 2013-05-08 戴育成 Human-derived serum-free culture medium and preparation method thereof
KR101639988B1 (en) * 2011-05-11 2016-07-15 (주)안트로젠 Method for preparation of mesenchymal stem cell culture comprising high concentration of cell growth factors and composition prepared therefrom
WO2013086029A1 (en) 2011-12-05 2013-06-13 Primorigen Biosciences Inc. Compositions and methods for differentiating pluripotent stem cells into primitive blood cells and uses thereof
GB201210857D0 (en) 2012-06-19 2012-08-01 Cambridge Entpr Ltd Transcription factor mediated programming towards megakaryocytes
US20140031772A1 (en) * 2012-07-30 2014-01-30 Next Healthcare, Inc. System and method for collecting stem cells
CN105101979B (en) 2012-12-21 2021-10-08 安斯泰来再生医药协会 Method for preparing platelets from pluripotent stem cells and composition thereof
WO2014113593A1 (en) * 2013-01-18 2014-07-24 Advanced Diagnostic Technologies, Llc Methods and compositions for preparing blood substitutes
JP6366944B2 (en) * 2014-01-24 2018-08-01 国立大学法人 東京大学 Method for producing pluripotent stem cells with improved chimera-forming ability from prime-type pluripotent stem cells, and composition and combination therefor
EP3074505B1 (en) 2014-02-14 2018-06-06 National University of Ireland Galway Serum-free medium
FR3018819A1 (en) * 2014-03-19 2015-09-25 Univ Bourgogne TREATMENT OF THE INFLAMMATORY AND DYSIMMUNITARY RESPONSE
JP2015188420A (en) * 2014-03-28 2015-11-02 セイコーエプソン株式会社 Support treatment method and treatment device
CN105368775B (en) * 2014-08-13 2019-05-24 苏州协和方舟生物医药研发有限公司 The method and system of megacaryocyte and blood platelet are prepared using human hematopoietic stem cell
CN105331575B (en) * 2014-08-13 2019-01-18 苏州方舟基因药业有限公司 A kind of efficient amplification cultivating system of human vascular endothelial progenitor cells
US11124766B2 (en) 2015-06-12 2021-09-21 Emory University Growth and survival compositions for cells capable of producing antibodies and methods related thereto
CN106906181A (en) * 2017-04-18 2017-06-30 北京康爱瑞浩生物科技股份有限公司 A kind of culture medium for cultivating human umbilical cord mesenchymal stem cells
US11125757B2 (en) 2017-05-26 2021-09-21 Emory University Methods of culturing and characterizing antibody secreting cells
EP3983542A2 (en) 2019-06-17 2022-04-20 CRISPR Therapeutics AG Methods and compositions for improved homology directed repair
US20220325336A1 (en) * 2019-09-05 2022-10-13 Bluebird Bio, Inc. Transduction efficiency assay
KR102337350B1 (en) 2019-11-22 2021-12-08 아주대학교산학협력단 Injectable hydrogel composition having endogenous progenitor or stem cell recruitment and induction of vascular differentiation of recruited cells
WO2022133246A1 (en) 2020-12-17 2022-06-23 Vertex Pharmaceuticals Incorporated Compositions and methods for editing beta-globin for treatment of hemaglobinopathies

Citations (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486329A (en) * 1991-11-25 1996-01-23 Kasai Kogyo Co., Ltd. Automotive door trim and method for fabricating the same
US5612211A (en) * 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
US5654186A (en) * 1993-02-26 1997-08-05 The Picower Institute For Medical Research Blood-borne mesenchymal cells
US5804446A (en) * 1993-02-26 1998-09-08 The Picower Institute For Medical Research Blood-borne mesenchymal cells
US5908782A (en) * 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US5925567A (en) * 1995-05-19 1999-07-20 T. Breeders, Inc. Selective expansion of target cell populations
US5945337A (en) * 1996-10-18 1999-08-31 Quality Biological, Inc. Method for culturing CD34+ cells in a serum-free medium
US6030836A (en) * 1998-06-08 2000-02-29 Osiris Therapeutics, Inc. Vitro maintenance of hematopoietic stem cells
US6037174A (en) * 1993-08-23 2000-03-14 Nexell Therapeutics, Inc. Preparation of serum-free suspensions of human hematopoietic cells or precursor cells
US6225119B1 (en) * 1998-05-22 2001-05-01 Osiris Therapeutics, Inc. Production of megakaryocytes by the use of human mesenchymal stem cells
US6261549B1 (en) * 1997-07-03 2001-07-17 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US6280718B1 (en) * 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US20020031757A1 (en) * 1997-01-24 2002-03-14 Asahi Medical Co., Ltd. Method of regenerating a tissue
US20020037278A1 (en) * 2000-07-26 2002-03-28 Takafumi Ueno Therapeutic angiogenesis by bone marrow-derived cell transplantation in myocardial ischemic tissue and skeletal muscle ischemic tissue
US20020051762A1 (en) * 1998-01-23 2002-05-02 Shahin Rafii Purified populations of endothelial progenitor cells
US6387369B1 (en) * 1997-07-14 2002-05-14 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
US20020061587A1 (en) * 2000-07-31 2002-05-23 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium
US6429012B1 (en) * 1997-10-06 2002-08-06 Viacell, Inc. Cell population containing non-fetal hemangioblasts and method for producing same
US20020114789A1 (en) * 1999-02-08 2002-08-22 Tony Peled Methods of controlling proliferation and differentiation of stem and progenitor cells
US20020142457A1 (en) * 1999-12-28 2002-10-03 Akihiro Umezawa Cell having the potentiality of differentiation into cardiomyocytes
US20020160512A1 (en) * 1999-05-14 2002-10-31 Chu-Chih Shih Ex vivo expansion of mammalian hematopoietic stem cells
US20020164794A1 (en) * 2000-11-03 2002-11-07 Peter Wernet Human cord blood derived unrestricted somatic stem cells (USSC)
US20020197717A1 (en) * 1994-07-20 2002-12-26 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoiesis
US20030032185A1 (en) * 2001-05-03 2003-02-13 Sharkis Saul J. Method of making a homogeneous preparation of hematopoietic stem cells
US20030031651A1 (en) * 2001-04-13 2003-02-13 Lee Ike W. Methods and reagents for cell transplantation
US20030036194A1 (en) * 2001-07-06 2003-02-20 Chunhui Xu Mesenchymal cells derived from human embryonic stem cells
US20030059414A1 (en) * 2001-09-21 2003-03-27 Ho Tony W. Cell populations which co-express CD49c and CD90
US20030077824A1 (en) * 2001-08-31 2003-04-24 Rossi Alexander B. Production of cultured human mast cells and basophils for high throughput small molecule drug discovery
US20030091547A1 (en) * 2001-08-09 2003-05-15 Edelberg Jay M. Platelet-derived growth factor protection of cardiac myocardium
US20030103951A1 (en) * 1997-07-14 2003-06-05 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
US20030113303A1 (en) * 1998-02-05 2003-06-19 Yitzhack Schwartz Homing of embryonic stem cells to a target zone in tissue using active therapeutics or substances
US20030125615A1 (en) * 1998-02-05 2003-07-03 Yitzhack Schwartz Homing of autologous cells to a target zone in tissue using active therapeutics or substances
US20030129750A1 (en) * 1998-02-05 2003-07-10 Yitzhack Schwartz Homing of donor cells to a target zone in tissue using active therapeutics or substances
US20030148512A1 (en) * 2001-12-21 2003-08-07 Fanslow William C. Endothelial stem cells, populations, methods of isolation and use thereof
US20030152558A1 (en) * 2001-11-09 2003-08-14 Christopher Luft Methods and compositions for the use of stromal cells to support embryonic and adult stem cells
US20030157078A1 (en) * 1999-07-20 2003-08-21 University Of Southern California, A California Corporation Identification of a pluripotent pre-mesenchymal, pre-hematopoietic progenitor cell
US20030166272A1 (en) * 1995-02-02 2003-09-04 Abuljadayel Ilham Saleh Method of preparing an undifferentiated cell
US20030180705A1 (en) * 1997-01-24 2003-09-25 Asahi Medical Co., Ltd. Method of regenerating blood vessels
US20030180265A1 (en) * 2002-03-21 2003-09-25 Scott Edward W. Modulating angiogenesis
US20030194802A1 (en) * 2002-04-16 2003-10-16 Technion Research And Development Foundation Ltd. Novel methods for the in-vitro identification, isolation and differentiation of vasculogenic progenitor cells
US20030199464A1 (en) * 2002-04-23 2003-10-23 Silviu Itescu Regeneration of endogenous myocardial tissue by induction of neovascularization
US20030203483A1 (en) * 2000-06-05 2003-10-30 Beerelli Seshi Human mesenchymal progenitor cell
US20030232050A1 (en) * 1999-01-11 2003-12-18 Isner Jeffrey M. Compositions and methods for regulating angiogenesis
US6667034B2 (en) * 1989-06-15 2003-12-23 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture, methods for assaying the effect of substances on lineage-specific cell production, and cell compositions produced by these cultures
US20030235563A1 (en) * 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20030235909A1 (en) * 2002-04-12 2003-12-25 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20040009589A1 (en) * 2002-03-26 2004-01-15 Shulamit Levenberg Endothelial cells derived from human embryonic stem cells
US20040018174A1 (en) * 2002-07-23 2004-01-29 Boston Scientific Corporation Cell therapy for regeneration
US20040052771A1 (en) * 2002-07-12 2004-03-18 Lim Sai Kiang Hemangioblast progenitor cells
US20040071665A1 (en) * 2000-09-05 2004-04-15 Yong-Fu Xiao Method for therapeutically treating a clinically recognized form of cardiopathology in a living mammal
US20040076620A1 (en) * 2002-09-03 2004-04-22 Donnie Rudd Method of repairing primate mammalian tissue
US20040076603A1 (en) * 2002-01-25 2004-04-22 Tony Peled Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US6733746B2 (en) * 1996-03-12 2004-05-11 Invitrogen Corporation Hematopoietic cell culture nutrient supplement
US20040121457A1 (en) * 2002-12-23 2004-06-24 Raquel Castellon Secondary sprouting for isolation and expansion of endothelial sprout cells and endothelial precursor cells from a mixed population and for screening substances
US20040121464A1 (en) * 2002-09-30 2004-06-24 Rathjen Peter David Method for the preparation of cells of mesodermal lineage
US20040121461A1 (en) * 2000-06-26 2004-06-24 Renomedix Institute Inc. Cell fractions containing cells capable of differentiating into neural cells
US20040126879A1 (en) * 2002-08-29 2004-07-01 Baylor College Of Medicine Heart derived cells for cardiac repair
US20040131585A1 (en) * 2000-06-05 2004-07-08 Silviu Itescu Identification and use og human bone marrow-derived endothelial progenitor cells to improve myocardial function after ischemic injury
US20040136973A1 (en) * 2002-11-07 2004-07-15 Eliezer Huberman Human stem cell materials and methods
US20040161419A1 (en) * 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
US20040197310A1 (en) * 2003-02-12 2004-10-07 Sanberg Paul R. Compositions and methods for using umbilical cord progenitor cells in the treatment of myocardial infarction
US20040215334A1 (en) * 2003-04-25 2004-10-28 Brian Fernandes Cellular therapy to heal vascular tissue
US20040228847A1 (en) * 2003-02-28 2004-11-18 Duke University Progenitor cells and methods of using same
US20040235160A1 (en) * 2001-08-07 2004-11-25 Mitsuo Nishikawa Process for preparing hematopoietic stem cells
US20040247575A1 (en) * 2003-06-03 2004-12-09 Caplice Noel M. Smooth muscle progenitor cells
US20040248796A1 (en) * 2003-02-04 2004-12-09 Kari Alitalo VEGF-B and PDGF modulation of stem cells
US20040258670A1 (en) * 2002-12-05 2004-12-23 Case Western Reserve University Cell-based therapies for ischemia
US20050026220A1 (en) * 2001-08-10 2005-02-03 Shahin Rafii Isolation and mobilization of stem cells expressing vegfr-1
US6875430B2 (en) * 1998-03-18 2005-04-05 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US20050276793A1 (en) * 2002-11-15 2005-12-15 The Board Of Trustees Of The University Of Illinois Methods for in vitro expansion of hematopoietic stem cells
US20060281174A1 (en) * 2004-03-09 2006-12-14 Gang Xu Methods for generating insulin-producing cells

Family Cites Families (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4965204A (en) 1984-02-06 1990-10-23 The Johns Hopkins University Human stem cells and monoclonal antibodies
US5130144B1 (en) 1984-02-06 1995-08-15 Univ Johns Hopkins Human stem cells and monoclonal antibodies
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5081030A (en) 1989-04-25 1992-01-14 The Johns Hopkins University Release of cells from affinity matrices
US5605822A (en) 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5635387A (en) 1990-04-23 1997-06-03 Cellpro, Inc. Methods and device for culturing human hematopoietic cells and their precursors
US5409825A (en) 1991-04-09 1995-04-25 Indiana University Foundation Expansion of human hematopoietic progenitor cells in a liquid medium
DK0670735T3 (en) 1992-11-13 1997-07-28 Univ Washington Peripheral Hematopoietic Stem Cells.
EP0669974A1 (en) 1992-11-16 1995-09-06 Rhone-Poulenc Rorer Pharmaceuticals Inc. Pluripotential quiescent stem cell population
DE4240635C2 (en) 1992-12-03 1997-07-10 Lothar Prof Dr Kanz Multiplication of hematopoietic progenitor cells ex vivo and compositions of hematopoietic growth factors
WO1995005843A1 (en) 1993-08-25 1995-03-02 Systemix, Inc. Method for producing a highly enriched population of hematopoietic stem cells
US5972627A (en) 1994-06-15 1999-10-26 Systemix, Inc. Method of purifying a population of cells enriched for dendritic and/or lymphoid progenitors and populations of cells obtained thereby
DE4422667A1 (en) 1994-06-30 1996-01-04 Boehringer Ingelheim Int Process for the production and cultivation of hematopoietic progenitor cells
US5827742A (en) 1994-09-01 1998-10-27 Beth Israel Deaconess Medical Center, Inc. Method of selecting pluripotent hematopioetic progenitor cells
US5677136A (en) 1994-11-14 1997-10-14 Systemix, Inc. Methods of obtaining compositions enriched for hematopoietic stem cells, compositions derived therefrom and methods of use thereof
US5665557A (en) 1994-11-14 1997-09-09 Systemix, Inc. Method of purifying a population of cells enriched for hematopoietic stem cells populations of cells obtained thereby and methods of use thereof
US5876956A (en) 1995-05-15 1999-03-02 Johns Hopkins University School Of Medicine Methods for identification or purification of cells containing an enzymatic intracellular marker
US6015554A (en) 1995-06-07 2000-01-18 Systemix, Inc. Method of reconstituting human lymphoid and dendritic cells
AU6223296A (en) 1995-06-07 1996-12-30 Novartis Ag Methods for obtaining compositions enriched for hematopoieti c stem cells and antibodies for use therein
US5728581A (en) 1995-06-07 1998-03-17 Systemix, Inc. Method of expanding hematopoietic stem cells, reagents and bioreactors for use therein
US5877299A (en) 1995-06-16 1999-03-02 Stemcell Technologies Inc. Methods for preparing enriched human hematopoietic cell preparations
US6306575B1 (en) 1995-06-16 2001-10-23 Stemcell Technologies, Inc. Methods for preparing enriched human hematopoietic cell preparations
US6060052A (en) 1995-10-30 2000-05-09 Systemix, Inc. Methods for use of Mpl ligands with primitive human hematopoietic stem cells
US5980887A (en) 1996-11-08 1999-11-09 St. Elizabeth's Medical Center Of Boston Methods for enhancing angiogenesis with endothelial progenitor cells
DE69841320D1 (en) 1997-02-10 2010-01-07 Harvard College METHOD FOR MODULATING HEMATOPOESIS AND VASCULAR GROWTH
US20020142462A1 (en) 1997-11-26 2002-10-03 Ildstad Suzanne T. Methods for mobilizing hematopoietic facilitating cells and hematopoietic stem cells into the peripheral blood
EP1034288A4 (en) 1997-12-04 2002-11-27 Univ Duke Methods of isolating and using cd7+cd34-lin-hematopoietic cells
CA2311729A1 (en) 1998-01-23 1999-07-29 Imclone Systems Incorporated Purified populations of stem cells
US20030044978A1 (en) 1998-02-05 2003-03-06 Novartis Corporation Expanded and genetically modified populations of human hematopoietic stem cells
US6676937B1 (en) 1998-03-09 2004-01-13 Caritas St. Elizabeth's Medical Center Of Boston Inc. Compositions and methods for modulating vascularization
JP2002516085A (en) 1998-05-29 2002-06-04 トーマス・ジェファーソン・ユニバーシティ Compositions and uses that affect hematopoietic stem cell populations in mammals
US6767737B1 (en) 1998-08-31 2004-07-27 New York University Stem cells bearing an FGF receptor on the cell surface
GB9819912D0 (en) 1998-09-11 1998-11-04 Univ Edinburgh Propagation and/or derivation of embryonic stem cells
CA2343571C (en) * 1998-09-22 2012-01-03 Neuralstem Biopharmaceuticals, Ltd. Stable neural stem cell lines
EP1129176A4 (en) 1998-11-09 2002-10-30 Es Cell Int Pte Ltd Embryonic stem cells
DE69933579D1 (en) * 1998-11-09 2006-11-23 Consorzio Per La Gestione Del SERUM-FREE MEDIUM FOR CHONDROCYTE-SIMILAR CELLS
US20030082152A1 (en) * 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
WO2001000019A1 (en) 1999-06-29 2001-01-04 The Board Of Trustees Of The Leland Stanford Junior University Mammalian myeloid progenitor cell subsets
US6761883B2 (en) 1999-06-29 2004-07-13 The Board Of Trustees Of The Leland Stanford Junior University Mammalian myeloid progenitor cell subsets
AU1100001A (en) 1999-10-29 2001-05-14 Children's Hospital Of Los Angeles Gammac negative hematopoietic progenitor cells
US20020159984A1 (en) 1999-11-12 2002-10-31 Quality Biological, Inc. Cultivation of cells for long term engraftment
US20010049139A1 (en) 2000-03-23 2001-12-06 Eric Lagasse Hepatic regeneration from hematopoietic stem cells
US6936281B2 (en) 2001-03-21 2005-08-30 University Of South Florida Human mesenchymal progenitor cell
US20020132343A1 (en) 2001-03-19 2002-09-19 Clark Lum System and method for delivering umbilical cord-derived tissue-matched stem cells for transplantation
US7432104B2 (en) 2001-08-06 2008-10-07 Bresgen Inc. Methods for the culture of human embryonic stem cells on human feeder cells
US20030124091A1 (en) 2001-10-26 2003-07-03 Large Scale Biology Corporation Endothelial cell derived hematopoietic growth factor
EP1445311A4 (en) 2001-10-30 2005-01-12 Daiichi Seiyaku Co Method of amplyfying hematopoietic stem cells
US20040151700A1 (en) 2001-10-30 2004-08-05 Harlan David M. Ex-vivo rescue of transplantable hematopoietic stem cells following myeloablative injury
DE10158680B4 (en) * 2001-11-30 2004-04-08 Universitätsklinikum Hamburg-Eppendorf Process for ex vivo expansion and differentiation of multipotent stem cells
US20040224403A1 (en) 2001-12-07 2004-11-11 Robarts Research Institute Reconstituting hematopoietic cell function using human embryonic stem cells
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
US20040077985A1 (en) 2002-09-03 2004-04-22 Donnie Rudd Method of replenishing cells damaged by treatment for cancer
US20050054097A1 (en) * 2002-11-17 2005-03-10 Tony Peled EX-VIVO expansion of hematopoietic system cell populations in mononuclear cell cultures
AU2003298775B2 (en) 2002-11-26 2008-07-17 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US20040110286A1 (en) 2002-12-06 2004-06-10 The John P. Robarts Research Institute Method for making hematopoietic cells
CA2515594A1 (en) 2003-02-13 2004-08-26 Anthrogenesis Corporation Use of umbilical cord blood to treat individuals having a disease, disorder or condition
US20040203142A1 (en) 2003-04-14 2004-10-14 Reliance Life Sciences Pvt. Ltd. Growth of neural precursor cells using umbilical cord blood serum and a process for the preparation thereof for therapeutic purposes
US7332334B2 (en) 2003-04-18 2008-02-19 Oklahoma Medical Research Foundation Hematopoietic stem cells treated by in vitro fucosylation and methods of use
DE10326750B4 (en) 2003-06-13 2006-07-27 Gerlach, Jörg, Dr.med. Process for the preparation of a cell preparation and cell preparations prepared in this way
US20050032122A1 (en) 2003-08-06 2005-02-10 Shiaw-Min Hwang Optimizing culture medium for CD34<+> hematopoietic cell expansion
US20050059083A1 (en) 2003-09-15 2005-03-17 Becton Dickinson And Company High throughput method to identify ligands for cell attachment

Patent Citations (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6667034B2 (en) * 1989-06-15 2003-12-23 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture, methods for assaying the effect of substances on lineage-specific cell production, and cell compositions produced by these cultures
US5612211A (en) * 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
US5817773A (en) * 1990-06-08 1998-10-06 New York University Stimulation, production, culturing and transplantation of stem cells by fibroblast growth factors
US5486329A (en) * 1991-11-25 1996-01-23 Kasai Kogyo Co., Ltd. Automotive door trim and method for fabricating the same
US6174526B1 (en) * 1993-02-26 2001-01-16 The Picower Institute For Medical Research Blood-borne mesenchymal cells
US5654186A (en) * 1993-02-26 1997-08-05 The Picower Institute For Medical Research Blood-borne mesenchymal cells
US5804446A (en) * 1993-02-26 1998-09-08 The Picower Institute For Medical Research Blood-borne mesenchymal cells
US6037174A (en) * 1993-08-23 2000-03-14 Nexell Therapeutics, Inc. Preparation of serum-free suspensions of human hematopoietic cells or precursor cells
US20020197717A1 (en) * 1994-07-20 2002-12-26 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoiesis
US20030166272A1 (en) * 1995-02-02 2003-09-04 Abuljadayel Ilham Saleh Method of preparing an undifferentiated cell
US5925567A (en) * 1995-05-19 1999-07-20 T. Breeders, Inc. Selective expansion of target cell populations
US20020022216A1 (en) * 1995-05-19 2002-02-21 T. Breeders, Inc. Selective expansion of target cell populations
US5908782A (en) * 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US6733746B2 (en) * 1996-03-12 2004-05-11 Invitrogen Corporation Hematopoietic cell culture nutrient supplement
US6372210B2 (en) * 1996-10-18 2002-04-16 Quality Biological, Inc. Method for repopulating human bone marrow comprising culturing CD34+ cells in a serum free medium
US5945337A (en) * 1996-10-18 1999-08-31 Quality Biological, Inc. Method for culturing CD34+ cells in a serum-free medium
US6224860B1 (en) * 1996-10-18 2001-05-01 Quality Biological, Inc. Method for repopulating human bone marrow comprising culturing CD34+ cells in a serum free medium
US20020031757A1 (en) * 1997-01-24 2002-03-14 Asahi Medical Co., Ltd. Method of regenerating a tissue
US20030180705A1 (en) * 1997-01-24 2003-09-25 Asahi Medical Co., Ltd. Method of regenerating blood vessels
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6261549B1 (en) * 1997-07-03 2001-07-17 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US6387369B1 (en) * 1997-07-14 2002-05-14 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
US20030103951A1 (en) * 1997-07-14 2003-06-05 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
US20020150561A1 (en) * 1997-10-06 2002-10-17 Morey Kraus Cell population containing non-fetal hemangioblasts and method for producing same
US6429012B1 (en) * 1997-10-06 2002-08-06 Viacell, Inc. Cell population containing non-fetal hemangioblasts and method for producing same
US20020051762A1 (en) * 1998-01-23 2002-05-02 Shahin Rafii Purified populations of endothelial progenitor cells
US20030125615A1 (en) * 1998-02-05 2003-07-03 Yitzhack Schwartz Homing of autologous cells to a target zone in tissue using active therapeutics or substances
US20030129750A1 (en) * 1998-02-05 2003-07-10 Yitzhack Schwartz Homing of donor cells to a target zone in tissue using active therapeutics or substances
US20030113303A1 (en) * 1998-02-05 2003-06-19 Yitzhack Schwartz Homing of embryonic stem cells to a target zone in tissue using active therapeutics or substances
US6875430B2 (en) * 1998-03-18 2005-04-05 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US6225119B1 (en) * 1998-05-22 2001-05-01 Osiris Therapeutics, Inc. Production of megakaryocytes by the use of human mesenchymal stem cells
US20010005591A1 (en) * 1998-05-22 2001-06-28 Osiris Therapeutics, Inc. Production of megakaryocytes by the use of human mesenchymal stem cells
US6030836A (en) * 1998-06-08 2000-02-29 Osiris Therapeutics, Inc. Vitro maintenance of hematopoietic stem cells
US20030232050A1 (en) * 1999-01-11 2003-12-18 Isner Jeffrey M. Compositions and methods for regulating angiogenesis
US20020114789A1 (en) * 1999-02-08 2002-08-22 Tony Peled Methods of controlling proliferation and differentiation of stem and progenitor cells
US20020159981A1 (en) * 1999-02-08 2002-10-31 Gamida-Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
US20020160512A1 (en) * 1999-05-14 2002-10-31 Chu-Chih Shih Ex vivo expansion of mammalian hematopoietic stem cells
US20030157078A1 (en) * 1999-07-20 2003-08-21 University Of Southern California, A California Corporation Identification of a pluripotent pre-mesenchymal, pre-hematopoietic progenitor cell
US6280718B1 (en) * 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
US20020142457A1 (en) * 1999-12-28 2002-10-03 Akihiro Umezawa Cell having the potentiality of differentiation into cardiomyocytes
US20040131585A1 (en) * 2000-06-05 2004-07-08 Silviu Itescu Identification and use og human bone marrow-derived endothelial progenitor cells to improve myocardial function after ischemic injury
US20030203483A1 (en) * 2000-06-05 2003-10-30 Beerelli Seshi Human mesenchymal progenitor cell
US20040121461A1 (en) * 2000-06-26 2004-06-24 Renomedix Institute Inc. Cell fractions containing cells capable of differentiating into neural cells
US20020037278A1 (en) * 2000-07-26 2002-03-28 Takafumi Ueno Therapeutic angiogenesis by bone marrow-derived cell transplantation in myocardial ischemic tissue and skeletal muscle ischemic tissue
US20040136966A1 (en) * 2000-07-31 2004-07-15 The Govt. Of The Usa As Represented By The Secretary Of The Dept. Of Health & Human Services Methods and compositions for the repair and/or regeneration of damaged myocardium
US20040076619A1 (en) * 2000-07-31 2004-04-22 The Government Of The United States Of America As Represented By The Department Of Health And Methods and compositions for the repair and/or regeneration of damaged myocardium
US20020061587A1 (en) * 2000-07-31 2002-05-23 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium
US20040071665A1 (en) * 2000-09-05 2004-04-15 Yong-Fu Xiao Method for therapeutically treating a clinically recognized form of cardiopathology in a living mammal
US20020164794A1 (en) * 2000-11-03 2002-11-07 Peter Wernet Human cord blood derived unrestricted somatic stem cells (USSC)
US20030031651A1 (en) * 2001-04-13 2003-02-13 Lee Ike W. Methods and reagents for cell transplantation
US20030032185A1 (en) * 2001-05-03 2003-02-13 Sharkis Saul J. Method of making a homogeneous preparation of hematopoietic stem cells
US20030036194A1 (en) * 2001-07-06 2003-02-20 Chunhui Xu Mesenchymal cells derived from human embryonic stem cells
US20040235160A1 (en) * 2001-08-07 2004-11-25 Mitsuo Nishikawa Process for preparing hematopoietic stem cells
US20040001807A1 (en) * 2001-08-09 2004-01-01 Jay Edelberg Endothelial precursor cells for enhancing and restoring vascular function
US20030091547A1 (en) * 2001-08-09 2003-05-15 Edelberg Jay M. Platelet-derived growth factor protection of cardiac myocardium
US20050026220A1 (en) * 2001-08-10 2005-02-03 Shahin Rafii Isolation and mobilization of stem cells expressing vegfr-1
US20030077824A1 (en) * 2001-08-31 2003-04-24 Rossi Alexander B. Production of cultured human mast cells and basophils for high throughput small molecule drug discovery
US20030059414A1 (en) * 2001-09-21 2003-03-27 Ho Tony W. Cell populations which co-express CD49c and CD90
US20030152558A1 (en) * 2001-11-09 2003-08-14 Christopher Luft Methods and compositions for the use of stromal cells to support embryonic and adult stem cells
US20030148512A1 (en) * 2001-12-21 2003-08-07 Fanslow William C. Endothelial stem cells, populations, methods of isolation and use thereof
US20040076603A1 (en) * 2002-01-25 2004-04-22 Tony Peled Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US20030180265A1 (en) * 2002-03-21 2003-09-25 Scott Edward W. Modulating angiogenesis
US20040009589A1 (en) * 2002-03-26 2004-01-15 Shulamit Levenberg Endothelial cells derived from human embryonic stem cells
US20030235909A1 (en) * 2002-04-12 2003-12-25 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20030194802A1 (en) * 2002-04-16 2003-10-16 Technion Research And Development Foundation Ltd. Novel methods for the in-vitro identification, isolation and differentiation of vasculogenic progenitor cells
US20040161419A1 (en) * 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
US20030235563A1 (en) * 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20030199464A1 (en) * 2002-04-23 2003-10-23 Silviu Itescu Regeneration of endogenous myocardial tissue by induction of neovascularization
US20040052771A1 (en) * 2002-07-12 2004-03-18 Lim Sai Kiang Hemangioblast progenitor cells
US20040018174A1 (en) * 2002-07-23 2004-01-29 Boston Scientific Corporation Cell therapy for regeneration
US20040126879A1 (en) * 2002-08-29 2004-07-01 Baylor College Of Medicine Heart derived cells for cardiac repair
US20040076620A1 (en) * 2002-09-03 2004-04-22 Donnie Rudd Method of repairing primate mammalian tissue
US20040121464A1 (en) * 2002-09-30 2004-06-24 Rathjen Peter David Method for the preparation of cells of mesodermal lineage
US20040136973A1 (en) * 2002-11-07 2004-07-15 Eliezer Huberman Human stem cell materials and methods
US20050276793A1 (en) * 2002-11-15 2005-12-15 The Board Of Trustees Of The University Of Illinois Methods for in vitro expansion of hematopoietic stem cells
US20040258670A1 (en) * 2002-12-05 2004-12-23 Case Western Reserve University Cell-based therapies for ischemia
US20040121457A1 (en) * 2002-12-23 2004-06-24 Raquel Castellon Secondary sprouting for isolation and expansion of endothelial sprout cells and endothelial precursor cells from a mixed population and for screening substances
US20040248796A1 (en) * 2003-02-04 2004-12-09 Kari Alitalo VEGF-B and PDGF modulation of stem cells
US20040197310A1 (en) * 2003-02-12 2004-10-07 Sanberg Paul R. Compositions and methods for using umbilical cord progenitor cells in the treatment of myocardial infarction
US20040228847A1 (en) * 2003-02-28 2004-11-18 Duke University Progenitor cells and methods of using same
US20040215334A1 (en) * 2003-04-25 2004-10-28 Brian Fernandes Cellular therapy to heal vascular tissue
US20040247575A1 (en) * 2003-06-03 2004-12-09 Caplice Noel M. Smooth muscle progenitor cells
US20060281174A1 (en) * 2004-03-09 2006-12-14 Gang Xu Methods for generating insulin-producing cells

Cited By (131)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8829166B2 (en) 2002-06-26 2014-09-09 Zimmer Orthobiologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
US8834914B2 (en) 2003-12-11 2014-09-16 Zimmer, Inc. Treatment methods using a particulate cadaveric allogenic juvenile cartilage particles
US8652507B2 (en) 2003-12-11 2014-02-18 Zimmer, Inc. Juvenile cartilage composition
US8765165B2 (en) 2003-12-11 2014-07-01 Zimmer, Inc. Particulate cartilage system
US8784863B2 (en) 2003-12-11 2014-07-22 Zimmer, Inc. Particulate cadaveric allogenic cartilage system
US8518433B2 (en) 2003-12-11 2013-08-27 Zimmer, Inc. Method of treating an osteochondral defect
US8524268B2 (en) 2003-12-11 2013-09-03 Zimmer, Inc. Cadaveric allogenic human juvenile cartilage implant
US20050261189A1 (en) * 2004-04-16 2005-11-24 Hydra Biosciences, Inc. Methods of promoting cardiac cell proliferation
US20080078000A1 (en) * 2004-10-19 2008-03-27 Regeneron Pharmaceuticals, Inc. Conditioned culture media
US10039269B2 (en) 2004-10-19 2018-08-07 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating a mouse
US8816150B2 (en) * 2004-10-19 2014-08-26 Regeneron Pharmaceuticals, Inc. Conditioned culture media
US9414575B2 (en) 2004-10-19 2016-08-16 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating a mouse
US9730434B2 (en) 2004-10-19 2017-08-15 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating a mouse
US8480757B2 (en) 2005-08-26 2013-07-09 Zimmer, Inc. Implants and methods for repair, replacement and treatment of disease
US8945535B2 (en) 2005-09-20 2015-02-03 Zimmer Orthobiologics, Inc. Implant for the repair of a cartilage defect and method for manufacturing the implant
US20080269895A1 (en) * 2005-09-20 2008-10-30 Steinwachs Matthias R Implant for the Repair of a Cartilage Defect and Method for Manufacturing the Implant
US9074176B2 (en) 2006-01-13 2015-07-07 Two Cells Co., Ltd. Culture medium additive for use in serum-free culturing of animal cell, kit and use thereof
CN105296415A (en) * 2006-03-23 2016-02-03 普拉里斯坦有限公司 Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
AU2007228341B2 (en) * 2006-03-23 2013-10-17 Pluri Biotech Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
EP3091071B1 (en) 2006-03-23 2019-07-03 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US20160022738A1 (en) * 2006-03-23 2016-01-28 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US20110293583A1 (en) * 2006-03-23 2011-12-01 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US20110129447A1 (en) * 2006-03-23 2011-06-02 Pluristem Ltd. Methods for Cell Expansion and Uses of Cells and Conditioned Media Produced Thereby for Therapy
US20110171182A1 (en) * 2006-03-23 2011-07-14 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US20100047211A1 (en) * 2006-05-19 2010-02-25 The Johns Hopkins University Method of growth of mesenchymal cells under non-adherent conditions for clinical applications
WO2007136760A3 (en) * 2006-05-19 2008-02-14 Univ Johns Hopkins Method of growth of mesenchymal cells under non-adherent conditions for clinical applications
WO2007136760A2 (en) * 2006-05-19 2007-11-29 The Johns Hopkins University Method of growth of mesenchymal cells under non-adherent conditions for clinical applications
US20100040582A1 (en) * 2006-05-31 2010-02-18 Nadir Askenasy Methods of selecting stem cells and uses thereof
US8435786B2 (en) 2006-05-31 2013-05-07 Cellect Biotechnology Ltd. Methods of selecting stem cells and uses thereof
US20070292949A1 (en) * 2006-06-20 2007-12-20 Genzyme Corporation Serum-free media and their uses for chondrocyte expansion
US8497121B2 (en) 2006-12-20 2013-07-30 Zimmer Orthobiologics, Inc. Method of obtaining viable small tissue particles and use for tissue repair
US9873862B2 (en) 2007-01-22 2018-01-23 The Board Of Trustees Of The University Of Arkansas Maintenance and propagation of mesenchymal stem cells
US10676718B2 (en) 2007-01-22 2020-06-09 The Board Of Trustees Of The University Of Arkansas Maintenance and propagation of mesenchymal stem cells
US9138318B2 (en) 2007-04-12 2015-09-22 Zimmer, Inc. Apparatus for forming an implant
WO2008130650A1 (en) * 2007-04-19 2008-10-30 University Of Massachusetts Medical School Thermal responsive polymer siloxanes, compositions, and methods and applications related thereto
US20090061517A1 (en) * 2007-05-31 2009-03-05 Kisaalita William S Cell culture apparatus and methods of making and using same
CN107050057A (en) * 2007-09-19 2017-08-18 普拉里斯坦有限公司 Adherent cell from fat or placenta tissue and its purposes in the treatment
EP3103463B1 (en) 2007-09-19 2020-01-01 Pluristem Ltd. Adherent cells from adipose or placenta tissues and use thereof in therapy
CN107028981A (en) * 2007-09-19 2017-08-11 普拉里斯坦有限公司 Adherent cell from fat or placenta tissue and its purposes in the treatment
US8563312B2 (en) 2008-01-30 2013-10-22 Geron Corporation Synthetic surfaces for culturing stem cell derived cardiomyocytes
US20090191634A1 (en) * 2008-01-30 2009-07-30 Arthur W Martin (meth)acrylate surfaces for cell culture, methods of making and using the surfaces
US8513009B2 (en) 2008-01-30 2013-08-20 Geron Corporation Synthetic surfaces for culturing stem cell derived oligodendrocyte progenitor cells
US8241907B2 (en) 2008-01-30 2012-08-14 Geron Corporation Synthetic surfaces for culturing stem cell derived cardiomyocytes
US9745550B2 (en) 2008-01-30 2017-08-29 Asterias Biotherapeutics, Inc. Synthetic surfaces for culturing stem cell derived cardiomyocytes
US8168433B2 (en) 2008-01-30 2012-05-01 Corning Incorporated Cell culture article and screening
US8354274B2 (en) 2008-01-30 2013-01-15 Geron Corporation Synthetic surfaces for culturing cells in chemically defined media
US10221390B2 (en) 2008-01-30 2019-03-05 Asterias Biotherapeutics, Inc. Synthetic surfaces for culturing stem cell derived oligodendrocyte progenitor cells
US20090191632A1 (en) * 2008-01-30 2009-07-30 Fadeev Andrei G Swellable (meth)acrylate surfaces for culturing cells in chemically defined media
US8329469B2 (en) 2008-01-30 2012-12-11 Geron Corporation Swellable (meth)acrylate surfaces for culturing cells in chemically defined media
US20090191627A1 (en) * 2008-01-30 2009-07-30 Andrei Gennadyevich Fadeev Synthetic surfaces for culturing cells in chemically defined media
US20090191633A1 (en) * 2008-01-30 2009-07-30 Christopher Bankole Shogbon Synthetic Surfaces for Culturing Stem Cell Derived Cardiomyocytes
US20090191626A1 (en) * 2008-01-30 2009-07-30 Christopher Bankole Shogbon Synthetic Surfaces for Culturing Stem Cell Derived Oligodendrocyte Progenitor Cells
US20090203065A1 (en) * 2008-01-30 2009-08-13 Jennifer Gehman Cell culture article and screening
EP2098244A1 (en) * 2008-03-04 2009-09-09 Medizinische Hochschule Hannover Pharmaceutical composition for the treatment of myocardial infarction
US8932858B2 (en) 2008-03-07 2015-01-13 Corning Incorporated Modified polysaccharide for cell culture and release
US20110111498A1 (en) * 2008-03-17 2011-05-12 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
US8716018B2 (en) 2008-03-17 2014-05-06 Agency For Science, Technology And Research Microcarriers for stem cell culture
US9340770B2 (en) 2008-03-17 2016-05-17 Agency For Science, Technology And Research Microcarriers for stem cell culture
US9458431B2 (en) 2008-03-17 2016-10-04 Agency For Science, Technology And Research Microcarriers for stem cell culture
US20110129919A1 (en) * 2008-03-17 2011-06-02 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
US20120219531A1 (en) * 2008-03-17 2012-08-30 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
US8691569B2 (en) 2008-03-17 2014-04-08 Agency For Science, Technology And Research Microcarriers for stem cell culture
US8828720B2 (en) 2008-03-17 2014-09-09 Agency For Science, Technology And Research Microcarriers for stem cell culture
US8637309B2 (en) 2008-03-17 2014-01-28 Agency For Science, Technology And Research Microcarriers for stem cell culture
WO2009120043A2 (en) * 2008-03-28 2009-10-01 경희대학교 산학협력단 Use for cd45+ cells or a cd45+ cell culture fluid for promoting the growth of mesenchymal stem cells
WO2009120043A3 (en) * 2008-03-28 2009-12-30 경희대학교 산학협력단 Use for cd45+ cells or a cd45+ cell culture fluid for promoting the growth of mesenchymal stem cells
US20100015710A1 (en) * 2008-04-25 2010-01-21 Sunghoon Jung Methods and Compositions for Isolating, Maintaining and Serially Expanding Human Mesenchymal Stem Cells
US20100035344A1 (en) * 2008-06-11 2010-02-11 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
US9682173B2 (en) * 2008-06-11 2017-06-20 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
US9758762B2 (en) 2008-09-02 2017-09-12 Pluristem Ltd. Perfusion bioreactor for culturing CD200—placenta adherent cells
US20110212523A1 (en) * 2008-11-11 2011-09-01 Yukio Kato Differentiation-inducing culture medium additive and use thereof
US10131877B2 (en) 2008-11-11 2018-11-20 Two Cells Co., Ltd. Differentiation-inducing culture medium additive and use thereof
US11278573B2 (en) * 2008-12-05 2022-03-22 Regenexx, LLC Methods and compositions to facilitate repair of avascular tissue
EP3441458A1 (en) * 2009-02-03 2019-02-13 Koninklijke Nederlandse Akademie van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
US10947510B2 (en) 2009-02-03 2021-03-16 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium for epithelial stem cells and organoids comprising the stem cells
EP4253529A3 (en) * 2009-02-03 2024-01-17 Koninklijke Nederlandse Akademie van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
CN102449141A (en) * 2009-03-31 2012-05-09 德克萨斯大学系统董事会 Isolation of human umbilical cord blood-derived mesenchymal stem cells
AU2009343787B2 (en) * 2009-03-31 2015-04-02 The Board Of Regents Of The University Of Texas System Isolation of human umbilical cord blood-derived mesenchymal stem cells
US20120142102A1 (en) * 2009-03-31 2012-06-07 The Board Of Regents Of The University Of Texas System Isolation of human umbilical cord blood-derived mesenchymal stem cells
US20100278783A1 (en) * 2009-05-01 2010-11-04 Biocardia, Inc. Method of Preparing Autologous Cells and Method of Use for Therapy
US10035982B2 (en) 2009-05-01 2018-07-31 Biocardia, Inc. Method of preparing autologous cells and methods of use for therapy
US9301975B2 (en) * 2009-05-01 2016-04-05 Biocardia, Inc. Method of preparing autologous cells and method of use for therapy
US9752123B2 (en) 2009-05-01 2017-09-05 Biocardia, Inc. Method of preparing autologous cells and methods of use for therapy
US20110065809A1 (en) * 2009-09-15 2011-03-17 Medtronic, Inc. Polymerization of Multifunctional Azides, and Polymers Therefrom
US20110207166A1 (en) * 2009-11-06 2011-08-25 Sarah Rivkah Vaiselbuh Human bone marrow microenvironments and uses thereof
US9394521B2 (en) 2010-03-10 2016-07-19 Two Cells Co., Ltd. Cell preparation containing mesenchymal stem cells, and method for producing same
US11034935B2 (en) 2010-07-29 2021-06-15 Koninklijke Nederlandse Akademie Van Wetenschappen Liver organoid, uses thereof and culture method for obtaining them
US9617511B2 (en) 2010-09-07 2017-04-11 The Board Of Regents Of The University Of Texas System Tissue-specific differentiation matrices and uses thereof
US10722541B2 (en) 2011-03-22 2020-07-28 Pluristem Ltd. Methods for treating radiation or chemical injury
US9193954B2 (en) 2011-03-31 2015-11-24 University Of Rochester Methods and compositions for mesenchymal stem cell proliferation
WO2012135813A1 (en) * 2011-03-31 2012-10-04 University Of Rochester Methods and compositions for mesenchymal stem cell proliferation
US11674958B2 (en) 2011-06-29 2023-06-13 Academia Sinica Capture, purification, and release of biological substances using a surface coating
US9541480B2 (en) 2011-06-29 2017-01-10 Academia Sinica Capture, purification, and release of biological substances using a surface coating
WO2013165252A1 (en) * 2012-05-03 2013-11-07 Erasmus University Medical Center Rotterdam Culturing of mesenchymal stem cells
US10167447B2 (en) 2012-12-21 2019-01-01 Zimmer, Inc. Supports and methods for promoting integration of cartilage tissue explants
WO2014120014A1 (en) * 2013-02-01 2014-08-07 Conradus Ghosal Gho Composition and method for preserving, transporting and storing living biological materials
NL2010222C2 (en) * 2013-02-01 2014-08-04 Conradus Ghosal Gho Composition and method for generating a desired cell type and/or tissue type from hair follicular stem cells.
NL2010225C2 (en) * 2013-02-01 2014-08-04 Conradus Ghosal Gho Composition and method for preserving, transporting and storing living biological materials.
US11840706B2 (en) 2013-02-01 2023-12-12 Conradus Ghosal Gho Composition and method for generating a desired cell type and/or tissue type from hair follicular stem cells
US10017741B2 (en) * 2013-03-15 2018-07-10 Stemcell Technologies Inc. Compositions and methods for obtaining enriched mesenchymal stem cell cultures
US20160032248A1 (en) * 2013-03-15 2016-02-04 Stemcell Technologies, Inc. Compositions and methods for obtaining enriched mesenchymal stem cell cultures
NL2011170C2 (en) * 2013-07-15 2015-01-21 Univ Erasmus Medical Ct Method and culture medium for in vitro culturing of stem cells.
WO2015009146A1 (en) * 2013-07-15 2015-01-22 Erasmus University Medical Center Rotterdam Method and culture medium for in vitro culturing of stem cells
CN105452446A (en) * 2013-07-15 2016-03-30 鹿特丹伊拉斯姆斯大学医疗中心 Method and culture medium for in vitro culturing of stem cells
US10150950B2 (en) * 2013-09-05 2018-12-11 Medipost Co., Ltd Method for culturing mesenchymal stem cells according to cell size
US10495644B2 (en) 2014-04-01 2019-12-03 Academia Sinica Methods and systems for cancer diagnosis and prognosis
US10112198B2 (en) 2014-08-26 2018-10-30 Academia Sinica Collector architecture layout design
RU2717983C2 (en) * 2014-10-01 2020-03-27 Санбио, Инк. Induction medium and methods of stem cell cultivation and therapy
US9321994B1 (en) 2014-10-01 2016-04-26 Commence Bio, Inc. Induction medium and methods for stem cell culture and therapy
WO2016053758A1 (en) * 2014-10-01 2016-04-07 Commence Bio, Inc. Induction medium and methods for stem cell culture and therapy
EP4234032A3 (en) * 2014-10-01 2023-09-27 SanBio, Inc. Induction medium and methods for stem cell culture and therapy
CN107002029A (en) * 2014-10-01 2017-08-01 科蒙斯生物公司 The inducing culture and method cultivated and treated for stem cell
US10273449B2 (en) 2014-10-01 2019-04-30 Sanbio, Inc. Induction medium and methods for stem cell culture and therapy
US11046929B2 (en) 2014-10-01 2021-06-29 Sanbio, Inc. Induction medium and methods for stem cell culture and therapy
US10626365B2 (en) 2015-01-26 2020-04-21 Ube Industries, Ltd. Long-term cell-cultivation using polyimide porous membrane and cell-cryopreservation method using polyimide porous membrane
US10443037B2 (en) * 2015-01-26 2019-10-15 Ube Industries, Ltd. Long-term cell-cultivation using polyimide porous membrane and cell-cryopreservation method using polyimide porous membrane
US10246679B2 (en) 2015-03-02 2019-04-02 The University Of Tokyo Method for producing pluripotent stem cell having improved chimera forming ability from primed pluripotent stem cell, and composition and combination therefor
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane
US10605708B2 (en) 2016-03-16 2020-03-31 Cellmax, Ltd Collection of suspended cells using a transferable membrane
US10456423B2 (en) 2016-06-13 2019-10-29 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10213463B2 (en) 2016-06-13 2019-02-26 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US11369640B2 (en) 2016-06-13 2022-06-28 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10426796B2 (en) 2016-06-13 2019-10-01 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10363271B2 (en) 2016-06-13 2019-07-30 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US20190224244A1 (en) * 2016-07-11 2019-07-25 Bonus Therapeutics Ltd. Cell compositions for tissue regeneration
CN109715785A (en) * 2016-07-25 2019-05-03 宇部兴产株式会社 Inhibit the method for the method of the differentiation of stem cell, the method and induction stem cell of modulating stem cell
CN110662833A (en) * 2017-04-25 2020-01-07 北海道公立大学法人札幌医科大学 Method for producing mesenchymal stem cell, marker for treatment effect of mesenchymal stem cell, method for determining treatment effect, and cell preparation containing mesenchymal stem cell
WO2018226435A1 (en) * 2017-06-10 2018-12-13 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US20190025286A1 (en) * 2017-07-20 2019-01-24 Seqvera Ltd. Oy In vitro method for measurement and model-free evaluation of time-invariant biomaterials functions
US10379106B2 (en) * 2017-07-20 2019-08-13 Seqvera Ltd. Oy In vitro method for measurement and model-free evaluation of time-invariant biomaterials functions
CN109609443A (en) * 2019-01-14 2019-04-12 上海银海圣生物科技有限公司 A kind of mouse liver primitive cell culture base, cultural method and incubator

Also Published As

Publication number Publication date
WO2005113749A2 (en) 2005-12-01
WO2005113751A1 (en) 2005-12-01
JP2007536935A (en) 2007-12-20
EP1745125A1 (en) 2007-01-24
US7790458B2 (en) 2010-09-07
EP1756267A2 (en) 2007-02-28
JP2007536936A (en) 2007-12-20
US20050272152A1 (en) 2005-12-08
WO2005113749A3 (en) 2006-08-10

Similar Documents

Publication Publication Date Title
US20050265980A1 (en) Cell culture environments for the serum-free expansion of mesenchymal stem cells
US11198849B2 (en) Method for stem cell culture and cells derived therefrom
Valonen et al. In vitro generation of mechanically functional cartilage grafts based on adult human stem cells and 3D-woven poly (ɛ-caprolactone) scaffolds
Liu et al. Biomimetic three-dimensional cultures significantly increase hematopoietic differentiation efficacy of embryonic stem cells
JP5670053B2 (en) In vitro expansion of postpartum-derived cells using microcarriers
US8192348B2 (en) Engineered blood vessels
Boo et al. Expansion and preservation of multipotentiality of rabbit bone-marrow derived mesenchymal stem cells in dextran-based microcarrier spin culture
US20120295347A1 (en) Methods and Compositions for Producing Endothelial Progenitor Cells from Pluripotent Stem Cells
US20110207175A1 (en) Multi-culture bioreactor system
WO2007103442A1 (en) Biocompatible scaffolds and adipose-derived stem cells
JP2010508851A5 (en)
US20140147419A1 (en) Compositions and methods for formation of bone tissue
JPWO2006093172A1 (en) In vitro amplification of adult stem cells
WO2009097657A1 (en) Method of producing progenitor cells from differentiated cells
WO2009080794A1 (en) Method for preparing cell-specific extracellular matrices
Welter et al. Chondrogenesis from human mesenchymal stem cells: Role of culture conditions
Nandakumar et al. Bioengineered 3D microfibrous‐matrix modulates osteopontin release from MSCs and facilitates the expansion of hematopoietic stem cells
US20120207715A1 (en) Methods and systems for storing and prolonging viability of matrix dependent cells
Gallo et al. Human embryonic stem cell-derived cardiomyocytes: inducing strategies
Gautrot et al. Biomimetic Artificial Bone Marrow Niches for the Scale Up of Hematopoietic Stem Cells
WO2023032945A1 (en) Therapeutic agent for arthropathy, and method for producing therapeutic agent for arthropathy
WO2021039943A1 (en) Method for producing mesenchymal stem cells from living body-derived cell sample containing mesenchymal stem cells
Bosch-Fortea et al. Biomimetic Artificial Bone Marrow Niches for the Scale Up of Hematopoietic Stem Cells
Srinivasan et al. Substrates and surfaces for control of pluripotent stem cell fate and function
Marklein et al. Biomanufacturing of Mesenchymal Stromal Cells for Therapeutic Applications

Legal Events

Date Code Title Description
AS Assignment

Owner name: BECTON, DICKINSON AND COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEN, CHANG;XU, RUILING;HAALAND, PERRY D.;AND OTHERS;REEL/FRAME:016623/0407;SIGNING DATES FROM 20050801 TO 20050808

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION