CA2192064A1 - Three-dimensional cartilage cultures - Google Patents

Three-dimensional cartilage cultures

Info

Publication number
CA2192064A1
CA2192064A1 CA002192064A CA2192064A CA2192064A1 CA 2192064 A1 CA2192064 A1 CA 2192064A1 CA 002192064 A CA002192064 A CA 002192064A CA 2192064 A CA2192064 A CA 2192064A CA 2192064 A1 CA2192064 A1 CA 2192064A1
Authority
CA
Canada
Prior art keywords
cells
stromal
dimensional
framework
living
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
CA002192064A
Other languages
French (fr)
Inventor
Anthony F. Purchio
Michael Zimber
Noushin Dunkelman
Gail K. Naughton
Brian A. Naughton
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.)
Advanced Tissue Sciences Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2192064A1 publication Critical patent/CA2192064A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3886Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells comprising two or more cell types
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B90/00Instruments, implements or accessories specially adapted for surgery or diagnosis and not covered by any of the groups A61B1/00 - A61B50/00, e.g. for luxation treatment or for protecting wound edges
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • A61L27/3629Intestinal tissue, e.g. small intestinal submucosa
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3637Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the origin of the biological material other than human or animal, e.g. plant extracts, algae
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3641Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the site of application in the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3839Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by the site of application in the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3839Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by the site of application in the body
    • A61L27/3843Connective tissue
    • A61L27/3852Cartilage, e.g. meniscus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3895Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells using specific culture conditions, e.g. stimulating differentiation of stem cells, pulsatile flow conditions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/60Materials for use in artificial skin
    • 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
    • C12N5/0655Chondrocytes; Cartilage
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • G01N33/5088Supracellular entities, e.g. tissue, organisms of vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B10/00Other methods or instruments for diagnosis, e.g. instruments for taking a cell sample, for biopsy, for vaccination diagnosis; Sex determination; Ovulation-period determination; Throat striking implements
    • A61B10/02Instruments for taking cell samples or for biopsy
    • A61B10/0233Pointed or sharp biopsy instruments
    • A61B10/025Pointed or sharp biopsy instruments for taking bone, bone marrow or cartilage samples
    • A61B2010/0258Marrow samples
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/0063Implantable repair or support meshes, e.g. hernia meshes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/04Hollow or tubular parts of organs, e.g. bladders, tracheae, bronchi or bile ducts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/04Hollow or tubular parts of organs, e.g. bladders, tracheae, bronchi or bile ducts
    • A61F2/06Blood vessels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/08Muscles; Tendons; Ligaments
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/06Materials or treatment for tissue regeneration for cartilage reconstruction, e.g. meniscus
    • 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
    • C12N2500/34Sugars
    • 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
    • C12N2501/15Transforming 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
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • 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
    • C12N2501/335Glucagon; Glucagon-like peptide [GLP]; Exendin
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/04Screening or testing on artificial tissues
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2533/30Synthetic polymers
    • C12N2533/40Polyhydroxyacids, e.g. polymers of glycolic or lactic acid (PGA, PLA, PLGA); Bioresorbable polymers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Abstract

The present invention relates to a method of stimulating the proliferation and appropriate cell maturation of a variety of different cells and tissues in three-dimensional cultures in vitro using TGF-.beta. in the culture medium. In accordance with the invention, stromal cells, including, but not limited to, chondrocytes, chondrocyte-progenitors, fibroblasts, fibroblast-like cells, umbilical cord cells or bone marrow cells from umbilical cord blood are inoculated and grown on a three-dimensional framework in the presence of TGF-.beta.. Stromal cells may also include other cells found in loose connective tissue such as endothelial cells, macrophages/monocytes, adipocytes, pericytes, reticular cells found in bone marrow stroma, etc. The stromal cells and connective tissue proteins naturally secreted by the stromal cells attach to and substantially envelope the framework composed of a biocompatible non-living material formed into a three-dimensional structure having interstitial spaces bridged by the stromal cells. The living stromal tissue so formed provides the support, growth factors, and regulatory factors necessary to sustain long-term active proliferation of cells in culture and/or cultures implanted in vivo. When grown in this three-dimensional system, the proliferating cells mature and segregate properly to form components of adult tissues analogous to counterparts in vivo.

Description

V~0 9!~133821 2 l 9 ~ ~ ~ 4 .

T~REE - DI~bnoll ~T. CARTIL~GB CULTURES

The invention relates to growing stromal cells, such as uhondLuuyLes~ progenito~ -_hUI~dL ~Cy Les, fibroblasts and/or fibroblast-like cells on a three-dimensional scaffold or rL ~L~ in vitro under conditions which enhance the formation of cartilage in culture. A variety of biodegradable and nonbiodegradable matrices treated with sterilizing agents and/or ploaeduL~s can be used as the scaffold in accordance with the invention. A variety of culture conditions can be adjusted, ~nrll-~;ng the control of physical conditions such as yLas~uLe and/or the addition of growth factors. Additionally, the cultured cells can be genetically engineered to express gene products beneficial to growth, transplantation and/or amelioration of disease conditions.
The resulting three-ta~ ionAl cultures and biological rt~plAt --t cartilage tissue constructs have a variety of applications ranging from transplantztion or implantation in vivo, to screening the effectiveness of cytotoxic ,- a_ and pharmaceutical agents in vitro. The invention is " LLtlted by way of ~- ~loc describing the three-dimensional culture of ohund~u~yLes.
2 . ~r~'v~P. _ OF TXT;: 1L~
Articular cartilages are rt~cpt~ncihlr for providing moveable joints the ability for smooth gliding mction. The articular cartilages are firmly attached to the underlying bones and measure less than Smm in th1r~n~cc in human joints, with considerable variation at~pt~nt~;ng on joint and site within the joint. The articular cartilages are aneural, avascular, and alymphatic. In adult humans, they WO gC,3382l ~ 1 9 2 ~ ~ ~ PC~'flJg~S/0~96 derive their nutrition by a double diffusion system through the synovial membrane and through the dense matrix of the cartilage to reach the chul.dLv~yLe.
The bio~hem; QR ~ composition of articular cartilage includes up to 65-80% water (~r~; ng on the cartilage~, with collagen as the most prevalent organic constituent. Articular cartilage consists of highly speriRli7pd chondrocytes ~u,Lu~l.ded by a dense extracellular matri~ consisting mainly of type II
collagen, proteoglycan and water. Collagen (mainly type II) accounts for about 15-25% of the wet weight and about half the dry weight, except in the superficial zone where it accounts for most of the dry weight. Its concentration is usually ployLessively reduced with increasing depth from the articular surface. The proteoglycan content accounts for up to 10~ of the wet weight or about a quarter of the dry weight. Proteoglycans consist of a protein core to which linear sulfated polysaccharides are attached, mostly in the form of chondroitin sulfate and keratin sulfate. Tn addition to type II collagen, articular collR~n contains several other collagen types tIV, V, IX and X) with distinct structures. There are a variety of interactions between these individual macromolecules, which include both noncovalent associations between proteoglycans and collagens, and covalent bonds between different collagen species.
Resistance of the extracellular matrix to water flow gives cartilage its ability to ~iep~ee high joint loads. It absorbs shûck and ml~;m;7~e stress on s~ l bone (Now et al., 1984, J. Biomech.
17:377-394). Adult cartilage and bone have a limited ability of repair.
Damage of cartilage produced by disease, such as rheumatoid and/or osteoarthritis, or trauma can lead to serious physical deformity and debilitation. As human articular cartilage ages, its tensile properties WO9~1338ZI 2 1 9 2 0 ~ 4 PCTIUS9~107Z96 .

change. The superficial zone of the knee articular cartilage exhibits an increase in ten6ile strength up to the third decade of life, after which it decreases ~ markedly with age as detec~hle damage to type II
collagen occurs at the articular surface. The deep ~ zone cartilage also exhibits a ~Lv~Lessive decrease in tensile strength with increasing age, although coll~gPn content does not decrease. These oLseLvGtions indicate that there are changes in r- '-n;r~l and, hence, ~L-u~LuLGl organization of cartilage with aging that, if sufficiently developed, can prP~icpnce cartilage to traumatic damage. In osteoarthritic cartilage there is excessive damage to type II collagen, resulting in crimping of collagen fibrils. In rheumatoid arthritis, the combined actions of free radicals and pro~P;n~ces released from polymorpholeukocytes cause much of the damage seen at the articular surface. (Tiku et al., l99o, J.
Immunol. 145:690-696). Induction of cartilage matrix degradation and prote;n~CPc by chondrocytes is probably induced primarily by interleukin-1 ~IL-l) or tumor necrosis factor-~ (TNF-~) (Tyler, 1985, Biochem.
J. 225:493-507).
The current therapy for loss of cartilage is repl~r L with a prosthetic material, for example, cilicnnP for jr repairs, or metal alloys for ~oint r~l in L. Pl~ t of prosthetic devices i6 usually associated with loss of underlying tissue and bone without I~CV~LY of the full function allowed by the original cartilage. Serious long-term complications associated with the ~L ~~enCe of a peL~anel.L foreign body can include infection, erosion and instability.
Use of sterilized bone or bone powder or surgical steel seeded with bone cells which were eventually implanted have been largely lln uvv~''ful because of the nvn deyL~dable nature of the cell support.

WO95/33X2l 2 1 ~ 2 ~ 6 4 r c ~ 5 According to one procedure fibroblasts are exposed n vitro for a minimum of three days, to a soluble bone protein capable of stimulating in vitro and/or in vivo a ~L~-dLv~ ic ~espunse. The activated fibroblasts are then transferred in vivo by ~;nin~ them with a blodegradable matrix, or by intra-articular injection or atf~ ~ to allografts and prss~hetic devices.
The disadvantage of this method is that chondLocJ~ ic is not allowed to develop in the short-term cultures and there is an unduly heavy reliance for cartilage synthesis by the exposed fibroblasts at the implant site. Caplan, A., U.S. Pat. No. 4,609,551, issued September 2, 1986.
U.S. Pat. No. 5,041,138 to J.P. Vacanti et al., issued August 20, 1991, describes growth of cartilaginous ~LL~ULUL~R seeding ul.ondLouyLes on biodegradable matrices for ~JhC~ t implantation L~
vivo. Although this system offers the advantage of a greater surface area and exposure to nutrients, the conditions employed for culturing the chondrocytes are routine and no efforts have been made to optimize the conditions for the ~hul-dLuuyLes to produce collagen and other cartilage-type macromolecules.

2.1. Growtb Factors r-' H~ ~e~
Growth factors have paracrine or autocrine ef$ects on cell metabolism and can retard or enhance chondrocyte division, matrix synthesis, and degradation.

2.1.1. Transformin~ Growth Factor-~
TGF-~ refers to a growing family of related dimeric proteins which regulate the growth and differentiation of many cell types (Barnard et al., 1990, Biochem. Biophys. Acta. 1032:79-87; M~C5~
1990, Annu. Rev. Cell. Biol. 6:597-619; Roberts and Sporn, 1990, pp. 419-472 M.~. Sporn and A.B. Roberts W095133821 2 1 9 2 0 6 4 r~

(eds.), Peptide Growth Factors and Their Receptors I, Springer-Verlag, Berlin). Members of this family include TGF~-1 (Derynck et al., 1985, Nature 316: 701-705; Moses et al., 1981, Cancer Res. 41:2842-2848;
Roberts et al., 1981, Proc. Natl. Acad. Sci. USA 78, 5339-5343; Sharples et al., 1987, DNA 6:239-244), TGF-~2 (DeNartin et al., 1987, ENBO J. 6:3676-3677; Hanks et al., 1988, Proc. Natl. Acad. Sci. USA 85, 79-82;
Ikeda et al., 1987, Biorh~mi-stry 26, 2406-2410;
Madisen et al., 1988, DNA 7, 1-8; Marquardt et al., 1987, Biol. Chem. 262:12127-12131, Seyedin et al., 1987, J. Biol. Chem. 262:1946-1949), TGF-~3 (Derynck et al., 1988, EMBO J. 7:3737-3743; Jakowlew et al., 1988, Endocrinnol. 2, 747-755, TGF-~4 (Jakowlew et al., 1988, Mol. Endocrinnol. 2:1064-1069), TGF-~5 (~n~aiAh et al., 1990, J. Biol. Chem. 265:1089-1093), and the more distantly related Nullerian inhibitory substance (Cate et al., 1986, Cell. 45:685-698), the ;nh;hinc (Mason et al., 1985, Nature 318:659-663), the bone morphogenetic proteins (Wozney et al., 1988, Science 242:1528-1534) and OP-1 (Ozkaynak et al., 1990, EMBO J. 9:2085-2093). Newly discov~ed members include OP-2 (Ozkaynak et al., 1992, J. Biol. Chem.
267:25220-25227), GDF-l (Lee, 1990, Mol. Endocrinnol.
4:1034-1040); GDF-3 and GDF-9 (McPherron and Lee, 1993, J. Biol. Chem. 268:3444-3449) and Nodal (Zhou çt al., 1993, Nature 361:S43-546).
TGF-~ was first characterized for its effects on cell proliferation. It both stimulated the anchorage-in~ l~n~r~r~ growth of rat kidney fibroblasts (Roberts et al., 1981), and inhibited the growth of monkey kidney cells (Tucker et al., 1984, Proc. Natl.
Acad. Sci. USA 81:6757-6761). Since then, it has been shown to have many diverse biological effects: it stimulates bone formation (Noda and C~mi 11 ir~re, 1989, Endocrinnol. 124:2991-2995; Joyce et al., 1990, J.
Cell. Biol. 110:2195-2207; Marcelli et al., 1990, J.

Wo95133X21 2 ~ 9 2 0 6 ~ ~5 Bone Mineral Res. 5:1087-1096; Beck Ç~_B11, 1991, J.
Bone Mineral Res. 6:961; Mackie and Trechsel, 1990, J.
Cell. Biol. 110, 2195-2207), induces rat muscle cells to produce cartilag_ _~ecific macromolecules tSeyedin et al., 1984, J. Biol. Chem. 261:5693-5695; Seyedin et al., 1986, J. Biol. Chem. 261:56g3-5695; and Seyedin et al., 1987, J. Biol. Chem. 262:1946-1949~, inhibits the growth of early he~atopoietic progenitor cells (Goey et al., 1989, J. Immunol. 143:877-880), T cells (Kehrl et al., 1986, J. Exp. Med. 163:1037-1050), 8 cells (Kasid et al., 1988, J. Immunol. 141, 690-698), mouse keratinocytes (Pietenpol et al., 1990, Cell 61:777-785; Coffey et al., 1988, Cancer Res.
48:1596-1602) and several human cancer cell lines ~Roberts et al., 1985, Proc. Natl. Acad. Sci. USA
82:119-123; Ranrh~li~ et al., 1987, Biophys. Res.
Co~mun. 148:783-789). It increases the synthesis and secretion of c~ q~n and fibronectin (Ignotz and Massague, 1986, J. Biol. Chem. 261:4337-4345;
Centrella et ~1., 1987, J. Biol. Chem. 262:2869-2874;
Malemud et nl., 1991, J. Cell Physio. 149:152-159;
Galera et al., 1992, J. Cell Physio. 153:59~-606;
Phillips ç~_gl_, 1994, soc. Inv. Derm. 103-2:228-232), accelerates healing of incisional wounds t~ustoe et al., 1987, Science 237:1333-1335~, ~u~Less~s casein synthesis in mouse mammary eYrl~nt~ i n~on et al., 1993, J. Cell. Biol. 120:245-251), inhibits DNA
synthesis and pho~huLylation of pRb in rat liver epithelial aells (~hitson and Itakura, 1992, J. Cell.
Biochem. 48:305-315~, stimulates the production of BFGF binding proteoglycans (Nugent and Edelman, 1992, J. Biol. Chem. 267:21256-21264), modulates rh~srhorylation of the EGF receptor and proliferation of epidermoid carcinoma cells (Goldkorn and M~n~t~l5t~hn, 1992, Cell Growth and Differentiation) and can lead to apoptosis in uterine epithelial cells (Rotello et al., 1991, Proc. Natl. Acad. Sci. USA

WO9S/33821 2 1 9 2 0 6 ~ PCT/U595l07296 .

88:3412-3415), cultured hepatocytes and regressing liver (Oberhammer et al., 1992, Proc. Natl. Acad. Sci.
USA 89:5408-5412). It can mediate cardioprotection against reperfusion injury tLefer et al., 1990, Science 249, 61-64) by inhibiting neutrophil adherence to endothelium (Lefer et al., 1993, Proc. Natl. Acad.
Sci. USA 90:1018-1022), and it protects against experimental autoimmune ~i ~P~D~ in mice (Kuruvilla et al., 1991, Proc. Natl. Acad. Sci. USA 88:2918-2921).
In contrast to the foregoing reports of the ability of TGF-~ to induce the production of cartilage-speci~ic macromolecules in muscle cells and chondL~yLes~ TGF-~ was found to act synergistically with fibroblast growth factor to inhibit the synthesis of collagen type II by chicken sternal chondrocytes (Horton et al., 1989, J. Cell Physio. 141:8-15) and TGF-~ inhibited production of type II collagen in rat chondrocytes (Rosen et al., 1988, J. Cell Physio.
134:337-346). In fact, TGF-~ has emerged as the prototypical inhibitor of the proliferation of most normal cell types in culture as well as in vivo, exhibiting a remarkable diversity o~ biological activity (Alexandrow, M.G., and Moses, H.L., 1995, Cancer Res. 55:1452-1457).
TGF-~1 has been purified from human and porcine blood platelets, Assoian et al., 1983, from human pl~cPnta, Frolick et al., 1983, and rP~hin~nt TGF-~1 is currently available, Gentry et al., 1988, Mol.
- Cell. Biol. 7:3418-3427.

2.1.2. Inculin-like Growth Factors I ~n~ IGF-I ana IGF-II) - Insulin alone is much less potent than IGF-I in stimulating collagen matrix synthesis. Insulin, ~ however, Pnh~nCP~ proteoglycan synthesis in the pIes~n-e of a low concenLL~tion of serum (1%). IGF-I, previously designated somatomedin c, is a potent ~ sl3382l 21 9 ~ G ~
.

inducer of collagen and proteoglycan synthesis in vitro. tLindahl et al., 1987, J. Endocrinnol.
115:263-271; Markower et al., 1989, Cell. Biol. Int.
Rep. 13:259-270).
IGF-II stlmulates DNA and ~NA synthesis and is more potent than IGF-I in stimulating clonal growth in fetal cells, whereas IGF-I is more effective on adult chul.dLu~yLes. IGF-II can stimulate proteoglycan synthesis, but, like insulin, is much less effective than IGF-I (~cQuillan et al., 1986, Biochem. J.
240:423-430).

2.1.3. Growth Normone (GH~
Parenteral administration of GH can stimulate localized growth plate development in vivo.
Hypophysectomy leads to disappearance of IGF-I in growth plate chondrocytes, indicating a cessation of synthesis. On the other hand, treatment with GH, syst~ic~lly or locally, results in the appearance of IGF-I. Reports of direct stimulatory effects of GH on cell growth in vitro (Maro et al., 1989, Endocrinnology 125:1239-1445) conflict with reports that it has no effect (Burch et al., 1985, J. Clin.
Endccrinnol. Metab. 60:747-750).

2.1.~. Qther Growth Factors Epidermal growth factor (EGF) alone has no effect on ~I.ol.dLu~yLe proliferation. Together with insulin, EGF synergistically stimulates proteoglycan synthesis and induces prcliferation of chondLu~yLes. (Osborn et ~ , 1989, J. Orthop. Res. 7:35-42). 8asic fibroblast growth factor (bFGF) inhibits proteoglycan synthesis in fetal articular cartilage (~2 -- -n et al., 1986, J. Cell. Physiol. 127:317-322), but it appears to function additively with IGF-I in adult articular cartilage and stimulates proteoglycan synthesis (Osborn, K.D., et al., 1989, J. Orthop. Res. 7:35-42~.

W095/33821 2 ~ ~ ~ 0 ~ 4 ra~

Platelet-derived growth factor (PDGF) also r~nh Inr~r~c proteoglycan synthesis ~Prins et al., 1982, Arthritis Rheum. 25:1228-1238).
3. 8~MMaRY OF THE ~
The present invention relates to the growth and p~epaL~Lion of cartilage m vitro which can be used for a variety of ~u.~oses in vivo. In accordance with the invention, stromal cells which elaborate cartilage-specific macromolecules and extracellular matrix proteins, are inoculated and grown on three-dimensional f~ -~L~S or biodegradable scaffolds.
The stromal cells, which are inoculated onto the scaffold, may include ch~ndLu~yLes, chol.dLu~yLe-progenitors, fibroblasts, fibroblast-like cells and/or cells capable of producing collagen type II and other collagen types, and proteoglycans which are typically ~L~duaed in cartilaginous tissues tSee Table I, iB~E~). The stromal cells and connective tissue proteins secreted by the stromal cells attach to and substantially envelope the three-dimensional rL JL~
or construct, _ e' of a biocompatible non-living material formed into a three-dimensional ~LLU~LULe~
having interstitial spaces bridged by the stromal cells. The living stromal tissue so formed provides the support, growth factors, and regulatory factors nr~r~r~Sc~ry to sustain long-term active proliferation of stromal cells in culture and/or cultures implanted ln v vo. When grown in this three-dimensional system, the proliferating cells mature and segregate properly to form e ts of adult tissue analogous to counterparts n vivo.
In another ~mho~i t of the invention, the stromal cells are inoculated and grown on a three-n~l rL JLk placed in any container that can be manipulated to allow intermittent p~essuLe changes or in a bioreactor system specially designed for the _ g _ W'O 95/33821 2 1 ~ 2 ~ 6 ~ PC 1'11~95/07296 .

n yitro production of cartilage tissue ~n~LLu~Ls, which allows for pressurization of the chamber during growth and an adequate supply of nutrients to stromal cells by convection.
In yet another c-~o~ of the invention, the stromal cells are stirulated to produce cartilage using ~ g~ cly added growth factors, e.q., TGF-~with or without ascorbate, in culture. Alternatively, the stromal cells can be genetically engineered to express the gsnes for specific types of TGF-~ se.q., TGF-~) for ,uccesDrul and/or i ~v~d LULII~V~L o~
cartilage production post-transplantation.
In yet another P~ho~i L of the invention, the stromal cells can be genetically engineered to express a gene product beneficial for successful and/or improved transplantation. For exanple, the stromal cells can be genetically engineered to express anti-inflammatory gene products to reduce the risk of degen~Lative ~ic~lc~c like rheumatoid arthritis resulting in failure of cartilage due to inflammatory reactions; ç~g., the stromal cells can be engineered to express peptides or polypeptides C~LL --L~ ;ng to the idiotype of neutralizing antibodies for granulocyte-m3~L~hag~ colony stimulating factor (GM-CSF), tumor necrosis factor (TNF), interleukin-2 SIL-2), or other inflammatory cytokines and mediators.
Preferably, the cells are engineered to express such gene products transiently and/or under inducible control during the post-operative Lec~v~Ly period, or as a chimeric fusion protein anchored to the stromal cell, e.a., a chimeric 1ecllle -FZ~ of an intracellular andlor tr~ a~-e domain of a receptor or receptor-like molecule, fused to the gene product as the extracellular domain.
In another alternative embodiment, the stromal cells can be genetically engineered to "knock outH
expression of factors that promote re~ection or WO95/33821 2 ~ 9~0~ PCT~S95/07296 degenerative changes in articular cartilage due to aging, rheumatoid disease or inflammation. For example, expression of pro-inflammatory mediators such as GM-CSF, TNF, IL-l, IL-2 and cytokines can be knocked out in the stromal cells to reduce the risk of ~ inflammation. Likewise, the expression of MHC class II molecules can be knocked out in order to reduce the risk of rejection of the cartilage graft.
In yet another Pmho~ of the invention, the three-dimensional culture system of the invention may afford a vehicle for introducing genes and gene pLoduuLs i vivo to assist or improve the results of the transplantation and/or the use in gene therapies.
For example, genes that prevent or ameliorate symptoms of degenerative changes in cartilage such as rheumatoid disease or inflammatory reactions and bone resorption, may be undële~Lessed or overexpressed in disease conditions and/or due to aging. Thus, the level of gene activity in the patient may be increased or de~Leased, respectively, by gene repl~r t therapy by adjusting the level of the active gene product in gpnptic~lly Pngin~red stromal cells.
In a specific Pmho~i~~~t exemplified by the examples in Sections 6-8, infra, chondrocytes from articular cartilage of New Zealand rabbits or cows were grown in culture in monolayer or on three-dimensional biodegradable, bi- ltible fibrous fL ~Lk or scaffold formed of sterilized polymers such as polyglycolic acid, polylactic acid or other polymers. The ~L~ ~LkS were designed to allow adequate nutrient and gas PYrh~ng~c to the cells until engraftment at the site of engraftment takes place.
Particular benefits were achieved by maintaining the cultures under sterile conditions without inhibiting the growth of cartilage in biodegradable polymers sterilized by rhP~ir~l methods or radiation.
~ TGF-~ was added to the three-dimensional 2 1 9206~
WO95/33821 PCT~95~729~
.

cultures to achieve a greatly increased proliferation and differentiation of chondrocyte cells. The cultured cartilage was characterized by analyzing the cartilage constructs for glycn~min~glycan, collagen I
and II by histology and i ~ tonh~mi~try, bioch~m~ç~l quantitation, Northern-Blot analysis and i -blotting.
4. ~RIEF D~o~It~lON OF THE DRAWING
Figure l is a photograph of rabbit cartilage tissue grown Ln Yitro with or without TGF-~1.

Figure 2 is a photograph of Hematoxylin and Eosin stained cartilage tissue grown n yitro without TGF-~1.

Figure 3 is a photograph of Hematoxylin and Eosin stained cartilage tissue grown n vitro with TGF-~1.

Figure 4 is a photograph o~ Trichrome stained cartilage tissue grown in vitro without TGF-~l to show the ~Lesence o~ collagen.

Figure 5 is a photograph of Trichrome stained cartilage tissue grown n yitro with TGF-~1 to show the pL ~s~-.ce of collagen.

Figure 6 is a photograph of Alcan Blue stained cartilage tissue grown i yitro without TGF-~1 to show~
the presence of glycos~minnglycan.

Figure 7 is a photograph of Alcan Blue stained cartilage tissue grown L~ vitro with TGF-~1 to show the presence of GAG.

Figure 8 is a photograph of cartilage after growth in yitro for eight weeks with or without TGF-~1.

WO95/~3821 2 1 9 2 0 6 4 PCT~US9~1~7296 Figure 9 is a photograph of cartilage grown on radiation sterilized mesh in control, FGF-, and TGF-~l-treated cultures of cartilage.

Figure lO: TGF-~ and ascorbate increase the proliferation of bovine articular chondrocytes.

Figure ll: Detection of collagen type II and GAGs in bovine articular ~1.ondLu~yLe lysates.
A. Bovine articular chondrocytes were grown in complete media containing no additives (lanes l and 5), ascorbate t50 ug/ml, lanes 2 and 6~, TGF-~ (20 ngjm, lanes 3 and 7), or TGF-~ +
ascorbate (lanes 4 and 8). Cell lysates were prepared, fractionated by SDS-PAGE and analyzed by ;mmnn~hlotting using anti-chondroitin sulfate antibody (anti-CS; lanes 1-4) or normal rabbit serum (NRS; lanes 5-8).
B. Bovine articular ch~ndL~yLe lysates were fractionated as in panel A and i nnhlotted with antibody against collagen type II (lanes 1-4) or normal rabbit serum (lanes 5-8). Lanes l and 5, no additives; lanes 2 and 6 plus ascorbate; lanes 3 and 7, plus TGF-~; lanes 4 and 8, plus TGF-~and ascorbate.
C. RNA was prepared from ch~..dLo~yLes which were untreated (lane l), treated with ascorbate (50 ug/ml, lane 2), treated with TGF-~ (20 ng/ml, lane 3), or TGF-~ plus ascorbate (lane 4) and analyzed by Northern blotting using anti-collagen type II probe as described in Materials and Methods.

Figure 12: Immuno-histo~h~ic~l staining of cartilage constructs. Samples were stained with normal rabbit serum (panel A and B), anti-collagen type I (panel C
and D), or anti-collagen type II (panel E and F).

WO95/33821 ~ /9Z ~6 y PCT~S95/07296 Samples shown in panel A, C, and E were grown without TGF-~ while samples shown in panels B, D, and F were grown with TGF-~.

Figure 13: Cartilage-like tissue produced by bovine chu..d~o~yLes grown on PGA scaffolds. Bovine ~h~l~dL~yLes were seeded onto PGA scaffolds and grown for three weeks as described in Materials and Methods with (panel A) or without (panel 8) TGF-~ (20 ng/ml).

Figure 14: ~ematoxylin and eosin staining of 'n Yi~LQ
cartilage tissue. The samples were sectioned and stained with hematoxylin and eosin. Samples shown in panel A and B were grown without TGF-~ and samples shown in panel C and D were grown with TGF-~.

Figure 15 i5 a photograph of cartilage constructs A-grown under static conditions and B - grown in bioreactors.

Figure 16 describes cartilage produced in a bioreactor and stained with: A and B: Hematoxylin/eosin; C and D: Trichrome stain; E and F: Alcan Blue; and G and H: Safranin O. Panels I and J show hematoxylin/eosin and trichrome stain IeD~e~Lrully of a cartilage sample grown under static conditions.

Figure 17: Immunoblotting of bovine articular ~1.o1.d.~yLe lysate. Cell-lysates from bovine ..d,~yLes were PYAminPd by Western blotting using anti-type I collagen (lane l), anti-type II collagen (lane 2), normal goat serum (lane 3), anti-versican (lane 4), normal rabbit serum (lane 5), anti-chondroitin sulfate (lane 6), normal mouse serum (lane 7).

W0~5133821 PCT~SsS1072g6 ~ 21 92064 Figure 18 T -' i .cto~hpmi cal staining of cartilage constructs - cartilage uu.-sLL~uLs produced in a bioreactor was processed for ;- nh;ctochemiCal analysis and stained with: A - antitype II collagen;
B - anti-chondroitin; C - anti-type I collagen; and D
normal rabbit serum.
5, DT~TTT.!n DE~KI~-lON OF THE l~V
THE 8TTMHT~ OF CELL PROLIFERATION
AND APPROPRIATE CELL MATURATION
In accordance with the invention, stromal cells are inoculated onto a three-dimensional framework network or scaffold, and grown in culture to form a living cartilaginous material. The stromal cells may comprise chondrocytes, chondrocyte-progenitors, fibroblasts or fibroblast-like cells with or without additional cells and/or elements described more fully herein. The chondrocytes, fibroblast-like cells and other cells and/or elements that comprise the stroma may be fetal or adult in origin, and may be derived from convenient sources such as cartilage, skin, etc.
Such tissues and/or organs can be obtained by ~L V~L iate biopsy or upon autopsy; cadaver organs may be used to provide a generous supply of stromal cells and elements. Alternatively, umbilical cord and placenta tissue or umbilical cord blood may serve as an advantageous source of fetal-type stromal cells, e.q., chondLuuyLe-progenitors and/or fibroblast-like cells for use in the three-~ nc;nn~l system of the invention.~
Fetal fibroblasts and/or chundLuurLes can be inoculated onto the fL ..JLk to form a "generic"
living stromal tissue for culturing any of a variety of cells and tissues. However, in certain instances, it may be preferable to use a "specific" rather than "generic" stromal system, in which case stromal cells and elements can be obtained from a particular tissue, WO 95/33821 PCI'~US9~/07296 21 92~64 organ, or individual. For example, where the three-dimensional culture is to be used for ~u-yOSeb of transplantation or implantation in vivo, it may be preferable to obtain the stromal cells and elements from the individual who is to receive the transplant or implant. This approach might be ~cp~ri~lly advantageous where immunological rejection of the transplant and/or gra~t versus host disease is likely.
Once inoculated onto the three-~ ion~l matrix or LL . ~ k, the stromal cells will proliferate on the f- - JL~ and form the living stromal tissue which can be used n yivo. The three-dimensional living stromal tissue will sustain active proliferation of the culture for long periods of time. Because openings in the mesh permit the exit of stromal cells in culture, confluent stromal cultures do not exhibit contact inhibition, and the stromal cells continue to grow, divide, and remain functionally active.
The production of cartilage in the three-dimensional culture is i ov~d by the application of intermittent pressurization and adequate supply of nutrients to stromal cells by convection.
Growth factors are not n~r~Cc~ry since they are elaborated by the stromal support matrix. However, growth regulatory factors including, but not limited to, TGF-~ and ascorbate, may be added to the culture.
Because, according to the invention, it is i L~nt to recreate, in culture, the cellular microenvironment found i vivo for cartilage, the extent to which the stromal cells are grown prior to implantation L~ vivo or use n vitro may vary. In addition, the stromal cells grown in the system may be genetically engineered to produce gene products bene~icial to transplantation, e.a. anti-inflammatory factors, e,~., anti-GM-CSF, anti-TNF, anti-IL-1, anti-IL-2, etc.
Alternatively, the stromal cells may be genetically engineered to "knock out" expression o~ native gene W09~l33~21 P~
~ 2 1 ~0~4 products that promote inflammation, e.q., GM-CSF, TNF, IL-l, IL-2, or "knock out" expression of MHC in order to lower the risk of rejection. In addition, the stromal cells may be genetically ~ngin~red for use in gene therapy to adjust the level of gene activity in a patient to assist or improve the results of the cartilage transplantation.
The three-dimensional cultures may also be used Ln vitro for testing the effectiveness or cytotoxicity of pharmaceutical agents, and screening ~ uu--ds.
In yet another application, the three-dimensional culture system may be used in a "bioreactor" to produce cartilage tissue constructs which possess critical bio~h~mical~ physical and structural properties of native human cartilage tissue by culturing the tissue under environmental conditions which are typically experienced by native cartilage tissue. The three-dimensional culture system may be maintained under intermittent and periodic pressurization and chondrocytes are provided an adequate supply of nutrients by convection. Pressure facilitates flow of fluid through the mi~LupuI~Us three-dimensional cartilage construct, thereby improving the supply of nutrients and removal of waste from cells c ~ in the ~ol-aLLu~L.
Although the ApplicAnts are under no duty or obligation to explain the r- ' Ani~m by which the invention works, a number of factors inherent in the three-d;- -iqnAl culture system may contribute to its success:
la) The three-dimensional matrix provides a greater surface area for protein att~- L, and ccnsequently, for the adherence of stromal cells.
(b) Because of the three-dimensionality of the matrix, stromal cells continue to actively grow, in contrast to cells in monolayer cultures, which grow to confluence, exhibit contact inhibition, and cease to ~095/33821 2 1 9 2 064 PCT~SgS10729~

grow and divide. The elaboration of growth andregulatory factors by replicating stromal cells may be partially r~cp~nc1hle for stimulating proliferation and regulating differentiation of cells in culture.
(c) The three~ ;on~l matrix allows for a spatial distribution of c~ r elements which is more analogous to that found in the counterpart tissue ln vivo.
(d) The increase in potential volume for cell growth in the three-~;n~ n~l system may allow the establi6hment of localized microenviLv ~ conducive to c~ r maturation.
(e~ The three~ inn~l matrix r-Y;~iz~c cell-cell interactions by allowing greater potential for - ~ L of migratory cells, such as macrophages, monocytes and possibly lymphocytes in the adherent layer.
(f) It has been recogn;zed that maintenance of a differentiated cellular pheno-y~e requires not only growth/differentiation factors but also the appropriate c~ interactions. The present invention effectively recreates the tissue microenvironment.
The three-~;r-~cinn~l stromal support, the culture system itself, and its maintenance, as well as various uses of the three-dimensional cultures are described in greater detail in the subsections below.
The three-dimensional uLv~Lv~yLe cultures can be subjected to intermittent pL~s~uLlzation by creating elevated ~ essive forces through the plastic bag in which the cultures are housed by merely p;n~h;ng or clamping the outlet valve. The chvlldLv~yLes respond to the ambient ples~ule at the level of cell division.
There i5 an increase in the level of proteoglycans which a: ,~n;~c increases in D~A synthesis.
The three-~ n~l cartilage cultures of the invention are maintained in a bioreactor, a special WO95/33821 PCT~S95107296 2 ~ 9?06~
device for creating intermittent and periodic pressurization and chu.,dLouy-es are provided an adequate supply of nutrients by convection.
M~;ntAininq an adequate supply of nutrients to -undLo~yLe cells tl,Luu~LuuL a r~plAc L cartilage tissue constrUct of approximately 2-5mm ~h; r~n~5 iS
e~LL. -ly important as the apparent density of the construct increases. The bioreactors may include a number of designs inrln~;nq, but not limited to, the "piston-style," hard plastic bioreactor; bellows;
soft plastic bag with "pressure plate"; and soft plastic bag with "roller pins".

5.1. EstAhli~l t Of Three-dimensional Stromal Ti55ue The three-dimensional framework may be of any mater;al and/or shape that: (a) allows cells to attach to it (or can be modified to allow cells to attach to it); and (b~ allows cells to grow in more than one layer. A number of different materials may be used to form the matrix, including but not limited to: nylon (polyamides), dacron (polyestersj, poly~LyLene, polypropylene, polyacrylates, polyvinyl , ~c (ç.a., polyvinylchloride), poly~dlbo~ldte tPVC), polytetrafluorethylene (PTFE, teflon), thermanox ~TPX), nitrocellulose, cotton, polyglycolic acid (PGA), collagen (in the form of sponges, braids, or woven threads, etc.), cat gut sutures, cellulose, gelatin, or other naturally occurring biodegradable materials or synthetic materials, inrln~;ng, for example, a variety of polyl-yd-u~y~lkanoates. Any of these materials may be woven into a mesh, for example, to form the three-~ inn~l fL ~Lk or scaffold.
Certain materials, such as nylon, polystyrene, etc.
are poor substrates for cellular attachment. When these materials are used as the three-dimensional CL 'Olk~ it is advisable to pre-treat the matrix ~09~38~1 P~l~sssl~7~g6 2~9~0~ ~

prior to inoculation of stromal cells in order to enhance the att~l ~ of stromal cells to the matrix.
For example, prior to inoculation wlth stromal cells, nylon matrices could be treated with 0.1 M acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon. Polystyrene could be similarly treated using sulfuric acid. Where the cultures are to be maintained for long periods of time or u~yupleserYed, non-degradable materials such as nylon, dacron, polystyrene, polyacrylates, polyvinyls, teflons, cotton, etc., may be preferred. A convenient nylon mesh which could be used in accordance with the invention is Nitex, a nylon filtration mesh having an average pore cize of 210 ~m and an average nylon riber diameter of 90 ~m (#3-210/36 Tetko, Inc., N.Y.).
Where the three-dimensional culture is itself to be implanted in vivo, it may be preferable to use biodegradable matrices such as polyglycolic acid, catgut suture material, collagen, or gelatin, for example. The polyglycolic acid is commonly sterilized in preparation for long-term in vitro, with ethylene oxide or by irradiating with an electron beam.
Unfortunately, both these p~uceduLes have deleteriou3 effects on the cells growing on the three-dimensional culture matrices. For exa~ple, ethylene oxide is toxic to the cells in culture and therefore, electron beam LL~al L is preferred. However, treatment with the electron beam results in cells falling off the rL ~ before depositing adequate extracellular matrix. The addition of TGF-~ to the cultured cells uve~s -- this problem.
Stromal cells comprising chondrocytes chondLu~yLe-progenitors~ fibroblasts or fibroblast-like cells, with or without other stromal cells and Pl~ c described below, are inoculated onto the framework. Growth factors, such as TGF-~ may be added to the culture prior to, during or 5llhc~qunnt to wo ss/33s2l r~ 5 2~ 9206~

inoculation of the stromal cells. The concenLL~tion of TGF-~ maintained in the cultures can be monitored and adjusted to optimize growth. Alternatively, host cells that are genetically engineered to express and produce TGF-~ may be included in the innC~llnm; such cells can include genetically engineered stromal cells. These cells would serve as a source of TGF-~
or other protein factor(s) in the culture.
Preferably, the gene or coding sequence for TGF-~
would be placed under the control of a regulated promoter, so that production of TGF-~ in culture can be controlled. The genetically engineered cells will be s~L~ened to select those cell types: 1) that bring about the amelioration of symptoms of rheumatoid disease or inflammatory reactions i~ v vo, and 2) escape immunoloyical surveillance and rejection.
Stromal cells such as ch~ndLo~yLes may be derived from articular cartilage, costal cartilage, etc. which can be obtained by biopsy (where appropriate) or upon autopsy. Fibroblasts can be obtained in quantity rather conveniently from foreskin or, alternatively, any a~L~pIiate cadaver organ. Fetal cells, ;n~ ;nq fibroblast-like cells, ~I.ol,d~o~yLe pL~y~llitors, may be obtained from umbilical cord or placenta tissue or nmhili~l cord blood. Such fetal stromal cells can be used to prepare a "generic" stromal or cartilaginous tissue. However, a "specific" stromal tissue may be prepaIed by inoculating the three-dimensional matrix with fibroblasts derived a particular individual who is later to receive the cells and/or tissues grown in culture in accordance with the three-dimensional system of the invention.
Fibroblasts may be readily isolated by di~ ey~ting an appropriate organ or tissue which is to serve as the source of the fibroblasts. This may be readily accomplished using techniques known to those skilled in the art. For example, the tissue or _ . .. _ . . . . . .

wo g5133821 2 1 9 2 3 6 ~ r organ can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that weaken the connections between nQ;ghh~ring cells making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage. ~nzy_atic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination. These include but are not limited to trypsin, ~ILY -tLy~sinl coll~gQnl~e, elastase, and/or hyaluronidase, ~nase, pronase, etc. MQ~h~nic~l disruption can also be accomplished by a number of methods including, but not limited to the use of grinders, blenders, sieves, h~ Qrs~ pressure cells, or sonicators to name but a few. For a review of tissue disaggregation techniques, see Freshney, Culture of Animal Cells. A
~anual of Basic Technique, 2d Ed., A.R. Liss, Inc., New York, 1987, Ch. 9, pp. 107-126.
Fibroblast-like cells may also be isolated from human umbilical cords ~33-44 weeks~. Fresh tissues may be minced into pieces and washed with medium or snap-frozen in liquid nitrogen until further use. The t~mhiljr~l tissues may be disaggregated as described above.
Once the tissue has been reduced to a suspension of individual cells, the s~lcpon~io~ can be fractionated into sllhpop~ tions from which the fibroblasts and/or other stromal cells and/or elements can be ~ht~ i nQ~ . This also may be accomplished using standard te~hni~lQc for cell separation in~ ng but not limited to cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection), separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adl.e~ence properties of the cells in the mixed WO95~33821 2 1 ~ 2 0 6 4 PCT~JS9~/~7296 population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter-streaming centrifugation), unit gravity separation, counter current distribution, electrophoresis and fluu~escence activated cell sorting. For a review of clonal selection and cell separation techniques, see Freshney, Culture of Animal Cells. A Manual of Basic T~rhniqll~c~ 2d Ed., A.R. Liss, Inc., New York, 1987, Ch. 11 and 12, pp. 137-168.
The isolation of chundIouyLes~ chondLuuyLe-progenitors, fibroblasts or fibroblast-like cells may, for example, be carried out as follows: fresh tissue samples are thoroughly washed and minced in Hanks bA1Anc~ salt solution (HBSS) in order to remove serum. The minced tissue is incubated from l-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the ~iCsociAted cells are snSp~n~d, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown. The isolated stromal cells can then be grown to confluency, lifted from the confluent culture and inoculated onto the three-~;r -i~nAl support (see, Naughton et al., 1587, J. Med. 18(3&4):219-250).
Inoculation of the three-dimensional matrix with a high cu..c~n~5~tion of stromal cells, e.a., approximately 106 to 5 x 107 cells/ml, will result in the establishment of the three-dimensional stromal support in shorter periods of time.
In addition to chondrocytes, chondrocyte-progenitors, fibroblasts or fibroblast-like cells, other cells may be added to form the three-~ir -ionAl stromal tissue required to support long term growth in culture. For example, other cells found in loose c~nn~c1ive tissue may be inoculated onto the three-dimensional support along with chondrocytes or WO95/3~21 PCT~S9~0729C
21 92~6~ --fibroblasts. Such cells include but are not limited to endothelial cells, pericytes, ma~Lu~ha~s, monocytes, plasma cells, mast cells, adipocytes, etc.
These stromal cells may readily be derived from appropriate organs including umbilical cord or placenta or umbilical cord blood using methods known in the art such as those ~i~c~lcced above.
Again, where the cultured cells are to be used for transplantation or implantation n vivo it is preferable to obtain the stromal cells from the patient's own tissues. The growth of cells on the three-dimensional support may be further ~nh~nc~d by adding to the fL vlk or coating the fL s~Lk with proteins (e.q., collagens, elastic fibers, reticular fibers) glycoproteins, glycos~min~glycans te-q-~heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc.), a celt~ r matrix, and~or other materials.
After inoculation of the stromal cells, the three-dimensional matrix should be incubated in an appropriate nutrient medium. Many commercially available mQdia such as DMEM, RPMI 1640. Fisher~s Iscove's, McCoy's, and the like may be suitable for use. It is important that the three-dimensional stromal matrix be Sn~p~n~ or floated in the mediu~
during the incubation period in order to ~ ize proliferative activity. In addition, the culture should be "fed" periodically to remove the spent media, ~pop~ te released cells, and add fresh media.
The ~ tion of TGF-~ may be adjusted during these steps. In ~1ulldLo~yL~ cultures, proline, a non-essential amino acid and ascorbate are also included in the cultures.
These procedures are greatly facilitated when carried out using a bioreactor, which i8 a closed system housing the three-dimensional framework inoculated with stromal cells. A bioreactor reduces W0~5/33821 PCT~S95/07296 ~ 2~92064 the possibility of contamination, maintains the cultures under intermittent and periodic pressurization to create environmental conditions that maintain an adequate supply of nutrients to ~h~..dLo~yLe cells thL~u~l.ou~ the cartilage tissue construct by convection.
During the incubation period, the stromal cells will grow linearly along and envelop and colonize the three~ -ci~n~l matrix before b~g;nn;ng to grow into the opr~n; ngc of the matrix. It is important to grow the cells to an appropriate degree which reflects the amount of stromal cells present in the in vivo tissue prior to inoculation of the stromal tissue with the tissue-specific cells.
The opPn;ngs of the fL ~Lk should be of an ~.u~Liate size to allow the stromal cells to stretch across the op~n;ngS. Maintaining actively growing stromal cells which stretch across the fL s~Lk onh~ncoc the production of growth factors which are elaborated by the stromal cells, and hence will support long term cultures. For example, if the opon;ngc are too small, the stromal cells may rapidly achieve confluence but be unable to easily exit from the mesh; trapped cells may exhibit contact inhibition and cease production of the appropriate factors n~cc~ry to support proliferation and maintain long term cultures. If the opon;ngc are too large, the stromal cells may be unable to stretch across the opening; this will also decrease stromal cell production of the ~Lu~.iate factors n~c~cq~ry to support proliferation and maintain long term cultures.
When using a mesh type of matrix, as exemplified herein we have found that op~n;nqC ranging from abut 150 ~m to about 220 ~m will work satisfactory.
However, dorr~n~; ng upon the three-dimensional structure and intricacy of the fL il~Lh, other sizes may work equally well. In fact, any shape or WO95/33821 2 1 9 2 0 6 4 PCT~lS95107296 structure that allow the stromal cells to stretch and continue to replicate and grow for lengthy time periods will work in accordance with the invention.
Different proportions of the various types of collagen deposited on the LL ..~Lk can affect the growth of later inoculated tissue-specific pa~ 1.y."al cells. For three-dimensional skin culture systems, collAqDn types I and III are preferably deposited in the initial matrix. The proportions of collagen types deposited can be manipulated or ~nh~n~efl by selecting fibroblasts which elaborate the appropriate collagen type. This can be ~ h~d using monoclonal ant;bo~i~e of an appropriate isotypes or subclass that is capable of acti~ating complement, and which define particular collagen type. These antibodies and complement can be used to negatively select the fibrcblasts which express the desired collagen type.
Alternatively, the stroma used to inoculate the matrix can be a mixture of cells which synthesize the appropriate collagen types desired. The distribution and origins o~ the five types of collagen is shown ln Table I.

W09~/33821 P~
21 9~06~

TABLE I
DISTRIBUTIONS AND ORIGINS OF
THE FIVE TYPES oF CnT~T~A~.EN
Collagen Principal Tissue Distribution Cells of origin connective tissue; reticular cells;
collagen fibers smooth muscle cells Fibrocartilage 80ne Osteoblast Dentin Odontoblasts II Hyaline and elastic Chondrocytes cartilage Vitreous body of eye Retinal cells III Loose ccnnective tissue; Fibrcblasts and reticular fibers reticular cells Papillary layer of dermis Blood vessels Smooth muscle cells;
endothelial cells IV Basement membranes Epithelial and endothelial cells Lens capsule of eye Lens fibers V Fetal membranes; Fibroblast placenta Basement membranes Bone Smooth muscle Smooth muscle cells Thus, d~p~n~ i ng upon the tissue to be cultured and the collagen types desired, the appropriate stromal cell(s) may be selected to inoculate the three-~i- ion~l matrix. For example, for the growth and preparation of cartilage, ~LondL~yLes~ chondrocyte-W0~33821 2 ~ 9 2 0 ~ ~ r~l,u~

progenitors, fibroblasts or fibroblast-like cells should be used.
During incubation of the three-di~ensional stromal support, proliferating cells may be released from the matrix. These released cells may stick to the walls of the culture vessel where they may continue to proliferate and form a confluent monolayer. This should be prevented or ~;n;~;7~A~ for example, by removal of the released cells during feeding, or by transferring the three-dimensional stromal matrix to a new culture vessel. The ~L~Ecnce of a confluent monolayer in the vessel will "shut down" the growth of cells in the thrce-~; ~; on~ 1 matrix an~/or culture. Removal of the confluent monolayer or transfer of the matrix to fresh media in a new vessel will restore proliferative activity of the three-dimensional culture system. 5uch removal or transfers should be done in any culture vessel which has a stromal monolayer ~x~e~in9 25~ confluency.
Alternatively, the culture system could be agitated to prevent the released cells from sticking, or instead of perio~ically feeding the cultures, the culture system could be set up so that fresh media contimloncly flows through the system by convection.
The flow rate could be adjusted to both maximize proliferation within the three~ ion~l culture, and to wash out and remove cells released from the matrix, so that they will not stick to the walls of the vessel and grow to confluence. In any case, the released stromal cells can be collected and es~rv~d for future use.

5.2. Uses of the Three-Dimension~l Culture _vstem The three-dimensional culture system of the invention can be used in a variety of applications.
These include but are not limited to transplantation or implantation of either the cultured cells obtained W095/33821 2 1 9 2 ~ 6 4 PCT~S95/~7296 from the matrix, or the cultured matrix itself in vivo; screening the effectiveness and cytotoxicity of _ ~c, allergens, growth/regulatory factors, phAr~-~outical ~ , etc., n vitro; elucidating the - ~n;c~ of certain A;ce~c~c; studying the n; c~ by which drugs and/or growth factors operate; diagnosing and monitoring cancer in a patient; gene therapy; and the production of biologically active products, to name but a few.

5.2.l. Trans~lantation In Vivo The biological replacement cartilage tissue co1lDLLu~Ls p~uduced in the three-dimensional culture system of the invention can be used to replace or augment existing cartilage tissue, to introduce new or altered tissue, to modify artificial prostheses, or to join biological tissues or sLLuuLuLes. For example, and not by way of limitation, specific ~ -';r-nts of the invention would include i) hip prostheses coated with replA~ - t cartilage tissue oull~LLuuLs grown in three-dimensional cultures; ii~ knee leuu1l~LL~ction with cartilage tissue co1l~LLuuLs; and iii) prostheses of other joints requiring reconstruction and/or replacement of articular cartilage.
The evaluation of internal derangements of articular cartilage in several articulations, ;nr1uA;ng the knee, hip, elbow, ankle and the gl -~ ~l joint, has been made possible by arthroscopic techniques. Arthroscopic surgery has become increasingly popular as well as , ~c~c~rul, e.q., uus small cutting tools, 3 to 4mm in diameter can be used in the knee. Triangulation, in which the operating in~LL, Ls are brought into the visual field provided by the aLLhLoscu~e, requires multiple portals of entry; alternatively, the cutting tools can be passed through a channel in the aLLh~oscope itself in which case only one opening in wo gS133X21 2 1 9 2 0 6 4 .

the joint i5 n~u-SS~y (Jackson, R.W., 1983, J. Bone Joint Surg. [AM] 65:416. Selective removal of the injured or deteriorated portion with arthroscopic surgery, followed by cartilage grafting can be employed successfully. Cartilage tissue constructs can also be employed in major reconstructive surgQry for different types of joints. Detailed ~LuaeduL~5 have been described in Resnick, D., and Niwayama, G., eds., 1988, Dia~nosis of Bone and Joint Disorders, 2d ed.l W.B. Sanders Co.
Three-dimensional tissue culture implants may, according to the inventions, be used to replace or augment existing tissue, to il--Luduce new or altered tissue, or to join together biological tissues or ~LL U.;LUr~=S .

S.2.2. 6~ S~ ing Effectiveness and cvtotoxicitY o~ Comoouna~ In vitro The three-dimensional cultures may be used n vitro to screen a wide variety o~ compounds, for effectiveness and cytotoxicity of pharmaceutical agents, growth/regulatory factors, anti-inflammatory agents, etc. To this end, the cultures are maintainea n vitro and exposed to the ~ __ to be tested.
The activity of a cytotoxic compound can be measured by its ability to damage or kill cells in culture.
This may readily be AcR~Rs~A by vital staining techniques. The effect of growth/requlatory factors may be Acsa~ced by analyzing the cellular content of the matrix, e.a., by total cell counts, and differential cell counts. Thi8 may be accomplished using standard cytological and/or histological techniques ;n~lnAing the use of immunocytochemical techniques employing antibodies that define type-specific callulAr antiqens. The effect of various drugs on normal cells cultured in the three-di~~~~i~nAl gystem may be ARsP-se~A.

W09S/3382l 2 1 9 2 0 6 ~ PCT~S9S/~7~96 .

The three~ in~l cultures of the invention may be used as model systems for the study of physiologic or pathologic conditions. For example, joints that are immobilized suffer relatively quickly in a number of ~ea~evLL. The metabolic activity of chund~v~y~es appears affected, as loss of proteoglycans and an increase in water content are soon observed. The normal white, glistening appearance of the cartilage changes to a dull, bluish color, and the cartilage thickness is reduced.
However, how much of this process is due to nutritional deficiency and how much is due to upset in the stress-~pP~d~nt metabolic homeostasis is not yet clear. The three-~ ion~l chondLv~yLe culture system may be used to determine the nutritional requirements of cartilage under different physical conditions, e.q., intermittent pressurization and by pumping action of nutrient medium into and out of the cartilage c~naLLuu~. This may be especially useful in studying underlying causes for age-related, or injury-related decrease in tensile strength of articular cartilage, e.a., in the knee, that pr~ pnses the weakened cartilage to traumatic damage.
According to the present invention, the three-dimensional chondrocyte cultures may also be used to study the -echAnicp of action of cytokines and other pro-inflammatory mediators released in rheumatic disease in the synovial fluid, ç.q., IL-1, TNF and prostaglandins. The patient's own joint fluid eould be used Ln vitro to study the effect of these _ '- on chundlvvyLe growth and to screen cytotoxic andjor pharmaceutical agents that are most efficacious for a particular patient; i.e., those that prevent resorption of cartilage and enhance the b~lAnred growth of articular cartilage. Those agents could then be used to therapeutically treat the patient.

Wog~/33821 2 1 9 2 0 6 4 PCT~$~5/07296 .

5.2.3. GeneticallY En~ineered Cartil~~e The three~ n~;~nAl culture sy~tem of the invention may afford a vehicle for introducing genes and gene products n Yivo to assist or improve the results of the transplantation and/or for use in gene therapies. For example, the stromal cells can be genetically engineered to express anti-infla~at~Ly gene products to reduce the risk of failure or degenerative changes in the cartilage due to rheumatoid disease of inflammatory reactions. In thi~
regard, the stromal cells can be genetically ~nqin~ored to express anti-inflammatory gene products, for example, peptides or polypeptides corrPcpnn~ing to the idiotype of neutralizing an~i h9d; ~q for granulocyte-macrophage colony stimulating factor ~M-CSF), TNF, IL-1, IL-2, or other inflammatory cytokines. Il-1 has been shown to decrease the synthesis of proteoglycans and collagens type II, IX, and XI (Tyler et al., 1985, Biochem. J. 227:869-878;
Tyier et al., 1988, Coll. Relat. Res. 82: 393-405;
Goldring et al., 1988, J. Clin. Invest. 82:2026-203~;
and Lefebvre ct al., 1gso, siophys. Acta. 1052:366-372. TNF also inhibits synthesis of proteoglycans and type II collagen although it is much less potent than IL-l tYaron, I., et al., 1989, Arthritis Rheu~.
32:173-180; Ikebe, T., et al., 1988, J. Im~unol.
140:827-831; and Saklatvala, J., 1986, Nature 322:547-5~9.
Preferably, the cells are ~qin~red to exprsss such gene products transiently and/or under in~ ihl~
control during the post-operative Leau~Ly period, or as a chimeric fusion protein anul.uL~d to the stromal cells, for example, a chimeric molecule e -6?~ of an intracellular andlor tr~r ' ane domain of a receptor or receptor-like molecule, fused to the gene product as the extrRc~ r domain. In another n~, the stromal cells could be genetically W095l3 21 92064 3821 r~ Il-J~iY:lIU/~,_ PnginPered to express a gene for which a patient is deficient, or which would exert a therapeutic effect, e.~., TGF-~ to stimulate cartilage production, etc.
The genes of interest engineered into the stromal cells need to be related to rheumatoid or joint disease.
The stromal cells can be engineered using a ~ ~in~nt DNA cul,~Ll~L containing the gene used to transform or transfect a host cell which is cloned and then clonally PYp~nAPd in the three-dimensional culture system. The three-dimensional culture which expresses the active gene product, could be implanted into an individual who is deficient for that product.
For example, genes that prevent or ameliorate symptoms of various types of rheumatoid or joint diseases may be und~ .~ssed or down regulated under disease conditions. Specifically, expression of genes involved in preventing inflammatory reactions in rheumatoid or joint diseases may be down-regulated.
Alternatively, the activity of gene products may be fliminichPfl, leading to the manifestations of some or all of the above pathological conditions and eventual devPl~, L of symptoms of rheumatoid or joint ~ir~~ . Thus, the level of gene activity may be increased by either increasing the level of gene product present or by increasing the level of the active gene product which is present in the three-dimensional culture system. The three-dimensional culture which ex~l~s6es the active target gene product can then be implanted into the rheumatoid or joint disease patient who is deficient for that product.
"Target gene," as used herein, refers to a gene involved in rheumatoid or joint fl~cP~cPc in a manner by which modulation of the level of target gene expression or of target gene product activity may act to ameliorate symptoms of rheumatoid or joint fliCPICPq Wo9~133~1 2 1 9 2 ~ 6 4 PCT~9.~0729~) by preventing resorption of cartilage ~nd production of inflammatory mediators by ~hol.dLv~yLes.
Further, patients may be treated by gene rDpl~ t therapy during the post-recovery period after cartilage transplantation. Repl~~
cartilage tissue COI1~LU~LS or sheets may be ~iq specifically to meet the requirements of an individual patient, for example, the stromal cells may be genetically engineered to regulate one or more genes;
or the regulation of gene expression may be transient or long-term; or the gene activity may be non-inducible or inducible. For example, one or more copies of a normal target gene, or a portion of the gene that directs the production of a normal target gene protein product with target gene function, may be inserted into human cells that populate the three-dimensional consLLu~ using either non-inducible vectors including, but are not limited to, adenovirus, adeno-associated virus, and retrovirus vectors, or inducible promoters, including metallothionien, or heat shock protein, in addition to other particles that il~LL~uc~ DNA into cells, such as li,-- -- or direct DNA injection or in gold particles. For example, the gene Dnco~ing the human complement regulatory protein, which prevents re~ection of the graft by the host, may be inserted into human fibroblasts. ~cCurry et al., 1995, Nature M~icin~
1:423-427.
The three-~i inn~l cultures containing such genetically DnginD~red stromal cells, e~a,, either mixtures of stromal cells each expressing a different desired gene product, or a stromal cell engineered to express several specific genes are then implanted into the patient to allow for the amelioration of the symptoms of rheumatoid or joint di5ease. The gene expression may be under the control of a non-inducible (i.e., constitutive) or i"~llcihl~ promoter. The level W0~5/338~1 2 1 q 2 ~ ~ 4 ~ ~ I/L~ JG
.

of gene expression and the type of gene regulated can be controlled ~PpPnrl;ng upon the treatment modality being followed for an individual patient.
The use of the three~ ncion~l culture in gene therapy has a number of advantages. Firstly, since the culture comprises eukaryotic cells, the gene product will be properly ~x~ssed and pLocessed in culture to form an active product. Secondly, gene therapy techniques are useful only if the number of transfected cells can be substantially ~nh~nrP~ to be of clinical value, relevance, and utility; the three-dimensional cultures of the invention allow for PYp~n~i~n of the number of transfected cells and amplification (via cell division) of transfected cells.
A variety of methods may be used to obtain the constitutive or transient expression of gene products engineered into the stromal cells. For example, the transkaryotic implantation technique described by Seldon et al., 1987, Science 236:714-718 can be used.
"Transkaryotic", as used herein, suggests that the nuclei of the implanted cells have been altered by the addition of DNA sequences by stable or transient transfection. The c811s can be engineered using any of the variety of vectors including, but not limited to, integrating viral vectors, e.q., retrovirus vector or adeno-associated viral vectors, or non-integrating replicating vectors, e.~., papilloma virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors. Where transient expression is desired, non-integrating vectors and replication defective vectors may be preferred, since either j~r~l1r;hlP or constitutive promoters can be used in these systems to control expression of the gene of interest. Alternatively, integrating vectors can be used to obtain transient expression, provided the gene of interest is controlled by an ; n~nri hle promoter.

~V095~1 2 ~ ~ 2 ~ ~ 4 PCT~S9~10729C
.

Preferably, the expression control elements used should allow for the regulated expression of the gene so that the product is synthesized only when needed in vivo. The promoter chosen would depend, in part upon the type of tissue and cells cultured. Cells and tissues which are capable of secreting proteins (e.q., those characterized by abundant rough Gn~t~F~ic reticulum, and golgi complex) are preferable. Hosts cells can be transformed with DNA controlled by appropriate expression control elements (e,g., promoter, Gnh~nrGr, s~q~1Gn~G~, transcription terminators, polyadenylation sites, etc.~ and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their ch. -- -- and grow to form foci which, in turn, can be cloned and PYp~n~G~ into cell lines. This method can advantageously be used to engineer cell lines which express the gene protein product.
Any promoter may be used to drive the expression of the inserted gene. For example, viral promoters include but are not limited to the CMV
promoter/Pnh~n~Pr~ SV 40, papillomavirus, ~pstein-Barr virus, elastin gene promoter and ~-globin. If transient expression is desired, such constitutive promoters are preferably used in a non-integrating and/or replication-defective vector. Alternatively, inducible promoters could be used to drive the expression of the inserted gene when necG~ry. For example, in~n~ihlP promoters include, but are not limited to, metallothionien and heat shock protein.
Examples of transcriptional control regions that exhibit tissue specificity which have been described WO95/~3821 2 ~ 6~ PCT~S9~/07296 .

and could be used, include but are nct limited to:
elastase I gene control region which is active in pancreatic acinar cells (Swit et al., 1984, Cell 38:639-646; Ornitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol. 50:399-409; MacDonald, 1987, Hepatology 7:425-515); insulin gene control region which is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115-122); immunoglobulin gene control region which is active in lymphoid cells (GL~JSS~
et al., 1984, Cell 3S:647-658: Adams et al., 1985, Nature 318:533-538; AlPY~n~r et al., 1987, Mol. Cell.
Biol. 7:1436-1444); myelin basic protein gene control region which is active in oliqod~rocyte cells in the brain (R~heAd et al., 1987, Cell 48:703-712); myosin light chain-2 gene control region which is active in skeletal muscle (Shani, 1985, Nature 314:283-286~; and gonadotropic releasing hormone gene control region which is active in the hypo~h~l (Mason et al., 1986, Science 234:1372-1378).
Once genetically engineered cells are implanted into an individual, the presence of the anti-inflammatory gene products, for example, peptides or polypeptides VV1L~LJOn~1;nq to the idiotype of neutralizing an~iho~i~c for GM-CSF, TNF, IL-1, IL-2, or other inflammatory cytokines, can bring about amelioration of the inflammatory reactions associated with rheumatoid or joint disease. IL-1 is a potent stimulator or cartilage resorption and of the production of inflammatory mediators by chu-.d~vvyLes (C -ll et al., 1991, J. Immun. 147:1238-1246).
The stromal cells used in the three-dimensional culture system of the invention may be genetically engineered to "knock cut" expression of factors that promote inflammation or rejection at the implant site.
Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are diccl~cs~d below. "Negative ~0~5l33x2t ~ PCTIllX~51~729~
2192064 ~

modulation", as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level andJor activity of the target gene product in the absence of the modulatory t~e , -nt. The expression of a gene native to stromal cell can be reduced or knocked out using a number of technigues, for example, expression may be inhiblted by inactivating the gene completely ~commonly termed "knockout") using the homologous I~ ~ ;n~tiO!I
technique. Usually, an exon ~nro~ i ng an important region of the protein tor an exon 5' to that region) is interrupted by a positive s~ler~hle marker (for example neo), preventing the production of normal mRNA
from the target gene and resulting in inactivation of the gene. A qene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the s_~uen~es intervening the two regions can be deleted. Mombaerts et al., 1991, Proc. ~at. Acad.
Sci. U.S.A. 88:3084-3087.
Antisense and ribozyme molecules which inhibit expression of the target gene can also be used in accordance with the invention to reduce the level of tar~et gene activity. For example, antisense RNA
molecules which inhibit the expression of major hi~- tibility gene complexes tHLA~ shown to be most versatile with respect to immune r~Cponc~.
Still further, triple helix ~lect~lPc can be utilized in reducing the level of target gene activity. These techniques are described in detail by L.G. Davis et al., eds, Basic Methods in Molecular Bioloqy, 2nd ed., Appleton & Lange, ~orwalk, Conn. 1994.
Using any of the foregoing techniques, the expression of IL-1 can be knocked out in the ~I.ol.dLu~y~es to reduce the risk of resorption of cartilage and production of inflammatory mediators by _ _ _ _ _ _ _ _ _ .

wo gsl33a2l 21 ~ 2 0 ~ ~ rcrluas~lo72sG

the chondrocytes. Likewise, the expression of MHC
class II molecules can be knocked out in order to reduce the risk of rejection of the graft.
In yet another ~ho~i L of the invention, the three-dimensional culture system could be used n vitro to produce biological products in high yield.
For example, a cell which naturally produces large quantities of a particular biological product (e.a., a growth factor, regulatory factor, peptide hormone, antibody, etc.), or a host cell genetically engineered to produce a foreign gene product, could be clonally ~YpAn~d using the three-dimensional culture system in vitro. If the transformed cell excretes the gene product into the nutrient medium, the product may be readily isolated from the spent or conditioned medium using standard separation techniques (e.c., HPLC, column chromatography, electrophoretic techniques, to name but a few). A "bioreactor" has been devised which takes advantage of the flow method for feeding the three-dimensional cultures in vitro. Essentially, as fresh media is passed through the three-dimensional culture, the gene product is washed out of the culture along with the cells released from the culture. The gene product is isolated (e.q., by HPLC column chromatography, electrophoresis, etc.) from the outflow of spent or conditioned media. The bioreactor system is specially designed to allow for YL ~S~UL ization of the chamber during growth of the cartilage tissue and supply nutrients to stromal cells by convection.
6. EXAMPLE: THREB-Dl~L~AI~ ~T~ O -_ L~
CULTURE SYSTBM
The three-dimensional culture of the present invention provides for the replication and colonization of cholldLo~yLes n vitro, in a system comparable to physiologic conditions. Importantly, WO 95133821 2 1 9 2 0 ~ 4 pCT/us9~)729~
.

the chondrocyte cells replicated in this system include all of the cells present in normal cartilage tissue, Acc~.~ing all cell types were present in the original chondrocyte inoculum used to initiate the cultures. Cartilage implants can be of one or more types of cartilage, ~p~n~ing primarily on the location of the implant and the type of cartilage cells seeded onto the polymeric matrix. The following ~ pl~s describe: (i) a method of gro~ing rabbit chondrocytes seeded on a biodegradable polyglycolic acid matrix sterilized by ethylene oxide or electron bea~ with or without TGF-~; (ii) a method of growing bovine chondrocytes on a polyglycolic acid matrix in a culture containing TGF-~ with or without ascorbate;
and iii) a method of growing rabbit chondrocytes on a polyglycolic acid matrix placed in a bioreactor.
Specific conditions used are described below.

6.1. Material an~ ~e~
G.1.1. Growth FActors Recombinant TGF-~ prepared according to Gentry ct al., 1987, Mol. Cell. Biol. 7:3418-3427, was u~ed at the conc~ L~tion of 20ng/ml. Recombinant human beta fibroblast growth factor ~FGF), Pepro Tech., Inc., Rocky Hill, N.J., was used at the concentration of lOmglml. Ascorbic acid or ascorbate was used at the con~ntration of 50~g/ml.

6.1.2. Cells -Cartilage was harvested from articular surfaces of healthy mature (2-3 years old) cows or New Zealand white rabbits (4-8 months old). The cartilage pieces were digested with collagenase (0.2~ weight/volume) in complete media DMEM containing 10% fetal bovine serum, 2mM L-glutamine, non-essential amino acids, 50mg~ml proline, l~M sodium pyruvate and 35 ~g/ml gentamicin for 20 hr. at 37~C. Liberated chondrocytes W0951338~1 2 1 9 2 0 6 ~ ~ r " .
.

were spun, rpcllcpDn~ in complete medium, counted and plated at 106 cells per T-150 flask. Cells were routinely passed at confluence (every 5-7 days).

6.1.3. Cell 8eedina Polyglycolic acid mesh t45mg/cc; non-heat plated;
2mm thick; lcm diameter) was sterilized by ethylene oxide or electron beam (E-beam, 3MR~ treatment and presoaked overnight in complete medium. The mesh was seeded in 6-well dishes with 3-4 x 10~ cells per mesh for 2 days at 37~C in complete medium and placed in an incubator or bioreactor ~p~n~; ng on the protocol used. Medium containing ascorbate was changed two-three times per week. Samples were then washed with PBS and processed for histology after a specific period of growth.

6.1.4. HistoloqY ana ~ ~~iqto~h~mictry Tissues were washed in PBS at harvesting and photographed. They were fixed in 10% buffered formalin, paraffin ~ e~ and stained with hematoxylin and eosin (HhE) and/or trichrome and/or Safranin 0. For some samples, i ~~ictorh~mir~
staining was also performed on paraffin . '~''ed tissue using Biotin-Streptavidin Amplified System tBiogenex Corp.) to unmask antigenic sites. They were then incubated with 3% hydrogen peroxide to block endo~enuus peroxidase activity, rinsed with tris-saline buffer and incubated with biotin block serum (Dako, Inc.) to reduce n~ncpecl~$c bauk~Luu..d.
Blo~ng serum was tapped off and primary ant~ho~i~c to collagen type I and collagen type II were added.
Sections were then incubated with biotinlated anti-lgG, and washed with tris-saline buffer. They were then incubated with horseradish peroxidase-conjugated ~LLe~ idin and washed with tris-saline. Sections WogSl3382l 2 ~ 9 2 0 6 4 PCT~59~/07296 were then incubated with 3,3~ ~;n~b~n~;~ine substrate and counterstained with hematoxylin.

6.1.5. ~ -hlottina Confluent monolayers of articular chondrocytes were scraped in PBS containing 1~ Tween 20. Lupis buffer (as described in Laemmli, U.R., 1970, Nature 227:680-685j was added and cell lysates were fractionated by SDS-PAGE and by ; ~hlotting as described in Burnette, W.N., 1981, Anal. Chem.
112:195-203. Antibodies to collagen type I and II
were obtained from Southern Biochemicals, Inc.
(Birmingham, AL) and anti-chondroitin sulfate antibody was from Sigma (St. Louis, MO).

6.1.6. Ouantitation of CollarJen ana R~a Constructs were frozen lyophilized and stored at -70~C until analysis. The CUI~LL~LS were digested with papain ~lmy/ml) in lOOmM phosphate buffer (pH
6.5) containing 5mM cysteine and 5mm EDTA at 65~C
overnight. Quantitztion of collagen and GAG were det~rmi n~d according to the methods described (Farndale et al., 1986, Biochem. Biophys. Acta 883:73-177; h~c-- er, J.F., 1961, Archiv. Biochem. Biophys.
93:440-447.

6.1.7. ~orthern Blot ~1Ysis RNA was isolated, fractionated on agarose-formaldehyde gels and probed with [37P}-labelled type II collagen cDNA as d,escribed (r~ _y~lski, P., and Sacchi, N., 1987, Anal. Bi-~' 162; Lehrach et al., 1977, Biochemistry 16:4743-4751; and Madisen et al., 1986, DNA 7:1-8).

WO95~33~21 ~ 92~4 PCT~S9~/07296 6.2. Effect of Ethylene Oxide or Electron Beam 8terilization on Rabblt Chond~v~yLe Cultures Polyglycolic acid mesh sterilized with: a~
ethylene oxide or b) electron beam was seeded in six well plates with 3-4 x 106 cells per mesh in a total volume of 10ml (50ml per side) and incubated for 3-4 hr. at 37~C in a tissue culture incubator. At this time, 1.5ml of media were added. The seeded mesh were incubated overnight. 5ml of media were added the next day. Media was changed three times per week for four weeks. Samples were then removed, washed with PBS, and processed for histology: hematoxylin/eosin, trichrome (for collagen); and Alcan blue (for glycosamineglycans).

6.3. Results 6.3.1. Effect of Ethylene Oxide or rl~ L v~ Bea~ 8terilization on Rabbit ~hol~aLv~yLe Cultures ~a) Ethylene Oxide Treated PolvqlYcolic Acid Mesh Cl.ol.dLv~yLes grown in the three-dLmensional matrix in the presence of TGF-~ produced cartilage tissue which was smoother, more glistening and had a more solid consistency than the tissue grown in cultures without the TGF-~ (Fig. 1).
Histological examination of the cartilage tissue using the hematoxylin-eosin stain showed an increase in c~ rity in the cultures without TGF-~ (Fig. 2) compared with those with TGF-~ (Fig. 3).
Staining with trichrome, an indicator of the plesellce of collagen, showed an increase in collagen deposition in cultures grown without TGF-~ (Fig. 4) compared with those grown in cultures with TGF-~(Figure 5).
Alcan blue staining indicated an even distribution and increase in GAG deposition throughout wo ~/33~21 r~

the cartilage tissue when samples were grown in the presence of TGF-~ ~Fig. 7) compared with those without TGF-~ (Fig. 6).
The gross appearance of the tis6ue after 8 weeks indicated that the cartilage grown in the presence of TGF-~ was substantially larger than that grown without TGF~ ig. 8~.
There was a two-fold increase in the dry weight of cartilage grown in cultures containing TGF-~, 20%
more collagen and 80% more GAG. See Table II below.

TABLE II - RABBIT CARTILAGE
Static: 8 Week Wet Dry Total Total% Collagen % GAG
Weight WeightCollagen GAG (Dry (Dry~
Sample(mg) (mg) (mg) (rng) Weight) Weight) TGF-~
64 (n=2) _ 4.8 1.4 273 (n=2) 8.Z 6.0 1.1 73.3 13 - TGF-,B
35(n=2) _ 1.9 0.3 2 50 (n=2) 4.2 2.6 0.3 61.9 7.1 Normal Rabbit Cartilage GAG 15-40%
Collagen 55-80%

(b) Bovine ~I.vdLv~y~es seeded on mesh sterilized by E-beam showed poor growth after four weeks. This is most likely due to the more rapid degradation of PGA after radiation treatment, causing the cells to fall off before being able to deposit adequate extrac~llu~ ~r matrix.

W095/338~l 2 1 9 2 0 6 4 ~CTIUS95/07296 Addition of fibroblast growth factor B (bFGF) had no effect. However, addition of TGF-~ resulted in growth of chondLo~yLes and formation of cartilage tissue by increasing the production of extracellular matrix by the uI.ondLu~yLes. Thus, addition of TGF-~~v~L~es the deleterious effects of sterilizing the mesh by the E-beam and ~nh~nrec the production of cartilage in the three-dimensional culture matrix.
(Fig. 9).
7. Effect of TGF-~ on Growth of Bovine Chondrocytes in ~onolayer Culture ~nd on Three-D;r---io--l FL ~ ..~r~ With or Without ascorb~te a) 80vine chondrocytes were seeded at 2.0 x 105 cells per T-25 flask. Twenty-four hours later they were treated with ascorbate, TGF-~ or both.
b) Polyglycolic acid mesh were seeded with 3 x 106 bovine ul~ulldlu~y~es and cultured in 6-well dishes for two days at 37~C in complete media containing TGF-and with 50mg/ml ascorbate or no ascorbate.

7.1. ESULTS
7.1.1. Effect of TGF-~ on Growth of Bovine Chondhv~es in ~onolayer Culture an~ on Three-D~- -ic--l FL .. ~L k~ With or Without ascorbate ~ a) TGP-~ increases Prolifer~tion A
Bovine Chondrocvtes in ~onolaYer Figure lOA shows that TGF-~ stimulated the growth of bovine chondrocytes in monolayer. Addition of ascorbate had a stimulating effect which was additive with TGF-~. Figure lOB shows a time course for the proliferative effect of TGF-~ in the ~ ence of ascorbate. Stimulation of chondrocyte growth was observed at O.lmg/ml TGF-~ (Figure lOC) and ~FGF had a slight inhibiting effect.
TGF-~ treat~ent also resulted in substantial increase in GAG synthesis (Figure llA) and had no W095~3~2t 2 1 9 2 0 6 4 PCTIU895~7~96 detectable ef~ect on collagen type II (Figure llB).
Northern blot analysis showed that TGF-~ had no effect on collagen type II mRNA levels (Figure llC).
T ~ tting with anti-type I collagen antibody was negative (Figure 12).
~b) Growth of Cartilage C~ns8Lu~Ls On ~kree-Dimension~l Fr~mewor~
Figure 13 shows the cartilage cu~laL~uL5 to be smooth and glistening and three-times larger in the presence of TGF-~ (Figure 13 and Table III). Cells were mostly cu..~enLL~ted on the outside edge while the centers were less dense and contained more undegraded polyglycolic acid fibers (Figure 14). Constructs stained positively for type II collagen (Figures 12E
and F) but not for collagen type I (Figures 12C and D); Table III shows that TGF-~ treatment resulted in about 2.5 fold increase in the dry ~eight of the constructs as well as in collagen and GAG.

TABLE III
BOVINE CARTILAGE
Total Tot~l G~Q
Dry Weight Collagen contont 8Ample TGF-~ (mg) ~g) ~g) 1 - 4.5 31.5 71.0 2 - ~.2 29.3 71.6 3 - 3.6 2S.2 69.3 ~ ~ 11.7 81.8 171.C
~ 11.0 76.9 180.5 6 ~ lt.2 78.3 189.~

These results indicate that TGF-~ is capable of increasing the proliferation of chondrocytes in monolayers as well as increasing cartilage production on three-~;r-n~lonAl fL JL~

Wos~3821 2 1 9 2 0 6 4 r 8. Cartilage Production by Rabbit ~ho~d~v~Le3 on Polyglycolic Ac$d FL - ..Or~ in a Closed Bioreactor 8vstem Polyglycolic acid mesh were seeded with 4 x 106 cells in 6-well dishes for two days and then placed into bioreactors where they were fed continuously using a 16-channel pump (Cole-Parmer: Masterflex with computerized drive, model no. 7550-90) with complete media (250ml for 5 bioreactors) at a flow rate of 50~g/ml for 28 days at 37~C with no media change.
Fresh ascorbate (50~g/ml final concentration) was added every three days. As a separate group of experiments, seeded mesh were cultured statically in 6 well dishes with 5ml of media which was changed twice weekly. The media used for growing cells on polyglycolic acid LL ~I~S was exactly the same as for cell growth in monolayer except for addition of ascorbate.

8.1. RESULTS
8.1.1. Cartilage Pro~v~t~n by Rabbit Chondrocytes on Polyglycolic Acid PrA~ ~L~S in a Closed Bioreactor 8YStem Figure 15A shows the gross appearance of seeded mesh grcwn statically. They were thinner than the c~nsLL~Ls grown in the bioreactor system, which were glistening and about 2mm in th i rkn~eS . Histologic examination with Hematoxylin/Eosin or Trichrome revealed cell growth and deposition of exacellular-matrix throughout the mesh (Figures 16A and 16C).
Alcan blue and Safranin 0 staining showed deposition of GAGs (Figures 16E-16H). Staining of COnSLLU~LS
grown statically showed far less matrix deposition (Figures 16I and J). T 7sLaining showed positive reactivity for type II collagen and chondroitin sulfate and no reactivity for type I collagen (Figure 17). Cartilage produced in the bioreactors stained positively with anti-type II collagen and anti-WO ')5133g21 PCT1115!1~1072g6 2 1 920~4 chondroitin sulfate (Figures 18A and B) but not withanti-type I collagen lFigure 18C). Bior~
analyses showea collagen and GAG values to be 15~ and 25~ dry weiqht, respectively. These values in con~LLuuLs compare favorably with respective publLshed values in rabbit articular cartilage of 30-70~ and 10-30~ of dry weight, respectively.
The present invention is not to be limited in scope by the specific embodiments described which are intended as single illustrations o~ individual aspects of the inventicn, and functionally equivalent methods and ~ t~ are within the scope of the invention, in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and ~rc~~p~nying drawings. Such modifications are intended to ~all within the scope of the appended claims.

Claims (30)

What is claimed is:
1. A method for preparing a living stromal tissue in vitro, comprising culturing stromal cells inoculated onto a three-dimensional framework in a culture medium containing an effective amount of growth factors, so that the stromal cells and connective tissue proteins naturally secreted by the stromal cells attach to and substantially envelope the framework of a biocompatible, non-living material formed into a three dimensional structure having interstitial spaces bridged by the stromal cells to form into a three-dimensional construct.
2. The method of Claim 1 in which the stromal cells are chondrocytes.
3. The method of Claim 1 in which the stromal cells are fibroblasts or fibroblast-like cells.
4. The method of Claim 1 in which the stromal cells are umbilical cord cells or bone marrow cells from umbilical cord blood.
5. The method of claim 1 in which the stromal cells are a combination of chondrocytes, chondrocyte-progenitors, and fibroblasts, fibroblast-like cells, endothelial cells, pericytes, macrophages, monocytes, leukocytes, plasma cells, mast cells, adipocytes, umbilical cord cells, and bone marrow cells from umbilical cord blood.
6. The method of Claim 1 in which the framework is composed of a biodegradable material.
7. The method of Claim 6 in which the biodegradable material is polyglycolic acid, cotton, cat gut sutures, cellulose, gelatin, collagen or polyhydroxyalkanoates.
8. The method of Claim 7 in which the polyglycolic acid is treated with ethylene oxide.
9. The method of Claim 7 in which the polyglycolic acid is treated with an electron beam.
10. The method of Claim 1 in which the framework is composed of a non-biodegradable material.
11. The method of Claim 8 in which the non-biodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, or a nitrocellulose compound.
12. The method of Claim 1 in which the framework is a mesh.
13. The method of Claim 1 further comprising culturing parenchymal cells inoculated onto the living stromal tissue.
14. The method of Claim 13 in which the parenchymal cells comprise chondrocytes, chondrocyte-progenitors, fibroblasts, fibroblast-like cells umbilical cord cells, or bone marrow cells from umbilical cord blood.
15. The method of Claim 1 in which the culture medium further comprises of an effective amount of ascorbate.
16. The method of Claim 1 in which the culture medium is kept under static conditions.
17. The method of Claim 1 in which the culture medium is kept in dynamic state by convection and under periodic pressurization.
18. A method for transplantation or implantation of a living cartilage tissue construct comprising, (a) inoculating stromal cells on a three-dimensional framework;
(b) culturing the stromal cells so that they proliferate in vitro; and (c) implanting the cartilage tissue construct in vivo.
19. A living stromal cell-colonized three-dimensional framework prepared in vitro, comprising stromal cells and connective tissue proteins naturally secreted by the stromal cells which are inoculated upon a three-dimensional framework such that the three-dimensional framework becomes populated with viable cells to form a three-dimensional structure having interstitial spaces bridged by the stromal cells.
20. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the stromal cells are chondrocytes.
21. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the stromal cells are fibroblasts or fibroblast-like cells.
22. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the stromal cells are umbilical cord cells or bone marrow cells from umbilical cord blood.
23. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the framework is treated with ethylene oxide.
24. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the framework is treated with an electron beam.
25. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the framework is composed of a biodegradable material.
26. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the biodegradable material is polyglycolic acid, cotton, cat gut sutures, cellulose, gelatin, collagen or polyhydroxyalkanoates.
27. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the framework is composed of a non-biodegradable material.
28. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the non-biodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, or a nitrocellulose compound.
29. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the framework is a mesh.
30. The living stromal cell-colonized three-dimensional framework of Claim 19 in which the stromal cells comprise chondrocyte-progenitors, fibroblasts, fibroblast-like cells, muscle cells, umbilical cord cells or bone marrow cells from umbilical cord blood.
CA002192064A 1994-06-06 1995-06-06 Three-dimensional cartilage cultures Abandoned CA2192064A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US25409694A 1994-06-06 1994-06-06
US254,096 1994-06-06
US08/463,566 US5902741A (en) 1986-04-18 1995-06-05 Three-dimensional cartilage cultures
US08/463,566 1995-06-05

Publications (1)

Publication Number Publication Date
CA2192064A1 true CA2192064A1 (en) 1995-12-14

Family

ID=26943819

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002192064A Abandoned CA2192064A1 (en) 1994-06-06 1995-06-06 Three-dimensional cartilage cultures

Country Status (8)

Country Link
US (2) US5902741A (en)
EP (1) EP0812351A4 (en)
JP (1) JP3599341B2 (en)
AU (1) AU689605B2 (en)
CA (1) CA2192064A1 (en)
IL (1) IL114017A0 (en)
NZ (1) NZ288467A (en)
WO (1) WO1995033821A1 (en)

Families Citing this family (311)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0781116A4 (en) 1994-09-12 1999-08-25 Advanced Tissue Sciences Inc Three-dimensional human cell cultures on cardiac valve frameworks and their uses
US5648088A (en) 1995-03-06 1997-07-15 Ethicon, Inc. Blends of absorbable polyoxaesters containing amines and/or amide groups
US5906934A (en) 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US6528483B2 (en) 1995-06-07 2003-03-04 André Beaulieu Method of producing concentrated non-buffered solutions of fibronectin
DE69631402T2 (en) 1995-11-06 2004-12-09 Ethicon, Inc. Polymer blends containing polyoxaesters and Lactonpolymeren
CA2236807A1 (en) * 1995-11-06 1997-05-15 Rita Kandel Reconstituted mineralized cartilage tissue
US20020111695A1 (en) * 1995-11-06 2002-08-15 Mount Sinai Hospital Corporation Reconstituted mineralized cartilage tissue
US6569172B2 (en) * 1996-08-30 2003-05-27 Verigen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
US20060025786A1 (en) * 1996-08-30 2006-02-02 Verigen Transplantation Service International (Vtsi) Ag Method for autologous transplantation
US20020173806A1 (en) * 1996-08-30 2002-11-21 Verigen Transplantation Service International (Vtsi) Ag Method for autologous transplantation
US5989269A (en) * 1996-08-30 1999-11-23 Vts Holdings L.L.C. Method, instruments and kit for autologous transplantation
EP0959879B1 (en) * 1996-10-10 2004-05-06 The General Hospital Corporation Photodynamic therapy for the treatment of osteoarthritis
US5919702A (en) * 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
WO1998044874A1 (en) * 1997-04-04 1998-10-15 Barnes-Jewish Hospital Neocartilage and methods of use
DE69714035T2 (en) 1997-08-14 2003-03-06 Sulzer Innotec Ag Composition and device for repairing cartilage tissue in vivo consisting of nanocapsules with osteoinductive and / or chondroinductive factors
US6316522B1 (en) 1997-08-18 2001-11-13 Scimed Life Systems, Inc. Bioresorbable hydrogel compositions for implantable prostheses
AU747166B2 (en) * 1997-10-31 2002-05-09 Children's Medical Center Corporation Bladder reconstruction
CA2221195A1 (en) * 1997-11-14 1999-05-14 Chantal E. Holy Biodegradable polymer matrix
AU1588099A (en) * 1997-11-17 1999-06-07 Beth Israel Deaconess Medical Center Hybrid tissues for tissue engineering
DE19752900A1 (en) * 1997-11-28 1999-06-02 Biotechnolog Forschung Gmbh Bone matrix and / or osteogenic cells, their use, therapeutic preparation and kit
DE19755023C2 (en) * 1997-12-11 2000-03-09 Celanese Chem Europe Gmbh Catalyst and process for making vinyl acetate
US6197586B1 (en) 1997-12-12 2001-03-06 The Regents Of The University Of California Chondrocyte-like cells useful for tissue engineering and methods
US6291240B1 (en) * 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
US6129757A (en) 1998-05-18 2000-10-10 Scimed Life Systems Implantable members for receiving therapeutically useful compositions
IL124722A0 (en) * 1998-06-02 1999-01-26 Oron Amir Ischemia laser treatment
AU771701B2 (en) * 1998-08-14 2004-04-01 Genzyme Corporation Methods, instruments and materials for chondrocyte cell transplantation
IL142914A0 (en) * 1998-11-09 2002-04-21 Consorzio Per La Gestione Del Serum free medium for chondrocyte-like cells
US7001746B1 (en) * 1999-01-29 2006-02-21 Artecel Sciences, Inc. Methods and compositions for the differentiation of human preadipocytes into adipocytes
US20040018226A1 (en) * 1999-02-25 2004-01-29 Wnek Gary E. Electroprocessing of materials useful in drug delivery and cell encapsulation
US6592623B1 (en) 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US20020081732A1 (en) * 2000-10-18 2002-06-27 Bowlin Gary L. Electroprocessing in drug delivery and cell encapsulation
US7615373B2 (en) * 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering
US20040116032A1 (en) * 1999-02-25 2004-06-17 Bowlin Gary L. Electroprocessed collagen
US6197061B1 (en) * 1999-03-01 2001-03-06 Koichi Masuda In vitro production of transplantable cartilage tissue cohesive cartilage produced thereby, and method for the surgical repair of cartilage damage
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US6315992B1 (en) * 1999-06-30 2001-11-13 Tissuegene Co. Generating cartilage in a mammal using fibroblasts transfected with a vector encoding TGF-β-1
US6287340B1 (en) 1999-05-14 2001-09-11 Trustees Of Tufts College Bioengineered anterior cruciate ligament
US6372494B1 (en) 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
EP1180037A2 (en) * 1999-05-26 2002-02-20 The Brigham and Women's Hospital Therapeutic uses of agents that modulate the activity of alpha-smooth muscle actin
US20040167634A1 (en) * 1999-05-26 2004-08-26 Anthony Atala Prosthetic kidney and its use for treating kidney disease
US6710025B1 (en) 1999-05-26 2004-03-23 The Brigham And Women's Hospital, Inc. Treatment of damaged tissue using agents that modulate the activity of alpha-smooth muscle actin
US6521431B1 (en) * 1999-06-22 2003-02-18 Access Pharmaceuticals, Inc. Biodegradable cross-linkers having a polyacid connected to reactive groups for cross-linking polymer filaments
US20040059416A1 (en) * 1999-06-22 2004-03-25 Murray Martha M. Biologic replacement for fibrin clot
US6964685B2 (en) * 1999-06-22 2005-11-15 The Brigham And Women's Hospital, Inc. Biologic replacement for fibrin clot
US7338655B1 (en) 1999-06-30 2008-03-04 Tissuegene, Inc. Gene therapy using TGF-β
US20020095157A1 (en) 1999-07-23 2002-07-18 Bowman Steven M. Graft fixation device combination
US6179840B1 (en) 1999-07-23 2001-01-30 Ethicon, Inc. Graft fixation device and method
US20020116063A1 (en) * 1999-08-02 2002-08-22 Bruno Giannetti Kit for chondrocyte cell transplantation
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US6555374B1 (en) 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US7078232B2 (en) 1999-08-19 2006-07-18 Artecel, Inc. Adipose tissue-derived adult stem or stromal cells for the repair of articular cartilage fractures and uses thereof
AU776260B2 (en) * 1999-08-30 2004-09-02 I.D.M. Immuno-Designed Molecules New humanized biomaterials, a process for their preparation and their applications
US7005274B1 (en) * 1999-09-15 2006-02-28 Migenix Corp. Methods and compositions for diagnosing and treating arthritic disorders and regulating bone mass
IT1309453B1 (en) * 1999-10-01 2002-01-23 Consorzio Per Gli Studi Uni BIOARTIFICIAL SUBSTRATE FOR THE REALIZATION OF FABRICS AND ORGANIANIMALS, IN PARTICULAR HUMAN.
EP1099443A1 (en) * 1999-11-11 2001-05-16 Sulzer Orthopedics Ltd. Transplant/implant device and method for its production
ES2491866T3 (en) * 1999-11-15 2014-09-08 Piramal Healthcare (Canada) Limited Temperature-controlled, pH-dependent, self-gelling aqueous biopolymer solution
US6503273B1 (en) * 1999-11-22 2003-01-07 Cyograft Tissue Engineering, Inc. Tissue engineered blood vessels and methods and apparatus for their manufacture
US20030158302A1 (en) * 1999-12-09 2003-08-21 Cyric Chaput Mineral-polymer hybrid composition
AU1979201A (en) * 1999-12-09 2001-06-18 Bio Syntech Canada Inc Mineral-polymer hybrid composition
KR100394430B1 (en) 1999-12-14 2003-08-09 (주)이노테크 메디칼 Medium for culturing human cell comprising human serum and method for culturing human cell using same
US6623963B1 (en) 1999-12-20 2003-09-23 Verigen Ag Cellular matrix
US6368859B1 (en) 1999-12-29 2002-04-09 Children's Medical Center Corporation Methods and compositions for producing a fascial sling
US6428802B1 (en) 1999-12-29 2002-08-06 Children's Medical Center Corp. Preparing artificial organs by forming polylayers of different cell populations on a substrate
US6376244B1 (en) * 1999-12-29 2002-04-23 Children's Medical Center Corporation Methods and compositions for organ decellularization
US6479064B1 (en) 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
EP1918366A1 (en) 2000-02-26 2008-05-07 Artecel, Inc. Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof
US7078230B2 (en) 2000-02-26 2006-07-18 Artecel, Inc. Adipose tissue-derived stromal cell that expresses characteristics of a neuronal cell
US7582292B2 (en) 2000-02-26 2009-09-01 Artecel, Inc. Adipose tissue derived stromal cells for the treatment of neurological disorders
US7182781B1 (en) 2000-03-02 2007-02-27 Regeneration Technologies, Inc. Cervical tapered dowel
US6723335B1 (en) 2000-04-07 2004-04-20 Jeffrey William Moehlenbruck Methods and compositions for treating intervertebral disc degeneration
JP5684963B2 (en) * 2000-04-14 2015-03-18 ユニバーシティ オブ ピッツバーグ オブ ザ コモンウェルス システム オブ ハイヤー エデュケイション Muscle tissue-derived progenitor cells, their compositions, and soft tissue and bone growth and thickening using treatments
AU2001255767A1 (en) * 2000-04-28 2001-11-12 Curis, Inc. Methods and reagents for tissue engineering of cartilage in vitro
US7585323B2 (en) * 2000-05-05 2009-09-08 Medlden, Llc In vitro mechanical loading of musculoskeletal tissues
DE20019809U1 (en) * 2000-05-31 2001-07-12 Fraunhofer Ges Forschung Cartilage replacement and biomatrix for the cultivation of cells
ATE308610T1 (en) 2000-05-31 2005-11-15 Fraunhofer Ges Forschung THREE-DIMENSIONAL SKIN MODEL
JP5089006B2 (en) * 2000-06-29 2012-12-05 ピラマル ヘルスケア (カナダ) リミテッド Compositions and methods for repair and regeneration of cartilage and other tissues
US6719970B1 (en) 2000-07-10 2004-04-13 Alkermes Controlled Therapeutics, Inc. Method of generating cartilage
US9452238B2 (en) 2000-07-29 2016-09-27 Smith & Nephew LLP Tissue implant
AU2007203472B2 (en) * 2000-07-29 2011-07-14 Smith & Nephew Plc Tissue implant for cartilage repair
CA2457162A1 (en) * 2000-09-01 2002-03-07 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed fibrin-based matrices and tissues
US20040091540A1 (en) * 2000-11-15 2004-05-13 Desrosiers Eric Andre Method for restoring a damaged or degenerated intervertebral disc
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
DE10061704A1 (en) * 2000-12-12 2002-06-20 Hans Joerg Bauer Method and device for producing biological tissue in a growth chamber
US20020127265A1 (en) * 2000-12-21 2002-09-12 Bowman Steven M. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US6852330B2 (en) * 2000-12-21 2005-02-08 Depuy Mitek, Inc. Reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
CA2365376C (en) * 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US7192604B2 (en) 2000-12-22 2007-03-20 Ethicon, Inc. Implantable biodegradable devices for musculoskeletal repair or regeneration
KR101132545B1 (en) 2001-02-14 2012-04-02 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
EP1362095B1 (en) 2001-02-14 2015-05-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
AU2002248484A1 (en) * 2001-02-21 2002-09-12 Drexel University Muscle-polymer constructs for bone tissue engineering
WO2002067867A2 (en) * 2001-02-23 2002-09-06 The University Of Pittsburgh Rapid preparation of stem cell matrices for use in tissue and organ treatment and repair
US20060094112A1 (en) * 2001-03-07 2006-05-04 Omotunde Babalola Biological scaffold
US7252982B2 (en) * 2001-03-15 2007-08-07 Massachusetts Institute Of Technology Tissue engineering enhanced by the transfer of a growth factor gene
EP2522304B1 (en) 2001-03-23 2014-01-08 Histogenics Corporation Composition and methods for the production of biological tissues and tissue constructs
US20040151705A1 (en) 2002-03-22 2004-08-05 Shuichi Mizuno Neo-cartilage constructs and a method for preparation thereof
US7029838B2 (en) 2001-03-30 2006-04-18 Arizona Board Of Regents On Behalf Of The University Of Arizona Prevascularized contructs for implantation to provide blood perfusion
US7030127B2 (en) 2001-06-29 2006-04-18 Ethicon, Inc. Composition and medical devices utilizing bioabsorbable polymeric waxes
US7034037B2 (en) 2001-06-29 2006-04-25 Ethicon, Inc. Compositions and medical devices utilizing bioabsorbable polymeric waxes and rapamycin
US6967234B2 (en) 2002-12-18 2005-11-22 Ethicon, Inc. Alkyd-lactone copolymers for medical applications
KR100494265B1 (en) * 2001-08-14 2005-06-13 메디포스트(주) Composition for treatment of articular cartilage damage
JP2005506860A (en) 2001-09-15 2005-03-10 ラツシユ−プレスビテリアン−セント・リユークズ・メデイカル・センター Layered cartilage tissue and engineering method thereof
US9993659B2 (en) * 2001-11-01 2018-06-12 Pthera, Llc Low level light therapy for enhancement of neurologic function by altering axonal transport rate
US10683494B2 (en) 2001-11-01 2020-06-16 Pthera LLC Enhanced stem cell therapy and stem cell production through the administration of low level light energy
US8308784B2 (en) * 2006-08-24 2012-11-13 Jackson Streeter Low level light therapy for enhancement of neurologic function of a patient affected by Parkinson's disease
US7303578B2 (en) * 2001-11-01 2007-12-04 Photothera, Inc. Device and method for providing phototherapy to the brain
US7534255B1 (en) 2003-01-24 2009-05-19 Photothera, Inc Low level light therapy for enhancement of neurologic function
US20030165473A1 (en) * 2001-11-09 2003-09-04 Rush-Presbyterian-St. Luke's Medical Center Engineered intervertebral disc tissue
US20030170285A1 (en) * 2001-11-13 2003-09-11 Veazey William S. Delivery of tissue engineering media
US20030166274A1 (en) * 2001-11-15 2003-09-04 Hewitt Charles W. Three-dimensional matrix for producing living tissue equivalents
BR0214217A (en) * 2001-11-16 2004-09-21 Childrens Medical Center Increased Organ Function
US7048963B2 (en) * 2001-11-30 2006-05-23 Cambridge Polymers Group, Inc. Layered aligned polymer structures and methods of making same
US20050019488A1 (en) * 2001-11-30 2005-01-27 Cambridge Polymer Group, Inc., Boston, Ma Layered aligned polymer structures and methods of making same
AU2002364558A1 (en) * 2001-12-11 2003-06-23 Cytograft Tissue Engineering, Inc. Tissue engineered cellular sheets, methods of making and use thereof
US20030144712A1 (en) * 2001-12-20 2003-07-31 Jackson Streeter, M.D. Methods for overcoming organ transplant rejection
US10695577B2 (en) * 2001-12-21 2020-06-30 Photothera, Inc. Device and method for providing phototherapy to the heart
US7316922B2 (en) * 2002-01-09 2008-01-08 Photothera Inc. Method for preserving organs for transplant
US20030138873A1 (en) * 2002-01-22 2003-07-24 Koichi Masuda Tissue engineered cartilage for drug discovery
DE10203644A1 (en) * 2002-01-30 2003-08-07 Fraunhofer Ges Forschung Cryopreservation on textile fabrics
EP2292091A1 (en) 2002-02-13 2011-03-09 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
AU2003219830A1 (en) * 2002-02-22 2003-09-09 Ebi, L.P. Methods and compositions for treating bone or cartilage defects
US7005136B2 (en) 2002-03-29 2006-02-28 Ethicon, Inc. Bone replacement materials utilizing bioabsorbable liquid polymers
US7326426B2 (en) 2002-03-29 2008-02-05 Ethicon, Inc. Compositions and medical devices utilizing bioabsorbable liquid polymers
CA2853267C (en) 2002-04-08 2018-06-26 Octane Biotech Inc. Automated tissue engineering system comprising sensors linked to a microprocessor
CN1309428C (en) * 2002-05-13 2007-04-11 东芝陶瓷株式会社 Member for regenerating joint cartilage and process for producing the same, method of regenerating joint cartilage and artificial cartilage for transplantation
JPWO2003094985A1 (en) * 2002-05-14 2005-09-08 北海道ティー・エル・オー株式会社 Artificial extracellular matrix and method for producing the same
DE10222896A1 (en) * 2002-05-23 2003-12-11 Biotissue Technologies Ag Process for the production of a three-dimensional and flat tissue graft
US20040153130A1 (en) * 2002-05-29 2004-08-05 Amir Oron Methods for treating muscular dystrophy
US7299805B2 (en) 2002-06-07 2007-11-27 Marctec, Llc Scaffold and method for implanting cells
US6943021B2 (en) * 2002-06-07 2005-09-13 Mattek Corporation Three dimensional vaginal tissue model containing immune cells
US7026374B2 (en) 2002-06-25 2006-04-11 Aruna Nathan Injectable microdispersions for medical applications
US7622562B2 (en) 2002-06-26 2009-11-24 Zimmer Orthobiologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
US7101566B2 (en) 2002-06-28 2006-09-05 Ethicon, Inc. Polymer coated microparticles for sustained release
ATE481110T1 (en) * 2002-07-16 2010-10-15 Biosyntech Canada Inc COMPOSITION FOR THE PRODUCTION OF CELL-COMPATIBLE, INJECTABLE, SELF-GELELLING CHITOSAN SOLUTIONS FOR ENCAPSULATING AND ADMINISTERING LIVING CELLS OR BIOLOGICALLY ACTIVE FACTORS
JP4353510B2 (en) 2002-09-09 2009-10-28 株式会社カネカ Tissue regeneration support and method for producing the same
US20040132002A1 (en) * 2002-09-17 2004-07-08 Jackson Streeter Methods for preserving blood
US20040136968A1 (en) * 2002-09-27 2004-07-15 Verigen Ag Autologous cells on a support matrix for tissue repair
US20040062753A1 (en) * 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
US7824701B2 (en) * 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
US20040078090A1 (en) 2002-10-18 2004-04-22 Francois Binette Biocompatible scaffolds with tissue fragments
WO2004047770A2 (en) 2002-11-26 2004-06-10 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
AT500418B1 (en) * 2002-12-05 2009-12-15 Winkler Heinz Dr CULTIVATED CELLULAR CELLS CONTAINING THE IMPLANT AND METHOD FOR THE PRODUCTION THEREOF
US20050031598A1 (en) * 2002-12-10 2005-02-10 Shulamit Levenberg Engineering three-dimensional tissue structures using differentiating embryonic stem cells
US6872799B2 (en) 2002-12-18 2005-03-29 Ethicon, Inc. Functionalized polymers for medical applications
US6866860B2 (en) 2002-12-19 2005-03-15 Ethicon, Inc. Cationic alkyd polyesters for medical applications
WO2004061092A1 (en) * 2002-12-27 2004-07-22 Mitsubishi Heavy Industries, Ltd. Method of culturing pluripotent stem cells and culture apparatus therefor
US20050059153A1 (en) * 2003-01-22 2005-03-17 George Frank R. Electromagnetic activation of gene expression and cell growth
ES2405080T3 (en) * 2003-02-26 2013-05-30 Zimmer Orthobiologics, Inc. Prepared for the repair of a cartilaginous tissue, in particular of articular cartilage defects
US7642092B2 (en) * 2003-03-03 2010-01-05 Technion Research & Development Foundation Ltd. Cultured cartilage/bone cells/tissue, method of generating same and uses thereof
US8197837B2 (en) 2003-03-07 2012-06-12 Depuy Mitek, Inc. Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
US7344555B2 (en) 2003-04-07 2008-03-18 The United States Of America As Represented By The Department Of Health And Human Services Light promotes regeneration and functional recovery after spinal cord injury
CN103230415B (en) * 2003-04-25 2016-05-04 匹兹堡大学联邦制高等教育 For promoting and strengthen the neural muscle derived cell (MDC) of repairing and regenerating
JP4566008B2 (en) * 2003-05-23 2010-10-20 ホイゼルマン・ハンス・イェルク Method and apparatus for mechanically activating tissue in vitro
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US7413734B2 (en) * 2003-06-27 2008-08-19 Ethicon, Incorporated Treatment of retinitis pigmentosa with human umbilical cord cells
US7875272B2 (en) * 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8491883B2 (en) * 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8790637B2 (en) * 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US8226715B2 (en) 2003-06-30 2012-07-24 Depuy Mitek, Inc. Scaffold for connective tissue repair
DE602004030254D1 (en) 2003-08-01 2011-01-05 Two Cells Co Ltd SCAFFOLD FREE, SELF ORGANIZED, 3 DIMENSIONAL, SYNTHETIC TISSUE
WO2007083504A1 (en) * 2003-08-01 2007-07-26 Norimasa Nakamura Scaffold-free self-organized 3d synthetic tissue
US10583220B2 (en) * 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
US20050054100A1 (en) * 2003-09-08 2005-03-10 Rennard Stephen I. Methods for fibroblast differentiation
EP1529543A1 (en) * 2003-11-04 2005-05-11 Institut National De La Sante Et De La Recherche Medicale (Inserm) Use of a hydrogel for culturing chondrocytes
US7316822B2 (en) 2003-11-26 2008-01-08 Ethicon, Inc. Conformable tissue repair implant capable of injection delivery
US7901461B2 (en) 2003-12-05 2011-03-08 Ethicon, Inc. Viable tissue repair implants and methods of use
WO2005058207A1 (en) * 2003-12-11 2005-06-30 Isto Technologies, Inc. Particulate cartilage system
AU2004308927A1 (en) * 2003-12-19 2005-07-14 Viacell, Inc. Use of human cord blood-derived pluripotent cells for the treatment of disease
WO2005073365A1 (en) * 2004-01-29 2005-08-11 Japan Tissue Engineering Co., Ltd. Method of treating cells for transplantation, cell suspension, prosthesis for transplantation and method of treating injured site
US11395865B2 (en) * 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
US8352031B2 (en) 2004-03-10 2013-01-08 Impulse Dynamics Nv Protein activity modification
EP1576957A1 (en) * 2004-03-18 2005-09-21 Universiteit Twente Tissue repair using pluripotent cells
US8221780B2 (en) * 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8657881B2 (en) * 2004-04-20 2014-02-25 Depuy Mitek, Llc Meniscal repair scaffold
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
MX2007001937A (en) 2004-08-16 2007-08-07 Cellres Corp Pte Ltd Isolation of stem/progenitor cells from amniotic membrane of umbilical cord.
US20060134074A1 (en) * 2004-08-30 2006-06-22 Naughton Gail K Compositions and methods for promoting hair growth
AU2005279934A1 (en) * 2004-08-30 2006-03-09 Theregen, Inc. Compositions and methods of promoting hair growth
US8697139B2 (en) 2004-09-21 2014-04-15 Frank M. Phillips Method of intervertebral disc treatment using articular chondrocyte cells
US20060111778A1 (en) * 2004-10-29 2006-05-25 Michalow Alexander E Methods of promoting healing of cartilage defects and method of causing stem cells to differentiate by the articular chondrocyte pathway
WO2006062989A1 (en) * 2004-12-07 2006-06-15 Bacterin International, Inc. Three-dimensional cell culsture system
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
WO2006101548A2 (en) * 2004-12-21 2006-09-28 Ethicon, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
PL1831356T3 (en) * 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
AU2005322068B2 (en) 2004-12-23 2011-09-01 Ethicon Incorporated Treatment of stroke and other acute neural degenerative disorders using postpartum derived cells
JP2008546686A (en) * 2005-06-17 2008-12-25 セレゲン,インコーポレーテッド Method for treating ischemic tissue
KR100731940B1 (en) * 2005-06-27 2007-06-25 미츠비시 쥬고교 가부시키가이샤 Method of culturing pluripotent stem cells and culture apparatus therefor
EP1916964A4 (en) 2005-08-26 2015-11-04 Zimmer Inc Implants and methods for repair, replacement and treatment of joint disease
AU2006292224B2 (en) 2005-09-19 2013-08-01 Histogenics Corporation Cell-support matrix and a method for preparation thereof
EP1764117A1 (en) 2005-09-20 2007-03-21 Zimmer GmbH Implant for the repair of a cartilage defect and method for manufacturing the implant
EP2530145A1 (en) 2005-10-13 2012-12-05 Anthrogenesis Corporation Immunomodulation using placental stem cells
CN103555655B (en) * 2005-10-21 2018-02-27 细胞研究私人有限公司 The application of ancestral cells and its cell of differentiation is separated and cultivated from umbilical cord amniotic membrane
US20090149421A1 (en) * 2005-11-04 2009-06-11 Bio Syntech Canada Inc. Gel formation of polyelectrolyte aqueous solutions by thermally induced changes in ionization state
US20100209397A1 (en) * 2005-11-10 2010-08-19 Carticure Ltd. Method for non-autologous cartilage regeneration
WO2007070870A1 (en) * 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
WO2007073552A1 (en) * 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
WO2007076522A2 (en) * 2005-12-28 2007-07-05 Ethicon, Incorporated Treatment of peripheral vascular disease using postpartum-derived cells
EP1976978A2 (en) 2005-12-29 2008-10-08 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
US20090306776A1 (en) 2006-01-25 2009-12-10 Children's Medical Center Corporation Methods and procedures for ligament repair
US10357662B2 (en) * 2009-02-19 2019-07-23 Pthera LLC Apparatus and method for irradiating a surface with light
US7575589B2 (en) * 2006-01-30 2009-08-18 Photothera, Inc. Light-emitting device and method for providing phototherapy to the brain
US20070179570A1 (en) * 2006-01-30 2007-08-02 Luis De Taboada Wearable device and method for providing phototherapy to the brain
US20090254154A1 (en) 2008-03-18 2009-10-08 Luis De Taboada Method and apparatus for irradiating a surface with pulsed light
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
PT2366775E (en) 2006-03-23 2015-07-20 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
ITTO20060282A1 (en) * 2006-04-14 2007-10-15 Univ Degli Studi Torino MEDIUM OF CULTURE AND PHARMACEUTICAL COMPOSITION FOR THE REGENERATION OF THE RELATIVE PAPER FABRIC PROCEDURE RELATED TO USES AND PRODUCTS
WO2008127256A1 (en) * 2006-06-01 2008-10-23 Massachusetts Institute Of Technology Control of cells and cell multipotentiality in three dimensional matrices
KR100834718B1 (en) * 2006-09-20 2008-06-02 세원셀론텍(주) bone recovery cell composition and manufacture method thereof
EP4162936A1 (en) 2006-09-28 2023-04-12 The Children's Medical Center Corporation Methods and collagen products for tissue repair
EP2097513A4 (en) * 2006-11-17 2012-11-28 Cytograft Tissue Engineering Inc Preparation and use of cell-synthesized threads
US8163549B2 (en) 2006-12-20 2012-04-24 Zimmer Orthobiologics, Inc. Method of obtaining viable small tissue particles and use for tissue repair
US20080221211A1 (en) * 2007-02-02 2008-09-11 Jackson Streeter Method of treatment of neurological injury or cancer by administration of dichloroacetate
NZ597779A (en) 2007-02-12 2013-07-26 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
US20090012629A1 (en) 2007-04-12 2009-01-08 Isto Technologies, Inc. Compositions and methods for tissue repair
EP2155860B1 (en) 2007-05-03 2014-08-27 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US9580688B2 (en) * 2007-06-08 2017-02-28 Wake Forest University Health Sciences Kidney structures and methods of forming the same
EP2162529B1 (en) 2007-06-08 2019-03-27 Wake Forest University Health Sciences Selective cell therapy for the treatment of renal failure
US10590391B2 (en) * 2007-06-08 2020-03-17 Wake Forest University Health Sciences Selective cell therapy for the treatment of renal failure
WO2008156659A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US20090054984A1 (en) 2007-08-20 2009-02-26 Histogenics Corporation Method For Use Of A Double-Structured Tissue Implant For Treatment Of Tissue Defects
US8685107B2 (en) 2007-07-03 2014-04-01 Histogenics Corporation Double-structured tissue implant and a method for preparation and use thereof
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
EP2182887B1 (en) 2007-08-20 2016-12-14 Histogenics Corporation A method for improvement of differentiation of mesenchymal stem cells using a double-structured tissue implant
PL2200622T5 (en) 2007-09-19 2016-08-31 Pluristem Ltd Adherent cells from adipose or placenta tissues and use thereof in therapy
EP3524253A1 (en) 2007-09-28 2019-08-14 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
WO2009046377A2 (en) * 2007-10-04 2009-04-09 Medistem Laboratories, Inc. Compositions and methods of stem cell therapy for autism
UA99152C2 (en) * 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US9393273B2 (en) 2008-05-27 2016-07-19 Pluristem Ltd. Methods of treating inflammatory colon diseases
WO2010005753A1 (en) * 2008-06-16 2010-01-14 Cytograft Tissue Engineering, Inc. Arterial implants
CA2729576A1 (en) * 2008-07-02 2010-01-07 Allergan, Inc. Compositions and methods for tissue filling and regeneration
AU2009282619B2 (en) * 2008-08-18 2015-08-20 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Bone augmentation utilizing muscle-derived progenitor compositions in biocompatible matrix, and treatments thereof
KR20180108887A (en) * 2008-08-20 2018-10-04 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
EP3539380A3 (en) 2008-08-20 2019-12-18 Celularity, Inc. Improved cell composition and methods of making the same
CA2734446C (en) 2008-08-22 2017-06-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
SG190659A1 (en) 2008-09-02 2013-06-28 Pluristem Ltd Adherent cells from placenta tissue and use thereof in therapy
US7848035B2 (en) * 2008-09-18 2010-12-07 Photothera, Inc. Single-use lens assembly
US20110294215A1 (en) * 2008-10-22 2011-12-01 Regenemed, Inc. Culture systems
WO2010056378A2 (en) 2008-11-14 2010-05-20 Histogen, Inc. Extracellular matrix compositions for the treatment of cancer
RU2562154C2 (en) 2008-11-19 2015-09-10 Антродженезис Корпорейшн Amniotic adhesive cells
US9192695B2 (en) * 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
CA2744533A1 (en) * 2008-11-24 2010-05-27 Bioe Llc Implantable compositions for repairing osteochondral defects
CA2747794C (en) 2008-12-19 2018-10-30 Advanced Technologies And Regenerative Medicine, Llc Treatment of lung and pulmonary diseases and disorders
ES2520393T3 (en) * 2008-12-19 2014-11-11 DePuy Synthes Products, LLC Cells derived from umbilical cord tissue to treat neuropathic pain and spasticity
JP6095893B2 (en) * 2008-12-19 2017-03-15 デピュイ・シンセス・プロダクツ・インコーポレイテッド Regeneration and repair of nerve tissue after injury
US10179900B2 (en) * 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
US8771677B2 (en) * 2008-12-29 2014-07-08 Vladimir B Serikov Colony-forming unit cell of human chorion and method to obtain and use thereof
US9511093B2 (en) * 2009-03-23 2016-12-06 The Texas A & M University System Compositions of mesenchymal stem cells to regenerate bone
BRPI1013409A2 (en) 2009-03-26 2018-01-16 Advanced Tech And Regenerative Medicine Llc human umbilical cord tissue cells as therapy for alzheimer's disease
CA2767014C (en) 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
NZ600651A (en) 2009-11-30 2014-07-25 Pluristem Ltd Adherent cells from placenta and use of same in disease treatment
ES2646750T3 (en) 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
EP2542249A4 (en) 2010-03-05 2013-08-07 Tissue Genesis Inc Methods and compositions to support tissue integration and inosculation of transplanted tissue and transplanted engineered penile tissue with adipose stromal cells
TWI578993B (en) 2010-04-07 2017-04-21 安瑟吉納西斯公司 Angiogenesis using placental stem cells
TW201138792A (en) 2010-04-08 2011-11-16 Anthrogenesis Corp Treatment of sarcoidosis using placental stem cells
EP2561066B1 (en) 2010-04-23 2016-03-30 Pluristem Ltd. Adherent stromal cells derived from plancentas of multiple donors and uses thereof
KR20130093091A (en) 2010-07-13 2013-08-21 안트로제네시스 코포레이션 Methods of generating natural killer cells
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
NZ612801A (en) 2010-12-27 2014-12-24 Intellicell Biosciences Inc Ultrasonic cavitation derived stromal or mesenchymal vascular extracts and cells derived therefrom obtained from adipose tissue and use thereof
EP2658557A1 (en) 2010-12-31 2013-11-06 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
KR101903339B1 (en) 2011-03-22 2018-10-01 플루리스템 리미티드 Methods for treating radiation or chemical injury
AU2012241521B2 (en) 2011-04-15 2016-07-14 Pluri Biotech Ltd Methods and systems for harvesting cells
TWI602570B (en) 2011-06-01 2017-10-21 安瑟吉納西斯公司 Treatment of pain using placental stem cells
FR2976587B1 (en) * 2011-06-20 2015-04-03 Basf Beauty Care Solutions F IN VITRO DOSING METHOD BY IMMUNOLOGICAL TECHNIQUE
WO2013012616A2 (en) 2011-07-15 2013-01-24 Orbotech Ltd. Electrical inspection of electronic devices using electron-beam induced plasma probes
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
MX362198B (en) 2011-12-23 2019-01-08 Depuy Synthes Products Llc Detection of human umbilical cord tissue-derived cells.
WO2013116744A1 (en) 2012-02-01 2013-08-08 Children's Medical Center Corporation Biomaterial for articular cartilage maintenance and treatment of arthritis
EP2841010B1 (en) 2012-04-24 2023-08-23 Harvard Apparatus Regenerative Technology, Inc. Supports for engineered tissue scaffolds
AU2013299505B2 (en) * 2012-08-10 2016-12-22 Spinalcyte, Llc Generation of cartilage ex vivo from fibroblasts
CA2882687A1 (en) 2012-09-04 2014-03-13 Pluristem Ltd. Methods for prevention and treatment of preeclampsia
US9512393B2 (en) 2012-09-06 2016-12-06 Pluristem Ltd. Devices and methods for culture of cells
US20140178343A1 (en) 2012-12-21 2014-06-26 Jian Q. Yao Supports and methods for promoting integration of cartilage tissue explants
WO2014110300A1 (en) 2013-01-09 2014-07-17 Harvard Apparatus Regenerative Technology Synthetic scaffolds
EP3798226A1 (en) 2013-02-01 2021-03-31 Children's Medical Center Corporation Collagen scaffolds
EP3622960A1 (en) 2013-02-05 2020-03-18 Celularity, Inc. Natural killer cells from placenta
US10351910B2 (en) 2013-02-20 2019-07-16 Pluristem Ltd Gene and protein expression properties of adherent stromal cells cultured in 3D
WO2014141111A1 (en) 2013-03-14 2014-09-18 Pluristem Ltd. Methods for prevention and treatment of graft-versus-host disease
EP2970882B1 (en) 2013-03-15 2018-11-28 AlloSource Cell repopulated collagen matrix for soft tissue repair and regeneration
WO2015004609A2 (en) 2013-07-09 2015-01-15 Pluristem Ltd. Adherent cells from placenta and use thereof in treatment of injured tendons
CN105793716A (en) * 2013-10-03 2016-07-20 奥宝科技有限公司 Application of electron-beam induced plasma probes to inspection, test, debug and surface modifications
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
WO2016019168A1 (en) * 2014-08-01 2016-02-04 Isto Technologies, Inc. Cell expansion methods and systems
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
US10077420B2 (en) 2014-12-02 2018-09-18 Histogenics Corporation Cell and tissue culture container
US10258652B2 (en) 2014-12-18 2019-04-16 Pluristem Ltd. Methods and compositions for treating and preventing muscle wasting disorders
CN107864627A (en) 2015-03-23 2018-03-30 普拉里斯坦有限公司 Include the method and composition of adhering substrate cell
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
JP6884792B2 (en) 2016-02-18 2021-06-09 プルリステム リミテッド Treatment methods and therapeutic compositions for cancer and neoplasms
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
IL270114B2 (en) 2017-04-24 2024-01-01 Pluri Biotech Ltd Methods and compositions for treating neurological disorders
CA3067558A1 (en) 2017-06-16 2018-12-20 Avery Therapeutics, Inc. Three dimensional tissue compositions and methods of use
IL260253A (en) 2017-07-03 2019-01-31 Pluristem Ltd Methods and compositions for detaching adherent cells
IT201700087978A1 (en) 2017-07-31 2019-01-31 Univ Degli Studi Genova Three-dimensional hydrogel scaffold for cell cultures and its production method
CN111373030A (en) 2017-09-01 2020-07-03 隆萨沃克斯维尔股份有限公司 End-to-end cell therapy automation
US11518971B2 (en) 2018-11-27 2022-12-06 Research Triangle Institute Method and apparatus for spatial control of cellular growth
CN113366099A (en) 2018-12-21 2021-09-07 奥克泰生物科技股份有限公司 Carousel for modular bio-production units
JP2022514761A (en) 2018-12-21 2022-02-15 ロンザ ウォーカーズヴィル,インコーポレーテッド Automatic production method of viral vector
SG11202106652WA (en) 2018-12-28 2021-07-29 Octane Biotech Inc Cell culture and tissue engineering systems with controlled environmental zones
KR20210125510A (en) 2019-02-08 2021-10-18 론자 워커스빌 아이엔씨. Cell Concentration Methods and Apparatus for Use in Automated Bioreactors
AU2021289205A1 (en) 2020-06-11 2023-02-02 Mesoblast International Sarl 3D culture of mesenchymal lineage precursor or stem cells

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4485096A (en) * 1982-02-26 1984-11-27 Massachusetts Institute Of Technology Tissue-equivalent and method for preparation thereof
IL68218A (en) * 1983-03-23 1985-12-31 Univ Ramot Compositions for cartilage repair comprising embryonal chondrocytes
US4609551A (en) * 1984-03-20 1986-09-02 Arnold Caplan Process of and material for stimulating growth of cartilage and bony tissue at anatomical sites
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US4721096A (en) * 1986-04-18 1988-01-26 Marrow-Tech Incorporated Process for replicating bone marrow in vitro and using the same
US5032508A (en) * 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US4963489A (en) * 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
HU202578B (en) * 1986-04-18 1991-03-28 Marrow Tech Inc Process for repricating bone marrow celles in three-dimension celle-cultivating systhem, three-dimension vehicle for the systhem and process for testing citotoxicity of the systhem
US5041138A (en) * 1986-11-20 1991-08-20 Massachusetts Institute Of Technology Neomorphogenesis of cartilage in vivo from cell culture
EP0282746A1 (en) * 1987-02-19 1988-09-21 Takeda Chemical Industries, Ltd. Method for producing artificial cultured tissue
DE01201546T1 (en) * 1988-04-08 2004-07-08 Stryker Corp., Kalamazoo Osteogenic facilities
US4837379A (en) * 1988-06-02 1989-06-06 Organogenesis Inc. Fibrin-collagen tissue equivalents and methods for preparation thereof
EP0381490B1 (en) * 1989-02-02 1994-09-21 Joel S. Greenberger Gene therapy using stromal cells
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5326357A (en) * 1992-03-18 1994-07-05 Mount Sinai Hospital Corporation Reconstituted cartridge tissue
WO1995030742A1 (en) * 1994-05-05 1995-11-16 Genzyme Corporation Methods and compositions for the repair of articular cartilage defects in mammals
ES2266715T3 (en) * 1995-03-28 2007-03-01 Thomas Jefferson University IMPLANTS THAT INCLUDE IMMUNOLOGICALLY ISOLATED THERMAL CELLS AND ITS USE.

Also Published As

Publication number Publication date
IL114017A0 (en) 1995-11-27
NZ288467A (en) 1998-10-28
US5902741A (en) 1999-05-11
US5962325A (en) 1999-10-05
JP3599341B2 (en) 2004-12-08
AU689605B2 (en) 1998-04-02
EP0812351A1 (en) 1997-12-17
AU2769695A (en) 1996-01-04
JP2002502226A (en) 2002-01-22
EP0812351A4 (en) 1998-07-08
WO1995033821A1 (en) 1995-12-14

Similar Documents

Publication Publication Date Title
CA2192064A1 (en) Three-dimensional cartilage cultures
US5919702A (en) Production of cartilage tissue using cells isolated from Wharton's jelly
JP3709487B2 (en) Compositions and methods for naturally secreted extracellular matrix
US6022743A (en) Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
EP1242129B1 (en) Biological joint construct
RU2523339C2 (en) Extracellular matrix compositions for treating cancer
EP0955959A1 (en) Method for making and/or repairing cartilage
US9434923B2 (en) Preparation of parental cell bank from foetal tissue
Nicolin et al. In vitro exposure of human chondrocytes to pulsed electromagnetic fields
AU729774B2 (en) Stromal cell-based three-dimensional culture system for forming tubes, tendons, ligaments and corrective structures
Testa et al. Haemopoietic growth factors: Their relevance in osteoclast formation and function

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued
FZDE Discontinued

Effective date: 20030606