US20040203146A1 - Composite scaffolds and methods using same for generating complex tissue grafts - Google Patents

Composite scaffolds and methods using same for generating complex tissue grafts Download PDF

Info

Publication number
US20040203146A1
US20040203146A1 US10/476,064 US47606404A US2004203146A1 US 20040203146 A1 US20040203146 A1 US 20040203146A1 US 47606404 A US47606404 A US 47606404A US 2004203146 A1 US2004203146 A1 US 2004203146A1
Authority
US
United States
Prior art keywords
scaffold
tissue
polymer
cell
tissue type
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
US10/476,064
Other languages
English (en)
Inventor
Dan Gazit
Avraham Domb
Gudi Turgeman
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.)
Yissum Research Development Co of Hebrew University of Jerusalem
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
Priority to US10/476,064 priority Critical patent/US20040203146A1/en
Assigned to YISSUM RESEARCH DEVELOPMENT COMPANY reassignment YISSUM RESEARCH DEVELOPMENT COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PELED, GADI, TURGEMAN, GADI, DOMB, AVRAHAM, GAZIT, DAN, AZZAM, TONY
Publication of US20040203146A1 publication Critical patent/US20040203146A1/en
Priority to US12/285,107 priority patent/US8197553B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61L27/3604Materials 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
    • A61L27/14Macromolecular materials
    • A61L27/18Macromolecular materials obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds
    • 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
    • A61L27/14Macromolecular materials
    • A61L27/20Polysaccharides
    • 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
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • 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
    • A61L27/3625Vascular tissue, e.g. heart valves
    • 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
    • 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
    • A61L27/3886Materials 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
    • 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
    • A61L27/3891Materials 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 as distinct cell layers
    • 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
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • 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
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L27/44Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • A61L27/48Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix with macromolecular fillers
    • 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
    • A61L27/507Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials for artificial blood vessels
    • 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
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • 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
    • A61L27/58Materials at least partially resorbable by the body
    • 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
    • C12N5/0068General culture methods using substrates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • 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
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/412Tissue-regenerating or healing or proliferative agents
    • A61L2300/414Growth factors
    • 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
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/426Immunomodulating agents, i.e. cytokines, interleukins, interferons
    • 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
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/602Type of release, e.g. controlled, sustained, slow
    • A61L2300/604Biodegradation
    • 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
    • A61L2430/36Materials or treatment for tissue regeneration for embolization or occlusion, e.g. vaso-occlusive compositions or devices
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M25/00Means for supporting, enclosing or fixing the microorganisms, e.g. immunocoatings
    • C12M25/14Scaffolds; Matrices
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/28Vascular endothelial 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • C12N2533/72Chitin, chitosan

Definitions

  • the present invention relates to composite scaffolds capable of supporting growth of complex tissue and to methods of manufacturing and using same.
  • the basic concept of tissue engineering employs a scaffold (matrix) which provides a support upon which seeded cells can organize and develop into desired tissue prior to implantation.
  • the scaffold provides an initial biomechanical profile for the replacement tissue until the cells can produce an adequate extracellular matrix.
  • the scaffold is either degraded or metabolized, eventually leaving engrafted tissue in its place.
  • scaffolds are manufactured from biocompatible materials such that implantation thereof does not result in an adverse immune response or induced toxicity.
  • scaffolds are typically manufactured with predetermined porosity so as to facilitate loading thereof of drugs or nutrients useful in promoting the growth of implanted cells.
  • Fabrication of an appropriate scaffold is determined by the type of tissue to be generated.
  • Various scaffold manufacturing procedures rely on fabrication and casting of polymeric foams.
  • Most of the polymeric foams used for tissue engineering applications are made from polylactides (PLA), polyglycolides) (PGA), or a combination of the two (PLGA).
  • Fiber bonding is a technique commonly for preparing structural interconnecting fiber networks for organ implants. Utilizing this process non-woven fibers are bonded together by immersing a non-bonded fiber structure of polymer A, such as PGA with a solution of polymer B (e.g., poly-L-lactic acid) (PLLA) using a solvent which does not dissolve polymer A. The solvent is then allowed to evaporate. The composite consisting of polymer A fibers embedded in a matrix of polymer B is heated above the melting temperature of polymer A to bond the fibers at their cross-points, and then polymer B is selectively dissolved (Mikos, et al. 1993 J. Biomed. Matl. Res. 27:183-189). The resultant bonded fiber structure of Polymer A has substantial rigidity, but the number of pores and their distribution is limited by that of the fiber mesh used in the fabrication.
  • polymer A such as PGA
  • polymer B e.g., poly-L-lactic acid
  • Solvent-casting and particulate-leading In this technique, sieved salt particles, such as sodium chloride crystals, are spread in a PLLA/chloroform solution which is then used to cast a membrane. After evaporating the solvent, the PLLA/salt composite membranes are heated above the PLLA melting temperature and then quenched or annealed by cooling at controlled rates to yield amorphous or semi-crystalline forms with regulated crystallinity. The salt particles are eventually leached out by selective dissolution to produce a porous polymer matrix (Mikos, et al. 1992 Biodegradable Materials Research Society Symposium Proceedings, 252:352-358).
  • Melt molding uses a TeflonTM mold, a mixture of fine PLGA powder and gelatin microspheres is heated above the glass-transition temperature of the polymer. The PLGA-gelatin composite is then removed from the mold and gelatin microspheres are leached out by selective dissolution in distilled de-ionized water.
  • tissue engineered using such scaffolds typically lack the full functional capabilities of natural tissues. This is due to the fact that such scaffolds are either incapable of generating complex tissues having a fully functional architecture (e.g., vascularized), or are incapable of supporting growth of complex tissues altogether.
  • scaffolds which can be used to generate complex tissue grafts, such as, for example, vascularized tissue grafts, either ex-vivo or in-vivo while being devoid of the limitations inherent to prior art scaffolds.
  • a composite scaffold for engineering a heterogeneous tissue comprising: (a) a first scaffold being capable of supporting formation of a first tissue type thereupon; and (b) a second scaffold being capable of supporting formation of a second tissue type thereupon; wherein the first scaffold and the second scaffold are arranged with respect to each other such that when the first scaffold supports the first tissue type and the second scaffold supports the second tissue type, a distance between any cell of the second tissue type and the first tissue type does not exceed 200 ⁇ m.
  • an engineered tissue graft generated using the composite scaffold described above.
  • a method of inducing the formation of a heterogeneous tissue comprising: (a) providing a first scaffold being capable of supporting formation of a first tissue type thereupon; (b) providing a second scaffold being capable of supporting formation of a second tissue type thereupon; (c) embedding the first scaffold in the second scaffold thereby forming a composite scaffold; and (d) implanting the composite scaffold in an individual.
  • the step of embedding is effected such that when the first scaffold supports the first tissue type and the second scaffold supports the second tissue type, a distance between any cell of the second tissue type and the first tissue type does not exceed 200 ⁇ m.
  • the first scaffold is a filamentous scaffold having filaments of a diameter selected from the range of 4-500 ⁇ m.
  • the second scaffold is a porous continuous scaffold.
  • the filamentous scaffold is selected so as to enable the first tissue type to form substantially tubular structures thereupon.
  • the first tissue type is vascular tissue.
  • the second tissue type is structural tissue selected from the group consisting of bone tissue, cartilage tissue, adipose tissue, connective tissue and muscle tissue.
  • first scaffold and/or the second scaffold further include a bioactive agent associated therewith.
  • the bioactive agent is selected from the group consisting of a cell proliferation factor, a cell differentiation factor, a cell attracting factor and a pharmacologically active factor.
  • the method further comprising growing the second tissue type on the second scaffold prior to step (c) or step (d).
  • the first scaffold is selected so as to enable colonization and/or proliferation of at least one cell type composing the first tissue type.
  • the second scaffold is selected so as to enable colonization and/or proliferation of at least one cell type composing the second tissue type.
  • first scaffold and/or second scaffold are degradable upon exposure to predetermined environmental conditions.
  • the predetermined environmental conditions are selected from the group consisting of presence of hydrolytic enzymes, presence of proteasomal enzymes, pH lower than 5 and reducing conditions.
  • composition of matter comprising: (a) a linker molecule attached to a first polymer backbone; (b) a stereoisomer of the linker molecule attached to a second polymer backbone; wherein when exposed to polymerizing conditions the first and the second polymer backbones cross-link with at least one molecule of the first and/or the second polymer backbones via at least one of the linker molecule and the stereoisomer of the linker molecule to thereby form a scaffold structure.
  • the first polymer backbone is identical to the second polymer backbone.
  • composition of matter comprising a polymer backbone attached to: (a) a linker molecule; and (b) a stereoisomer of the linker molecule; wherein when exposed to polymerizing conditions the polymer backbone cross links with at least an additional polymer backbone via at least one of the linker molecule and the stereoisomer of the linker molecule, to thereby form a scaffold structure.
  • the linker molecule is a co-polymer of lactic acid.
  • the scaffold structure is three-dimensional.
  • a scaffold comprising a plurality of molecules of a polymeric backbone cross-linked therebetween via L and D stereoisomers of a linker molecule.
  • the polymer backbone is a hydrophilic polymer.
  • the hydrophilic polymer is selected from the group consisting of a natural polysaccharide, a protein, an ethylene glycol based polymer and a propylene glycol based polymer.
  • the plurality of molecules of the polymeric backbone are hydrophilic polymers.
  • the hydrophilic polymers are selected from the group consisting of natural polysaccharides, proteins, ethylene glycol based polymers and a propylene glycol based polymers.
  • a scaffold capable of releasing a bioactive agent, the scaffold comprising a polymeric backbone and the bioactive agent, wherein the polymeric backbone is selected such that exposure thereof to predetermined environmental conditions leads to release of the bioactive agent from the scaffold.
  • the bioactive agent is selected from the group consisting of a cell proliferating factor, a cell differentiating factor, a cell attracting factor and a pharmacologically active factor.
  • the polymeric backbone is selected from the group consisting of cellulose, hydroxy alkyl acid polyester, polyphosphazene, polycarbonate, lactide acid and glycolide acid.
  • the bioactive agent is incorporated within the polymeric backbone, and whereas the bioactive agent is released following degradation and/or disintegration of the polymeric backbone in the environmental conditions.
  • the bioactive agent is a negatively charged bioactive agent, and whereas the negatively charged bioactive agent is incorporated within pre-cationized regions of the polymeric backbone.
  • the polymeric backbone is designed and constructed so as to enable timed release of the bioactive agent from the scaffold.
  • the predetermined environmental conditions are selected from the group consisting of presence of hydrolytic enzymes, presence of proteasomal enzymes, presence of pH lower than 5 and presence of reducing conditions
  • a scaffold comprising a filamentous polymer including: (a) a hydrophilic molecule being capable of promoting degradation of the filamentous polymer when exposed to predetermined environmental conditions; (b) a plasticizing agent being capable of rendering the filamentous polymer flexible; and (c) a co-polymeric stereocomplex being capable of cross linking the filamentous polymer with at least one additional filamentous polymer to thereby form the scaffold.
  • the filamentous polymer has a diameter selected from a range of 4-500 ⁇ m.
  • the filamentous polymer is designed and configured for supporting formation of a tube shaped tissue structure thereupon
  • the filamentous polymer is selected from the group consisting of hydroxy alkyl acid polyester, polyphosphazene, poly carbonate and poly phosphate ester.
  • the filamentous polymer is degradable upon exposure to predetermined environmental conditions.
  • the predetermined environmental conditions are selected from the group consisting of presence of hydrolytic enzymes, presence of proteasomal enmines, pH lower than 5 and reducing conditions.
  • the tube shaped tissue is vascular tissue.
  • the hydrophilic molecule is poly ethylene glycol and poly ethylene propylene glycol.
  • the plasticizing agent is selected from the group consisting of a tributyl citrate, a tributyl citrate acetate, a phospholipids and an oleate ester.
  • the co-polymeric stereocomplex includes lactide acid stereoisomers.
  • the scaffold further comprising a bioactive agent associated therewith.
  • the bioactive agent is selected from the group consisting of a cell proliferation factor, a cell differentiation factor, a cell attracting factor and a pharmacologically active factor.
  • the present invention successfully addresses the shortcomings of the presently known configurations by providing novel scaffold configurations which can be used, ex-vivo and/or in-vivo, to generate complex tissue, such as, for example, vascularized tissues and organs like liver, pancreas, kidney and the like.
  • FIG. 1 depicts formation of a cross-linked polymeric cellular scaffold generated by stereocomplexation of dextran with poly-D and poly-L lactic acids.
  • FIG. 2 depicts various combinations of in vitro and in vivo modalities for tissue replacement therapy using cellular scaffolds composed of a filamentous polymer for tubular growth of vascular tissues integrally embedded within a continuous sponge matrix polymer for three-dimensional growth of replacement tissues.
  • FIGS. 3 a - c depicts the genetic modification of a murinemesenchymal stem cell line transfected in vitro with a liposome-complexed plasmid encoding a ⁇ -galactosidase marker gene.
  • FIG. 3 a is a schematic depicting liposome complexation of a plasmid encoding ⁇ -galactosidase and transfection of a target cell.
  • 3 b and 3 c are low- and high-power magnification photomicrographs, respectively, demonstrating ⁇ -galactosidase production by a murine mesenchymal stem cell line transfected in vitro with a liposome-complexed plasmid encoding a ⁇ -galactosidase marker gene.
  • FIGS. 4 a - c depicts the genetic modification of a murine mesenchymal stem cell line transfected in vitro with a dextran polycation-complexed plasmid encoding a ⁇ -galactosidase marker gene.
  • FIG. 4 a is a schematic depicting dextran polycation complexation of a plasmid encoding ⁇ -galactosidase and transfection of a target cell.
  • 4 b and 4 c are low- and high-power magnification photomicrographs, respectively, demonstrating ⁇ -galactosidase production by a murine mesenchymal stem cell line transfected in vitro with a dextran polycation-complexed plasmid encoding a ⁇ -galactosidase marker gene.
  • FIG. 5 is a series of photomicrographs depicting the in vivo genetic transformation of mesenchymal stem cells colonizing arabinogalactan-chitosan cellular scaffolds (AG) loaded with liposome-complexed and dextran-complexed plasmid pNGVL1-ntbGAL encoding ⁇ -galactosidase.
  • AG arabinogalactan-chitosan cellular scaffolds
  • FIG. 6 is a series of photomicrographs depicting chondrogenesis and osteogenesis induced by in vivo implantation of arabinogalactan-chitosan cellular scaffolds loaded with naked and liposome-complexed plasmid encoding human BMP-2.
  • FIG. 7 is a series of photomicrographs depicting in vitro growth of either osseous or combined osseous and vascular tissues in arabinogalactan-chitosan cellular scaffolds seeded with MSCs genetically modified to express either BMP-2 alone or in combination with endothelial cells, respectively.
  • FIGS. 8 a - o depict ectopic growth and differentiation of cartilage (C), bone (B) and bone marrow (BM) tissues derived from MSCs genetically modified to express BMP-2 seeded in collagen matrices and implanted in abdominal muscle and depicts doxycycline-mediated inhibition of BMP-2 mRNA transcription and BMP-2-induced osteogenesis by MSCs expressing BMP-2 under the regulatory control of a tetracycline/doxycycline-inhibited promoter.
  • FIGS. 8 a and 8 f are macrophotographs depicting growth and differentiation of tissue in the absence ( ⁇ Dox) and presence (+Dox) of doxycycline, respectively.
  • FIGS. 8 b and 8 g are X-ray photographs depicting growth of calcified tissues in the absence of doxycycline compared with non in the presence of doxycycline, respectively.
  • FIGS. 8 c and 8 h are photomicrographs depicting growth and differentiation of osteogenic tissues composed of cartilage (C), bone (B) and bone marrow (BM) in the muscle tissue M) in the absence of doxycycline compared with non in the presence of doxycycline, respectively.
  • FIGS. 8 j and 8 k are photomicrographs depicting growth and differentiation of engrafted engineered cells lining trabecular tissue displaying chondrocytic and osteoblastic morphologies, respectively, as determined by X-gal staining, on Day 20.
  • FIG. 8 l and 8 m are photomicrographs depicting expression of BMP-2 and ⁇ -galactosidase, respectively, in chondrocytes derived from cells genetically modified to express both genes, as determined by immunohistochemical analyses.
  • FIG. 8 o depicts the presence and absence of BMP-2 mRNA transcription in the absence and presence of doxycycline, respectively, in engrafted cells (C9) on Day 10, as determined by RT-PCR analysis.
  • FIG. 9 depicts the orthotopic regeneration of bone tissue by implantation of collagen matrices seeded with MSCs genetically modified to express BMP-2 ⁇ C9).
  • FIGS. 9 a and 9 b are CT micro-CT imaging data depicting bone regeneration in the absence ( ⁇ Dox) or presence (+Dox) of doxycycline, respectively.
  • FIG. 9 c is a series of morphometric analysis diagrams depicting regenerating bone volume, surface area and surface area to volume ratio in the absence ( ⁇ Dox) or presence (+Dox) of doxycycline.
  • FIG. 10 depicts in vivo vascularization induced in collagen matrix implants seeded with MSCs genetically modified to express BMP-2 under the regulatory control of a tetracyclineldoxycycline-inhibited promoter, angiogenesis induced by 10 ⁇ g BMP-2 in chick CAM assays and angiogenesis induced in vivo in bone tissue formed by culturing cellular scaffolds seeded with MSCs genetically modified to express BMP-2.
  • FIGS. 10 a and 10 c are photomicrographs depicting blood vessel formation in the absence and presence of doxycycline, respectively, in collagen matrix implants seeded with MSCs genetically modified to express BMP-2.
  • FIGS. 10 a and 10 c are photomicrographs depicting blood vessel formation in the absence and presence of doxycycline, respectively, in collagen matrix implants seeded with MSCs genetically modified to express BMP-2.
  • FIGS. 10 b and 10 d are photomicrographs depicting blood vessel formation, in the absence and presence of doxycycline, respectively, in collagen matrix implants seeded with MSCs genetically modified to express BMP-2, as determined histologically.
  • FIG. 10 e is a diagram depicting blood vessel formation, in the absence or presence of doxycycline in collagen matrix implants seeded with MSCs genetically modified to express BMP-2 (C9), as determined by morphometric analysis of blood vessel surface area.
  • FIG. 10 f is a photomicrograph depicting blood vessel formation in collagen matrix implants seeded with MSCs genetically modified to express BMP-2, as determined by immunohistochemical staining of the endothelial marker PECAM.
  • FIGS. 10 g and 10 h are photomicrographs depicting angiogenesis induced by 10 ⁇ g of human BMP-2 protein and vehicle as control in chick CAM assays, respectively.
  • FIGS. 11 a - b depicts angiogenesis induced in vivo in bone tissue formed by culturing cellular scaffolds seeded with MSCs genetically modified to express BMP-2, as determined by microscopic and MRI analyses.
  • FIG. 11 a is a low photomicrograph of the implant (arrows indicate blood vessels).
  • FIG. 11 b represents MRI data of the area including the implant (sBV: small blood vessels, mBv: large blood vessel, L: hind limb, V: vertebral column).
  • FIG. 12 is a photomicrograph depicting formation of vascular tissue upon filamentous cellular scaffolds cultured in vitro with endothelial cells.
  • FIGS. 13 a - b depicts vascularization of in vivo implants of filamentous cellular scaffolds cultured in vitro with endothelial cells.
  • FIG. 13 a depicts low and high-power photomicrographs of seeded implants two weeks after incubation in rotating bioreactor (endothelial cells depicted by arrows).
  • FIG. 13 b depicts low power photomicrographs of a seeded/non-implanted scaffold (“Ex Vivo”) and seeded and non-seeded implants two weeks after implantation.
  • P polymer.
  • FIGS. 14 a - g illustrate staining and RT-PCR analysis of genetically engineered AMSCs (C9 cells) conditionally expressing rhBMP2 under tet regulation.
  • the cells were grown under static culture conditions till confluency and then seeded on arabinogalactan-chitosan scaffolds in 96 well-plate (2 ⁇ 106 cells/scaffold; each scaffold: 5 mm in diameter and 2-3 mm in width) for 24 hours.
  • the scaffolds were placed in a rotary biorecator (4 scaffolds per 50 ml vessel). The vessels were rotated at 15 to 18 RPM.
  • FIG. 14 a empty scaffold
  • FIG. 14 b C9 cells on scaffold
  • FIG. 14 c collagen II detection via RT-PCR
  • FIG. 14 d H&E staining
  • FIG. 14 e anti Osteocalcin staining
  • FIG. 14 f anti R galactosidase staining
  • FIG. 14 g anti collagen X staining.
  • FIGS. 15 a - b illustrate a scanning electron micrograph (FIG. 15 a ) and a computerized regeneration (FIG. 15 b ) of genetically engineered AMSCs (C9 cells) which were grown as described in FIG. 14 for 30 days.
  • FIG. 2 a clearly illustrates cells attached to the polymeric structure. The cells were stained with a fluorescent stain (propidium iodide) and were seen in different optic sections within the scaffold.
  • the image presented in FIG. 15 b is a computerized reconstruction of all optic sections (the different colors represent different distance from the surface of the scaffold).
  • FIGS. 16 a - c illustrate tissue sections taken from cell seeded scaffolds implanted in immune deficient mice (CD-1 nude mice) for six weeks. The tissue sections were stained with Hematoxiline & Eosin (FIG. 16 a and c ), and Masson's Trichrome (FIG. 16 b ). The staining clearly demonstrates the formation of bone trabecules within the implant.
  • FIGS. 17 a - f illustrate scaffolds seeded with genetically engineered AMSCs (C9 cells) conditionally expressing rhBMP2 under tet regulation.
  • the cells were grown in static culture conditions till confluency.
  • the cells were then mixed with arabinogalactan-chitosan beads and cultured in rotary bioreactor for one week.
  • 2 ⁇ 106 B-END-2 cells endothelial cell-line
  • the arabinogalactan beads and the PLA filaments were then co-cultured in a single vessel of the rotary bioreactor and were allowed to form a hybrid structure (FIG.
  • the present invention is of novel polymeric compositions and methods of using same for engineering complex tissue. Specifically, the present invention can be used to generate complex tissue grafts composed of at least two tissue types co-arranged in a functional architecture.
  • the bioartificial implant is one proposed solution.
  • presently fabricated bioartificial implants has proven to be unsatisfactory for a variety of reasons, including, poor biocompatibility, engraftment failure or tissue dysfunction.
  • One of the major problems of bioartificial implants is the need for a well branched vascular network, which can providing the engineered tissue with continuous supply of oxygen and nutrients at the transplantation site.
  • the present invention provides a novel approach for engineering complex tissues, such as vascularized structural tissues, thus substantially enhancing the acceptance rate of engrafted engineered tissue.
  • the complex tissues of the present invention are generated using novel composite scaffolds and in-vivo and/or ex-vivo tissue engineering approaches.
  • a composite scaffold for engineering a heterogeneous tissue there is provided a composite scaffold for engineering a heterogeneous tissue.
  • the term “scaffold” refers to an engineered platform which serves as a physical substrate for cell colonization and/or proliferation.
  • a “composite scaffold” refers to a substrate which is engineered in order to support colonization and/or proliferation of two or more tissue types which together comprise a “heterogeneous tissue”.
  • the composite scaffold of the present invention includes a first scaffold for supporting formation of a first tissue type thereupon and a second scaffold for supporting formation of a second tissue type thereupon.
  • the first scaffold and the second scaffold are arranged with respect to each other such that when the first scaffold supports the first tissue type and the second scaffold supports the second tissue type, a distance between any cell of the second tissue type and the first tissue type does not exceed 200 ⁇ m. Such an arrangement ensures that a functional architecture is maintained within the heterogeneous tissue formed upon the scaffold.
  • such scaffold arrangement ensures cell viability by providing diffusion of nutrients and gases such as oxygen to every cell in the tissue graft formed, as well as diffusion of cellular waste out of the graft so as to minimize cellular toxicity and concomitant death due to localization of the waste within graft tissues.
  • the design of the composite scaffold of the present invention is dictated by cellular organization of natural tissue which is to mimicked by the scaffold-engineered tissue.
  • organ tissue is arranged into functional units composed of cells, each no more than 225 ⁇ m away from a source of nutrients (see, PCT/US98/00594).
  • Such organization ensures efficient gas and nutrient exchange and thus cell viability and functionality.
  • such a composite scaffold will have at least two scaffold components arranged such that tissue formed thereupon would mimic the cellular arrangement of an organ functional unit.
  • the composite scaffold of the present invention is designed such that tissue engineered thereupon would exhibit a spatial distribution of cellular elements which closely approximates that found in the counterpart tissue in vivo. This would enable rapid engraftment of the engineered tissue while ensuring a high rate of survival of graft tissue during the engraftment process.
  • the composite scaffold of the present invention is generated from at least two different scaffold structures.
  • Each scaffold structure is composed of scaffold material and microstructure suitable for organizing and stimulating the growth of a specific cell type.
  • the composite scaffold of the present invention includes a filamentous scaffold for supporting colonization/proliferation of vascular tissue forming cells and a continuous scaffold for supporting colonization/proliferation of cell types which form the structural tissue.
  • the composite scaffold of the present invention is designed so as to allow complete co-integration of the two different scaffolds, enabling the formation of a heterogeneous tissue which maintains a functional architecture.
  • the continuous scaffold is preferably arranged around the filamentous scaffold such that the structural tissue encapsulates the vascular tissue.
  • the various scaffold components of the composite scaffold of the present invention are preferably generated from biocompatible material especially in cases where the scaffold is utilized for in-vivo generation of heterogeneous tissue as is described in detail hereinbelow.
  • biocompatible refers to a substance which does not induce an immune response or fibrosis.
  • examples of biocompatible materials include, but are not limited to, polysaccharides, alginates, polyalcohols, organic acids, agarose, agarose/poly(styrene sulfonic acid), hydroxyethyl methacrylate-methyl methacrylate copolymer, polyvinyl alcohol and protamine-heparin.
  • the various scaffold components of the composite scaffold of the present invention are also preferably generated from biodegradable material especially in cases where it is important to get rid of scaffold material following tissue formation.
  • biodegradable refers to material which is chemically degraded by the action of hydrolytic enzymes, proteolytic enzymes, extreme pH conditions, and the like.
  • biodegradable materials include polymer compositions such as polyhydroxy acids, modified polysaccharides and combinations thereof.
  • a composite scaffold for supporting vascularized structural tissue preferably includes two scaffold structures, a filamentous scaffold for the formation of vascular tissue and a continuous scaffold for the formation of the structural tissue. Following is a detailed description of these two scaffold structures.
  • the filamentous scaffold of the present invention is a solid scaffold capable of supporting cell growth thereupon.
  • Such a scaffold can mimic a blood vessel lumen and form a blood vessel having even small: capillary diameter of 4-50 microns.
  • the filamentous scaffold must be strong and flexible enough to allow formation of flexible thin filaments having a diameter ranging between 4-500 microns.
  • biodegradable polymers meet these criteria, including, for example, thin cellulose fibers.
  • Cellulose fibbers can be modified by oxidation with, for example, periodate in aqueous medium, rendering the fibers more susceptible to hydrolytic degradation (biodegradation). The degree of oxidation determines the strength of the fiber and its degradation profile.
  • These oxidized fibers can be further modified by impregnation with a biodegradable polymer such as poly (lactide-glycolide) so as to be more susceptible to biological degradation.
  • fiber aldehyde groups can be reacted with amino containing hydrophilic or hydrophobic safe molecules including amino acids.
  • polymers and copolymers based on hydroxy alkyl acid polyesters, polyphosphazene, poly (carbonates) and poly (phosphate esters), can also be used.
  • Polymers based on lactide and glycolide acids are better suited for use with the filamentous scaffold of the present invention since it has been previously shown that such materials are capable of supporting cell growth and can be safely transplanted in humans (Shand and Heggie 2000).
  • polymers can also be modified to meet the requirements described above.
  • block and random copolymers of lactide acid and glycolic acid having a molecular weight greater than 10,000 can be spun into thin filaments.
  • copolymers including 30 to 70% lactic acid may be used to delay degradation to a few weeks post transplantation.
  • Increased flexibility of the filaments can be obtained by adding plasticizing agents such as, for example, tributyl citrate, tributyl citrate acetate, phospholipids, oleate esters and the like to the polymer blend or by incorporating agents, such as, for example, ricinoleic acid into the polymer chain.
  • plasticizing agents such as, for example, tributyl citrate, tributyl citrate acetate, phospholipids, oleate esters and the like to the polymer blend or by incorporating agents, such as, for example, ricinoleic acid into the polymer chain.
  • the mechanical properties of the filamentous scaffold must be maximized when supporting formation of a blood vessel such as an artery, which has to exhibit resistance to high blood pressure. This can be achieved by various cross-linking methods, interlinking the filamentous polymers, described herein above.
  • cross linking is achieved via stereocomplexation which utilizes stereoisomers, such as the stereoisomers of copolymer of lactic acid, as linker molecules for stereo-cross-linking the polymer backbone (see Example 1 of the Examples section for further detail).
  • stereoisomers such as the stereoisomers of copolymer of lactic acid
  • the continuous scaffold of the present invention is designed so as to support tissue formation in and around the filamentous scaffold.
  • the continuous scaffold can be composed of any of the polymers described hereinabove and/or any other polymers suitable for supporting structural tissue colonization/proliferation.
  • polysaccharide such as dextran, arabinogalactan, chitosan, alginates, pullulan, hyaluronic acid, and the like
  • proteins such as gelatine, collagen, fibrin, fibrinogen, albumin, and the like
  • synthetic polymers such as, lactide and glycolide foams can also be used
  • the continuous scaffold component is generated under mild conditions. This enables to form the continuous scaffold component over a filamentous scaffold component which is already seeded with cells.
  • Compositions based on viscous hyaluronic acid solutions, alginates cross-linked by calcium salts and proteins cross-linked by denaturation or non-harmful molecules can be used to form the continuous scaffold component over an already seeded filamentous scaffold component.
  • stereocomplexed hydrophilic polymers including, natural polysaccharides, proteins, and polymers based on ethylene and propylene glycol and mixtures thereof can also be used.
  • the composite scaffold or any of its components may also include a biologically active (bioactive) agent.
  • bioactive agents can be incorporated into, or attached to, scaffold material. Incorporation or attachment can be configured for slow or timed release of one or more bioactive agents under suitable conditions.
  • Bioactive agents used are slightly water-soluble, preferably moderately water-soluble, and are diffusible through the polymeric composition. They can be acidic, basic, or salts. They can be neutral molecules, polar molecules, or molecular complexes capable of hydrogen bonding. They can be in the form of ethers, esters, amides and the like, which are biologically activated when injected into the human or animal body.
  • Bioactive agents suitable for use with the composite scaffold of the present invention include cytokines and growth factors for inducing proliferation/differentiation of various cell types, therapeutic agents used for the treatment, prevention, diagnosis, cure or mitigation of disease or illness, and diagnostic agents.
  • cytokines and growth factors include, VEGF, Ang1, Ang.-2, Ang3, PDGF, members of the Transforming Growth Factor b family, members of the Bone Morphogenetic Proteins family, members of the Fibroblasts Growth Factors family.
  • Therapeutic agents include, but are not limited to, anabolic agents, antacids, anti-asthmatic agents, anti-cholesterolemic and anti-lipid agents, anti-coagulants, anti-convulsants, anti-diarrheals, anti-emetics, anti-infective agents, anti-inflammatory agents, anti-manic agents, anti-nauseants, anti-neoplastic agents, anti-obesity agents, anti-pyretic and analgesic agents, anti-spasmodic agents, anti-thrombotic agents, anti-uricemic agents, anti-anginal agents, antihistamines, anti-tussives, appetite suppressants, biologicals, cerebral dilators, coronary dilators, decongestants, diuretics, diagnostic agents, erythropoietic agents, expectorants, gastrointestinal sedatives hyperglycemic agents, hypnotics, hypoglycemic agents, ion exchange resins, laxatives, mineral supplements, mucolytic agents,
  • the bioactive agent can be provided in the form of a chemical, a peptide or polypeptide, or a nucleic acid molecule which can form a part of a tissue expressible construct.
  • Incorporation of the active agent in the scaffolds can be performed during scaffold preparation.
  • a negatively charged bioactive agent such as a DNA molecule
  • a pre-cationized scaffold material prior to, during or following scaffold formation.
  • the bioactive agent can be encapsulated within a delivery system (e.g., microspheres, liposomes) followed by attachment of the delivery system to the prepared scaffold.
  • a delivery system e.g., microspheres, liposomes
  • the scaffold material can be formulated to degrade following implantation thereby promoting the release of the bioactive material from the scaffold.
  • Release of a material having a low solubility in water typically requires the degradation of a substantial part of the polymer matrix to expose the material directly to the surrounding tissue fluids.
  • the release of the biologically active material from the scaffold can be varied by, for example, the solubility of the bioactive material in water, the distribution of the bioactive material within the scaffold, or the size, shape, porosity, solubility and biodegradability of the scaffold material, among other factors.
  • the release of the bioactive material from the scaffold can also be controlled by varying the molecular weight of the scaffold material and by adding rate modifying agents, such as oxidizing agents or hydrophilic molecules.
  • a specific rate and/or time of release may be configured for each bioactive agent. Such specific release control is particularly desired in cases where the bioactive agents are used for inducing tissue formation.
  • the biologically active agents are used in amounts that are therapeutically effective.
  • the effective amount of a biologically active agent will depend on the particular material being used, delivery system and releasing rate.
  • heterogeneous tissue ex-vivo.
  • the heterogeneous tissue is represented by vascularized structural tissue.
  • the first step typically involves building the vascular infrastructure.
  • Vascular cells can be any cell type which can differentiate and give rise to cells lining blood vessels and capillaries, i.e., endothelial cells or pericytes from large blood vessels, skin tissue, foreskin tissue bone marrow and the like.
  • the vascular cells are seeded onto the polymeric filaments of the filamentous scaffold, described hereinabove. This step can be effected using a spinner flask or an agitating tube. Seeded filamentous scaffold is incubated in a 37 ⁇ C incubator for 7-14 days. This results in the cover of the filamentous scaffold with vascular cells and forming a complex of branched tubular vascular structures termed as vascular bed system or vascular infrastructure (see Example 6 of the Examples section).
  • vascular endothelial growth factor VEGF
  • VEGF vascular endothelial growth factor
  • a timed release of these factors mimics the biological state, hence enabling angiogenic differentiation and blood vessel formation.
  • the vascular bed formed is embedded in a continuous scaffold.
  • Embedding can be effected, for example, by using aqueous solution of natural or semi-natural polysaccharides or proteins such as hyaluronic acid, alginates, oxidized cellulose, high molecular weight dextran, pullulan or arabinogalactan.
  • tissue cells such as bone forming or muscle forming cells, can be mixed in to the colloidal dispersion and cast onto the vascular infrastructure to form a seeded composite scaffold.
  • the cell containing continuous scaffold material can be solidified by adding gelling ingredients such as calcium salts, in case of acidic polysaccharides (i.e., alginates, hyaluronic acid, and the like) or oxidized aldehyde containing polysaccharides in case of primary amino containing proteins (i.e., gelatin, albumin, collagen, fibrin and the like) or chitosan.
  • gelation can be obtained by stereocomplexation of a polysaccharide based continuous scaffold as described hereinabove.
  • the continuous scaffold solution can be cast over the filamentous scaffold and the cells seeded thereupon following polymerization.
  • Example 4 of the Examples section which follows illustrates scaffold co-culturing of two cellular components ex-vivo.
  • the composite scaffold of the present invention can also be used for in-vivo tissue generation.
  • In-vivo tissue generation can be effected by implanting the composite scaffold of the present invention in an individual (see, for example, FIG. 2, Paradigm 1 ).
  • scaffold materials preferably include bioactive agents (e.g., angiogenic factors) for promoting growth of one or more tissue types in-vivo.
  • bioactive agents e.g., angiogenic factors
  • the composite scaffold of the present invention can also be used for combined ex-vivo/in-vivo tissue generation.
  • a vascular infrastructure scaffold carrying vascular cells is embedded in the tissue continuous scaffold and the composite scaffold formed is transplanted within an individual. Transplantation of such a scaffold provides a vascular infrastructure ready to integrate with the host vascular system and as such provides optimal setting for ingrowth of tissue and regeneration.
  • structural tissue can be formed ex-vivo on a continuous scaffold surrounding a filamentous scaffold which includes angiogenic factor(s). Following implantation, the angiogenic factor(s) will promote ingrowth of capillaries and blood vessels into the engrafted structural tissue.
  • the present invention provides composite scaffolds and methods of using same for ex-vivo/in-vivo generation of heterogeneous tissue.
  • Two types of dextran-based block copolymers one containing segments of enantiomeric D-lactic acid of at least 10 monomer units and the other composed of segments of enantiomeric L-lactic acid of at least 10 monomer units were prepared. These two copolymers were then mixed together to form specific stereocomplex interactions between the complementary D and L enantiomeric blocks along the polymer chains. Stereocomplexation of a polymeric backbone was effected by separately conjugating copolymeric chains of poly-D and -L lactic acid (PLA), having a molecular weight of 1500, to dextran 40,000 or 500,000 via the dextran hydroxyl groups.
  • PPA poly-D and -L lactic acid
  • PLA (1 eq.), p-nitrophenylchloroformate (4 eq.) and triethylamine (6 eq.) were dissolved in 1 ml dichloromethane per 100 mg PLA. The mixture was stirred at room temperature overnight following which the dichloromethane was evaporated until a concentrated solution was obtained which was precipitated in isopropanol to yield PLA-O-p-nitrophenyl (>90% yield).
  • a filamentous cellular scaffold based on poly(lactide-glycolide) polymer capable of supporting optimal growth of vascular tissue when embedded within a continuous sponge matrix cellular scaffold was synthesized.
  • the use of such a polymer further permits the incorporation of agents, such as polypeptides, nucleic acids or lipids promoting the growth and differentiation of vascular cells therein.
  • the random filaments were slightly compressed so as to form a three dimensional network of filamental bundles that were employed for cell-seeding.
  • Incorporation of a bioactive agent into such a scaffold was effected by mixing the active agent into the polymeric material prior to spooning it into filaments.
  • the active agent can be added as free powdery compound or in a formulated form such as pre-encapsulated in a biodegradable polymeric nano or microspheres.
  • the active agent can also be applied onto the formed filament either by coating the filament with a polymeric solution that contains the active agent which after solvent evaporation forms a thin coating containing the active agent.
  • Reconstitution of body tissues with tissues grown in three-dimensional artificial cellular scaffolds represents a highly desirable goal for replacement of diseased, defective or absent tissues or organs and is of particular benefit for avoiding rejection of transplanted donor tissues when such reconstitution is not effected with self-tissues.
  • a major obstacle preventing the achievement of this goal is the requirement for vascularization of three-dimensional, biologically-engineered replacement tissues.
  • an artificial cellular scaffold enabling the combined and regulated growth both of target replacement tissues and of their supporting vascular tissues has been constructed.
  • the architecture of such a scaffold is composed of a first filamentous polymeric component designed for the tubular growth of supporting vasculature by endothelial cells which is integrally embedded within a three-dimensional continuous matrix sponge polymer designed to support the growth of the target replacement tissues. Both of these polymeric components were further designed for the regulated delivery of factors promoting specific differentiation of cells desired for each component.
  • a filamentous polymer scaffold for growth of vascular cells is integrally embedded within a continuous matrix scaffold for growth of target replacement tissues. This is achieved by inducing polymerization of the continuous matrix polymer scaffold around the filamentous polymer scaffold.
  • the polymers constituting the scaffold were pre-treated so as to optimize the in vivo implantation, growth and/or differentiation of their respective tissues. This was effected by pre-treating the scaffold components to express chemoattractants, adhesion molecules, differentiation factors and/or growth factors and/or to incorporate nucleic acids leading to the production of such factors.
  • VEGF vascular endothelial growth factor
  • an integrated cellular scaffold was implanted in vivo without pre-seeding with precursor cells to promote therein the differential colonization and growth of vascular and target replacement tissues derived from colonizing cells of the recipient (FIG. 2, Paradigm 1 ).
  • the target replacement tissue-specific and the vasculature-specific polymers constituting the scaffold are pre-treated so as to optimize the implantation, growth and/or differentiation of their respective tissues, however in this case the vasculature-specific polymers are pre-treated so as to optimize the in vitro growth of vasculature which is followed by in vivo growth of target replacement tissues (FIG. 2, Paradigm 2 ).
  • vascular tissues is first performed ex vivo by seeding the scaffold with vascular cells, such as endothelial cells, followed by culturing in a bioreactor under conditions optimal for growth of vascular cells (Vailhe B. et al., Lab Invest. 2001, 81:439) as follows:
  • Scaffolds are seeded with 24 million vascular cells per scaffold by adding a cell suspension in medium of vascular cells on to the scaffold in a 96-well in tubes followed by culturing in spinner flasks or in agitated tubes for 20 hours.
  • These vascular cells can include endothelial cells or pericytes harvested from large blood vessels, skin tissue, foreskin tissue or bone marrow from the patient or from a donor.
  • These pre-seeded angiogenic scaffolds are then further cultured in a bioreactor or in a spinner flask for 7-14 days. Following formation of this angiogenic component, the resulting vascular bed is embedded in the target replacement tissue polymer solution which is induced to polymerize around the vascular infrastructure.
  • the pre-vascularized scaffold is implanted in a recipient at the desired anatomical location for colonization, growth and differentiation of the target replacement tissue.
  • both the target replacement tissues and the supporting vascular tissues were grown ill the integrated cellular scaffold in vitro prior to implantation in vivo (FIG. 2, Paradigm 3 ).
  • the target replacement tissue-specific and the vasculature-specific polymers constituting the scaffold were pre-treated so as to optimize the implantation, growth and/or differentiation of their respective tissues, however in this case these were pre-treated so as to optimize their growth and differentiation in vitro prior to implantation in the recipient.
  • the growth of both vascular and target replacement tissues were performed ex vivo by seeding the scaffold with vascular cells and the seeded scaffolds were cultured in a bioreactor under conditions permitting the growth and differentiation of both tissue types as follows:
  • Filamentous polymer cellular scaffolds were seeded with 2-4 million endothelial cells per scaffold and seeded scaffolds were incubated in spinner flasks or in agitated tubes for 20 hours, following which scaffolds were further incubated in a rotating bioreactor or in a spinner flask for 7-14 days.
  • the resulting vascular bed was embedded immersion in a polymerizing continuous matrix polymer solution containing target replacement tissue cells.
  • the integrated cellular scaffold can be seeded with target replacement tissue precursor cells following polymerization by culture in spinner flasks or in agitating tubes for 4-10 hours. Growth of target replacement tissues with the scaffold was effected by culturing the seeded scaffold in a bioreactor for an additional 7 to 14 days.
  • scaffolds containing reconstituted and vascularized tissues were implanted in recipients at the desired anatomical location in vivo for further colonization and growth of the component tissues.
  • MSCs were transfected with a liposome-DNA complex, as depicted in FIG. 3 a, to determine the feasibility of this transfection modality prior to performing such a transfection with cellular scaffold loaded liposome-DNA complex.
  • DNA-liposome complex was prepared by mixing 2.2 ml of 2.2 mM pNGVL1-ntbGAL with 98 ml serum-free medium followed by brief vortexing. To this mixture was added 82 ml of “Superfect” (QUIAGEN) liposome suspension and this mixture was allowed to sit for 5-10 min to allow complexation of plasmid and liposomes after which 486 ml of complete medium was added to the complexation mixture. The cell monolayer was subsequently washed in 1 ml PBS and incubated with liposome-plasmid complex for 2-3 hours.
  • 1.3 ⁇ 10 5 MSCs cells were cultured in 2 ml complete medium (DMEM supplemented with 2 mM L-glutamine, 100 units/ml penicillin, 100 units/ml streptomycin and 10% FCS) in 6-well plates for 24 hours in a 37° C. humidified incubator with 5% CO 2 , resulting in growth of a cell monolayer.
  • a transfection mixture consisting of 30 ml of 0.2 mM pNGVL1-ntbGAL, 24 ml dextran polycation and 29 ml HBSS was prepared and allowed to sit for 30 min to allow complexation of DNA and dextran. The cell monolayer was washed with 1 ml of 1 ⁇ PBS.
  • Arabinogalactan-chitosan scaffolds were loaded with dextran-complexed or liposome-complexed DNA in order to test the ability of such scaffolds to induce cellular colonization and to serve as a DNA delivery system, when loaded with DNA, capable of leading to the up-take and expression of transgenes in colonizing cells.
  • Non-complexed, dextran-complexed and liposome-complexed plasmid was then prepared for incorporation into arabinogalactan-chitosan scaffolds as follows:
  • compositions of plasmid DNA for loading in cellular scaffolds were prepared as follows:
  • Non-complexed plasmid 51 ml of 1.5 mM pNGVL1-ntbGAL was mixed with 49 ml of 5% dextrose
  • Liposome-complexed plasmid 21 ml of 7.4 mM DOTAP-cholesterol (ROCHE) solution was mixed with 51 ml of 1.5 mM plasmid DNA and 28 ml of 5% dextrose
  • Dextran-complexed plasmid 27 ml of dextran polymer (4 mg/ml) was mixed with 51 ml plasmid DNA and 22 ml of 5% dextrose
  • Plasmid-loaded scaffolds were transplanted subcutaneously in C3H/HeN mice and the in vivo-passaged scaffold samples were harvested at two and four weeks post-implantation for analysis of ⁇ -galactosidase expression.
  • Arabinogalactan-chitosan cellular scaffolds were loaded with naked, or liposome-complexed human BMP-2-encoding plasmid. DNA complexation and loading to scaffolds was performed as described above. Scaffolds were processed as described above, with the modification that these were loaded with 200 mg of BMP-2-encoding plasmid DNA per scaffold. Plasmid-loaded cellular scaffolds were then implanted subcutaneously into C3H/HeN and CD Nude mice and, four weeks post-implantation, samples were harvested and processed for histological and immunohistochemical analyses.
  • tissue-replacement is to seed and culture cellular scaffolds in vitro with precursor cells specific for the target replacement tissue which have been genetically modified to express a factor promoting the growth and differentiation of such a tissue. Such seeded scaffolds may then be reimplanted in vivo for efficient reconstitution of target replacement tissues.
  • a major obstacle to tissue replacement effected by in vitro culture of three-dimensional replacement tissues is the requirement for such tissues to be vascularized.
  • this can be achieved by employing an integrated cellular scaffold containing a filamentous polymer component for growth of vascular cells embedded within a continuous sponge matrix polymer supporting the growth and differentiation of the target replacement tissues.
  • Such an integrated cellular scaffold can thus be seeded with both target replacement tissue precursor cells and vasculogenic cells to thereby form the target replacement tissue with supporting vascularization.
  • Arabinogalactan-chitosan scaffolds prepared as described in Example 4, were hydrated by agitation in 15 ml complete medium for 6 hours. Afterwards, hydrated scaffolds were seeded with 2 ⁇ 10 6 MSCs genetically modified to express BMP-2 and ⁇ -galactosidase in a volume 1 ml complete medium per scaffold and cultured with agitation at 100 RPM for 20 hours. Seeded scaffolds were then further cultured in a rotating bioreactor (SYNTHECON) at 4 scaffolds per 50 ml vessel for 2 weeks with medium replacement every 48 h.
  • SYNTHECON rotating bioreactor
  • cellular scaffolds were seeded with both endothelial cells and MSCs genetically-modified to express BMP-2 and ⁇ -galactosidase and were cultured in vitro as follows:
  • Arabinogalactan-chitosan cellular scaffolds were prepared, hydrated and seeded with genetically modified MSCs together with murine endothelial cell lines ⁇ -END-2 or MA 2.1 as described above using 10 6 cells each of genetically modified MSCs and endothelial cells per scaffold (2 ⁇ 10 2 total). Seeded scaffolds were cultured in a rotating bioreactor (Synthecon) at 4 scaffolds per 50 ml vessel for 1 week and were then harvested and processed for histological analysis. Frozen 15 mm sections of cultured scaffolds were stained with either H&E or X-gal for detection of cartilage tissue and cells derived from the genetically modified MSCs, respectively.
  • tissue-replacement is to seed cellular scaffolds with precursor cells specific for the target replacement tissue which have been genetically modified to express a factor specific for the growth and/or differentiation of such a tissue. Such seeded scaffolds may then be implanted in vivo in order to reconstitute the target replacement tissue following in vitro culture. This can be achieved by either implanting such scaffolds ectopically, either in the intended recipient or in a donor so as to enable in vivo culturing of replacement tissues. Alternatively, seeded scaffolds can be directly implanted orthotopically within the anatomical location requiring the replacement tissues.
  • a regulatable transgene promoter system can be employed to enable shutting off of transgene expression. This constitutes an important safety feature when implanting genetically modified cells in the context of tissue-replacement therapy.
  • Precut collagen sponges (3 ⁇ 3 ⁇ 2 mm, Colastat® #CP-3n, Vitaphore Corp.) were seeded with approximately 10 6 BMP-2 and ⁇ -galactosidase coexpressing.
  • MSCs cultured in DUEM supplemented with 2 mM L-glutamine, 100 units/ml penicillin, 100 units/ml streptomycin and 10% FCS. Seeded matrices were then implanted into abdominal muscle of C3H/HEN mice. One control group of mice was maintained with water containing 0.5 mg/ml doxycycline.
  • Detection of BMP-2 DNA and mRNA in implants was performed, respectively, by PCR and RT-PCR analysis of 1 mg genomic DNA and 2 mg mRNA harvested from abdominal muscle tissues. Transplanted cells were detected immunohistochemically with antibodies recognizing BMP-2 protein.
  • Bone regeneration was analyzed by micro-CT morphometric analysis 20 days post-implantation.
  • Ectopic growth and differentiation of osseous tissues derived from MSCs genetically modified to express BMP-2 implanted in vivo BMP-2 and ⁇ -galactosidase co-expressing MSCs were implanted in abdominal muscle of BALB/c mice were observed to develop into tissues displaying cartilage and bone phenotypes on Day 10 (FIGS. 6 a - e ,) and to develop into tissues displaying a bone marrow phenotype on Day 20 (FIG. 6 c and e ).
  • Genetically modified cells in cartilage tissue and in bone-lining trabecular tissue were found to display chondrocytic and osteoblastic morphologies, respectively, as determined by X-gal staining (FIGS.
  • mesenchymal stem cells genetically modified to express the osteogenic factor BMP-2 are capable of forming organized osseous tissue including cartilage, bone and bone marrow when implanted ectopically in muscle tissue. Such a method can thus be utilized to grow replacement bony tissues ectopically for bone replacement therapy.
  • MSCs were genetically modified with a transgene under the regulatory control of a tetracycline-inhibited promoter in order to provide a negative control for expression of BMP-2, thus enabling determination of its effect on differentiation of MSCs and also to provide an inducible shut-off system to terminate expression of a transgene by a genetically modified precursor cell employed in tissue replacement therapy.
  • Doxycycline, a tetracycline analog was administered in the drinking water of in one control group of animals having received an implant of genetically modified cells.
  • mesenchymal stem cells genetically modified to express BMP-2 have the capacity to efficiently regenerate damaged or missing bone tissues in vivo.
  • MSCs respond to growth factors to differentiate and give rise to cartilage and bone cells (Triffitt, J T. Principles of bone biology. Eds. Bilezikian, J P. et al., Acad. Press Inc. NY. 1996, p. 39).
  • a major obstacle to replacement of body tissues with replacement tissues grown within cellular scaffolds is the requirement for such tissues to be vascularized.
  • Vascularization of such cellular scaffold-grown replacement tissues may be induced in vivo by factors promoting angiogenesis, such as factors secreted by the replacement tissue cells themselves, for example, by replacement tissue precursor cells transfected to express such factors.
  • angiogenesis of cellular scaffold-grown replacement tissues can be effected in vivo by pre-seeding of scaffolds with angiogenic cells prior to implantation.
  • An important means of optimizing angiogenesis of replacement tissues grown in cellular scaffolds is to employ scaffolds, as described above, in which a filamentous polymer for the tubular growth of vascular tissues is embedded integrally within a continuous, three dimensional sponge matrix supporting the growth of target replacement tissues.
  • Vitrogen Collagen gels (Vitrogen 100; Collagen Corp., U.S.A.) were seeded with MSCs genetically modified to express the osteogenic factor BMP-2 under the regulatory control of a tetracycline/doxycycline-inhibited promoter and cultured in vitro. Cultured matrices were then implanted subcutaneously in C3H/HeN mice. Twenty days following implantation, implant samples were harvested and analyzed for blood vessel formation by histology and by immunohistochemical staining for detection of the endothelial marker PECAM. The surface area of the blood vessels formed in the implants was quantified via computerized histomorphometry.
  • Chick chorioallantoic membrane (CAM) angiogenesis assays In order to determine the angiogenic potential of BMP-2, discs loaded with 25 mg of BMP-2 protein were analyzed by chick CAM assay, as previously described (O'Reilly, M. S. et al. Cell 1994, 79:315).
  • Filamentous cellular scaffolds prepared as described in Example 2 were seeded with cells of the B-END-2 endothelial cell line by culturing scaffolds and cells for 20 hours with agitation after which these were transferred a rotating bioreactor for further culturing. Scaffold samples were harvested and processed for analysis of cartilage formation by H&E staining after 3 days of bioreactor culture. After 2 weeks of bioreactor incubation, seeded scaffolds were implanted subcutaneously in CD-nude mice.
  • Angiogenesis induced by 10 mg BMP-2 in chick CAM assays A chick CAM assay of angiogenesis induced by BMP-2 clearly indicated that this protein has an angiogenic effect at a quantity of 10 mg comparted to non in vehicle injection (FIGS. 10 g and 10 h respectively) providing further support for the observation that this factor is angiogenic and can be employed to generate both bone tissue and supporting vasculature when implanted within a scaffold in vivo for bone tissue replacement therapy.
  • Angiogenesis and chondrogenesis induced in vivo by osseous tissue formed by culturing cellular scaffolds seeded with MSCs genetically modified to express BMP-2 Intra-muscular implants of osseous tissue formed in vitro by seeding cellular scaffolds with MSCs genetically modified to express BMP-2 were shown to, engraft and to undergo very high levels of vascularization on Day 12, as evidenced by the numerous blood vessels visualized by microscopy and by MRI (FIGS. 11 a and 11 b respectively). Histological analysis of H&E stained frozen sections furthermore demonstrated significant growth of cartilage in the transplant area (FIG. 16 a,c ).
  • filamentous cellular scaffolds seeded with cells of the B-END-2 endothelial cell line displayed massive vascularization (FIG. 13).
  • Angiostatin a novel angiogenesis inhibitor that mediates the suppression of metastases by Lewis lung carcinoma. Cell. 79,315-328.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Transplantation (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Dermatology (AREA)
  • Zoology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Botany (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Vascular Medicine (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Composite Materials (AREA)
  • Materials Engineering (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Rheumatology (AREA)
  • Materials For Medical Uses (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Prostheses (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US10/476,064 2001-04-30 2002-04-30 Composite scaffolds and methods using same for generating complex tissue grafts Abandoned US20040203146A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/476,064 US20040203146A1 (en) 2001-04-30 2002-04-30 Composite scaffolds and methods using same for generating complex tissue grafts
US12/285,107 US8197553B2 (en) 2001-04-30 2008-09-29 Composite scaffolds and methods using same for generating complex tissue grafts

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28700301P 2001-04-30 2001-04-30
PCT/IL2002/000336 WO2002087411A2 (fr) 2001-04-30 2002-04-30 Squelettes composites et procedes d'utilisation associes pour generer des greffes de tissus complexes
US10/476,064 US20040203146A1 (en) 2001-04-30 2002-04-30 Composite scaffolds and methods using same for generating complex tissue grafts

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/285,107 Continuation US8197553B2 (en) 2001-04-30 2008-09-29 Composite scaffolds and methods using same for generating complex tissue grafts

Publications (1)

Publication Number Publication Date
US20040203146A1 true US20040203146A1 (en) 2004-10-14

Family

ID=23101055

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/476,064 Abandoned US20040203146A1 (en) 2001-04-30 2002-04-30 Composite scaffolds and methods using same for generating complex tissue grafts
US10/234,707 Abandoned US20030068817A1 (en) 2001-04-30 2002-09-05 Vascular tissue engineering
US12/285,107 Expired - Fee Related US8197553B2 (en) 2001-04-30 2008-09-29 Composite scaffolds and methods using same for generating complex tissue grafts

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/234,707 Abandoned US20030068817A1 (en) 2001-04-30 2002-09-05 Vascular tissue engineering
US12/285,107 Expired - Fee Related US8197553B2 (en) 2001-04-30 2008-09-29 Composite scaffolds and methods using same for generating complex tissue grafts

Country Status (7)

Country Link
US (3) US20040203146A1 (fr)
EP (1) EP1409647A4 (fr)
JP (1) JP2004528101A (fr)
AU (1) AU2002307791A1 (fr)
CA (1) CA2445523A1 (fr)
IL (1) IL158622A0 (fr)
WO (1) WO2002087411A2 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030068817A1 (en) * 2001-04-30 2003-04-10 Dan Gazit Vascular tissue engineering
US20040156878A1 (en) * 2003-02-11 2004-08-12 Alireza Rezania Implantable medical device seeded with mammalian cells and methods of treatment
US20040228897A1 (en) * 2003-05-16 2004-11-18 Zhang Ping Ye Methods and apparatus for in vivo cell localization
US20050197304A1 (en) * 2003-07-08 2005-09-08 Dicesare Paul Nucleic acid therapy to enhance cartilage repair
US20060122696A1 (en) * 2004-11-22 2006-06-08 Zhang Ping Y Methods and apparatus for in vivo cell therapy
WO2009022133A2 (fr) 2007-08-14 2009-02-19 Smith & Nephew Plc Échafaudages
US20090162415A1 (en) * 2007-12-25 2009-06-25 National Taiwan University Gel Scaffolds for Tissue Engineering
US20090214649A1 (en) * 2008-01-31 2009-08-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Scaffolds with oxygen carriers, and their use in tissue regeneration
US20090318955A1 (en) * 2008-03-14 2009-12-24 Vipul Bhupendra Dave Vascular closure device
WO2010009320A1 (fr) * 2008-07-18 2010-01-21 Cornell University Façonnage d’un système vasculaire en utilisant des structures sacrificielles
US10195644B2 (en) 2012-02-14 2019-02-05 Board Of Regents, The University Of Texas System Tissue engineering device and construction of vascularized dermis

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
GB2397824A (en) * 2003-01-31 2004-08-04 Himedica Ltd Three dimensional cell culture
JP4674288B2 (ja) * 2004-03-31 2011-04-20 恵雄 岡畑 歯科用材料。
US20070233274A1 (en) * 2004-04-12 2007-10-04 Hideyuki Miyake Artificial Tissue and Process for Producing the Same
CA2578951A1 (fr) * 2004-08-30 2006-03-09 Theregen, Inc. Tissus tridimensionnels de culture et utilisations de ceux-ci
US20090232784A1 (en) * 2005-03-10 2009-09-17 Dale Feldman Endothelial predecessor cell seeded wound healing scaffold
EP1887870B1 (fr) * 2005-06-02 2018-09-05 Arizona Board of Regents on behalf of the University of Arizona Dispositifs prévascularisés et méthodes connexes
WO2007001351A1 (fr) * 2005-06-17 2007-01-04 Iken Tissue Therapeutics, Inc. Méthodes de traitement d'un tissu ischémique
US8354258B2 (en) 2005-12-14 2013-01-15 The Invention Science Fund I, Llc Diatom device
US8198080B2 (en) 2005-12-14 2012-06-12 The Invention Science Fund I, Llc Bone delivery device
US8367384B2 (en) 2005-12-14 2013-02-05 The Invention Science Fund I, Llc Bone semi-permeable device
US8734823B2 (en) * 2005-12-14 2014-05-27 The Invention Science Fund I, Llc Device including altered microorganisms, and methods and systems of use
US8278094B2 (en) 2005-12-14 2012-10-02 The Invention Science Fund I, Llc Bone semi-permeable device
US9061075B2 (en) 2005-12-14 2015-06-23 The Invention Science Fund I, Llc Bone delivery device
US8682619B2 (en) * 2005-12-14 2014-03-25 The Invention Science Fund I, Llc Device including altered microorganisms, and methods and systems of use
US8900865B2 (en) 2005-12-14 2014-12-02 The Invention Science Fund I, Llc Blood brain barrier device
US9005944B2 (en) * 2005-12-14 2015-04-14 The Invention Science Fund I, Llc Bone cell delivery device
US7862830B2 (en) * 2006-07-13 2011-01-04 Abbott Cardiovascular Systems Inc. Stereocomplex-forming composition and implantable medical device comprising same
US20120121719A1 (en) * 2009-04-22 2012-05-17 University Of Utah Research Foundation Methods of making and using three-dimensional extracellular matrices
US8551749B2 (en) * 2009-04-23 2013-10-08 The Invention Science Fund I, Llc Device including bone cage and method for treatment of disease in a subject
KR101744040B1 (ko) * 2010-03-01 2017-06-07 후지필름 가부시키가이샤 생체 친화성을 갖는 고분자 블록과 세포로 이루어지는 세포 구조체
US9968705B2 (en) 2011-09-30 2018-05-15 Wake Forest University Health Sciences Bioscaffolds for formation of motor endplates and other specialized tissue structures
RU2519326C2 (ru) 2011-12-29 2014-06-10 Общество с ограниченной ответственностью "НекстГен" Биокомпозит для обеспечения восстановительных процессов после повреждения у млекопитающего, способ его получения (варианты) и применения
EP2809269B1 (fr) 2012-01-31 2020-05-13 Wake Forest University Health Sciences Innervation de structures synthétisées
US11311367B2 (en) 2012-01-31 2022-04-26 Wake Forest University Health Sciences Tissue-engineered gut-sphincter complexes and methods of making the same
BR112017009505A2 (pt) * 2014-11-10 2018-02-06 Ethris Gmbh composição farmacêutica, e, matriz ou suporte.
BR112017013298B1 (pt) * 2014-12-22 2021-12-21 Celwise Ab Ferramenta ou parte de ferramenta, método e sistema para moldar um produto
US20190015190A1 (en) 2015-12-30 2019-01-17 Wake Forest University Health Sciences Tissue-engineered gut-sphincter complexes and methods of making the same
US20220016316A1 (en) * 2018-11-16 2022-01-20 Wake Forest University Health Sciences Methods of bioengineering internal anal sphincter constructs
CN116159184A (zh) * 2023-03-14 2023-05-26 青岛安融生物健康科技有限公司 一种人工血管及其体外制备方法
CN116251232B (zh) * 2023-03-14 2023-11-28 暨南大学 骨组织修复复合支架及其制备方法和应用

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4670286A (en) * 1983-09-20 1987-06-02 Allied Corporation Method of forming prosthetic devices
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5948429A (en) * 1994-11-22 1999-09-07 Tissue Engineering, Inc. Methods for preparing biopolymer foams
US6143293A (en) * 1998-03-26 2000-11-07 Carnegie Mellon Assembled scaffolds for three dimensional cell culturing and tissue generation
US6176874B1 (en) * 1993-10-18 2001-01-23 Masschusetts Institute Of Technology Vascularized tissue regeneration matrices formed by solid free form fabrication techniques
US6365173B1 (en) * 1999-01-14 2002-04-02 Efrat Biopolymers Ltd. Stereocomplex polymeric carriers for drug delivery
US6365149B2 (en) * 1999-06-30 2002-04-02 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6368356B1 (en) * 1996-07-11 2002-04-09 Scimed Life Systems, Inc. Medical devices comprising hydrogel polymers having improved mechanical properties
US20030068817A1 (en) * 2001-04-30 2003-04-10 Dan Gazit Vascular tissue engineering
US6596296B1 (en) * 1999-08-06 2003-07-22 Board Of Regents, The University Of Texas System Drug releasing biodegradable fiber implant

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1340581C (fr) * 1986-11-20 1999-06-08 Joseph P. Vacanti Neomorphogenese chimerique d'organes par implatation cellulaire controlee, utilisant des matrices artificielles
CA2111199C (fr) 1991-06-21 2008-08-05 Eyal Ron Formulations pharmaceutiques de proteines osteogenes
JP2001510330A (ja) * 1996-11-15 2001-07-31 オシリス セラピューティクス,インコーポレイテッド MSC―メガカリオサイト前駆体組成物およびメガカリオサイト分離による分離メガカリオサイトと会合するMSC▲下s▼の分離方法
WO1999011771A1 (fr) * 1997-09-04 1999-03-11 Science Research Laboratory, Inc. Separation de cellules par utilisation de champs electriques
DK1082410T3 (da) * 1998-05-29 2007-11-26 Osiris Therapeutics Inc Humane CD45 - og/eller fibroblast mesenchymale stamceller
US6177527B1 (en) * 1998-09-08 2001-01-23 Union Carbide Chemical & Plastics Technology Corporation Process for the preparation of polyethylene or polypropylene
US7776359B1 (en) 1999-02-19 2010-08-17 Universiteit Utrecht Stereocomplex hydrogels
EP2348104A1 (fr) 1999-08-05 2011-07-27 Mcl Llc Cellules souches adultes multipotentes et procédés d'isolation
US20020142457A1 (en) * 1999-12-28 2002-10-03 Akihiro Umezawa Cell having the potentiality of differentiation into cardiomyocytes
WO2002067978A1 (fr) * 2001-02-23 2002-09-06 Wyeth Utilisation des protéines bmp pour potentialiser la chondrogenèse par les cellules cd105+ tirées de la moelle osseuse humaine
EP1551955A4 (fr) * 2002-07-16 2006-11-29 Yissum Res Dev Co Procedes servant a implanter des cellules souches mesenchymateuses pour la reparation et le developpement de tissus

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4670286A (en) * 1983-09-20 1987-06-02 Allied Corporation Method of forming prosthetic devices
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US6176874B1 (en) * 1993-10-18 2001-01-23 Masschusetts Institute Of Technology Vascularized tissue regeneration matrices formed by solid free form fabrication techniques
US5948429A (en) * 1994-11-22 1999-09-07 Tissue Engineering, Inc. Methods for preparing biopolymer foams
US6368356B1 (en) * 1996-07-11 2002-04-09 Scimed Life Systems, Inc. Medical devices comprising hydrogel polymers having improved mechanical properties
US6143293A (en) * 1998-03-26 2000-11-07 Carnegie Mellon Assembled scaffolds for three dimensional cell culturing and tissue generation
US6365173B1 (en) * 1999-01-14 2002-04-02 Efrat Biopolymers Ltd. Stereocomplex polymeric carriers for drug delivery
US6365149B2 (en) * 1999-06-30 2002-04-02 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6596296B1 (en) * 1999-08-06 2003-07-22 Board Of Regents, The University Of Texas System Drug releasing biodegradable fiber implant
US20030068817A1 (en) * 2001-04-30 2003-04-10 Dan Gazit Vascular tissue engineering

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8197553B2 (en) 2001-04-30 2012-06-12 Yissum Research Development Company Of The Hebrew University Of Jerusalem Composite scaffolds and methods using same for generating complex tissue grafts
US20090035349A1 (en) * 2001-04-30 2009-02-05 Yissum Research Development Company Of The Hebrew University Of Jerusalem Composite scaffolds and methods using same for generating complex tissue grafts
US20030068817A1 (en) * 2001-04-30 2003-04-10 Dan Gazit Vascular tissue engineering
US20040156878A1 (en) * 2003-02-11 2004-08-12 Alireza Rezania Implantable medical device seeded with mammalian cells and methods of treatment
US20040228897A1 (en) * 2003-05-16 2004-11-18 Zhang Ping Ye Methods and apparatus for in vivo cell localization
US20050197304A1 (en) * 2003-07-08 2005-09-08 Dicesare Paul Nucleic acid therapy to enhance cartilage repair
US20060122696A1 (en) * 2004-11-22 2006-06-08 Zhang Ping Y Methods and apparatus for in vivo cell therapy
WO2009022133A2 (fr) 2007-08-14 2009-02-19 Smith & Nephew Plc Échafaudages
EP2187985B1 (fr) * 2007-08-14 2016-09-28 Smith & Nephew PLC Échafaudages
EP2187985A2 (fr) * 2007-08-14 2010-05-26 Smith & Nephew, PLC Échafaudages
US20110064809A1 (en) * 2007-08-14 2011-03-17 Kelly Langford Scaffolds
AU2008288260B2 (en) * 2007-08-14 2014-10-02 Smith & Nephew Plc Scaffolds
US20090162415A1 (en) * 2007-12-25 2009-06-25 National Taiwan University Gel Scaffolds for Tissue Engineering
US20090214649A1 (en) * 2008-01-31 2009-08-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Scaffolds with oxygen carriers, and their use in tissue regeneration
US9381273B2 (en) * 2008-01-31 2016-07-05 Yissum Research Development Company Of The Hebrew University Of Jerusalem Scaffolds with oxygen carriers, and their use in tissue regeneration
US8876860B2 (en) * 2008-03-14 2014-11-04 Cordis Corporation Vascular closure device
US20090318955A1 (en) * 2008-03-14 2009-12-24 Vipul Bhupendra Dave Vascular closure device
US9242027B2 (en) 2008-07-18 2016-01-26 Cornell University Fabrication of a vascular system using sacrificial structures
WO2010009320A1 (fr) * 2008-07-18 2010-01-21 Cornell University Façonnage d’un système vasculaire en utilisant des structures sacrificielles
US10195644B2 (en) 2012-02-14 2019-02-05 Board Of Regents, The University Of Texas System Tissue engineering device and construction of vascularized dermis

Also Published As

Publication number Publication date
US20030068817A1 (en) 2003-04-10
CA2445523A1 (fr) 2002-11-07
EP1409647A4 (fr) 2009-09-09
US8197553B2 (en) 2012-06-12
WO2002087411A3 (fr) 2004-02-26
WO2002087411A2 (fr) 2002-11-07
JP2004528101A (ja) 2004-09-16
AU2002307791A1 (en) 2002-11-11
EP1409647A2 (fr) 2004-04-21
IL158622A0 (en) 2004-05-12
US20090035349A1 (en) 2009-02-05

Similar Documents

Publication Publication Date Title
US8197553B2 (en) Composite scaffolds and methods using same for generating complex tissue grafts
Fan et al. Macroporous hydrogel scaffolds for three-dimensional cell culture and tissue engineering
US11110148B2 (en) Silk fibroin materials and use thereof
Ikada Tissue engineering: fundamentals and applications
Lih et al. Biomimetic porous PLGA scaffolds incorporating decellularized extracellular matrix for kidney tissue regeneration
Yoon et al. Natural and synthetic polymeric scaffolds
US8815276B2 (en) Three-dimensional nanostructured hybrid scaffold and manufacture thereof
JP4624800B2 (ja) 細胞の成長を促進するための工学設計骨格
EP1835949B1 (fr) Dispositifs d'ingenierie tissulaire destines a reparer et a regenerer des tissus
US6991652B2 (en) Tissue engineering composite
Yao et al. Biomimetic injectable HUVEC‐adipocytes/collagen/alginate microsphere co‐cultures for adipose tissue engineering
Kundu et al. Biomaterials for biofabrication of 3D tissue scaffolds
Sarker et al. Bioprinting of vascularized tissue scaffolds: influence of biopolymer, cells, growth factors, and gene delivery
AU2005279771A1 (en) Cultured three dimensional tissues and uses thereof
Spadaccio et al. Drug releasing systems in cardiovascular tissue engineering
US20050260179A1 (en) Stem and progenitor cell capture for tissue regeneration
De Vries et al. Bioengineering, biomaterials, and β-cell replacement therapy
Ahadian et al. Biomaterials in tissue engineering
Esmaeili et al. Exploring the evolution of tissue engineering strategies over the past decade: From cell-based strategies to gene-activated matrix
KR20210042827A (ko) 신규한 다공성 스캐폴드 및 이의 제조방법
Gupta et al. Tissue engineering and artificial organ
Salimi et al. Application of nanoscaffolds and mesenchymal stem cells in tissue engineering
Hosseinkhani et al. Tissue engineered Scaffolds for stem cells and regenerative medicine
Ko et al. Tissue engineering of the urogenital system
Roether et al. Polymers for Myocardial Tissue Engineering

Legal Events

Date Code Title Description
AS Assignment

Owner name: YISSUM RESEARCH DEVELOPMENT COMPANY, ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GAZIT, DAN;DOMB, AVRAHAM;TURGEMAN, GADI;AND OTHERS;REEL/FRAME:015367/0379;SIGNING DATES FROM 20040501 TO 20040513

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION