WO1997030662A1 - Method for making and/or repairing cartilage - Google Patents

Method for making and/or repairing cartilage Download PDF

Info

Publication number
WO1997030662A1
WO1997030662A1 PCT/US1997/002909 US9702909W WO9730662A1 WO 1997030662 A1 WO1997030662 A1 WO 1997030662A1 US 9702909 W US9702909 W US 9702909W WO 9730662 A1 WO9730662 A1 WO 9730662A1
Authority
WO
WIPO (PCT)
Prior art keywords
scaffold
cells
stromal cells
tissue
fact
Prior art date
Application number
PCT/US1997/002909
Other languages
French (fr)
Inventor
Gail K. Naughton
Jane Willoughby
Original Assignee
Advanced Tissue Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Advanced Tissue Sciences, Inc. filed Critical Advanced Tissue Sciences, Inc.
Priority to JP9530388A priority Critical patent/JP2000505338A/en
Priority to NZ331517A priority patent/NZ331517A/en
Priority to AU19731/97A priority patent/AU715282B2/en
Priority to EP97907835A priority patent/EP0955959A1/en
Publication of WO1997030662A1 publication Critical patent/WO1997030662A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • A61L27/3834Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/30Joints
    • A61F2/30756Cartilage endoprostheses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3641Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the site of application in the body
    • A61L27/3645Connective tissue
    • A61L27/3654Cartilage, e.g. meniscus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials 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 subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment
    • A61L27/3687Materials 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 subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment characterised by the use of chemical agents in the treatment, e.g. specific enzymes, detergents, capping agents, crosslinkers, anticalcification agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3839Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by the site of application in the body
    • A61L27/3843Connective tissue
    • A61L27/3852Cartilage, e.g. meniscus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/30Joints
    • A61F2002/30001Additional features of subject-matter classified in A61F2/28, A61F2/30 and subgroups thereof
    • A61F2002/30003Material related properties of the prosthesis or of a coating on the prosthesis
    • A61F2002/3006Properties of materials and coating materials
    • A61F2002/30062(bio)absorbable, biodegradable, bioerodable, (bio)resorbable, resorptive
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/30Joints
    • A61F2002/30001Additional features of subject-matter classified in A61F2/28, A61F2/30 and subgroups thereof
    • A61F2002/30108Shapes
    • A61F2002/30199Three-dimensional shapes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/30Joints
    • A61F2/30756Cartilage endoprostheses
    • A61F2002/30766Scaffolds for cartilage ingrowth and regeneration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2210/00Particular material properties of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2210/0004Particular material properties of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof bioabsorbable
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2230/00Geometry of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2230/0063Three-dimensional shapes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/06Materials or treatment for tissue regeneration for cartilage reconstruction, e.g. meniscus
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S623/00Prosthesis, i.e. artificial body members, parts thereof, or aids and accessories therefor
    • Y10S623/902Method of implanting

Definitions

  • the present invention relates to methods for making and/or repairing cartilage in vivo. More specifically, the invention relates to methods of making and/or repairing cartilage comprising implanting into a patient, at a site of cartilage damage or loss, a biocompatible, non-living three- dimensional scaffold or framework structure, in combination with periosteal/perichondrial tissue, and administering a preparation of chondrocytes and/or other stromal cells, such as chondrocyte progenitor cells, to the site of the implant before, during or after implantation of the scaffold and/or the periosteal/perichondrial tissue.
  • the periosteal/perichondrial tissue can be used to hold the scaffold in place at the site of implantation and also provides a source of stromal cells, e.g., chondrocytes and/or chondrocyte progenitor cells, for attachment to the scaffold in vivo.
  • stromal cells e.g., chondrocytes and/or chondrocyte progenitor cells
  • the preparation of stromal cells seeded directly into the implantation site in vivo provides not only a readily-accessible source of chondrocytes and/or other stromal cells for attachment to the scaffold but also provides a rapid and efficient means of inducing chondrogenesis as well as migration of stromal cells from the surrounding in vivo environment to the scaffold via factors produced by the stromal cells of the preparation.
  • the seeded stromal cells can be genetically engineered to express gene products beneficial to growth, implantation and/or amelioration of disease conditions. The methods of the invention therefore result in the efficient production
  • the methods of this invention are useful in the production/repair of articular cartilage in patients suffering from degenerative connective tissue diseases such as rheumatoid and/or osteoarthritis as well as in patients who have cartilage defects due to trauma.
  • the methods of this invention can be used to replace or augment existing cartilage tissue, to introduce new or altered tissue or to join together biological tissues or structures.
  • cartilage e.g., articular or hyaline cartilage, elastic cartilage and fibrocartilage.
  • Articular cartilage is found at the articular surfaces of bones, e.g., in the joints, and is responsible for providing the smooth gliding motion characteristic of moveable joints. Articular cartilage is firmly attached to the underlying bones and measures less than 5mm in thickness in human joints, with considerable variation depending on joint and site within the joint.
  • articular cartilage is aneural, avascular, and alymphatic. In adult humans, this cartilage derives its nutrition by a double diffusion system through the synovial membrane and through the dense matrix of the cartilage to reach the chondrocyte, the cells that are found in the connective tissue of cartilage.
  • articular cartilage consists of highly specialized chondrocyte cells surrounded by a dense extracellular matrix consisting mainly of type II collagen, proteoglycan and water. While the biochemical composition of articular cartilage includes up to 65-80% water (depending on the cartilage) , the collagen component of the cartilage is the most prevalent organic constituent.
  • the collagen (mainly type II) accounts for about 15-25% of the wet weight or about half the dry weight, except in the superficial zone where it accounts for most of the dry weight. Its concentration is usually progressively reduced with increasing depth from the articular surface.
  • the proteoglycan content accounts for up to 10% of the wet weight or about a quarter of the dry weight.
  • Proteoglycans consist of a protein core to which linear sulfated polysaccharides are attached, mostly in the form of chondroitin sulfate and keratin sulfate.
  • articular collagen contains several other collagen types (IV, V, IX and X) with distinct structures.
  • IV, V, IX and X collagen types
  • chondrocytes Induction of cartilage matrix degradation and proteinases by chondrocytes is probably induced primarily by interleukin-1 (IL-1) or tumor necrosis factor- ⁇ (TNF- ) (Tyler, 1985, Bioche . J. 225:493- 507) .
  • IL-1 interleukin-1
  • TNF- tumor necrosis factor- ⁇
  • the current therapy for damage or loss of cartilage is replacement with a prosthetic material, for example, silicone for cosmetic repairs, or metal alloys for joint realignment.
  • Implantation of prosthetic devices is usually associated with loss of underlying tissue and bone without recovery of the full function allowed by the original cartilage. Serious long-term complications associated with the presence of a permanent foreign body can include infection, erosion and instability.
  • Use of sterilized bone or bone powder or surgical steel seeded with bone cells that are eventually implanted have been largely unsuccessful because of the non-degradable nature of the cell support.
  • fibroblasts are exposed in vitro for a minimum of three days to a soluble bone protein capable of stimulating a chondrogenic response.
  • the activated fibroblasts are then transferred in vivo by combining them with a biodegradable matrix or by intra-articular injection or attachment to allografts or prosthetic devices.
  • the disadvantage of this method is that chondrogenesis is not allowed to develop in the short-term cultures and there is an unduly heavy reliance on cartilage synthesis by the exposed fibroblasts at the implant site. See Caplan, A., United States Pat. No. 4,609,551, issued September 2, 1986. United States Pat. No. 5,041,138 to J.P. Vacanti et al.. issued August 20, 1991, describes the growth of cartilaginous structures by seeding chondrocytes on biodegradable matrices in vitro for subsequent implantation in vivo.
  • United States Patents Nos. 5,197,985 and 5,226,914, to Caplan et al.. issued March 30, 1993 and July 13, 1993, respectively, relate to culturing marrow-derived mesenchymal stem cells in vitro in the presence of growth factors, applying these cells to a carrier, e.g., a porous ceramic vehicle, to promote round cell morphology, and implanting the carrier containing the cells into damaged articular cartilage.
  • a carrier e.g., a porous ceramic vehicle
  • Biomaterials 14 (No. 7): 513-521 relate to the use of free autogenous periosteal grafts that are placed directly into full-thickness articular defects for the repair of such defects in animals.
  • the present invention relates to methods of making and/or repairing cartilage in vivo comprising implanting into a patient, at a site of cartilage damage or loss, a biocompatible, non-living three-dimensional scaffold or framework structure in combination with periosteal/perichondrial tissue that can be used to hold the scaffold in place and provides a source of chondrocyte progenitor cells, chondrocytes and other stromal cells for attachment to the scaffold in vivo.
  • a preparation of cells that can include chondrocytes, chondrocyte progenitor cells or other stromal cells is administered, either before, during or after implantation of the scaffold and/or the periosteal/perichondrial tissue; the cells are administered directly into the site of the implant in vivo and promote chondrogenesis and the production of factors that induce the migration of chondrocytes, progenitor cells and other stromal cells from the adjacent in vivo environment into the scaffold for the production of new cartilage at the site of implantation.
  • the three-dimensional scaffold contains interstitial spaces into which progenitor cells, chondrocytes and other stromal cells from the adjacent in vivo environment, including the implanted periosteal/perichondrial tissue, migrate for attachment and growth on and within the scaffold structure.
  • the preparation of stromal cells seeded in combination with the scaffold and periosteal/perichondrial tissue provides a ready source of chondrocytes and other stromal cells which produce biological factors that promote chondrogenesis and the migration of stromal cells from, e.g., the periosteal/perichondrial tissue to the scaffold for attachment and/or differentiation thereon.
  • the stromal cell preparation also provides a direct source of stromal cells, e.g., chondrocytes and/or progenitor cells, that are capable of migrating into the scaffold and attaching thereto.
  • stromal cells e.g., chondrocytes and/or progenitor cells
  • the stromal cells in the scaffold whether derived from the periosteal/perichondrial tissue, from the exogenous stromal cell preparation or from the in vivo environment adjacent to the implant site, grow on the scaffold to form a cellular matrix and provide the support, growth factors and regulatory factors required for cartilage formation at a cartilage defect site in vivo.
  • the methods of this invention thus result in the production of new cartilage in vivo at the implant site.
  • the periosteal/perichondrial tissue is placed over the implanted scaffold at the site of cartilage damage or loss ("the defect site") and affixed, e.g. , by sutures, to that site, thus holding the scaffold in place.
  • the scaffold is composed of a biodegradable material such that, upon successful engraftment, the scaffold structure is completely absorbed in vivo, resulting in new cartilage having no foreign, non ⁇ living material encompassed within it.
  • the preparation of chondrocytes and/or other stromal cells is administered in vivo to the site of the implant after the scaffold and periosteal/perichondrial tissue have been implanted.
  • bioactive agents such as cellular growth factors (e.g. , TGF- ⁇ ) , factors that stimulate chondrogenesis (e.g., bone morphogeni ⁇ proteins (BMPs) that promote cartilage formation) , factors that stimulate migration of stromal cells and/or matrix deposition, anti-inflammatories or immunosuppressants, are included at the implantation site.
  • these factors can be incorporated into the scaffold material to provide for release at the site of implantation; the scaffold can also be comprised of, or coated with, one or more of these bioactive agents.
  • the factor(s) can be administered into or adjacent to the scaffold, either before, during or after seeding of the stromal cells, e.g., the bioactive agent(s) can be administered to the site, either as a separate preparation or as part of the stromal cell preparation.
  • the stromal cells seeded at the defect site can be genetically engineered to express the genes for these bioactive agents, e.g. , specific types of TGF- ⁇ such as TGF-S1 or specific types of BMPs such as BMP-13. Exposure of the defect site to these bioactive agents promotes the successful and/or improved production of new cartilage and/or improves the success of implantation, for example, by reducing the risk of rejection or inflammation associated with the implant.
  • the stromal cells can be genetically engineered to express an i-inflammatory gene products to ameliorate the effects of degenerative diseases like rheumatoid arthritis which result in cartilage damage due to inflammatory reactions; e.g. , the stromal cells can be engineered to express peptides or polypeptides corresponding to the idiotype of neutralizing antibodies for granulocyte- macrophage colony stimulating factor (GM-CSF) , tumor necrosis factor (TNF) , interleukin-2 (IL-2) , or other inflammatory cytokines and mediators.
  • GM-CSF granulocyte- macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • IL-2 interleukin-2
  • the stromal cells can be genetically engineered to express tissue factors that enhance migration of stromal cells from the adjacent in vivo environment into the scaffold at the implantation site.
  • the cells are engineered to express such gene products transiently and/or under inducible control during the post-operative recovery period, or as a chimeric fusion protein anchored to the stromal cell, e.g. , a chimeric molecule composed of an intracellular and/or transmembrane domain of a receptor or receptor-like molecule, fused to the gene product as the extracellular domain.
  • the stromal cells can be genetically engineered to "knock out" expression of factors that promote rejection of the implant or degenerative changes in articular cartilage due to aging, rheumatoid disease or inflammation.
  • factors that promote rejection of the implant or degenerative changes in articular cartilage due to aging, rheumatoid disease or inflammation For example, expression of pro- inflammatory mediators such as GM-CSF, TNF, IL-1, IL-2 and cytokines can be knocked out in the exogenously-administered stromal cells or on the implanted periosteal or perichondrial tissue to reduce the risk of inflammation.
  • the expression of MHC class II molecules on the cells or tissues can be knocked out in order to reduce the risk of rejection of the implant.
  • the methods of the invention may afford a vehicle for introducing genes and gene products in vivo to assist or improve the results of the implantation and/or for use in gene therapies.
  • genes that prevent or ameliorate symptoms of degenerative changes in cartilage such as rheumatoid disease or inflammatory reactions and bone resorption may be underexpressed or overexpressed in disease conditions and/or due to aging.
  • the level of gene activity in the patient may be increased or decreased, respectively, by gene replacement therapy by adjusting the level of the active gene product in genetically engineered stromal cells.
  • the cartilage defect site into which the implant will be placed is treated, preferably prior to implantation, to degrade the pre-existing cartilage at the defect site, freeing cells to migrate into the scaffold of the implant and promoting the orderly deposition of new cartilage.
  • Methods of such treatment include enzymatic treatment, abrasion or microdrilling.
  • the preparation of stromal cells of the invention can be injected into the degraded cartilage at the defect site, e.g., into the surrounding cells or into the walls of the defect, providing a source of biological factors that induce migration of stromal cells from the degraded cartilage to the implant.
  • the present invention involves methods of making and/or repairing cartilage in vivo comprising the implantation in vivo of a three-dimensional scaffold or framework structure made of a biocompatible, non-living material in combination with periosteal/perichondrial tissue and the administration of a preparation of stromal cells, such as chondrocytes or chondrocyte progenitor cells, which cells are seeded at the site of implantation in vivo.
  • stromal cells such as chondrocytes or chondrocyte progenitor cells
  • chondrocyte progenitor cell refers to either (1) a pluripotent, or lineage-uncommitted, progenitor cell, typically referred to in the art as a “stem cell” or “mesenchymal stem cell”, which is potentially capable of an unlimited number of mitotic divisions to either renew its line or to produce progeny cells which will differentiate into chondrocytes; or (2) a lineage-committed progenitor cell produced from the mitotic division of a stem cell which will eventually differentiate into a chondrocyte.
  • the lineage- committed progenitor is generally considered to be incapable of an unlimited number of mitotic divisions and will eventually differentiate into a chondrocyte.
  • cartilage or “cartilage tissue” as used herein is generally recognized in the art, and refers to a specialized type of dense connective tissue comprising cells embedded in an extracellular matrix (ECM) (see, for example, Cormack, 1987, Ham's Histology, 9th Ed., J.B. Lippincott Co., pp. 266-272) .
  • ECM extracellular matrix
  • the biochemical composition of cartilage differs according to type; however, the general composition of cartilage comprises chondrocytes surrounded by a dense ECM consisting of collagen, proteoglycans and water.
  • cartilage Several types are recognized in the art, including, for example, hyaline or articular cartilage such as that found within the joints, fibrous cartilage such as that found within the meniscus and costal regions, and elastic cartilage.
  • the production of any type of cartilage is intended to fall within the scope of the invention.
  • the invention is directed predominantly to methods for the production of new cartilage tissue in humans, the invention may also be practiced so as to produce new cartilage tissue in any mammal in need thereof, including horses, dogs, cats, sheep, pigs, among others. The treatment of such animals is intended to fall within the scope of the invention.
  • the invention is divided into the following sections solely for the purpose of description: (a) the three- dimensional scaffold; (b) the periosteal/perichondrial tissue and its implantation in combination with the scaffold; (c) the stromal cell preparation, including genetically engineered stromal cells; (d) administration of the stromal cells in vivo and (e) uses of the methods of the invention.
  • the three-dimensional scaffold or framework structure may be of any material and/or shape that: (a) allows cells to attach to it (or can be modified to allow the cells to attach to it); and (b) allows cells to grow in more than one layer. Because the three-dimensional structure is to be implanted in vivo, it may be preferable to use biodegradable materials such as polyglycolic acid (PGA) , polylactic acid (PLA) , hyaluronic acid, catgut suture material, gelatin, cellulose, nitrocellulose, collagen, cotton, or other naturally- occurring biodegradable materials.
  • PGA polyglycolic acid
  • PLA polylactic acid
  • catgut suture material gelatin
  • gelatin cellulose
  • nitrocellulose collagen, cotton, or other naturally- occurring biodegradable materials.
  • the three-dimensional structure prior to implantation, e.g., by treatment with ethylene oxide or by gamma irradiation or irradiation with an electron beam.
  • a number of other materials may be used to form the scaffold or framework structure, including but not limited to: nylon (polyamides) , dacron (polyesters) , polystyrene, polypropylene, polyacrylates, polyvinyl compounds (e.g., polyvinylchloride) , polycarbonate (PVC) , polytetrafluorethylene (PTFE, teflon) , thermanox (TPX) , and a variety of polyhydroxyalkanoates.
  • nylon, polystyrene, etc. may be poor substrates for cellular attachment, when these materials are used as the three-dimensional framework, it is advisable to pre-treat the framework prior to implantation in order to enhance the attachment of chondrocytes and other stromal cells to the scaffold.
  • nylon matrices can be treated with 0.1 M acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon.
  • Polystyrene could be similarly treated using sulfuric acid.
  • any of the above-listed materials may be formed into a mesh or a felt, for example, to produce the three-dimensional framework or scaffold for use in the methods of this invention.
  • the openings of the framework should be of an appropriate size to allow the chondrocytes and other stromal cells to stretch across the openings. Maintaining actively growing stromal cells which stretch across the framework enhances the production of growth factors which are elaborated by the stromal cells, and further enhances new cartilage formation in vivo.
  • the openings of the framework must allow for adequate diffusion of nutrients and waste products into and out of the structure and for vascularization at the site of implantation.
  • the stromal cells may rapidly achieve confluence but be unable to easily exit from the matrix; trapped cells may exhibit contact inhibition and cease production of the appropriate factors necessary to support proliferation. If the openings are too large, the stromal cells may be unable to stretch across the opening; this will also decrease stromal cell production of the appropriate factors necessary to support proliferation.
  • openings ranging from about 150 ⁇ m to about 220 ⁇ m are satisfactory. However, depending upon the three- dimensional structure and intricacy of the framework, other sizes may work equally well. In fact, any shape or structure that allows the stromal cells to stretch and continue to replicate and grow for lengthy time periods will work in accordance with the invention. For example, for felt-type frameworks, openings ranging from about 80 ⁇ m to about 120 ⁇ m are preferred.
  • the scaffold is a felt, which can be composed of a multifilament yarn made from a bioabsorbable material, e.g., PGA, PLA, polygluconate (PLGA) or hyaluronic acid.
  • the yarn is made into a felt using standard textile processing techniques consisting of crimping, cutting, carding and needling.
  • the porosity of the felt ranges from 80-98%, the density of the felt ranges from 30-60 mg/cc and the thickness of the felt ranges from 1-7 mm.
  • the collagen may be in the form of a sponge, a braid or woven threads, etc.
  • a convenient nylon mesh is Nitex, a nylon filtration mesh having an average pore size of 210 ⁇ m and an average nylon fiber diameter of 90 ⁇ m (#3-210/36 Tetko, Inc. , N.Y.) .
  • the three-dimensional framework provides a greater surface area for protein attachment, and consequently, for the adherence of stromal cells in vivo.
  • the stromal cells that attach to the framework continue to actively grow and produce growth and regulatory factors which promote new cartilage formation in vivo, and are less likely to exhibit contact inhibition.
  • the three-dimensional framework allows for a spatial distribution of cellular elements which is analogous to that found in vivo.
  • the increase in potential volume for cell growth in the three-dimensional structure may allow the establishment of localized microenvironments conducive to cellular differentiation and maturation in the production of new cartilage in vivo.
  • the three-dimensional matrix maximizes cell-cell interactions by allowing greater potential for movement of migratory cells, such as macrophages, monocytes and possibly lymphocytes.
  • the scaffold may comprise or be modified, e.g. , coated or impregnated, prior to implantation with certain substances to enhance the attachment and growth of chondrocytes and other stromal cells on the scaffold in vivo.
  • bioactive agents such cellular growth factors (e.g., TGF- / 3) , substances that stimulate chondrogenesis (e.g., BMPs that stimulate cartilage formation such as BMP-2, BMP-12 and BMP-13) , factors that stimulate migration of stromal cells to the scaffold, factors that stimulate matrix deposition, anti-inflammatories (e.g., non-steroidal anti-inflammatories) , immunosuppressants (e.g., cyclosporins) , as well as other proteins, such as collagens, elastic fibers, reticular fibers, glycoproteins or glycosaminoglycans, such as heparin sulfate, chondroitin-4- sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc.
  • bioactive agents such as cellular growth factors (e.g., TGF- / 3)
  • substances that stimulate chondrogenesis e.g.
  • chondrocyte differentiation and cartilage formation by chondrocytes have been found to trigger chondrocyte differentiation and cartilage formation by chondrocytes.
  • hyaluronic acid is a good substrate for the attachment of chondrocytes and other stromal cells and can be incorporated as part of the scaffold or coated onto the scaffold.
  • bioactive agents may also be included in or on the scaffold for local, sustained release of the agents.
  • sustained release formulations include composites comprising the bioactive agent and a biocompatible polymer, such as poly(lactic acid), poly(lactic-co-glycolic acid) , methylcellulose, hyaluronic acid, collagen, and the like.
  • a biocompatible polymer such as poly(lactic acid), poly(lactic-co-glycolic acid) , methylcellulose, hyaluronic acid, collagen, and the like.
  • degradable polymers in drug delivery vehicles have been reviewed in several publications, including, A. Domb et al.. 1992, Polymers for Advanced Technologies 3:279-292. Additional guidance in selecting and using polymers in pharmaceutical formulations can be found in the text by M. Chasin and R.
  • the scaffold of the invention as described above is implanted into the defect site in vivo in combination with periosteal tissue, perichondrial tissue or a combination of the two tissues.
  • Periosteal tissue is derived from the periosteum, a fibrous membrane localized at the surfaces of bones, and can be obtained from the periosteum/bone interface of any suitable bone of the patient (or subject) or a histoco patible donor, e.g., ileum, scapula, tibia, fibula, femur, etc.
  • the periosteal tissue contains a variety of stromal cells including osteocytes, chondrocytes and fibroblasts as well as mesenchymal stem cells having the potential to differentiate into osteogenic or chondrogenic cells.
  • Perichondrial tissue is derived from the perichondrium, the fibrous connective tissue covering cartilage, except articular surfaces.
  • Perichondrial tissue contains chondrogenic progenitor cells and chondrocytes.
  • the periosteal/perichondrial tissue can be in the form of a segment or layer of tissue of any size or shape, preferably of a size and shape that fits within or corresponds to the defect site.
  • the tissue can be laid over or under the scaffold at the implantation site and can optionally be mechanically fixed to the scaffold and/or the defect site, e.g., by sutures or glue fixation, e.g., fibrin glue.
  • the periosteal/perichondrial tissue may be autologous (i.e., derived from the subject receiving the implant)
  • the tissue may be derived from a heterologous source.
  • the three-dimensional scaffold is implanted at the defect site in vivo and a piece of periosteal/perichondrial tissue is placed over the implanted scaffold and sutured in place so that the tissue overlays and lies adjacent to the scaffold structure.
  • a segment of periosteum or perichondrium may be implanted directly into the defect site and the scaffold placed on top of the tissue such that the stromal cells of the tissue can migrate from the tissue into the scaffold.
  • the periosteal/perichondrial tissue should be situated with respect to the scaffold in such a way as to allow the stromal cells from the tissue to migrate into the scaffold and proliferate thereon and therein.
  • the scaffold and/or periosteal/perichondrial tissue can be implanted using surgical techniques well known in the art, e.g., arthroscopy.
  • the periosteal tissue is situated or oriented such that the cambium layer of the tissue is facing into the defect; thus, in the embodiment wherein the scaffold is placed directly into the defect site and the periosteal tissue is placed on top of the scaffold, the periosteal tissue is oriented in relation to the top of the scaffold such that the cambium layer is facing the top of the scaffold.
  • the perichondrial tissue is also placed into the defect site or oriented with respect to the scaffold such that its cambium or inner transition layers faces the defect or scaffold. It is these layers that contain chondrogenic stem cells and/or chondrocytes that can migrate into the scaffold for the production of new cartilage at the defect site.
  • a bioresorbable patch e.g., film, mesh or felt
  • a bioresorbable patch can be used in place of the periosteal/perichondrial tissue and situated or oriented adjacent to the scaffold within the defect site.
  • a film e.g., it may be comprised of PGA or polygluconate; if a mesh or felt is used, they may be comprised of vicryl or PLA.
  • the preparation of stromal cells is seeded into the defect site as described herein.
  • the defect site is treated, preferably prior to implantation, to degrade the cartilage at the site of the defect, freeing cells (e.g., stromal cells) from that area to migrate into the scaffold of the implant and promoting the orderly deposition of new cartilage.
  • freeing cells e.g., stromal cells
  • enzymes include but are not limited to trypsin, chymotrypsin, collagenase, elastase, and/or hyaluronidase, Dnase, pronase, chondroitinase, etc.
  • Alternative methods of treating the defect site to degrade the cartilage include abrasion, debridement, shaving or icrodrilling.
  • the surface of the cartilage may be serrated, e.g., via wire wool.
  • a drilling device is used to create small defects or channels in the cartilage. Treatment of the defect site to degrade or disrupt the pre-existing cartilage reduces the chances of scar tissue forming at the site and promotes the orderly deposition of new cartilage at the defect site.
  • a preparation of stromal cells is additionally administered at the implantation site, which cells produce biological factors that promote chondrogenesis and the migration of cells such as chondrogenic stem cells or chondrocytes, from the in vivo environment adjacent to the implant, including from the periosteal/perichondrial tissue, to the scaffold for attachment and/or differentiation thereon and therein.
  • the stromal cell preparation also provides a direct source of stromal cells, e.g. , chondrocytes and/or chondrocyte progenitor cells, that are capable of migrating to the scaffold, attaching thereto, and elaborating cartilage- specific macromolecules and extracellular matrix proteins for the production of new cartilage at the defect site.
  • the cells described herein can be administered before, during or after implantation of the scaffold and/or periosteal/ perichondrial tissue, as discussed in Section 4.4, infra.
  • the stromal cells of the preparation may include chondrocytes, chondrocyte progenitor cells including mesenchymal stem cells, fibroblasts, fibroblast-like cells and/or cells capable of producing collagen type II and other collagen types, and proteoglycans which are typically produced in cartilaginous tissues.
  • the stromal cells can be obtained from the patient (or subject) or a histocompatible donor.
  • chondrocytes, progenitor cells, fibroblast-like cells and other cells and/or elements that comprise the stroma may be fetal or adult in origin, and may be derived from convenient sources such as cartilage, bone, skin, ligaments, tendons, muscles, placenta, umbilical cord, etc.
  • stromal cells such as chondrocytes may be derived from any type of cartilage, including but not limited to, hyaline cartilage, costal cartilage, fibrous cartilage, etc. , which can be obtained by biopsy (where appropriate) or upon autopsy.
  • Chondrocyte progenitor cells may be derived from various sources including bone marrow, periosteum, perichondrium or various sources of undifferentiated human mesenchyme. Fibroblasts can be obtained in quantity rather conveniently from foreskin, preferably fetal foreskin, or, alternatively, any appropriate cadaver organ. Fetal cells, including fibroblast-like cells and chondrocyte progenitor cells, may be obtained from umbilical cord or placenta tissue or umbilical cord blood.
  • stromal cells from a variety of sources may be used in the claimed methods, it is preferable that, for implantation in vivo, the stromal cells be derived from the individual who is to receive the implant or from cells of fetal origin which may be viewed as "universal donors" so as to minimize the risk of immunological rejection of the implant.
  • the stromal cells may be readily isolated by disaggregating an appropriate tissue which is to serve as the source of the cells. This may be readily accomplished using techniques known to those skilled in the art.
  • the tissue can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that weaken the connections between neighboring cells making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage.
  • Enzymatic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination.
  • the suspension can be fractionated into subpopulations from which chondrocytes, fibroblasts and/or other stromal cells and/or elements can be obtained.
  • This also may be accomplished using standard techniques for cell separation including but not limited to cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection) , separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter- streaming centrifugation) , unit gravity separation, counter current distribution, electrophoresis and fluorescence- activated cell sorting.
  • clonal selection and cell separation techniques see Freshney, supra, Ch. 11 and 12, pp. 137-168.
  • chondrocytes For example, the isolation of chondrocytes, chondrocyte progenitors, fibroblasts or fibroblast-like cells is carried out as follows: fresh human cartilage tissue can be thoroughly washed and minced in Hanks balanced salt solution (HBSS) in order to remove serum. The minced tissue is incubated from 1-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the dissociated cells are suspended, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown.
  • HBSS Hanks balanced salt solution
  • the isolated stromal cells can then be grown to confluency, lifted from the confluent culture and administered to the cartilage defect site in vivo (see, e.g., Naughton et al.. 1987, J. Med. 18(3&4) :219-250) .
  • Fibroblast- like cells may also be isolated from human umbilical cords (33-44 weeks) .
  • Fresh tissues may be minced into pieces and washed with medium or snap-frozen in liquid nitrogen until further use.
  • the umbilical tissues may be disaggregated as described above.
  • chondrocytes or chondrocyte progenitor cells have been isolated, their population can be expanded mitotically in vitro in order to obtain the cell preparation for implantation.
  • Methods for the selection of the most appropriate culture medium, medium preparation, and cell culture techniques are well known in the art and are described in a variety of sources, including Doyle et al.. (eds.), 1995, Cell & Tissue Culture: Laboratory Procedures. John Wiley & Sons, Chichester; and Ho and Wang (eds.), 1991, Animal Cell Bioreactors. Butterworth-Heinemann, Boston.
  • the cells should be transferred or "passaged" to fresh medium when they reach an appropriate density, such as 3 to 6.5 x 10 /cm 2 , or, for example, when they reach a defined percentage of confluency on the surface of a culture dish.
  • an appropriate density such as 3 to 6.5 x 10 /cm 2
  • the stromal cells may stick to the walls of the culture vessel where they can continue to proliferate and form a confluent monolayer. This should be prevented or minimized, for example, by transferring a portion of the cells to a new culture vessel having fresh medium, since the presence of a confluent monolayer in the culture vessel will tend to "shut down" the growth of cells in the culture.
  • Removal of the confluent monolayer or transfer of a portion of the cells to fresh media in a new vessel will usually restore proliferative activity of the cells. Such removal or transfer should be done in any culture vessel which has a monolayer exceeding about 25% confluency.
  • the liquid culture can be agitated, for example, on an orbital shaker or in roller bottles, to prevent or minimize the cells from sticking to the vessel walls.
  • stromal cells may be maintained or stored in cell "banks" comprising either continuous in vitro cultures of cells requiring regular transfer, or, preferably, cells which have been cryopreserved. Cryopreservation of the cells may be carried out according to known methods, such as those described in Doyle et al.. 1995, supra.
  • cells may be suspended in a "freeze medium” such as, for example, culture medium further comprising 20% FBS and 9% dimethylsulfoxide (DMSO) , with or without 5-10% glycerol, at a density, for example, of about 4-10 x 10 6 cells/ml.
  • DMSO dimethylsulfoxide
  • the cells are dispensed into glass or plastic ampoules (Nunc) which are then sealed and transferred to the freezing chamber of a programmable freezer.
  • the optimal rate of freezing may be determined empirically. For example, a freezing program that gives a change in temperature of -l°C/min through the heat of fusion may be used.
  • a freezing program that gives a change in temperature of -l°C/min through the heat of fusion may be used.
  • Once the ampoules have reached -180°C they are transferred to a liquid nitrogen storage area. Cryopreserved cells can be stored for a period of years, though they should be checked at least every 5 years for maintenance of viability.
  • the cryopreserved cells constitute a bank of cells, portions of which can be "withdrawn” by thawing and then used to produce new cartilage tissue as needed.
  • Thawing should generally be carried out rapidly, for example, by transferring an ampoule from liquid nitrogen to a 37°C water bath.
  • the thawed contents of the ampoule should be immediately transferred under sterile conditions to a culture vessel containing an appropriate medium such as RPMI 1640 conditioned with 10% FBS and 5% ES.
  • an appropriate medium such as RPMI 1640 conditioned with 10% FBS and 5% ES. It is advisable that the cells in the culture medium be adjusted to an initial density of about 3-6 x 10 5 cells/ml so that the cells can condition the medium as soon as possible, thereby preventing a protracted lag phase.
  • the cells may be examined daily, for example, with an inverted microscope to detect cell proliferation, and subcultured as soon as they reach an appropriate density.
  • chondrocytes In addition to chondrocytes, chondrocyte progenitors, fibroblasts or fibroblast-like cells, other cells may be added to the cell preparation for implantation in vivo, which other cells aid in the production of the stromal matrix on the scaffold of the invention.
  • other cells found in loose connective tissue may be seeded along with chondrocytes or fibroblasts.
  • Such cells include but are not limited to endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, etc.
  • These stromal cells may readily be derived from appropriate organs including umbilical cord or placenta or umbilical cord blood using methods known in the art such as those discussed above.
  • the stromal cell preparation may further comprise one or more other components, including selected extracellular matrix components, such as one or more types of collagen known in the art, as well as growth factors and/or drugs.
  • Growth factors which may be usefully incorporated into the cell preparation include one or more tissue growth or stimulatory factors known in the art or to be identified in the future, including but not limited to any member of the TGF-3 family, BMPs that stimulate cartilage formation, e.g., BMP-2, BMP-12, and BMP-13, factors that stimulate migration of stromal cells and/or matrix deposition, insulin-like growth factor (IGF)-I and -II, growth hormone, etc.
  • IGF insulin-like growth factor
  • Drugs which may be usefully incorporated into the cell preparation include anti-inflammatory compounds such as non-steroidal anti-inflammatories, immunosuppressants such as the cyclosporins, as well as local anesthetics.
  • Other components may also be included in the preparation, including but not limited to any of the following: (1) buffers to provide appropriate pH and isotonicity; (2) lubricants; (3) viscous materials to retain the cells at or near the site of administration, including, for example, alginates, agars and plant gums; and (4) other cell types that may produce a desired effect at the site of administration, such as, for example, enhancement or modification of the formation of cartilage tissue or its physicochemical characteristics, or support for the viability of the cells, or inhibition of inflammation or rejection.
  • the stromal cells may be obtained from the patient's own tissues or from a fetal source ("universal donor") .
  • the growth of cells on the three-dimensional scaffold may be further enhanced by including in or on the framework or coating the framework with proteins (e.g., collagens, elastic fibers, reticular fibers) , glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4- sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc.), and/or other bioactive materials such as growth factors.
  • proteins e.g., collagens, elastic fibers, reticular fibers
  • glycoproteins e.g., glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4- sulfate, chondroitin-6-sulfate, dermatan
  • growth regulatory or stimulatory factors including, but not limited to, TGF-/S and ascorbate, or BMPs that stimulate cartilage formation such as BMP-2, BMP-12, and BMP-13 may be added to the implantation site before, during or after implantation of either the scaffold and/or the periosteal/perichondrial tissue, in order to promote the production of new cartilage at the site.
  • growth regulatory factors can be administered to the site at the time of administration of the stromal cells, either as a separate preparation or, as noted supra. as part of the stromal cell preparation.
  • the stromal cells may be genetically engineered to produce growth factors such as TGF- / S as well as other biological factors such as factors that stimulate chondrogenesis or the migration of chondrogenic and other stromal cells to the scaffold of this invention.
  • the stromal cells administered to or in combination with the scaffold and periosteal/perichondrial tissue of the invention can also be genetically engineered to produce gene products that promote the successful production or repair of cartilage at a defect site and/or for use in gene therapies.
  • the stromal cells can be genetically engineered to express anti-inflammatory factors, e.g., anti-GM-CSF, anti-TNF, anti-IL-1, anti-IL-2, etc., to reduce the risk of rejection of the implant or to reduce the risk of degenerative changes in the cartilage due to rheumatoid disease or other inflammatory reactions.
  • the stromal cells can be genetically engineered to express peptides or polypeptides corresponding to the idiotype of neutralizing antibodies for granulocyte-macrophage colony stimulating factor (GM-CSF), TNF, IL-1, IL-2, or other inflammatory cytokines.
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • TNF granulocyte-macrophage colony stimulating factor
  • IL-1 has been shown to decrease the synthesis of proteoglycans and collagens type II, IX, and XI (Tyler et al. , 1985, Biochem. J. 227:869-878; Tyler et al.. 1988, Coll. Relat. Res. 82: 393-405; Goldring et al.. 1988, J. Clin. Invest.
  • TNF also inhibits synthesis of proteoglycans and type II collagen, although it is much less potent than IL-1 (Yaron, I., et al.. 1989, Arthritis Rheum. 32:173-180; Ikebe, T. , et al.. 1988, J. Immunol. 140:827-831; and Saklatvala, J. , 1986, Nature 322:547-549).
  • the presence of the anti- inflammatory gene products can bring about amelioration of immunological rejection or the inflammatory reactions associated with rheumatoid or joint disease.
  • the stromal cells can be genetically engineered to express a gene which would exert a therapeutic effect, e.g., in the production of TGF- ⁇ to stimulate cartilage production, or other factors such as BMP- 13 to promote chondrogenesis and/or prevent bone formation or stimulatory factors that promote migration of stromal cells and/or matrix deposition.
  • the stromal cells can be genetically engineered to express a gene for which a patent is deficient.
  • genes that prevent or ameliorate symptoms of various types of rheumatoid or joint diseases may be underexpressed or down-regulated under disease conditions. Specifically, expression of genes involved in preventing inflammatory reactions in rheumatoid or joint diseases may be down-regulated.
  • the activity of gene products may be diminished, leading to the manifestations of some or all of the above pathological conditions and eventual development of symptoms of rheumatoid or joint diseases.
  • the level of gene activity may be increased by either increasing the level of gene product present or by increasing the level of the active gene product present at the defect site.
  • Target gene refers to a gene involved in rheumatoid or joint diseases in a manner by which modulation of the level of target gene expression or of target gene product activity may act to ameliorate symptoms of rheumatoid or joint diseases by preventing resorption of cartilage and production of inflammatory mediators by chondrocytes.
  • patients may be treated by gene replacement therapy by means of the stromal cells administered according to the methods of this invention.
  • replacement or repaired cartilage may be designed specifically to meet the requirements of an individual patient; for example, the stromal cells may be genetically engineered to regulate one or more genes; or the regulation of gene expression may be transient or long-term; or the gene activity may be non- inducible or inducible.
  • the gene encoding the human complement regulatory protein, which prevents rejection of an implant by the host may be inserted into human fibroblasts. McCurry et al.. 1995, Nature Medicine 1:423- 427.
  • the stromal cells used in the methods of the invention can also be genetically engineered to "knock out” expression of factors that promote inflammation or rejection at the implant site. Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are discussed below. "Negative modulation”, as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level and/or activity of the target gene product in the absence of the modulatory treatment.
  • the expression of a gene native to stromal cell can be reduced or knocked out using a number of techniques, for example, expression may be inhibited by inactivating the gene completely (commonly termed "knockout") using standard homologous recombination techniques.
  • an exon encoding an important region of the protein is interrupted by a positive selectable marker (for example neo) , preventing the production of normal mRNA from the target gene and resulting in inactivation of the gene.
  • a gene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the sequences intervening the two regions can be deleted. Mombaerts et al.. 1991, Proc. Nat. Acad. Sci. U.S.A. 88:3084-3087.
  • Antisense and ribozyme molecules which inhibit expression of the target gene can also be used in accordance with the invention to reduce the level of target gene activity.
  • antisense RNA molecules which inhibit the expression of major histocompatibility gene complexes (HLA) have been shown to be most versatile with respect to immune responses.
  • appropriate ribozyme molecules can be designed as described, e.g. , by Haseloff et al. , 1988, Nature 334:585-591; Zaug et al.. 1984, Science 224:574-578; and Zaug and Cech, 1986, Science 231:470-475.
  • triple helix molecules can be utilized in reducing the level of target gene activity.
  • the expression of IL-1 can be knocked out in the chondrocytes to reduce the risk of resorption of cartilage and production of inflammatory mediators by the chondrocytes.
  • the expression of MHC class II molecules can be knocked out in order to reduce the risk of rejection of the implant.
  • a recombinant DNA construct or vector containing the gene of interest may be constructed and used to transform or transfect the stromal cells of the invention.
  • Such transformed or transfected cells that carry the gene of interest, and that are capable of expressing said gene are selected and clonally expanded in culture.
  • Methods for preparing DNA constructs containing the gene of interest, for transforming or transfecting cells, and for selecting cells carrying and expressing the gene of interest are well-known in the art. See, for example, the techniques described in Maniatis et al. , 1989, Molecular Cloning.
  • the cells can be engineered using any of a variety of vectors including, but not limited to, integrating viral vectors, e.g. , retrovirus vector or adeno-associated viral vectors; or non-integrating replicating vectors, e.g., papillo a virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors.
  • integrating viral vectors e.g. , retrovirus vector or adeno-associated viral vectors
  • non-integrating replicating vectors e.g., papillo a virus vectors, SV40 vectors, adenoviral vectors
  • replication-defective viral vectors e.g., papillo a virus vectors, SV40 vectors, adenoviral vectors
  • replication-defective viral vectors e.g., papillo a virus vectors, SV40 vectors, adenoviral vectors
  • replication-defective viral vectors e.
  • Hosts cells are preferably transformed or transfected with DNA controlled by, i.e., in operative association with, one or more appropriate expression control elements such as promoter or enhancer sequences, transcription terminators, polyadenylation sites, among others, and a selectable marker.
  • engineered cells may be allowed to grow in enriched media and then switched to selective media.
  • the selectable marker in the foreign DNA confers resistance to the selection and allows cells to stably integrate the foreign DNA as, for example, on a plasmid, into their chromosomes and grow to form foci which, in turn, can be cloned and expanded into cell lines. This method can be advantageously used to engineer cell lines which express the gene product.
  • any promoter may be used to drive the expression of the inserted gene.
  • viral promoters include but are not limited to the CMV promoter/enhancer, SV40, papillo avirus, Epstein-Barr virus, elastin gene promoter and ⁇ -globin.
  • the control elements used to control expression of the gene of interest should allow for the regulated expression of the gene so that the product is synthesized only when needed in vivo.
  • constitutive promoters are preferably used in a non-integrating and/or replication-defective vector.
  • inducible promoters could be used to drive the expression of the inserted gene when necessary.
  • Inducible promoters can be built into integrating and/or replicating vectors.
  • inducible promoters include, but are not limited to, metallothionien and heat shock protein.
  • transkaryotic as used herein, suggests that the nuclei of the implanted cells have been altered by the addition of DNA sequences by stable or transient transfection.
  • the stromal cells are engineered to express such gene products transiently and/or under inducible control during the post ⁇ operative recovery period, or as a chimeric fusion protein anchored to the stromal cells, for example, as a chimeric molecule composed of an intracellular and/or transmembrane domain of a receptor or receptor-like molecule, fused to the gene product as the extracellular domain.
  • a chimeric fusion protein anchored to the stromal cells for example, as a chimeric molecule composed of an intracellular and/or transmembrane domain of a receptor or receptor-like molecule, fused to the gene product as the extracellular domain.
  • the preparation of stromal cells can be administered either before, during or after implantation of the scaffold and/or the periosteal/perichondrial tissue.
  • the cells can be seeded into the defect site before implantation of either the scaffold or the periosteal/perichondrial tissue.
  • the stromal cells can be administered to the site after either one or both of the scaffold and tissue have been implanted; e.g. , by injection into the site after suturing the periosteal/perichondrial tissue to the scaffold.
  • the cells are seeded between the periosteal/perichondrial tissue and the scaffold at the defect site.
  • the cells are seeded directly into the scaffold.
  • the stromal cells can also be seeded into the degraded cartilage, e.g., into the surrounding cells or directly into the cartilage wall.
  • the stromal cells act therein to induce the migration of stromal cells from the degraded cartilage to the implant.
  • the stromal cells can be seeded by any means that allows administration of the cells to the defect site, e.g., by injection.
  • administration of the cells e.g., by injection.
  • such injection can be achieved by any means that maintains the viability of the cells, e.g., via syringe or more preferably, via an arthroscope.
  • the number of cells administered can range from approximately 1 x IO 6 to 30 x 10 6 stromal cells.
  • the defect site is surgically sealed.
  • the stromal cells to be seeded are surgically obtained from the patient, e.g., from ear cartilage and/or bone marrow, in a separate surgical procedure, cultured in vitro to obtain an appropriate amount of cells and administered to the patient at the time of a second surgery wherein the scaffold and periosteal/perichondrial tissue are implanted.
  • the periosteal/perichondrial tissue is preferably surgically obtained from the patient at the time that the scaffold is implanted, although the tissue can be cultured ij vitro prior to implantation (see, e.g., O'Driscoll et al. , 1994, J. Bone and Joint Surg. 76-A:1042-1051) .
  • the stromal cells from the periosteal/perichondrial tissue and from the added stromal preparation populate the scaffold structure to form a stromal matrix that resembles the in vivo microenvironment of cartilage tissue, allowing for the production of new cartilage at the defect site.
  • the stromal cells administered to the site provide important biological factors that promote chondrogenesis and the migration of stromal cells, whether from the implanted tissue, the in vivo cartilage environment of the defect or from the stromal preparation, to the scaffold, thus promoting the production of a living stromal tissue that provides the support, growth factors, and regulatory factors necessary to sustain long- term active proliferation of the stromal cells in vivo.
  • the stromal cells may additionally provide factors that promote the deposition of the living stromal matrix at the defect site. The proliferating cells mature and segregate properly within the matrix to form new cartilage tissue at the defect site in vivo.
  • the successful repair or replacement of damaged cartilage can be enhanced if the new cartilage tissue can be fixed in place at the site of repair. Post-implantation movement may cause the new cartilage tissue to become dislodged from the site if a pro-active fixation technique is not employed.
  • Various methods can be used to fix the new cartilage tissue in place, including: patches derived from a bioresorbable polymer or biocompatible tissues, which can be placed over the site and sutured; bioabsorbable sutures or other fasteners, e.g., pins, staples, tacks, screws, anchors, glues, e.g., fibrin glue; non-absorbable fixation devices, e.g., sutures, pins, screws and anchors; adhesives; and the use of interference fit geometries.
  • the methods of this invention are useful to replace or augment existing cartilage tissue in vivo, to introduce new or altered cartilage tissue or to join together biological tissues or structures.
  • the present methods find use in a number of specific areas.
  • the evaluation of internal derangements of articular cartilage in several articulations, including the knee, hip, elbow, ankle and the glenohumeral joint has been made possible by arthroscopic techniques.
  • Such derangements can be caused by physical trauma to the cartilage, by various connective tissues diseases and/or by increased age of the individual.
  • Arthroscopic surgery has become increasingly popular as well as successful, e.g., numerous small cutting tools, 3 to 4mm in diameter can be used in the knee.
  • Triangulation in which the operating instruments are brought into the visual field provided by the arthroscope, requires multiple portals of entry; alternatively, the cutting tools can be passed through a channel in the arthroscope itself in which case only one opening in the joint is necessary (Jackson, R.W. , 1983, J. Bone Joint Surg. [AM] 65:416) .
  • Selective removal of damaged or deteriorated cartilage via arthroscopic surgery results in cartilage loss, which can be repaired using the methods of the present invention.
  • the present methods can also be employed to repair or augment cartilage loss that results from major reconstructive surgery for different types of joints, the procedures for which surgery have been described in Resnick, D. , and Niwayama, G.
  • the present invention is useful for the production or repair of cartilage in vivo in the treatment of degenerative connective tissue diseases, such as rheumatoid or osteoarthritis, or in the treatment of physical trauma, wherein cartilage is damaged or lost.
  • a three-dimensional scaffold comprising felt derived from a PGA multifilament yarn was generated.
  • the yarn is commercially available from Davis and Geek/Sherwood Medical under the trade name DexonTM.
  • the particular type of DexonTM yarn is 56/123.
  • the DexonTM yarn is processed into felt via standard textile processing techniques.
  • the felt sheet which has a porosity of 97%, a density of 45 g/cc and a thickness of 2-7 mm after processing, is cut into the appropriate size for implantation in vivo. Standard serialization techniques (radiation or ethylene oxide gas) for medical products are used to sterilize the felt.
  • the felt can be implanted dry or pre-soaked using DMEM medium containing 10% fetal bovine serum, 2 mM L-glutamine, non- essential amino acids, 50 mg/ml proline, 1 mM sodium pyruvate, 35 ⁇ g/ml gentamicin sulfate and 50 ⁇ g/ml ascorbate. 5.2. PREPARATION OF PERIOSTEAL/ PERICHONDRIAL TISSUE
  • Periosteal tissue to be used in the methods of this invention is obtained by removing, e.g. , by arthroscopy, a section of periosteum from the tibia or femur of the patient.
  • Perichondrial tissue can be obtained by removing a section of the perichondrium from the rib of the patient.
  • the periosteal/perichondrial tissue is of such size and/or shape so as to correspond to the defect site.
  • the excision of the periosteal/perichondrial tissue from the patient is preferably performed at the time of the implantation of the scaffold.
  • cartilage slices e.g., 300- 500 mg by weight
  • cartilage slices can be obtained by arthroscopy from a minor load-bearing area of a joint such as the femoral condyle of the knee.
  • the cartilage tissue is placed in a chilled sterile solution of sodium chloride (0.9 % weight per volume) and cells are harvested within 2-5 hr by mincing and washing in Ham's F12 medium (Gibco Labs, Grand Island, NY) containing HEPES buffer (10 mmol/1) , gentamicin sulfate (50 ⁇ g/ml) , amphotericin B (2 ⁇ g/ml) , and L-ascorbic acid (50 ⁇ g/ml) .
  • the cells can similarly be harvested using the complete DMEM medium described in Section 5.1, supra.
  • the cartilage is then digested with collagenase (0.2% weight/volume) for approximately 16 hr and the cells are filtered through a nylon mesh, washed with medium and resuspended in medium supplemented with the patient's serum.
  • the cells are seeded at a density of approximately 1 X 10 6 cells per T-150 flask and cultured in an incubator at 37°C, 5% C0 2 . Cells are passed at confluence every 5-7 days.
  • chondrocyte progenitor cells can be obtained using methods similar to those described in Wakitani et al.. 1994, J. Bone and Joint Surg. 76-A (No.
  • the cells are then trypsinized (0.25% trypsin, 1 mM EDTA) for 5 min, to release the cells from the culture dish.
  • Chondrocyte progenitor cells can also be obtained from periosteum harvested, e.g., from the tibia or femur. The periosteal tissue is incubated with 0.25% collagenase in
  • the stromal cell preparation Prior to administration in vivo, the stromal cell preparation is suspended by treatment with trypsin, centrifuged and the pellet washed in culture medium
  • the cell preparation is then aspirated into a 1 ml tuberculin syringe with a 1.2 mm needle for administration to the implant site.
  • the cartilage defect site is cleaned/sterilized prior to implantation by scrubbing or rinsing with a sterilizing solution, e.g., iodine surgical scrub, e.g., 0.75% titratable iodine, or 0.9% sodium chloride solution.
  • a sterilizing solution e.g., iodine surgical scrub, e.g., 0.75% titratable iodine, or 0.9% sodium chloride solution.
  • __ can also be enzymatically treated to degrade the cartilage surface area of the defect by treatment with trypsin in the range of 0.1-100 Units/ml for 1-30 min depending upon the concentration of enzyme used.
  • the felt scaffold as described above, tailored to fit the defect site, is placed into the site and the periosteal tissue as described above is placed on top of the felt scaffold with the cambium layer of the periosteum facing toward the scaffold and into the defect.
  • the periosteal tissue may optionally be sewn onto the ends of the felt scaffold using resorbable vicryl sutures.
  • the flap is also sutured to the surrounding cartilage at the defect site.
  • the stromal cells of the preparation described above can be administered, e.g., by an arthroscope, into the defect site.
  • an arthroscope For a defect 5 mm thick and 1.5 cm in diameter, a total of 14 x 10 6 stromal cells in 200 ⁇ l of a nutrient-based solution such as DMEM (with or without serum) is administered.
  • the stromal cells are injected between the periosteal tissue and the scaffold.
  • the defect site is closed in layers with sutures and the site bandaged.
  • a post-surgical analgesic may be administered.

Abstract

The present invention relates to methods of making and/or repairing cartilage in vivo comprising implanting into a patient, at a site of cartilage damage or loss, a biocompatible, non-living three-dimensional scaffold or framework structure in combination with periosteal/perichondrial tissue that can be used to hold the scaffold in place and provides a source of chondrocyte progenitor cells, chondrocytes and other stromal cells for attachment to the scaffold in vivo. In addition, a preparation of cells that can include chondrocytes, chondrocyte progenitor cells or other stromal cells is administered, either before, during or after implantation of the scaffold and/or the periosteal perichondrial tissue; the cells are administered directly into the site of the implant in vivo and promote the induction of factors that enhance chondrogenesis and the migration of chondrocytes, progenitor cells and other stromal cells from the adjacent in vivo environment into the scaffold for the production of new cartilage at the site of implantation.

Description

METHOD FOR MAKING AND /OR REPAIRING CARTILAGE
1. INTRODUCTION
The present invention relates to methods for making and/or repairing cartilage in vivo. More specifically, the invention relates to methods of making and/or repairing cartilage comprising implanting into a patient, at a site of cartilage damage or loss, a biocompatible, non-living three- dimensional scaffold or framework structure, in combination with periosteal/perichondrial tissue, and administering a preparation of chondrocytes and/or other stromal cells, such as chondrocyte progenitor cells, to the site of the implant before, during or after implantation of the scaffold and/or the periosteal/perichondrial tissue. The periosteal/perichondrial tissue can be used to hold the scaffold in place at the site of implantation and also provides a source of stromal cells, e.g., chondrocytes and/or chondrocyte progenitor cells, for attachment to the scaffold in vivo. The preparation of stromal cells seeded directly into the implantation site in vivo provides not only a readily-accessible source of chondrocytes and/or other stromal cells for attachment to the scaffold but also provides a rapid and efficient means of inducing chondrogenesis as well as migration of stromal cells from the surrounding in vivo environment to the scaffold via factors produced by the stromal cells of the preparation. Additionally, the seeded stromal cells can be genetically engineered to express gene products beneficial to growth, implantation and/or amelioration of disease conditions. The methods of the invention therefore result in the efficient production of new cartilage in vivo.
The methods of this invention are useful in the production/repair of articular cartilage in patients suffering from degenerative connective tissue diseases such as rheumatoid and/or osteoarthritis as well as in patients who have cartilage defects due to trauma. The methods of this invention can be used to replace or augment existing cartilage tissue, to introduce new or altered tissue or to join together biological tissues or structures.
2. BACKGROUND OF THE INVENTION There are various types of cartilage, e.g., articular or hyaline cartilage, elastic cartilage and fibrocartilage. Articular cartilage is found at the articular surfaces of bones, e.g., in the joints, and is responsible for providing the smooth gliding motion characteristic of moveable joints. Articular cartilage is firmly attached to the underlying bones and measures less than 5mm in thickness in human joints, with considerable variation depending on joint and site within the joint. In addition, articular cartilage is aneural, avascular, and alymphatic. In adult humans, this cartilage derives its nutrition by a double diffusion system through the synovial membrane and through the dense matrix of the cartilage to reach the chondrocyte, the cells that are found in the connective tissue of cartilage.
In fact, articular cartilage consists of highly specialized chondrocyte cells surrounded by a dense extracellular matrix consisting mainly of type II collagen, proteoglycan and water. While the biochemical composition of articular cartilage includes up to 65-80% water (depending on the cartilage) , the collagen component of the cartilage is the most prevalent organic constituent. The collagen (mainly type II) accounts for about 15-25% of the wet weight or about half the dry weight, except in the superficial zone where it accounts for most of the dry weight. Its concentration is usually progressively reduced with increasing depth from the articular surface. The proteoglycan content accounts for up to 10% of the wet weight or about a quarter of the dry weight. Proteoglycans consist of a protein core to which linear sulfated polysaccharides are attached, mostly in the form of chondroitin sulfate and keratin sulfate. In addition to type II collagen, articular collagen contains several other collagen types (IV, V, IX and X) with distinct structures. There are a variety of interactions between these individual macromolecules, which include both noncovalent associations between proteoglycans and collagens, and covalent bonds between different collagen species. Resistance of the extracellular matrix to water flow gives cartilage its ability to dispense high joint loads. It absorbs shock and minimizes stress on subchondral bone (Mow et al.. 1984, J. Biomech. 17:377-394). However, adult cartilage and bone have a limited ability of repair; thus, damage to cartilage produced by disease, such as rheumatoid and/or osteoarthritis, or trauma can lead to serious physical deformity and debilitation. Furthermore, as human articular cartilage ages, its tensile properties change. The superficial zone of the knee articular cartilage exhibits an increase in tensile strength up to the third decade of life, after which it decreases markedly with age as detectable damage to type II collagen occurs at the articular surface. The deep zone cartilage also exhibits a progressive decrease in tensile strength with increasing age, although collagen content does not decrease. These observations indicate that there are changes in mechanical and, hence, structural organization of cartilage with aging that, if sufficiently developed, can predispose cartilage to traumatic damage. In osteoarthritic cartilage, there is excessive damage to type II collagen, resulting in crimping of collagen fibrils. In rheumatoid arthritis, the combined actions of free radicals and proteinases released from polymorpholeukocytes cause much of the damage seen at the articular surface (Tiku et al.. 1990, J. Immunol. 145:690-696). Induction of cartilage matrix degradation and proteinases by chondrocytes is probably induced primarily by interleukin-1 (IL-1) or tumor necrosis factor-α (TNF- ) (Tyler, 1985, Bioche . J. 225:493- 507) .
The current therapy for damage or loss of cartilage is replacement with a prosthetic material, for example, silicone for cosmetic repairs, or metal alloys for joint realignment. Implantation of prosthetic devices is usually associated with loss of underlying tissue and bone without recovery of the full function allowed by the original cartilage. Serious long-term complications associated with the presence of a permanent foreign body can include infection, erosion and instability. Use of sterilized bone or bone powder or surgical steel seeded with bone cells that are eventually implanted have been largely unsuccessful because of the non-degradable nature of the cell support. According to one procedure, fibroblasts are exposed in vitro for a minimum of three days to a soluble bone protein capable of stimulating a chondrogenic response. The activated fibroblasts are then transferred in vivo by combining them with a biodegradable matrix or by intra-articular injection or attachment to allografts or prosthetic devices. The disadvantage of this method is that chondrogenesis is not allowed to develop in the short-term cultures and there is an unduly heavy reliance on cartilage synthesis by the exposed fibroblasts at the implant site. See Caplan, A., United States Pat. No. 4,609,551, issued September 2, 1986. United States Pat. No. 5,041,138 to J.P. Vacanti et al.. issued August 20, 1991, describes the growth of cartilaginous structures by seeding chondrocytes on biodegradable matrices in vitro for subsequent implantation in vivo. United States Patents Nos. 5,197,985 and 5,226,914, to Caplan et al.. issued March 30, 1993 and July 13, 1993, respectively, relate to culturing marrow-derived mesenchymal stem cells in vitro in the presence of growth factors, applying these cells to a carrier, e.g., a porous ceramic vehicle, to promote round cell morphology, and implanting the carrier containing the cells into damaged articular cartilage. Finally, United
States Pat. No. 4,520,821, to Schmidt et al.. issued June 4, 1985 relates to a method of growing a biological tissue correction structure using a bioabsorbable mesh or gauze. According to a preferred embodiment, biological tissue is removed from a defective area of the body and grown in vitro on a bioabsorbable mesh and the tissue/mesh structure is placed into the area of the defect in vivo for a time long enough to allow the mesh to be completely bioabsorbed. In all of the approaches cited above, cells are applied to a carrier in vitro for implantation into the site of a cartilage lesion. United States Pat. No. 4,846,835 to Grande et al.. issued July 11, 1989, also describes a grafting technique for promoting articular cartilage healing by culturing autologous chondrocytes in vitro and seeding the chondrocytes on a collagen matrix in vitro. Grande further describes implanting the chondrocyte/collagen matrix into a cartilage lesion and mechanically fixing the graft by suturing a periosteal flap to the cartilage. Grande teaches that the periosteum has very little chondrogenic potential and that the periosteal flap utilized therein could have been substituted by a resorbable biocompatible polymer to achieve the same fixation purpose. While von Schroeder et al.. 1991, J. Biomed. Mat. Res. 25: 329-339, relates to periosteal grafts in combination with a polylactic acid matrix for the repair of full-thickness osteochondral defects in animals, Messner, 1994, Biomaterials 15 (No. 3): 223-230 describes experiments in which periosteal grafts in combination with Teflon and Dacron felts for the repair of full-thickness osteochondral defects were unsatisfactory in achieving normal cartilage repair in animals. O'Driscoll et al. , 1986, J. Bone and Joint Surg. 68-A (No. 7): 1017-1035; O'Driscoll et al. , 1985, 31st Annual ORS, p. 292, Las Vegas, Nevada, Jan. 21-24, 1985; Rubak, 1982, Acta. Orthop. Scand. 53: 175-180; and Messner et al.. 1993, Biomaterials 14 (No. 7): 513-521 relate to the use of free autogenous periosteal grafts that are placed directly into full-thickness articular defects for the repair of such defects in animals.
In addition, Thoma et al.. 1993, Plast. Reconstr. Surg. 91 (No. 2): 307-315 and Bruns et al. , 1992, Virchows. Arch. A. Pathol. Anat. Histopathol. 421 (No. 1): 1-8 relate to the use of autogenous perichondrial grafts for the repair of full-thickness articular defects in animals. Thoma noted marked degenerative changes resembling osteoarthritis in the perichondrial grafts and concluded that spontaneous repair of such large defects may result in a more normal new articular cartilage than the perichondrial grafts attempted. Bruns noted hyaline-like cartilage in non-weight-bearing areas of the defect, but not in weight-bearing areas. Finally, Bean et al. , 1994, ORL J. Otorhinolaryngol. Relat. Spec. 56: 224- 229 reports the use of a composite graft of demineralized bovine bone matrix and autogenous perichondrium for the reconstruction of the anterior laryngeal wall in rabbits.
3. SUMMARY OF THE INVENTION The present invention relates to methods of making and/or repairing cartilage in vivo comprising implanting into a patient, at a site of cartilage damage or loss, a biocompatible, non-living three-dimensional scaffold or framework structure in combination with periosteal/perichondrial tissue that can be used to hold the scaffold in place and provides a source of chondrocyte progenitor cells, chondrocytes and other stromal cells for attachment to the scaffold in vivo. In addition, a preparation of cells that can include chondrocytes, chondrocyte progenitor cells or other stromal cells is administered, either before, during or after implantation of the scaffold and/or the periosteal/perichondrial tissue; the cells are administered directly into the site of the implant in vivo and promote chondrogenesis and the production of factors that induce the migration of chondrocytes, progenitor cells and other stromal cells from the adjacent in vivo environment into the scaffold for the production of new cartilage at the site of implantation.
More specifically, the three-dimensional scaffold contains interstitial spaces into which progenitor cells, chondrocytes and other stromal cells from the adjacent in vivo environment, including the implanted periosteal/perichondrial tissue, migrate for attachment and growth on and within the scaffold structure. The preparation of stromal cells seeded in combination with the scaffold and periosteal/perichondrial tissue provides a ready source of chondrocytes and other stromal cells which produce biological factors that promote chondrogenesis and the migration of stromal cells from, e.g., the periosteal/perichondrial tissue to the scaffold for attachment and/or differentiation thereon. The stromal cell preparation also provides a direct source of stromal cells, e.g., chondrocytes and/or progenitor cells, that are capable of migrating into the scaffold and attaching thereto. The stromal cells in the scaffold, whether derived from the periosteal/perichondrial tissue, from the exogenous stromal cell preparation or from the in vivo environment adjacent to the implant site, grow on the scaffold to form a cellular matrix and provide the support, growth factors and regulatory factors required for cartilage formation at a cartilage defect site in vivo. The methods of this invention thus result in the production of new cartilage in vivo at the implant site.
In a preferred embodiment of the methods of the invention, the periosteal/perichondrial tissue is placed over the implanted scaffold at the site of cartilage damage or loss ("the defect site") and affixed, e.g. , by sutures, to that site, thus holding the scaffold in place. According to a further preferred embodiment, the scaffold is composed of a biodegradable material such that, upon successful engraftment, the scaffold structure is completely absorbed in vivo, resulting in new cartilage having no foreign, non¬ living material encompassed within it.
According to another preferred embodiment, the preparation of chondrocytes and/or other stromal cells is administered in vivo to the site of the implant after the scaffold and periosteal/perichondrial tissue have been implanted. In yet a further embodiment of the invention, bioactive agents such as cellular growth factors (e.g. , TGF- β) , factors that stimulate chondrogenesis (e.g., bone morphogeniσ proteins (BMPs) that promote cartilage formation) , factors that stimulate migration of stromal cells and/or matrix deposition, anti-inflammatories or immunosuppressants, are included at the implantation site. For example, these factors can be incorporated into the scaffold material to provide for release at the site of implantation; the scaffold can also be comprised of, or coated with, one or more of these bioactive agents.
Alternatively, the factor(s) can be administered into or adjacent to the scaffold, either before, during or after seeding of the stromal cells, e.g., the bioactive agent(s) can be administered to the site, either as a separate preparation or as part of the stromal cell preparation. In addition, the stromal cells seeded at the defect site can be genetically engineered to express the genes for these bioactive agents, e.g. , specific types of TGF-β such as TGF-S1 or specific types of BMPs such as BMP-13. Exposure of the defect site to these bioactive agents promotes the successful and/or improved production of new cartilage and/or improves the success of implantation, for example, by reducing the risk of rejection or inflammation associated with the implant. For example, according to one embodiment of the invention, the stromal cells can be genetically engineered to express an i-inflammatory gene products to ameliorate the effects of degenerative diseases like rheumatoid arthritis which result in cartilage damage due to inflammatory reactions; e.g. , the stromal cells can be engineered to express peptides or polypeptides corresponding to the idiotype of neutralizing antibodies for granulocyte- macrophage colony stimulating factor (GM-CSF) , tumor necrosis factor (TNF) , interleukin-2 (IL-2) , or other inflammatory cytokines and mediators.
According to another embodiment, the stromal cells can be genetically engineered to express tissue factors that enhance migration of stromal cells from the adjacent in vivo environment into the scaffold at the implantation site. Preferably, the cells are engineered to express such gene products transiently and/or under inducible control during the post-operative recovery period, or as a chimeric fusion protein anchored to the stromal cell, e.g. , a chimeric molecule composed of an intracellular and/or transmembrane domain of a receptor or receptor-like molecule, fused to the gene product as the extracellular domain. In another alternative embodiment, the stromal cells can be genetically engineered to "knock out" expression of factors that promote rejection of the implant or degenerative changes in articular cartilage due to aging, rheumatoid disease or inflammation. For example, expression of pro- inflammatory mediators such as GM-CSF, TNF, IL-1, IL-2 and cytokines can be knocked out in the exogenously-administered stromal cells or on the implanted periosteal or perichondrial tissue to reduce the risk of inflammation. Likewise, the expression of MHC class II molecules on the cells or tissues can be knocked out in order to reduce the risk of rejection of the implant.
In another embodiment of the invention, the methods of the invention may afford a vehicle for introducing genes and gene products in vivo to assist or improve the results of the implantation and/or for use in gene therapies. For example, genes that prevent or ameliorate symptoms of degenerative changes in cartilage such as rheumatoid disease or inflammatory reactions and bone resorption, may be underexpressed or overexpressed in disease conditions and/or due to aging. Thus, the level of gene activity in the patient may be increased or decreased, respectively, by gene replacement therapy by adjusting the level of the active gene product in genetically engineered stromal cells.
In yet another preferred embodiment, the cartilage defect site into which the implant will be placed is treated, preferably prior to implantation, to degrade the pre-existing cartilage at the defect site, freeing cells to migrate into the scaffold of the implant and promoting the orderly deposition of new cartilage. Methods of such treatment include enzymatic treatment, abrasion or microdrilling.
According to a further embodiment, the preparation of stromal cells of the invention can be injected into the degraded cartilage at the defect site, e.g., into the surrounding cells or into the walls of the defect, providing a source of biological factors that induce migration of stromal cells from the degraded cartilage to the implant.
4. DETAILED DESCRIPTION OF THE INVENTION The present invention involves methods of making and/or repairing cartilage in vivo comprising the implantation in vivo of a three-dimensional scaffold or framework structure made of a biocompatible, non-living material in combination with periosteal/perichondrial tissue and the administration of a preparation of stromal cells, such as chondrocytes or chondrocyte progenitor cells, which cells are seeded at the site of implantation in vivo. The term "chondrocyte progenitor cell" as used herein refers to either (1) a pluripotent, or lineage-uncommitted, progenitor cell, typically referred to in the art as a "stem cell" or "mesenchymal stem cell", which is potentially capable of an unlimited number of mitotic divisions to either renew its line or to produce progeny cells which will differentiate into chondrocytes; or (2) a lineage-committed progenitor cell produced from the mitotic division of a stem cell which will eventually differentiate into a chondrocyte. Unlike the stem cell from which it is derived, the lineage- committed progenitor is generally considered to be incapable of an unlimited number of mitotic divisions and will eventually differentiate into a chondrocyte.
The term "cartilage" or "cartilage tissue" as used herein is generally recognized in the art, and refers to a specialized type of dense connective tissue comprising cells embedded in an extracellular matrix (ECM) (see, for example, Cormack, 1987, Ham's Histology, 9th Ed., J.B. Lippincott Co., pp. 266-272) . The biochemical composition of cartilage differs according to type; however, the general composition of cartilage comprises chondrocytes surrounded by a dense ECM consisting of collagen, proteoglycans and water. Several types of cartilage are recognized in the art, including, for example, hyaline or articular cartilage such as that found within the joints, fibrous cartilage such as that found within the meniscus and costal regions, and elastic cartilage. The production of any type of cartilage is intended to fall within the scope of the invention. Furthermore, although the invention is directed predominantly to methods for the production of new cartilage tissue in humans, the invention may also be practiced so as to produce new cartilage tissue in any mammal in need thereof, including horses, dogs, cats, sheep, pigs, among others. The treatment of such animals is intended to fall within the scope of the invention.
The invention is divided into the following sections solely for the purpose of description: (a) the three- dimensional scaffold; (b) the periosteal/perichondrial tissue and its implantation in combination with the scaffold; (c) the stromal cell preparation, including genetically engineered stromal cells; (d) administration of the stromal cells in vivo and (e) uses of the methods of the invention.
4.1. THE THREE-DIMENSIONAL SCAFFOLD The three-dimensional scaffold or framework structure may be of any material and/or shape that: (a) allows cells to attach to it (or can be modified to allow the cells to attach to it); and (b) allows cells to grow in more than one layer. Because the three-dimensional structure is to be implanted in vivo, it may be preferable to use biodegradable materials such as polyglycolic acid (PGA) , polylactic acid (PLA) , hyaluronic acid, catgut suture material, gelatin, cellulose, nitrocellulose, collagen, cotton, or other naturally- occurring biodegradable materials. Furthermore, it may be preferable to sterilize the three-dimensional structure prior to implantation, e.g., by treatment with ethylene oxide or by gamma irradiation or irradiation with an electron beam. In addition, a number of other materials may be used to form the scaffold or framework structure, including but not limited to: nylon (polyamides) , dacron (polyesters) , polystyrene, polypropylene, polyacrylates, polyvinyl compounds (e.g., polyvinylchloride) , polycarbonate (PVC) , polytetrafluorethylene (PTFE, teflon) , thermanox (TPX) , and a variety of polyhydroxyalkanoates. Because certain of these materials, such as nylon, polystyrene, etc. may be poor substrates for cellular attachment, when these materials are used as the three-dimensional framework, it is advisable to pre-treat the framework prior to implantation in order to enhance the attachment of chondrocytes and other stromal cells to the scaffold. For example, nylon matrices can be treated with 0.1 M acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon. Polystyrene could be similarly treated using sulfuric acid.
Any of the above-listed materials may be formed into a mesh or a felt, for example, to produce the three-dimensional framework or scaffold for use in the methods of this invention. Regardless of the shape of the framework, the openings of the framework should be of an appropriate size to allow the chondrocytes and other stromal cells to stretch across the openings. Maintaining actively growing stromal cells which stretch across the framework enhances the production of growth factors which are elaborated by the stromal cells, and further enhances new cartilage formation in vivo. In addition, the openings of the framework must allow for adequate diffusion of nutrients and waste products into and out of the structure and for vascularization at the site of implantation. For example, if the openings are too small, the stromal cells may rapidly achieve confluence but be unable to easily exit from the matrix; trapped cells may exhibit contact inhibition and cease production of the appropriate factors necessary to support proliferation. If the openings are too large, the stromal cells may be unable to stretch across the opening; this will also decrease stromal cell production of the appropriate factors necessary to support proliferation. When using a mesh type of framework, openings ranging from about 150 μm to about 220 μm are satisfactory. However, depending upon the three- dimensional structure and intricacy of the framework, other sizes may work equally well. In fact, any shape or structure that allows the stromal cells to stretch and continue to replicate and grow for lengthy time periods will work in accordance with the invention. For example, for felt-type frameworks, openings ranging from about 80 μm to about 120 μm are preferred.
According to a preferred embodiment, the scaffold is a felt, which can be composed of a multifilament yarn made from a bioabsorbable material, e.g., PGA, PLA, polygluconate (PLGA) or hyaluronic acid. The yarn is made into a felt using standard textile processing techniques consisting of crimping, cutting, carding and needling. According to a further preferred embodiment, the porosity of the felt ranges from 80-98%, the density of the felt ranges from 30-60 mg/cc and the thickness of the felt ranges from 1-7 mm.
In an embodiment wherein the scaffold is made of collagen, the collagen may be in the form of a sponge, a braid or woven threads, etc. In an embodiment wherein the scaffold is made of nylon, a convenient nylon mesh is Nitex, a nylon filtration mesh having an average pore size of 210 μm and an average nylon fiber diameter of 90 μm (#3-210/36 Tetko, Inc. , N.Y.) .
Although Applicants are under no duty or obligation to explain the mechanism by which the invention works, a number of factors inherent in the three-dimensional framework may contribute to its successful use in the present invention:
(a) The three-dimensional framework provides a greater surface area for protein attachment, and consequently, for the adherence of stromal cells in vivo.
(b) Because of the three-dimensionality of the matrix, as noted above, the stromal cells that attach to the framework continue to actively grow and produce growth and regulatory factors which promote new cartilage formation in vivo, and are less likely to exhibit contact inhibition. (c) The three-dimensional framework allows for a spatial distribution of cellular elements which is analogous to that found in vivo.
(d) The increase in potential volume for cell growth in the three-dimensional structure may allow the establishment of localized microenvironments conducive to cellular differentiation and maturation in the production of new cartilage in vivo.
(e) The three-dimensional matrix maximizes cell-cell interactions by allowing greater potential for movement of migratory cells, such as macrophages, monocytes and possibly lymphocytes.
(f) It has been recognized that maintenance of a differentiated cellular phenotype requires not only growth/differentiation factors but also the appropriate cellular interactions. The present invention effectively recreates the in vivo tissue microenvironment.
According to one embodiment of the invention, the scaffold may comprise or be modified, e.g. , coated or impregnated, prior to implantation with certain substances to enhance the attachment and growth of chondrocytes and other stromal cells on the scaffold in vivo. These substances include, but are not limited to, bioactive agents such cellular growth factors (e.g., TGF-/3) , substances that stimulate chondrogenesis (e.g., BMPs that stimulate cartilage formation such as BMP-2, BMP-12 and BMP-13) , factors that stimulate migration of stromal cells to the scaffold, factors that stimulate matrix deposition, anti-inflammatories (e.g., non-steroidal anti-inflammatories) , immunosuppressants (e.g., cyclosporins) , as well as other proteins, such as collagens, elastic fibers, reticular fibers, glycoproteins or glycosaminoglycans, such as heparin sulfate, chondroitin-4- sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc. For example, growth factors such as TGF-3, with ascorbate, have been found to trigger chondrocyte differentiation and cartilage formation by chondrocytes. In addition, hyaluronic acid is a good substrate for the attachment of chondrocytes and other stromal cells and can be incorporated as part of the scaffold or coated onto the scaffold.
These bioactive agents may also be included in or on the scaffold for local, sustained release of the agents. Examples of such sustained release formulations include composites comprising the bioactive agent and a biocompatible polymer, such as poly(lactic acid), poly(lactic-co-glycolic acid) , methylcellulose, hyaluronic acid, collagen, and the like. The structure, selection and use of degradable polymers in drug delivery vehicles have been reviewed in several publications, including, A. Domb et al.. 1992, Polymers for Advanced Technologies 3:279-292. Additional guidance in selecting and using polymers in pharmaceutical formulations can be found in the text by M. Chasin and R.
Langer (eds.), 1990, "Biodegradable Polymers as Drug Delivery Systems, Vol. 45 of Drugs and the Pharmaceutical Sciences. M. Dekker, New York.
4.2. THE PERIOSTEAL/PERICHONDRIAL TISSUE AND ITS
IMPLANTATION IN COMBINATION WITH THE SCAFFOLD
The scaffold of the invention as described above is implanted into the defect site in vivo in combination with periosteal tissue, perichondrial tissue or a combination of the two tissues. Periosteal tissue is derived from the periosteum, a fibrous membrane localized at the surfaces of bones, and can be obtained from the periosteum/bone interface of any suitable bone of the patient (or subject) or a histoco patible donor, e.g., ileum, scapula, tibia, fibula, femur, etc. The periosteal tissue contains a variety of stromal cells including osteocytes, chondrocytes and fibroblasts as well as mesenchymal stem cells having the potential to differentiate into osteogenic or chondrogenic cells. Perichondrial tissue is derived from the perichondrium, the fibrous connective tissue covering cartilage, except articular surfaces. Perichondrial tissue contains chondrogenic progenitor cells and chondrocytes. The periosteal/perichondrial tissue can be in the form of a segment or layer of tissue of any size or shape, preferably of a size and shape that fits within or corresponds to the defect site. The tissue can be laid over or under the scaffold at the implantation site and can optionally be mechanically fixed to the scaffold and/or the defect site, e.g., by sutures or glue fixation, e.g., fibrin glue. Although it may be preferable for the periosteal/perichondrial tissue to be autologous (i.e., derived from the subject receiving the implant) , the tissue may be derived from a heterologous source. When periosteal/perichondrial tissue from a heterologous source is used, it may be preferable to add anti-inflammatory factors or immunosuppressants to the defect site, e.g., attached to or within the scaffold or exogenously administered to the site, to minimize the risk of immunological rejection.
According to a preferred embodiment of the invention, the three-dimensional scaffold is implanted at the defect site in vivo and a piece of periosteal/perichondrial tissue is placed over the implanted scaffold and sutured in place so that the tissue overlays and lies adjacent to the scaffold structure. Alternatively, a segment of periosteum or perichondrium may be implanted directly into the defect site and the scaffold placed on top of the tissue such that the stromal cells of the tissue can migrate from the tissue into the scaffold. In any case, the periosteal/perichondrial tissue should be situated with respect to the scaffold in such a way as to allow the stromal cells from the tissue to migrate into the scaffold and proliferate thereon and therein. The scaffold and/or periosteal/perichondrial tissue can be implanted using surgical techniques well known in the art, e.g., arthroscopy.
According to a preferred embodiment, the periosteal tissue is situated or oriented such that the cambium layer of the tissue is facing into the defect; thus, in the embodiment wherein the scaffold is placed directly into the defect site and the periosteal tissue is placed on top of the scaffold, the periosteal tissue is oriented in relation to the top of the scaffold such that the cambium layer is facing the top of the scaffold. When perichondrial tissue is used in the claimed methods, the perichondrial tissue is also placed into the defect site or oriented with respect to the scaffold such that its cambium or inner transition layers faces the defect or scaffold. It is these layers that contain chondrogenic stem cells and/or chondrocytes that can migrate into the scaffold for the production of new cartilage at the defect site.
In an alternative embodiment of the invention, a bioresorbable patch, e.g., film, mesh or felt, can be used in place of the periosteal/perichondrial tissue and situated or oriented adjacent to the scaffold within the defect site. For example, if a film is used, it may be comprised of PGA or polygluconate; if a mesh or felt is used, they may be comprised of vicryl or PLA. The preparation of stromal cells is seeded into the defect site as described herein.
According to a further embodiment of the invention, the defect site is treated, preferably prior to implantation, to degrade the cartilage at the site of the defect, freeing cells (e.g., stromal cells) from that area to migrate into the scaffold of the implant and promoting the orderly deposition of new cartilage. When enzymes are used to treat the defect site, such enzymes include but are not limited to trypsin, chymotrypsin, collagenase, elastase, and/or hyaluronidase, Dnase, pronase, chondroitinase, etc. Alternative methods of treating the defect site to degrade the cartilage include abrasion, debridement, shaving or icrodrilling. Where abrasion techniques are utilized, the surface of the cartilage may be serrated, e.g., via wire wool. In microdrilling, a drilling device is used to create small defects or channels in the cartilage. Treatment of the defect site to degrade or disrupt the pre-existing cartilage reduces the chances of scar tissue forming at the site and promotes the orderly deposition of new cartilage at the defect site. 4.3. THE STROMAL CELL PREPARATION
According to the methods of this invention, a preparation of stromal cells is additionally administered at the implantation site, which cells produce biological factors that promote chondrogenesis and the migration of cells such as chondrogenic stem cells or chondrocytes, from the in vivo environment adjacent to the implant, including from the periosteal/perichondrial tissue, to the scaffold for attachment and/or differentiation thereon and therein. The stromal cell preparation also provides a direct source of stromal cells, e.g. , chondrocytes and/or chondrocyte progenitor cells, that are capable of migrating to the scaffold, attaching thereto, and elaborating cartilage- specific macromolecules and extracellular matrix proteins for the production of new cartilage at the defect site. The cells described herein can be administered before, during or after implantation of the scaffold and/or periosteal/ perichondrial tissue, as discussed in Section 4.4, infra. The stromal cells of the preparation may include chondrocytes, chondrocyte progenitor cells including mesenchymal stem cells, fibroblasts, fibroblast-like cells and/or cells capable of producing collagen type II and other collagen types, and proteoglycans which are typically produced in cartilaginous tissues. The stromal cells can be obtained from the patient (or subject) or a histocompatible donor. The chondrocytes, progenitor cells, fibroblast-like cells and other cells and/or elements that comprise the stroma may be fetal or adult in origin, and may be derived from convenient sources such as cartilage, bone, skin, ligaments, tendons, muscles, placenta, umbilical cord, etc. For example, stromal cells such as chondrocytes may be derived from any type of cartilage, including but not limited to, hyaline cartilage, costal cartilage, fibrous cartilage, etc. , which can be obtained by biopsy (where appropriate) or upon autopsy. Chondrocyte progenitor cells may be derived from various sources including bone marrow, periosteum, perichondrium or various sources of undifferentiated human mesenchyme. Fibroblasts can be obtained in quantity rather conveniently from foreskin, preferably fetal foreskin, or, alternatively, any appropriate cadaver organ. Fetal cells, including fibroblast-like cells and chondrocyte progenitor cells, may be obtained from umbilical cord or placenta tissue or umbilical cord blood. Although stromal cells from a variety of sources may be used in the claimed methods, it is preferable that, for implantation in vivo, the stromal cells be derived from the individual who is to receive the implant or from cells of fetal origin which may be viewed as "universal donors" so as to minimize the risk of immunological rejection of the implant.
The stromal cells may be readily isolated by disaggregating an appropriate tissue which is to serve as the source of the cells. This may be readily accomplished using techniques known to those skilled in the art. For example, the tissue can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that weaken the connections between neighboring cells making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage. Enzymatic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination. These include but are not limited to trypsin, chymotrypsin, collagenase, elastase, and/or hyaluronidase, Dnase, pronase, etc. Mechanical disruption can also be accomplished by a number of methods including, but not limited to the use of grinders, blenders, sieves, homogenizers, pressure cells, or sonicators to name but a few. For a review of tissue disaggregation techniques, see Freshney, Culture of Animal Cells. A Manual of Basic Technique . 2d Ed., A.R. Liss, Inc., New York, 1987, Ch. 9, pp. 107-126.
Once the tissue has been reduced to a suspension of individual cells, the suspension can be fractionated into subpopulations from which chondrocytes, fibroblasts and/or other stromal cells and/or elements can be obtained. This also may be accomplished using standard techniques for cell separation including but not limited to cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection) , separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter- streaming centrifugation) , unit gravity separation, counter current distribution, electrophoresis and fluorescence- activated cell sorting. For a review of clonal selection and cell separation techniques, see Freshney, supra, Ch. 11 and 12, pp. 137-168.
For example, the isolation of chondrocytes, chondrocyte progenitors, fibroblasts or fibroblast-like cells is carried out as follows: fresh human cartilage tissue can be thoroughly washed and minced in Hanks balanced salt solution (HBSS) in order to remove serum. The minced tissue is incubated from 1-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the dissociated cells are suspended, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown. The isolated stromal cells can then be grown to confluency, lifted from the confluent culture and administered to the cartilage defect site in vivo (see, e.g., Naughton et al.. 1987, J. Med. 18(3&4) :219-250) . Fibroblast- like cells may also be isolated from human umbilical cords (33-44 weeks) . Fresh tissues may be minced into pieces and washed with medium or snap-frozen in liquid nitrogen until further use. The umbilical tissues may be disaggregated as described above.
Once chondrocytes or chondrocyte progenitor cells have been isolated, their population can be expanded mitotically in vitro in order to obtain the cell preparation for implantation. Methods for the selection of the most appropriate culture medium, medium preparation, and cell culture techniques are well known in the art and are described in a variety of sources, including Doyle et al.. (eds.), 1995, Cell & Tissue Culture: Laboratory Procedures. John Wiley & Sons, Chichester; and Ho and Wang (eds.), 1991, Animal Cell Bioreactors. Butterworth-Heinemann, Boston.
The cells should be transferred or "passaged" to fresh medium when they reach an appropriate density, such as 3 to 6.5 x 10 /cm2, or, for example, when they reach a defined percentage of confluency on the surface of a culture dish. During incubation, the stromal cells may stick to the walls of the culture vessel where they can continue to proliferate and form a confluent monolayer. This should be prevented or minimized, for example, by transferring a portion of the cells to a new culture vessel having fresh medium, since the presence of a confluent monolayer in the culture vessel will tend to "shut down" the growth of cells in the culture. Removal of the confluent monolayer or transfer of a portion of the cells to fresh media in a new vessel will usually restore proliferative activity of the cells. Such removal or transfer should be done in any culture vessel which has a monolayer exceeding about 25% confluency. Alternatively, the liquid culture can be agitated, for example, on an orbital shaker or in roller bottles, to prevent or minimize the cells from sticking to the vessel walls.
In addition, once the stromal cells have been established in culture, they may be maintained or stored in cell "banks" comprising either continuous in vitro cultures of cells requiring regular transfer, or, preferably, cells which have been cryopreserved. Cryopreservation of the cells may be carried out according to known methods, such as those described in Doyle et al.. 1995, supra. For example, but not by way of limitation, cells may be suspended in a "freeze medium" such as, for example, culture medium further comprising 20% FBS and 9% dimethylsulfoxide (DMSO) , with or without 5-10% glycerol, at a density, for example, of about 4-10 x 106 cells/ml. The cells are dispensed into glass or plastic ampoules (Nunc) which are then sealed and transferred to the freezing chamber of a programmable freezer. The optimal rate of freezing may be determined empirically. For example, a freezing program that gives a change in temperature of -l°C/min through the heat of fusion may be used. Once the ampoules have reached -180°C, they are transferred to a liquid nitrogen storage area. Cryopreserved cells can be stored for a period of years, though they should be checked at least every 5 years for maintenance of viability.
The cryopreserved cells constitute a bank of cells, portions of which can be "withdrawn" by thawing and then used to produce new cartilage tissue as needed. Thawing should generally be carried out rapidly, for example, by transferring an ampoule from liquid nitrogen to a 37°C water bath. The thawed contents of the ampoule should be immediately transferred under sterile conditions to a culture vessel containing an appropriate medium such as RPMI 1640 conditioned with 10% FBS and 5% ES. It is advisable that the cells in the culture medium be adjusted to an initial density of about 3-6 x 105 cells/ml so that the cells can condition the medium as soon as possible, thereby preventing a protracted lag phase. Once in culture, the cells may be examined daily, for example, with an inverted microscope to detect cell proliferation, and subcultured as soon as they reach an appropriate density.
In addition to chondrocytes, chondrocyte progenitors, fibroblasts or fibroblast-like cells, other cells may be added to the cell preparation for implantation in vivo, which other cells aid in the production of the stromal matrix on the scaffold of the invention. For example, other cells found in loose connective tissue may be seeded along with chondrocytes or fibroblasts. Such cells include but are not limited to endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, etc. These stromal cells may readily be derived from appropriate organs including umbilical cord or placenta or umbilical cord blood using methods known in the art such as those discussed above.
Moreover, the stromal cell preparation may further comprise one or more other components, including selected extracellular matrix components, such as one or more types of collagen known in the art, as well as growth factors and/or drugs. Growth factors which may be usefully incorporated into the cell preparation include one or more tissue growth or stimulatory factors known in the art or to be identified in the future, including but not limited to any member of the TGF-3 family, BMPs that stimulate cartilage formation, e.g., BMP-2, BMP-12, and BMP-13, factors that stimulate migration of stromal cells and/or matrix deposition, insulin-like growth factor (IGF)-I and -II, growth hormone, etc. Drugs which may be usefully incorporated into the cell preparation include anti-inflammatory compounds such as non-steroidal anti-inflammatories, immunosuppressants such as the cyclosporins, as well as local anesthetics. Other components may also be included in the preparation, including but not limited to any of the following: (1) buffers to provide appropriate pH and isotonicity; (2) lubricants; (3) viscous materials to retain the cells at or near the site of administration, including, for example, alginates, agars and plant gums; and (4) other cell types that may produce a desired effect at the site of administration, such as, for example, enhancement or modification of the formation of cartilage tissue or its physicochemical characteristics, or support for the viability of the cells, or inhibition of inflammation or rejection. Again, because the cells are to be used for implantation in vivo, it is preferable to obtain the stromal cells from the patient's own tissues or from a fetal source ("universal donor") . The growth of cells on the three-dimensional scaffold may be further enhanced by including in or on the framework or coating the framework with proteins (e.g., collagens, elastic fibers, reticular fibers) , glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4- sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc.), and/or other bioactive materials such as growth factors.
According to one embodiment of the invention, growth regulatory or stimulatory factors including, but not limited to, TGF-/S and ascorbate, or BMPs that stimulate cartilage formation such as BMP-2, BMP-12, and BMP-13 may be added to the implantation site before, during or after implantation of either the scaffold and/or the periosteal/perichondrial tissue, in order to promote the production of new cartilage at the site. Moreover, such growth regulatory factors can be administered to the site at the time of administration of the stromal cells, either as a separate preparation or, as noted supra. as part of the stromal cell preparation. In addition, the stromal cells may be genetically engineered to produce growth factors such as TGF-/S as well as other biological factors such as factors that stimulate chondrogenesis or the migration of chondrogenic and other stromal cells to the scaffold of this invention.
4.3.1. GENETICALLY ENGINEERED STROMAL CELLS The stromal cells administered to or in combination with the scaffold and periosteal/perichondrial tissue of the invention can also be genetically engineered to produce gene products that promote the successful production or repair of cartilage at a defect site and/or for use in gene therapies. For example, the stromal cells can be genetically engineered to express anti-inflammatory factors, e.g., anti-GM-CSF, anti-TNF, anti-IL-1, anti-IL-2, etc., to reduce the risk of rejection of the implant or to reduce the risk of degenerative changes in the cartilage due to rheumatoid disease or other inflammatory reactions. For example, the stromal cells can be genetically engineered to express peptides or polypeptides corresponding to the idiotype of neutralizing antibodies for granulocyte-macrophage colony stimulating factor (GM-CSF), TNF, IL-1, IL-2, or other inflammatory cytokines. IL-1 has been shown to decrease the synthesis of proteoglycans and collagens type II, IX, and XI (Tyler et al. , 1985, Biochem. J. 227:869-878; Tyler et al.. 1988, Coll. Relat. Res. 82: 393-405; Goldring et al.. 1988, J. Clin. Invest. 82:2026-2037; and Lefebvre et al.. 1990, Biophys. Acta. 1052:366-372) and is a potent stimulator of cartilage resorption and of the production of inflammatory mediators by chondrocytes (Campbell et al.. 1991, J. Immunol. 147:1238-1246). TNF also inhibits synthesis of proteoglycans and type II collagen, although it is much less potent than IL-1 (Yaron, I., et al.. 1989, Arthritis Rheum. 32:173-180; Ikebe, T. , et al.. 1988, J. Immunol. 140:827-831; and Saklatvala, J. , 1986, Nature 322:547-549).
Once the genetically engineered stromal cells are implanted into an individual, the presence of the anti- inflammatory gene products can bring about amelioration of immunological rejection or the inflammatory reactions associated with rheumatoid or joint disease.
In another embodiment, the stromal cells can be genetically engineered to express a gene which would exert a therapeutic effect, e.g., in the production of TGF-β to stimulate cartilage production, or other factors such as BMP- 13 to promote chondrogenesis and/or prevent bone formation or stimulatory factors that promote migration of stromal cells and/or matrix deposition. In addition, the stromal cells can be genetically engineered to express a gene for which a patent is deficient. For example, genes that prevent or ameliorate symptoms of various types of rheumatoid or joint diseases may be underexpressed or down-regulated under disease conditions. Specifically, expression of genes involved in preventing inflammatory reactions in rheumatoid or joint diseases may be down-regulated. Alternatively, the activity of gene products may be diminished, leading to the manifestations of some or all of the above pathological conditions and eventual development of symptoms of rheumatoid or joint diseases. Thus, the level of gene activity may be increased by either increasing the level of gene product present or by increasing the level of the active gene product present at the defect site. By implanting stromal cells genetically engineered to express the active target gene product into the defect site of a rheumatoid or joint disease patient who is deficient for that product, the level of the target gene product and/or the activity of that product can be modulated to prevent or ameliorate the symptoms of rheumatoid or joint diseases. "Target gene," as used herein, refers to a gene involved in rheumatoid or joint diseases in a manner by which modulation of the level of target gene expression or of target gene product activity may act to ameliorate symptoms of rheumatoid or joint diseases by preventing resorption of cartilage and production of inflammatory mediators by chondrocytes.
In addition, patients may be treated by gene replacement therapy by means of the stromal cells administered according to the methods of this invention. Thus, replacement or repaired cartilage may be designed specifically to meet the requirements of an individual patient; for example, the stromal cells may be genetically engineered to regulate one or more genes; or the regulation of gene expression may be transient or long-term; or the gene activity may be non- inducible or inducible. For example, the gene encoding the human complement regulatory protein, which prevents rejection of an implant by the host, may be inserted into human fibroblasts. McCurry et al.. 1995, Nature Medicine 1:423- 427.
The stromal cells used in the methods of the invention can also be genetically engineered to "knock out" expression of factors that promote inflammation or rejection at the implant site. Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are discussed below. "Negative modulation", as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level and/or activity of the target gene product in the absence of the modulatory treatment. The expression of a gene native to stromal cell can be reduced or knocked out using a number of techniques, for example, expression may be inhibited by inactivating the gene completely (commonly termed "knockout") using standard homologous recombination techniques. Usually, an exon encoding an important region of the protein (or an exon 5' to that region) is interrupted by a positive selectable marker (for example neo) , preventing the production of normal mRNA from the target gene and resulting in inactivation of the gene. A gene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the sequences intervening the two regions can be deleted. Mombaerts et al.. 1991, Proc. Nat. Acad. Sci. U.S.A. 88:3084-3087. Antisense and ribozyme molecules which inhibit expression of the target gene can also be used in accordance with the invention to reduce the level of target gene activity. For example, antisense RNA molecules which inhibit the expression of major histocompatibility gene complexes (HLA) have been shown to be most versatile with respect to immune responses. Furthermore, appropriate ribozyme molecules can be designed as described, e.g. , by Haseloff et al. , 1988, Nature 334:585-591; Zaug et al.. 1984, Science 224:574-578; and Zaug and Cech, 1986, Science 231:470-475. Still further, triple helix molecules can be utilized in reducing the level of target gene activity. These techniques are described in detail by L.G. Davis et al.. eds, Basic Methods in Molecular Biology. 2nd ed. , Appleton & Lange, Norwalk, Conn. 1994. Using any of the foregoing techniques, the expression of IL-1 can be knocked out in the chondrocytes to reduce the risk of resorption of cartilage and production of inflammatory mediators by the chondrocytes. Likewise, the expression of MHC class II molecules can be knocked out in order to reduce the risk of rejection of the implant.
Methods that may be useful to genetically engineer the cells of the invention are well-known in the art. For example, a recombinant DNA construct or vector containing the gene of interest may be constructed and used to transform or transfect the stromal cells of the invention. Such transformed or transfected cells that carry the gene of interest, and that are capable of expressing said gene, are selected and clonally expanded in culture. Methods for preparing DNA constructs containing the gene of interest, for transforming or transfecting cells, and for selecting cells carrying and expressing the gene of interest are well-known in the art. See, for example, the techniques described in Maniatis et al. , 1989, Molecular Cloning. A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Ausubel et al.. 1989, Current Protocols in Molecular Biology. Greene Publishing Associates & Wiley Interscience, N.Y.; and Sambrook et al.. 1989, Molecular Cloning: A Laboratory Manual. 2nd Ed. , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.
The cells can be engineered using any of a variety of vectors including, but not limited to, integrating viral vectors, e.g. , retrovirus vector or adeno-associated viral vectors; or non-integrating replicating vectors, e.g., papillo a virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors. Where transient expression is desired, non-integrating vectors and replication defective vectors may be preferred, since either inducible or constitutive promoters can be used in these systems to control expression of the gene of interest. Alternatively, integrating vectors can be used to obtain transient expression, provided the gene of interest is controlled by an inducible promoter. Other methods of introducing DNA into cells include the use of liposomes, lipofection, electroporation, a particle gun, or by direct DNA injection.
Hosts cells are preferably transformed or transfected with DNA controlled by, i.e., in operative association with, one or more appropriate expression control elements such as promoter or enhancer sequences, transcription terminators, polyadenylation sites, among others, and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow in enriched media and then switched to selective media. The selectable marker in the foreign DNA confers resistance to the selection and allows cells to stably integrate the foreign DNA as, for example, on a plasmid, into their chromosomes and grow to form foci which, in turn, can be cloned and expanded into cell lines. This method can be advantageously used to engineer cell lines which express the gene product.
Any promoter may be used to drive the expression of the inserted gene. For example, viral promoters include but are not limited to the CMV promoter/enhancer, SV40, papillo avirus, Epstein-Barr virus, elastin gene promoter and β-globin. Preferably, the control elements used to control expression of the gene of interest should allow for the regulated expression of the gene so that the product is synthesized only when needed in vivo. If transient expression is desired, constitutive promoters are preferably used in a non-integrating and/or replication-defective vector. Alternatively, inducible promoters could be used to drive the expression of the inserted gene when necessary. Inducible promoters can be built into integrating and/or replicating vectors. For example, inducible promoters include, but are not limited to, metallothionien and heat shock protein.
A variety of methods may be used to obtain the constitutive or transient expression of gene products engineered into the stromal cells. For example, the transkaryotic implantation technique described by Seldon et al. , 1987, Science 236:714-718 can be used. "Transkaryotic", as used herein, suggests that the nuclei of the implanted cells have been altered by the addition of DNA sequences by stable or transient transfection. Preferably, the stromal cells are engineered to express such gene products transiently and/or under inducible control during the post¬ operative recovery period, or as a chimeric fusion protein anchored to the stromal cells, for example, as a chimeric molecule composed of an intracellular and/or transmembrane domain of a receptor or receptor-like molecule, fused to the gene product as the extracellular domain. Once the stromal cells have been genetically engineered, they may be administered to the patient to produce new cartilage in cases of degenerative connective tissue disease or trauma.
4.4. ADMINISTRATION OF THE STROMAL CELLS IN VIVO
The preparation of stromal cells can be administered either before, during or after implantation of the scaffold and/or the periosteal/perichondrial tissue. For example, the cells can be seeded into the defect site before implantation of either the scaffold or the periosteal/perichondrial tissue. Alternatively, the stromal cells can be administered to the site after either one or both of the scaffold and tissue have been implanted; e.g. , by injection into the site after suturing the periosteal/perichondrial tissue to the scaffold. According to a preferred embodiment, the cells are seeded between the periosteal/perichondrial tissue and the scaffold at the defect site. According to another preferred embodiment, the cells are seeded directly into the scaffold. In the embodiment of the invention wherein the cartilage at the defect site is treated so as to degrade the cartilage, the stromal cells can also be seeded into the degraded cartilage, e.g., into the surrounding cells or directly into the cartilage wall. The stromal cells act therein to induce the migration of stromal cells from the degraded cartilage to the implant.
The stromal cells can be seeded by any means that allows administration of the cells to the defect site, e.g., by injection. When the cells are injected into the site, such injection can be achieved by any means that maintains the viability of the cells, e.g., via syringe or more preferably, via an arthroscope. According to a preferred embodiment of the invention, the number of cells administered can range from approximately 1 x IO6 to 30 x 106 stromal cells.
After the scaffold, periosteal/perichondrial tissue and stromal cells are implanted, the defect site is surgically sealed. According to a preferred embodiment of the invention, the stromal cells to be seeded are surgically obtained from the patient, e.g., from ear cartilage and/or bone marrow, in a separate surgical procedure, cultured in vitro to obtain an appropriate amount of cells and administered to the patient at the time of a second surgery wherein the scaffold and periosteal/perichondrial tissue are implanted. The periosteal/perichondrial tissue is preferably surgically obtained from the patient at the time that the scaffold is implanted, although the tissue can be cultured ij vitro prior to implantation (see, e.g., O'Driscoll et al. , 1994, J. Bone and Joint Surg. 76-A:1042-1051) .
According to the methods of this invention, the stromal cells from the periosteal/perichondrial tissue and from the added stromal preparation populate the scaffold structure to form a stromal matrix that resembles the in vivo microenvironment of cartilage tissue, allowing for the production of new cartilage at the defect site. The stromal cells administered to the site provide important biological factors that promote chondrogenesis and the migration of stromal cells, whether from the implanted tissue, the in vivo cartilage environment of the defect or from the stromal preparation, to the scaffold, thus promoting the production of a living stromal tissue that provides the support, growth factors, and regulatory factors necessary to sustain long- term active proliferation of the stromal cells in vivo. The stromal cells may additionally provide factors that promote the deposition of the living stromal matrix at the defect site. The proliferating cells mature and segregate properly within the matrix to form new cartilage tissue at the defect site in vivo.
The successful repair or replacement of damaged cartilage can be enhanced if the new cartilage tissue can be fixed in place at the site of repair. Post-implantation movement may cause the new cartilage tissue to become dislodged from the site if a pro-active fixation technique is not employed. Various methods can be used to fix the new cartilage tissue in place, including: patches derived from a bioresorbable polymer or biocompatible tissues, which can be placed over the site and sutured; bioabsorbable sutures or other fasteners, e.g., pins, staples, tacks, screws, anchors, glues, e.g., fibrin glue; non-absorbable fixation devices, e.g., sutures, pins, screws and anchors; adhesives; and the use of interference fit geometries.
4.5. USES OF THE METHODS OF THE INVENTION
The methods of this invention are useful to replace or augment existing cartilage tissue in vivo, to introduce new or altered cartilage tissue or to join together biological tissues or structures. The present methods find use in a number of specific areas. For example, the evaluation of internal derangements of articular cartilage in several articulations, including the knee, hip, elbow, ankle and the glenohumeral joint, has been made possible by arthroscopic techniques. Such derangements can be caused by physical trauma to the cartilage, by various connective tissues diseases and/or by increased age of the individual. Arthroscopic surgery has become increasingly popular as well as successful, e.g., numerous small cutting tools, 3 to 4mm in diameter can be used in the knee. Triangulation, in which the operating instruments are brought into the visual field provided by the arthroscope, requires multiple portals of entry; alternatively, the cutting tools can be passed through a channel in the arthroscope itself in which case only one opening in the joint is necessary (Jackson, R.W. , 1983, J. Bone Joint Surg. [AM] 65:416) . Selective removal of damaged or deteriorated cartilage via arthroscopic surgery results in cartilage loss, which can be repaired using the methods of the present invention. The present methods can also be employed to repair or augment cartilage loss that results from major reconstructive surgery for different types of joints, the procedures for which surgery have been described in Resnick, D. , and Niwayama, G. , eds., 1988, Diagnosis of Bone and Joint Disorders, 2d ed. , W.B. Sanders Co. Furthermore, the present methods are useful for reconstruction of the temporal andibular joint as well as for other facial and cranial reconstructions.
As such, the present invention is useful for the production or repair of cartilage in vivo in the treatment of degenerative connective tissue diseases, such as rheumatoid or osteoarthritis, or in the treatment of physical trauma, wherein cartilage is damaged or lost.
5. EXAMPLE The subsections below describe the components to be used in the methods of this invention as well as the in vivo implantation of a scaffold, periosteal tissue and stromal cells at a cartilage defect site according to the methods of the invention.
5.1. PREPARATION OF THREE-DIMENSIONAL SCAFFOLD A three-dimensional scaffold comprising felt derived from a PGA multifilament yarn was generated. The yarn is commercially available from Davis and Geek/Sherwood Medical under the trade name Dexon™. The particular type of Dexon™ yarn is 56/123. The Dexon™ yarn is processed into felt via standard textile processing techniques. The felt sheet, which has a porosity of 97%, a density of 45 g/cc and a thickness of 2-7 mm after processing, is cut into the appropriate size for implantation in vivo. Standard serialization techniques (radiation or ethylene oxide gas) for medical products are used to sterilize the felt. The felt can be implanted dry or pre-soaked using DMEM medium containing 10% fetal bovine serum, 2 mM L-glutamine, non- essential amino acids, 50 mg/ml proline, 1 mM sodium pyruvate, 35 μg/ml gentamicin sulfate and 50 μg/ml ascorbate. 5.2. PREPARATION OF PERIOSTEAL/ PERICHONDRIAL TISSUE
Periosteal tissue to be used in the methods of this invention is obtained by removing, e.g. , by arthroscopy, a section of periosteum from the tibia or femur of the patient. Perichondrial tissue can be obtained by removing a section of the perichondrium from the rib of the patient. The periosteal/perichondrial tissue is of such size and/or shape so as to correspond to the defect site. The excision of the periosteal/perichondrial tissue from the patient is preferably performed at the time of the implantation of the scaffold.
5.3. PREPARATION OF STROMAL CELL PREPARATION A preparation of human chondrocytes is obtained as described in Brittberg et al.. 1994, New Eng. J. Med. 331 (No. 14): 890-895. For example, cartilage slices (e.g., 300- 500 mg by weight) can be obtained by arthroscopy from a minor load-bearing area of a joint such as the femoral condyle of the knee. The cartilage tissue is placed in a chilled sterile solution of sodium chloride (0.9 % weight per volume) and cells are harvested within 2-5 hr by mincing and washing in Ham's F12 medium (Gibco Labs, Grand Island, NY) containing HEPES buffer (10 mmol/1) , gentamicin sulfate (50 μg/ml) , amphotericin B (2 μg/ml) , and L-ascorbic acid (50 μg/ml) .
The cells can similarly be harvested using the complete DMEM medium described in Section 5.1, supra. The cartilage is then digested with collagenase (0.2% weight/volume) for approximately 16 hr and the cells are filtered through a nylon mesh, washed with medium and resuspended in medium supplemented with the patient's serum. The cells are seeded at a density of approximately 1 X 106 cells per T-150 flask and cultured in an incubator at 37°C, 5% C02. Cells are passed at confluence every 5-7 days. In addition, chondrocyte progenitor cells can be obtained using methods similar to those described in Wakitani et al.. 1994, J. Bone and Joint Surg. 76-A (No. 4): 579-592. Briefly, blood is aspirated from bone marrow and the aspirate is washed and centrifuged at 180 x g for 5 min. After resuspension in Ham's F12 medium supplemented with 10% FCS and antibiotics, the red blood cells are disrupted by 5 treatment with 4% acetic acid and the remaining cells are cultured in complete medium (10% FCS) at 37 °C in a humidified atmosphere, 5% C02. At day 5, the medium is changed, which removes essentially all of the non-adherent cells. The adherent cells are grown as a monolayer until confluence.
10 The cells are then trypsinized (0.25% trypsin, 1 mM EDTA) for 5 min, to release the cells from the culture dish.
Chondrocyte progenitor cells can also be obtained from periosteum harvested, e.g., from the tibia or femur. The periosteal tissue is incubated with 0.25% collagenase in
15 Ham's F12 medium for 3 h at 37°C, applying periodic agitation. The tissue is then disrupted by vortexing and the cells are filtered through a 100-micrometer Nitex filter, washed twice and centrifuged. The cells are then cultured in complete medium, which is replaced every other day. Adherent cells
20 are grown to confluence and released with trypsin as described above for the bone-marrow derived cells.
Prior to administration in vivo, the stromal cell preparation is suspended by treatment with trypsin, centrifuged and the pellet washed in culture medium
25 supplemented with autologous serum. The cell preparation is then aspirated into a 1 ml tuberculin syringe with a 1.2 mm needle for administration to the implant site.
5.4. IMPLANTATION OF SCAFFOLD, PERIOSTEAL 30 TISSUE AND STROMAL CELLS IN VIVO
The cartilage defect site is cleaned/sterilized prior to implantation by scrubbing or rinsing with a sterilizing solution, e.g., iodine surgical scrub, e.g., 0.75% titratable iodine, or 0.9% sodium chloride solution. The defect site
__ can also be enzymatically treated to degrade the cartilage surface area of the defect by treatment with trypsin in the range of 0.1-100 Units/ml for 1-30 min depending upon the concentration of enzyme used.
The felt scaffold as described above, tailored to fit the defect site, is placed into the site and the periosteal tissue as described above is placed on top of the felt scaffold with the cambium layer of the periosteum facing toward the scaffold and into the defect. The periosteal tissue may optionally be sewn onto the ends of the felt scaffold using resorbable vicryl sutures. The flap is also sutured to the surrounding cartilage at the defect site.
Either prior to implantation of the scaffold and/or the periosteal tissue, the stromal cells of the preparation described above can be administered, e.g., by an arthroscope, into the defect site. For a defect 5 mm thick and 1.5 cm in diameter, a total of 14 x 106 stromal cells in 200 μl of a nutrient-based solution such as DMEM (with or without serum) is administered. According to a preferred embodiment, the stromal cells are injected between the periosteal tissue and the scaffold. The defect site is closed in layers with sutures and the site bandaged. A post-surgical analgesic may be administered.
The present invention is not to be limited in scope by the specific embodiments described which are intended as single illustrations of individual aspects of the invention, and functionally equivalent methods and components are within the scope of the invention, in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims.

Claims

What is claimed is:
1. A method of producing cartilage at a cartilage defect site in vivo comprising: (a) implanting into the defect site a biocompatible, non-living three-dimensional scaffold structure in combination with periosteal tissue, perichondrial tissue or a combination of said tissues; and
(b) additionally administering into the defect site a preparation of stromal cells for attachment to the scaffold and/or for inducing chondrogenesis or migration of stromal cells from the in vivo environment adjacent to the defect site to the scaffold.
2. The method of claim 1, wherein the scaffold is implanted into the defect site and the periosteal or perichondrial tissue is placed on top of and adjacent to the scaffold.
3. The method of claim 1, wherein the periosteal or perichondrial tissue is implanted into the defect site and the scaffold is placed on top of and adjacent to the tissue.
4. The method of claim 1, wherein the periosteal or perichondrial tissue is situated with respect to the scaffold such that stromal cells from the tissue can migrate from the tissue to the scaffold.
5. The method of claim 1, wherein the periosteal tissue is situated with respect to the scaffold such that the cambium layer of the tissue faces the scaffold.
6. The method of claim 1, wherein the perichondrial tissue is situated with respect to the scaffold such that the cambium or inner transition layers of the tissue face the scaffold.
7. The method of claim 1, wherein the preparation of stromal cells is administered prior to, during or after implantation of the scaffold structure.
8. The method of claim 1, wherein the preparation of stromal cells is administered prior to, during or after implantation of the periosteal or perichondrial tissue.
9. The method of claim 1, wherein the preparation of stromal cells is physically placed between the scaffold and the periosteal or perichondrial tissue.
10. The method of claim 1, wherein the scaffold structure is composed of a biodegradable material.
11. The method of claim 10, wherein the biodegradable material is polyglycolic acid, polylactic acid, cotton, cat gut sutures, cellulose, nitrocellulose, gelatin, collagen or polyhydroxyalkanoates.
12. The method of claim 1, wherein the scaffold structure is composed of a non-biodegradable material.
13. The method of claim 12, wherein the non- biodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, thermanox or a polyhydroxyalkanoate.
14. The method of claim 1, wherein the framework is a felt.
15. The method of claim 1, wherein the framework is a mesh.
16. The method of claim 1, wherein the scaffold is treated with ethylene oxide prior to implantation.
17. The method of claim 1, wherein the scaffold is treated with an electron beam prior to implantation.
18. The method of claim 1, wherein the scaffold comprises or is modified to contain at least one substance capable of enhancing the attachment or growth of stromal cells on the scaffold.
19. The method of claim 18, wherein the substance is a bioactive agent selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants.
20. The method of claim 19, wherein the bioactive agent is a TGF-S, with or without ascorbate.
21. The method of claim 19, wherein the bioactive agent is a bone morphogenetic protein that stimulates cartilage formation.
22. The method of claim 19, wherein the bioactive agent comprises a sustained release formulation.
23. The method of claim 22, wherein the formulation comprises a composite of the bioactive agent and a biocompatible polymer.
24. The method of claim 23, wherein the biocompatible polymer is selected from the group consisting of polylactic acid, poly(lactic-co-glycolic acid) , methylcellulose, hyaluronic acid, and collagen.
25. The method of claim 18, wherein the substance is selected from the group consisting of collagens, elastic fibers, reticular fibers, heparin sulfate, chondroitin-4- sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate and hyaluronic acid.
26. The method of claim 1, further comprising the step of administering to the defect site at least one substance capable of enhancing the attachment or growth of stromal cells on the scaffold.
27. The method of claim 26, wherein the substance is a bioactive agent selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants.
28. The method of claim 27, wherein the substance is a TGF-/3, with or without ascorbate.
29. The method of claim 27, wherein the bioactive agent is a bone morphogenetic protein that stimulates cartilage formation.
30. The method of claim 1, wherein the periosteal or perichondrial tissue is autologous to the defect site.
31. The method of claim 1, wherein the preparation of stromal cells comprises chondrocytes.
32. The method of claim 1, wherein the preparation of stromal cells comprises chondrocyte progenitor cells.
33. The method of claim 1, wherein the preparation of stromal cells comprises fibroblasts or fibroblast-like cells.
34. The method of claim 1, wherein the preparation of stromal cells comprises a combination of chondrocytes, chondrocyte progenitor cells, fibroblasts, fibroblast-like cells, endothelial cells, pericytes, macrophages, monocytes, leukocytes, plasma cells, mast cells, adipocytes, umbilical cord cells, or bone marrow cells from umbilical cord blood.
35. The method of claim 1, wherein the preparation of stromal cells comprises at least one bioactive agent.
36. The method of claim 35, wherein the bioactive agent is selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants.
37. The method of claim 36, wherein the bioactive agent is a TGF-/S, with or without ascorbate.
38. The method of claim 36, wherein the bioactive agent is a bone morphogenetic protein that stimulates cartilage formation.
39. The method of claim 1, wherein the stromal cells of the preparation are genetically engineered to produce at least one bioactive agent.
40. The method of claim 39, wherein the bioactive agent is selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants.
41. The method of claim 1, wherein the stromal cells of the preparation are genetically engineered to express a gene that is deficiently expressed in vivo.
42. The method of claim 1, wherein the stromal cells of the preparation are genetically engineered to prevent or reduce the expression of a gene normally expressed by the stromal cells.
43. The method of claim 1, wherein the cartilage defect site is treated to degrade the existing cartilage at the site.
44. The method of claim 43, wherein the treatment is selected from the group consisting of enzyme treatment, abrasion, debridement, shaving, and microdrilling.
45. The method of claim 44, wherein the enzyme treatment utilizes at least one enzyme selected from the group consisting of trypsin, chy otrypsin, collagenase, elastase, hyaluronidase, Dnase, pronase and chondroitinase.
46. The method of claim 43, wherein the cartilage defect site is enzymatically treated prior to implantation of the scaffold or the periosteal or perichondrial tissue.
AMENDED CLAIMS
[received by the International Bureau on 9 Ouly 1997 (09.07.97); original claims 1-46 replaced by new claims 1-47 (8 pages)]
1. The use of a biocompatible, non-living three- dimensional scaffold structure in combination with periosteal tissue, perichondrial tissue or a combination of said tissues in conjunction with a preparation of stromal cells for the production, subsequent to implantation, of cartilage.
2. The use according to claim 1, characterized by the fact that said stromal cells attach to the scaffold and/or induce chondrogenesis or migration of stromal cells.
3. The use according to claim 1 or 2, characterized by the fact that the periosteal or perichondrial tissue is located on top or at the bottom of and adjacent to the scaffold.
4. The use according to claim 1 or 2, characterized by the fact that the periosteal or perichondrial tissue is situated with respect to the scaffold such that stromal cells from the tissue can migrate from the tissue to the scaffold.
5. The use according to claim 1 or 2, characterized by the fact that the periosteal or perichondrial tissue is situated with respect to the scaffold such that the cambium or inner transition layers of the tissue faces the scaffold.
6. The use according to claim 1 or 2, characterized by the fact that the preparation of stromal cells is physically placed between the scaffold and the periosteal or perichondrial tissue.
7. The use according to any one of claims l to 6, characterized by the fact that the scaffold structure is composed of a biodegradable or non-biodegradable material. 8. The use according to claim 7, characterized by the fact that the biodegradable material is polyglycolic acid, polylactic acid, cotton, cat gut sutures, cellulose, nitrocellulose, gelatin, collagen or polyhydroxyalkanoates.
9. The use according to claim 7, characterized by the fact that the non-biodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, thermanox, cotton, cellulose or a polyhydroxyalkanoate.
10. The use according to any one of claims l to 9, characterized by the fact that the framework is a felt or a mesh.
11. The use according to any one of claims 1 to 10, characterized by the fact that the scaffold is treated with ethylene oxide or with an electron beam prior to implantation.
12. The use according to any one of claims l to 11, characterized by the fact that the scaffold comprises or is modified to contain at least one substance capable of enhancing the attachment or growth of stromal cells on the scaffold.
13. The use according to claim 12, characterized by the fact that the substance is (a) a bioactive agent selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants, or (b) is selected from the group consisting of collagens, elastic fibers, reticular fibers, heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate and hyaluronic acid. 14. The use according to claim 13, characterized by the fact that the bioactive agent (a) is a TGF-/3, with or without ascorbate, or (b) is a bone morphogenetic protein that stimulates cartilage formation, or (c) comprises a sustained release formulation.
15. The use according to claim 14, characterized by the fact that the formulation comprises a composite of the bioactive agent and a biocompatible polymer.
16. The use according to claim 15, characterized by the fact that the biocompatible polymer is selected from the group consisting of polylactic acid, poly(lactic-co-glycolic acid) , methylcellulose, hyaluronic acid, and collagen.
17. The use according to any one of claims 1 to 16, characterized by the fact that the preparation of stromal cells comprises chondrocytes, or chondrocyte progenitor cells, or fibroblasts or fibroblast-like cells, or a combination of chondrocytes, chondrocyte progenitor cells, fibroblasts, fibroblast-like cells, endothelial cells, pericytes, macrophages, monocytes, leukocytes, plasma cells, mast cells, adipocytes, umbilical cord cells, or bone marrow cells from umbilical cord blood.
18. The use according to claim 17, characterized by the fact that the chondrocyte progenitor cells comprise mesenchymal stem cells.
19. The use according to any one of claims 1 to 18, characterized by the fact that the preparation of stromal cells further comprise at least one bioactive agent or said cells are genetically engineered to produce at least one bioactive agent.
20. The use according to claim 19, characterized by the fact that the bioactive agent is selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants.
21. The use according to claim 20, characterized by the fact that the bioactive agent is a TGF-/3, with or without ascorbate, or a bone morphogenetic protein that stimulates cartilage formation.
22. The use according to any one of claims 1 to 18, characterized by the fact that the stromal cells of the preparation are genetically engineered to express a gene that is deficiently expressed in vivor or to prevent or reduce the expression of a gene normally expressed by the stromal cells.
23. A method of producing cartilage at a cartilage defect site in vivo comprising:
(a) implanting into the defect site a biocompatible, non-living three-dimensional scaffold structure in combination with periosteal tissue, perichondrial tissue or a combination of said tissues; and
(b) additionally administering into the defect site a preparation of stromal cells for attachment to the scaffold and/or for inducing chondrogenesis or migration of stromal cells from the in vivo environment adjacent to the defect site to the scaffold.
24. The method of claim 1, wherein the scaffold is implanted into the defect site and the periosteal or perichondrial tissue is placed on top of and adjacent to the scaffold.
25. The method of claim 1, wherein the periosteal or perichondrial tissue is implanted into the defect site and the scaffold is placed on top of and adjacent to the tissue. 26. The method of claim 1, wherein the periosteal or perichondrial tissue is situated with respect to the scaffold such that stromal cells from the tissue can migrate from the tissue to the scaffold.
27. The method of claim 1, wherein the periosteal tissue is situated with respect to the scaffold such that the cambium layer of the tissue faces the scaffold.
28. The method of claim 1, wherein the perichondrial tissue is situated with respect to the scaffold such that the cambium or inner transition layers of the tissue face the scaffold.
29. The method of claim 1, wherein the preparation of stromal cells is administered prior to, during or after implantation of the scaffold structure.
30. The method of claim 1, wherein the preparation of stromal cells is administered prior to, during or after implantation of the periosteal or perichondrial tissue.
31. The method of claim 1, wherein the preparation of stromal cells is physically placed between the scaffold and the periosteal or perichondrial tissue.
32. The method according to any one of claims 23 to 31, characterized by the fact that the scaffold structure is composed of a biodegradable or a non-biodegradable material.
33. The method according to claim 32, characterized by the fact that the biodegradable material is polyglycolic acid, polylactic acid, cotton, cat gut sutures, cellulose, nitrocellulose, gelatin, collagen or polyhydroxyalkanoates.
34. The method according to claim 32, characterized by the fact that the non-biodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, thermanox, cotton, cellulose or a polyhydroxyalkanoate.
35. The method according to any one of claims 23 to 34, characterized by the fact that the framework is a felt or a mesh.
36. The method according to any one of claims 23 to 35, characterized by the fact that the scaffold is treated with ethylene oxide or with an electron beam prior to implantation.
37. The method according to any one of claims 23 to 36, characterized by the fact that the scaffold comprises or is modified to contain at least one substance capable of enhancing the attachment or growth of stromal cells on the scaffold.
38. The method according to claim 37, characterized by the fact that the substance is (a) a bioactive agent selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants, or (b) is selected from the group consisting of collagens, elastic fibers, reticular fibers, heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate and hyaluronic acid.
39. The method according to claim 38, characterized by the fact that the bioactive agent (a) is a TGF-β, with or without ascorbate, or (b) is a bone morphogenetic protein that stimulates cartilage formation, or (c) comprises a sustained release formulation. 40. The method according to claim 39, characterized by the fact that the formulation comprises a composite of the bioactive agent and a biocompatible polymer.
41. The method according to claim 40, characterized by the fact that the biocompatible polymer is selected from the group consisting of polylactic acid, poly(lactic-co-glycolic acid) , methylcellulose, hyaluronic acid, and collagen.
42. The method according to any one of claims 23 to 41, characterized by the fact that the preparation of stromal cells comprises chondrocytes, or chondrocyte progenitor cells, or fibroblasts or fibroblast-like cells, or a combination of chondrocytes, chondrocyte progenitor cells, fibroblasts, fibroblast-like cells, endothelial cells, pericytes, macrophages, monocytes, leukocytes, plasma cells, mast cells, adipocytes, umbilical cord cells, or bone marrow cells from umbilical cord blood.
43. The method according to claim 42, characterized by the fact that the chondrocyte progenitor cells comprise mesenchymal stem cells.
44. The method according to any one of claims 23 to 43, characterized by the fact that the preparation of stromal cells further comprise at least one bioactive agent or said cells are genetically engineered to produce at least one bioactive agent.
45. The method according to claim 44, characterized by the fact that the bioactive agent is selected from the group consisting of cellular growth factors, factors that stimulate migration of stromal cells, factors that stimulate chondrogenesis, factors that stimulate matrix deposition, anti-inflammatories, and immunosuppressants. 46. The method according to claim 45, characterized by the fact that the bioactive agent is a TGF-/3, with or without ascorbate, or a bone morphogenetic protein that stimulates cartilage formation.
47. The method according to any one of claims 23 to 43, characterized by the fact that the stromal cells of the preparation are genetically engineered to express a gene that is deficiently expressed in vivo, or to prevent or reduce the expression of a gene normally expressed by the stromal cells.
PCT/US1997/002909 1996-02-21 1997-02-20 Method for making and/or repairing cartilage WO1997030662A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP9530388A JP2000505338A (en) 1996-02-21 1997-02-20 Method of forming and / or repairing cartilage
NZ331517A NZ331517A (en) 1996-02-21 1997-02-20 A method for making and/or repairing cartilage in vivo by implanting a scaffold or framework in combination with periosteal/perichondrial tissue that holds the scaffold in place
AU19731/97A AU715282B2 (en) 1996-02-21 1997-02-20 Method for making and/or repairing cartilage
EP97907835A EP0955959A1 (en) 1996-02-21 1997-02-20 Method for making and/or repairing cartilage

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/604,284 US5842477A (en) 1996-02-21 1996-02-21 Method for repairing cartilage
US08/604,284 1996-02-21

Publications (1)

Publication Number Publication Date
WO1997030662A1 true WO1997030662A1 (en) 1997-08-28

Family

ID=24418997

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/002909 WO1997030662A1 (en) 1996-02-21 1997-02-20 Method for making and/or repairing cartilage

Country Status (8)

Country Link
US (1) US5842477A (en)
EP (1) EP0955959A1 (en)
JP (1) JP2000505338A (en)
KR (1) KR19990087147A (en)
AU (1) AU715282B2 (en)
CA (1) CA2247158A1 (en)
NZ (1) NZ331517A (en)
WO (1) WO1997030662A1 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998042389A1 (en) * 1997-03-25 1998-10-01 Peter Villeneuve Materials for healing cartilage and bone defects
WO1999011789A1 (en) * 1997-09-04 1999-03-11 North Shore University Hospital Research Corporation Genetic engineering of cells to enhance healing and tissue regeneration
WO1999025396A2 (en) * 1997-11-17 1999-05-27 The Regents Of The University Of Michigan Hybrid tissues for tissue engineering
WO2000009179A2 (en) * 1998-08-14 2000-02-24 Verigen Transplantation Service International (Vtsi) Ag Methods, instruments and materials for chondrocyte cell transplantation
US6283980B1 (en) 1996-08-30 2001-09-04 Verigen Transplantation Services Internt'l Method, instruments, and kit for autologous transplantation
WO2002010348A2 (en) * 2000-07-29 2002-02-07 Smith & Nephew Plc Tissue implant for cartilage repair
US6569172B2 (en) 1996-08-30 2003-05-27 Verigen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
EP1416879A2 (en) * 2001-07-16 2004-05-12 Depuy Products, Inc. Unitary surgical device and method
EP1416888A2 (en) * 2001-07-16 2004-05-12 Depuy Products, Inc. Meniscus regeneration device and method
EP1656960A1 (en) * 1998-08-14 2006-05-17 Verigen AG Methods, instruments and materials for chondrocyte cell transplantation
US7445793B2 (en) 2002-09-09 2008-11-04 Kaneka Corporation Support for tissue regeneration and process for producing the same
JP2009102393A (en) * 1997-02-07 2009-05-14 Stryker Corp Matrix-free osteogenic devices, implants and methods of use thereof
US20100189712A1 (en) * 2006-11-17 2010-07-29 Cytograft Tissue Engineering, Inc. Preparation And Use Of Cell-Synthesized Threads
EP1276486B1 (en) * 2000-04-25 2010-11-24 Osiris Therapeutics, Inc. Joint repair using mesenchymal stem cells
US7871440B2 (en) 2006-12-11 2011-01-18 Depuy Products, Inc. Unitary surgical device and method
AU2007203472B2 (en) * 2000-07-29 2011-07-14 Smith & Nephew Plc Tissue implant for cartilage repair
US8137689B1 (en) 1999-11-11 2012-03-20 Zimmer Gmbh Transplant/implant device and method for its production
US8173162B2 (en) 2003-02-26 2012-05-08 Zimmer Orthobiologics, Inc. Preparation for repairing cartilage tissue, especially articular cartilage defects
US8691259B2 (en) 2000-12-21 2014-04-08 Depuy Mitek, Llc Reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US8895045B2 (en) 2003-03-07 2014-11-25 Depuy Mitek, Llc Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
US8945535B2 (en) 2005-09-20 2015-02-03 Zimmer Orthobiologics, Inc. Implant for the repair of a cartilage defect and method for manufacturing the implant
US9211362B2 (en) 2003-06-30 2015-12-15 Depuy Mitek, Llc Scaffold for connective tissue repair
US9238090B1 (en) 2014-12-24 2016-01-19 Fettech, Llc Tissue-based compositions
US9486558B2 (en) 2003-03-27 2016-11-08 Locate Therapeutics Limited Porous matrix
US9511171B2 (en) 2002-10-18 2016-12-06 Depuy Mitek, Llc Biocompatible scaffolds with tissue fragments
US10583220B2 (en) 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue

Families Citing this family (374)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050186673A1 (en) * 1995-02-22 2005-08-25 Ed. Geistlich Soehne Ag Fuer Chemistrie Industrie Collagen carrier of therapeutic genetic material, and method
GB9503492D0 (en) * 1995-02-22 1995-04-12 Ed Geistlich S Hne A G F R Che Chemical product
US6974571B2 (en) * 1995-03-28 2005-12-13 Thomas Jefferson University Isolated stromal cells and methods of using the same
US6653134B2 (en) * 1995-03-28 2003-11-25 Cp Hahnemann University Isolated stromal cells for use in the treatment of diseases of the central nervous system
WO1998004681A2 (en) * 1996-07-25 1998-02-05 Genzyme Corporation Chondrocyte media formulations and culture procedures
US6187053B1 (en) * 1996-11-16 2001-02-13 Will Minuth Process for producing a natural implant
US7618451B2 (en) 2001-05-25 2009-11-17 Conformis, Inc. Patient selectable joint arthroplasty devices and surgical tools facilitating increased accuracy, speed and simplicity in performing total and partial joint arthroplasty
US8556983B2 (en) 2001-05-25 2013-10-15 Conformis, Inc. Patient-adapted and improved orthopedic implants, designs and related tools
US8617242B2 (en) 2001-05-25 2013-12-31 Conformis, Inc. Implant device and method for manufacture
US7468075B2 (en) 2001-05-25 2008-12-23 Conformis, Inc. Methods and compositions for articular repair
US9603711B2 (en) 2001-05-25 2017-03-28 Conformis, Inc. Patient-adapted and improved articular implants, designs and related guide tools
US8771365B2 (en) 2009-02-25 2014-07-08 Conformis, Inc. Patient-adapted and improved orthopedic implants, designs, and related tools
US8480754B2 (en) 2001-05-25 2013-07-09 Conformis, Inc. Patient-adapted and improved articular implants, designs and related guide tools
US8083745B2 (en) 2001-05-25 2011-12-27 Conformis, Inc. Surgical tools for arthroplasty
US8735773B2 (en) 2007-02-14 2014-05-27 Conformis, Inc. Implant device and method for manufacture
US8882847B2 (en) 2001-05-25 2014-11-11 Conformis, Inc. Patient selectable knee joint arthroplasty devices
US7534263B2 (en) 2001-05-25 2009-05-19 Conformis, Inc. Surgical tools facilitating increased accuracy, speed and simplicity in performing joint arthroplasty
US8545569B2 (en) 2001-05-25 2013-10-01 Conformis, Inc. Patient selectable knee arthroplasty devices
US7767452B2 (en) * 1997-02-20 2010-08-03 Kleinsek Don A Tissue treatments with adipocyte cells
WO1998036704A1 (en) * 1997-02-20 1998-08-27 Keller Gregory S Augmentation and repair of dermal, subcutaneous, and vocal cord tissue defects
PT981381E (en) * 1997-05-12 2007-04-30 Metabolix Inc Polyhydroxyalkanoates for in vivo applications
US6110209A (en) * 1997-08-07 2000-08-29 Stone; Kevin R. Method and paste for articular cartilage transplantation
DE69714035T2 (en) 1997-08-14 2003-03-06 Sulzer Innotec Ag Composition and device for repairing cartilage tissue in vivo consisting of nanocapsules with osteoinductive and / or chondroinductive factors
US7637948B2 (en) 1997-10-10 2009-12-29 Senorx, Inc. Tissue marking implant
US20050186283A1 (en) * 1997-10-10 2005-08-25 Ed. Geistlich Soehne Ag Fuer Chemistrie Industrie Collagen carrier of therapeutic genetic material, and method
US8668737B2 (en) 1997-10-10 2014-03-11 Senorx, Inc. Tissue marking implant
US8858981B2 (en) * 1997-10-10 2014-10-14 Ed. Geistlich Soehne Fuer Chemistrie Industrie Bone healing material comprising matrix carrying bone-forming cells
EP1030676B1 (en) * 1997-10-30 2005-09-14 The General Hospital Corporation Bonding of cartilaginous matrices using isolated chondrocytes
US20030099620A1 (en) * 1997-10-30 2003-05-29 The General Hospital Corporation Bonding of cartilaginous matrices using isolated chondrocytes
US6110487A (en) * 1997-11-26 2000-08-29 Keraplast Technologies Ltd. Method of making porous keratin scaffolds and products of same
US5932552A (en) * 1997-11-26 1999-08-03 Keraplast Technologies Ltd. Keratin-based hydrogel for biomedical applications and method of production
WO1999041403A1 (en) * 1998-02-12 1999-08-19 The Regents Of The University Of California Compositions for receptor/liposome mediated transfection and methods of using same
US6224630B1 (en) 1998-05-29 2001-05-01 Advanced Bio Surfaces, Inc. Implantable tissue repair device
ATE439806T1 (en) 1998-09-14 2009-09-15 Univ Leland Stanford Junior DETERMINING THE CONDITION OF A JOINT AND PREVENTING DAMAGE
US7239908B1 (en) 1998-09-14 2007-07-03 The Board Of Trustees Of The Leland Stanford Junior University Assessing the condition of a joint and devising treatment
US7184814B2 (en) 1998-09-14 2007-02-27 The Board Of Trustees Of The Leland Stanford Junior University Assessing the condition of a joint and assessing cartilage loss
US6727224B1 (en) * 1999-02-01 2004-04-27 Genetics Institute, Llc. Methods and compositions for healing and repair of articular cartilage
US7983734B2 (en) * 2003-05-23 2011-07-19 Senorx, Inc. Fibrous marker and intracorporeal delivery thereof
US6862470B2 (en) 1999-02-02 2005-03-01 Senorx, Inc. Cavity-filling biopsy site markers
US8498693B2 (en) 1999-02-02 2013-07-30 Senorx, Inc. Intracorporeal marker and marker delivery device
US9820824B2 (en) 1999-02-02 2017-11-21 Senorx, Inc. Deployment of polysaccharide markers for treating a site within a patent
US6725083B1 (en) 1999-02-02 2004-04-20 Senorx, Inc. Tissue site markers for in VIVO imaging
US8361082B2 (en) 1999-02-02 2013-01-29 Senorx, Inc. Marker delivery device with releasable plug
US20090216118A1 (en) 2007-07-26 2009-08-27 Senorx, Inc. Polysaccharide markers
US7651505B2 (en) 2002-06-17 2010-01-26 Senorx, Inc. Plugged tip delivery for marker placement
EP2305324B1 (en) 1999-03-25 2014-09-17 Metabolix, Inc. Medical devices and applications of polyhydroxyalkanoate polymers
US6315992B1 (en) * 1999-06-30 2001-11-13 Tissuegene Co. Generating cartilage in a mammal using fibroblasts transfected with a vector encoding TGF-β-1
US6575991B1 (en) 1999-06-17 2003-06-10 Inrad, Inc. Apparatus for the percutaneous marking of a lesion
US7338655B1 (en) * 1999-06-30 2008-03-04 Tissuegene, Inc. Gene therapy using TGF-β
US6849594B1 (en) 1999-06-30 2005-02-01 John Lawler Purification and use of human recombinant cartilage oligomeric matrix protein
US20020095157A1 (en) 1999-07-23 2002-07-18 Bowman Steven M. Graft fixation device combination
US6179840B1 (en) 1999-07-23 2001-01-30 Ethicon, Inc. Graft fixation device and method
US7078232B2 (en) * 1999-08-19 2006-07-18 Artecel, Inc. Adipose tissue-derived adult stem or stromal cells for the repair of articular cartilage fractures and uses thereof
US6429013B1 (en) * 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US6783546B2 (en) 1999-09-13 2004-08-31 Keraplast Technologies, Ltd. Implantable prosthetic or tissue expanding device
US7025980B1 (en) * 1999-09-14 2006-04-11 Tepha, Inc. Polyhydroxyalkanoate compositions for soft tissue repair, augmentation, and viscosupplementation
FR2798671A1 (en) * 1999-09-16 2001-03-23 Univ Paris Curie CHONDROCYTE COMPOSITIONS, PREPARATION AND USES
US8632590B2 (en) 1999-10-20 2014-01-21 Anulex Technologies, Inc. Apparatus and methods for the treatment of the intervertebral disc
US7935147B2 (en) 1999-10-20 2011-05-03 Anulex Technologies, Inc. Method and apparatus for enhanced delivery of treatment device to the intervertebral disc annulus
US6592625B2 (en) 1999-10-20 2003-07-15 Anulex Technologies, Inc. Spinal disc annulus reconstruction method and spinal disc annulus stent
US7004970B2 (en) 1999-10-20 2006-02-28 Anulex Technologies, Inc. Methods and devices for spinal disc annulus reconstruction and repair
US7615076B2 (en) 1999-10-20 2009-11-10 Anulex Technologies, Inc. Method and apparatus for the treatment of the intervertebral disc annulus
US7052516B2 (en) 1999-10-20 2006-05-30 Anulex Technologies, Inc. Spinal disc annulus reconstruction method and deformable spinal disc annulus stent
US8128698B2 (en) 1999-10-20 2012-03-06 Anulex Technologies, Inc. Method and apparatus for the treatment of the intervertebral disc annulus
US7951201B2 (en) 1999-10-20 2011-05-31 Anulex Technologies, Inc. Method and apparatus for the treatment of the intervertebral disc annulus
US20080267923A2 (en) * 1999-11-05 2008-10-30 Donald Kleinsek Hair undifferentiated cells
US7799325B2 (en) * 1999-11-05 2010-09-21 Kleinsek Donald A Removal of hypertrophic scars
JP2003517858A (en) * 1999-11-05 2003-06-03 ジェリジーン メディカル コーポレーション Augmentation and repair of age-related soft tissue defects
US20090074729A2 (en) * 1999-11-05 2009-03-19 Donald Kleinsek Augmentation and repair of spincter defects with cells including fibroblasts
US20080286242A2 (en) * 1999-11-05 2008-11-20 Donald Kleinsek Augmentation and repair of spincter defects with cells including mesenchymal cells
ES2491866T3 (en) 1999-11-15 2014-09-08 Piramal Healthcare (Canada) Limited Temperature-controlled, pH-dependent, self-gelling aqueous biopolymer solution
US20030158302A1 (en) * 1999-12-09 2003-08-21 Cyric Chaput Mineral-polymer hybrid composition
AU1979201A (en) * 1999-12-09 2001-06-18 Bio Syntech Canada Inc Mineral-polymer hybrid composition
US6623963B1 (en) * 1999-12-20 2003-09-23 Verigen Ag Cellular matrix
US6702821B2 (en) 2000-01-14 2004-03-09 The Bonutti 2003 Trust A Instrumentation for minimally invasive joint replacement and methods for using same
US7635390B1 (en) 2000-01-14 2009-12-22 Marctec, Llc Joint replacement component having a modular articulating surface
US6805695B2 (en) 2000-04-04 2004-10-19 Spinalabs, Llc Devices and methods for annular repair of intervertebral discs
JP5684963B2 (en) * 2000-04-14 2015-03-18 ユニバーシティ オブ ピッツバーグ オブ ザ コモンウェルス システム オブ ハイヤー エデュケイション Muscle tissue-derived progenitor cells, their compositions, and soft tissue and bone growth and thickening using treatments
US6454803B1 (en) 2000-05-23 2002-09-24 Romo, Iii Thomas External nasal valve batten implant device and method
JP5089006B2 (en) * 2000-06-29 2012-12-05 ピラマル ヘルスケア (カナダ) リミテッド Compositions and methods for repair and regeneration of cartilage and other tissues
US6719970B1 (en) 2000-07-10 2004-04-13 Alkermes Controlled Therapeutics, Inc. Method of generating cartilage
US6440141B1 (en) 2000-07-24 2002-08-27 Oratec Interventions, Inc. Method and apparatus for treating osteochondral pathologies
US8366787B2 (en) * 2000-08-04 2013-02-05 Depuy Products, Inc. Hybrid biologic-synthetic bioabsorbable scaffolds
US6638312B2 (en) 2000-08-04 2003-10-28 Depuy Orthopaedics, Inc. Reinforced small intestinal submucosa (SIS)
US6599526B2 (en) * 2000-08-18 2003-07-29 The University Of North Texas Health Science Center At Fort Worth Pericardial anti-adhesion patch
US7671018B2 (en) 2000-08-30 2010-03-02 University Of Delaware Delivery system for heparin-binding growth factors
ATE426357T1 (en) 2000-09-14 2009-04-15 Univ Leland Stanford Junior ASSESSING THE CONDITION OF A JOINT AND PLANNING TREATMENT
US7560280B2 (en) * 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
US20090130066A1 (en) * 2000-11-06 2009-05-21 Gerigene Medical Corporation Augmentation and repair of sphincter defects with cells including muscle cells
US20040091540A1 (en) * 2000-11-15 2004-05-13 Desrosiers Eric Andre Method for restoring a damaged or degenerated intervertebral disc
WO2002041786A2 (en) 2000-11-20 2002-05-30 Senorx, Inc. Tissue site markers for in vivo imaging
US6852330B2 (en) 2000-12-21 2005-02-08 Depuy Mitek, Inc. Reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
US7553662B2 (en) * 2000-12-22 2009-06-30 Keele University Culturing tissue using magnetically generated mechanical stresses
US7192604B2 (en) * 2000-12-22 2007-03-20 Ethicon, Inc. Implantable biodegradable devices for musculoskeletal repair or regeneration
AU2002219226B2 (en) * 2000-12-28 2007-03-22 Fidia Advanced Biopolymers S.R.L. Use of a biological material containing three-dimensional scaffolds of hyaluronic acid derivatives for the preparation of implants in arthroscopy and kit for instruments for implanting said biological materials by arthroscopy
US6911202B2 (en) * 2001-02-06 2005-06-28 Abraham Amir Cosmetic repair using cartilage producing cells and medical implants coated therewith
WO2002067867A2 (en) * 2001-02-23 2002-09-06 The University Of Pittsburgh Rapid preparation of stem cell matrices for use in tissue and organ treatment and repair
US6827743B2 (en) * 2001-02-28 2004-12-07 Sdgi Holdings, Inc. Woven orthopedic implants
KR20010044624A (en) * 2001-03-12 2001-06-05 정재호 A process for preparing the scaffold and tissue engineered cartilage made from the scaffold
US7029838B2 (en) 2001-03-30 2006-04-18 Arizona Board Of Regents On Behalf Of The University Of Arizona Prevascularized contructs for implantation to provide blood perfusion
US8951260B2 (en) 2001-05-25 2015-02-10 Conformis, Inc. Surgical cutting guide
US8439926B2 (en) 2001-05-25 2013-05-14 Conformis, Inc. Patient selectable joint arthroplasty devices and surgical tools
JP2005504563A (en) 2001-05-25 2005-02-17 イメージング セラピューティクス,インコーポレーテッド Methods and compositions for resurfacing joints
AU2002313694B2 (en) * 2001-07-16 2007-08-30 Depuy Products, Inc. Cartilage repair apparatus and method
EP1416866A4 (en) 2001-07-16 2007-04-18 Depuy Products Inc Devices form naturally occurring biologically derived
US8025896B2 (en) 2001-07-16 2011-09-27 Depuy Products, Inc. Porous extracellular matrix scaffold and method
WO2003007790A2 (en) 2001-07-16 2003-01-30 Depuy Products, Inc. Hybrid biologic/synthetic porous extracellular matrix scaffolds
US7201917B2 (en) 2001-07-16 2007-04-10 Depuy Products, Inc. Porous delivery scaffold and method
US7819918B2 (en) 2001-07-16 2010-10-26 Depuy Products, Inc. Implantable tissue repair device
EP1416886A4 (en) * 2001-07-16 2007-04-18 Depuy Products Inc Cartilage repair and regeneration scaffold and method
IL144446A0 (en) * 2001-07-19 2002-05-23 Prochon Biotech Ltd Plasma protein matrices and methods for their preparation
KR100494265B1 (en) * 2001-08-14 2005-06-13 메디포스트(주) Composition for treatment of articular cartilage damage
US7708741B1 (en) 2001-08-28 2010-05-04 Marctec, Llc Method of preparing bones for knee replacement surgery
WO2003028655A2 (en) * 2001-10-04 2003-04-10 Massachusetts Institute Of Technology Effect of bone morphogenetic proteins on engineered cartilage
US7303578B2 (en) 2001-11-01 2007-12-04 Photothera, Inc. Device and method for providing phototherapy to the brain
US10683494B2 (en) 2001-11-01 2020-06-16 Pthera LLC Enhanced stem cell therapy and stem cell production through the administration of low level light energy
US9993659B2 (en) 2001-11-01 2018-06-12 Pthera, Llc Low level light therapy for enhancement of neurologic function by altering axonal transport rate
US7534255B1 (en) 2003-01-24 2009-05-19 Photothera, Inc Low level light therapy for enhancement of neurologic function
US8308784B2 (en) 2006-08-24 2012-11-13 Jackson Streeter Low level light therapy for enhancement of neurologic function of a patient affected by Parkinson's disease
US10695577B2 (en) 2001-12-21 2020-06-30 Photothera, Inc. Device and method for providing phototherapy to the heart
US7316922B2 (en) 2002-01-09 2008-01-08 Photothera Inc. Method for preserving organs for transplant
AU2003219830A1 (en) * 2002-02-22 2003-09-09 Ebi, L.P. Methods and compositions for treating bone or cartilage defects
CN1653179B (en) * 2002-03-12 2012-07-25 组织基因股份有限公司 Cartilage regeneration using chondrocyte and TGF-beta
US7431922B2 (en) * 2002-03-29 2008-10-07 Tissuegene, Inc. Bioadhesive directed somatic cell therapy
US7005127B2 (en) * 2002-03-29 2006-02-28 Tissuegene, Inc. Mixed-cell gene therapy
CA2853267C (en) 2002-04-08 2018-06-26 Octane Biotech Inc. Automated tissue engineering system comprising sensors linked to a microprocessor
US7223289B2 (en) * 2002-04-16 2007-05-29 Warsaw Orthopedic, Inc. Annulus repair systems and techniques
PL373554A1 (en) * 2002-05-01 2005-09-05 Verigen Ag Injectable chondrocyte implant
JP4067880B2 (en) * 2002-06-17 2008-03-26 オリンパス株式会社 Cell culture method
US7622562B2 (en) 2002-06-26 2009-11-24 Zimmer Orthobiologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
ATE481110T1 (en) * 2002-07-16 2010-10-15 Biosyntech Canada Inc COMPOSITION FOR THE PRODUCTION OF CELL-COMPATIBLE, INJECTABLE, SELF-GELELLING CHITOSAN SOLUTIONS FOR ENCAPSULATING AND ADMINISTERING LIVING CELLS OR BIOLOGICALLY ACTIVE FACTORS
US20040136968A1 (en) * 2002-09-27 2004-07-15 Verigen Ag Autologous cells on a support matrix for tissue repair
US20040062753A1 (en) * 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
EP1555962B1 (en) 2002-10-07 2011-02-09 Conformis, Inc. Minimally invasive joint implant with 3-dimensional geometry matching the articular surfaces
US7824701B2 (en) 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
JP2006505366A (en) 2002-11-07 2006-02-16 コンフォーミス・インコーポレイテッド Method of determining meniscus size and shape and devised treatment
US20060036158A1 (en) 2003-11-17 2006-02-16 Inrad, Inc. Self-contained, self-piercing, side-expelling marking apparatus
US20040127402A1 (en) * 2002-12-27 2004-07-01 Vad Vijay B. Injectible composition and method for treating degenerative animal joints
US8551100B2 (en) 2003-01-15 2013-10-08 Biomet Manufacturing, Llc Instrumentation for knee resection
US7789885B2 (en) 2003-01-15 2010-09-07 Biomet Manufacturing Corp. Instrumentation for knee resection
US7837690B2 (en) 2003-01-15 2010-11-23 Biomet Manufacturing Corp. Method and apparatus for less invasive knee resection
US7887542B2 (en) 2003-01-15 2011-02-15 Biomet Manufacturing Corp. Method and apparatus for less invasive knee resection
AU2004208038B2 (en) 2003-01-30 2007-09-06 Prochon Biotech Ltd. Freeze-dried fibrin matrices and methods for preparation thereof
AU2003900620A0 (en) * 2003-02-12 2003-02-27 Australian Surgical Design And Manufacture Pty Limited Arthroscopic chondrocyte implantation method and device
US7642092B2 (en) * 2003-03-03 2010-01-05 Technion Research & Development Foundation Ltd. Cultured cartilage/bone cells/tissue, method of generating same and uses thereof
US7344555B2 (en) 2003-04-07 2008-03-18 The United States Of America As Represented By The Department Of Health And Human Services Light promotes regeneration and functional recovery after spinal cord injury
JP2006524072A (en) * 2003-04-21 2006-10-26 ベリーゲン アーゲー Seeded tear-resistant scaffold
CN103230415B (en) * 2003-04-25 2016-05-04 匹兹堡大学联邦制高等教育 For promoting and strengthen the neural muscle derived cell (MDC) of repairing and regenerating
US7067123B2 (en) * 2003-04-29 2006-06-27 Musculoskeletal Transplant Foundation Glue for cartilage repair
US8034048B2 (en) 2003-05-05 2011-10-11 Boston Scientific Scimed, Inc. Tissue patches and related delivery systems and methods
US7416546B2 (en) * 2003-05-05 2008-08-26 Boston Scientific Scimed, Inc. Tissue patches and related delivery systems and methods
WO2004101002A2 (en) 2003-05-08 2004-11-25 Tepha, Inc. Polyhydroxyalkanoate medical textiles and fibers
US7553827B2 (en) * 2003-08-13 2009-06-30 Depuy Spine, Inc. Transdiscal administration of cycline compounds
US20040229878A1 (en) * 2003-05-13 2004-11-18 Depuy Spine, Inc. Transdiscal administration of specific inhibitors of P38 kinase
US7344716B2 (en) * 2003-05-13 2008-03-18 Depuy Spine, Inc. Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US7429378B2 (en) * 2003-05-13 2008-09-30 Depuy Spine, Inc. Transdiscal administration of high affinity anti-MMP inhibitors
US8273347B2 (en) * 2003-05-13 2012-09-25 Depuy Spine, Inc. Autologous treatment of degenerated disc with cells
US7901457B2 (en) 2003-05-16 2011-03-08 Musculoskeletal Transplant Foundation Cartilage allograft plug
US7877133B2 (en) 2003-05-23 2011-01-25 Senorx, Inc. Marker or filler forming fluid
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US7413734B2 (en) 2003-06-27 2008-08-19 Ethicon, Incorporated Treatment of retinitis pigmentosa with human umbilical cord cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US8361467B2 (en) 2003-07-30 2013-01-29 Depuy Spine, Inc. Trans-capsular administration of high specificity cytokine inhibitors into orthopedic joints
US7897384B2 (en) * 2003-09-08 2011-03-01 Ethicon, Inc. Chondrocyte therapeutic delivery system
US8257963B2 (en) 2007-06-01 2012-09-04 Depuy Mitek, Inc. Chondrocyte container and method of use
US7927599B2 (en) * 2003-09-08 2011-04-19 Ethicon, Inc. Chondrocyte therapeutic delivery system
DE10349722A1 (en) * 2003-10-23 2005-06-16 Beschorner, Katharina, Dr. Composition for arthritis / arthritis treatment, in particular of joints
US20050273002A1 (en) 2004-06-04 2005-12-08 Goosen Ryan L Multi-mode imaging marker
US8895540B2 (en) 2003-11-26 2014-11-25 DePuy Synthes Products, LLC Local intraosseous administration of bone forming agents and anti-resorptive agents, and devices therefor
US7316822B2 (en) 2003-11-26 2008-01-08 Ethicon, Inc. Conformable tissue repair implant capable of injection delivery
US7901461B2 (en) * 2003-12-05 2011-03-08 Ethicon, Inc. Viable tissue repair implants and methods of use
US7488324B1 (en) 2003-12-08 2009-02-10 Biomet Manufacturing Corporation Femoral guide for implanting a femoral knee prosthesis
WO2005058207A1 (en) * 2003-12-11 2005-06-30 Isto Technologies, Inc. Particulate cartilage system
GB0329310D0 (en) * 2003-12-18 2004-01-21 Univ Keele Method
AU2004308927A1 (en) * 2003-12-19 2005-07-14 Viacell, Inc. Use of human cord blood-derived pluripotent cells for the treatment of disease
GB0402838D0 (en) * 2004-02-10 2004-03-17 Univ Belfast Method
US20070185585A1 (en) * 2004-03-09 2007-08-09 Brat Bracy Implant Scaffold Combined With Autologous Tissue, Allogenic Tissue, Cultured Tissue, or combinations Thereof
CA2555586A1 (en) * 2004-03-09 2005-09-22 Osteobiologics, Inc. Implant scaffold combined with autologous or allogenic tissue
RU2252252C1 (en) * 2004-04-09 2005-05-20 Тепляшин Александр Сергеевич Method for isolation of mesenchymal stem cells
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8657881B2 (en) 2004-04-20 2014-02-25 Depuy Mitek, Llc Meniscal repair scaffold
US8221780B2 (en) 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
WO2005105992A1 (en) * 2004-04-21 2005-11-10 New York Eye & Ear Infirmary Chondrocyte culture formulations
EP1740123A4 (en) * 2004-04-26 2008-09-03 Howmedica Osteonics Corp Stent for avascular meniscal repair and regeneration
US20050288796A1 (en) * 2004-06-23 2005-12-29 Hani Awad Native soft tissue matrix for therapeutic applications
US7632284B2 (en) * 2004-07-06 2009-12-15 Tyco Healthcare Group Lp Instrument kit and method for performing meniscal repair
US8637065B2 (en) * 2004-07-09 2014-01-28 William Marsh Rice University Dermis-derived cells for tissue engineering applications
JP4949241B2 (en) * 2004-07-12 2012-06-06 イスト・テクノロジーズ・インコーポレイテッド Tissue matrix system
US8512730B2 (en) * 2004-07-12 2013-08-20 Isto Technologies, Inc. Methods of tissue repair and compositions therefor
US20090181092A1 (en) * 2004-07-16 2009-07-16 Spinal Restoration, Inc. Methods for Treating Joints and Discs with a Carrier Matrix and Cells
US7335508B2 (en) * 2004-07-22 2008-02-26 Prochon Biotech Ltd. Porous plasma protein matrices and methods for preparation thereof
EP1778305B1 (en) 2004-08-03 2010-07-07 Tepha, Inc. Non-curling polyhydroxyalkanoate sutures
US7785582B2 (en) * 2004-09-07 2010-08-31 Johnson Lanny L Use of synovium and omentum for tissue engineering
US8182806B2 (en) * 2004-09-07 2012-05-22 Johnson Lanny L Synovial villi for use with tissue engineering
GB2432845A (en) * 2004-09-21 2007-06-06 Massachusetts Inst Technology Gradient scaffolding and methods of producing the same
US7837740B2 (en) 2007-01-24 2010-11-23 Musculoskeletal Transplant Foundation Two piece cancellous construct for cartilage repair
US20060111778A1 (en) * 2004-10-29 2006-05-25 Michalow Alexander E Methods of promoting healing of cartilage defects and method of causing stem cells to differentiate by the articular chondrocyte pathway
US7513866B2 (en) 2004-10-29 2009-04-07 Depuy Products, Inc. Intestine processing device and associated method
US20060097422A1 (en) * 2004-11-08 2006-05-11 Diamond Andrew J Method for performing surgery and appliances produced thereby
US9981063B2 (en) * 2004-11-24 2018-05-29 Mayo Foundation For Medical Education And Research Biosynthetic composite for osteochondral defect repair
PL1831356T3 (en) 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US7695479B1 (en) 2005-04-12 2010-04-13 Biomet Manufacturing Corp. Femoral sizer
US10357328B2 (en) 2005-04-20 2019-07-23 Bard Peripheral Vascular, Inc. and Bard Shannon Limited Marking device with retractable cannula
US7815926B2 (en) 2005-07-11 2010-10-19 Musculoskeletal Transplant Foundation Implant for articular cartilage repair
KR100774089B1 (en) * 2005-07-20 2007-11-06 세원셀론텍(주) Simple Method of Autologous Chondrocyte Transplantation ? Injectable Chondrocyte Transplantation
US8048297B2 (en) 2005-08-23 2011-11-01 Biomet Biologics, Llc Method and apparatus for collecting biological materials
EP1916964A4 (en) 2005-08-26 2015-11-04 Zimmer Inc Implants and methods for repair, replacement and treatment of joint disease
KR20080065606A (en) * 2005-09-02 2008-07-14 인터페이스 바이오텍 에이/에스 A method for cell implantation
US7927630B2 (en) * 2005-09-12 2011-04-19 Johnson Lanny L Use of autologous sediment from fluid aspirates as vehicles for drug delivery
US8518349B2 (en) 2005-09-12 2013-08-27 Lanny Johnson Use of autologous sediment from fluid aspirates as vehicles for drug delivery
US20070065415A1 (en) * 2005-09-16 2007-03-22 Kleinsek Donald A Compositions and methods for the augmentation and repair of defects in tissue
AU2006292224B2 (en) 2005-09-19 2013-08-01 Histogenics Corporation Cell-support matrix and a method for preparation thereof
EP1928359A4 (en) 2005-09-30 2010-10-13 Conformis Inc Joint arthroplasty devices
CA2562580C (en) 2005-10-07 2014-04-29 Inrad, Inc. Drug-eluting tissue marker
US20090149421A1 (en) * 2005-11-04 2009-06-11 Bio Syntech Canada Inc. Gel formation of polyelectrolyte aqueous solutions by thermally induced changes in ionization state
US20100209397A1 (en) * 2005-11-10 2010-08-19 Carticure Ltd. Method for non-autologous cartilage regeneration
EP1951158A4 (en) 2005-11-21 2010-03-31 Vertegen Inc Devices and methods for treating facet joints, uncovertebral joints, costovertebral joints and other joints
CA2631520A1 (en) * 2005-12-07 2007-06-14 Isto Technologies, Inc. Cartilage repair methods
WO2007070870A1 (en) 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
WO2007073552A1 (en) 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
CA2640185A1 (en) * 2006-01-24 2007-08-02 Christopher J. Centeno Mesenchymal stem cell isolation and transplantation method and system to be used in a clinical setting
US7776043B2 (en) * 2006-01-26 2010-08-17 Warsaw Orthopedic, Inc. Osteochondral implant fixation procedure and bone dilator used in same
US7575589B2 (en) 2006-01-30 2009-08-18 Photothera, Inc. Light-emitting device and method for providing phototherapy to the brain
US10357662B2 (en) 2009-02-19 2019-07-23 Pthera LLC Apparatus and method for irradiating a surface with light
US20090254154A1 (en) 2008-03-18 2009-10-08 Luis De Taboada Method and apparatus for irradiating a surface with pulsed light
WO2007092841A2 (en) 2006-02-06 2007-08-16 Conformis, Inc. Patient selectable joint arthroplasty devices and surgical tools
US8623026B2 (en) 2006-02-06 2014-01-07 Conformis, Inc. Patient selectable joint arthroplasty devices and surgical tools incorporating anatomical relief
US8070752B2 (en) 2006-02-27 2011-12-06 Biomet Manufacturing Corp. Patient specific alignment guide and inter-operative adjustment
US9907659B2 (en) 2007-04-17 2018-03-06 Biomet Manufacturing, Llc Method and apparatus for manufacturing an implant
US9173661B2 (en) 2006-02-27 2015-11-03 Biomet Manufacturing, Llc Patient specific alignment guide with cutting surface and laser indicator
US10278711B2 (en) 2006-02-27 2019-05-07 Biomet Manufacturing, Llc Patient-specific femoral guide
US7780672B2 (en) 2006-02-27 2010-08-24 Biomet Manufacturing Corp. Femoral adjustment device and associated method
US9113971B2 (en) 2006-02-27 2015-08-25 Biomet Manufacturing, Llc Femoral acetabular impingement guide
US8591516B2 (en) 2006-02-27 2013-11-26 Biomet Manufacturing, Llc Patient-specific orthopedic instruments
US9289253B2 (en) 2006-02-27 2016-03-22 Biomet Manufacturing, Llc Patient-specific shoulder guide
US20150335438A1 (en) 2006-02-27 2015-11-26 Biomet Manufacturing, Llc. Patient-specific augments
US9345548B2 (en) 2006-02-27 2016-05-24 Biomet Manufacturing, Llc Patient-specific pre-operative planning
US9918740B2 (en) 2006-02-27 2018-03-20 Biomet Manufacturing, Llc Backup surgical instrument system and method
US8407067B2 (en) 2007-04-17 2013-03-26 Biomet Manufacturing Corp. Method and apparatus for manufacturing an implant
US8603180B2 (en) 2006-02-27 2013-12-10 Biomet Manufacturing, Llc Patient-specific acetabular alignment guides
US9339278B2 (en) 2006-02-27 2016-05-17 Biomet Manufacturing, Llc Patient-specific acetabular guides and associated instruments
US7695520B2 (en) 2006-05-31 2010-04-13 Biomet Manufacturing Corp. Prosthesis and implementation system
US9795399B2 (en) 2006-06-09 2017-10-24 Biomet Manufacturing, Llc Patient-specific knee alignment guide and associated method
KR100803576B1 (en) * 2006-06-14 2008-02-15 주식회사 인피트론 A composition for transplant comprising adipose stem cells and adipocytes
US20080294039A1 (en) * 2006-08-04 2008-11-27 Senorx, Inc. Assembly with hemostatic and radiographically detectable pellets
CA2660117C (en) * 2006-08-07 2012-07-03 Howmedica Osteonics Corp. Insertion system for implanting a medical device and surgical methods
US20080033487A1 (en) * 2006-08-07 2008-02-07 Bioduct, Llc Medical device for repair of tissue and method for implantation and fixation
US20100040667A1 (en) * 2006-09-07 2010-02-18 Ed. Geistlich Soehne Ag Fuer Chemische Industrie Method of treating bone cancer
ES2443526T3 (en) 2006-10-23 2014-02-19 C.R. Bard, Inc. Breast marker
US7943683B2 (en) 2006-12-01 2011-05-17 Tepha, Inc. Medical devices containing oriented films of poly-4-hydroxybutyrate and copolymers
US20080138414A1 (en) * 2006-12-08 2008-06-12 Smith & Nephew, Inc. Methods of Regenerating Cartilage
US9579077B2 (en) 2006-12-12 2017-02-28 C.R. Bard, Inc. Multiple imaging mode tissue marker
US8323642B2 (en) * 2006-12-13 2012-12-04 Depuy Mitek, Inc. Tissue fusion method using collagenase for repair of soft tissue
EP2101670B1 (en) 2006-12-18 2013-07-31 C.R.Bard, Inc. Biopsy marker with in situ-generated imaging properties
US8163549B2 (en) * 2006-12-20 2012-04-24 Zimmer Orthobiologics, Inc. Method of obtaining viable small tissue particles and use for tissue repair
US20080193424A1 (en) * 2007-02-09 2008-08-14 Biomet Biologics, Inc. Treatment of tissue defects with a therapeutic composition
US8435551B2 (en) 2007-03-06 2013-05-07 Musculoskeletal Transplant Foundation Cancellous construct with support ring for repair of osteochondral defects
US8034014B2 (en) 2007-03-06 2011-10-11 Biomet Biologics, Llc Angiogenesis initation and growth
US20090012629A1 (en) 2007-04-12 2009-01-08 Isto Technologies, Inc. Compositions and methods for tissue repair
EP2155860B1 (en) * 2007-05-03 2014-08-27 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US20080281419A1 (en) * 2007-05-10 2008-11-13 Matheny Robert G Breast implants and compositions of extracellular matrix
US20090004253A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Composite device for the repair or regeneration of tissue
US9095562B2 (en) * 2007-07-05 2015-08-04 Regenerative Sciences, Inc. Methods and compositions for optimized expansion and implantation of mesenchymal stem cells
UA99152C2 (en) 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
CN101945994A (en) * 2007-12-19 2011-01-12 再生科学有限责任公司 Compositions and methods to promote implantation and engrafment of stem cells
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US8986696B2 (en) 2007-12-21 2015-03-24 Depuy Mitek, Inc. Trans-capsular administration of p38 map kinase inhibitors into orthopedic joints
WO2009099767A2 (en) 2008-01-31 2009-08-13 C.R. Bard, Inc. Biopsy tissue marker
US8753690B2 (en) * 2008-02-27 2014-06-17 Biomet Biologics, Llc Methods and compositions for delivering interleukin-1 receptor antagonist
EP2567692B1 (en) 2008-02-27 2016-04-06 Biomet Biologics, LLC Use of a device for obtaining interleukin-1 receptor antagonist rich solutions
CA2717725A1 (en) 2008-03-05 2009-09-11 Musculoskeletal Transplant Foundation Cancellous constructs, cartilage particles and combinations of cancellous constructs and cartilage particles
US8682052B2 (en) 2008-03-05 2014-03-25 Conformis, Inc. Implants for altering wear patterns of articular surfaces
EP2257176B1 (en) 2008-03-14 2013-09-18 Regenerative Sciences, LLC Compositions and methods for cartilage repair
EP2303193A4 (en) 2008-05-12 2012-03-21 Conformis Inc Devices and methods for treatment of facet and other joints
AU2009282619B2 (en) * 2008-08-18 2015-08-20 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Bone augmentation utilizing muscle-derived progenitor compositions in biocompatible matrix, and treatments thereof
US7848035B2 (en) 2008-09-18 2010-12-07 Photothera, Inc. Single-use lens assembly
US9327061B2 (en) 2008-09-23 2016-05-03 Senorx, Inc. Porous bioabsorbable implant
US8163022B2 (en) 2008-10-14 2012-04-24 Anulex Technologies, Inc. Method and apparatus for the treatment of the intervertebral disc annulus
WO2010048418A1 (en) * 2008-10-22 2010-04-29 The Trustees Of Columbia University In The City Of New York Cartilage regeneration without cell transplantation
CN102292092B (en) 2008-11-25 2014-05-07 组织基因公司 Primed cell therapy
US20110245804A1 (en) 2008-12-05 2011-10-06 Regenerative Sciences, Llc Methods and Compositions to Facilitate Repair of Avascular Tissue
US20100168022A1 (en) * 2008-12-11 2010-07-01 Centeno Christopher J Use of In-Vitro Culture to Design or Test Personalized Treatment Regimens
US20100151114A1 (en) * 2008-12-17 2010-06-17 Zimmer, Inc. In-line treatment of yarn prior to creating a fabric
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
CA2747794C (en) 2008-12-19 2018-10-30 Advanced Technologies And Regenerative Medicine, Llc Treatment of lung and pulmonary diseases and disorders
WO2010077244A1 (en) 2008-12-30 2010-07-08 C.R. Bard Inc. Marker delivery device for tissue marker placement
US20100291182A1 (en) * 2009-01-21 2010-11-18 Arsenal Medical, Inc. Drug-Loaded Fibers
WO2010099231A2 (en) 2009-02-24 2010-09-02 Conformis, Inc. Automated systems for manufacturing patient-specific orthopedic implants and instrumentation
US9017334B2 (en) 2009-02-24 2015-04-28 Microport Orthopedics Holdings Inc. Patient specific surgical guide locator and mount
US8808297B2 (en) 2009-02-24 2014-08-19 Microport Orthopedics Holdings Inc. Orthopedic surgical guide
US8808303B2 (en) 2009-02-24 2014-08-19 Microport Orthopedics Holdings Inc. Orthopedic surgical guide
BRPI1013409A2 (en) 2009-03-26 2018-01-16 Advanced Tech And Regenerative Medicine Llc human umbilical cord tissue cells as therapy for alzheimer's disease
SG175229A1 (en) 2009-04-16 2011-11-28 Conformis Inc Patient-specific joint arthroplasty devices for ligament repair
US20100316614A1 (en) * 2009-06-16 2010-12-16 Northwestern University Compositions and methods for urinary bladder regeneration
US9173817B2 (en) 2009-08-24 2015-11-03 Arsenal Medical, Inc. In situ forming hemostatic foam implants
US10420862B2 (en) 2009-08-24 2019-09-24 Aresenal AAA, LLC. In-situ forming foams for treatment of aneurysms
US9044580B2 (en) 2009-08-24 2015-06-02 Arsenal Medical, Inc. In-situ forming foams with outer layer
US20110202016A1 (en) * 2009-08-24 2011-08-18 Arsenal Medical, Inc. Systems and methods relating to polymer foams
US9763875B2 (en) 2009-08-27 2017-09-19 Biomet Biologics, Llc Implantable device for production of interleukin-1 receptor antagonist
US20110052561A1 (en) * 2009-08-27 2011-03-03 Biomet Biologics,LLC Osteolysis treatment
US20110054929A1 (en) * 2009-09-01 2011-03-03 Cell Solutions Colorado Llc Stem Cell Marketplace
US8377432B2 (en) * 2009-09-02 2013-02-19 Khay-Yong Saw Method and composition for neochondrogenesis
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
EP2509539B1 (en) 2009-12-11 2020-07-01 ConforMIS, Inc. Patient-specific and patient-engineered orthopedic implants
US8652153B2 (en) 2010-01-11 2014-02-18 Anulex Technologies, Inc. Intervertebral disc annulus repair system and bone anchor delivery tool
KR20120113265A (en) 2010-01-15 2012-10-12 유니버시티 오브 메디신 앤드 덴티스트리 오브 뉴 저지 Use of vanadium compounds to accelerate bone healing
EP2542249A4 (en) 2010-03-05 2013-08-07 Tissue Genesis Inc Methods and compositions to support tissue integration and inosculation of transplanted tissue and transplanted engineered penile tissue with adipose stromal cells
US8697111B2 (en) * 2010-05-12 2014-04-15 Covidien Lp Osteochondral implant comprising osseous phase and chondral phase
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
US9968376B2 (en) 2010-11-29 2018-05-15 Biomet Manufacturing, Llc Patient-specific orthopedic instruments
US20140322292A1 (en) 2010-12-10 2014-10-30 Rutgers, The State University Of New Jersey Insulin-mimetics as therapeutic adjuncts for bone regeneration
WO2013006798A1 (en) * 2011-07-06 2013-01-10 University Of Medicine And Dentistry Of New Jersey Vandium compounds as therapeutic adjuncts for cartilage regeneration and repair
NZ612801A (en) 2010-12-27 2014-12-24 Intellicell Biosciences Inc Ultrasonic cavitation derived stromal or mesenchymal vascular extracts and cells derived therefrom obtained from adipose tissue and use thereof
US9194058B2 (en) 2011-01-31 2015-11-24 Arsenal Medical, Inc. Electrospinning process for manufacture of multi-layered structures
US9034240B2 (en) 2011-01-31 2015-05-19 Arsenal Medical, Inc. Electrospinning process for fiber manufacture
US8968626B2 (en) 2011-01-31 2015-03-03 Arsenal Medical, Inc. Electrospinning process for manufacture of multi-layered structures
EP2754419B1 (en) 2011-02-15 2024-02-07 ConforMIS, Inc. Patient-adapted and improved orthopedic implants
US9241745B2 (en) 2011-03-07 2016-01-26 Biomet Manufacturing, Llc Patient-specific femoral version guide
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
CN103930542A (en) 2011-06-29 2014-07-16 生物修复疗法有限公司 Brown fat cell compositions and methods
KR101360162B1 (en) * 2011-06-30 2014-02-11 김지형 A Method for Proliferating Chondrocytes For Meniscus Transplantation
US8993831B2 (en) 2011-11-01 2015-03-31 Arsenal Medical, Inc. Foam and delivery system for treatment of postpartum hemorrhage
MX362198B (en) 2011-12-23 2019-01-08 Depuy Synthes Products Llc Detection of human umbilical cord tissue-derived cells.
US9486226B2 (en) 2012-04-18 2016-11-08 Conformis, Inc. Tibial guides, tools, and techniques for resecting the tibial plateau
US9675471B2 (en) 2012-06-11 2017-06-13 Conformis, Inc. Devices, techniques and methods for assessing joint spacing, balancing soft tissues and obtaining desired kinematics for joint implant components
US10245306B2 (en) 2012-11-16 2019-04-02 Isto Technologies Ii, Llc Flexible tissue matrix and methods for joint repair
US20140178343A1 (en) 2012-12-21 2014-06-26 Jian Q. Yao Supports and methods for promoting integration of cartilage tissue explants
US9737294B2 (en) 2013-01-28 2017-08-22 Cartiva, Inc. Method and system for orthopedic repair
WO2014117107A1 (en) 2013-01-28 2014-07-31 Cartiva, Inc. Systems and methods for orthopedic repair
US9895418B2 (en) 2013-03-15 2018-02-20 Biomet Biologics, Llc Treatment of peripheral vascular disease using protein solutions
US10143725B2 (en) 2013-03-15 2018-12-04 Biomet Biologics, Llc Treatment of pain using protein solutions
US20140271589A1 (en) 2013-03-15 2014-09-18 Biomet Biologics, Llc Treatment of collagen defects using protein solutions
US9950035B2 (en) 2013-03-15 2018-04-24 Biomet Biologics, Llc Methods and non-immunogenic compositions for treating inflammatory disorders
US9878011B2 (en) 2013-03-15 2018-01-30 Biomet Biologics, Llc Treatment of inflammatory respiratory disease using biological solutions
US9758806B2 (en) 2013-03-15 2017-09-12 Biomet Biologics, Llc Acellular compositions for treating inflammatory disorders
US10208095B2 (en) 2013-03-15 2019-02-19 Biomet Manufacturing, Llc Methods for making cytokine compositions from tissues using non-centrifugal methods
USD716450S1 (en) 2013-09-24 2014-10-28 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
USD715442S1 (en) 2013-09-24 2014-10-14 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
USD715942S1 (en) 2013-09-24 2014-10-21 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
USD716451S1 (en) 2013-09-24 2014-10-28 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
US9833474B2 (en) 2013-11-26 2017-12-05 Biomet Biologies, LLC Methods of mediating macrophage phenotypes
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
US10500303B2 (en) 2014-08-15 2019-12-10 Tepha, Inc. Self-retaining sutures of poly-4-hydroxybutyrate and copolymers thereof
AU2015320722A1 (en) 2014-09-23 2017-04-13 Cytex Therapeutics, Inc. Articular cartilage repair
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
US10179191B2 (en) 2014-10-09 2019-01-15 Isto Technologies Ii, Llc Flexible tissue matrix and methods for joint repair
US10441635B2 (en) 2014-11-10 2019-10-15 Biomet Biologics, Llc Methods of treating pain using protein solutions
US10077420B2 (en) 2014-12-02 2018-09-18 Histogenics Corporation Cell and tissue culture container
US10626521B2 (en) 2014-12-11 2020-04-21 Tepha, Inc. Methods of manufacturing mesh sutures from poly-4-hydroxybutyrate and copolymers thereof
CA2969429C (en) 2014-12-11 2020-10-27 Tepha, Inc. Methods of orienting multifilament yarn and monofilaments of poly-4-hydroxybutyrate and copolymers thereof
US9763800B2 (en) 2015-03-18 2017-09-19 Biomet C. V. Implant configured for hammertoe and small bone fixation
EP3297694A1 (en) 2015-05-21 2018-03-28 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US10022231B2 (en) * 2016-07-22 2018-07-17 Cytex Therapeutics, Inc. Articular cartilage repair
US10722310B2 (en) 2017-03-13 2020-07-28 Zimmer Biomet CMF and Thoracic, LLC Virtual surgery planning system and method
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
CN111373030A (en) 2017-09-01 2020-07-03 隆萨沃克斯维尔股份有限公司 End-to-end cell therapy automation
JP2022514761A (en) 2018-12-21 2022-02-15 ロンザ ウォーカーズヴィル,インコーポレーテッド Automatic production method of viral vector
CN113366099A (en) 2018-12-21 2021-09-07 奥克泰生物科技股份有限公司 Carousel for modular bio-production units
SG11202106652WA (en) 2018-12-28 2021-07-29 Octane Biotech Inc Cell culture and tissue engineering systems with controlled environmental zones
KR20210125510A (en) 2019-02-08 2021-10-18 론자 워커스빌 아이엔씨. Cell Concentration Methods and Apparatus for Use in Automated Bioreactors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4846835A (en) * 1987-06-15 1989-07-11 Grande Daniel A Technique for healing lesions in cartilage
WO1993015694A1 (en) * 1992-02-14 1993-08-19 Board Of Regents, The University Of Texas System Multi-phase bioerodible implant/carrier and method of manufacturing and using same
US5376118A (en) * 1989-05-10 1994-12-27 United States Surgical Corporation Support material for cell impregnation
US5460959A (en) * 1987-09-11 1995-10-24 Whitehead Institute For Biomedical Research Transduced fibroblasts

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4520821A (en) * 1982-04-30 1985-06-04 The Regents Of The University Of California Growing of long-term biological tissue correction structures in vivo
US4609551A (en) * 1984-03-20 1986-09-02 Arnold Caplan Process of and material for stimulating growth of cartilage and bony tissue at anatomical sites
US4963489A (en) * 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5160490A (en) * 1986-04-18 1992-11-03 Marrow-Tech Incorporated Three-dimensional cell and tissue culture apparatus
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US5032508A (en) * 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5510254A (en) * 1986-04-18 1996-04-23 Advanced Tissue Sciences, Inc. Three dimensional cell and tissue culture system
US5041138A (en) * 1986-11-20 1991-08-20 Massachusetts Institute Of Technology Neomorphogenesis of cartilage in vivo from cell culture
US5306311A (en) * 1987-07-20 1994-04-26 Regen Corporation Prosthetic articular cartilage
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5226914A (en) * 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5206023A (en) * 1991-01-31 1993-04-27 Robert F. Shaw Method and compositions for the treatment and repair of defects or lesions in cartilage
US5478739A (en) * 1992-10-23 1995-12-26 Advanced Tissue Sciences, Inc. Three-dimensional stromal cell and tissue culture system
US5723331A (en) * 1994-05-05 1998-03-03 Genzyme Corporation Methods and compositions for the repair of articular cartilage defects in mammals

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4846835A (en) * 1987-06-15 1989-07-11 Grande Daniel A Technique for healing lesions in cartilage
US5460959A (en) * 1987-09-11 1995-10-24 Whitehead Institute For Biomedical Research Transduced fibroblasts
US5376118A (en) * 1989-05-10 1994-12-27 United States Surgical Corporation Support material for cell impregnation
WO1993015694A1 (en) * 1992-02-14 1993-08-19 Board Of Regents, The University Of Texas System Multi-phase bioerodible implant/carrier and method of manufacturing and using same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CALCIFIED TISSUE INTERNATIONAL, 1993, Vol. 53, TANIGUCHI Y. et al., "Transforming Growth Factor beta1-Induced Cellular Heterogeneity in the Periosteum of Rat Bones", pages 122-126. *
See also references of EP0955959A4 *

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6569172B2 (en) 1996-08-30 2003-05-27 Verigen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
US7137989B2 (en) 1996-08-30 2006-11-21 Verigen Ag Method, instruments, and kit for autologous transplantation
US7048750B2 (en) 1996-08-30 2006-05-23 Verigen Ag Method, instruments, and kits for autologous transplantation
US6599301B2 (en) 1996-08-30 2003-07-29 Verrgen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
US6599300B2 (en) 1996-08-30 2003-07-29 Verigen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
US6283980B1 (en) 1996-08-30 2001-09-04 Verigen Transplantation Services Internt'l Method, instruments, and kit for autologous transplantation
US6592598B2 (en) 1996-08-30 2003-07-15 Verigen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
US6379367B1 (en) 1996-08-30 2002-04-30 Verigen Transplantation Service International (Vtsi) Ag Method instruments and kit for autologous transplantation
US6592599B2 (en) 1996-08-30 2003-07-15 Verigen Transplantation Service International (Vtsi) Method, instruments, and kit for autologous transplantation
JP2009102393A (en) * 1997-02-07 2009-05-14 Stryker Corp Matrix-free osteogenic devices, implants and methods of use thereof
WO1998042389A1 (en) * 1997-03-25 1998-10-01 Peter Villeneuve Materials for healing cartilage and bone defects
US6077987A (en) * 1997-09-04 2000-06-20 North Shore-Long Island Jewish Research Institute Genetic engineering of cells to enhance healing and tissue regeneration
US6398816B1 (en) 1997-09-04 2002-06-04 North Shore-Long Island Jewish Research Institute Genetic engineering of cells to enhance healing and tissue regeneration
WO1999011789A1 (en) * 1997-09-04 1999-03-11 North Shore University Hospital Research Corporation Genetic engineering of cells to enhance healing and tissue regeneration
WO1999025396A3 (en) * 1997-11-17 1999-07-29 Univ Michigan Hybrid tissues for tissue engineering
WO1999025396A2 (en) * 1997-11-17 1999-05-27 The Regents Of The University Of Michigan Hybrid tissues for tissue engineering
WO2000009179A3 (en) * 1998-08-14 2000-06-02 Verigen Transplantation Serv Methods, instruments and materials for chondrocyte cell transplantation
US6866668B2 (en) 1998-08-14 2005-03-15 Verigen Transplantation Service International (“VTSL”) AG Methods, instruments and materials for chondrocyte cell transplantation
EP1656960A1 (en) * 1998-08-14 2006-05-17 Verigen AG Methods, instruments and materials for chondrocyte cell transplantation
WO2000009179A2 (en) * 1998-08-14 2000-02-24 Verigen Transplantation Service International (Vtsi) Ag Methods, instruments and materials for chondrocyte cell transplantation
US8137689B1 (en) 1999-11-11 2012-03-20 Zimmer Gmbh Transplant/implant device and method for its production
JP2015128633A (en) * 2000-04-25 2015-07-16 メゾブラスト・インターナショナル・ソシエテ・ア・レスポンサビリテ・リミテ Usage of mesenchymal stem cell to pharmaceutical composition preparation for recovery of animal joint
US9050178B2 (en) 2000-04-25 2015-06-09 Mesoblast International Sàrl Joint repair using mesenchymal stem cells
JP2016216487A (en) * 2000-04-25 2016-12-22 メゾブラスト・インターナショナル・ソシエテ・ア・レスポンサビリテ・リミテ Method of using mesenchymal stem cells for preparing pharmaceutical compositions for joint repair in animal
US9814580B2 (en) 2000-04-25 2017-11-14 Mesoblast International Sarl Joint repair using mesenchymal stem cells
JP2012210437A (en) * 2000-04-25 2012-11-01 Osiris Therapeutics Inc Method for using mesenchymal stem cells in preparing medical composition for mammal joint repair
EP1276486B1 (en) * 2000-04-25 2010-11-24 Osiris Therapeutics, Inc. Joint repair using mesenchymal stem cells
EP2071020A3 (en) * 2000-07-29 2014-12-31 Smith&Nephew PLC Tissue Implant
WO2002010348A2 (en) * 2000-07-29 2002-02-07 Smith & Nephew Plc Tissue implant for cartilage repair
WO2002010348A3 (en) * 2000-07-29 2002-09-06 Smith & Nephew Tissue implant for cartilage repair
AU2007203472B2 (en) * 2000-07-29 2011-07-14 Smith & Nephew Plc Tissue implant for cartilage repair
AU2001275746B2 (en) * 2000-07-29 2007-04-26 Smith & Nephew Plc Tissue implant for cartilage repair
US9452238B2 (en) 2000-07-29 2016-09-27 Smith & Nephew LLP Tissue implant
US8691259B2 (en) 2000-12-21 2014-04-08 Depuy Mitek, Llc Reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
EP1416888A2 (en) * 2001-07-16 2004-05-12 Depuy Products, Inc. Meniscus regeneration device and method
AU2002354911B2 (en) * 2001-07-16 2007-08-30 Depuy Products, Inc. Meniscus regeneration device and method
EP1416888A4 (en) * 2001-07-16 2007-04-25 Depuy Products Inc Meniscus regeneration device and method
EP1416879A4 (en) * 2001-07-16 2007-04-25 Depuy Products Inc Unitary surgical device and method
EP1416879A2 (en) * 2001-07-16 2004-05-12 Depuy Products, Inc. Unitary surgical device and method
US7445793B2 (en) 2002-09-09 2008-11-04 Kaneka Corporation Support for tissue regeneration and process for producing the same
US9511171B2 (en) 2002-10-18 2016-12-06 Depuy Mitek, Llc Biocompatible scaffolds with tissue fragments
US10603408B2 (en) 2002-10-18 2020-03-31 DePuy Synthes Products, Inc. Biocompatible scaffolds with tissue fragments
US8173162B2 (en) 2003-02-26 2012-05-08 Zimmer Orthobiologics, Inc. Preparation for repairing cartilage tissue, especially articular cartilage defects
US8895045B2 (en) 2003-03-07 2014-11-25 Depuy Mitek, Llc Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
US9486558B2 (en) 2003-03-27 2016-11-08 Locate Therapeutics Limited Porous matrix
US9211362B2 (en) 2003-06-30 2015-12-15 Depuy Mitek, Llc Scaffold for connective tissue repair
US10583220B2 (en) 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
US8945535B2 (en) 2005-09-20 2015-02-03 Zimmer Orthobiologics, Inc. Implant for the repair of a cartilage defect and method for manufacturing the implant
US20100189712A1 (en) * 2006-11-17 2010-07-29 Cytograft Tissue Engineering, Inc. Preparation And Use Of Cell-Synthesized Threads
US7871440B2 (en) 2006-12-11 2011-01-18 Depuy Products, Inc. Unitary surgical device and method
US9238090B1 (en) 2014-12-24 2016-01-19 Fettech, Llc Tissue-based compositions
US11938246B2 (en) 2014-12-24 2024-03-26 Fettech, Llc Tissue-based compositions and methods of use thereof

Also Published As

Publication number Publication date
EP0955959A4 (en) 1999-11-17
EP0955959A1 (en) 1999-11-17
US5842477A (en) 1998-12-01
JP2000505338A (en) 2000-05-09
NZ331517A (en) 2000-01-28
CA2247158A1 (en) 1997-08-28
KR19990087147A (en) 1999-12-15
AU715282B2 (en) 2000-01-20
AU1973197A (en) 1997-09-10

Similar Documents

Publication Publication Date Title
US5842477A (en) Method for repairing cartilage
US11357889B2 (en) Native soft tissue matrix for therapeutic applications
US7078232B2 (en) Adipose tissue-derived adult stem or stromal cells for the repair of articular cartilage fractures and uses thereof
US4904259A (en) Compositions and methods for repair of cartilage and bone
AU635025B2 (en) Neomorphogenesis of cartilage in vivo from cell culture
Radice et al. Hyaluronan‐based biopolymers as delivery vehicles for bone‐marrow‐derived mesenchymal progenitors
US5919702A (en) Production of cartilage tissue using cells isolated from Wharton's jelly
US8796019B2 (en) Tissue repair with multipotent cells
US6482231B1 (en) Biological material for the repair of connective tissue defects comprising mesenchymal stem cells and hyaluronic acid derivative
US5736372A (en) Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
JP2003510108A (en) Biological joint structures
CA2435767A1 (en) Compositions and methods for the treatment and repair of defects or lesions in articular cartilage using synovial-derived tissue or cells
Clark et al. Porous implants as drug delivery vehicles to augment host tissue integration
Rotter et al. Behavior of tissue-engineered human cartilage after transplantation into nude mice
Vacanti et al. Structural tissue engineering
Petite et al. Marrow stromal stem cells for repairing the skeleton
Yannas Regenerative medicine II: clinical and preclinical applications
CA1341078C (en) Compositions and methods for repairs of cartilage and bone

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GE HU IL IS JP KG KP KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TR TT UA UZ VN YU AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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

Ref document number: 2247158

Country of ref document: CA

Ref country code: CA

Ref document number: 2247158

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1019980706539

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 331517

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1997907835

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997907835

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1019980706539

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1997907835

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1019980706539

Country of ref document: KR