US20050025838A1 - Conditioned compositions for tissue restoration - Google Patents

Conditioned compositions for tissue restoration Download PDF

Info

Publication number
US20050025838A1
US20050025838A1 US10/876,299 US87629904A US2005025838A1 US 20050025838 A1 US20050025838 A1 US 20050025838A1 US 87629904 A US87629904 A US 87629904A US 2005025838 A1 US2005025838 A1 US 2005025838A1
Authority
US
United States
Prior art keywords
matrix
composition
cells
tissue
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/876,299
Inventor
Stephen Badylak
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Acell Inc
Original Assignee
Badylak Stephen F.
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 Badylak Stephen F. filed Critical Badylak Stephen F.
Priority to US10/876,299 priority Critical patent/US20050025838A1/en
Publication of US20050025838A1 publication Critical patent/US20050025838A1/en
Assigned to ACELL, INC. reassignment ACELL, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BADYLAK, STEPHEN F.
Priority to US12/351,757 priority patent/US8409625B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/24Mucus; Mucous glands; Bursa; Synovial fluid; Arthral fluid; Excreta; Spinal fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/36Skin; Hair; Nails; Sebaceous glands; Cerumen; Epidermis; Epithelial cells; Keratinocytes; Langerhans cells; Ectodermal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/38Stomach; Intestine; Goblet cells; Oral mucosa; Saliva
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/39Pancreas; Islets of Langerhans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • A61L27/3633Extracellular matrix [ECM]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/20Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials
    • A61L2300/252Polypeptides, proteins, e.g. glycoproteins, lipoproteins, cytokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/412Tissue-regenerating or healing or proliferative agents
    • A61L2300/414Growth factors

Definitions

  • the present invention relates to compositions comprising matrices that are conditioned for remodeling, replacement, restoration or repair of tissue within a host, methods of making and methods of using the compositions.
  • Tissue graft constructs such as regenerative scaffolds, are commonly utilized in tissue engineering applications to repair, replace, restore and/or remodel damaged or diseased tissue.
  • tissue scaffolds often include matrices, for example, a naturally occurring extracellular matrix (ECM), to provide structure to the scaffold.
  • ECM extracellular matrix
  • Naturally occurring ECM is a mixture of structural and functional molecules arranged within a complex three-dimensional ultrastructure that surrounds and supports cells that are found within tissues and organs.
  • a matrix may be manufactured from structural or functional components of a naturally occurring ECM, such as collagen.
  • Naturally occurring ECMs are composed of both structural and functional biologically active molecules, including cytokines and growth factors, which play an important role in replication, differentiation, maturation and organization of cells in contact with the matrix.
  • the composition and structure of a naturally occurring ECM is a function of age of the host, location of the ECM within specific tissues and organs, and the demands placed upon the ECM as a result of environmental stressors.
  • naturally occurring musculotendinous ECM becomes stronger as a result of collagen deposition and collagen fiber reorganization in response to repeated uniaxial or multiaxial stress or compressive loading.
  • Naturally occurring hepatic ECM shows increased concentration of laminin, fibronectin and collagen IV within hours of the onset of hypoxia.
  • the naturally occurring ECM has been recognized as a critical component in the host response to tissue injury just as it is an essential element of normal tissue development.
  • ECM derived from the small intestinal submucosa
  • SIS small intestinal submucosa
  • compositions comprising matrices can be conditioned to augment the repair, replacement, remodeling, or restoration of a tissue in a patient.
  • the invention features a composition including a decellularized matrix.
  • the matrix is conditioned by cells cultured on the matrix in vitro whereby the decellularized conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in a patient in need of restoration, remodeling, replacement or repair.
  • the matrix comprises a naturally occurring extracellular matrix (ECM).
  • ECM comprises at least a portion of tunica submucosa, at least a portion of epithelial basement membrane, or at least a portion of tunica intestinal.
  • at least a portion of the ECM comprises dermis, subcutaneous tissue, pancreatic connective tissue, tissue harvested from the stomach, tissue harvested from the intestine, tissue harvested from the urinary bladder, tissue harvested from the skin or tissue harvested from a tissue comprising a mucous membrane.
  • the matrix comprises a structural or functional component of a naturally occurring ECM, or the matrix comprises a degradation product of a structural or functional component of a naturally occurring ECM.
  • the cells for conditioning the matrix can be selected from the group consisting of a primary, secondary, and immortalized cell population.
  • the matrix conditioning cells are selected from the group consisting of fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof; and hepatocyte and precursor cells thereof
  • the matrix conditioning cells express a biologically active molecule of interest.
  • the biologically active molecule of interest comprises a protein, and in further embodiments, the protein is selected from the group consisting of angiogenin, angiopoietin-1, Del-1, acidic-fibroblast growth factor (aFGF), basic-fibroblast growth factor (bFGF), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), Interleukin-8 (IL-8), leptin, placental growth factor (PlGF), platelet-derived endothelial cell growth factor (PD-ECGF), platelet-derived growth factor-BB (PDGF-BB), pleiotrophin (PTN), proliferin, transforming growth factor-alpha (TGF-alpha), transforming growth factor-beta (TGF-beta), tumor necrosis factor-alpha (TGF-alpha), tumor
  • the protein according to one embodiment is a growth factor, and in related embodiments, the growth factor is an angiogenic growth factor or an osteogenic growth factor.
  • the matrix conditioning cells are genetically-modified cells.
  • the genetically-modified cells are transfected cells comprising an exogenous nucleic acid and which express a protein of interest.
  • the genetically-modified cells are transfected with a nucleic acid that encodes a protein of interest.
  • the genetically-modified cells can express a biologically active molecule of interest.
  • the biologically active molecule of interest is a VEGF protein, a bFGF protein, a BMP protein, or a stromal derived growth factor-1 protein.
  • the biologically active molecule of interest is a recombinant protein.
  • the matrix comprises a naturally occurring ECM and the ECM is harvested from a tissue of a vertebrate.
  • the tissue in need of restoration, remodeling, replacement or repair comprises a tissue in a patient selected from the group consisting of bone, skin, heart, a tissue of the urogenital tract, a tissue of the gastrointestinal tract, nervous tissue, joint tissue and connective tissue.
  • the matrix conditioning cells are exposed to at least one stressor.
  • the at least one stressor can comprise hypoxia, hypercarbia, or an electrical current.
  • the matrix is exposed to at least one stressor, and in a related embodiment the stressor comprises mechanical loading.
  • a further aspect of the invention generally involves a composition
  • a composition comprising a matrix conditioned by cells cultured on the matrix in vitro and by exposure to at least one stressor.
  • the conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
  • the cells for conditioning the matrix are selected from the group consisting of fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof, and hepatocyte and precursor cells thereof.
  • the matrix conditioning cells are autologous cells.
  • the at least one stressor comprises hypoxia, hypercarbia, electrical current, or mechanical loading.
  • the matrix conditioning cells express a biologically active molecule of interest, and in a related embodiment, the biologically active molecule of interest comprises a protein.
  • the matrix conditioning cells are genetically-modified cells and the genetically-modified cells express a biologically active molecule of interest.
  • the biologically active molecule of interest can be a protein, and the protein can be a recombinant protein.
  • the invention features a method for inducing restoration, remodeling, replacement or repair of a tissue in a mammal.
  • the method comprises providing a decellularized matrix, conditioning the matrix by culturing cells on the matrix in vitro, and decellularizing the matrix wherein the decellularized conditioned matrix induces restoration, remodeling, replacement or repair of the tissue.
  • the method includes the step of exposing the cells for conditioning the matrix to at least one stressor. In another embodiment, the method includes the step of exposing the matrix to at least one stressor.
  • the tissue comprises a tissue selected from the group consisting of blood vessel tissue, heart tissue and bone tissue.
  • the cells for culturing the matrix express a biologically active molecule of interest.
  • the biologically active molecule of interest can comprise a growth factor.
  • the biologically active molecule of interest comprises a VEGF protein, a stromal derived growth factor-1 protein, or a bone morphogenic protein.
  • the matrix conditioning cells are genetically-modified cells, and in a related embodiment, the genetically-modified cells express a biologically active molecule of interest. In a further embodiment, the biologically active molecule of interest is a recombinant protein.
  • the matrix is harvested from a tissue of a vertebrate.
  • a further aspect of the invention generally involves a composition for tissue replacement, repair, restoration, or remodeling in a patient.
  • the composition comprises a decellularized matrix comprising a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof.
  • the matrix is conditioned by culturing cells on the matrix in vitro. In one embodiment, the cells for conditioning the matrix are exposed to at least one stressor, and in another embodiment, the matrix is exposed to at least one stressor.
  • a further aspect of the invention features a composition comprising a matrix.
  • the matrix is conditioned for tissue restoration, remodeling, replacement or repair by exposure to at least one stressor.
  • the stressor is an electric current.
  • biologically active molecule is meant a molecule capable of causing an effect on, interaction with, or response from living matter.
  • contacting includes both direct and indirect contact, including fluid communication.
  • genetically modified cell is meant a cell that includes an exogenous nucleic acid sequence.
  • exogenous nucleic acid sequence is meant a nucleic acid which is introduced into a cell by transduction.
  • the exogenous nucleic acid can be a gene which encodes a protein of interest or can be a nucleic acid sequence which alters the expression of a protein of interest, for example, upregulates the expression and production of the protein of interest.
  • An exogenous nucleic acid sequence includes a nucleic acid that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.
  • transfection is meant a process of introducing genes or gene fragments into cells.
  • transduced is meant a process of inserting exogenous nucleic acids into cells.
  • the insertion may, for example, be effected by transfection or transformation, viral infection, injection, transfection, gene bombardment, electroporation or any other means effective in introducing nucleic acids into cells.
  • the exogenous nucleic acid is either integrated wholly or in part, to the cell's genome (DNA), or remains external to the cell's genome, thereby providing stably transduced or transiently transduced cells.
  • primary cell includes cells present in a suspension of cells isolated from a tissue source (prior to their being plated, i.e., attached to a tissue culture substrate such as a dish or flask), cells present in an explant derived from tissue, both of the previous types of cells plated for the first time, and cell suspensions derived from these plated cells.
  • Secondary cells refers to cells at all subsequent steps in culturing. That is, the first time a plated primary cell is removed from the culture substrate and replated (passaged), it is referred to as a secondary cell, as are all cells in subsequent passages.
  • immortalized cell is meant a cell from an established cell line that is capable of cell division producing progeny that are capable of cell division in culture.
  • the present invention is directed to a composition that is conditioned for replacing, repairing, restoring or remodeling tissue when implanted in a host.
  • the composition includes a matrix derived from naturally occurring ECM or from structural or functional components of a naturally occurring ECM or their degradation products.
  • the composition can be conditioned in various ways, including culturing cells on the matrix, exposing the cells and/or the matrix to altered environmental and/or physiological conditions, or a combination thereof. Culturing cells on the matrix and/or exposing the cultured cells and matrix to various stressors provides a conditioned composition with enhanced biological properties compared to matrices that are merely combined with various cells and not decellularized or combined with bioactive factors by chemical or physical methods.
  • the conditioned matrices according to the invention retain the properties integrated into the matrix by the cultured cells, or by the application of stressors, and are able to deliver the resulting molecular properties of the conditioned matrix when implanted in a host in a manner that more closely simulates the natural state.
  • the conditioned composition according to the invention supports the replication, differentiation, maturation and spatial organization of cells, therefore augmenting regeneration, replacement, repair, restoration and/or remodeling of an organ or tissue in a patient.
  • the invention includes a composition comprising a decellularized matrix.
  • the matrix is conditioned by cells cultured on the matrix in vitro whereby the decellularized conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
  • the invention provides a composition comprising a matrix.
  • the matrix is conditioned by cells cultured on the matrix in vitro and by exposure to at least one stressor, whereby the conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
  • the invention further provides a method for inducing restoration, remodeling, replacement, or repair of a tissue in a mammal.
  • the method comprises the steps of providing a decellularized matrix, then conditioning the matrix by culturing cells on the matrix, and lastly, decellularizing the matrix wherein the decellularized, conditioned matrix induces restoration, remodeling, replacement, or repair of the tissue.
  • the invention includes a composition for tissue replacement, repair, restoration, or remodeling, comprising a decellularized matrix including a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof.
  • the matrix is conditioned by culturing cells on the matrix in vitro.
  • the invention features a composition comprising a matrix and the matrix is conditioned for tissue restoration, remodeling, replacement or repair by exposure to at least one stressor.
  • the matrix for example, comprises a naturally occurring ECM isolated from the tissues of vertebrates, including, for example, alimentary, respiratory, intestinal, urinary or genital tracts of mammals.
  • the naturally occurring ECM can comprise at least a portion of tunica submucosa, epithelial basement membrane, tunica muscular, or dermis.
  • the naturally occurring ECM comprises, for example, at least a portion of subcutaneous tissue, respiratory tissue, pancreatic connective tissue, hepatic connective tissue, tissue harvested from the stomach, tissue harvested from the intestine, tissue harvested from the urinary bladder, tissue harvested from the urinary tract, tissue harvested from the liver, tissue harvested from the genital tract, tissue harvested from the skin, or tissue harvested from a tissue comprising a mucous membrane.
  • the naturally occurring ECM comprises a combination of the above, for example, the ECM may comprise tissue harvested from the urinary bladder including a portion of the epithelial basement membrane and a portion of the tunica basement membrane.
  • the matrix comprises structural or functional components of naturally occurring ECMs, degradation products of structural or functional components of naturally occurring ECMs, or a combination thereof.
  • the structural or functional components comprise, for example, collagen (i.e., collagen types I-XIX), hyaluronic acid, laminin, fibronectin, elastin, purified components of naturally occurring ECMs, or a combination thereof.
  • Matrixes can include highly conserved collagens, glycoproteins, proteoglycans and glycosaminoglycans in their natural configuration and natural concentration.
  • cells are cultured on and/or within the matrix, and the cells comprise a primary, secondary and/or immortalized cell population.
  • the cells are, for example, fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof, and hepatocyte and precursor cells thereof.
  • the cells cultured on and/or within the matrix remain on and/or within the matrix when the matrix is implanted into a host.
  • primary, secondary or immortalized cells including autologous cells, are added to a conditioned matrix. These cells can be grown on or within the conditioned matrix and implanted with the conditioned composition at an anatomical site in a patient.
  • the cells express a biologically active molecule of interest.
  • the biologically active molecule of interest is produced by the cells and is integrated within the matrix while the cells are cultured on or within the matrix.
  • the biologically active molecule of interest is, for example, a protein, including, for example, a growth factor.
  • the growth factor is, for example, an angiogenic or osteogenic growth factor.
  • the protein includes angiogenin, angiopoietin-1, Del-1, acidic-fibroblast growth factor (aFGF), basic-fibroblast growth factor (bFGF), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), Interleukin-8 (IL-8), leptin, placental growth factor (PlGF), platelet-derived endothelial cell growth factor (PD-ECGF), platelet-derived growth factor-BB (PDGF-BB), pleiotrophin (PTN), proliferin, transforming growth factor-alpha (TGF-alpha), transforming growth factor-beta (TGF-beta), tumor necrosis factor-alpha (TNF-alpha), vascular endothelial growth factor (VEGF), bone morphogenetic protein-2 (BMP-2), and/or stromal derived growth factor-1.
  • aFGF acid
  • the cells are genetically modified.
  • the genetically modified cells are transfected cells that comprise an exogenous nucleic acid and that express a protein of interest, or the genetically modified cells are cells transfected with a nucleic acid that encodes a protein of interest.
  • the genetically modified cells express one or more biologically active molecules of interest, such as, for example, a VEGF protein, a bFGF protein, a BMP protein, a stromal derived growth factor-1 protein, or a recombinant protein.
  • Cells cultured on the matrix can also be exposed to at least one stressor or a combination of stressors to enhance the cells' ability to condition the matrix. Exposure to certain stressors can stimulate the release of biologically active molecules from the cells and contribute to the conditioning of the composition for various purposes. Exposure to certain stressors can also prevent the release of undesired biologically active molecules from the cells and contribute to the conditioning of the composition to avoid undesired effects when implanted in a host. According to one embodiment, the stressor is hypoxia, and in another embodiment, the stressor is hypercarbia.
  • Exposure to hypoxia and/or hypercarbia can assist the cells in producing biologically active molecules that are incorporated in the matrix on which the cells are cultured and support, for example, the growth of new blood vessels and assist in the restoration of blood flow to tissue in and surrounding the implantation site.
  • Exposure time to each stressor can vary, for example, from 10 minutes to 1 month, depending on the intended use of the conditioned composition, the type of tissue involved and/or the type of matrix being conditioned.
  • the cells are exposed to stressors such as mechanical loading.
  • Exposure to mechanical loading for example, enhances cellular production of structural proteins and other biologically active molecules, integrating them into the matrix to improve its tensile strength.
  • exposure to mechanical loading is achieved by consistently applying strain (i.e., applying a force that causes a change in length) or by applying stress (i.e., applying a mechanical load) to the matrix over a predetermined period of time, for example, for a total time period of 168 hours.
  • mechanical conditioning can be achieved by applying stress or strain to the matrix in a cyclical pattern, for example, applying stress or strain for 1 minute every 5 minutes over a total time of 48 hours.
  • the stress or strain on the matrix is constant throughout the time period, or alternatively, the stress or strain can vary.
  • the amount of stress or strain can vary, for example, from 5% to 15% using an appropriate device, such as a load cell.
  • the stress or strain can be applied in a uniaxial or multiaxial direction.
  • the amount of strain applied can be measured in the cell culture systems by any means known in the art, for example, by laser measurements using laser beams and deflection times.
  • the cells cultured on the matrix are exposed to an electric current.
  • Exposure to electrical current can, for example, stimulate the release of growth factors and other biologically active molecules from the cell into the matrix, resulting in the matrix exhibiting accelerated tissue regenerative and wound healing properties.
  • the cells are exposed to an electric current for a consistent period of time, or the cultured cells are exposed to an electric current in a cyclical pattern, for example, applying the electric current or electrical potential for 10 seconds every 1 minute over a total time of 1 hour.
  • the electric current potential remains constant over the entire exposure time period, or, alternatively, the magnitude of the electric current potential can alternate between, for example, a positive and negative potential within, for example, a 100 mV-300 mV range.
  • cells are not cultured on the matrix, and the matrix is exposed to at least one stressor to condition the matrix for its intended use.
  • the stressor can stimulate the release of biologically active molecules or, alternatively, the stressor can be applied to decrease or prevent the release of undesired biologically active molecules into the matrix.
  • the matrix can be exposed, for example, to an electric current or electrical potential. Exposure to electric current or electrical potential can decrease or prevent the release of certain growth factors, such as TGF-beta, from cells existing on the matrix, resulting in a conditioned matrix that decreases the risk of adverse events, such as, for example, the formation of scar tissue, when implanted in a host.
  • the conditioned composition is useful in replacing, repairing, restoring or remodeling various types of tissue, for example, bone, skin, heart, tissue of the urogenital tract, tissue of the gastrointestinal tract, nervous tissue, joint tissue and connective tissue.
  • tissue for example, bone, skin, heart, tissue of the urogenital tract, tissue of the gastrointestinal tract, nervous tissue, joint tissue and connective tissue.
  • the decellularized, conditioned composition can be used to promote vascularization by providing a decellularized scaffold including a matrix conditioned by cells cultured on the matrix that express an angiogenic or vasculogenic protein of interest to the anatomical site in the host requiring vascularization.
  • the decellularized, conditioned composition can also be used, for example, for musculotendinous tissue reconstruction by providing a decellularized composition conditioned with cultured cells that express a structural protein of interest to the anatomical site in the host requiring musculotendinous reconstruction.
  • the decellularized, conditioned composition can be conditioned with cells to express a growth factor of interest to stimulate growth of the host's tissues surrounding the composition when that composition is introduced to the anatomical site in the host that is in need of treatment.
  • the following provides exemplary methods for preparing a matrix for use according to the invention.
  • a particular example illustrates preparation of a matrix comprising a naturally occurring ECM.
  • Preparation of the naturally occurring ECM for use according to the invention is also described in U.S. Pat. Nos. 4,902,508, 4,956,178, 5,554,389, 6,576,265 and 6,579,538, the entirety of each patent is incorporated by reference herein.
  • intestinal submucosal tissue can be prepared by harvesting tissue from a vertebrate such as porcine, ovine or bovine species.
  • the tissue is subjected to abrasion using a longitudinal wiping motion to remove the outer layers, smooth muscle tissues, and the innermost layer, i.e., the luminal portion of the tunica mucosa.
  • the remaining submucosal tissue or any portion of it is then rinsed with saline.
  • a naturally occurring ECM including epithelial basement membrane derived from urinary bladder can be prepared by removing the urinary bladder tissue from a vertebrate, for example, a pig, and delaminating the tissue by first soaking the tissue in a deepithelializing solution, for example, hypertonic saline, most preferably 1.0 N saline, for periods of time ranging from 10 minutes to 4 hours. Exposure to a hypertonic saline solution effectively removes the epithelial cells from the underlying basement membrane. The tissue remaining after the initial delamination procedure includes epithelial basement membrane and the tissue layers abluminal to the epithelial basement membrane.
  • a deepithelializing solution for example, hypertonic saline, most preferably 1.0 N saline
  • This tissue is next subjected to further treatment to remove the majority of abluminal tissues but not the epithelial basement membrane.
  • the outer serosal, adventitial, smooth muscle tissues, submucosa and abluminal portion of the tunica intestinal are removed from the remaining deepithelialized tissue by mechanical abrasion or by a combination of enzymatic treatment, hydration, and abrasion.
  • Mechanical removal of these tissues is accomplished by removal of mesenteric tissues with, for example, Adson-Brown forceps and Metzenbaum scissors and wiping away the tunica muscularis and abluminal tunica intestinal using a longitudinal wiping motion with a scalpel handle or other rigid object wrapped in moistened gauze.
  • the resulting ECM consists of epithelial basement membrane and subjacent tunica intestinal.
  • the tissues may be further processed by rinsing in hypertonic saline, peracetic acid or sterile water. Other methods for removing tissue layers, a microtome, for example, may also be used to obtain the tissue composition of the invention.
  • the matrix according to the invention comprises a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof.
  • the structural and functional components can include, for example, collagen (i.e., any of collagen types I-XIX), hyaluronic acid, laminin, fibronectin, elastin, purified components of the naturally occurring ECM, or a combination thereof.
  • Matrixes can include, for example, highly conserved collagens, glycoproteins, proteoglycans and glycosaminoglycans in their natural configuration and natural concentration.
  • the matrix can be stored in a hydrated or dehydrated state. Lyophilized or air dried matrix can be rehydrated and used in accordance with this invention.
  • the matrix can be sterilized prior to the addition of cells using conventional sterilization techniques including glutaraldehyde tanning, formaldehyde tanning at acidic pH, propylene oxide treatment, ethylene oxide, gas plasma sterilization, gamma radiation, electron beam, peracetic acid sterilization.
  • Sterilization techniques which do not adversely affect the mechanical strength, structure, and biotropic properties of the tissue are preferred. For instance, strong gamma radiation may cause loss of strength of the sheets of submucosal tissue.
  • Preferred sterilization techniques include exposing the graft to peracetic acid, 1-4 Mrads gamma irradiation (more preferably 1-2.5 Mrads of gamma irradiation) or gas plasma sterilization; peracetic acid sterilization is the most preferred sterilization method.
  • the tissue is subjected to two or more sterilization processes. After the tissue is sterilized, for example by chemical treatment, the tissue may be wrapped in a plastic or foil wrap and sterilized again using electron beam or gamma irradiation sterilization techniques.
  • compositions comprising submucosal tissue can be used for supporting growth or proliferation of eukaryotic cells in vitro.
  • Submucosal tissue can be used in accordance with this invention as a cell growth substrate in a variety of forms, including its naturally occurring sheet-like configuration, as a gel matrix, as an addition for art-recognized cell/tissue culture media, or as coating for culture-ware to provide a more physiologically relevant substrate that supports and enhances the proliferation of cells in contact with the matrix.
  • the submucosal tissue provides surfaces for cell adhesion and also helps induce cell differentiation.
  • nonsterile matrix can be used if antibiotics are included in the cell culture system.
  • the matrix can be decellularized after cells have been cultured on the matrix using any technique known in the art.
  • the matrix can be decellularized by placing the matrix in a decellularizing solution such as in hypertonic saline, for example, 1.0N saline or in Hanks buffered saline solution (HBSS).
  • a decellularizing solution such as in hypertonic saline, for example, 1.0N saline or in Hanks buffered saline solution (HBSS).
  • primary, secondary or immortalized cells including autologous cells
  • the type of primary, secondary, and/or immortalized cell is chosen based upon the intended use of the conditioned composition, the anatomical site of implantation of the conditioned composition or the desired physiological properties of the conditioned composition.
  • the following provides exemplary sources and methods for the preparation of matrix conditioning cells that may be cultured on the matrix to make the conditioned matrix according to the invention.
  • the composition and configuration of the matrix provides a unique cell growth substrate that promotes the attachment and proliferation of various types of cells.
  • methods involve contacting the cells with the matrix under conditions conducive to cell growth and which are well known in the art.
  • the matrix conditioning cells can be prokaryotic or eukaryotic cells such as vertebrate cells (particularly mammalian cells, such as cells derived from a human, chimpanzee, mouse, rat, hamster, guinea pig, rabbit, cow, horse, pig, goat, sheep, dog, or cat).
  • the matrix conditioning cells can be primary cells, secondary cells, and/or immortalized cells, and the matrix conditioning cells can be autologous cells.
  • the types of matrix conditioning cells include fibroblasts, keratinocytes, epithelial cells (e.g., mammary epithelial cells, intestinal epithelial cells), endothelial cells, adipocytes, astrocytes, cardiac muscle cells, chondrocytes, glial cells, neural cells formed elements of the blood (e.g., lymphocytes, bone marrow cells), muscle cells, gangliocytes, glandular cells, glial cells, hematopoietic cells, mesenchymal cells, hepatocytes and precursors of these somatic cell types.
  • epithelial cells e.g., mammary epithelial cells, intestinal epithelial cells
  • endothelial cells e.g., endothelial cells
  • adipocytes e.g., astrocytes, cardiac muscle cells
  • chondrocytes e.g., glial cells
  • neural cells formed elements of the blood e.g.,
  • the matrix conditioning cells can be genetically modified cells.
  • Various genetically modified mammalian host cell lines can be used, for example, L cells, C127, 3T3 fibroblasts, T-84 cells, Chinese hamster ovary (CHO), HeLa, BHK cell lines, CV-1 cells (ATCC CCL70), COS-7 cells and CV-1/EBNA.
  • a matrix conditioning cell of choice can be genetically modified by transducing into the cell exogenous nucleic acid sequences.
  • the exogenous nucleic acid can encode a biologically active molecule of interest, such as a protein, or alternatively, the exogenous DNA can be a regulatory sequence that will activate expression of an endogenous gene (for example, using homologous recombination) to produce a biologically active molecule of interest.
  • a biologically active molecule of interest such as a protein
  • the exogenous DNA can be a regulatory sequence that will activate expression of an endogenous gene (for example, using homologous recombination) to produce a biologically active molecule of interest.
  • any desired biologically active molecule can be selected for secretion onto the matrix.
  • the biologically active molecule of interest is chosen based on where the conditioned composition is to be located in the body and/or the physiological requirements of the recipient. For example, if the decellularized, conditioned composition is used to repair bone, the biologically active molecule of interest which is produced on the matrix can be a bone growth factor, such as a bone morphogenic protein, e.g., BMP-2.
  • the biologically active molecule of interest can include enzymes, hormones, cytokines, colony stimulating factors, vaccine antigens, antibodies, clotting factors, angiogenesis factors, regulatory proteins, transcription factors, receptors, and structural proteins, for example, epithelial basement membrane proteins such as type IV collagen, laminin, and fibronectin.
  • the biologically active molecule of interest can also include human growth hormone, Factor VIII, Factor IX, erythropoietin, and insulin.
  • nucleic acid sequences and amino acid sequences for biologically active molecules of interest can be readily obtained from National Center for Biotechnology Information. (http://www.ncbi.nlm.nih.gov/).
  • a biologically active molecule of interest is expressed in the genetically modified matrix conditioning cell using an expression vector such as a mammalian expression vector.
  • Mammalian expression vectors typically contain non-transcribed elements such as an origin of replication, a suitable promoter, such as, for example, the ROSA promoter, and an enhancer linked to the gene to be expressed, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences, such as necessary ribosome binding sites, a poly-adenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • the transcriptional and translational control sequences in expression vectors to be used in transforming vertebrate cells may be provided by viral sources.
  • promoters and enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus.
  • DNA sequences derived from the SV40 viral genome for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of an exogenous DNA sequence.
  • the early and late promoters are particularly useful because both are obtained easily from the virus as a fragment that also contains the SV40 viral origin or replication.
  • the expression vector may comprise DNA encoding a signal or leader peptide such as the native signal sequence of IL-7 or interleukin-4.
  • the biologically active molecule of interest can include a recombinant protein.
  • High-yield production of recombinant proteins can be produced by cells that stably express a protein of interest.
  • Cell lines which stably express the protein of interest may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • Vector DNA can be introduced into eukaryotic matrix conditioning cells via conventional transduction techniques such as using calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation.
  • engineered matrix conditioning cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • Preferred selectable markers include G418, hygromycin and methotrexate.
  • Such engineered cell lines can then be added to the matrix and cultured, and a biologically active molecule of interest can be secreted onto and integrate into the matrix.
  • Matrix conditioning cells can be cultured, in vitro, on and/or within the matrix, using a matrix prepared by any of the methods described above.
  • the cells are cultured under conditions conducive to cell growth and if desired, under conditions conducive to expression of a biologically active molecule of interest.
  • Conditions for cell culturing are well known in the art and are dependent on the particular cell type. While not wishing to be bound by theory, it is believed that a biologically active molecule that is expressed by matrix conditioning cells on the matrix, in vitro, is incorporated into the matrix similar to the manner by which molecules, such as proteins, are integrated into a naturally occurring extracellular matrix in vivo.
  • the incorporation of the molecule produced by the cultured cells into the matrix imparts superior restorative properties to this conditioned composition compared to the mere addition of, for example, a protein to a decellularized matrix. It is believed that the incorporated biologically active molecule is released in the recipient of the conditioned composition as a function of composition degradation. Additionally, the conditioned composition can be substantially devoid of cellular elements that may lead to rejection of the tissue implant by the recipient.
  • composition of the invention can be implanted into any anatomical location in the body of the host where there exists a tissue in need of regeneration, repair, replacement, restoration or remodeling.
  • Angiogenesis and vasculogenesis are an integral part of the restoration of blood flow to transplanted tissue.
  • the process of angiogenesis involves the migration and proliferation of capillary endothelial cells in the early post-injury phase, and the differentiation of the capillary endothelial cells in the later post-injury phase to organize endothelial cells into a microvascular network.
  • Angiogenesis requires dynamic interactions between cells such as fibroblasts, epithelial and endothelial cells, and a wide array of angiogenic and vasculogenic factors and cytokines that exist within the matrix, such as FGF, VEGF, TGF ⁇ , and others. These cells and matrix composites orchestrate the proliferative, inhibitory, and differentiation phases of cells to re-populate, repair and restore damaged tissues and its vascular supply.
  • the following experiment relates to the use of the conditioned composition of the invention in relation to vasculogenesis and myelogenesis.
  • a naturally occurring ECM derived from urinary bladder was cultured with matrix conditioning transfected cardiac myofibroblasts that constitutively express SDF-1.
  • SDF-1 concentration in the media and in the matrix was measured over a six day time period. Results showed that the concentration of SDF-1 in the scaffold increased more than 100 fold in just six days (from less than 5 pg/ml to 625 pg/ml).
  • mice The SDF-1 conditioned matrix was decellularized as described above and implanted subcutaneously into mice that have been genetically altered to express green fluorescent protein in all bone marrow-derived cells.
  • Control mice were similarly treated using non-conditioned naturally occurring ECM or simply a sham operation. Blood vessel formation in the mice was measured and quantified by performing morphometric assays on cross sectional samples taken from predetermined sites in low magnification fields. Following 7 and 14 days, mice treated with the decellularized, conditioned scaffold of the invention had increased blood vessel formation and increased numbers of bone marrow-derived cells compared with control mice. Results demonstrated a percentage increase in blood vessel cross sectional area of 48%+/ ⁇ 13%, p ⁇ 0.05 compared to a control (non-conditioned) matrix.
  • the conditioned compositions of the invention are used to improve angiogenesis in the composition after implantation.
  • Exposure of the matrix conditioning endothelial cells to a hypoxic environment assists the matrix conditioning cells in releasing a natural complement of angiogenic factors into the matrix that will enhance the matrix's specificity.
  • the period of exposure time can range from one minute to thirty minutes.
  • Such angiogenesis and vasculogenesis factors can include one or more of the following proteins: FGF, VEGF, TGF ⁇ , and Angiopoietin.
  • the matrix is decellularized but retains the angiogenic factors that have become integrated in the matrix.
  • the conditioned matrix is then implanted within a recipient in need of vascular assistance and results in enhanced vascularization properties in vivo.
  • ECM a naturally occurring ECM was prepared as explained above.
  • Matrix conditioning endothelial cells EC were delivered to the matrix and cultured on the matrix under normoxic conditions for a period of 72 hours, with the last 24 hours in serum-free media. The cultured cells were then exposed to a hypoxic environment having 2% oxygen for a period of 48 hours. The concentration of the 165 and 189 isoforms of VEGF protein was measured in both the matrix and the supernatant. Results showed a 5-fold increase of VEGF concentration in the matrix and a 10-fold increase of VEGF in the supernatant as compared to a control in which EC cells were cultured under the same conditions but not exposed to a hypoxic environment.
  • One illustrative experiment pertains to use of the invention in regenerating heart tissue.
  • a naturally occurring ECM derived from the small intestine measuring approximately 1 cm ⁇ 2 cm is prepared as described above and is seeded with matrix conditioning fibroblasts at 0.5 ⁇ 106 cells/cm 2 .
  • the matrix conditioning fibroblasts are genetically engineered to express stromal derived growth factor-1 (SDF-1).
  • SDF-1 stromal derived growth factor-1
  • the matrix conditioning genetically modified cells are cultured with the matrix during a time period ranging from 24 to 168 hours.
  • the presence of SDF-1 in the conditioned matrix is determined by ELISA.
  • the matrix is then decellularized by placing the matrix in HBSS. Following decellularization, the conditioned matrix is implanted by a surgical procedure to a damaged area of the myocardium in the recipient.
  • the protein of interest for example, SDF-1
  • the conditioned composition attracts circulating multi-potential cells that are influenced, for example, to differentiate towards cardiomyocytes.
  • the period of exposure time can range from one minute to thirty minutes.
  • a naturally occurring ECM is prepared as described above and is seeded with matrix conditioning cells that are genetically modified to produce factors which improve angiogenesis and/or vasculogenesis.
  • matrix conditioning cells that are genetically modified to produce factors which improve angiogenesis and/or vasculogenesis.
  • angiogenesis and vasculogenesis factors can include one or more of the following proteins: FGF, VEGF, TGF ⁇ , and Angiopoietin.
  • the experiments can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • the matrix is prepared as explained above, and matrix conditioning fibroblast cells are introduced to the matrix and exposed to mechanical loading.
  • a cyclic loading system that allows for independent control of stress and strain is applied to the matrix in a bioreactor environment and a cell culture system provides for sterile conditions while simultaneously monitoring the stress and strain load in the matrix.
  • the cell culture system includes, for example, a load cell with 25 N capacity and a linear actuator with a 30 mm travel capacity.
  • the system allows the user to apply a load to each matrix and define the displacement profile by specifying the amplitude and frequency of oscillation or by defining a function to prescribe the displacement.
  • Exposure of the matrix and/or matrix conditioning fibroblast cells to mechanical loading assists in the release of structural proteins and other biologically active molecules from the cells and into the matrix.
  • the conditioned composition including the matrix is decellularized prior to implantation in a host.
  • a matrix comprising a naturally occurring ECM is prepared as described above and genetically modified cells that secrete structural proteins, such as collagen, after a suitable incubation period are cultured on the matrix.
  • the matrix is decellularized prior to implantation into a host to form the conditioned composition according to the invention.
  • fibroblasts were introduced to a naturally occurring ECM derived from the small intestine measuring approximately 1 cm ⁇ 2 cm.
  • the cells were cultured on the matrix for approximately 12 hours, during which time a preload amount of 0.25 N was applied. A strain of 10% was then applied to the matrix at 1 Hz for 24 hours.
  • the load placed upon the matrix was monitored continuously during the experiment, and the cell number and pH of the media were determined at the conclusion of the experiment.
  • Cell viability was determined using the MTT assay to stain metabolically active cells.
  • the presence of procollagen Type I was determined with the use of an ELISA kit.
  • Results showed that the fibroblasts rapidly aligned along the lines of applied stress and assumed an elongated, spindloid morphology. Such results indicate that the strength of the conditioned composition will be enhanced compared to compositions not similarly conditioned when implanted in a host.
  • the experiments described above can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • the conditioned composition of the invention is used to promote bone formation.
  • a matrix comprising a naturally occurring ECM is prepared as described above.
  • Matrix conditioning genetically modified cells that secrete the bone growth factor, bone morphogenetic protein-2 (BMP-2), are cultured on the matrix for a suitable incubation period.
  • the matrix is then decellularized prior to implantation into a host.
  • the same experiment can be performed using a matrix comprising structural or functional components of a naturally occurring ECM.
  • a matrix comprising a naturally occurring ECM is prepared as described above and matrix conditioning cells that produce growth factors and other biologically active molecules after a suitable incubation period are cultured on the matrix.
  • the cultured cells are exposed to either constant or alternating electrical potential for the purpose of stimulating the release of growth factors and other biologically active molecules from the cells into the matrix prior to implantation.
  • the matrix is seeded with a population of matrix conditioning endothelial cells prior to its exposure to electrical potential.
  • An electric potential in the range of 25 mV to about 500 mV, preferably 300 mV, is applied across the matrix for 24 hours to 72 hours, preferably for 48 hours.
  • growth factors and other biologically active molecules such as, for example, proteins, enzymes, hormones, and cytokines, are released from the cells and are incorporated into the matrix.
  • the matrix is then decellularized prior to its implantation into the host.
  • the method can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • a matrix comprising a naturally occurring ECM is prepared as described above.
  • the matrix is exposed to an electric potential that depletes the matrix of certain biologically active molecules that can result in undesired properties when the matrix is implanted in a host.
  • the matrix is exposed for a period of 72 hours to a constant electric current potential known to reduce or prevent the release of TGF-beta growth factors.
  • the conditioned matrix is then implanted in a host and the risk of undesired scar tissue formation at the site of implantation is decreased.
  • a matrix comprising a naturally occurring ECM is prepared as described above and conditioned in vitro by the addition of a specific population of matrix conditioning cells, such as astroglial cells, chosen to bestow upon the matrix characteristics known to be unique to the naturally occurring ECM of a particular tissue type, such as tissues of the central nervous system (CNS).
  • matrix conditioning hepatocytes are introduced and cultured on the matrix to confer upon it the properties inherent to the naturally occurring ECM that surrounds liver tissue. The cultured cells are removed before implantation into a recipient.
  • the decellularized conditioned matrix can direct endothelial cell differentiation, for example, hepatic sinusoidal endothelial cell (HSEC) differentiation and brain endothelial cell (BEC) differentiation, upon transplant of the conditioned matrix into the target tissue of the host.
  • HSEC hepatic sinusoidal endothelial cell
  • BEC brain endothelial cell
  • the method can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • a naturally occurring ECM is a dynamic structure that reflects the products of the resident cells and is tissue and organ specific.
  • the composition conditioned according to the exemplary methods described above comprises a specialized matrix, in that, once decellularized, the matrix maintains the properties associated with the cellular subtypes present during the conditioning and/or culturing phase.
  • the conditioned composition can be primed for implantation into a target region and will demonstrate improved restorative capabilities as compared with an unconditioned matrix.
  • Endothelial cells for example, show diverse phenotypic potential. EC phenotype is related to EC function, and function is site dependent. For example, renal glomerular endothelial cells are well-suited for a glomerular filtration function.
  • Hepatic sinusoidal endothelial cells have the unique phenotypic characteristic of fenestrations or “sieve plates” that are essential for normal hepatic blood filtration functions.
  • endothelial cells in the brain are a critical component of the blood-brain barrier that is virtually impermeable to most molecules.
  • Vascular networks within the CNS have no fenestrations (i.e., blood-brain barrier) and express zona occludens 1, 2 and 3 proteins characteristic of tight junctions.

Abstract

The invention provides a composition conditioned for the remodeling, restoration, repair, or replacement of tissue within a host. The composition is conditioned by culturing cells on the matrix and/or by exposing the cultured cells or matrix to one or more stressors.

Description

    RELATED APPLICATIONS
  • This application claims priority to and the benefit of U.S. Ser. No. 60/482,480, filed Jun. 25, 2003, and U.S. Ser. No. 60/538,385, filed Jan. 21, 2004, the contents of which are incorporated by reference herein.
  • FIELD OF THE INVENTION
  • The present invention relates to compositions comprising matrices that are conditioned for remodeling, replacement, restoration or repair of tissue within a host, methods of making and methods of using the compositions.
  • BACKGROUND OF THE INVENTION
  • Tissue graft constructs, such as regenerative scaffolds, are commonly utilized in tissue engineering applications to repair, replace, restore and/or remodel damaged or diseased tissue. These tissue scaffolds often include matrices, for example, a naturally occurring extracellular matrix (ECM), to provide structure to the scaffold. Naturally occurring ECM is a mixture of structural and functional molecules arranged within a complex three-dimensional ultrastructure that surrounds and supports cells that are found within tissues and organs. Alternatively, a matrix may be manufactured from structural or functional components of a naturally occurring ECM, such as collagen. Naturally occurring ECMs are composed of both structural and functional biologically active molecules, including cytokines and growth factors, which play an important role in replication, differentiation, maturation and organization of cells in contact with the matrix.
  • The composition and structure of a naturally occurring ECM is a function of age of the host, location of the ECM within specific tissues and organs, and the demands placed upon the ECM as a result of environmental stressors. For example, naturally occurring musculotendinous ECM becomes stronger as a result of collagen deposition and collagen fiber reorganization in response to repeated uniaxial or multiaxial stress or compressive loading. Naturally occurring hepatic ECM shows increased concentration of laminin, fibronectin and collagen IV within hours of the onset of hypoxia. The naturally occurring ECM has been recognized as a critical component in the host response to tissue injury just as it is an essential element of normal tissue development.
  • Selected forms of the naturally occurring ECM, for example, ECM derived from the small intestinal submucosa (SIS), have been successfully used as a scaffold for tissue engineering applications in both pre-clinical animal studies and in human clinical applications. Experience with patients has suggested that the degree of success and the morphologic changes that occur within the scaffold over time are a direct result of local environmental stimuli, such as mechanical loading (rehabilitation), the viability of surrounding tissue, and the surrounding tissue pH and ionic concentrations. Although these scaffold compositions have provided a starting point for tissue engineering applications, there exists a need in the art for compositions that have improved ability to support the replication, differentiation, maturation and spatial organization of numerous cell types.
  • SUMMARY OF THE INVENTION
  • The present invention is based on the finding that compositions comprising matrices can be conditioned to augment the repair, replacement, remodeling, or restoration of a tissue in a patient. According to one aspect, the invention features a composition including a decellularized matrix. The matrix is conditioned by cells cultured on the matrix in vitro whereby the decellularized conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in a patient in need of restoration, remodeling, replacement or repair.
  • In one embodiment according to this aspect of the invention, the matrix comprises a naturally occurring extracellular matrix (ECM). In related embodiments, the ECM comprises at least a portion of tunica submucosa, at least a portion of epithelial basement membrane, or at least a portion of tunica propria. In other related embodiments according to this aspect of the invention, at least a portion of the ECM comprises dermis, subcutaneous tissue, pancreatic connective tissue, tissue harvested from the stomach, tissue harvested from the intestine, tissue harvested from the urinary bladder, tissue harvested from the skin or tissue harvested from a tissue comprising a mucous membrane.
  • In other embodiments of the invention, the matrix comprises a structural or functional component of a naturally occurring ECM, or the matrix comprises a degradation product of a structural or functional component of a naturally occurring ECM.
  • The cells for conditioning the matrix can be selected from the group consisting of a primary, secondary, and immortalized cell population. In one embodiment, the matrix conditioning cells are selected from the group consisting of fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof; and hepatocyte and precursor cells thereof
  • In a further embodiment, the matrix conditioning cells express a biologically active molecule of interest. The biologically active molecule of interest according to one embodiment comprises a protein, and in further embodiments, the protein is selected from the group consisting of angiogenin, angiopoietin-1, Del-1, acidic-fibroblast growth factor (aFGF), basic-fibroblast growth factor (bFGF), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), Interleukin-8 (IL-8), leptin, placental growth factor (PlGF), platelet-derived endothelial cell growth factor (PD-ECGF), platelet-derived growth factor-BB (PDGF-BB), pleiotrophin (PTN), proliferin, transforming growth factor-alpha (TGF-alpha), transforming growth factor-beta (TGF-beta), tumor necrosis factor-alpha (TNF-alpha), vascular endothelial growth factor (VEGF), bone morphogenetic protein-2 (BMP-2) and stromal derived growth factor-1.
  • The protein according to one embodiment is a growth factor, and in related embodiments, the growth factor is an angiogenic growth factor or an osteogenic growth factor.
  • In another embodiment of the invention, the matrix conditioning cells are genetically-modified cells. In one embodiment, the genetically-modified cells are transfected cells comprising an exogenous nucleic acid and which express a protein of interest. In another embodiment, the genetically-modified cells are transfected with a nucleic acid that encodes a protein of interest. The genetically-modified cells can express a biologically active molecule of interest. In a related embodiment, the biologically active molecule of interest is a VEGF protein, a bFGF protein, a BMP protein, or a stromal derived growth factor-1 protein. In another related embodiment, the biologically active molecule of interest is a recombinant protein.
  • In a further embodiment, the matrix comprises a naturally occurring ECM and the ECM is harvested from a tissue of a vertebrate.
  • The tissue in need of restoration, remodeling, replacement or repair according to an embodiment of the invention comprises a tissue in a patient selected from the group consisting of bone, skin, heart, a tissue of the urogenital tract, a tissue of the gastrointestinal tract, nervous tissue, joint tissue and connective tissue.
  • In another embodiment, the matrix conditioning cells are exposed to at least one stressor. The at least one stressor can comprise hypoxia, hypercarbia, or an electrical current. In a further embodiment, the matrix is exposed to at least one stressor, and in a related embodiment the stressor comprises mechanical loading.
  • A further aspect of the invention generally involves a composition comprising a matrix conditioned by cells cultured on the matrix in vitro and by exposure to at least one stressor. The conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
  • In one embodiment according to this aspect of the invention, the cells for conditioning the matrix are selected from the group consisting of fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof, and hepatocyte and precursor cells thereof. In another embodiment, the matrix conditioning cells are autologous cells.
  • According to further embodiments of the invention, the at least one stressor comprises hypoxia, hypercarbia, electrical current, or mechanical loading. In another embodiment, the matrix conditioning cells express a biologically active molecule of interest, and in a related embodiment, the biologically active molecule of interest comprises a protein. In other embodiments, the matrix conditioning cells are genetically-modified cells and the genetically-modified cells express a biologically active molecule of interest. The biologically active molecule of interest can be a protein, and the protein can be a recombinant protein.
  • In another aspect, the invention features a method for inducing restoration, remodeling, replacement or repair of a tissue in a mammal. The method comprises providing a decellularized matrix, conditioning the matrix by culturing cells on the matrix in vitro, and decellularizing the matrix wherein the decellularized conditioned matrix induces restoration, remodeling, replacement or repair of the tissue.
  • In one embodiment of this aspect of the invention, the method includes the step of exposing the cells for conditioning the matrix to at least one stressor. In another embodiment, the method includes the step of exposing the matrix to at least one stressor. In a further embodiment, the tissue comprises a tissue selected from the group consisting of blood vessel tissue, heart tissue and bone tissue.
  • According to another embodiment, the cells for culturing the matrix express a biologically active molecule of interest. The biologically active molecule of interest can comprise a growth factor. In one embodiment, the biologically active molecule of interest comprises a VEGF protein, a stromal derived growth factor-1 protein, or a bone morphogenic protein.
  • In another embodiment, the matrix conditioning cells are genetically-modified cells, and in a related embodiment, the genetically-modified cells express a biologically active molecule of interest. In a further embodiment, the biologically active molecule of interest is a recombinant protein.
  • In yet another embodiment of the invention, the matrix is harvested from a tissue of a vertebrate.
  • A further aspect of the invention generally involves a composition for tissue replacement, repair, restoration, or remodeling in a patient. The composition comprises a decellularized matrix comprising a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof. The matrix is conditioned by culturing cells on the matrix in vitro. In one embodiment, the cells for conditioning the matrix are exposed to at least one stressor, and in another embodiment, the matrix is exposed to at least one stressor.
  • A further aspect of the invention features a composition comprising a matrix. The matrix is conditioned for tissue restoration, remodeling, replacement or repair by exposure to at least one stressor. In one embodiment, the stressor is an electric current.
  • By “biologically active molecule” is meant a molecule capable of causing an effect on, interaction with, or response from living matter.
  • The term “contacting” includes both direct and indirect contact, including fluid communication.
  • By “genetically modified cell” is meant a cell that includes an exogenous nucleic acid sequence.
  • By “exogenous nucleic acid sequence” is meant a nucleic acid which is introduced into a cell by transduction. The exogenous nucleic acid can be a gene which encodes a protein of interest or can be a nucleic acid sequence which alters the expression of a protein of interest, for example, upregulates the expression and production of the protein of interest. An exogenous nucleic acid sequence includes a nucleic acid that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.
  • By “transfection” is meant a process of introducing genes or gene fragments into cells.
  • By “transduced” is meant a process of inserting exogenous nucleic acids into cells. The insertion may, for example, be effected by transfection or transformation, viral infection, injection, transfection, gene bombardment, electroporation or any other means effective in introducing nucleic acids into cells. Following transduction, the exogenous nucleic acid is either integrated wholly or in part, to the cell's genome (DNA), or remains external to the cell's genome, thereby providing stably transduced or transiently transduced cells.
  • The term “primary cell” includes cells present in a suspension of cells isolated from a tissue source (prior to their being plated, i.e., attached to a tissue culture substrate such as a dish or flask), cells present in an explant derived from tissue, both of the previous types of cells plated for the first time, and cell suspensions derived from these plated cells. “Secondary cells” refers to cells at all subsequent steps in culturing. That is, the first time a plated primary cell is removed from the culture substrate and replated (passaged), it is referred to as a secondary cell, as are all cells in subsequent passages. By “immortalized cell” is meant a cell from an established cell line that is capable of cell division producing progeny that are capable of cell division in culture.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to a composition that is conditioned for replacing, repairing, restoring or remodeling tissue when implanted in a host. The composition includes a matrix derived from naturally occurring ECM or from structural or functional components of a naturally occurring ECM or their degradation products. The composition can be conditioned in various ways, including culturing cells on the matrix, exposing the cells and/or the matrix to altered environmental and/or physiological conditions, or a combination thereof. Culturing cells on the matrix and/or exposing the cultured cells and matrix to various stressors provides a conditioned composition with enhanced biological properties compared to matrices that are merely combined with various cells and not decellularized or combined with bioactive factors by chemical or physical methods. The conditioned matrices according to the invention retain the properties integrated into the matrix by the cultured cells, or by the application of stressors, and are able to deliver the resulting molecular properties of the conditioned matrix when implanted in a host in a manner that more closely simulates the natural state. Thus, the conditioned composition according to the invention supports the replication, differentiation, maturation and spatial organization of cells, therefore augmenting regeneration, replacement, repair, restoration and/or remodeling of an organ or tissue in a patient.
  • According to one aspect, the invention includes a composition comprising a decellularized matrix. The matrix is conditioned by cells cultured on the matrix in vitro whereby the decellularized conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
  • In another aspect, the invention provides a composition comprising a matrix. The matrix is conditioned by cells cultured on the matrix in vitro and by exposure to at least one stressor, whereby the conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
  • The invention further provides a method for inducing restoration, remodeling, replacement, or repair of a tissue in a mammal. The method comprises the steps of providing a decellularized matrix, then conditioning the matrix by culturing cells on the matrix, and lastly, decellularizing the matrix wherein the decellularized, conditioned matrix induces restoration, remodeling, replacement, or repair of the tissue.
  • In another aspect, the invention includes a composition for tissue replacement, repair, restoration, or remodeling, comprising a decellularized matrix including a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof. The matrix is conditioned by culturing cells on the matrix in vitro.
  • In yet another aspect, the invention features a composition comprising a matrix and the matrix is conditioned for tissue restoration, remodeling, replacement or repair by exposure to at least one stressor.
  • According to embodiments of the invention, the matrix, for example, comprises a naturally occurring ECM isolated from the tissues of vertebrates, including, for example, alimentary, respiratory, intestinal, urinary or genital tracts of mammals. In addition, the naturally occurring ECM can comprise at least a portion of tunica submucosa, epithelial basement membrane, tunica propria, or dermis. In other embodiments, the naturally occurring ECM comprises, for example, at least a portion of subcutaneous tissue, respiratory tissue, pancreatic connective tissue, hepatic connective tissue, tissue harvested from the stomach, tissue harvested from the intestine, tissue harvested from the urinary bladder, tissue harvested from the urinary tract, tissue harvested from the liver, tissue harvested from the genital tract, tissue harvested from the skin, or tissue harvested from a tissue comprising a mucous membrane. Alternatively, the naturally occurring ECM comprises a combination of the above, for example, the ECM may comprise tissue harvested from the urinary bladder including a portion of the epithelial basement membrane and a portion of the tunica propria.
  • Alternatively, in another embodiment, the matrix comprises structural or functional components of naturally occurring ECMs, degradation products of structural or functional components of naturally occurring ECMs, or a combination thereof. The structural or functional components comprise, for example, collagen (i.e., collagen types I-XIX), hyaluronic acid, laminin, fibronectin, elastin, purified components of naturally occurring ECMs, or a combination thereof. Matrixes can include highly conserved collagens, glycoproteins, proteoglycans and glycosaminoglycans in their natural configuration and natural concentration.
  • According to one embodiment of the invention, cells are cultured on and/or within the matrix, and the cells comprise a primary, secondary and/or immortalized cell population. The cells are, for example, fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof, and hepatocyte and precursor cells thereof. In a particular embodiment, the cells cultured on and/or within the matrix remain on and/or within the matrix when the matrix is implanted into a host. In another embodiment, primary, secondary or immortalized cells, including autologous cells, are added to a conditioned matrix. These cells can be grown on or within the conditioned matrix and implanted with the conditioned composition at an anatomical site in a patient.
  • In one embodiment, the cells express a biologically active molecule of interest. The biologically active molecule of interest is produced by the cells and is integrated within the matrix while the cells are cultured on or within the matrix. The biologically active molecule of interest is, for example, a protein, including, for example, a growth factor. The growth factor is, for example, an angiogenic or osteogenic growth factor. In additional embodiments, the protein includes angiogenin, angiopoietin-1, Del-1, acidic-fibroblast growth factor (aFGF), basic-fibroblast growth factor (bFGF), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), Interleukin-8 (IL-8), leptin, placental growth factor (PlGF), platelet-derived endothelial cell growth factor (PD-ECGF), platelet-derived growth factor-BB (PDGF-BB), pleiotrophin (PTN), proliferin, transforming growth factor-alpha (TGF-alpha), transforming growth factor-beta (TGF-beta), tumor necrosis factor-alpha (TNF-alpha), vascular endothelial growth factor (VEGF), bone morphogenetic protein-2 (BMP-2), and/or stromal derived growth factor-1.
  • In another embodiment, the cells are genetically modified. In one embodiment according to the invention, the genetically modified cells are transfected cells that comprise an exogenous nucleic acid and that express a protein of interest, or the genetically modified cells are cells transfected with a nucleic acid that encodes a protein of interest. Additionally, the genetically modified cells express one or more biologically active molecules of interest, such as, for example, a VEGF protein, a bFGF protein, a BMP protein, a stromal derived growth factor-1 protein, or a recombinant protein.
  • Cells cultured on the matrix can also be exposed to at least one stressor or a combination of stressors to enhance the cells' ability to condition the matrix. Exposure to certain stressors can stimulate the release of biologically active molecules from the cells and contribute to the conditioning of the composition for various purposes. Exposure to certain stressors can also prevent the release of undesired biologically active molecules from the cells and contribute to the conditioning of the composition to avoid undesired effects when implanted in a host. According to one embodiment, the stressor is hypoxia, and in another embodiment, the stressor is hypercarbia. Exposure to hypoxia and/or hypercarbia can assist the cells in producing biologically active molecules that are incorporated in the matrix on which the cells are cultured and support, for example, the growth of new blood vessels and assist in the restoration of blood flow to tissue in and surrounding the implantation site. Exposure time to each stressor can vary, for example, from 10 minutes to 1 month, depending on the intended use of the conditioned composition, the type of tissue involved and/or the type of matrix being conditioned.
  • In other embodiments, the cells are exposed to stressors such as mechanical loading. Exposure to mechanical loading, for example, enhances cellular production of structural proteins and other biologically active molecules, integrating them into the matrix to improve its tensile strength. According to one embodiment, exposure to mechanical loading is achieved by consistently applying strain (i.e., applying a force that causes a change in length) or by applying stress (i.e., applying a mechanical load) to the matrix over a predetermined period of time, for example, for a total time period of 168 hours. Alternatively, mechanical conditioning can be achieved by applying stress or strain to the matrix in a cyclical pattern, for example, applying stress or strain for 1 minute every 5 minutes over a total time of 48 hours. The stress or strain on the matrix is constant throughout the time period, or alternatively, the stress or strain can vary. The amount of stress or strain can vary, for example, from 5% to 15% using an appropriate device, such as a load cell. The stress or strain can be applied in a uniaxial or multiaxial direction. The amount of strain applied can be measured in the cell culture systems by any means known in the art, for example, by laser measurements using laser beams and deflection times.
  • In another embodiment of the conditioned composition, the cells cultured on the matrix are exposed to an electric current. Exposure to electrical current can, for example, stimulate the release of growth factors and other biologically active molecules from the cell into the matrix, resulting in the matrix exhibiting accelerated tissue regenerative and wound healing properties. The cells are exposed to an electric current for a consistent period of time, or the cultured cells are exposed to an electric current in a cyclical pattern, for example, applying the electric current or electrical potential for 10 seconds every 1 minute over a total time of 1 hour. The electric current potential remains constant over the entire exposure time period, or, alternatively, the magnitude of the electric current potential can alternate between, for example, a positive and negative potential within, for example, a 100 mV-300 mV range.
  • In another embodiment, cells are not cultured on the matrix, and the matrix is exposed to at least one stressor to condition the matrix for its intended use. The stressor can stimulate the release of biologically active molecules or, alternatively, the stressor can be applied to decrease or prevent the release of undesired biologically active molecules into the matrix. The matrix can be exposed, for example, to an electric current or electrical potential. Exposure to electric current or electrical potential can decrease or prevent the release of certain growth factors, such as TGF-beta, from cells existing on the matrix, resulting in a conditioned matrix that decreases the risk of adverse events, such as, for example, the formation of scar tissue, when implanted in a host.
  • The conditioned composition is useful in replacing, repairing, restoring or remodeling various types of tissue, for example, bone, skin, heart, tissue of the urogenital tract, tissue of the gastrointestinal tract, nervous tissue, joint tissue and connective tissue. The anatomical site in which the conditioned composition will be placed in the body, and/or the other desired physiological effects, e.g., angiogenesis and osteogenesis, play a role in selecting which, if any, cells are cultured on the matrix, as well as which, if any, biologically active molecule is of interest and what, if any, stressor the cells and/or matrix will be exposed to.
  • For example, the decellularized, conditioned composition can be used to promote vascularization by providing a decellularized scaffold including a matrix conditioned by cells cultured on the matrix that express an angiogenic or vasculogenic protein of interest to the anatomical site in the host requiring vascularization. The decellularized, conditioned composition can also be used, for example, for musculotendinous tissue reconstruction by providing a decellularized composition conditioned with cultured cells that express a structural protein of interest to the anatomical site in the host requiring musculotendinous reconstruction. Additionally, the decellularized, conditioned composition can be conditioned with cells to express a growth factor of interest to stimulate growth of the host's tissues surrounding the composition when that composition is introduced to the anatomical site in the host that is in need of treatment.
  • The following examples provide further details of practicing the invention. While certain methods useful for practicing the present invention are exemplified below, the invention is not so limited and the skilled artisan will appreciate its wide range of application upon consideration thereof.
  • EXAMPLE 1 Sources and Preparation of the Matrix
  • The following provides exemplary methods for preparing a matrix for use according to the invention. A particular example illustrates preparation of a matrix comprising a naturally occurring ECM. Preparation of the naturally occurring ECM for use according to the invention is also described in U.S. Pat. Nos. 4,902,508, 4,956,178, 5,554,389, 6,576,265 and 6,579,538, the entirety of each patent is incorporated by reference herein. For example, intestinal submucosal tissue can be prepared by harvesting tissue from a vertebrate such as porcine, ovine or bovine species. The tissue is subjected to abrasion using a longitudinal wiping motion to remove the outer layers, smooth muscle tissues, and the innermost layer, i.e., the luminal portion of the tunica mucosa. The remaining submucosal tissue or any portion of it is then rinsed with saline.
  • Alternatively, a naturally occurring ECM including epithelial basement membrane derived from urinary bladder (UBM) can be prepared by removing the urinary bladder tissue from a vertebrate, for example, a pig, and delaminating the tissue by first soaking the tissue in a deepithelializing solution, for example, hypertonic saline, most preferably 1.0 N saline, for periods of time ranging from 10 minutes to 4 hours. Exposure to a hypertonic saline solution effectively removes the epithelial cells from the underlying basement membrane. The tissue remaining after the initial delamination procedure includes epithelial basement membrane and the tissue layers abluminal to the epithelial basement membrane. This tissue is next subjected to further treatment to remove the majority of abluminal tissues but not the epithelial basement membrane. The outer serosal, adventitial, smooth muscle tissues, submucosa and abluminal portion of the tunica propria are removed from the remaining deepithelialized tissue by mechanical abrasion or by a combination of enzymatic treatment, hydration, and abrasion. Mechanical removal of these tissues is accomplished by removal of mesenteric tissues with, for example, Adson-Brown forceps and Metzenbaum scissors and wiping away the tunica muscularis and abluminal tunica propria using a longitudinal wiping motion with a scalpel handle or other rigid object wrapped in moistened gauze. After these tissues are removed, the resulting ECM consists of epithelial basement membrane and subjacent tunica propria. The tissues may be further processed by rinsing in hypertonic saline, peracetic acid or sterile water. Other methods for removing tissue layers, a microtome, for example, may also be used to obtain the tissue composition of the invention.
  • Alternatively, the matrix according to the invention comprises a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof. The structural and functional components can include, for example, collagen (i.e., any of collagen types I-XIX), hyaluronic acid, laminin, fibronectin, elastin, purified components of the naturally occurring ECM, or a combination thereof. Matrixes can include, for example, highly conserved collagens, glycoproteins, proteoglycans and glycosaminoglycans in their natural configuration and natural concentration. Methods of preparing a matrix comprising structural or functional components of a naturally occurring ECM and their degradation products are well known in the art, for example, as described in U.S. Pat. Nos. 6,572,650 and 6,051,750, the entirety of each patent is incorporated by reference herein.
  • The matrix can be stored in a hydrated or dehydrated state. Lyophilized or air dried matrix can be rehydrated and used in accordance with this invention.
  • In one embodiment, the matrix can be sterilized prior to the addition of cells using conventional sterilization techniques including glutaraldehyde tanning, formaldehyde tanning at acidic pH, propylene oxide treatment, ethylene oxide, gas plasma sterilization, gamma radiation, electron beam, peracetic acid sterilization. Sterilization techniques which do not adversely affect the mechanical strength, structure, and biotropic properties of the tissue are preferred. For instance, strong gamma radiation may cause loss of strength of the sheets of submucosal tissue. Preferred sterilization techniques include exposing the graft to peracetic acid, 1-4 Mrads gamma irradiation (more preferably 1-2.5 Mrads of gamma irradiation) or gas plasma sterilization; peracetic acid sterilization is the most preferred sterilization method. Typically, the tissue is subjected to two or more sterilization processes. After the tissue is sterilized, for example by chemical treatment, the tissue may be wrapped in a plastic or foil wrap and sterilized again using electron beam or gamma irradiation sterilization techniques.
  • Compositions comprising submucosal tissue can be used for supporting growth or proliferation of eukaryotic cells in vitro. Submucosal tissue can be used in accordance with this invention as a cell growth substrate in a variety of forms, including its naturally occurring sheet-like configuration, as a gel matrix, as an addition for art-recognized cell/tissue culture media, or as coating for culture-ware to provide a more physiologically relevant substrate that supports and enhances the proliferation of cells in contact with the matrix. The submucosal tissue provides surfaces for cell adhesion and also helps induce cell differentiation.
  • While the matrix is preferably sterilized prior to the addition of cells, nonsterile matrix can be used if antibiotics are included in the cell culture system.
  • The matrix can be decellularized after cells have been cultured on the matrix using any technique known in the art. For example, the matrix can be decellularized by placing the matrix in a decellularizing solution such as in hypertonic saline, for example, 1.0N saline or in Hanks buffered saline solution (HBSS).
  • After the matrix has been conditioned according to embodiments of the invention, primary, secondary or immortalized cells, including autologous cells, can be added to and grown on and/or within the conditioned matrix and implanted with the conditioned composition at an anatomical site in a patient. The type of primary, secondary, and/or immortalized cell is chosen based upon the intended use of the conditioned composition, the anatomical site of implantation of the conditioned composition or the desired physiological properties of the conditioned composition.
  • EXAMPLE 2 Sources and Preparation of Cells
  • The following provides exemplary sources and methods for the preparation of matrix conditioning cells that may be cultured on the matrix to make the conditioned matrix according to the invention. The composition and configuration of the matrix provides a unique cell growth substrate that promotes the attachment and proliferation of various types of cells. Generally, methods involve contacting the cells with the matrix under conditions conducive to cell growth and which are well known in the art.
  • The matrix conditioning cells can be prokaryotic or eukaryotic cells such as vertebrate cells (particularly mammalian cells, such as cells derived from a human, chimpanzee, mouse, rat, hamster, guinea pig, rabbit, cow, horse, pig, goat, sheep, dog, or cat). The matrix conditioning cells can be primary cells, secondary cells, and/or immortalized cells, and the matrix conditioning cells can be autologous cells. The types of matrix conditioning cells include fibroblasts, keratinocytes, epithelial cells (e.g., mammary epithelial cells, intestinal epithelial cells), endothelial cells, adipocytes, astrocytes, cardiac muscle cells, chondrocytes, glial cells, neural cells formed elements of the blood (e.g., lymphocytes, bone marrow cells), muscle cells, gangliocytes, glandular cells, glial cells, hematopoietic cells, mesenchymal cells, hepatocytes and precursors of these somatic cell types.
  • Additionally, the matrix conditioning cells can be genetically modified cells. Various genetically modified mammalian host cell lines can be used, for example, L cells, C127, 3T3 fibroblasts, T-84 cells, Chinese hamster ovary (CHO), HeLa, BHK cell lines, CV-1 cells (ATCC CCL70), COS-7 cells and CV-1/EBNA. A matrix conditioning cell of choice can be genetically modified by transducing into the cell exogenous nucleic acid sequences. The exogenous nucleic acid can encode a biologically active molecule of interest, such as a protein, or alternatively, the exogenous DNA can be a regulatory sequence that will activate expression of an endogenous gene (for example, using homologous recombination) to produce a biologically active molecule of interest. These techniques are well known and may be found in, e.g., Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y.) and other laboratory manuals.
  • EXAMPLE 3 Biologically Active Molecules of Interest
  • Any desired biologically active molecule can be selected for secretion onto the matrix. The biologically active molecule of interest is chosen based on where the conditioned composition is to be located in the body and/or the physiological requirements of the recipient. For example, if the decellularized, conditioned composition is used to repair bone, the biologically active molecule of interest which is produced on the matrix can be a bone growth factor, such as a bone morphogenic protein, e.g., BMP-2. The biologically active molecule of interest can include enzymes, hormones, cytokines, colony stimulating factors, vaccine antigens, antibodies, clotting factors, angiogenesis factors, regulatory proteins, transcription factors, receptors, and structural proteins, for example, epithelial basement membrane proteins such as type IV collagen, laminin, and fibronectin. The biologically active molecule of interest can also include human growth hormone, Factor VIII, Factor IX, erythropoietin, and insulin. For genetically modified cells, nucleic acid sequences and amino acid sequences for biologically active molecules of interest can be readily obtained from National Center for Biotechnology Information. (http://www.ncbi.nlm.nih.gov/).
  • In a particular example, a biologically active molecule of interest is expressed in the genetically modified matrix conditioning cell using an expression vector such as a mammalian expression vector. Mammalian expression vectors typically contain non-transcribed elements such as an origin of replication, a suitable promoter, such as, for example, the ROSA promoter, and an enhancer linked to the gene to be expressed, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences, such as necessary ribosome binding sites, a poly-adenylation site, splice donor and acceptor sites, and transcriptional termination sequences. The transcriptional and translational control sequences in expression vectors to be used in transforming vertebrate cells may be provided by viral sources. Commonly used promoters and enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of an exogenous DNA sequence. The early and late promoters are particularly useful because both are obtained easily from the virus as a fragment that also contains the SV40 viral origin or replication.
  • For secretion of the biologically active molecule of interest, for example, a protein of interest, the expression vector may comprise DNA encoding a signal or leader peptide such as the native signal sequence of IL-7 or interleukin-4.
  • The biologically active molecule of interest can include a recombinant protein. High-yield production of recombinant proteins can be produced by cells that stably express a protein of interest. Cell lines which stably express the protein of interest may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • Vector DNA can be introduced into eukaryotic matrix conditioning cells via conventional transduction techniques such as using calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation.
  • Following the introduction of the exogenous DNA, engineered matrix conditioning cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. Preferred selectable markers include G418, hygromycin and methotrexate. Such engineered cell lines can then be added to the matrix and cultured, and a biologically active molecule of interest can be secreted onto and integrate into the matrix.
  • EXAMPLE 4 Culturing Cells on the Matrix
  • Matrix conditioning cells can be cultured, in vitro, on and/or within the matrix, using a matrix prepared by any of the methods described above. The cells are cultured under conditions conducive to cell growth and if desired, under conditions conducive to expression of a biologically active molecule of interest. Conditions for cell culturing are well known in the art and are dependent on the particular cell type. While not wishing to be bound by theory, it is believed that a biologically active molecule that is expressed by matrix conditioning cells on the matrix, in vitro, is incorporated into the matrix similar to the manner by which molecules, such as proteins, are integrated into a naturally occurring extracellular matrix in vivo. The incorporation of the molecule produced by the cultured cells into the matrix imparts superior restorative properties to this conditioned composition compared to the mere addition of, for example, a protein to a decellularized matrix. It is believed that the incorporated biologically active molecule is released in the recipient of the conditioned composition as a function of composition degradation. Additionally, the conditioned composition can be substantially devoid of cellular elements that may lead to rejection of the tissue implant by the recipient.
  • EXAMPLE 5 Uses of the Conditioned Composition of the Invention
  • The composition of the invention can be implanted into any anatomical location in the body of the host where there exists a tissue in need of regeneration, repair, replacement, restoration or remodeling.
  • A. Conditioned Composition for Enhanced Vascularization
  • Angiogenesis and vasculogenesis are an integral part of the restoration of blood flow to transplanted tissue. The process of angiogenesis involves the migration and proliferation of capillary endothelial cells in the early post-injury phase, and the differentiation of the capillary endothelial cells in the later post-injury phase to organize endothelial cells into a microvascular network. Angiogenesis requires dynamic interactions between cells such as fibroblasts, epithelial and endothelial cells, and a wide array of angiogenic and vasculogenic factors and cytokines that exist within the matrix, such as FGF, VEGF, TGFβ, and others. These cells and matrix composites orchestrate the proliferative, inhibitory, and differentiation phases of cells to re-populate, repair and restore damaged tissues and its vascular supply.
  • The following experiment relates to the use of the conditioned composition of the invention in relation to vasculogenesis and myelogenesis. A naturally occurring ECM derived from urinary bladder was cultured with matrix conditioning transfected cardiac myofibroblasts that constitutively express SDF-1. SDF-1 concentration in the media and in the matrix was measured over a six day time period. Results showed that the concentration of SDF-1 in the scaffold increased more than 100 fold in just six days (from less than 5 pg/ml to 625 pg/ml).
  • The SDF-1 conditioned matrix was decellularized as described above and implanted subcutaneously into mice that have been genetically altered to express green fluorescent protein in all bone marrow-derived cells. Control mice were similarly treated using non-conditioned naturally occurring ECM or simply a sham operation. Blood vessel formation in the mice was measured and quantified by performing morphometric assays on cross sectional samples taken from predetermined sites in low magnification fields. Following 7 and 14 days, mice treated with the decellularized, conditioned scaffold of the invention had increased blood vessel formation and increased numbers of bone marrow-derived cells compared with control mice. Results demonstrated a percentage increase in blood vessel cross sectional area of 48%+/−13%, p<0.05 compared to a control (non-conditioned) matrix.
  • According to exemplary methods, the conditioned compositions of the invention are used to improve angiogenesis in the composition after implantation. Exposure of the matrix conditioning endothelial cells to a hypoxic environment assists the matrix conditioning cells in releasing a natural complement of angiogenic factors into the matrix that will enhance the matrix's specificity. The period of exposure time can range from one minute to thirty minutes. Such angiogenesis and vasculogenesis factors can include one or more of the following proteins: FGF, VEGF, TGFβ, and Angiopoietin. The matrix is decellularized but retains the angiogenic factors that have become integrated in the matrix. The conditioned matrix is then implanted within a recipient in need of vascular assistance and results in enhanced vascularization properties in vivo.
  • In a particular experiment, a naturally occurring ECM was prepared as explained above. Matrix conditioning endothelial cells (EC) were delivered to the matrix and cultured on the matrix under normoxic conditions for a period of 72 hours, with the last 24 hours in serum-free media. The cultured cells were then exposed to a hypoxic environment having 2% oxygen for a period of 48 hours. The concentration of the 165 and 189 isoforms of VEGF protein was measured in both the matrix and the supernatant. Results showed a 5-fold increase of VEGF concentration in the matrix and a 10-fold increase of VEGF in the supernatant as compared to a control in which EC cells were cultured under the same conditions but not exposed to a hypoxic environment.
  • One illustrative experiment pertains to use of the invention in regenerating heart tissue. A naturally occurring ECM derived from the small intestine measuring approximately 1 cm×2 cm is prepared as described above and is seeded with matrix conditioning fibroblasts at 0.5×106 cells/cm2. The matrix conditioning fibroblasts are genetically engineered to express stromal derived growth factor-1 (SDF-1). The matrix conditioning genetically modified cells are cultured with the matrix during a time period ranging from 24 to 168 hours. The presence of SDF-1 in the conditioned matrix is determined by ELISA. The matrix is then decellularized by placing the matrix in HBSS. Following decellularization, the conditioned matrix is implanted by a surgical procedure to a damaged area of the myocardium in the recipient. As the decellularized, conditioned composition is degraded by the recipient, the protein of interest, for example, SDF-1, is released along with other products of matrix degradation. Thereafter, the conditioned composition attracts circulating multi-potential cells that are influenced, for example, to differentiate towards cardiomyocytes. The period of exposure time can range from one minute to thirty minutes.
  • In another illustrative example, a naturally occurring ECM is prepared as described above and is seeded with matrix conditioning cells that are genetically modified to produce factors which improve angiogenesis and/or vasculogenesis. Such angiogenesis and vasculogenesis factors can include one or more of the following proteins: FGF, VEGF, TGFβ, and Angiopoietin. After a suitable incubation period, the matrix is decellularized by methods described above. After decellularization, the conditioned composition is implanted at a location in the host that is in need of new blood vessel formation, for example, into heart tissue for the treatment of ischemic myocardium caused by, for example, a myocardial infarct.
  • Although the examples provided above refer to the use of a naturally occurring ECM, the experiments can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • B. Conditioned Composition for Enhanced Strength
  • The following is an exemplary method for using the invention to enhance the strength of a composition for tissue repair, replacement, restoration or remodeling. The matrix is prepared as explained above, and matrix conditioning fibroblast cells are introduced to the matrix and exposed to mechanical loading. A cyclic loading system that allows for independent control of stress and strain is applied to the matrix in a bioreactor environment and a cell culture system provides for sterile conditions while simultaneously monitoring the stress and strain load in the matrix. The cell culture system includes, for example, a load cell with 25 N capacity and a linear actuator with a 30 mm travel capacity. The system allows the user to apply a load to each matrix and define the displacement profile by specifying the amplitude and frequency of oscillation or by defining a function to prescribe the displacement. Exposure of the matrix and/or matrix conditioning fibroblast cells to mechanical loading assists in the release of structural proteins and other biologically active molecules from the cells and into the matrix. The conditioned composition including the matrix is decellularized prior to implantation in a host.
  • In one illustrative example, a matrix comprising a naturally occurring ECM is prepared as described above and genetically modified cells that secrete structural proteins, such as collagen, after a suitable incubation period are cultured on the matrix. The matrix is decellularized prior to implantation into a host to form the conditioned composition according to the invention.
  • In a particular example, 0.5×106 cm2 of fibroblasts were introduced to a naturally occurring ECM derived from the small intestine measuring approximately 1 cm×2 cm. The cells were cultured on the matrix for approximately 12 hours, during which time a preload amount of 0.25 N was applied. A strain of 10% was then applied to the matrix at 1 Hz for 24 hours. The load placed upon the matrix was monitored continuously during the experiment, and the cell number and pH of the media were determined at the conclusion of the experiment. Cell viability was determined using the MTT assay to stain metabolically active cells. The presence of procollagen Type I was determined with the use of an ELISA kit. Results showed that the fibroblasts rapidly aligned along the lines of applied stress and assumed an elongated, spindloid morphology. Such results indicate that the strength of the conditioned composition will be enhanced compared to compositions not similarly conditioned when implanted in a host.
  • The experiments described above can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • C. Conditioned Composition for Bone Formation
  • In another illustrative example, the conditioned composition of the invention is used to promote bone formation. A matrix comprising a naturally occurring ECM is prepared as described above. Matrix conditioning genetically modified cells that secrete the bone growth factor, bone morphogenetic protein-2 (BMP-2), are cultured on the matrix for a suitable incubation period. The matrix is then decellularized prior to implantation into a host. The same experiment can be performed using a matrix comprising structural or functional components of a naturally occurring ECM.
  • D. Conditioned Composition for Inducing Growth of a Tissue
  • According to the following exemplary method, a matrix comprising a naturally occurring ECM is prepared as described above and matrix conditioning cells that produce growth factors and other biologically active molecules after a suitable incubation period are cultured on the matrix. The cultured cells are exposed to either constant or alternating electrical potential for the purpose of stimulating the release of growth factors and other biologically active molecules from the cells into the matrix prior to implantation. In one illustrative example, the matrix is seeded with a population of matrix conditioning endothelial cells prior to its exposure to electrical potential. An electric potential in the range of 25 mV to about 500 mV, preferably 300 mV, is applied across the matrix for 24 hours to 72 hours, preferably for 48 hours. Upon the cells exposure to electric current, growth factors and other biologically active molecules, such as, for example, proteins, enzymes, hormones, and cytokines, are released from the cells and are incorporated into the matrix. The matrix is then decellularized prior to its implantation into the host. The method can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • Scattered reports exist that applied electrical potential can facilitate various forms of wound healing. Applying low level electrical potential to severely ischemic tissue in the rabbit has been reported to accelerate healing and promote neovascularization of the tissue. Chekanov et al. (2002), Electrical stimulation promotes angiogenesis in rabbi hind-limb ischemia model, Vasc. Endovascular Surg. Vol. 36, pp. 357-366. Electrical potential has been used to accompany acupuncture treatment of wounds and to treat non-healing bone fractures, ulcers and to stimulate nerve regeneration. Sisken et al. (1993), Prospects on clinical applications of electrical stimulation for nerve regeneration, J. Cell Biochem., vol. 51, pp. 404-409; Evans et al. (2001), Electrical stimulation with bone and wound healing, Clin. Podiatr. Med. Surg., vol. 18, pp. 79-95. Data suggests that in these studies, the applied electrical potential stimulates the release of biologically active factors in vivo, either from cells, from the matrices or from both.
  • E. Conditioned Composition for Reducing Undesired Properties
  • According to the following exemplary method, a matrix comprising a naturally occurring ECM is prepared as described above. The matrix is exposed to an electric potential that depletes the matrix of certain biologically active molecules that can result in undesired properties when the matrix is implanted in a host. In one particular example, the matrix is exposed for a period of 72 hours to a constant electric current potential known to reduce or prevent the release of TGF-beta growth factors. The conditioned matrix is then implanted in a host and the risk of undesired scar tissue formation at the site of implantation is decreased.
  • F. Conditioned Composition for Tissue-Specific Compatibility
  • According to the following illustrative method, a matrix comprising a naturally occurring ECM is prepared as described above and conditioned in vitro by the addition of a specific population of matrix conditioning cells, such as astroglial cells, chosen to bestow upon the matrix characteristics known to be unique to the naturally occurring ECM of a particular tissue type, such as tissues of the central nervous system (CNS). In one example, matrix conditioning hepatocytes are introduced and cultured on the matrix to confer upon it the properties inherent to the naturally occurring ECM that surrounds liver tissue. The cultured cells are removed before implantation into a recipient. The decellularized conditioned matrix can direct endothelial cell differentiation, for example, hepatic sinusoidal endothelial cell (HSEC) differentiation and brain endothelial cell (BEC) differentiation, upon transplant of the conditioned matrix into the target tissue of the host. The method can also be performed using a matrix comprising structural or functional components of a naturally occurring ECM and/or degradation products thereof.
  • A naturally occurring ECM is a dynamic structure that reflects the products of the resident cells and is tissue and organ specific. The composition conditioned according to the exemplary methods described above comprises a specialized matrix, in that, once decellularized, the matrix maintains the properties associated with the cellular subtypes present during the conditioning and/or culturing phase. The conditioned composition can be primed for implantation into a target region and will demonstrate improved restorative capabilities as compared with an unconditioned matrix. Endothelial cells (EC), for example, show diverse phenotypic potential. EC phenotype is related to EC function, and function is site dependent. For example, renal glomerular endothelial cells are well-suited for a glomerular filtration function. Hepatic sinusoidal endothelial cells have the unique phenotypic characteristic of fenestrations or “sieve plates” that are essential for normal hepatic blood filtration functions. Alternatively, endothelial cells in the brain are a critical component of the blood-brain barrier that is virtually impermeable to most molecules. Vascular networks within the CNS have no fenestrations (i.e., blood-brain barrier) and express zona occludens 1, 2 and 3 proteins characteristic of tight junctions.
  • Variations and modifications of what is described herein will occur to those of ordinary skill in the art without departing from the spirit and the scope of the invention as claimed. Accordingly, the invention is to be defined not by the preceding illustrative description but instead by the spirit and scope of the following claims.

Claims (66)

1. A composition, comprising:
a decellularized matrix, the matrix conditioned by cells cultured on the matrix in vitro whereby the decellularized conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
2. The composition of claim 1, wherein the matrix comprises a naturally occurring extracellular matrix (ECM).
3. The composition of claim 2 wherein the ECM comprises at least a portion of tunica submucosa.
4. The composition of claim 2 wherein the ECM comprises at least a portion of epithelial basement membrane.
5. The composition of claim 2 wherein the ECM comprises at least a portion of tunica propria.
6. The composition of claim 2 wherein at least a portion of the ECM comprises dermis.
7. The composition of claim 2 wherein at least a portion of the ECM comprises subcutaneous tissue.
8. The composition of claim 2 wherein at least a portion of the ECM comprises pancreatic connective tissue.
9. The composition of claim 2 wherein at least a portion of the ECM comprises tissue harvested from the stomach.
10. The composition of claim 2 wherein at least a portion of the ECM comprises tissue harvested from the intestine.
11. The composition of claim 2 wherein at least a portion of the ECM comprises tissue harvested from the urinary bladder.
12. The composition of claim 2 wherein at least a portion of the ECM comprises tissue harvested from the skin.
13. The composition of claim 2 wherein at least a portion of the ECM comprises tissue harvested from a tissue comprising a mucous membrane.
14. The composition of claim 1 wherein the matrix comprises a structural or functional component of a naturally occurring ECM.
15. The composition of claim 1 wherein the matrix comprises a degradation product of a structural or functional component of a naturally occurring ECM.
16. The composition of claim 1 wherein the matrix conditioning cells are selected from the group consisting of a primary, secondary, and immortalized cell population.
17. The composition of claim 1 wherein the matrix conditioning cells are selected from the group consisting of fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof, and hepatocyte and precursor cells thereof.
18. The composition of claim 1 wherein the matrix conditioning cells express a biologically active molecule of interest.
19. The composition of claim 18 wherein the biologically active molecule of interest comprises a protein.
20. The composition of claim 19 wherein the protein is selected from the group consisting of angiogenin, angiopoietin-1, Del-1, acidic-fibroblast growth factor (aFGF), basic-fibroblast growth factor (bFGF), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), Interleukin-8 (IL-8), leptin, placental growth factor (PlGF), platelet-derived endothelial cell growth factor (PD-ECGF), platelet-derived growth factor-BB (PDGF-BB), pleiotrophin (PTN), proliferin, transforming growth factor-alpha (TGF-alpha), transforming growth factor-beta (TGF-beta), tumor necrosis factor-alpha (TNF-alpha), vascular endothelial growth factor (VEGF), bone morphogenetic protein-2 (BMP-2) and stromal derived growth factor-1.
21. The composition of claim 19 wherein the protein is a growth factor.
22. The composition of claim 21 wherein the growth factor is an angiogenic growth factor.
23. The composition of claim 21 wherein the growth factor is an osteogenic growth factor.
24. The composition of claim 1 wherein the matrix conditioning cells are genetically-modified cells.
25. The composition of claim 24 wherein the genetically-modified cells are transfected cells comprising an exogenous nucleic acid and which express a protein of interest.
26. The composition of claim 24 wherein the genetically-modified cells are transfected with a nucleic acid that encodes a protein of interest.
27. The composition of claim 24 wherein the genetically-modified cells express a biologically active molecule of interest.
28. The composition of claim 27 wherein the biologically active molecule of interest is a VEGF protein, a bFGF protein, a BMP protein, or a stromal derived growth factor-1 protein.
29. The composition of claim 27 wherein the biologically active molecule of interest is a recombinant protein.
30. The composition of claim 2 wherein the ECM is harvested from a tissue of a vertebrate.
31. The composition of claim 1 wherein the tissue in need of restoration, remodeling, replacement or repair comprises a tissue selected from the group consisting of bone, skin, heart, a tissue of the urogenital tract, a tissue of the gastrointestinal tract, nervous tissue, joint tissue and connective tissue.
32. The composition of claim 1 wherein the matrix conditioning cells are exposed to at least one stressor.
33. The composition of claim 32 wherein the at least one stressor comprises hypoxia.
34. The composition of claim 32 wherein the at least one stressor comprises hypercarbia.
35. The composition of claim 32 wherein the at least one stressor comprises an electrical current.
36. The composition of claim 1 wherein the matrix is exposed to at least one stressor.
37. The composition of claim 36 wherein the stressor comprises mechanical loading.
38. A composition, comprising:
a matrix conditioned by cells cultured on the matrix in vitro and by exposure to at least one stressor, whereby the conditioned matrix has enhanced restorative, remodeling, replacement or repair properties when placed in contact with a tissue in need of restoration, remodeling, replacement or repair.
39. The composition of claim 38 wherein the matrix conditioning cells are selected from the group consisting of fibroblasts, keratinocytes, astroglial cells, epithelial cells, endothelial cells, glial cells, neural cells, cells of the blood and precursors thereof, and hepatocyte and precursor cells thereof.
40. The composition of claim 38 wherein the matrix conditioning cells are autologous cells.
41. The composition of claim 38 wherein the at least one stressor comprises hypoxia.
42. The composition of claim 38 wherein the at least one stressor comprises hypercarbia.
43. The composition of claim 38 wherein the at least one stressor comprises an electrical current.
44. The composition of claim 38 wherein the at least one stressor comprises mechanical loading.
45. The composition of claim 38 wherein the matrix conditioning cells express a biologically active molecule of interest.
46. The composition of claim 45 wherein the biologically active molecule of interest comprises a protein.
47. The composition of claim 38 wherein the matrix conditioning cells are genetically-modified cells.
48. The composition of claim 47 wherein the genetically-modified cells express a biologically active molecule of interest.
49. The composition of claim 48 wherein the biologically active molecule of interest is a protein.
50. The composition of claim 49 wherein the protein is a recombinant protein.
51. A method for inducing restoration, remodeling, replacement or repair of a tissue in a mammal, the method comprising:
providing a decellularized matrix;
conditioning the matrix by culturing cells on the matrix in vitro; and
decellularizing the matrix wherein the decellularized conditioned matrix induces restoration, remodeling, replacement or repair of the tissue.
52. The method of claim 51 further comprising the step of exposing the matrix conditioning cells to at least one stressor.
53. The method of claim 51 further comprising the step of exposing the matrix to at least one stressor.
54. The method of claim 51 wherein the tissue comprises a tissue selected from the group consisting of blood vessel tissue, heart tissue and bone tissue.
55. The method of claim 51 wherein the matrix conditioning cells express a biologically active molecule of interest.
56. The method of claim 55 wherein the biologically active molecule of interest comprises a growth factor.
57. The method of claim 55 wherein the biologically active molecule of interest comprises a VEGF protein, a stromal derived growth factor-1 protein, or a bone morphogenic protein.
58. The method of claim 51 wherein the matrix conditioning cells are genetically-modified cells.
59. The method of claim 58 wherein the genetically-modified cells express a biologically active molecule of interest.
60. The method of claim 59 wherein the biologically active molecule of interest is a recombinant protein.
61. The method of claim 51 wherein the matrix is harvested from a tissue of a vertebrate.
62. A composition for tissue replacement, repair, restoration, or remodeling, comprising:
a decellularized matrix comprising a structural or functional component of a naturally occurring ECM, a degradation product of a structural or functional component of a naturally occurring ECM, or a combination thereof, wherein the matrix is conditioned by culturing cells on the matrix in vitro.
63. The composition of claim 62 wherein the matrix conditioning cells are exposed to at least one stressor.
64. The composition of claim 62 wherein the matrix is exposed to at least one stressor.
65. A composition, comprising:
a matrix, the matrix conditioned for tissue restoration, remodeling, replacement or repair by exposure to at least one stressor.
66. The composition of claim 65, wherein the stressor is electric current.
US10/876,299 2003-06-25 2004-06-24 Conditioned compositions for tissue restoration Abandoned US20050025838A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/876,299 US20050025838A1 (en) 2003-06-25 2004-06-24 Conditioned compositions for tissue restoration
US12/351,757 US8409625B2 (en) 2003-06-25 2009-01-09 Conditioned decellularized native tissues for tissue restoration

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US48248003P 2003-06-25 2003-06-25
US53838504P 2004-01-21 2004-01-21
US10/876,299 US20050025838A1 (en) 2003-06-25 2004-06-24 Conditioned compositions for tissue restoration

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/351,757 Continuation US8409625B2 (en) 2003-06-25 2009-01-09 Conditioned decellularized native tissues for tissue restoration

Publications (1)

Publication Number Publication Date
US20050025838A1 true US20050025838A1 (en) 2005-02-03

Family

ID=33567649

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/876,299 Abandoned US20050025838A1 (en) 2003-06-25 2004-06-24 Conditioned compositions for tissue restoration
US12/351,757 Active 2026-07-26 US8409625B2 (en) 2003-06-25 2009-01-09 Conditioned decellularized native tissues for tissue restoration

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/351,757 Active 2026-07-26 US8409625B2 (en) 2003-06-25 2009-01-09 Conditioned decellularized native tissues for tissue restoration

Country Status (6)

Country Link
US (2) US20050025838A1 (en)
EP (1) EP1644011A1 (en)
JP (1) JP2007526017A (en)
AU (1) AU2004253508A1 (en)
CA (1) CA2530490A1 (en)
WO (1) WO2005002601A1 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050192637A1 (en) * 2004-02-27 2005-09-01 Girouard Steven D. Method and apparatus for device controlled gene expression
US20060134071A1 (en) * 2004-12-20 2006-06-22 Jeffrey Ross Use of extracellular matrix and electrical therapy
US20060136028A1 (en) * 2004-12-20 2006-06-22 Jeffrey Ross Epicardial patch including isolated extracellular matrix with pacing electrodes
US20060136027A1 (en) * 2004-12-20 2006-06-22 Randy Westlund Lead electrode incorporating extracellular matrix
US20060147433A1 (en) * 2003-09-04 2006-07-06 Cook Biotech Incorporated Extracellular matrix composite materials, and manufacture and use thereof
US20070014871A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US20070014869A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for reconstruction, replacement or repair of intracardiac tissue
WO2009065093A2 (en) * 2007-11-17 2009-05-22 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
US20100247494A1 (en) * 2009-03-23 2010-09-30 The Texas A&M University System Compositions of Mesenchymal Stem Cells to Regenerate Bone
US20140087000A1 (en) * 2006-01-18 2014-03-27 Robert G. Matheny Method and System for Treatment of Biological Tissue
US20140335046A1 (en) * 2013-05-13 2014-11-13 Robert G. Matheny Compositions, Methods and Systems for Regenerative Cosmetics
US20140342984A1 (en) * 2007-10-19 2014-11-20 Robert G. Matheny Compositions for Regenerating Defective or Absent Myocardium
US20150093353A1 (en) * 2006-01-18 2015-04-02 Cormatrix Cardiovascular, Inc. Method and System for Treatment of Damaged Biological Tissue
WO2013102048A3 (en) * 2011-12-30 2015-06-18 University Of Miami Regenerative tissue matrix
WO2015160952A1 (en) * 2014-04-15 2015-10-22 Pittenger Mark Frings Apparatus or device for cellular therapy
US9238090B1 (en) 2014-12-24 2016-01-19 Fettech, Llc Tissue-based compositions
WO2023059011A1 (en) * 2021-10-08 2023-04-13 에이치엘비셀 주식회사 Method for construction of extracellular matrix through cell culture and use of constructed extracellular matrix
US11679178B2 (en) 2019-02-25 2023-06-20 University Of Rochester Methods for improving mechanical properties of a tissue or for regenerating an injured or diseased tissue

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160082153A1 (en) * 2006-01-18 2016-03-24 Cormatrix Cardiovascular, Inc. Method and System for Treatment of Damaged Biological Tissue
JP5218955B2 (en) * 2007-10-15 2013-06-26 独立行政法人物質・材料研究機構 Porous scaffold for regeneration and method for producing the same
US8257434B2 (en) 2007-12-18 2012-09-04 Cormatrix Cardiovascular, Inc. Prosthetic tissue valve
US8679176B2 (en) 2007-12-18 2014-03-25 Cormatrix Cardiovascular, Inc Prosthetic tissue valve
WO2009098698A2 (en) * 2008-02-07 2009-08-13 Shahar Cohen Compartmental extract compositions for tissue engineering
US8415159B2 (en) * 2008-07-30 2013-04-09 Mesynthes Ltd. Tissue scaffolds derived from forestomach extracellular matrix
JPWO2011142425A1 (en) * 2010-05-12 2013-07-22 国立大学法人 香川大学 Regeneration material and evaluation method for epithelial layer-containing tissue
US9102913B2 (en) 2010-07-02 2015-08-11 The University Of North Carolina At Chapel Hill Biomatrix scaffolds for industrial scale dispersal
CA2856553C (en) 2011-12-09 2017-08-29 Acell, Inc. Hemostatic device
US9801910B2 (en) 2014-03-17 2017-10-31 Ethicon, Inc. Decellularized pleural matrix
JP6681872B2 (en) 2014-03-21 2020-04-15 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 Method for preparing final sterile hydrogel derived from extracellular matrix
US11291688B2 (en) 2017-03-02 2022-04-05 University of Pittsburgh—of the Commonwealth System of Higher Education Extracellular matrix (ECM) hydrogel and soluble fraction thereof for the treatment of cancer
AU2018226867B2 (en) 2017-03-02 2023-11-02 University Of Pittsburgh - Of The Commonwealth System Of Higher Education ECM hydrogel for treating esophageal inflammation
EP3618841B1 (en) 2017-05-05 2023-03-22 University of Pittsburgh - of The Commonwealth System of Higher Education Ocular applications of matrix bound vesicles (mbvs)
US11826490B1 (en) 2020-12-29 2023-11-28 Acell, Inc. Extracellular matrix sheet devices with improved mechanical properties and method of making

Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2127903A (en) * 1936-05-05 1938-08-23 Davis & Geck Inc Tube for surgical purposes and method of preparing and using the same
US3562820A (en) * 1966-08-22 1971-02-16 Bernhard Braun Tubular sheet and strip form prostheses on a basis of biological tissue
US4439521A (en) * 1981-10-21 1984-03-27 Ontario Cancer Institute Method for producing self-reproducing mammalian pancreatic islet-like structures
US4703108A (en) * 1984-03-27 1987-10-27 University Of Medicine & Dentistry Of New Jersey Biodegradable matrix and methods for producing same
US4743552A (en) * 1984-09-26 1988-05-10 Sloan-Kettering Institute For Cancer Research Method for growth in tissue culture of normal colonic epithelial cells and method for determination of preneoplastic color cells
US4776853A (en) * 1986-07-28 1988-10-11 Hsc Research Development Corporation Process for preparing biological mammalian implants
US4801299A (en) * 1983-06-10 1989-01-31 University Patents, Inc. Body implants of extracellular matrix and means and methods of making and using such implants
US4829000A (en) * 1985-08-30 1989-05-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Reconstituted basement membrane complex with biological activity
US4902508A (en) * 1988-07-11 1990-02-20 Purdue Research Foundation Tissue graft composition
US4956178A (en) * 1988-07-11 1990-09-11 Purdue Research Foundation Tissue graft composition
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US5275826A (en) * 1992-11-13 1994-01-04 Purdue Research Foundation Fluidized intestinal submucosa and its use as an injectable tissue graft
US5281422A (en) * 1991-09-24 1994-01-25 Purdue Research Foundation Graft for promoting autogenous tissue growth
US5336616A (en) * 1990-09-12 1994-08-09 Lifecell Corporation Method for processing and preserving collagen-based tissues for transplantation
US5352463A (en) * 1992-11-13 1994-10-04 Badylak Steven F Tissue graft for surgical reconstruction of a collagenous meniscus and method therefor
US5478739A (en) * 1992-10-23 1995-12-26 Advanced Tissue Sciences, Inc. Three-dimensional stromal cell and tissue culture system
US5480424A (en) * 1993-11-01 1996-01-02 Cox; James L. Heart valve replacement using flexible tubes
US5543894A (en) * 1994-07-18 1996-08-06 Xerox Corporation Correction for surface velocity mismatch in multiple servo paper paths
US5554389A (en) * 1995-04-07 1996-09-10 Purdue Research Foundation Urinary bladder submucosa derived tissue graft
US5618312A (en) * 1995-10-31 1997-04-08 Bio-Engineering Laboratories, Ltd. Medical materials and manufacturing methods thereof
US5641518A (en) * 1992-11-13 1997-06-24 Purdue Research Foundation Method of repairing bone tissue
US5695998A (en) * 1995-02-10 1997-12-09 Purdue Research Foundation Submucosa as a growth substrate for islet cells
US5711969A (en) * 1995-04-07 1998-01-27 Purdue Research Foundation Large area submucosal tissue graft constructs
US5753276A (en) * 1995-10-30 1998-05-19 Cimenti; Antonio Machine for rounding pieces of dough
US5755791A (en) * 1996-04-05 1998-05-26 Purdue Research Foundation Perforated submucosal tissue graft constructs
US5762966A (en) * 1995-04-07 1998-06-09 Purdue Research Foundation Tissue graft and method for urinary tract urothelium reconstruction and replacement
US5855620A (en) * 1995-04-19 1999-01-05 St. Jude Medical, Inc. Matrix substrate for a viable body tissue-derived prosthesis and method for making the same
US5866415A (en) * 1997-03-25 1999-02-02 Villeneuve; Peter E. Materials for healing cartilage and bone defects
US5891617A (en) * 1993-09-15 1999-04-06 Organogenesis Inc. Cryopreservation of harvested skin and cultured skin or cornea equivalents by slow freezing
US5899936A (en) * 1994-03-14 1999-05-04 Cryolife, Inc. Treated tissue for implantation and methods of preparation
US5916266A (en) * 1995-10-31 1999-06-29 Bio-Engineering Laboratories, Ltd. Raw membranous material for medical materials and manufacturing methods thereof
US6022887A (en) * 1996-12-13 2000-02-08 Osteoscreen, Inc. Compositions and methods for stimulating bone growth
US6051750A (en) * 1992-08-07 2000-04-18 Tissue Engineering, Inc. Method and construct for producing graft tissue from an extracellular matrix
US6096347A (en) * 1996-11-05 2000-08-01 Purdue Research Foundation Myocardial graft constructs
US6126686A (en) * 1996-12-10 2000-10-03 Purdue Research Foundation Artificial vascular valves
US6171344B1 (en) * 1996-08-16 2001-01-09 Children's Medical Center Corporation Bladder submucosa seeded with cells for tissue reconstruction
US6206931B1 (en) * 1996-08-23 2001-03-27 Cook Incorporated Graft prosthesis materials
US6322593B1 (en) * 1999-04-09 2001-11-27 Sulzer Carbomedics Inc. Method for treating cross-linked biological tissues
US6376244B1 (en) * 1999-12-29 2002-04-23 Children's Medical Center Corporation Methods and compositions for organ decellularization
US6432712B1 (en) * 1999-11-22 2002-08-13 Bioscience Consultants, Llc Transplantable recellularized and reendothelialized vascular tissue graft
US6455311B1 (en) * 1999-04-30 2002-09-24 The General Hospital Corporation Fabrication of vascularized tissue
US6479064B1 (en) * 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
US20020172705A1 (en) * 1998-11-19 2002-11-21 Murphy Michael P. Bioengineered tissue constructs and methods for producing and using thereof
US6485723B1 (en) * 1995-02-10 2002-11-26 Purdue Research Foundation Enhanced submucosal tissue graft constructs
US6485969B1 (en) * 1997-12-23 2002-11-26 Purdue Research Foundation Biomaterial derived from follicle basement membranes
US6572650B1 (en) * 1998-06-05 2003-06-03 Organogenesis Inc. Bioengineered vascular graft support prostheses
US6576265B1 (en) * 1999-12-22 2003-06-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6579538B1 (en) * 1999-12-22 2003-06-17 Acell, Inc. Tissue regenerative compositions for cardiac applications, method of making, and method of use thereof
US20030148510A1 (en) * 2002-01-15 2003-08-07 Mitrani Eduardo N. Methods of inducing differentiation in stem cells, methods of generating tissue using scaffold matrices derived from micro-organs and stem cells, methods of producing adult stem cells and methods of continuously generating stem cells by implantation of micro-organs as sources of stem cells
US20030211130A1 (en) * 2002-02-22 2003-11-13 Sanders Joan E. Bioengineered tissue substitutes
US20040043006A1 (en) * 2002-08-27 2004-03-04 Badylak Stephen F. Tissue regenerative composition
US20040175366A1 (en) * 2003-03-07 2004-09-09 Acell, Inc. Scaffold for cell growth and differentiation
US20040176855A1 (en) * 2003-03-07 2004-09-09 Acell, Inc. Decellularized liver for repair of tissue and treatment of organ deficiency
US6877495B2 (en) * 2002-10-29 2005-04-12 Wetherill Associates, Inc. Vehicle ignition system using ignition module with reduced heat generation

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE215605T1 (en) 1991-06-24 2002-04-15 Pacific Biomedical Res Inc HORMONE-SECRETING PANCREATIC CELLS MAINTAINED IN LONG-TERM CULTURE
GB9413724D0 (en) 1994-07-07 1994-08-24 Wellcome Found Therapeutic nucleosides
US6095148A (en) * 1995-11-03 2000-08-01 Children's Medical Center Corporation Neuronal stimulation using electrically conducting polymers
AU1051597A (en) 1995-11-09 1997-05-29 University Of Massachusetts Tissue re-surfacing with hydrogel-cell compositions
US5869041A (en) * 1996-01-12 1999-02-09 The Miriam Hospital Delivery of bioactive compounds to an organism
BR9712671A (en) 1996-12-10 1999-10-19 Purdue Research Foundation Tubular submucosal graft constructions
CA2271658C (en) 1996-12-10 2009-02-03 Purdue Research Foundation Stent with reduced thrombogenicity
AU732726B2 (en) 1996-12-10 2001-04-26 Purdue Research Foundation Biomaterial derived from vertebrate liver tissue
AU746318B2 (en) 1997-04-11 2002-04-18 Cryolife, Inc. Tissue decellularization
JP2002507907A (en) 1997-06-27 2002-03-12 バーダー、アウグスチヌス Biosynthetic implant and method for producing the same
JP2002525043A (en) 1998-09-11 2002-08-13 パーデュー・リサーチ・ファウンデーション Improved submucosal tissue graft composition
US6454804B1 (en) * 1999-10-08 2002-09-24 Bret A. Ferree Engineered tissue annulus fibrosis augmentation methods and apparatus
DE60031461T2 (en) 1999-12-22 2007-08-23 Acell, Inc., Cambridge Composition for tissue regeneration
US20020098167A1 (en) * 2000-07-31 2002-07-25 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium
CA2419817C (en) 2000-08-16 2014-11-18 Duke University Decellularized tissue engineered constructs and tissues
US6591123B2 (en) * 2000-08-31 2003-07-08 Mallinckrodt Inc. Oximeter sensor with digital memory recording sensor data
CA2438501C (en) * 2001-02-14 2014-09-16 Leo T. Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
JP5007476B2 (en) * 2001-03-23 2012-08-22 オリンパス株式会社 Artificial aggregate
JP2005509495A (en) 2001-11-16 2005-04-14 チルドレンズ メディカル センター コーポレーション Enhancement of organ function

Patent Citations (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2127903A (en) * 1936-05-05 1938-08-23 Davis & Geck Inc Tube for surgical purposes and method of preparing and using the same
US3562820A (en) * 1966-08-22 1971-02-16 Bernhard Braun Tubular sheet and strip form prostheses on a basis of biological tissue
US4439521A (en) * 1981-10-21 1984-03-27 Ontario Cancer Institute Method for producing self-reproducing mammalian pancreatic islet-like structures
US4801299A (en) * 1983-06-10 1989-01-31 University Patents, Inc. Body implants of extracellular matrix and means and methods of making and using such implants
US4703108A (en) * 1984-03-27 1987-10-27 University Of Medicine & Dentistry Of New Jersey Biodegradable matrix and methods for producing same
US4743552A (en) * 1984-09-26 1988-05-10 Sloan-Kettering Institute For Cancer Research Method for growth in tissue culture of normal colonic epithelial cells and method for determination of preneoplastic color cells
US4829000A (en) * 1985-08-30 1989-05-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Reconstituted basement membrane complex with biological activity
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US4776853A (en) * 1986-07-28 1988-10-11 Hsc Research Development Corporation Process for preparing biological mammalian implants
US4902508A (en) * 1988-07-11 1990-02-20 Purdue Research Foundation Tissue graft composition
US4956178A (en) * 1988-07-11 1990-09-11 Purdue Research Foundation Tissue graft composition
US5336616A (en) * 1990-09-12 1994-08-09 Lifecell Corporation Method for processing and preserving collagen-based tissues for transplantation
US5281422A (en) * 1991-09-24 1994-01-25 Purdue Research Foundation Graft for promoting autogenous tissue growth
US5372821A (en) * 1991-09-24 1994-12-13 Purdue Research Foundation Graft for promoting autogenous tissue growth
US5445833A (en) * 1991-09-24 1995-08-29 Purdue Research Foundation Tendon or ligament graft for promoting autogenous tissue growth
US5573784A (en) * 1991-09-24 1996-11-12 Purdue Research Foundation Graft for promoting autogenous tissue growth
US6051750A (en) * 1992-08-07 2000-04-18 Tissue Engineering, Inc. Method and construct for producing graft tissue from an extracellular matrix
US5478739A (en) * 1992-10-23 1995-12-26 Advanced Tissue Sciences, Inc. Three-dimensional stromal cell and tissue culture system
US5641518A (en) * 1992-11-13 1997-06-24 Purdue Research Foundation Method of repairing bone tissue
US5352463A (en) * 1992-11-13 1994-10-04 Badylak Steven F Tissue graft for surgical reconstruction of a collagenous meniscus and method therefor
US5516533A (en) * 1992-11-13 1996-05-14 Purdue Research Foundation Fluidized intestinal submucosa and its use as an injectable tissue graft
US5275826A (en) * 1992-11-13 1994-01-04 Purdue Research Foundation Fluidized intestinal submucosa and its use as an injectable tissue graft
US5891617A (en) * 1993-09-15 1999-04-06 Organogenesis Inc. Cryopreservation of harvested skin and cultured skin or cornea equivalents by slow freezing
US5480424A (en) * 1993-11-01 1996-01-02 Cox; James L. Heart valve replacement using flexible tubes
US5899936A (en) * 1994-03-14 1999-05-04 Cryolife, Inc. Treated tissue for implantation and methods of preparation
US5543894A (en) * 1994-07-18 1996-08-06 Xerox Corporation Correction for surface velocity mismatch in multiple servo paper paths
US5695998A (en) * 1995-02-10 1997-12-09 Purdue Research Foundation Submucosa as a growth substrate for islet cells
US6087157A (en) * 1995-02-10 2000-07-11 Clarian Health Partners Device and method for analyzing tumor cell invasion of an extracellular matrix
US5866414A (en) * 1995-02-10 1999-02-02 Badylak; Stephen F. Submucosa gel as a growth substrate for cells
US6485723B1 (en) * 1995-02-10 2002-11-26 Purdue Research Foundation Enhanced submucosal tissue graft constructs
US5711969A (en) * 1995-04-07 1998-01-27 Purdue Research Foundation Large area submucosal tissue graft constructs
US5762966A (en) * 1995-04-07 1998-06-09 Purdue Research Foundation Tissue graft and method for urinary tract urothelium reconstruction and replacement
US5885619A (en) * 1995-04-07 1999-03-23 Purdue Research Foundation Large area submucosal tissue graft constructs and method for making the same
US5554389A (en) * 1995-04-07 1996-09-10 Purdue Research Foundation Urinary bladder submucosa derived tissue graft
US5855620A (en) * 1995-04-19 1999-01-05 St. Jude Medical, Inc. Matrix substrate for a viable body tissue-derived prosthesis and method for making the same
US5753276A (en) * 1995-10-30 1998-05-19 Cimenti; Antonio Machine for rounding pieces of dough
US5618312A (en) * 1995-10-31 1997-04-08 Bio-Engineering Laboratories, Ltd. Medical materials and manufacturing methods thereof
US5916266A (en) * 1995-10-31 1999-06-29 Bio-Engineering Laboratories, Ltd. Raw membranous material for medical materials and manufacturing methods thereof
US5755791A (en) * 1996-04-05 1998-05-26 Purdue Research Foundation Perforated submucosal tissue graft constructs
US6171344B1 (en) * 1996-08-16 2001-01-09 Children's Medical Center Corporation Bladder submucosa seeded with cells for tissue reconstruction
US6206931B1 (en) * 1996-08-23 2001-03-27 Cook Incorporated Graft prosthesis materials
US6096347A (en) * 1996-11-05 2000-08-01 Purdue Research Foundation Myocardial graft constructs
US6126686A (en) * 1996-12-10 2000-10-03 Purdue Research Foundation Artificial vascular valves
US6022887A (en) * 1996-12-13 2000-02-08 Osteoscreen, Inc. Compositions and methods for stimulating bone growth
US5866415A (en) * 1997-03-25 1999-02-02 Villeneuve; Peter E. Materials for healing cartilage and bone defects
US6485969B1 (en) * 1997-12-23 2002-11-26 Purdue Research Foundation Biomaterial derived from follicle basement membranes
US6572650B1 (en) * 1998-06-05 2003-06-03 Organogenesis Inc. Bioengineered vascular graft support prostheses
US20020172705A1 (en) * 1998-11-19 2002-11-21 Murphy Michael P. Bioengineered tissue constructs and methods for producing and using thereof
US6322593B1 (en) * 1999-04-09 2001-11-27 Sulzer Carbomedics Inc. Method for treating cross-linked biological tissues
US6455311B1 (en) * 1999-04-30 2002-09-24 The General Hospital Corporation Fabrication of vascularized tissue
US6432712B1 (en) * 1999-11-22 2002-08-13 Bioscience Consultants, Llc Transplantable recellularized and reendothelialized vascular tissue graft
US6869619B2 (en) * 1999-12-22 2005-03-22 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6861074B2 (en) * 1999-12-22 2005-03-01 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6576265B1 (en) * 1999-12-22 2003-06-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6579538B1 (en) * 1999-12-22 2003-06-17 Acell, Inc. Tissue regenerative compositions for cardiac applications, method of making, and method of use thereof
US20030133916A1 (en) * 1999-12-22 2003-07-17 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6783776B2 (en) * 1999-12-22 2004-08-31 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6890564B2 (en) * 1999-12-22 2005-05-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6890562B2 (en) * 1999-12-22 2005-05-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6893666B2 (en) * 1999-12-22 2005-05-17 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6890563B2 (en) * 1999-12-22 2005-05-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6852339B2 (en) * 1999-12-22 2005-02-08 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6849273B2 (en) * 1999-12-22 2005-02-01 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6376244B1 (en) * 1999-12-29 2002-04-23 Children's Medical Center Corporation Methods and compositions for organ decellularization
US6479064B1 (en) * 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
US20030148510A1 (en) * 2002-01-15 2003-08-07 Mitrani Eduardo N. Methods of inducing differentiation in stem cells, methods of generating tissue using scaffold matrices derived from micro-organs and stem cells, methods of producing adult stem cells and methods of continuously generating stem cells by implantation of micro-organs as sources of stem cells
US20030211130A1 (en) * 2002-02-22 2003-11-13 Sanders Joan E. Bioengineered tissue substitutes
US20040043006A1 (en) * 2002-08-27 2004-03-04 Badylak Stephen F. Tissue regenerative composition
US6877495B2 (en) * 2002-10-29 2005-04-12 Wetherill Associates, Inc. Vehicle ignition system using ignition module with reduced heat generation
US20040176855A1 (en) * 2003-03-07 2004-09-09 Acell, Inc. Decellularized liver for repair of tissue and treatment of organ deficiency
US20040175366A1 (en) * 2003-03-07 2004-09-09 Acell, Inc. Scaffold for cell growth and differentiation

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9186435B2 (en) 2003-09-04 2015-11-17 Cook Biotech, Incorporated Extracellular matrix composite materials, and manufacture and use thereof
US20060147433A1 (en) * 2003-09-04 2006-07-06 Cook Biotech Incorporated Extracellular matrix composite materials, and manufacture and use thereof
US7795027B2 (en) 2003-09-04 2010-09-14 Cook Biotech Incorporated Extracellular matrix composite materials, and manufacture and use thereof
US7840263B2 (en) 2004-02-27 2010-11-23 Cardiac Pacemakers, Inc. Method and apparatus for device controlled gene expression
US20050192637A1 (en) * 2004-02-27 2005-09-01 Girouard Steven D. Method and apparatus for device controlled gene expression
US20060134071A1 (en) * 2004-12-20 2006-06-22 Jeffrey Ross Use of extracellular matrix and electrical therapy
US20060136028A1 (en) * 2004-12-20 2006-06-22 Jeffrey Ross Epicardial patch including isolated extracellular matrix with pacing electrodes
US20060136027A1 (en) * 2004-12-20 2006-06-22 Randy Westlund Lead electrode incorporating extracellular matrix
US8060219B2 (en) 2004-12-20 2011-11-15 Cardiac Pacemakers, Inc. Epicardial patch including isolated extracellular matrix with pacing electrodes
US7981065B2 (en) 2004-12-20 2011-07-19 Cardiac Pacemakers, Inc. Lead electrode incorporating extracellular matrix
US20070014871A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US20070014869A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for reconstruction, replacement or repair of intracardiac tissue
US20070014874A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US20070014872A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US20140242184A1 (en) * 2005-07-15 2014-08-28 Robert G Matheny Compositions for Regenerating Defective or Absent Myocardium
US20070014873A1 (en) * 2005-07-15 2007-01-18 Cormatrix Cardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US8771737B2 (en) * 2005-07-15 2014-07-08 Cormatrix Cardiovascular, Inc Compositions for regenerating defective or absent myocardium
US8784891B2 (en) * 2005-07-15 2014-07-22 CorMatrixCardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US20130116198A1 (en) * 2005-07-15 2013-05-09 Robert G. Matheny Compositions for Regenerating Defective or Absent Myocardium
US20130129831A1 (en) * 2005-07-15 2013-05-23 Francis Law Group Compositions for Regenerating Defective or Absent Myocardium
US8568761B2 (en) 2005-07-15 2013-10-29 Cormatrix Cardiovascular, Inc. Compositions for regenerating defective or absent myocardium
US20150093353A1 (en) * 2006-01-18 2015-04-02 Cormatrix Cardiovascular, Inc. Method and System for Treatment of Damaged Biological Tissue
US20140087000A1 (en) * 2006-01-18 2014-03-27 Robert G. Matheny Method and System for Treatment of Biological Tissue
US20140342984A1 (en) * 2007-10-19 2014-11-20 Robert G. Matheny Compositions for Regenerating Defective or Absent Myocardium
WO2009065093A3 (en) * 2007-11-17 2009-12-30 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
US20110020291A1 (en) * 2007-11-17 2011-01-27 Debrabrata Banerjee Use of stem cells for wound healing
WO2009065093A2 (en) * 2007-11-17 2009-05-22 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
US9452185B2 (en) * 2009-03-23 2016-09-27 The Texas A&M University System Mesenchymal stem cells and supports for tissue regeneration, repair and reconstruction
US20100247494A1 (en) * 2009-03-23 2010-09-30 The Texas A&M University System Compositions of Mesenchymal Stem Cells to Regenerate Bone
US20120100114A1 (en) * 2009-03-23 2012-04-26 Gregory Carl A Mesenchymal stem cells and supports for tissue regeneration, repair and reconstruction
US9511093B2 (en) 2009-03-23 2016-12-06 The Texas A & M University System Compositions of mesenchymal stem cells to regenerate bone
WO2013102048A3 (en) * 2011-12-30 2015-06-18 University Of Miami Regenerative tissue matrix
US20140335046A1 (en) * 2013-05-13 2014-11-13 Robert G. Matheny Compositions, Methods and Systems for Regenerative Cosmetics
WO2015160952A1 (en) * 2014-04-15 2015-10-22 Pittenger Mark Frings Apparatus or device for cellular therapy
US9238090B1 (en) 2014-12-24 2016-01-19 Fettech, Llc Tissue-based compositions
US11679178B2 (en) 2019-02-25 2023-06-20 University Of Rochester Methods for improving mechanical properties of a tissue or for regenerating an injured or diseased tissue
WO2023059011A1 (en) * 2021-10-08 2023-04-13 에이치엘비셀 주식회사 Method for construction of extracellular matrix through cell culture and use of constructed extracellular matrix

Also Published As

Publication number Publication date
US8409625B2 (en) 2013-04-02
AU2004253508A1 (en) 2005-01-13
EP1644011A1 (en) 2006-04-12
US20090118166A1 (en) 2009-05-07
JP2007526017A (en) 2007-09-13
CA2530490A1 (en) 2005-01-13
WO2005002601A1 (en) 2005-01-13

Similar Documents

Publication Publication Date Title
US8409625B2 (en) Conditioned decellularized native tissues for tissue restoration
Kc et al. Cardiac tissue-derived extracellular matrix scaffolds for myocardial repair: advantages and challenges
Miyagi et al. Biodegradable collagen patch with covalently immobilized VEGF for myocardial repair
Kuo et al. Bioengineering vascularized tissue constructs using an injectable cell-laden enzymatically crosslinked collagen hydrogel derived from dermal extracellular matrix
RU2645473C2 (en) Tissue structures obtained by bioengineering, and methods for their production and application
Reing et al. Degradation products of extracellular matrix affect cell migration and proliferation
Choi et al. Full-thickness skin wound healing using human placenta-derived extracellular matrix containing bioactive molecules
CA2859714C (en) Organoids comprising decellularized and repopulated placental vascular scaffold
US6733747B2 (en) Prosthetic grafts
US20040176855A1 (en) Decellularized liver for repair of tissue and treatment of organ deficiency
US20090074732A1 (en) Scaffold for Cell Growth and Differentiation
Han et al. Microencapsulated VEGF gene–modified umbilical cord mesenchymal stromal cells promote the vascularization of tissue-engineered dermis: an experimental study
TW200813225A (en) Biocompatible scaffolds and adipose-derived stem cells
Gugatschka et al. Regenerative medicine of the larynx. Where are we today? A review
Kim et al. Effect of decellularized extracellular matrix bioscaffolds derived from fibroblasts on skin wound healing and remodeling
US20150359852A1 (en) Methods and compositions for bone formation
US20070081983A1 (en) Isolation of smooth muscle cells and tissue-engineered vasculature containing the isolated cells
EP1887080A1 (en) Endothelized artificial matrix comprising a fibrin gel, which is a superproducer of proangiogenic factors
Roberts et al. Tissue engineering for wound and organ repair: Angiogenesis as a mechanism of action
Congress et al. SPU RESEARCH GRANT UPDATE

Legal Events

Date Code Title Description
AS Assignment

Owner name: ACELL, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BADYLAK, STEPHEN F.;REEL/FRAME:018628/0084

Effective date: 20051221

STCB Information on status: application discontinuation

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