US20040259190A1 - Compositions and methods for production and use of an injectable naturally secreted extracellular matrix - Google Patents

Compositions and methods for production and use of an injectable naturally secreted extracellular matrix Download PDF

Info

Publication number
US20040259190A1
US20040259190A1 US10/851,773 US85177304A US2004259190A1 US 20040259190 A1 US20040259190 A1 US 20040259190A1 US 85177304 A US85177304 A US 85177304A US 2004259190 A1 US2004259190 A1 US 2004259190A1
Authority
US
United States
Prior art keywords
cells
framework
stromal
injectable formulation
tissue
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
US10/851,773
Inventor
Gail Naughton
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Advanced BioHealing Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/470,101 external-priority patent/US5830708A/en
Application filed by Individual filed Critical Individual
Priority to US10/851,773 priority Critical patent/US20040259190A1/en
Publication of US20040259190A1 publication Critical patent/US20040259190A1/en
Assigned to ADVANCED BIOHEALING, INC. reassignment ADVANCED BIOHEALING, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SMITH & NEPHEW WOUND MANAGEMENT (LA JOLLA)
Assigned to SMITH & NEPHEW WOUND MANGEMENT (LA JOLLA), SMITH & NEPHEW, INC. reassignment SMITH & NEPHEW WOUND MANGEMENT (LA JOLLA) SECURITY AGREEMENT Assignors: ADVANCED BIOHEALING, INC.
Assigned to HORIZON TECHNOLOGY FUNDING COMPANY LLC reassignment HORIZON TECHNOLOGY FUNDING COMPANY LLC SECURITY AGREEMENT Assignors: ADVANCED BIOHEALING, INC.
Assigned to ADVANCED BIOHEALING, INC. reassignment ADVANCED BIOHEALING, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: HORIZON TECHNOLOGY FUNDING COMPANY LLC
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • 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
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3839Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by the site of application in the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/60Materials for use in artificial skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/0059Cosmetic or alloplastic implants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/10Hair or skin implants
    • A61F2/105Skin implants, e.g. artificial skin
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T442/00Fabric [woven, knitted, or nonwoven textile or cloth, etc.]
    • Y10T442/20Coated or impregnated woven, knit, or nonwoven fabric which is not [a] associated with another preformed layer or fiber layer or, [b] with respect to woven and knit, characterized, respectively, by a particular or differential weave or knit, wherein the coating or impregnation is neither a foamed material nor a free metal or alloy layer
    • Y10T442/2525Coating or impregnation functions biologically [e.g., insect repellent, antiseptic, insecticide, bactericide, etc.]

Definitions

  • the present invention relates to compositions and methods for the treatment and repair of soft tissue and skin defects such as wrinkles and scars. More particularly, the invention relates to an injectable composition of human extracellular matrix components and methods of preparing and using same.
  • the injectable preparation is obtained from three-dimensional living stromal tissues that are prepared in vitro.
  • bovine collagen has gained widespread use as an injectable material for soft tissue augmentation.
  • Collagen is the principal extracellular structural protein of the animal body.
  • mammalian collagen At least fourteen types of mammalian collagen have been described. The common characteristic amongst them is a three stranded helix, consisting of three polypeptide chains, called alpha-chains. All alpha-chains have the same configuration, but differ in the composition and sequence of their amino acids. Although this leads to different types of alpha-chains, however, they all have glycine at every third position in the amino acid sequence. The glycine at every third position allows for the helical structure of the alpha-chains.
  • Type I collagen is composed of two alpha 1 -chains and one alpha 2 -chain and is the principal extracellular material of skin, tendon and bone.
  • collagen When clinicians mention “collagen”, they are usually referring to type I collagen. See Table I, infra, for a detailed listing of collagen types I-V and in which tissues they are found.
  • Collagen has been used as an implant material to replace or augment hard or soft connective tissue, such as skin, tendon, cartilage, bone and interstitium. Additionally, collagen implants have been used for cosmetic purposes for a number of years since collagen can help cellular ingrowth at the placement site. Early collagen implants were often solid collagen masses which were cross-linked with chemical agents, radiation or other means to improve mechanical properties, decrease immunogenicity and/or increase resistance to resorption. The-collagen utilized was in a variety of forms, including cross-linked and non-cross-linked fibrillar collagens, gelatins, and the like and sometimes was combined with various other components, such as lubricants, osteogenic factors and the like, depending on use. A major disadvantage of solid cross-linked collagen implants is the requirement for surgical implantation by means of incision. In addition, lack of deformability and flexibility are other disadvantages of solid collagen implants.
  • Schechter, et al., Br. J. Plas. Surg. (1975) 28:198-202 disclose glutaraldehyde cross-linked skin that was soaked in L-alanine after cross-linking. The article postulates that the exposure of the skin to L-alanine blocked residual reactive groups of the aldehyde, thereby preventing the release of toxic molecules generated by such groups.
  • U.S. Pat. No. 3,949,073 describes the use of atelopeptide solutions of bovine collagen as an injectable implant material for augmenting soft tissue.
  • the bovine collagen is reconstituted before implantation and forms a fibrous mass of tissue when implanted.
  • the patent suggests adding particles of insoluble bovine collagen microfibrils to control the shrinkage of the fibrous mass formed at the augmentation site.
  • the commercial embodiment of the material described in the patent is composed of reconstituted atelopeptide bovine collagen in saline that contains a small amount of local anesthetic.
  • the implant shrinks in volume after implantation due primarily to absorption of its fluid component by the body.
  • volume consistency is essential, an additional injection or injections of supplemental implant material is required.
  • This specific composition has many serious drawbacks, e.g., the collagen is from a bovine source, not human, and the preparation process is not only lengthy and expensive but also requires the addition of microfibrils.
  • U.S. Pat. No. 4,424,208 describes an injectable dispersion of cross-linked atelopeptide bovine collagen and reconstituted atelopeptide bovine collagen fibers in an aqueous carrier which exhibited improved volume consistency over the material of U.S. Pat. No. 3,949,073.
  • U.S. Pat. No. 4,582,640 discloses an improved injectable implant over U.S. Pat. Nos. 3,949,073 and 4,424,208 in which the improvement consists of improved volume consistency and resistance to physical deformation, improved injectability as compared to the dispersion of U.S. Pat. No. 4,424,208 and that the bovine collagen contains only a single physical form of collagen as compared to the two physical forms found in U.S. Pat. No. 4,424,208.
  • U.S. Pat. No. 4,803,075 describes bovine collagen compositions including a lubricant material to enhance injectability through narrow diameter needles for soft tissue repair.
  • the present invention relates to injectable materials for soft tissue augmentation and methods for use and manufacture of the same, which overcome the shortcomings of bovine injectable collagen and other injectable materials, including silicone, of the prior art.
  • the injectable materials used in accordance with the present invention comprise naturally secreted extracellular matrix preparations as well as preparations derived from naturally secreted extracellular matrix. These preparations are biocompatible, biodegradable and are capable of promoting connective tissue deposition, angiogenesis, reepithilialization and fibroplasia, which is useful in the repair of skin and other tissue defects. These extracellular matrix preparations may be used to repair tissue defects by injection at the site of the defect.
  • the injectable preparations of the present invention have many advantages over conventional injectable collagen preparations used for the repair of skin defects.
  • the extracellular matrix preparations of the present invention contain only human proteins, therefore, there is a reduced risk of an immune response due to foreign proteins or peptides, especially the type of immune response seen with bovine collagen found in conventional injectable collagen preparations.
  • the injected preparations of the present invention should persist longer and even if multiple injections are required, the injections should not be subject to the “no more than three injections per year” rule of bovine collagen-based preparations due to the lack of immunogenicity.
  • Another advantage provided by the present invention is that the preparations of native extracellular matrix contain a mixture of extracellular matrix proteins which closely mimics the compositions of physiologically normal conditions, for example, in an extracellular matrix derived from dermal cells, type I and III collagens, hyaluronic acid as well as various glycosaminoglycans and natural growth factors are present. Many of these extracellular matrix proteins and growth factors have been studied extensively and have been shown to be critical for wound healing and tissue restoration.
  • the preparations can be used in highly improved systems for in vitro tissue culture.
  • naturally secreted extracellular matrix coated three-dimensional frameworks can be used to culture cells which require attachment to a support in order to grow but do not attach to conventional tissue culture vessels.
  • the extracellular matrix secreted by the cells onto the framework can be collected and used to coat vessels for use in tissue culture.
  • the extracellular matrix, acting as a base substrate, may allow cells normally unable to attach to conventional tissue culture dish base substrates to attach and subsequently grow.
  • Yet another embodiment of the present invention is directed to a novel method for determining the ability for cellular taxis of a particular cell.
  • the method involves inoculating one end of a native extracellular matrix coated three-dimensional framework with the cell type in question and over time measure the distance traversed across the framework by the cell. Because the extracellular matrix is secreted naturally by the cells onto the framework, it is an excellent in vitro equivalent of extracellular matrix found in the body.
  • Such an assay may inform whether isolated tumor cells are metastatic or whether certain immune cells can migrate across or even chemotact across the framework, thus, indicating that the cell has such cellular taxis ability.
  • Adherent Layer [0019] Adherent Layer
  • an aqueous medium at physiological isotonicity and pH may contain other elements such as local anesthetics and/or fluid lubricants.
  • fibroblasts with or without other cells and/or elements found in loose connective tissue including but not limited to, endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, chondrocytes, etc.
  • a three dimensional support composed of any material and/or shape that (a) allows cells to attach to it (or can be modified to allow cells to attach to it); and (b) allows cells to grow in more than one layer.
  • This support is inoculated with stromal cells to form the living stromal matrix.
  • stromal cells [0028] a three dimensional framework which has been inoculated with stromal cells. Whether confluent or subconfluent, stromal cells according to the invention continue to grow and divide.
  • the living stromal tissue prepared in vitro is the source of the extracellular matrix proteins used in the injectable formulations of the invention.
  • FIG. 1 is a scanning electron micrograph depicting fibroblast attachment to the three-dimensional matrix and extension of cellular processes across the mesh opening. Fibroblasts are actively secreting matrix proteins and are at the appropriate stage of subconfluency which should be obtained prior to inoculation with tissue-specific cells.
  • FIGS. 2 A-D are transmission electron micrographs of collagen isolated from extracellular matrix prepared from dermal tissue grown in vitro (FIG. 2A-B) or from a normal adult human dermal sample (FIG. 2C-D).
  • One embodiment of the present invention involves the preparation and use of an injectable extracellular matrix composition for the treatment of skin defects.
  • the extracellular matrix proteins are derived from a living stromal tissue prepared in vitro by growing stromal cells on a three-dimensional framework resulting in a multi-layer cell culture system. In conventional tissue culture systems, the cells were grown in a monolayer. Cells grown on a three-dimensional framework support, in accordance with the present invention, grow in multiple layers, forming a cellular matrix. This matrix system approaches physiologic conditions found in vivo to a greater degree than previously described monolayer tissue culture systems.
  • the three-dimensional cell culture system is applicable to the proliferation of different types of stromal cells and formation of a number of different stromal tissues, including but not limited to dermis, bone marrow stroma, glial tissue, cartilage, to name but a few.
  • the pre-established living stromal tissue comprises stromal cells grown on a three-dimensional framework or network.
  • the stromal cells can comprise fibroblasts with or without additional cells and/or elements described more fully herein.
  • the fibroblasts and other cells and/or elements that comprise the stroma can be fetal or adult in origin, and can be derived from convenient sources such as skin, liver, pancreas, etc.
  • Such tissues and/or organs can be obtained by appropriate biopsy or upon autopsy.
  • cadaver organs may be used to provide a generous supply of stromal cells and elements.
  • the stromal cells will proliferate on the framework, and elaborate growth factors, regulatory factors and extracellular matrix proteins that are deposited on the support.
  • the living stromal tissue will sustain active proliferation of the culture for long periods of time. Growth and regulatory factors can be added to the culture, but are not necessary since they are elaborated by the stromal support matrix.
  • the naturally secreted extracellular matrix is collected from the three-dimensional framework and is processed further with a pharmaceutically acceptable aqueous carrier and placed in a syringe for precise placement of the biomaterial into tissues, such as the facial dermis.
  • the present invention is based, in part, on the discovery that during the growth of human stromal cells on a biodegradable or non-biodegradable three-dimensional support framework, the cells synthesize and deposit on the three-dimensional support framework a human extracellular matrix as produced in normal human tissue.
  • the extracellular matrix is secreted locally by cells and not only binds cells and tissues together but also influences the development and behavior of the cells it contacts.
  • the extracellular matrix contains various fiber-forming proteins interwoven in a hydrated gel composed of a network of glycosaminoglycan chains.
  • the glycosaminoglyeans are a heterogeneous group of long, negatively charged polysaccharide chains, which (except for hyaluronic acid) are covalently linked to protein to form proteoglycan molecules.
  • the fiber-forming proteins are of two functional types: mainly structural (collagens and elastin) and mainly adhesive (such as fibronectin and laminin).
  • the fibrillar collagens (types I, II, and III) are rope-like, triple-stranded helical molecules that aggregate into long cable-like fibrils in the extracellular space; these in turn can assemble into a variety of highly ordered arrays.
  • Type IV collagen molecules assemble into a sheetlike meshwork that forms the core of all basal laminae.
  • Elastin molecules form an extensive cross-linked network of fibers and sheets that can stretch and recoil, imparting elasticity to the matrix.
  • Fibronectin and laminin are examples of large adhesive glycoproteins in the matrix; fibronectin is widely distributed in connective tissues, whereas laminin is found mainly in basal laminae. By means of their multiple binding domains, such proteins help cells adhere to and become organized by the extracellular matrix.
  • a naturally secreted human dermal extracellular matrix contains type I and type III collagens, fibronectin, tenascin, glycosaminoglycans, acidic and basic FGF, TGF- ⁇ and TGF- ⁇ , KGF, decorin and various other secreted human dermal matrix proteins.
  • the various extracellular matrix proteins are produced in the quantities and ratios similar to that existing in vivo.
  • growth of the stromal cells in three dimensions will sustain active proliferation of cells in culture for much longer time periods than will monolayer systems.
  • the three-dimensional system supports the maturation, differentiation, and segregation of cells in culture in vitro to form components of adult tissues analogous to counterparts found in vivo.
  • the extracellular matrix created by the cells in culture is more analogous to native tissues.
  • the three-dimensional framework provides a greater surface area for protein attachment, and consequently, for the adherence of stromal cells.
  • stromal cells continue to actively grow in contrast to cells in monolayer cultures, which grow to confluence, exhibit contact inhibition, and cease to grow and divide.
  • the elaboration of growth and regulatory factors by replicating stromal cells may be partially responsible for stimulating proliferation and regulating differentiation of cells in culture.
  • the increase in potential volume for cell growth in the three-dimensional system may allow the establishment of localized microenvironments analogous to native counterparts found in vivo.
  • the three-dimensional matrix maximizes cell-cell interactions by allowing greater potential for movement of migratory cells, such as macrophages, monocytes and possibly lymphocytes in the adherent layer.
  • the three-dimensional stromal support, the culture system itself, and its maintenance, as well as various uses of the three-dimensional cultures and of the naturally secreted extracellular matrix are described in greater detail in the subsections below. Solely for ease of explanation, the detailed description of the invention is divided into the three sections, (i) growth of the three-dimensional stromal cell culture, (ii) isolation of the naturally secreted human extracellular matrix, and (iii) formulation of the isolated extracellular matrix into preparations for injection at the site of soft tissue defects.
  • the three-dimensional support used to culture stromal tissue may be of any material and/or shape that:
  • non-biodegradable materials include but are not limited to: nylon (polyamides), dacron (polyesters), polystyrene, polypropylene, polyacrylates, polyvinyl compounds (e.g., polyvinylchloride), polycarbonate (PVC), polytetrafluorethylene (PTFE; teflon), thermanox (TPX), etc.
  • biodegradable material may also be utilized, including but not limited to: nitrocellulose, cotton, polyglycolic acid (PGA), cat gut sutures, cellulose, gelatin, dextran, collagen, chitosan, hyaluronic acid, etc. Any of these materials, bio- or non-biodegradable, can be woven into a mesh to form a three-dimensional framework. Alternatively, the materials can be used to form other types of three-dimensional frameworks, for example, sponges, such as collagen sponges.
  • nylon polystyrene, etc.
  • nylon frameworks can be treated with 0.1 M acetic acid, and incubated in polylysine, FBS, and/or collagen to coat the nylon.
  • Polystyrene can be similarly treated using sulfuric acid.
  • a convenient nylon mesh which can be used in accordance with the invention is Nitex, a nylon filtration mesh having an average pore size of 210 ⁇ m and an average nylon fiber diameter of 90 ⁇ m (#3-210/36, Tetko, Inc., N.Y.).
  • fibroblasts derived from adult or fetal tissue, with or without other cells and elements described below, are inoculated onto the framework.
  • fibroblasts may be derived from organs, such as skin, liver, pancreas, etc. which can be obtained by biopsy, where appropriate, or upon autopsy.
  • fibroblasts can be obtained in quantity rather conveniently from any appropriate cadaver organ.
  • fetal fibroblasts can be obtained in high quantity from foreskin.
  • Fibroblasts may be readily isolated by disaggregating an appropriate organ or tissue which is to serve as the source of the fibroblasts. This can be readily accomplished using techniques known to those skilled in the art.
  • the tissue or organ can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that-weaken the connections between neighboring cells making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage.
  • Enzymatic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination.
  • the suspension can be fractionated into subpopulations from which the fibroblasts and/or other stromal cells and/or elements can be obtained. This also may be accomplished using standard techniques for cell separation including, but not limited to, cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection), separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter-streaming centrifugation), unit gravity separation, countercurrent distribution, electrophoresis and fluorescence-activated cell sorting.
  • standard techniques for cell separation including, but not limited to, cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection), separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elu
  • fibroblasts for example, can be carried out as follows: fresh tissue samples are thoroughly washed and minced in Hanks' balanced salt solution (HBSS) in order to remove serum. The minced tissue is incubated from 1-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the dissociated cells are suspended, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown. The isolated fibroblasts can then be grown to confluency, lifted from the confluent culture and inoculated onto the three-dimensional framework, see Naughton et al., 1987, J. Med.
  • HBSS Hanks' balanced salt solution
  • stromal cell culture-producing extracellular matrix can be added to form the three-dimensional stromal cell culture-producing extracellular matrix.
  • other cells found in loose connective tissue may be inoculated onto the three-dimensional support framework along with fibroblasts.
  • Such cells include, but are not limited to, endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, chondrocytes, etc.
  • These stromal cells can be readily derived from appropriate organs such as skin, liver, etc., using methods known, such as those discussed above.
  • stromal cells which are specialized for the particular tissue to be cultured can be added to the fibroblast stroma for the production of a tissue type specific extracellular matrix.
  • dermal fibroblasts can be used to form the three-dimensional subconfluent stroma for the production of skin-specific extracellular matrix in vitro.
  • stromal cells of hematopoietic tissue including, but not limited to, fibroblast endothelial cells, macrophages/monocytes, adipocytes and reticular cells, can be used to form the three-dimensional subconfluent stroma for the production of a bone marrow-specific extracellular matrix in vitro, see infra.
  • Hematopoietic stromal cells can be readily obtained from the “buffy coat” formed in bone marrow suspensions by centrifugation at low forces, e.g., 3000 ⁇ g.
  • Stromal cells of liver may include fibroblasts, Kupffer cells, and vascular and bile duct endothelial cells.
  • glial cells can be used as the stroma to support the proliferation of neurological cells and tissues. Glial cells for this purpose can be obtained by trypsinization or collagenase digestion of embryonic or adult brain. Ponten and Westermark, 1980, In Federof, S. Hertz, L., eds, “Advances in Cellular Neurobiology,” Vol.1, New York, Academic Press, pp.209-227.
  • the stromal cells For certain uses in vivo it is preferable to obtain the stromal cells from the patient's own tissues.
  • the growth of cells in the presence of the three-dimensional stromal support framework can be further enhanced by adding to the framework, or coating the framework support with proteins, e.g., collagens, elastic fibers, reticular fibers, glycoproteins; glycosaminoglycans, e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratan sulfate, etc.; a cellular matrix, and/or other materials.
  • proteins e.g., collagens, elastic fibers, reticular fibers, glycoproteins
  • glycosaminoglycans e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulf
  • the three-dimensional framework is incubated in an appropriate nutrient medium under physiologic conditions favorable for cell growth, i.e., promoting mitosis, i.e., cell division.
  • an appropriate nutrient medium such as RPMI 1640, Fisher's, Iscove's, McCoy's, and the like may be suitable for use. It is important that the three-dimensional stromal culture be suspended or floated in the medium during the incubation period in order to maximize proliferative activity. In addition, the culture should be “fed” periodically to remove the spent media, depopulate released cells, and to add fresh media.
  • the stromal cells will grow linearly along and envelop the three-dimensional framework before beginning to grow into the openings of the framework.
  • the cells are grown to an appropriate degree to allow for adequate deposition of extracellular matrix proteins.
  • the openings of the framework should be of an appropriate size to allow the stromal cells to stretch across the openings. Maintaining actively growing stromal cells which stretch across the framework enhances the production of growth factors which are elaborated by the stromal cells, and hence, will support long term cultures. For example, if the openings are too small, the stromal cells may rapidly achieve confluence but be unable to easily exit from the mesh. Trapped cells can exhibit contact inhibition and cease production of the appropriate factors necessary to support proliferation and maintain long term cultures. If the openings are too large, the stromal cells are unable to stretch across the opening. This will also decrease stromal cell production of the appropriate factors necessary to support proliferation and maintain long term cultures.
  • openings ranging from about 150 ⁇ m to about 220 ⁇ m will work satisfactorily.
  • other sizes may work equally well.
  • any shape or structure that allows the stromal cells to stretch and continue to replicate and grow for lengthy time periods will work in accordance with the present invention.
  • Different proportions of the various types of collagen deposited on the framework can be achieved by inoculating the framework with different tissue-specific cells.
  • the matrix should preferably contain collagen types III, IV and I in an approximate ratio of 6:3:1 in the initial matrix.
  • collagen types I and III are preferably deposited in the initial matrix.
  • the proportions of collagen types deposited can be manipulated or enhanced by selecting fibroblasts which elaborate the appropriate extracellular matrix proteins. This can be accomplished using monoclonal antibodies of an appropriate isotype or subclass which are capable of activating complement, and which define particular collagen types. These antibodies in combination with complement can be used to negatively select the fibroblasts which express the desired collagen type.
  • the stroma used to inoculate the framework can be a mixture of cells which synthesize the appropriate collagen types desired.
  • the distribution and origins of the five types of collagen is shown in Table I. TABLE I DISTRIBUTIONS AND ORIGINS OF THE FIVE TYPES OF COLLAGEN Collagen Principal Tissue Type Distribution Cells of Origin I Loose and dense ordinary Fibroblasts and connective tissue; collagen reticular cells; fibers smooth muscle cells Fibrocartilage Bone Osteoblast Dentin Odontoblasts II Hyaline and elastic Chondrocytes cartilage Retinal Cells Vitreous body of eye III Loose connective tissue; Fibroblasts and reticular fibers reticular cells Papillary layer of dermis Smooth muscle cells; Blood vessels endothelial cells IV Basement membranes Epithelial and endothelial cells Lens capsule of eye Lens fibers V Fetal membranes; placenta Fibroblasts Basement membranes Bone Smooth muscle Smooth muscle cells
  • the appropriate stromal cell(s) can be selected to inoculate the three-dimensional framework.
  • the three-dimensional extracellular matrix producing culture of the present invention affords a vehicle for introducing gene products in vivo.
  • the cells may be genetically engineered to express the gene product, or altered forms of the gene product that are immobilized in the extracellular matrix laid down by the stromal cells.
  • a gene of interest can be placed under the control of an inducible promoter.
  • the recombinant DNA construct containing the gene can be used to transform or transfect a host cell which is cloned and then clonally expanded in the three-dimensional culture system.
  • the use of the-three-dimensional culture in this regard has a number of advantages.
  • the number of transfected cells can be substantially enhanced to be of clinical value, relevance, and utility.
  • the three-dimensional cultures of the present invention allow for expansion of the number of transfected cells and amplification (via cell division) of transfected cells.
  • the expression control elements used should allow for the regulated expression of the gene so that the product can be over synthesized in culture.
  • the transcriptional promoter chosen, generally, and promoter elements specifically, depends, in part, upon the type of tissue and cells cultured. Cells and tissues which are capable of secreting proteins (e.g., those characterized by abundant rough endoplasmic reticulum and golgi complex) are preferable.
  • proliferating cells are released from the framework. These released cells can stick to the walls of the culture vessel where they can continue to proliferate and form a confluent monolayer. This should be prevented or minimized, for example, by removal of the released cells during feeding, or by transferring the three-dimensional framework to a new culture vessel. The presence of a confluent monolayer in the vessel will “shut down” the growth of cells in the three-dimensional framework and/or culture. Removal of the confluent monolayer or transfer of the stromal culture to fresh media in a new vessel will restore proliferative activity of the three-dimensional culture system. Such removal or transfers should be done in any culture vessel which has a stromal monolayer exceeding 25% confluency.
  • the culture system can be agitated to prevent the released cells from sticking, or instead of periodically feeding the cultures, the culture system could be set up so that fresh media continuously flows through the system.
  • the flow rate can be adjusted to both maximize proliferation within the three-dimensional culture, and to wash out and remove cells released from the matrix, so that they will not stick to the walls of the vessel and grow to confluence.
  • the released stromal cells can be collected and crypreserved for future use.
  • fibroblasts Once inoculated onto the three-dimensional framework, adherence of the fibroblasts is seen quickly (e.g., within hours) and the fibroblasts begin to stretch across the framework openings within days. These fibroblasts are metabolically active, secrete extracellular matrix and rapidly form a dermal equivalent consisting of active fibroblasts and collagen.
  • FIG. 1 illustrates the ability of the fibroblasts to arrange themselves into parallel layers between the naturally-secreted collagen bundles. These fibroblasts exhibit a rapid rate of cell division and protein secretion.
  • the cells can be killed by flash-freezing the living stromal tissue prepared in vitro in liquid nitrogen without a cryopreservative.
  • Another way to kill the cells is to irrigate the inoculated three-dimensional framework with sterile water, such that the cells burst in response to osmotic pressure.
  • the framework can be subjected to enzymatic digestion and/or extracting with reagents that break down the cellular membranes and allow removal of cell contents.
  • detergents include non-ionic detergents (for example, TRITON X-100, octylphenoxy polyethoxyethanol, (Rohm and Haas); BRIJ-35, a polyethoxyethanol lauryl ether (Atlas Chemical Co.), TWEEN 20, a polyethoxyethanol sorbitan monolaureate (Rohm and Haas), LUBROL-PX, or polyethylene lauryl ether (Rohm and Haas)); and ionic detergents (for example, sodium dodecyl sulphate, sulfated higher aliphatic alcohol, sulfonated alkane and sulfonated alkylarene containing 7 to 22 carbon atoms in a branched or unbranched chain).
  • non-ionic detergents for example, TRITON X-100, octylphenoxy polyethoxyethanol, (Rohm and Haas); BRIJ-35, a
  • Enzymes can be used also and can include nucleases (for example, deoxyribonuclease and ribonuclease), phospholipases and lipases.
  • nucleases for example, deoxyribonuclease and ribonuclease
  • phospholipases for example, phospholipases and lipases.
  • the collection of the naturally secreted human extracellular matrix can be accomplished in a variety of ways which depends on whether the three-dimensional framework is composed of material that is biodegradable or non-biodegradable. For example, if the framework is composed of non-biodegradable material, one can remove the extracellular matrix from a non-biodegradable support by subjecting the three-dimensional framework to sonication and/or to high pressure water jets and/or to mechanical scraping and/or to a mild treatment with detergents and/or enzymes to remove the attached extracellular matrix from the framework.
  • the extracellular matrix is deposited on a biodegradable three-dimensional framework, after killing and removing the cells and cellular debris, the extracellular matrix can be recovered, for example, by simply allowing the framework to degrade in solution, i.e., allow the framework to dissolve, thus freeing the extracellular matrix.
  • the biodegradable support is composed of a material which can be injected, like the extracellular matrix itself, one can process the entire extracellular matrix coated framework into syringes for injection.
  • the matrix can be removed by the same methods as if the matrix had been deposited on a non-biodegradable support, i.e., by subjecting the three-dimensional framework to sonication and/or to high pressure water jets and/or to mechanical scraping and/or to a mild treatment with detergents and/or enzymes to remove the attached extracellular matrix from the framework. None of the removal processes are designed to damage and/or denature the naturally secreted human extracellular matrix produced by the cells.
  • the extracellular matrix can be homogenized to fine particles, such that it can pass through a surgical needle. Homogenization is well known in the art, for example, by sonication. Further, the extracellular matrix can be cross-linked by gamma irradiation without the use of chemical cross-linking agents, such as glutaraldehyde, which are toxic. The gamma irradiation should be a minimum of 20 M rads to sterilize the material since all bacteria, fungi, and viruses are destroyed at 0.2 M rads. Preferably, the extracellular matrix can be irradiated from 0.25 to 2 M rads to sterilize and cross-link the extracellular matrix.
  • the amounts and/or ratios of the collagens and other proteins may be adjusted by mixing extracellular matrices secreted by other cell types prior to placing the material in a syringe.
  • biologically active substances such as proteins and drugs
  • exemplary biologically active substances can include tissue growth factors, such as TGF- ⁇ , and the like which promote healing and tissue repair at the site of the injection.
  • Final formulation of the aqueous suspension of naturally secreted human extracellular matrix will typically involve adjusting the ionic strength of the suspension to isotonicity (i.e., about 0.1 to 0.2) and to physiological pH (i.e., about pH 6.8 to 7.5) and adding a local anesthetic, such as lidocaine, (usually at a concentration of about 0.3% by weight) to reduce local pain upon injection.
  • the final formulation will also typically contain a fluid lubricant, such as maltose, which must be tolerated by the body.
  • Exemplary lubricant components include glycerol, glycogen, maltose and the like organic polymer base materials, such as polyethylene glycol and hyaluronic acid as well as non-fibrillar collagen, preferably succinylated collagen, can also act as lubricants.
  • Such lubricants are generally used to improve the injectability, intrudability and dispersion of the injected biomaterial at the site of injection and to decrease the amount of spiking by modifying the viscosity of the compositions.
  • This final formulation is by definition the processed extracellular matrix in a pharmaceutically acceptable carrier.
  • the matrix is subsequently placed in a syringe or other injection apparatus for precise placement of the matrix at the site of the tissue defect.
  • injectable means the formulation can be dispensed from syringes having a gauge as low as 25 under normal conditions under normal pressure without substantial spiking. Spiking can cause the composition to ooze from the syringe rather than be injected into the tissue.
  • needles as fine as 27 gauge (200 ⁇ I.D.) or even 30 gauge (150 ⁇ I.D.) are desirable.
  • the maximum particle size that can be extruded through such needles will be a complex function of at least the following: particle maximum dimension, particle aspect ratio (length:width), particle rigidity, surface roughness of particles and related factors affecting particle:particle adhesion, the viscoelastic properties of the suspending fluid, and the rate of flow through the needle.
  • particle maximum dimension particle aspect ratio (length:width)
  • particle rigidity particle rigidity
  • surface roughness of particles and related factors affecting particle:particle adhesion the viscoelastic properties of the suspending fluid
  • the rate of flow through the needle Rigid spherical beads suspended in a Newtonian fluid represent the simplest case, while fibrous or branched particles in a viscoelastic fluid are likely to be more complex.
  • the above described steps in the process for preparing injectable naturally secreted human extracellular matrix are preferably carried out under sterile conditions using sterile materials.
  • the processed extracellular matrix in a pharmaceutically acceptable carrier can be injected intradermally or subcutaneously to augment soft tissue, to repair or correct congenital anomalies, acquired defects or cosmetic defects.
  • congenital anomalies as hemifacial microsomia, malar and zygomatic hypoplasia, unilateral mammary hypoplasia, pectus excavatum, pectoralis agenesis (Poland's anomaly) and velopharyngeal incompetence secondary to cleft palate repair or submucous cleft palate (as a retropharyngeal implant); acquired defects (post-traumatic, post-surgical, post-infectious) such as depressed scars, subcutaneous atrophy (e.g., secondary to discoid lupis erythematosus), keratotic lesions, enophthalmos in the unucleated eye (also superior sulcus syndrome), acne pitting of the face, linear scleroderma with subcutaneous atrophy, saddle-nose deformity, Romberg's disease and unilateral vocal cord paralysis; and cosmetic defects such as glabellar frown lines, deep nasolabial creases, circum-oral geographical
  • the three-dimensional culture system of the invention is described based upon the type of tissue and cells used in various systems. These descriptions specifically include but are not limited to bone marrow, skin, epithelial cells, and cartilage but it is expressly understood that the three-dimensional culture system can be used with other types of cells and tissues.
  • the invention is also illustrated by way of examples, which demonstrate characteristic data generated for each system described.
  • Skin fibroblasts were isolated by mincing dermal tissue, trypsinization for 2 hours, and separation of cells into a suspension by physical means. Fibroblasts were grown to confluency in 25 cm 2 Falcon tissue culture dishes and fed with RPMI 1640 (Sigma, MO) supplemented with 10% fetal bovine serum (FBS), fungizone, gentamicin, and penicillin/streptomycin. Fibroblasts were lifted by mild trypsinization and cells were plated onto nylon filtration mesh, the fibers of which are approximately 90 ⁇ m in diameter and are assembled into a square weave with a mesh opening of 210 ⁇ m (Tetko, Inc., NY).
  • FIG. 1 is a scanning electron micrograph depicting fibroblast attachment and extension of cellular processes across the mesh opening.
  • Bone marrow was aspirated from multiple sites on the posterior iliac crest of hematologically normal adult volunteers after informed consent was obtained. Specimens were collected into heparinized tubes and suspended in 8 ml of RPMI 1640 medium which was conditioned with 10% FBS and 5-10% HS and supplemented with hydrocortisone, fungizone, and streptomycin. The cell clumps were disaggregated and divided into aliquots of 5 ⁇ 10 6 nucleated cells.
  • Nylon filtration screen (#3-210/36, Tetko Inc., NY) was used as a three-dimensional framework to support all stromal cell cultures described in the examples below.
  • the screen consisted of fibers, which were 90 ⁇ m in diameter, assembled into a square weave pattern with sieve openings of 210 ⁇ m.
  • Stromal cells were inoculated using the protocols described in Section 6.1. Adherence and subsequent growth of the stromal elements was monitored using inverted phase contrast microscopy and scanning electron microscopy (SEM).
  • Tissues were removed and placed in fresh trypsin solution, and gently agitated until cell appeared to form a single-cell suspension.
  • the single-cell suspension was then diluted in MEM containing 10% heat inactivated fetal bovine serum and centrifuged at 1400 ⁇ g for 7 minutes. The supernatant was decanted and the pellet containing mucosal epithelial cells was placed into seeding medium.
  • Medium consisted of DMEM with 2% Ultrosen G, 1 ⁇ L-glutamine, 1 ⁇ non-essential amino acids, penicillin and streptomycin. The cells were seeded onto a three-dimensional framework.
  • the three-dimensional stromal culture was generated using oral fibroblasts and 8 mm ⁇ 45 mm pieces of nylon filtration screen (#3-210/36, Tetko Inc., NY). The mesh was soaked in 0.1 M acetic acid for 30 minutes and treated with 10 mM polylysine suspension for 1 hour. The meshes were placed in a sterile petri dish and inoculated with 1 ⁇ 10 6 oral fibroblasts collected as described above in DMEM complete medium. After 1-2 hours of incubation at 5% CO 2 the meshes were placed in a Corning 25 cm 2 tissue culture flask, floated with an additional 5 ml of medium, and allowed to reach subconfluence, being fed at 3 day intervals. Cultures were maintained in DMEM complete medium at 37° C. and 5% CO 2 in a humidified atmosphere and were fed with fresh medium every 3 days.
  • Small vessel endothelial cells isolated from the brain according to the method of Larson et al., 1987, Microvasc. Res. 34:184 were cultured in vitro using T-75 tissue culture flasks.
  • the cells were maintained in Dulbecco's Modified Eagle Medium/Hams-F-12 medium combination (the solution is available as a 1:1 mixture).
  • the medium was supplemented with 20% heat-inactivated fetal calf serum (FCS), glutamine, and antibiotics.
  • FCS heat-inactivated fetal calf serum
  • the cells were seeded at a concentration of 1 ⁇ 10 6 cells per flask, and reached a confluent state within one week.
  • the cells were passaged once a week, and, in addition, were fed once a week with DMEM/Hams-F-12 containing FCS, glutamine, and antibiotics as described.
  • flasks were rinsed twice with 5 ml of PBS (without Ca ++ or Mg ++ ) and trypsinized with 3 ml of 0.05% Trypsin and 0.53 mM EDTA.
  • the cells were pelleted, resuspended, and tested for viability by trypan blue exclusion, seeded and fed with 25 ml of the above mentioned DMEM/Hams-F-12 supplemented medium.
  • a factor VIII related antigen assay Grulnick et al., 1977, Ann. Int. Med. 86:598-616, is used to positively identify endothelial cells, and silver staining was used to identify tight junctional complexes, specific to only small vessel endothelium.
  • Nylon filtration screen mesh (#3-210/36, Tetko, Inc., NY) was prepared essentially as described above. The mesh was soaked in an acetic acid solution (1 ml glacial acetic acid plus 99 ml distilled H 2 O) for thirty minutes, was rinsed with copious amounts of distilled water and then autoclaved. Meshes were coated with 6 ml fetal bovine serum per 8 ⁇ 8 cm mesh and incubated overnight. The meshes were then stacked, three high, and 3 ⁇ 10 7 small vessel endothelial cells (cultured as described) were seeded onto the stack, and incubated for three hours at 37° C. under 5% CO 2 in a humidified atmosphere. The inoculated meshes were fed with 10 ml of DME/Hams-F-12 medium every 3-4 days until complete confluence was reached (in approximately two weeks).
  • Cartilage was harvested from articular surfaces of human joints.
  • the cartilage pieces were digested with collagenase (0.2% w/v) in complete medium (DMEM with 10% fetal bovine serum, glutamine, non-essential amino acids, sodium pyruvate, 50 ⁇ g/ml ascorbate and 35 ⁇ g/ml gentamicin) for 20 hours at 37° C.
  • DMEM complete medium
  • Liberated chondrocytes were spun, resuspended in complete medium, counted and plated at 1 ⁇ 10 6 cells per T-150 flask. Cells were routinely passed at confluence (every 5-7 days).
  • Polyglycolic acid mesh (1 mm diameter ⁇ 2 mm thick) was sterilized by ethylene oxide or electron beam treatment and presoaked overnight in complete medium. The mesh was seeded in 6 well plates with 3-4 ⁇ 10 6 cells per mesh in a total volume of 10 ⁇ l and incubated for 3-4 hours at 37° C. in a tissue culture incubator. At this time, 1.5 ml of media was added. The seeded mesh was incubated overnight. 5 ml of media was added the next day. Media was changed three times per week until confluence is reached.
  • the extracellular matrix has been characterized by a number of analytic methods to determine its content of matrix proteins, each value is the average of at least two independent determinations.
  • the matrix contained type I and type III collagens, fibronectin, tenascin, sulfated glycosaminoglycans, decorin and various other secreted human extracellular matrix proteins. Additionally, the secreted matrix proteins were found throughout the three-dimensional support framework.
  • the extracellular matrix contained a total protein amount of 292 mg/cm 2 ⁇ 0.06; fibronectin was present at 3.4 mg/cm 2 ⁇ 1.2; and tenascin at 1.7 mg/cm 2 ⁇ 0.6. Both fibronectin and tenascin showed the expected molecular weight distributions on immunoblots.
  • Collagen derived from the dermal tissue grown in vitro and collagen derived from a normal adult human dermal sample were processed and visualized by transmission electron microscopy (TEM). Briefly, the respective collagens were weighed and placed in a sterile 50 ml centrifuge tube with 30 ml 0.05 M Tris buffer, pH 8.0. After mixing for two hours on a wrist shaker, the Tris buffer was removed and the specimen placed in a homogenization cylinder along with 30 ml fresh 0.05 M Tris buffer. The sample was homogenized for 30 seconds in buffer alone and then for two 30 second bursts following the addition of a dispersing agent as described in U.S. Pat. Nos. 4,969,912 and 5,332,802.
  • the temperature was maintained at 5-10° C. during the mechanical disruption process.
  • the dispersing agent was added at a concentration of 0.05% (wet weight of the collagen).
  • the homogenized preparation was centrifuged at 3500 rpm for 6 minutes to separate the dispersed collagenous material from the yet undispersed material.
  • the undispersed residue was again treated with dispersing agent at 0.05% (wet weight of the collagen) and homogenized for two 30 second bursts. The dispersion was again centrifuged to recover dispersed collagenous material which was added to the first recovery.
  • the collagenous dispersion was filtered through a 100 micron filter, centrifuged at 3500 rpm and the pellet was washed 3 times with 0.004 M phosphate buffer, pH 7.4. The last centrifugation was conducted at 10,000 rpm to pack the collagenous pellet. Samples were then collected for TEM. As shown in FIGS. 2 A-D, the collagen fibers isolated from either the extracellular matrix prepared from dermal tissue grown in vitro (FIGS. 2 A-B) or from normal adult human dermis (FIGS. 2 C-D) appeared identical in that intact collagen fibers with typical collagen banding and normal periodicity in both preparations.
  • Glycosaminoglycans have been shown to play a variety of structural and functional roles in the body and their presence in the secreted extracellular matrix is important. Table II lists a number of examples of glycosaminoglycans which have been determined to be found in the extracellular matrix as well as their functional importance in normal dermis.
  • the extracellular matrix was found to contain a total of 2.8 mg/cm 2 ⁇ 0.1 sulfated glycosaminoglycans.
  • the cells producing and depositing the extracellular matrix also expressed a number of different growth factors. Growth factors are important in the extracellular matrix for two reasons. During the growth of and deposition of the extracellular matrix, naturally seeded growth factors help to control cell proliferation and activity. Further, growth factors remain attached to the extracellular matrix. A variety of growth factors have been determined to be expressed during the deposition of the matrix.
  • acidic and basic FGF, TGF- ⁇ and TGF- ⁇ , and KGF mRNA transcripts were present as were several others as shown in Table III, including PDGF, amphiregulin, HBEGF, IGF, SPARC and VEGF.
  • PDGF and TGF- ⁇ 3 are thought to be involved in regulation of cell proliferation and matrix deposition in culture, while TGF- ⁇ 1, HBEGF, KGF, SPARC, VEGF and decorin are deposited in the matrix.
  • Amphiregulin, IGF-1, IGF-2 and IL-1 were not expressed at the sensitivity used in these experiments.

Abstract

The present invention discloses compositions containing natural human extracellular matrices and methods for the use thereof. More particularly, the present invention provides compositions and methods for the repair of skin defects using natural human extracellular matrix by injection.

Description

  • This application is a continuation-in-part application of U.S. patent application Ser. No. 08/470,101 filed Jun. 6, 1995, which is incorporated by reference herein in its entirety.[0001]
  • 1. INTRODUCTION
  • The present invention relates to compositions and methods for the treatment and repair of soft tissue and skin defects such as wrinkles and scars. More particularly, the invention relates to an injectable composition of human extracellular matrix components and methods of preparing and using same. The injectable preparation is obtained from three-dimensional living stromal tissues that are prepared in vitro. [0002]
  • 2. BACKGROUND OF THE INVENTION
  • The idea of using an injectable material for soft tissue augmentation and repair developed soon after the invention of the hypodermic needle. Various products have been injected into the human body for correction of soft tissue and skin defects including paraffin, petrolatum, vegetable oils, lanolin, bees wax, and silicone. Injectable liquid silicone has been used extensively, however, due to long term side effects, such as nodules, recurring cellulitis and skin ulcers which are now being followed more closely, the use of injectable silicone is on the decline. Further, in the State of Nevada it is a felony to use injectable silicone in a human. Orange, Skin and Allergy News (1992) Vol.23, No.6, pg. 1. More recently, bovine collagen has gained widespread use as an injectable material for soft tissue augmentation. Collagen is the principal extracellular structural protein of the animal body. At least fourteen types of mammalian collagen have been described. The common characteristic amongst them is a three stranded helix, consisting of three polypeptide chains, called alpha-chains. All alpha-chains have the same configuration, but differ in the composition and sequence of their amino acids. Although this leads to different types of alpha-chains, however, they all have glycine at every third position in the amino acid sequence. The glycine at every third position allows for the helical structure of the alpha-chains. Type I collagen is composed of two alpha[0003] 1-chains and one alpha2-chain and is the principal extracellular material of skin, tendon and bone. When clinicians mention “collagen”, they are usually referring to type I collagen. See Table I, infra, for a detailed listing of collagen types I-V and in which tissues they are found.
  • Collagen has been used as an implant material to replace or augment hard or soft connective tissue, such as skin, tendon, cartilage, bone and interstitium. Additionally, collagen implants have been used for cosmetic purposes for a number of years since collagen can help cellular ingrowth at the placement site. Early collagen implants were often solid collagen masses which were cross-linked with chemical agents, radiation or other means to improve mechanical properties, decrease immunogenicity and/or increase resistance to resorption. The-collagen utilized was in a variety of forms, including cross-linked and non-cross-linked fibrillar collagens, gelatins, and the like and sometimes was combined with various other components, such as lubricants, osteogenic factors and the like, depending on use. A major disadvantage of solid cross-linked collagen implants is the requirement for surgical implantation by means of incision. In addition, lack of deformability and flexibility are other disadvantages of solid collagen implants. [0004]
  • Oliver et al., Clinical Orthopaedics & Related Research (1976) 115:291-302; Br. J. Exp. Path. (1980) 61:544-549; and Conn. Tissue Res. (1981) 9:59-62 describe implants made by treating skin with trypsin followed by cross-linking with an aldehyde. The resulting solid collagen implants were reported to maintain their original mass after prolonged implantation. A main problem with such solid implants is that they must be implanted surgically. Other disadvantages are that they are not as deformable as injectable implants and residual glutaraldehyde may cause the implant to lose its flexibility due to continuing cross-linking in situ. [0005]
  • Schechter, et al., Br. J. Plas. Surg. (1975) 28:198-202 disclose glutaraldehyde cross-linked skin that was soaked in L-alanine after cross-linking. The article postulates that the exposure of the skin to L-alanine blocked residual reactive groups of the aldehyde, thereby preventing the release of toxic molecules generated by such groups. [0006]
  • An alternative to surgically implanted solid collagen material is disclosed in U.S. Pat. No. 3,949,073. U.S. Pat. No. 3,949,073 describes the use of atelopeptide solutions of bovine collagen as an injectable implant material for augmenting soft tissue. According to the patent, the bovine collagen is reconstituted before implantation and forms a fibrous mass of tissue when implanted. The patent suggests adding particles of insoluble bovine collagen microfibrils to control the shrinkage of the fibrous mass formed at the augmentation site. The commercial embodiment of the material described in the patent is composed of reconstituted atelopeptide bovine collagen in saline that contains a small amount of local anesthetic. While effective, the implant shrinks in volume after implantation due primarily to absorption of its fluid component by the body. Thus, if volume consistency is essential, an additional injection or injections of supplemental implant material is required. This specific composition has many serious drawbacks, e.g., the collagen is from a bovine source, not human, and the preparation process is not only lengthy and expensive but also requires the addition of microfibrils. [0007]
  • U.S. Pat. No. 4,424,208 describes an injectable dispersion of cross-linked atelopeptide bovine collagen and reconstituted atelopeptide bovine collagen fibers in an aqueous carrier which exhibited improved volume consistency over the material of U.S. Pat. No. 3,949,073. [0008]
  • U.S. Pat. No. 4,582,640 discloses an improved injectable implant over U.S. Pat. Nos. 3,949,073 and 4,424,208 in which the improvement consists of improved volume consistency and resistance to physical deformation, improved injectability as compared to the dispersion of U.S. Pat. No. 4,424,208 and that the bovine collagen contains only a single physical form of collagen as compared to the two physical forms found in U.S. Pat. No. 4,424,208. [0009]
  • U.S. Pat. No. 4,803,075 describes bovine collagen compositions including a lubricant material to enhance injectability through narrow diameter needles for soft tissue repair. [0010]
  • Despite the advantages and overall usefulness of the injectable collagen implant materials disclosed above, problems associated with producing and injecting the materials have been encountered. For example, for soft tissue repair, suspensions of fibrillar collagen have often been used by injecting the composition to a treatment site through a fine gauge needle. The use of fibrillar collagen as the primary matrix material in injectable soft and hard tissue implant compositions has several limitations. The preparation of fibrillar collagen suitable for human use is relatively time consuming and expensive. In particular, the complete removal of contaminating and potentially immunogenic substances to produce atelocollagen is a relatively complex and expensive procedure. Moreover, the persistence, shape retention, cohesiveness, stability, elasticity, toughness-and intrudability of the fibrillar collagen compositions are not optimal. [0011]
  • In addition to the problems associated with producing and injecting the collagen implant materials, problems with the actual use of the above mentioned patented injectable implants are also abundant. For instance, since the above patented injectables derive collagen from xenogeneic sources, usually bovine collagen, the collagen must be modified to reduce its immunogenicity. Even with modified collagen, the implant material is still quite immunogenic to which some people are either already naturally allergic or develop an allergic reaction over time to the bovine collagen. Due to these allergic reactions the injectable collagen implants described above cannot be given to many people and others are limited to receiving only three injections per year. Severe allergic reactions include symptoms of rheumatoid arthritis, while less severe reactions include redness and swelling at the site-of injection which may lead to permanent scarring. Because of these severe side effects, the above described collagen injectables are no longer used for lip augmentation. Further, the problems associated with injecting xenogeneic collagen seem so intractable that rather than injecting collagen, biocompatible ceramic matrices have been injected to achieve similar results as described in U.S. Pat. No. 5,204,382. [0012]
  • In summary, due to the shortcomings of the above-described injectable compositions for the repair of soft tissue defects, such as the lack of persistence, the need for repeated injections and serious concern over adverse reactions, newer injectable materials for soft tissue augmentation are needed. [0013]
  • 3. SUMMARY OF THE INVENTION
  • The present invention relates to injectable materials for soft tissue augmentation and methods for use and manufacture of the same, which overcome the shortcomings of bovine injectable collagen and other injectable materials, including silicone, of the prior art. The injectable materials used in accordance with the present invention comprise naturally secreted extracellular matrix preparations as well as preparations derived from naturally secreted extracellular matrix. These preparations are biocompatible, biodegradable and are capable of promoting connective tissue deposition, angiogenesis, reepithilialization and fibroplasia, which is useful in the repair of skin and other tissue defects. These extracellular matrix preparations may be used to repair tissue defects by injection at the site of the defect. [0014]
  • The injectable preparations of the present invention have many advantages over conventional injectable collagen preparations used for the repair of skin defects. The extracellular matrix preparations of the present invention contain only human proteins, therefore, there is a reduced risk of an immune response due to foreign proteins or peptides, especially the type of immune response seen with bovine collagen found in conventional injectable collagen preparations. Additionally, the injected preparations of the present invention should persist longer and even if multiple injections are required, the injections should not be subject to the “no more than three injections per year” rule of bovine collagen-based preparations due to the lack of immunogenicity. Another advantage provided by the present invention is that the preparations of native extracellular matrix contain a mixture of extracellular matrix proteins which closely mimics the compositions of physiologically normal conditions, for example, in an extracellular matrix derived from dermal cells, type I and III collagens, hyaluronic acid as well as various glycosaminoglycans and natural growth factors are present. Many of these extracellular matrix proteins and growth factors have been studied extensively and have been shown to be critical for wound healing and tissue restoration. [0015]
  • In another aspect of the invention, the preparations can be used in highly improved systems for in vitro tissue culture. In this embodiment, naturally secreted extracellular matrix coated three-dimensional frameworks can be used to culture cells which require attachment to a support in order to grow but do not attach to conventional tissue culture vessels. In addition to culturing cells on a coated framework, the extracellular matrix secreted by the cells onto the framework can be collected and used to coat vessels for use in tissue culture. The extracellular matrix, acting as a base substrate, may allow cells normally unable to attach to conventional tissue culture dish base substrates to attach and subsequently grow. [0016]
  • Yet another embodiment of the present invention is directed to a novel method for determining the ability for cellular taxis of a particular cell. The method involves inoculating one end of a native extracellular matrix coated three-dimensional framework with the cell type in question and over time measure the distance traversed across the framework by the cell. Because the extracellular matrix is secreted naturally by the cells onto the framework, it is an excellent in vitro equivalent of extracellular matrix found in the body. Such an assay, for example, may inform whether isolated tumor cells are metastatic or whether certain immune cells can migrate across or even chemotact across the framework, thus, indicating that the cell has such cellular taxis ability. [0017]
  • 3.1. DEFINITIONS AND ABBREVIATIONS
  • The following terms used herein shall have the meanings indicated: [0018]
  • Adherent Layer: [0019]
  • cells attached directly to the three-dimensional framework or connected indirectly by attachment to cells that are themselves attached directly to the matrix. [0020]
  • Pharmaceutically Acceptable Carrier: [0021]
  • an aqueous medium at physiological isotonicity and pH and may contain other elements such as local anesthetics and/or fluid lubricants. [0022]
  • Stromal Cells: [0023]
  • fibroblasts with or without other cells and/or elements found in loose connective tissue, including but not limited to, endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, chondrocytes, etc. [0024]
  • Three-Dimensional Framework: [0025]
  • a three dimensional support composed of any material and/or shape that (a) allows cells to attach to it (or can be modified to allow cells to attach to it); and (b) allows cells to grow in more than one layer. This support is inoculated with stromal cells to form the living stromal matrix. [0026]
  • Living Stromal Tissue: [0027]
  • a three dimensional framework which has been inoculated with stromal cells. Whether confluent or subconfluent, stromal cells according to the invention continue to grow and divide. The living stromal tissue prepared in vitro is the source of the extracellular matrix proteins used in the injectable formulations of the invention. [0028]
  • The following abbreviations shall have the meanings indicated: [0029]
  • EDTA ethylene diamine tetraacetic acid [0030]
  • FBS fetal bovine serum [0031]
  • HBSS Hank's balanced salt solution [0032]
  • HS horse serum [0033]
  • MEM minimal essential medium [0034]
  • PBS phosphate buffered saline [0035]
  • RPMI 1640 Roswell Park Memorial Institute Medium No. 1640 (GIBCO, Inc., Grand Island, N.Y.) [0036]
  • SEM scanning electron microscopy [0037]
  • The present invention may be more fully understood by reference to the following detailed description, examples of specific embodiments and appended figures which are offered for purposes of illustration only and not by way of limitation.[0038]
  • 4. BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. FIG. 1 is a scanning electron micrograph depicting fibroblast attachment to the three-dimensional matrix and extension of cellular processes across the mesh opening. Fibroblasts are actively secreting matrix proteins and are at the appropriate stage of subconfluency which should be obtained prior to inoculation with tissue-specific cells. [0039]
  • FIG. 2A-D. FIGS. [0040] 2A-D are transmission electron micrographs of collagen isolated from extracellular matrix prepared from dermal tissue grown in vitro (FIG. 2A-B) or from a normal adult human dermal sample (FIG. 2C-D).
  • 5. DETAILED DESCRIPTION OF THE INVENTION
  • One embodiment of the present invention involves the preparation and use of an injectable extracellular matrix composition for the treatment of skin defects. The extracellular matrix proteins are derived from a living stromal tissue prepared in vitro by growing stromal cells on a three-dimensional framework resulting in a multi-layer cell culture system. In conventional tissue culture systems, the cells were grown in a monolayer. Cells grown on a three-dimensional framework support, in accordance with the present invention, grow in multiple layers, forming a cellular matrix. This matrix system approaches physiologic conditions found in vivo to a greater degree than previously described monolayer tissue culture systems. The three-dimensional cell culture system is applicable to the proliferation of different types of stromal cells and formation of a number of different stromal tissues, including but not limited to dermis, bone marrow stroma, glial tissue, cartilage, to name but a few. [0041]
  • In accordance with the present invention, the pre-established living stromal tissue comprises stromal cells grown on a three-dimensional framework or network. The stromal cells can comprise fibroblasts with or without additional cells and/or elements described more fully herein. The fibroblasts and other cells and/or elements that comprise the stroma can be fetal or adult in origin, and can be derived from convenient sources such as skin, liver, pancreas, etc. Such tissues and/or organs can be obtained by appropriate biopsy or upon autopsy. In fact, cadaver organs may be used to provide a generous supply of stromal cells and elements. [0042]
  • Once inoculated onto the three-dimensional framework, the stromal cells will proliferate on the framework, and elaborate growth factors, regulatory factors and extracellular matrix proteins that are deposited on the support. The living stromal tissue will sustain active proliferation of the culture for long periods of time. Growth and regulatory factors can be added to the culture, but are not necessary since they are elaborated by the stromal support matrix. [0043]
  • The naturally secreted extracellular matrix is collected from the three-dimensional framework and is processed further with a pharmaceutically acceptable aqueous carrier and placed in a syringe for precise placement of the biomaterial into tissues, such as the facial dermis. [0044]
  • The present invention is based, in part, on the discovery that during the growth of human stromal cells on a biodegradable or non-biodegradable three-dimensional support framework, the cells synthesize and deposit on the three-dimensional support framework a human extracellular matrix as produced in normal human tissue. The extracellular matrix is secreted locally by cells and not only binds cells and tissues together but also influences the development and behavior of the cells it contacts. The extracellular matrix contains various fiber-forming proteins interwoven in a hydrated gel composed of a network of glycosaminoglycan chains. The glycosaminoglyeans are a heterogeneous group of long, negatively charged polysaccharide chains, which (except for hyaluronic acid) are covalently linked to protein to form proteoglycan molecules. [0045]
  • The fiber-forming proteins are of two functional types: mainly structural (collagens and elastin) and mainly adhesive (such as fibronectin and laminin). The fibrillar collagens (types I, II, and III) are rope-like, triple-stranded helical molecules that aggregate into long cable-like fibrils in the extracellular space; these in turn can assemble into a variety of highly ordered arrays. Type IV collagen molecules assemble into a sheetlike meshwork that forms the core of all basal laminae. Elastin molecules form an extensive cross-linked network of fibers and sheets that can stretch and recoil, imparting elasticity to the matrix. Fibronectin and laminin are examples of large adhesive glycoproteins in the matrix; fibronectin is widely distributed in connective tissues, whereas laminin is found mainly in basal laminae. By means of their multiple binding domains, such proteins help cells adhere to and become organized by the extracellular matrix. [0046]
  • As an example, a naturally secreted human dermal extracellular matrix contains type I and type III collagens, fibronectin, tenascin, glycosaminoglycans, acidic and basic FGF, TGF-α and TGF-β, KGF, decorin and various other secreted human dermal matrix proteins. As naturally secreted products, the various extracellular matrix proteins are produced in the quantities and ratios similar to that existing in vivo. Moreover, growth of the stromal cells in three dimensions will sustain active proliferation of cells in culture for much longer time periods than will monolayer systems. Further, the three-dimensional system supports the maturation, differentiation, and segregation of cells in culture in vitro to form components of adult tissues analogous to counterparts found in vivo. Thus, the extracellular matrix created by the cells in culture is more analogous to native tissues. [0047]
  • Although the applicants are under no duty or obligation to explain the mechanism by which the invention works, a number of factors inherent in the three-dimensional culture system may contribute to these features of the three dimensional culture system: [0048]
  • (a) The three-dimensional framework provides a greater surface area for protein attachment, and consequently, for the adherence of stromal cells. [0049]
  • (b) Because of the three-dimensionality of the framework, stromal cells continue to actively grow in contrast to cells in monolayer cultures, which grow to confluence, exhibit contact inhibition, and cease to grow and divide. The elaboration of growth and regulatory factors by replicating stromal cells may be partially responsible for stimulating proliferation and regulating differentiation of cells in culture. [0050]
  • (c) The three-dimensional framework allows for a spatial distribution of cellular elements which is more analogous to that found in the counterpart tissue in vivo. [0051]
  • (d) The increase in potential volume for cell growth in the three-dimensional system may allow the establishment of localized microenvironments analogous to native counterparts found in vivo. [0052]
  • (e) The three-dimensional matrix maximizes cell-cell interactions by allowing greater potential for movement of migratory cells, such as macrophages, monocytes and possibly lymphocytes in the adherent layer. [0053]
  • (f) It has been recognized that maintenance of a differentiated cellular phenotype requires not only growth/differentiation factors but also the appropriate cellular interactions. The present invention effectively recreates the stromal tissue microenvironment. [0054]
  • The three-dimensional stromal support, the culture system itself, and its maintenance, as well as various uses of the three-dimensional cultures and of the naturally secreted extracellular matrix are described in greater detail in the subsections below. Solely for ease of explanation, the detailed description of the invention is divided into the three sections, (i) growth of the three-dimensional stromal cell culture, (ii) isolation of the naturally secreted human extracellular matrix, and (iii) formulation of the isolated extracellular matrix into preparations for injection at the site of soft tissue defects. [0055]
  • 5.1. Preparing the Living Stromal Tissue In Vitro
  • The three-dimensional support used to culture stromal tissue may be of any material and/or shape that: [0056]
  • (a) allows cells to attach to it (or can be modified to allow cells to attach to it); and [0057]
  • (b) allows cells to grow in more than one layer. [0058]
  • A number of different materials may be used to form the framework, such as non-biodegradable or biodegradable materials. For example, non-biodegradable materials include but are not limited to: nylon (polyamides), dacron (polyesters), polystyrene, polypropylene, polyacrylates, polyvinyl compounds (e.g., polyvinylchloride), polycarbonate (PVC), polytetrafluorethylene (PTFE; teflon), thermanox (TPX), etc. Additionally, biodegradable material may also be utilized, including but not limited to: nitrocellulose, cotton, polyglycolic acid (PGA), cat gut sutures, cellulose, gelatin, dextran, collagen, chitosan, hyaluronic acid, etc. Any of these materials, bio- or non-biodegradable, can be woven into a mesh to form a three-dimensional framework. Alternatively, the materials can be used to form other types of three-dimensional frameworks, for example, sponges, such as collagen sponges. [0059]
  • Certain materials, such as nylon, polystyrene, etc., are poor substrates for cellular attachment. When these materials are used as the three-dimensional support framework, it is advisable to pre-treat the framework prior to inoculation of stromal cells in order to enhance the attachment of stromal cells to the framework. For example, prior to inoculation with stromal cells, nylon frameworks can be treated with 0.1 M acetic acid, and incubated in polylysine, FBS, and/or collagen to coat the nylon. Polystyrene can be similarly treated using sulfuric acid. A convenient nylon mesh which can be used in accordance with the invention is Nitex, a nylon filtration mesh having an average pore size of 210 μm and an average nylon fiber diameter of 90 μm (#3-210/36, Tetko, Inc., N.Y.). [0060]
  • Stromal cells comprising fibroblasts derived from adult or fetal tissue, with or without other cells and elements described below, are inoculated onto the framework. These fibroblasts may be derived from organs, such as skin, liver, pancreas, etc. which can be obtained by biopsy, where appropriate, or upon autopsy. In fact, fibroblasts can be obtained in quantity rather conveniently from any appropriate cadaver organ. In a preferred embodiment, fetal fibroblasts can be obtained in high quantity from foreskin. [0061]
  • Fibroblasts may be readily isolated by disaggregating an appropriate organ or tissue which is to serve as the source of the fibroblasts. This can be readily accomplished using techniques known to those skilled in the art. For example, the tissue or organ can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that-weaken the connections between neighboring cells making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage. Enzymatic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination. These include but are not limited to trypsin, chymotrypsin, collagenase, elastase, hyaluronidase, DNase, pronase, and/or dispase etc. Mechanical disruption can also be accomplished by a number of methods including, but not limited to the use of grinders, blenders, sieves, homogenizers, pressure cells, or insonators to name but a few. For a review of tissue disaggregation techniques, see Freshney, Culture of Animal Cells. A Manual of Basic Technique, 2d Ed., A. R. Liss, Inc., New York, 1987, Ch. 9, pp. 107-126. [0062]
  • Once the tissue has been reduced to a suspension of individual cells, the suspension can be fractionated into subpopulations from which the fibroblasts and/or other stromal cells and/or elements can be obtained. This also may be accomplished using standard techniques for cell separation including, but not limited to, cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection), separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter-streaming centrifugation), unit gravity separation, countercurrent distribution, electrophoresis and fluorescence-activated cell sorting. For a review of clonal selection and cell separation techniques, see Freshney, Culture of Animal Cells. A Manual of Basic Techniques, 2d Ed., A. R. Liss, Inc., New York, 1987, Ch. 11 and 12, pp. 137-168. [0063]
  • The isolation of fibroblasts, for example, can be carried out as follows: fresh tissue samples are thoroughly washed and minced in Hanks' balanced salt solution (HBSS) in order to remove serum. The minced tissue is incubated from 1-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the dissociated cells are suspended, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown. The isolated fibroblasts can then be grown to confluency, lifted from the confluent culture and inoculated onto the three-dimensional framework, see Naughton et al., 1987, J. Med. 18(3&4):219-250. Inoculation of the three-dimensional framework with a high concentration of stromal cells, e.g., approximately 10[0064] 6 to 5×107 cells/ml, will result in the establishment of the three-dimensional stromal support in shorter periods of time.
  • In addition to fibroblasts, other cells can be added to form the three-dimensional stromal cell culture-producing extracellular matrix. For example, other cells found in loose connective tissue may be inoculated onto the three-dimensional support framework along with fibroblasts. Such cells include, but are not limited to, endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, chondrocytes, etc. These stromal cells can be readily derived from appropriate organs such as skin, liver, etc., using methods known, such as those discussed above. [0065]
  • In one embodiment of the present invention, stromal cells which are specialized for the particular tissue to be cultured can be added to the fibroblast stroma for the production of a tissue type specific extracellular matrix. For example, dermal fibroblasts can be used to form the three-dimensional subconfluent stroma for the production of skin-specific extracellular matrix in vitro. Alternatively, stromal cells of hematopoietic tissue including, but not limited to, fibroblast endothelial cells, macrophages/monocytes, adipocytes and reticular cells, can be used to form the three-dimensional subconfluent stroma for the production of a bone marrow-specific extracellular matrix in vitro, see infra. Hematopoietic stromal cells can be readily obtained from the “buffy coat” formed in bone marrow suspensions by centrifugation at low forces, e.g., 3000×g. Stromal cells of liver may include fibroblasts, Kupffer cells, and vascular and bile duct endothelial cells. Similarly, glial cells can be used as the stroma to support the proliferation of neurological cells and tissues. Glial cells for this purpose can be obtained by trypsinization or collagenase digestion of embryonic or adult brain. Ponten and Westermark, 1980, In Federof, S. Hertz, L., eds, “Advances in Cellular Neurobiology,” Vol.1, New York, Academic Press, pp.209-227. [0066]
  • For certain uses in vivo it is preferable to obtain the stromal cells from the patient's own tissues. The growth of cells in the presence of the three-dimensional stromal support framework can be further enhanced by adding to the framework, or coating the framework support with proteins, e.g., collagens, elastic fibers, reticular fibers, glycoproteins; glycosaminoglycans, e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratan sulfate, etc.; a cellular matrix, and/or other materials. [0067]
  • After inoculation of the stromal cells, the three-dimensional framework is incubated in an appropriate nutrient medium under physiologic conditions favorable for cell growth, i.e., promoting mitosis, i.e., cell division. Many commercially available media such as RPMI 1640, Fisher's, Iscove's, McCoy's, and the like may be suitable for use. It is important that the three-dimensional stromal culture be suspended or floated in the medium during the incubation period in order to maximize proliferative activity. In addition, the culture should be “fed” periodically to remove the spent media, depopulate released cells, and to add fresh media. [0068]
  • During the incubation period, the stromal cells will grow linearly along and envelop the three-dimensional framework before beginning to grow into the openings of the framework. The cells are grown to an appropriate degree to allow for adequate deposition of extracellular matrix proteins. [0069]
  • The openings of the framework should be of an appropriate size to allow the stromal cells to stretch across the openings. Maintaining actively growing stromal cells which stretch across the framework enhances the production of growth factors which are elaborated by the stromal cells, and hence, will support long term cultures. For example, if the openings are too small, the stromal cells may rapidly achieve confluence but be unable to easily exit from the mesh. Trapped cells can exhibit contact inhibition and cease production of the appropriate factors necessary to support proliferation and maintain long term cultures. If the openings are too large, the stromal cells are unable to stretch across the opening. This will also decrease stromal cell production of the appropriate factors necessary to support proliferation and maintain long term cultures. When using a mesh type of matrix, as exemplified herein, we have found that openings ranging from about 150 μm to about 220 μm will work satisfactorily. However, depending upon the three-dimensional structure and intricacy of the framework, other sizes may work equally well. In fact, any shape or structure that allows the stromal cells to stretch and continue to replicate and grow for lengthy time periods will work in accordance with the present invention. [0070]
  • Different proportions of the various types of collagen deposited on the framework can be achieved by inoculating the framework with different tissue-specific cells. For example, for hematopoietic cells, the matrix should preferably contain collagen types III, IV and I in an approximate ratio of 6:3:1 in the initial matrix. For skin, collagen types I and III are preferably deposited in the initial matrix. The proportions of collagen types deposited can be manipulated or enhanced by selecting fibroblasts which elaborate the appropriate extracellular matrix proteins. This can be accomplished using monoclonal antibodies of an appropriate isotype or subclass which are capable of activating complement, and which define particular collagen types. These antibodies in combination with complement can be used to negatively select the fibroblasts which express the desired collagen type. Alternatively, the stroma used to inoculate the framework can be a mixture of cells which synthesize the appropriate collagen types desired. The distribution and origins of the five types of collagen is shown in Table I. [0071]
    TABLE I
    DISTRIBUTIONS AND ORIGINS OF THE
    FIVE TYPES OF COLLAGEN
    Collagen Principal Tissue
    Type Distribution Cells of Origin
    I Loose and dense ordinary Fibroblasts and
    connective tissue; collagen reticular cells;
    fibers smooth muscle cells
    Fibrocartilage
    Bone Osteoblast
    Dentin Odontoblasts
    II Hyaline and elastic Chondrocytes
    cartilage Retinal Cells
    Vitreous body of eye
    III Loose connective tissue; Fibroblasts and
    reticular fibers reticular cells
    Papillary layer of dermis Smooth muscle cells;
    Blood vessels endothelial cells
    IV Basement membranes Epithelial and
    endothelial cells
    Lens capsule of eye Lens fibers
    V Fetal membranes; placenta Fibroblasts
    Basement membranes
    Bone
    Smooth muscle Smooth muscle cells
  • Thus, depending upon the collagen types desired, the appropriate stromal cell(s) can be selected to inoculate the three-dimensional framework. [0072]
  • The three-dimensional extracellular matrix producing culture of the present invention affords a vehicle for introducing gene products in vivo. In certain situations, it may be desirable to prepare an extracellular matrix containing a foreign gene product, growth factor, regulatory factor, etc. In such cases, the cells may be genetically engineered to express the gene product, or altered forms of the gene product that are immobilized in the extracellular matrix laid down by the stromal cells. For example, using recombinant DNA techniques, a gene of interest can be placed under the control of an inducible promoter. The recombinant DNA construct containing the gene can be used to transform or transfect a host cell which is cloned and then clonally expanded in the three-dimensional culture system. The use of the-three-dimensional culture in this regard has a number of advantages. First, since the culture comprises eukaryotic cells, the gene product will be properly expressed and processed in culture to form an active product. Second, the number of transfected cells can be substantially enhanced to be of clinical value, relevance, and utility. The three-dimensional cultures of the present invention allow for expansion of the number of transfected cells and amplification (via cell division) of transfected cells. [0073]
  • Preferably, the expression control elements used should allow for the regulated expression of the gene so that the product can be over synthesized in culture. The transcriptional promoter chosen, generally, and promoter elements specifically, depends, in part, upon the type of tissue and cells cultured. Cells and tissues which are capable of secreting proteins (e.g., those characterized by abundant rough endoplasmic reticulum and golgi complex) are preferable. [0074]
  • During incubation of the three-dimensional culture, proliferating cells are released from the framework. These released cells can stick to the walls of the culture vessel where they can continue to proliferate and form a confluent monolayer. This should be prevented or minimized, for example, by removal of the released cells during feeding, or by transferring the three-dimensional framework to a new culture vessel. The presence of a confluent monolayer in the vessel will “shut down” the growth of cells in the three-dimensional framework and/or culture. Removal of the confluent monolayer or transfer of the stromal culture to fresh media in a new vessel will restore proliferative activity of the three-dimensional culture system. Such removal or transfers should be done in any culture vessel which has a stromal monolayer exceeding 25% confluency. Alternatively, the culture system can be agitated to prevent the released cells from sticking, or instead of periodically feeding the cultures, the culture system could be set up so that fresh media continuously flows through the system. The flow rate can be adjusted to both maximize proliferation within the three-dimensional culture, and to wash out and remove cells released from the matrix, so that they will not stick to the walls of the vessel and grow to confluence. In any case, the released stromal cells can be collected and crypreserved for future use. [0075]
  • Once inoculated onto the three-dimensional framework, adherence of the fibroblasts is seen quickly (e.g., within hours) and the fibroblasts begin to stretch across the framework openings within days. These fibroblasts are metabolically active, secrete extracellular matrix and rapidly form a dermal equivalent consisting of active fibroblasts and collagen. [0076]
  • FIG. 1 illustrates the ability of the fibroblasts to arrange themselves into parallel layers between the naturally-secreted collagen bundles. These fibroblasts exhibit a rapid rate of cell division and protein secretion. [0077]
  • 5.2. Removal of the Extracellular Matrix from the Framework
  • After the cells have been inoculated onto the framework and cultured under conditions favoring cellular growth, such that a desired amount of extracellular matrix is secreted on to the three-dimensional framework, the cells are killed and the naturally secreted extracellular matrix is processed further. [0078]
  • This involves first killing the cells and removing the killed cells and any cellular debris from the three-dimensional framework. This process is carried out in a number of different ways. For example, the cells can be killed by flash-freezing the living stromal tissue prepared in vitro in liquid nitrogen without a cryopreservative. Another way to kill the cells is to irrigate the inoculated three-dimensional framework with sterile water, such that the cells burst in response to osmotic pressure. Once the cells have been killed, one can, for example, disrupt the cellular membranes and remove the cellular debris by a mild detergent rinse, such as EDTA, CHAPS or a zwitterionic detergent, followed by treatment with a cryoprotectant such as DMSO, propylene glycol, butanediol, raffinose, polyvinyl pyrrolidone, dextran or sucrose and vitrified in liquid nitrogen. Alternatively, the framework can be subjected to enzymatic digestion and/or extracting with reagents that break down the cellular membranes and allow removal of cell contents. Examples of detergents include non-ionic detergents (for example, TRITON X-100, octylphenoxy polyethoxyethanol, (Rohm and Haas); BRIJ-35, a polyethoxyethanol lauryl ether (Atlas Chemical Co.), TWEEN 20, a polyethoxyethanol sorbitan monolaureate (Rohm and Haas), LUBROL-PX, or polyethylene lauryl ether (Rohm and Haas)); and ionic detergents (for example, sodium dodecyl sulphate, sulfated higher aliphatic alcohol, sulfonated alkane and sulfonated alkylarene containing 7 to 22 carbon atoms in a branched or unbranched chain). Enzymes can be used also and can include nucleases (for example, deoxyribonuclease and ribonuclease), phospholipases and lipases. An advantage to using a mild detergent rinse is that it will solubilize membrane-bound proteins, which are often highly antigenic. [0079]
  • Once the cells have been killed and the cellular debris has been removed, the collection of the naturally secreted human extracellular matrix can be accomplished in a variety of ways which depends on whether the three-dimensional framework is composed of material that is biodegradable or non-biodegradable. For example, if the framework is composed of non-biodegradable material, one can remove the extracellular matrix from a non-biodegradable support by subjecting the three-dimensional framework to sonication and/or to high pressure water jets and/or to mechanical scraping and/or to a mild treatment with detergents and/or enzymes to remove the attached extracellular matrix from the framework. [0080]
  • If the extracellular matrix is deposited on a biodegradable three-dimensional framework, after killing and removing the cells and cellular debris, the extracellular matrix can be recovered, for example, by simply allowing the framework to degrade in solution, i.e., allow the framework to dissolve, thus freeing the extracellular matrix. Additionally, if the biodegradable support is composed of a material which can be injected, like the extracellular matrix itself, one can process the entire extracellular matrix coated framework into syringes for injection. Further, if the extracellular matrix is deposited on a biodegradable support, the matrix can be removed by the same methods as if the matrix had been deposited on a non-biodegradable support, i.e., by subjecting the three-dimensional framework to sonication and/or to high pressure water jets and/or to mechanical scraping and/or to a mild treatment with detergents and/or enzymes to remove the attached extracellular matrix from the framework. None of the removal processes are designed to damage and/or denature the naturally secreted human extracellular matrix produced by the cells. [0081]
  • 5.3. Formulation and Use of Injectable Preparations
  • Once the naturally secreted extracellular matrix has been collected, it is processed further. The extracellular matrix can be homogenized to fine particles, such that it can pass through a surgical needle. Homogenization is well known in the art, for example, by sonication. Further, the extracellular matrix can be cross-linked by gamma irradiation without the use of chemical cross-linking agents, such as glutaraldehyde, which are toxic. The gamma irradiation should be a minimum of 20 M rads to sterilize the material since all bacteria, fungi, and viruses are destroyed at 0.2 M rads. Preferably, the extracellular matrix can be irradiated from 0.25 to 2 M rads to sterilize and cross-link the extracellular matrix. [0082]
  • Further, the amounts and/or ratios of the collagens and other proteins may be adjusted by mixing extracellular matrices secreted by other cell types prior to placing the material in a syringe. For example, biologically active substances, such as proteins and drugs, can be incorporated in the compositions of the present invention for release or controlled release of these active substances after injection of the composition. Exemplary biologically active substances can include tissue growth factors, such as TGF-β, and the like which promote healing and tissue repair at the site of the injection. [0083]
  • Final formulation of the aqueous suspension of naturally secreted human extracellular matrix will typically involve adjusting the ionic strength of the suspension to isotonicity (i.e., about 0.1 to 0.2) and to physiological pH (i.e., about pH 6.8 to 7.5) and adding a local anesthetic, such as lidocaine, (usually at a concentration of about 0.3% by weight) to reduce local pain upon injection. The final formulation will also typically contain a fluid lubricant, such as maltose, which must be tolerated by the body. Exemplary lubricant components include glycerol, glycogen, maltose and the like organic polymer base materials, such as polyethylene glycol and hyaluronic acid as well as non-fibrillar collagen, preferably succinylated collagen, can also act as lubricants. Such lubricants are generally used to improve the injectability, intrudability and dispersion of the injected biomaterial at the site of injection and to decrease the amount of spiking by modifying the viscosity of the compositions. This final formulation is by definition the processed extracellular matrix in a pharmaceutically acceptable carrier. [0084]
  • The matrix is subsequently placed in a syringe or other injection apparatus for precise placement of the matrix at the site of the tissue defect. In the case of formulations for dermal augmentation, the term “injectable” means the formulation can be dispensed from syringes having a gauge as low as 25 under normal conditions under normal pressure without substantial spiking. Spiking can cause the composition to ooze from the syringe rather than be injected into the tissue. For this precise placement, needles as fine as 27 gauge (200μ I.D.) or even 30 gauge (150μ I.D.) are desirable. The maximum particle size that can be extruded through such needles will be a complex function of at least the following: particle maximum dimension, particle aspect ratio (length:width), particle rigidity, surface roughness of particles and related factors affecting particle:particle adhesion, the viscoelastic properties of the suspending fluid, and the rate of flow through the needle. Rigid spherical beads suspended in a Newtonian fluid represent the simplest case, while fibrous or branched particles in a viscoelastic fluid are likely to be more complex. [0085]
  • The above described steps in the process for preparing injectable naturally secreted human extracellular matrix are preferably carried out under sterile conditions using sterile materials. The processed extracellular matrix in a pharmaceutically acceptable carrier can be injected intradermally or subcutaneously to augment soft tissue, to repair or correct congenital anomalies, acquired defects or cosmetic defects. Examples of such conditions are congenital anomalies as hemifacial microsomia, malar and zygomatic hypoplasia, unilateral mammary hypoplasia, pectus excavatum, pectoralis agenesis (Poland's anomaly) and velopharyngeal incompetence secondary to cleft palate repair or submucous cleft palate (as a retropharyngeal implant); acquired defects (post-traumatic, post-surgical, post-infectious) such as depressed scars, subcutaneous atrophy (e.g., secondary to discoid lupis erythematosus), keratotic lesions, enophthalmos in the unucleated eye (also superior sulcus syndrome), acne pitting of the face, linear scleroderma with subcutaneous atrophy, saddle-nose deformity, Romberg's disease and unilateral vocal cord paralysis; and cosmetic defects such as glabellar frown lines, deep nasolabial creases, circum-oral geographical wrinkles, sunken cheeks and mammary hypoplasia. The compositions of the present invention can also be injected into internal tissues, such as the tissues defining body sphincters to augment such tissues. [0086]
  • Various sample embodiments of the invention are described in the sections below. For purposes of description only, and not by way of limitation, the three-dimensional culture system of the invention is described based upon the type of tissue and cells used in various systems. These descriptions specifically include but are not limited to bone marrow, skin, epithelial cells, and cartilage but it is expressly understood that the three-dimensional culture system can be used with other types of cells and tissues. The invention is also illustrated by way of examples, which demonstrate characteristic data generated for each system described. [0087]
  • EXAMPLES 6. Example Three-Dimensional Skin Stromal Culture System
  • The subsections below describe the three-dimensional culture system of the invention for culturing different stromal cells in vitro. Briefly, cultures of fibroblasts were established on nylon mesh which had been previously sterilized. Within 6-9 days of incubation, adherent fibroblasts began to grow into the meshwork openings and deposited parallel bundles of collagen. Indirect immunofluorescence using monoclonal antibodies showed predominantly type I collagen with some type III as well. [0088]
  • 6.1. Establishment of the Three-Dimensional Stroma of Skin Fibroblasts
  • Skin fibroblasts were isolated by mincing dermal tissue, trypsinization for 2 hours, and separation of cells into a suspension by physical means. Fibroblasts were grown to confluency in 25 cm[0089] 2 Falcon tissue culture dishes and fed with RPMI 1640 (Sigma, MO) supplemented with 10% fetal bovine serum (FBS), fungizone, gentamicin, and penicillin/streptomycin. Fibroblasts were lifted by mild trypsinization and cells were plated onto nylon filtration mesh, the fibers of which are approximately 90 μm in diameter and are assembled into a square weave with a mesh opening of 210 μm (Tetko, Inc., NY). The mesh was pretreated with a mild acid wash and incubated in polylysine and FBS. Adherence of the fibroblasts was seen within 3 hours, and fibroblasts began to stretch across the mesh openings within 5-7 days of initial inoculation. These fibroblasts were metabolically active, secreted an extracellular matrix, and rapidly formed a dermal equivalent consisting of active fibroblasts and collagen. FIG. 1 is a scanning electron micrograph depicting fibroblast attachment and extension of cellular processes across the mesh opening.
  • 6.2 Establishment of the Three-Dimensional Bone Marrow Stromal Cultures
  • Bone marrow was aspirated from multiple sites on the posterior iliac crest of hematologically normal adult volunteers after informed consent was obtained. Specimens were collected into heparinized tubes and suspended in 8 ml of RPMI 1640 medium which was conditioned with 10% FBS and 5-10% HS and supplemented with hydrocortisone, fungizone, and streptomycin. The cell clumps were disaggregated and divided into aliquots of 5×10[0090] 6 nucleated cells.
  • Nylon filtration screen (#3-210/36, Tetko Inc., NY) was used as a three-dimensional framework to support all stromal cell cultures described in the examples below. The screen consisted of fibers, which were 90 μm in diameter, assembled into a square weave pattern with sieve openings of 210 μm. Stromal cells were inoculated using the protocols described in Section 6.1. Adherence and subsequent growth of the stromal elements was monitored using inverted phase contrast microscopy and scanning electron microscopy (SEM). [0091]
  • 6.3 Preparation of the Three-Dimensional Oral Mucosal Epithelial Stromal Matrix
  • Samples of oral mucosal tissue were obtained from orthodontic surgical specimens. Tissue was washed three times with fresh MEM containing antibiotics (2 ml of antibiotic antimycotic solution from GIBCO, Cat. #600-5240 AG; and 0.01 ml of gentamicin solution from GIBCO Cat. #600-5710 AD per 100 cc MEM), cut into small pieces, then washed with 0.02% EDTA (w/v). 0.25% trypsin (in PBS without Ca[0092] ++ or Mg++) was added; after a few seconds, the tissue pieces were removed and placed in fresh trypsin (in PBS without Ca++ or Mg++) and refrigerated at 4° C. overnight. Tissues were removed and placed in fresh trypsin solution, and gently agitated until cell appeared to form a single-cell suspension. The single-cell suspension was then diluted in MEM containing 10% heat inactivated fetal bovine serum and centrifuged at 1400×g for 7 minutes. The supernatant was decanted and the pellet containing mucosal epithelial cells was placed into seeding medium. Medium consisted of DMEM with 2% Ultrosen G, 1×L-glutamine, 1×non-essential amino acids, penicillin and streptomycin. The cells were seeded onto a three-dimensional framework. The three-dimensional stromal culture was generated using oral fibroblasts and 8 mm×45 mm pieces of nylon filtration screen (#3-210/36, Tetko Inc., NY). The mesh was soaked in 0.1 M acetic acid for 30 minutes and treated with 10 mM polylysine suspension for 1 hour. The meshes were placed in a sterile petri dish and inoculated with 1×106 oral fibroblasts collected as described above in DMEM complete medium. After 1-2 hours of incubation at 5% CO2 the meshes were placed in a Corning 25 cm2 tissue culture flask, floated with an additional 5 ml of medium, and allowed to reach subconfluence, being fed at 3 day intervals. Cultures were maintained in DMEM complete medium at 37° C. and 5% CO2 in a humidified atmosphere and were fed with fresh medium every 3 days.
  • 6.4 Establishment of the Three Dimensional Small Vessel Endothelial Stromal Cell Culture
  • Small vessel endothelial cells isolated from the brain according to the method of Larson et al., 1987, Microvasc. Res. 34:184 were cultured in vitro using T-75 tissue culture flasks. The cells were maintained in Dulbecco's Modified Eagle Medium/Hams-F-12 medium combination (the solution is available as a 1:1 mixture). The medium was supplemented with 20% heat-inactivated fetal calf serum (FCS), glutamine, and antibiotics. The cells were seeded at a concentration of 1×10[0093] 6 cells per flask, and reached a confluent state within one week. The cells were passaged once a week, and, in addition, were fed once a week with DMEM/Hams-F-12 containing FCS, glutamine, and antibiotics as described. To passage the cells, flasks were rinsed twice with 5 ml of PBS (without Ca++ or Mg++) and trypsinized with 3 ml of 0.05% Trypsin and 0.53 mM EDTA. The cells were pelleted, resuspended, and tested for viability by trypan blue exclusion, seeded and fed with 25 ml of the above mentioned DMEM/Hams-F-12 supplemented medium. A factor VIII related antigen assay, Grulnick et al., 1977, Ann. Int. Med. 86:598-616, is used to positively identify endothelial cells, and silver staining was used to identify tight junctional complexes, specific to only small vessel endothelium.
  • Nylon filtration screen mesh (#3-210/36, Tetko, Inc., NY) was prepared essentially as described above. The mesh was soaked in an acetic acid solution (1 ml glacial acetic acid plus 99 ml distilled H[0094] 2O) for thirty minutes, was rinsed with copious amounts of distilled water and then autoclaved. Meshes were coated with 6 ml fetal bovine serum per 8×8 cm mesh and incubated overnight. The meshes were then stacked, three high, and 3×107 small vessel endothelial cells (cultured as described) were seeded onto the stack, and incubated for three hours at 37° C. under 5% CO2 in a humidified atmosphere. The inoculated meshes were fed with 10 ml of DME/Hams-F-12 medium every 3-4 days until complete confluence was reached (in approximately two weeks).
  • 6.5 Establishment of the Three Dimensional Chondrocyte Stromal Cell Culture
  • Cartilage was harvested from articular surfaces of human joints. The cartilage pieces were digested with collagenase (0.2% w/v) in complete medium (DMEM with 10% fetal bovine serum, glutamine, non-essential amino acids, sodium pyruvate, 50 μg/ml ascorbate and 35 μg/ml gentamicin) for 20 hours at 37° C. Liberated chondrocytes were spun, resuspended in complete medium, counted and plated at 1×10[0095] 6 cells per T-150 flask. Cells were routinely passed at confluence (every 5-7 days).
  • Polyglycolic acid mesh (1 mm diameter×2 mm thick) was sterilized by ethylene oxide or electron beam treatment and presoaked overnight in complete medium. The mesh was seeded in 6 well plates with 3-4×10[0096] 6 cells per mesh in a total volume of 10 μl and incubated for 3-4 hours at 37° C. in a tissue culture incubator. At this time, 1.5 ml of media was added. The seeded mesh was incubated overnight. 5 ml of media was added the next day. Media was changed three times per week until confluence is reached.
  • 7. Example Extracellular Matrix Composition
  • The extracellular matrix has been characterized by a number of analytic methods to determine its content of matrix proteins, each value is the average of at least two independent determinations. The matrix contained type I and type III collagens, fibronectin, tenascin, sulfated glycosaminoglycans, decorin and various other secreted human extracellular matrix proteins. Additionally, the secreted matrix proteins were found throughout the three-dimensional support framework. The extracellular matrix contained a total protein amount of 292 mg/cm[0097] 2±0.06; fibronectin was present at 3.4 mg/cm2±1.2; and tenascin at 1.7 mg/cm2±0.6. Both fibronectin and tenascin showed the expected molecular weight distributions on immunoblots.
  • 7.1. Collagen Content of the Extracellar Matrix
  • Collagen content of the extracellular matrix was determined using the Sirius Red assay. The binding of Sirius Red F3BA in saturated picric acid solution has been used widely to estimate fibrotic collagen deposition. Bedossa et al., 1989, Digestion 44(1):7-13; Finkelstein et al., 1990, Br. J. Ophthalmol. 74(5):280-282; James et al., 1990, Liver 10(1):1-5. The specificity of Sirius Red binding to collagen is based largely on its use as a histological stain. In rat liver with various degrees of cholestatic fibrosis, collagen content measured by Sirius Red binding shows strong correlation with hydroxyproline content. Walsh et al., 1992, Analyt. Biochem. 203:187-190. In addition, histological staining with Sirius Red is birefringent, indicating directional binding related to the orientation of the collagen strands. Sirius Red is known to bind to proteins other than the classical collagens that contain collagen-like triple helices, such as the complement component C1. Some minor binding to serum albumin has also been found, although control experiments using bovine serum albumin standard showed no interference with the assay. The interference is estimated to represent less than 2% of the collagen signal in the extracellular matrix and the use of Sirius Red assay gives a reproducible method for measuring collagens. The extracellular matrix contained a collagen content of 0.61 mg/cm[0098] 2±0.09. Further, collagens I and III showed the expected molecular weight distributions on immunoblots
  • 7.2. Collagen Fibers Visualized Via Electron Microscopy
  • Collagen derived from the dermal tissue grown in vitro and collagen derived from a normal adult human dermal sample were processed and visualized by transmission electron microscopy (TEM). Briefly, the respective collagens were weighed and placed in a sterile 50 ml centrifuge tube with 30 ml 0.05 M Tris buffer, pH 8.0. After mixing for two hours on a wrist shaker, the Tris buffer was removed and the specimen placed in a homogenization cylinder along with 30 ml fresh 0.05 M Tris buffer. The sample was homogenized for 30 seconds in buffer alone and then for two 30 second bursts following the addition of a dispersing agent as described in U.S. Pat. Nos. 4,969,912 and 5,332,802. The temperature was maintained at 5-10° C. during the mechanical disruption process. The dispersing agent was added at a concentration of 0.05% (wet weight of the collagen). The homogenized preparation was centrifuged at 3500 rpm for 6 minutes to separate the dispersed collagenous material from the yet undispersed material. The undispersed residue was again treated with dispersing agent at 0.05% (wet weight of the collagen) and homogenized for two 30 second bursts. The dispersion was again centrifuged to recover dispersed collagenous material which was added to the first recovery. [0099]
  • The collagenous dispersion was filtered through a 100 micron filter, centrifuged at 3500 rpm and the pellet was washed 3 times with 0.004 M phosphate buffer, pH 7.4. The last centrifugation was conducted at 10,000 rpm to pack the collagenous pellet. Samples were then collected for TEM. As shown in FIGS. [0100] 2A-D, the collagen fibers isolated from either the extracellular matrix prepared from dermal tissue grown in vitro (FIGS. 2A-B) or from normal adult human dermis (FIGS. 2C-D) appeared identical in that intact collagen fibers with typical collagen banding and normal periodicity in both preparations.
  • 7.3. Glycosaminoglycans Present in the Extracellular Matrix
  • Glycosaminoglycans have been shown to play a variety of structural and functional roles in the body and their presence in the secreted extracellular matrix is important. Table II lists a number of examples of glycosaminoglycans which have been determined to be found in the extracellular matrix as well as their functional importance in normal dermis. [0101]
    TABLE II
    NAME LOCATION GLYCAN FUNCTION MECHANISM
    Versican Matrix 12-15 Structural Binds hyaluronic
    Chondroitin sulfate acid and collagen
    Decorin Matrix 1 Chondroitin/dermatan Binding TGF-β Inactivates
    sulfate and other growth factors
    growth factors;
    binds to
    collagen
    Betaglycan Cell  1-4 TGF-β Type III Adjunct receptor
    membrane Chondroitin/heparan receptor for TGFβ
    sulfate
    Syndecan Cell  1-3 Chondroitin Growth factor
    membrane sulfate, 1-2 heparan binding
    sulfate
  • Further, the extracellular matrix was found to contain a total of 2.8 mg/cm[0102] 2±0.1 sulfated glycosaminoglycans.
  • 7.4. Growth Factors Present in the Extracellular Matrix
  • The cells producing and depositing the extracellular matrix also expressed a number of different growth factors. Growth factors are important in the extracellular matrix for two reasons. During the growth of and deposition of the extracellular matrix, naturally seeded growth factors help to control cell proliferation and activity. Further, growth factors remain attached to the extracellular matrix. A variety of growth factors have been determined to be expressed during the deposition of the matrix. [0103]
  • The expression of growth factors has been examined by polymerase chain reaction of reverse transcripts (RT-PCR) of total RNA. Briefly, RNA was extracted from the growing cells by an SDS precipitation and organic solvent partition procedure. The RNA was transcribed using superscript reverse transcriptase and random hexamer primers. The same batch of reverse transcript was used for detection of all the growth factors. PCR was performed under standard conditions, using 4 μl reverse transcript, corresponding to 200 ng RNA in a total volume of 20 μl. [0104]
  • Based on this assay, acidic and basic FGF, TGF-α and TGF-β, and KGF mRNA transcripts were present as were several others as shown in Table III, including PDGF, amphiregulin, HBEGF, IGF, SPARC and VEGF. Of these, PDGF and TGF-β3 are thought to be involved in regulation of cell proliferation and matrix deposition in culture, while TGF-β1, HBEGF, KGF, SPARC, VEGF and decorin are deposited in the matrix. Amphiregulin, IGF-1, IGF-2 and IL-1 were not expressed at the sensitivity used in these experiments. [0105]
    TABLE III
    Messenger
    RNA Full Name Function Expression
    PDGF-A Chain Platelet- Mitogen for ++
    derived growth fibroblasts,
    factor, A granulation
    chain tissue,
    chemotactic
    PDGF-B Chain Platelet- Mitogen for 0 − (+)*
    derived growth fibroblasts,
    factor, B granulation
    chain tissue,
    chemotactic
    IGF-1 Insulin-like Mitogen for 0
    growth factor-1 fibroblasts
    IGF-2 Insulin-like Mitogen for (+)
    growth factor-2 fibroblasts
    TGF-α Transforming Mitogen for +
    growth factor-α fibroblasts,
    keratinocytes
    Amphiregulin Amphiregulin Mitogen for 0
    fibroblasts,
    keratinocytes
    KGF Keratinocyte Mitogen for ++
    growth factor keratinocytes
    HBEGF Heparin- Mitogen for +++
    binding fibroblasts,
    epidermal keratinocytes
    growth factor-
    like growth
    factor
    TGF-β1 Transforming Stimulates +
    growth factor- matrix
    β1 deposition
    TGF-β3 Transforming Stimulates ++ − ++
    growth factor- matrix
    β3 deposition
    VEGF Vascular Angiogenic ++
    endothelial factor
    growth factor
    SPARC Secreted Complex anti- ++++
    protein acidic angiogenic,
    and rich in angiogenic
    cysteine
    ICAM-1 Intercellular Lymphocyte +
    adhesion adhesion,
    molecule-1 mobility
    VCAM Vascular Lymphocyte +++
    cellular adhesion,
    adhesion mobility
    molecule
    GAPDh Glyceraldehyde Glycolytic +++
    3-phosphate housekeeping
    dehydrogenase gene
    β2- β2- Antigen +++
    microglobulin microglobulin presentation
  • The invention described and claimed herein is not to be limited in scope by the specific embodiments herein disclosed since these embodiments are intended as illustrations of several aspects of the invention. Any equivalent embodiments are intended to be within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. [0106]
  • A number of references are cited herein, the entire disclosures of which are incorporated herein, in their entirety, by reference. [0107]

Claims (31)

1-29. (cancelled).
30. A composition comprising a three-dimensional framework, said framework coated with a naturally secreted extracellular matrix material composed of human proteins.
31. The composition according to claim 30, wherein the framework is composed of a biodegradable material.
32. The composition according to claim 31, wherein the biodegradable material is cotton, polyglycolic acid, cat gut sutures, cellulose, gelatin, or dextran.
33. The composition according to claim 30, wherein the framework is composed of a non-biodegradable material.
34. The composition according to claim 33, wherein the nonbiodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, or a nitrocellulose compound.
35. The composition according to claim 30, wherein the three-dimensional framework has pore spaces of about 150 μm to about 220 μm.
36. The composition according to claim 30, wherein the extracellular matrix is secreted by human stromal cells.
37. The composition according to claim 36, wherein the stromal cells are fibroblasts.
38. The composition according to claim 36, wherein the stromal cells are found in loose connective tissue or bone marrow.
39. The composition according to claim 38, wherein the stromal cells are endothelial cells, pericytes, macrophages, monocytes, leukocytes, plasma cells, mast cells or adipocytes.
40. An injectable formulation for the treatment of a skin or tissue defect, comprising a cell-free, injectable formulation of naturally secreted human extracellular matrix components synthesized by cells in vitro and a pharmaceutically acceptable carrier formulated for in vivo administration by injection via a syringe.
41. The injectable formulation according to claim 40, in which the matrix is produced by a method comprising:
(a) providing a living stromal tissue prepared in vitro comprising human stromal cells and connective tissue proteins naturally secreted by the stromal cells attached to and substantially enveloping a framework, said framework composed of a biocompatible, non-living material formed into a three-dimensional structure having interstitial spaces bridged by the stromal cells;
(b) killing the cells in the living stromal tissue;
(c) removing the killed cells and any cellular contents from the framework;
(d) collecting the connective tissue proteins naturally secreted by the stromal cells attached to the framework; and
(e) processing the collected connective tissue proteins of step (d) with a pharmaceutically acceptable carrier into a formulation that is suitable for in vivo administration by injection via a syringe.
42. The injectable formulation according to claim 41, in which the collected connective tissue proteins of step (d) are processed by homogenizing, cross-linking, or suspending the collected connective tissue proteins in a physiological acceptable carrier prior to step (e).
43. The injectable formulation according to claim 41 in which the collected connective tissue proteins of step (d) are processed by adjusting ratios of collagen types I-V, respective to each other, prior to step (e).
44. The injectable formulation according to claim 41, in which the stromal cells of the living stromal tissue are fibroblasts.
45. The injectable formulation according to claim 41 in which the stromal cells of the living stromal tissue are cells found in loose connective tissue or bone marrow.
46. The injectable formulation according to claim 45 in which the stromal cells of the living stromal tissue are endothelial cells, pericytes, macrophages, monocytes, leukocytes, plasma cells, mast cells, chondrocytes or adipocytes.
47. The injectable formulation according to claim 41, in which the framework is composed of a biodegradable material.
48. The injectable formulation according to claim 47, in which the biodegradable material is cotton, polyglycolic acid, cat gut sutures, cellulose, gelatin, or dextran.
49. The injectable formulation according to claim 41, in which the framework is composed of a non-biodegradable material.
50. The injectable formulation according to claim 49, in which the non-biodegradable material is a polyamide, a polyester, a polystyrene, a polypropylene, a polyacrylate, a polyvinyl, a polycarbonate, a polytetrafluorethylene, or a nitrocellulose compound.
51. The injectable formulation according to claim 41, in which the framework is a mesh
52. The injectable formulation according to claim 51 in which the mesh has pore spaces of about 150 μm to about 220 μm.
53. The injectable formulation according to claim 41, in which the pharmaceutically acceptable carrier contains an anesthetic agent, a lubricating agent, a tissue growth factor or combinations thereof.
54. The injectable formulation according to claim 40 produced by a method comprising:
(a) providing a living stromal tissue prepared in vitro comprising human stromal cells and connective tissue proteins naturally secreted by the stromal cells attached to and substantially enveloping a framework, said framework composed of a biocompatible, non-living biodegradable material formed into a three-dimensional structure having interstitial spaces bridged by the stromal cells;
(b) killing the cells in the living stromal tissue;
(c) removing the killed cells and any cellular contents from the framework; and
(d) processing the connective tissue proteins naturally secreted by the stromal cells attached to the framework and the biodegradable framework with a pharmaceutically acceptable carrier into a formulation suitable for in vivo administration by injection via a syringe.
55. The injectable formulation according to claim 41, in which the stromal cells recombinantly express a gene product.
56. The injectable formulation according to claim 55, in which expression of the gene product is under the control of an inducible promoter.
57. The injectable formulation according to claim 41, in which steps (b) and (c) are carried out concurrently.
58. The injectable formulation according to claim 54, in which steps (b) and (c) are carried out concurrently.
59. The injectable formulation according to claim 30 in which the cells are grown in vitro on a three-dimensional framework.
US10/851,773 1995-06-06 2004-05-21 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix Abandoned US20040259190A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/851,773 US20040259190A1 (en) 1995-06-06 2004-05-21 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/470,101 US5830708A (en) 1995-06-06 1995-06-06 Methods for production of a naturally secreted extracellular matrix
US66078796A 1996-06-06 1996-06-06
US09/948,379 US20020038152A1 (en) 1995-06-06 2001-09-07 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US10/805,774 US20040180431A1 (en) 1995-06-06 2004-03-22 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US10/851,773 US20040259190A1 (en) 1995-06-06 2004-05-21 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/805,774 Continuation US20040180431A1 (en) 1995-06-06 2004-03-22 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Publications (1)

Publication Number Publication Date
US20040259190A1 true US20040259190A1 (en) 2004-12-23

Family

ID=27042969

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/182,822 Expired - Lifetime US6284284B1 (en) 1995-06-06 1998-10-29 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US09/947,131 Abandoned US20020019339A1 (en) 1995-06-06 2001-09-04 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US09/948,379 Abandoned US20020038152A1 (en) 1995-06-06 2001-09-07 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US10/805,774 Abandoned US20040180431A1 (en) 1995-06-06 2004-03-22 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US10/851,773 Abandoned US20040259190A1 (en) 1995-06-06 2004-05-21 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US09/182,822 Expired - Lifetime US6284284B1 (en) 1995-06-06 1998-10-29 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US09/947,131 Abandoned US20020019339A1 (en) 1995-06-06 2001-09-04 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US09/948,379 Abandoned US20020038152A1 (en) 1995-06-06 2001-09-07 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US10/805,774 Abandoned US20040180431A1 (en) 1995-06-06 2004-03-22 Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Country Status (1)

Country Link
US (5) US6284284B1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9062288B2 (en) 2008-08-22 2015-06-23 Regeneus Ltd Therapeutic methods using adipose tissue-derived cell suspensions comprising adipocytes
WO2018191672A1 (en) * 2017-04-14 2018-10-18 President And Fellows Of Harvard College Methods for generation of cell-derived microfilament network
US10494662B2 (en) 2013-03-12 2019-12-03 President And Fellows Of Harvard College Method for generating a three-dimensional nucleic acid containing matrix
US10501791B2 (en) 2011-10-14 2019-12-10 President And Fellows Of Harvard College Sequencing by structure assembly
US10640826B2 (en) 2012-06-05 2020-05-05 President And Fellows Of Harvard College Spatial sequencing of nucleic acids using DNA origami probes
US11021737B2 (en) 2011-12-22 2021-06-01 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11085072B2 (en) 2016-08-31 2021-08-10 President And Fellows Of Harvard College Methods of generating libraries of nucleic acid sequences for detection via fluorescent in situ sequencing
US11118220B2 (en) 2015-11-03 2021-09-14 President And Fellows Of Harvard College Method and apparatus for volumetric imaging of a three-dimensional nucleic acid containing matrix
US11193163B2 (en) 2018-07-30 2021-12-07 Readcoor, Llc Methods and systems for sample processing or analysis
US11293054B2 (en) 2011-12-22 2022-04-05 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11447807B2 (en) 2016-08-31 2022-09-20 President And Fellows Of Harvard College Methods of combining the detection of biomolecules into a single assay using fluorescent in situ sequencing
US11713485B2 (en) 2016-04-25 2023-08-01 President And Fellows Of Harvard College Hybridization chain reaction methods for in situ molecular detection

Families Citing this family (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6284284B1 (en) * 1995-06-06 2001-09-04 Advanced Tissue Sciences, Inc. Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
WO1998036704A1 (en) * 1997-02-20 1998-08-27 Keller Gregory S Augmentation and repair of dermal, subcutaneous, and vocal cord tissue defects
US7192984B2 (en) * 1997-06-17 2007-03-20 Fziomed, Inc. Compositions of polyacids and polyethers and methods for their use as dermal fillers
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US6485489B2 (en) 1999-10-02 2002-11-26 Quantum Cor, Inc. Catheter system for repairing a mitral valve annulus
US20030069570A1 (en) * 1999-10-02 2003-04-10 Witzel Thomas H. Methods for repairing mitral valve annulus percutaneously
US7229469B1 (en) 1999-10-02 2007-06-12 Quantumcor, Inc. Methods for treating and repairing mitral valve annulus
US9080146B2 (en) 2001-01-11 2015-07-14 Celonova Biosciences, Inc. Substrates containing polyphosphazene as matrices and substrates containing polyphosphazene with a micro-structured surface
KR100527623B1 (en) * 2002-06-01 2005-11-15 라이프코드인터내셔날 주식회사 Biodegradable polymer scaffold containing extracellular matrix used for artificial organs and method for preparing same
CN1671400A (en) * 2002-07-03 2005-09-21 派瑞克科学公司 Compositions of hyaluronic acid and methods of use
WO2004047622A2 (en) 2002-11-26 2004-06-10 Crosscart, Inc. Substantially non-immunogenic injectable collagen
US7655048B2 (en) * 2003-04-02 2010-02-02 Furlow Jr Leonard T Materials and methods for soft tissue augmentation
IL156945A0 (en) * 2003-07-15 2004-02-08 Itzhak Tavori Device and a method for orthopedic delivery of bone reconstruction medium
US20050013870A1 (en) * 2003-07-17 2005-01-20 Toby Freyman Decellularized extracellular matrix of conditioned body tissues and uses thereof
WO2005046445A2 (en) 2003-11-07 2005-05-26 University Of Connecticut Artificial tissue systems and uses thereof
US20070190101A1 (en) * 2004-03-31 2007-08-16 Chunlin Yang Flowable bone grafts
US8377484B1 (en) 2004-05-06 2013-02-19 Maria V. Tsiper Tumor encapsulation for prevention and treatment of metastatic cancer disease
CA2566765C (en) * 2004-05-11 2011-08-23 Synthasome Inc. Hybrid tissue scaffolds containing extracellular matrix
AU2005279934A1 (en) * 2004-08-30 2006-03-09 Theregen, Inc. Compositions and methods of promoting hair growth
US9114162B2 (en) 2004-10-25 2015-08-25 Celonova Biosciences, Inc. Loadable polymeric particles for enhanced imaging in clinical applications and methods of preparing and using the same
US9107850B2 (en) 2004-10-25 2015-08-18 Celonova Biosciences, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
KR101153785B1 (en) 2004-10-25 2012-07-09 셀로노바 바이오사이언시스 저머니 게엠베하 Loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US20210299056A9 (en) 2004-10-25 2021-09-30 Varian Medical Systems, Inc. Color-Coded Polymeric Particles of Predetermined Size for Therapeutic and/or Diagnostic Applications and Related Methods
JP2008546686A (en) * 2005-06-17 2008-12-25 セレゲン,インコーポレーテッド Method for treating ischemic tissue
WO2007064305A1 (en) * 2005-12-01 2007-06-07 Agency For Science, Technology And Research Three-dimensional reconstituted extracellular matrices as scaffolds for tissue engineering
US20070178137A1 (en) * 2006-02-01 2007-08-02 Toby Freyman Local control of inflammation
JP5101489B2 (en) 2006-03-08 2012-12-19 杏林製薬株式会社 Process for producing aminoacetylpyrrolidinecarbonitrile derivative and its production intermediate
WO2008041909A1 (en) 2006-10-02 2008-04-10 Norrfors Searl A method of producing native components, such as growth factors or extracellular matrix proteins, through cell culturing of tissue samples for tissue repair
US8084023B2 (en) 2007-01-22 2011-12-27 The Board Of Trustees Of The University Of Arkansas Maintenance and propagation of mesenchymal stem cells
US8143427B2 (en) * 2007-03-22 2012-03-27 Kyorin Pharmaceutical Co., Ltd. Method for producing aminoacetylpyrrolidinecarbonitrile derivative
WO2009042514A1 (en) * 2007-09-25 2009-04-02 Integra Lifesciences Corporation Flowable wound matrix and its preparation and use
EP2222159B1 (en) 2007-11-20 2018-02-21 Pioneer Surgical Orthobiologics, Inc. Cryopreservation of cells using cross-linked bioactive hydrogel matrix particles
SG10201906494PA (en) 2008-01-30 2019-09-27 Histogen Inc Extracellular matrix compositions
US20090202642A1 (en) * 2008-02-08 2009-08-13 Xiao Huang Drug Delivery System Comprising Microparticles and Gelation System
CA2729576A1 (en) * 2008-07-02 2010-01-07 Allergan, Inc. Compositions and methods for tissue filling and regeneration
US8357795B2 (en) 2008-08-04 2013-01-22 Allergan, Inc. Hyaluronic acid-based gels including lidocaine
JP5476305B2 (en) * 2008-08-07 2014-04-23 杏林製薬株式会社 Method for producing bicyclo [2.2.2] octylamine derivative
JPWO2010018866A1 (en) * 2008-08-14 2012-01-26 杏林製薬株式会社 Stabilized pharmaceutical composition
FR2938187B1 (en) * 2008-11-07 2012-08-17 Anteis Sa INJECTABLE COMPOSITION BASED ON HYALURONIC ACID OR ONE OF ITS HEAT-STERILIZED SALTS, POLYOLS AND LIDOCAINE
US20100297182A1 (en) * 2009-05-20 2010-11-25 Tozzi Donna M Injectable Amino-acid Composition
US9101538B2 (en) 2009-05-20 2015-08-11 Donna M. Tozzi Injectable amino-acid composition
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
CA2810444A1 (en) * 2010-09-07 2012-03-15 The Board Of Regents Of The University Of Texas System Tissue-specific differentiation matrices and uses thereof
NZ612801A (en) 2010-12-27 2014-12-24 Intellicell Biosciences Inc Ultrasonic cavitation derived stromal or mesenchymal vascular extracts and cells derived therefrom obtained from adipose tissue and use thereof
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
US20130178874A1 (en) * 2011-07-19 2013-07-11 Hilton Becker Composite implant
ES2559467T3 (en) 2011-12-23 2016-02-12 Pioneer Surgical Technology, Inc. Continuous matrix with osteoconductive particles dispersed therein, formation procedure, and its use to regenerate the bone with it
US10130288B2 (en) 2013-03-14 2018-11-20 Cell and Molecular Tissue Engineering, LLC Coated sensors, and corresponding systems and methods
US10405961B2 (en) 2013-03-14 2019-09-10 Cell and Molecular Tissue Engineering, LLC Coated surgical mesh, and corresponding systems and methods
WO2014185945A1 (en) * 2013-05-13 2014-11-20 Cormatrix Cardiovascular, Inc. Compositions, methods and systems for regenerative cosmetics
US20150037436A1 (en) 2013-07-30 2015-02-05 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11352606B2 (en) 2014-02-10 2022-06-07 The Johns Hopkins University Low oxygen tension enhances endothelial fate of human pluripotent stem cells
US10004824B2 (en) 2015-05-11 2018-06-26 Laboratoires Vivacy Compositions comprising at least one polyol and at least one anesthetic
EP3297694A1 (en) 2015-05-21 2018-03-28 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US20190184060A1 (en) * 2016-08-19 2019-06-20 Regentys Corporation Extracellular matrix for tissue reconstruction of mucosal tissue
BE1024733B1 (en) 2016-11-09 2018-06-14 Univercells Sa CELL GROWTH MATRIX
EP3921405A1 (en) 2019-02-05 2021-12-15 Corning Incorporated Woven cell culture substrates
CN110638684A (en) * 2019-09-02 2020-01-03 深圳兰度生物材料有限公司 Injectable collagen composition and preparation method thereof
US11118151B2 (en) 2019-11-05 2021-09-14 Corning Incorporated Fixed bed bioreactor and methods of using the same
CN111282022A (en) * 2020-02-14 2020-06-16 昆明医科大学 Method for assisting in constructing collagen ultrathin membrane tissue engineering bone
CN112503731B (en) * 2020-11-25 2022-02-08 珠海格力电器股份有限公司 Method and device for controlling air conditioning equipment and air conditioning equipment

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3949073A (en) * 1974-11-18 1976-04-06 The Board Of Trustees Of Leland Stanford Junior University Process for augmenting connective mammalian tissue with in situ polymerizable native collagen solution
US4424208A (en) * 1982-01-11 1984-01-03 Collagen Corporation Collagen implant material and method for augmenting soft tissue
US4582640A (en) * 1982-03-08 1986-04-15 Collagen Corporation Injectable cross-linked collagen implant material
US4721096A (en) * 1986-04-18 1988-01-26 Marrow-Tech Incorporated Process for replicating bone marrow in vitro and using the same
US4801229A (en) * 1986-07-31 1989-01-31 Nippon Yusen Kaisha and Nippon Fruehauf Company, Ltd. Transportation container
US4803075A (en) * 1986-06-25 1989-02-07 Collagen Corporation Injectable implant composition having improved intrudability
US4837285A (en) * 1984-03-27 1989-06-06 Medimatrix Collagen matrix beads for soft tissue repair
US4963489A (en) * 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US4969912A (en) * 1988-02-18 1990-11-13 Kelman Charles D Human collagen processing and autoimplant use
US5002071A (en) * 1987-04-03 1991-03-26 Research Development Foundation Injectable soft tissue augmentation materials from the placenta and their method of manufacture
US5032508A (en) * 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5160490A (en) * 1986-04-18 1992-11-03 Marrow-Tech Incorporated Three-dimensional cell and tissue culture apparatus
US5204382A (en) * 1992-02-28 1993-04-20 Collagen Corporation Injectable ceramic compositions and methods for their preparation and use
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US5288617A (en) * 1984-10-31 1994-02-22 Commonwealth Scientific And Industrial Research Organization Method of producing an antigenic preparation
US5342765A (en) * 1991-08-01 1994-08-30 Occidental Chemical Corporation Method of producing extracellular products from aerobic microorganisms
US5397352A (en) * 1993-08-27 1995-03-14 Burres; Steven Method of recollagenation
US5460939A (en) * 1986-04-18 1995-10-24 Advanced Tissue Sciences, Inc. Temporary living skin replacement
US5478739A (en) * 1992-10-23 1995-12-26 Advanced Tissue Sciences, Inc. Three-dimensional stromal cell and tissue culture system
US5516532A (en) * 1994-08-05 1996-05-14 Children's Medical Center Corporation Injectable non-immunogenic cartilage and bone preparation
US5541106A (en) * 1993-04-05 1996-07-30 Desmos, Inc. Cell matrix stimulated attachment and hemidesmosome assembly
US5830708A (en) * 1995-06-06 1998-11-03 Advanced Tissue Sciences, Inc. Methods for production of a naturally secreted extracellular matrix
US6284284B1 (en) * 1995-06-06 2001-09-04 Advanced Tissue Sciences, Inc. Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
CA2045222A1 (en) 1990-07-12 1992-01-13 Norman R. Weldon Composite biosynthetic graft
US5428022A (en) * 1992-07-29 1995-06-27 Collagen Corporation Composition of low type III content human placental collagen
US5656478A (en) * 1994-02-25 1997-08-12 The Regents Of The University Of California Smooth muscle tissue formation in vivo using cultured smooth muscle cells combined with an extracellular matrix
US5716394A (en) 1994-04-29 1998-02-10 W. L. Gore & Associates, Inc. Blood contact surfaces using extracellular matrix synthesized in vitro
AU2517395A (en) 1994-05-20 1995-12-18 Vec Tec, Inc. Methods rendering grafts nonthrombogenic and substantially nonimmunogenic
EP0781116A4 (en) 1994-09-12 1999-08-25 Advanced Tissue Sciences Inc Three-dimensional human cell cultures on cardiac valve frameworks and their uses

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3949073A (en) * 1974-11-18 1976-04-06 The Board Of Trustees Of Leland Stanford Junior University Process for augmenting connective mammalian tissue with in situ polymerizable native collagen solution
US4424208A (en) * 1982-01-11 1984-01-03 Collagen Corporation Collagen implant material and method for augmenting soft tissue
US4582640A (en) * 1982-03-08 1986-04-15 Collagen Corporation Injectable cross-linked collagen implant material
US4837285A (en) * 1984-03-27 1989-06-06 Medimatrix Collagen matrix beads for soft tissue repair
US5288617A (en) * 1984-10-31 1994-02-22 Commonwealth Scientific And Industrial Research Organization Method of producing an antigenic preparation
US4721096A (en) * 1986-04-18 1988-01-26 Marrow-Tech Incorporated Process for replicating bone marrow in vitro and using the same
US5460939A (en) * 1986-04-18 1995-10-24 Advanced Tissue Sciences, Inc. Temporary living skin replacement
US5443950A (en) * 1986-04-18 1995-08-22 Advanced Tissue Sciences, Inc. Three-dimensional cell and tissue culture system
US5160490A (en) * 1986-04-18 1992-11-03 Marrow-Tech Incorporated Three-dimensional cell and tissue culture apparatus
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US4803075A (en) * 1986-06-25 1989-02-07 Collagen Corporation Injectable implant composition having improved intrudability
US4801229A (en) * 1986-07-31 1989-01-31 Nippon Yusen Kaisha and Nippon Fruehauf Company, Ltd. Transportation container
US5002071A (en) * 1987-04-03 1991-03-26 Research Development Foundation Injectable soft tissue augmentation materials from the placenta and their method of manufacture
US4963489A (en) * 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5332802A (en) * 1988-02-18 1994-07-26 Autogenesis Technologies, Inc. Human collagen processing and autoimplant use
US4969912A (en) * 1988-02-18 1990-11-13 Kelman Charles D Human collagen processing and autoimplant use
US5032508A (en) * 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5342765A (en) * 1991-08-01 1994-08-30 Occidental Chemical Corporation Method of producing extracellular products from aerobic microorganisms
US5204382A (en) * 1992-02-28 1993-04-20 Collagen Corporation Injectable ceramic compositions and methods for their preparation and use
US5478739A (en) * 1992-10-23 1995-12-26 Advanced Tissue Sciences, Inc. Three-dimensional stromal cell and tissue culture system
US5541106A (en) * 1993-04-05 1996-07-30 Desmos, Inc. Cell matrix stimulated attachment and hemidesmosome assembly
US5397352A (en) * 1993-08-27 1995-03-14 Burres; Steven Method of recollagenation
US5516532A (en) * 1994-08-05 1996-05-14 Children's Medical Center Corporation Injectable non-immunogenic cartilage and bone preparation
US5830708A (en) * 1995-06-06 1998-11-03 Advanced Tissue Sciences, Inc. Methods for production of a naturally secreted extracellular matrix
US6284284B1 (en) * 1995-06-06 2001-09-04 Advanced Tissue Sciences, Inc. Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US20020019339A1 (en) * 1995-06-06 2002-02-14 Advanced Tissue Sciences, Inc. Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US20020038152A1 (en) * 1995-06-06 2002-03-28 Advanced Tissue Sciences, Inc. Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
US20040180431A1 (en) * 1995-06-06 2004-09-16 Naughton Gail K. Compositions and methods for production and use of an injectable naturally secreted extracellular matrix

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9062288B2 (en) 2008-08-22 2015-06-23 Regeneus Ltd Therapeutic methods using adipose tissue-derived cell suspensions comprising adipocytes
US10501791B2 (en) 2011-10-14 2019-12-10 President And Fellows Of Harvard College Sequencing by structure assembly
US11312992B2 (en) 2011-10-14 2022-04-26 President And Fellows Of Harvard College Sequencing by structure assembly
US11293051B2 (en) 2011-12-22 2022-04-05 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11639518B2 (en) 2011-12-22 2023-05-02 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11566276B2 (en) 2011-12-22 2023-01-31 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11566277B2 (en) 2011-12-22 2023-01-31 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11021737B2 (en) 2011-12-22 2021-06-01 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11549136B2 (en) 2011-12-22 2023-01-10 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11111521B2 (en) 2011-12-22 2021-09-07 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11293054B2 (en) 2011-12-22 2022-04-05 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11293052B2 (en) 2011-12-22 2022-04-05 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11473139B2 (en) 2012-06-05 2022-10-18 President And Fellows Of Harvard College Spatial sequencing of nucleic acids using DNA origami probes
US10640826B2 (en) 2012-06-05 2020-05-05 President And Fellows Of Harvard College Spatial sequencing of nucleic acids using DNA origami probes
US11078520B2 (en) 2013-03-12 2021-08-03 President And Fellows Of Harvard College Method for generating a three-dimensional nucleic acid containing matrix
US10494662B2 (en) 2013-03-12 2019-12-03 President And Fellows Of Harvard College Method for generating a three-dimensional nucleic acid containing matrix
US11299767B2 (en) 2013-03-12 2022-04-12 President And Fellows Of Harvard College Method for generating a three-dimensional nucleic acid containing matrix
US11118220B2 (en) 2015-11-03 2021-09-14 President And Fellows Of Harvard College Method and apparatus for volumetric imaging of a three-dimensional nucleic acid containing matrix
US11542554B2 (en) 2015-11-03 2023-01-03 President And Fellows Of Harvard College Method and apparatus for volumetric imaging
US11713485B2 (en) 2016-04-25 2023-08-01 President And Fellows Of Harvard College Hybridization chain reaction methods for in situ molecular detection
US11718874B2 (en) 2016-04-25 2023-08-08 President And Fellows Of Harvard College Hybridization chain reaction methods for in situ molecular detection
US11085072B2 (en) 2016-08-31 2021-08-10 President And Fellows Of Harvard College Methods of generating libraries of nucleic acid sequences for detection via fluorescent in situ sequencing
US11447807B2 (en) 2016-08-31 2022-09-20 President And Fellows Of Harvard College Methods of combining the detection of biomolecules into a single assay using fluorescent in situ sequencing
US11185568B2 (en) 2017-04-14 2021-11-30 President And Fellows Of Harvard College Methods for generation of cell-derived microfilament network
WO2018191672A1 (en) * 2017-04-14 2018-10-18 President And Fellows Of Harvard College Methods for generation of cell-derived microfilament network
US11193163B2 (en) 2018-07-30 2021-12-07 Readcoor, Llc Methods and systems for sample processing or analysis

Also Published As

Publication number Publication date
US20040180431A1 (en) 2004-09-16
US20020019339A1 (en) 2002-02-14
US20020038152A1 (en) 2002-03-28
US6284284B1 (en) 2001-09-04

Similar Documents

Publication Publication Date Title
US6284284B1 (en) Compositions and methods for production and use of an injectable naturally secreted extracellular matrix
AU708090B2 (en) Compositions and methods for naturally secreted extracellular matrix
US5919702A (en) Production of cartilage tissue using cells isolated from Wharton's jelly
Rotter et al. Cartilage reconstruction in head and neck surgery: comparison of resorbable polymer scaffolds for tissue engineering of human septal cartilage
US5041138A (en) Neomorphogenesis of cartilage in vivo from cell culture
EP0641223B1 (en) Biotherapeutic cell-coated microspheres
US5736372A (en) Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
AU715282B2 (en) Method for making and/or repairing cartilage
US20070154462A1 (en) Augmentation and repair of tissue defects with in vitro cultured fibroblasts
WO1999052572A1 (en) Methods and compositions for tissue regeneration
EP1196206A1 (en) Human naturally secreted extracellular matrix-coated device
AU740113B2 (en) Augmentation and repair of dermal, subcutaneous, and vocal cord tissue defects
AU668959C (en) Biotherapeutic cell-coated microspheres
Yannas Regenerative medicine II: clinical and preclinical applications

Legal Events

Date Code Title Description
AS Assignment

Owner name: ADVANCED BIOHEALING, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SMITH & NEPHEW WOUND MANAGEMENT (LA JOLLA);REEL/FRAME:017730/0160

Effective date: 20060525

AS Assignment

Owner name: SMITH & NEPHEW, INC., TENNESSEE

Free format text: SECURITY AGREEMENT;ASSIGNOR:ADVANCED BIOHEALING, INC.;REEL/FRAME:017882/0698

Effective date: 20060525

Owner name: SMITH & NEPHEW, INC. (A DELAWARE CORPORATION), TEN

Free format text: SECURITY AGREEMENT;ASSIGNOR:ADVANCED BIOHEALING, INC.;REEL/FRAME:017882/0698

Effective date: 20060525

Owner name: SMITH & NEPHEW WOUND MANGEMENT (LA JOLLA), ENGLAND

Free format text: SECURITY AGREEMENT;ASSIGNOR:ADVANCED BIOHEALING, INC.;REEL/FRAME:017882/0698

Effective date: 20060525

AS Assignment

Owner name: HORIZON TECHNOLOGY FUNDING COMPANY LLC, CONNECTICU

Free format text: SECURITY AGREEMENT;ASSIGNOR:ADVANCED BIOHEALING, INC.;REEL/FRAME:018806/0123

Effective date: 20061221

AS Assignment

Owner name: ADVANCED BIOHEALING, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:HORIZON TECHNOLOGY FUNDING COMPANY LLC;REEL/FRAME:019193/0646

Effective date: 20070321

STCB Information on status: application discontinuation

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