USRE42479E1 - Engineering of strong, pliable tissues - Google Patents

Engineering of strong, pliable tissues Download PDF

Info

Publication number
USRE42479E1
USRE42479E1 US10/782,750 US78275004A USRE42479E US RE42479 E1 USRE42479 E1 US RE42479E1 US 78275004 A US78275004 A US 78275004A US RE42479 E USRE42479 E US RE42479E
Authority
US
United States
Prior art keywords
matrix
cells
cell
seeded
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.)
Expired - Lifetime
Application number
US10/782,750
Inventor
Joseph P. Vacanti
Christopher K. Breuer
Berverly E. Chaignaud
Toshiraru Shin'oka
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.)
Childrens Medical Center Corp
Original Assignee
Childrens Medical Center Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Childrens Medical Center Corp filed Critical Childrens Medical Center Corp
Priority to US10/782,750 priority Critical patent/USRE42479E1/en
Priority to US11/529,691 priority patent/USRE42575E1/en
Application granted granted Critical
Publication of USRE42479E1 publication Critical patent/USRE42479E1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3886Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells comprising two or more cell types
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/18Macromolecular materials obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials 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
    • A61L27/3641Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the site of application in the body
    • A61L27/3645Connective tissue
    • 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
    • A61L27/3843Connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/507Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials for artificial blood vessels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61F2/24Heart valves ; Vascular valves, e.g. venous valves; Heart implants, e.g. passive devices for improving the function of the native valve or the heart muscle; Transmyocardial revascularisation [TMR] devices; Valves implantable in the body
    • A61F2/2412Heart valves ; Vascular valves, e.g. venous valves; Heart implants, e.g. passive devices for improving the function of the native valve or the heart muscle; Transmyocardial revascularisation [TMR] devices; Valves implantable in the body with soft flexible valve members, e.g. tissue valves shaped like natural valves
    • A61F2/2415Manufacturing methods

Definitions

  • This invention is generally in the field of reconstruction and augmentation of flexible, strong connective tissue such as arteries and heart valves.
  • Tissue engineering is a multidisciplinary science that utilizes basic principles from the life sciences and engineering sciences to create cellular constructs for transplantation.
  • the first attempts to culture cells on a matrix for use as artificial skin, which requires formation of a thin three dimensional structure, were described by Yannas and Bell (See, for example, U.S. Pat. Nos. 4,060,081, 4,485,097 and 4,458,678). They used collagen type structures which were seeded with cells, then placed over the denuded area.
  • a problem with the use of the collagen matrices was that the rate of degradation is not well controlled.
  • Another problem was that cells implanted into the interior of thick pieces of the collagen matrix failed to survive.
  • Vacanti and Langer recognized that there was a need to have two elements in any matrix used to form organs: adequate structure and surface area to implant a large volume of cells into the body to replace lost function and a matrix formed in a way that allowed adequate diffusion of gases and nutrients throughout the matrix as the cells attached and grew to maintain viability in the absence of vascularization. Once implanted and vascularized, the porosity required for diffusion of the nutrients and gases was no longer critical.
  • WO 93/08850 Prevascularized Polymeric Implants for Organ Transplantation
  • Massachusetts Institute of Technology and Children's Medical Center Corporation disclosed implantation of relatively rigid, non-compressible porous matrices which are allowed to become vascularized, then seeded with cells. It was difficult to control the extent of ingrowth of fibrous tissue, however, and to obtain uniform distribution of cells throughout the matrix when they were subsequently injected into the matrix.
  • connective tissue such as bone and cartilage
  • soft tissue such as hepatocytes, intestine, endothelium
  • specific structures such as ureters.
  • Valve replacement is the state-of-the art therapy for end-stage valve disease.
  • Heart valve replacement with either nonliving xenografts or mechanical protheses is an effective therapy for valvular heart disease.
  • both types of heart valve replacements have limitations, including finite durability, foreign body reaction or rejection and the inability of the non-living structures to grow, repair and remodel, as well as the necessity of life-long anticoagulation for the mechanical prothesis.
  • the construction of a tissue engineered living heart valve could eliminate these problems.
  • Tissue engineered blood vessels may offer a substitute for small caliber vessels for bypass surgery and replacement of diseased vessels.
  • the matrices must have sufficient surface area and exposure to nutrients such that cellular growth and differentiation can occur prior to the ingrowth of blood vessels following implantation. This is not a limiting feature where the matrix is implanted and ingrowth of tissue from the body occurs, prior to seeding of the matrix with dissociated cells.
  • the organization of the tissue may be regulated by the microstructure of the matrix. Specific pore sizes and structures may be utilized to control the pattern and extent of fibrovascular tissue ingrowth from the host, as well as the organization of the implanted cells.
  • the surface geometry and chemistry of the matrix may be regulated to control the adhesion, organization, and function of implanted cells or host cells.
  • the matrix should be a pliable, non-toxic, injectable porous template for vascular ingrowth.
  • the pores should allow vascular ingrowth and the injection of cells in a desired density and region(s) of the matrix without damage to the cells. These are generally interconnected pores in the range of between approximately 100 and 300 microns.
  • the matrix should be shaped to maximize surface area, to allow adequate diffusion of nutrients and growth factors to the cells and to allow the ingrowth of new blood vessels and connective tissue.
  • the overall, or external, matrix configuration is dependent on the tissue which is to reconstructed or augmented.
  • the shape can also be obtained using struts, as described below, to impart resistance to mechanical forces and thereby yield the desired shape. Examples include heart valve “leaflets” and tubes.
  • bioerodible or “biodegradable”, as used herein refers to materials which are enzymatically or chemically degraded in vivo into simpler chemical species. Either natural or synthetic polymers can be used to form the matrix, although synthetic biodegradable polymers are preferred for reproducibility and controlled release kinetics.
  • Synthetic polymers that can be used include bioerodible polymers such as poly(lactide) (PLA), poly(glycolic acid) (PGA), poly(lactide-co-glycolide) (PLGA), poly(caprolactone), polycarbonates, polyamides, polyanhydrides, polyamino acids, polyortho esters, polyacetals, polycyanoacrylates and degradable polyurethanes, and non-erodible polymers such as polyacrylates, ethylene-vinyl acetate polymers and other acyl substituted cellulose acetates and derivatives thereof, non-erodible polyurethanes, polystyrenes, polyvinyl chloride, polyvinyl fluoride, poly(vinyl imidazole), chloro-sulphonated polyolifins, polyethylene oxide, polyvinyl alcohol, teflon®, and nylon.
  • bioerodible polymers such as poly(lactide) (PLA), poly(glycolic
  • non-degradable materials can be used to form the matrix or a portion of the matrix, they are not preferred.
  • the preferred non-degradable material for implantation of a matrix which is prevascularized prior to implantation of dissociated cells is a polyvinyl alcohol sponge, or alkylation, and acylation derivatives thereof, including esters.
  • a non-absorbable polyvinyl alcohol sponge is available commercially as IvalonTM, from Unipoint Industries. Methods for making this material are described in U.S. Pat. Nos.
  • natural polymers include proteins such as albumin, collagen, synthetic polyamino acids, and prolamines, and polysaccharides such as alginate, heparin, and other naturally occurring biodegradable polymers of sugar units. These are not preferred because of difficulty with quality control and lack of reproducible, defined degradation characteristics.
  • PLA, PGA and PLA/PGA copolymers are particularly useful for forming the biodegradable matrices. These are synthetic, biodegradable ⁇ -hydroxy acids with a long history of medical use.
  • PLA polymers are usually prepared from the cyclic esters of lactic acids. Both L(+) and D( ⁇ ) forms of lactic acid can be used to prepare the PLA polymers, as well as the optically inactive DL-lactic acid mixture of D( ⁇ ) and L(+) lactic acids. Methods of preparing polylactides are well documented in the patent literature. The following U.S.
  • PGA is the homopolymer of glycolic acid (hydroxyacetic acid).
  • glycolic acid hydroxyacetic acid
  • glycolic acid is initially reacted with itself to form the cyclic ester glycolide, which in the presence of heat and a catalyst is converted to a high molecular weight linear-chain polymer.
  • PGA polymers and their properties are described in more detail in Cyanamid Research Develops World's First Synthetic Absorbable Suture”, Chemistry and Industry, 905 (1970).
  • the erosion of the matrix is related to the molecular weights of the polymer, for example, PLA, PGA or PLA/PGA.
  • a preferred material is poly(lactide-co-glycolide) (50:50), which degrades in about six weeks following implantation (between one and two months) and poly(glycolic acid).
  • All polymers for use in the matrix must meet the mechanical and biochemical parameters necessary to provide adequate support for the cells with subsequent growth and proliferation.
  • the polymers can be characterized with respect to mechanical properties such as tensile strength using an Instron tester, for polymer molecular weight by gel permeation chromatography (GPC), glass transition temperature by differential scanning calorimetry (DSC) and bond structure by infrared (IR) spectroscopy, with respect to toxicology by initial screening tests involving Ames assays and in vitro teratogenicity assays, and implantation studies in animals for immunogenicity, inflammation, release and degradation studies.
  • GPC gel permeation chromatography
  • DSC differential scanning calorimetry
  • IR infrared
  • attachment of the cells to the polymer is enhanced by coating the polymers with compounds such as basement membrane components, agar, agarose, gelatin, gum arabic, collagens types I, II, III, IV, and V, fibronectin, laminin, glycosaminoglycans, polyvinyl alcohol, mixtures thereof, and other hydrophilic and peptide attachment materials known to those skilled in the art of cell culture.
  • a preferred material for coating the polymeric matrix is polyvinyl alcohol or collagen.
  • struts can be biodegradable or non-degradable polymers which are inserted to form a more defined shape than is obtained using the cell-matrices.
  • An analogy can be made to a corset, with the struts acting as “stays” to push the surrounding tissue and skin up and away from the implanted cells.
  • the struts are implanted prior to or at the time of implantation of the cell-matrix structure.
  • the struts are formed of a polymeric material of the same type as can be used to form the matrix, as listed above, having sufficient strength to resist the necessary mechanical forces.
  • the bioactive factors are growth factors, angiogenic factors, compounds selectively inhibiting ingrowth of fibroblast tissue such as antiinflammatories, and compounds selectively inhibiting growth and proliferation of transformed (cancerous) cells. These factors may be utilized to control the growth and function of implanted cells, the ingrowth of blood vessels into the forming tissue, and/or the deposition and organization of fibrous tissue around the implant.
  • growth factors examples include heparin binding growth factor (hbgf), transforming growth factor alpha or beta (TGF ⁇ ), alpha fibroblastic growth factor (FGF), epidermal growth factor (TGF), vascular endothelium growth factor (VEGF), some of which are also angiogenic factors.
  • Other factors include hormones such as insulin, glucagon, and estrogen.
  • NGF nerve growth factor
  • MMP muscle morphogenic factor
  • Steroidal antiinflammatories can be used to decrease inflammation to the implanted matrix, thereby decreasing the amount of fibroblast tissue growing into the matrix.
  • Bioactive molecules can be incorporated into the matrix and released over time by diffusion and/or degradation of the matrix, they can be suspended with the cell suspension, they can be incorporated into microspheres which are suspended with the cells or attached to or incorporated within the matrix, or some combination thereof.
  • Microspheres would typically be formed of materials similar to those forming the matrix, selected for their release properties rather than structural properties. Release properties can also be determined by the size and physical characteristics of the microspheres.
  • Cells are preferably autologous cells, obtained by biopsy and expanded in culture, although cells from close relatives or other donors of the same species may be used with appropriate immunosuppression.
  • Immunologically inert cells such as embryonic or fetal cells, stem cells, and cells genetically engineered to avoid the need for immunosuppression can also be used. Methods and drugs for immunosuppression are known to those skilled in the art of transplantation.
  • a preferred compound is cyclosporin using the recommended dosages.
  • cells are obtained by biopsy and expanded in culture for subsequent implantation.
  • Cells can be easily obtained through a biopsy anywhere in the body, for example, skeletal muscle biopsies can be obtained easily from the arm, forearm, or lower extremities, and smooth muscle can be obtained from the area adjacent to the subcutaneous tissue throughout the body.
  • the area to be biopsied can be locally anesthetized with a small amount of lidocaine injected subcutaneously.
  • a small patch of lidocaine jelly can be applied over the area to be biopsied and left in place for a period of 5 to 20 minutes, prior to obtaining biopsy specimen.
  • the biopsy can be effortlessly obtained with the use of a biopsy needle, a rapid action needle which makes the procedure extremely simple and almost painless. With the addition of the anesthetic agent, the procedure would be entirely painless.
  • This small biopsy core of either skeletal or smooth muscle can then be transferred to media consisting of phosphate buffered saline.
  • the biopsy specimen is then transferred to the lab where the muscle can be grown utilizing the explant technique, wherein the muscle is divided into very pieces which are adhered to culture plate, and serum containing media is added.
  • the muscle biopsy can be enzymatically digested with agents such as trypsin and the cells dispersed in a culture plate with any of the routinely used medias. After cell expansion within the culture plate, the cells can be easily passaged utilizing the usual technique until an adequate number of cells is achieved.
  • the present method uses the recipient or an animal as the initial bioreactor to form a fibrous tissue-polymeric construct which optionally can be seeded with other cells and implanted.
  • the matrix becomes infiltrated with fibrous tissue and/or blood vessels over a period ranging from between one day and a few weeks, most preferably one and two weeks.
  • the matrix is then removed and implanted at the site where it is needed.
  • the matrix is formed of polymer fibers having a particular desired shape, that is implanted subcutaneously.
  • the implant is retrieved surgically, then one or more defined cell types distributed onto and into the fibers.
  • the matrix is seeded with cells of a defined type, implanted until fibrous tissue has grown into the matrix, then the matrix removed, optionally cultured further in vitro, then reimplanted at a desired site.
  • the resulting structures are dictated by the matrix construction, including architecture, porosity (% void volume and pore diameter), polymer nature including composition, crystallinity, molecular weight, and degradability, hydrophobicity, and the inclusion of other biologically active molecules.
  • valves are heart valves and valves of the type used for ventricular shunts for treatment of hydrocephaly.
  • a similar structure could be used for an ascites shunt in the abdomen where needed due to liver disease or in the case of a lymphatic obstructive disease.
  • tubular structures include blood vessels, intestine, ureters, and fallopian tubes.
  • the structures are formed at a site other than where they are ultimately required. This is particularly important in the case of tubular structures and valves, where integrity to fluid is essential, and where the structure is subjected to repeated stress and strain.
  • Valvular heart disease is a significant cause of morbidity and mortality. Construction of a tissue engineered valve using living autologous cells offers advantages over currently used mechanical or glutaraldehyde fixed xenograft valves.
  • a PGA mesh (density 76.9 mg/ml and thickness 0.68 mm) was seeded with the mixed cell population and grown in culture. When the myofibroblasts reached confluence, endothelial cells were seeded onto the surface of the fibroblast/mesh constructs and grown into a single monolayer.
  • vascular smooth muscle tubular structures using a biodegradable polyglycolic acid polymer scaffold have been developed.
  • the technique involves the isolation and culture of vascular smooth muscle cells, the reconstruction of a vascular wall using biodegradable polymer, and formation of the neo-tissue tubes in vitro.
  • the feasibility of engineering vascular structures by coculturing endothelial cells with fibroblasts and smooth muscle cells on a synthetic biodegradable matrix in order to create tubular constructs which histologically resemble native vascular structures was also demonstrated.
  • PGA centimeter polyglycolic acid
  • PGA Polyglycolic acid
  • Bovine periosteum harvested from fresh calf limbs, was placed either directly onto PGA polymer (1 ⁇ 1 cm) or onto tissue culture dishes for periosteal cell isolation.
  • the periosteum/PGA construct was cultured for one week in MEM 199 culture media with antibiotics and ascorbic acid, then implanted into the dorsal subcutaneous space of nude mice.
  • Periosteal cell cultured from pieces of periosteum for two weeks, were isolated into cell suspension and seeded (approximately 1 to 3 ⁇ 10 7 cells) onto PGA polymer (1 ⁇ 1 cm); after one week in culture, the periosteal cell seeded polymer was implanted into the subcutaneous space of nude mice. Specimens, harvested at 4, 8, and 14 week intervals, were evaluated grossly and histologically.
  • the periosteum/PGA constructs showed an organized cartilage matrix with early evidence of bone formation at four weeks, a mixture of bone and cartilage at 8 weeks, and a complete bone matrix at 14 weeks. Constructs created from periosteal cells seeded onto polymer showed presence of disorganized cartilage at 4 and 8 weeks, and a mixture of bone and cartilage at 14 weeks. Periosteum placed directly onto polymer will form an organized cartilage and bone matrix earlier than constructs formed from periosteal cell seeded polymer. This data indicates that PGA is an effective scaffold for periosteal cell attachment and migration to produce bone, which may offer new approaches to reconstructive surgery.
  • the aim of this study was to determine if new vascularized bone could be engineered by transplantation of osteoblast around existing vascular pedicle using biodegradable polymers as cell delivery devices, to be used to reconstruct weight bearing bony defects.
  • Osteoblast and chondryocytes were isolated from calf periosteum and articular cartilage, cultured in vitro for three weeks, then seeded onto a 1 ⁇ 1 cm non-woven polyglycolic acid (PGA) mesh. After maintenance in vitro for one week, cell-polymer constructs were wrapped around saphenous vessels, and implanted into athymic rats for 8 weeks. The implants showed gross and histological evidence of vascularized bone or cartilage. At this time, bilateral 0.8 cm femoral shaft defect were created in the same rat, and fixed in position with a 3 cm craniofacial titanium miniplate. The new engineered bone/cartilage construct was then transferred to the femoral defect on its bilateral vascular pedicle.
  • PGA polyglycolic acid
  • a total of 30 femoral defects were repaired in three groups of animals (each group composed of five animals with defects).
  • Animals in Group 1 received implants composed of vascularized bone constructs, animals in Group 2 with vascular cartilage constructs, and Group 3 animals with blank polymer only.
  • Group 1 defect showed evidence of new bone formation around the defect. Neither Group 2 nor Group 3 defect showed any radiographic evidence of healing or bone formation. Grossly, Group 1 animals developed exuberant bony callus formation and healing of the defect. The animals in Group 2 showed filling of the bony defect with cartilaginous tissue, whereas all of the animals in Group 3 either developed a fibrous non-union or simple separation of both bony fragments with soft tissue invasion of the defect. The histological studies showed new bone formation in all Group 1 animals, new cartilage formation in all Group 2 animals, and fibrous tissue invasion in all Group 3 animals.
  • All implants formed into cylindrical shapes, flattened at the ends.
  • the central portion of the implant formed into a bony matrix and the ends of the specimens formed into cartilage, approximately where the periosteum and chondrocytes were placed. Histological sections showed an organized matrix of bone and cartilage with a distinct transition between bone and cartilage.
  • periosteum and chondrocytes placed onto a biodegradable polymer will form into a composite tissue of bone and cartilage. Moreoever, bone and cartilage composite formation with selective placement of periosteum and chondrocytes on a biodegradable polymer scaffold was shown.
  • a PGA mesh as described in Example 1 or 2 was implanted subcutaneously in an animal, then removed after a period of one to two weeks. Fibroblasts migrated into the polymeric mesh while it was implanted. The implant was then seeded with other cells such as chondrocytes or endothelial cells and cultured in vitro for an additional period of time.
  • the resulting implant was shown to have greater mechanical strength and pliability than implants formed solely by seeding of dissociated cells.

Abstract

It has been discovered that improved yields of engineered tissue following implantation, and engineered tissue having enhanced mechanical strength and flexibility or pliability, can be obtained by implantation, preferably subcutaneously, of a fibrous polymeric matrix for a period of time sufficient to obtain ingrowth of fibrous tissue and/or blood vessels, which is the removed for subsequent implantation at the site where the implant is desired. The matrix is optionally seeded prior to the first implantation, after ingrowth of the fibrous tissue, or at the time of reimplantation. The time required for fibrous ingrowth typically ranges from days to weeks. The method is particularly useful in making valves and tubular structures, especially heart valves and blood vessels.

Description

RELATED APPLICATIONS
This is a divisional of U.S. Ser. No. 08/445,280 filed May 19, 1995 now U.S. Pat. No. 5,855,610.
BACKGROUND OF THE INVENTION
This invention is generally in the field of reconstruction and augmentation of flexible, strong connective tissue such as arteries and heart valves.
Tissue engineering is a multidisciplinary science that utilizes basic principles from the life sciences and engineering sciences to create cellular constructs for transplantation. The first attempts to culture cells on a matrix for use as artificial skin, which requires formation of a thin three dimensional structure, were described by Yannas and Bell (See, for example, U.S. Pat. Nos. 4,060,081, 4,485,097 and 4,458,678). They used collagen type structures which were seeded with cells, then placed over the denuded area. A problem with the use of the collagen matrices was that the rate of degradation is not well controlled. Another problem was that cells implanted into the interior of thick pieces of the collagen matrix failed to survive.
U.S. Pat. No. 4,520,821 to Schmidt describes the use of synthetic polymeric meshes to form linings to repair defects in the urinary tract. Epithelial cells were implanted onto the synthetic matrices, which formed a new tubular lining as the matrix degraded. The matrix served a two fold purpose—to retain liquid while the cells replicated, and to hold and guide the cells as they replicated.
In European Patent Application No. 88900726.6 “Chimeric Neomorphogenesis of Organs by Controlled Cellular Implantation Using Artificial Matrices” by Children's Hospital Center Corporation and Massachusetts Institute of Technology, a method of culturing dissociated cells on biocompatible, biodegradable matrices for subsequent implantation into the body was described. This method was designed to overcome a major problem with previous attempts to culture cells to form three dimensional structures having a diameter of greater than that of skin. Vacanti and Langer recognized that there was a need to have two elements in any matrix used to form organs: adequate structure and surface area to implant a large volume of cells into the body to replace lost function and a matrix formed in a way that allowed adequate diffusion of gases and nutrients throughout the matrix as the cells attached and grew to maintain viability in the absence of vascularization. Once implanted and vascularized, the porosity required for diffusion of the nutrients and gases was no longer critical.
To overcome some of the limitations inherent in the design of the porous structures which support cell growth throughout the matrix solely by diffusion, WO 93/08850 “Prevascularized Polymeric Implants for Organ Transplantation” by Massachusetts Institute of Technology and Children's Medical Center Corporation disclosed implantation of relatively rigid, non-compressible porous matrices which are allowed to become vascularized, then seeded with cells. It was difficult to control the extent of ingrowth of fibrous tissue, however, and to obtain uniform distribution of cells throughout the matrix when they were subsequently injected into the matrix.
Many tissues have now been engineered using these methods, including connective tissue such as bone and cartilage, as well as soft tissue such as hepatocytes, intestine, endothelium, and specific structures, such as ureters. There remains a need to improve the characteristic mechanical and physical properties of the resulting tissues, which in some cases does not possess the requisite strength and pliability to perform its necessary function in vivo. Examples of particular structures include heart valves and blood vessels.
Despite major advances in its treatment over the past thirty-five years, valvular heart disease is still a major cause of morbidity and mortality in the United States. Each year 10,000 Americans die as a direct result of this problem. Valve replacement is the state-of-the art therapy for end-stage valve disease. Heart valve replacement with either nonliving xenografts or mechanical protheses is an effective therapy for valvular heart disease. However, both types of heart valve replacements have limitations, including finite durability, foreign body reaction or rejection and the inability of the non-living structures to grow, repair and remodel, as well as the necessity of life-long anticoagulation for the mechanical prothesis. The construction of a tissue engineered living heart valve could eliminate these problems.
Atherosclerosis and cardiovascular disease are also major causes of morbidity and mortality. More than 925,000 Americans died from heart and blood vessels disease in 1992, and an estimated 468,000 coronary artery bypass surgeries were performed on 393,000 patients. This does not include bypass procedures for peripheral vascular disease. Currently, internal mammary and saphenous vein grafts are the most frequently used native grafts for coronary bypass surgery. However, with triple and quadruple bypasses and often the need for repeat bypass procedures, sufficient native vein grafts can be a problem. Surgeons must frequently look for vessels other than the internal mammary and saphenous vessels. While large diameter (0.5 mm internal diameter) vascular grafts of dacron or polytetraflorethylene (PTFE) have been successful, small caliber synthetic vascular grafts frequently do not remain patent over time. Tissue engineered blood vessels may offer a substitute for small caliber vessels for bypass surgery and replacement of diseased vessels.
It is therefore an object of the present invention to provide a method for making tissue engineered constructs which have improved mechanical strength and flexibility.
It is a further object of the present invention to provide a method and materials for making valves and vessels which can withstand repeated stress and strain.
It is another object of the present invention to provide a method improving yields of engineered tissues following implantation.
SUMMARY OF THE INVENTION
It has been discovered that improved yields of engineered tissue following implantation, and engineered tissue having enhanced mechanical strength and flexibility or pliability, can be obtained by implantation, preferably subcutaneously, of a fibrous polymeric matrix for a period of time sufficient to obtain ingrowth of fibrous tissue and/or blood vessels, which is then removed for subsequent implantation at the site where the implant is desired. The matrix is optionally seeded prior to the first implantation, after ingrowth of the fibrous tissue, or at the time of reimplantation. The time required for fibrous ingrowth typically ranges from days to weeks. The method is particularly useful in making valves and tubular structures, especially heart valves and blood vessels.
Examples demonstrate construction of blood vessels, heart valves and bone and cartilage composite structures.
DETAILED DESCRIPTION OF THE INVENTION
As described herein, structures are created by seeding of fibrous or porous polymeric matrices with dissociated cells which are useful for a variety of applications, ranging from soft tissues formed of parenchymal cells such as hepatocytes, to tissues having structural elements such as heart valves and blood vessels, to cartilage and bone. In a particular improvement over the prior art methods, the polymeric matrices are implanted into a human or animal to allow ingrowth of fibroblastic tissue, then implanted at the site where the structure is needed, either alone or seeded with defmed defined cell populations.
I. Matrix Fabrication
The synthetic matrix serves several purposes. It functions as a cell delivery system that enables the organized transplantation of large numbers of cells into the body. The matrix acts as a scaffold providing three-dimensional space for cell growth. The matrix functions as a template providing structural cues for tissue development. In the case of tissues have specific requirements for structure and mechanical strength, the polymer temporarily provides the biomechanical properties of the final construct, giving the cells time to lay down their own extracellular matrix which ultimately is responsible for the biomechanical profile of the construct. The scaffold also determines the limits of tissue growth and thereby determines the ultimate shape of tissue engineered construct. Cells implanted on a matrix proliferate only to the edges of the matrix; not beyond.
Matrix Architecture
As previously described, for a tissue to be constructed, successfully implanted, and function, the matrices must have sufficient surface area and exposure to nutrients such that cellular growth and differentiation can occur prior to the ingrowth of blood vessels following implantation. This is not a limiting feature where the matrix is implanted and ingrowth of tissue from the body occurs, prior to seeding of the matrix with dissociated cells.
The organization of the tissue may be regulated by the microstructure of the matrix. Specific pore sizes and structures may be utilized to control the pattern and extent of fibrovascular tissue ingrowth from the host, as well as the organization of the implanted cells. The surface geometry and chemistry of the matrix may be regulated to control the adhesion, organization, and function of implanted cells or host cells.
In the preferred embodiment, the matrix is formed of polymers having a fibrous structure which has sufficient interstitial spacing to allow for free diffusion of nutrients and gases to cells attached to the matrix surface. This spacing is typically in the range of 100 to 300 microns, although closer spacings can be used if the matrix is implanted, blood vessels allowed to infiltrate the matrix, then the cells are seeded into the matrix. As used herein, “fibrous” includes one or more fibers that is entwined with itself, multiple fibers in a woven or non-woven mesh, and sponge like devices.
The matrix should be a pliable, non-toxic, injectable porous template for vascular ingrowth. The pores should allow vascular ingrowth and the injection of cells in a desired density and region(s) of the matrix without damage to the cells. These are generally interconnected pores in the range of between approximately 100 and 300 microns. The matrix should be shaped to maximize surface area, to allow adequate diffusion of nutrients and growth factors to the cells and to allow the ingrowth of new blood vessels and connective tissue.
The overall, or external, matrix configuration is dependent on the tissue which is to reconstructed or augmented. The shape can also be obtained using struts, as described below, to impart resistance to mechanical forces and thereby yield the desired shape. Examples include heart valve “leaflets” and tubes.
Polymers
The term “bioerodible”, or “biodegradable”, as used herein refers to materials which are enzymatically or chemically degraded in vivo into simpler chemical species. Either natural or synthetic polymers can be used to form the matrix, although synthetic biodegradable polymers are preferred for reproducibility and controlled release kinetics. Synthetic polymers that can be used include bioerodible polymers such as poly(lactide) (PLA), poly(glycolic acid) (PGA), poly(lactide-co-glycolide) (PLGA), poly(caprolactone), polycarbonates, polyamides, polyanhydrides, polyamino acids, polyortho esters, polyacetals, polycyanoacrylates and degradable polyurethanes, and non-erodible polymers such as polyacrylates, ethylene-vinyl acetate polymers and other acyl substituted cellulose acetates and derivatives thereof, non-erodible polyurethanes, polystyrenes, polyvinyl chloride, polyvinyl fluoride, poly(vinyl imidazole), chloro-sulphonated polyolifins, polyethylene oxide, polyvinyl alcohol, teflon®, and nylon. Although non-degradable materials can be used to form the matrix or a portion of the matrix, they are not preferred. The preferred non-degradable material for implantation of a matrix which is prevascularized prior to implantation of dissociated cells is a polyvinyl alcohol sponge, or alkylation, and acylation derivatives thereof, including esters. A non-absorbable polyvinyl alcohol sponge is available commercially as Ivalon™, from Unipoint Industries. Methods for making this material are described in U.S. Pat. Nos. 2,609,347 to Wilson; 2,653,917 to Hammon, 2,659,935 to Hammon, 2,664,366 to Wilson, 2,664,367 to Wilson, and 2,846,407 to Wilson, the teachings of which are incorporated by reference herein. These materials are all commercially available.
Examples of natural polymers include proteins such as albumin, collagen, synthetic polyamino acids, and prolamines, and polysaccharides such as alginate, heparin, and other naturally occurring biodegradable polymers of sugar units. These are not preferred because of difficulty with quality control and lack of reproducible, defined degradation characteristics.
PLA, PGA and PLA/PGA copolymers are particularly useful for forming the biodegradable matrices. These are synthetic, biodegradable α-hydroxy acids with a long history of medical use. PLA polymers are usually prepared from the cyclic esters of lactic acids. Both L(+) and D(−) forms of lactic acid can be used to prepare the PLA polymers, as well as the optically inactive DL-lactic acid mixture of D(−) and L(+) lactic acids. Methods of preparing polylactides are well documented in the patent literature. The following U.S. Patents, the teachings of which are hereby incorporated by reference, describe in detail suitable polylactides, their properties and their preparation: 1,995,970 to Dorough; 2,703,316 to Schneider; 2,758,987 to Salzberg; 2,951,828 to Zeile; 2,676,945 to Higgins; and 2,683,136; 3,531,561 to Trehu.
PGA is the homopolymer of glycolic acid (hydroxyacetic acid). In the conversion of glycolic acid to poly(glycolic acid), glycolic acid is initially reacted with itself to form the cyclic ester glycolide, which in the presence of heat and a catalyst is converted to a high molecular weight linear-chain polymer. PGA polymers and their properties are described in more detail in Cyanamid Research Develops World's First Synthetic Absorbable Suture”, Chemistry and Industry, 905 (1970).
The erosion of the matrix is related to the molecular weights of the polymer, for example, PLA, PGA or PLA/PGA. The higher molecular weights, weight average molecular weights of 90,000 or higher, result in polymer matrices which retain their structural integrity for longer periods of time; while lower molecular weights, weight average molecular weights of 30,000 or less, result in both slower release and shorter matrix lives. A preferred material is poly(lactide-co-glycolide) (50:50), which degrades in about six weeks following implantation (between one and two months) and poly(glycolic acid).
All polymers for use in the matrix must meet the mechanical and biochemical parameters necessary to provide adequate support for the cells with subsequent growth and proliferation. The polymers can be characterized with respect to mechanical properties such as tensile strength using an Instron tester, for polymer molecular weight by gel permeation chromatography (GPC), glass transition temperature by differential scanning calorimetry (DSC) and bond structure by infrared (IR) spectroscopy, with respect to toxicology by initial screening tests involving Ames assays and in vitro teratogenicity assays, and implantation studies in animals for immunogenicity, inflammation, release and degradation studies.
Polymer Coatings
In some embodiments, attachment of the cells to the polymer is enhanced by coating the polymers with compounds such as basement membrane components, agar, agarose, gelatin, gum arabic, collagens types I, II, III, IV, and V, fibronectin, laminin, glycosaminoglycans, polyvinyl alcohol, mixtures thereof, and other hydrophilic and peptide attachment materials known to those skilled in the art of cell culture. A preferred material for coating the polymeric matrix is polyvinyl alcohol or collagen.
Struts
In some embodiments it may be desirable to create additional structure using devices provided for support, referred to herein as “struts”. These can be biodegradable or non-degradable polymers which are inserted to form a more defined shape than is obtained using the cell-matrices. An analogy can be made to a corset, with the struts acting as “stays” to push the surrounding tissue and skin up and away from the implanted cells. In a preferred embodiment, the struts are implanted prior to or at the time of implantation of the cell-matrix structure. The struts are formed of a polymeric material of the same type as can be used to form the matrix, as listed above, having sufficient strength to resist the necessary mechanical forces.
Additives to Polymer Matrices
In some embodiments it may be desirable to add bioactive molecules to the cells. A variety of bioactive molecules can be delivered using the matrices described herein. These are referred to generically herein as “factors” or “bioactive factors”.
In the preferred embodiment, the bioactive factors are growth factors, angiogenic factors, compounds selectively inhibiting ingrowth of fibroblast tissue such as antiinflammatories, and compounds selectively inhibiting growth and proliferation of transformed (cancerous) cells. These factors may be utilized to control the growth and function of implanted cells, the ingrowth of blood vessels into the forming tissue, and/or the deposition and organization of fibrous tissue around the implant.
Examples of growth factors include heparin binding growth factor (hbgf), transforming growth factor alpha or beta (TGFβ), alpha fibroblastic growth factor (FGF), epidermal growth factor (TGF), vascular endothelium growth factor (VEGF), some of which are also angiogenic factors. Other factors include hormones such as insulin, glucagon, and estrogen. In some embodiments it may be desirable to incorporate factors such as nerve growth factor (NGF) or muscle morphogenic factor (MMP).
Steroidal antiinflammatories can be used to decrease inflammation to the implanted matrix, thereby decreasing the amount of fibroblast tissue growing into the matrix.
These factors are known to those skilled in the art and are available commercially or described in the literature. In vivo dosages are calculated based on in vitro release studies in cell culture; an effective dosage is that dosage which increases cell proliferation or survival as compared with controls, as described in more detail in the following examples. Preferably, the bioactive factors are incorporated to between one and 30% by weight, although the factors can be incorporated to a weight percentage between 0.01 and 95 weight percentage.
Bioactive molecules can be incorporated into the matrix and released over time by diffusion and/or degradation of the matrix, they can be suspended with the cell suspension, they can be incorporated into microspheres which are suspended with the cells or attached to or incorporated within the matrix, or some combination thereof. Microspheres would typically be formed of materials similar to those forming the matrix, selected for their release properties rather than structural properties. Release properties can also be determined by the size and physical characteristics of the microspheres.
II. Cells to Be Implanted
Cells to be implanted are dissociated using standard techniques such as digestion with a collagenase, trypsin or other protease solution. Preferred cell types are mesenchymal cells, especially smooth or skeletal muscle cells, myocytes (muscle stem cells), fibroblasts, chondrocytes, adipocytes, fibromyoblasts, and ectodermal cells, including ductile and skin cells, hepatocytes, Islet cells, cells present in the intestine, and other parenchymal cells, osteoblasts and other cells forming bone or cartilage. In some cases it may also be desirable to include nerve cells. Cells can be normal or genetically engineered to provide additional or normal function. Methods for genetically engineering cells with retroviral vectors, polyethylene glycol, or other methods known to those skilled in the art can be used.
Cells are preferably autologous cells, obtained by biopsy and expanded in culture, although cells from close relatives or other donors of the same species may be used with appropriate immunosuppression. Immunologically inert cells, such as embryonic or fetal cells, stem cells, and cells genetically engineered to avoid the need for immunosuppression can also be used. Methods and drugs for immunosuppression are known to those skilled in the art of transplantation. A preferred compound is cyclosporin using the recommended dosages.
In the preferred embodiment, cells are obtained by biopsy and expanded in culture for subsequent implantation. Cells can be easily obtained through a biopsy anywhere in the body, for example, skeletal muscle biopsies can be obtained easily from the arm, forearm, or lower extremities, and smooth muscle can be obtained from the area adjacent to the subcutaneous tissue throughout the body. To obtain either type of muscle, the area to be biopsied can be locally anesthetized with a small amount of lidocaine injected subcutaneously. Alternatively, a small patch of lidocaine jelly can be applied over the area to be biopsied and left in place for a period of 5 to 20 minutes, prior to obtaining biopsy specimen. The biopsy can be effortlessly obtained with the use of a biopsy needle, a rapid action needle which makes the procedure extremely simple and almost painless. With the addition of the anesthetic agent, the procedure would be entirely painless. This small biopsy core of either skeletal or smooth muscle can then be transferred to media consisting of phosphate buffered saline. The biopsy specimen is then transferred to the lab where the muscle can be grown utilizing the explant technique, wherein the muscle is divided into very pieces which are adhered to culture plate, and serum containing media is added. Alternatively, the muscle biopsy can be enzymatically digested with agents such as trypsin and the cells dispersed in a culture plate with any of the routinely used medias. After cell expansion within the culture plate, the cells can be easily passaged utilizing the usual technique until an adequate number of cells is achieved.
III. Methods for Implantation
Unlike other prior art methods for making implantable matrices, the present method uses the recipient or an animal as the initial bioreactor to form a fibrous tissue-polymeric construct which optionally can be seeded with other cells and implanted. The matrix becomes infiltrated with fibrous tissue and/or blood vessels over a period ranging from between one day and a few weeks, most preferably one and two weeks. The matrix is then removed and implanted at the site where it is needed.
In one embodiment, the matrix is formed of polymer fibers having a particular desired shape, that is implanted subcutaneously. The implant is retrieved surgically, then one or more defined cell types distributed onto and into the fibers. In a second embodiment, the matrix is seeded with cells of a defined type, implanted until fibrous tissue has grown into the matrix, then the matrix removed, optionally cultured further in vitro, then reimplanted at a desired site.
The resulting structures are dictated by the matrix construction, including architecture, porosity (% void volume and pore diameter), polymer nature including composition, crystallinity, molecular weight, and degradability, hydrophobicity, and the inclusion of other biologically active molecules.
This methodology is particularly well suited for the construction of valves and tubular structures. Examples of valves are heart valves and valves of the type used for ventricular shunts for treatment of hydrocephaly. A similar structure could be used for an ascites shunt in the abdomen where needed due to liver disease or in the case of a lymphatic obstructive disease. Examples of tubular structures include blood vessels, intestine, ureters, and fallopian tubes.
The structures are formed at a site other than where they are ultimately required. This is particularly important in the case of tubular structures and valves, where integrity to fluid is essential, and where the structure is subjected to repeated stress and strain.
The present invention will be further understood by reference to the following non-limiting examples.
EXAMPLE 1
Tissue Engineering of Heart Valves
Valvular heart disease is a significant cause of morbidity and mortality. Construction of a tissue engineered valve using living autologous cells offers advantages over currently used mechanical or glutaraldehyde fixed xenograft valves.
Methods and Materials
A tissue engineered valve was constructed by seeding a synthetic polyglycolic acid (PGA) fiber based matrix with dissociated fibroblasts and endothelial cells harvested from a donor sheep heart valve. The cells were grown to confluence and split several times to increase the cell number. A mixed cell population including myofibroblasts and endothelial cells was obtained. The endothelial cells were labeled with an Ac-Dil-LDL fluorescent antibody obtained from a commercial source and sorted in a cell-sorting machine to yield a nearly pure endothelial cell population (LDL+) and a mixed cell population containing myofibroblasts and endothelial cells (LDL−). A PGA mesh (density 76.9 mg/ml and thickness 0.68 mm) was seeded with the mixed cell population and grown in culture. When the myofibroblasts reached confluence, endothelial cells were seeded onto the surface of the fibroblast/mesh constructs and grown into a single monolayer.
Immunohistochemical evaluation of constructs with antibodies against factor VIII, a specific marker for endothelial cells, revealed that tissue engineered valves histologically resemble native valve tissue. The effects of physiological flow on elastin and collagen production within the ECM were examined in a bioreactor and implanted in a sheep to determine if the constructs had the required pliability and mechanical strength for use in patients.
EXAMPLE 2
Tissue Engineering of Vascular Structures
Vascular smooth muscle tubular structures using a biodegradable polyglycolic acid polymer scaffold have been developed. The technique involves the isolation and culture of vascular smooth muscle cells, the reconstruction of a vascular wall using biodegradable polymer, and formation of the neo-tissue tubes in vitro. The feasibility of engineering vascular structures by coculturing endothelial cells with fibroblasts and smooth muscle cells on a synthetic biodegradable matrix in order to create tubular constructs which histologically resemble native vascular structures was also demonstrated.
Methods
In a first set of studies, bovine and ovine endothelial cells, smooth muscle cells, and fibroblasts were isolated using a combination of standard techniques including collagenase digestion and explantation. These cells were then expanded in tissue culture. All cells were grown in Delbecco's modified Eagle's media supplemented with 10% fetal bovine serum, 1% antibiotic solution, and basic fibroblast growth factor. Mixed colonies were purified using dilutional cloning. Thirty (N=30) two by two centimeter polyglycolic acid (PGA) fiber meshes (thickness=0.68 mm, density=76.9 mg/cc) were then serially seeded with 5×105 fibroblasts and smooth muscle cells and placed in culture. Five (N=5) 85% PGA, 15% polylatic acid tubular constructs (length=2 cm, diameter=0.8 cm) were seeded in a similar fashion. After the fibroblasts and smooth muscle cell constructs had grown to confluence (mean time 3 weeks), 1×106 endothelial cells were seeded onto them and they were placed in culture for one week. These vascular constructs were then fixed in a paraffin, sectioned and analyzed using immunohistochemical staining for factor VIII (specific for endothelial cells) and desmin (specific for muscle cells).
In a second set of studies, smooth muscle cells were obtained by harvesting the media from the artery of a lamb using standard explant techniques. Cells were expanded in culture through repeated passages and then seeded on the biodegradable polymer scaffold at a density of 1×106 cells per cm2 of polymer. The cell-polymer constructs were formed into tubes with internal diameters ranging from 2 mm to 5 mm and maintained in vitro for 6 to 8 weeks.
Results
Microscopic examination of all constructs in the first study (N=30/N=5) revealed that both types of constructs had achieved the proper histological architecture and resembled native vessels after one week. Immunohistochemical staining confirmed that endothelially lined smooth muscle/fibroblast tubes had been created. The extracellular matrices (ECM) of the vascular constructs were examined in order to determine the composition of elastin and collagen types I and III, the ECM molecules which determine the physical characteristics of native vascular tissues.
The results of the second study show that vascular smooth muscle tubes which retain their structure can be successfully formed using a polyglycolic acid polymer scaffold. The biodegradable polymer was absorbed over time, leaving a neo-tissue vascular smooth muscle tube.
EXAMPLE 3
Engineered bone from PGA Polymer Scaffold and Periosteum
The ability to create bone from periosteum and biodegradable polymer may have significant utility in reconstructive orthopedic and plastic surgery. Polyglycolic acid (PGA) is a preferred material for forming a biodegradable matrix which can be configured to a desirable shape and structure. This study was conducted to determine whether new bone constructs can be formed from periosteum or periosteal cells placed onto PGA polymer.
Materials and Methods
Bovine periosteum, harvested from fresh calf limbs, was placed either directly onto PGA polymer (1×1 cm) or onto tissue culture dishes for periosteal cell isolation. The periosteum/PGA construct was cultured for one week in MEM 199 culture media with antibiotics and ascorbic acid, then implanted into the dorsal subcutaneous space of nude mice. Periosteal cell, cultured from pieces of periosteum for two weeks, were isolated into cell suspension and seeded (approximately 1 to 3×107 cells) onto PGA polymer (1 ×1 cm); after one week in culture, the periosteal cell seeded polymer was implanted into the subcutaneous space of nude mice. Specimens, harvested at 4, 8, and 14 week intervals, were evaluated grossly and histologically.
Results
The periosteum/PGA constructs showed an organized cartilage matrix with early evidence of bone formation at four weeks, a mixture of bone and cartilage at 8 weeks, and a complete bone matrix at 14 weeks. Constructs created from periosteal cells seeded onto polymer showed presence of disorganized cartilage at 4 and 8 weeks, and a mixture of bone and cartilage at 14 weeks. Periosteum placed directly onto polymer will form an organized cartilage and bone matrix earlier than constructs formed from periosteal cell seeded polymer. This data indicates that PGA is an effective scaffold for periosteal cell attachment and migration to produce bone, which may offer new approaches to reconstructive surgery.
EXAMPLE 4
Bone Reconstruction with Tissue Engineered Vascularized Bone
The aim of this study was to determine if new vascularized bone could be engineered by transplantation of osteoblast around existing vascular pedicle using biodegradable polymers as cell delivery devices, to be used to reconstruct weight bearing bony defects.
Methods
Osteoblast and chondryocytes were isolated from calf periosteum and articular cartilage, cultured in vitro for three weeks, then seeded onto a 1×1 cm non-woven polyglycolic acid (PGA) mesh. After maintenance in vitro for one week, cell-polymer constructs were wrapped around saphenous vessels, and implanted into athymic rats for 8 weeks. The implants showed gross and histological evidence of vascularized bone or cartilage. At this time, bilateral 0.8 cm femoral shaft defect were created in the same rat, and fixed in position with a 3 cm craniofacial titanium miniplate. The new engineered bone/cartilage construct was then transferred to the femoral defect on its bilateral vascular pedicle. A total of 30 femoral defects were repaired in three groups of animals (each group composed of five animals with defects). Animals in Group 1 received implants composed of vascularized bone constructs, animals in Group 2 with vascular cartilage constructs, and Group 3 animals with blank polymer only.
At six months after surgery, the animals were studied radiographically for evidence of new bone formation at the site of the defect. Euthanasia was then performed by anesthetic overdose and each experimented femur was removed. Gross appearance was recorded and histological studies performed using hematoxylin and eosin (H & E) staining.
Results
Group 1 defect showed evidence of new bone formation around the defect. Neither Group 2 nor Group 3 defect showed any radiographic evidence of healing or bone formation. Grossly, Group 1 animals developed exuberant bony callus formation and healing of the defect. The animals in Group 2 showed filling of the bony defect with cartilaginous tissue, whereas all of the animals in Group 3 either developed a fibrous non-union or simple separation of both bony fragments with soft tissue invasion of the defect. The histological studies showed new bone formation in all Group 1 animals, new cartilage formation in all Group 2 animals, and fibrous tissue invasion in all Group 3 animals.
Conclusion
In conclusion, it was possible to engineer vascularized bone and cartilage grafts, which could be used to repair bone defects in the rat femur. Engineered tissue maintained the characteristics of the tissues form which the cells were originally isolated.
EXAMPLE 5
Engineering of Composite Bone and Cartilage
The ability to construct a composite structure of bone and cartilage offers a significant modality in reconstructive plastic and orthopedic surgery. The following study was conducted to engineer a bone and cartilage composite structure using periosteum, chondrocytes and biodegradable polymer and to direct bone and cartilage formation by selectively placing periosteum and chondrocytes onto the polymer scaffold.
Methods and materials
Bovine periosteum and cartilage were harvested from newborn calf limbs. Periosteum (1.5×2.0 cm) was wrapped around a polyglycolic acid/poly L-lactic acid co-polymer tube (3 cm in length, 3 mm in diameter), leaving the ends exposed. The cartilage pieces were enzymatically digested with collagenase, and chondrocytes (2×107 cells) were seeded onto each end of the exposed polymer. The composite construct was cultured for seven days in Medium 199 with antibiotics, fetal bovine serum, and ascorbic acid at 37° C. with 5% CO2. Eight constructs were then implanted into the dorsal subcutaneous space of eight nude mice. After 8 to 14 weeks in vivo, the implants were harvested and evaluated grossly and histologically.
Results
All implants formed into cylindrical shapes, flattened at the ends. The central portion of the implant formed into a bony matrix and the ends of the specimens formed into cartilage, approximately where the periosteum and chondrocytes were placed. Histological sections showed an organized matrix of bone and cartilage with a distinct transition between bone and cartilage.
Conclusions
The results show that periosteum and chondrocytes placed onto a biodegradable polymer will form into a composite tissue of bone and cartilage. Moreoever, bone and cartilage composite formation with selective placement of periosteum and chondrocytes on a biodegradable polymer scaffold was shown.
EXAMPLE 6
Implantation of Matrix for Ingrowth of Fibrous Tissue to Increase Mechanical Properties and Cell Survival
The following study was conducted to increase the mechanical strength and pliability of the heart valve leaflets or other engineered tissues such as those for use as blood vessels.
Methods
A PGA mesh as described in Example 1 or 2 was implanted subcutaneously in an animal, then removed after a period of one to two weeks. Fibroblasts migrated into the polymeric mesh while it was implanted. The implant was then seeded with other cells such as chondrocytes or endothelial cells and cultured in vitro for an additional period of time.
Results
The resulting implant was shown to have greater mechanical strength and pliability than implants formed solely by seeding of dissociated cells.
Modifications and variations of the method and compositions described herein will be obvious to those skilled in the art from the foregoing detailed description. Such modifications and variations are intended to come within the scope of the appended claims.

Claims (14)

1. A method for making a cell-matrix construct for use as in the shape of a heart valve or blood vessel or heart valve leaflet comprising implanting into an animal at a first site a cell-matrix construct comprising
(a) a fibrous matrix formed in the shape of a heart valve or heart value leaflet, wherein the matrix consists of a synthetic, biocompatible, chemically biodegradable polymer, and
(b) having seeded therein a mixture of cells selected from the group selected from consisting of endothelial cells, myofibroblasts, skeletal muscle cells, vascular smooth muscle cells, myocytes, fibromyoblasts, and ectodermal cells, seeded thereon,
wherein the synthetic chemically biodegradable polymer provides the biochemical properties of a heart valve or leaflet until the seeded cells can lay down their own extracellular matrix, and
matrix is formed of a biocompatible, biodegradable polymer, and implanting into an animal or human the matrix at a site where the resulting cell-construct is neededthe matrix is formed so that the cells attach to and proliferate on it to the edges of the matrix.
2. The method of claim 1 further comprising seeding wherein the matrix is seeded with dissociated parenchymal or connective tissue cells.
3. The method of claim 1 wherein the matrix is first cultured at a first site in a patient prior to being implanted at transplanted to a second site.
4. The method of claim 1 wherein the matrix is in the form of a heart valve and is implanted in the heart leaflet.
5. The method of claim 1 wherein the cell-matrix construct is seeded with vascular smooth muscle cells and endothelial cells is and implanted to form a heart valve.
6. The method of claim 5 wherein the valve is a heart valve.
7. The method of claim 1 wherein the cell-matrix construct is seeded with endothelial cells and implanted to form a blood vessel.
8. The method of claim 1 wherein the cell-matrix construct is formed of a potymer selected from the group consisting of poly(lactide) (PLA), poly(glycolic acid) (PGA), poly(lactide-co-glycolide) (PLGA), poly(caprolactone), polyanhydrides, polyamino acids, and polyortho esters.
9. The method of claim 1 wherein the cell-matrix construct contains interconnected pores in the range of between approximately 100 and 300 microns.
10. The method of claim 1 wherein the cell-matrix construct includes growth factors.
11. The method of claim 10 wherein the growth factors are selected from the group consistin of heparin binding growth factor (hbgf), transforming growth factor alpha or beta (TGFβ), alpha fibroblastic growt.h...factor (FGF),.epidermal growth factor (TGF), vascular endothelium growth factor (VEGF), insulin, glucagon, estrogen, nerve growth factor (NGF) and muscle morphogenic factor (MMP).
12. The method of claim 1 wherein the cell-matrix further comprises bioactive factors incorporated to between one and 30% by weight.
13. The method of claim 1 wherein the cell-matrix is first cultured in a bioreactor to form a fibrous tissue-polymeric construct before implantation.
14. The method of claim 13 wherein the bioreactor is an animal.
US10/782,750 1995-05-19 2004-02-19 Engineering of strong, pliable tissues Expired - Lifetime USRE42479E1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/782,750 USRE42479E1 (en) 1995-05-19 2004-02-19 Engineering of strong, pliable tissues
US11/529,691 USRE42575E1 (en) 1995-05-19 2006-09-28 Engineering of strong, pliable tissues

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/445,280 US5855610A (en) 1995-05-19 1995-05-19 Engineering of strong, pliable tissues
US09/185,360 US6348069B1 (en) 1995-05-19 1998-11-03 Engineering of strong, pliable tissues
US10/782,750 USRE42479E1 (en) 1995-05-19 2004-02-19 Engineering of strong, pliable tissues

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US08/445,280 Division US5855610A (en) 1995-05-19 1995-05-19 Engineering of strong, pliable tissues
US09/185,360 Reissue US6348069B1 (en) 1995-05-19 1998-11-03 Engineering of strong, pliable tissues

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US09/185,360 Division US6348069B1 (en) 1995-05-19 1998-11-03 Engineering of strong, pliable tissues

Publications (1)

Publication Number Publication Date
USRE42479E1 true USRE42479E1 (en) 2011-06-21

Family

ID=23768289

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/445,280 Expired - Lifetime US5855610A (en) 1995-05-19 1995-05-19 Engineering of strong, pliable tissues
US09/185,360 Ceased US6348069B1 (en) 1995-05-19 1998-11-03 Engineering of strong, pliable tissues
US10/782,750 Expired - Lifetime USRE42479E1 (en) 1995-05-19 2004-02-19 Engineering of strong, pliable tissues
US11/529,691 Expired - Lifetime USRE42575E1 (en) 1995-05-19 2006-09-28 Engineering of strong, pliable tissues

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/445,280 Expired - Lifetime US5855610A (en) 1995-05-19 1995-05-19 Engineering of strong, pliable tissues
US09/185,360 Ceased US6348069B1 (en) 1995-05-19 1998-11-03 Engineering of strong, pliable tissues

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/529,691 Expired - Lifetime USRE42575E1 (en) 1995-05-19 2006-09-28 Engineering of strong, pliable tissues

Country Status (1)

Country Link
US (4) US5855610A (en)

Families Citing this family (187)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030086975A1 (en) * 2001-11-08 2003-05-08 Timothy Ringeisen Method for making a porous Polymeric material
US6528483B2 (en) 1995-06-07 2003-03-04 André Beaulieu Method of producing concentrated non-buffered solutions of fibronectin
US20030008396A1 (en) * 1999-03-17 2003-01-09 Ku David N. Poly(vinyl alcohol) hydrogel
US7524335B2 (en) * 1997-05-30 2009-04-28 Smith & Nephew, Inc. Fiber-reinforced, porous, biodegradable implant device
US5980564A (en) * 1997-08-01 1999-11-09 Schneider (Usa) Inc. Bioabsorbable implantable endoprosthesis with reservoir
AU747166B2 (en) * 1997-10-31 2002-05-09 Children's Medical Center Corporation Bladder reconstruction
US6872387B1 (en) 1998-02-24 2005-03-29 The Regents Of The University Of Michigan Three-dimensional hydrogel/cell system
US6660301B1 (en) 1998-03-06 2003-12-09 Biosphere Medical, Inc. Injectable microspheres for dermal augmentation and tissue bulking
US6171610B1 (en) * 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
US6027744A (en) * 1998-04-24 2000-02-22 University Of Massachusetts Medical Center Guided development and support of hydrogel-cell compositions
US20080077251A1 (en) * 1999-06-07 2008-03-27 Chen Silvia S Cleaning and devitalization of cartilage
US20100030340A1 (en) * 1998-06-30 2010-02-04 Wolfinbarger Jr Lloyd Plasticized Grafts and Methods of Making and Using Same
US8563232B2 (en) 2000-09-12 2013-10-22 Lifenet Health Process for devitalizing soft-tissue engineered medical implants, and devitalized soft-tissue medical implants produced
US6293970B1 (en) * 1998-06-30 2001-09-25 Lifenet Plasticized bone and soft tissue grafts and methods of making and using same
US6551355B1 (en) * 1998-08-14 2003-04-22 Cambridge Scientific, Inc. Tissue transplant coated with biocompatible biodegradable polymer
US7001746B1 (en) * 1999-01-29 2006-02-21 Artecel Sciences, Inc. Methods and compositions for the differentiation of human preadipocytes into adipocytes
DE19919625C2 (en) * 1999-04-29 2002-10-31 Symetis Ag Zuerich In vitro method for producing a homologous heart valve and valve that can be produced by this method
GB9910377D0 (en) * 1999-05-05 1999-06-30 Univ Westminster Tissue engineering
US20040167634A1 (en) * 1999-05-26 2004-08-26 Anthony Atala Prosthetic kidney and its use for treating kidney disease
JP5398941B2 (en) 1999-08-05 2014-01-29 エイビーティー ホールディング カンパニー Pluripotent adult stem cell and method for isolating the same
JP3603179B2 (en) * 1999-09-09 2004-12-22 グンゼ株式会社 Cardiovascular tissue culture substrate and tissue regeneration method
AU6132700A (en) * 1999-09-30 2001-04-05 Chienna B.V. Polymers loaded with bioactive agents
US6391049B1 (en) 1999-10-06 2002-05-21 Board Of Regents The University Of Texas System Solid biodegradable device for use in tissue repair
JP2001120582A (en) 1999-10-22 2001-05-08 Gunze Ltd Artificial cardiac valve and method of manufacturing the same
EP1099443A1 (en) * 1999-11-11 2001-05-16 Sulzer Orthopedics Ltd. Transplant/implant device and method for its production
DE19959750C1 (en) * 1999-12-11 2001-05-31 Axel Thierauf Production of skin implant, e.g. for treating burn or other open wound, uses flat element of fine biocompatible, biodegradable or bioresorbable fibers of hydrolytically condensed silicon compound on nutrient solution
US6479064B1 (en) 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
US6376244B1 (en) * 1999-12-29 2002-04-23 Children's Medical Center Corporation Methods and compositions for organ decellularization
US6368859B1 (en) 1999-12-29 2002-04-09 Children's Medical Center Corporation Methods and compositions for producing a fascial sling
US6428802B1 (en) 1999-12-29 2002-08-06 Children's Medical Center Corp. Preparing artificial organs by forming polylayers of different cell populations on a substrate
US7575921B2 (en) * 1999-12-30 2009-08-18 Vbi Technologies, L.L.C. Spore-like cells and uses thereof
US7560275B2 (en) * 1999-12-30 2009-07-14 Vbi Technologies, L.L.C. Compositions and methods for generating skin
EP1918366A1 (en) 2000-02-26 2008-05-07 Artecel, Inc. Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof
US7078230B2 (en) 2000-02-26 2006-07-18 Artecel, Inc. Adipose tissue-derived stromal cell that expresses characteristics of a neuronal cell
US7582292B2 (en) 2000-02-26 2009-09-01 Artecel, Inc. Adipose tissue derived stromal cells for the treatment of neurological disorders
US7338657B2 (en) * 2001-03-15 2008-03-04 Biosphere Medical, Inc. Injectable microspheres for tissue construction
EP1274472A2 (en) 2000-03-20 2003-01-15 Biosphere Medical, Inc. Injectable and swellable microspheres for tissue bulking
US6436424B1 (en) 2000-03-20 2002-08-20 Biosphere Medical, Inc. Injectable and swellable microspheres for dermal augmentation
US20020133229A1 (en) 2000-03-24 2002-09-19 Laurencin Cato T. Ligament and tendon replacement constructs and methods for production and use thereof
JP2003528130A (en) * 2000-03-24 2003-09-24 バイオスフィアー メディカル,インク. Microspheres for active embolization
US20030212022A1 (en) * 2001-03-23 2003-11-13 Jean-Marie Vogel Compositions and methods for gene therapy
US6652583B2 (en) * 2000-04-07 2003-11-25 Rhode Island Hospital Cardiac valve replacement
JP5684963B2 (en) * 2000-04-14 2015-03-18 ユニバーシティ オブ ピッツバーグ オブ ザ コモンウェルス システム オブ ハイヤー エデュケイション Muscle tissue-derived progenitor cells, their compositions, and soft tissue and bone growth and thickening using treatments
US7108721B2 (en) * 2000-05-11 2006-09-19 Massachusetts Institute Of Technology Tissue regrafting
GB0011244D0 (en) * 2000-05-11 2000-06-28 Smith & Nephew Inc Tissue regrafting
DE10026482A1 (en) * 2000-05-29 2001-12-13 Augustinus Bader Process for the production of a bioartificial graft
DE10026480A1 (en) * 2000-05-29 2001-12-13 Augustinus Bader Method of making a recipient-specific tissue graft or implant
WO2002026939A2 (en) 2000-09-25 2002-04-04 The Board Of Trustees Of The University Of Illinois Microfabrication of membranes for the growth of cells
US20020151050A1 (en) * 2000-10-30 2002-10-17 Vacanti Charles A. Isolation of spore-like cells from tissues exposed to extreme conditions
US6835390B1 (en) * 2000-11-17 2004-12-28 Jon Vein Method for producing tissue engineered meat for consumption
US6696074B2 (en) * 2000-12-04 2004-02-24 Tei Biosciences, Inc. Processing fetal or neo-natal tissue to produce a scaffold for tissue engineering
US6669725B2 (en) 2000-12-28 2003-12-30 Centerpulse Biologics Inc. Annuloplasty ring for regeneration of diseased or damaged heart valve annulus
US6899873B2 (en) * 2001-02-02 2005-05-31 The Regents Of The University Of Michigan Method of forming micro-tubular polymeric materials
DE50111147D1 (en) * 2001-02-13 2006-11-16 Corlife Gbr Bioartificial primary vascularized tissue matrix and bioartificial primary vascularized tissue
US7326564B2 (en) 2001-02-20 2008-02-05 St. Jude Medical, Inc. Flow system for medical device evaluation and production
WO2002067867A2 (en) * 2001-02-23 2002-09-06 The University Of Pittsburgh Rapid preparation of stem cell matrices for use in tissue and organ treatment and repair
ES2328460T3 (en) 2001-04-24 2009-11-13 Dolores Baksh POPULATIONS OF PROGENITING CELLS, EXPANSION OF THE SAME AND GROWTH OF TYPES OF NON HEMATOPOYETIC CELLS AND FABRICS FROM THE SAME.
US6913762B2 (en) * 2001-04-25 2005-07-05 Mayo Foundation For Medical Education And Research Stent having non-woven framework containing cells
US20030129751A1 (en) * 2001-05-16 2003-07-10 Grikscheit Tracy C. Tissue-engineered organs
DE10126137C1 (en) * 2001-05-29 2002-11-07 Andreas Haisch In vitro preparation of cells, tissues and organs, useful e.g. as wound dressing, as model or implant, by growing on resorbable matrix of hydrolyzed silane
WO2002102237A2 (en) * 2001-06-15 2002-12-27 The Cleveland Clinic Foundation Tissue engineered mitral valve chrodae and methods of making and using same
EP1416888A4 (en) 2001-07-16 2007-04-25 Depuy Products Inc Meniscus regeneration device and method
US7819918B2 (en) * 2001-07-16 2010-10-26 Depuy Products, Inc. Implantable tissue repair device
EP1416886A4 (en) * 2001-07-16 2007-04-18 Depuy Products Inc Cartilage repair and regeneration scaffold and method
EP1416866A4 (en) * 2001-07-16 2007-04-18 Depuy Products Inc Devices form naturally occurring biologically derived
WO2003007790A2 (en) * 2001-07-16 2003-01-30 Depuy Products, Inc. Hybrid biologic/synthetic porous extracellular matrix scaffolds
US7163563B2 (en) * 2001-07-16 2007-01-16 Depuy Products, Inc. Unitary surgical device and method
AU2002320610A1 (en) * 2001-07-16 2003-03-03 Altertek/Bio Inc Tissue engineered heart valve
US8025896B2 (en) * 2001-07-16 2011-09-27 Depuy Products, Inc. Porous extracellular matrix scaffold and method
US7049057B2 (en) * 2001-11-16 2006-05-23 Children's Medical Center Corporation Tissue engineered uterus
BR0214217A (en) * 2001-11-16 2004-09-21 Childrens Medical Center Increased Organ Function
AU2002364558A1 (en) * 2001-12-11 2003-06-23 Cytograft Tissue Engineering, Inc. Tissue engineered cellular sheets, methods of making and use thereof
US7297540B2 (en) * 2002-01-15 2007-11-20 Yissum Research Development Company Of The Hebrew University Of Jerusalem Methods of generating tissue using devitalized, acellular scaffold matrices derived from micro-organs
US7622299B2 (en) * 2002-02-22 2009-11-24 University Of Washington Bioengineered tissue substitutes
US20050260259A1 (en) * 2004-04-23 2005-11-24 Bolotin Elijah M Compositions for treatment with glucagon-like peptide, and methods of making and using the same
DE60335608D1 (en) * 2002-02-27 2011-02-17 Pharmain Corp COMPOSITIONS FOR THE DELIVERY OF THERAPEUTICS AND OTHER MATERIALS AND METHOD FOR THE PRODUCTION AND USE THEREOF
US7635463B2 (en) * 2002-02-27 2009-12-22 Pharmain Corporation Compositions for delivery of therapeutics and other materials
US7396537B1 (en) * 2002-02-28 2008-07-08 The Trustees Of The University Of Pennsylvania Cell delivery patch for myocardial tissue engineering
US20040254640A1 (en) * 2002-03-01 2004-12-16 Children's Medical Center Corporation Needle punched textile for use in growing anatomical elements
US7776600B2 (en) 2002-04-18 2010-08-17 Carnegie Mellon University Method of manufacturing hydroxyapatite and uses therefor in delivery of nucleic acids
AU2003218271A1 (en) * 2002-04-18 2003-11-03 Carnegie Mellon University Method of manufacturing hydroxyapatite and uses therefor in delivery of nucleic acids
KR100484636B1 (en) * 2002-04-30 2005-04-20 경북대학교 산학협력단 Matrix for cultivating hepatocyte
US7299805B2 (en) 2002-06-07 2007-11-27 Marctec, Llc Scaffold and method for implanting cells
DE10235237A1 (en) * 2002-08-01 2004-02-12 Symetis Ag In vitro preparation of homologous heart valves, useful for replacement of diseased valves, by inoculating biodegradable carrier with fibroblasts and attachment to a non-degradable stent
AU2003284875A1 (en) * 2002-10-17 2004-05-04 Vacanti, Joseph P. Biological scaffolding material
KR100539371B1 (en) * 2002-10-21 2005-12-27 메디칸(주) In vitro cultured human preadipocyte for human soft tissue volume replacement with injectable material as a scaffold
US7718189B2 (en) 2002-10-29 2010-05-18 Transave, Inc. Sustained release of antiinfectives
CN1747738B (en) * 2002-10-29 2010-11-24 川塞夫有限公司 Sustained release of antifectives
US7879351B2 (en) * 2002-10-29 2011-02-01 Transave, Inc. High delivery rates for lipid based drug formulations, and methods of treatment thereof
US7627373B2 (en) * 2002-11-30 2009-12-01 Cardiac Pacemakers, Inc. Method and apparatus for cell and electrical therapy of living tissue
US20040158289A1 (en) * 2002-11-30 2004-08-12 Girouard Steven D. Method and apparatus for cell and electrical therapy of living tissue
US8940292B2 (en) 2003-01-28 2015-01-27 Wake Forest University Health Sciences Enhancement of angiogenesis to grafts using cells engineered to produce growth factors
EP1608292A4 (en) * 2003-03-13 2007-10-31 Univ Rice William M Composite injectable and pre-fabricated bone replacement material and method for the production of such bone replacement material
JP2006524072A (en) * 2003-04-21 2006-10-26 ベリーゲン アーゲー Seeded tear-resistant scaffold
CN103230415B (en) * 2003-04-25 2016-05-04 匹兹堡大学联邦制高等教育 For promoting and strengthen the neural muscle derived cell (MDC) of repairing and regenerating
DE10322024A1 (en) * 2003-05-16 2004-12-02 Symetis Ag Bioreactor for manufacturing a tissue prosthesis, in particular a heart valve
US7384786B2 (en) * 2003-07-16 2008-06-10 Scimed Life Systems, Inc. Aligned scaffolds for improved myocardial regeneration
US20050013870A1 (en) * 2003-07-17 2005-01-20 Toby Freyman Decellularized extracellular matrix of conditioned body tissues and uses thereof
DE602004030254D1 (en) 2003-08-01 2011-01-05 Two Cells Co Ltd SCAFFOLD FREE, SELF ORGANIZED, 3 DIMENSIONAL, SYNTHETIC TISSUE
WO2007083504A1 (en) * 2003-08-01 2007-07-26 Norimasa Nakamura Scaffold-free self-organized 3d synthetic tissue
US20070087959A1 (en) * 2003-08-19 2007-04-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Method of inducing biomineralization method of inducing bone regeneration and methods related thereof
US7261732B2 (en) * 2003-12-22 2007-08-28 Henri Justino Stent mounted valve
WO2005077013A2 (en) 2004-02-06 2005-08-25 Georgia Tech Research Corporation Surface directed cellular attachment
CA2558661C (en) * 2004-02-06 2012-09-04 Georgia Tech Research Corporation Load bearing biocompatible device
US7840263B2 (en) * 2004-02-27 2010-11-23 Cardiac Pacemakers, Inc. Method and apparatus for device controlled gene expression
EP1729675A4 (en) * 2004-03-05 2011-05-18 Univ Columbia Multi-phased, biodegradable and osteointegrative composite scaffold for biological fixation of musculoskeletal soft tissue to bone
EP1744794A2 (en) * 2004-03-05 2007-01-24 The Trustees Of Columbia University In The City Of New York Polymer-ceramic-hydrogel composite scaffold for osteochondral repair
US20050249772A1 (en) * 2004-05-04 2005-11-10 Prasanna Malaviya Hybrid biologic-synthetic bioabsorbable scaffolds
US7764995B2 (en) 2004-06-07 2010-07-27 Cardiac Pacemakers, Inc. Method and apparatus to modulate cellular regeneration post myocardial infarct
US20050278025A1 (en) * 2004-06-10 2005-12-15 Salumedica Llc Meniscus prosthesis
US7351423B2 (en) 2004-09-01 2008-04-01 Depuy Spine, Inc. Musculo-skeletal implant having a bioactive gradient
US20060097422A1 (en) * 2004-11-08 2006-05-11 Diamond Andrew J Method for performing surgery and appliances produced thereby
US20080118561A1 (en) * 2004-12-08 2008-05-22 Helen Marie Nugent Materials and Methods for Minimally-Invasive Administration of a Cell-Containing Flowable Composition
US8060219B2 (en) * 2004-12-20 2011-11-15 Cardiac Pacemakers, Inc. Epicardial patch including isolated extracellular matrix with pacing electrodes
US7981065B2 (en) 2004-12-20 2011-07-19 Cardiac Pacemakers, Inc. Lead electrode incorporating extracellular matrix
US8874204B2 (en) * 2004-12-20 2014-10-28 Cardiac Pacemakers, Inc. Implantable medical devices comprising isolated extracellular matrix
US7354627B2 (en) * 2004-12-22 2008-04-08 Depuy Products, Inc. Method for organizing the assembly of collagen fibers and compositions formed therefrom
EP1693025A1 (en) * 2005-02-17 2006-08-23 Universität Zürich Method of manufacturing a tissue-engineered prosthesis
US20060204445A1 (en) * 2005-03-11 2006-09-14 Anthony Atala Cell scaffold matrices with image contrast agents
US20060204539A1 (en) * 2005-03-11 2006-09-14 Anthony Atala Electrospun cell matrices
CA2602029C (en) * 2005-03-11 2014-07-15 Wake Forest University Health Sciences Tissue engineered blood vessels
WO2006099332A2 (en) 2005-03-11 2006-09-21 Wake Forest University Health Sciences Production of tissue engineered digits and limbs
WO2006099334A2 (en) 2005-03-11 2006-09-21 Wake Forest University Health Sciences Production of tissue engineered heart valves
CA2605080A1 (en) * 2005-04-21 2006-11-02 Massachusetts Institute Of Technology Materials and methods for altering an immune response to exogenous and endogenous immunogens, including syngeneic and non-syngeneic cells, tissues or organs
CA2607228C (en) * 2005-05-09 2014-09-16 Biosphere Medical S.A. Compositions and methods using microspheres and non-ionic contrast agents
SG195537A1 (en) 2005-06-21 2013-12-30 Shire Regenerative Medicine Inc Methods and compositions for enhancing vascular access
SG184751A1 (en) * 2005-10-26 2012-10-30 Jan-Eric W Ahlfors Acellular bioabsorbable tissue regeneration matrices produced by incubating acellular blood products
US20070098798A1 (en) * 2005-10-28 2007-05-03 Medtronic, Inc. Method and article to restore function to a flow control location in a patient
US20100204783A1 (en) * 2005-12-06 2010-08-12 Helen Marie Nugent Methods and compositions for enhancing vascular access
DK1962805T3 (en) 2005-12-08 2016-09-26 Insmed Inc Lipid-based compositions of the anti-infective agents for the treatment of lung infections
KR101872061B1 (en) 2005-12-19 2018-06-27 파마인 코포레이션 Hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
US20070178137A1 (en) * 2006-02-01 2007-08-02 Toby Freyman Local control of inflammation
JP5372764B2 (en) * 2006-11-07 2013-12-18 シャイア リジェネラティブ メディシン, インコーポレイテッド Materials and methods for the treatment and management of angiogenesis-related diseases
US20100047309A1 (en) * 2006-12-06 2010-02-25 Lu Helen H Graft collar and scaffold apparatuses for musculoskeletal tissue engineering and related methods
US7871440B2 (en) * 2006-12-11 2011-01-18 Depuy Products, Inc. Unitary surgical device and method
US20080176206A1 (en) * 2007-01-18 2008-07-24 Toshiharu Shinoka Cardiovascular tissue culture substrate
US8753391B2 (en) 2007-02-12 2014-06-17 The Trustees Of Columbia University In The City Of New York Fully synthetic implantable multi-phased scaffold
WO2008137717A1 (en) * 2007-05-04 2008-11-13 Transave, Inc. Compositions of multicationic drugs for reducing interactions with polyanionic biomolecules and methods and uses thereof
US9114081B2 (en) 2007-05-07 2015-08-25 Insmed Incorporated Methods of treating pulmonary disorders with liposomal amikacin formulations
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US9333214B2 (en) 2007-05-07 2016-05-10 Insmed Incorporated Method for treating pulmonary disorders with liposomal amikacin formulations
US9580688B2 (en) * 2007-06-08 2017-02-28 Wake Forest University Health Sciences Kidney structures and methods of forming the same
US10590391B2 (en) * 2007-06-08 2020-03-17 Wake Forest University Health Sciences Selective cell therapy for the treatment of renal failure
EP2162529B1 (en) 2007-06-08 2019-03-27 Wake Forest University Health Sciences Selective cell therapy for the treatment of renal failure
WO2008156705A1 (en) * 2007-06-13 2008-12-24 Pervasis Therapeutics, Inc. Methods and devices for minimally-invasive delivery of cell-containing flowable compositions
US20090024224A1 (en) * 2007-07-16 2009-01-22 Chen Silvia S Implantation of cartilage
US9125743B2 (en) * 2007-07-16 2015-09-08 Lifenet Health Devitalization and recellularization of cartilage
US7960336B2 (en) * 2007-08-03 2011-06-14 Pharmain Corporation Composition for long-acting peptide analogs
CA2965384C (en) 2007-08-09 2021-06-22 The Board Of Regents Of The University Of Texas System Bi-layered bone-like scaffolds
US8563527B2 (en) * 2007-08-20 2013-10-22 Pharmain Corporation Oligonucleotide core carrier compositions for delivery of nucleic acid-containing therapeutic agents, methods of making and using the same
WO2009042671A1 (en) * 2007-09-24 2009-04-02 The Board Of Trustees Of The University Of Illinois Three-dimensional microfabricated bioreactors with embedded capillary network
US20090176892A1 (en) * 2008-01-09 2009-07-09 Pharmain Corporation Soluble Hydrophobic Core Carrier Compositions for Delivery of Therapeutic Agents, Methods of Making and Using the Same
US11051733B2 (en) * 2008-01-18 2021-07-06 Wake Forest University Health Sciences Isolating and purifying cells for therapy
WO2009102466A1 (en) * 2008-02-14 2009-08-20 The General Hospital Corporation Well-based flow system for cell culture
AU2009282619B2 (en) * 2008-08-18 2015-08-20 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Bone augmentation utilizing muscle-derived progenitor compositions in biocompatible matrix, and treatments thereof
KR101724639B1 (en) 2008-11-04 2017-04-07 리젠메드 (케이만) 엘티디. Cell-scaffold constructs
US9277999B2 (en) * 2009-02-27 2016-03-08 University of Pittsburgh—of the Commonwealth System of Higher Education Joint bioscaffolds
US9511093B2 (en) 2009-03-23 2016-12-06 The Texas A & M University System Compositions of mesenchymal stem cells to regenerate bone
DE102009032218A1 (en) * 2009-07-06 2011-01-13 Aesculap Aktiengesellschaft Elevated implant for the reconstruction of meniscal defects or meniscal duodenal defects
WO2011060135A1 (en) 2009-11-12 2011-05-19 Vbi Technologies, Llc Subpopulations of spore-like cells and uses thereof
KR20130061685A (en) * 2010-05-03 2013-06-11 텐젼, 인코포레이티드 Smooth muscle cell constructs
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
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
EP2757964B1 (en) 2011-05-26 2016-05-04 Cartiva, Inc. Tapered joint implant and related tools
CN103930066A (en) * 2011-09-12 2014-07-16 奥加诺沃公司 Platform for engineered implantable tissues and organs and methods of making the same
WO2013074099A1 (en) 2011-11-16 2013-05-23 Empire Technology Development Llc Three-dimensional porous biodegradable cell scaffold
EP2841010B1 (en) 2012-04-24 2023-08-23 Harvard Apparatus Regenerative Technology, Inc. Supports for engineered tissue scaffolds
LT2852391T (en) 2012-05-21 2022-03-10 Insmed Incorporated Systems for treating pulmonary infections
US10350072B2 (en) 2012-05-24 2019-07-16 Cartiva, Inc. Tooling for creating tapered opening in tissue and related methods
AU2013352259B2 (en) 2012-11-29 2018-06-14 Insmed Incorporated Stabilized vancomycin formulations
WO2014110300A1 (en) 2013-01-09 2014-07-17 Harvard Apparatus Regenerative Technology Synthetic scaffolds
EP2983643A4 (en) 2013-04-12 2016-12-28 Univ Columbia Methods for host cell homing and dental pulp regeneration
US20150037436A1 (en) 2013-07-30 2015-02-05 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US10092679B2 (en) 2013-10-18 2018-10-09 Wake Forest University Health Sciences Laminous vascular constructs combining cell sheet engineering and electrospinning technologies
US9801910B2 (en) 2014-03-17 2017-10-31 Ethicon, Inc. Decellularized pleural matrix
ES2926985T3 (en) 2014-05-15 2022-10-31 Insmed Inc Methods for treating nontuberculous mycobacterial lung infections
DE102014008476A1 (en) 2014-06-05 2015-12-17 Michael Jagodzinski Production of a Component for Cultivation of a Joint Surface Implant, Cultivation and Implantation of a Joint Surface Implant
CN105983134A (en) * 2015-03-05 2016-10-05 刘畅 Artificial blood vessel and preparation method thereof
WO2016161025A1 (en) 2015-03-31 2016-10-06 Cartiva, Inc. Hydrogel implants with porous materials and methods
EP3892241A1 (en) 2015-03-31 2021-10-13 Cartiva, Inc. Drill bit for carpometacarpal implant
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
CA3067558A1 (en) * 2017-06-16 2018-12-20 Avery Therapeutics, Inc. Three dimensional tissue compositions and methods of use
EP3773505A4 (en) 2018-03-30 2021-12-22 Insmed Incorporated Methods for continuous manufacture of liposomal drug products

Citations (138)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1995970A (en) 1931-04-04 1935-03-26 Du Pont Polymeric lactide resin
US2609347A (en) 1948-05-27 1952-09-02 Wilson Christopher Lumley Method of making expanded polyvinyl alcohol-formaldehyde reaction product and product resulting therefrom
US2653917A (en) 1950-06-15 1953-09-29 Christopher L Wilson Method of making an expanded material and the product resulting therefrom
US2659935A (en) 1950-03-18 1953-11-24 Christopher L Wilson Method of making compressed sponges
US2664366A (en) 1949-09-19 1953-12-29 Wilson Christopher Lumley Plasticized sponge material and method of making same
US2676945A (en) 1950-10-18 1954-04-27 Du Pont Condensation polymers of hydroxyacetic acid
US2683136A (en) 1950-10-25 1954-07-06 Du Pont Copolymers of hydroxyacetic acid with other alcohol acids
US2703316A (en) 1951-06-05 1955-03-01 Du Pont Polymers of high melting lactide
US2758987A (en) 1952-06-05 1956-08-14 Du Pont Optically active homopolymers containing but one antipodal species of an alpha-monohydroxy monocarboxylic acid
US2846407A (en) 1954-01-13 1958-08-05 Wilson Christopher Lumley Method of making a detergent and solvent resistant sponge material
US2951828A (en) 1957-03-04 1960-09-06 Boehringer Sohn Ingelheim Process for the production of bead polymerizates from cyclic internal esters of alpha-hydroxy carboxylic acids
US3514791A (en) 1967-07-25 1970-06-02 Charles H Sparks Tissue grafts
US3531561A (en) 1965-04-20 1970-09-29 Ethicon Inc Suture preparation
US3826241A (en) 1972-10-16 1974-07-30 Investors In Ventures Inc Implanting method
US3880991A (en) 1969-03-24 1975-04-29 Brook David E Polymeric article for dispensing drugs
US3883393A (en) 1972-05-18 1975-05-13 Us Health Education & Welfare Cell culture on semi-permeable tubular membranes
US3902497A (en) 1974-03-25 1975-09-02 American Cyanamid Co Body absorbable sponge and method of making
US3935065A (en) 1971-09-02 1976-01-27 Roland Karl Doerig Procedure for conservation of living organs and apparatus for the execution of this procedure
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
US3960150A (en) 1971-09-09 1976-06-01 Alza Corporation Bioerodible ocular device
US3974526A (en) 1973-07-06 1976-08-17 Dardik Irving I Vascular prostheses and process for producing the same
US3992725A (en) 1973-11-16 1976-11-23 Homsy Charles A Implantable material and appliances and method of stabilizing body implants
US3995444A (en) 1974-11-08 1976-12-07 American Hospital Supply Corporation Organ perfusion system
US4026304A (en) 1972-04-12 1977-05-31 Hydro Med Sciences Inc. Bone generating method and device
US4060081A (en) 1975-07-15 1977-11-29 Massachusetts Institute Of Technology Multilayer membrane useful as synthetic skin
US4069307A (en) 1970-10-01 1978-01-17 Alza Corporation Drug-delivery device comprising certain polymeric materials for controlled release of drug
US4137921A (en) 1977-06-24 1979-02-06 Ethicon, Inc. Addition copolymers of lactide and glycolide and method of preparation
US4141087A (en) 1977-01-19 1979-02-27 Ethicon, Inc. Isomorphic copolyoxalates and sutures thereof
US4144126A (en) 1975-05-21 1979-03-13 Beecham Group Limited Cell culture method
DE2853614A1 (en) 1978-01-25 1979-07-26 Bentley Lab IMPLANT
US4186448A (en) 1976-04-16 1980-02-05 Brekke John H Device and method for treating and healing a newly created bone void
US4192827A (en) 1974-06-27 1980-03-11 Ciba-Geigy Corporation Water-insoluble hydrophilic copolymers
US4205399A (en) 1977-06-13 1980-06-03 Ethicon, Inc. Synthetic absorbable surgical devices of poly(alkylene oxalates)
US4228243A (en) 1978-07-13 1980-10-14 Toray Industries, Inc. Cell culture propagation apparatus
US4239664A (en) 1978-10-31 1980-12-16 Research Corporation Anti-thrombogenic PVP-heparin polymer
US4243775A (en) 1978-11-13 1981-01-06 American Cyanamid Company Synthetic polyester surgical articles
US4277582A (en) 1978-03-03 1981-07-07 Ciba-Geigy Corporation Water-insoluble hydrophilic copolymers
US4280954A (en) 1975-07-15 1981-07-28 Massachusetts Institute Of Technology Crosslinked collagen-mucopolysaccharide composite materials
US4304866A (en) 1979-11-14 1981-12-08 Massachusetts Institute Of Technology Transplantable sheets of living keratinous tissue
US4304591A (en) 1978-01-25 1981-12-08 Ciba-Geigy Corporation Water-insoluble hydrophilic copolymers used as carriers for medicaments and pesticides
US4328204A (en) 1977-03-02 1982-05-04 Ethicon, Inc. Absorbable polymer-drug compounds and method for making same
US4348329A (en) 1979-12-20 1982-09-07 Dennis Chapman Biocompatible surfaces
US4347847A (en) 1980-06-06 1982-09-07 Usher Francis C Method of hernia repair
US4352883A (en) 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4356261A (en) 1980-04-22 1982-10-26 Rush-Presbyterian-St. Luke's Medical Center Anti-invasion factor containing cultures
US4391797A (en) 1977-01-05 1983-07-05 The Children's Hospital Medical Center Systems for the controlled release of macromolecules
US4416986A (en) 1981-01-16 1983-11-22 Merck & Co., Inc. Methods of producing HBsAg
US4427808A (en) 1980-07-21 1984-01-24 Ceskoslovenska Akademie Ved Composite polymeric material for biological and medical applications and the method for its preparation
US4431428A (en) 1981-09-30 1984-02-14 Trimedyne, Inc. Bio-artificial organ using microencapsulated enzymes
US4438198A (en) 1981-09-30 1984-03-20 Trimedyne, Inc. Biochemically active matrix for use in a bio-artificial organ
US4439152A (en) 1982-03-04 1984-03-27 Small Irwin A Method of jawbone abutment implant for dental prostheses and implant device
US4440921A (en) 1982-06-21 1984-04-03 Research Corporation Coupling of polyorganophosphazenes to carboxylic acid
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4446229A (en) 1982-12-30 1984-05-01 Indech Robert B Method of tissue growth
US4446234A (en) 1981-10-23 1984-05-01 The United States Of America As Represented By The Department Of Health And Human Services Vitro cellular interaction with amnion membrane substrate
US4450150A (en) 1973-05-17 1984-05-22 Arthur D. Little, Inc. Biodegradable, implantable drug delivery depots, and method for preparing and using the same
US4456687A (en) 1978-11-16 1984-06-26 President And Fellows Of Harvard College Agents for promoting growth of epithelial cells
US4458678A (en) 1981-10-26 1984-07-10 Massachusetts Institute Of Technology Cell-seeding procedures involving fibrous lattices
US4485096A (en) 1982-02-26 1984-11-27 Massachusetts Institute Of Technology Tissue-equivalent and method for preparation thereof
US4485097A (en) 1982-05-26 1984-11-27 Massachusetts Institute Of Technology Bone-equivalent and method for preparation thereof
US4489056A (en) 1982-06-30 1984-12-18 Merck & Co., Inc. Acid anhydrides as rate controlling agent for the erosion of polymers which latter polymers have beneficial substances dispersed throughout their matrix or where the polymer matrix surrounds the beneficial substance
US4494385A (en) 1982-06-04 1985-01-22 Hoxan Corporation Method of preserving organ and apparatus for preserving the same
US4495174A (en) 1982-06-21 1985-01-22 Research Corporation Anesthetic polyorganophosphazenes
US4505266A (en) * 1981-10-26 1985-03-19 Massachusetts Institute Of Technology Method of using a fibrous lattice
US4520821A (en) * 1982-04-30 1985-06-04 The Regents Of The University Of California Growing of long-term biological tissue correction structures in vivo
US4528265A (en) 1982-05-11 1985-07-09 Becker Robert O Processes and products involving cell modification
US4544516A (en) 1982-07-28 1985-10-01 Battelle Development Corporation Collagen orientation
US4545082A (en) 1982-03-25 1985-10-08 Vascutek Limited Vascular prosthesis
US4553272A (en) 1981-02-26 1985-11-19 University Of Pittsburgh Regeneration of living tissues by growth of isolated cells in porous implant and product thereof
US4559298A (en) 1982-11-23 1985-12-17 American National Red Cross Cryopreservation of biological materials in a non-frozen or vitreous state
US4559304A (en) 1982-08-09 1985-12-17 Koken Co., Ltd. Substratum for cell culture and a method for culturing and isolating cells using same
US4563350A (en) 1984-10-24 1986-01-07 Collagen Corporation Inductive collagen based bone repair preparations
US4563489A (en) 1984-02-10 1986-01-07 University Of California Biodegradable organic polymer delivery system for bone morphogenetic protein
US4576608A (en) 1980-11-06 1986-03-18 Homsy Charles A Porous body-implantable polytetrafluoroethylene
US4595713A (en) 1985-01-22 1986-06-17 Hexcel Corporation Medical putty for tissue augmentation
US4609551A (en) 1984-03-20 1986-09-02 Arnold Caplan Process of and material for stimulating growth of cartilage and bony tissue at anatomical sites
DE3518150C1 (en) 1985-05-21 1986-10-23 Karl Prof. Dr.med. 7302 Ostfildern Theurer Process for the production of suspendible corpuscular particles of adhesive materials with molecular long-fibre crosslinking
US4627853A (en) 1985-05-29 1986-12-09 American Hospital Supply Corporation Method of producing prostheses for replacement of articular cartilage and prostheses so produced
US4637931A (en) 1984-10-09 1987-01-20 The United States Of America As Represented By The Secretary Of The Army Polyactic-polyglycolic acid copolymer combined with decalcified freeze-dried bone for use as a bone repair material
US4642120A (en) 1983-03-23 1987-02-10 Ramot University Authority For Applied Research And Industrial Development Ltd. Repair of cartilage and bones
US4645669A (en) 1982-10-04 1987-02-24 Albert Einstein College Of Medicine Of Yeshiva University Culturing and emplacement of differentiated cells in vivo
US4675189A (en) 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
US4675284A (en) 1984-08-22 1987-06-23 Leevy Carroll M Process and apparatus for evaluating liver disease
US4681763A (en) 1985-06-11 1987-07-21 University Of Medicine And Dentistry Of New Jersey Composition for stimulating bone growth
US4689293A (en) 1983-06-06 1987-08-25 Connaught Laboratories Limited Microencapsulation of living tissue and cells
WO1987006120A1 (en) 1986-04-18 1987-10-22 Marrow-Tech Incorporated Process for replicating bone marrow
US4713070A (en) 1978-11-30 1987-12-15 Sumitom Electric Industries, Ltd. Porous structure of polytetrafluoroethylene and process for production thereof
US4721096A (en) 1986-04-18 1988-01-26 Marrow-Tech Incorporated Process for replicating bone marrow in vitro and using the same
US4734373A (en) 1986-06-24 1988-03-29 Bartal Arie H Apparatus for enhancing cell growth, preservation and transport
WO1988003785A1 (en) 1986-11-20 1988-06-02 Vacanti Joseph P Chimeric neomorphogenesis of organs by controlled cellular implantation using artificial matrices
US4757128A (en) 1986-08-01 1988-07-12 Massachusetts Institute Of Technology High molecular weight polyanhydride and preparation thereof
US4757017A (en) 1984-09-14 1988-07-12 Mcw Research Foundation, Inc. In vitro cell culture system
EP0282746A1 (en) 1987-02-19 1988-09-21 Takeda Chemical Industries, Ltd. Method for producing artificial cultured tissue
US4778749A (en) 1984-06-01 1988-10-18 Karyon Technology, Inc. Tissue culture and production in permeable gels
US4795459A (en) * 1987-05-18 1989-01-03 Rhode Island Hospital Implantable prosthetic device with lectin linked endothelial cells
WO1989000413A1 (en) 1987-07-20 1989-01-26 Stone Kevin R Prosthetic meniscus
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
US4846835A (en) 1987-06-15 1989-07-11 Grande Daniel A Technique for healing lesions in cartilage
US4853324A (en) 1985-12-02 1989-08-01 Viles Joseph M Liver assist device employing transformed cell lines
WO1989007944A1 (en) 1988-02-24 1989-09-08 American National Red Cross Device for site directed neovascularization and method for same
US4868121A (en) 1985-02-07 1989-09-19 Mcdonnell Douglas Corporation Islet isolation process
US4880622A (en) 1986-05-20 1989-11-14 Research Corporation Technologies, Inc. Water-soluble phosphazene polymers having pharmacological applications
US4886870A (en) 1984-05-21 1989-12-12 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US4888176A (en) 1984-05-21 1989-12-19 Massachusetts Institute Of Technology Controlled drug delivery high molecular weight polyanhydrides
US4891225A (en) 1984-05-21 1990-01-02 Massachusetts Institute Of Technology Bioerodible polyanhydrides for controlled drug delivery
US4902289A (en) * 1982-04-19 1990-02-20 Massachusetts Institute Of Technology Multilayer bioreplaceable blood vessel prosthesis
EP0344924A3 (en) 1988-06-02 1990-03-07 Organogenesis Inc. Fibrin-collagen tissue equivalents and methods of preparation thereof
EP0361957A1 (en) 1988-09-30 1990-04-04 Organogenesis Inc. Tissue equivalents and methods of preparation thereof
US4916193A (en) * 1987-12-17 1990-04-10 Allied-Signal Inc. Medical devices fabricated totally or in part from copolymers of recurring units derived from cyclic carbonates and lactides
US4946938A (en) 1989-08-01 1990-08-07 The University Of Pittsburgh A process for the catalytic synthesis of polyphosphazenes
EP0248246B1 (en) 1986-06-02 1990-09-19 GebràœDer Sulzer Aktiengesellschaft Vascular graft wall
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
WO1990012604A1 (en) 1989-04-25 1990-11-01 Vacanti Joseph P Method for implanting large volumes of cells on polymeric matrices
WO1990012603A1 (en) 1989-04-17 1990-11-01 Vacanti Joseph P Neomorphogenesis of cartilage in vivo from cell culture
US4988761A (en) 1988-09-22 1991-01-29 Dow Corning K.K. Process for producing a low water content PVA hydrogel
WO1991001720A1 (en) 1989-08-07 1991-02-21 Herman Wade Schlameus Composition and method of promoting hard tissue healing
US5032508A (en) 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5078744A (en) 1987-09-04 1992-01-07 Bio-Products, Inc. Method of using tendon/ligament substitutes composed of long, parallel, non-antigenic tendon/ligament fibers
EP0248247B1 (en) 1986-06-02 1992-01-22 GebràœDer Sulzer Aktiengesellschaft Vascular graft wall
WO1992006702A1 (en) 1990-10-19 1992-04-30 New York University A method for transplanting cells into the brain and therapeutic uses therefor
WO1992007525A1 (en) 1990-10-31 1992-05-14 Baxter International Inc. Close vascularization implant material
EP0153896B1 (en) 1984-02-21 1992-05-27 BIOETICA, Société Anonyme Biocompatible implantable containers based on collagen for the conservation and/or the cultivation of cells and/or the controlled liberation of active compounds
EP0339607B1 (en) 1988-04-29 1993-02-10 Samuel Dr. Itay Composition for repair of cartilage and bone and method for their preparation as skeletal tissue implant
WO1993007913A1 (en) 1991-10-24 1993-04-29 Children's Medical Center Corporation Neomorphogenesis of urological structures in vivo from cell culture
US5207705A (en) * 1988-12-08 1993-05-04 Brigham And Women's Hospital Prosthesis of foam polyurethane and collagen and uses thereof
WO1993008850A1 (en) 1991-10-30 1993-05-13 Massachusetts Institute Of Technology Prevascularized polymeric implants for organ transplantation
US5219361A (en) 1988-09-16 1993-06-15 Clemson University Soft tissue implant with micron-scale surface texture to optimize anchorage
WO1993016687A1 (en) 1992-02-28 1993-09-02 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
EP0226061B1 (en) 1985-12-17 1994-02-16 United States Surgical Corporation High molecular weight bioresorbable polymers and implantation devices thereof
US5324519A (en) 1989-07-24 1994-06-28 Atrix Laboratories, Inc. Biodegradable polymer composition
WO1994021299A1 (en) 1993-03-19 1994-09-29 Medinvent A composition and a method for tissue augmentation
WO1994025079A1 (en) 1993-04-23 1994-11-10 Massachusetts Institute Of Technology Porous biodegradable polymeric materials for cell transplantation
US5512600A (en) 1993-01-15 1996-04-30 Massachusetts Institute Of Technology Preparation of bonded fiber structures for cell implantation
US5514378A (en) * 1993-02-01 1996-05-07 Massachusetts Institute Of Technology Biocompatible polymer membranes and methods of preparation of three dimensional membrane structures
US5709854A (en) * 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
US5720981A (en) 1985-08-14 1998-02-24 Sloan-Kettering Institute For Cancer Research Epidermal cell extracts and method to enhance wound healing and regenerate epidermis
US5772695A (en) 1991-03-05 1998-06-30 Colorado State University Research Foundation Treated tissue for implantation and methods of treatment and use
US5855608A (en) 1994-05-13 1999-01-05 Thm Biomedical, Inc. Device and methods for in vivo culturing of diverse tissue cells

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE511546A (en) * 1951-10-23
US3531551A (en) * 1967-06-09 1970-09-29 American Cyanamid Co Dielectric curing of spandex
GB1279915A (en) * 1969-03-18 1972-06-28 Knoll Ag 0-aryl-2,3,4,5-tetrahydro-1h-1,5-benzodiazepine-2-ones and a process for their preparation
DE2406693C3 (en) * 1974-02-13 1981-01-22 Benteler-Werke Ag Werk Neuhaus, 4790 Paderborn Device for the transmission of direct current of high amperage and low harmonics to the rotating electrode disk during roller seam resistance butt welding
GB1522603A (en) * 1974-08-07 1978-08-23 Nat Res Dev Prosthetic devices
FR2435050A1 (en) * 1978-08-29 1980-03-28 Commissariat Energie Atomique DEVICE FOR ANALYZING THE ACTIVITY OF A RADIOACTIVE AEROSOL AS A FUNCTION OF THE GRANULOMETRY OF ITS PARTICLES
US4494386A (en) * 1982-03-15 1985-01-22 Rovac Corporation Vapor refrigeration cycle system with constrained rotary vane compressor and low vapor pressure refrigerant
SE430669B (en) * 1982-04-30 1983-12-05 Lindskog K J E SET FOR MANUFACTURING FIBER REFINED PLASTIC BODIES AND APPARATUS FOR USE IN SUCH MANUFACTURING
DE3318150C2 (en) 1983-05-18 1985-07-11 Vsesojuznyj zaočnyj politechničeskij institut, Moskva Working container for blow molding and sand shooting machines
FR2560490B1 (en) 1984-03-02 1986-12-19 Boton Freres CUTTING HEAD WITH MEANS OF REGULATING THE CUTTING PLANE FOR MATERIAL
US4732636A (en) 1986-05-23 1988-03-22 International Business Machines Corporation Method for producing a prepreg having improved dimensional stability

Patent Citations (142)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1995970A (en) 1931-04-04 1935-03-26 Du Pont Polymeric lactide resin
US2609347A (en) 1948-05-27 1952-09-02 Wilson Christopher Lumley Method of making expanded polyvinyl alcohol-formaldehyde reaction product and product resulting therefrom
US2664366A (en) 1949-09-19 1953-12-29 Wilson Christopher Lumley Plasticized sponge material and method of making same
US2659935A (en) 1950-03-18 1953-11-24 Christopher L Wilson Method of making compressed sponges
US2653917A (en) 1950-06-15 1953-09-29 Christopher L Wilson Method of making an expanded material and the product resulting therefrom
US2676945A (en) 1950-10-18 1954-04-27 Du Pont Condensation polymers of hydroxyacetic acid
US2683136A (en) 1950-10-25 1954-07-06 Du Pont Copolymers of hydroxyacetic acid with other alcohol acids
US2703316A (en) 1951-06-05 1955-03-01 Du Pont Polymers of high melting lactide
US2758987A (en) 1952-06-05 1956-08-14 Du Pont Optically active homopolymers containing but one antipodal species of an alpha-monohydroxy monocarboxylic acid
US2846407A (en) 1954-01-13 1958-08-05 Wilson Christopher Lumley Method of making a detergent and solvent resistant sponge material
US2951828A (en) 1957-03-04 1960-09-06 Boehringer Sohn Ingelheim Process for the production of bead polymerizates from cyclic internal esters of alpha-hydroxy carboxylic acids
US3531561A (en) 1965-04-20 1970-09-29 Ethicon Inc Suture preparation
US3514791A (en) 1967-07-25 1970-06-02 Charles H Sparks Tissue grafts
US3880991A (en) 1969-03-24 1975-04-29 Brook David E Polymeric article for dispensing drugs
US4069307A (en) 1970-10-01 1978-01-17 Alza Corporation Drug-delivery device comprising certain polymeric materials for controlled release of drug
US3935065A (en) 1971-09-02 1976-01-27 Roland Karl Doerig Procedure for conservation of living organs and apparatus for the execution of this procedure
US3960150A (en) 1971-09-09 1976-06-01 Alza Corporation Bioerodible ocular device
US4026304A (en) 1972-04-12 1977-05-31 Hydro Med Sciences Inc. Bone generating method and device
US3883393A (en) 1972-05-18 1975-05-13 Us Health Education & Welfare Cell culture on semi-permeable tubular membranes
US3826241A (en) 1972-10-16 1974-07-30 Investors In Ventures Inc Implanting method
US4450150A (en) 1973-05-17 1984-05-22 Arthur D. Little, Inc. Biodegradable, implantable drug delivery depots, and method for preparing and using the same
US3974526A (en) 1973-07-06 1976-08-17 Dardik Irving I Vascular prostheses and process for producing the same
US3992725A (en) 1973-11-16 1976-11-23 Homsy Charles A Implantable material and appliances and method of stabilizing body implants
US3902497A (en) 1974-03-25 1975-09-02 American Cyanamid Co Body absorbable sponge and method of making
US4192827A (en) 1974-06-27 1980-03-11 Ciba-Geigy Corporation Water-insoluble hydrophilic copolymers
US3995444A (en) 1974-11-08 1976-12-07 American Hospital Supply Corporation Organ perfusion system
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
US4144126A (en) 1975-05-21 1979-03-13 Beecham Group Limited Cell culture method
US4060081A (en) 1975-07-15 1977-11-29 Massachusetts Institute Of Technology Multilayer membrane useful as synthetic skin
US4280954A (en) 1975-07-15 1981-07-28 Massachusetts Institute Of Technology Crosslinked collagen-mucopolysaccharide composite materials
US4186448A (en) 1976-04-16 1980-02-05 Brekke John H Device and method for treating and healing a newly created bone void
US4391797A (en) 1977-01-05 1983-07-05 The Children's Hospital Medical Center Systems for the controlled release of macromolecules
US4141087A (en) 1977-01-19 1979-02-27 Ethicon, Inc. Isomorphic copolyoxalates and sutures thereof
US4328204A (en) 1977-03-02 1982-05-04 Ethicon, Inc. Absorbable polymer-drug compounds and method for making same
US4205399A (en) 1977-06-13 1980-06-03 Ethicon, Inc. Synthetic absorbable surgical devices of poly(alkylene oxalates)
US4137921A (en) 1977-06-24 1979-02-06 Ethicon, Inc. Addition copolymers of lactide and glycolide and method of preparation
DE2853614A1 (en) 1978-01-25 1979-07-26 Bentley Lab IMPLANT
US4304591A (en) 1978-01-25 1981-12-08 Ciba-Geigy Corporation Water-insoluble hydrophilic copolymers used as carriers for medicaments and pesticides
US4277582A (en) 1978-03-03 1981-07-07 Ciba-Geigy Corporation Water-insoluble hydrophilic copolymers
US4228243A (en) 1978-07-13 1980-10-14 Toray Industries, Inc. Cell culture propagation apparatus
US4239664A (en) 1978-10-31 1980-12-16 Research Corporation Anti-thrombogenic PVP-heparin polymer
US4243775A (en) 1978-11-13 1981-01-06 American Cyanamid Company Synthetic polyester surgical articles
US4456687A (en) 1978-11-16 1984-06-26 President And Fellows Of Harvard College Agents for promoting growth of epithelial cells
US4713070A (en) 1978-11-30 1987-12-15 Sumitom Electric Industries, Ltd. Porous structure of polytetrafluoroethylene and process for production thereof
US4352883A (en) 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4304866A (en) 1979-11-14 1981-12-08 Massachusetts Institute Of Technology Transplantable sheets of living keratinous tissue
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4348329A (en) 1979-12-20 1982-09-07 Dennis Chapman Biocompatible surfaces
US4356261A (en) 1980-04-22 1982-10-26 Rush-Presbyterian-St. Luke's Medical Center Anti-invasion factor containing cultures
US4347847A (en) 1980-06-06 1982-09-07 Usher Francis C Method of hernia repair
US4427808A (en) 1980-07-21 1984-01-24 Ceskoslovenska Akademie Ved Composite polymeric material for biological and medical applications and the method for its preparation
US4563490A (en) 1980-07-21 1986-01-07 Czechoslovenska Akademie Ved Of Praha Composite polymeric material for biological and medical application and the method for its preparation
US4576608A (en) 1980-11-06 1986-03-18 Homsy Charles A Porous body-implantable polytetrafluoroethylene
US4675189A (en) 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
US4416986A (en) 1981-01-16 1983-11-22 Merck & Co., Inc. Methods of producing HBsAg
US4553272A (en) 1981-02-26 1985-11-19 University Of Pittsburgh Regeneration of living tissues by growth of isolated cells in porous implant and product thereof
US4438198A (en) 1981-09-30 1984-03-20 Trimedyne, Inc. Biochemically active matrix for use in a bio-artificial organ
US4431428A (en) 1981-09-30 1984-02-14 Trimedyne, Inc. Bio-artificial organ using microencapsulated enzymes
US4446234A (en) 1981-10-23 1984-05-01 The United States Of America As Represented By The Department Of Health And Human Services Vitro cellular interaction with amnion membrane substrate
US4458678A (en) 1981-10-26 1984-07-10 Massachusetts Institute Of Technology Cell-seeding procedures involving fibrous lattices
US4505266A (en) * 1981-10-26 1985-03-19 Massachusetts Institute Of Technology Method of using a fibrous lattice
US4485096A (en) 1982-02-26 1984-11-27 Massachusetts Institute Of Technology Tissue-equivalent and method for preparation thereof
US4439152A (en) 1982-03-04 1984-03-27 Small Irwin A Method of jawbone abutment implant for dental prostheses and implant device
US4545082A (en) 1982-03-25 1985-10-08 Vascutek Limited Vascular prosthesis
US4902289A (en) * 1982-04-19 1990-02-20 Massachusetts Institute Of Technology Multilayer bioreplaceable blood vessel prosthesis
US4520821A (en) * 1982-04-30 1985-06-04 The Regents Of The University Of California Growing of long-term biological tissue correction structures in vivo
US4528265A (en) 1982-05-11 1985-07-09 Becker Robert O Processes and products involving cell modification
US4485097A (en) 1982-05-26 1984-11-27 Massachusetts Institute Of Technology Bone-equivalent and method for preparation thereof
US4494385A (en) 1982-06-04 1985-01-22 Hoxan Corporation Method of preserving organ and apparatus for preserving the same
US4495174A (en) 1982-06-21 1985-01-22 Research Corporation Anesthetic polyorganophosphazenes
US4440921A (en) 1982-06-21 1984-04-03 Research Corporation Coupling of polyorganophosphazenes to carboxylic acid
US4489056A (en) 1982-06-30 1984-12-18 Merck & Co., Inc. Acid anhydrides as rate controlling agent for the erosion of polymers which latter polymers have beneficial substances dispersed throughout their matrix or where the polymer matrix surrounds the beneficial substance
US4544516A (en) 1982-07-28 1985-10-01 Battelle Development Corporation Collagen orientation
US4559304A (en) 1982-08-09 1985-12-17 Koken Co., Ltd. Substratum for cell culture and a method for culturing and isolating cells using same
US4645669A (en) 1982-10-04 1987-02-24 Albert Einstein College Of Medicine Of Yeshiva University Culturing and emplacement of differentiated cells in vivo
US4559298A (en) 1982-11-23 1985-12-17 American National Red Cross Cryopreservation of biological materials in a non-frozen or vitreous state
US4446229A (en) 1982-12-30 1984-05-01 Indech Robert B Method of tissue growth
US4642120A (en) 1983-03-23 1987-02-10 Ramot University Authority For Applied Research And Industrial Development Ltd. Repair of cartilage and bones
US4689293A (en) 1983-06-06 1987-08-25 Connaught Laboratories Limited Microencapsulation of living tissue and cells
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
US4563489A (en) 1984-02-10 1986-01-07 University Of California Biodegradable organic polymer delivery system for bone morphogenetic protein
EP0153896B1 (en) 1984-02-21 1992-05-27 BIOETICA, Société Anonyme Biocompatible implantable containers based on collagen for the conservation and/or the cultivation of cells and/or the controlled liberation of active compounds
US4609551A (en) 1984-03-20 1986-09-02 Arnold Caplan Process of and material for stimulating growth of cartilage and bony tissue at anatomical sites
US4888176A (en) 1984-05-21 1989-12-19 Massachusetts Institute Of Technology Controlled drug delivery high molecular weight polyanhydrides
US4886870A (en) 1984-05-21 1989-12-12 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US4891225A (en) 1984-05-21 1990-01-02 Massachusetts Institute Of Technology Bioerodible polyanhydrides for controlled drug delivery
US4778749A (en) 1984-06-01 1988-10-18 Karyon Technology, Inc. Tissue culture and production in permeable gels
US4675284A (en) 1984-08-22 1987-06-23 Leevy Carroll M Process and apparatus for evaluating liver disease
US4757017A (en) 1984-09-14 1988-07-12 Mcw Research Foundation, Inc. In vitro cell culture system
US4637931A (en) 1984-10-09 1987-01-20 The United States Of America As Represented By The Secretary Of The Army Polyactic-polyglycolic acid copolymer combined with decalcified freeze-dried bone for use as a bone repair material
US4563350A (en) 1984-10-24 1986-01-07 Collagen Corporation Inductive collagen based bone repair preparations
US4595713A (en) 1985-01-22 1986-06-17 Hexcel Corporation Medical putty for tissue augmentation
US4868121A (en) 1985-02-07 1989-09-19 Mcdonnell Douglas Corporation Islet isolation process
DE3518150C1 (en) 1985-05-21 1986-10-23 Karl Prof. Dr.med. 7302 Ostfildern Theurer Process for the production of suspendible corpuscular particles of adhesive materials with molecular long-fibre crosslinking
US4627853A (en) 1985-05-29 1986-12-09 American Hospital Supply Corporation Method of producing prostheses for replacement of articular cartilage and prostheses so produced
US4681763A (en) 1985-06-11 1987-07-21 University Of Medicine And Dentistry Of New Jersey Composition for stimulating bone growth
US5720981A (en) 1985-08-14 1998-02-24 Sloan-Kettering Institute For Cancer Research Epidermal cell extracts and method to enhance wound healing and regenerate epidermis
US4853324A (en) 1985-12-02 1989-08-01 Viles Joseph M Liver assist device employing transformed cell lines
EP0226061B1 (en) 1985-12-17 1994-02-16 United States Surgical Corporation High molecular weight bioresorbable polymers and implantation devices thereof
WO1987006120A1 (en) 1986-04-18 1987-10-22 Marrow-Tech Incorporated Process for replicating bone marrow
US4721096A (en) 1986-04-18 1988-01-26 Marrow-Tech Incorporated Process for replicating bone marrow in vitro and using the same
US4880622A (en) 1986-05-20 1989-11-14 Research Corporation Technologies, Inc. Water-soluble phosphazene polymers having pharmacological applications
EP0248247B1 (en) 1986-06-02 1992-01-22 GebràœDer Sulzer Aktiengesellschaft Vascular graft wall
EP0248246B1 (en) 1986-06-02 1990-09-19 GebràœDer Sulzer Aktiengesellschaft Vascular graft wall
US4734373A (en) 1986-06-24 1988-03-29 Bartal Arie H Apparatus for enhancing cell growth, preservation and transport
US4757128A (en) 1986-08-01 1988-07-12 Massachusetts Institute Of Technology High molecular weight polyanhydride and preparation thereof
US5770193A (en) * 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Preparation of three-dimensional fibrous scaffold for attaching cells to produce vascularized tissue in vivo
WO1988003785A1 (en) 1986-11-20 1988-06-02 Vacanti Joseph P Chimeric neomorphogenesis of organs by controlled cellular implantation using artificial matrices
US5041138A (en) 1986-11-20 1991-08-20 Massachusetts Institute Of Technology Neomorphogenesis of cartilage in vivo from cell culture
EP0282746A1 (en) 1987-02-19 1988-09-21 Takeda Chemical Industries, Ltd. Method for producing artificial cultured tissue
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US4795459A (en) * 1987-05-18 1989-01-03 Rhode Island Hospital Implantable prosthetic device with lectin linked endothelial cells
US4846835A (en) 1987-06-15 1989-07-11 Grande Daniel A Technique for healing lesions in cartilage
WO1989000413A1 (en) 1987-07-20 1989-01-26 Stone Kevin R Prosthetic meniscus
AU2424588A (en) 1987-07-20 1989-02-13 Kevin R. Stone Prosthetic meniscus
US5078744A (en) 1987-09-04 1992-01-07 Bio-Products, Inc. Method of using tendon/ligament substitutes composed of long, parallel, non-antigenic tendon/ligament fibers
US4916193A (en) * 1987-12-17 1990-04-10 Allied-Signal Inc. Medical devices fabricated totally or in part from copolymers of recurring units derived from cyclic carbonates and lactides
WO1989007944A1 (en) 1988-02-24 1989-09-08 American National Red Cross Device for site directed neovascularization and method for same
EP0339607B1 (en) 1988-04-29 1993-02-10 Samuel Dr. Itay Composition for repair of cartilage and bone and method for their preparation as skeletal tissue implant
EP0344924A3 (en) 1988-06-02 1990-03-07 Organogenesis Inc. Fibrin-collagen tissue equivalents and methods of preparation thereof
US5032508A (en) 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5219361A (en) 1988-09-16 1993-06-15 Clemson University Soft tissue implant with micron-scale surface texture to optimize anchorage
US4988761A (en) 1988-09-22 1991-01-29 Dow Corning K.K. Process for producing a low water content PVA hydrogel
EP0361957A1 (en) 1988-09-30 1990-04-04 Organogenesis Inc. Tissue equivalents and methods of preparation thereof
US5207705A (en) * 1988-12-08 1993-05-04 Brigham And Women's Hospital Prosthesis of foam polyurethane and collagen and uses thereof
WO1990012603A1 (en) 1989-04-17 1990-11-01 Vacanti Joseph P Neomorphogenesis of cartilage in vivo from cell culture
WO1990012604A1 (en) 1989-04-25 1990-11-01 Vacanti Joseph P Method for implanting large volumes of cells on polymeric matrices
US5324519A (en) 1989-07-24 1994-06-28 Atrix Laboratories, Inc. Biodegradable polymer composition
US4946938A (en) 1989-08-01 1990-08-07 The University Of Pittsburgh A process for the catalytic synthesis of polyphosphazenes
WO1991001720A1 (en) 1989-08-07 1991-02-21 Herman Wade Schlameus Composition and method of promoting hard tissue healing
WO1992006702A1 (en) 1990-10-19 1992-04-30 New York University A method for transplanting cells into the brain and therapeutic uses therefor
WO1992007525A1 (en) 1990-10-31 1992-05-14 Baxter International Inc. Close vascularization implant material
US5772695A (en) 1991-03-05 1998-06-30 Colorado State University Research Foundation Treated tissue for implantation and methods of treatment and use
WO1993007913A1 (en) 1991-10-24 1993-04-29 Children's Medical Center Corporation Neomorphogenesis of urological structures in vivo from cell culture
WO1993008850A1 (en) 1991-10-30 1993-05-13 Massachusetts Institute Of Technology Prevascularized polymeric implants for organ transplantation
WO1993016687A1 (en) 1992-02-28 1993-09-02 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5512600A (en) 1993-01-15 1996-04-30 Massachusetts Institute Of Technology Preparation of bonded fiber structures for cell implantation
US5514378A (en) * 1993-02-01 1996-05-07 Massachusetts Institute Of Technology Biocompatible polymer membranes and methods of preparation of three dimensional membrane structures
WO1994021299A1 (en) 1993-03-19 1994-09-29 Medinvent A composition and a method for tissue augmentation
WO1994025079A1 (en) 1993-04-23 1994-11-10 Massachusetts Institute Of Technology Porous biodegradable polymeric materials for cell transplantation
US5709854A (en) * 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
US5855608A (en) 1994-05-13 1999-01-05 Thm Biomedical, Inc. Device and methods for in vivo culturing of diverse tissue cells

Non-Patent Citations (209)

* Cited by examiner, † Cited by third party
Title
"Brain Graft Seeks to Relieve Huntington Disease Patient," New York Times (Mar. 4, 1988).
Allcock & Kwon, "An Ionically Cross-Linkable Polyphosphazene: Poly[bis(carboxylatophenoxy)phosphazene] and Its Hydrogels and Membranes," Macromolecules 22:75-79 (1989).
Allcock & Kwon, "Glyceryl Phosphazenes: Synthesis, Properties, and Hydrolysis," Macromolecules 21(7):1980-1985 (1988).
Allcock & Scopelianos, "Synthesis of Sugar-Substituted Cyclic and Polymeric Phosphazenes and Their Oxidation, Reduction, and Acetylation Reactions," Macromolecules 16(5)715-719 (1983).
Allcock, H. R., et al., "Amphilphilic Polyphosphazenes as Membrane Materials: Influence of Side Group on Radiation Cross Linking," Biomaterials 9(6):500-508 (Nov. 1988).
Allcock, H. R., et al., "Phosphonitrilic Compounds. IV. High Molecular Weight Poly(bis(amino)phosphazenes) and Mixed-Substituent Poly(aminophosphazenes)," Inorg. Chem. 11(11):2584-2590 (1972).
Allcock, H. R., et al., "Polyphosphazenes with Etheric Side Groups: Prospective Biomedical and Solid Electrolyte Polymers," Macromolecules 19:1508-1512 (1986).
Allcock, H. R., et al., "Synthesis of Poly[(Amino Acid Alkyl Ester)Phosphazenes]1-3", Macromolecules, 10(4) (Jul./Aug. 1977).
Allcock, H.R., et al., "Hydrolysis Pathways for Aminophosphazenes", Inorg. Chem. 21(1):515-521 (Jan. 1982).
Atala and Casale, "Management of Primary Vesicoureteral Reflux," Infections in Urology pp. 39-43 (Mar./Apr. 1990).
Atala, "Endoscopic Treatment of Reflux with Autologous Bladder Muscle Cells," American Academy of Pediatrics Meeting held in Dalls, TX Oct. 23, 1994 (Abstract).
Atala, "Laparoscopic Treatment of Vesicoureteral Reflux," Dia. Ped. Urol. 14:212 (1993).
Atala, et al., "Endoscopic Treatment of Vesicoureteral Reflux with a Chondrocyte-Alginate Suspension," The Journal of Urology 152:641-643 (Aug. 1994).
Atala, et al., "Endoscopic Treatment of Vesicoureteral Reflux with a Self-Detachable Balloon System," The Journal of Urology 148:724-728 (Aug. 1992).
Atala, et al., "Formation of Urothelial Structures in Vivo from Dissociated Cells Attached to Biodegradable Polymer Scaffolds in Vitro," The Journal of Urology 148:658-662 (Aug. 1992).
Atala, et al., "Implantation in Vivo and Retrieval of Artificial Structures Consisting of Rabbit and Human Urothelium and Human Bladder Muscle," The Journal of Urology 150:608-612 (Aug. 1993).
Atala, et al., "Injectable Alginate Seeded with Chondrocytes as a Potential Treatment for Vesicoureteral Reflux," The Journal of Urology 150:745-747 (Aug. 1993).
Atala, et al., "Laparoscopic Correction of Vesicoureteral Reflux," The Journal of Urology 150:748-751 (Aug. 1983).
Atala, et al., "Sonography with Sonicated Albumin in the Detection of Vesicoureteral Reflux," The Journal of Urology 150:756-758 (Aug. 1993).
Baklund, et al. "Toward a Transplantation Therapy in Parkinson's Disease," Annals of the N.Y. Acad. of Sci. 495:658-673 (1987).
Bazeed, et al. "New Surgical Procedure for Management of Peyronie Disease," Urology 21(5), 501-504 (1983).
Bennett & Hirt, "A History of Tissue Expansion," Dermatol. Surg. Oncol. 19:1066-1073 (1993).
Ben-Ze'Ev, et al. "Cell-Cell and Cell-Matrix Interactions Differentially Regulate the Expression of Hepatic and Cytoskeletal Genes in Primary Cultures of Rat Hepatocytes," Proc. Natl. Acad. Sci. USA 85:2161-2165 (Apr. 1988).
Berrino, et al. "Surgical Correction of Breast Deformities Following Long-Lasting Complications of Polyurethane-Covered Implants," Ann. Plast. Surg., 24:481 (1990).
Biers, "Organogenesis' Human Artery Equivalent May Revolutionize Vascular Grafts," Genetic Engineering News (Nov./Dec. 1987).
Bissell, "Support of Cultured Hepatocytes by a Laminin-Rich Gel," J. Clin. Invest. 79:801-812 (1987).
Bissell, et al. "The Role of Extracellular Matrix in Normal Liver," Scand. J. Gastroenterol. 23:107 (1988).
Bissell, et al., "Interactions of Rat Hepatocytes with Type IV Collagen, Fibronectin and Laminin Matrices, Distinct Matrix-Controlled Modes of Attachment and Spreading," European Journ. of Cell Biology 40:72-78 (1986).
Bjorklund, Annals of the N.Y. Academy of Science 495:676-686 (1987).
Blaivas, et al., "When Sphincter Failure is the Cause of Female Stress Incontinence," Contemporary Urology 5(3):33-54 (Mar. 1993).
Bohn, et al., "Adrenal Medulla Grafts Enhance Recovery of Striatal Dopaminergic Fibers," Science 238(4817):913-916 (Aug. 21, 1987).
Breuer, et al., "Tissue Engineering Heart Valves," American Chemical Society Spring Meeting, (Apr. 2-6, 1995).
Brown, "Fibrin-Collagen Nerve Repair Device," Inventors: Russ Griffiths, Larry Stensaas & Ken Horch, Letter dated May 10, 1988.
Burke, "The Effects of the Configuration of an Artificial Extracellular Matrix on the Development of a Functional Dermis," The Role of Extracellular Matrix in Development 351-355 (Alan R. Liss, Inc., NY 1984).
Cao, at al., "Bone Reconstruction with Tissue Engineered Vascularized Bone," (Abstract) Apr. 30-May 3, 1995.
Cao, et al., "The Generation of Neo-Tendon Using Synthetic Polymers seeded with Tenocytes," Transplantation Proceedings, 26(6):3390-3392 (1994).
Chaikin, "Tissue Engineering: Science Non-Fiction," Medical Industry Executive pp. 6-7 (May 1993).
Chuang, et al., "Sheath Needle for Liver Biopsy in High-Risk Patients," RSNA pp. 261-262 (1988).
Cilento, et al., "Phenotypic and Cytogenetic Characterization of Human Bladder Urothelia Expanded in Vitro," Microbiology & Immunology 152:665-670 (Aug. 1994).
Claes, et al., "Pulmonary Migration Following Periurethral Polytetrafluorethylene Injection for Urinary Incontinence," The Journal of Urology 142:821-822 (Sep. 1989).
Cohen, "Navigating Through Tissue Expansion Terminology," J. Dermatol. Surg. Oncol. 19:614-615 (1993).
Collier, et al., "Norepinephrine Deficiency and Behavioral Senescence in Aged Rats: Transplanted Locus Ceruleus Neurons as an Experimental Replacement Therapy," Annals of the New York Academy of Science 495:396-403 (1987).
Cosimi, et al., "Transplantation of Skin," Surgical Clinics of N.A. 58(2), 435-451 (Apr. 1978).
Culliton, "Gore Tex Organoids and Genetic Drugs," Science 246:747-749 (1989).
Da Silva, "An In Vivo Model to Quantify Motor and Sensory Peripheral Nerve Regeneration Using Bioresorbable Nerve Guide Tubes," Brain Research, 342:307-315 (1985).
Davis., et al., "Human Amnion Membrane Serves as a Substratum for Growing Axons in Vitro and in Vivo," Science 236:1106-1109 (May 29, 1987).
Del Cerro, et al., "Retinal Transplants into One Anterior Chamber of the Rat Eye," Neuroscience 21:(3)707-23 (Jun. 1987).
Doillon, et al., "Collagen-Based Wound Dressings: Control of the Pore Structure and Morphology," Journal of Biomedical Materials Research, 20:1219-1228 (1986).
Doillon, et al., "Epidermal Cells Cultured on a Collagen-Based Material," G. W. Bailey, Editor, Proceedings of the 44th Annual Meeting of the Electron Microscopy Society of America, (1986).
Ebata, et al. "Liver Regeneration Utilizing Isolated Hepatocytes Transplanted into the Rat Spleen," Surg. Forum 29: 338-340 (1978).
Elkowitz, et al., "Various Methods of Breast Reconstruction After Mastectomy: An Economic Comparison," Plastic and Reconstructive Surgery, 92(1):77-83 (Jul. 1993).
Erickson, "Material Help," Scientific American pp. 114-116 (Aug. 1992).
Ferro, et al., "Periurethral Granuloma: Unusual Complication of Teflon Periurethral Injection," Urology 31(5):422-423 (May 1988).
Folkman, et al., "Angiogenic Factors," Science 235:442-447 (Jan. 23, 1987).
Fontaine, et al., "Optimization Studies on Retroviral Mediated Gene Transfer into Rat Hepatocytes: Implications for Gene Therapy," The Society of University Surgeons, Resident's Program, Cincinnati, Ohio (Feb. 15, 1992).
Fontaine, et al., "Transplantation of Genetically Altered Hepatocytes Using Cell-Polymer Constructs," Transplantation Proceedings, 25:1002-1004 (Feb. 1993).
Freshney, "The Culture Environment: I. Substrate, Gas Phase, and Temperature," Culture of Animal Cells pp. 55-56 (Alan R. Liss, NY 1983).
Gash, "Neural Transplantation: Potential Therapy for Alzheimer's Disease," J. Neural Trans. [ Suppl] 24:301-8 (1987).
Gash, et al., "Amitiotic Neuroblastoma Cells Used for Neural Implants in Monkeys," Science 233(4771):1420-2 (Sep. 26, 1986).
Geiss, et al., "Multicenter Survey of Endoscopic Treatment of Vesidoureteral Reflux in Children," Eur. Urol 17:328-329 (1990).
Gilbert, et al., "Cell Transplantation of Genetically Altered Cells on Biodegradable Polymer Scaffolds in Syngeneic Rats," Transplantation, 56(2): 423-427 (Aug. 1993).
Grande, et al., "Healing of Experimentally Produced Lesions in Articular Cartilage Following Chondrocyte Transplantation," The Anatomical Record 218:142-148 (1987).
Grande, et al., "The Repair of Experimentally Produced Defects in Rabbit Articular Cartilage by Autologous Chondrycyte Transplantation," (May 11, 1988).
Green, "Growth of Cultured Human Epidermal Cells into Multiple Epithelia Suitable for Grafting," Proc. Natl. Acad. Sci. USA 76(11):5665-5668 (Nov. 1979).
Grolleman, et al., "Studies on a Bioerodible Drug Carrier System Based on Polyphosphazene," Journal of Controlled Release 3:143-154 (1986).
Groth, et al., "Correction of Hyperbilirubinemia in the Glucoronyltransferase-Deficient Rat by Intraportal Hepatocyte Transplantation ," Transplant. Proc. 9:313-316 (1977).
Hammond, et al., "Morphologic Analysis of Tissue-Expander Shape Using a Biomechanical Model," Plastic and Reconstructive Surgery 92(2):255-259 (Aug. 1993).
Harris, et al. "Silicone Rubber Substrata: A New Wrinkle in the Study of Cell Locomotion," Science 208:177-179 (1980).
Hendren & Atala, "Use of Bowel for Vaginal Reconstruction," The Journal of Urology 152:752-755 (Aug. 1994).
Henly, et al., "Particulate Silicone for Use in Periurethral Injections: A Study of Local Tissue Effects and a Search for Migration," The Journal of Urology 147(4):376A (Apr. 1992).
Henry, et al., "Nerve Regeneration Through Biodegradable Polyester Tubes," Exp. Neurol. 90(3): 652-676 (Dec. 1985).
Ingber, "How Does Extracellular Matrix Control Capillary Morphogenesis?" Cell 58:803-805 (Sep. 8, 1989).
Ingber, et al., "Cells as Tensecrity Structures: Architectural Regulation of Histodifferentiation by Physical Forces Transduced Over Basement Membrane," Gene Expression During Normal and Malignant Differentiation L.-C. Anderson, et al., editors, pp. 13-32 (Academic Press, Orlando, FL 1985).
Ingber, et al., "Control of Capillary Morphogenesis: A Molecular System of Mechanical Switches," J. Cell Biol., 107:797a (1988).
Ingber, et al., "Endothelial Growth Factors and Extracellular Matrix Regulate DNA Synthesis Through Modulation of Cell and Nuclear Expansion," In Vitro Cellular and Developmental Biology 23(5):387-394 (May 1987).
Ingber, et al., "Mechanochemical Switching Between Growth Factor-Stimulated Angiogenesis in Vitro: Role of Extracellular Matrix," J. Cell. Biol., 109:317-330 (1989).
Ingber, Growth Control Through Fibronectin-Dependent Modulation of Cell Shape,: (1987). J. Cell Biol. 105:219a (1987).
Jacksic, et al., "The Use of 'Artificial Skin' for Burns," Ann. Rev. Med, 38:107-116 (1987).
Jauregyu, et al., "Attachment and Long Term Survival of Adult Rat Hepatocytes in Primary Monolayer Cultures: Comparison of Different Substrata and Tissue Culture Medial Formulations," In vitro Cellular & Development Biology, 22(1):13-22 (Jan. 1986).
Jones, "Degradation of Artificial Tissue Substrates," Cancer Invasion and Metastesis: Biologic and Therapeutic Aspects, 177-185 (Raven Press, NY 1984).
Junquiera, et al., 1989 Basic Histology, Prentice Hall, pp. 282-283 (1989).
Junquiera, et al., 1989 Basic Histology, Prentice Hall, pp. 282-283 (1989).
Kehna, et al., "Diffusion of Antibiotics Across Tissue Expanders: An in Vitro Study," Annals of Plastic Surgery 32(4);346-349 (Apr. 1994).
Klagsburn, "Large-Scale Preparation of Chondrocytes," Methods in Enzymology vol. LVIII, Academic Press, New York, 1979.
Kleinman, et al., "Use of Extracellular Matrix Components and Cell Culture," Analytical Biochemistry 166:1-13 (1987).
Klompmaker, et al, "Porous Polymer Implants for Repair of Full-Thickness Defects of Articular Cartilage: An Experimental Study in Rabbit and Dog," Biomaterials 13(9):625-634 (1992).
Kolata, "Parkinson Procedure: Fervor Turns to Disillusion," The New York Times, (Apr. 21, 1988).
Kordower, et al., "An In Vivo and In Vitro Assessment of Differentiated Neuroblastoma Cells as a Source of Donor Tissue for Transplantation," Annals of the New York Academy of Sciences, 495:606-622 (new York 1987).
Kordower, et al., "Neuroblastoma Cells in Neural Transplants: A Neuroanatomical and Behavioral Analysis," Brain Research, 417:85-98 (1987).
Kretschmer, at al, "Autotransplantation of Pancreatic Fragments to the Portal Vein and Spleen of Total Pancreatectomized Dogs," Ann. Surg. 187:79-86 (Jan. 1978).
Kretschmer, at al, "Autotransplantation of Pancreatic Fragments to the Portal Vein and Spleen of Total Pancreatectomized Dogs," Ann. Surg. 187:79-86 (Jan. 1978).
Kusano, et al., Acta Japoni Hepato 63:345-351 (1989).
Langer & Moses, "Biocompatible Controlled Release Polymers for Delivery of Polypeptides and Growth Factors," Journal of Cellular Biochemistry, 45:340-345 (1991).
Langer and Vacanti, "Tissue Engineering," Science 260:920-926 (May 14, 1993).
Leonard, et al., "Endoscopic Injection of Glutaraldehyde Cross-Linked Bovine Dermal Collagen for Correction of Vesidoureteral Reflux," The Journal of Urology 145:115-119 (Jan. 1991).
Leong, et al., "Bioerodible Polyanhydrides as Drug-Carrier Matrices. I: Characterization, Degradation, and Release Characteristics," Journal of Biomedical Materials Research 19:941-955 (1985).
Letourneau, "Possible Roles of Cell-to-Sutstratus Adhesion in Neuronal Morphogenesis," Developmental Biology, 44:77-91 (1975).
Lewin, "Cloud Over Parkinson's Therapy," Science News, 240: 390-392 (1988).
Lewin, "Disappointing Brain Graft Results," Science p. 1407 (Jun. 10, 1988).
Li, et al., Influence of a Reconstituted Basement Membrane and its Components of Casein Gene Expression and Secretion in Mouse Mammary Epithelial Cells,: Proc. Natl. Acad. Sci. USA, 84:136-140 (1987).
Lucas, et al, "Ectopic Induction of Cartilage and Bone by Water-Soluble Proteins from Bovine Bone Using a Ppolyanhydride Delivery Vehicle," Journal of Biomedical Materials Research 24(7):901-911 (1990).
Macklis, et al., "Cross-Linked Collagen Surface for Cell Culture that is Stable, Uniform, and Optically Superior to Conventional Surfaces," In Vitro Cellular & Developmental Biology, 21(3)(1): 189-194 (Mar. 1985).
Madison, at al., "Nontoxic Nerve Guide Tubes Support Neovascular Growth in Transected Rat Optic Nerve," Exp. Neurol. 86:448-461 (1984).
Madison, et al., "Increased Rate of Peripheral Nerve Regeneration Using Bioresorbable Nerve Guides and Lamin-Containing Gel," Exp. Neurol. 88(3) 767-772 (Jun. 1985).
Madison, rt al. "Peripheral Nerve Regeneration with Entubulation Repair: Comparison of Biodegradeable Nerve Guides Versus Polyethylene Tubes and the Effects of a Laminin-Containing Gel," Exp. Neurol. 95(2)387-390 (Feb. 1987).
Malizia, et al., "Migration and Granulomatous Reaction After Periurethral Injection of Polytef (Teflon)," JAMA, 251(24):3277-3281 (Jun. 1984).
Marciano and Gash, "Structural and Functional Relationships of Grafted Vasopressin Neurons," Brain Res., 370(2):338-342 (Apr. 9, 1986).
Matas, et al., "Hepatocellular Transplantation for Metabolic Deficiencies: Decrease of Plasma Bilirubin in Gunn Rats," Science 192:892-894 (1976).
Matouschek, "Die Behandlung des vesikorenalen Refluxes durth Transurethrale Einspritzung von Teflonpaste," Urologe A 20:263-264 (1981).
Mesnil, et al., "Cell Contact but Not Junctional Communication (Dye Coupling) with Biliary Epithelial Cells is Required for Hepatocytes to Maintain Differentiated Functions," Exper. Cell. Res. 173:524-533 (1987).
Michapoulos & Pitot, "Primary Culture of Parenchymal Liver Cells on Collagen Membranes," Exper. Cell. Res. 94:70-78 (1975).
Millaruelo, "Role of Plasminogen Activator and its Inhibitors in Axonal Outgrowth and Regeneration in Vivo," Caltech Biology, (1987).
Minato, et al., "Transplantation of Hepatocytes for Treatment of Surgically Induced Acute Hepatic Failure in the Rat," Eur. Surg. Res., 16:162-169 (1984).
Mito, et al, "Hepatocellular Transplantation," Department of Surgery, Asahikawa Medical College 078 4-5 Nishi-Kagura, Asahikawa, Japan.
Mittleman & Marraccini, "Pulmonary Teflon Granulomas Following Periurethral Teflon Injection for Urinary Incontinence," Arch. Pathol. Lab. Med. 107:611-612 (Nov. 1983).
Mooney, "Control of Hepatocyte Function Through Polymer-Substrate Modulation," Thesis Proposal—Department of Chemical Engineering, Massuchusetts Institute of Technology (Sep. 22, 1989).
Mooney, "Switching from Differentiation to Growth in Hepatocytes: Control by Extracellular Matrix," J. Cell. Phys. (151):497-505 (1992).
Mooney, et al., "Integrating Cell Transplantation and Controlled Drug Delivery Technologies to Engineer Liver Tissue," (abstract) Materials Research Society, (Apr. 17-21, 1995).
Mooney, et al., "Tissue Engineering Using Cells and Synthetic Polymers," Transplantation Reviews 7(3):153-162 (1993).
Mooney, et at., "Induction of Hepatocyte Differentiation by the Extracellular Matrix and an RGD-Containing Synthetic Peptide," Mat. Res. Soc. Symp. Proc. 252 (1992).
Mounzer, at al., "Polyglycolic Acid Mesh in Repair of Renal Injury," Urology 28(2):172-130 (1986).
Movitz, "Accessory Spleens and Experimental Splenosis Principles of Growth," The Chicago Medical School Quarterly, 26(4):183-187 (Winter-Spring 1967).
Nahi, at al., "Successful Islet Transplantation in Spontaneous Diabetes," Surgery 86:218-226 (1979).
Nastelin, "Pancreatic Islet Cell Transplantation: Optimization of Islet Cell Adhesion by Altering Polymer Surface Characteristics," Harvard—M.I.T. Division of Health Sciences and Technology (Feb. 1990).
Naughton, at al., Granulopoiesis and Colony Stimulating Factor Production in Regenerating Liver, Exp. Hematol., 10(5):451-458 (May 1982).
Naughton, et al, "Erythropoietin Production by Macrophages in the Regenerating Liver," Journal of Surgical Oncology 30:184-197 (1985).
Naughton, et al., "Long-Term Growth of Rat Bone Marrow Cells in a Three-Dimensional Matrix," Medical Laboratory Sciences Department, Hunter College School of Health Sciences, New York, The Anatomical Record, 218(1):97a (May 1987).
Notter, et al, "Neuronal Properties of Monkey Adrenal Medulla in vitro," Cell Tissue Res., 244(1):69-76 (1986).
Nyilas, et al, "Peripheral Nerve Repair with Bioresorbable Prosthese," Trans. Am. Soc. Artif. Intern. Organs, 29:307-313 (1983).
O'C. Hamilton, "Miracle Cures May be in Your Cells," BusinessWeek (Dec. 6, 1993).
O'Connor, et al., "Grafting of Burns with Cultured Epithelium Prepared from Autologous Epidermal Cells," The Lancet, I(8210):75-78 (Jan. 1981).
O'Donell & Puri, "Treatment of Vesicoureteric Reflux by Endoscopic Injection of Teflon," British Medical Jornal 289:7-9 (Jul. 7, 1984).
Oellrich, R. G., et al. "Biliary Atresia," Neonatal Network pp. 25-30 (Apr. 1987).
Oliwenstein, "The Power of Plastics," Discover p. 18 (Dec. 1989).
Omery, "A Nursing Perspective of the Ethical Issues Surrounding Liver Transplantation," Heart & Lung 17(6):626-630 (Nov. 1988).
Paige, at al, "De Novo Cartilage Generation Using Calcium Alginate-Chondrocyte Constructs," Children's Hospital, Boston, MA, Plastic Surgery Research Council Meeting; N.E. Society of Plastic and Reconstructive Surgeons Meeting; and North Eastern Society of Plastic Surgeons Meeting pp. 1-34 (1993).
Pasik, Annals of the N.Y. Academy of Science, 495:674-675 (1987).
Patterson, et al., "Adrenal Chromaffin Cell-Derived Cholinergic Neurons for Brain Transplants," Cattech Biology pp. 201-202 (1987).
Perlow, M. J., "Brain Grafting as a Treatment for Parkinson's Disease," Neurosurgery 20(2):335-342 (1987).
Pimpl, et al., "Experimentelle Studie zur Frage der Transplantatkonditionierung and Transplantatgrofe bei heterotoper autologer Milztransplantation," Lagenbecks Archiv 37215-36218 (Salzburg 1984).
Pimpl, et al., "Perfusion of Autologous Splenic Grafts in Correlation with Specific Immunological Functions An Experimental Study in Pigs," Eur. Surg. Res. 19:53-61 (1987).
Pitman, et al., "The Use of Adhesives in Chondrocyte Transplantation Surgery: In-Vivo Studies," Bulletin of the Hospital for Joint Diseases Orthopaedic Institute 49(2):213-220 (1989).
Ptasinska-Urbanska, et al, "Intrascleral Introduction of Isolated Allogeneic Chondrocytes Capable of Cartilage Reformation in Rabbits; Possible Procedure in Treatment of Detachment of the Retina," Exp. Eye. Res., 24(3):241-247 (1977).
Puelacher, W. C., et al., "Tissue-Engineered Growth of Cartilage: the Effect of Varying the Concentration of Chondrocytes Seeded onto Synthetic Polymer Matrices," Int. J. Oral Maxillofac. Surg., 23:49-53 (1994).
Rames & Aartonson, "Migration of Polytef Paste to the Lung and Brain Following Intravesical Injection for the Correction of Reflux," Pediatric Surgery 6(1):239-240 (Jan. 1991).
Redmond, et al., "Fetal Neuronal Grafts in Monkeys Given Methyphenyltetrahydropyridine," The Lancet, pp. 1125-1127 (May 17, 1986).
Redmond, et al., "Transplants of Primate Neurons," The Lancet 2(8510):1046 (Nov. 1, 1986).
Reid, et al. "Long-Term Cultures of Normal Rat Hepatocytes on Liver Biomatrix," Ann. NY Acad. Sci. 349:70-76 (1980).
Report of the International Reflux Study Committee, "Medical Versus Surgical Treatment of Primary Vesicoureteral Reflux: A Prospective International Reflux Study in Children," The Journal of Urology 125:277-283 (Mar. 1981).
Retik, et al., "Management of Severe Hypospadias with a 2-Stage Repair," Microbiology & Immunology 152:749-751 (Aug. 1984).
Rhine, at al, "Polymers for Sustained Macromolecule Release: Procedures to Fabricate Reproducible Delivery Systems and Control Release Kinetics," Journal of Pharmaceutical Sciences, 69(3):265-269 (Mar. 1980).
Rosen, "Bioerodible Polyanhydrides for Controlled Drug Delivery," 1983 Butterworth & Co. (Publishers) Ltd.
Rosen, "Bioerodible Polymers for Controlled Release Systems," Controlled Release Systems: Fabrication Technology, II:83-110.
Sapozhnikova, et al., "Morphological Changes in Splenic Autografts Following Splenectomy: Experimental and Clinical Findings," Biological Abstracts, 86(76896) (1987).
Sasaki, "Neovascularization in the Splenic Autograft Transplanted into Rat Omentum as Studied by Scanning Electron Microscopy of Vascular Casts," Virchows Arch. 409:325-334 (1986).
Sawada, et al., "Effects of Extracellular Matrix Components on the Growth and Differentiation of Cultured Rat Hepatocytes," In Vitro Cellular & Development Biology 23(4): 267-273 (Apr. 1987).
Schmeck, "Doctors try to Capitalize on the Liver's Ability to Regenerate Itself," The New York Times Medical Science, (May 16, 1989).
Schubert & Baird, "Multiple Influences of a Heparin-Binding Growth Factor for Neuronal Development," The Journal of Cell Biology 104:635-643 (Mar. 1987).
Seckle, "Nerve Regeneration Through Synthetic Biodegradable Nerve Guides: Regulation by the Target Organ," Plast. Reconstr. Surg., 74(2):173-81 (Aug. 1974).
Selden, et al., "The Pulmonary Vascular Bed as a Site for Implantation of Isolated Liver Cells in Inbred Rats," Transplantation 38(1):81-83 (Jul. 1984).
Shine, et al., "Cultured Peripheral Nervous System Cells Support Peripheral Nerve Regeneration Through Tubes in the Absence of Distal Nerve Stump," Journal of Neuroscience Research 14:393-401 (1985).
Siegel & Langer, "Controlled Release of Polypeptides and Other Macromolecules," Pharmaceutical Research, pp. 2-10 (1984).
Sirica, A., et al, "Use of Primary Cultures of Adult Rat Hepatocytes on Collagen Gel-Nylon Mesh to Evaluate Carcinogen-Induced Unscheduled DNA Synthesis," Cancer Research, 40:3259-3267 (Sep. 1980).
Sirica, et al., "Fetal Phenotypic Expression by Adult Rat Hepatocytes on Collagen Gel/Nyton Meshes," Proc. Natl. Acad. Sci. USA, 76(1):283-287 (Jan. 1979).
Sladek & Shoulson, "Neural Transplantation: A Call for Patience Rather Than Patients," Science, 240:386-388 (Jun. 10, 1988).
Sladek, "Transplantation of Fetal Dopamine Neurons in Primate Brain Reverses MPTP Induced Parkinsonism," Progress in Brain Research 71:309-323 (1987).
Sladek, et al, "Reversal of Parkinsonism by Fetal Nerve Cell Transplants in Primate Brain," Annals of the New York Academy of Sciences, 495:641-657 (1987).
Sladek, et al., "Survival and Growth of Fetal Catecholamine Neurons Transplanted Into Primate Brain," Brain Research Bulletin, 17:809-818 (1986).
Sommer, et al., "Hepatocellular Transplantation for Treatment of D-Galactosamine-Induced Acute Liver Failure in Rats," Transplant. Proc. 11(1):578-584 (Mar. 1979).
Stein, "Three Dimensional Tissue Reorganization in Hepatocyte Transplantation," American College of Surgeons, Surgical Forum, (Oct. 1991).
Stemple, Altech Biology (1987).
Strom, et al., "Isolation, Culture, and Transplantation of Human Hepatocytes," JNCL, 68(5):771-778 (May 1982).
Sudhakaran, et al., "Modulation of Protein Synthesis and Secretion by Substratum in Primary Cultures of Rat Hepatocytes," Exper. Cell Res. 167:505-516 (1986).
Sullivan, "Spinal Injury Research Yields a Glimmer of Hope," The New York Times p. C6 (Jul. 14, 1987).
Sutherland, D. E., et al., "Hepatocellular Transplantation in Acute Liver Failure," Surgery 82(1):124-132 (Jul. 1977).
Tachibana, "Ureteral Replacement Using Collagen Sponge Tube Grafts," The Journal of Urology 133(4):866-869 (Apr. 1985).
Takeda, et al,, "Hepatocyte Transplantation in Biodegradable Polymer Scaffolds Using the Dalmatian Dog Model of Hyperuricosuria," Transplantation Proceedings 27(1):635-636 (Feb. 1995).
Tavassoli, et al., "Studies on Regeneration of Heterotopic Splenic Autotransplants," Blood, 41(5):701-709 (May 1973).
Thompson, "Implantable Bioreactors: Modern Concepts of Gene Therapy," Current Communications in Molecular Biology, Daniel Marshak, et al., editors, pp. 143-147 (Cold Spring Harbor Laboratory, 1989).
Thompson, J. A., "Heparin-Binding Growth Factor 1 Induces the Formation of Organoid Neovascular Structures in Vivo," Proc. Natl. Acad. Sci USA 86:7928-27932 (Oct. 1989).
Thuroff, et al., "Cultured Rabbit Vesical Smooth Muscle Cells for Lining of Dissolvable Synthetic Prosthesis," Urology, 21(2):155-158 (1983).
Tomomura, et al, "The Control of DNA Synthesis in Primary Cultures of Hepatocytes From Adult and Young Rats: Interactions of Extracellular Matrix Components, Epidermal Growth Factor, and the Cell Cycle," © 1987 Alan R. Liss, Inc.
Unipoint Industries, Inc., "Polyvinyl Alcohol Foam for Surgical And Industrial Use," Product Review.
UNOS Update, "National Cooperative Transplantation Study Completed," 7(10) (Oct./Nov. 1991).
Upton, et al., Neocartilage Derived from Transplanted Perichondrium: What is it? Plastic and Reconstructive Surgery 68(2): 166-174 (1981).
Uyama, et al., "Delivery of Whole Liver-Equivalent Hepatocyte Mass Using Polymer Devices and Hepatotrophic Stimulation," Transplantation 55:932-935 (Apr. 1993).
Vacanti, "Beyond Transplantation," Arch. Surgery 123:545-549 (May 1988).
Vacanti, "Tissue Engineered Composites of Bone and Cartilage Using Synthetic Polymers Seeded with Two Cell Types," 39th Annual Meeting of the Orthopaedic Research Society (Feb. 15-18, 1993).
Vacanti, at al., "Synthetic Polymers Seeded with Chondrocytes Provide a Template for New Cartilage Formation," Journal of the American Society of Plastic and Reconstructive Surgeons, Inc., 88(5):753-759 (Nov. 1991).
Vacanti, et al., "Engineered Bone from Polyglycolic Acid Polymer Scaffold and Periosteum," (abstract) Materials Research Society, (Apr. 17-21, 1985).
Vacanti, et al., "Formation of New Cartilage in Vivo by Implantation of Cell-Polymer Constructs Created in Vitro,".
Vacanti, J.P., et at., Selective Cell Transplantation Using Bioabsorbable Artificial Polymers as Matrices, Journal of Pediatric Surgery, 23(1):3-9 (Jan. 1988).
Van Der Kwast, et al., "Establishment and Characterization of Long-Term Primary Mouse Urothelial Cell Cultures," Urological Researach, 17(1):290-293 (1989).
Vargo, "Infection as a Complication of Liver Transplant," Critical Care Nurse 9(4):52-62.
Vijg, et al., "UV-Induced DNA Excision Repair in Rat Fibroblasts During Immortalization and Terminal Differentiation in Vitro," Exp. Cell. Res. 167:517-530 (1986).
Vorstman, et al., "Polytetrafluoroethylene Injection for Urinary Incontinence in Children," The Journal of Urology 133(2):248-250 (Feb. 1985).
Vroemen, at al., "Hepatocyte Transplantation for Enzyme Deficiency Disease in Congenic Rats," Transplantation 42(2):130-135 (1986).
Walker, et al., "Injectable Bioglass as a Potential Substitute for Injectable Polytetrafluoroethylene," The Journal of Urology 148(1):645-647.
Walton, et al., "Tissue Engineering of Biomaterials for Composite Reconstruction: An Experimental Model," Annals of Plastic Surgery 30(2):104-110 (Feb. 1993).
Whitaker, "Scientists Growing Tissue From ‘Seed’," The Boston Globe (Monday, Feb. 22, 1993).
Wozney, at al., "Novel Regulators of Bone Formation: Molecular Clones and Activities," Science, 242:1528-1534 (Dec. 16, 1988).
Yannas, "What Criteria Should be Used for Designing Artifical Skin Replacement and How Well do the Current Grafting Materials Meet These Criteria?" J. of Trauma, 24(9):S29-S39 (1984).
Yannas, "Wound Tissue Can Utilize a Polymeric Template to Synthesize a Functional Extension of Skin," Science 215:174-176 (1982).
Yannas, at al., "Design of an Artificial Skin. I. Basic Design Principles," Journal of Biomedical Materials Research 14:65-81 (1980).
Yannas, at al., "Polymeric Template Facilitates Regeneration of Sciatic Nerve Across 15 MM Gap," Polymer. Material Sci. Eng. 53:216-218 (1985).
Yannas, at al., "Suppression of In Vivo Degradability and of Immunogenicity of Collagen by Reaction with Glycosaminoglycans," Polymer. Prepar. Am. Chem. Soc. Div. Polym. Chem., 16(2): 209-214 (1975).
Yannas, et al, "Artifical Skin: A Fifth Route to Organ Repair and Replacement," Iss. Polym. Biomaterial, 106:221-230 (1986).
Yannas, et al., "Regeneration of Sciatic Nerve Across 15 mm Gap by Use of a Polymeric Template," Polym. Sci. Technol. Iss. Adv. Biomed. Polymer 35:109 (1987).
Zund, et al., "A New Approach for a Bioprothetic-Heart Valve," The European Association for Cardio-Thoracic Surgery, (Jan. 31, 1995).

Also Published As

Publication number Publication date
USRE42575E1 (en) 2011-07-26
US5855610A (en) 1999-01-05
US6348069B1 (en) 2002-02-19

Similar Documents

Publication Publication Date Title
USRE42479E1 (en) Engineering of strong, pliable tissues
EP0739631B1 (en) Laminar bone support for cartilage growth
US5041138A (en) Neomorphogenesis of cartilage in vivo from cell culture
Shum-Tim et al. Tissue engineering of autologous aorta using a new biodegradable polymer
US5567612A (en) Genitourinary cell-matrix structure for implantation into a human and a method of making
US7914579B2 (en) Tissue engineered tendons and ligaments
US5736372A (en) Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
US5851833A (en) Neomorphogenesis of urological structures in vivo from cell culture
EP0299010B1 (en) Chimeric neomorphogenesis of organs by controlled cellular implantation using artificial matrices
EP0707498B1 (en) Implantable prosthesis, kit and device for manufacturing the same
US8663675B2 (en) Injectable matrix having a polymer and a stem cell niche composed of cup-shaped nanoparticles containing growth factors or physiological agents for organ reconstruction
JP4854670B2 (en) Structural skeletal engineering
JPH10505250A (en) Biomatrix for tissue regeneration
CA2330104A1 (en) Creation of three-dimensional tissues
WO2001003750A1 (en) Human naturally secreted extracellular matrix-coated device
Stock et al. Valves in development for autogenous tissue valve replacement
Goulet et al. Tissue-engineered ligament
RU2240135C1 (en) Cell culture comprising precursor cells of osteogenesis, implant based on thereof and its applying for recovery bone integrity
Ikada Why is the Clinical Application of Tissue Engineering So Slow?
Shin'oka et al. Current status of tissue engineering for cardiovascular structures
Kim The role of mechanical stimuli in engineered smooth muscle tissue development
Daniel Eberli et al. COMPOSITE SCAFFOLDS FOR THE ENGINEERING OF HOLLOW ORGANS AND