CA2627907C - Osteoinductive composition comprising osteoinductive factors recovered from mineralized bone and a carrier - Google Patents

Osteoinductive composition comprising osteoinductive factors recovered from mineralized bone and a carrier Download PDF

Info

Publication number
CA2627907C
CA2627907C CA2627907A CA2627907A CA2627907C CA 2627907 C CA2627907 C CA 2627907C CA 2627907 A CA2627907 A CA 2627907A CA 2627907 A CA2627907 A CA 2627907A CA 2627907 C CA2627907 C CA 2627907C
Authority
CA
Canada
Prior art keywords
osteoinductive
bone
carrier
composition
factors
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 - Fee Related
Application number
CA2627907A
Other languages
French (fr)
Other versions
CA2627907A1 (en
Inventor
Keyvan Behnam
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.)
Warsaw Orthopedic Inc
Original Assignee
Warsaw Orthopedic Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Warsaw Orthopedic Inc filed Critical Warsaw Orthopedic Inc
Publication of CA2627907A1 publication Critical patent/CA2627907A1/en
Application granted granted Critical
Publication of CA2627907C publication Critical patent/CA2627907C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61L27/3604Materials 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
    • A61L27/3608Bone, e.g. demineralised bone matrix [DBM], bone powder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/32Bones; Osteocytes; Osteoblasts; Tendons; Tenocytes; Teeth; Odontoblasts; Cartilage; Chondrocytes; Synovial membrane
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/01Hydrolysed proteins; Derivatives thereof
    • A61K38/012Hydrolysed proteins; Derivatives thereof from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61L27/02Inorganic materials
    • A61L27/12Phosphorus-containing materials, e.g. apatite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3641Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the site of application in the body
    • A61L27/3645Connective tissue
    • A61L27/365Bones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment
    • A61L27/3687Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment characterised by the use of chemical agents in the treatment, e.g. specific enzymes, detergents, capping agents, crosslinkers, anticalcification agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L27/42Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having an inorganic matrix
    • A61L27/425Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having an inorganic matrix of phosphorus containing material, e.g. apatite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L27/44Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • A61L27/48Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix with macromolecular fillers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • 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
    • A61F2/28Bones
    • 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
    • A61F2/28Bones
    • A61F2002/2817Bone stimulation by chemical reactions or by osteogenic or biological products for enhancing ossification, e.g. by bone morphogenetic or morphogenic proteins [BMP] or by transforming growth factors [TGF]
    • 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
    • A61F2/28Bones
    • A61F2002/2835Bone graft implants for filling a bony defect or an endoprosthesis cavity, e.g. by synthetic material or biological material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/30Joints
    • A61F2002/30001Additional features of subject-matter classified in A61F2/28, A61F2/30 and subgroups thereof
    • A61F2002/30667Features concerning an interaction with the environment or a particular use of the prosthesis
    • A61F2002/30677Means for introducing or releasing pharmaceutical products, e.g. antibiotics, into the body
    • 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
    • A61F2310/00Prostheses classified in A61F2/28 or A61F2/30 - A61F2/44 being constructed from or coated with a particular material
    • A61F2310/00005The prosthesis being constructed from a particular material
    • A61F2310/00359Bone or bony 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
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/20Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials
    • A61L2300/252Polypeptides, proteins, e.g. glycoproteins, lipoproteins, cytokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/412Tissue-regenerating or healing or proliferative agents
    • A61L2300/414Growth factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/80Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special chemical form
    • A61L2300/802Additives, excipients, e.g. cyclodextrins, fatty acids, surfactants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/02Materials or treatment for tissue regeneration for reconstruction of bones; weight-bearing implants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/40Preparation and treatment of biological tissue for implantation, e.g. decellularisation, cross-linking

Abstract

An osteoinductive composition, corresponding osteoimplants, and methods for making the osteoinductive composition are disclosed. The osteoinductive composition comprises osteoinductive factors, such as may be extracted from demineralized bone, and a carrier. The osteoinductive composition is prepared by providing demineralized bone, extracting osteoinductive factors from the demineralized bone, and adding the extracted osteoinductive factors to a carrier. Further additives such as bioactive agents may be added to the osteoinductive composition. The carrier and osteoinductive factors may form an osteogenic osteoimplant. The osteoimplant, when implanted in a mammalian body, can induce at the locus of the implant the full developmental cascade of endochondral bone formation including vascularization, mineralization, and bone marrow differentiation. Also, in some embodiments, the osteoinductive composition can be used as a delivery device to administer bioactive agents.

Description

OSTEOINDUCTIVE COMPOSITION COMPRISING OSTEOINDUCTIVE
FACTORS RECOVERED FROM MINERALIZED BONE AND A CARRIER
BACKGROUND
[001] Introduction [002] Mammalian bone tissue is known to contain one or more proteinaceous materials, presumably active during growth and natural bone healing, that can induce a developmental cascade of cellular events resulting in endochondral bone formation. The active factors are variously been referred to in the literature as bone morphogenetic or morphogenic proteins (BMPs), bone inductive proteins, bone growth or growth factors, osteogenic proteins, or osteoinductive proteins. These active factors are collectively referred to herein as osteoinductive factors.
10031 It is well kaown that bone contains these osteoinductive factors.
These osteoinductive factors are present within the compound structure of cortical bone and are present at very low concentrations, e.g., 0.003%. Osteoinductive factors direct the differentiation of pluripotential mesenchymal cells into osteoprogenitor cells flint form osteoblasts. Based upon the work of Marshall Urist as-shown in U_S. Pat. No. 4,294,753, issued Oct. 13, 1981, proper demineralization of cortical bone exposes the osteoinductive factors, rendering it osteoinductive, as discussed more fully below.
[004] Overview of Bone Grafts [005] The rapid and effective repair of bone defects caused by injury, disease, wounds, or surgery has long been a goal of orthopaedic surgery.
Toward this end, a number of compositions and materials have been used or proposed for use in the repair of bone defects. The biological, physical, and mechanical properties of the compositions and materials are among the major factors influ.encing their suitability and performance in various orthopaedic applications.
(006] Autologous cancellous bone ("ACB") long has been considered the gold standard for bone grafts. ACB is osteoinductive and nonimmunogenic, and, by definition, it has all of the appropriate structural and functional characteristics appropriate for the particular recipient. Unfortunately, ACB is only available in a limited number of circumstances. Some individuals lack ACB of appropriate dimensions and quality for transplantation, and donor site pain and morbidity can pose serious problems for patients and their physicians.
[007] Much effort has been invested in the identification and development of altenaative bone graft materials. Urist has published seminal articles on the theory of bone induction and a method for decalcifying bone, i.e., making demineralized bone matrix (DBM). Urist M.R, Bone Formation by Autoinduction, Science 1965; 150(698):893-9; Urist M.R. et al., The Bone Induction Principle, Clin. Orthop. Rel. Res. 53:243-2.83, 1967. As mentioned above, DBM is an osteoinductive material, in that it induces bone growth when implanted in an ectopic site of a rodent, owing to the osteoinductive factors contained within the DBM. It is now known that there are numerous osteoinductive factors, e.g., BMP 1-15, which are part of the transforming growth factor-beta (TGF-beta) superfarnily.
131VIP-2 has become the most important and widely studied of the BMP family of proteins. There are also other proteins present in DBM that are not osteoinductive alone but still contribute to bone growth, including fibroblast growth factor-(FGF-2), insulin-like growth factor-I and 41 (IGF-I and IGF-I1), platelet derived growth factor (F'DGF), and transforming growth factor-beta 1 (TGF-beta.1).
[0081 DBM implants have been reported to be particularly useful (see, for example, U.S. Patent Nos. 4,394,370, 4,440,750, 4,485,097, 4,6'78,470, and 4,743,259; Mulliken et al., Caleif Tissue Int. 33:71, 1981; Neigel et al., Opthal.
Plast Reconstr. Surg. 12:108, 1996; Whiteman et al., J. Hand. Surg. 18B:487, 1993; Xiaobo et al., Clin. Orthop. 293:360, 1993).
DBM typically is derived from cadavers. The bone is removed aseptically and treated to kill any infectious agents. The bone is particulated by milling or grinding, and then the mineral component is extracted by various methods, such as by soaking the bone in an acidic solution. The remaining matrix is malleable and cari be further processed and/or formed and shaped for implantation into a particular site in the recipient. Deminerali2ed bone _9_ prepared in this manner contains a variety of components including proteins, glycoproteins, growth factors, and proteoglycans. Following implantation, the presenoe of DBM induces cellular recruitment to the site of injury. The recruited cells may eventually differentiate into bone forming cells. Such recruitment of cells leads to an increase in the rate of wound healing and, therefore, to faster recovery for the patient.
[009] Some studies indicate that the osteoinductive capabilities of demineralized bone from higher order species in higher order species is relatively low. One study compared the osteoinductivity of rat and canine bone matrix, and of cortical and cancellous bone. Rat bone matrix consistently induced new bone and high phosphatase levels when implanted ectopically in rat. Canine matrix induced small amounts of bone and lower phosphatase levels when implanted in dog and in rat, with cortical matrix being somewhat more inductive than cancellous matrix. Demineralized cancellous bone matrix from dog was the only material tested not showing any osteoinductivity. Schwarz et al., Acta.
Orthop.
Scan. 60(6):693-695, 1989.
[010] Similarly, another study determined that monkey bone matrix induces ectopic bone formation in the athymic rat but not in adult monkeys. It was concluded that adult monkey bone matrix contains bone inductive properties but that these properties are not sufficient to induce bone formation in adult monkey muscle sites. Aspenberg et al., J. of Orthop. Res. 9:20-25, 1991.
[011] Yet another study evaluated bone and cementum regeneration following guided tissue regeneration (GTR) in periodontal fenestration defects.
Specifically, the adjunctive effect of allogenic, freeze-dried DBM implant was evaluated and found to exhibit no discernible adjunctive effect to GTR in the defect model. The critical findings were 1) the DBM particles remained embedded in dense connective tissue without evidence of bone metabolic activity;
and 2) limited and similar amounts of bone and cementum regeneration were observed for both GTR plus DBM and GTR defects. Caplanis et al., J Periodontal 851-856, August, 1998.
[012] Current DBM Emulations have various drawbacics. First, while the collagen-based matrix of DBM is relatively stable, the osteoinductive factors within the DBM matrix are rapidly degraded. The osteogenic activity of the DBM may be significantly degraded within 24 hours atter implantation, and in some instances the osteogenic activity may be inactivated within 6 hours.
Therefore, the osteoinductive factors associated with the DBM are only available to recruit cells to the site-of injury for a short time after transplantation.
For much of the healing process, which may take weeks to months, the implanted material ma.y provide little or no assistance in recruiting cells. In addition to the osteoinductive factors present within the DBM, the overall structure of the DBM
implant is also believed to contribute to the bone healing capabilities of the implant.
[013] Extracting Proteins [014] The potential utility of osteoinductive factors has been recognized = widely. It has been contemplated that the availability of osteoinductive factors could revolutionize orthopedic medicine and certain types of plastic surgery, dental, and various periodontal and cratulofacial reconstructive procedures.
[015] Urist's U.S. Patent No. 4,294,753,.
was the first of his many patents on a process for extracting BMP from DBM. At the time of the Urist /53 patent, BMP was referred to generally.
However, as mentioned above, now it is known that there are multiple forms of BMP. The Urist process became widely adopted, and though different users may use different chemical agents from those disclosed in the basic Urist process, the basic layout of the steps of the process remains widely used today as one of the main methods of extracting BMP from DBM. See, e.g., U.S. Pub 2003/0065392 (2003); U.S. Pub 2002/0197297 (2002). Urist reported that his basic process actually results in generally low yields of BMP per unit weight of DBM.
[016] The observed properties of osteoinductive factors have induced an intense research effort in several laboratories directed to isolating and identifying the pure factor or factors responsible for osteogenic activity. A modified process for purification of osteogenic protein from mammalian bone is disclosed by San3path et al. (1987) Proc. Natl. Acad. Sol. USA 84:7109-7113. Urist et al.
(1983), Proc. Soc. Exp. Biol. Med. 173:194-199, disclose a human osteogenic protein fraction which was extracted from deroinerali7ed cortical bone by means of a calcium chloride-urea inorganic-organic solvent mixture, and retrieved by differential precipitation in guanidine-hydrochloride and piwarative gel electrophoresis. The authors report that the protein fraction has an amino acid composition of an acidic polypeptide and a molecular weight in a range of 17-kDa_ This material was said to be distinct from a protein called. "bone derived growth factor" disclosed by Canalis et al. (1980 Science 210:1021-1023) and by Farley et al. (1982) Biochem 21:3508-351.3.
[0171 Urist et al., (1984) Proc. Nail. Acad. Sci. USA 81:371-375, disclose a bovine BMP extract having dae properties of an acidic polypeptide and a molecular weight of approximately 18 Is.13a. The authors report that the protein was present in a fraction separated by hydroxyapatite chromatography, and that it induced bone formation in mouse hindquarter muscle and bone regeneration in trephine defects in rat and dog skulls. Their method of obtaining the extract from bone results in ill-defined and impure preparations.
[018] European Patent Application Serial No. 148,155, published Oct. 7, 1985. purports to disclose osteogenic proteins derived from bovine, porcine, and human origin. One of the proteins, designated by the inventors as a P3 protein having a molecular weight of 22-24 kDa, is said to have been purified to an essentially homogeneous state. This material is reported to induce bone formation when implanted into aninaals.
[019] International Application No. PCT/087/01537 (Int. Pub. No.
W088100205) discloses an impure fraction from bovine bone with bone induction qualities. =The named applicants also disclose putative "bone inductive factors"
produced by recombinant DNA techniques. Four DNA sequences were retrieved from human or bovine genornic or cDNA libraries and expressed in recombinant host cells. While the applicants stated that the expressed proteins may be bone morphogenic proteins, bone induction was not demonstrated. This same group reported subsequently ((1988) Science 242:1528-1534) that three of the four factors induce cartilage formation, and postulate that bone formation activity "is due to a mixture of regulatory molecules" and that "bone formation is most likely controlled. . . by the interaction of these molecules." Again, no bone induction was attributed to the products of expression of the cDNAs. See also Urist et al., EPO 0,212,474, entitled "Bone Morphogenic Agents."
[020] Wang et al., (1988) Proc. Nat. Acad. Sci. USA 85: 9484-9488, disclose the partial purification of a bovine bone morphogenetic protein from guanidine extracts of demineralized bone having cartilage and bone formation activity as a basic protein corresponding to a molecular weight of 30 kDa determined from gel elution. Separation of the 30 kDa fraction yielded proteins of 30, 18, and 16 kDa, which, upon separation, were inactive. In view of this result, the authors acknowledge that the exact identity of the active material had not been determined.
[021] Wang et al., (1990) Proc. Nat. Acad. Sci. USA 87: 2220-2224, describe the expression and partial purification of one of the cDNA sequences described in PCT 87/01537. Consistent cartilage and/or bone formation with their protein requires a minimum of 600 ng of 50% pure material.
[022] International Application No. PCT/89/04458 (Int. Pub. No.
W090/003733) describes the purification and analysis of a family of osteogenic factors called "P3 OF 31-34.". The protein family contains at least four proteins, which are characterized by peptide fragment sequences. The impure mixture P3 OF 31-34 is assayed for osteogenic activity. The activity of the individual proteins is neither assessed nor discussed.
[023] Implanting Extracted Proteins [024] Successful implantation of the osteoinductive factors for endochondral bone formation requires association of the proteins vvith a suitable carrier material capable of maintaining the proteins at an in vivo site of application. The carrier should be biocompatible, in vivo biodegradable, and porous enough to allow cell infiltration. Insoluble collagen particles that remain after guanidine extraction and delipidation of pulverized bone generally have been found effective in allogenic implants in some species. However, studies have shown that while osteoinductive proteins are useful cross species, the collagenous bone matrix generally used for inducing endochondral bone formation is species-specific. Sarnpath and Reddi, (1983) Proc. Nat. Acad. Sci. USA 80: 6591-6594.
Demineralized, delipidated, extracted xenogenic bone matrix carriers implanted in vivo invariably fail to induce osteogenesis, presumably due to inhibitory or immunogenic components in the bone matrix. Even the use of allogenic bone matrix in osteogenic devices may not be sufficient for osteoinductive bone formation in many species, as discimed above.
[025] U.S. Patent No. 4,563,350 discloses the use of trypsini7ed bovine bone matrix as a xenogenic matrix to effect osteogenic activity when implanted with extracted, partially p-urified bone-inducing protein preparations. Bone formation is said to require the presence of at least 5%, and preferably at least 10%, non-fibrillar collagen. The named inventors claim that removal of telopeptides that are responsible in part for the imnogenicity of collagen preparations is more suitable for xenogenic implants.
[0261 European Patent Application Serial No. 309,241, published March 29, 1989, discloses a device for inducing endochondral bone formation comprising an osteogenic protein preparation, and a matrix carrier comprising 60-98% of either mineral component or bone collagen powder and 2-40% atelopepiide hypoimmunogenic collagen.
[0271 Deatherage et al., (1987) Collagen Rel. Res. 7: 2225-2231, purport to disclose an apparently xenogenic implantable device comprising a bovine bone matrix extract that has been minimally purified by a one-step ion exchange column and reconstituted with highly purified h-urnan Type-1 placental collagen.
[0-281 U.S. Patent No. 3,394,370 describes a matrix of reconstituted collagen purportedly useful in xenogenic implants. The collagen fibers are treated enzymatically to remove potentially immunogenic telopeptides (also the primary source of interfibril crosslinIcs), and are dissolved to remove associated noncollagenenous components. The matrix is formulated by dis' persing the reconstituted collagen in acetic acid to form a disordered matrix of elementary collagen molecules that is then mixed with an osteogenic substance and lyophilized to form a. "semi-rigid foam or sponge"
that is preferably crosslinked. The formulated matrix is not tested in vivo.
[029] U.S. Patent No. 4,172,128 describes a method for degrading and regenerating bone-like material of reduced inmxtmogenicity, said to be useful cross-species. Demineralized bone particles are treated with a swelling agent to dissolve any associated mucopolysaccharides (glycosaminoglycann), and the collagen fibers subsequently dissolved to form a homogenous colloidal solution. A gel of reconstituted fibers then can be formed using physiologically inert mucopolysaceharides and an electrolyte to aid in fibril formation.
[030] The use of pulverized exogenous bone growth material, e.g., derived from deanineralized allogenic or xenogenic bone, in the surgical repair or reconstruction of defective or diseased bone is known. See, in this regard, the disclosures of U.S. Patent Nos. 4,394,370, 4,440,750, 4,472,840, 4,485,097, 4,678,470, and 4,743,259; Bolander et al., "The Use of Demineralized Bone Matrix in the Repair of Segmental Defects," The Journal of Bone and Joint Surgery, Vol. 68-A, No. 8, pp. 1264-1273; Olowacki et al, "Demineralized Bone Implants," Symposium on Horizons in Plastic Surgery, Vol. 12, No. 2; pp. 233-241 (1985); Gepstein et al., "Bridging Large Defects in Bone by Demineralized Bone Matrix in the Form of a Powder," The Journal of Bone and Joint Surgery, VoI. 69-A, No, 7, pp. 984-991 (1987); /v1ellonig, "Decalcified Freeze-Dried Bone Allograft as an Implant Material In Human Periodontal Defects," The International Journal of Periodontics and Restorative Dentistry, pp'. 41-45 (June 1984); Kahan et al., "Treatment of Jaw Defects with Demineralized Bone Implants," Journal of Oral and Maxillofacial Surgery, pp.623-626 (Jun. 6, 1989);
and Todescan et at., "A Small Animal Model for Investigating Endosseous Dental Implants: Effect of Graft Materials on Healing of Endosseous, Porous-Surfaced Implants Placed in a Fresh Extraction Socket," The International Journal of Oral & Maxillofacial Implants Vol. 2, No. 4, pp. 217-223 (1987).
1031] A. variety of approaches have been explored in an attempt to recruit progenitor cells or chondrocytes into an osteochondral or chondral defect. For example, penetration of subchondral bone has been performed in order to access rnesenchymal stem cells (1V1SCs) in the bone marrow, which have the potential to diffezentiate into cartilage and bone. Steadman, et al., "Microfracture:
Surgical Technique and Rehabilitation to Treat Chondral Defects," Clin. Orthop., 391 S:362-369 (2001). In addition, some factors in the body are believed to aid in the repair of cartilage. For example, transforming growth factors beta (TGF-B) have the capacity to recruit progenitor cells into a chondral defect from the synoviurn or elsewhere when loaded in the defect. Hunziker, et al., "Repair of Partial Thickness Defects in Articular Cartilage: Cell Recruitment From the Synovial Membrane," J Bone Joint Surg., 78-A:721-733 (1996). However, the application of growth factors to bone and cartilage implants has not resulted in the expected increase in osteoinductive or chondrogenic activity.
[032] Each of U.S. Patent Nos. 5,270,300 and 5,041,138 describes a method for treating defects or lesions in cartilage that provides a matrix, possibly composed of collagen, with pores large enough to allow cell population and contnin growth factors (TGF-B or other factors (such as angiogenesis factors)) appropriate for the type of tissue desired to be regenerated.
[033] U.S. Patent Publication No. 2003/0044445 describes an osteogenic composition prepared by a process including the steps of subjecting demineralized bone to an extraction medium to produce an insoluble extraction product and a soluble extraction product, separating the insoluble extraction product and the soluble extraction product, drying the soluble extraction product to remove all or substantially all of the moisture in the soluble extraction product, and combining the dried soluble extraction product with demineralized bone particles. This process involves several steps and is quite laborious. Studies using the process have shown that the formed osteogenic composition does not have appreciably increased osteoinductive properties when compared to the demineralized bone particles to which the dried soluble extraction product is added. It was further determined that the demineralized bone from which the extraction products are extracted does not exhibit = appreciably decreased osteoinductive properties when compared with its properties prior to extraction. It is thus theorized that the extraction process withdraws only a small fraction of available tissue repair factors.
[034] Overall, current bone and cartilage graft formulations have various drawbacks. The osteoinductive factors within the matrices can be rapidly degraded and, thus, factors associated with the matrix are only available to recruit cells to the site of injury for a short time after implantation. Further, when added to a matrix, the active factors often do not appreciably increase the osteoinductive activity of the matrix or, at least, do not increase the osteoinductive activity of the matrix as much as is desirable.
[035] Thus, it would be useful to provide increased osteoinductive activity from an osteogenic composition in more concentrated form such that increased osteoinductive activity can be seen even with little bone void space.
BRIEF SUMMARY
[036] Osteoinductive compositions, and implants and methods for their production, are provided. According to certain embodiments, a carrier is exposed to a treatment or condition that increases at least one biological activity of the carrier. More specifically, osteoinductive factors are added to the carrier.
The biological activities that may be increased include, but are not limited to, bone forming; bone healing; osteoinductive activity, osteogenic activity, chondrogenic activity, wound healing activity, neurogenic activity, contraction-inducing activity, mitosisinducing activity, differentiation-inducing activity, chemotactic activity, angiogenic or vasculogenic activity, and exocytosis or endocytosis-inducing activity.
[037] Thus, an osteoinductive composition is provided comprising osteoinductive factors, such as may be extracted from demineralized bone, and a carrier. The osteoinductive composition provides concentrated or enhanced osteoinductive activity. The osteoinductive composition is prepared by providing demineralized bone, extracting osteoinductive factors from the demineralized bone, and adding the extracted osteoinductive factors to a carrier. The carrier and osteoinductive factors may form an osteogenic osteoimplant. The osteoimplant, when implanted in a mammalian body, can induce at the locus of the implant the full developmental cascade of endochondral bone formation including vascularization, mineralization, and bone marrow differentiation. Also, in some embodiments, the osteoinductive composition can be used as a delivery device to administer additional bioactive agents.
[038] The demineralized bone from which the osteoinductive factors are extracted may be provided in any suitable manner. In a one demineralization procedure, the bone is subjected to an acid demineralization step followed by a defatting/disinfecting step.
[039] A simple and economically viable method for extracting osteoinductive factors from bone is provided herein. The method comprises extracting osteoinductive factors such as noncollagenous proteins (including osteogenic growth factors) from demineralized bone matrix using a chaotropic solvent (e.g., 4M guanidine hydrochloride) or a detergent (e.g., 1% sodium dodecylsulfate), removing the chemical used for extraction in an efficient manner that preserves the biological activity of the growth factors, concentrating the biologically active components by purifying away nonessential proteins and inhibitors of bone morphogenetic protein, and combining the protein extracts with a biologically compatible delivery vehicle. Optionally, methods and materials for =
the preservation of activity during storage may be utilized with the present invention. Alternate methods of extracting osteoinductive factors from bone may =
be used.
[040] Thus, the extracted osteoinductive factors are added to a carrier.
When the osteoinductive factors are added to a carrier, the carrier acts as a scaffold and aids in controlling release kinetics. Any suitable shape, size, and porosity of carrier may be used. Suitable carriers include demineralized bone;

surface demineralized bone; mineralized bone; cancellous scaffolds (mineralized or deraineraLized); particulate, dernineralized, guanidine extracted, species-specific (allogenic) bone; specially treated particulate, calcium phosphates, fatty acids, protein extracted, deminerali7ed, xenogenic bone; collagen; synthetic hydroxyapatites; polymers; hydrogels; starches; polyethylene glycol, tricalcium phosphate, sintered hydroxyapatite, settable hydroxyapatite; polylactic acid;
tyrosine polycarbonate; calcium sulfate; collagen sheets; settable calcium phosphate; settable polymers; polymeric cements; settable poly vinyl alcohols;

polyurethanes; and other biocompatible settable materials. Further, the carrier may comprise combinations, modifications, or derivatives of these or others.
Optionally, xenogenic bone powder carriers also may be treated with protenses such as trypsin. The osteoinductive factors and carrier (or delivery or support system) together form an osteoimplant useful in clinical applications.
[0411 Any suitable method for adding, or dispersing, the osteoinductive factors to the carrier may be used. Exactly how this occurs can influence on the biological activity of the final formulation. The extracted osteoinductive factors naay have been lyophilized, resulting in a powder composition. For obvious reasons, adding a powder to a bone matrix may be challenging. Thus, it may be desirable to process the osteoinductive factors to form a homogenous mixture that maybe more easily added to a carrier. This can have a significant impact on the release kinetics of the osteoinductive factors.
[042] Optionally, other additives may be included in the osteoinductive coinposition. For example, radiopaque substances, angiogenesis promoting materials, cytokine inhibitors, bioactive agents, other medically/surgically useful substances, binding agents, or other osteoinducing agents may be added [043] The thus formed osteogenic osteoimplant is intended to be applied at a bone repair site, or a site for bone augmentation or ectopic bone formation (e.g., lateral spine fusion). Examples include a site resulting from injury, defect brought about during the course of surgery, infection, malignancy or developmental malformation. The osteoinductive compositions may also be used as drug delivery devices.

=

[043a] Specific aspects of the invention include:
- an osteoinductive composition comprising: a carrier; osteoinductive factors, the osteoinductive factors being supported by the carrier, the osteoinductive factors being recovered from acid used to demineralize mineralized bone; wherein the osteoinductive composition exhibits increased osteoinductive activity when compared to a carrier not supporting osteoinductive factors;
- an osteoinductive material comprising: a mineralized particulated material;
osteoinductive growth factors, the osteoinductive factors being recovered from acid used to demineralize mineralized bone; and a demineralized bone matrix scaffold;
wherein the osteoinductive growth factors are solubilized and combined with the mineralized particulated material to form a growth factor/mineral composite and wherein the growth factor/mineral composite is distributed in the demineralized bone matrix scaffold;
- an osteoinductive composition with reduced immunogenicity comprising:
noncollagenous proteins recovered from acid used to demineralize bone; and a carrier;
wherein the osteoinductive composition exhibits an ability to induce formation of heterotopic bone in a euthymic mouse;
- an osteoinductive composition comprising: a carrier; and a protein composition comprising a protein recovered from acid used to demineralize bone wherein the protein composition has less than 90% by weight inorganic components; and - a method for producing an osteoinductive composition, the method comprising: providing bone; providing osteoinductive factors; providing a carrier; and adding the osteoinductive factors to the carrier, wherein the providing osteoinductive factors step comprises extracting the osteoinductive factors from acid used to demineralize the bone.
-12a-[044] This application refers to various patents, patent applications, journal articles, and other publications , including the following:
PCT/U804/43999; PCT/1J305/003092; US 2003/0143258 Al; PCT/US02/32941;
Current Protocols in Molecular Biology, Current Protocols in Immunology, Current Protocols in Protein Science, and Current Protocols in Cell Biology, John Wiley 8c Sons, N.Y., edition hs ofInly 2002; Sambrook, Russell, and Sambrook, Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 2001; Rodd 1989 "Chemistry of Carbon Comp:ma," vols. 1-5 and supps, Elsevier Science Publishers, 1989;
"Organic Reactions," vols 1-40, John. Wiley and Sons, New York, NY, 1991;
March 2001i "Advanced Organic Chemistry," 5th ed. John. Wiley and Sons, New York, NY. In the event of a conflict between the specification and any of the cited references, the specification shall control. Where numerical values herein are expressed as a range, endpoints are included.
[045] While multiple embodiments are disclosed, still other embodiments of the present invention will become apparent to those skilled in the art from the following detailed description. As will be apparent, the invention. is capable of modifications in various obvious aspects, all without departing from the scope of the claims. Accordingly, the detailed description is to be regarded as illustrative in nature and not restrictive.
DEFINMONS
[046] Bloactive Agent or Bioactiye Compound is used herein to refer to a compound or entity that alters, inhibits, activates, or otherwise affects biological or chemical *events. For example, bioactive agents may include, but are not limited to, osteogenic or chondrogenic proteins or peptides, anti-AIDS
substances, anti-cancer substances, antibiotics, immun.osuppressants, anti-viral substances, enzyme inhibitors, hormones, neumtoxins, opioids, hypnotics, anti-histamines, lubricants, tranquilizers, anti-convulsants, muscle relaxants and anti-Parlcinson substances, anti-spasmodics and muscle contractants including channel blockers, miotics and anti-cholinergics, anti-glaucoma compounds; anti-parasite and/or anti-protozoal compounds, modulators of cell-extracellular matrix interactions including cell growth inhibitors and antiadhesiou molecules, vasodilating agents, inhibitors of DNA, RNA or protein synthesis, anti-hypertensives, analgesics, anti-pyretics, steroidal and non-steroidal anti-inflammatory agents, anti-angiogenic factors, angiogenic factors, anti-secretory factors, anticoagulants and/or antithrombotic agents, local anesthetics, oplithalrnics, prostaglandins, anti-depressants, anti-psychotic substances, anti-eraetics, and imaging agents. In certain embodiments, the bioactive agent is a. drug. In some embodiments, the bioactive agent is a growth factor, cytokine, extracellular matrix molecule or a fragment or derivative thereof, for example, a cell attachment sequence such as RGD. A more complete listing of bioactive agents and specific drugs suitable for use in the present invention ma.y be found in "Pharmaceutieal Substances:
Syntheses, Patents, Applications" by Axel Kleeman-n and Jurgen Engel, Thieme Medical Publishing, 1999; the "Merck Index: An Encyclopedia of Chemicals, Drugs, and Biologicals", Edited by Susan Budavari et al., CRC Press, 1996; and the United States Phannacopeia-25/National Formulary-20, published by the United States Pharmcopeial Convention, Inc., Rockville MD, 2001, (0471 Biocornpatible, as used herein, is intended to describe materials that, upon administration in vivo, do not induce undesirable long-term effects.
[0481 Bone as used herein refers to bone that is cortical, cancellous or cortico-cancellous of autogcnous, allogenic, xenogenic, or transgenic (0491 Demineralized, as used herein, refers to any material generated by removing mineral material from tissue, e.g., bone tissue. In certain embodiments, the demineralized compositions described herein include preparations containing less than 5% calcium and preferably less than 1% calcium by weight. Partially demineralized bone (e.g., preparations with greater than 5% calcium by weight but containing less than 100% of the original starting amount of calcitun) is also considered within the scope of the invention. In some embodiments, demineralized bon.e has less than 95% of its original mineral content.

Demineralized is intended to encompass such expressions as "substantially demineralized," "partially denaineralized," and "fully deminerali7ed."
[050] Demineralized bone matrix, as used herein, refers to any material generated by removing mineral material from bone tissue. In preferred embodiments, the DBM compositions as used herein include preparations containing less than 5% calcium and preferably less than 1% calcium by weight.

Partially der1iners1i7ed bone (e.g., preparations with greater than 5% calcium by weight but contairMig less than 100% of the original starting amount of calcium) are also considered vvithin the scope of the invention.
[051] Osteoconductive is used herein to refer to the ability of a non-osteoinductive substance to serve as a suitable template or substance along which bone may grow.
[052] Osteogenic is used herein to refer to the ability of an agent, material, or implant to enhance or accelerate the growth of new bone tissue by one or 111011, mechanisms such as osteogenesis, osteoconductioia, and/or osteoinduction.
[053] Osteoimplant as used herein refers to any bone-derived implant prepared in accordance with the embodiments of this invention and therefore is intended to include expressions such as bone membrane, bone graft, etc.
[054] Osteoinductive, as used herein, refers to the gonlity of being able to recruit cells from the host that have the potential to stimulate new bone formation.
Any material that can induce the formation of ectopie bone in the soft tissue of an animal is considered osteoinductive. For example, most oste,oinductive materials induce bone formation in atb.ymic rats when assayed according to the method of Edwards et al., "Osteoinduction of Human Derninergized Bone: Characterization in a Rat Model," Clinical Orthopaedics & Rel. Res., 357:219-228, December 1998. In other instances, osteoinductionis considered to occur through cellular recruitment and induction of the recruited cells to an osteogenic phenotype. Osteoinductivity score refers to a score ranging from 0 to 4 as determined according to the method of Edwards et al. (1998) or an equivalent calibrated test. In the method of Edwards et al., a score of "0"

represents no new bone formation; "1" represents 1%-25% of implant involved in new bone formation; "2" represents 26-50% of implant involved in new bone formation; "3" represents 51%-75% of implant involved in new bone formation;
and "4" represents >75% of implant involved in new bone formation. In most instances, the score is assessed 28 days after implantation. However, the osteoinductivity score may be obtained at earlier tinae points such as 7, 14, or 21 days following implantation. In these instances it may be desirable to include a normal DBM control such as DBM powder without a carrier, and if possible, a positive control such as BMP. Occasionally osteoinductivity may also be scored at later timepoints such as 40, 60, or even 100 days following implantation.
Percentage of osteoinductivity refers to an osteoinductivity score at a given time point expressed as a percentage of activity, of a specified reference score.
[055] Proteases, as used herein, are protein-cleaving enzymes that cleave peptide bonds that link amino acids in protein molecules to generate peptides and protein fragments. A large collection of proteases and protease families has been identified_ Some exemplary proteases include serine proteases, aspartyl proteases, acid proteases, alkaline proteases, metalloproteases, carboxypeptidase, arninopeptidase, cysteine protease, coliagenase, etc. An exemplary family of proteases is the proprotein convertase family, which includes furin. Dubois et al., American .Journal of Pathology (2001) 158(1):305316. Members of the proprotein corivertase family of proteases are lcnowti to proteolytically process proTGFs and proBMPs to their active mature forms. Dubois et al., Anzerican Journal of Pathology (2001) 158(1):305-316; Cui et al., The Embo Journal (1998) 17(16):4735-4743; Cui et al., Genes & Development (2001) 15:2797-2802.
[056] Protease inhibitors, as used herein, refers to chc-mical compounds capable of inhibiting the enzymatic activity of protein cleaving enzymes (i.e., proteases). The proteases inhibited by these compounds include serine proteases, acid proteases, metalloproteases (exanaples of some matrix metalloprotease inhibitors are shown in Figure 6), carboxypeptidase, aminopeptidase, cysteine protease, etc. The protease inhibitor may act specifically to inhibit only a specific protease or class of proteases, or it may act more generally by inhibiting most if not all proteases. Preferred protease inhibitors are protein or peptide based and are commercially available from chemical companies such as Aldrich-Sigma.
Protein or peptide-based inhibitors which adhere to the DBM (or calcium phosphate or ceramic carrier) are particularly preferred as they remain associated with the matrix providing a stabilizing effect for a longer period of time than freely diffusible inhibitors. Examples of protease inhibitors include aprotinin, 4-(2aminoethyl) benzenesulfonyl fluoride (AEBSF), amastatin-HC1, alphal-antichymotrypsin, antithrombin 111, alphal-antitrypsin, 4-arninophenylmethane sulfonyl-fluoride (APMSF), arphamenine A, arphamenine B, E-64, bestatin, CA-074, CA-074-Me, calpain inhibitor I, calpain inhibitor 11, cathepsin inhibitor, chymostatin, diisopropylfluorophosphate (DFP), dipeptidylpeptidase IV
inhibitor, diprotin A, E-64c, E-64d, E-64, ebelactone A, ebelactone B, EGTA, elastatinal, foroxymithine, hirudin, leuhistin, leupeptin, alpha2macroglobulin, phenylmethylsulfonyl nuo4de (PMSF), pepstatin A, phebestin, 1,10phenanthroline, phosphorarnidon, chymostatin, benzamidine HCI, antipain, epsilon aminocaproic acid, N-ethylmaleimide, trypsin inhibitor, 1-chloro-3-tosylamido-7-amino2-heptanone (TLCK), 1-chloro-3-tosylamido-4-pheny1-2-butanone (TPCK), trypsin inhibitor, and sodium EDTA. Stabilizing agent is any chemical entity that, when included in an inventive composition comprising DBM

and/or a growth factor, enhances the osteoinductivity of the composition as measured against a specified reference sample. In most cases, the reference sample will not contain the stabilizing agent, but in all other respects will be the same as the composition with stabilizing agent. The stabilizing agent also generally has little or no osteoinductivity of its own and works either by increasing the half-life of one or more of the active entities within the inventive composition as compared with an otherwise identical composition lacking the stabilizing agent, or by prolonging or delaying the release of an active factor. In certain embodiments, the stabilizing agent may act by providing a barrier between proteases and sugar-degrading enzymes thereby protecting the osteoinductive factors found in or on the matrix from degradation and/or release. In other .

embodiments, the stabilizing agent may be a chemical compound that inhibits the activity of proteases or sugar-degrading enzymes. In a preferred embodiment, the stabilizing agent retards the access of enzymes known to release and solubilize the active factors. Half-life may be determined by immunolgical or enzymatic assay of a specific factor, either as attached to the matrix or extracted there from.
Alternatively, measurement of an increase in osteoinductivity half-life, or measurement of the enhanced appearance of products of the osteoinductive process (e.g., bone, cartilage or osteogenic cells, products or indicators thereof) is a useful indicator of stabilizing effects for an enhanced osteoinductive matrix composition. The measurement of prolonged or delayed appearance of a strong osteoinductive response will generally be indicative of an increase in stability of a factor coupled with a delayed unmasking of the factor activity.
[057] Superficially demineralized as used herein refers to bone-derived elements possessing at least about 90 weight percent of their original inorganic mineral content, the expression "partially demineralized" as used herein refers to bone-derived elements possessing from about 8 to about 90 weight percent of their original inorganic mineral content and the expression "fully dernineralized"
as used herein refers to bone containing less than 8% of its original mineral context.
DETAILED DESCRIPTION
[058] I. INTRODUCTION
[059] The present invention provides osteoinductive compositions and implants and methods for their production. According to certain embodiments, a carrier is exposed to a treatment or condition that increases at least one biological activity of the carrier, as described above. In certain embodiments, the carrier contains peptides or protein fragments that increase the osteoinductive or chondrogenic properties of the carrier. Those of ordinary skill will appreciate that a variety of embodiments or versions of the invention are not specifically discussed below but are nonetheless within the scope of the present invention, as defined by the appended claims.

[060] Bone is made up principally of cells, and also of collagen, minerals, and other noncollagenous proteins. Bone matrices can be nondemineralized, partially demineralized, demineralized, deorganified, anorga.nic, or mixtures of these. DBM is comprised principally of proteins and glycoproteins, collagen being the primary protein component of DBM. While collagen is relatively stable, normally being degraded only by the relatively rare collagenase enzymes, various other proteins and active factors present in DBM
are quickly degraded by enzymes present in the host. These host-derived enzymes include proteases and sugar-degrading enzymes (e.g., endo- and exoglycosidases, glycanases, glycolases, amylase, pectinases, galacatosidases, etc.). Many of the active growth factors responsible for the osteoinductive activity of DBM exist in cryptic form, in the matrix until activated.
Activation can involve the change of a pre or pro function of the factor, or release of the function from a second factor or entity that binds to the first growth factor.
Thus, growth factor proteins in a DBM or added to a DBM may have a limited osteoinductive effect because they are rapidly inactivated by the proteolytic environment of the implant site, or even within the DBM itself.
[061] A number of endogenous factors that play important roles in the development and/or repair of bone and/or cartilage have been identified. BMPs such as BMP-2 and BMP-4 induce differentiation of mesenchymal cells towards cells of the osteoblastic lineage, thereby increasing the pool of mature cells, and also enhance the functions characteristic of differentiated osteoblasts.
Canalis et al., Endocrine Rev. 24(2):218-235, 2003. In addition, BMPs induce endochondral ossification and chondrogenesis. BMPs act by binding to specific receptors, which results in phosphorylation of a class of proteins referred to as SMADs.
Activated SMADs enter the nucleus, where they regulate transcription of particular target genes. BMPs also activate SMAD-independent pathways such as those involving Ras/MAPK signaling. Unlike most BMPs such as BMP-2 and BMP-4, certain BMPs (e.g., BMP-3) act as negative regulators (inhibitors) of osteogenesis. In addition, BMP-1 is distinct both structurally and in terms of its mechanism of action from other BMPs, which are members of the TGF-13 superfarnily. Unlike certain other BlVIPs (e.g., BMP-2, BMP-4), BMP-1 is not osteoinductive. Instead, BMP-1 is a collagenolytic protein that has also been shown to cleave chordin (an endogenous inhibitor of BMP-2 and BMP-4).
Tolloid is a metalloprotease that is structurally related to BMP-1 and has proteolytic activity towards chordin. See Canalis, supra, for further details regarding the activities of BMPs and their roles in osteogenesis and chondrogenesis.
[062] A
variety of endogenous inhibitors of BMPs have been discovered in addition to chordin. These proteins act as BMP antagonists and include pseudoreceptors (e.g., Bambi) that compete with signaling receptors, inhibitory SMADs that block signaling, intracellular binding proteins that bind to activating SMADs, factors that induce ubiquitination and proteolysis of activating SMADs, and extracellular proteins that bind BMPs and prevent their binding to signaling receptors. Among the extracellular proteins are noggin, chordin, follistatin, members of the Dan/Cerberus family, and twisted gastrulation. These proteins and their sequences are known and readily available to one of ordinary skill in the art.
[0631 11.
INCREASING THE BIOLOGICAL ACTIVITY OF A
CARRIER
[0641 Methods for increasing the biologic activity of a bone, cartilage, or other carrier are provided. Osteoinductive osteoimplants are further provided.

The osteoinductive osteoimplants comprise carriers and osteoinductive factors, wherein the carrier has increased biological activity relative to a carrier that has not been exposed to a treatment or condition. The biological activities that may be increased include but are not limited to osteoinductive activity, osteogenic activity, chondrogenic activity, wound healing activity, neurogenic activity, contraction-inducing activity, mitosis-inducing activity, differentiation-inducing activity, chemotactic activity, angiogenic or vasculogenic activity, and exocytosis or endocytosis-inducing activity. It will be appreciated that bone formation processes frequently include a first stage of cartilage formation that creates the basic shape of the bone, which then becomes mineralized (endochondral bone fornaation.). Thus, in many instances, chondrogenesis may be considered an early stage of osteogenesis, though of course it may also occur in other contexts.
[065] An osteoinductive composition is provided comprising osteoinductive factors, such as may be extracted from demineralized bone, and a carrier. The osteoinductive composition provides concentrated or enhanced osteoinductive activity. The osteoinductive composition is prepared by providing demineralized bone, extracting osteoinductive factors from the dernineralized bone, and adding the extracted osteoinductive factors to a carrier. The carrier and osteoinductive factors may form an osteogenic osteoimplant. The osteoimplant, when implanted in a mammalian body, can induce at the locus of the implant the full developmental cascade of endochondral bone formation including vascularization, mineralization, and bone marrow differentiation. Also, in some embodiments, the osteoinductive composition can be used as a delivery device to administer bioactive agents.
[066) In certain embodiments, the carrier contains peptides or protein fragments that increase its osteoinductive or chondrogenic properties. The peptides or protein fragments may be exogenously added to the carrier.
Further, other agents may be added to the carrier, e.g., agents that improve the osteogenic ancVor chondrogenic activity of the matrix by either transcriptional or post-transcriptional regulation of the synthesis of bone or cartilage enhancing or inhibiting factors by cells within the carrier.
[067] III. PROVIDE DEMINERALIZPD BONE
[068] The clenaineralized bone from which the osteoinductive factors are extracted may be provided in any suitable manner. The bone useful in the invention herein is obtained utilizing methods well known in the art, e.g., allogenic donor bone. Bone-derived elements can be readily obtained from donor bone by various suitable methods, e.g., as described in U.S. Patent No.
6,616,698.
The bone may be cortical, cancellous, or cortico-cancellous of autogenous, allogenic, xenogenic, or transgenic 1069) DBM preparations have been used for many years in orthopedic medicine to promote the formation of bone. For example, DBM has found use in the repair of fractures, in the fusion of vertebrae, in joint replacement surgery, and in treating bone destruction due to underlying disease such as rheumatoid arthritis.
DBM is thought to promote bone formation in vivo by osteoconductive and osteoinductive processes. The osteoinductive effect of implanted DBM
compositions is thought to result from the presence of active growth factors present on the isolated collagen-based matrix. These factors include members of the TGF-13, IGF, and BMP protein families. Particular examples of osteoinductive factors include TGF-13, IGF-1, IGF-2, BMP-2, BMP-7, parathyroid hormone (PTH), and angiogenic factors. Other osteoinductive factors such as osteocalcin and osteopontin are also likely to be present in DBM preparations as well.
There are also likely to be other unnamed or undiscovered osteoinductive factors present in DBM.
[070] In one demineralization procedure, the bone is subjected to an acid demineralization step followed by a defatting/disinfecting step. The bone is immersed in acid over time to effect demineralization. Acids that can be employed in this step include inorganic acids such as hydrochloric acid and as well as organic acids such as formic acid, acetic acid, peracetic acid, citric acid, propionic acid, etc. The depth of demineralization into the bone surface can be controlled by adjusting the treatment time, temperature of the demineralizing solution, concentration of the demineralizing solution, and agitation intensity during treatment.
(071] The demineralized bone is rinsed with sterile water and/or buffered solution(s) to remove residual amounts of acid and thereby raise the pH. A
suitable defatting/disinfectant solution is an aqueous solution of ethanol, the ethanol being a good solvent for lipids and the water being a good hydrophilic carrier to enable the solution to penetrate more deeply into the bone particles.
The aqueous ethanol solution also disinfects the bone by killing vegetative microorganisms and viruses. Ordinarily, at least about 10 to 40 percent by weight of water (i.e., about 60 to 90 weight percent of defatting agent such as alcohol) should be present in the defatting disinfecting solution to produce optimal lipid removal and disinfection within the shortest period of time. The preferred concentration range of the defatting solution is from about 60 to about 85 weight percent alcohol and most preferably about 70 weight percent alcohol.
[072] IV. EXTRACT OSTEOINDUCTIVE FACTORS FROM
DBM
[073] A simple and economically viable method for extracting osteoinductive factors from bone is provided herein. The method comprises extracting osteoinductive factors such as noncollagenous proteins (including osteogenic growth factors) from DBM using a chaotropic solvent or a detergent.

The chaotropic solvent may be guanidine hydrochloride of any suitable concentration, such as 4M. The detergent may be sodium dodecylsulfate in any suitable concentration, such as 1%. The chemical used for extraction is removed in an efficient manner that preserves the biological activity of the growth factors.
The biologically active components are concentrated by purifying away nonessential proteins and inhibitors of bone morphogenetic protein, and the protein extracts are then combined with a biologically compatible delivery vehicle.
[074] Most of the previous work published on the subject uses complicated extraction schemes that are expensive and difficult to implement in a commercial or industrial application. Using the method described, the process is optimized by using less costly chaotropic agents, and detergents that are easier to remove, than those previously used. Innovative methods to increase the speed of renaturing the extracted proteins are further provided. Typically, dialysis against water is used to remove the detergent or chaotropic agent. However, by precipitating the proteins with ethanol, ammonium sulfate, or polyethylene glycol, dialysis against water is not necessary. Further, ultrafiltration may be used, thereby avoiding dialysis.
[075] Generally, extracted osteoinductive factors have lower specific bone fonning activity when compared to the starting material. This may be caused by protein denaturation that results from the extraction. For example, when guanidine is used to extract the hydrophobic osteoinductive proteins, the proteins lose their native three-dimensional conformation. As a result, unless they regain their normal shape upon removal of the guanidine, they no longer are active. The addition of chemical chaperones to the guanidine solution may prevent this irreversible protein denaturation. Suitable chemical chaperones include glycerol, trehalose, proline, gIycine betaine, and dextrose, along with mixtures of these. These chemical chaperones enable the osteoinductive proteins to regain their native three-dimensional conformation when the guanidine is removed. They also prevent protein denaturation during lyophilization.
[076] Thus, a method for extracting osteoinductive factors from the mineral component of bone is provided to recover growth factor activity that is normally lost during the demineralization process. It is known that 4 M
guanidine hydrochloride can extract osteoinductive factors from finely powdered mineralized bone. Additionally, osteoinductive factors can be recovered from the acid that is typically used to demineralized bone, such as 0.6 N HC1. These osteoinductive factors are normally lost during the demineralization process and treated as waste.
[077] Growth factors may be extracted from the mineral phase of bone using, for example, the following procedure. As previously described, bone is at least partially demineralized. The bone may comprise powder, fibers, chips, or other. The bone may be demineralized in an acid, for example 1M citric acid, citric acid, or 0.6N HC1, at temperatures ranging from, for example 1 C to 28 C
for time period of, for example 10 minutes to 96 hours. In one embodiment, the bone is demineralized in an acid at a temperature of 4 C. After demineralization, the acid used for demineralization contains growth factors and mineral. The acid may be dialyzed against water to cause the mineral phase and the protein growth factors to co-precipitate. This biphasic (protein and mineral) material may then be collected by filtration or centrifutation and combined with a carrier or lyophilized.
[078] In alternative embodiments, the protein and mineral material in the acid may be separated by dialyzing the acid, also referred to as the demineralization bath, against a weak acid, for example. 0.25M citric acid. In such embodiment, the mineral phase passes through the dialysis bag and the protein phase (collagen fragments, growth factors, etc.) is left within the bag. The protein phase can then be recovered by dialyzing against water and separating water soluble and water insoluble proteins from one another.
[079] In one embodiment, the method for extracting growth factors comprises demineralizing powdered bone with dilute acid within a dialysis bag.

Suitable dilute acid includes 0.05 M to 1.0 M HC1 and 1M or 2M citric acid.
After removing the demineralized bone, the contents of the bag may be further dialyzed against dilute acid to remove the mineral components. A volatile acid, such as acetic acid, can be used to facilitate recovery by lyophilization.
[080] Thus, mineralized bone or bone mineral recovered from demineralization acid may be used as a means of purifying recovered proteins.
The protein phase recovered from the demineralization bath may be solubilized in urea or other form of detergent solution. The bone stimulating growth factors may then be purified, for example using a hydroxyapatite affinity chromatogrphay scheme, described below.
[081] In one embodiment a protein composition substantially free from inorganic components is provided. The protein composition may comprise less than 5% inorganic components by weight. In an alternative embodiment, a protein composition comprising organic components ranging from approximately 6% to approximately 20% by weight is provided. In another embodiment, a protein composition comprising organic components ranging from approximately 21% to approximately 50% may be provided. In yet a further embodiment, a protein composition comprising organic components ranging from approximately 51% to approximately 90% may be provided. The protein composition may be recovered from acid used to demineralize bone. The proteinaceous material of the protein composition may be purified by chromatography, electrophoresis, or other chemical or physical means. The protein composition may be combined with another material such as demineralized bone, hydroxyapatite, tricalcimn phosophate,dicalcium phosphate or other. In some embodiments, the protein composition may exhibit the ability to induce heterotopic bone formation in an athymic animal. In other embodiments the protein composition can serve as a source of collagen Type I and other extracellular matrix proteins that can support tissue repair processes such as angiogenesis, osteocond-uction and wound healing.
As the protein material has desirable handling properties when combined with water or glycerol, the protein can also serve as a carrier for a variety of bone forming matrices including DBM.
[082] In some embodiments, the protein composition may be solubalized in an appropriate medium, such as 6M urea, exposed to hydroxyapatite, TCP, DCP, mineralized bone, surface demineralized bone, or mineral recovered from acid used to demineralize bone. The protein may further be permitted to adsorb onto the mineral sufaces and be washed with a solution comprising, for example, sodium phosphate ranging from approximately ltnIVI to 50mM in concentration.
The proteins may then be eluted with a solution comprising, for example, sodium phosophate ranging in concentrations from between approximately 100m1VI to approximately 500m_M.
[083] As is described below, growth factors recovered from the mineral phase of bone may be recombined with an osteoinductive carrier.
[084] Proteases may reduce the activity of the osteoinductive factors in demineralized bone by breaking down those osteoinductive factors. This negative effect may be reduced or eliminated by adding protease inhibitors to the Hel solution. Suitable protease inhibitors for use in the present invention include N-ethyl maleimide, benzamidine HC1, cysteine, or iodoacetic acid. Alternatively, the bone may be heated briefly to inactivate the proteases, which are relatively more heat sensitive than the growth factors. A suitable heating regimen is 5 Minutes at 60 C, or 1 minute at 90 C.
[085] In alternative embodiments, extraction of osteoinductive factors from mineralized or demineralized bone may be done in any suitable manner.
Further, during extraction, coprecipitates may be used. Thus, for example, bone may be treated with a chaotropic solvent such as guanidine hydrochloride. The bone and chaotropic solvent are dialyzed against water. As the chaotropic solvent decreases, it is replaced by water. Precipitates are then extracted.
Coprecipitates, such as protein, collagen, collagen fragments, albumen, or protein with RGD

sequences, may be extracted. The extracted osteoinductive factors and coprecipitates may then be blended into a homogenous mixture.
[086] Proteins in bone matrix tend to be insoluble and may associate with the bone matrix. Generally, collagens are among the most insoluble osteoinductive factors. Extraction methods may be used to increase the solubility of the osteoindUctive factors to facilitate extraction of the osteoinductive factors.
Generally, growth factors are hydrophobic and are not readily soluble. The growth factors may be treated to improve solubility.
[087] The solubility of the demineralized bone in one or more solvents (e.g., an aqueous medium) may be changed, e.g., increased, relative, for example, to the solubility of a standard DBM not exposed to the treatment. Preferably, the aqueous medium is at physiological conditions, e.g., pH, osmotic pressure, salt concentration, etc. are within physiologically appropriate ranges. For example, the pH may be approximately 7.2-8.0, or preferably 7.4-7.6. The osmotic pressure may be approximately 250-350 mosrn/kg, 280-300 mosm/kg, etc. More generally, the pH may be between approximately 3-11, 4-10, 5-9, 6-8.5, etc.
The osmotic pressure may be between 50-500 rnosm/kg, 100-350 mosm/kg, etc. The salt concentration may be approximately 100-300 mIVI NaC1, e.g., approximately 150 mM NaCl. The aqueous medium may be tissue culture medium, blood, extracellular fluid, etc., and the physiological conditions may be conditions such as are typically found within these fluids and/or within a body tissue such as muscle. The solubility may be increased at any temperature, e.g., room temperature, body temperature of a subject such as a human or animal, etc.
[088] Collagenase treatment of standard human DBM significantly increases its solubility relative to that of untreated standard human DBM. The solubility of the DBM may be increased by exposure to an appropriate treatment or condition, e.g., collagenase treatment, radiation, heat, etc. The extent to which ' the solubility is increased may be varied by varying the nature of the treatment (e.g., the enzyme concentration) and/or the time over which it is applied. A
combination of treatments may be used. In certain embodiments of the invention, the solubility of the human DBM composition is greater than that of a standard =
DBM composition by between 10% and 4000% percent. For example, the solubility may be greater by between 10% and 100%, 100% and 500%, 500% and 1000%, 1000% and 2000%, 2000% and 3000%, 3000% and 4000% or any other range between 10% and 4000%. The solubility may be assessed at any time following the treatment. For example, the DBM may be placed in aqueous medium for a period of time such as 244-8 hours, 3, 4, 5, 6, or 7 days, 10 days, 14 days, etc. The amount of DBM remaining after the period of time is quantitated (e.g., . dry weight is measured) and compared with the amount that was present initially. The extent to which the amount decreases after a period of time serves as an indicator of the extent of solubilization.
[089] Extraction may extract both osteoinductive factors and their inhibitors. If the inhibitors are extracted, it may be desirable in some instances to separate out the osteoinductive factors. This may be referred to as removal of the inhibitors or concentration of the osteoinductive factors.
[090] As a general matter, both the osteoinductive factors and the inhibitors may be extracted and both the osteoinductive factors and the inhibitors may be used for manufacturing the osteogenic osteoimplant. Alternately, only the osteoinductive factors (and not their inhibitors) are extracted and only the osteoinductive factors are used for manufacturing the osteogenic osteoimplant.

Lastly, both the osteoinductive factors and the inhibitors may be extracted and only the osteoinductive factors may be used for manufacturing the osteogenic osteoimplant. The embodiment of extraction and resultant use of osteoinductive factors with or without inhibitors is not a limiting feature of the present invention.
[091] In one embodiment, a simplified extraction process is used that is amenable to batch processing. K. Behnam, E. Brochmann, and S. Murray; Alkali-urea extraction of demineralized bone matrix removes noggin, an inhibitor of bone morphogenetic proteins; Connect Tissue Res. 2004, 45(4-5):257-60.
[092] In certain embodiments, the bone is exposed to a treatment or condition that generates peptides and protein fragments having osteoinductive or chondrogenic activity. In contrast to various longer proteins, certain peptides and protein fragments are less susceptible to proteolytic degradation and more likely to maintain their osteoinductive or chondrogenic properties in the proteolytic environment of the matrix or implant site. Many osteoinductive and chondrogenic proteins, for example, growth factors such as BMPs, cell signaling molecules, transcription factors, hormones, etc., have domains that are responsible for binding to receptors and/or initiating signal transduction in bone and cartilage growth pathways. These dorriains are capable of functioning independently as peptides and protein fragments. In certain embodiments, the osteoinductive or =
chondrogenic activity of bone and cartilage matrices is increased by cleaving the osteoinductive and chondrogenic factors present in the matrix to generate active peptides or protein fragments and/or to generate active peptides or protein fragments that are less susceptible to degradation than their longer precursors.
The increased number of factors in the matrix results in increased bone or cartilage formation.
[093] If desired, the osteoinductive factors can be modified in one or more ways, e.g., their protein content can-be augmented or modified as described in U.S. Patent Nos. 4,743,259 and 4,902,296.
As discussed more fully below, the osteoinductive factors can be admixed with one or more optional substances such as binders, fillers, fibers, meshes, substances providing radiopacity, plasticizers, biostatic/biocidal agents, surface active agents, and the like, prior to, during, or after adding to the carrier.
[094] A nurnber of naturally occurring proteins from bone or recombinant osteoinductive factors have been described in the literature and are suitable for use in the osteoinductive composition. RecombinantIy produced osteoinductive factors have been produced by several entities. Creative Biornolecules of Hopkinton, Mass., produces an osteoinductive factor referred to as Osteogenic Protein 1, or OP1. Genetics Institute of Cambridge, Mass., produces a series of osteoinductive factors referred to as Bone Morphogenetic Proteins 1-13 (i.e., BMP 1-13), some of which are described in 'U.S. Patent Nos.
5,106,748 and 5,658,882 and in PCT Publication No. WO 96/39,170.
Purified osteoinductive factors have been developed by several entities. Collagen Corporation of Palo Alto, Calif., developed a purified protein mixture that is purported to have osteogenic activity, as described in U.S. Patent Nos. 4,774,228, 4,774,322, 4,810,691, and 4,843,063.
Urist developed a purified protein mixture which is p-urported to be osteogenic, as described in U.S. Patent Nos. 4,455,256, 4,619,989, 4,761,471, 4,789,732, and 4,795,804, International Genetic Engineering, Inc. of Santa Monica, Ca if., developed a p-urified protein mixture that is purported to be osteogenic, as described in U.S.
, Patent No. 4,804,744.
[0951 One osteoinductive factor that may be used in the osteoinductive composition is described in detail in U.S. Patent No. 5,290,763, This osteoinductive factor has a high osteogenic activity and degree of purity. The osteoinduefive factor of the '763 patent exhibits osteoinductive activity at about 3 micrograms when deposited onto a suitable carrier and implante,r1 subcutaneously into a rat. In one embodiment, the osteoinductive factor is an, osteoinductively active mixture of proteins that exhibit the gel separation profile shown in Fig. 1 of U.S. Patent No. 5,563,124.
[090] In some embodiments, bone stimulating growth factors, for example recovered from the mineral phase of bone, may be puriBed using a apatite affinity chromatography scheme. Thus, mineralized or surface = demineralized bone may be used as a chromatography resin. Bone minaret comprises calcium phosphate sales similar to hydroxyapatite. To use mineralized or surface denainerati7ed bone as a clu-omatography resin, excess lipid and protein = may be removed from the surfaces of the bone. In other embodiments, a similar scheme maybe done using demine-ralized bone matrix as a resin. In yet fiuther embodiments, recovered inorganic bone mineral (sintered or unsintered) may be = used as the chromatography resin_ [097] In one embodiment, the protocol for such schenae may be as follows. Mineralized bone particles, for example ranging from 100/tm to 5nun, are prepared. The surface of the mineralized bone particles is cleaned, for example by soaking or stirring the bone particles in a dilute base such as 0.1M
NaOH for several minutes. Generally, such surface cleaning removes proteins as well as lipids. In alternative embodiments, surface cleaning may be performed using supercritical CO2. Growth factor extracts from the mineral phase may be solubalized in a chaotropic solvent such as 6M urea. The growth factor solution may then be mixed with the mineralized bone particles, for example, for several minutes. During such mixing, proteins having an affinity for hydroxyapatite bind to the bone surfaces. The bone-protein complex is then precipitated and the supernatant removed. The bone-protein complex may be treated to remove weakly bound proteins such as collagen fragments while retaining osteoinductive proteins (the osteoinductive proteins remain bound to the material). Such treatment may comprise treatingthe bone-protein complex with a 6M urea containing low concentrations of sodium phosphate. The treated bone-protein complex may be centrifuged and the supernatant aspirated. In some embodiments, the bone-protein complex may be treated with urea containing higher concentrations of sodium phosphate (e.g., 100m.M, 180mM, or 250mM) to release bound osteoinductive proteins. Alternatively, the bone-osteoinductive protein complex may be lyophilized and formulated with a carrier, for example for orthopedic applications. Further, the bone protein comples may be used as a growth factor microcarrier that can. be distributed in a DBM macrocarrier.
[098] In yet a further embodiment, an osteoinductive composition with reduced immunogenicity is provided. The osteoinductive composition comprises noncollagenous proteins and mineral recovered acid. The noncollagenous proteins may be extracted from demineralized bone or recovered from acid used to demineralize bone. The mineral recovered acid may be from acid used to demineralize bone. The noncallegnous proteins and mineral recovered acid may be combined such that the osteoinductive composition exhibits the ability to induce the formation of heterotopic bone in a normal (euthymic) mouse.
[099] V. OPTIONAL PROCESSING
[0100] As mentioned above, in some instances it may be desirable to remove inhibitors or concentrate the osteoinductive factors. This is optional and may be done by any suitable method. Generally, it may be desirable to remove the inhibitors quickly without denaturing the osteoinductive factors.
[0101] Suitable osteoinductive factors may be obtained by purification of naturally occurring proteins from bone or by recombinant DNA techniques. As used herein, the term recombinantly produced osteoinductive factors refers to the production of osteoinductive factors using recombinant DNA technology. For example, nucleic acids encoding proteins having osteogenic activity can be identified by producing antibodies that bind to the proteins. The antibodies can. be used to isolate, by affinity chromatography, purified populations of a particular osteogenic protein. The amino acid sequence can be identified by sequencing the purified protein. It is possible to synthesize DNA oligon-ucleotides from the known amino acid sequence. The oligonucleotides can be used to screen either a gen.omic DNA and/or cDNA library made from, for example, bovine DNA, to identify nucleic acids encoding the osteogenic protein. The correct oligonucleotide will hybridize to the appropriate cDNA, thereby identifying the cDNA encoding the osteogenic protein encoding gene.
0102] In other embodiments, the DBM may include and/or be treated with agents that inhibit the activity of one or more activating enzymes, proteases, or glycosidases. Such inhibitory agents are expected to reduce the activity of specific enzymes (whether derived from the host or from the DBM) that would otherwise interact with osteoinductive agents or other active agents in the DBM, thereby reducing osteoinductivity or wound healing.
10103] The treatment or condition may cleave an inhibitory factor that would otherwise inhibit a positively acting agent (by which is meant an agent that enhances a biological activity of the bone matrix). For example, a variety of proteins or protein fragments are known to inhibit the osteoinductive and/or osteogenic activity of certain bone morphogenetic proteins such as BMP-2. In certain embodiments the inhibitory effect of a protein or protein fragment is reduced by exposing to a treatment or condition that causes the cleavage or degradation of the inhibitory agent. The treatment or condition may block the interaction of the inhibitory agent with its target (e.g., BMP-2) or may inhibit synthesis, secretion, post-translational modification, transport, etc., of the inhibitory agent. For example, the bone may be exposed to antibody to an inhibitory agents or the antibody can be added to the bone.
[0104] As will be appreciated by those skilled in the art, factors having osteoinductive, osteogenic, and/or chondrogenic activity can be inhibited by a variety of mechanisms including proteolytic degradation, binding or sequestration of the factor, etc. A variety of proteins or protein fragments inhibit the osteoinductive and/or osteogenic activity of certain bone m.orphogenetic proteins, such as BlVIPs -2, -4, -5, -6, and -7. Among these inhibitory agents are noggin, chordin, gremlin, Dan, Cerberus, the protein related to Dan and Cerberus (PRDC), caronte, Dante, sclerostin, follistatin, follistatin-related gene (FLRG), ventroptin, and alpha2 HSglycoprotein. Noggin blocks the effect of BMPs in cells of the osteoblastic lineage, and the addition of noggin to osteoblasts in culture blocks BMP-induced synthesis of collagen and noncollagen proteins, and also inhibits the stimulatory effect of BMPs on alkaline phosphatase activity. Chordin acts in a similar fashion. Further details regarding these inhibitory agents are found in Canalis, supra, and references cited therein.
[0105] Certain collagen fragments are also believed to inhibit BMPs.
For example, a potentially inhibitory collagen fragment corresponds to the C-terminal end of procollagen that is released during extracellular matrix remodeling and collagen assembly. The sequence for a chordin-like collagen fragment (from Type I collagen) is YVEFQEAGSC VQDGQRYNDK DVWKPEPCRI
CVCDTGTVLC DDIICEDVKD CLSPEIPFGECCPICPADLAAAA (SEQ JD
NO: 1).
[0106] Bone or cartilage inhibitory factors (BCIF) can be inactivated or inhibited by a variety of methods. For example, a specific protease that cleaves or degrades the BCIF can be used. Another approach is to use an antibody that binds to the BCIF and blocks its interaction with a positively acting factor such as BMP-2 or BMP-4. The antibody may inhibit post-translational modification, transport, etc., of the inhibitory agent. Antibodies to the inhibitory agents mentioned herein (and others) are known in the art or could be generated using known methods without undue experimentation. Either polyclonal or monoclonal antibodies, or antigen-binding fragments thereof, can be used. Other agents having specific binding ability (e.g., affibodies) could likewise be used. One of ordinary skill in the art will be able to generate appropriate antibodies, affibodies, etc., based upon the known sequences of the inhibitory proteins.
[0107] VI. PROVIDE A CARRIER
[0108] Thus, the extracted osteoinductive factors (and possibly inhibitors) may be added to a carrier. For ease of reference, unless otherwise noted, reference to osteoinductive factors refers to osteoinductive factors with or without inhibitors. When the osteoinductive factors are added to a carrier, the carrier acts first as a bulking means for applying a small amount of extracted material.
The carrier also may serve as a scaffold, and may aid in controlling release kinetics.
Suitable carriers include DBM, including surface demineralized bone;
mineralized bone; nondemineralized cancellous scaffolds; demineralized cancellous scaffolds; particulate, demineralized, guanidine extracted, species-specific (allogenic) bone; specially treated particulate, protein extracted, demineralized, xenogenic bone; collagen; synthetic hydroxyapatites; polymers;
hydrogels; starches; polyethylene glycol, tricalcium phosphate, sintered hydroxyapatite, sellable hydroxyapatite; polylactic acid; tyrosine polycarbonate;
calcium sulfate; collagen sheets; sellable calcium phosphate; polymeric cements;
settable poly vinyl alcohols, polyurethanes; resorbable polymers;
polysaccharides and other large polymers; liquid sellable polymers; and other biocompatible sellable materials. Settable materials may be used, and they may set up either in situ, or prior to implantation. Optionally, xenogenic bone powder carriers also may be treated with proteases such as trypsin. Preferably, xenogenic carriers are treated with one or more fibril modifying agents to increase the intraparticle intrusion volume (porosity) and surface area. Useful agents include solvents such as dichloromethane, trichloroacetic acid, acetonitrile and acids such as trifluoroacetic acid and hydrogen fluoride. The osteoinductive factors and carrier (or delivery or support system) together form an osteoimplant useful in clinical applications.

[0109] Any suitable shape, size, and porosity of carrier may be used. Rat studies show that the new bone is formed essentially having the dimensions of the device implanted. Generally, particle size influences the quantitative response of new bone; particles between 70p.rn and 420pm elicit the maximum response.
However, other particle sizes may be used. Contamination of the carrier with bone mineral may inhibit bone formation.
[0110] In some embodiments, the carrier may be settable and/or injectable. Such carrier may be, for example, a polymeric cement, a settable calcium phosphate, a settable poly vinyl alcohol, a polyurethane, or a liquid settable polymer. Suitable settable calcium phosphates are disclosed in U.S.
Patent Nos. 5,336,264 and 6,953,594.
[0111] A successful carrier for osteoinductive factors performs several functions_ It carries osteoinductive factors and allows appropriate release kinetics. A successful carrier also appropriately accommodates each step of the cellular response during bone development, and in some cases protects the osteoinductive factors from nonspecific proteolysis. In addition, selected materials must be biocompatible in vivo and optionally biodegradable. In some uses, the carrier must act as a temporary scaffold until feplaced completely by new bone. Polylactic acid (PLA), polyglycolic acid (PGA), and various combinations. have different dissolution rates in vivo_ In bone, the dissolution rates can vary according to whether the implant is placed in cortical or trabecular bone.
[0112] The carrier may comprise a shape-retaining solid made of loosely adhered particulate material, e.g., with collagen. It may also comprise a molded, porous solid, or simply an aggregation of close-packed particles held in place by surrounding tissue. Masticated muscle or other tissue may also be used. Large allogenic bone implants may act as a carrier if their marrow cavities are cleaned and packed with particles and the osteoinductive factors.
[0113] The osteoimplant resulting from the carrier and the osteoinductive factors may assume a determined or regular form or configuration such as a sheet, plate, disk, tunnel, cone, or tube, to name but a few. Prefabricated geometry -3s-would include, but not be limited to, a crescent apron for single site use, an I-.
shape to be placed between teeth for intra-bony defects, a rectangular bib for defects involving both the buccal and lingual alveolar ridges, neutralization plates, reconstructive plates, buttress plates, T-buttress plates, spoon plates, clover leaf plates, condylar plates, compression plates, bridge plates, or wave plates.
Partial tubular as well as flat plates can be fabricated from the osteoimplant. Such plates may include such conformations as, e.g., concave contoured, bowl shaped, or defect shaped. The osteoimplant can be machined or shaped by any suitable mechanical shaping means. Computerized modeling can provide for the intricately-shaped three-dimensional architecture of an osteoimplant custom-fitted to the bone repair site with great precision.
[0114] In one embodiment, the osteoimplant induces endochondral bone formation reliably and reproducibly in a mammalian body. The carrier comprises particles of porous materials. The pores must be of a dimension to permit progenitor cell migration into the carrier and subsequent differentiation and proliferation. The particle size should be within the range of 70 µm-850 preferably 70 ,um-420 pm, most preferably 150 inn-420 Am. It may be fabricated by close packing particulate material into a shape spanning the bone defect, or by otherwise structuring as desired a material that is biocompatible, and preferably biodegradable in vivo to serve as a "temporary scaffold" and substratum for recruitment of migratory progenitor cells, and as a base for their subsequent anchoring and proliferation. Useful carrier materials include collagen;
homopolymers or copolymers of glycolic acid, lactic acid, and butyric acid, including derivatives thereof; and ceramics, such as hydroxyapatite, tricalcium phosphate and other calcium phosphates. Combinations of these carrier materials also may be used.
[0115] Mineralized or surface deminralized bone particles may alternatively be used as a carrier. Thus, an osteoinductive composition comprising mineralized or surface demienralized bone particles and extracts of DBM is provided. The DBM extracts may be adsorbed to the surfaces of the mineralized or partially demineralized bone particles. Weakly bound components may be eluted using, for example, low concentrations of sodium phosphate (for example, 5m.M to 50mM), thereby concentrating the growth factors. In some embodiments, analysis of the proteins bound to the surfaces of the mineralized or surface demineralized bone particles indicaes a ratio of Histone H2A to total protein bound elevated by a factor of 2 to 10,000 times over the normal ratio found in extracts of demineralized bone matrix or protein recovered from acid used to demineralize bone. In some embodiments, analysis of the proteins bound to the surfaces of the mineralized or surface demineralized bone particles indicates a ratio of Secreted Phosphoprotein 24 to total protein bound elevated by a factor of 2 to 10,000 times over the normal ratio found in extracts of demineralized bone matrix or protein recovered from acid used to demineralize bone. In some embodiments, analysis of the proteins bound to the surfaces of the mineralized or surface demineralized bone particles indicates a ratio of BMP-2 to total protein bound elevated by a factor of 2 to 10,000 times over the normal ratio found in extracts of demineralized bone matrix or protein recovered from acid used to demineralize bone. In some embodiments, analysis of -the proteins bound to the surfaces of the mineralized or surface demineralized bone particles indicates a ratio of BMP-4 to total protein bound elevated by a factor of 2 to 10,000 times over the normal ratio found in extracts of demineralized bone matrix or protein recovered from acid used to demineralize bone.. In some embodiments, analysis of -the proteins bound to the surfaces of the mineralized or surface demineralized bone particles indicates a ratio of TGF-Beta to total protein bound elevated by a factor of 2 to 10,000 times over the normal ratio found in extracts of demineralized bone matrix or protein recovered from acid used to demineralize bone.
[0116] USE OF DBM AS CARRIER
[0117] Any of a variety of DBM preparations may be used as a carrier.
DBM prepared by any method may be employed, including particulate or fiber-based preparations, mixtures of fiber and particulate preparations, fully or partially demineralized preparations, mixtures of fully and partially demineralized preparations, and surface demineralized preparations. See U.S. Patent 6,326,018, Reddi et al., Proc. Natl. Acad. Sci. USA (1972) 69:1601-1605; Lewandrowski et al., Clin. Ortho. Rel. Res., (1995) 317;254-262; Lewandroski et al., J. Blamed Mater. Res. (1996) 31:365-372; Lewandro-wski et al. Calctfied Tiss. Int., (1997) 61;294-297; Lewandrowslci et aL,.1. Ortho. Res. (1997) 15:748-756.
Preferred demineralized bone matrix compositions 'are described in U.S. Patent No. 5,507,813.
The DBM may be in the form of a section that substantially retains the shape of the original bone (or a portion thereof) from which it was derived.
Also , useful are DBM preparations comprising ailditives or carriers such as polyhydroxy compounds, polysaccharides, glycosanainoglyean proteins, nucleic acids, po/ymers, polaxomers, resins, clays, calcium salts, and/or derivatives thereof.
[0118] The DBM component may be ground or otherwise processed into particles of an appropriate size before or after demineralization. In certain embodiments, the particle size is greater than 75 microns, more preferably ranging from about..100 to about 3000 microns, and most preferably from about 200 to about 2000 microns. After grinding the DBM component, the mixhire may be sieved to select those particles of a desired size. In certain 'embodiments, the DBM particles may be sieved though a 50 micron sieve, more preferably a 75 micron sieve,. and most preferably a 100 micron sieve.
[0119) One way to protect small size particles from cellular ingestion and/or provide a diffusion barrier is to embed them in a monolithic bioabsorbable matrix, and then fragment the particle-containing monolithic matrix into particle sizes greater than 70 microns, preferably greater than 100 microns, and most preferably greater than 150 microns in their smallest dimension. Preferred raatrices for embedding small DBM particles include biocompatibIe polymers and setting calcium phosphate cements. Generally the particulate DBM/polymer weight ratio will range from. about 1:5 to about 1:3. ln the case of calcium phosphate, the DBM will be present up to 75% by weight. Particulation of the monolith can be accomplished by conventional milling or grinding, Or through the use of cryomilling, or freezing followed by pulverization. In one embodiment, lyophilized DBM is embedded in a resorbable polymer. In a further embodiment, lyophilized DBM
is embedded in one of the setting calcium phosphates known to the art.
101201 Following particulation, the DBM is treated to remove mineral from the bone. While hydrochloric acid is the industry-recognized demineralization agent of choice, the literature contains numerous reports of methods for preparing DBM (see, for example, Russell et al., Orthopaedics 22(5):524-53 1, May 1999). Any material that provides a scaffolding containing active osteoinductive factors is considered DBM. The DBM may be prepared by methods known in the art or by other methods that can be developed by those of ordinary skill in the art without undue experimentation. In some instances, large fragments or even whole bone may be demineralized, and then paiticulated following demineralization.
101211 In one embodiment, an osteoinductive composition comprising partially deminalized bone matrix particles is provided. The partially demineralized bone matrix particles may, for example, range in size from 500 pm to 4 mm. In one embodiment 10-80% of the mineral of the mineral content of the bone is removed. The partially demineralized bone may be heated to temperatures ranging from approximately 40 C to approximately 120 C for period of time ranging from approximately 1 minute to approximately 96 hours. Heating may be done with the partially demineralized bone in a dry state, in distilled water, in a neutral buffer solution, or other. The osteoinductive composition may exhibit the ability to induce the formation of heterotopic bone in a higher order animal such as a dog, human, or sheep. In some embodiments, the osteoinductive composition may be combined with osteoinductive growth factors extracted from bone, recovered from acid used to demineralized bone, or other.
101221 Mixtures of one or more types of demineralized bone-derived elements can be employed. Moreover, one or more of types of demineralized bone-derived elements can be employed in combination with non-demineralized bone-derived elements, i.e., bone-derived elements that have not been subjected to a demineralization process. Thus, e.g., the weight ratio of non-demineralized to demineralized bone elements can broadly range from about 0:1 to about 1:0.
Suitable amounts can be readily determined by those skilled in the art on a case-by-case basis by routine experimentation.
[0123] An osteoi3aductive dcmineralized bone matrix scaffold is thus further provided. In one embodiment, the osteoinductive denaineralized bone matrix scaffold comprises bone derived components and exhibits, without collagensae pretreatement, the ability to induce specific alkaline phosphotase activity levels higher than those of standard derruneralized bone matrix preparations, for example, 2 to 100,000,000 higher than standard deranneralized bone matrix preparations in cultured C2C12 cells, In another embodiment, the osteoinductive demineralized bone matrix scaffold comprises osteoinductive growth factors, for example extmcte,d from DBM or recovered from acid baths used for demineralization of the bone matrix, and deminerali7ed bone matrix and exhibits the ability to induce specific alkaline phosphotase activity levels higher than those of standard demineralized bone naatrix preparations, for example, 2 to 100,000,000 higher than standard dermineralized bone matrix preparations in cultured C2C12 cells.
[01241 In addition to the demineralizing step, the bone is optionally subjected to a configuring step to form an implant The configuring step can be employed using conventional equipment known to those skilled in the art to produce a wide variety of geometries, e.g., concave or convex surfaces, stepped surfaces, cylindrical dowels, wedges, blocks, screws, and the Like. A
surgically implantable material fabricated from elongated bone particles that have been demineralized, which rnay be shaped as a sheet, and processes for fabricating shaped materials from demineralized bone particles are disclosed in U.S.
Patent Nos. 5,507,813 and 6,436,138, respectively.
Suitable sheets include those sold under the trade name Graf-Ion DBM Flex, which must be wetted/hydrated prior to use to be useful for implantation. Such sheets have recently been reported as effective in seeding hurnan bone marrow stromal cells (BMSCs), which may be useful in the repair of large bone defects. Kasten et al., "Comparison of Human Bone Marrow Stromal Cells Seeded on Calcium-Deficient Hydroxyapatite, Betatricalcium Phosphate and Deraineraliz.ed Bone Matrix," Biomaterlals, 24(15):2593-603, 2003. Also useful are demineralized bone and other matrix preparations comprising additives or carriers such as binders, fillers, plasticizers, wetting agents, surface active agents, biostatic agents, biocidal agents, and the like. Some exemplary additives and carriers include polyhydroxy compounds, polysaccharides, glycosarainoglycan proteins, nucleic acids, polymers, polaxoraers, resins, clays, calcium salts, and/or derivatives thereof.
[0125] The bone used in creating the bone matrix may be obtained from any source of living or dead tissue. Often, it will be preferred that the source of bone be matched to the eventual recipient of the inventive composition. At a minimum, it is ofteu desirable tbat the donor and recipient are of the same species, though even xenogenic sources are permitted. Thus for use in humans, it is generally preferred to use DBM derived at least in part from human bone. For example, the bone material maybe at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more human bone material. In certain embodiments 100% of the bone material is human bone material.
[0125] The matrix may be completely insoluble or may be slowly solubilized after implantation. Following implantation, preferred matrices resorb or degrade, xemaining substantially intact for at least one to seven days, most preferably for two or four weeks or longer and often longer than 60 days.
Bioactive agents may be endogenously present in the matrix as in the case of most demineralized bone, or they may be exogenously added to the matrix.. Matrices may also comprise cornbinations of endogenous and exogenous bioactive agents.
[0127] The matrix may comprise a number of materials in combination, some or all of which may be in the form of fibers and/or particles. The matrix may comprise calcium phosphates. Driessens et aL "Calcium phosphate bone cements," Wise, D.L., Ed., Encyclopedic Handbook of Biomaterials and Bioengineering, Part B, Applications New York: Marcel Decker; Elliott, Structure and Chemistry of the Apatites and Other Calcium Phosphates Elsevier, Amsterdam, 1994. Calcium phosphate matrices include, but are not limited to, dicalcium phosphate dihydrate, monetite, tricalcium phospate, tetracalcium phosphate, hydroxyapatite, nanocrystalline hydroxyapatite, poorly crystalline hydroxyapatite, substituted hydroxyapatite, and calcium deficient hydroxyapatites.
[0128] In one embodiment, an osteoinductive material is provided comprising a mineralized particulated material, osteoinductive growth factors, and a demineralized bone matrix scaffold. The mineralized particulated material may be TCP, hydroxyapatite, mineral recovered from bone, cancellous chips, cortical chips, surface demineralized bone, or other material. The osteoinductive growth factors may be synthetically derived, extracted from demineralized bone matrix, recovered from demineralization acid bath, or other. The demineralized bone matrix scaffold may be combined with a carrier such as starch or glycerol. In various embodiments, the osteoinductive growth factors may be solubilized and combined with the mineralized particulate material, thus allowing adsorption of the growth factors onto the mineral surfaces, as previously described. The growth factor/mineral composite may then be distributed throughout the demineralized bone matrix scaffold. The growth factor/mineral composite thus may comprise a microcarrier and the demineralized bone matrix scaffold may comprise a macrocanier. The size of the microcarrier mineralized particles may range, for example, from 50 nm to 5 mm.
[0129] In another embodiment, an osteoinductive composition with reduced immunogenicity is provided comprising noncollagenous proteins and a demineralized bone matrix scaffold. The noncaollagenous proteins may be extracted, for example, from demineralized bone or recovered from acid used to demineralize bone. The noncollagenous proteins and the demineralized bone matrix scaffold may be combined such that the osteoinductive composition exhibits the ability to induce the formation of heterotopic bone in a normal (euthyrnic) mouse.
[0130] TREATMENT OF CARRIER
[0131] In other embodiments, the present invention further provides methods of increasing the osteoinductivity of a carrier by exposing the carrier to at least one treatment (e.g., a biological or chemical agent). In addition to dispersion of the extracted osteoinductive factors onto the carrier, the carrier may be exposed to a chemical or condition that selectively degrades inhibitors of osteogenic activity and/or to a chemical or condition that activates osteoinductive factors in the carrier. Thus, the resulting carrier has an increased osteoinductivity, osteogenic, or chondrogenic activity compared to a similar carrier not exposed to the treatment or condition, because inhibition of an osteoinductive, osteogenic, or chondrogenic factor is blocked. In general, agents that inhibit or reduce osteoinductive, osteogenic, or chondrogenic activity may be referred to as bone/cartilage inhibitory factors (BCIF).
[0132] Reduction of Inhibitors [0133] As stated above, the carrier may be treated to reduce inhibitors of osteoinductive factors. As will be discussed more fully below, the extracted osteoinductive factors are added to the carrier. The addition of the osteoinductive factors and the treatment of the carrier to reduce inhibitors may be performed in combination or sequentially. One or more rounds of treatment may be used, i.
e., the treatments may alternate.
[0134] Thus, the carrier may be treated to cleave or degrade a specific protein such as an inhibitor of BMP. The carrier is treated such that a specific protein that is not a major structural component of the carrier is affected.
The specific protein generally makes up less than 1% of the dry weight of the matrix, e.g., less than 0.5%, less than 0.1%, etc. The specific protein can be a negatively acting factor, e.g., an inhibitor of a BMP or an inhibitor of a BMP signaling pathway, wherein cleavage or degradation of the inhibitor allows increased activity of the protein that it would otherwise inhibit.
[0135] Alternatively or additionally, the carrier may include inhibitory agents presented in a time-release formulation (e.g., encapsulated in a biodegradable polymer). In the case of activating enzymes, i.e., enzymes that lead to the release, presentation, or creation of osteoinductive factors), inhibitory agents that reduce the activity of activating enzymes preferably lead to increased osteoinductivity over an extended period of time rather than just a burst just after implantation.
[0136] Activation [0137] Certain of the osteoinductive or chondrogenic factors found in a bone or cartilage matrix are in cryptic form and must be "activated" or "released"
to be osteoinductive. The activation of osteoinductive factors may involve a confonnational change, a post-translational modification, protein cleavage, a change in tertiary or quaternary structure, or release from a binding protein.
The osteoinductive factors, either those extracted and added to the carrier or those already present in the carrier, may be in a pre- or pro-form, which requires proteolytic cleavage to be active. The osteoinductive factors also may be associated with a binding protein or a protein of a bone or cartilage matrix.
Proteolysis may also be involved in the activation or inactivation of a binding protein, which could result in activation of the osteoinductive peptide or protein fragment. Therefore, all treatments of a bone or cartilage matrix with any specific or non-specific condition may affect activation rates of osteoinductive peptides and protein fragments.
[0138] The presence or activation of peptides and/or protein fragments having osteoinductive or chondrogenic activity may compensate for degradation of osteoinductive or chondrogenic proteins in the carrier, which may occur during preparation of the carrier. In certain embodiments it may be desirable to both inhibit the degradation of osteoinductive or chondrogenic factor and. activate or add osteoinductive or chondrogenic factors such as osteoinductive or chondrogenic peptides or protein fragments. Variables such as pH and ion concentration may affect protein function and/or folding of the peptide or protein fragment, and may affect the activation of osteoinductive or chondrogenic factors.
These variables also may affect the release of an osteoinductive factor from its binding protein. For example, where pH plays a role in the activation of a factor, the carrier may include a chemical compound such as a polymer that will break down over time and release an acid by-product; thereby activating the osteoinductive factors within the carrier. Alternatively, a biodegradable polymer may release ions or a protease that is able to "activate" the osteoinductive factors of the carrier.
[0139] In certain embodiments, one or more enzymes, such as proteases, lipases, and glycosidases, are added to the carrier to activate the osteoinductive or chondrogenic factors already present (e.g., to convert one or more osteoinductive factors from an inactive to an active form or from an active form to a more active form, or from a form that is susceptible to degradation to a form that is less susceptible to degradation, e.g., a form that has a longer half-life).
[0140] Many of the growth factors responsible for the osteoinductive or chondrogenic activity of a carrier, such as a bone matrix carrier, exist in cryptic form, in the carrier, until activated. Activation can involve the change of a pre or pro function of a factor, or release of the function from a second factor or entity, which binds to the first growth factor. For example, proteolytic cleavage results in separation of the inactive proprotein (e.g., a proprotein from the TGF
superfamily of proproteins, e.g., TGF-(3) and release of an active, mature peptide). As proteins of bone and cartilage matrices degrade naturally or artificially, they break down into peptides and protein fragments that contain active domains and function as receptor ligands and signal transducers in bone and cartilage growth signaling pathways. These reactions may be promoted to enhance osteoinductive and chondrogenic signaling in the carrier.
[0141] Generate Osteoinductive Peptides and/or Protein Factors in the Carrier [0142] A wide variety of agents, selected from biological agents such as enzymes, chemicals, and conditions, can be used to generate osteoinductive peptides and protein fragments, and these are well known in the art. The proteases, chemicals, and conditions of the present invention can be site specific, amino acid site specific, protein specific, semi-site-specific, lipid specific, or sugar specific. Enzymes may be obtained from endogenous, exogenous, autogenic (autologous), allogenic, or xenogenic sources. They may be purified from natural sources or produced recombinantly. In many embodiments the enzymes are purchased from commercial sources (Worthington Biochemical Industries, Sigma, etc.) and either used directly or subsequently purified to be free of contaminants that may negatively affect the activity of the final product.
Enzymes, peptides, or protein fragments (e.g., generated by particular proteases) and other treatments may also be purified by conventional methods. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y.;
Ausubel et al. "Current Protocols in Molecular Biology, Greene Publishing Associates, New York, V 1 & 2 1996. Purification can be carried out by a variety of chromatographic techniques. Size exclusion chromatography is commonly used. Other methods include ion exchange, hydrophobic interaction, and affinity chromatography. Peptides or protein fragments may be used in the bone or cartilage repair matrix as unpurified preparations as long as the peptides or protein fragments maintain their osteoinductive or chondrogenic activity.
Alternatively, the enzymes, peptides, or protein fragments can be synthesized artificially using conventional techniques, produced recombinantly, etc. It may be preferable to use preparations having a high degree of purity. For example, an enzyme preparation may contain at least 90%, at least 95%, at least 98%, at least 99%
of the enzyme by weight. The preparation may be essentially free of bacterial components, particularly bacterial components that could cause inflammatory or immunological reactions in a host, such as endotoxin, lipopolysaccharide, etc.

Preparations having a purity greater than 99.5% can be used.
[0143] One suitable protease is a collagenase. Collagenases and their activity on collagens of various types have been extensively studied. A number of collagenase preparations are available from Worthington Biochemical Corporation, Lakewood, NJ. As described on the company's web site and known in the art, collagen consists of fibrils composed of laterally aggregated, polarized tropocollagen molecules (MW 300,000). Each tropocollagen unit consists of three helically wound polypeptide a-chains around a single axis. The strands have repetitive glycine residues at every third position and numerous proline and hydroxyproline residues, with the particular amino acid sequence being characteristic of the tissue of origin. Tropocollagen units combine uniformly to create an axially repeating periodicity. Cross linkages continue to develop and collagen becomes progressively more insoluble and resistant to lysis on aging.

Gelatin results when soluble topocollagen is denatured, for example on mild heating, and the polypeptide chains become randomly dispersed. In this state the strands may readily be cleaved by a wide variety of proteases.
[01441 In general, a variety of different collagenases known in the art can be used. Collagenases are classified in section 3.4.24 under the International Union of Biochemistry and Molecular Biology (NC-IUBMB) enzyme nomenclature recommendations (see, e.g., 3.4.24.3, 3.4.24.7, 3,4.24.19). The collagenase can be of eukaryotic (e.g., mammalian) or prokaryotic (bacterial) origin. Bacterial enzymes differ from mammalian collagenases in that they attack many sites along the helix. Collagenase may cleave simultaneously across all three chains or attack a single strand. Preferably the collagenase cleaves Type I
collagen, e.g., degrades the helical regions in native collagen, preferentially at the Y-Gly bond in the sequence Pro-Y-Gly-Pro-, where Y is most frequently a neutral amino acid. This cleavage yields products susceptible to further peptidase digestion. Any protease having one or more of these activities associated with collagenase may be used as a collagenase in accordance with the present invention.
[01451 It will be appreciated that crude collagenase preparations contain not only several collagenases, but also a sulfhydryl protease, clostripain, a trypsin-like enzyme, and an aminopeptidase. This combination of collagenolytic and proteolytic activities is effective at breaking down intercellular matrices, the essential part of tissue disassociation. Crude collagenase is inhibited by metal chelating agents such as cysteine, EDTA, or o-phenanthroline, but not DFP. It is also inhibited by aa-macroglobulin, a large plasma glycoprotein. Ca2+ is required for enzyme activity. Therefore, it may be desirable to avoid collagenase inhibiting agents when treating bone matrix with collagenase. In addition, although the additional proteases present in some collagenase preparations may aid in breaking down tissue, they may also cause degradation of desired matrix constituents such as growth factors. Therefore, it may be desirable to use a highly purified collagenase that contains minimal secondary proteolytic activities along with high collagenase activity. For example, a collagenase preparation may contain at least 90%, at least 95%, at least 98%, at least 99% collagenase by weight. The preparation may be essentially free of bacterial components, particularly bacterial components that could cause inflammatory or immunological reactions in a host, such as endotoxin, lipopolysaccharide, etc.

Preparations having a purity greater than 99.5% can be used. A suitable preparation is chromatographically purified CLSPA collagenase from Worthington Biochemical Corporation. It may be desirable to include various protease inhibitors that do not inhibit collagenase but that inhibit various proteases that digest BMP. For example, protease inhibitors that are known to protect BMP activity from degradation include N-ethyl maleimide, benzamidine hydrochloride, iodoacetic acid, PMSF, AEBSF, E-64. Bestatin may also be used, particularly if the preparation contains aminopeptidase activity. Any of these protease inhibitors (or others) can be included in a carrier, such as a bone matrix composition, or in any composition that is used to treat a carrier.
[0146] Another suitable protease bone morphogenetic protein I (BMP-1).
BMP-1 is a collagenolytic protein that has also been shown to cleave chordin (an inhibitor of BMP-2 and BMP-4). Thus, BMP- I may be of use to alter the physical structure of the carrier (e.g., by breaking down collagen) and/or to cleave specific inhibitory protein(s), e.g., chordin or noggin. Proteins related to any of the proteases described herein, i.e., proteins or protein fragments having the same cleavage specificity, can also be used. It will be appreciated that variants having substantial sequence identity to naturally occurring protease can be used. For example, variants at least 80% identical over at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the length of naturally occurring protease (or any known active fragment thereof that retains cleavage specificity) when aligned for maximum identity allowing gaps can be used.
[1)147] Certain preferred proteases include members of the proprotein convertase (PPC) family of proteases, such as furin and related proteases.
Members of this family of cellular enzymes cleave most prohorrnones and neuropeptide precursors. Numerous other cellular proteins, some viral proteins, and bacterial toxins that are transported by the constitutive secretory pathway are also targeted for maturation by PCs. Furin and other PC family members share structural similarities that include a heterogeneous ¨10 kDa amino-terminal proregion, a highly conserved ¨55 lcDa subtilisin-like catalytic domain, and carboxyl-terminal donaain that is heterogeneous in length and sequence. These enzymes become catalytically active following proregion cleavage within the appropriate cellular compartment. Furin is the major processing enzyme of the secretory pathway and is locali7ed in the trans-golgi network. van den Ouweland, A. M. W. et al. (1990) Nucl. Acid Res. 18, 664; Steiner, D. F. (1998) Curr.
Opin.
Chem. Biol. 2, 31.-39. Substrates of furin include blood clotting factors, serum proteins, and growth factor receptors such as the insulin-like growth factor receptor. Bravo D. A. et al_ (1994) J. Biol. Chem. 269, 25830-25873. The minimal cleavage site for furin is Arg-X-X-Arg. However, the enzyme prefers the site Arg-X-(Lys/Arg)-Arg. An additional arginine at the P6 position appears to enhance cleavage. Krysan. D. J. et al. (1999) J. Biol. Chem. 274, 2322923234.
Fuxin is inhibited by EGTA, antitrypsin Portland, Jean, F. et al. (1998) Proc.
Natl. Acad. Sci. USA 95, 7293-7298, and polyarginine compounds, Cameron, A.
et al. (2000) J. Biol. Chem. 275, 36741-36749. Furin has been shown to proteolytically process both proTGF and proBIVIP proteins, for example, proTGF-g and proBMP-4, respectively, resulting in the release of the active mature form for cach molecule. Dubois et al., American Journal of Pathology (2001) 158(1):305-316; Cui et aL, The Embo Journal (1998) 17(16):47354743; Cui et al., Genes & Development (2001) 15:2797-2802, Furin has also been shown to cleave BMP-2, I3MP-6, and BMP-7. For example, furin cleaves between amino acids 282 and 283 in rnattu-e human BMP-2. Newly synthesized htunan BMP-2 contains a signal sequence (amino acids 1-23), a propeptide (amino acids 24-282), and an active portion (amino acids 283-396). Furin cleaves mature BMP-2 (amino acids 24-396) between amino acids 282 and 283 to release the propeptide and the active molecule.
[0148] Thus, the carrier, such as a DBM matrix, may be treated with PPCs such as furin and/or other proteases, which process immature TGF43 and/or BM_P
-49.

superfamily propeptides into their active mature forms and/or process active or inactive TGF-13 and/or BMP superfamily polypeptides into smaller active fragments that are resistant to degradation or inactivation relative to the longer polypeptide, generates a carrier with increased osteoinductivity compared to a canier lacking the protease, resulting in improved bone formation. The higher titers of the mature and/or degradation resistant species in these preparations increase the osteoinductive capacity of the carrier.
[0149] Allogenic cancellous demineralized bone is known not to be osteoinductive. When treated with collagenase enzymes for periods routinely used in the art, such as 24 hours, allogenic cancellous demineralized bone remains nonosteoinductive. Applicants have made the surprising discovery that allogenic cancellous bone, when treated with collagenase enzymes for approximately one hour, becomes osteoinductive, approximately as osteoinductive as allogenic cortical demineralized bone. While not desiring to be bound by any particular scientific theory, applicants state that it is believed that the disruption of the collagen matrix makes osteoinductive factors bioavailable. Many types of collagenalytic enzymes, such as those set forth herein, would be expected to render allogenic cancellous demineralized bone osteoinductive when treated as described herein. Other treatments also are expected to provide the same result, including the use of salts or ionizing or electromagnetic radiation, or various categories of enzymes, so long as the enzymes disrupt the collagen without damaging the osteoinductive factors.
[0150] DELIVERY SYSTEM
[0151] The carrier and the method of adding osteoinductive factors to the carrier, discussed more fully below, may result in an enhanced delivery system.
More specifically, the osteoinductive factors may be added to a carrier such that the osteoinductive factors are generally evenly dispersed in three dimensions as opposed to superficially coating a carrier. Dispersion throughout the carrier affects control of release kinetics of the osteoinductive factors from the carrier. In one embodiment, a plurality of thin sheets of carrier are provided. Each sheet of carrier is layered with osteoinductive factors. These sheets of carrier layered with osteoinductive factors are stacked.
[0152] Thus, optionally, the osteoimplant is formed as a laminate. A
laminate osteoimplant may advantageously be shaped in three dimensions, as in the introduction of a concave surface shape. Further, each layer of the laminate is continuous, without requiring binding of the joints between the pieces.
[0153] Assembling the superimposed layers into a strong unitary structure may be accomplished by a variety of means/procedures, e.g., application of known and conventional biologically compatible adhesives such as the cyanoacrylates; epoxy-based compounds, dental resin sealants, dental resin cements, glass ionorner cements, polymethyl methacrylate, gelatin-resorcinol-formaldehyde glues, collagen-based glues, inorganic bonding agents such as zinc phosphate, magnesium phosphate or other phosphate-based cements, zinc carboxylate, etc., and protein-based binders such as fibrin glues and mussel-derived adhesive proteins; the use of mechanical fasteners such as pins, screws, dowels, etc., which can be fabricated from natural or synthetic materials and bioabsorbable as well as nonbioabsorbable materials; laser tissue welding;
and, ultrasonic bonding. If desired, the layers of the osteogenic osteoimplant can be provided with mechanically interengaging features, e.g., tongue-and-groove, mortise-and-tenon, or dove-tail elements, to facilitate their assembly into the final product and/or to fix the layers to each other in a more secure fashion. The optimal method of assembly would be determined on a case-by-case basis through routine experimentation. In addition to its carrier and osteoinductive layers, the osteoimplant of this embodiment can optionally possess one or more layers formed from one or more other materials or substances.
[0154] In another embodiment, the carrier may comprise a single thin sheet of material. The delivery systems thus may comprise a single thin sheet of material coated with osteoinductive factors. The coated sheet of material may be rolled or folded over itself such that the growth factor content in the interior of the sheet approximates the growth factor levels at the surfaces.

[0155] VII. DISPERSION OF OSTEOINDUCTIVE FACTORS
ONTO CARRIER
[0156] The osteoinductive factors extracted are combined with the appropriate carrier. Exactly how this occurs can have a significant influence on the biological activity of the fmal formulation. The extracted osteoinductive factors may have been lyophilized, resulting in a powder composition. In some situations, adding a powder to a bone matrix may be challenging. Thus, it may be desirable to process the osteoinductive factors to form a homogenous mixture that may be more easily added to a carrier. This can have a significant impact on the release kinetics of the growth factor.
[0157] Thus, in a specific example, if the osteoinductive factors are lyophilized and then added to a DBM carrier, the solution is likely to be inhomogeneous, with most of the osteoinductive factors concentrated on the outside of the DBM carrier. If the osteoinductive factors are added to a carrier comprising very thin sheets of collagen and the carrier is then folded in on itself, the distribution of growth factor is more homogenous. The collagen sheets in such an embodiment can be very thin, on the order of microns.
[0158] Any suitable method for adding, or dispersing, the osteoinductive factors to the carrier may be used. Generally, the procedures used to formulate or disperse osteoinductive factors onto the carrier are sensitive to the physical and chemical state of both the osteoinductive factors and the carrier. The carriers could potentially be added to the extracts in their denatured state, such as in guanidine, allowing the growth factors to be precipitated directly onto the carriers.
[0159] In one embodiment, the osteoinductive factors are blended with a bulking agent to form a homogenous mixture. This mixture is added to the carrier.
[0160] Alternatively, the osteoinductive factors may be blended with coprecipitates (described more fully above) and this blend may be added to the carrier. For example, the osteoinductive factor and coprecipitate blend may be added to a carrier such as cancellous chips, synthetic calcium phosphate, or a liquid settable polymer. Generally, calcium phosphate is a liquid solid slurry while a polymer is a liquid. Thus, the choice of carrier may depend on the desired characteristics of the composition. Further, a lubricant, such as water, glycerol, or polyethylene glycol may be added.
[0161] In an alternative embodiment, the extracted osteoinductive factors may act as their own carrier. In a further embodiment, the above osteoimplant can be combined in various ways with other similar osteoinaplants or other materials to form an osteoimplant of laminate-type construction. For example, layers of the osteoimplant of the invention herein can be, through chemical or mechanical means, caused to adhere to each other; or, optionally, with other materials, e.g., reinforcing fibers, fabrics, meshes, etc., between some or all of the osteoimplant layers. Such laminate materials will differ from known osteoimplant laminates such as those disclosed in U.S. Patent No. 5,899,939, in that the final size and architecture will be determined by the total amount of starting donor material available rather than the specifi' c size or shape of the usable donor material available_ [0162] FORMULATION
[0163] The carrier, the osteoinductive composition, or the osteoimplant may be formulated for a particular use. The formulation may be used to alter the physical, biological, or chemical properties of the carrier. A physician would readily be able to determine the formulation needed for a particular application, taking into account such factors as the type of injury, the site of injury, the patient's heath, and the risk of infection. In various embodiments, the osteoinductive composition may comprise, for example less than approximately 0.5% water, less than approximately 1% water, or less than approximately 5%
water.
[0164] Carriers, osteoinductive compositions, or osteoiruplants therefore may be prepared to have selected resorption/loss of osteoinductivity rates, or even to have different rates in different portions of an implant. For example, the formulation process may include the selection of DBM particles of a particular size or composition, combined with the selection of a particular stabilizing agent or agents, and the amounts of such agents.

101651 In one example, it may be desirable to provide an osteoimplant whose osteoinductive factors are active in a relatively constant amount over a given period of time. A DBM carrier comprising factors with longer half-lives can be prepared using a less biodegradable polymer or a larger amount (e.g., a thicker coating) of polymeric compound. Altematively or additionally, the particle size may be important in determining the half-life ofthe osteoimplant. In certain embodiments, an inventive formulation may include a mixture of particles, each with a different half-life. Such a mixture could provide the steady or possible unmasking of osteoinductive factors over an extended period of time ranging from days to weeks to months depending on the needs ofthe injury.
Compositions such as this can be formulated to stimulate bone growth in a human patient comparable to the bone growth induced by treatment with 10 pg of rhBMP on a collagen sponge, and preferably comparable to 100 pg, and most preferably 1-10 mg rhBMP. When the degradation of the osteoimplant is of concern, it may be desirable to test the shelf-life of the osteoimplant to determine shelf-life at, for example, 1, 2, or 3 years.
This may be done by storing the osteoimplant at, for example, room temperature or, for accelerated testing, 38 degrees Celsius, and periodically checking the inductivity of the osteoimplant. Reference is made to PCT/US05/003092. Implants with enhanced shelf lives may retain more than about 75% and about 80% of their osteoinductivity after as long as, or longer than, three years.
[0166] Physical properties such as deformability and viscosity of the carrier may also be chosen depending on the particular clinical application. If DBM is used as a carrier, the particles of the DBM may be mixed with other materials and factors to improve other characteristics of the implant.
For example, the improved DBM material may be mixed with other agents to improve wound healing.
These agents may include drugs, proteins, peptides, polynucleotides, solvents, chemical compounds, and biological molecules.
[01671 Further, the composition may be formulated to be settable and/or injectable. Thus, for example, the composition may be injectable through a 10-gauge, a 12-gauge, or an 18-gauge needle.

O1 68] Particles of the carrier may also be formed into various shapes and configurations. The particles can be formed into any suitable configuration, including rods, strings, sheets, weaves, solids, cones, discs, fibers, and wedges. In certain embodiments, the shape and size of the particles in the carrier affect the time course of osteoixtductivity. For example, in a cone or wedge shape, the tapered end will result in osteoinduciivity shortly after implantation of the osteoimpIant, whereas the thicker end will lead to osteoinductivity later in the healing process (hours to days to weeks later). In certain embodiments, the , particle have a length of weater than 2 mm, greater than 1.5 nun, greater than 1 mm, preferably greater than 500 microns, and most preferably greater than 200 microns autoss its widest dimension_ Also, larger particle size vvill have induce bone formation over a longer time course than smaller particles. Particles of different characteristics (e.g., composition, size, shape) may be used in the formation of these different shapes and configurations. For example, in a sheet of DBM a layer of long half-life particles may be alternated between layers of shorter haLf-life particles. See U.S. Patent No. 5,899,939 for suitable examples. In a weave, strands composed of short half-life particles may be woven together with strands of longer half-lives.
[0169] In one embodiment, fibrous DBM is shaped into a matrix form carrier as described in U.S. Patent No. 5,507,813.
The shaped DBM is then embedded within a diffusion barrier type matrix, such that a portion of the matrix is left exposed free of the matrix material.
Particularly preferred blocking matrices are starch, phosphatidyl choline, tyrosine polyearbonates, tyrosine poiyarylates, polylaetides, polygalactides, or other resorbable polymers or copolymers. Devices prepared in this way front these matrices have a combination of immediate and longer lasting osteoinductive properties and are particularly useful in promoting bone mass formation in human posterolateral spine fusion indications.
101701 In another embodiment, carriers having a pre-selected three-dimensional shape are prepared by repeated application of individual layers of DBM, for exaniple by 3-D printing as described by 'U.S. Patent Nos. 5,490,962, 5,518,680, and 5,807,437. Different layers may comprise individual stabilized DBM preparations, or alternatively may comprise DBM layers treated with stabilizing agents after deposition of multiple layers.
t01711 In the process of pieparing the osteoimplant, the materials may be produced entirely aseptically or be sterilized to eliminate any infectious agents such as HIV, hepatitis B, or hepatitis C. The sterilization may be accomplished -using antibiotics, irradiation, chemical sterilization (e.g., ethylene oxide), or thermal sterilization. Other methods known in the art of preparing DBM such as defatting, sonication, and lyophilization may also be used in preparing a DBM
carrier. Since the biological activity of demineralized bone is known to be detrimentally affected by most terminal sterilization processes, care must be taken when sterilizing the inventive conapositions.
[0172 VJJI. OPTIONAL ADDITIVES
f0173] Optionally, other additives may be included ixt the osteoinductive bone matrix. It will be appreciated that the ammuit of additive used will vary depending upon the type of additive, the specific activity of the particular additive preparation employed, and the intended use of the composition. The desired amount is readily determinable by the user. Any of a variety of medically and/or surgically useful optional substances can be incorporated in, or associated with, the osteoinductive factors either before, during, or after preparation of the osteogenie composition.
[01743 In certain embodiments, the additive is adsorbed to or otherwise associated with the osteoimplant. The additive may be associated with the osteoirnplant through specific or non-specific interactions, or covalent or noncovalent interactions. Examples of specific interactions include those between a ligand and a receptor, an epitope and an antibody, etc. Examples of nonspecific interactions include hydrophobic interactions, electrostatic interactions, magnetic interactions, dipole interactions, van der Weals interactions, hydrogen bonding, etc. In certain embodiments, the additive is attached to the osteoimplant, for example, to the carrier, using a linker so that the additive is free to associate with its receptor or site of action in vivo. In other embodiments the additive is either covalently or non-covalently attached to the carrier. In certain embodiments, the additive may be attached to a chemical compound such as a peptide that is recognized by the carrier. In another embodiment, the additive is attached to an antibody, or fragment thereof, that recognizes an epitope found -within the carrier.
In certain embodiments at least additives are attached to the osteoimplant. In other embodiments at least three additives are attached to the osteoimplant.
An additive may be provided within the osteoimplant in a sustained release format.
For example, the additive may be encapsulated within biodegradable nanospheres, microspheres, etc.
[0175] It will be understood by those skilled in the art that the lists of optional substances herewith included are not intended to be exhaustive and that other materials may be admixed with bone-derived elements within the practice of the present invention.
[0176] Radiopaque Substances [0177] Radiopaque substances may be added to impart radiopacity to the composition. Examples of substances imparting radiopacity include for example, fully mineralized bone particles, Barium and Iodine containing compounds or compositions, e.g., Barium Sulfate and Barium Sulfate for Suspension, lopanoic Acid, and the like. When employed, substances imparting radiopacity will typically represent from about l to about 25 weight percent of the bone particle containing composition, calculated prior to forming the shaped material.
[0178] Angiogenesis Promoting Materials [0179] Development of a vasculature around the implant site may also be important to forming new bone ancl/or cartilage tissues. Angiogenesis may be an important contributing factor for the replacement of new bone and cartilage tissues. In certain embodiments, angiogenesis is promoted so that blood vessels are formed at the site to allow efficient transport of oxygen and other nutrients and growth factors to the developing bone or cartilage tissue. Thus, angiogenesis promoting factors may be included in the osteoimplant to increase angiogenesis in that region. For example, class 3 semaphorins, e.g., SEMA3, controls vascular morphogenesis by inhibiting integrin function in the vascular system, Serini et al., Nature, (July 2003) 424:391-397, and may be included in the osteoimplant [0180] Bioactive Agents [0181] The osteoconchictive composition may provide a system for delivering bioactive agents, such as osteoinductive factors, to a host animal.

Thus, the osteoimplant enables an improved healing response to the implant without the need to administer separately the bioactive agent. A problem with the introduction of the bioactive agent at the site is that it is often diluted and redistributed during the healing process by the circulatory systems (e.g., blood, lymph) of the recipient before complete healing has occurred. A solution to this problem of redistribution is to affix the bioactive components to the osteoimplant.
Some preferred bioactive agents that can be delivered using a DBM composition include agents that promote the natural healing process, i.e., resorption, vascularization, angiogenesis, new growth, etc. In one embodiment, the osteoimplant is provided in which DBM, together with a stabilizing agent, is used to deliver the biologically active agent. It is expected that the stabilizing agent will protect the biologically active agent from degradation, and therefore will extend its active life after delivery into the recipient animal. In certain embodiments, the bioactive agent is an osteoinductive agent, and in certain embodiments, the DBM may be used to deliver more than one bioactive agent, preferably more than two, and more preferably sometimes more than three bioactive agents. The bioactive agent may be associated with the DBM. For example, the bioactive agent may be associated with the DBM through electrostatic interactions, hydrogen bonding, pi stacking, hydrophobic interactions, van dcr Waals interactions, etc. In certain embodiments, the bioactive agent is attached to the DBM through specific interactions such as those between a receptor and its ligand or between an antibody and its antigen_ In other embodiments, the bioactive agent is attached to the DBM through non-specific interactions (e_g., hydrophobic interactions).

[0182] Medically/surgically useful substances include physiologically or pharmacologically active substance,s that act locally or systemically in the host.
Generally, these substances may include bioactive substances which can be readily incorporated into the osteoimplant and include, e.g., demineralized bone powder as described in U.S. Pat. No. 5,073,373;
collagen, insoluble collagen derivatives, etc., and soluble solids and/or liquids dissolved therein; antiviricides, particularly those effective against HIV and hepatitis; antimicrobials and/or antibiotics such as erythromycin, bacitracinõ neomycin, penicillin, polyinycin B, tetracyclines, biomycin, chloromycetin, and streptomycins, cefazolin, ampicillin, 07aetam, tobramycin, clindamycin and gentamycin, etc.; biocidaUbiostatic sugars such as dextren, glucose, etc.; amino acids; peptides; vitamins; inorganic elements;
co-factors for protein synthesis; hormones; endocrine tissue or tissue fragments;

synthesizers; enzymes such as alkaline phosphatase, coliagenase, peptidases, oxidases, etc.; polymer cell scaffolds with parenchymal cells; angiogenic agents and polymeric carriers containing sucb agents; collagen lattices; antigenic agents;
cytoskeletal agents; cartilage fragments; living cells such as chortdrocytes, bone marrow cells, mesenchyroal stem cells; natural extracts; genetically engineered living cells or otherwise modified living cells; expanded or cultured cells;
DNA
delivered by plasn-rid, viral vectors or other means; tissue transplants;
clemineralized bone powder; autogenous tissues such as blood, serum, soft tissue, bone marrow, etc.; bioadhesives; bone morphogenic proteins (BMPs);
osteoinductive factor (IF0); fibronectin (FN); endothelial cell growth factor (ECGF); vascular endothelial growth factor (VEGF); ceutentum attachment extracts (CAE); ketanserin; human growth hormone (HGH); animal growth hormones; epidermal growth factor (EGF); intcrleukins, e.g., interleukin-1 (IL-1), interleukin-2 (IL-2); human alpha thrombin; transforming growth factor (TGF-beta); insulin-like growth factors (IGF-1, IGF-2); platelet derived growth factors (PDGF); fibroblast growth factors (FGF, BFGF, etc.); periodontal ligament chemotactic factor (PDLGF); enamel matrix proteins; growth and differentiation factors (GDF); hedgehog family of proteins; protein receptor molecules; small peptides derived from growth factors above; bone promoters; cytokines;
somatotropin; bone digesters; antitumor agents; cellular attractants and attachment agents; immuno-suppressants; permeation enhancers, e.g., fatty acid esters such as laureate, myristate and stearate monoesters of polyethylene glycol, enamine derivatives, alpha-keto aldehydes, etc.; and nucleic acids. The amounts of such optionally added substances can vary widely with optimum levels being readily determined in a specific case by routine experimentation.
[0183] In certain embodiments, the agent to be delivered is adsorbed to or otherwise associated with the osteoimplant. The agent may be associated with the osteoimplant through specific or non-specific interactions; or covalent or non-covalent interactions. Examples of specific interactions include those between a ligand and a receptor, a epitope and an antibody, etc. Examples of non-specific interactions include hydrophobic interactions, electrostatic interactions, magnetic interactions, dipole interactions, van der Waals interactions, hydrogen bonding, etc. In certain embodiments, the agent is attached to the osteoimplant using a linker so that the agent is free to associate with its receptor or site of action. in vivo. Iri certain embodiments, the agent to be delivered may be attached to a chemical compound such as a peptide that is recognized by the matrix of the DBM composition. In another embodiment, the agent to be delivered is attached to an antibody, or fragment thereof, that recognizes an epitope found within the matrix of the DBM composition. In a further embodiment, the agent is a BMP, TGF-0, IGF, parathyroid hormone (PTH), growth factors, or angiogenic factors.
In certain embodiments at least two bioactive agents are attached to the DBM
composition. In other embodiments at least three bioactive agents are attached to the DBM composition.
[0184] Osteoinducing Agents [0185] Other osteoinducing agents besides the extracted osteoinductive factors may be added to the carrier. These agents may be added in an activated or non-activated form. These agents may be added at anytime during the preparation of the inventive material. For example, in the case of a DBM carrier, the osteoinducing agent may be added after the demineralization step and prior to the addition of the stabilizing agents so that the added osteoinducing agent is protected from. exogenous degrading enzymes once implanted. In some embodiments the DBM is lyophilized in a solution containing the osteoinducing agent. In certain other embodiments, the osteoinducing agents are adhered onto the hydrated deminere1i7ed bone matrix and are not freely soluble. In other instances, the osteoinducing agent is added after addition of a stabilizing agent so that the osteoinducing agent is available immediately upon implantation of the DBM, [0188] Osteoinducing agents include any agent that leads to or enhances the formation of bone. The osteoinducing agent may do this in any manner, for example, the agent may lead to the recruitment of cells responsible for bone formation, the agent may lead to the secretion of matrix which may subsequently undergo mineralization, the agent may lead to the decreaSed resorption of bone, etc. Suitable osteoinducing agents include bone morphogenic proteins (BMPs), transforming growth factor (TGF-0), insulin-like growth factor (IGF-l), parathyroid hornaone (PTH), and angiogenic factors such as 'VEGF. In one embodiment, the inducing agent is genetically engineered to eonaprise ari amino acid sequence which promotes the binding of the inducing agent to the DBM or the carrier. Sebald et al., PCT/EP00/00637 describe the production of exemplary en ineered growth factors suitable for use with DBM.
[0187] VIII. TREATMENT OF COMPOSITIONS
[0188] In the process of preparing improved inventive bone and cartilage matrix materials, the materials may be produced entirely aseptically or be sterilized to eliminate any infectious agents such as HIV, hepatitis B, or hepatitis C. The sterilization may be accomplished using antibiotics, irradiation, chemical sterilization (e.g., ethylene oxide), or thermal sterilization. Other methods known in the art of preparing bone and cartilage matrices, such as defatting, sonication, and lyophilization may also be used in preparing the carrier. Since the biological activity of various materials including demincralized bone is lmown to be detrimentally affected by most terminal sterilization processes, care must be taken when sterilizing the inventive compositions. In some embodiments, the osteoimplants described herein will be prepared. aseptically or sterilized.
[0189] Ix. EXAMPLE COMPOSITIONS
[0190] In one embodiment, the osteoimplant comprises a combination of DBM and osteoinductive factors from bone that has 2 to150 fold greater activity than DBM that has not been supplemented, as measured by the ability of the composition to induce heterotopic bone formation in a rat or mouse. Various ratios of DBM to osteoinductive factors may be used, ranging from 1 gram DBM:1 ug osteoinductive factors to 1 gram DBM:100 mg osteoinductive factors.
The osteoimplant may comprise osteoinductive growth factors extracted from DBM or recovered from acid baths used for demineralization of bone matrix and demineralized bone matrix and may exhibit the ability to induce specific alkaline phosphatase activity higher than. those of standard demineralized bone matrix preparations, for example, 2 to 100,000,000 higher than standard dermineralized bone matrix preparations in cultured C2C12.
[0191] In another embodiment, the osteoimplant comprises a combination of DBM, osteoinductive factors, and a carrier in various ratios. Any suitable carrier may be used, including polyethylene glycol, lecithin, starch, collagen, ' hydroxyapatite, or hyalurounic acid. Suitable ratios include 1 gram DBM:10 ug to 100 mg osteoinductive factors:100 mg to 10 grams carrier.
[0192] In a further embodiment, the osteoimplant comprises a combination of nondemineralized bone with osteoinductive factors from bone that has the ability to induce posterolateral spine fusion in higher animals, such as humans and dogs, without the addition of recombinant growth factors or iliac crest autograft. Various ratios of bone to osteoinductive factors ranging from gram bone:1 ug osteoinductive factors to 1 gram bone:100 mg osteoinductive factors can be used.
[0193] In yet a furtherembodiment, the osteoimplant comprises a combination of mineralized bone, osteoinductive factors, and a carrier in various ratios. Any suitable carrier may be used, as set forth above. Suitable ratios include 1 gram DBM:10 ug to 100 mg osteoinductive factors:100 mg to 10 grams carrier, with the ability to induce posterolateral spine fusion in humans or dogs without the addition of recombinant growth factors or iliac crest autograft.
[0194] As previously described, in another embodiment, aia osteoinductive material comprising a mineralized particulated naaterial, osteoinductive growth factors, and a demineraliz,ed bone matrix scaffold is provided.
[01951 In a further previously described embodiment, an osteoinductive composition comprising noncollagenous proteins extracted from demineralized bone or recovered from acid used to demineralize bone and mineral recovered acid used to demineralize bone. Alternatively, the osteoinductive eomposition may comprise noncollagenous prOtthIS extracted from demineralized bone or recovered from acid used to clemineralize bone and a demineralized bone matrix scaffold.
[0190] In yet a further previously described emobidment, an osteoinductive composition comprising mineralized or surface demineralized bone particles and extracts of DBM may be provided.
[0197] X. A.SSESSIVIENT OF OSTEOGENIC ACTIVITY
[0198] Induction of bone formation Gall be determined by a histological evaluation showing the de novo formation of bone with accompanying osteoblasts, osteoclasts, and osteoid matrix. For example, osteoinductive activity of an osteoinductive factor can be demonstrated by a test using a substrate onto which material to be tested is deposited. The substrate with deposited material is implanted subcutaneously in a test animal. The implant is subsequently removed and examined microscopically for the presence of bone formation including the presence of osteoblasts, osteoclacts, and osteoid matrix. A suitable procedure for assessing osteoinductive activity is illustrated in Example 5 of U.S. Patent No.
5,290,763. Although there is no generally accepted scale of evaluating the degree of osteogenic activity, certain factors are widely recognized as indicating bone formation. Such factors are referenced in the scale of 0-8 which is provided in Table 3 of exaLuple 1 of U.S. Patent No.
5,563,124. The 0 ______ portion of this scale corresponds to the scoring system described in U.S. Patent No. 5,290,763, which is limited to scores of 0-4. The remaining portion of the scale, scores 5-8, references additional levels of maturation. of bone formation. The expanded scale also includes consideration of resorption of collagen, a factor which is not described in the '763 patent.
[01991 In studies, a typical amount of DBM for implantation is 20 mg in a mouse and 40 mg in a rat. Significant increases in the growth factor dose, for example, 150x dose (or 150 times the growth factor found in normal DBM), lead to significantly more and potentially faster bone growth with larger volume bone growth, more dense bone growth, larger nodules of bone growth, higher x-ray density, and, generally, a higher osteoinductive score. Associated with this increase in osteoinductivity can be a cortical shell surrounding the nodule and some level of vascularizatiort in the nodule. However, the ability to quantitatively measure is generally limited by the method used, and generally measured increases in osteoinductive activity are not linear with the increase in.
dosage.
Thus, if 20 mg of DBM gives an osteoinductive activity of 1, 100 times the growth factor dose (2000 mg of DBM growth factors) does not give an osteoinductive activity of 100. Instead, it may result in an osteoinductive activity of about 20. A limitation of measurement using osteoinductive scores is that, in some situations, the system's ability to respond may be saturated. Thus, for example, if the score ranges only from 1 to 4, two samples may have the same score (4) but may not, in fact, be comparable. This is particularly the Case when the bone resulting from one method or implant is qualitatively better than the bone resulting from another method or implant. That is, both methods or implants may result in an osteoinductive score of 4 but one may result in qualitatively better bone than the other. Thus, in some situations it may be desirable to test speed of growth, density, presence of cortical bone, shelling, and/or other factors showing an. increase over normal demineralized bone matrix. Further, in addition to, or in lieu of, testing at 28 days, it may be desirable to test inductivity at 21 days Generally, inductivity may be measured histornorphometrically by methods known in art.

[0200] Further, delivering 100 times the growth factor dose may be challenging. In filling a bone defect, only as much filler may be used as there is bone void space.
[0201] XI. USES
[0202] Therapeutic Uses [0203] The osteogenic osteoimplant is intended to be applied at a bone repair site, for example, a site resulting from injury, defect brought about during the course of surgery, infection, malignancy, or developmental malformation.
The osteoimplant can be utilized in a wide variety of orthopedic, periodontal, neurosurgical, and oral and maxillofacial surgical procedures.
[0204] At the time just prior to when the osteoimplant of the invention is to be placed in. a defect site, optional materials, e.g., autograft bone marrow aspirate, autograft bone, preparations of selected autograft cells, autograft cells containing genes encoding bone promoting action, etc., can be combined with the osteoimplant. The osteoimplant can be implanted at the bone repair site, if desired, using any suitable affixation means, e.g., sutures, staples, bioadhesives, screws, pins, rivets, other fasteners and the like or it may be retained in place by the closing of the soft tissues around it.
[0205] The osteoinductive compositions may also be used. as drug delivery devices. In certain embodiments, association with the osteoinductive compositions increases the half-life of the relevant biologically active agent(s).
Particularly preferred inventive drug delivery devices are used to deliver osteoinductive growth factors. Other preferred agents to be delivered include factors or agents that promote wound healing. However, the osteoinductive compositions may alternatively or additionally be used to deliver other pharmaceutical agents including antibiotics, anti-neoplastic agents, growth factors, hernatopoietic factors, nutrients, an other bioactive agents described above. The amount of the bioactive agent included with the DBM composition can vary widely and will depend on such factors as the agent being delivered, the site of administration, and the patient's physiological condition. The optimum levels is determined in a specific case based upon the intended use of the implant.

102061 Non-Therapeutie Uses [02071 In addition to therapeutic uses involving implantation into a subject, the osteoinductive compositions have a number of other uses. For example, they can be used to generate cell lines, tissues, or organs having osteogenic or chondrogenic properties. In particular, cells can be removed from a donor and cultured in the presence of an osteoinductive composition_ The invention includes such cells as well as tissues and organs derived therefrom.
The cells, tissues, or organs may be implanted into the original donor after a period of culture in vitro or may be implanted into a different subject.
[02081 XII. CO1MCLUSION-[0209] In certain embodiments, the osteoinductive compositions and associated osteoimplants produce bone or cartilage in an animal model and/or in human patients with similar timing and at a level at least 10%, 20%, 35%, 50%, 100%, 200%, 300%, or 400% or greater osteogenic, osteoinductive or chondrogenic activity than a corollary carrier that has not been exposed to a treatment or condition as described herein. Of course, one slthiled in the art will appreciate that these values may vary slightly depending on the type of test used to measure the osteoinductivity or osteogenic or chondrogenic activity described above. The test results may fall within the range of 10% to 35%, 35% to 50%, 50% to 100%, 100% to 200%, and 200% to 400%. In certain embodiments, when an osteoinaplant is implanted into a bone defect site, the osteoimplant has an osteoinductivity score of at least 1, 2, 3, or 4 in an animal model and/or in humans.
[0210i Although the invention has been described with reference to preferred embodiments, persons skilled in the art will recognize that changes may be made in form and detail without departing from the scope of the invention.

Claims (73)

1. An osteoinductive composition comprising:
a carrier;
osteoinductive factors, the osteoinductive factors being supported by the carrier, the osteoinductive factors being recovered from acid used to demineralize mineralized bone;
wherein the osteoinductive composition exhibits increased osteoinductive activity when compared to a carrier not supporting osteoinductive factors.
2. The composition of claim 1, wherein the osteoinductive factors comprise noncollagenous proteins.
3. The composition of claim 1, further comprising a coprecipitate, wherein the coprecipitate and the osteoinductive factors form a homogenous mixture supported by the carrier.
4. The composition of claim 3, wherein the coprecipitate comprises collagen.
5. The composition of claim 3, wherein the coprecipitate comprises hydroxyapatite.
6. The composition of claim 3, wherein the coprecipitate comprises mineralized components of bone recovered during demineralization.
7. The composition of claim 3, wherein the coprecipitate is selected from the group consisting of a protein, a collagenous fragment, albumen and a protein with RGD
sequences.
8. The composition of claim 1, further comprising a bioactive agent, wherein the carrier acts as a delivery device to administer the bioactive agent.
9. The composition of claim 1, and further comprising a bioactive agent.
10. The composition of claim 9, wherein the bioactive agent is selected from the group consisting of osteogenic or chondrogenic proteins or peptides, anti-AIDS
substances, anti-cancer substances, antibiotics, immunosuppressants, anti-viral substances, enzyme inhibitors, hormones, neurotoxins, opioids, hypnotics, anti-histamines, lubricants, tranquilizers, anti-convulsants, muscle relaxants and anti-Parkinson substances, anti-spasmodics and muscle contractants, miotics and anti-cholinergics, anti-glaucoma compounds, anti-parasite compounds, anti-protozoal compounds, modulators of cell-extracellular matrix interactions, vasodilating agents, inhibitors of DNA, RNA
or protein synthesis, anti-hypertensives, analgesics, anti-pyretics, steroidal and non-steroidal anti-inflammatory agents, anti-angiogenic factors, angiogenic factors, anti-secretory factors, anticoagulant agents, antithrombotic agents, local anesthetics, ophthalmics, prostaglandins, anti-depressants, anti-psychotic substances, anti-emetics, and imaging agents.
11. The composition of claim 10, wherein the bioactive agent is a channel blocker.
12. The composition of claim 10, wherein the bioactive agent is a cell growth inhibitor or an antiadhesion molecule.
13. The composition of claim 1, wherein the carrier is demineralized bone.
14. The composition of claim 1, wherein the carrier is surface demineralized bone.
15. The composition of claim 1, wherein the carrier is mineralized bone.
16. The composition of claim 1, wherein the carrier is collagen.
17. The composition of claim 1, wherein the carrier is mineral recovered from bone.
18. The composition of claim 1, wherein the carrier is polyethylene glycol.
19. The composition of claim 1, wherein the carrier is selected from the group consisting of mineralized cancellous scaffolds; demineralized cancellous scaffolds;
particulate, demineralized, guanidine extracted, species-specific bone;
specially treated particulate, protein extracted, demineralized, xenogenic bone; synthetic hydroxyapatites;
polymers; hydrogels; starches; tricalcium phosphate, sintered hydroxyapatite, settable hydroxyapatite; polylactic acid; tyrosine polycarbonate; calcium sulfate;
collagen sheets;
settable calcium phosphate; polymeric cements; settable poly vinyl alcohols;
and polyurethanes.
20. The composition of claim 1, wherein the composition is settable.
21. The composition of claim 1, wherein the composition is injectable.
22. The composition of claim 1, wherein the carrier and osteoinductive factors comprise a delivery system.
23. The composition of claim 22, wherein the carrier comprises a plurality of sheets, each sheet being coated with osteoinductive factors, wherein the sheets are stacked, thereby forming a laminate.
24. The composition of claim 23, further comprising an adhesive layer between each sheet.
25. The composition of claim 22, wherein the carrier comprises a sheet, the sheet being coated with osteoinductive factors, wherein the sheet is rolled or folded such that a growth factor content in an interior of the sheet approximates a growth factor content at a surface of the sheet.
26. The composition of claim 1, wherein the osteoinductive factors comprise an homogenous mixture.
27. The composition of claim 26, wherein the homogenous mixture further comprises a bulking agent.
28. The composition of claim 1, wherein the carrier is demineralized bone matrix and the osteoinductive factors are osteoinductive growth factors recovered from acid baths used for demineralization of bone matrix, and wherein the growth factors and the demineralized bone matrix are combined such that the osteoinductive composition exhibits an ability to induce specific alkaline phosphatase activity levels 2 to 100,000,000 times higher than standard demineralized bone matrix preparations in cultured C2Cl2 cells.
29. The composition of claim 1, wherein the carrier comprises mineralized or surface demineralized bone particles and wherein the osteoinductive factors are adsorbed to surfaces of the mineralized or partially demineralized bone particles and wherein weakly bound extracts are eluted.
30. An osteoinductive material comprising:
a mineralized particulated material;
osteoinductive growth factors, the osteoinductive factors being recovered from acid used to demineralize mineralized bone; and a demineralized bone matrix scaffold;
wherein the osteoinductive growth factors are solubilized and combined with the mineralized particulated material to form a growth factor/mineral composite and wherein the growth factor/mineral composite is distributed in the demineralized bone matrix scaffold.
31. The osteoinductive material of claim 30, wherein the mineralized particulated material is mineral recovered from bone.
32. The osteoinductive material of claim 30, wherein the mineralized particulated material is surface demineralized bone.
33. The osteoinductive material of claim 30, wherein the osteoinductive growth factors are recovered from demineralization acid bath.
34. An osteoinductive composition with reduced immunogenicity comprising:
noncollagenous proteins recovered from acid used to demineralize bone; and a carrier;
wherein the osteoinductive composition exhibits an ability to induce formation of heterotopic bone in a euthymic mouse.
35. The osteoinductive composition of claim 34, wherein the carrier is mineral recovered from acid used to demineralize bone.
36. The osteoinductive composition of claim 34, wherein the carrier is a demineralized bone matrix scaffold.
37. An osteoinductive composition comprising:
a carrier; and a protein composition comprising a protein recovered from acid used to demineralize bone wherein the protein composition has less than 90% by weight inorganic components.
38. The osteoinductive composition of claim 37, wherein the carrier is demineralized bone.
39. The osteoinductive composition of claim 37, wherein the carrier is hydroxyapatite.
40. A method for producing an osteoinductive composition, the method comprising:
providing bone;
providing osteoinductive factors;

providing a carrier; and adding the osteoinductive factors to the carrier, wherein the providing osteoinductive factors step comprises extracting the osteoinductive factors from acid used to demineralize the bone.
41. The method of claim 40, wherein the extracting the osteoinductive factors step comprises using acid, removing the acid, and concentrating the osteoinductive factors.
42. The method of claim 40, wherein extracting the osteoinductive factors further comprises extracting a coprecipitate.
43. The method of claim 42, further comprising blending the osteoinductive factors and coprecipitate into a homogenous mixture.
44. The method of claim 43, wherein adding the osteoinductive factors to the carrier comprises blending the homogenous mixture with the carrier.
45. The method of claim 44, wherein the carrier is selected from the group consisting of cancellous bone chips, cortical chips, synthetic calcium phosphate, mineral recovered from acid bath during demineralization process and liquid settable polymers.
46. The method of claim 40, wherein providing osteoinductive factors comprises recovering the osteoinductive factors from a demineralization acid bath.
47. The method of claim 40, further comprising adding a bioactive agent to the composition.
48. The method of claim 47, wherein adding a bioactive agent to the composition comprises adding the bioactive agent to the carrier.
49. The method of claim 40, wherein the providing bone step comprises subjecting bone to an acid demineralization step followed by a defatting/disinfecting step.
50. The method of claim 40, further comprising combining the osteoinductive factors with a biologically compatible delivery vehicle prior to adding the osteoinductive factors to the carrier.
51. The method of claim 40, further comprising adding an additive to the composition wherein the additive is selected from the group consisting of radioopaque substances, angiogenesis promoting materials, cytokine inhibitors, bioactive agents, other medically/surgically useful substances, and other osteoinducing agents.
52. The method of claim 40, wherein the osteoinductive factors are supported by the carrier; and wherein the osteoinductive composition, comprising the carrier and the osteoinductive factors, exhibits increased osteoinductive activity when compared to a carrier not supporting osteoinductive factors.
53. The method of claim 52, wherein the osteoinductive factors comprise noncollagenous proteins.
54. The method of claim 52 or 53, further comprising providing a coprecipitate, wherein the coprecipitate and the osteoinductive factors form a homogenous mixture supported by the carrier.
55. The method of claim 54, wherein the coprecipitate comprises collagen, hydroxyapatite, or mineralized components of bone recovered during demineralization.
56. The method of claim 54, wherein the coprecipitate is selected from the group consisting of a protein, a collagenous fragment, albumen, and a protein with RGD sequences.
57. The method of claim 52, further comprising providing a bioactive agent, wherein the carrier acts as a delivery device to administer the bioactive agent.
58. The method of claim 52, and further comprising providing a bioactive agent.
59. The method of claim 57 or 58, wherein the bioactive agent is selected from the group consisting of osteogenic or chondrogenic proteins or peptides, anti-AIDS
substances, anti-cancer substances, antibiotics, immunosuppressants, antiviral substances, enzyme inhibitors, hormones, neurotoxins, opioids, hypnotics, anti-histamines, lubricants, tranquilizers, anti-convulsants, muscle relaxants and anti-Parkinson substances, anti-spasmodics and muscle contractants, miotics and anti-cholinergics, anti-glaucoma compounds, anti-parasite compounds, anti-protozoal compounds, modulators of cell-extracellular matrix interactions, vasodilating agents, inhibitors of DNA, RNA
or protein synthesis, anti-hypertensives, analgesics, anti-pyretics, steroidal and non-steroidal anti-inflammatory agents, anti-angiogenic factors, angiogenic factors, anti-secretory factors, anticoagulants agents, antithrombotic agents, local anesthetics, ophthalmics, prostaglandins, anti-depressants, anti-psychotic substances, anti-emetics, and imaging agents.
60. The method of claim 59, wherein the bioactive agent is a channel blocker.
61. The method of claim 59, wherein the bioactive agent is a cell growth inhibitor or an antiadhesion molecule.
62. The method of any one of claims 52 to 61, wherein the carrier is demineralized bone, surface demineralized bone, mineralized bone, collagen, mineral recovered from bone or polyethylene glycol.
63. The method of any one of claims 52 to 61, wherein the carrier is selected from the group consisting of mineralized cancellous scaffolds; demineralized cancellous scaffolds;
particulate, demineralized, guanidine extracted, species-specific bone;
specially treated particulate, protein extracted, demineralized, xenogenic bone; synthetic hydroxyapatites;
polymers; hydrogels; starches; tricalcium phosphate, sintered hydroxyapatite, settable hydroxyapatite; polylactic acid; tyrosine polycarbonate; calcium sulfate;
collagen sheets;
settable calcium phosphate; polymeric cements; settable poly vinyl alcohols;
and polyurethanes.
64. The method of any one of claims 52 to 63, wherein the composition is settable.
65. The method of any one of claims 52 to 64, wherein the composition is injectable.
66. The method of any one of claims 52 to 65, wherein the carrier and osteoinductive factors comprise a delivery system.
67. The method of claim 66, wherein the carrier comprises a plurality of sheets, each sheet being coated with osteoinductive factors, wherein the sheets are stacked, thereby forming a laminate.
68. The method of claim 67, further comprising providing an adhesive layer between each sheet.
69. The method of claim 66, wherein the carrier comprises a sheet, the sheet being coated with osteoinductive factors, wherein the sheet is rolled or folded such that a growth factor content in an interior of the sheet approximates a growth factor content at a surface of the sheet.
70. The method of any one of claims 52 to 69, wherein the osteoinductive factors comprise a homogenous mixture.
71. The method of claim 70, wherein the homogenous mixture further comprises a bulking agent.
72. The method of claim 52, wherein the carrier is demineralized bone matrix and the osteoinductive factors are osteoinductive growth factors recovered from acid baths used for demineralization of bone matrix, and wherein the growth factors and the demineralized bone matrix are combined such that the osteoinductive composition exhibits an ability to induce specific alkaline phosphatase activity levels 2 to 100,000,000 times higher than standard demineralized bone matrix preparations in cultured C2C12 cells.
73. The method of claim 52, wherein the carrier comprises mineralized or surface demineralized bone particles and wherein the osteoinductive factors are adsorbed to surfaces of the mineralized or partially demineralized bone particles and wherein weakly bound extracts are eluted.
CA2627907A 2005-11-01 2006-11-01 Osteoinductive composition comprising osteoinductive factors recovered from mineralized bone and a carrier Expired - Fee Related CA2627907C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US73267505P 2005-11-01 2005-11-01
US60/732,675 2005-11-01
PCT/US2006/060452 WO2007053850A2 (en) 2005-11-01 2006-11-01 Bone matrix compositions and methods

Publications (2)

Publication Number Publication Date
CA2627907A1 CA2627907A1 (en) 2007-05-10
CA2627907C true CA2627907C (en) 2014-06-17

Family

ID=37831682

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2627907A Expired - Fee Related CA2627907C (en) 2005-11-01 2006-11-01 Osteoinductive composition comprising osteoinductive factors recovered from mineralized bone and a carrier

Country Status (7)

Country Link
US (3) US8911759B2 (en)
EP (1) EP1942960B1 (en)
CN (1) CN101365499A (en)
AU (1) AU2006308534B2 (en)
CA (1) CA2627907C (en)
ES (1) ES2393756T3 (en)
WO (1) WO2007053850A2 (en)

Families Citing this family (108)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020114795A1 (en) 2000-12-22 2002-08-22 Thorne Kevin J. Composition and process for bone growth and repair
US8740987B2 (en) 2001-06-04 2014-06-03 Warsaw Orthopedic, Inc. Tissue-derived mesh for orthopedic regeneration
WO2003066000A2 (en) * 2002-02-08 2003-08-14 Wyeth Formulation comprising bioactive agents and method of using same
US7166133B2 (en) 2002-06-13 2007-01-23 Kensey Nash Corporation Devices and methods for treating defects in the tissue of a living being
US7241874B2 (en) 2002-06-26 2007-07-10 Zimmer Ortho Biologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
US7622562B2 (en) 2002-06-26 2009-11-24 Zimmer Orthobiologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
CA2535169A1 (en) 2003-12-31 2005-07-21 Osteotech, Inc. Improved bone matrix compositions and methods
US20070231788A1 (en) * 2003-12-31 2007-10-04 Keyvan Behnam Method for In Vitro Assay of Demineralized Bone Matrix
CN101365499A (en) 2005-11-01 2009-02-11 骨骼技术股份有限公司 Bone matrix compositions and methods
AU2007234612B2 (en) * 2006-12-14 2013-06-27 Johnson & Johnson Regenerative Therapeutics, Llc Protein stabilization formulations
DE102006060958A1 (en) 2006-12-20 2008-06-26 Jennissen, Herbert P., Prof. Dr. Process for the preparation of a polymer matrix, implants made thereof and their use
US7718616B2 (en) 2006-12-21 2010-05-18 Zimmer Orthobiologics, Inc. Bone growth particles and osteoinductive composition thereof
US20080206299A1 (en) * 2007-02-27 2008-08-28 Shimp Lawrence A Method for Recovering Minerals From Bone and Use of Same
WO2008157492A2 (en) 2007-06-15 2008-12-24 Osteotech, Inc. Osteoinductive demineralized cancellous bone
CA2690457C (en) 2007-06-15 2018-02-20 Osteotech, Inc. Bone matrix compositions and methods
US9554920B2 (en) * 2007-06-15 2017-01-31 Warsaw Orthopedic, Inc. Bone matrix compositions having nanoscale textured surfaces
US8642061B2 (en) * 2007-06-15 2014-02-04 Warsaw Orthopedic, Inc. Method of treating bone tissue
US7678764B2 (en) * 2007-06-29 2010-03-16 Johnson & Johnson Regenerative Therapeutics, Llc Protein formulations for use at elevated temperatures
US20110054408A1 (en) * 2007-07-10 2011-03-03 Guobao Wei Delivery systems, devices, tools, and methods of use
US9358113B2 (en) * 2007-07-10 2016-06-07 Warsaw Orthopedic, Inc. Delivery system
CA2695697A1 (en) * 2007-08-07 2009-02-12 Advanced Technologies And Regenerative Medicine, Llc Protein formulations comprising gdf-5 in aqueous acidic solution
ES2446544T3 (en) * 2007-10-19 2014-03-10 Warsaw Orthopedic, Inc. Demineralized bone matrix compositions and methods
DE102007051914A1 (en) * 2007-10-29 2009-05-07 Herbert Prof. Dr. Jennissen Process for the preparation of particles loaded with growth factors and the particles thus obtained
EP3366318B1 (en) 2008-03-28 2020-02-05 Warsaw Orthopedic, Inc. Delivery system attachment
AU2009236459B2 (en) * 2008-04-14 2013-07-25 Advanced Technologies And Regenerative Medicine, Llc Liquid buffered GDF-5 formulations
US9492375B2 (en) * 2008-07-23 2016-11-15 Warsaw Orthopedic, Inc. Foam carrier for bone grafting
KR101019741B1 (en) 2008-09-09 2011-03-08 주식회사 세라젬바이오시스 Method for preparation of composite materials for Bone Defect Filling and Bone Replacement
EP2358352B1 (en) * 2008-10-24 2018-08-29 Warsaw Orthopedic, Inc. Compositions and methods for promoting bone formation
US10660986B2 (en) * 2008-12-13 2020-05-26 Advanced Biologics, Llc Bioactive grafts and composites
US9101475B2 (en) 2009-02-12 2015-08-11 Warsaw Orthopedic, Inc. Segmented delivery system
WO2011017284A2 (en) 2009-08-03 2011-02-10 Osteotech, Inc. Bone matrix compositions and methods
US9649408B1 (en) * 2009-11-05 2017-05-16 Lifecell Corporation Systems and methods for sterilization of bone or bone components
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
EP2585083B1 (en) 2010-06-28 2015-09-09 BBS - Bioactive Bone Substitutes Oy A bone protein preparation in a polyethylene glycol/glycerol matrix
US8926710B2 (en) 2010-10-25 2015-01-06 Warsaw Orthopedic, Inc. Osteoinductive bone graft injectable cement
US9265830B2 (en) 2011-04-20 2016-02-23 Warsaw Orthopedic, Inc. Implantable compositions and methods for preparing the same
US20120294898A1 (en) * 2011-05-19 2012-11-22 Warsaw Orthopedic, Inc. Injectable dbm for soft tissue repair
EP2768542A4 (en) * 2011-10-21 2015-08-05 Univ Maryland Bone pastes comprising biofunctionalized calcium phosphate cements with enhanced cell functions for bone repair
US8992628B2 (en) 2012-01-20 2015-03-31 Warsaw Orthopedic, Inc. Bone delivery system
US9198758B2 (en) 2012-01-26 2015-12-01 Warsaw Orthopedic, Inc. Delivery systems
US9642935B1 (en) * 2012-01-27 2017-05-09 Robert L. Bundy Synthetic, composite osteogenic bone graft
US9775862B2 (en) 2012-01-30 2017-10-03 Warsaw Orthopedic, Inc. Modification of reactivity of bone constructs
US8926622B2 (en) 2012-04-03 2015-01-06 Warsaw Orthopedic, Inc. Bone delivery systems including holding and filling devices and methods
US9283089B2 (en) 2012-04-05 2016-03-15 Warsaw Orthopedic, Inc. Interbody bone implant device
US9730801B2 (en) 2012-04-17 2017-08-15 Warsaw Orthopedic, Inc. Interbody bone implant device
US9101606B2 (en) 2012-04-19 2015-08-11 Warsaw Orthopedic, Inc. Bone delivery system having a therapeutic agent
US9220608B2 (en) 2012-04-24 2015-12-29 Warsaw Orthopedic, Inc. Facet joint implant device
US8771368B2 (en) 2012-04-24 2014-07-08 William F. McKay Interspinous bone implant device
US9480567B2 (en) 2012-05-07 2016-11-01 Warsaw Orthopedic, Inc. Bone implants and methods comprising demineralized bone material
US10207027B2 (en) 2012-06-11 2019-02-19 Globus Medical, Inc. Bioactive bone graft substitutes
CN102698321A (en) * 2012-06-29 2012-10-03 上海大学 Preparation method of bone repairing body with drug loading function
US9655994B2 (en) 2012-07-25 2017-05-23 William F. McKay Delivery systems
US9585764B2 (en) 2012-07-26 2017-03-07 Warsaw Orthopedic, Inc. Bone implant device
US10172651B2 (en) 2012-10-25 2019-01-08 Warsaw Orthopedic, Inc. Cortical bone implant
BR112015010877A2 (en) * 2012-11-14 2017-07-11 Orthopaedic Innovation Center Inc antimicrobial articles produced by additive manufacturing
US9265609B2 (en) 2013-01-08 2016-02-23 Warsaw Orthopedic, Inc. Osteograft implant
US9034052B2 (en) 2013-01-14 2015-05-19 Warsaw Orthopedic, Inc. Delivery systems containing bioactive materials
AU2014225458A1 (en) 2013-03-07 2015-07-09 Allosource Consistent calcium content bone allograft systems and methods
US9283013B2 (en) 2013-03-14 2016-03-15 Warsaw Orthopedic, Inc. Filling systems for bone delivery devices
CA2917093A1 (en) * 2013-07-01 2015-01-08 Amit Prakash Govil Soft tissue implant
US9861724B2 (en) * 2013-08-06 2018-01-09 Fusion Innovations, Llc Method for fusion of fractured or voided sternum post sternotomy
US20150050358A1 (en) * 2013-08-15 2015-02-19 Regents Of The University Of Minnesota Implant support composition and methods of use
US9539286B2 (en) 2013-10-18 2017-01-10 Globus Medical, Inc. Bone grafts including osteogenic stem cells, and methods relating to the same
US9486483B2 (en) 2013-10-18 2016-11-08 Globus Medical, Inc. Bone grafts including osteogenic stem cells, and methods relating to the same
US9757330B2 (en) 2013-10-18 2017-09-12 Industrial Technology Research Institute Recipe for in-situ gel, and implant, drug delivery system formed thereby
CN103800945B (en) * 2014-01-17 2015-05-20 北京大清生物技术有限公司 Moldable bone repairing material for bone repairing and preparation method thereof
US9579421B2 (en) * 2014-02-07 2017-02-28 Globus Medical Inc. Bone grafts and methods of making and using bone grafts
US9463264B2 (en) 2014-02-11 2016-10-11 Globus Medical, Inc. Bone grafts and methods of making and using bone grafts
US20150258245A1 (en) * 2014-03-13 2015-09-17 Warsaw Orthopedic. Inc. Method of delipidation and/or terminal sterilization for bone material
US9364583B2 (en) 2014-04-25 2016-06-14 Warsaw Orthopedic, Inc. Osteoinductive demineralized bone implant
FR3020273B1 (en) * 2014-04-25 2021-04-16 Biobank BONE PASTE MANUFACTURING PROCESS
CN103990180A (en) * 2014-05-26 2014-08-20 中国人民解放军第四军医大学 Preparation method and application of deproteinated decalcification bone matrix implantable microcarrier
US9610143B2 (en) * 2014-06-19 2017-04-04 Osteolife Biomedical, Llc Compressed decalcified trabecular bone grafts and tooth socket repair
US9238090B1 (en) 2014-12-24 2016-01-19 Fettech, Llc Tissue-based compositions
DE102015100806A1 (en) * 2015-01-20 2016-07-21 Antonis Alexakis Biocompatible molding
US10006705B2 (en) 2015-02-09 2018-06-26 Warsaw Orthopedic, Inc. Methods for treating tissue materials
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US11426489B2 (en) 2015-06-10 2022-08-30 Globus Medical, Inc. Biomaterial compositions, implants, and methods of making the same
US10016529B2 (en) 2015-06-10 2018-07-10 Globus Medical, Inc. Biomaterial compositions, implants, and methods of making the same
US10549011B2 (en) 2015-10-26 2020-02-04 Osteolife Biomedical, Llc Bone putty and gel systems and methods
US20170128633A1 (en) 2015-11-10 2017-05-11 Theodore Malinin Bioactive Implants and Methods of Making and Using
CN105837682A (en) * 2015-11-25 2016-08-10 天津中津生物发展有限公司 BMP-3 protein, and preparation method and application thereof as bone repair injection
US11090411B2 (en) 2016-01-28 2021-08-17 Warsaw Orthopedic, Inc. Electron beam irradiated osteoinductive bone implant
US10383731B2 (en) 2016-02-24 2019-08-20 Warsaw Orthopedic, Inc. Spinal implant system and method
CN105944148A (en) * 2016-04-29 2016-09-21 苏州蔻美新材料有限公司 Preparation method of bone filling material
CN105999412A (en) * 2016-06-13 2016-10-12 深圳市光明创博生物制品发展有限公司 Method for preparing bone induced osteogenesis preparation
CN106075581A (en) * 2016-06-13 2016-11-09 深圳市光明创博生物制品发展有限公司 A kind of method preparing self-bone grafting osteogenic formulation
US10813763B2 (en) * 2016-07-26 2020-10-27 Warsaw Orthopedic, Inc. Implantable mesh
CN106492281B (en) * 2016-11-17 2022-02-08 温州医科大学 Biocompatible bone graft and preparation method thereof
CN106691525A (en) * 2017-01-06 2017-05-24 南京市六合区人民医院 Fixing system for transplant in anterior cruciate ligament reconstruction
RU2665962C1 (en) * 2017-03-17 2018-09-05 Общество с ограниченной ответственностью "Матрифлекс" Bioresorable biological matrix for substitution of bone tissue defects and method of its obtaining
CN109663149A (en) * 2017-10-17 2019-04-23 天津大学 The composite material and preparation method of non-viral vector delivery MicroRNA and bone meal
CN107823711B (en) * 2017-11-09 2020-10-30 华中科技大学同济医学院附属协和医院 Preparation of core-shell structure composite material and method for constructing tissue engineering micro-tissue by using same
RU2693606C1 (en) * 2018-06-05 2019-07-03 Общество с ограниченной ответственностью "Матрифлекс" Method of producing and using a highly purified mineral matrix in the form of segments and granules with osteoinductive properties for bone defect replacement
CN109529106B (en) * 2018-11-14 2021-09-10 山东隽秀生物科技股份有限公司 Preparation method of chronic wound repair matrix
RU2704114C1 (en) * 2019-04-24 2019-10-24 Общество с ограниченной ответственностью "ЛИОСЕЛЛ" Method of producing a mineral-organic component of bone tissue
US11583402B2 (en) 2019-07-16 2023-02-21 William Baumgartl Method for treating joint pain
US20210022872A1 (en) 2019-07-25 2021-01-28 D2 Medical Llc Bone-derived thermoplastic filament and method of manufacture
CN111012947B (en) * 2019-12-30 2021-11-12 南京财经大学 Injectable and self-healing starch-based hydrogel and preparation method and application thereof
US11896736B2 (en) 2020-07-13 2024-02-13 Globus Medical, Inc Biomaterial implants and methods of making the same
CN111848741B (en) * 2020-07-22 2021-06-15 中国人民解放军陆军军医大学第一附属医院 Peptide, peptide-modified DBM (DBM) scaffold, and preparation method and application thereof
CN111905146B (en) * 2020-08-01 2021-11-16 北京欧亚铂瑞科技有限公司 Acellular bone matrix hydrogel retaining natural hydroxyapatite and preparation method thereof
CN112089891A (en) * 2020-09-22 2020-12-18 天津中津生物发展有限公司 Bone induction active extract and preparation method thereof
CN112023112A (en) * 2020-09-22 2020-12-04 天津中津生物发展有限公司 Bone hemostatic material with osteogenesis inducing activity and preparation method thereof
CN113209368A (en) * 2021-04-26 2021-08-06 深圳市迈捷生命科学有限公司 Animal-derived mineralized bone for treating bone diseases and cosmetic plastic and preparation method thereof
CA3177746A1 (en) * 2021-09-29 2023-03-29 Musculoskeletal Transplant Foundation Modified grafts
CN114259604B (en) * 2021-12-17 2022-12-27 上海纳米技术及应用国家工程研究中心有限公司 Preparation method of 3D printing ordered vascularization promoting drug-loaded bone repair scaffold, product and application thereof

Family Cites Families (198)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2253086C3 (en) 1972-10-25 1979-02-15 Martin Prof. Dr. 1000 Berlin Wenzel Preserving solutions for infusion and for storage of tissues and tissue parts
US4172128A (en) 1975-03-26 1979-10-23 Erhard Thiele Process of degrading and regenerating bone and tooth material and products
US4294753A (en) * 1980-08-04 1981-10-13 The Regents Of The University Of California Bone morphogenetic protein process
US4761471A (en) 1980-08-04 1988-08-02 The Regents Of The University Of California Bone morphogenetic protein composition
US4619989A (en) 1981-05-05 1986-10-28 The Regents Of The University Of Cal. Bone morphogenetic protein composition
US4789732A (en) 1980-08-04 1988-12-06 Regents Of The University Of California Bone morphogenetic protein composition
US4455256A (en) * 1981-05-05 1984-06-19 The Regents Of The University Of California Bone morphogenetic protein
US4430760A (en) * 1981-12-18 1984-02-14 Collagen Corporation Nonstress-bearing implantable bone prosthesis
US4472840A (en) 1981-09-21 1984-09-25 Jefferies Steven R Method of inducing osseous formation by implanting bone graft material
US4394370A (en) * 1981-09-21 1983-07-19 Jefferies Steven R Bone graft material for osseous defects and method of making same
US4440750A (en) * 1982-02-12 1984-04-03 Collagen Corporation Osteogenic composition and method
US4485097A (en) 1982-05-26 1984-11-27 Massachusetts Institute Of Technology Bone-equivalent and method for preparation thereof
US4440370A (en) * 1982-09-28 1984-04-03 Rood Robert M Securing stake
US4795804A (en) * 1983-08-16 1989-01-03 The Regents Of The University Of California Bone morphogenetic agents
NZ210699A (en) 1984-01-04 1989-06-28 Int Genetic Eng Isolation of an osteogenic protein of the p3 immunologically related family
US4804744A (en) 1984-01-04 1989-02-14 International Genetic Engineering, Inc. Osteogenic factors
US4563489A (en) * 1984-02-10 1986-01-07 University Of California Biodegradable organic polymer delivery system for bone morphogenetic protein
US4789663A (en) 1984-07-06 1988-12-06 Collagen Corporation Methods of bone repair using collagen
DE3588058T3 (en) 1984-07-16 2005-04-07 Celtrix Pharmaceuticals, Inc., Palo Alto Cartilage-inducing polypeptide factors from bone
US4843063A (en) * 1984-07-16 1989-06-27 Collagen Corporation Polypeptide cartilage-inducing factors found in bone
US4657548A (en) * 1984-09-11 1987-04-14 Helitrex, Inc. Delivery system for implantation of fine particles in surgical procedures
US4888366A (en) 1984-10-24 1989-12-19 Collagen Corporation Inductive collagen-based bone repair preparations
US4563350A (en) 1984-10-24 1986-01-07 Collagen Corporation Inductive collagen based bone repair preparations
US4678470A (en) 1985-05-29 1987-07-07 American Hospital Supply Corporation Bone-grafting material
US4645503A (en) * 1985-08-27 1987-02-24 Orthomatrix Inc. Moldable bone-implant material
US4755184A (en) * 1986-01-09 1988-07-05 Mark Silverberg Bone augmentation implant
US6311690B1 (en) 1986-03-27 2001-11-06 Gensci Orthobiologics, Inc. Bone repair material and delayed drug delivery system
US5106748A (en) * 1986-07-01 1992-04-21 Genetics Institute, Inc. Dna sequences encoding 5 proteins
IL83003A (en) 1986-07-01 1995-07-31 Genetics Inst Osteoinductive factors
US5108922A (en) * 1986-07-01 1992-04-28 Genetics Institute, Inc. DNA sequences encoding BMP-1 products
FI81010C (en) 1986-09-05 1990-09-10 Biocon Oy Benomplaceringsimplants
US4902296A (en) * 1986-10-29 1990-02-20 The University Of Virginia Alumni Patents Foundation Use of demineralized bone matrix in the repair of segmental defects
US4743259A (en) * 1986-10-29 1988-05-10 The University Of Virginia Alumni Patents Foundation Use of demineralized bone matrix in the repair of segmental defects
US5041138A (en) 1986-11-20 1991-08-20 Massachusetts Institute Of Technology Neomorphogenesis of cartilage in vivo from cell culture
US4787906A (en) 1987-03-02 1988-11-29 Haris Andras G Controlled tissue growth and graft containment
FR2616318A1 (en) 1987-06-15 1988-12-16 Centre Nat Rech Scient ARTIFICIAL SKIN AND PROCESS FOR PREPARING THE SAME
US5320970A (en) 1987-11-06 1994-06-14 Washington Research Foundation Detection of collagen degradation in vivo
JPH01179689A (en) 1988-01-08 1989-07-17 Res Dev Corp Of Japan Stabilizer for organism catalyst
IL85312A (en) 1988-02-03 1991-08-16 Israel State Injectable pharmaceutical compositions having improved stability
US5266683A (en) * 1988-04-08 1993-11-30 Stryker Corporation Osteogenic proteins
US5354557A (en) 1988-04-08 1994-10-11 Stryker Corporation Osteogenic devices
US4880610A (en) 1988-04-20 1989-11-14 Norian Corporation In situ calcium phosphate minerals--method and composition
US5106626A (en) 1988-10-11 1992-04-21 International Genetic Engineering, Inc. Osteogenic factors
US5073373A (en) 1989-09-21 1991-12-17 Osteotech, Inc. Flowable demineralized bone powder composition and its use in bone repair
US5290558A (en) 1989-09-21 1994-03-01 Osteotech, Inc. Flowable demineralized bone powder composition and its use in bone repair
US5236456A (en) * 1989-11-09 1993-08-17 Osteotech, Inc. Osteogenic composition and implant containing same
US5204055A (en) 1989-12-08 1993-04-20 Massachusetts Institute Of Technology Three-dimensional printing techniques
US5139520A (en) 1990-01-31 1992-08-18 American Cyanamid Company Method for acl reconstruction
US5378469A (en) 1990-04-06 1995-01-03 Organogenesis, Inc. Collagen threads
US6197325B1 (en) 1990-11-27 2001-03-06 The American National Red Cross Supplemented and unsupplemented tissue sealants, methods of their production and use
US5563124A (en) 1991-04-22 1996-10-08 Intermedics Orthopedics/ Denver, Inc. Osteogenic product and process
US5290763A (en) * 1991-04-22 1994-03-01 Intermedics Orthopedics/Denver, Inc. Osteoinductive protein mixtures and purification processes
US6503277B2 (en) * 1991-08-12 2003-01-07 Peter M. Bonutti Method of transplanting human body tissue
US5270300A (en) 1991-09-06 1993-12-14 Robert Francis Shaw Methods and compositions for the treatment and repair of defects or lesions in cartilage or bone
US5211664A (en) 1992-01-14 1993-05-18 Forschungsinstitut, Davos Laboratorium Fur Experimentelle Chirugie Shell structure for bone replacement
US5314476A (en) 1992-02-04 1994-05-24 Osteotech, Inc. Demineralized bone particles and flowable osteogenic composition containing same
US7060287B1 (en) * 1992-02-11 2006-06-13 Bioform Inc. Tissue augmentation material and method
DE69329108T2 (en) * 1992-03-02 2001-03-22 Bioeng Inc METHOD FOR INACTIVATING VIRUSES
CA2093836A1 (en) 1992-04-24 1993-10-25 Wayne Gombotz Biodegradable tgf-.beta. delivery system for bone regeneration
FR2699408B1 (en) 1992-12-21 1995-03-24 Bioland Method for treating bone tissue and corresponding implantable biomaterials.
FR2702660B1 (en) 1993-03-17 1995-05-24 Karl Simpson Stabilized therapeutic compositions and process for their preparation.
EP0621020A1 (en) 1993-04-21 1994-10-26 SULZER Medizinaltechnik AG Intervertebral prosthesis and method of implanting such a prosthesis
US5385887A (en) * 1993-09-10 1995-01-31 Genetics Institute, Inc. Formulations for delivery of osteogenic proteins
US5490962A (en) 1993-10-18 1996-02-13 Massachusetts Institute Of Technology Preparation of medical devices by solid free-form fabrication methods
US6610656B1 (en) * 1993-12-30 2003-08-26 President And Fellows Of Harvard College Method of promoting chondrocyte differentiation with hedgehog related polypeptides
US5518680A (en) * 1993-10-18 1996-05-21 Massachusetts Institute Of Technology Tissue regeneration matrices by solid free form fabrication techniques
ATE319823T1 (en) 1993-12-07 2006-03-15 Inst Genetics Llc BMP-12, BMP-13 AND TENDON-INDUCING COMPOSITIONS CONTAINING SAME
US5723012A (en) * 1993-12-09 1998-03-03 Bioland Uses for a current of supercritical carbon dioxide as an antiviral agent
US5507813A (en) * 1993-12-09 1996-04-16 Osteotech, Inc. Shaped materials derived from elongate bone particles
US20030032963A1 (en) 2001-10-24 2003-02-13 Kyphon Inc. Devices and methods using an expandable body with internal restraint for compressing cancellous bone
US5571189A (en) 1994-05-20 1996-11-05 Kuslich; Stephen D. Expandable fabric implant for stabilizing the spinal motion segment
SE9402528D0 (en) * 1994-07-19 1994-07-19 Astra Ab Hard tissue stimulant with electricity
US5531735A (en) 1994-09-27 1996-07-02 Hercules Incorporated Medical devices containing triggerable disintegration agents
US6180606B1 (en) 1994-09-28 2001-01-30 Gensci Orthobiologics, Inc. Compositions with enhanced osteogenic potential, methods for making the same and uses thereof
TW369414B (en) 1994-09-30 1999-09-11 Yamanouchi Pharma Co Ltd Bone formation transplant
US5501706A (en) 1994-11-29 1996-03-26 Wildflower Communications, Inc. Medical implant structure and method for using the same
US6958150B2 (en) 1994-12-15 2005-10-25 Advance Biofactures Of Curacao, N.V. Reduction of adipose tissue
US6599515B1 (en) 1995-01-16 2003-07-29 Baxter International Inc. Fibrin porous structure
US5788959A (en) 1995-04-24 1998-08-04 University Of Maryland, Baltimore County Drug delivery device and method for employing the same
US5902785A (en) 1995-06-06 1999-05-11 Genetics Institute, Inc. Cartilage induction by bone morphogenetic proteins
CA2224253A1 (en) 1995-06-09 1996-12-27 Martin J. Macphee Chitin hydrogels, methods of their production and use
FI101933B1 (en) 1995-06-13 1998-09-30 Biocon Oy A joint prosthesis
JP3881707B2 (en) 1995-07-20 2007-02-14 学校法人松本歯科大学 Method for producing osteogenesis promoter and method for producing osteogenic composition using osteogenesis promoter
US5776193A (en) 1995-10-16 1998-07-07 Orquest, Inc. Bone grafting matrix
US5607269A (en) * 1995-11-21 1997-03-04 Osteotech, Inc. Bone milling apparatus
US5902562A (en) 1995-12-21 1999-05-11 Sandia Corporation Method for the preparation of high surface area high permeability carbons
JP3476631B2 (en) 1995-12-21 2003-12-10 株式会社アムニオテック Medical material composed of human-derived natural collagen membrane
EP0880345B1 (en) 1996-01-17 2006-04-05 Osteotech, Inc. Process for producing flexible sheets from demineralized, elongate, bone particles
FR2749756B1 (en) * 1996-06-14 1998-09-11 Bioland PROCESS FOR THE PREPARATION OF AN IMPLANTABLE COMPOSITE MATERIAL, MATERIAL OBTAINED, IMPLANT COMPRISING SUCH MATERIAL, AND IMPLEMENTATION KIT
FR2749864B1 (en) 1996-06-18 1998-09-11 Bioland METHODS OF MANUFACTURING AND TREATMENT OF A TEXTILE PART AND APPLICATIONS
US6953594B2 (en) 1996-10-10 2005-10-11 Etex Corporation Method of preparing a poorly crystalline calcium phosphate and methods of its use
US5846484A (en) 1997-03-20 1998-12-08 Osteotech, Inc. Pressure flow system and method for treating a fluid permeable workpiece such as a bone
PT981381E (en) 1997-05-12 2007-04-30 Metabolix Inc Polyhydroxyalkanoates for in vivo applications
US20010031254A1 (en) 1998-11-13 2001-10-18 Bianchi John R. Assembled implant
US6117646A (en) 1997-09-22 2000-09-12 Osteometer Biotech A/S Assaying protein fragments in body fluids
JP3483753B2 (en) 1997-12-29 2004-01-06 タキロン株式会社 Biodegradable absorbent plastic adhesive
US5899939A (en) 1998-01-21 1999-05-04 Osteotech, Inc. Bone-derived implant for load-supporting applications
US6123731A (en) * 1998-02-06 2000-09-26 Osteotech, Inc. Osteoimplant and method for its manufacture
US6326018B1 (en) 1998-02-27 2001-12-04 Musculoskeletal Transplant Foundation Flexible sheet of demineralized bone
US7019192B2 (en) 1998-02-27 2006-03-28 Musculoskeletal Transplant Foundation Composition for filling bone defects
USRE38522E1 (en) * 1998-02-27 2004-05-25 Musculoskeletal Transplant Foundation Malleable paste for filling bone defects
US6030635A (en) * 1998-02-27 2000-02-29 Musculoskeletal Transplant Foundation Malleable paste for filling bone defects
US7045141B2 (en) * 1998-02-27 2006-05-16 Musculoskeletal Transplant Foundation Allograft bone composition having a gelatin binder
US6911212B2 (en) 1998-02-27 2005-06-28 Musculoskeletal Transplant Foundation Malleable putty and flowable paste with allograft bone having residual calcium for filling bone defects
US7208011B2 (en) * 2001-08-20 2007-04-24 Conor Medsystems, Inc. Implantable medical device with drug filled holes
US6149864A (en) 1998-06-25 2000-11-21 Massachusetts Institute Of Technology Supercritical fluid sterilization method
SE515572C2 (en) 1998-09-09 2001-09-03 Lanka Ltd Implants, ways of making it and using it
JP4058180B2 (en) 1998-11-20 2008-03-05 キヤノン株式会社 Image forming apparatus and method
CA2355046A1 (en) 1998-12-14 2000-06-22 Osteotech, Inc. Bone graft and guided bone regeneration method
WO2000045870A1 (en) * 1999-02-04 2000-08-10 Sdgi Holdings, Inc. Osteogenic paste compositions and uses thereof
DE19904785A1 (en) 1999-02-05 2000-08-10 Ulrich Zimmermann Process for the production of stable alginate material
DE19906096A1 (en) 1999-02-13 2000-08-17 Walter Sebald Protein with a heparin-binding epitope
US6294187B1 (en) 1999-02-23 2001-09-25 Osteotech, Inc. Load-bearing osteoimplant, method for its manufacture and method of repairing bone using same
US6696073B2 (en) 1999-02-23 2004-02-24 Osteotech, Inc. Shaped load-bearing osteoimplant and methods of making same
US6143030A (en) 1999-03-26 2000-11-07 Bristol-Myers Squibb Co. Impaction allograft form and method of orthopaedic surgery using same
US6468543B1 (en) 1999-05-03 2002-10-22 Zymogenetics, Inc. Methods for promoting growth of bone using ZVEGF4
US6291041B1 (en) 1999-05-10 2001-09-18 Curwood, Inc. Heat resistant nylon multi-layer film
US6969404B2 (en) 1999-10-08 2005-11-29 Ferree Bret A Annulus fibrosis augmentation methods and apparatus
WO2001087369A2 (en) * 2000-05-12 2001-11-22 Osteotech, Inc. Osteoimplant and method for making same
AU782394B2 (en) 1999-06-29 2005-07-21 J. Alexander Marchosky Compositions and methods for forming and strengthening bone
US6618698B1 (en) 1999-08-12 2003-09-09 Quickturn Design Systems, Inc. Clustered processors in an emulation engine
US6352667B1 (en) 1999-08-24 2002-03-05 Absorbable Polymer Technologies, Inc. Method of making biodegradable polymeric implants
US6162258A (en) 1999-08-25 2000-12-19 Osteotech, Inc. Lyophilized monolithic bone implant and method for treating bone
US6783546B2 (en) 1999-09-13 2004-08-31 Keraplast Technologies, Ltd. Implantable prosthetic or tissue expanding device
US6648919B2 (en) 1999-10-14 2003-11-18 Bret A. Ferree Transplantation of engineered meniscus tissue to the intervertebral disc
ATE305276T1 (en) 1999-10-20 2005-10-15 Sdgi Holdings Inc IMMEDIATE ORTHOPEDIC BONE SUPPORT IMPLANT
US6740093B2 (en) 2000-02-28 2004-05-25 Stephen Hochschuler Method and apparatus for treating a vertebral body
DE60115215T2 (en) 2000-03-22 2006-08-03 Synthes (U.S.A.) GUN FOR FILLING BONE DEFECTS
US6884778B2 (en) 2000-04-14 2005-04-26 William Marsh Rice University Biocompatible macromers
US6682760B2 (en) 2000-04-18 2004-01-27 Colbar R&D Ltd. Cross-linked collagen matrices and methods for their preparation
US7025771B2 (en) * 2000-06-30 2006-04-11 Spineology, Inc. Tool to direct bone replacement material
DE60141653D1 (en) * 2000-07-21 2010-05-06 Spineology Group Llc A STRONG, POROUS NET BAG DEVICE AND ITS USE IN BONE SURGERY
DE10046119A1 (en) * 2000-09-15 2002-03-28 Inst Textil & Faserforschung Medical bioresorbable implant, method of manufacture and use
DE60121017T2 (en) * 2000-10-24 2007-01-11 SDGI Holdings, Inc., Wilmington DEVICE AND METHOD FOR BONE CONSTRUCTION FULL PLUG
EP1341484B1 (en) * 2000-12-08 2009-05-06 Osteotech, Inc. Implant for orthopedic applications
US6752831B2 (en) * 2000-12-08 2004-06-22 Osteotech, Inc. Biocompatible osteogenic band for repair of spinal disorders
US7323193B2 (en) 2001-12-14 2008-01-29 Osteotech, Inc. Method of making demineralized bone particles
US6712853B2 (en) * 2000-12-15 2004-03-30 Spineology, Inc. Annulus-reinforcing band
CA2365376C (en) * 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US20020082697A1 (en) * 2000-12-22 2002-06-27 Damien Christopher J. Implantable osteogenic material
FR2819715B1 (en) 2001-01-25 2004-01-02 Cousin Biotech BONE REPAIR DEVICE
US6855169B2 (en) 2001-02-28 2005-02-15 Synthes (Usa) Demineralized bone-derived implants
US6595998B2 (en) 2001-03-08 2003-07-22 Spinewave, Inc. Tissue distraction device
US7211271B2 (en) 2001-03-12 2007-05-01 The Regents Of The University Of California Method to improve hydroxyapatite implantation and stimulate bone regeneration
ES2305246T3 (en) * 2001-06-01 2008-11-01 Wyeth COMPOSITIONS FOR THE SYSTEMIC ADMINISTRATION OF SEQUENCES CODING OSEAS MORPHOGENETIC PROTEINS.
US8740987B2 (en) 2001-06-04 2014-06-03 Warsaw Orthopedic, Inc. Tissue-derived mesh for orthopedic regeneration
US20030008328A1 (en) 2001-07-03 2003-01-09 Wironen John F. Quantitative in vitro bone induction assay
US7132110B2 (en) * 2001-08-30 2006-11-07 Isotis Orthobiologics, Inc. Tissue repair compositions and methods for their manufacture and use
US7758895B2 (en) * 2001-09-04 2010-07-20 Perth Bone and Tissue Bank Methods for purifying insoluble bone gelatin
JP2003082535A (en) 2001-09-12 2003-03-19 Shigenori Kuga Minute fibrous carbon material derived from cellulose raw material and method for producing the same
KR20040047746A (en) 2001-10-12 2004-06-05 오스테오테크, 인코포레이티드 Improved bone graft
US7008591B2 (en) 2001-10-17 2006-03-07 Edwards Lifesciences Corporation Supercritical fluid extraction process for tissue preparation
US7629388B2 (en) 2001-11-20 2009-12-08 William Marsh Rice University Synthesis and characterization of biodegradable cationic poly(propylene fumarate-co-ethylene glycol) copolymer hydrogels modified with agmatine for enhanced cell adhesion
US6478825B1 (en) 2001-11-28 2002-11-12 Osteotech, Inc. Implant, method of making same and use of the implant for the treatment of bone defects
DE50200594D1 (en) 2002-02-08 2004-08-12 Storz Karl Gmbh & Co Kg Anchor element for anchoring a ligament graft
US20030194708A1 (en) 2002-04-10 2003-10-16 Minke Binnerts Human homolog of crossveinless materials and methods
WO2003099325A1 (en) * 2002-05-21 2003-12-04 Schering-Plough Ltd. Methods for the in vitro culture of sporozoea sp. and user thereof
US7241874B2 (en) 2002-06-26 2007-07-10 Zimmer Ortho Biologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
US7622562B2 (en) * 2002-06-26 2009-11-24 Zimmer Orthobiologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
US7744597B2 (en) * 2002-06-26 2010-06-29 Lifenet Health Device and process for producing fiber products and fiber products produced thereby
US7244445B2 (en) * 2002-07-29 2007-07-17 Poly Med, Inc Conformable, absorbable, solid composite preforms and their use for bone tissue engineering
SG145565A1 (en) * 2002-08-12 2008-09-29 Osteotech Inc Synthesis of a bone-polymer composite material
WO2004032988A2 (en) 2002-10-08 2004-04-22 Osteotech, Inc. Coupling agents for orthopedic biomaterials
EP1578957B1 (en) * 2002-12-12 2015-04-15 Warsaw Orthopedic, Inc. Formable and settable polymer bone composite and method of production thereof
US20050251267A1 (en) 2004-05-04 2005-11-10 John Winterbottom Cell permeable structural implant
JP2006516467A (en) 2003-02-04 2006-07-06 オステオテック,インコーポレイテッド Polyurethane for bone implants
US7985414B2 (en) 2003-02-04 2011-07-26 Warsaw Orthopedic, Inc. Polyurethanes for osteoimplants
AU2004212942A1 (en) 2003-02-14 2004-09-02 Depuy Spine, Inc. In-situ formed intervertebral fusion device
TW587933B (en) 2003-04-30 2004-05-21 A Spine Holding Group Corp Device for anchoring bone tissue
US7351262B2 (en) 2003-06-05 2008-04-01 Warsaw Orthopedic, Inc. Bone implants and methods of making same
US7108832B2 (en) 2003-06-23 2006-09-19 Novasterilis Inc. Sterialization methods and apparatus which employ additive-containing supercritical carbon dioxide sterilant
US20050020506A1 (en) * 2003-07-25 2005-01-27 Drapeau Susan J. Crosslinked compositions comprising collagen and demineralized bone matrix, methods of making and methods of use
US20050131417A1 (en) 2003-08-22 2005-06-16 Ahern James W. Kit for treating bony defects
EP1744793A2 (en) 2003-10-22 2007-01-24 University of Florida Biomimetic organic/inorganic composites, processes for their production, and methods of use
US20070231788A1 (en) 2003-12-31 2007-10-04 Keyvan Behnam Method for In Vitro Assay of Demineralized Bone Matrix
CA2535169A1 (en) 2003-12-31 2005-07-21 Osteotech, Inc. Improved bone matrix compositions and methods
CA2554571A1 (en) 2004-01-27 2005-08-11 Osteotech, Inc. Stabilized bone graft
US20050244450A1 (en) 2004-04-28 2005-11-03 Reddi A H Heat-treated implantable bone material
US7678385B2 (en) 2004-04-28 2010-03-16 Biomet Manufacturing Corp. Irradiated implantable bone material
US20060045902A1 (en) * 2004-09-01 2006-03-02 Serbousek Jon C Polymeric wrap for in vivo delivery of osteoinductive formulations
CN101141934B (en) 2005-01-14 2010-11-24 骨骼技术股份有限公司 Expandable osteoimplant
US20060216321A1 (en) 2005-03-24 2006-09-28 Sdgi Holdings, Inc. Solvent based processing technologies for making tissue/polymer composites
US20060287732A1 (en) 2005-06-20 2006-12-21 Pezeshkian Alex A Maxillary sinus bone augmentation with resorbable bone pack
US7371260B2 (en) 2005-10-26 2008-05-13 Biomet Sports Medicine, Inc. Method and instrumentation for the preparation and transplantation of osteochondral allografts
CN101365499A (en) * 2005-11-01 2009-02-11 骨骼技术股份有限公司 Bone matrix compositions and methods
WO2007062080A2 (en) 2005-11-21 2007-05-31 Philipp Lang Intervetebral devices and methods
US20070125700A1 (en) 2005-12-05 2007-06-07 Jiang Ding Nanoweb composite material and gelling method for preparing same
US8690957B2 (en) 2005-12-21 2014-04-08 Warsaw Orthopedic, Inc. Bone graft composition, method and implant
US20070162132A1 (en) 2005-12-23 2007-07-12 Dominique Messerli Flexible elongated chain implant and method of supporting body tissue with same
CA2637606C (en) 2006-01-19 2013-03-19 Osteotech, Inc. Porous osteoimplant
US8460860B2 (en) 2006-05-08 2013-06-11 Nuvasive, Inc. Cancellous bone treated with collagenase and essentially free of blood cells
EP2076220A2 (en) 2006-07-25 2009-07-08 Musculoskeletal Transplant Foundation Packed demineralized cancellous tissue forms for disc nucleus augmentation, restoration, or replacement and methods of implantation
US20080260794A1 (en) 2007-02-12 2008-10-23 Lauritzen Nels J Collagen products and methods for producing collagen products
CA2690457C (en) * 2007-06-15 2018-02-20 Osteotech, Inc. Bone matrix compositions and methods
WO2008157492A2 (en) 2007-06-15 2008-12-24 Osteotech, Inc. Osteoinductive demineralized cancellous bone
US8642061B2 (en) 2007-06-15 2014-02-04 Warsaw Orthopedic, Inc. Method of treating bone tissue
US9554920B2 (en) 2007-06-15 2017-01-31 Warsaw Orthopedic, Inc. Bone matrix compositions having nanoscale textured surfaces
US9358113B2 (en) 2007-07-10 2016-06-07 Warsaw Orthopedic, Inc. Delivery system

Also Published As

Publication number Publication date
US8992965B2 (en) 2015-03-31
EP1942960B1 (en) 2012-08-29
ES2393756T3 (en) 2012-12-27
EP1942960A2 (en) 2008-07-16
CA2627907A1 (en) 2007-05-10
WO2007053850A3 (en) 2007-07-12
CN101365499A (en) 2009-02-11
US8911759B2 (en) 2014-12-16
US20150190547A1 (en) 2015-07-09
AU2006308534B2 (en) 2013-02-07
US20070110820A1 (en) 2007-05-17
WO2007053850A2 (en) 2007-05-10
US10328179B2 (en) 2019-06-25
US20070098756A1 (en) 2007-05-03
AU2006308534A1 (en) 2007-05-10

Similar Documents

Publication Publication Date Title
US10328179B2 (en) Bone matrix compositions and methods
US10357511B2 (en) Bone matrix compositions and methods
US9415136B2 (en) Osteoinductive demineralized cancellous bone
EP1701729B1 (en) Improved bone matrix compositions and methods
US8435566B2 (en) Demineralized bone matrix compositions and methods

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20151102