WO2008157492A2 - Osteoinductive demineralized cancellous bone - Google Patents

Osteoinductive demineralized cancellous bone Download PDF

Info

Publication number
WO2008157492A2
WO2008157492A2 PCT/US2008/067117 US2008067117W WO2008157492A2 WO 2008157492 A2 WO2008157492 A2 WO 2008157492A2 US 2008067117 W US2008067117 W US 2008067117W WO 2008157492 A2 WO2008157492 A2 WO 2008157492A2
Authority
WO
WIPO (PCT)
Prior art keywords
partially demineralized
bone matrix
demineralized bone
treating
bone
Prior art date
Application number
PCT/US2008/067117
Other languages
French (fr)
Other versions
WO2008157492A3 (en
Inventor
Keyvan Behnam
Nanette Forsyth
James Russell
John M. Winterbottom
Todd M. Boyce
Original Assignee
Osteotech, 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 Osteotech, Inc. filed Critical Osteotech, Inc.
Publication of WO2008157492A2 publication Critical patent/WO2008157492A2/en
Publication of WO2008157492A3 publication Critical patent/WO2008157492A3/en

Links

Classifications

    • 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
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment
    • A61L27/3687Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment characterised by the use of chemical agents in the treatment, e.g. specific enzymes, detergents, capping agents, crosslinkers, anticalcification agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3641Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the site of application in the body
    • A61L27/3645Connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials 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
    • A61L27/3691Materials 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 physical conditions of the treatment, e.g. applying a compressive force to the composition, pressure cycles, ultrasonic/sonication or microwave treatment, lyophilisation
    • 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
    • A61L27/3695Materials 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 function or physical properties of the final product, where no specific conditions are defined to achieve this
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3839Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by the site of application in the body
    • A61L27/3878Nerve tissue, brain, spinal cord, nerves, dura mater
    • 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
    • A61F2/30Joints
    • A61F2002/30001Additional features of subject-matter classified in A61F2/28, A61F2/30 and subgroups thereof
    • A61F2002/30003Material related properties of the prosthesis or of a coating on the prosthesis
    • A61F2002/30004Material related properties of the prosthesis or of a coating on the prosthesis the prosthesis being made from materials having different values of a given property at different locations within the same prosthesis
    • A61F2002/30059Material related properties of the prosthesis or of a coating on the prosthesis the prosthesis being made from materials having different values of a given property at different locations within the same prosthesis differing in bone mineralization, e.g. made from both mineralized and demineralized adjacent parts
    • 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
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/02Materials or treatment for tissue regeneration for reconstruction of bones; weight-bearing implants

Definitions

  • 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 variously have 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.
  • BMPs bone morphogenetic or morphogenic proteins
  • osteoinductive proteins are collectively referred to herein as osteoinductive factors.
  • 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 that form osteoblasts. Based upon the work of Marshall Urist as shown in U.S. Patent No. 4,294,753, issued October 13, 1981, proper demineralization of cortical bone exposes the osteoinductive factors, rendering it osteoinductive, as discussed more fully below. [005] Overview of Bone Grafts
  • DBM demineralized bone matrix
  • BMP 1-15 which are part of the transforming growth factor-beta (TGF-beta) superfamily BMP-2 has become the most important and widely studied of the BMP family of proteins.
  • TGF-beta transforming growth factor-beta
  • proteins present in DBM that are not osteoinductive alone but still contribute to bone growth, including fibroblast growth factor-2 (FGF-2), insulin-like growth factor-I and -II (IGF-I and IGF-II), platelet derived growth factor (PDGF), and transforming growth factor-beta 1 (TGF-beta.1).
  • FGF-2 fibroblast growth factor-2
  • IGF-I and IGF-II insulin-like growth factor-I and -II
  • PDGF platelet derived growth factor
  • TGF-beta.1 transforming growth factor-beta 1
  • 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, 4,678,470, and 4,743,259; Mulliken et al., Calcif 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, each of which is incorporated herein by reference). 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 can be further processed and/or formed and shaped for implantation into a particular site in the recipient.
  • Demineralized bone prepared in this manner contains a variety of components including proteins, glycoproteins, growth factors, and proteoglycans.
  • the presence 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.
  • cortical 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.
  • the osteogenic activity of the DBM may be significantly degraded within 24 hours after 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 may 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.
  • U.S. Patent No. 4,563,350 herein incorporated by reference in its entirety, discloses the use of trypsinized bovine bone matrix as a xenogenic matrix to effect osteogenic activity when implanted with extracted, partially purified 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 immunogenicity of collagen preparations is more suitable for xenogenic implants. [016] European Patent Application Serial No. 309,241 , published March
  • the matrix is formulated by dispersing 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.
  • Demineralized bone particles are treated with a swelling agent to dissolve any associated mucopolysaccharides (glycosaminoglycans), and the collagen fibers subsequently dissolved to form a homogenous colloidal solution.
  • a gel of reconstituted fibers then can be formed using physiologically inert mucopolysaccharides and an electrolyte to aid in fibril formation.
  • Nogami and Urist also assessed the effect of various treatments of cortical bone, including collagenase treatment of cortical bone BMG, on cartilage tissue differentiation.
  • Nogami and Urist Substrata Prepared from Bone Matrix for Chondrogenesis in Tissue Culture, 62 J.of Cell Bio. 510-519 (1974).
  • MSCs mesenchymal stem cells
  • TGF- ⁇ transforming growth factors beta
  • the application of growth factors to bone and cartilage implants has not resulted in the expected increase in osteoinductive or chondrogenic activity.
  • At least partially demineralized bone matrix is treated to enhance the osteoinductive activity of the bone matrix. More specifically, the at least partially demineralized bone matrix may be treated with enzymes, chemicals, ionizing radiation, electromagnetic radiation, compaction, or other treatments to impart or enhance osteoinductive activity of the bone matrix.
  • This application refers to various patents, patent applications, journal articles, and other publications, all of which are incorporated herein by reference in their entireties. The following documents are incorporated herein by reference in their entireties: PCT Publications PCT/US04/43999 and PCT/US05/003092; US Patent Application Pub. No.
  • Figure 1 illustrates the results of Example 1.
  • Figure 2a illustrates a generally round bone particle wherein the bone particle has been surface demineralized in accordance with one embodiment.
  • Figure 2b illustrates an elongate bone particle wherein the bone particle has been surface demineralized in accordance with one embodiment.
  • Figure 3 illustrates formulations of compressed DBM.
  • Figure 4 illustrates a formulation of Figure 3 after 7 hours of submersion in water.
  • Figure 5 illustrates a formulation of Figure 3 after 72 hours of submersion in water.
  • Figure 6 illustrates a formulation of Figure 3 after 7 hours of submersion in water.
  • Bioactive Agent or Bioactive Compound refers to a compound or entity that alters, inhibits, activates, or otherwise affects biological or chemical events.
  • bioactive agents may include, but are not limited to, 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, antispasmodics and muscle contractants including channel blockers, miotics and anticholinergics, anti-glaucoma compounds, anti-parasite and/or anti-protozoal compounds, modulators of cell-extracellular matrix interactions including cell growth inhibitors and antiadhesion molecules, vasodilating agents, inhibitors of DNA, RNA or protein synthesis, anti-hypertensives, analgesic
  • the bioactive agent is a drug.
  • 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 cell attachment sequence such as RGD.
  • Biocompatible refers to materials that, upon administration in vivo, do not induce undesirable long-term effects.
  • Bone refers to bone that is cortical, cancellous or cortico-cancellous of autogenous, allogenic, xenogenic, or transgenic origin.
  • Demineralized refers to material generated by removing mineral material from tissue, e.g., bone tissue.
  • demineralized compositions as described herein may include preparations containing less than 5% calcium, or less than 1% calcium by weight.
  • Partially demineralized bone is intended to refer to preparations with greater than 5% calcium by weight but containing less than 100% of the original starting amount of calcium.
  • demineralized bone has less than 95% of its original mineral content. Percentage of demineralization may refer to percentage demineralized by weight, or to percentage demineralized by depth, as described with reference to Figures 4a and 4b.
  • Surface demineralized bone is a subset of partially demineralized bone and refers to compositions having a demineralized surface and a non-demineralized core.
  • Superficially demineralized refers to bone-derived elements possessing at least about 90 percent of their original inorganic mineral content.
  • Partially demineralized refers to bone-derived elements possessing from about 8 to about 90 percent of their original inorganic mineral content.
  • Fully demineralized refers to bone containing less than 8% of its original mineral context. Demineralized bone encompasses such expressions as "substantially demineralized,” “partially demineralized,” “surface demineralized,” and “fully demineralized.”
  • Osteoconductive refers to the ability of a non- osteoinductive substance to serve as a suitable template or substance along which bone may grow.
  • Osteogenic refers to the ability of an agent, material, or implant to enhance or accelerate the growth of new bone tissue by one or more mechanisms such as osteogenesis, osteoconduction, and/or osteoinduction.
  • Osteoimplant 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.
  • Osteoinductive refers to the quality 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 ectopic bone in the soft tissue of an animal is considered osteoinductive. For example, most osteoinductive materials induce bone formation in athymic rats when assayed according to the method of Edwards et al., Osteoinduction of Human Demineralized Bone: Characterization in a Rat Model, 357Clinical Orthopaedics & ReI. Res. 219-228 (1998), incorporated herein by reference in its entirety. In other instances, osteoinduction is 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., Osteoinduction of Human Demineralized Bone: Characterization in a Rat Model, 357 Clinical Orthopaedics & ReI. Res. 219-228 (1998) or an equivalent calibrated test.
  • a score of "0" represents no new bone formation; "1” represents l%-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.
  • the score is assessed 28 days after implantation.
  • the osteoinductivity score may be obtained at earlier time points such as 7, 14, or 21 days following implantation.
  • a normal DBM control such as DBM powder without a carrier
  • a positive control such as BMP.
  • osteoinductivity also may 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.
  • Proteases refers to protein-cleaving enzymes that cleave peptide bonds that link amino acids in protein molecules to generate peptides and protein fragments.
  • proteases and protease families have been identified.
  • Some exemplary proteases include serine proteases, aspartyl proteases, acid proteases, alkaline proteases, metalloproteases, carboxypeptidase, aminopeptidase, cysteine protease, collagenase, etc.
  • An exemplary family of proteases is the proprotein convertase family, which includes furin. Dubois et al., 158(1) Am. J. of Pathology 305-316 (2001) . Members of the proprotein convertase family of proteases are known to proteolytically process proTGFs and proBMPs to their active mature forms. Dubois et al., 158(1) Am. J.
  • proteases refers to chemical 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, 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.
  • Some protease inhibitors are protein- or peptide-based and are commercially available from chemical companies such as Aldrich-Sigma.
  • Protein or peptide-based inhibitors adhere to the DBM may provide particular benefits as they remain associated with the matrix providing a stabilizing effect for a longer period of time than freely diffusible inhibitors.
  • protease inhibitors include aprotinin, 4-(2aminoethyl) benzenesulfonyl fluoride (AEBSF), amastatin-HCl, alphal-antichymotrypsin, antithrombin III, alphal-antitrypsin, 4-aminophenylmethane sulfonyl-fluoride (APMSF), arphamenine A, arphamenine B, E-64, bestatin, CA-074, CA-074-Me, calpain inhibitor I, calpain inhibitor II, cathepsin inhibitor, chymostatin, diisopropylfluorophosphate (DFP), dipeptidylpeptidase IV inhibitor, diprotin A, E- 64c
  • Stabilizing agent refers to 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.
  • 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.
  • 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.
  • the stabilizing agent may be a chemical compound that inhibits the activity of proteases or sugar-degrading enzymes.
  • 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 therefrom.
  • 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.
  • Osteoinductive compositions and implants and methods for their production are provided.
  • at least partially demineralized bone is treated with one or more enzymes, chemicals, ionizing radiation, electromagnetic radiation, or compaction to impart or enhance osteoinductive activity of the bone.
  • the bone may be cancellous, corticocancellous, cortical, or a composite bone.
  • the bone may be fully demineralized, partially demineralized, or surface demineralized.
  • the bone is particulated, such as through grinding, and the particles are compacted.
  • Bone is made up principally of cells, and also of collagen, minerals, and other noncollagenous proteins. Cortical bone accounts for approximately eighty percent of skeletal bone mass. Cortical bone is structural and bears the majority of the body's weight. Cancellous bone is relatively porous and spongy and accounts for approximately twenty percent of skeletal bone mass. Cancellous bone contains bone marrow and elements required for bone to heal itself. The physical characteristics of cancellous bone make it an ideal material for a variety of orthopaedic applications including spinal fusions, maxillofacial and craniofacial reconstruction, and treatment of long bone defects. Despite desirable physical characteristics, cancellous bone may not be used in some applications because it is generally accepted that cancellous demineralized bone has minimal or no osteoinductive capacity.
  • Bone is living, growing tissue. In vivo, bone is constantly being renewed. The old bone is removed and new bone is laid down. There are two phases, involving cellular activities, in this process. The first phase, removal of old bone, is osteoclastic resorption. Osteoclasts dissolve some tissue on the surface of the bone and create a small cavity. Typically this process takes place over a few days. The second phase, laying down of new bone, is osteoblastic formation. Osteoblasts fill the cavities created by the osteoclasts with new bone. Typically this process takes a few months. Factors such as hormones, calcium, exercise, and other can affect the cells on the surface of bone and trigger the remodeling cycle.
  • An osteoinductive bone matrix is provided by altering the natural collagenous structure of the bone.
  • the osteoinductive bone matrix is an osteoinductive cancellous bone matrix, thus providing a graft material with desirable physical characteristics and osteoinductive capacity. In one embodiment, this is done by altering the collagenous structure of the bone such that the structure may be at least partially resorbed. In another embodiment, this is done by compacting the trabecular structure of the bone. In vivo, the osteoinductive bone matrix triggers the remodeling cycle. It is to be appreciate that, while the description herein may refer to cancellous bone for illustrative purposes, one skilled in the art would appreciate that the discussion may be applied to cortical, corticocancellous, or composite bone.
  • osteoinductive osteoimplants comprise at least partially demineralized bone having increased biological activity relative to at least partially demineralized bone that has not been exposed to a treatment or condition as described herein.
  • the osteoinductive osteoimplants may comprise at least partialy demineralized cancellous bone that has been exposed to a treatment or condition as described herein.
  • the biological activities that may be increased include but are not limited to osteoinductive activity, osteogenic activity, chondrogenic activity, triggering of bone remodeling, 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.
  • osteogenesis may be considered an early stage of osteogenesis, though of course it may also occur in other contexts.
  • the at least partially demineralized bone including wherein the at least partially demineralized bone is at least partially demineralized cancellous bone, provided herein exhibits osteoinductive activity.
  • the osteoinductive at least partially demineralized bone may be prepared by providing at least partially demineralized bone and altering the collagenous structure of the at least partially demineralized bone.
  • the osteoinductivity of the at least partially demineralized bone may be increased by either (1) altering the structure of the bone such that the trabecular density is increased while the collagen structure is decreased, or (2) by opening up the structure of the bone, thus breaking the collagen apart and making growth factors within the bone more accessible.
  • the at least partially demineralized bone is exposed to a biological or chemical agent or to a combination of agents.
  • the agent may be a cleavage agent, e.g., a protease such as collagenase(s), or a chemical agent such as cyanogen bromide.
  • the matrix may be exposed to multiple treatments either together or sequentially. While not wishing to be bound by any theory, the treatment may alter the primary, secondary, tertiary, and/or quaternary structure of a component of the bone matrix (e.g., collagen, a bone morphogenetic protein, etc.) so as to increase the biological activity of the matrix.
  • a component of the bone matrix e.g., collagen, a bone morphogenetic protein, etc.
  • An inventive treatment or condition may "open up" the structure of the matrix, e.g., so as to allow biologically active molecules to be more readily released from or diffuse within the matrix and/or to allow components such as nutrients or growth-stimulatory molecules to enter the matrix.
  • the treatment or condition cleaves proteins present in the DBM (e.g., proteins such as bone morphogenetic proteins), which may result in conversion of an inactive protein into an active form, and/or may generate an active molecule that is less susceptible to degradation than a longer molecule from which it is derived.
  • the bone is ground to a generally powder consistency and the powder is reaggregated into a dense structure through compression.
  • the at least partially demineralized bone triggers the remodeling cycle.
  • remodeling of bone may be directed to sites that show damage such as microcracks. It is theorized that the body recognizes tissue regions that need repair and then sends cells to repair those regions.
  • the at least partially demineralized bone is "marked” as requiring repair.
  • an implant formed from the cancellous at least partially demineralized bone is marked as requiring repair by cells.
  • inductive materials may be added to the implant to further stimulate bone remodeling.
  • native inductive materials in the bone may be exposed.
  • the bone may be compacted to concentrate the inductive materials (added or native).
  • the implant exhibiting tissue damage and, in some embodiments, increased and/or concentrated inductive material triggers a rapid biologic response.
  • the treatment or condition may cleave an inhibitory factor that would otherwise inhibit a positively acting agent (an agent that enhances a biological activity of the bone matrix).
  • a positively acting agent an agent that enhances a biological activity of the bone matrix.
  • proteins or protein fragments are known to inhibit the osteoinductive and/or osteogenic activity of certain bone morphogenetic proteins, such as BMP-2.
  • the inhibitory effect of a protein or protein fragment may be reduced by exposing a bone or cartilage matrix to a treatment or condition.
  • the treatment or condition may cause 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.
  • the bone matrix may be exposed to cleave antibody inhibitory agents, or the antibody may be added to the bone matrix.
  • the osteoinductive at least partially demineralized bone may form an osteogenic and osteoinductive osteoimplant, discussed more fully below.
  • the osteoimplant may have desirable characteristics of cancellous bone and yet also have osteoinductive capacity.
  • the osteoimplant when implanted in a mammalian body, may induce, at the locus of the implant, the full developmental cascade of endochondral bone formation including vascularization, mineralization, and bone marrow differentiation.
  • the osteoinductive at least partially demineralized bone may be used as a delivery device to administer bioactive agents.
  • the collagenous structure of the at least partially demineralized bone is disrupted such that the demineralized bone may be at least partially resorbed after implantation.
  • at least partially demineralized bone may be altered so that it has a solid structure at room temperature but substantially liquefies in the body or in tissue culture media such that it may have increased osteoinductive activity when compared to standard at least partially demineralized bone.
  • Cancellous femoral bone has been shown to contain osteoinductive growth factors that may be extracted from human bone.
  • cancellous DBM may be treated with, for example, collagenase, which is believed to open the structure of the cancellous DBM to make the growth factors within the bone more bioaccessible.
  • collagenase the osteoinductive cancellous DBM may substantially liquefy in the body and may be at least partially resorbed by the body when implanted.
  • Cortical bone includes dense lamellar structure.
  • the structure of cancellous bone is less dense than that of the lamellar structure of cortical bone. If demineralized bone is considered to be a growth factor source, the growth factor concentration gradients produced by implantation of cancellous and cortical allograft may differ significantly. By compressing the structure of cancellous bone, the osteoinductive potential may be increased. In some embodiments, the structure of cortical bone may be compacted.
  • Compression of the at least partially demineralized bone may be achieved via any suitable mechanism.
  • compression may be achieved by mechanical means, heat, chemical modification of the bone structure, any suitable type of compression processing, or combinations of these.
  • the bone is ground or otherwise processed into particles of an appropriate size and formed into a dense bone structure. Grinding may produce bone of a powder like consistency, the powder is wet, and the wet bone powder is smeared to a consistency of fibrous paste. The fibrous paste may be compressed and the paste may be permitted to dry.
  • the bone may be particulated before or after demineralization.
  • the bone may be particulated and thereafter partially demineralized.
  • the bone may be demineralized and then particulated.
  • the bone may be compressed by loading cancellous bone in constrained compression, for example via palletizing the bone, without grinding the bone.
  • the at least partially demineralized bone is mechanically compacted into a dense structure.
  • the at least partially demineralized bone is compacted by treatment with chemicals, such as lithium chloride (LiCl), thereby shrinking the collagenous structure.
  • demineralized bone that is substantially fully demineralized is used.
  • partially demineralized bone is used.
  • the surface demineralized bone is used.
  • nondemineralized bone may be used.
  • combinations of some of all of the above may be used. While many of the examples in this section refer to partially or surface demineralized bone, this is for illustrative purposes.
  • the bone is partially demineralized.
  • the at least partially demineralized bone 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 may be readily obtained from donor bone by various suitable methods, e.g., as described in U.S. Patent No. 6,616,698, incorporated herein by reference in its entirety.
  • the bone may be of autogenous, allogenic, xenogenic, or transgenic origin.
  • DBM preparations have been used for many years in orthopaedic medicine to promote the formation of bone.
  • DBM preparations for promoting the formation of bone have comprised cortical or corticocancellous DBM.
  • DBM has found use, for example, 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.
  • Cortical DBM is thought to promote bone formation in vivo by osteoconductive and osteoinductive processes.
  • the osteoinductive effect of implanted cortical 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- ⁇ , IGF, and BMP protein families.
  • osteoinductive factors include TGF- ⁇ , IGF-I, 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 also are likely to be other unnamed or undiscovered osteoinductive factors present in DBM. Cancellous DBM is understood to have little or no osteoinductive capacity.
  • the bone is subjected to an acid demineralization step and a defatting/disinfecting step.
  • the bone is immersed in acid over time to effect demineralization.
  • Acids that may 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.
  • Demineralization may be to a point where the bone is fully demineralized, partially demineralized, or surface demineralized.
  • the depth of demineralization into the bone surface may be controlled by adjusting the treatment time, temperature of the demineralizing solution, concentration of the demineralizing solution, and agitation intensity during treatment.
  • the at least partially 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.
  • the concentration range of the defatting solution is from about 60 to about 85 weight percent alcohol.
  • the defatting solution is about 70 weight percent alcohol.
  • the cancellous bone is defatted in a solution of 1 : 1 chloroform:methanol at room temperature and then demineralized in 0.6 N HCl at 4 0 C.
  • the at least partially demineralized bone may be ground or otherwise processed into particles of an appropriate size before or after demineralization.
  • the particle size is greater than 75 microns, ranges from about 100 to about 3000 microns, or ranges from about 100 to about 800 microns.
  • the mixture may be sieved to select those particles of a desired size.
  • the bone particles may be sieved though a 50 micron sieve, a 75 micron sieve, or a 100 micron sieve.
  • demineralization conditions may affect the osteoinductivity of DBM. Proteases that degrade the osteoinductive activity of cortical bone have been described. Proteases present in cancellous bone have been shown to differ from those responsible for cortical bone remodeling. Thus, demineralization conditions utilized for cortical bone processing may not be ideal for cancellous bone. Specific protease inhibitor cocktails and lower temperatures may be used.
  • the bone may be treated to remove mineral from the bone as discussed above.
  • hydrochloric acid is commonly used as a demineralization agent
  • there are other methods for preparing DBM which vary widely and include choices regarding the concentration of the demineralization agent; the temperature and duration of the demineralization step; the inclusion or exclusion at various points in the demineralization process of solvents or solvent combinations such as ethanol, methanol, and chloroform: ether; the extent to which the matrix is washed following the demineralization step; and whether the resulting DBM is stored frozen or is lyophilized and stored at room temperature. See, for example, Russell et al., 22(5) Orthopaedics 524-531 (1999), incorporated herein by reference.
  • 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 No. 6,326,018; Reddi et al., 69 Proc. Natl. Acad. Sci. USA 1601-1605 (1972) ; Lewandrowski et al., 317 Clin. Ortho. ReI. Res. 254-262 (1995) ; Lewandroski et al., 31 J. Biomed. Mater. Res.
  • the bone may be surface demineralized.
  • the surface may be an inner surface, such as inside trabeculae or inside a Halversion canal.
  • the surface may be an outer surface.
  • surface demineralized refers to the bone comprising at least one outer surface, or zone of an outer surface, that is demineralized and possessing a non- demineralized core.
  • the entirety of the surface may be partially demineralized.
  • a portion of the surface may be demineralized, such as by exposing only a portion of a particle to the demineralization process, by exposing a portion of the surface to a greater or lesser extent of the demineralization process, by masking, etc.
  • Demineralization may be done to a certain percentage. In some embodiments, that percentage relates to weight percentage. In other embodiments, that percentage relates to percentage of the size of the bone being demineralized, or to the depth of demineralization. The depth of demineralization of the at least one outer surface thus may be viewed as a percentage of the size of the bone being demineralized or may be viewed as an absolute number. [072] Demineralization thus may be carried out to a percentage depth of the size of the bone being demineralized.
  • Figures 4a and 4b illustrate surface demineralized bone particles.
  • the bone particle 100 of Figure 2a is substantially spherical.
  • the bone particle 110 of Figure 4b is somewhat elongate.
  • the bone particle 100 of Figure 2a has a demineralized surface region 106 and a non-demineralized core 108.
  • the bone particle 100 includes a length 102 along its longest dimension and a length 104 along its shortest dimension.
  • the length 102 in the longest dimension comprises first and second demineralized portions 103 a and 103b and a nondemineralized portion 105.
  • a percentage of demineralization in the longest dimension may be determined by summing the length of the first and second demineralized portions 103 a and 103b and dividing that total by the length 102 (comprising 103a, 103b and 105).
  • the length 104 in the shortest dimension likewise comprises first and second demineralized portions 107a and 107b and a nondemineralized portion 109.
  • a percentage of demineralization in the shortest dimension may be determined by summing the length of the first and second demineralized portions 107a and 107b and dividing that total by the length 104 (comprising 107a, 107b and 109).
  • a total percentage demineralization may be determined by averaging the percent demineralization in the longest dimension with the percent demineralization in the shortest dimension.
  • the bone particle 110 of Figure 2b has a demineralized surface region 116 and a non-demineralized core 118.
  • the bone particle 110 includes a length 112 along its longest dimension and a length 114 along its shortest dimension.
  • the longest dimension and shortest dimension are taken as those measuring largest and smallest, respectively, such as by a micrometer o r using other by suitable manner and generally going through the center of the bone particle 110.
  • the length 112 in the longest dimension comprises first and second demineralized portions 113a and 113b and a nondemineralized portion 115.
  • a percentage of demineralization in the longest dimension may be determined by summing the length of the first and second demineralized portions 113a and 113b and dividing that total by the length 112 (comprising 113a, 113b, and 115).
  • the length 114 in the shortest dimension likewise comprises first and second demineralized portions 117a and 117b and a nondemineralized portion 119.
  • a percentage of demineralization in the shortest dimension may be determined by summing the length of the first and second demineralized portions 117a and 117b and dividing that total by the length 114 (comprising 117a, 117b, and 119).
  • a total percentage demineralization may be determined by averaging the percent demineralization in the longest dimension with the percent demineralization in the shortest dimension. [075] Alternatively, percentage demineralization may be based on weight percent demineralized of total weight of the bone particle.
  • demineralization may be carried out to a depth of, for example, at least about 100 microns.
  • Surface demineralization may alternatively be done to a depth less than or more than about 100 microns.
  • surface demineralization may be done to a depth of at least 50 microns, at least 100 microns, at least 200 microns, or other.
  • the demineralized bone comprises at least one outer surface possessing at least one demineralized zone and a non-demineralized core, wherein the demineralized zone of the outer surface of the bone may be, for example, at least about 100 microns thick.
  • the demineralized zone may alternatively be less than or more than about 100 microns thick.
  • the demineralized zone of the surface of the bone is osteoinductive, and therefore promotes rapid new ingrowth of native host bone tissue into an osteoimplant comprising surface demineralized bone.
  • the osteoimplant may comprise surface demineralized monolithic bone or an aggregate of surface demineralized bone particles, and may be substantially solid, fiowable, or moldable.
  • the demineralized zone of the surface of the bone can be any surface portion.
  • the extent and regions of demineralization of the monolithic bone may be controlled.
  • depth of demineralization may range from at least about 100 microns to up to about 7000 microns or more, depending on the intended application and graft site. In some embodiments, the depth of demineralization is between 100 to about 5000 microns, between about 150 to about 2000 microns, or between about 200 microns to about 1000 microns. In alternative embodiments, depth of demineralization may be less than about 100 microns.
  • U.S. Patent No. 7,179,299 herein incorporated by reference for discussion of surface demineralization.
  • a benefit of surface demineralized bone is that the demineralized zone(s) can elastically yield under applied force while the mineralized core has strength and load bearing capacity exceeding that of demineralized bone.
  • the demineralized zones can conform to contours of adjacent bone tissue and thereby minimize voids or spaces between the osteoimplant and adjacent bone tissue. This can be useful because host bone tissue will not grow to bridge large voids or spaces.
  • an osteoimplant comprising surface demineralized monolithic bone exhibits enhanced biological properties such as, for example, incorporation and remodeling.
  • the non- demineralized inner core imparts mechanical strength and allows the monolithic osteoimplant to bear loads in vivo.
  • Other non-demineralized zones provide improved tolerances when engaged with other objects such as, for example, insertion instruments, other implants or implant devices, etc. It is noted that some of these characteristics may also be exhibited by an osteoimplant comprising an aggregate of surface-demineralized bone particles.
  • the treatment or condition alters a biological activity of the at least partially demineralized bone such that the at least partially demineralized bone displays osteoinductive, osteogenic, and/or chondrogenic activity in a species in which a control matrix (e.g., an inactivated matrix or a matrix not exposed to the treatment or condition) does not show such activity, or shows it in a lesser amount.
  • a control matrix e.g., an inactivated matrix or a matrix not exposed to the treatment or condition
  • at least partially demineralized bone exposed to the treatment or condition may display increased osteoinductive, osteogenic, and/or chondrogenic activity in human, dog, squirrel monkey, etc., as assessed either in vitro or in vivo.
  • cancellous bone which is understood to have little to no osteoinductivity, as treated herein displays osteoinductive capacity.
  • the structure of the at least partially demineralized bone may be altered to make growth factors within the bone more accessible.
  • the at least partially demineralized bone may be contacted with a cleavage agent, e.g., a protease such as collagenase(s) or a chemical agent such as cyanogen bromide.
  • the cleavage agents may be applied either together or sequentially, optionally washing the matrix between application of different agents to remove residual agent.
  • the biological and chemical agents may be used in an effective amount and for a time sufficient to achieve a desired outcome, e.g., a desired increase in a biological activity of the matrix.
  • treatment may be by limited digestion with purified bacterial 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, of Lakewood, NJ.
  • 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.
  • the cancellous DBM may be partially solubilized by the collagenase, while in others, the cancellous DBM may be completely solubilized by the collagenase.
  • a combination of treatments may be used.
  • the partially digested at least partially demineralized bone may be treated with heat or pepsin or another protease to disrupt cross-links not disrupted by the collagenase.
  • the at least partially demineralized bone may be exposed to a variety of biological agents in addition to, or instead of, one or more proteases.
  • 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 maybe of eukaryotic (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.
  • the collagenase cleaves Type I collagen, e.g., degrades the helical regions in native collagen, at the Y-GIy 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.
  • collagenase inhibiting agents when treating at least partially demineralized bone with collagenase.
  • additional proteases present in some collagenase preparations may aid in breaking down tissue, they also may 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.
  • a collagenase preparation may contain at least 90%, at least 95%, at least 98%, or 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% may be used. It may be desirable to include various protease inhibitors that do not inhibit collagenase but that inhibit various proteases that digest BMP.
  • protease inhibitors that are known to protect BMP activity from degradation include N-ethyl maleimide, benzamidine hydrochloride, iodoacetic acid, PMSF, AEBSF, and E-64. Bestatin may also be used, particularly if the preparation contains aminopeptidase activity. Any of these protease inhibitors (or others) may be included in a carrier, such as a bone matrix composition, or in any composition that is used to treat a carrier. [089] As discussed above, collagenase disrupts crosslinks between collagen.
  • 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 may be used.
  • Newly synthesized human 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.
  • the at least partially demineralized bone may be treated with
  • At least partially demineralized bone may be exposed to any of the enzymes, e.g., proteases described herein (and others) at a range of different concentrations, e.g., between 1 pg/ml - 100 ⁇ g/ml.
  • a protease maybe used at between 1 pg/ml - 100 pg/ml, between 100 pg/ml and 1 ng/ml, between 1 ng/ml and 100 ng/ml, between 100 ng/ml and 1 ⁇ g/ml, between 1 ⁇ g/ml and 100 ⁇ g/ml, etc.
  • a variety of different digestion buffers may be used.
  • the time of digestion may vary according to the protease, amount of DBM, and desired degree of digestion. In general, suitable times range between 30 minutes to 72 hours, e.g., between 30 minutes to 1 hour, between 1 and 12 hours, between 12 and 24 hours, between 24 and 48 hours, between 48 and 72 hours, etc. It will be appreciated that these times are approximate. Determination of the optimal treatment times for any preparation may involve assay of the treated tissue preparation in one of the biological activity assays described herein or others known in the art. [095] Allogenic cancellous demineralized bone is known not to be osteoinductive. Schwarz et al., 111(1) Arch Orthop Trauma Surg. 47-50 (1991).
  • Structural modifications of demineralized bone collagen may serve to activate the latent osteoinductive potential of DBM preparations in higher order species.
  • Such products irrespective of whether derived from cancellous or cortical bone, may have significantly enhanced clinical efficacy in a variety of orthopaedic applications as observed by more rapid bone healing and spinal fusion.
  • the at least partially demineralized bone may be exposed to a physical condition instead of, or in addition to, a biological or chemical agent.
  • the at least partially demineralized bone may be exposed to heat or cold for a suitable period of time, e.g., minutes, hours, or up to several days, where "heat” refers to temperatures above room temperature (about 23-25 degrees C) and "cold” refers to temperatures below room temperature. Cycles of temperature change may be used, e.g., the at least partially demineralized bone may be heated and cooled a plurality of times.
  • the temperature may, for example, be at least 37 degrees C, at least 40, 50, 60, 70, 80, or 90 degrees C.
  • the heat treatment may be relatively gentle to avoid denaturing growth factors and other factors, typically proteins or peptides, that contribute to the osteogenic, osteoinductive, or chondrogenic activity of the matrix.
  • the temperature may be 20 degrees C or below, 15 degrees C or below, 10 degrees C or below, 0 degrees C or below, etc.
  • the at least partially demineralized bone may be exposed to any desired temperature in the presence or absence of other agents, solvents, or other suitable media.
  • the at least partially demineralized bone may be exposed to electromagnetic energy of any type, e.g., X-rays and microwaves. Gamma-rays, beta-rays, or any suitable ionizing radiation may be used.
  • the treatment may be performed in the absence of oxygen or in a reduced oxygen environment.
  • the treatment or condition alters the physical structure of the matrix so as to increase its biological activity.
  • the treatment or condition may alter the structure of the at least partially demineralized bone so as to facilitate the presentation of such molecules, e.g., on a surface of the at least partially demineralized bone.
  • the treatment or condition may alter the conformation of such molecules in a manner that facilitates interactions with target cells, e.g., cells that migrate towards or into the bone matrix.
  • the treatment or condition may alter release kinetics of agents such as growth factors, differentiation factors, chemotactic factors, etc., from the matrix.
  • collagenase may cut across the framework and thereby potentially allow access to the NCPs.
  • the amount of collagen (or other structural protein) that is cleaved and/or degraded may vary. For example, in certain embodiments, at least 10%, at least 25%, at least 50%, at least 75%, or at least 90% of the collagen originally present in the DBM is cleaved or degraded. Between 10-25%, 25-50%, 50-75%, 75-90%, 90- 100%, or any other suitable range such as 10-90%, 25-75%, etc., of the collagen maybe cleaved or degraded.
  • a polypeptide is considered to be cleaved if it is cleaved at a single site or at multiple sites.
  • a bone matrix may be exposed to any of a variety of different biological or chemical agents or conditions for different time periods in order to achieve a desired degree of cleavage or degradation of a structural component of the matrix such as collagen.
  • the invention therefore provides a modified bone matrix comprising a collagen-containing bone matrix, wherein at least a portion of the collagen is cleaved or degraded. Matrices in which at least a portion of a different structural component of the matrix is cleaved or degraded are also provided.
  • the at least partially demineralized bone may be exposed to a treatment or condition that generates peptides and protein fragments having osteoinductive or chondrogenic activity.
  • 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 domains are capable of functioning independently as peptides and protein fragments.
  • the present invention increases the osteoinductive or chondrogenic activity of bone and cartilage matrices 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.
  • the osteoimplant may be rehydrated to expand to an increased size, for example as far as permitted by the confines of the intervertebral body fusion device and spinal endplates, thereby aiding in greater vertebral endplate contact and conformity in spinal surgery. In addition or alternatively, rehydration may take place in vivo.
  • the osteoimplant may comprise fully or partially demineralized cancellous bone and fully and/or partially demineralized cortical bone.
  • the cancellous bone is provided in chips, and the cortical bone in fibers.
  • the demineralized cancellous bone may comprise, in whole or part, osteoinductive cancellous DBM as taught herein.
  • the expandable osteoimplant may comprise, in whole or in part, cancellous DBM, some or all of which may be made osteoinductive pursuant to the present invention.
  • the expandable osteoimplant may comprise a monolithic piece of bone treated as provided herein, such as by at least partially demineralizing and treating the monolithic piece of bone.
  • the compressed at least partially demineralized bone is provided by grinding the bone, for example into a fine powder, and molding it into a cohesive shape.
  • the bone may be wetted and processed to a consistency of a fibrous paste before molding.
  • the molding is done via hand molding and drying the material at ambient temperature.
  • the molding is done by putting the bone in a mold, such as a polyurethane or PTFE mold, putting the mold under pressure, for example by clamping the mold, and allowing the bone to dry, for example via vacuum drying the material at 4O 0 C for 72 hours.
  • the molding is done by putting the bone into a stainless steel mold, pressing the mold with 500 pounds and vacuum drying the mold at 4O 0 C for 72 hours with weighted dehydrothermal plungers.
  • the DBM may be ground to a powder- like consistency, processed into a fibrous paste, and the fibrous paste compressed and dried. Compression and drying may be done in any suitable manner. For example, compression may be done by applying manual pressure, by clamping (powered manually, through dehydrothermal plungers, or other), or other means. Drying may be done in air at ambient temperature, in an oven, vacuum dried, or in any other suitable manner.
  • the at least partially demineralized bone is compacted by treatment with LiCl to shrink the collagenous structure.
  • at least partially demineralized bone may be treated with 8M LiCl to increase the osteoinductivity of the at least partially demineralized bone. Treatment with LiCl may further increase the radioopacity of the at least partially demineralized bone.
  • the at least partially demineralized bone matrix provided herein may be used as a structural graft, an osteogenic graft, or a graft suitable for both structural and osteogenic uses.
  • Any suitable shape, size, and porosity of at least partially demineralized bone may be used.
  • the bone may be monolithic or may be composite. Rat studies show that the new bone is formed essentially having the dimensions of the device implanted. A successful osteoimplant for encouraging bone development appropriately accommodates each step of the cellular response during bone development, and in some cases, protects osteoinductive factors from nonspecific proteolysis. In some uses, the osteoimplant acts as a temporary scaffold until replaced completely by new bone. In bone, the dissolution rates may vary according to whether the implant is placed in cortical or trabecular bone.
  • the osteoimplant resulting from the at least partially demineralized bone may assume a determined or regular form or configuration such as a sheet, plate, disk, tunnel, cone, tube, a rod, string, weave, solid, fiber, or wedge, to name but a few.
  • Prefabricated geometries include, but are not 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, wave plates.
  • Partial tubular as well as flat plates may be fabricated from the osteoimplant.
  • Such plates may include such conformations as, e.g., concave contoured, bowl shaped, or defect shaped.
  • the shape and size of the particles in the carrier affect the time course of osteoinductivity. For example, in a cone or wedge shape, the tapered end will result in osteoinductivity shortly after implantation of the osteoimplant, whereas the thicker end will lead to osteoinductivity later in the healing process (hours to days to weeks later). Also, larger particle size will have induce bone formation over a longer time course than smaller particles. Particles of different characteristics (e.g., composition, size, shape) maybe used in the formation of these different shapes and configurations.
  • Suitable sheets include those sold under the trade name Grafton ® 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 human bone marrow stromal cells (BMSCs), which may be useful in the repair of large bone defects.
  • BMSCs human bone marrow stromal cells
  • 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.
  • additives or carriers include polyhydroxy compounds, polysaccharides, glycosaminoglycan proteins, nucleic acids, polymers, polaxomers, resins, clays, calcium salts, and/or derivatives thereof. See, for example, U.S. Patent No. 5,290,558, incorporated by reference herein in its entirety.
  • the bone used in creating the at least partially demineralized bone may be obtained from any source of living or dead tissue. In many instances, the source of bone may be matched to the eventual recipient of the inventive composition.
  • DBM derived at least in part from human bone may be used.
  • 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.
  • the osteoinductive at least partially demineralized bone 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 DBM.
  • 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 health, and the risk of infection.
  • the at least partially demineralized bone may comprise, for example, less than approximately 0.5% water, less than approximately 1% water, or less than approximately 5% water.
  • the osteoinductive at least partially demineralized bone may be configured to be suturable.
  • the osteoinductive at least partially demineralized bone or an osteoimplant formed from the osteoinductive at least partially demineralized bone may be sutured in place in vivo.
  • An osteoimplant formed from osteoinductive cancellous DBM may be configured for expansion.
  • Cancellous DBM has a sponge-like texture and expands and conforms.
  • an osteoimplant may be formed that is able to expand and conform to a site in vivo.
  • the osteoimplant so formed may comprise monolithic cancellous bone or particulated (and reaggregated) cancellous bone.
  • an osteoimplant having a pre-selected three- dimensional shape is prepared by repeated application of individual layers of DBM, for example by 3-D printing as described by U.S. Patent Nos. 5,490,962, 5,518,680, and 5,807,437, each incorporated herein by reference in their entireties.
  • Different layers may comprise individual stabilized DBM preparations, or alternatively may comprise DBM layers treated with stabilizing agents after deposition of multiple layers.
  • 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), super critical CO 2 treatment, thermal sterilization, or combinations thereof.
  • Other methods known in the art of preparing bone and cartilage matrices, such as defatting, sonication, lyophilization, and combinations thereof may also be used. Since the biological activity of various materials including demineralized bone is known to be detrimentally affected by most terminal sterilization processes, care must be taken when sterilizing the inventive compositions.
  • the osteoimplants described herein will be prepared aseptically or sterilized.
  • the present invention provides methods of increasing the osteoinductivity of the at least partially demineralized bone by further exposing the at least partially demineralized bone to at least one treatment (e.g., a biological or chemical agent).
  • at least one treatment e.g., a biological or chemical agent
  • the at least partially demineralized bone 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.
  • An additive may be provided within the osteoimplant in a sustained release format.
  • the additive may be encapsulated within biodegradable nanospheres, microspheres, or the like.
  • Osteoinductive factors may be added to the treated at least partially demineralized bone, thus further increasing the osteoinductivity of the at least partially demineralized bone.
  • Osteoinductive factors include any agent that leads to or enhances the formation of bone.
  • the osteoinductive factors may do this in any manner.
  • the osteoinductive factors may lead to the recruitment of cells responsible for bone formation, the osteoinductive factors may lead to the secretion of matrix which may subsequently undergo mineralization, the osteoinductive factors may lead to the decreased resorption of bone, etc.
  • Suitable osteoinductive factors include bone morphogenic proteins (BMPs), transforming growth factor (TGF-O), insulin- like growth factor (IGF-I), parathyroid hormone (PTH), and angiogenic factors such as VEGF.
  • osteoinductive factors also may be added to the at least partially demineralized bone. These osteoinductive factors may be added in an activated or nonactivated form, and they may be added at anytime during the preparation of the inventive material. For example, the osteoinductive factors may be added after the demineralization step and prior to the addition of the stabilizing agents so that the added osteoinductive factors are protected from exogenous degrading enzymes once implanted.
  • the at least partially demineralized bone is lyophilized in a solution containing the osteoinductive factors.
  • the osteoinductive factors are adhered onto the hydrated demineralized bone matrix and are not freely soluble.
  • the osteoinductive factors are added after addition of a stabilizing agent so that the osteoinductive factors are available immediately upon implantation of the at least partially demineralized bone.
  • Radiopaque substances may be added to impart radiopacity to the at least partially demineralized bone.
  • 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.
  • substances imparting radiopacity will typically represent from about 1 to about 25 weight percent of the bone particle containing composition, calculated prior to forming the shaped material.
  • the at least partially demineralized bone may have some inherent radiopacity due to remaining mineralization.
  • class 3 semaphorins e.g., SEMA3, controls vascular morphogenesis by inhibiting integrin function in the vascular system, Serini et al., 424 Nature 391-397 (2003), incorporated herein by reference in its entirety, and may be included in the osteoimplant.
  • the osteoconductive at least partially demineralized bone may provide a system for delivering bioactive agents, such as osteoinductive factors, to a host animal.
  • bioactive agents such as osteoinductive factors
  • 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.
  • bioactive agents that may be delivered using an at least partially demineralized bone composition include agents that promote the natural healing process, i.e., resorption, vascularization, angiogenesis, new growth, or the like.
  • the osteoimplant is provided in which the treated at least partially demineralized bone, 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.
  • the bioactive agent is an osteoinductive agent, and in certain embodiments, the at least partially demineralized bone may be used to deliver more than one bioactive agent, more than two, and sometimes more than three bioactive agents.
  • the bioactive agent may be associated with the at least partially demineralized bone.
  • the bioactive agent may be associated with the at least partially demineralized bone through electrostatic interactions, hydrogen bonding, pi stacking, hydrophobic interactions, van der Waals interactions, etc.
  • the bioactive agent is attached to the at least partially demineralized bone through specific interactions such as those between a receptor and its ligand or between an antibody and its antigen.
  • the bioactive agent is attached to the at least partially demineralized bone through non-specific interactions (e.g., hydrophobic interactions).
  • Medically/surgically useful substances include physiologically or pharmacologically active substances that act locally or systemically in the host. Generally, these substances may include bioactive substances which may be readily incorporated into the osteoimplant and include, e.g., demineralized bone powder as described in U.S. Patent No.
  • 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, an 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.
  • 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.
  • BMG bone matrix gelatin
  • Glacial acetic acid add 248.56 ml of deionized water to 1.44 ml of 17.4 M Glacial Acetic Acid, Sterile filtered).
  • collagenase treatment comprised placing sections of demineralized cancellous dowels weighing between 50 and 100 mg in a 5 ml digestion buffer containing 80 units/ml collagenase. The samples were incubated at 37 0 C for 90 minutes. Each sample was washed for 30 minutes with 30 ml 0.1N acetic acid in the cold. The samples were then rinsed for 10 minutes with phosphate buffered saline ("PBS") and 10 minutes with deionized water prior to lyophilization.
  • PBS phosphate buffered saline
  • Figure 1 is a graph illustrating the results of Example 1. As shown, the collagenase treated DBM had higher histologic scores for both the long and short samples than did the control DBM. The long collagenase treated BMG sample had a lower histologic score than did the long control BMG sample. The short collagenase treated BMG sample had a higher histologic score than did the short control BMG sample. [0160] The results demonstrate that collagenase treatment enhances the osteoinductive activity of cancellous DBM scaffolds. More specifically, the results indicate that the osteinductive activity of cancellous DBM scaffolds, as measured by the histologic score, was measurable, and increased above that of untreated DBM, in all samples.
  • Collagenase treated human demineralized cancellous scaffolds have potent osteoinductive activity as measured by heterotopic bone formation in athymic rats.
  • the negligible osteoinductive activity of human cancellous demineralized scaffolds in athymic mice may be markedly enhanced by treatment with bacterial collagenase, without requiring treatment with LiCl prior to treatment with bacterial collagenase.
  • the activity of these preparations is comparable to that of human cortical DBM.
  • Cancellous bone was demineralized in 0.6 N HCl at room temperature. The demineralized bone was washed, and the wet demineralized bone was smeared to a consistency of a fibrous paste. The fibrous paste was then treated using one of three methods:
  • Figure 3 illustrates the formulations thus produced.
  • Formulation 200 corresponds to method 1
  • formulation 202 corresponds to method 2
  • formulation 204 corresponds to method 3.
  • Each of the formulations 200, 202, and 204 were submerged in deionized water and observed for cohesiveness.
  • Figure 4 illustrates formulation 200 at 7 hours of submersion in water.
  • Figure 5 illustrates formulation 202 submerged in water for 72 hours.
  • Figure 6 illustrates formulation 204 submerged in water for 72 hours. From Figure 4, formulation 200 swelled when submerged in water but did not dissociate. From Figures 5 and 6, formulations 202 and 204 each generally retained their shape in water.
  • 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 may be utilized in a wide variety of orthopaedic, periodontal, neurosurgical, and oral and maxillofacial surgical procedures.
  • Demineralized cancellous bone has a sponge-like structure. The material may be compressed significantly and will substantially regain its original structure in the absence of the compressive force. An osteoinductive cancellous scaffold may be forced into a space or defect through a small opening. The structure will then expand and conform to the shape of the environment.
  • the implant may expand in vivo over the course of between a few hours to a few weeks depending on, for example, the density of the implant, other materials incorporated into the compressed implant, the size and shape of the implant, or other aspects of the implant that would be readily understandable by those skilled in the art.
  • Activated scaffolds comprising the osteoinductive cancellous DBM may reduce or eliminate the need for harvesting iliac crest autograft in challenging orthopaedic applications such as posterolateral spinal fusion and the treatment of critical sized nonunion fractures.
  • the osteoinductive at least partially demineralized bone may also be used as a drug delivery device.
  • Demineralized cancellous bone has the capacity to absorb significant amounts of fluids, and it has a high surface area to volume ratio. These properties indicate that the material would be an ideal growth scaffold for bioactive cells.
  • Osteoinductive at least partially demineralized bone may be used as a carrier for osteogenic cells, chrondrogenic cells, stem cells, bone marrow aspirates, fat cells, and other cell types that are involved in tissue formation or that have the capability to differentiate into tissue forming cells when exposed to appropriate signals.
  • the at least partially demineralized bone may also serve as a carrier for a variety of native or recombinant growth and differentiation factors.
  • association with the osteoinductive at least partially demineralized bone increases the half-life of the relevant biologically active agent(s).
  • Certain inventive drug delivery devices are used to deliver osteoinductive growth factors.
  • Other agents that may be delivered include factors or agents that promote wound healing.
  • the osteoinductive at least partially demineralized bone may alternatively or additionally be used to deliver other pharmaceutical agents including antibiotics, anti-neoplastic agents, growth factors, hematopoietic factors, nutrients, an other bioactive agents described above.
  • the amount of the bioactive agent included with the at least partially demineralized bone 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.
  • the osteoinductive at least partially demineralized bone 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 at least partially demineralized bone that has not been exposed to a treatment or condition as described herein.
  • these values may vary 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%.
  • the osteoimplant when an osteoimplant 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, hi some embodiments, when an osteoimplant comprising osteoinductive cancellous DBM is implanted in a bone defect site, the osteoimplant has an osteoinductivity score comparable to a cortical DBM implant that has not been treated.

Abstract

An osteoinductive demineralized bone matrix, corresponding osteoimplants, and methods for making the osteoinductive demineralized bone matrix are disclosed. The osteoinductive demineralized bone matrix may be prepared by providing demineralized bone and altering the collagenous structure of the bone. The osteoinductive demineralized bone matrix may also be prepared by providing demineralized bone and compacting the bone, for example via mechanical compaction, grinding into a particulate, or treatment with a chemical. Additives such as growth factors or bioactive agents may be added to the osteoinductive demineralized bone matrix. The osteoinductive demineralized bone matrix may form an osteogenic osteoimplant. The osteoimplant, when implanted in a mammalian body, may induce at the locus of the implant the full developmental cascade of endochondral bone formation including vascularization, mineralization, and bone marrow differentiation. The osteoinductive demineralized bone matrix may also be used as a delivery device to administer bioactive agents.

Description

OSTEOINDUCTIVE DEMINERALIZED CANCELLOUS BONE
BACKGROUND
[001] This application application claims benefit of U.S. Serial No.
60/944,417, filed on June 15, 2007 and U.S. Serial No. 60/986,843 filed on November 9, 2007, each of which are hereby incorporated by reference in their entireties.
[002] Introduction
[003] 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 variously have 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. [004] It is well known 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 that form osteoblasts. Based upon the work of Marshall Urist as shown in U.S. Patent No. 4,294,753, issued October 13, 1981, proper demineralization of cortical bone exposes the osteoinductive factors, rendering it osteoinductive, as discussed more fully below. [005] Overview of Bone Grafts
[006] 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 influencing their suitability and performance in various orthopaedic applications. [007] 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.
[008] Much effort has been invested in the identification and development of alternative 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, 150 Science 698, 893-899 (1965); Urist M.R. et al., The Bone Induction Principle, 53 Clin. Orthop. ReI. Res. 243-283 (1967). As mentioned above, it is known that DBM that is derived from cortical bone 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 also known that there are numerous osteoinductive factors, e.g., BMP 1-15, which are part of the transforming growth factor-beta (TGF-beta) superfamily BMP-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-2 (FGF-2), insulin-like growth factor-I and -II (IGF-I and IGF-II), platelet derived growth factor (PDGF), and transforming growth factor-beta 1 (TGF-beta.1).
[009] 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, 4,678,470, and 4,743,259; Mulliken et al., Calcif 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, each of which is incorporated herein by reference). 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 can be further processed and/or formed and shaped for implantation into a particular site in the recipient. Demineralized bone prepared in this manner contains a variety of components including proteins, glycoproteins, growth factors, and proteoglycans. Following implantation, the presence 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. [010] It is generally accepted that cancellous demineralized bone matrix does not have osteoinductive capacity. One study found that allogeneic cancellous bone blocks, demineralized or not, have no osteoinductive capacity and no osteoconductive function that promotes healing of mid-diaphyseal bone defects in dogs. Schwarz et al., Arch Orthop Trauma Surg., 1991 ; 11 l(l):47-50; see also Nade et al., J Bone Joint Surg Br 1997 May 59(2): 189-96. [011] 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. The study looked at rat cortical bone matrix, canine cortical bone matrix, and canine cancellous bone matrix. Cortical 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.
[012] Another study, looking at cortical bone matrix from monkeys, 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.
[013] 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 GTR plus DBM and GTR defects. Caplanis et al., J Periodontal 851-856, August, 1998. [014] Current DBM formulations have various drawbacks. 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 after 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 may 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.
[015] U.S. Patent No. 4,563,350, herein incorporated by reference in its entirety, discloses the use of trypsinized bovine bone matrix as a xenogenic matrix to effect osteogenic activity when implanted with extracted, partially purified 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 immunogenicity of collagen preparations is more suitable for xenogenic implants. [016] European Patent Application Serial No. 309,241 , published March
29, 1989, herein incorporated by reference in its entirety, 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% atelopeptide hypoimmunogenic collagen. [017] U.S. Patent No. 3,394,370, herein incorporated by reference in its entirety, 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 interfϊbril crosslinks), and are dissolved to remove associated noncollagenenous components. The matrix is formulated by dispersing 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.
[018] U.S. Patent No. 4,172,128, herein incorporated by reference in its entirety, describes a method for degrading and regenerating bone-like material of reduced immunogenicity, said to be useful cross-species. Demineralized bone particles are treated with a swelling agent to dissolve any associated mucopolysaccharides (glycosaminoglycans), and the collagen fibers subsequently dissolved to form a homogenous colloidal solution. A gel of reconstituted fibers then can be formed using physiologically inert mucopolysaccharides and an electrolyte to aid in fibril formation.
[019] Various papers have looked at cartilage tissue differentiation of bone matrix gelatin (BMG) from cortical bone. Terashima and Urist found that cortical bone BMG is chemically more reactive than whole bone matrix. Terashima et al., Chondrogenesis in Outgrowths of Muscle Tissue onto Modified Bone Matrix in Tissue Culture, Ml Clin. Orthop. and ReI. Res. 248-256 (1977). A later study found that rat BMG may induce chondrogenesis in cell culture. Urist et al., Cartilage Tissue Differentiation from Mesenchymal Cells Derived from Mature Muscle in Tissue Culture, 14(8) In Vitro 697-706 (1978). Nogami and Urist also assessed the effect of various treatments of cortical bone, including collagenase treatment of cortical bone BMG, on cartilage tissue differentiation. Nogami and Urist, Substrata Prepared from Bone Matrix for Chondrogenesis in Tissue Culture, 62 J.of Cell Bio. 510-519 (1974). [020] 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 mesenchymal stem cells (MSCs) in the bone marrow, which have the potential to differentiate into cartilage and bone. Steadman, et al., Microfracture: Surgical Technique and Rehabilitation to Treat Chondral Defects 391 S Clin. Orthop. 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-β) have the capacity to recruit progenitor cells into a chondral defect from the synovium or elsewhere when loaded in the defect. Hunziker, et al., Repair of Partial Thickness Defects in Articular Cartilage: Cell Recruitment From the Synovial Membrane, 78-A J. Bone Joint Surg., 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.
[021] Each of U.S. Patent Nos. 5,270,300 and 5,041,138, each herein incorporated by reference in its entirety, 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 contain growth factors (TGF-β or other factors (such as angiogenesis factors)) appropriate for the type of tissue desired to be regenerated.
BRIEF SUMMARY
[022] Osteoinductive compositions, and implants and methods for their production, are provided. At least partially demineralized bone matrix is treated to enhance the osteoinductive activity of the bone matrix. More specifically, the at least partially demineralized bone matrix may be treated with enzymes, chemicals, ionizing radiation, electromagnetic radiation, compaction, or other treatments to impart or enhance osteoinductive activity of the bone matrix. [023] This application refers to various patents, patent applications, journal articles, and other publications, all of which are incorporated herein by reference in their entireties. The following documents are incorporated herein by reference in their entireties: PCT Publications PCT/US04/43999 and PCT/US05/003092; US Patent Application Pub. No. 2003/0143258 Al; PCT Publication 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 & Sons 2002); Sambrook, Russell, and Sambrook, Molecular Cloning: A Laboratory Manual, (Cold Spring Harbor Laboratory Press2001); Rodd , Chemistry of Carbon Compounds, vols. 1-5 and supps. (Elsevier Science Publishers 1989); Organic Reactions, vols. 1-40 (John Wiley & Sons 1991); Advanced Organic Chemistry, (John Wiley & Sons 2001). In the event of a conflict between the specification and any of the incorporated references, the specification shall control. [024] 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 spirit and scope of the present invention. Accordingly, the detailed description is to be regarded as illustrative in nature and not restrictive.
BRIEF DESCRIPTION OF THE DRAWINGS
[025] Figure 1 illustrates the results of Example 1.
[026] Figure 2a illustrates a generally round bone particle wherein the bone particle has been surface demineralized in accordance with one embodiment.
[027] Figure 2b illustrates an elongate bone particle wherein the bone particle has been surface demineralized in accordance with one embodiment.
[028] Figure 3 illustrates formulations of compressed DBM.
[029] Figure 4 illustrates a formulation of Figure 3 after 7 hours of submersion in water.
[030] Figure 5 illustrates a formulation of Figure 3 after 72 hours of submersion in water.
[031] Figure 6 illustrates a formulation of Figure 3 after 7 hours of submersion in water.
DEFINITIONS
[032] Bioactive Agent or Bioactive Compound, as used herein, refers 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, immunosuppressants, anti-viral substances, enzyme inhibitors, hormones, neurotoxins, opioids, hypnotics, anti-histamines, lubricants, tranquilizers, anti-convulsants, muscle relaxants and anti-Parkinson substances, antispasmodics and muscle contractants including channel blockers, miotics and anticholinergics, anti-glaucoma compounds, anti-parasite and/or anti-protozoal compounds, modulators of cell-extracellular matrix interactions including cell growth inhibitors and antiadhesion 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, ophthalmics, prostaglandins, anti-depressants, antipsychotic substances, anti-emetics, 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 may be found in Axel Kleemann and Jurgen Engel, Pharmaceutical Substances: Syntheses, Patents, Applications (Thieme Medical Publishing 1999); the Merck Index: An Encyclopedia of Chemicals, Drugs, and Biologicals (Susan Budavari et al., CRC Press 1996); and the United States Pharmacopeia-25/National Formulary-20(United States Pharmcopeial Convention, Inc. 2001), each of which is incorporated herein by reference in their entireties.
[033] Biocompatible, as used herein, refers to materials that, upon administration in vivo, do not induce undesirable long-term effects. [034] Bone, as used herein, refers to bone that is cortical, cancellous or cortico-cancellous of autogenous, allogenic, xenogenic, or transgenic origin. [035] Demineralized, as used herein, refers to material generated by removing mineral material from tissue, e.g., bone tissue. In certain embodiments, demineralized compositions as described herein may include preparations containing less than 5% calcium, or less than 1% calcium by weight.Partially demineralized bone is intended to refer to preparations with greater than 5% calcium by weight but containing less than 100% of the original starting amount of calcium. In some embodiments, demineralized bone has less than 95% of its original mineral content. Percentage of demineralization may refer to percentage demineralized by weight, or to percentage demineralized by depth, as described with reference to Figures 4a and 4b. Surface demineralized bone is a subset of partially demineralized bone and refers to compositions having a demineralized surface and a non-demineralized core. Superficially demineralized refers to bone-derived elements possessing at least about 90 percent of their original inorganic mineral content. Partially demineralized refers to bone-derived elements possessing from about 8 to about 90 percent of their original inorganic mineral content. Fully demineralized refers to bone containing less than 8% of its original mineral context. Demineralized bone encompasses such expressions as "substantially demineralized," "partially demineralized," "surface demineralized," and "fully demineralized."
[036] Osteoconductive, as used herein, refers to the ability of a non- osteoinductive substance to serve as a suitable template or substance along which bone may grow. [037] Osteogenic, as used herein, refers to the ability of an agent, material, or implant to enhance or accelerate the growth of new bone tissue by one or more mechanisms such as osteogenesis, osteoconduction, and/or osteoinduction. [038] 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. [039] Osteoinductive, as used herein, refers to the quality 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 ectopic bone in the soft tissue of an animal is considered osteoinductive. For example, most osteoinductive materials induce bone formation in athymic rats when assayed according to the method of Edwards et al., Osteoinduction of Human Demineralized Bone: Characterization in a Rat Model, 357Clinical Orthopaedics & ReI. Res. 219-228 (1998), incorporated herein by reference in its entirety. In other instances, osteoinduction is 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., Osteoinduction of Human Demineralized Bone: Characterization in a Rat Model, 357 Clinical Orthopaedics & ReI. Res. 219-228 (1998) or an equivalent calibrated test. In the method of Edwards et al., a score of "0" represents no new bone formation; "1" represents l%-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 time 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 also may 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. [040] Proteases, as used herein, refers to 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, aminopeptidase, cysteine protease, collagenase, etc. An exemplary family of proteases is the proprotein convertase family, which includes furin. Dubois et al., 158(1) Am. J. of Pathology 305-316 (2001) . Members of the proprotein convertase family of proteases are known to proteolytically process proTGFs and proBMPs to their active mature forms. Dubois et al., 158(1) Am. J. of Pathology 305-316 (2001); Cui et al., 17(16) Embo J. 4735-4743 (1998); Cui et al., 15 Genes & Development 2797-2802 (2001), each incorporated by reference herein in their entireties.
[041] Protease inhibitors, as used herein, refers to chemical 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, 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. Some protease inhibitors are protein- or peptide-based and are commercially available from chemical companies such as Aldrich-Sigma. Protein or peptide-based inhibitors adhere to the DBM (or calcium phosphate or ceramic carrier) may provide particular benefits 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-HCl, alphal-antichymotrypsin, antithrombin III, alphal-antitrypsin, 4-aminophenylmethane sulfonyl-fluoride (APMSF), arphamenine A, arphamenine B, E-64, bestatin, CA-074, CA-074-Me, calpain inhibitor I, calpain inhibitor II, 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 fluo4de (PMSF), pepstatin A, phebestin, l,10phenanthroline, phosphoramidon, chymostatin, benzamidine HCI, antipain, epsilon aminocaproic acid, N-ethylmaleimide, trypsin inhibitor, l-chloro-3-tosylamido-7-amino2-heptanone (TLCK), l-chloro-3- tosylamido-4-phenyl-2-butanone (TPCK), trypsin inhibitor, and sodium EDTA. [042] Stabilizing agent, as used herein, refers to 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. According to certain embodiments, 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 therefrom. 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.
DETAILED DESCRIPTION
[043] I. INTRODUCTION
[044] Osteoinductive compositions and implants and methods for their production are provided. According to certain embodiments, at least partially demineralized bone is treated with one or more enzymes, chemicals, ionizing radiation, electromagnetic radiation, or compaction to impart or enhance osteoinductive activity of the bone. The bone may be cancellous, corticocancellous, cortical, or a composite bone. The bone may be fully demineralized, partially demineralized, or surface demineralized. In some embodiments, the bone is particulated, such as through grinding, and the particles are compacted. 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. [045] Bone is made up principally of cells, and also of collagen, minerals, and other noncollagenous proteins. Cortical bone accounts for approximately eighty percent of skeletal bone mass. Cortical bone is structural and bears the majority of the body's weight. Cancellous bone is relatively porous and spongy and accounts for approximately twenty percent of skeletal bone mass. Cancellous bone contains bone marrow and elements required for bone to heal itself. The physical characteristics of cancellous bone make it an ideal material for a variety of orthopaedic applications including spinal fusions, maxillofacial and craniofacial reconstruction, and treatment of long bone defects. Despite desirable physical characteristics, cancellous bone may not be used in some applications because it is generally accepted that cancellous demineralized bone has minimal or no osteoinductive capacity.
[046] Bone is living, growing tissue. In vivo, bone is constantly being renewed. The old bone is removed and new bone is laid down. There are two phases, involving cellular activities, in this process. The first phase, removal of old bone, is osteoclastic resorption. Osteoclasts dissolve some tissue on the surface of the bone and create a small cavity. Typically this process takes place over a few days. The second phase, laying down of new bone, is osteoblastic formation. Osteoblasts fill the cavities created by the osteoclasts with new bone. Typically this process takes a few months. Factors such as hormones, calcium, exercise, and other can affect the cells on the surface of bone and trigger the remodeling cycle. [047] An osteoinductive bone matrix is provided by altering the natural collagenous structure of the bone. In some embodiments, the osteoinductive bone matrix is an osteoinductive cancellous bone matrix, thus providing a graft material with desirable physical characteristics and osteoinductive capacity. In one embodiment, this is done by altering the collagenous structure of the bone such that the structure may be at least partially resorbed. In another embodiment, this is done by compacting the trabecular structure of the bone. In vivo, the osteoinductive bone matrix triggers the remodeling cycle. It is to be appreciate that, while the description herein may refer to cancellous bone for illustrative purposes, one skilled in the art would appreciate that the discussion may be applied to cortical, corticocancellous, or composite bone.
[048] II. OVERVIEW OF INCREASING THE
OSTEOINDUCTIVE POTENTIAL OF BONE MATRIX [049] Methods for increasing the biological activity of an at least partially demineralized bone matrix are provided. Osteoinductive osteoimplants are further provided. The osteoinductive osteoimplants comprise at least partially demineralized bone having increased biological activity relative to at least partially demineralized bone that has not been exposed to a treatment or condition as described herein. In some embodiments, the osteoinductive osteoimplants may comprise at least partialy demineralized cancellous bone that has been exposed to a treatment or condition as described herein. The biological activities that may be increased include but are not limited to osteoinductive activity, osteogenic activity, chondrogenic activity, triggering of bone remodeling, 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. U.S. Patent Application No. to Bone Matrix Compositions and Methods, filed June 16,
2008, is herein incorporated by reference in its entirety for the purposes of all that is disclosed therein. 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 formation). Thus, in many instances, chondrogenesis may be considered an early stage of osteogenesis, though of course it may also occur in other contexts.
[050] The at least partially demineralized bone, including wherein the at least partially demineralized bone is at least partially demineralized cancellous bone, provided herein exhibits osteoinductive activity. The osteoinductive at least partially demineralized bone may be prepared by providing at least partially demineralized bone and altering the collagenous structure of the at least partially demineralized bone. Generally speaking, the osteoinductivity of the at least partially demineralized bone may be increased by either (1) altering the structure of the bone such that the trabecular density is increased while the collagen structure is decreased, or (2) by opening up the structure of the bone, thus breaking the collagen apart and making growth factors within the bone more accessible. Thus, in certain embodiments, the at least partially demineralized bone is exposed to a biological or chemical agent or to a combination of agents. The agent may be a cleavage agent, e.g., a protease such as collagenase(s), or a chemical agent such as cyanogen bromide. The matrix may be exposed to multiple treatments either together or sequentially. While not wishing to be bound by any theory, the treatment may alter the primary, secondary, tertiary, and/or quaternary structure of a component of the bone matrix (e.g., collagen, a bone morphogenetic protein, etc.) so as to increase the biological activity of the matrix. An inventive treatment or condition may "open up" the structure of the matrix, e.g., so as to allow biologically active molecules to be more readily released from or diffuse within the matrix and/or to allow components such as nutrients or growth-stimulatory molecules to enter the matrix. In certain embodiments the treatment or condition cleaves proteins present in the DBM (e.g., proteins such as bone morphogenetic proteins), which may result in conversion of an inactive protein into an active form, and/or may generate an active molecule that is less susceptible to degradation than a longer molecule from which it is derived. In certain embodiments, the bone is ground to a generally powder consistency and the powder is reaggregated into a dense structure through compression.
[051] In certain embodiments, it is thought that the at least partially demineralized bone provided herein triggers the remodeling cycle. Specifically, remodeling of bone may be directed to sites that show damage such as microcracks. It is theorized that the body recognizes tissue regions that need repair and then sends cells to repair those regions. Thus, by treating or damaging the at least partially demineralized bone by, for example, treating or damaging the collagen, the at least partially demineralized bone is "marked" as requiring repair. Thus, an implant formed from the cancellous at least partially demineralized bone is marked as requiring repair by cells. In some embodiments, inductive materials may be added to the implant to further stimulate bone remodeling. Alternatively, or additionally, native inductive materials in the bone may be exposed. The bone may be compacted to concentrate the inductive materials (added or native). The implant exhibiting tissue damage and, in some embodiments, increased and/or concentrated inductive material triggers a rapid biologic response.
[052] The treatment or condition may cleave an inhibitory factor that would otherwise inhibit a positively acting agent (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 of the invention, the inhibitory effect of a protein or protein fragment may be reduced by exposing a bone or cartilage matrix to a treatment or condition. The treatment or condition may cause 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 matrix may be exposed to cleave antibody inhibitory agents, or the antibody may be added to the bone matrix.
[053] The osteoinductive at least partially demineralized bone may form an osteogenic and osteoinductive osteoimplant, discussed more fully below. In embodiments wherein the at least partially demineralized bone is at least partially demineralized cancellous bone, the osteoimplant may have desirable characteristics of cancellous bone and yet also have osteoinductive capacity. The osteoimplant, when implanted in a mammalian body, may 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 at least partially demineralized bone may be used as a delivery device to administer bioactive agents.
[054] Altering the Structure of the at least partially demineralized
Bone
[055] In one embodiment, the collagenous structure of the at least partially demineralized bone is disrupted such that the demineralized bone may be at least partially resorbed after implantation. For example, at least partially demineralized bone may be altered so that it has a solid structure at room temperature but substantially liquefies in the body or in tissue culture media such that it may have increased osteoinductive activity when compared to standard at least partially demineralized bone.
[056] Cancellous femoral bone has been shown to contain osteoinductive growth factors that may be extracted from human bone. In one embodiment, cancellous DBM may be treated with, for example, collagenase, which is believed to open the structure of the cancellous DBM to make the growth factors within the bone more bioaccessible. After treatment with collagenase, the osteoinductive cancellous DBM may substantially liquefy in the body and may be at least partially resorbed by the body when implanted. [057] Compacting the Structure
[058] Cortical bone includes dense lamellar structure. The structure of cancellous bone is less dense than that of the lamellar structure of cortical bone. If demineralized bone is considered to be a growth factor source, the growth factor concentration gradients produced by implantation of cancellous and cortical allograft may differ significantly. By compressing the structure of cancellous bone, the osteoinductive potential may be increased. In some embodiments, the structure of cortical bone may be compacted.
[059] Compression of the at least partially demineralized bone may be achieved via any suitable mechanism. For example, compression may be achieved by mechanical means, heat, chemical modification of the bone structure, any suitable type of compression processing, or combinations of these. In one embodiment, the bone is ground or otherwise processed into particles of an appropriate size and formed into a dense bone structure. Grinding may produce bone of a powder like consistency, the powder is wet, and the wet bone powder is smeared to a consistency of fibrous paste. The fibrous paste may be compressed and the paste may be permitted to dry. It is to be appreciated that the bone may be particulated before or after demineralization. Thus, for example, the bone may be particulated and thereafter partially demineralized. Alternatively, the bone may be demineralized and then particulated.
[060] In some embodiments, the bone may be compressed by loading cancellous bone in constrained compression, for example via palletizing the bone, without grinding the bone. Thus, in various embodiments, the at least partially demineralized bone is mechanically compacted into a dense structure. In yet other embodiments, the at least partially demineralized bone is compacted by treatment with chemicals, such as lithium chloride (LiCl), thereby shrinking the collagenous structure.
[061] III. PROVIDING DEMINERALIZED BONE
[062] In some embodiments, demineralized bone that is substantially fully demineralized is used. In other embodiments, partially demineralized bone is used. In other embodiments, the surface demineralized bone is used. In other embodiments, nondemineralized bone may be used. In other embodiments, combinations of some of all of the above may be used. While many of the examples in this section refer to partially or surface demineralized bone, this is for illustrative purposes.
[063] In one embodiment, the bone is partially demineralized. The at least partially demineralized bone 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 may be readily obtained from donor bone by various suitable methods, e.g., as described in U.S. Patent No. 6,616,698, incorporated herein by reference in its entirety. The bone may be of autogenous, allogenic, xenogenic, or transgenic origin.
[064] DBM preparations have been used for many years in orthopaedic medicine to promote the formation of bone. Typically, DBM preparations for promoting the formation of bone have comprised cortical or corticocancellous DBM. DBM has found use, for example, 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. Cortical DBM is thought to promote bone formation in vivo by osteoconductive and osteoinductive processes. The osteoinductive effect of implanted cortical 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-β, IGF, and BMP protein families. Particular examples of osteoinductive factors include TGF-β, IGF-I, 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 also are likely to be other unnamed or undiscovered osteoinductive factors present in DBM. Cancellous DBM is understood to have little or no osteoinductive capacity.
[065] In one demineralization procedure of the present invention, the bone is subjected to an acid demineralization step and a defatting/disinfecting step. The bone is immersed in acid over time to effect demineralization. Acids that may 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. Demineralization may be to a point where the bone is fully demineralized, partially demineralized, or surface demineralized. The depth of demineralization into the bone surface may be controlled by adjusting the treatment time, temperature of the demineralizing solution, concentration of the demineralizing solution, and agitation intensity during treatment.
[066] The at least partially 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. In a particular embodiment, the concentration range of the defatting solution is from about 60 to about 85 weight percent alcohol. In a further embodiment, the defatting solution is about 70 weight percent alcohol. In yet another embodiment, the cancellous bone is defatted in a solution of 1 : 1 chloroform:methanol at room temperature and then demineralized in 0.6 N HCl at 40C.
[067] The at least partially demineralized bone 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, ranges from about 100 to about 3000 microns, or ranges from about 100 to about 800 microns. After grinding the bone to the desired size, the mixture may be sieved to select those particles of a desired size. In certain embodiments, the bone particles may be sieved though a 50 micron sieve, a 75 micron sieve, or a 100 micron sieve. [068] Generally, demineralization conditions may affect the osteoinductivity of DBM. Proteases that degrade the osteoinductive activity of cortical bone have been described. Proteases present in cancellous bone have been shown to differ from those responsible for cortical bone remodeling. Thus, demineralization conditions utilized for cortical bone processing may not be ideal for cancellous bone. Specific protease inhibitor cocktails and lower temperatures may be used.
[069] Following particulation, the bone may be treated to remove mineral from the bone as discussed above. While hydrochloric acid is commonly used as a demineralization agent, there are other methods for preparing DBM, which vary widely and include choices regarding the concentration of the demineralization agent; the temperature and duration of the demineralization step; the inclusion or exclusion at various points in the demineralization process of solvents or solvent combinations such as ethanol, methanol, and chloroform: ether; the extent to which the matrix is washed following the demineralization step; and whether the resulting DBM is stored frozen or is lyophilized and stored at room temperature. See, for example, Russell et al., 22(5) Orthopaedics 524-531 (1999), incorporated herein by reference.
[070] Any of a variety of at least partially demineralized bone preparations may be used with the present invention. 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 No. 6,326,018; Reddi et al., 69 Proc. Natl. Acad. Sci. USA 1601-1605 (1972) ; Lewandrowski et al., 317 Clin. Ortho. ReI. Res. 254-262 (1995) ; Lewandroski et al., 31 J. Biomed. Mater. Res. 365-372 (1996) ; Lewandrowski et al. 61 Calcified Tiss. Int., 294-297 (1997); Lewandrowski et al., 15 I Ortho. Res. 748-756 (1997), each of which are incorporated herein by reference in their entireties. Suitable demineralized bone matrix compositions are described in U.S. Patent No. 5,507,813, hereby incorporated by reference in its entirety. In some instances, large fragments or even whole bone may be demineralized, and then particulated following demineralization.
[071] In some embodiments, the bone may be surface demineralized. The surface may be an inner surface, such as inside trabeculae or inside a Halversion canal. In other embodiments the surface may be an outer surface. In some embodiments, surface demineralized refers to the bone comprising at least one outer surface, or zone of an outer surface, that is demineralized and possessing a non- demineralized core. In some embodiments, the entirety of the surface may be partially demineralized. In other embodiments, a portion of the surface may be demineralized, such as by exposing only a portion of a particle to the demineralization process, by exposing a portion of the surface to a greater or lesser extent of the demineralization process, by masking, etc. Demineralization may be done to a certain percentage. In some embodiments, that percentage relates to weight percentage. In other embodiments, that percentage relates to percentage of the size of the bone being demineralized, or to the depth of demineralization. The depth of demineralization of the at least one outer surface thus may be viewed as a percentage of the size of the bone being demineralized or may be viewed as an absolute number. [072] Demineralization thus may be carried out to a percentage depth of the size of the bone being demineralized. Figures 4a and 4b illustrate surface demineralized bone particles. The bone particle 100 of Figure 2a is substantially spherical. The bone particle 110 of Figure 4b is somewhat elongate. [073] As shown, the bone particle 100 of Figure 2a has a demineralized surface region 106 and a non-demineralized core 108. The bone particle 100 includes a length 102 along its longest dimension and a length 104 along its shortest dimension. The length 102 in the longest dimension comprises first and second demineralized portions 103 a and 103b and a nondemineralized portion 105. A percentage of demineralization in the longest dimension may be determined by summing the length of the first and second demineralized portions 103 a and 103b and dividing that total by the length 102 (comprising 103a, 103b and 105). The length 104 in the shortest dimension likewise comprises first and second demineralized portions 107a and 107b and a nondemineralized portion 109. A percentage of demineralization in the shortest dimension may be determined by summing the length of the first and second demineralized portions 107a and 107b and dividing that total by the length 104 (comprising 107a, 107b and 109). A total percentage demineralization may be determined by averaging the percent demineralization in the longest dimension with the percent demineralization in the shortest dimension.
[074] As shown, the bone particle 110 of Figure 2b has a demineralized surface region 116 and a non-demineralized core 118. The bone particle 110 includes a length 112 along its longest dimension and a length 114 along its shortest dimension. The longest dimension and shortest dimension are taken as those measuring largest and smallest, respectively, such as by a micrometer o r using other by suitable manner and generally going through the center of the bone particle 110. The length 112 in the longest dimension comprises first and second demineralized portions 113a and 113b and a nondemineralized portion 115. A percentage of demineralization in the longest dimension may be determined by summing the length of the first and second demineralized portions 113a and 113b and dividing that total by the length 112 (comprising 113a, 113b, and 115). The length 114 in the shortest dimension likewise comprises first and second demineralized portions 117a and 117b and a nondemineralized portion 119. A percentage of demineralization in the shortest dimension may be determined by summing the length of the first and second demineralized portions 117a and 117b and dividing that total by the length 114 (comprising 117a, 117b, and 119). A total percentage demineralization may be determined by averaging the percent demineralization in the longest dimension with the percent demineralization in the shortest dimension. [075] Alternatively, percentage demineralization may be based on weight percent demineralized of total weight of the bone particle.
[076] In some embodiments, demineralization may be carried out to a depth of, for example, at least about 100 microns. Surface demineralization may alternatively be done to a depth less than or more than about 100 microns. Generally, surface demineralization may be done to a depth of at least 50 microns, at least 100 microns, at least 200 microns, or other. Accordingly, in some embodiments, the demineralized bone comprises at least one outer surface possessing at least one demineralized zone and a non-demineralized core, wherein the demineralized zone of the outer surface of the bone may be, for example, at least about 100 microns thick. The demineralized zone may alternatively be less than or more than about 100 microns thick. The demineralized zone of the surface of the bone is osteoinductive, and therefore promotes rapid new ingrowth of native host bone tissue into an osteoimplant comprising surface demineralized bone. The osteoimplant may comprise surface demineralized monolithic bone or an aggregate of surface demineralized bone particles, and may be substantially solid, fiowable, or moldable. The demineralized zone of the surface of the bone can be any surface portion.
[077] When it is desirable to provide an osteoimplant having improved biological properties while still substantially maintaining the strength present in the osteoimplant prior to demineralization, for example where monolithic bone is used, the extent and regions of demineralization of the monolithic bone may be controlled. For example, depth of demineralization may range from at least about 100 microns to up to about 7000 microns or more, depending on the intended application and graft site. In some embodiments, the depth of demineralization is between 100 to about 5000 microns, between about 150 to about 2000 microns, or between about 200 microns to about 1000 microns. In alternative embodiments, depth of demineralization may be less than about 100 microns. Reference is made to U.S. Patent No. 7,179,299, herein incorporated by reference for discussion of surface demineralization.
[078] A benefit of surface demineralized bone is that the demineralized zone(s) can elastically yield under applied force while the mineralized core has strength and load bearing capacity exceeding that of demineralized bone. Thus, when the surface demineralized bone is subjected to an applied load, the demineralized zones can conform to contours of adjacent bone tissue and thereby minimize voids or spaces between the osteoimplant and adjacent bone tissue. This can be useful because host bone tissue will not grow to bridge large voids or spaces. Thus, by conforming to the contours of adjacent bone tissue, an osteoimplant comprising surface demineralized monolithic bone exhibits enhanced biological properties such as, for example, incorporation and remodeling. The non- demineralized inner core imparts mechanical strength and allows the monolithic osteoimplant to bear loads in vivo. Other non-demineralized zones provide improved tolerances when engaged with other objects such as, for example, insertion instruments, other implants or implant devices, etc. It is noted that some of these characteristics may also be exhibited by an osteoimplant comprising an aggregate of surface-demineralized bone particles.
[079] IV. ENHANCING OSTEOINDUCTIVE PROPERTIES
[080] In certain embodiments, the treatment or condition alters a biological activity of the at least partially demineralized bone such that the at least partially demineralized bone displays osteoinductive, osteogenic, and/or chondrogenic activity in a species in which a control matrix (e.g., an inactivated matrix or a matrix not exposed to the treatment or condition) does not show such activity, or shows it in a lesser amount. For example, at least partially demineralized bone exposed to the treatment or condition may display increased osteoinductive, osteogenic, and/or chondrogenic activity in human, dog, squirrel monkey, etc., as assessed either in vitro or in vivo. Specifically, in some embodiments, cancellous bone, which is understood to have little to no osteoinductivity, as treated herein displays osteoinductive capacity.
[081] Altering the Structure of the at least partially demineralized
Bone
[082] In one embodiment, the structure of the at least partially demineralized bone may be altered to make growth factors within the bone more accessible. Thus, the at least partially demineralized bone may be contacted with a cleavage agent, e.g., a protease such as collagenase(s) or a chemical agent such as cyanogen bromide. The cleavage agents may be applied either together or sequentially, optionally washing the matrix between application of different agents to remove residual agent. In general, the biological and chemical agents may be used in an effective amount and for a time sufficient to achieve a desired outcome, e.g., a desired increase in a biological activity of the matrix. [083] In one embodiment, treatment may be by limited digestion with purified bacterial 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, of Lakewood, NJ. As is 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 tropocollagen is denatured, for example on mild heating, and the polypeptide chains become randomly dispersed. In this state the strands readily may be cleaved by a wide variety of proteases. [084] In the past, digestion of bone with purified bacterial collagenase has been used for altering the solubility of human DBM in cell culture media. Digestion of bone with purified bacterial collagenase has also been used to enhance the response of myoblastic cells to cortical human DBM.
[085] As taught herein, cancellous DBM treated with collagenase has enhanced in vivo osteoinductive capacity. Collagenase treatment of cancellous human DBM increases its solubility relative to that of untreated cancellous human DBM. The solubility of the cancellous DBM may be increased by exposure to an appropriate treatment or condition, e.g. , collagenase treatment, radiation, heat, or any other suitable treatment and/or condition, 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. In some implementations, the cancellous DBM may be partially solubilized by the collagenase, while in others, the cancellous DBM may be completely solubilized by the collagenase. [086] According to various embodiments, a combination of treatments may be used. For example, the partially digested at least partially demineralized bone may be treated with heat or pepsin or another protease to disrupt cross-links not disrupted by the collagenase. The at least partially demineralized bone may be exposed to a variety of biological agents in addition to, or instead of, one or more proteases. Other enzymes include methylases, acylases, lipases, phospholipases, endo- and exo-glycosidases, glycanases, glycolases, amylase, pectinases, galacatosidases, etc. Chemical agents that perform similar reactions may be used. For example, a number of different alkylating agents are known. A variety of salts present in high concentrations (e.g., at least 6 M, 7M, 8M, etc.) may be used. Exemplary salts include salts of various Group I elements, e.g., LiCl. Denaturing agents, e.g., denaturing salts such as guanidinium HCl, may be used. Where denaturing agents are used, care should be taken to avoid denaturing desired components present in the matrix, e.g., growth factors.
[087] A variety of different collagenases known in the art may 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 maybe of eukaryotic (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. Generally, the collagenase cleaves Type I collagen, e.g., degrades the helical regions in native collagen, at the Y-GIy 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.
[088] 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 α2- macroglobulin, a large plasma glycoprotein. Ca2+ is required for enzyme activity. Therefore, it may be desirable to avoid collagenase inhibiting agents when treating at least partially demineralized bone with collagenase. In addition, although the additional proteases present in some collagenase preparations may aid in breaking down tissue, they also may 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%, or 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% may be used. 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, and E-64. Bestatin may also be used, particularly if the preparation contains aminopeptidase activity. Any of these protease inhibitors (or others) may be included in a carrier, such as a bone matrix composition, or in any composition that is used to treat a carrier. [089] As discussed above, collagenase disrupts crosslinks between collagen. Using highly purified collagenase, it is possible that not all the crosslinks are disrupted. Generally, different grades of collagenase may disrupt different ranges of crosslinks. Thus, other treatments, such as pepsin treatment or heat treatment, may be used to disrupt crosslinks not affected by the collagenase. [090] In alternative embodiments, any suitable compound for altering the structure of the DBM may be used. For example, enzymes (e.g., pepsin) or chemicals may be used. Pepsin alters the structure of Type I collagen by cleaving the associated telopeptides. Further, mechanical means such as ionizing radiation or electromagnetic radiation may be used.
[091] Another suitable protease is bone morphogenetic protein 1 (BMP-I).
BMP-I 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 DBM (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, also may be used. It will be appreciated that variants having substantial sequence identity to naturally occurring protease may 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 may be used.
[092] Certain proteases that may provide desirable effects 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 prohormones 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 kDa subtilisin-like catalytic domain, and carboxyl-terminal domain 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 localized in the trans-golgi network, van den Ouweland, A. M. W. et al, 18 Nucl. Acid Res. 664 (1990); Steiner, D. F.,2 Curr. Opin. Chem. Biol. 31-39 (1998). 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., 269 J. Biol. Chem. 25830-25873 (1994). 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, 274 J. Biol. Chem. 23229-23234 (1999). Furin is inhibited by EGTA, αl- antitrypsin Portland, Jean, F. et al., 95 Proc. Natl. Acad. Sci. USA 7293-7298 (1998), and polyarginine compounds, Cameron, A. et al., 275 J. Biol. Chem. 36741-36749 (2000). Furin has been shown to proteolytically process both proTGF and proBMP proteins, for example, proTGF-β and proBMP-4, respectively, resulting in the release of the active mature form for each molecule. Dubois et al., 158(1) Am. J. of Pathology 305-316 (2001); Cui et al., 17(16) Embo Journal 4735-4743 (1998); Cui et al., 15 Genes & Development 2797-2802 (2001), each incorporated by reference herein in their entireties. Furin has also been shown to cleave BMP-2, BMP-6, and BMP-7. For example, furin cleaves between amino acids 282 and 283 in mature human BMP-2. Newly synthesized human 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.
[093] Thus, the at least partially demineralized bone may be treated with
PPCs such as furin and/or other proteases, which process immature TGF-β and/or BMP superfamily propeptides into their active mature forms and/or process active or inactive TGF-β and/or BMP superfamily polypeptides into smaller active fragments that are resistant to degradation or inactivation relative to the longer polypeptide, generating at least partially demineralized bone with increased osteoinductivity compared to at least partially demineralized bone 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 at least partially demineralized bone.
[094] At least partially demineralized bone may be exposed to any of the enzymes, e.g., proteases described herein (and others) at a range of different concentrations, e.g., between 1 pg/ml - 100 μg/ml. For example, a protease maybe used at between 1 pg/ml - 100 pg/ml, between 100 pg/ml and 1 ng/ml, between 1 ng/ml and 100 ng/ml, between 100 ng/ml and 1 μg/ml, between 1 μg/ml and 100 μg/ml, etc. A variety of different digestion buffers may be used. The time of digestion may vary according to the protease, amount of DBM, and desired degree of digestion. In general, suitable times range between 30 minutes to 72 hours, e.g., between 30 minutes to 1 hour, between 1 and 12 hours, between 12 and 24 hours, between 24 and 48 hours, between 48 and 72 hours, etc. It will be appreciated that these times are approximate. Determination of the optimal treatment times for any preparation may involve assay of the treated tissue preparation in one of the biological activity assays described herein or others known in the art. [095] Allogenic cancellous demineralized bone is known not to be osteoinductive. Schwarz et al., 111(1) Arch Orthop Trauma Surg. 47-50 (1991). 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. Thus, the bone, whether cancellous, corticocancellous, or cortical, may be treated for any suitable time period to enhance its osteoinductivity, including from about 15 minute to about 3 hours, from about 30 minutes to about 90 minutes, or for about 60 minutes. 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 at least partially demineralized bone osteoinductive when treated as described herein. Other treatments alternatively may be used and 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.
[096] In addition, the osteoinductive capacity of DBM preparations in higher order species, such as squirrel monkeys, dogs, and humans, has been questioned. For example, allogeneic cancellous bone blocks, demineralized or not, have no osteoinductive capacity and no osteoconductive function that promotes healing of mid-diaphyseal bone defects in dogs. Schwarz et al., 111(1) Arch. Orthop. Trauma Surg. 47-50 (1991). Adult monkey bone matrix contains bone inductive properties, but these properties are not sufficient to induce bone formation in adult monkey muscle sites. Aspenberg et al., 63(6) Acta Orthop Scand. 619-622 (1992); see also Aspenberg et al., 9(1) J. Orthop. Res. 20-5 (1991); Schmid et al., 19(1) Unfallchirurgie 1-8 (1993); Toriumi et al., 1 16(6) Arch Otolaryngol Head Neck Surg. 676-680 (1990); and Ousterhout, DK, 15(5) Ann. Plast Surg. 367-373 (1985). Structural modifications of demineralized bone collagen may serve to activate the latent osteoinductive potential of DBM preparations in higher order species. Such products, irrespective of whether derived from cancellous or cortical bone, may have significantly enhanced clinical efficacy in a variety of orthopaedic applications as observed by more rapid bone healing and spinal fusion. [097] The at least partially demineralized bone may be exposed to a physical condition instead of, or in addition to, a biological or chemical agent. For example, the at least partially demineralized bone may be exposed to heat or cold for a suitable period of time, e.g., minutes, hours, or up to several days, where "heat" refers to temperatures above room temperature (about 23-25 degrees C) and "cold" refers to temperatures below room temperature. Cycles of temperature change may be used, e.g., the at least partially demineralized bone may be heated and cooled a plurality of times. The temperature may, for example, be at least 37 degrees C, at least 40, 50, 60, 70, 80, or 90 degrees C. In some embodiments, the heat treatment may be relatively gentle to avoid denaturing growth factors and other factors, typically proteins or peptides, that contribute to the osteogenic, osteoinductive, or chondrogenic activity of the matrix. The temperature may be 20 degrees C or below, 15 degrees C or below, 10 degrees C or below, 0 degrees C or below, etc. In general, the at least partially demineralized bone may be exposed to any desired temperature in the presence or absence of other agents, solvents, or other suitable media. The at least partially demineralized bone may be exposed to electromagnetic energy of any type, e.g., X-rays and microwaves. Gamma-rays, beta-rays, or any suitable ionizing radiation may be used. The treatment may be performed in the absence of oxygen or in a reduced oxygen environment. U.S. Patent Application
No. to Method of Treating Tissue, filed June 16, 2008, is herein incorporated by reference in its entirety for the purposes of all that is disclosed therein.
[098] In certain embodiments, the treatment or condition alters the physical structure of the matrix so as to increase its biological activity. The treatment or condition may alter the structure of the at least partially demineralized bone so as to facilitate the presentation of such molecules, e.g., on a surface of the at least partially demineralized bone. The treatment or condition may alter the conformation of such molecules in a manner that facilitates interactions with target cells, e.g., cells that migrate towards or into the bone matrix. The treatment or condition may alter release kinetics of agents such as growth factors, differentiation factors, chemotactic factors, etc., from the matrix. Exemplary factors that up-regulate collagen synthesis by osteoblasts include TGF-β, PDGF, IGF, IL-I, PGE2, and certain BMPs. Certain treatments may alter, e.g., increase, the affinity of bone and/or cartilage forming cells and/or undifferentiated cells capable of differentiation into bone and/or cartilage forming cells for the matrix. For example, the treatment or condition may alter integrin binding sites (such as RGD sequences), e.g., by making them more available to cells.
[099] In certain embodiments, alteration of the structure involves cleavage or partial degradation of one or more major structural component of the matrix such as collagen, e.g., components that typically make up at least 1%, 5%, 10%, 25%, 50%, 75%, 90% etc., of the dry weight of the at least partially demineralized bone. In certain embodiments, the secondary, tertiary, and/or quaternary structure of a major structural component of the matrix is altered. The alteration may include destruction of bonds that normally maintain the triple helical structure of collagen, bonds that hold collagen fibrils together, etc. DBM is a dense structure held together by cross-linked collagen. Most of the noncollagenous proteins (NCPs) are trapped within and/or attached to this framework. Certain agents such as collagenase may cut across the framework and thereby potentially allow access to the NCPs. The amount of collagen (or other structural protein) that is cleaved and/or degraded may vary. For example, in certain embodiments, at least 10%, at least 25%, at least 50%, at least 75%, or at least 90% of the collagen originally present in the DBM is cleaved or degraded. Between 10-25%, 25-50%, 50-75%, 75-90%, 90- 100%, or any other suitable range such as 10-90%, 25-75%, etc., of the collagen maybe cleaved or degraded. A polypeptide is considered to be cleaved if it is cleaved at a single site or at multiple sites. In certain embodiments, the cleavage cleaves a crosslink. In certain embodiments, at least a portion of the collagen is present as collagen fragments. For example, at least 10%, at least 25%, at least 50%, at least 75%, at least 90%, etc., of the collagen is present as collagen fragments in certain embodiments. Between 10-25%, 25-50%, 50-75%, 75-90%, 90-100%, or any other range such as 10-90%, 25-75%, etc., of the collagen may be present as collagen fragments. The fragments may remain associated with or present in or on the bone matrix or may diffuse away. A bone matrix may be exposed to any of a variety of different biological or chemical agents or conditions for different time periods in order to achieve a desired degree of cleavage or degradation of a structural component of the matrix such as collagen. The invention therefore provides a modified bone matrix comprising a collagen-containing bone matrix, wherein at least a portion of the collagen is cleaved or degraded. Matrices in which at least a portion of a different structural component of the matrix is cleaved or degraded are also provided.
[0100] In certain embodiments, the at least partially demineralized bone may be 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 domains are capable of functioning independently as peptides and protein fragments. In certain embodiments, the present invention increases the osteoinductive or chondrogenic activity of bone and cartilage matrices 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. [0101] Compacting the Structure
[0102] The at least partially demineralized bone may be compacted to increase the growth factor concentration gradient of the at least partially demineralized bone. Thus, by compressing the structure of bone, the osteoinductive potential may be increased. Compression may be achieved via any suitable mechanism. For example, compression may be achieved by mechanical means, heat, chemical modification of the collagenous structure or any suitable type of compression processing, In one embodiment, the bone is compacted by grinding or otherwise processing the bone into particles of an appropriate size and formed into a dense bone structure . In another embodiment, the bone is compacted by treatment with LiCl, thereby shrinking the structure.
[0103] Cancellous DBM has a sponge-like structure. Accordingly, in some embodiments, use of compression may be particularly suitable for cancellous bone. While the DBM may be compressed, typically when contacted with a liquid the DBM returns to its pre-compressed state. To prevent the DBM from returning to its pre-compressed state, the DBM may be provided in a container for maintaining force on it. Thsu, for example, it may be placed in a defect, mesh, chamber, cage, etc. In accordance with some embodiments, the pressed DBM structure may be specifically configured to retain its shape in water for a predetermined amount of time. For example, the pressed DBM structure may be configured to retain its shape in water for one or more hours, up to seven hours, up to several days, e.g., 48, 72 or 96 hours, or up to one week or several weeks.
[0104] The osteoinductive at least partially demineralized bone may be used in an expandable osteoimplant such as taught by PCT Application No. PCT/US2006/001540, filed January 17, 2006, hereby incorporated by reference in its entirety. As disclosed in that application, an osteoimplant comprising demineralized bone particles has a first state and an expanded state. The osteoimplant may be used with another device or on its own. In the first state, the osteoimplant may be inserted into a device such as an intervertebral body fusion device. The osteoimplant may be rehydrated to expand to an increased size, for example as far as permitted by the confines of the intervertebral body fusion device and spinal endplates, thereby aiding in greater vertebral endplate contact and conformity in spinal surgery. In addition or alternatively, rehydration may take place in vivo. The osteoimplant may comprise fully or partially demineralized cancellous bone and fully and/or partially demineralized cortical bone. In one embodiment, the cancellous bone is provided in chips, and the cortical bone in fibers. The demineralized cancellous bone may comprise, in whole or part, osteoinductive cancellous DBM as taught herein. In another embodiment, the expandable osteoimplant may comprise, in whole or in part, cancellous DBM, some or all of which may be made osteoinductive pursuant to the present invention. In yet a further embodiment, the expandable osteoimplant may comprise a monolithic piece of bone treated as provided herein, such as by at least partially demineralizing and treating the monolithic piece of bone.
[0105] In one embodiment, the compressed at least partially demineralized bone is provided by grinding the bone, for example into a fine powder, and molding it into a cohesive shape. The bone may be wetted and processed to a consistency of a fibrous paste before molding. In one embodiment, the molding is done via hand molding and drying the material at ambient temperature. In another embodiment, the molding is done by putting the bone in a mold, such as a polyurethane or PTFE mold, putting the mold under pressure, for example by clamping the mold, and allowing the bone to dry, for example via vacuum drying the material at 4O0C for 72 hours. In yet another embodiment, the molding is done by putting the bone into a stainless steel mold, pressing the mold with 500 pounds and vacuum drying the mold at 4O0C for 72 hours with weighted dehydrothermal plungers. [0106] Thus, in various embodiments, the DBM may be ground to a powder- like consistency, processed into a fibrous paste, and the fibrous paste compressed and dried. Compression and drying may be done in any suitable manner. For example, compression may be done by applying manual pressure, by clamping (powered manually, through dehydrothermal plungers, or other), or other means. Drying may be done in air at ambient temperature, in an oven, vacuum dried, or in any other suitable manner.
[0107] In another embodiment, the at least partially demineralized bone is compacted by treatment with LiCl to shrink the collagenous structure. Specifically, at least partially demineralized bone may be treated with 8M LiCl to increase the osteoinductivity of the at least partially demineralized bone. Treatment with LiCl may further increase the radioopacity of the at least partially demineralized bone. [0108] V. OSTEOIMPLANT
[0109] Bone grafting applications are differentiated by the requirements of the skeletal site. Certain applications generally use a "structural graft" in which one role of the graft is to provide mechanical or structural support to the site. Such grafts contain a substantial portion of mineralized bone tissue to provide the strength needed for load-bearing. Examples of applications requiring a "structural graft" include intercalary grafts, spinal fusion, joint plateaus, joint fusions, large bone reconstructions, etc. Other applications generally use an "osteogenic graft," in which one role of the graft is to enhance or accelerate the growth of new bone tissue at the site. Such grafts contain a substantial portion of demineralized bone tissue to improve the osteoinductivity needed for growth of new bone tissue. Examples of applications requiring "osteogenic graft" include deficit filling, spinal fusions, joint fusions, etc. Grafts may also have other beneficial biological properties, such as, for example, serving as delivery vehicles for bioactive substances. Bioactive substances include physiologically or pharmacologically active substances that act locally or systemically in the host.
[0110] In accordance with various embodiments, the at least partially demineralized bone matrix provided herein may be used as a structural graft, an osteogenic graft, or a graft suitable for both structural and osteogenic uses. [0111] Any suitable shape, size, and porosity of at least partially demineralized bone may be used. In various embodiments, the bone may be monolithic or may be composite. Rat studies show that the new bone is formed essentially having the dimensions of the device implanted. A successful osteoimplant for encouraging bone development appropriately accommodates each step of the cellular response during bone development, and in some cases, protects osteoinductive factors from nonspecific proteolysis. In some uses, the osteoimplant acts as a temporary scaffold until replaced completely by new bone. In bone, the dissolution rates may vary according to whether the implant is placed in cortical or trabecular bone.
[0112] The osteoimplant resulting from the at least partially demineralized bone may assume a determined or regular form or configuration such as a sheet, plate, disk, tunnel, cone, tube, a rod, string, weave, solid, fiber, or wedge, to name but a few. Prefabricated geometries include, but are not 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, wave plates. Partial tubular as well as flat plates may be fabricated from the osteoimplant. Such plates may include such conformations as, e.g., concave contoured, bowl shaped, or defect shaped. [0113] In certain embodiments, the shape and size of the particles in the carrier affect the time course of osteoinductivity. For example, in a cone or wedge shape, the tapered end will result in osteoinductivity shortly after implantation of the osteoimplant, whereas the thicker end will lead to osteoinductivity later in the healing process (hours to days to weeks later). Also, larger particle size will have induce bone formation over a longer time course than smaller particles. Particles of different characteristics (e.g., composition, size, shape) maybe 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, herein incorporated by reference in its entirety, for suitable examples. In a weave, strands composed of short half-life particles may be woven together with strands of longer half-lives. [0114] The osteoimplant may be machined or shaped by any suitable mechanical shaping means. Computerized modeling may provide for the intricately- shaped three-dimensional architecture of an osteoimplant custom-fitted to the bone repair site with precision.
[0115] Thus, the at least partially demineralized bone may optionally be subjected to a configuring step to form an implant. In an embodiment wherein the bone is compressed to enhance osteoinductivity, the configuring step may be done during compression. The configuring step may 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 may 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, the contents of which are incorporated by reference herein in their entireties. Suitable sheets include those sold under the trade name Grafton® 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 human 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 Hydroxy apatite, B etatricalcium Phosphate and Demineralized Bone Matrix, 24(15) Biomaterials 2593-2603 (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, glycosaminoglycan proteins, nucleic acids, polymers, polaxomers, resins, clays, calcium salts, and/or derivatives thereof. See, for example, U.S. Patent No. 5,290,558, incorporated by reference herein in its entirety. [0116] The bone used in creating the at least partially demineralized bone may be obtained from any source of living or dead tissue. In many instances, the source of bone may be matched to the eventual recipient of the inventive composition. At a minimum, it is often desirable that the donor and recipient are of the same species, though even xenogenic sources are permitted. Thus, for use in humans, generally DBM derived at least in part from human bone may be used. For example, 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.
[0117] In one embodiment, the osteoimplant induces endochondral bone formation reliably and reproducibly in a mammalian body. The at least partially demineralized bone osteoimplant 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 may be within the range of 70 μm-850 μm, or 70 μm-420 μm, or 150 μm-420 μm, though any suitable size may be used. 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 in some embodiments 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. [0118] VI. FORMULATION
[0119] The osteoinductive at least partially demineralized bone 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 DBM. 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 health, and the risk of infection. In various embodiments, the at least partially demineralized bone may comprise, for example, less than approximately 0.5% water, less than approximately 1% water, or less than approximately 5% water. [0120] The osteoinductive at least partially demineralized bone may be configured to be suturable. Thus, in certain embodiments, the osteoinductive at least partially demineralized bone or an osteoimplant formed from the osteoinductive at least partially demineralized bone may be sutured in place in vivo. [0121] An osteoimplant formed from osteoinductive cancellous DBM may be configured for expansion. Cancellous DBM has a sponge-like texture and expands and conforms. Thus, an osteoimplant may be formed that is able to expand and conform to a site in vivo. The osteoimplant so formed may comprise monolithic cancellous bone or particulated (and reaggregated) cancellous bone. [0122] Osteoinductive at least partially demineralized bone or osteoimplants 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 bone particles of a particular size or composition, combined with the selection of a particular stabilizing agent or agents and the amounts of such agents.
[0123] Physical properties such as deformability and viscosity may also be chosen depending on the particular clinical application. The particles of bone may be mixed with other materials and factors to improve other characteristics of the implant. For example, the at least partially demineralized bone 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.
[0124] In another embodiment, an osteoimplant having a pre-selected three- dimensional shape is prepared by repeated application of individual layers of DBM, for example by 3-D printing as described by U.S. Patent Nos. 5,490,962, 5,518,680, and 5,807,437, each incorporated herein by reference in their entireties. Different layers may comprise individual stabilized DBM preparations, or alternatively may comprise DBM layers treated with stabilizing agents after deposition of multiple layers.
[0125] In the process of preparing improved inventive bone 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), super critical CO2 treatment, thermal sterilization, or combinations thereof. Other methods known in the art of preparing bone and cartilage matrices, such as defatting, sonication, lyophilization, and combinations thereof may also be used. Since the biological activity of various materials including demineralized bone is known 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. [0126] VII. OPTIONAL TREATMENTS
[0127] In other embodiments, the present invention provides methods of increasing the osteoinductivity of the at least partially demineralized bone by further exposing the at least partially demineralized bone to at least one treatment (e.g., a biological or chemical agent). In addition to treatment with collagenase (or other suitable compound) or compaction, the at least partially demineralized bone 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 at least partially demineralized bone has an increased osteoinductivity, osteogenic, or chondrogenic activity compared to a similar at least partially demineralized bone 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). [0128] Generally, it may be desirable to remove the inhibitors quickly without denaturing the osteoinductive factors. As will be appreciated by those skilled in the art, factors having osteoinductive, osteogenic, and/or chondrogenic activity may be inhibited by a variety of mechanisms including proteolytic degradation, binding or sequestration of the factor, etc. [0129] VIII. OPTIONAL ADDITIVES
[0130] Optionally, other additives may be included in the osteoinductive at least partially demineralized bone. It will be appreciated that the amount 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 at least partially demineralized bone. The desired amount is readily determinable by the user. Any of a variety of medically and/or surgically useful optional substances may be incorporated in, or associated with, the osteoinductive factors either before, during, or after preparation of the at least partially demineralized bone. [0131] In certain embodiments, the additive is adsorbed to or otherwise associated with the osteoimplant. The additive may be associated with the osteoimplant 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 Waals 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 DBM. In another embodiment, the additive is attached to an antibody, or fragment thereof, that recognizes an epitope found within the DBM. An additive may be provided within the osteoimplant in a sustained release format. For example, the additive may be encapsulated within biodegradable nanospheres, microspheres, or the like. [0132] 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. [0133] Osteoinductive Factors
[0134] Osteoinductive factors may be added to the treated at least partially demineralized bone, thus further increasing the osteoinductivity of the at least partially demineralized bone. U.S. Patent Application No. 11/555,608, entitled Bone Matrix Compositions and Methods, filed November 1 , 2006, which is herein incorporated by reference in its entirety, teaches suitable methods for adding osteoinductive factors to the at least partially demineralized bone and for otherwise enhancing the osteoinductivity of the bone.
[0135] Osteoinductive factors include any agent that leads to or enhances the formation of bone. The osteoinductive factors may do this in any manner. For example, the osteoinductive factors may lead to the recruitment of cells responsible for bone formation, the osteoinductive factors may lead to the secretion of matrix which may subsequently undergo mineralization, the osteoinductive factors may lead to the decreased resorption of bone, etc. Suitable osteoinductive factors include bone morphogenic proteins (BMPs), transforming growth factor (TGF-O), insulin- like growth factor (IGF-I), parathyroid hormone (PTH), and angiogenic factors such as VEGF. In one embodiment, the osteoinductive factors is genetically engineered to comprise an amino acid sequence which promotes the binding of the inducing agent to the DBM or the carrier. Sebald et al, PCT/EPOO/00637, incorporated herein by reference in its entirety, describe the production of exemplary engineered growth factors suitable for use with DBM.
[0136] Other osteoinductive factors also may be added to the at least partially demineralized bone. These osteoinductive factors may be added in an activated or nonactivated form, and they may be added at anytime during the preparation of the inventive material. For example, the osteoinductive factors may be added after the demineralization step and prior to the addition of the stabilizing agents so that the added osteoinductive factors are protected from exogenous degrading enzymes once implanted. In some embodiments, the at least partially demineralized bone is lyophilized in a solution containing the osteoinductive factors. In certain other embodiments, the osteoinductive factors are adhered onto the hydrated demineralized bone matrix and are not freely soluble. In other instances, the osteoinductive factors are added after addition of a stabilizing agent so that the osteoinductive factors are available immediately upon implantation of the at least partially demineralized bone.
[0137] Radiopaque Substances
[0138] Radiopaque substances may be added to impart radiopacity to the at least partially demineralized bone. 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 1 to about 25 weight percent of the bone particle containing composition, calculated prior to forming the shaped material. In some embodiments, the at least partially demineralized bone may have some inherent radiopacity due to remaining mineralization. [0139] Angiogenesis Promoting Materials
[0140] Development of a vasculature around the implant site also may help form new bone and/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., 424 Nature 391-397 (2003), incorporated herein by reference in its entirety, and may be included in the osteoimplant.
[0141 ] Bioactive Agents
[0142] The osteoconductive at least partially demineralized bone 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 bioactive agents that may be delivered using an at least partially demineralized bone composition include agents that promote the natural healing process, i.e., resorption, vascularization, angiogenesis, new growth, or the like. In one embodiment, the osteoimplant is provided in which the treated at least partially demineralized bone, 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 at least partially demineralized bone may be used to deliver more than one bioactive agent, more than two, and sometimes more than three bioactive agents. The bioactive agent may be associated with the at least partially demineralized bone. For example, the bioactive agent may be associated with the at least partially demineralized bone through electrostatic interactions, hydrogen bonding, pi stacking, hydrophobic interactions, van der Waals interactions, etc. In certain embodiments, the bioactive agent is attached to the at least partially demineralized bone 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 at least partially demineralized bone through non-specific interactions (e.g., hydrophobic interactions).
[0143] Medically/surgically useful substances include physiologically or pharmacologically active substances that act locally or systemically in the host. Generally, these substances may include bioactive substances which may be readily incorporated into the osteoimplant and include, e.g., demineralized bone powder as described in U.S. Patent No. 5,073,373, the contents of which are incorporated herein by reference in its entirety; 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, bacitracin, neomycin, penicillin, polymycin B, tetracyclines, biomycin, Chloromycetin, and streptomycins, cefazolin, ampicillin, azactam, tobramycin, clindamycin and gentamycin, etc.; biocidal/biostatic sugars such as dextran, 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, collagenase, peptidases, oxidases, etc.; polymer cell scaffolds with parenchymal cells; angiogenic agents and polymeric carriers containing such agents; collagen lattices; antigenic agents; cytoskeletal agents; cartilage fragments; living cells such as chondrocytes, bone marrow cells, mesenchymal stem cells; natural extracts; genetically engineered living cells or otherwise modified living cells; expanded or cultured cells; DNA delivered by plasmid, viral vectors or other means; tissue transplants; demineralized bone powder; autogenous tissues such as blood, serum, soft tissue, bone marrow, etc.; bioadhesives; BMPs; osteoinductive factor (IFO); fϊbronectin (FN); endothelial cell growth factor (ECGF); vascular endothelial growth factor (VEGF); cementum attachment extracts (CAE); ketanserin; human growth hormone (HGH); animal growth hormones; epidermal growth factor (EGF); interleukins, e.g., interleukin-1 (IL-I), interleukin-2 (IL-2); human alpha thrombin; transforming growth factor (TGF-beta); IGF-I and 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; immunosuppressants; 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 may vary widely with optimum levels being readily determined in a specific case by routine experimentation.
[0144] 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, an 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. In 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 at least partially demineralized bone 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-β, IGF, parathyroid hormone (PTH), growth factors, or angiogenic factors. In certain embodiments at least two bioactive agents are attached to the at least partially demineralized bone composition. In other embodiments at least three bioactive agents are attached to the at least partially demineralized bone composition.
[0145] IX. EXAMPLES
[0146] Aspects of the present teachings may be further understood in light of the following examples, which should not be construed as limiting the scope of the present teachings in any way. [0147] Example 1
[0148] Preparation of Cancellous Dowels. Corticocancellous dowels were drilled from the femoral condyle of a human donor using 12 mm Codman dowel cutter. Any attached cartilage or cortical bone was removed, leaving only dense and diffuse cancellous structures. The bone was drilled while in a frozen state and the drilled dowels were immediately placed in a cold solution of 0.2 mM, 1 mM NaN3, and 0.1 mM Benzamidine HCl (40C). The bones were removed after two hours on ice. In order to remove the bone marrow elements, the dowels were lavaged using semi-gentle (nondisruptive to cancellous structure) cool water. The material was stored at -700C until defatting and demineralization.
[0149] Defatting and Demineralization. The dowels were defatted in a solution of 1 :1 chlorofornrmethanol for 15 hours at room temperature. The solution was then poured off and any organic residue was allowed to evaporate from the bone for 6 hours under a fume hood. The cancellous matrix was demineralized in 0.6 N HCl at 40C overnight and washed extensively with water prior to lyophilization. Specifically, the following protocol was followed:
1. Extraction with 400 ml 1 : 1 chloroform:methanol for 15 hours at room temperature.
2. Evaporate 6 hours under fume hood.
3. 300 ml 0.6 N HCl at 40C overnight.
4. Change acid, remove air bubbles by pressing cancellous structure, continue demineralization at 40C for 6 hours.
5. Wash with water using 800 ml at 40C. Perform 4 times for 10 minutes each.
6. Lyophilize two dowels not treated with collagenase (one large, one small) overnight -30, 230C. 3 days. The untreated dowels are control dowels.
[0150] Collagen Denaturation. The collagen structure of two dowels for treatment with collagenase was gelatinized by the following sequence:
1. Extract 1 hour, 2 M CaCl2 40C (400 ml).
2. Wash 3x10 minutes using cold water.
3. Extract 1 hour 0.5 M EDTA pH 7.4 at 40C (400 ml).
4. Wash 3x10 minutes using cold water.
5. Extract 8 M LiCl overnight at 40C (400 ml).
6. Wash 1 hour using 400 ml water at 40C.
7. Wash with 300 ml sterile water at 550C for 90 minutes.
8. Lyophilize after freezing. [0151] The dowels so treated are referred to as bone matrix gelatin (BMG) samples.
[0152] Preparation of Samples. Each of the dowels were cut to produce 2 long DBM of approximately 100 mg each, 2 short DBM of approximately 50 mg each, 2 long BMG of approximately 100 mg each, and 2 short BMG of approximately 60 mg each
[0153] Each sample was washed and rehydrated in enough Alloprep to cover the sample (approximately 750 μl).
[0154] Collagenase Treatment. Four samples were treated with collagenase.
Two samples were BMG samples. Two samples were DBM samples.
[0155] Materials:
Digestion buffer (50 mM Tris-HCl pH 7.4, containing 5 niM CaCl2, sterile filtered);
Chromatographically purified collagenase (Worthington CLSPA; Source: Clostridium Histolyticum; 10,000 units); and
0.1N Glacial acetic acid (Add 248.56 ml of deionized water to 1.44 ml of 17.4 M Glacial Acetic Acid, Sterile filtered).
[0156] Generally, collagenase treatment comprised placing sections of demineralized cancellous dowels weighing between 50 and 100 mg in a 5 ml digestion buffer containing 80 units/ml collagenase. The samples were incubated at 370C for 90 minutes. Each sample was washed for 30 minutes with 30 ml 0.1N acetic acid in the cold. The samples were then rinsed for 10 minutes with phosphate buffered saline ("PBS") and 10 minutes with deionized water prior to lyophilization. The specific protocol was as follows:
1. Cut 2 cancellous BMG pieces, one from the BMG-long which weighed 100 mg and one from the BMG-short which weighed 60 mg.
2. Cut 2 cancellous DBM pieces, one from the DBM-long which weighed 100 mg and one from the DBM-short which weighed 50 mg.
3. Add 4.8 ml digestion buffer to each sample.
4. Add 200 μl of stock collagenase to each sample.
5. Vortex each sample.
6. Incubate each sample for 1.5 hours in a 37 0C water bath.
7. Add 30 ml 0.1N glacial acetic acid to each sample. Wash for 1 hour at 40C using a magnetic stir bar and plate.
8. Pour off the glacial acetic acid.
9. Wash with 45 ml PBS for 10 minutes at 4 0C. Pour off PBS. Repeat.
10. Wash with 45 ml deionized water for 10 minutes at 4 0C. Pour off deionized water. 11. Press samples into a syringe and freeze-dry using a 24 hour cycle. [0157] Implantation. The following pieces were implanted, aiming for a range of 10-20 mg (all samples were implanted in athymic rats except for those where mouse is indicated):
Collagenase short-BMG Collagenase long-BMG
9 mg 14 mg
8 mg 17 mg
12 mg 18 mg
14 mg (mouse)
Collagenase short-DBM Collagenase long-DBM
7 mg 11 mg
9 mg 17 mg
18 mg 18 mg
9 mg (mouse)
Control Short BMG Control Long BMG
10 mg 10 mg
11 mg 10 mg
12 mg 11 mg
Control Short DBM Control Long DBM
10 mg 10 mg
10 mg 11 mg
11 mg 12 mg
[0158] Assay. The osteoinductive potential of collagenase treated and untreated (control) cancellous bone samples was evaluated by implanting the materials intramuscularly in athymic rats and an athymic mouse. After 28 days the explants were evaluated histologically and radiographically for evidence of heterotopic bone formation.
[0159] Results. Figure 1 is a graph illustrating the results of Example 1. As shown, the collagenase treated DBM had higher histologic scores for both the long and short samples than did the control DBM. The long collagenase treated BMG sample had a lower histologic score than did the long control BMG sample. The short collagenase treated BMG sample had a higher histologic score than did the short control BMG sample. [0160] The results demonstrate that collagenase treatment enhances the osteoinductive activity of cancellous DBM scaffolds. More specifically, the results indicate that the osteinductive activity of cancellous DBM scaffolds, as measured by the histologic score, was measurable, and increased above that of untreated DBM, in all samples. The increase may be noted both histologically and radiographically. Collagenase treated human demineralized cancellous scaffolds have potent osteoinductive activity as measured by heterotopic bone formation in athymic rats. The negligible osteoinductive activity of human cancellous demineralized scaffolds in athymic mice may be markedly enhanced by treatment with bacterial collagenase, without requiring treatment with LiCl prior to treatment with bacterial collagenase. The activity of these preparations is comparable to that of human cortical DBM. [0161] Example 2
[0162] Cancellous bone was demineralized in 0.6 N HCl at room temperature. The demineralized bone was washed, and the wet demineralized bone was smeared to a consistency of a fibrous paste. The fibrous paste was then treated using one of three methods:
1. Hand manipulated with slight pressure, dried at ambient temperature for approximately 72 hours;
2. Placed in a PTFE mold and compressed between plungers. The loaded hand clamped mold was placed in a vacuum oven and dried at 4O0C for approximately 72 hours;
3. Loaded into a mold and compressed between stainless steel dehydro thermal plungers. Initially, 500 pounds of force was applied to the plungers to press out excess water and compress the paste. After initial compression, dead weights were applied to continue to keep the paste under compression while a vacuum was drawn through the dehydrothermal plunters. During vacuum dryin g(via the plungers), the mold was kept at approximately 4O0C for 72 hours.
After processing, each sample was placed in a beaker containing room temperature deionized water, and observed for signs of disassociation.
Figure 3 illustrates the formulations thus produced. Formulation 200 corresponds to method 1, formulation 202 corresponds to method 2, and formulation 204 corresponds to method 3. Each of the formulations 200, 202, and 204 were submerged in deionized water and observed for cohesiveness. Figure 4 illustrates formulation 200 at 7 hours of submersion in water. Figure 5 illustrates formulation 202 submerged in water for 72 hours. Figure 6 illustrates formulation 204 submerged in water for 72 hours. From Figure 4, formulation 200 swelled when submerged in water but did not dissociate. From Figures 5 and 6, formulations 202 and 204 each generally retained their shape in water.
[0163] X. ASSESSMENT OF OSTEOGENIC ACTIVITY
[0164] Induction of bone formation may 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 may 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, osteoclasts, and osteoid matrix. A suitable procedure for assessing osteoinductive activity is illustrated in Example 5 of U.S. Patent No. 5,290,763 ("the '763 patent"), herein incorporated by reference in its entirety. 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 example 1 of U.S. Patent No. 5,563,124, herein incorporated by reference in its entirety. The 0-4 portion of this scale corresponds to the scoring system described in the '763 patent, 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.
[0165] Human DBM has not been proven to have osteoinductive activity in higher order species. Studies indicate that dog DBM is osteoinductive when tested in athymic rats but not in dogs. Further, studies have been published stating that BMP -2 has osteoinductive activity in squirrel monkeys but bone matrix does not. Collagenase treated DBM slowly solubilizes in tissue culture media (in the presence of cells) at 37°C. Without being bound by theory, it is possible that the lack of osteoinductive activity of DBM in higher species may be related to the failure of DBM to present growth factors to the host in an efficient manner. Collagenase treatment provides a DBM capable of osteoinductive activity in higher species. Such formation may serve as a basis for eliminating the need for autograft in many orthopaedic applications, including posterior spinal fusion. [0166] XI. USES
[0167] 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 may be utilized in a wide variety of orthopaedic, periodontal, neurosurgical, and oral and maxillofacial surgical procedures. [0168] Demineralized cancellous bone has a sponge-like structure. The material may be compressed significantly and will substantially regain its original structure in the absence of the compressive force. An osteoinductive cancellous scaffold may be forced into a space or defect through a small opening. The structure will then expand and conform to the shape of the environment. This characteristic may be particularly useful in embodiments wherein the collagenous structure has been altered, for example, by treatment with collagenase. Thus, in one application, an osteoinductive cancellous scaffold is placed into a compromised vertebral element in a procedure similar to vertebroplasty. It is expected that the cancellous scaffold would expand in the collapsed vertebrae and, over time, new bone formed in the interior of the vertebral element would restore its structural integrity. The procedure could be used in conjunction with or as a replacement for Kyphoplasty. [0169] In addition or alternatively, demineralized cancellous bone may be ground into a particulate and formed into a compressed implant having a denser structure compared to the implant described above. The dense implant may be used alone or in combination with implant scaffolding. The implant may expand in vivo over the course of between a few hours to a few weeks depending on, for example, the density of the implant, other materials incorporated into the compressed implant, the size and shape of the implant, or other aspects of the implant that would be readily understandable by those skilled in the art.
[0170] 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., may be combined with the osteoimplant. The osteoimplant may 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.
[0171] Activated scaffolds comprising the osteoinductive cancellous DBM may reduce or eliminate the need for harvesting iliac crest autograft in challenging orthopaedic applications such as posterolateral spinal fusion and the treatment of critical sized nonunion fractures.
[0172] The osteoinductive at least partially demineralized bone may also be used as a drug delivery device. Demineralized cancellous bone has the capacity to absorb significant amounts of fluids, and it has a high surface area to volume ratio. These properties indicate that the material would be an ideal growth scaffold for bioactive cells. Osteoinductive at least partially demineralized bone may be used as a carrier for osteogenic cells, chrondrogenic cells, stem cells, bone marrow aspirates, fat cells, and other cell types that are involved in tissue formation or that have the capability to differentiate into tissue forming cells when exposed to appropriate signals. The at least partially demineralized bone may also serve as a carrier for a variety of native or recombinant growth and differentiation factors. [0173] Collagenase treatment of at least partially demineralized bone may have further applications. For example, cells harvested from a recipient may be treated with collagenase treated at least partially demineralized bone in a manner that induces their differentiation into bone, cartilage, or other types of tissues such as bone marrow, neural cells, or vasculature. The tissues/organs could then be transplanted into the recipient after they have developed to a certain point in vitro. In addition, at least partially demineralized bone contains a variety of growth factors. The type of growth factors harvested, and the manipulation of cell culture conditions or specific growth factor levels in at least partially demineralized bone, may be optimized for the tissue of interest.
[0174] In certain embodiments, association with the osteoinductive at least partially demineralized bone increases the half-life of the relevant biologically active agent(s). Certain inventive drug delivery devices are used to deliver osteoinductive growth factors. Other agents that may be delivered include factors or agents that promote wound healing. However, the osteoinductive at least partially demineralized bone may alternatively or additionally be used to deliver other pharmaceutical agents including antibiotics, anti-neoplastic agents, growth factors, hematopoietic factors, nutrients, an other bioactive agents described above. The amount of the bioactive agent included with the at least partially demineralized bone 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.
[0175] XII. CONCLUSION
[0176] In certain embodiments, the osteoinductive at least partially demineralized bone 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 at least partially demineralized bone that has not been exposed to a treatment or condition as described herein. Of course, one skilled in the art will appreciate that these values may vary 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 osteoimplant 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, hi some embodiments, when an osteoimplant comprising osteoinductive cancellous DBM is implanted in a bone defect site, the osteoimplant has an osteoinductivity score comparable to a cortical DBM implant that has not been treated.
[0177] Although the invention has been described with reference to various embodiments, persons skilled in the art will recognize that changes may be made in form and detail without departing from the spirit and scope of the invention.

Claims

CLAIMSWhat is claimed is:
1. A method for forming an osteoinductive composition comprising: providing bone matrix; at least partially demineralizing the bone matrix; and treating the at least partially demineralized bone matrix with a chemical agent to impart or enhance osteoinductive activity of the at least partially demineralized bone matrix.
2. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with a collagenase preparation.
3. The method of claim 2, wherein the at least partially demineralized bone matrix is substantially completely solubilized by the collagenase preparation.
4. The method of claim 2, wherein the at least partially demineralized bone matrix is partially solubilized by the collagenase preparation.
5. The method of claim 4, further comprising treating the partially solubilized at least partially demineralized bone matrix to disrupt cross-links not disrupted by the collagenase preparation.
6. The method of claim 5, wherein the further treatment comprises treatment with heat.
7. The method of claim 5, wherein the further treatment comprises treatment with pepsin.
8. The method of claim 2, wherein the collagenase preparation is a eukaryotic collagenase.
9. The method of claim 2, wherein the collagenase preparation is a prokaryotic collagenase.
10. The method of claim 2, wherein the collagenase preparation is highly purified.
11. The method of claim 2, wherein the collagenase preparation is substantially free of bacterial components.
12. The method of claim 2, wherein the collagenase preparation further comprises at least one protease inhibitor.
13. The method of claim 1 , wherein providing bone matrix comprises providing cancellous bone.
14. The method of claim 13, further comprising compacting the at least partially demineralized cancellous bone.
15. The method of claim 14, wherein the at least partially demineralized cancellous bone is compacted mechanically.
16. The method of claim 1, wherein treating the at least partially demineralized cancellous bone compacts the at least partially demineralized cancellous bone.
17. The method of claim 14, wherein compacting the at least partially demineralized cancellous bone is done by chemical modification of a structure of the at least partially demineralized cancellous bone.
18. The method of claim 14, wherein compacting the at least partially demineralized cancellous bone comprises loading the at least partially demineralized cancellous bone in a constrained compression.
19. The method of claim 18, wherein loading the at least partially demineralized cancellous bone matrix in a constrained compression comprises palletizing the at least partially demineralized cancellous bone.
20. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises altering a structure of the at least partially demineralized bone structure such that trabecular density is increased while collagen structure is decreased.
21. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises opening up a structure of the at least partially demineralized bone matrix and breaking apart collagen within the at least partially demineralized bone matrix.
22. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with an enzyme.
23. The method of claim 22, wherein treating the at least partially demineralized bone matrix with an enzyme comprises treating the at least partially demineralized bone matrix with a protease.
24. The method of claim 23, wherein treating the at least partially demineralized bone matrix with a protease comprises treating the at least partially demineralized bone matrix with a protease of the proprotein convertase family of proteases.
25. The method of claim 22, wherein treating the at least partially demineralized bone matrix with an enzyme comprises treating the at least partially demineralized bone matrix with pepsin.
26. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with bone morphogenetic protein- 1.
27. The method of claim 26, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with cyanogen bromide.
28. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with a cleaving agent.
29. The method of claim 28, wherein treating the at least partially demineralized bone matrix with a cleaving agent cleaves an inhibitory factor.
30. The method of claim 29, wherein the cleaving agent cleaves an antibody inhibitory agent.
31. The method of claim 29, wherein the cleaving agent cleaves a protein that inhibits osteoinductive and/or osteogenic activity.
32. The method of claim 1 , further comprising treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with heat.
33. The method of claim 32, wherein treating the at least partially demineralized bone matrix further comprises treatment with cold.
34. The method of claim 1 , further comprising treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with cold.
35. The method of claim 1 , wherein treating the at least partially demineralized bone matrix inhibits synthesis of an agent that inhibits the biological activity of the partially demineralized bone matrix.
36. The method of claim 1, wherein treating the at least partially demineralized bone matrix inhibits secretion of an agent that inhibits the biological activity of the partially demineralized bone matrix.
37. The method of claim 1, wherein treating the at least partially demineralized bone matrix inhibits post-translational modification of an agent that inhibits the biological activity of the partially demineralized bone matrix.
38. The method of claim 1, wherein treating the at least partially demineralized bone matrix inhibits transport of an agent that inhibits the biological activity of the partially demineralized bone matrix.
39. The method of claim 1 , further comprising treating the at least partially demineralized bone matrix with ionizing radiation.
40. The method of claim 39, wherein treating the at least partially demineralized bone matrix with ionizing radiation comprises treating the at least partially demineralized bone matrix with gamma rays.
41. The method of claim 39, wherein treating the at least partially demineralized bone matrix with ionizing radiation comprises treating the at least partially demineralized bone matrix with beta rays.
42. The method of claim 1 , further comprising treating the at least partially demineralized bone matrix with electromagnetic radiation.
43. The method of claim 42, wherein treating the at least partially demineralized bone matrix with electromagnetic radiation comprises treating the at least partially demineralized bone matrix with x-rays.
44. The method of claim 42, wherein treating the at least partially demineralized bone matrix with electromagnetic radiation comprises treating the at least partially demineralized bone matrix with microwaves.
45. The method of claim 1 , wherein treating the at least partially demineralized bone matrix comprises compacting the at least partially demineralized bone matrix with or in the presence of the chemical agent.
46. The method of claim 45, wherein compacting the at least partially demineralized bone matrix with a chemical agent comprises compacting the at least partially demineralized bone matrix with or in the presence of lithim chloride.
47. The method of claim 1, further comprising compacting the at least partially demineralized bone matrix.
48. The method of claim 45, wherein the at least partially demineralized bone matrix is compacted manually.
49. The method of claim 45, wherein the at least partially demineralized bone matrix is compacted with heat.
50. The method of claim 1 , further comprising particulating the at least partially demineralized bone matrix and compacting the particulated at least partially demineralized bone matrix.
51. The method of claim 50, wherein particulating the at least partially demineralized bone matrix is done to a generally powder consistency and provides a powder.
52. The method of claim 51 , wherein compacting the at least partially demineralized bone matrix reaggregates the powder.
53. The method of claim 51 , further comprising wetting the powder and smearing the powder to a consistency of fibrous paste, and wherein compacting the at least partially demineralized bone matrix comprises compacting the fibrous paste.
54. The method of claim 53, further comprising molding the fibrous paste.
55. The method of claim 54, wherein the molded fibrous paste is dried at generally ambient temperature.
56. The method of claim 54, wherein molding is done by putting the fibrous paste in a mold and putting the mold under pressure.
57. The method of claim 56, further comprising vacuum drying the molded fibrous paste.
58. The method of claim 1 , wherein treating the at least partially demineralized bone matrix marks the partially demineralized bone matrix for repair.
59. The method of claim 1 , wherein treating the at least partially demineralized bone matrix alters integrin binding sites of the at least partially demineralized bone matrix.
60. The method of claim 1 , wherein treating the at least partially demineralized bone matrix substantially exposes native inductive materials in the at least partially demineralized bone matrix.
61. The method of claim 1 , further comprising adding at least one inductive material to the treated at least partially demineralized bone matrix.
62. The method of claim 1 , further comprising treating the at least partially demineralized bone matrix a second time to impart or enhance osteoinductive activity of the at least partially demineralized bone matrix.
63. The method of claim 62, further comprising washing the at least partially demineralized bone matrix between the first and second treatments.
64. The method of claim 63, wherein the second treatment is concurrent with the first treatment.
65. The method of claim 63, wherein the second treatment is after the first treatment.
66. The method of claim 1 , wherein providing bone matrix comprises providing corticocancellous bone.
67. The method of claim 1, wherein providing bone matrix comprises providing cortical bone.
68. The method of claim 1, wherein providing bone matrix comprises providing composite bone.
69. The method of claim 1 , wherein at least partially demineralizing the bone matrix comprises substantially fully demineralizing the bone matrix.
70. The method of claim 1 , wherein at least partially demineralizing the bone matrix comprises surface demineralizing the bone matrix.
71. The method of claim 1 , further comprising adding a bioactive agent to the treated at least partially demineralized bone matrix.
72. The method of claim 1 , wherein treating the at least partially demineralized bone matrix alters one or more structural components of the at least partially demineralized bone matrix.
73. An osteoinductive composition comprising: at least partially demineralized cancellous bone, the at least partially demineralized cancellous bone having been treated to increase the osteoinductive activity of the bone; wherein the at least partially demineralized cancellous bone exhibits increased osteoinductive activity when compared to untreated at least partially demineralized cancellous bone.
74. The osteoinductive composition of claim 73, wherein the composition has a histologic osteoinductivity score of at least 1 on a 4 point scale.
75. The osteoinductive composition of claim 73, wherein the composition induces bone formation in higher order species.
76. The osteoinductive composition of claim 73, wherein the composition exhibits a solid structure at room temperature and substantially liquefies when implanted in a body.
77. The osteoinductive composition of claim 73, wherein the composition has enhanced solubility when compared to solubility of untreated at least partially demineralized bone matrix.
78. The osteoinductive composition of claim 73, wherein trabecular structure of the treated at least partially demineralized cancellous bone is compacted when compared to trabecular structure of untreated at least partially demineralized cancellous bone.
79. The osteoinductive composition of claim 73, wherein the at least partially demineralized cancellous bone exhibits a trabecular density that is increased when compared to untreated at least partially demineralized cancellous bone.
80. The osteoinductive composition of claim 73, wherein collagen of the treated at least partially demineralized cancellous bone is disrupted when compared to collagen of untreated at least partially demineralized cancellous bone.
81. The osteoinductive composition of claim 73, wherein a portion of collagen present in the treated at least partially demineralized cancellous bone is present as collagen fragments.
82. The osteoinductive composition of claim 73, wherein the at least partially demineralized cancellous bone is compacted and wherein the composition retains its shape in water for a predetermined period of time.
83. The osteoinductive composition of claim 73, wherein the at least partially demineralized cancellous bone is particulated and reformed into an implant.
84. The osteoinductive compositon of claim 83, wherein the implant further comprises a carrier.
85. The osteoinductive composition of claim 84, wherein the carrier is a polymer and wherein the cancellous bone is treated after blending with the polymer.
86. The osteoinductive composition of claim 73, wherein the at least partially demineralized cancellous bone is compacted.
87. The osteoinductive composition of claim 73, further comprising at least partially demineralized cortical bone that has been treated to increase the osteoinductive activity of the bone.
88. The osteoinductive composition of claim 73, further comprising at least partially demineralized corticocancellous bone that has been treated to increase the osteoinductive activity of the bone.
89. The osteoinductive composition of claim 73, further comprising a bioactive agent.
90. The osteoinductive composition of claim 73, wherein native inductive materials of the at least partially demineralized bone matrix are substantially exposed.
91. The osteoinductive composition of claim 73, further comprising at least one inductive material.
92. The osteoinductive composition of claim 73, wherein one or more structural components of the treated at least partially demineralized bone matrix is altered.
93. A method for treating a bone condition, the method comprising: providing bone matrix; at least partially demineralizing the bone matrix; treating the at least partially demineralized bone matrix to impart or enhance osteoinductive activity of the at least partially demineralized bone matrix; forming the treated at least partially demineralized bone matrix into an implant; and implanting the implant.
94. The method of claim 93, wherein the method is used to treat a metabolic bone disease, for bone healing, for cartilage repair, for spinal disc repair, for tendon repair, for repair of a defect created by disease or surgery, or for dural repair.
95. The method of claim 93, wherein the treated implant is at least partially resorbed after implantation.
96. The method of claim 93, wherein forming the treated at least partially demineralized bone matrix into an implant includes configuring the implant.
97. The method of claim 95, wherein configuring is done under compression.
98. The method of claim 93, wherein the implant expands after implantation.
99. The method of claim 93, wherein the osteoimplant has a solid structure at room temperature but substantially liquifies in the body.
100. The method of claim 93, wherein a portion of collagen present in the treated at least partially demineralized bone is present as collagen fragments.
101. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with a collagenase preparation.
102. The method of claim 101, wherein the at least partially demineralized bone matrix is substantially completely solubilized by the collagenase preparation.
103. The method of claim 101, wherein the at least partially demineralized bone matrix is partially solubilized by the collagenase preparation.
104. The method of claim 103, further comprising treating the partially solubilized at least partially demineralized bone matrix to disrupt cross-links not disrupted by the collagenase preparation.
105. The method of claim 104, wherein further treatment comprises treatment with heat.
106. The method of claim 104, wherein further treatment comprises treatment with pepsin.
107. The method of claim 101, wherein the collagenase preparation is highly purified.
108. The method of claim 101, wherein the collagenase preparation is substantially free of bacterial components.
109. The method of claim 101, wherein the collagenase preparation further comprises at least one protease inhibitor.
110. The method of claim 93, wherein providing bone matrix comprises providing cancellous bone.
111. The method of claim 110, wherein treating the at least partially demineralized bone matrix comprises compacting the at least partially demineralized cancellous bone and wherein compacting the at least partially demineralized cancellous bone may form the treated at least partially demineralized bone matrix into the implant.
112. The method of claim 111, wherein the at least partially demineralized cancellous bone is compacted mechanically.
113. The method of claim 111, wherein the at least partially demineralized cancellous bone is compacted by chemical modification of a structure of the at least partially demineralized cancellous bone.
114. The method of claim 111, wherein compacting the at least partially demineralized cancellous bone comprises loading the at least partially demineralized cancellous bone matrix in a constrained compression.
115. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises altering a structure of the at least partially demineralized bone such that trabecular density is increased while collagen structure is decreased.
116. The method of claim 93 , wherein treating the at least partially demineralized bone matrix comprises opening up a structure of the at least partially demineralized bone matrix and breaking apart collagen within the at least partially demineralized bone matrix.
117. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with an enzyme.
118. The method of claim 117, wherein treating the at least partially demineralized bone matrix with an enzyme comprises treating the at least partially demineralized bone matrix with a protease.
119. The method of claim 118, wherein treating the at least partially demineralized bone matrix with a protease comprises treating the at least partially demineralized bone matrix with bone morphogenetic protein- 1.
120. The method of claim 117, wherein treating the at least partially demineralized bone matrix with an enzyme comprises treating the at least partially demineralized bone matrix with pepsin.
121. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with a chemical agent.
122. The method of claim 121, wherein the chemical agent is cyanogen bromide.
123. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with a cleaving agent.
124. The method of claim 23, wherein treating the at least partially demineralized bone matrix with a cleaving agent cleaves an inhibitory factor that inhibits the biological activity of the at least partially demineralized bone matrix.
125. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with heat.
126. The method of claim 125, wherein treating the at least partially demineralized bone matrix further comprises treatment with cold.
127. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with cold.
128. The method of claim 93, wherein treating the at least partially demineralized bone matrix inhibits synthesis of an agent that inhibits the biological activity of the partially demineralized bone matrix.
129. The method of claim 93, wherein treating the at least partially demineralized bone matrix inhibits secretion of an agent that inhibits the biological activity of the partially demineralized bone matrix.
130. The method of claim 93, wherein treating the at least partially demineralized bone matrix inhibits post-translational modification of an agent that inhibits the biological activity of the partially demineralized bone matrix.
131. The method of claim 93 , wherein treating the at least partially demineralized bone matrix inhibits transport of an agent that inhibits the biological activity of the partially demineralized bone matrix.
132. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with ionizing radiation.
133. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with electromagnetic radiation.
134. The method of claim 93, wherein treating the at least partially demineralized bone matrix comprises compacting the at least partially demineralized bone matrix.
135. The method of claim 134, wherein compacting the at least partially demineralized bone matrix assists in the forming of the treated at least partially demineralized bone matrix into the implant.
136. The method of claim 134, wherein compacting the at least partially demineralized bone matrix comprises treating the at least partially demineralized bone matrix with a chemical.
137. The method of claim 134, wherein the at least partially demineralized bone matrix is compacted mechanically.
138. The method of claim 134, wherein the at least partially demineralized bone matrix is compacted with heat.
139. The method of claim 93, further comprising particulating the at least partially demineralized bone matrix and wherein treating the at least partially demineralized bone matrix comprises compacting the particulated at least partially demineralized bone matrix.
140. The method of claim 139, wherein the at least partially demineralized bone matrix is particulated to a generally powder consistency.
141. The method of claim 140, further comprising wetting the powder and smearing the powder to a consistency of fibrous paste, and wherein compacting the at least partially demineralized bone matrix comprises compacting the fibrous paste.
142. The method of claim 141, further comprising molding the fibrous paste into the implant.
143. The method of claim 142, wherein molding is done by putting the fibrous paste in a mold and putting the mold under pressure.
144. The method of claim 93, wherein treating the at least partially demineralized bone matrix substantially exposes native inductive materials in the at least partially demineralized bone matrix.
145. The method of claim 93, further comprising adding at least one inductive material to the treated at least partially demineralized bone matrix.
146. The method of claim 93, further comprising treating the at least partially demineralized bone matrix a second time to impart or enhance osteoinductive activity of the at least partially demineralized bone matrix.
147. The method of claim 146, further comprising washing the at least partially demineralized bone matrix between the first and second treatments.
148. The method of claim 93 , wherein providing bone matrix comprises providing corticocancellous bone.
149. The method of claim 93, wherein providing bone matrix comprises providing cortical bone.
150. The method of claim 93, wherein providing bone matrix comprises providing composite bone.
151. The method of claim 93, wherein at least partially demineralizing the bone matrix comprises substantially fully demineralizing the bone matrix.
152. The method of claim 93, wherein at least partially demineralizing the bone matrix comprises surface demineralizing the bone matrix.
153. The method of claim 93, further comprising adding a bioactive agent to the treated at least partially demineralized bone matrix.
154. The method of claim 93, wherein treating the at least partially demineralized bone matrix alters one or more structural components of the at least partially demineralized bone matrix.
155. An osteoinductive osteoimplant comprising: at least partially demineralized bone matrix; wherein the at least partially demineralized bone matrix has been treated to impart or enhance osteoinductive activity of the at least partially demineralized bone matrix; and wherein the treated at least partially demineralized bone matrix is part of an implant for implantation into the body.
156. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized bone matrix is at least partially demineralized cancellous bone.
157. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized cancellous bone is compacted.
158. The osteoinductive osteoimplant of claim 157, wherein the at least partially demineralized cancellous bone is particulated and compacted.
159. The osteoinductive osteoimplant of claim 158, wherein the at least partially demineralized cancellous bone is particulated, wet, compacted, and dried.
160. The osteoinductive osteoimplant of claim 158, wherein the at least partially demineralized cancellous bone is molded.
161. The osteoinductive osteoimplant of claim 157, wherein the compacted at least partially demineralized cancellous bone retains its shape in water for one or more hours.
162. The osteoinductive osteoimplant of claim 155, wherein the osteoimplant is able to expand and substantially conform to an in vivo site.
163. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized bone matrix is at least partially demineralized cortical bone.
164. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized bone matrix is at least partially demineralized corticocancellous bone.
165. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized bone matrix is at least partially demineralized composite bone.
166. The osteoinductive osteoimplant of claim 155, wherein the osteoimplant has a solid structure at room temperature but substantially liquifies in the body.
167. The osteoinductive osteoimplant of claim 155, wherein the osteoimplant has enhanced solubility when compared to untreated at least partially demineralized bone matrix.
168. The osteoinductive osteoimplant of claim 155, wherein a portion of collagen present in the treated at least partially demineralized bone matrix is present as collagen fragments.
169. The osteoinductive osteoimplant of claim 155, further comprising at least one bioactive agent wherein the osteoimplant comprises a delivery device to administer the at least one bioactive agent.
170. The osteoinductive osteoimplant of claim 169, wherein the treatment alters release kinetics of the bioactive agent.
171. The osteoinductive osteoimplant of claim 155, further comprising at least one osteoinductive factor.
172. The osteoinductive osteoimplant of claim 155, further comprising a radiopaque substance.
173. The osteoinductive osteoimplant of claim 155, wherein trabecular structure of the treated at least partially demineralized bone matrix is compacted when compared to trabecular structure of untreated at least partially demineralized bone matrix.
174. The osteoinductive osteoimplant of claim 155, wherein the bone exhibits increased trabecular density.
175. The osteoinductive osteoimplant of claim 155, wherein collagen of the treated bone is disrupted when compared to collagen of untreated demineralized cancellous bone.
176. The osteoinductive osteoimplant of claim 155, wherein integrin binding sites of the at least partially demineralized bone matrix are altered.
177. The osteoinductive osteoimplant of claim 155, wherein native inductive materials of the at least partially demineralized bone matrix are substantially exposed.
178. The osteoinductive osteoimplant of claim 155, further comprising at least one inductive material.
179. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized bone matrix is substantially fully demineralized.
180. The osteoinductive osteoimplant of claim 155, wherein the at least partially demineralized bone matrix is surface demineralized.
181. The osteoinductive osteoimplant of claim 155, wherein the osteoimplant is a structural implant.
182. The osteoinductive osteoimplant of claim 155, wherein the osteoimplant is an osteogenic implant.
183. An expandable osteoimplant comprising: demineralized cancellous bone particles, at least a portion of the demineralized cancellous bone particles having been treated to increase the osteoinductive activity of the bone particles; wherein the osteoimplant exhibits a first state and an expanded state, the expanded state being achieved upon implantation of the osteoimplant.
184. The osteoimplant of claim 183, wherein the demineralized cancellous bone particles are formed into an implant having an increased density of demineralized cancellous bone when compared to implants formed of non-parti culate demineralized cancellous bone.
PCT/US2008/067117 2007-06-15 2008-06-16 Osteoinductive demineralized cancellous bone WO2008157492A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US94441707P 2007-06-15 2007-06-15
US60/944,417 2007-06-15
US98684307P 2007-11-09 2007-11-09
US60/986,843 2007-11-09

Publications (2)

Publication Number Publication Date
WO2008157492A2 true WO2008157492A2 (en) 2008-12-24
WO2008157492A3 WO2008157492A3 (en) 2009-12-10

Family

ID=39968001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/067117 WO2008157492A2 (en) 2007-06-15 2008-06-16 Osteoinductive demineralized cancellous bone

Country Status (2)

Country Link
US (2) US8734525B2 (en)
WO (1) WO2008157492A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011017284A3 (en) * 2009-08-03 2011-05-05 Osteotech, Inc. Bone matrix compositions and methods
WO2013079443A1 (en) * 2011-12-01 2013-06-06 Antonis Alexakis Regeneration aid for bone defects
WO2017132038A1 (en) * 2016-01-28 2017-08-03 Warsaw Orthopedic, Inc. Electron beam irradiated osteoinductive bone implant

Families Citing this family (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
CN101365499A (en) * 2005-11-01 2009-02-11 骨骼技术股份有限公司 Bone matrix compositions and methods
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
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
ES2446544T3 (en) * 2007-10-19 2014-03-10 Warsaw Orthopedic, Inc. Demineralized bone matrix compositions and methods
US9192695B2 (en) 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
US9101475B2 (en) 2009-02-12 2015-08-11 Warsaw Orthopedic, Inc. Segmented delivery system
CN102781487B (en) 2009-12-13 2016-11-16 阿米特·普拉卡什·戈维 Biological activity graft and complex
US9486500B2 (en) 2010-01-28 2016-11-08 Warsaw Orthopedic, Inc. Osteoimplant and methods for making
US9050274B2 (en) * 2010-01-28 2015-06-09 Warsaw Orthopedic, Inc. Compositions and methods for treating an intervertebral disc using bulking agents or sealing agents
US9125902B2 (en) 2010-01-28 2015-09-08 Warsaw Orthopedic, Inc. Methods for treating an intervertebral disc using local analgesics
US20110189253A1 (en) * 2010-01-29 2011-08-04 Warsaw Orthopedic, Inc. Biomaterial composition and method
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
WO2012061024A1 (en) * 2010-10-25 2012-05-10 Musculoskeletal Transplant Foundation Demineralized cortical bone implants
US8920511B2 (en) 2011-11-17 2014-12-30 Allosource Multi-piece machine graft systems and methods
US9162011B2 (en) 2011-12-19 2015-10-20 Allosource Flowable matrix compositions 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
US8771368B2 (en) 2012-04-24 2014-07-08 William F. McKay Interspinous bone implant device
US9220608B2 (en) * 2012-04-24 2015-12-29 Warsaw Orthopedic, Inc. Facet joint 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
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
AU2013342255B2 (en) * 2012-11-08 2017-05-04 Smith & Nephew, Inc. Methods and compositions suitable for improved reattachment of detached cartilage to subchondral bone
AU2013342257B2 (en) 2012-11-08 2018-08-30 Smith & Nephew, Inc. Improved reattachment of detached cartilage to subchondral bone
KR102258565B1 (en) 2012-11-15 2021-05-28 알로소스 Minced cartilage systems and methods
US9265609B2 (en) 2013-01-08 2016-02-23 Warsaw Orthopedic, Inc. Osteograft implant
CA2894750C (en) 2013-02-22 2021-12-14 Allosource Cartilage mosaic compositions and methods
AU2014225458A1 (en) 2013-03-07 2015-07-09 Allosource Consistent calcium content bone allograft systems and methods
EP2967874B1 (en) 2013-03-15 2019-11-20 AlloSource Perforated osteochondral allograft compositions
CA2899713C (en) 2013-03-15 2022-07-19 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
WO2014151091A2 (en) 2013-03-15 2014-09-25 Theracell, Inc. Compositions of and methods for cancellous bone matrix
US9486483B2 (en) 2013-10-18 2016-11-08 Globus Medical, Inc. Bone grafts including osteogenic stem cells, and methods relating to the same
US9539286B2 (en) 2013-10-18 2017-01-10 Globus Medical, Inc. Bone grafts including osteogenic stem cells, and methods relating to the same
US9913929B2 (en) 2013-10-18 2018-03-13 Fortus Medical, Inc. Bone marrow aspirate enhanced bone graft
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
US9730796B2 (en) 2014-05-16 2017-08-15 Allosource Composite bone constructs and methods
US9610143B2 (en) * 2014-06-19 2017-04-04 Osteolife Biomedical, Llc Compressed decalcified trabecular bone grafts and tooth socket repair
US10279078B2 (en) * 2014-12-31 2019-05-07 Bacterin International, Inc. Crosslinkable 3D printed biomaterial-based implants and methods of manufacture thereof
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
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using 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
US9788950B1 (en) * 2016-04-22 2017-10-17 Vivex Biomedical, Inc. Cohesive bone composition
US20170319626A1 (en) * 2016-05-09 2017-11-09 Warsaw Orthopedic, Inc. Osteoinductive fibrous bone chips
US10688222B2 (en) 2016-11-21 2020-06-23 Warsaw Orthopedic, Inc. Lyophilized moldable implants containing an oxysterol
WO2018232126A1 (en) * 2017-06-14 2018-12-20 Fabrico Technology Inc. Decellularized and demineralized bone matrices and methods for making same
US11452796B2 (en) 2017-06-30 2022-09-27 Allosource Cellular bone grafts, and methods of manufacture and use
US10966816B1 (en) 2019-10-18 2021-04-06 Sparta Biopharma LLC Connective tissue to bone interface scaffolds
US11896736B2 (en) 2020-07-13 2024-02-13 Globus Medical, Inc Biomaterial implants and methods of making the same
US11364324B2 (en) 2020-08-24 2022-06-21 Sparta Biopharma Inc. Methods of forming bone interface scaffolds

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5073373A (en) * 1989-09-21 1991-12-17 Osteotech, Inc. Flowable demineralized bone powder composition and its use in bone repair
WO2005065396A2 (en) * 2003-12-31 2005-07-21 Osteotech, Inc. Improved bone matrix compositions and methods
WO2007053850A2 (en) * 2005-11-01 2007-05-10 Osteotech, Inc. Bone matrix compositions and methods

Family Cites Families (181)

* 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
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
US4294753A (en) 1980-08-04 1981-10-13 The Regents Of The University Of California Bone morphogenetic protein process
US4619989A (en) 1981-05-05 1986-10-28 The Regents Of The University Of Cal. Bone morphogenetic protein composition
US4761471A (en) 1980-08-04 1988-08-02 The Regents Of The University Of California Bone morphogenetic protein composition
US4430760A (en) * 1981-12-18 1984-02-14 Collagen Corporation Nonstress-bearing implantable bone prosthesis
US4394370A (en) * 1981-09-21 1983-07-19 Jefferies Steven R Bone graft material for osseous defects and method of making same
US4472840A (en) 1981-09-21 1984-09-25 Jefferies Steven R Method of inducing osseous formation by implanting bone graft material
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
US4789663A (en) 1984-07-06 1988-12-06 Collagen Corporation Methods of bone repair using collagen
US4843063A (en) * 1984-07-16 1989-06-27 Collagen Corporation Polypeptide cartilage-inducing factors found in bone
DE3588058T3 (en) 1984-07-16 2005-04-07 Celtrix Pharmaceuticals, Inc., Palo Alto Cartilage-inducing polypeptide factors from bone
US4657548A (en) * 1984-09-11 1987-04-14 Helitrex, Inc. Delivery system for implantation of fine particles in surgical procedures
US4563350A (en) * 1984-10-24 1986-01-07 Collagen Corporation Inductive collagen based bone repair preparations
US4888366A (en) 1984-10-24 1989-12-19 Collagen Corporation Inductive collagen-based bone repair preparations
US4678470A (en) * 1985-05-29 1987-07-07 American Hospital Supply Corporation Bone-grafting 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
IL83003A (en) 1986-07-01 1995-07-31 Genetics Inst Osteoinductive factors
US5106748A (en) * 1986-07-01 1992-04-21 Genetics Institute, Inc. Dna sequences encoding 5 proteins
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
US5354557A (en) 1988-04-08 1994-10-11 Stryker Corporation Osteogenic devices
US5266683A (en) 1988-04-08 1993-11-30 Stryker Corporation Osteogenic proteins
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
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
US5290763A (en) * 1991-04-22 1994-03-01 Intermedics Orthopedics/Denver, Inc. Osteoinductive protein mixtures and purification processes
US5563124A (en) 1991-04-22 1996-10-08 Intermedics Orthopedics/ Denver, Inc. Osteogenic product and process
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
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
US5507813A (en) * 1993-12-09 1996-04-16 Osteotech, Inc. Shaped materials derived from elongate bone particles
US5723012A (en) * 1993-12-09 1998-03-03 Bioland Uses for a current of supercritical carbon dioxide as an antiviral agent
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
US7045141B2 (en) * 1998-02-27 2006-05-16 Musculoskeletal Transplant Foundation Allograft bone composition having a gelatin binder
US6030635A (en) * 1998-02-27 2000-02-29 Musculoskeletal Transplant Foundation Malleable paste for filling bone defects
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
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
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
US6696073B2 (en) * 1999-02-23 2004-02-24 Osteotech, Inc. Shaped load-bearing osteoimplant and methods of making same
US6294187B1 (en) 1999-02-23 2001-09-25 Osteotech, Inc. Load-bearing osteoimplant, method for its manufacture and method of repairing bone using 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
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
DE60121017T2 (en) 2000-10-24 2007-01-11 SDGI Holdings, Inc., Wilmington DEVICE AND METHOD FOR BONE CONSTRUCTION FULL PLUG
US6752831B2 (en) 2000-12-08 2004-06-22 Osteotech, Inc. Biocompatible osteogenic band for repair of spinal disorders
EP1341484B1 (en) * 2000-12-08 2009-05-06 Osteotech, Inc. Implant for orthopedic applications
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
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
US7241874B2 (en) * 2002-06-26 2007-07-10 Zimmer Ortho Biologics, 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
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
US20050251267A1 (en) 2004-05-04 2005-11-10 John Winterbottom Cell permeable structural implant
EP1578957B1 (en) * 2002-12-12 2015-04-15 Warsaw Orthopedic, Inc. Formable and settable polymer bone composite and method of production thereof
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
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
CA2554571A1 (en) 2004-01-27 2005-08-11 Osteotech, Inc. Stabilized bone graft
US7678385B2 (en) 2004-04-28 2010-03-16 Biomet Manufacturing Corp. Irradiated implantable bone material
US20050244450A1 (en) 2004-04-28 2005-11-03 Reddi A H Heat-treated implantable bone material
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
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
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

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5073373A (en) * 1989-09-21 1991-12-17 Osteotech, Inc. Flowable demineralized bone powder composition and its use in bone repair
WO2005065396A2 (en) * 2003-12-31 2005-07-21 Osteotech, Inc. Improved bone matrix compositions and methods
WO2007053850A2 (en) * 2005-11-01 2007-05-10 Osteotech, Inc. Bone matrix compositions and methods

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MALENE LAURSEN ET AL.: "Optimal handling of fresh cancellous bone graft-Different peroperative storing techniques evaluated by in vitro osteobalst-like cell metabolism" ACTA ORTHOP SCAND, vol. 74, no. 4, 2003, XP002550487 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011017284A3 (en) * 2009-08-03 2011-05-05 Osteotech, Inc. Bone matrix compositions and methods
EP3342432A1 (en) * 2009-08-03 2018-07-04 Warsaw Orthopedic, Inc. Bone matrix compositions and methods
WO2013079443A1 (en) * 2011-12-01 2013-06-06 Antonis Alexakis Regeneration aid for bone defects
CN104136051A (en) * 2011-12-01 2014-11-05 安东尼·阿莱克斯 Regeneration aid for bone defects
WO2017132038A1 (en) * 2016-01-28 2017-08-03 Warsaw Orthopedic, Inc. Electron beam irradiated osteoinductive bone implant
CN108495660A (en) * 2016-01-28 2018-09-04 华沙整形外科股份有限公司 The osteoinductive bone implantation material of electron beam irradiation
US11090411B2 (en) 2016-01-28 2021-08-17 Warsaw Orthopedic, Inc. Electron beam irradiated osteoinductive bone implant
CN108495660B (en) * 2016-01-28 2022-03-22 华沙整形外科股份有限公司 Electron beam irradiated osteoinductive bone implant
US11918705B2 (en) 2016-01-28 2024-03-05 Warsaw Orthopedic, Inc. Electron beam irradiated osteoinductive bone implant

Also Published As

Publication number Publication date
US9415136B2 (en) 2016-08-16
US8734525B2 (en) 2014-05-27
US20090155378A1 (en) 2009-06-18
WO2008157492A3 (en) 2009-12-10
US20140255506A1 (en) 2014-09-11
US20140065239A9 (en) 2014-03-06

Similar Documents

Publication Publication Date Title
US9415136B2 (en) Osteoinductive demineralized cancellous bone
US10357511B2 (en) Bone matrix compositions and methods
US10328179B2 (en) Bone matrix compositions and methods
EP1701729B1 (en) Improved bone matrix compositions and methods
WO2009052492A2 (en) Demineralized bone matrix compositions and methods

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08771188

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08771188

Country of ref document: EP

Kind code of ref document: A2